Regulation of the Hypothalamic-Pituitary

PHYSIOLOGICAL REVIEWS
Vol. 79, No. 1, January 1999
Printed in U.S.A.
Regulation of the Hypothalamic-Pituitary-Adrenal Axis by
Cytokines: Actions and Mechanisms of Action
ANDREW V. TURNBULL AND CATHERINE L. RIVIER
The Clayton Foundation Laboratories for Peptide Biology, The Salk Institute, La Jolla, California; and North
Western Injury Research Centre, University of Manchester, Manchester, United Kingdom
2
2
2
5
8
10
10
12
13
13
14
16
17
18
18
Downloaded from on April 23, 2014
I. Introduction
A. Hormones and cytokines: definitions
B. Concept of bidirectional communication between immune and neuroendocrine systems:
a historical perspective
C. Cytokines
D. Hypothalamic-pituitary-adrenal axis
II. Cytokine Influence on Hypothalamic-Pituitary-Adrenal Axis Secretory Activity In Vivo
A. Animal studies
B. Human studies
III. Physiological/Pathophysiological Circumstances in Which Endogenous Cytokines Play a Role in
Regulation of Hypothalamic-Pituitary-Adrenal Axis
A. Viral disease
B. Endotoxin treatment
C. Local inflammation
D. Physical and psychological stress
E. Basal hypothalamic-pituitary-adrenal activity
IV. Cytokine Actions on the Central Nervous System, Pituitary, and Adrenal
A. Evidence that cytokines activate the hypothalamic-pituitary-adrenal axis primarily at the level of the
central nervous system
B. Evidence for direct effects of cytokines on pituitary adrenocorticotropic hormone secretion
C. Evidence for direct actions of cytokines on adrenal glucocorticoid secretion
V. Mechanisms of Hypothalamic-Pituitary-Adrenal Axis Activation by Interleukin-1
A. Direct actions on pituitary and adrenal
B. Penetration of cytokines into brain
C. Role of readily diffusible intermediates
D. Induction of intermediates at blood-brain barrier interface
E. Actions at circumventricular organs
F. Potential role of catecholamines and evidence of activation of medullary cell groups
G. Activation of vagal afferent fibers
H. Cytokine synthesis within brain
I. Local interleukin-1 induction of circulating interleukin-6
VI. Conclusions
19
26
30
31
31
32
33
36
37
39
40
41
43
43
Turnbull, Andrew V., and Catherine L. Rivier. Regulation of the Hypothalamic-Pituitary-Adrenal Axis by Cytokines: Actions and Mechanisms of Action. Physiol. Rev. 79: 1–71, 1999.—Glucocorticoids are hormone products
of the adrenal gland, which have long been recognized to have a profound impact on immunologic processes. The
communication between immune and neuroendocrine systems is, however, bidirectional. The endocrine and immune
systems share a common ‘‘chemical language,’’ with both systems possessing ligands and receptors of ‘‘classical’’
hormones and immunoregulatory mediators. Studies in the early to mid 1980s demonstrated that monocyte-derived
or recombinant interleukin-1 (IL-1) causes secretion of hormones of the hypothalamic-pituitary-adrenal (HPA) axis,
establishing that immunoregulators, known as cytokines, play a pivotal role in this bidirectional communication
between the immune and neuroendocrine systems. The subsequent 10–15 years have witnessed demonstrations
that numerous members of several cytokine families increase the secretory activity of the HPA axis. Because this
neuroendocrine action of cytokines is mediated primarily at the level of the central nervous system, studies investigating the mechanisms of HPA activation produced by cytokines take on a more broad significance, with findings
relevant to the more fundamental question of how cytokines signal the brain. This article reviews published findings
that have documented which cytokines have been shown to influence hormone secretion from the HPA axis,
0031-9333/99 $15.00 Copyright q 1999 the American Physiological Society
/ 9j0c$$oc11
P13-8
11-25-98 11:16:36
1
pra
APS-Phys Rev
2
ANDREW V. TURNBULL AND CATHERINE L. RIVIER
Volume 79
determined under what physiological/pathophysiological circumstances endogenous cytokines regulate HPA axis
activity, established the possible sites of cytokine action on HPA axis hormone secretion, and identified the potential
neuroanatomic and pharmacological mechanisms by which cytokines signal the neuroendocrine hypothalamus.
I. INTRODUCTION
A. Hormones and Cytokines: Definitions
/ 9j0c$$oc11
P13-8
B. Concept of Bidirectional Communication
Between Immune and Neuroendocrine Systems:
a Historical Perspective
Regulation of the immune system by the adrenal
gland was observed as early as the middle of the 19th
century when Thomas Addison (3) documented that a
patient with adrenal insufficiency had an excess of circulating lymphocytes. In agreement with this observation,
removal of the adrenal gland of the rat was found to produce hypertrophy of the thymus (an organ responsible for
the manufacture of mature lymphocytes) (363). Perhaps
the best known of early experimental studies were those
of Hans Selye (764, 765), who found that enlargement
of the adrenal gland and involution of the thymus were
communal features of an animal’s response to stress, regardless of the nature of the injurious insult. These early
studies clearly suggested a close association between adrenal gland physiology and immune activity.
The isolation of the active principal of the adrenal
cortex, cortisone, by Kendall and Reichstein in the late
11-25-98 11:16:36
pra
APS-Phys Rev
Downloaded from on April 23, 2014
Defining what is meant by the term hormone, and
what is meant by the term cytokine, is certainly no easy
task. Indeed, the work reviewed in this article has changed
many people’s opinion as to what our definitions of these
classes of cell-cell signaling molecules should be. However, we are faced at the outset with conveying to the
reader what ‘‘we’’ mean when we use the terms cytokines
and hormones throughout this review.
A classical endocrinology textbook definition of a
hormone is ‘‘a biomolecule, which is produced by a specialized cell type, is secreted from a ductless gland directly into the bloodstream, and acts on distant target
cells/tissues, to regulate pre-existing cellular activities.’’
Chemically, hormones are small to large polypeptides,
proteins, glycoproteins, derivatives of aromatic amino
acids, or steroids. In the case of the pituitary peptide
hormone adrenocorticotropin (ACTH), it is produced by
corticotropes (specialized cell type) within the anterior
pituitary (a ductless gland), is secreted into the general
circulation, and acts predominantly on the adrenal cortex
(a distant target) to enhance glucocorticoid secretion.
This is perhaps a narrow view of a hormone, because
many ‘‘classical’’ hormones are also synthesized and act
within the brain and are produced and act locally within
the periphery. On the other hand, it is perhaps too broad,
because under this definition CO2 produced by exercising
muscle and stimulating respiration might have to be considered a hormone also. However, it is a fairly accurate
description of what most people understand when thinking of a hormone acting in a ‘‘classical endocrine fashion.’’
In contrast to hormones that have most commonly
been associated with the ‘‘endocrine system,’’ cytokines
have been classically associated with the ‘‘immune system.’’ Defining a cytokine is even more difficult than defining a hormone. For the purposes of the work described
within this review, we found the definition used in The
Cytokine Handbook (862) most useful. Here cytokines are
defined as ‘‘regulatory proteins secreted by white blood
cells and a variety of other cells in the body; the pleiotropic actions of cytokines include numerous effects on cells
of the immune system and modulation of inflammatory
responses.’’ This definition is somewhat narrow in that it
probably overemphasizes the importance of the immune
system as a source and target, but probably reflects most
accurately people’s first thoughts when they think of cyto-
kines. It certainly reflects best the definition that would
have been applied at the time when the majority of the
work described in this article was performed. Under any
definition, the term cytokine encompasses the ‘‘monokines’’ (monocyte/macrophage-derived mediators) and
‘‘lymphokines’’ (lymphocyte-derived mediators), which
were terms commonly used in the earlier studies described in this review.
Table 1 was compiled of what is presently known of
the features of hormones and cytokines. Although this is
again not definitive, it is fair to say that if the majority of
characteristics of the substance under consideration fit in
the cytokine column, then the substance is a cytokine,
and if the majority of characteristics fit in the hormone
column, then it is a hormone. It should be pointed out
that the same chemical substance could be classified differently depending on the ‘‘setting’’ under consideration.
For example, prolactin produced by the pituitary and acting on the mammary gland is clearly acting in an ‘‘endocrine hormone’’ fashion. However, prolactin can also be
produced by, and act on, lymphocytes, a situation in
which it might be better classified as a cytokine. Perhaps
the key difference between cytokines and hormones that
we indicate in Table 1 is that cytokines are regulators of
predominantly local tissue processes, whereas hormones
function as regulators predominantly of ‘‘systemic’’ or
‘‘whole body’’ homeostasis.
January 1999
TABLE
REGULATION OF HPA AXIS BY CYTOKINES
3
1. Features of cytokines and classical endocrine hormones
Structure
Cell sources
Concentrations in healthy,
stress-free subjects
Location of action relative
to secretion
Range of activities
‘‘Redundancy’’
Cytokine
Classical Endocrine Hormone
Large polypeptides, proteins, or
glycoproteins
Secreted by white blood cells (and many
other cells in numerous types of tissues
and organs)
Very low (virtually absent); increase
markedly during tissue disease, injury, or
repair
Act predominantly locally, in a paracrine or
autocrine manner
‘‘Pleiotropic,’’ multiple target cell types and
broad spectrum of actions
Small to large polypeptides, proteins, glycoproteins, derivatives of
aromatic amino acids, or steroids
Secreted by one type of specialized cell within a ductless gland
(an ‘‘endocrine gland’’)
Display great overlap of biological activities
(i.e., ‘‘redundancy’’)
Function predominantly as regulators of
local tissue processes
Function
Usually measurable and commonly display pulsatile and circadian
patterns of secretion
Act on distant target cells
Breadth of actions highly variable: many anterior pituitary
hormones (e.g., TSH, ACTH) have highly limited actions, but
‘‘target gland’’ hormones (e.g., T3, T4, and glucocorticoids) have
very broad range of activities
Far less overlap of biological activities; deficiency in single
hormone usually produces marked abnormalities
Function predominantly as regulators of systemic or ‘‘whole
body’’ homeostasis
TSH, thyroid-stimulating hormone; ACTH, adrenocorticotropin; T3, 3,3*,5-triiodothyronine; T4, thyroxine.
/ 9j0c$$oc11
P13-8
of an immune response to a foreign antigen (sheep red
blood cells), rats mount a parallel endocrine response
characterized by elevated plasma levels of GC (corticosterone) (73). Furthermore, mice injected with supernatants
from concanavalin A-stimulated peripheral blood or
spleen cells produce one or more GC-increasing factors
(GIF) that increase the blood concentrations of corticosterone (68). These studies therefore suggested the existence of an immune-neuroendocrine regulatory feedback
mechanism in which immune cells limited their own activity by secreting molecules that stimulate the secretion of
adrenal GC.
The concept of ‘‘bidirectional communication’’ between immune and endocrine systems became firmly established with the seminal works of Edwin Blalock and
co-workers in the early 1980s. These workers began to
describe molecular basis for such bidirectional communication (reviewed in Refs. 80–82, 952, 953). Their early
studies showed a commonality in the pathways of action
of immunoregulators (e.g., interferon) and hormones
(e.g., norepinephrine) (84). This group went on to discover that a number of classical hormones are not only
secreted by classical endocrine glands (e.g., pituitary) but
are also made by cells of the immune system (e.g., lymphocytes). For example, they showed that lymphocytes
synthesize ACTH, the pituitary hormone which is the major physiological regulator of adrenal GC secretion (792).
In addition, they demonstrated that not only do lymphocytes produce hormones such as ACTH, endorphins, thyrotropin, and growth hormone (792, 794, 951) but that
these hormones were able to influence immunologic processes (79, 83, 370, 371). Subsequent studies have demonstrated that a number of hormones (e.g., prolactin, insulinlike growth factor) are produced by lymphocytes (372,
556, 557, 621).
11-25-98 11:16:36
pra
APS-Phys Rev
Downloaded from on April 23, 2014
1940s, and the demonstration of its ability to suppress
inflammation (335), gave support to the hypothesis that
adrenal glucocorticoid (GC) secretion plays a significant
role in regulating immunologic processes. However, even
though marked elevations in the plasma blood concentration of GC are observed after all types of stressful stimuli,
studies showing that GC produce immunosuppression
were assumed (by the majority of workers) to be of pharmacological, rather than of physiological, significance.
These findings advented the widespread use of GC-based
therapies for autoimmune and inflammatory disease. It
was not until the late 1970s and the pioneering work of
Besedovsky, del Rey, Sorkin, and colleagues that a physiological role for GC in preventing overactivity, and preserving the specificity, of immune reactions became established (67, 191). In a significant review article of the early
1980s, Munck et al. (567) reinforced this concept. These
authors proposed the now commonly held view that endogenous GC act to prevent ‘‘overshoot’’ of immune/inflammatory responses, thus limiting the host defense response to fighting the aggressor (e.g., invading pathogen)
without the deleterious effects to the host of a hyperactive
immune system (e.g., autoimmunity). More recent work
has indicated that the influence of GC on immunologic
processes is more complex than a generalized suppression of immune activity and depends on the type of immune activity and the subset of immunologic cells involved (see Ref. 531 for extensive review). However, it is
clear that endogenous GC are key regulators of immune
system function.
Further work by Besedovsky et al. (73) suggested
that not only do GC have a profound impact on immune
activity, but that the converse is also true and immune
activity influences GC secretion. This hypothesis grew out
of experimental observations that during the development
4
ANDREW V. TURNBULL AND CATHERINE L. RIVIER
1
Interleukin literally means ‘‘between leukocytes.’’
/ 9j0c$$oc11
P13-8
resulted in articles being published back to back in a 1987
issue of Science (55, 62, 730). One of these studies (62)
demonstrated a direct action of IL-1 on ACTH secretion
from primary cell cultures of rat anterior pituitary cells,
thus supporting the earlier studies by Blalock and coworkers (966) suggesting a direct action of IL-1 on the
pituitary gland to secrete ACTH. Conversely, the other
two Science papers (55, 730) found that IL-1 does not
stimulate ACTH secretion from anterior pituitary cells in
primary culture, despite the fact that IL-1 in vivo elevates
plasma ACTH and GC concentrations. These latter two
groups (55, 730) showed that IL-1-induced ACTH secretion in vivo is dependent on the secretion and action of
the hypothalamic 41-amino acid peptide CRF, which is the
major hypothalamic ACTH secretagogue. These findings
clearly implicate the hypothalamus as the site at which
the HPA axis response to IL-1 is mediated and gave great
support to the idea that immunoregulators could influence
the activity of the central nervous system (CNS).
Controversy over the likely primary site of IL-1 action
(CNS, pituitary gland, or possibly adrenal glands) in stimulating pituitary-adrenal secretion has continued for many
years and is considered in detail in sections IV and V.
However, a large body of evidence has now accumulated
that indicates that IL-1 and other cytokines can signal the
brain. In parallel with studies investigating the relationship between the immune system and HPA axis, a large
number of studies indicated that fever caused by invading
pathogens occurs as a result of the elaboration from immune cells of an ‘‘endogenous pyrogen’’ capable of signaling the CNS (reviewed in Refs. 23, 420). This endogenous
pyrogen was putatively identified as IL-1 (reviewed in Ref.
420). Furthermore, administration of IL-1 produces many
CNS-mediated changes including changes in behavior (reduced exploration, reproductive activity, food-motivated
behavior, and increased sleep), changes in autonomic outflow, metabolic rate, and the activity of a number of neuroendocrine axes (see Refs. 54, 65, 408, 409, 439, 440, 529,
683, 708 for relevant reviews). Collectively, these studies
have provided strong evidence for the regulation by IL-1 of
CNS responses to peripheral changes in immune activity.
Furthermore, the common mediator of the effects on various CNS responses provides a molecular basis for the
observations of the stereotypical responses to immune
challenges of diverse origins. This ‘‘acute phase response’’
to sickness is characterized by fever, appetite suppression, anorexia, alterations in plasma cation concentrations, synthesis of specific liver proteins (known as acute
phase proteins), and changes in neuroendocrine secretions (441). It is now firmly established that acute phase
responses are produced by the actions of, and complex
interactions between, IL-1 and numerous other cytokines.
Since the landmark studies by the groups of Besedovsky and Blalock, it has become apparent that IL-1 has
potent effects on the secretion of the majority of hor-
11-25-98 11:16:36
pra
APS-Phys Rev
Downloaded from on April 23, 2014
The finding that lymphocytes are able to synthesize
an ACTH-like molecule, combined with the demonstration
that mice whose pituitary gland had been removed (hypophysectomy) still produced a corticosterone response
to infection with the Newcastle disease virus (NDV), led
Blalock and co-workers (793) to propose the concept of
lymphoid-adrenal axis. According to this hypothesis,
ACTH produced by virus-stimulated lymphocytes acts on
the adrenal to increase corticosterone secretion. However, subsequent studies have failed to replicate the persistance of an NDV-induced corticosterone response in
hypophysectomized mice (64, 218, 221, 604), and the hypothesis of a lymphoid-adrenal axis involving lymphoid
production of ACTH molecule has fallen out of favor.
Furthermore, Besedovsky et al. (71) showed that stimulated lymphocytes secrete GIF that increase plasma ACTH
and corticosterone levels in rats and that this corticosterone response was prevented by hypophysectomy. Given
that the electrical and neurochemical activities of the hypothalamus are also altered during the course of an immune response (69, 72), Besedovsky et al. (71) proposed
that the effects of such GIF on adrenal GC secretion were
probably mediated at the hypothalamic component of the
hypothalamic-pituitary-adrenal (HPA) axis, rather than on
the pituitary or adrenal glands directly.
The chemical identity of putative ‘‘GIF’’ became apparent with the recognition that classical endocrine hormones are not the only class of mediators involved in
immune-endocrine communication. Indeed, in the mid
1980s, it became readily apparent that immunoregulatory
cytokines also form a key link between immune and neuroendocrine systems (70, 331, 532, 966). Blalock and coworkers (966) showed that the monokines interleukin
(IL)-1 and IL-6 (or hepatocyte-stimulating factor) stimulate ACTH secretion from the corticotropic tumor cell line
AtT20.1 A year later, Besedovsky et al. (70) demonstrated
that systemic administration of monocyte-derived or recombinant IL-1 increases plasma ACTH and GC concentrations in normal mice. Furthermore, Besedovsky et al.
(70) demonstrated that neutralization of endogenous IL1 inhibits the GC response to experimental viral infection
(NDV) in rats. This latter experiment clearly indicated
that the observations of stimulatory effects of cytokines
on neuroendocrine secretion were not merely pharmacological phenomena and suggested that IL-1 plays an important endogenous role in regulating the HPA axis during
viral disease. Indeed, these landmark studies by Besedovsky and Blalock indicated that cytokines could be the
extrahypothalamic corticotropin-releasing factors (CRF)
released by injured tissue which had previously been reported by Broddish and co-workers during the 1970s (112,
113, 497).
Subsequent work by three independent laboratories
Volume 79
January 1999
REGULATION OF HPA AXIS BY CYTOKINES
2. Influence of interleukin-1 on neuroendocrine
secretion
TABLE
Neuroendocrine System
Hypothalamic-pituitaryadrenal axis
Hypothalamic-pituitarygonadal axis
Hypothalamic-pituitarythyroid axis
Somatotropic axis
Prolactin secretion
Posterior pituitary
hormone secretion
Catecholamines
Effect
Reference No.
Increases secretion
863, 883, 959
Decreases secretion
683, 715, 882,
886
336, 358, 665,
886
634, 922
Decreases secretion
Increases or decreases
secretion
Increases or decreases
secretion
Increases secretion
671, 689
158, 159, 446,
992
54, 695
Increases secretion
C. Cytokines
1. Cytokines and cytokine receptor families
Cytokines are large (8–60 kDa), soluble polypeptide
mediators that regulate growth, differentiation, and function of many different cell types (see Table 3). The majority of cytokines have been classically associated with the
regulation of immune and/or inflammatory processes, and
within the immune system, their actions are generally exerted in paracrine or autocrine fashions. However, because of the demonstration that immune, central nervous,
and neuroendocrine systems share a common chemical
language, much more diverse actions of cytokines in host
/ 9j0c$$oc11
P13-8
defense are now recognized. Accordingly, the expression
of these polypeptides and their receptors is not restricted
to cells of the immune system but is also found in many
other tissues (including the brain and endocrine glands).
Furthermore, many cytokines exert potent actions on a
variety of physiological activities outside of immunoregulation; for example, many cytokines induce fever, sleep,
anorexia, malaise, and alterations in neuroendocrine secretions. Finally, the ability of some cytokines to regulate
homeostatic processes at tissues distant from their site
of production has firmly established cytokines as key regulators of coordinated local and systemic responses to
tissue trauma, infection, and disease.
The classification of cytokines into families has
proven somewhat arbitrary. With the exception of a few
homologous peptides (e.g., IL-1a and -1b; interferon-a
and -b; and tumor necrosis factor-a, -b) most cytokines
share little sequence similarity. Consequently, classification of cytokines has been based on either functional attributes, target receptors, or cells of origin. Most commonly,
cytokines have been classified into families of interleukins, tumor necrosis factors (TNF), interferons (IFN),
chemokines, hematopoietins (or neuropoietins), and colony-stimulating factors (CSF). Because of their similar
actions particularly within the CNS and peripheral nervous system, growth factors (GF) and neurotrophins (NT)
have also been considered to fall under the umbrella term
cytokine. Their overlapping actions lead to a number of
cytokines belonging to more than one family (see Table
3). For example, IL-6 is not only an interleukin, but also
a member of a family of either hematopoietic or (neuropoietic) factors that utilize an identical receptor subunit
(gp130) for cell signaling (416). Furthermore, IL-1, IL-3,
IL-5, and IL-6 are also CSF.
One of the striking features of cytokines is their ability to exert many different actions (a property known as
‘‘pleiotropy’’) and, conversely, that many different cytokines exert the same biological actions (a property known
as ‘‘redundancy’’) (162, 633). Cytokine pleiotropy presumably relates to the widespread distribution of cytokine
receptors on numerous cell types and the ability of signal
transduction mechanisms activated by cytokines to alter
expression of a wide variety of target genes. The functional redundancy of various cytokines has, at least partially, been explained by the identification and molecular
cloning of many cytokine receptors. Some, although certainly not all, cytokine receptors consist of a multiunit
complex, including a cytokine-specific ligand binding
component and a ‘‘class’’-specific signal transduction unit
(416, 733). In addition to the gp130 signaling cytokines,
common receptor subunits have also been demonstrated
for IL-2, IL-4, and IL-7 (733) and also for IL-3, IL-5, and
granulocyte-macrophage CSF which share the signal
transduction subunit KH7 (552). However, cytokine redundancy cannot be totally explained by the sharing of
11-25-98 11:16:36
pra
APS-Phys Rev
Downloaded from on April 23, 2014
mones under neuroendocrine control (see Table 2). Furthermore, more recent studies have shown that alterations
in neuroendocrine secretion are produced not only by IL1, but also by many other immunoreglatory cytokines.
The HPA axis has remained the most extensively studied
neuroendocrine system with respect to the influence of
cytokines, and the ability to increase the secretory activity
of this axis is a biological property of several interleukins,
tumor necrosis factors, chemokines, hematopoietins, interferons, growth factors, and neurotrophic factors.
This article reviews published findings that demonstrate 1) which cytokines influence hormone secretion
from the HPA axis, 2) under what physiological/pathophysiological circumstances endogenous cytokines may
influence HPA axis secretory activity, 3) at which level
(hypothalamus, pituitary, or adrenal) cytokines primarily
act, and 4) what anatomic and pharmacological pathways
mediate the actions of cytokines on the neuroendocrine
hypothalamus. To achieve this aim, we have divided this
article into sections corresponding to these overall objectives. We begin by providing brief introductions to relevant aspects of cytokine biology (see sect. IC) and the
functional anatomy of the HPA axis (see sect. ID).
5
6
ANDREW V. TURNBULL AND CATHERINE L. RIVIER
TABLE
Volume 79
3. Cytokine families
Family
Members
Major Ascribed Actions
Interleukins
IL-1 to IL-18
Tumor necrosis factors
TNF-a, TNF-b
Interferons
Chemokines
IFN-a, -b, -g
IL-8/cinc/gro/NAP-1,
MIP-1a, -b, RANTES
IL-6, CNTF, LIF, OM, IL-11,
CT-1
G-CSF, M-CSF, GM-CSF,
SCF, IL-3, IL-5
Hematopoietins (neuropoietins)
Colony stimulating factors
Neurotrophins
NGF, BDNF, GDNF, NT-3,
NT-6
IGF-I, IGF-II, EGF, aFGF,
bFGF, PDGF, TGF-a,
TGF-b, activin
Growth factors
Categorization as an IL does not imply function; IL have numerous and
diverse immunoregulatory actions; some IL have clearly proinflammatory
actions (e.g., IL-1a, IL-1b, IL-8, IL-9), whereas others have antiinflammatory effects (e.g., IL-1ra, IL-4, IL-10, IL-13). Many IL also induce
systemic aspects of acute phase response (e.g., fever)
Tumor cytotoxicity; broad-ranging immunologic activities; induction of many
other cytokines; immunostimulant; proximal mediator of inflammatory
response
Inhibit viral replication; regulation of specificity of immune responses
Chemotaxis; activation of cells at inflammatory sites
All utilize gp130 receptor subunit for signaling; various actions on B cells
and other immunoregulatory actions; promote survival of neurons
Promotion of growth and differentiation of multipotential progenitor cells in
bone marrow; increase numbers of, or enhance activity of, cells of
granulocyte, macrophage, and eosinophil lineages
Neuronal growth and differentiation
Cell growth and differentiation
receptor subunits, since a number of cytokines, for example, IL-1, IL-6, and TNF-a, have many common biological
activities despite the utilization of distinct cell surface
receptors (see Table 4).
Although there is some evidence that cytokines may
play a role in some ‘‘normal’’ physiological processes such
as sleep (605, 606, 838, 839), exercise (138, 139, 815, 920),
and ovulation (2, 137), the expression of most cytokines
in most normal healthy tissues is very low. However, cytokine production increases markedly during ‘‘tissue stress’’
produced by diverse cellular challenges, including periods
4. Shared biological activities of IL-1, IL-6,
and TNF-a
TABLE
Immunologic properties
T-cell activation
B-cell activation
Nonspecific resistance to infection
Neuroendocrine actions
Activation of the hypothalamic-pituitary-adrenal axis
Suppression of the hypothalamic-pituitary-gonadal axis
Suppression of the hypothalamic-pituitary-thyroidal axis
CNS activities
Fever
Hypermetabolism
Anorexia
Effects on liver
Acute phase protein synthesis
CNS, central nervous system.
/ 9j0c$$oc11
P13-8
of rapid cellular growth, tissue remodeling, disease, infection, or trauma. The particular cytokines produced in response to a threat to tissue homeostasis depends on the
nature of the threat (e.g., bacterial, viral, inflammatory),
the cellular or tissue type being threatened, the hormone
milieu, and to a large extent the profile of other cytokines
that are being produced.
A further striking feature of cytokines is the multiple
interactions between different individual cytokines. Many
types of interactions are apparent, including stimulatory
or inhibitory actions at the level of cytokine synthesis.
For example, the proinflammatory cytokines TNF-a and
IL-1 potently stimulate the production of a number of
other cytokines, including each other, as well as IL-6, IL8, IL-9, macrophage inflammatory protein (MIP), and CSF
(206). In contrast, anti-inflammatory cytokines such as
IL-4, IL-10, and IL-13 abrogate the production of many
proinflammatory cytokines (e.g., IL-1, IL-8, IL-12, TNF-a,
IFN-g, and CSF) (122, 563), whereas IL-6 inhibits the production of IL-1 and TNF-a but stimulates the production
of the endogenous IL-1 receptor antagonist IL-1ra (379,
743). Furthermore, there are numerous examples of a particular cytokine influencing the cellular responses to another cytokine. For example, nearly all biological responses to either TNF-a or IL-1 can be enhanced when
the two are administered together (206), whereas activin
functionally antagonizes the actions of gp130-signaling cytokines (114). The great propensity for cytokine-cytokine
11-25-98 11:16:36
pra
APS-Phys Rev
Downloaded from on April 23, 2014
aFGF, acidic fibroblast growth factor; BDNF, brain-derived neurotrophic factor; bFGF, basic fibroblast growth factor; cinc, cytokine-induced
neutrophil chemoattractant; CNTF, ciliary neurotrophic factor; CT-1, cardiotropin-1; EGF, epidermal growth factor; G-CSF, granulocyte colony
stimulating factor; GDNF, glial-derived neurotrophic factor; GM-CSF, granulocyte-macrophage colony stimulating factor; gro, growth-related oncogene; IGF, insulin-like growth factor; IL, interleukin; IL-1ra, IL-1 receptor antagonist; IFN, interferon; LIF, leukemia inhibitory factor; M-CSF,
macrophage colony stimulating factor; MIP, macrophage inflammatory protein; NAP, neutrophil activating protein; NGF, nerve growth factor; NT,
neurotrophin; OM, oncostatin M; PDGF, platelet-derived growth factor; RANTES, regulated upon activation normal and secreted; SCF, stem cell
factor; TGF, transforming growth factor; TNF, tumor necrosis factor.
January 1999
REGULATION OF HPA AXIS BY CYTOKINES
interactions is illustrated by the large number of different
cytokines that may be produced by a single threat to cellular/tissue homeostasis. For example, endotoxemia has
been reported to cause the increased synthesis and/or
secretion of IL-1a, IL-1b, IL-1ra, IL-6, IL-8, IL-10, IL-12,
TNF-a, MIP, macrophage migrating inhibitory factor
(MIF), IFN-g, leukemia inhibitory factor (LIF), and granulocyte-macrophage CSF. It is therefore apparent that during the course of a threat to tissue homeostasis, the physiological outcome is determined by the net effect of the
interactions between a number of cytokines.
Although many different cytokines have been shown
to influence HPA axis secretory activity, by far the majority of studies have focused on the cytokines IL-1, IL-6
and TNF-a. These three cytokines share many biological
activities (see Table 4). Section IC2 gives a brief outline
of their structure, biosynthesis, and receptors.
2. IL-1, IL-6, and TNF-a
/ 9j0c$$oc11
P13-8
munoglobulin supergene family and possess a single
transmembrane domain. Each receptor binds IL-1a, IL1b, and IL-1ra, but with differing affinities (784). It has
been proposed that the biological actions of IL-1 are mediated exclusively through IL-1R1 (442, 785), with IL-1R2
functioning solely as a decoy receptor that limits the availability of IL-1 for interaction with IL-1R1 (163, 784, 786).
In contrast, some studies have demonstrated that a monoclonal antibody (ALVA 42) raised against IL-1R2 inhibits
some actions of IL-1 within the brain (493, 548), although
the ability of this antibody to bind IL-1R2 has been questioned (282). In addition to the two receptor isoforms,
an accessory protein (IL-1RAcP) has been identified that
enhances binding of IL-1 to IL-1R1 (304, 482) and plays a
critical role in cell signaling through this receptor (348,
433, 994). Several additional members of the IL-1 receptor
family have been identified on the basis of sequence homology (53, 94, 278, 488, 489, 550, 627, 989). One of these
proteins (IL-1 receptor-related protein, IL-1Rrp) has recently been identified as a functional receptor for IGIF/
IL-18/IL-1g (869).
Tumor necrosis factor also occurs in a- and b-forms,
which share Ç50% homology. Tumor necrosis factor-b
(lymphotoxin-a) is produced predominantly by activated
lymphocytes. In contrast, TNF-a (also known as
cachectin) is expressed on a wide variety of hemopoietic
and nonhemopoietic cells as a 26-kDa membrane-associated molecule. This can be processed to give a secreted
17-kDa soluble form that mediates a range of inflammatory and cellular immune responses. Tumor necrosis factor is one of 10 known members of a family of ligands
that activate a family of structurally related receptors.
These include receptors for TNF-a and TNF-b, lymphotoxin-b, Fas ligand, nerve growth factor (NGF), and CD40
ligand (47). All the ligands for these receptors consist of
three polypeptide chains, and the majority are transmembrane proteins that act mainly through cell-to-cell contact.
However, TNF, as indicated, is also secreted.
Actions of TNF-a are exerted through interactions
with two distinct receptors: the 55-kDa (TNF-R1) and 75kDa (TNF-R2) receptors (47). These two receptors are
both transmembrane proteins with a single transmembrane span and are expressed at low levels on most cell
types. Although the extracellular domains of these two
receptors show a similar architecture, the intracellular
domains of these two receptors bear no significant homology, suggesting that they utilize separate signaling pathways (467). Indeed, studies of the effects of receptorspecific agonistic antibodies (233, 241, 283, 851, 968) and
of TNF-R-deficient mice (234, 642, 707) indicate that these
two receptors mediate effects that are largely, but not
exclusively, nonoverlapping. Recent molecular studies
have shed a considerable light on the activities of the two
receptors (reviewed in Ref. 182). Binding of TNF to either
receptor activates the proinflammatory transcription fac-
11-25-98 11:16:36
pra
APS-Phys Rev
Downloaded from on April 23, 2014
There are at least three distinct glycoproteins that
constitute the IL-1 family. The two agonists, IL-1a and
IL-1b, share Ç25% sequence homology, are distinct gene
products, and exhibit the same activities in numerous biological test systems (205). Both are synthesized as 31-kDa
precursor molecules. Pro-IL-1b is biologically inactive and
requires proteolytic cleavage by the IL-1b converting enzyme (ICE, also known as caspase-1) (401). In addition,
an endogenous antagonist at IL-1 receptors (IL-1ra) has
been described, which shares significant homology with
IL-1a and IL-1b, binds IL-1 receptors, but lacks intrinsic
biological activity (225, 318). Interleukin-1ra has been
used extensively as a pharmacological tool to explore the
role of IL-1-IL-1 receptor interactions in physiological responses (203). More interestingly, however, endogenous
IL-1ra is secreted by similar cell types, and in response
to similar stimuli, as those which produce the IL-1 agonists, and endogenous IL-1ra plays an important role in
regulating the physiological responses to endogenous IL1 (260, 485). Recently, a fourth member of the IL-1 family
has been proposed. Structural alignment of mouse IL-18
(also known as IFN-g inducing factor, IGIF) demonstrated
a 12 and 19% structural homology of this newly cloned
cytokine with IL-1b and IL-1a, respectively (46). Interleukin-18 was thus tentatively termed IL-1g (46). Although
there is, as yet, little information about this new cytokine
(850), it is known that lipopolysaccharide (LPS)-stimulated production of mature IL-18/IL-1g requires ICE (249,
287, 307). Furthermore, IL-18/IL-1g and IL-1 itself share
similar signaling pathways (428) and functional activities
(351), again indicating a close relationship between this
novel cytokine and the IL-1 family.
Two distinct mammalian, membrane-bound IL-1 receptors have been described and designated IL-1R1 and
IL-1R2 (784). Both are glycoproteins belonging to the im-
7
8
ANDREW V. TURNBULL AND CATHERINE L. RIVIER
/ 9j0c$$oc11
P13-8
and TNF soluble receptors to bind their ligands limits the
availability of either IL-1 or TNF-a for interaction with
their membrane-bound receptors and therefore confers
antagonistic properties to these truncated receptors. In
contrast, the soluble IL-6 receptor, upon binding its ligand,
can interact with gp130 and elicit a cellular response.
Indeed, coincubation of IL-6 with its soluble receptor has
been demonstrated to confer IL-6 sensitivity to previously
IL-6-insensitive cells and enhances the effectiveness of IL6 in vivo (506, 641, 747). Thus the biological activity of
a particular cytokine is determined not only by its own
concentration and the concentration of cytokines which
influence its activity but also by the presence of its soluble
receptor.
D. Hypothalamic-Pituitary-Adrenal Axis
1. HPA axis organization
Over the last 10–15 years, there have been over 1,000
published articles concerning the activation of the HPA
axis by cytokines. This relative abundance of work is due
to the large number of cytokines discovered, the complexity of the organization of the HPA axis (see Fig. 1), and
the functional importance of activation of the HPA axis
during stressful situations. Basal secretion of GC is necessary for the normal function of most tissues, and even
small deviations from normal circulating levels of these
steroids produce changes in a wide variety of physiological and biochemical parameters. Interactions between the
endocrine system and the CNS result in a diurnal rhythm
of GC secretion with a peak occurring at the time of awakening and a nadir during the first few hours of sleep. Blood
levels of circulating GC increase in response to virtually
any type of stimulus that poses, or is perceived to pose,
a threat to bodily homeostasis. Glucocorticoids act on
multiple targets to enhance or inhibit various cellular activities, actions that are aimed at providing the altered
metabolic, endocrine, nervous, cardiovascular, and immunologic needs that promote survival. Not surprisingly,
therefore, the regulation of blood levels of GC is subject
to diverse sensory inputs, and this information is integrated at the level of the hypothalamus.
The primary CNS nucleus involved in the regulation
of pituitary-adrenal axis is the paraventricular nucleus
(PVN) of the hypothalamus. The PVN is the principal CNS
source of the 41-amino acid peptide CRF, which is the
major physiological regulator of pituitary ACTH secretion
(691). The CRF hypophysiotropic neurons from the PVN
project to the external zone of the median eminence (ME)
and release CRF into a specialized capillary network. The
anterior pituitary (or adenohypophysis) is vascularized by
hypophysial portal vessels that arise from these median
eminence capillary beds. Within the anterior pituitary,
CRF interacts with a specific G protein-coupled receptor
11-25-98 11:16:36
pra
APS-Phys Rev
Downloaded from on April 23, 2014
tor NFkB. In the case of TNF-R2, signal transduction occurs via heteterodimerization of the receptor with two
TNF-R2 associated factors, TRAF1 and TRAF2, and it is
TRAF2 that appears to mediate TNF-R2-induced activation of NFkB. In contrast, TNF-R1, upon ligand binding,
recruits a protein called TRADD. Like TRAF2, TRADD
causes NFkB activation but, unlike TRAF2, also causes
apoptosis via an ICE-like protease. This explains why
TNF-R1, but not TNF-R2, causes apoptosis. However, the
NH2-terminal domain of TRADD interacts directly with
TRAF2, and overexpression of a dominant negative
TRAF2 blocks not only TNF-R2 but also TNF-R1-induced
NFkB activation. Thus activation of the two TNF receptors elicits separate signaling pathways that can interact
with one another, thus explaining the distinct and overlapping signals generated by the two TNF receptors.
Interleukin-6 is a single 21- to 28-kDa glycoprotein
produced by both lymphoid and nonlymphoid cells and
regulates immune responses, acute-phase protein synthesis, and hematopoiesis. Human IL-6 is synthesized as a
precursor polypeptide of 212 amino acids that is processed by cleavage of a 28-amino acid NH2-terminal signal
sequence into a mature form of 184 amino acids.
One IL-6 receptor has thus far been identified. This
IL-6 specific receptor (IL-6Ra) is responsible only for
binding of its ligand (IL-6). Interleukin-6 belongs to a family of cytokines that includes ciliary neurotropic factor
(CNTF), oncostatin M (OM), LIF, IL-11, and cardiotropin1 (CT-1), which share a common signal-transducing mechanism (reviewed in Refs. 415, 416). All these cytokines
are bound by receptors that interact with the common
cell-surface protein gp130. Ligand-receptor complexes
that share gp130 trigger signaling by the formation of either homodimers of gp130 or heterodimers between gp130
and LIFR. In the case of IL-6, signaling is initiated by
the homodimerization of gp130 induced by the interaction
with the IL-6/IL-6Ra complex. Either homodimerization
of gp130 or heterodimerization of gp130 with LIFR activates JAK kinases, followed by the tyrosine-specific phosphorylation and nuclear translocation of a member of the
STAT family (STAT3) of transcription factors. In addition,
there is another signaling pathway that involves the activation of the RAS-MAP kinase cascade followed by the
activation of transcription factors such as nuclear factorIL-6 (NF-IL-6). Such sharing of receptor complexes and
subsequent activation of similar signaling pathways by
members of the IL-6 family of cytokines is a clear example
of how a number of different cytokines display similar
biological activities (i.e., cytokine redundancy).
Receptors for IL-1, IL-6, and TNF-a occur not only
in membrane-bound forms, but also as truncated soluble
products, which are capable of binding their ligand (257,
332, 703). These receptors are generated either by proteolytic cleavage at the cell surface or are synthesized as
alternatively spliced mRNA species. The ability of IL-1
Volume 79
January 1999
REGULATION OF HPA AXIS BY CYTOKINES
(CRF-R1) on the corticotrope cell surface, resulting in the
stimulation of the synthesis of the ACTH precursor peptide proopiomelanocortin (POMC) and the secretion of
ACTH and other POMC-derived peptides (888, 898). Adrenocorticotropin hormone potently induces the secretion
of GC from the zona fasciculata of the adrenal cortex. In
humans, the major GC is cortisol, but in the rat and mouse,
corticosterone is the main steroid product of the zona
fasciculata. In a classical endocrine feedback manner,
these steroids inhibit the synthesis and secretion of CRF
within the hypothalamus and POMC-derived peptides in
the pituitary (404, 995).
The PVN has an anatomically discrete topography but
functionally and phenotypically overlapping organization.
The PVN is comprised of two major neurosecretory subdivisions; the magnocellular (mPVN) and parvocellular
(pPVN) divisions, as well as more caudal cell groups giv-
/ 9j0c$$oc11
P13-8
ing rise to long descending projections to brain stem autonomic structures (738). The mPVN together with the supraoptic nucleus (SON) constitute the magnocellular neurons that are the major cell sources of arginine
vasopressin (AVP) and oxytocin released into the general
circulation from neurons terminating in the posterior pituitary. The more medially situated pPVN is the major
source of hypophysiotropic CRF neurons, which release
CRF into the hypophysial portal circulation. The cell
groups projecting to autonomic structures contain all
three peptides (CRF, AVP, and oxytocin). However, although these subdivisions of PVN are anatomically discrete, the PVN does display considerable peptide phenotype plasticity. The CRF hypophysiotropic neurons also
produce a number of additional peptides, most notably
AVP (740, 864), which interacts synergistically with CRF
to stimulate ACTH secretion (693), and whose synthesis in
these neurons can be enhanced during increased pituitaryadrenal activity (738). Furthermore, AVP and oxytocin
derived from sources other than the pPVN may also contribute to the pool of ACTH secretagogues in hypophysial
portal blood (15, 648, 651). Therefore, although it is generally agreed that CRF arising from the pPVN is the major
means of stimulating ACTH secretion, this is not an absolute, with AVP (and possibly oxytocin) secretion from
either pPVN or magnocellular neurons contributing to an
extent that varies with the nature of the physiological
threat.
Consistent with the extreme diversity of stressful
stimuli that give rise to activation of the HPA axis, the
pPVN receives diverse inputs from regions of the brain
conveying visceral, somatosensory, auditory, nociceptive, and visual information and also from limbic regions
involved in the integration of cognitive and emotional
influences (738). These inputs include projections from
other nuclei within the hypothalamus (e.g., medial preoptic anterior hypothalamus). Extrahypothalamic inputs include areas both within [e.g., nucleus of the solitary tract (NTS) and other medullary catecholaminergic
cell groups] and outside [organum vascularis of the lamina terminalis (OVLT) and subfornical organ (SFO)] the
blood-brain barrier (BBB) (738). There are, therefore,
multiple levels at which the activity of the HPA axis may
be modulated, and indeed, virtually all the regulatory
processes described above have been proposed as sites
at which cytokines regulate HPA axis activity.
2. Experimental assessment of the HPA axis
secretory activity
The measurable end points and experimental methodologies that have been used to examine the secretory
activity of the HPA axis in response to cytokines are extremely diverse and shall be considered here briefly. The
net result of increased HPA axis secretory activity is the
11-25-98 11:16:36
pra
APS-Phys Rev
Downloaded from on April 23, 2014
FIG. 1. Functional anatomy of hypothalamic-pituitary-adrenal axis.
AVP, arginine vasopressin; CRF, corticotropin-releasing factor; I.H., inferior hypophysial; S.H., superior hypophysial.
9
10
ANDREW V. TURNBULL AND CATHERINE L. RIVIER
/ 9j0c$$oc11
P13-8
of either hypothalami, anterior pituitaries, or adrenals,
with the tissue being intact (whole), in segments or
slices, or in dispersed monolayer cell culture. These
methodologies produce a more isolated environment in
which to define direct actions of applied substances.
II. CYTOKINE INFLUENCE ON
HYPOTHALAMIC-PITUITARY-ADRENAL
AXIS SECRETORY ACTIVITY IN VIVO
A. Animal Studies
Numerous studies have confirmed and extended
the original findings that the administration of either
IL-1a or IL-1b to rats or mice stimulates ACTH and
GC secretion, as well as many other indices of HPA
activation (see Table 5). In addition, IL-1 has been demonstrated to increase the secretory activity of the HPA
axis of chickens (959), sheep (916), baboons (674), and
humans (see sect. IIB). Studies addressing interactions
between cytokines and neuroendocrine systems in an
invertebrate species (snail) have demonstrated the
presence of a rudimentary stress system involving CRF-,
ACTH-, and bioamine-like molecules in immunocytes
(613 – 616). The snail immunocyte stress system contains, and is responsive to, cytokines, including IL-1, IL2, and TNF-a (612, 615 – 617). Activation of the HPA axis
(or invertebrate equivalent) by IL-1 has therefore been
highly conserved throughout evolution in different species and taxa, indicating the importance of this adaptive
response to survival (615).
In mammals, the ACTH response to intravenous IL-1
is usually prompt, commencing within 5–10 min, and of
relatively short duration (Ç1 h). In comparison, the
plasma ACTH response to intraperitoneal injection of IL1b is slower in onset, but usually of longer duration (at
least 2 h). Finally, the response to IL-1 administered directly into the brain (intracerebroventricularly) is of intermediate latency and lasts for several hours (usually
greater than 3–4 h). The majority of studies have found
IL-1b to be more potent than IL-1a in the rat (525, 574,
690, 695). Studies utilizing a panel of monoclonal antibodies have demonstrated that amino acids in the domain
66–85 on the recombinant rat IL-1b molecule are critical
for its ACTH-releasing capacity (751). In the rat, IL-1b
stimulates ACTH secretion at all stages of postnatal development of both males and females, although the magnitude of the ACTH response depends on age and gender
(59, 192, 466, 598, 686).
A single administration of IL-1b not only acutely elevates plasma ACTH and corticosterone concentrations in
the rat, but has been demonstrated to produce a longlasting (at least 3 wk) increase in the coexpression of AVP
in hypothalamic CRF neurons and a hyperresponsiveness
11-25-98 11:16:36
pra
APS-Phys Rev
Downloaded from on April 23, 2014
elevated concentrations of ACTH and GC in blood. Temporal measures of immunoreactive levels of these hormones have been widely adopted as means of studying
the influence of cytokines on the HPA axis and are the
main method suitable for use in humans. Furthermore,
the determination in laboratory animals of the effects of
various surgical, pharmacological, or genetic manipulations on the plasma hormone response to a given cytokine
provides a valuable means to elucidate the anatomic and
neurochemical mechanisms involved in the activation of
the HPA axis by a particular cytokine.
To assess the activity of neuronal components of the
HPA axis in response to a particular cytokine, a number
of methods have been employed, including the use of
electrophysiological recordings and the histochemical determination of the expression of cellular immediate early
genes (cIEG), such as c-fos. The demonstration of c-fos
as an inducible and widely applicable marker of neuronal
activity (558) has afforded a means of mapping neuronal
activation in response to a variety of stimuli, including
the administration of cytokines. In particular, the stressinduced induction of c-fos mRNA and/or Fos protein
within the PVN has been thoroughly examined and validated as a means to identify activation of neurosecretory
neurons (152, 436). Another cIEG, NGFI-B, has been used
for similar purposes (152, 436). Identification of the peptide phenotype of cells within the PVN expressing cIEG
provides a powerful means to identify the activation of
particular neurosecretory neurons. However, it should be
noted that at present we do not know how the induction
of such transcription factors relates to transcriptional activity within the PVN. Indeed, of the three peptides CRF,
AVP, and oxytocin, only the AVP gene includes an AP-1
response element that binds Fos-Jun dimers, but all three
genes contain potential NGFI-B response elements (152).
Functional assessments of the relative neurosecretory
rates of peptides from hypophysial PVN neurons have also
been perfomed by measuring 1) peptide concentrations
directly in portal blood, 2) peptide concentrations in the
perfusates from push-pull cannulas or microdialysis
probes within the ME, and 3) peptide content of the ME
of either normal animals or animals pretreated with colchicine to block axonal transport. Finally, determination
of the expression of steady-state mRNA or primary transcript RNA (hnRNA) levels of either CRF or AVP within
the PVN have also been well documented.
To determine the direct effects of cytokines on particular components of the HPA axis, a number of in
vitro methodologies have also been used. Only one cell
line has been available to study the action of cytokines
on the HPA axis, namely, the AtT20 mouse corticotropic
tumor line, which has been used as a model for investigating the direct effects of cytokines on anterior pituitary corticotropes. By far the majority of in vitro studies have utilized static or perfused primary preparations
Volume 79
January 1999
TABLE
Cytokine
IL-1a
5. Acute effects of cytokines on the HPA axis of laboratory animals
Route
Effect
icv
iv
ip
icv
IL-2
iv
icv
ip
IL-4
IL-6
ip
iv
OM
CT-1
CNTF
TNF-a
ip
icv
ip
ip
ia
ip
ip
ip
ip
iv
IFN-a
Activin
icv
ip, icv
icv
IL-11
IL-12
LIF
EGF
NGF
SCF
iv
icv
iv
iv
Reference No.
CRF in portal blood
Plasma ACTH, corticosterone
CRF mRNA
CRF content of ME
POMC mRNA in anterior pituitary
Plasma ACTH
Plasma ACTH
PVN c-fos mRNA or Fos protein
PVN CRF mRNA
CRF secretion from ME
CRF content of ME
Plasma ACTH, corticosterone
PVN c-fos mRNA or Fos protein
CRF PVN mRNA
POMC mRNA in anterior pituitary
POMC hnRNA in anterior pituitary
ACTH content of anterior pituitary
Plasma ACTH, corticosterone
PVN c-fos mRNA or Fos protein
PVN CRF mRNA
PVN AVP mRNA
CRF secretion from ME
Plasma ACTH, corticosterone
ACTH
Electrical activity of PVN neurons
Plasma ACTH*, corticosterone*
POMC in anterior pituitary
Hypothalamic vasopressin mRNA
Hypothalamic oxytocin mRNA
POMC in anterior pituitary
PVN Fos protein
Plasma ACTH, corticosterone
Plasma ACTH, corticosterone
Plasma ACTH, corticosterone
Plasma corticosterone†
Plasma corticosterone
Plasma ACTH
Plasma corticosterone†
Plasma corticosterone†
Plasma corticosterone†
Plasma corticosterone†
CRF secretion from ME
Plasma ACTH, corticosterone
Plasma ACTH
or F Plasma corticosterone‡
CRF in portal blood
Plasma ACTH
Plasma ACTH, corticosterone
Plasma ACTH, corticosterone
Plasma ACTH, corticosterone
PVN Fos protein
Plasma ACTH, corticosterone
223, 525, 574, 629, 730
639, 640, 824
574, 695
55, 223, 237, 525, 527, 560, 824, 929, 932, 936, 940
63, 64, 70, 98, 187, 327, 466, 628, 686, 690, 752, 824
40, 187, 457, 681, 684, 692
574
76, 317
326, 623
326
527, 575, 589, 909
52, 327
499, 527, 859, 909
52
608
5
52
52
52
52, 246
58, 66, 223, 771, 772, 938
51, 673, 881
725, 726, 729
650
492, 551
551
618, 741, 836
435
Routes of administration: iv, intravenous; ia, intra-arterial; ip, intraperitoneal; icv, intracerebroventricular. PVN, paraventricular nucleus; ME,
median eminence; POMC, proopiomelanocortin. Other definitions are as in Table 3.
* Chronic (7-day) infusion. Significant increases in plasma
ACTH concentration observed only during latter period of dark cycle.
† At doses tested, each of these cytokines alone had no effect on plasma
corticosterone, but each potentiated the corticosterone response to a submaximal dose of IL-1.
‡ Stimulation of corticosterone secretion was
noted only at higher doses of IFN-a.
of the HPA axis (744). Long-term administration of IL-1b
to rats enhances CRF- and ACTH-like immunoreactivities
in the hypothalamus and pituitary, respectively, increases
adrenal weight (573), and elevates plasma ACTH concentrations for at least 7 days (573, 830, 908).
In addition to IL-1, a number of other cytokines have
/ 9j0c$$oc11
P13-8
been demonstrated to influence HPA axis secretory activity in experimental in vivo paradigms (see Table 5). Activation of the HPA axis is not restricted to cytokines produced predominantly by myeloid (e.g., monocyte, macrophage) cells (e.g. IL-1), but also by cytokines produced
by lymphoid (e.g., T lymphocytes) cells (e.g., IL-2). Where
11-25-98 11:16:36
pra
APS-Phys Rev
Downloaded from on April 23, 2014
F
F
F
f
F
F
F
F
F
F
f
F
F
F
F
F
f
F
F
F
F
F
F
F
F
F
F
F
F
f
F
F
F
F
F
F
F
F
F
F
F
F
F
F
f
F
F
F
F
F
F
F
iv
ip
IL-1b
11
REGULATION OF HPA AXIS BY CYTOKINES
12
ANDREW V. TURNBULL AND CATHERINE L. RIVIER
/ 9j0c$$oc11
P13-8
effects on the HPA axis of recombinant rat cytokines generated by this initiative had been published.
Although pharmacological differences of cytokines
from different species may seem to complicate interpretation of data, they can actually provide useful pharmacological tools. For example, the difference between the
effects of intracerebroventricular mouse and human TNFa on plasma ACTH concentration may reflect the differing
pharmacological profiles of the two identified TNF receptors, TNF-R1 and TNF-R2. Murine TNF-R1 has high affinity
for either mouse or human TNF-a, whereas only mouse
TNF-a is effective at TNF-R2 (334, 467, 505). Should the
same pharmacology be true at rat TNF-a receptors, as
has been suggested (817), then the increase in plasma
ACTH concentrations induced by intracerebroventricular
mouse, but not human, TNF-a indicates that TNF-R2 is
the major receptor isoform involved in cerebral TNF-ainduced activation of the rat HPA axis.
Although by far the majority of cytokines tested in
animal studies have been found to exert stimulatory actions on the HPA axis (see Table 5), two cytokines (IL-4
and IFN-g) have been suggested to inhibit HPA axis activity in vivo. The anti-inflammatory cytokine IL-4 dose-dependently inhibits POMC mRNA expression in the anterior pituitary, without affecting CRF mRNA in the pPVN,
suggesting a direct inhibitory action at the level of the
pituitary (326). Interferon-g has also been suggested to
inhibit HPA axis secretory activity, since the administration of low doses either intraperitoneally or intracerebroventricularly reduces plasma corticosterone levels and the
electrical activity of neurons within the PVN (725, 726,
729). However, higher doses of IFN-g given intracerebroventricularly enhance plasma corticosterone levels (726),
suggesting that the qualitative effects of IFN-g on HPA
axis activity are dependent on the dose used.
B. Human Studies
In addition to the numerous studies of the effects of
cytokines on the HPA axis of laboratory animals, the clinical trials of a number of cytokines as anticancer strategies
have afforded the opportunity to detail their effects on the
HPA axis of humans. Such clinical studies have permitted
investigation of the effects of cytokines on the HPA axis
in a homologous system (i.e., human cytokines in human
subjects). Either intravenous or subcutaneous administration of IL-1a (179, 795), IL-1b (176, 179), IL-2 (24, 194,
479–481, 812), IL-6 (519, 520, 809), TNF-a (596) IFN-a (41,
289, 337, 565, 566, 702, 758), IFN-b (596), and IFN-g (342,
596, 810) elevates plasma ACTH and/or cortisol concentrations. As in laboratory animals, the stimulation of
ACTH secretion produced by cytokines occurs rapidly:
within 1 h of intravenous infusion or within 1–4 h of
subcutaneous treatment.
11-25-98 11:16:36
pra
APS-Phys Rev
Downloaded from on April 23, 2014
studies have compared the potencies, IL-1b has generally
been found to be the cytokine most potent at stimulating
ACTH secretion (e.g., Refs. 66, 358, 527, 772). However,
this is not necessarily of physiological significance, since
when it is the levels of endogenous cytokines that are
elevated, the relative concentrations of each cytokine is
a major determinant of which cytokine is of greatest influence. For example, during local inflammation, IL-6,
which is generally agreed to be a less potent HPA axis
secretagogue than IL-1, is elevated to a greater extent and
for greater periods of time than is IL-1. Additionally, at
least some cytokines act synergistically to enhance ACTH
secretion. For example, all hematopoietic cytokines (IL6, LIF, OM, CNTF, IL-11, and CT-1) enhance ACTH and/
or corticosterone secretion produced by IL-1 to an extent
greater than can be accounted for by additive effects (52,
639, 1003). Similarly, TNF-a synergistically enhances
ACTH release produced by IL-1b (907).
A number of studies have produced contradictory
data with respect to the effects of various cytokines on
HPA axis secretory activity. Although a stimulatory effect
of TNF-a on the rat HPA axis has not been disputed when
the cytokine has been administered peripherally (58, 772,
909), contrasting data have been obtained when TNF-a
has been administered directly into the brain (intracerebroventricular). For example, although we showed that
intracerebroventricular TNF-a induces marked elevations
in plasma ACTH concentrations in rats (881), a number
of other investigators have found little or no effect (673,
772, 909). Such discrepancies may, at least in part, be
explained by the use of cytokines of different species
origin. In the example cited, we used murine TNF-a (881),
whereas those reporting little or no effect of intracerebroventricular TNF-a on plasma ACTH levels used human
TNF-a (673, 772, 909). Similarly, studies in mice (51) have
shown that plasma corticosterone concentrations are elevated to a greater extent by intracerebroventricular murine TNF-a than by intracerebroventricular human TNFa. Species differences have also been noted with IL-6, and
the human IL-6Ra recognizes human IL-6 but not mouse
IL-6 (173, 905). The use of human or mouse cytokine preparations has been common due to their wide availability,
yet the most commonly used species in the investigation
of HPA axis activity is the rat. However, the previous
limited availability of recombinant rat cytokines has
meant that not many studies have investigated the effects
of cytokines in a homologous system (rat cytokine in the
rat). When rat IL-1a (574) and rat IL-1b (751) have been
tested in rats, the intravenous injection of these cytokines
produces a marked elevation in plasma ACTH levels. Recombinant rat cytokines have now become more widely
available since the establishment of BIOMED 1 program
‘‘Cytokines in the brain’’ of the European Communities
(headed by Dr. R. Dantzer, Bordeaux, France). However,
at the time of submitting this review, no studies of the
Volume 79
January 1999
REGULATION OF HPA AXIS BY CYTOKINES
III. PHYSIOLOGICAL/PATHOPHYSIOLOGICAL
CIRCUMSTANCES IN WHICH ENDOGENOUS
CYTOKINES PLAY A ROLE IN REGULATION
OF HYPOTHALAMIC-PITUITARY-ADRENAL
AXIS
There are a number of examples of injury, infection,
and/or disease that are associated with increased cytokine
production and concomitant elevations in HPA axis activity: head trauma (427, 611, 976), cerebral ischemia
(stroke) (253, 367, 571), a number of autoimmune diseases
(175, 239, 322–325, 347, 455, 539, 654, 959), psychiatric
and dementia disorders (165, 302, 510, 896), acquired immune deficiency syndrome (AIDS) (636, 661, 763, 827,
954), and antigenic challenges (69, 72, 73). However, direct and clear evidence for the involvement of particular
/ 9j0c$$oc11
P13-8
cytokines in the HPA axis response to such insults has
been limited to only a few cases (viral or bacterial infection, local tissue damage and inflammation, and acute
physical/psychological stress). Furthermore, although numerous cytokines have been shown to influence the secretory activity of the HPA axis, the direct demonstration of
cytokine involvement in physiological or pathophysiological HPA axis responses has been restricted largely to the
cytokines IL-1, IL-6, and TNF-a. The following sections
outline the types of threats to homeostasis that have been
demonstrated to elicit activation of the HPA axis via
mechanisms that depend critically on the endogenous
elaboration of cytokines.
A. Viral Disease
1. Newcastle disease virus
The first direct evidence indicating that IL-1 participates in the HPA axis response to an immune challenge
came from studies investigating the neuroendocrine responses to inoculation with NDV (70). Newcastle disease
virus is a neutrotropic paramyxovirus which, when administered to rodents, produces symptoms of viral disease
without the potential hazard of replication. Within 1–2 h
of injection, NDV produces marked elevations in ACTH
and corticosterone concentrations in the blood of mice
(70, 219–221, 604, 793) or rats (685). The majority of studies demonstrate that the increase in corticosterone is
abolished by hypophysectomy (218, 220, 221, 604), indicating the importance of the pituitary in the adrenal response. Furthermore, the ACTH response is completely
prevented when rats are passively immunized against CRF
(685), indicating that hypothalamic CRF regulates the pituitary ACTH response to NDV. The stimulation of the
HPA axis by NDV appears to be produced not by the virus
itself, but by mediators released by immune cells exposed
to the virus. This is evidenced by the fact that the supernatants of cocultures of NDV with either human peripheral
blood leukocytes (HPBL) or mouse spleen cells also elevate plasma corticosterone concentrations. Indeed, intraperitoneal injection of supernatant from NDV plus HPBL
produces a fourfold increase in plasma corticosterone
concentrations in rats (70). This plasma corticosterone
response is prevented by preincubation of the NDV plus
HPBL supernatant with a rabbit neutralizing anti-human
IL-1 antibody (70). More recent studies have demonstrated that intraperitoneal administration of IL-1ra to
mice virtually abolishes the elevations in plasma ACTH
and corticosterone concentrations 2 h after NDV (221),
confirming the obligatory role of IL-1 in the generation of
the HPA axis response to this viral challenge.
2. Polyinosinic polycytidilic acid
Polyinosinic polycytidilic acid (Poly I:C) is a synthetic, double-stranded polyribonucleotide commonly
11-25-98 11:16:36
pra
APS-Phys Rev
Downloaded from on April 23, 2014
A series of experiments (519, 520) in which cancer
patients of good clinical performance were examined
showed that IL-6 is a particularly potent activator of the
HPA axis and that the human HPA axis is remarkably
responsive to this cytokine. On the first treatment day, IL6 (30 mg/kg sc) induced marked elevations in plasma
ACTH and cortisol concentrations, with peaks occurring
at 1 and 2 h, respectively. Plasma ACTH concentrations
returned to basal levels within 5 h, whereas plasma cortisol levels remained elevated for 24 h. By the seventh day
of treatment, the ACTH response to IL-6 was markedly
diminished, an effect that was probably due to increased
negative feedback produced by persistently elevated
plasma cortisol levels. The sustained secretory activity of
the adrenal was accompanied by gross enlargement of
the adrenal glands as assessed by computed tomographic
scans. Subsequent studies demonstrated that as little as
0.3 mg/kg (intravenous) is already a maximal dose of IL6 (520). The magnitude of the plasma ACTH response to
a first injection of IL-6 was greater than that reported with
the standard tests of pituitary-adrenal function such as
injection of ovine CRF or the insulin tolerance test. The
relatively mild toxic effects of IL-6 led the authors to propose that IL-6 may provide a new means of testing pituitary-adrenal function (519) and to conduct additional
studies in normal human subjects (876). In normal healthy
males, IL-6 (subcutaneous) again produced substantial elevations in plasma ACTH and cortisol, with peaks in
ACTH and cortisol levels observed at 60–90 and 90–120
min, respectively, and a minimal effective IL-6 dose of 1–
3 mg/kg (876).
Overall, human and animal studies agree that many
exogenously administered cytokines have marked stimulatory actions on HPA axis secretory activity and suggest
that endogenous production of cytokines during homeostatic threats may well play a causal role in the elaboration
of the accompanying HPA axis response.
13
14
ANDREW V. TURNBULL AND CATHERINE L. RIVIER
Volume 79
used to mimic viral exposure. Injection of Poly I:C, like
NDV, produces a rapid (within 1–2 h) activation of the
HPA axis (544, 716). Although there have been only a few
investigations of the HPA axis response to Poly I:C, it is
apparent that CRF is an important mediator of this response, because Poly I:C-induced increases in rabbit
plasma cortisol concentrations are abolished by pretreatment with a monoclonal anti-CRF antibody (544). Furthermore, the substantial plasma corticosterone response to
Poly I:C observed in normal mice is completely absent in
mice deficient in IL-6 (716), indicating that Poly I:C-induced activation of the HPA axis is dependent on the
elaboration of the cytokine IL-6.
/ 9j0c$$oc11
P13-8
11-25-98 11:16:36
pra
APS-Phys Rev
Downloaded from on April 23, 2014
derlying the neuroendocrine responses to bacterial infection and sepsis (214, 863, 883).
Although mice and rats are relatively insensitive to
LPS in comparison with many other species (including
humans), the intravenous administration of LPS to laboratory rodents produces marked elevations in ACTH and
corticosterone secretion within 30–60 min. It should be
emphasized that, quantitatively, physiological responses
to LPS can differ depending on its source and preparation
(345). We find that doses of Ç5 mg/kg LPS (Escherichia
coli serotype O26:B6; lot 20H4025, Sigma Chemical) produce a peak elevation in plasma ACTH concentration of
500–1,000 pg/ml (compared with õ20 pg/ml in controls)
at 90–120 min after intravenous injection in intact, male
3. Murine cytomegalovirus
rats (889). Similarly, LPS stimulates ACTH and corticosterone secretion in mice, and HPA activation can be obCytomegaloviruses (CMV) are herpes viruses that are
served in both rats and mice after either intravenous or
a major cause of mortality and morbidity in human transintraperitoneal administration. However, the precise
plant recipients, are a serious problem in patients with
mechanisms through which these two routes of LPS adAIDS, and are the most frequent viral cause of congenital
ministration influence the HPA axis may be substantially
abnormalities. Administration of murine CMV (MCMV)
different (144, 925).
productively infects mice. Recent studies by Ruzek et al.
In addition to elevated blood levels of ACTH and corti(716) showed that MCMV induces increased levels of ILcosterone, other indexes of HPA activation have been re12, IFN-g, TNF-a, IL-1a, and IL-6, but not IL-1b, in the
ported after peripheral administration of LPS. Intravenous
general circulation at 24–48 h of infection. During this
or intraperitoneal LPS produces increased pPVN expression
period, there are marked increases in the plasma concenof c-fos mRNA or Fos protein and of CRF hnRNA or mRNA
trations of corticosterone and smaller, but statistically sig(230, 387, 456, 680, 718, 924, 925). Rats in which the PVN
nificant, increases in plasma ACTH (716). The corticostehas been electrolytically lesioned can mount a detectable
rone response to MCMV in either normal mice treated
plasma ACTH response to an extremely large dose of LPS (2
with neutralizing anti-IFN-g antibodies, or in IFN-g-defimg/kg ip), but its magnitude is markedly diminished (227),
cient mice, is comparable to that in control mice infected
indicating that the PVN plays a pivotal role in LPS-stimuwith MCMV (716). However, IL-1 and IL-6 appear to play
lated increases in plasma ACTH. Indeed, it is clear that CRF
important roles in the activation of the HPA axis. The
is an important mediator of LPS-induced ACTH secretion.
corticosterone response to MCMV is markedly blunted in
This is evidenced by the increased secretion of CRF from
either normal mice treated with IL-1ra or in IL-6-deficient
the ME after systemic treatment with LPS (292) and by
mice, without either of these ‘‘treatments’’ having a sigthe marked attenuation, or abolition, of LPS-induced ACTH
nificant impact on viral load. The elevated IL-6 levels prosecretion produced by doses of LPS that are either very
duced by MCMV infection are dramatically reduced when
large (2.5 mg/kg ip) or more moderate (2.5 mg/kg ip or 50
mice are treated with IL-1ra, whereas IL-1a levels are
mg/kg iv), respectively (25, 750).
normally elevated in IL-6-deficient MCMV mice (716). ConThese actions of LPS on the HPA axis in vivo are
sequently, the authors concluded that IL-6 is the pivotal
not due to a direct pharmacological interaction of LPS
cytokine in the activation of the HPA axis in response to
with HPA axis tissues. Lipopolysaccharide has either no
MCMV and that IL-1a plays a secondary role by contributeffect or an inhibitory action on either CRF secretion
ing to IL-6 production (716).
from hypothalamic explants (545, 581, 652) or ACTH secretion from rat anterior pituitary cell cultures (117, 886).
B. Endotoxin Treatment
It also seems unlikely that LPS enhances the pituitary
ACTH response to CRF, since LPS reduces the expresEndotoxins are LPS constituents of the outermost sion of CRF receptors in the pituitary both in vivo and
part of a Gram-negative bacterial cell membrane that are in vitro (25), and ACTH secretion by rat anterior pituitary
released upon bacterial lysis. Administration of purified cell cultures stimulated with CRF is unaffected by copreparations of LPS mimics many of the acute phase re- treatment with LPS (886). Furthermore, mice (C3H/HeJ
sponses to Gram-negative infection without actively in- strain) that are deficient in their production of IL-1 in
fecting the host (123). Consequently, administration of response to LPS (373, 759, 856) exhibit markedly reduced
bacterial endotoxins to laboratory animals has been the elevations in the plasma concentrations of ACTH and
most commonly used model to study the mechanisms un- corticosterone after intraperitoneal LPS (216), sug-
January 1999
REGULATION OF HPA AXIS BY CYTOKINES
15
/ 9j0c$$oc11
P13-8
11-25-98 11:16:36
pra
APS-Phys Rev
Downloaded from on April 23, 2014
gesting that IL-1 is an important mediator of the effects or IL-1ra injected into rats (223, 752) reduce the ACTH
of LPS. Lipopolysaccharide is a potent inducer of the response to LPS admistered intravenously or intraperitosynthesis and secretion of a number of cytokines (see neally. A CNS site of IL-1 action has been suggested by
sect. IC). In particular, the cytokines IL-1, IL-6, and experiments showing that intracerebroventricular infuTNF-a are likely candidate mediators of the effects of sion of IL-1ra inhibits the increase in CRF mRNA in the
LPS on neuroendocrine secretion (863).
PVN of rats 8 h after intraperitoneal LPS (387).
After administration of LPS directly into the bloodAlthough the above findings seem to strongly implistream, the plasma concentrations of each of these three cate the cytokine IL-1 as a mediator of the activation of
cytokines are elevated in a regulated temporal manner the HPA axis by LPS, not all studies support this hypothewith TNF-a first, then IL-1b, and finally IL-6 (174, 189, sis. For example, mice injected with IL-1ra ip, at a dose
290). Not only are the secretions of these three cytokines that inhibited the corticosterone response to either IL-1a
temporally related, but there is also evidence that they or IL-1b, failed to inhibit the corticosterone response to
are also causally related. Administration of antibodies to LPS (213). Furthermore, the development of genetically
TNF-a blunts the secretion of IL-1 and IL-6 in response manipulated mice, with deficiencies in various compoto LPS (263), whereas immunoneutralization of IL-1 (487) nents of the IL-1 system, has raised interesting questions
or frequent administration of large doses of IL-1ra (494) regarding the absolute requirement of IL-1 in acute phase
abrogates LPS-induced IL-6 secretion. After local injection responses to LPS. For example, fever in response to LPS
of LPS (e.g., intraperitoneally or into an experimentally is inhibited by either anti-IL-1b antibodies (418, 461, 487)
constructed, subcutaneous air pouch), the local concen- or by IL-1ra (494, 791), clearly implicating IL-1 in the
trations of all three cytokines are also elevated. However, pathogenesis of fever due to LPS. However, mice deficient
their levels in blood appear to be dependent on the degree in IL-1b show only a slightly decreased (10) or even an
of ‘‘overspill’’ into the general circulation, with only IL-6 increased (438) fever in response to intraperitoneal LPS.
levels being consistently increased in systemic blood (541, Similarly, although IL-1b has been implicated in the induc542, 844, 998). Local administration of IL-1ra at the site tion of IL-6 and the cachexia after LPS, neither of these
of LPS injection inhibits the rise in plasma IL-6 levels responses are inhibited in IL-1b-deficient mice (10, 247,
(541), again suggesting a causal relationship between the 250, 438, 1001). Studies in IL-1R1-deficient mice demonproduction of IL-1 and IL-6 after LPS. In light of these strated that IL-1R1 is essential for all the IL-1 mediated
data, it is surprising that recent experiments performed signaling events examined (fever, induction of IL-6, inducin mice have shown that the plasma IL-6 response to LPS tion of E-selectin) (442), but these animals display normal
appears normal in mutant mice lacking either IL-1b (10, fever and cachexia induced by intraperitoneal LPS (442,
1001) or IL-1R1 (463). Whether this indicates important 463). Not surprisingly, therefore, investigations of the
roles for IL-1a (or IL-1g) and the IL-1R2 (or novel IL-1 HPA axis response to LPS in these mutant mice have also
receptors) clearly warrants investigation.
failed to confirm a role of IL-1 in the elaboration of this
Comparisons of the time courses of cytokine produc- HPA axis response (10, 247, 250).
tion and HPA axis activation produced by systemic LPS
Investigations of the effect of immunoneutralizing ILhave produced somewhat conflicting data. The concentra- 6 and TNF-a have also suggested physiological roles for
tions of TNF-a, IL-1b, and IL-6 in plasma have been re- these cytokines in the HPA axis secretory response to
ported to lag behind the rise in plasma ACTH after intra- LPS. Although inhibition of either IL-1, IL-6, or TNF-a
arterial LPS regardless of LPS dose (290). However, after abrogates the ACTH response to larger doses of LPS in
low doses of LPS administered intravenously, elevations mice, Perlstein et al. (640) found that only anti-IL-6 antiin plasma TNF-a coincide with the secretion of ACTH, bodies were completely effective at reducing plasma
whereas at higher doses, elevations in plasma TNF-a oc- ACTH concentrations produced by lower doses in mice.
cur after the initial rise in plasma ACTH (223). We find In contrast, IL-6-deficient mice show a normal plasma corthat after LPS (5 mg/kg iv), the TNF-a profile in blood ticosterone response to a high dose (1 mg/kg ip) of LPS
precedes that of ACTH, with a time of onset and peak (254). In rats, anti-TNF-a antisera inhibits the plasma
that occurs 15 min before those of plasma ACTH (889).
ACTH response to both low and high doses of intravenous
The importance of cytokines in the ACTH response LPS (223, 889), whereas TNF-R1-deficient mice display a
to LPS was first directly indicated by the pronounced inhi- normal corticosterone response to intracerebroventricubition of this response when mice were pretreated with lar LPS (2.5 mg).
IL-1 receptor antibodies (690). Further studies showed
From the above discussion, it is apparent that the
that destruction of macrophages produced a marked re- precise role played by IL-1, IL-6, and TNF-a in the elaboraduction in circulating IL-1 levels in response to a high tion of the HPA axis response to LPS is not fully underdose of LPS (2.5 mg/kg iv) and a 40% inhibition of the stood. That each of these three cytokines affects the synACTH response to a small dose of LPS (2.5 mg/kg iv) (195). thesis and secretion of the other, that each is capable of
Either anti-IL-1 receptor antibodies given to mice (640) enhancing the others’ effect in a synergistic manner, and
16
ANDREW V. TURNBULL AND CATHERINE L. RIVIER
Volume 79
the possible redundancy of each of these cytokines undoubtedly contribute to the lack of clarity of the various
studies described. Overall, however, the evidence presented seems to indicate that, depending on experimental
paradigm, one or more of IL-1, IL-6, and/or TNF-a may
contribute to the HPA axis secretory response to LPS.
C. Local Inflammation
/ 9j0c$$oc11
P13-8
FIG. 2. Plasma concentrations of ACTH, corticosterone, and interleukin-6 (IL-6) in rats after (intramuscular) injection of 50 ml/100 g
body wt of either saline (s) or turpentine (j). Thick, broken bar represents dark cycle (lights out). [From Turnbull and Rivier (884); q The
Endocrine Society.]
IL-6 response to turpentine is completely absent in either
of these mutants (463, 1001). Interestingly, however, ICEdeficient mice fail to generate mature IL-1b in response
to LPS (469) but exhibit normal production of mature
IL-1b and display normal plasma IL-6 responses, when
injected with turpentine (248). These latter studies clearly
indicate that distinct molecular mechanisms of IL-1b and
IL-6 elaboration are operative during systemic (LPS) and
local (turpentine) inflammation.
Interleukin-6 has been demonstrated to be an obligatory mediator of a number of acute phase responses to
local inflammation induced by turpentine, including fever,
cachexia, and increased hepatic protein synthesis (87,
247, 254, 432, 437, 603). Because IL-6 is the only identified
cytokine in the systemic circulation in significant quantities after turpentine, it seems likely that it is the major
circulating signal to the brain. Nevertheless, there is only
11-25-98 11:16:36
pra
APS-Phys Rev
Downloaded from on April 23, 2014
A number of studies have investigated the mechanisms by which acute local inflammation produces activation of the HPA axis (169, 879, 881, 884, 965). One muchstudied model consists of the subcutaneous or intramuscular injection of the irritant turpentine into the mouse
or rat. Injection of turpentine produces a localized inflammation that is characterized by a centrally necrotic,
well-defined abscess and increased vascular permeability
(476, 620, 829, 978). This paradigm has been used to investigate a multitude of acute phase responses to local inflammation including fever, hypermetabolism, cytokine
synthesis and secretion, protein metabolism, anorexia,
neuroendocrine alterations, and hepatic acute phase protein synthesis (27, 167–169, 247, 286, 432, 603, 965, 978,
1001).
The intramuscular injection of turpentine produces
a biphasic activation of the HPA axis in the rat (see Fig.
2). Increased ACTH secretion shortly after turpentine administration appears to be due to activation of nociceptive
sensory afferents (879) and is not related to cytokine synthesis or secretion (247, 254). By 3 h after turpentine,
plasma ACTH concentrations return temporarily to
around those of control animals (Fig. 2). This is followed
by a second rise in plasma ACTH and corticosterone concentrations that parallels the development of the local
inflammation, lasts for Ç24 h, and is the HPA axis response to the actual local inflammation per se (884). Turpentine-induced inflammation produces a long-lasting (at
least 12 h) increase in the expression of Fos in the PVN
(696), and the second rise in plasma ACTH concentration
is completely reversed by the administration of anti-CRF
antiserum (884), indicating the importance of hypothalamic CRF secretion to the plasma ACTH response.
The generation and role of cytokines during acute
phase responses produced by turpentine injection has
been well studied. Turpentine-induced local inflammation
elicits a marked elevation in the plasma levels of IL-6,
but not of IL-1 or TNF-a (167, 248, 495, 881). However,
concentrations of IL-1 are elevated at the site of local
inflammation induced by turpentine (248), and inhibition
of either TNF-a (167) or IL-1 (286, 495, 879) action markedly reduces the levels of IL-6 in blood, suggesting that
both these cytokines stimulate the secretion of IL-6 at the
local inflammatory site. Although IL-1b or IL-1R1-deficient
mice display normal IL-6 responses to LPS, the plasma
January 1999
REGULATION OF HPA AXIS BY CYTOKINES
D. Physical and Psychological Stress
The elaboration of cytokines appears not to be restricted to injurious, inflammatory, and infectious insults,
/ 9j0c$$oc11
P13-8
FIG. 3. Effects of turpentine-induced local inflammation on plasma
corticosterone in either wild-type (IL-6 ///) or IL-6-deficient (IL-6 0/
0) male mice. Plasma samples were collected at 9 h after either saline
or turpentine administration. *** P õ 0.002. (From A. V. Turnbull, S. J.
Hopkins, S. Prehar, and C. L. Rivier, unpublished data.)
since recent studies have indicated that IL-1, IL-6, and
brain-derived neurotrophic factor (BDNF) synthesis and/
or secretion are altered during acute physical or psychological stresses (462, 536, 546, 777, 778, 797, 1002). For
example, plasma IL-6 levels are elevated in rats by exposure to a novel environment (462, 561), conditioned aversive stimuli (1002), electroshock (883, 1002), or restraint
(1002). Furthermore, IL-6 mRNA is elevated in the midbrain and IL-6R mRNA is diminished in the midbrain and
hypothalamus 4–24 h after restraint stress in the rat (779),
whereas immobilization causes increases plasma IL-6 concentrations and elevated hepatic and splenic IL-6 mRNA
expression in mice (417). In humans, plasma IL-6 levels
increase rapidly after treadmill exercise with peak increases apparent at 15 and 45 min (622). The increases in
plasma IL-6 after physical and/or psychological stresses
in rats are much more rapid (within 15 min) than those
observed after either local (turpentine, 2–3 h) or systemic
(LPS, 45–60 min) inflammations and appear to be mediated by an action of catecholamines (196, 800, 852, 910).
However, increased blood levels of IL-6 do not appear to
directly contribute to the HPA axis response to acute
stress, since the rise in plasma IL-6 levels is only modest,
and although plasma IL-6 levels increase rapidly, they still
lag behind plasma ACTH responses (883, 1002). Furthermore, Ruzek et al. (716) reported that IL-6-deficient mice
display an increase in plasma corticosterone levels in response to restraint stress that is comparable to that found
in wild-type mice.
Acute restraint or immobilization has also been reported to increase hypothalamic IL-1b mRNA (546, 828),
IL-1 protein (778), and IL-1RA mRNA (828) within 30 min
of commencement of stress. In addition, chronic physical/
psychological stress elevates both plasma IL-1b concentrations in rats (536) and humans (814) and hypothalamic
11-25-98 11:16:36
pra
APS-Phys Rev
Downloaded from on April 23, 2014
limited evidence indicating that IL-6 mediates the HPA
axis response to turpentine. Plasma IL-6 concentrations
correlate well with the second rise in plasma ACTH and
corticosterone levels (879, 884) (see Fig. 2), which given
the known stimulatory effects of IL-6 on HPA axis secretory activity (327, 499, 519, 520, 527, 574, 575, 809) suggests that IL-6 is a likely circulating mediator of this neuroendocrine response to turpentine (879, 884). However,
one report shows that immunoneutralization of systemic
IL-6 does not influence the rise in plasma corticosterone
in mice 12 h after turpentine (603). It should be noted
that in this study (603), administration of IL-6 antibodies
resulted in enhanced rather than diminished biological
activity of IL-6 in plasma, a finding that has been commonly reported (reviewed in Ref. 760), and which casts
doubt on the significance of the lack of effect on plasma
corticosterone levels in response to turpentine. Studies in
both IL-1b- and IL-6-deficient mice have shown that neither IL-1b nor IL-6 is an obligatory mediator of the elevation in plasma corticosterone concentrations shortly (1.5–
2 h) after turpentine (247, 254). However, the time point
chosen for investigation by these authors corresponds to
the initial, pain-related increase in HPA axis secretory
activity (see Fig. 2), and no observation was made at later
time points corresponding to the HPA axis response to
the actual local inflammatory reaction (6–24 h). We have
recently obtained evidence that indicates that IL-6 is an
important mediator of the HPA axis response to local inflammation (A. V. Turnbull, S. J. Hopkins, S. Prehar, and
C. L. Rivier, unpublished data). At 9–12 h after turpentine,
when plasma IL-6 levels are maximally elevated in control
animals (see Fig. 2), IL-6-deficient mice display a very markedly reduced (two-thirds inhibited) plasma corticosterone
response (Fig. 3). These data together with those demonstrating reduced fever (437), acute phase protein synthesis
(432), and cachexia (432, 437) in IL-6-deficient mice collectively indicate that IL-6 is an important circulating mediator
during local inflammation, acting as an ‘‘SOS’’ signal and
inducing a variety of acute phase responses.
There is also evidence for the involvement of other
cytokines in this response. Although intramuscular turpentine in rats appears not to elevate mRNA for either IL1b, IL-6, or TNF-a within the CNS or pituitary, intracerebroventricular administration of either a neutralizing antiTNF-a antiserum or a dimeric soluble TNF-a receptor
construct markedly inhibits ACTH secretion 6–9 h after
turpentine in the rat (881). This indicates an important,
although as yet unexplained, role of cerebral TNF-a in
this response. The extent to which other CNS-derived cytokines may contribute to the enhanced secretory activity
of the HPA axis is presently unknown.
17
18
ANDREW V. TURNBULL AND CATHERINE L. RIVIER
E. Basal Hypothalamic-Pituitary-Adrenal Activity
Perhaps even more surprising than the demonstrated role of cytokines in the regulation of the HPA
axis in response to noninflammatory or noninfectious
stress is the recent finding that the cytokine LIF may
contribute to the regulation of HPA axis secretion under
basal, nonstress conditions. Although inhibition or genetic deletion of IL-1, IL-6, or TNF-a does not significantly affect basal plasma ACTH or corticosterone concentrations, recent studies by Melmed and colleagues
(6, 7, 667, 926) have demonstrated that a member of
the gp130 signaling family of cytokines, LIF, plays an
important role in the regulation of basal ACTH secretion. Either normal or 36-h fasted mutant LIF-deficient
mice have lower basal plasma concentrations of ACTH
than wild-type controls, an effect that can be reversed
/ 9j0c$$oc11
P13-8
by administration of exogenous LIF (6). This suggests
that either LIF is important in the development and
maturation of the HPA axis or that it represents a tonic
ACTH secretagogue in the adult animal.
IV. CYTOKINE ACTIONS ON THE CENTRAL
NERVOUS SYSTEM, PITUITARY,
AND ADRENAL
The effects of both administration of cytokines to
normal, healthy subjects and the consequences of inhibiting cytokine action during infectious, inflammatory, or
stressful threats imply that cytokines may play a physiological role in the regulation of the secretory activity of
the HPA axis. However, cytokines also produce a number
of systemic acute phase responses that themselves could
elicit HPA activation, raising the question of whether the
effects of cytokines on HPA axis secretory activity are
direct or secondary to stress produced by other acute
phase responses. For example, IL-1b causes fever, sickness behavior, increases in heart rate, increased blood
flow to certain vascular beds, activation of the sympathetic nervous system, and changes in intermediary metabolism. These physiological changes are themselves
challenges to the maintenance of homeostasis, and each
could, if pronounced, activate secretion by the HPA axis.
The effects of some cytokines on plasma ACTH concentrations have nevertheless been dissociated from a number of these other acute phase responses. For example,
in the original account by Besedovsky et al. (70), IL-1
induced activation of the HPA axis of mice kept at an
ambient temperature that did not result in these animals
mounting a febrile response. Indeed, IL-1b analogs with
markedly reduced pyrogenicity still stimulate ACTH secretion (572). In addition, enhanced ACTH secretion is
observed after peripheral administration of IL-1b at doses
of IL-1b that have no, or only modest, effects on the secretion of other hormones, such as luteinizing hormone, catecholamines, and prolactin, whose secretion is markedly
altered by many other types of stressors (399, 695, 886).
Similarly, doses of IL-6 that markedly elevate plasma
ACTH and cortisol concentrations in humans have only
moderate effects on other acute phase responses (519,
520). Finally, studies described in section IIIE clearly indicate that regulation of ACTH secretion by LIF can be
demonstrated in healthy animals. These findings suggest
cytokines target the HPA axis in a specific manner.
The original Science papers of 1987 suggested that
the influence of IL-1 on pituitary ACTH secretion was
attributable to actions either at the level of hypothalamic
CRF release (55, 730) or directly on the pituitary itself
(62) to stimulate ACTH secretion. Furthermore, although
original hypotheses regarding a lymphoid-adrenal axis
whereby ACTH produced by lymphocytes represented the
11-25-98 11:16:36
pra
APS-Phys Rev
Downloaded from on April 23, 2014
IL-1b mRNA in mice (837). On the basis of studies showing that intrahypothalamic administration of IL-1ra produces a significant reduction of the plasma ACTH response to immobilization in rats, a role for IL-1 in the
regulation of the HPA axis response to a stress unrelated
to infection or inflammation has now been proposed (777,
778). Indeed, other studies have shown that intracerebroventricular administration of IL-1ra before inescapable
shock blocks the subsequent interference with escape
learning and enhancement of fear conditioning normally
produced by such a stressor (511), suggesting that IL-1
may also mediate some behavioral effects of noninflammatory/infectious stresses.
The findings that cytokines may be elaborated quickly
(within minutes) in response to a stimulus that does not
appear to result in tissue damage or infection is novel
and suggests a role of cytokines in homeostasis that has
previously been unrecognized. It should be pointed out
that many of the physiological responses to inflammatory/
infectious stressors and psychological/physical stressors
are common (e.g., activation of the HPA axis, suppression
of reproduction, certain changes in behavior, fever, and
reduced appetite). Indeed, psychological/physical stress
produces many aspects of the acute phase response to
sickness. Furthermore, although humans experience psychological/physical stress commonly without the presence of inflammatory/infectious stress, psychological/
physical stress commonly precedes inflammatory/infectious stress in animals in the wild (e.g., predator-prey
experiences, shortage of food/water supply). That the
rapid elaboration of cytokines may contribute to the signal-generating physiological (e.g., neuroendocrine) responses to psychological/physical stressors (778) suggests that, mechanistically, responses to such stressors
may not differ markedly from responses to inflammatory/
infectious stressors.
Volume 79
January 1999
REGULATION OF HPA AXIS BY CYTOKINES
A. Evidence That Cytokines Activate the
Hypothalamic-Pituitary-Adrenal Axis Primarily
at the Level of the Central Nervous System
1. Cytokine receptors within the CNS
Receptors to many cytokines have been localized
within the CNS or described in primary cell cultures or
cell lines derived from brain tissue. These include receptors for IL-1, IL-2, IL-3, IL-4, IL-6, IL-7, interferons, TNF,
growth factor, CSF, growth factors, and neurotrophins
(for reviews, see Refs. 65, 344, 762) (Table 6). Of most
relevance to this review article is the distribution throughout the rodent CNS of the receptors for the cytokines IL-
/ 9j0c$$oc11
P13-8
1, IL-6, and TNF-a, and these are discussed in more detail
here.
A) IL-1 RECEPTORS IN THE CNS. Early studies investigating the localization of IL-1 receptors in the brain indicated
a fairly widespread distribution. High levels of specific
binding of 125I-labeled IL-1b were found in the choroid
plexus, dentate gyrus, hippocampus, cerebellum, and olfactory bulb, with low levels in the hypothalamus and ME
in rat brain slices (251). Specific binding of 125I-labeled IL1b to membrane preparations of rat hypothalamus and
cortex were also reported (398). Subsequent studies have
confirmed the existence of IL-1 receptors within the rodent CNS, and IL-1R1 mRNA or IL-1 binding have been
demonstrated on neurons (see below), astrocytes (31, 714,
868), cerebrovascular endothelia (904), neuroblastoma
cells (625), and glioblastoma cells (303), but not on microglia (31, 868). However, the localization within the rat
brain appears to differ somewhat from that which was
originally reported, in particular with respect to the presence of IL-1 receptors within the hypothalamus. Furthermore, there are marked differences in the distribution of
IL-1 receptors in rat and mouse brains.
Overall, the mouse brain exhibits very low densities
of IL-1 receptors as assessed by binding of 125I-labeled IL1a, IL-1b, or IL-1ra. However, very high levels of labeling
are found consistently in the hippocampus (dentate gyrus,
but not CA1 to CA4 pyramidal regions), choroid plexus,
and meninges (30, 841, 848). Within the dentate gyrus, IL1 binding appears to be predominantly to neurons (30,
848). Most notably, none of the more recent studies reported substantial IL-1 binding in the hypothalamus (30,
841, 848). In situ hybridization histochemical analyses of
mouse brains have indicated that IL-1R1 mRNA is expressed predominantly in the granule cell layer of the
dentate gyrus, the entire midline raphe system, the choroid plexus, and endothelial cells, but not in the hypothalamus (177, 178). In contrast, IL-1R2 mRNA has been undetectable in normal mouse brain using in situ hybridization
histochemical procedures (202).
As in the mouse, the rat choroid plexus, but not hypothalamus, shows significant IL-1 binding (516, 845, 848).
However, in marked contrast to the robust IL-1 binding
in the mouse hippocampus, the rat hippocampus displays
no binding of either IL-1a, IL-1b, or IL-1ra (using either
heterologous or homologous ligands) (516, 845, 848). Similarly, in situ hybridization signal for IL-1R1 mRNA over
the rat hippocampus has been described as either weak
(972, 982) or background (238), with no study demonstrating significant signal in the dentate gyrus. The IL-1R1
mRNA in rat brain is confined largely to nonneuronal cells,
with the ependymal cells lining the ventricular system,
the choroid plexus, the leptomeninges, and in particular
endothelial/perivascular cells being the major sites of IL1 receptor expression (178, 238, 969, 970, 972, 982). A few
neuronal groups in the rat brain do, however, display low
11-25-98 11:16:36
pra
APS-Phys Rev
Downloaded from on April 23, 2014
link between activation of the immune system and adrenal
hormone secretion (793) has fallen out of favor, other
studies (e.g., Ref. 698) suggest that cytokines may act on
the adrenal directly.
The following sections describe the evidence that indicates the potential site(s) of cytokine action on HPA
axis secretory activity. In presenting arguments for each
level of the HPA axis (CNS, pituitary, and adrenal), we
consider three lines of evidence. First, we describe the
localization of cytokine receptors within the HPA axis.
The a priori condition for a particular cytokine to influence HPA axis secretory activity by an action at a particular level is the expression of functional receptors for that
cytokine by the tissue under consideration (e.g., hypothalamus) or at least in functionally related tissues (e.g., other
regions of the CNS which send projections to the hypothalamus). Second, we describe the expression of cytokines by tissues of the HPA axis. It was originally hypothesized that the influence on neuroendocrine function of
endogenous cytokines resulted from the exposure of the
CNS and pituitary to cytokines produced by circulating
or tissue-resident cells such as macrophages, monocytes,
fibroblasts, and B and T lymphocytes. However, studies
over the last decade have demonstrated that many IL,
chemokines, TNF, IFN, CSF, and growth factors are produced by numerous cell types, including those found
within neuroendocrine systems. Therefore, it appears that
cytokines may play a paracrine or autocrine role in the
regulation on neuroendocrine function, as they do in the
immune system. In general, levels of cytokines in resting,
healthy, unstressed animals or humans are low, but the
expression of a number of cytokines can increase dramatically during injury, infection, disease, or physical/psychological stress. The following sections therefore outline
what is known about the synthesis of cytokines within
the CNS, pituitary, and adrenal and how this expression
may be regulated. Finally, we discuss the numerous in
vivo and in vitro studies that have investigated the effects
of cytokines on each component of the HPA axis.
19
20
ANDREW V. TURNBULL AND CATHERINE L. RIVIER
TABLE
Volume 79
6. Cytokines and their receptors in the CNS
Cytokine
IL-1a
IL-1b
IL-1ra
IL-2
IL-3
IL-4
IL-5
IL-6
IL-7
IL-8
IL-10
IL-11
IL-12
IL-13
IL-15
MIP-1b
LIF
CNTF
TNF-a
TNF-b
MIF
IFN-a
IFN-b
IFN-g
IGF-I
IGF-II
NGF
EGF
bFGF
TGF-a
TGF-b
Activin
G-CSF
M-CSF
GM-CSF
SCF
Neurotrophins (NGF,
BDNF, NT-3)
/
/
/
/
/
/
/
/
/
/
/
/
/
/
/
/
/
/
/
/
/
/
/
/
/
/
/
/
/
/
/
/
/
/
/
Induced by
Systemic LPS
Receptor
Reference No.
/
/
//0
/
/
/
/
/
/
/
/
/
//0
/
30, 178, 238, 277, 672
30, 101, 124, 178, 238, 277, 453, 454, 644, 901, 902
269, 277, 353, 472, 841, 971
18, 448
16, 252, 430
407, 490, 977
736
273, 274, 277, 453, 644, 746, 748, 749, 900, 988
538
473, 721, 834, 979
349, 971
210
624
971
316, 460
547
926, 987
208, 823
102, 277, 414, 453, 644, 967
559
590
99, 364, 834, 983, 984
834
403, 484, 644, 834
4, 43, 93, 391, 465, 515, 704, 723, 956
93, 391, 465, 533, 704, 723
32, 240, 288, 425, 670, 811
245, 259, 504
228, 360, 486
259, 502, 503, 960
486, 646
136, 697
12, 593, 834
8, 12, 154, 266, 595, 663, 834
12, 513, 593, 663, 834
313, 411, 514, 706, 834, 973, 1000
38, 125, 184, 185, 475, 797, 798
/
0
/
/
/
/
/
/
/
/
/
/
/
/
/
/
/
/
/
/
/
/
/
/
/
/
Definitions are as in Table 3. Reference numbers with regard to IL-8 include the homologous cytokines cinc/gro/NAP-1.
to moderate levels of IL-1R1 mRNA: the basolateral nucleus of the amygdala, the arcuate nucleus of the hypothalamus, the trigeminal and hypoglossal motor nuclei,
and the area postrema (238). As in the mouse, IL-1R2
mRNA is undetectable in the rat brain by in situ hybridization histohemistry (592), although the expression of IL1R2 mRNA is induced by systemic treatment with kainic
acid (592).
The low levels of IL-1R1 mRNA observed in the hippocampal formation of the rat are in apparent contrast to
the localization in the rat of IL-1RAcP. The IL-1RAcP
mRNA is highly expressed in both mouse (304) and rat
brain (482). In situ hybridization studies of rat brains demonstrated that in contrast to IL-1R1 mRNA, IL-1RAcP
mRNA is highly expressed in the granule cell layer of
dentate gyrus, a region in which IL-1R1 mRNA is expressed in mice but not in rats (482). Furthermore, IL1R1 and IL-1RAcP mRNA are differentially regulated by
peripheral LPS treatment (271, 353). It therefore seems
/ 9j0c$$oc11
P13-8
possible that the IL-1RAcP in the brain may also serve
as an accessory protein to novel IL-1 receptor(s) or has
functions unrelated to IL-1 receptor signaling.
In addition to the radioligand binding and in situ hybridization studies described above, RNase protection
assay (RPA) (238, 280, 352, 353) and RT-PCR (270, 625)
procedures have been used fairly extensively to investigate the distribution and regulation of IL-1R1 and IL-1R2
mRNA in both rat and mouse brain. Parnet et al. (625)
found transcripts for both IL-1R1 and IL-1R2 in whole
mouse brain but showed that the murine neuroblastoma
cell line C1300 expresses only IL-1R1 mRNA. Indeed, Gabellec et al. (270) also demonstrated transcripts for both
IL-1 receptors in the mouse brain but found that IL-1R1
mRNA was the predominant species in brain, whereas IL1R2 mRNA was the most abundant in the spleen. Although
previous work showed that systemic LPS reduces IL-1
binding in rat and mouse brain (29, 321, 517, 840, 842,
843), RT-PCR and RPA analyses have shown that either
11-25-98 11:16:36
pra
APS-Phys Rev
Downloaded from on April 23, 2014
Cytokine
January 1999
REGULATION OF HPA AXIS BY CYTOKINES
/ 9j0c$$oc11
P13-8
include the hypothalamus and, of particular interest, the
PVN (900). Gp130 mRNA is also expressed by cerebrovascular elements of LPS-treated rats (900).
C) TNF RECEPTORS IN THE CNS. Currently, there is little
known of the distribution or physiological role of either
TNF receptor within the CNS. Studies utilizing radioligand
binding of 125I-rmTNF-a to sections of whole mouse brain
have demonstrated only weak specific binding throughout
the entire surface of brain tissue (967), and binding assays
of tissue homogenates have shown specific binding in the
mouse brain stem, cortex, thalamus, basal ganglia, and
cerebellum (414). In vitro studies have shown that both
TNF-R1 and TNF-R2 mRNA are present in mouse cerebrovascular endothelium (49). In microglia, astrocytes, and
oligodendrocytes, both TNF-R1 and TNF-R2 can be expressed in humans (788, 823a, 834, 963), whereas at least
one receptor subtype is present in rats and mice (17, 209,
855). Both undifferentiated and differentiated clonal murine neuroblastoma cells (N1E cells) express TNF-R1, but
not TNF-R2, mRNA (787). The TNF-R1 immunoreactivity
has also been demonstrated in neurons in the substantia
nigra of humans and both TNF-R1 and TNF-R2 immunoreactivities shown in human hippocampal and striatal neurons (92). However, there is presently no evidence that
either TNF-a receptor subtype is localized to hypothalamic regions directly involved in the regulation of HPA
axis secretory activity.
2. Cytokine expression in the CNS
The production and actions of cytokines within the
CNS have been reviewed extensively in a number of recent excellent review articles (65, 344, 710, 711, 745, 762),
and only a brief overview pertinent to the discussion of
cytokine influence on the HPA axis is given here.
A) BASAL EXPRESSION. Many cytokines are synthesized
within the brain (see Table 6), although in most cases
their expression in healthy, stress-free subjects is low.
Nonetheless, a number of studies have reported the distributions of IL-1, IL-6, and TNF-a immunoreactive or biologically active protein or mRNA in the brains from normal
untreated subjects. In the human brain, IL-1b immunoreactivity is found within neuronal elements of the hypothalamus, including periventricular regions, the pPVN, and
the ME (101). This distribution is consistent with a role
of IL-1b as a neuroregulator of acute phase responses,
and in particular, of the HPA axis (101). Although studies
in rats also detected neuronal immunoreactive IL-1b in
similar hypothalamic regions, more prominent staining
was found in extrahypothalamic sites, particularly the hippocampus (454). Interleukin-1b immunoreactivity has
also been reported in the rat hypothalamus (672). Interleukin-1 biological activity in the rat brain has been either
undetectable (264, 655) or detected in brain stem, cerebral
cortex, diencephalon, and hippocampal homogenates (32,
11-25-98 11:16:36
pra
APS-Phys Rev
Downloaded from on April 23, 2014
LPS or IL-1b increases the expression of IL-1R1 and IL1R2 mRNA (270, 271, 281, 353, 354, 357, 646, 668). These
differences between binding studies and mRNA analyses
presumably relate to either increased receptor occupancy
after LPS treatment (see sect. IVA2B) or decreased translation of receptor mRNA into receptor protein.
B) IL-6 RECEPTORS IN THE CNS. Specific IL-6 binding sites
have been demonstrated in both astrocytoma and glioblastoma cell lines (835), as well as in extracts of bovine
hypothalamus (170). Messenger RNA encoding the a-subunit of the IL-6 receptor (IL-6Ra) has also been detected
in neurons, microglia, and astrocytes from either normal
brain tissue, primary cell cultures, or tumor cell lines (735,
834). The IL-6Ra mRNA is expressed in several brain regions in the untreated rat (273, 274, 746, 749, 900, 988).
The IL-6Ra mRNA is most abundant in the pyrimadal cells
of the CA1 and CA4 regions of the hippocampus and in
the granule cell layer of the dentate gyrus (900) and has
also been detected in the hypothalamus, cerebellum, hippocampus, striatum, neocortex, and pons/medulla (273,
274, 746, 749, 988). Specific hybridization signals are observed in glial cells of the lateral olfactory tract, in ependymal cells of the olfactory and anterior lateral ventricle,
and in neurons of the piriform cortex, medial habenular
nucleus, neocortex, hippocampus, and hypothalamus
(746). Within the hypothalamus, IL-6Ra mRNA is present
in the ventromedial and dorsomedial regions including the
periventricular hypothalamus and in the medial preoptic
nucleus. However, the anterior hypothalamus and PVN
display no specific hybridization signal for IL-6Ra mRNA
(746).
The expression of IL-6Ra mRNA in the rat brain appears to be developmentally regulated, with marked increases in its expression, particularly in the striatum, hypothalamus, hippocampus, and neocortex, between 2 and
20 days of age (273, 274). Furthermore, LPS administration markedly elevates IL-6Ra mRNA levels in several rat
brain regions (area postrema, bed nucleus of the stria
terminalis, amygdala, cerebral cortex, claustrum, hippocampus, ME, piriform cortex, septohippocampal nucleus,
PVN, SFO, and OVLT) (900). Furthermore LPS (intraperitoneal) or IL-1b (intravenous) stimulates the expression
of IL-6Ra mRNA over blood vessels throughout the brain
(900).
In addition to IL-6Ra, the signal-transducing component of the IL-6 receptor, gp130, has also been localized
within the rat CNS (900, 927). Gp130 is found in glial,
neuronal, oligodendrocyte, and ependymal cell types. The
distribution of immunoreactive gp130 in the rat brain
overlaps that which has been observed for IL-6Ra but is
more widespread, consistent with its role in signal transduction for other members of the IL-6 family (927). Indeed, gp130 mRNA has been detected throughout the normal rat brain, with positive hybridization signals detectable in almost all brain areas (900). These brain areas
21
22
ANDREW V. TURNBULL AND CATHERINE L. RIVIER
/ 9j0c$$oc11
P13-8
demonstrated, microglia appear to be the major ‘‘brainresident’’ cell type that synthesizes IL, chemokines, TNF,
and IFN, although vascular cells, astrocytes, and neurons
also contribute to cytokine production.
Of particular significance to the field of immune-nervous system interactions has been the hypothesis that the
synthesis of cytokines in brain may be induced by stimuli
other than those resulting in direct cellular challenge to
the CNS, and consequently that cytokines may act as neuroregulators within the brain in a manner akin to classical
neuropeptides. Indeed, in response to the peripheral administration of LPS, the CNS expression of a number of
cytokines is elevated (see Table 6). In response to large
doses of LPS (0.4–4 mg/kg ip or iv), the mRNA encoding
IL-1a, IL-1b, IL-1ra, IL-6, and TNF-a are elevated in homogenates of several regions of the mouse brain as assessed by RT-PCR and Northern blot hybridization methodologies (28, 269, 277, 453, 570). These elevations have
been noted within 1 h, and peak at Ç6 h. However, an
important question regarding the induction of cytokines
in the brain by LPS is whether the stimulus causing increased synthesis is of peripheral origin, because large
doses of LPS could, for example, penetrate the BBB in
sufficient quantities to stimulate cytokine synthesis directly. Indeed, LPS is a potent stimulus of IL-1, IL-6, and
TNF-a synthesis after its intracerebroventricular administration (197, 338, 655, 881) and induces cytokine synthesis
by glial cells in cell culture (770). In addition, large doses
of LPS may disrupt the BBB (91, 199, 483, 496, 781, 878),
thus permitting the entrance from the periphery of cells
(e.g., macrophages) that may contribute to the cytokine
signal. The majority of RT-PCR studies have been performed using high doses of LPS, and the fact that these
have not fully addressed issues regarding contamination
with blood cells and possible disruption of the BBB makes
conclusions difficult. However, recent studies by Pitossi
and co-workers (643, 644) used semi-quantitative RT-PCR
to measure cytokine production after the injection of LPS
to mice, at a dose (20 mg/kg ip) that is reported not to
influence BBB integrity. Importantly, these authors also
quantified possible contaminaton by peripheral blood
cells. This work showed marked induction of IL-1b, IL-6,
TNF-a, and IFN-a mRNA in several brain regions (cortex,
cerebellum, thalamus/striatum, hippocampus, brain stem,
and hypothalamus), with peak increases observed 2–4 h
after LPS administration (644). Because the authors concluded that these increased signals could not be accounted for by contamination with peripheral blood cells,
it is clear that these cytokines are induced in brain by
doses of LPS that do not disrupt the BBB.
A number of in situ hybridization and immunocytochemical studies have investigated the distribution of
IL-1b mRNA or IL-1b protein after systemic (iv or ip)
LPS in rats and rabbits and have yielded very similar
results. ‘‘Barrier-related’’ regions have been the most
11-25-98 11:16:36
pra
APS-Phys Rev
Downloaded from on April 23, 2014
656, 778). In the normal rat or mouse hypothalamus, IL1b is detectable using sensitive immunoassays (314).
However, the majority of in situ hybridization studies have
found that the brain parenchyma lacks a readily distinguishable IL-1b mRNA signal (124, 338, 546, 981), whereas
constitutive expression of IL-1b in the cerebrovasculature
of control animals is readily observable (969, 970). Similarly, the majority of Northern blot hybridization or RTPCR studies have found extremely low or undetectable
levels of IL-1a or IL-1b mRNA in the brains of control rats
or mice (197, 277, 338, 453, 507, 644), although sufficient
sensitivity to demonstrate small diurnal variations in rat
brain IL-1b mRNA expression was achieved in one study
(838).
Messenger RNA of the gene encoding ICE, the enzyme responsible for cleavage of pro-IL-1b to mature, active IL-1b, has been demonstrated in murine microglia
(990), whole brain homogenates (401), homogenates of
hypothalamus and hippocampus (452, 865, 866), and
blood vessels (arterioles and venules) throughout the
brain parenchyma (970) of control rats. Interleukin-1 receptor antagonist mRNA is also present within the rat
brain (269, 281, 353–357, 472, 485, 970, 971), with positive
in situ hybridization signal present in the hypothalamus
(particularly the PVN), hippocampus, cerebellum, choroid
plexus, and blood vessels throughout the brain (472, 970).
Tumor necrosis factor-a immunoreactivity is found
in the hypothalamus, caudal raphe nuclei, and along the
ventral surface of the brain in the pons and medulla in the
normal mouse brain (105). On the basis of morphological
observations, the majority of staining observed appears
neuronal, with two principal fiber pathways being noted:
a periventricular pathway which coursed along the ventricular system and a pathway associated with the medial
forebrain bundle (105). Within the hypothalamus, the PVN
represents one of the terminal fields of fibers originating
from the most intensely stained cell groups within the bed
nucleus of the stria terminalis. On the basis of in situ
hybridization studies, there is only a weak signal over
regions that coincided with immunoreactive TNF-a (105).
Finally, IL-6 mRNA has been either undetectable (900) or
shown to be colocalized with IL-6Ra mRNA within several
regions of the normal rat brain, including hypothalamus,
cerebellum, hippocampus, striatum, neocortex, and pons/
medulla (746).
B) INDUCED EXPRESSION. The expression of a number
of cytokines within the CNS increases dramatically upon
cellular damage. Accordingly, local concentrations of IL1b, IL-6, and TNF-a, in particular, are elevated during
CNS bacterial or viral infections, brain trauma, cerebral
ischemia, and convulsions (344, 745). In addition, their
expression is increased during a number of chronic CNS
disorders such as multiple sclerosis, Down’s syndrome,
and Alzheimer’s disease (344, 745). In general, when induction of cytokine synthesis within the brain has been
Volume 79
January 1999
REGULATION OF HPA AXIS BY CYTOKINES
/ 9j0c$$oc11
P13-8
tine-induced inflammation (Ref. 881 and unpublished observations).
Recent studies have suggested that cytokine synthesis in brain may also be induced by stressors unrelated
to infection or inflammation. Hypothalamic expression of
IL-1b mRNA (546, 828), IL-1ra mRNA (828), and IL-1 bioactivity (778) is increased within 30 min of immobilization
stress in the rat, and IL-6 mRNA is elevated in the midbrain
4–24 h after restraint stress (779). Similar paradigms produce increases in IFN-g mRNA expression in mouse brain
homogenates (849). Acute (2 h) or repeated immobilization stress in rats increases BDNF mRNA expression in
the pPVN and the lateral hypothalamus (797) and decreases BDNF mRNA in the hippocampus, whereas repeated stress increases NT-3, but not NT-4, mRNA in the
hippocampus (798).
3. Cytokine actions at the level of the CNS
Consistent with the CNS as a primary target of IL-1
action in eliciting pituitary ACTH secretion, administration of either IL-1a or IL-1b directly into the cerebroventricles (intracerebroventricular) of rats markedly elevates
plasma ACTH concentrations. Elevation in plasma ACTH
concentrations produced by intracerebroventricular IL-1
generally occurs at considerably lower (5- to 20-fold less)
doses than those required by intravenous IL-1 (399, 684,
695, 909). Similarly, IL-2, IL-6, TNF-a, and epidermal
growth factor (EGF) have been shown to elevate plasma
ACTH and/or corticosterone concentrations when administered via the intracerebroventricular route (see Table
5). Interleukin-1 infused directly into several brain sites,
including the PVN (40, 939), ME (525–527, 530, 939), and
hippocampus (477), also increases pituitary ACTH secretion.
The effectiveness of cytokines when administered directly into the brain, and in particular the fact that lower
doses are usually required to stimulate HPA axis secretory
activity than when administered peripherally, have been
interpreted as evidence that the activation of the HPA
axis by peripherally administered cytokines occurs via an
action within the CNS. This assumption based on such
dose-response studies alone is at least questionable, given
the differences in dilution of the cytokine when administered via these two routes. Indeed, at least in the cases
of IL-1b and TNF-a, activation of the HPA axis is associated with patterns of gene expression within the PVN
and/or pharmacological profiles that differ according to
whether the cytokine was administered peripherally or
centrally (457, 684, 688, 881). This indicates that the mechanisms by which these routes of cytokine administration
stimulate HPA axis secretory activity are distinct. However, a number of lines of evidence do suggest that the
stimulation of pituitary ACTH and adrenal GC secretion
by either centrally or peripherally administered cytokine
11-25-98 11:16:36
pra
APS-Phys Rev
Downloaded from on April 23, 2014
consistently observed sites of IL-1b expression after systemic LPS (124, 576 – 578, 971). These include the circumventricular organs [CVO; including OVLT, SFO, ME,
area postrema (AP), and pineal gland], meninges, and
choroid plexus. The cell types expressing IL-1b in these
regions include macrophages, microglia, and perivascular cells. Marked induction of IL-1b in microglia throughout the entire brain has also been observed, but this
tends to occur only at larger doses of LPS (ú2.5 mg/kg
vs. õ1 mg/kg for barrier related) (124, 902, 971). Furthermore, the induction of IL-1b expression in barrier-related regions seems to occur faster (by 1 – 2 h) than that
in brain parenchyma (peak Ç6 – 8 h). Immuno- and bioassays have also been used to document increases in
brain cytokine concentrations. Lipopolysaccharide administered intraperitoneally increases the immunoreactive concentration of IL-1b within the rat hypothalamus
(314, 340). The minimum doses of LPS required to measure significant increases in hypothalamic IL-1b have
been reported to be between 0.15 and 1 mg/kg (intraperitoneal), and elevations have been detected as early as 1
h after LPS, with a peak increase apparent at 4 – 10 h
(314, 340). Similarly bioactive IL-1 has been detected in
the brains of rats (655) and mice (264) after LPS treatment, and again the doses used were large and the time
required before a significant increase was measurable
was long (5 – 6 h).
The topographical, temporal, and cellular induction
of TNF-a mRNA by LPS treatment in mice appears to be
similar, but not identical, to that described for IL-1b (102).
At early time points (1.5 h), hybridization signal is most
prevalent over perivascular and neuronal elements in the
CVO (OVLT, AP, and ME) and in the meninges. Increased
hybridization signal within the brain parenchyma is not
obvious 6 h after LPS, with marked induction of TNF-a
mRNA in the hypothalamus and NTS being apparent only
at 9–18 h after LPS (102). Increased immunoreactive TNFa concentrations in rat CSF have been detected as early
as 0.5 h after a huge dose of LPS (30 mg/kg iv) (483), and
significant increases in the concentrations of bioactive
TNF-a and IL-6 in push-pull perfusates from the anterior
hypothalamus have also been demonstrated within 1–3 h
of LPS (20–50 mg/kg ip) injection in rats or guinea pigs
(365, 418, 419, 705). Similar to IL-1 and TNF-a, IL-6 mRNA
is induced in CVO (particularly the SFO, OVLT, ME, and
AP) and the choroid plexus 3–6 h after LPS (intraperitoneal) (900).
The effect of discrete localized inflammation within
the periphery on cytokine expression in the brain has
been less well studied. We have found no increase in
either IL-1b or TNF-a mRNA using in situ hybridization
or IL-1b, IL-6, or TNF-a mRNA by competitive RT-PCR at
5–8 h after intramuscular turpentine (881). Furthermore,
we have been unable to detect any increase in brain homogenate content of either IL-6 or TNF-a during turpen-
23
24
ANDREW V. TURNBULL AND CATHERINE L. RIVIER
/ 9j0c$$oc11
P13-8
to IL-1b, but not IL-6. However, this contrasts with the
findings of PVN lesioning studies that indicate an obligatory
role of the PVN in the elaboration of the plasma ACTH
response to a single injection of IL-6 (434). The relationship
between changes in mRNA levels of neuropeptides and
cIEG in the PVN and hypothalamic release of peptides and
subsequent pituitary ACTH secretion thus remains unclear.
Whereas ACTH secretion is observed within 5–10 min of
peripheral administration of IL-1, the earliest time point
reported for induction of c-fos mRNA in the PVN after
peripheral IL-1b has been 30 min (187), and the earliest
time reported for CRF mRNA is 1 h (98). Furthermore, the
minimum dose of IL-1b required to induce Fos expression
in CRF-containing neurons in the PVN is an order of magnitude greater than the minimum dose required to elicit
ACTH secretion (237). Finally, marked increases in plasma
ACTH concentrations (from õ20 pg/ml basal to ú700 pg/
ml) can be elicited by doses of peripheral IL-1b that do
not produce measurable changes in either CRF or AVP
mRNA in the pPVN (457). This disparity in minimum doses
could be due to differences in the sensitivity of the detection of elevations in plasma ACTH versus the detection of
induction of Fos or CRF mRNA. Alternatively, it may indicate that ACTH secretion can occur at doses of IL-1b that
do not elicit changes in gene expression in the PVN. Indeed,
the transcriptional/translational changes that have been observed within the pPVN may well occur as a consequence
of, rather than as a cause of, increased secretory activity
of PVN neurons. Consequently, the absence of detectable
changes in cIEG or neuropeptide mRNA expression in the
pPVN does not necessarily indicate a lack of PVN contribution to observed increases in HPA axis secretory activity.
Strong evidence that IL-1 stimulates the secretory activity of the HPA axis primarily by an action on the CNS
comes from studies that have demonstrated that IL-1 rapidly stimulates the secretion of CRF from the ME into
hypophysial portal blood vessels. Corticotropin-releasing
factor is depleted from the ME of colchicine-treated rats
within 1 h of intraperitoneal IL-1b (55), and CRF concentrations in portal blood are elevated within 30 min of
intravenous IL-1b (730). In the perfusates from push-pull
cannulas placed within the ME, CRF concentrations are
elevated within 5 min of either intracerebroventricular or
intra-PVN IL-1b (40) and precede the rise in plasma ACTH
after intravenous IL-1b (936, 940). Similarly, intravenous
TNF-a produces an immediate rise in CRF secretion (938).
Histological examination of hypophysiotropic nerve terminals suggests that AVP may (958) or may not (54) be
cosecreted with CRF in response to peripheral IL-1,
whereas electrophysiological data indicate that increased
activity in the PVN is selective for neurons containing
CRF only (728). However, in one study, the mean portal
blood concentrations of AVP were almost twofold elevated by 30 min after intravenous IL-1b, although this
increase did not achieve statistical significance (730). Fur-
11-25-98 11:16:36
pra
APS-Phys Rev
Downloaded from on April 23, 2014
is due to an action at or above the level of the hypothalamus. Indeed, surgical lesioning studies indicate the importance of an intact hypothalamus (619) to the elaboration
of a plasma ACTH responses to IL-1b in the rat. Electrolytic obliteration of the rat PVN also markedly reduces
the rise in plasma ACTH concentrations produced by a
number of cytokines, with inhibition being complete in
the cases of intracerebroventricular IL-1b (681) or intravenous IL-6 (434), Ç70% after intravenous TNF-a (434), and
Ç50% when IL-1b is injected intravenously (434).
Studies assessing the expression of cIEG and neuropeptide mRNA within the rat pPVN after administration
of IL-1b have also suggested a CNS site of action of IL-1
administered via either peripheral or central routes. Interleukin-1b administered intravenously (237, 588, 914) or
intraperitoneally (98, 128, 187, 831) induces c-fos mRNA
or Fos protein expression in the rat pPVN. The Fos signal
in the pPVN colocalizes with CRF immunoreactivity or
mRNA after either peripheral route of IL-1b treatment
(128, 237, 914), indicating cellular activation of CRF-containing neurons. Peripheral administration of large doses
of IL-1b also increases CRF mRNA in the PVN (98, 237,
327). Similarly, increases in c-fos mRNA or Fos protein
are elicited in the pPVN in response to intracerebroventricular IL-1b (153, 187, 588, 682, 684) and are accompanied by increased expression of not only CRF (457, 682)
but also AVP mRNA in the pPVN (457).
In addition to IL-1b, IL-1a also stimulates Fos expression in the pPVN after its intracerebroventricular administration (153, 381). Interleukin-1a administered intraperitoneally has been reported not to influence the expression
of pPVN CRF mRNA (327). However, the fact that the
doses of IL-1a used in this latter study were not sufficient
to produce significant increases in plasma ACTH concentrations casts doubt on the significance of these negative
findings (327). In contrast to the impact of a single injection of IL-1b on pPVN cIEG expression, a single injection
of IL-6 intravenously (899), intracerebroventricularly
(899), or intraperitoneally (128) has no impact on PVN
cIEG expression. It should be noted, however, that the
half-life of IL-6 (like other cytokines) in circulation after
intravenous injection is very short. Indeed, Castell et al.
(146) described a biphasic disappearance of human IL-6
from rat plasma, with an initial half-life of just 3 min.
However, during inflammation, plasma IL-6 concentrations are elevated for hours or days. Therefore, the findings that constant infusion of IL-6 (589) does induce Fos
expression in the pPVN (in a cyclooxygenase-dependent
fashion) are clearly of relevance and indicate that circulating IL-6 can influence pPVN activity.
The different effects of a single injection of IL-1b or
IL-6 on pPVN neuronal activity, as assessed by Fos and
CRF mRNA expression, would seem to indicate that activation of the pPVN may be a factor responsible for elevated
HPA axis secretory activity produced by acute exposure
Volume 79
January 1999
TABLE
Cytokine
IL-1a
IL-1b
IL-2
IL-6
IL-8
TNF-a
IFN-a
EGF
Activin
25
REGULATION OF HPA AXIS BY CYTOKINES
7. Cytokines increase CRF secretion from the hypothalamus in vitro
Preparation
Static explant
Superperfused hypothalami
Static explant
Superperfused hypothalami
Dispersed cell cultures
Static explant
Hypothalamic slices
Superperfused hypothalami
Static explant
Static explant
Static explant
Static explant
Hypothalamic slices
Static explant
Dispersed cell cultures
Minimum Doses
of Cytokine
Incubation Time
or Time to Effect
Reference No.
0.1–10 pM
100 pM
0.001–280 pM
0.6–100 pM
1 pM–10 nM
0.01–0.1 pM
20–30 min
6 min
20–45 min
5–10 min
4–20 h
30 min
20–40 min
5 min
20–40 min
30 min
20–40 min
20 min
20 min
20 min
24 h
56, 491, 854, 873
600
59, 121, 172, 315, 491, 501, 583, 653, 813, 854, 873, 999
130, 131, 133–135, 599, 600, 724
116, 346
392–394
659, 660
131
491, 500, 501, 545, 583, 813, 854
491, 854
58, 813
289
660
492
650
0.1–800 pM
30 pM
100 pM–6 nM
50 nM
1 pM
1 nM
Definitions are as in Table 3. The majority of studies have quoted cytokine concentrations in terms of molarity. Only a few studies quote
their concentrations in the more appropriate biological or international standard units, but these have not been included here, because without
their widespread usage, they do not afford easy comparison between different studies.
/ 9j0c$$oc11
P13-8
of their individual effects (131). Such synergistic effects
mirror the cytokine interactions that take place within the
immune system (see sect. IC) and underscore the importance of applying to experimental paradigms the in vivo
reality of multiple cytokine production.
Although the above discussion clearly indicates that
a number of cytokines influence hypothalamic neurosecretory activity, probably the best evidence that the CNS
is the primary target of cytokine action on the HPA axis
in vivo is derived from experiments where CRF (or AVP)
has been immunoneutralized. Immunoneutralization of
CRF in the rat inhibits the rise in plasma ACTH or corticosterone concentrations produced by intravenous IL-1a or
-1b, IL-6, and TNF-a (55, 58, 457, 575, 730, 894, 909). The
reduction in intravenous IL-1b-induced ACTH secretion
produced by anti-CRF antisera/antibodies has been found
to range from an 84% reduction to a complete blockade
(55, 457, 730, 894). Interleukin-1b administered either intraperitoneally or intracerebroventricularly elicits ACTH
secretion that can be inhibited by 85 and 73%, respectively
(457). When a complete blockade of intravenous IL-1binduced ACTH secretion has been achieved, the rise in
plasma corticosterone concentrations has also been totally abolished (730). However, some authors have reported only marginal effects of CRF antibodies on the
plasma corticosterone response to IL-1b (intravenous)
(909), but the absence of measurement of plasma ACTH
levels in this study precludes determination of whether
these findings can be interpreted as indicating direct actions of IL-1 on the pituitary and/or adrenal glands.
Corticotropin-releasing factor antisera/antibodies/receptor antagonists also inhibit the rise in plasma ACTH
concentrations produced by a number of other cytokines.
Consistent with the effects of PVN lesioning, anti-CRF
pretreatment totally abolishes elevations in both plasma
11-25-98 11:16:36
pra
APS-Phys Rev
Downloaded from on April 23, 2014
thermore, either intravenous, intra-PVN, or intra-ME IL1b causes prompt increases in both CRF and AVP concentrations in ME push-pull perfusates (939). In contrast, the
intravenous injection of IL-1b does not influence hypophysial portal blood oxytocin concentrations (730).
Despite the fact that no IL-1 receptors have been
demonstrated in the PVN, a number of studies demonstrate that IL-1 stimulates the secretion of CRF from the
hypothalamus in vitro (see Table 7). Incubation with subnanomolar doses of IL-1a or IL-1b has consistently been
reported to rapidly (within minutes) increase the release
of CRF from rat hypothalamic explants, superfused hypothalamic tissue, as well as dispersed hypothalamic cell
cultures (see Table 7). In addition, IL-1b increases the
CRF content of hypothalamic explants, indicating an increase in CRF peptide production (315). Most (121, 491,
545, 991, 992, 999), although not all, investigators (813)
have noted concomitant increases in AVP secretion. However, it should be noted that AVP release from the hypothalamus may be destined, in vivo, for secretion from
the posterior pituitary, rather than direct interaction with
corticotropes. Similarly, IL-2, IL-6, IL-8, TNF-a, IFN-g, and
EGF potently induce rapid increases in CRF (see Table
7) and/or AVP secretion (339, 658, 741) from rat hypothalami in vitro. A few studies have also demonstrated synergistic effects of various cytokine combinations on hypothalamic neuropeptide secretion. Buckingham et al. (121)
demonstrated that the release of CRF and AVP by conditioned media from LPS-stimulated rat peritoneal macrophages (which contains multiple cytokines) is far greater
than that observed with either IL-1a, IL-1b, IL-6, IL-8, or
TNF-a alone. Indeed, a subthreshold dose of TNF-a markedly potentiates the IL-1b-induced release of AVP from
hypothalamic explants (121), while the increase in CRF
secretion due to IL-1b plus IL-2 is greater than the addition
26
ANDREW V. TURNBULL AND CATHERINE L. RIVIER
B. Evidence for Direct Effects of Cytokines
on Pituitary Adrenocorticotropic
Hormone Secretion
1. Cytokine receptors within the pituitary
A number of cytokine receptors have been localized
in the pituitary (see Table 8). For example, 125I-labeled
human IL-1a (29, 30, 177, 843, 846, 847), IL-1b (30) or ILra (841), or rat IL-1b (516) bind specifically to the anterior
lobe of the mouse pituitary gland, with little or no IL-1
binding apparent in the mouse posterior pituitary (29, 30,
516). Pituitary IL-1 binding in the mouse is decreased by
systemic treatment with LPS (840, 843) and increased by
immobilization stress (29), ether-laparotomy stress (846,
847), and long-term treatment (7 day) with GC (29). Inter-
/ 9j0c$$oc11
P13-8
estingly, the increase in IL-1 binding observed in the pituitary after ether-laporotomy stress can be prevented by
treating mice with a CRF receptor antagonist (847), suggesting that activation of the HPA axis secretion per se
may upregulate pituitary IL-1 receptors. Although there
have been many studies investigating IL-1 binding in
mouse pituitary, there is only one report demonstrating
IL-1 binding in the rat (516). In contrast to the mouse,
both anterior and posterior lobes of the rat pituitary bind
125
I-labeled rat IL-1b (516).
In addition to studies demonstrating IL-1 binding
by the anterior pituitary, RT-PCR experiments have
demonstrated the presence of both IL-1R1 and IL-1R2
mRNA in the whole mouse pituitary (626). In situ hybridization histochemistry experiments agree with IL-1
binding studies and show that IL-1R1 mRNA is present
in the mouse anterior, but not posterior, pituitary (178).
Interleukin-1 binding and IL-1R1 and IL-1R2 mRNA have
also been demonstrated in the mouse corticotropic tumor cell line AtT20 (110, 111, 423, 872, 947). Interestingly, and in accordance with the stress-induced increases in IL-1 binding in the anterior pituitary of the
mouse (847), CRF increases IL-1a binding in AtT20 cells
(947), while IL-1b and TNF-a increases the expression
of both IL-1R1 and IL-1R2 mRNA (110). However, the
expression of IL-1R1 and IL-1R2 by AtT20 cells does not
necessarily indicate that normal corticotropes contain
cell-surface IL-1 receptors. Indeed, when IL-1R1 and
IL-1R2 immunoreactivities were localized to particular
endocrine cell types in the normal mouse anterior pituitary, no evidence of colocalization of IL-1 receptor with
ACTH was apparent (267). Immunolabeling using a
panel of IL-1 receptor antibodies demonstrated that
IL-1R1 and IL-1R2 were abundantly expressed in the
mouse anterior pituitary, were always coexpressed, and
were predominantly localized to a single cell type, the
somatotrope (growth hormone-producing cells) (267).
However, the possibility that IL-1 receptor expression
by corticotropes is below the limits of the immunohistochemical techniques employed, or that the level of
IL-1 receptor expression may be induced (as demonstrated in AtT20 cells), cannot be excluded.
Much less work has focused on the presence of
IL-6 and TNF-a receptors within the pituitary. Rodent
anterior pituitaries exhibit binding of 125I-labeled IL-6
(601), and IL-6Ra mRNA is expressed in normal rat (601,
915) and fetal and adult human pituitaries (774, 915).
Interleukin-6Ra is also expressed in human ACTH and
growth-hormone secreting tumors (675, 915). In addition, the IL-6R signaling subunit gp130 is present in human fetal pituitary cells (774). High concentrations of
binding sites for rmTNF-a have also been demonstrated
in the mouse and rat anterior pituitaries (967), AtT20
cells (422), and the folliculostelate cell line TtT/GF
(422).
11-25-98 11:16:36
pra
APS-Phys Rev
Downloaded from on April 23, 2014
ACTH and corticosterone concentrations produced by IL6 (575, 909), whereas inhibition of intravenous TNF-ainduced ACTH secretion by anti-CRF is nearly complete
(58), with only a small corticosterone response remaining
(58, 909). Administration of the CRF receptor antagonist
a-helical CRF-(9{41) inhibits the rise in plasma ACTH
due to EGF (551), but not NGF (741). However, the low
potency of this receptor antagonist at the pituitary CRF
receptor (888, 891) makes interpretation of incomplete
inhibition of ACTH secretion difficult.
The effect of immunoneutralization of AVP on cytokine-induced ACTH secretion has been less well studied.
We have consistently observed a 15–20% decrease in the
plasma ACTH response to peripherally administered IL1b when rats are pretreated with anti-AVP, although these
effects are not always statistically significant (457, 687;
unpublished observations). However, we also find that
immunoneutralization of AVP inhibits the ACTH response
to CRF itself by Ç20–30% (892). Thus the role of AVP in
peripheral IL-1b-induced ACTH secretion is probably only
permissive and is required only to permit the full expression of the ACTH secretagogue capacity of CRF. However,
when IL-1b is administered intracerebroventricularly, the
effects of AVP neutralization are more pronounced (40%
inhibition), a finding consistent with the stimulatory effects of intracerebroventricular IL-1b on pPVN AVP
mRNA and which suggests an activational role of AVP in
the ACTH response to intracerebroventricular IL-1b (457).
Collectively, the rapid effects of IL-1 and other cytokines on hypothalamic CRF secretion in vivo and in vitro,
together with the reduction of plasma ACTH responses
to cytokines produced by inhibiting the actions of CRF,
provide an extremely strong case for the CNS as a primary
site of cytokine action in the stimulation of HPA axis
secretory activity. Nevertheless, a large number of in vitro
studies have also indicated the possibility of direct effects
of cytokines on pituitary ACTH secretion and adrenal GC
secretion.
Volume 79
January 1999
TABLE
27
REGULATION OF HPA AXIS BY CYTOKINES
8. Cytokine receptors in pituitary
Cytokine Receptor
IL-1
IL-2
IL-6
TNF-a
LIF
OM
Activin
EGF
FGF
PDGF (a and b)
Neurotrophin receptors
Reference No.
AtT20 cells
Normal mouse anterior pituitary
Normal rat anterior and posterior pituitary
AtT20 cells
Human corticotropic adenoma
Rat anterior lobe, ACTH-containing cells
Normal rat anterior rat pituitary
Human fetal pituitary (IL-6Ra and gp130)
Normal human pituitary
Human pituitary tumors
Normal rat and mouse anterior lobe
AtT20 cells
Folliculostellate cells
Human pituitary fetal cells, ACTH cells
AtT20 cells
Human pituitary fetal cells
AtT20 cells
Normal rat pituitary
GH3 rat pituitary tumor cells
Rat anterior lobe
Sheep anterior lobe
Normal anterior and posterior lobes
Human pituitary adenomas
Normal human anterior lobe
Rat anterior lobe
Rat anterior pituitary, lactotropes
GH3 rat pituitary tumor cells
Rat anterior pituitary
Rat anterior and posterior
Rat anterior and intermediate
29, 30, 110, 111, 178, 267, 423, 516, 517, 626, 840, 843, 846, 847, 872, 947
22, 796
601, 675, 774, 775, 915
422, 967
7, 774
774
136, 155, 521
151, 244, 562
298
464
244
155, 188
431, 631
Definitions are as in Table 3.
2. Cytokine expression in the pituitary
The pituitary has been shown to produce a diverse
range of cytokines (see Table 9 and an excellent review
by Ray and Melmed, Ref. 666). Some of these cytokines
(e.g., IL-2, IL-10, LIF, and MIF) have been localized to
corticotropes or demonstrated in the corticotrope cell
line AtT20 (7, 22, 61, 119, 350). In vivo mRNA encoding
the cytokines IL-1a, IL-1b, IL-6, LIF, IFN-g, and TNF-a
mRNA in the pituitary are all elevated by 45 min to 6 h
after treatment with LPS (277, 424, 453, 570, 644, 748,
865, 926). Anterior pituitary IL-6 mRNA is also increased
by chronic, local inflammation (732, 818). The constitutive expression of IL-1ra in the pituitary (269, 277, 471,
971) is of particular interest given its ability to antagonize
the effects of IL-1 agonists, suggesting that the responsiveness of the pituitary to IL-1 may be modulated at a
local level. Interleukin-1ra mRNA has been reported to
be either unaffected (277) or induced (269, 471, 971) by
systemic treatment with LPS.
One cytokine in particular, MIF, has been proposed
to serve as a pituitary hormone (60, 118–120, 126). Bucala
and co-workers (60) isolated a 12.5-kDa protein from the
conditioned media of LPS-stimulated mouse pituitary
cells, which was subsequently sequenced, its cDNA
cloned, and identified as the mouse homolog of human
/ 9j0c$$oc11
P13-8
MIF (60, 61, 118, 119). The mouse pituitary contains large
amounts of preformed, intracellular pools of MIF (possibly located within corticotropes) which are released into
the systemic circulation after treatment with LPS. Macrophage migrating inhibitory factor appears in the blood of
normal mice within 2 h of LPS treatment, and its serum
concentration continues to rise for Ç20 h. No MIF is detectable in the serum of hypophysectomized mice 20 h
after LPS, suggesting a pituitary origin of MIF found in
serum. Macrophage migrating inhibitory factor appears to
play a critical role in host defense to endotoxemia, since
LPS-induced lethality in mice is potentiated by coadministration of MIF, whereas an anti-MIF antibody confers protection from lethal doses of LPS (60). Corticotropin-releasing factor increases the secretion of MIF from AtT20
cells, at doses lower than those required to stimulate
ACTH secretion, suggesting that MIF secretion may increase in parallel with activation of the HPA axis (591).
Although itself proposed as a pituitary hormone, the effects of MIF on the secretion of other pituitary hormones
(e.g., ACTH) have not been characterized.
The regulation of IL-6 secretion from anterior pituitary cell cultures has been investigated extensively. Anterior pituitary cells constitutively produce IL-6 (807), and
the secretion of this cytokine can be induced within 6 h
of treatment with LPS (801, 806, 886), IL-1a, IL-1b (801,
11-25-98 11:16:36
pra
APS-Phys Rev
Downloaded from on April 23, 2014
TGF-a
TGF-b
Localization
28
ANDREW V. TURNBULL AND CATHERINE L. RIVIER
TABLE
9. Cytokines in the pituitary
Cytokine
IL-1a
IL-1b
IL-1ra
IL-2
IL-6
IL-8/cinc/gro
IL-10
TNF-a
LIF
IFN-g
Activin
BDNF
EGF
bFGF
NGF
PDGF (A
and B
subunits)
TGF-a
TGF-b
Whole rat pituitary
Rat anterior pituitary
Whole rat or mouse pituitary
Human pituitary adenomas
Whole rat pituitary
AtT-20
Human corticotropic adenoma
Rat, mouse, pig, or sheep anterior
or whole pituitaries
Normal rat anterior pituitary cell
cultures
Human pituitary tumors
Folliculostellate cells
Rat neurointermediate lobe
Rat anterior pituitary
AtT-20
Mouse pituitary
Human pituitary
Whole mouse or rat pituitary
Human fetal pituitaries
Normal mouse pituitary
Bovine pituitary folliculostellate
cells
Normal sheep pituitary
Mouse anterior pituitary
Corticotrophs and thyrotrophs
Rat whole pituitary
Gonadotropes
Rat anterior and intermediate
lobes
Rat anterior pituitary
Rat folliculostellate cells
Rat anterior pituitary
Whole human pituitary
Rat anterior pituitary
Human pituitary adenomas
Normal human anterior pituitary
Rat anterior pituitary
Bovine anterior pituitary
Rat anterior pituitary
Reference No.
277
277, 424, 453, 644
471, 734, 971
22
1, 11, 277, 453, 570,
644, 818
145, 508, 579, 802–
806, 853, 986
375–378, 675, 874,
895
801, 912, 913
426
350, 662
277, 453, 644, 756
7, 258, 756, 774,
926
60, 118, 119
644
75
431, 797
244, 245, 562
13
298
518
164, 549, 631, 632
464
244
564
731
Definitions are as in Table 3.
805, 806), TNF-a (579), the IFN family (986), phorbol myrisate, and agents that elevate cAMP (803, 807). Accordingly, dibutyryl cAMP, prostaglandin E2, forskolin, and
cholera toxin all increase IL-6 secretion from rat anterior
pituitary cell cultures (145, 802, 805, 806). Some [vasoactive intestinal peptide (VIP), pituitary adenylate cyclase
activating peptide (PACAP), and calcitonin gene-related
peptide] but not all (CRF, growth hormone releasing factor) neuropeptides that utilize cAMP-coupled receptors
for signaling, therefore, also stimulate IL-6 secretion in
anterior pituitary cells (803, 853, 886). However, the induction of IL-6 secretion from anterior pituitary cells by IL1b does not appear to be mediated by cAMP-dependent
pathways, since in these cells, no increase in intracellular
cAMP is apparent after treatment with IL-1b (802, 804–
806, 808). Glucocorticoids inhibit basal (145) and IL-1bstimulated (806) IL-6 release from rat anterior pituitaries.
/ 9j0c$$oc11
P13-8
In addition to secretion from anterior pituitary cells, IL-6
is released by neurointermediate lobe cells in culture, with
IL-1b and LPS again being potent secretagogues (801).
The cell source within the normal anterior pituitary
does not appear to be a classical endocrine cell type.
Rather, folliculostellate (FS) cells are the major sources of
IL-6 (11, 912, 913). Folliculostellate cells are of monocytic
lineage and are thought to be involved in paracrine regulation of hormone secretion from the pituitary (26). Recently, an FS-like cell line has been isolated (TtT/GF), and
in accordance with previous studies on whole anterior
pituitaries, TtT/GF cells constitutively secrete IL-6, and
IL-6 secretion is enhanced by TNF-a, VIP, PACAP (422,
522) or IL-1b (L. Bilezikjian and A. V. Turnbull, unpublished observations).
3. Direct effects of cytokines on pituitary ACTH
secretion in vitro
With the exception of activin, which inhibits POMC
mRNA expression and ACTH secretion in the corticotropic tumor cell line AtT20 (74), and reduces ACTH secretion from primary cultures of rat anterior pituitary cells
(75), all other cytokines studied have been reported to
either enhance or have no effect on either ACTH secretion
or POMC mRNA expression in otherwise untreated pituitary cells (see Table 10). The effects of cytokines on
the mouse anterior pituitary tumor cell line AtT20 are
reasonably undisputed. Similarly, IL-1b, IFN-g, and GMCSF stimulate ACTH secretion from cultured human pituitary adenoma cells from patients with Cushing’s disease
(512). The duration of exposure of AtT20 cells and human
pituitary adenoma cells to cytokines required to elicit statistically significant increases in ACTH secretion has, in
general, been long. Although one report indicates increases in ACTH produced by either IL-1 or IL-6 within 2
h (966), the majority have used incubation times ranging
from 6 to 72 h (7, 115, 243, 268, 667, 816). The fact that
many cytokines have a stimulatory effect on pituitary tumor cells indicates the ability of signal transduction pathways activated by cytokine-cytokine receptor interaction
to influence POMC expression and/or ACTH secretion.
However, it is unclear to what extent corticotrope tumors
accurately represent normal anterior pituitary corticotropes. For example, although AtT20 cells possess both
IL-1R1 and IL-1R2 mRNA (110, 198, 872, 947), immunostaining of normal mouse pituitary does not show significant IL-1 receptor expression by corticotropes (267). Most
important to the present discussion, however, is that although most cytokines tested increase ACTH secretion
from AtT20 cells, their effects on normal rat anterior pituitaries have been extensively debated (see Table 10).
Although the effects of a number of cytokines on in
vitro anterior pituitary preparations have been investigated (see Table 10), by far the best studied has been the
11-25-98 11:16:36
pra
APS-Phys Rev
Downloaded from on April 23, 2014
MIF
Localization
Volume 79
January 1999
REGULATION OF HPA AXIS BY CYTOKINES
10. Direct effects of cytokines on ACTH secretion
from the pituitary
TABLE
AtT-20
Cytokine
Effect
IL-1a
115, 268
IL-1b
115, 243, 268,
312, 717
IL-2
IL-6
115, 796
268, 966
IL-8d
IL-10
LIF
OM
TNF-a
No effect
268
350
7, 95, 667,
816
7, 667, 816
422
Effect
No effect
402
825a
48, 62, 115,
116, 132,
329, 402,
893
825a
893b
115, 395
499, 774
527c
737c
491, 629, 730,
873, 893
25, 55, 96, 366,
491, 583,
628, 629,
873
583
491, 498, 583
491
774
774
540
58b
74
897
156
NGF
402, 583
275e
491e
492, 551
289, 583
583
913g
741
Reference numbers for corresponding studies are given. Unless
otherwise stated, effect of cytokines is to increase either ACTH secretion
a
or proopiomelanocortin (POMC) mRNA expression.
Neither IL-1a
nor IL-1b affected ACTH secretion or POMC mRNA expression after 3
h incubation, but either IL-1a or IL-1b increased ACTH secretion after
3 h, with only IL-1b producing a statistically significant increase in POMC
b
mRNA.
Significant increases observed only at doses of 100 nM and
c
d
greater.
Stimulation described as ‘‘weak’’ or ‘‘slight.’’
References with regard to IL-8 include the homologous cytokines cinc/gro/
e
NAP-1.
TNF-a had no effect on basal ACTH secretion but inhibited
f
CRF- or hypothalamic extract-induced ACTH secretion.
Unlike all
other cytokines listed, activin decreases POMC mRNA expression and
g
IFN-g
ACTH secretion in AtT20 cells and rat anterior pituitary cells.
did not affect basal but inhibited CRF-stimulated ACTH secretion. Definitions are as in Table 3.
influence of IL-1. However, this literature is confusing.
Indeed, of the three original articles demonstrating the
potent effects of IL-1 on ACTH secretion in the rat, one
described a direct effect on pituitary cells in culture (62),
whereas the other two found no effect (55, 730). Subsequent studies have not clarified this apparent discrepancy
(see Table 10). Although some have shown stimulatory
effects of IL-1b but not IL-1a (893), others have reported
that either will enhance ACTH secretion directly from the
anterior pituitary in vitro (402, 825). A greater proportion
of positive findings have been obtained when using pituitary tissue obtained from female rats (62, 402, 873, 893).
However, when directly compared, the male and female
pituitary responded to IL-1b in an identical manner (893),
and there have been reports of a stimulatory influence of
IL-1 in male pituitaries (116, 132, 329) and no effect in
females (55). The use of different anterior pituitary prepa-
/ 9j0c$$oc11
P13-8
rations does not seem a likely explanation for the contradictory data obtained, since either positive or negative
results have been obtained using either static dispersed
rat anterior pituitary cell cultures (55, 62, 402, 730), perifused dispersed rat anterior pituitary cells (132, 583, 873),
or perifused rat anterior pituitary segments (48, 116, 329,
628, 629). Furthermore, whether IL-1 potentiates the
ACTH response to CRF has also been disputed (55, 96,
132, 366, 629, 635, 873, 893). It should be noted that virtually all studies employed pituitary preparations that are
capable of enhanced ACTH secretion, as shown by a stimulatory effect of CRF.
Of particular interest to the issue of whether cytokines directly stimulate ACTH secretion in vitro is the
influence of time of incubation. The studies that have used
long incubation periods (7 h or more) have all reported a
significant effect of IL-1b on ACTH secretion. Indeed, IL1b has been found to enhance ACTH secretion at 8–24 h
but not at 4 h (402), at 15 h but not 3 h (825), and at 7–
30 h but not at 3 h (329). However, given the rapid effects
of IL-1 on ACTH secretion in vivo (within 5–10 min),
the majority of in vitro studies have focused on shorter
incubation times. Some studies have demonstrated increased ACTH secretion within minutes of IL-1b application to perifused anterior pituitary cells (48, 132), within
1 h of incubation in rat anterior pituitary cell cultures
(116), whereas others indicate effects on ACTH secretion
by rat anterior pituitary cell cultures at picomolar to nanomolar doses at 4 h (62), or only at high doses (100 nM)
at 2 h (893). The majority, however, report no effect of
IL-1 within 3 h of incubation of rat anterior pituitary preparations with either IL-1a or IL-1b (25, 55, 96, 329, 366, 402,
491, 583, 628, 629, 730, 825, 873). As with IL-1, studies
of the direct ACTH stimulating effects of IL-2, IL-6, IL-8,
TNF-a, and EGF in vitro have been contradictory (see
Table 10).
Although it may be contentious to equate the different in vitro preparations, it is interesting to note that
where hypothalamic explants and pituitary cell cultures
have been compared within the same study, hypothalamic
CRF and/or AVP release is induced more rapidly and at
much lower concentrations of IL-1b (873), IL-6 (583),
TNF-a (58), IFN-g (289), NGF (741), and EGF (492) than
those required to elicit pituitary ACTH secretion. However, it should be noted that, despite the well-known synergistic effects of cytokines within the immune system,
there is little known about possible synergistic effects of
cytokines on pituitary ACTH secretion in vitro. This may
be of particular relevance given that Buckingham et al.
(121) demonstrated that although neither IL-1a, IL-1b,
IL-6, IL-8, nor TNF-a alone affects ACTH secretion from
rat anterior pituitary fragments (491), conditioned media
from macrophages stimulated with LPS produce a
dose-dependent increase in ACTH secretion within just
30 min (121).
11-25-98 11:16:36
pra
APS-Phys Rev
Downloaded from on April 23, 2014
Activinf
EGF
IFN-a
IFN-g
Anterior Pituitary Cells
29
30
ANDREW V. TURNBULL AND CATHERINE L. RIVIER
C. Evidence for Direct Actions of Cytokines on
Adrenal Glucocorticoid Secretion
1. Cytokine receptors within the adrenal
To our knowledge, there are only very few published
studies of the presence of cytokine receptors within the
adrenal gland. The adrenal gland displays no positive in
situ hybridization signal for IL-1R1 mRNA (178), whereas
IL-6Ra mRNA has been detected mainly in the zona glomerulosa and fasciculata in human adrenals (630) and also
in the adrenal medulla (272).
2. Cytokine expression in the adrenal gland
/ 9j0c$$oc11
P13-8
produce IL-6, dexamethasone does not influence either
basal or IL-1b-stimulated IL-6 secretion in zona glomerulosa cells (383). Intriguingly, ACTH increases the release of IL-6 from zona glomerulosa cells but not zona
fasciculata/reticularis, despite similar ACTH-stimulated
cAMP levels in each cell type (383). Such ACTH-induced
IL-6 release from the adrenal suggests that activation
of the HPA axis per se may cause IL-6 secretion from
the adrenal (383). Like IL-6, TNF-a secretion is stimulated by protein kinase C activators and ionomycin
(384). However, in stark contrast to IL-6, basal and stimulated TNF-a release is dose dependently inhibited by
ACTH and dibutyryl cAMP (384). Similarly, differential
effects on IL-6 and TNF-a secretion from zona glomerulosa cells are observed with serotonin and adenosine
(676, 677).
3. Direct effects of cytokines on adrenal glucocorticoid
secretion
Despite a lack of evidence for the presence of IL-1,
IL-6, or TNF-a receptors within the adrenal gland, direct
actions of these cytokines on GC secretion have been
demonstrated. In vivo, peripheral administration of IL-1b
has been reported to either stimulate (14) or exert no
effect (310) on corticosterone secretion in hypophysectomized rats and to induce secretion of GC in anesthetized
rats with isolated adrenal glands (698). The doses required
to observe direct effects of IL-1b on adrenal corticosterone secretion in vivo have, however, been extremely high
(35 mg/rat, Ref. 698), which casts doubt on the physiological relevance of this effect.
In vitro studies have indicated that IL-1b does not
influence either basal or ACTH-stimulated GC production from human fetal adrenal tissue either in cell or
organ culture (328). However, either IL-1a or -1b increases GC secretion from rat quartered adrenals (311),
rat adrenal slices (14), and rat, bovine, and human dispersed adrenal cells (311, 528, 597, 867, 957, 964). Similarly, IL-2, IL-3, and IL-6 induce GC secretion from various adrenal cell preparations (180, 630, 867, 946), and
IL-6 potentiates corticosterone secretion induced by
low concentrations of ACTH (722). Tumor necrosis factor-a stimulates cortisol secretion from adult adrenocortical cells (180) but inhibits ACTH-induced cortisol
secretion from human fetal adrenal tissue (361) and
cultured cells (362). Interferon-g stimulates corticosterone secretion from primary dispersed rat adrenal cells
(289) and normal human adrenal slices (143), whereas
NGF does not influence either basal or ACTH-stimulated corticosterone production from dispersed rat adrenal cell cultures (741).
These in vitro studies provide strong evidence that
some cytokines may influence GC secretion directly.
However, it should be noted that of the studies cited,
11-25-98 11:16:36
pra
APS-Phys Rev
Downloaded from on April 23, 2014
In the adrenal gland, large constitutive pools of IL1a and IL-1b have been identified (42, 753, 754). Interleukin-1a, IL-1b, and IL-1ra immunoreactivties have
been demonstrated in adrenal chromaffin cells (42, 753,
754). In addition, IL-1a, IL-1b, ICE, and IL-1ra mRNA
are present in the adrenal cortex and are markedly elevated in both adrenal medulla and cortex 90 min after
intravenous or intraperitoneal LPS (594, 754, 866). Interestingly, the IL-1-related cytokine IL-18/IL-1g is also
synthesized within the rat adrenal cortex, mainly in the
zona reticularis and fasciculata, and is strongly induced
by acute cold stress (166). Interleukin-6 mRNA is also
present throughout the human adrenal cortex (299,
630), and rat adrenal gland extracts contain IL-6 mRNA
(272, 570, 748), which is markedly induced 2 h after
intraperitoneal LPS (570). Although TNF-a mRNA has
been described throughout the cortex of human adult
adrenals, particularly in steroid-producing cells (300),
TNF-a immunoreactive protein has been detected in
only 12 of 22 fetal, and in 0 of 7 adult, human adrenals
(361). A number of growth factors are also present in
the adrenal: basic fibroblast growth factor mRNA or
protein is present in whole human adrenals or in rat or
bovine cortex or medulla (44, 301, 306, 341, 537, 757,
955), TGF-b1 mRNA and protein in bovine adrenal cortex and mouse whole adrenal (255, 341, 410, 861, 961),
and NT-3 protein has been reported in whole rat adrenal (396).
Studies of primary cultures of dispersed adult rat
adrenal glands have indicated the likely cellular sources
and major secretagogues of the cytokines IL-6 and TNFa within the adrenal gland. Although the zona fasciculata/reticularis produce small amounts of TNF-a and IL6, the primary source of IL-6 in the rat adrenal is the
zona glomerulosa (382 – 385). The secretion of TNF-a
and IL-6 from adrenal glomerulosa cells is stimulated
in a dose-dependent manner by LPS, IL-1a, and IL-1b
(383 – 385). Furthermore, IL-6 secretion from these cells
is enhanced by protein kinase C activators, the calcium
ionophore ionomycin, prostaglandin E2 , forskolin, and
angiotensin II (382, 385). Unlike most cell types that
Volume 79
January 1999
REGULATION OF HPA AXIS BY CYTOKINES
the majority required incubations in excess of 12 h to
observe significant effects of cytokines on GC secretion.
Indeed, in a comprehensive series of studies by van der
Meer et al. (906), either IL-1a, IL-1b, IL-2, IL-6, or TNFa had no significant effect on GC secretion from isolated
rat adrenal cells inside 60 min, but a small stimulation
was apparent Ç4 h after completion of a 6-h incubation
with IL-1b. Thus, as with the effects of cytokines directly on the pituitary, it seems unlikely that a direct
action of a cytokine on the adrenal can account for
the rapid in vivo effects of administered cytokines on
plasma GC concentrations.
V. MECHANISMS OF HYPOTHALAMICPITUITARY-ADRENAL AXIS ACTIVATION
BY INTERLEUKIN-1
A. Direct Actions on Pituitary and Adrenal
The majority of in vivo data indicate that stimulation of pituitary ACTH and adrenal GC secretion in re-
/ 9j0c$$oc11
P13-8
sponse to the presence of IL-1 is produced by enhanced
secretion of hypothalamic ACTH secretagogues, in particular CRF. This is evidenced by the marked reductions
in plasma ACTH and corticosterone concentrations
when CRF is immunoneutralized. However, incomplete
inhibition of either ACTH or corticosterone secretion
after CRF immunoneutralization has been observed
(e.g., Ref. 909). Furthermore, marked elevations in
plasma corticosterone concentrations have been noted
even when ACTH secretion produced by IL-1 has been
markedly reduced (e.g., Ref. 390). However, decisive
evidence that CRF has been completely immunoneutralized or that the ACTH secretion remaining is unable
to produce marked elevations in plasma corticosterone
concentrations is often lacking. Although plasma ACTH
concentrations can be elevated to levels of 1,000 pg/ml
by intravenous IL-1b, much lower levels (100 – 200 pg/
ml) of ACTH are sufficient to stimulate corticosterone
secretion maximally. Indeed, the adrenals of both rats
and dogs have been shown to respond to very small
elevations (°10 pg/ml) in plasma ACTH with significant
elevations in plasma GC (388, 405, 975).
The data discussed in section IV indicate that a number of cytokines, and IL-1 in particular, may be capable
of having direct actions on the pituitary to enhance the
secretion of ACTH and on the adrenal cortex to increase
GC secretion. Receptors for IL-1 are clearly present in the
anterior pituitary, although it is unlikely that these are
expressed on normal corticotropes. The pituitary and adrenals clearly would be exposed to IL-1 if its concentrations in blood were elevated (e.g., severe endotoxemia).
In addition, IL-1 can be synthesized locally within these
tissues. However, as discussed above, it seems clear that
prolonged exposure of the pituitary or adrenals to IL-1 is
necessary to elicit the release of ACTH or GC, respectively. Therefore, direct actions of IL-1 on either the pituitary or adrenal do not appear to account for a significant
component of the hormone secretion observed in response to acute exposure to IL-1 in vivo. In contrast, circumstances involving prolonged increases in cytokines,
for example, during chronic inflammation (732, 818), may
well involve direct actions of IL-1 or other cytokines on
the pituitary ACTH and/or adrenal GC secretion. Furthermore, enhanced pituitary or adrenal synthesis of IL-1 may
regulate these glands’ growth and development (21, 669,
819, 1004). Similarly, a number of other interleukins (e.g.,
IL-2, IL-6) and growth factors (e.g., EGF) have been shown
to influence the growth of the pituitary or adrenal glands
(20, 21, 157, 637).
A large body of evidence indicates that the level of
the HPA axis primarily affected by IL-1 is the hypothalamus. This is true whether IL-1 has been administered
directly into the brain or into the periphery. This raises
the question of how a blood-borne, large, hydrophilic
peptide such as IL-1 accesses the CNS to influence hypo-
11-25-98 11:16:36
pra
APS-Phys Rev
Downloaded from on April 23, 2014
The above discussion details the ability of a number
of cytokines from various families to significantly influence the activity of the HPA axis. Furthermore, the distributions of various cytokines and their receptors
throughout the brain, pituitary, and to a lesser extent
the adrenal provide an anatomic basis for the hypothesis that cytokines influence the function of these organs.
However, in general, the evidence that cytokine receptors, and IL-1 receptors in particular, are expressed by
secretory cells (hypophysiotropic CRF neurons, normal
corticotropes, GC-producing adrenocortical cells) is
not strong. This has suggested the involvement of structural and/or pharmacological intermediates in activation of the HPA axis by cytokines. Studies aimed at
elucidating the mechanisms by which particular cytokines may activate the HPA axis during a particular
threat to homeostasis have focused on the cytokine IL1 and to a much lesser extent IL-6 and TNF-a. The following sections discuss the proposed anatomic and
pharmacological pathways by which IL-1 generated in
response to a particular threat to homeostasis may influence the HPA axis and are summarized in Figure 4.
The studies described in section IV indicate that the CNS
is probably the primary site of IL-1 action in eliciting
increased HPA axis secretory activity. Because the
question of how IL-1 signals the brain is relevant to all
CNS-mediated acute phase responses, such as fever,
hypermetabolism, cachexia, suppression of reproduction, and sickness behavior, knowledge derived from
studies of such responses is mentioned wherever pertinent. Furthermore, what information is available for
other cytokines, in particular IL-6 and TNF-a, is also
discussed.
31
32
ANDREW V. TURNBULL AND CATHERINE L. RIVIER
Volume 79
FIG. 4 Proposed models by which interleukin-1 influences secretory activity of
hypothalamic-pituitary-adrenal axis. AP,
area postrema; BBB, blood-brain barrier;
CVO, circumventricular organs; ME, median eminence; OVLT, organum vascularis
of lateral terminalis; PVN, paraventricular
nucleus; CNS, central nervous system;
NTS, nucleus tractus solitarius.
Downloaded from on April 23, 2014
thalamic secretions, and the models which have been
proposed to address this question are discussed below.
These include the possibilities that IL-1 penetrates the
BBB to enter brain parenchyma (see sect. VB) or that
IL-1 stimulates the production of intermediary signals
(see sect. VC), by actions at the BBB interface (see sect.
VD), at regions of the brain relatively devoid of a BBB
(see sect. VE), at brain stem medullary cell groups (see
sect. VF), and/or peripheral afferent nerves (see sect.
VG). The hypothesis that IL-1 generated within the brain
itself produces activation of the HPA axis is also considered (see sect. VH). Finally, the possibility that increased secretion of IL-1 locally in damaged or diseased
tissue can activate the HPA axis indirectly by inducing
the synthesis and secretion of a circulating factor is
also discussed (see sect. VI).
/ 9j0c$$oc11
P13-8
B. Penetration of Cytokines Into Brain
The transport of solutes out of vascular compartments and into perivascular tissue (or vice versa) occurs
via either paracellular or transcellular mechanisms.
Within the cerebrovaculature, the paracellular route is
particularly impermeable due to the presence of the BBB.
The BBB consists primarily of nonfenestrated endothelial
cells that are connected by tight junctions and thus form
a continuous cell layer that has the permeability properties of a continuous plasma membrane (664). The paracellular ultrafiltration of solutes into and out of tissues that
occurs in peripheral vascular beds does therefore not occur in most cerebrovascular beds, at least while the BBB
remains intact. Furthermore, the large molecular size (8–
65 kDa) and hydrophilic nature of cytokines preclude
11-25-98 11:16:36
pra
APS-Phys Rev
January 1999
REGULATION OF HPA AXIS BY CYTOKINES
their movement transcellularly by simple diffusion to any
appreciable extent. Indeed, early studies concluded that
the BBB was impermeable to IL-1 (85, 161, 204). However,
transport of cytokines via the paracellular route can occur
when BBB integrity is compromised (see sect. VB1), and
saturable transcellular transport mechanisms for a number of cytokines have now been described (see sect. VB2).
1. Cytokines and blood-brain barrier integrity
2. Carrier-mediated transport of cytokines across the
blood-brain barrier
Work by Banks et al. (34) has shown that transcellular, saturable transport mechanisms afford a means of
/ 9j0c$$oc11
P13-8
cytokine entry into the brain, even when BBB integrity
is not compromised. These include saturable transport
mechanisms for IL-1a (35), IL-1b (37), IL-1ra (309), IL-6
(36), and TNF-a (308) but not IL-2 (923). Such transport
mechanisms have been described by injecting 125I-labeled
cytokines intravenously in mice and measuring radioactivity in either whole brain, whole brain perfused free of
blood contamination, CSF, or brain parenchymal homogenates depleted of brain capillaries. It should be noted that
at the doses of cytokines used, no perturbation of BBB
integrity was apparent as indicated by the low and unaltered rates of 125I-labeled albumin entry into the CNS. Not
all radioactive material found in brain after intravenous
cytokine chromatographically elutes as authentic cytokine. In particular, only 16% of radioactivity in brain parenchyma after intravenous 125I-labeled IL-6 was intact IL6 (36), which raises the question of how much of the
125
I-labeled IL-6 entering the brain is actually biologically
active. Nevertheless, multiple time regression analyses
and competition with unlabeled cytokines has demonstrated that such transport of radioactivity into brain is
saturable. The members of the IL-1 family, IL-1a, IL-b,
and IL-1ra share the same transporter(s) (309, 647), but
IL-6 entry into brain is not inhibited by the presence of
unlabeled IL-1a or TNF-a (36), whereas competition studies with IL-1a, IL-1b, IL-6, and MIP-1a have demonstrated
selectivity of the TNF-a transporter (308).
These studies by Banks et al. (34) have demonstrated
that peak values between 0.05 and 0.3% of the total dose
of either 125I-labeled IL-1a, IL-1b, IL-1ra, IL-6, or TNF-a is
found in each gram of whole brain tissue by 20–60 min
after their intravenous injection. Many have questioned
whether this amount of cytokine entry into brain is physiologically significant (678, 711, 943). Indeed, it seems unlikely that such a small proportion of cytokine entering
the brain via a saturable transport mechanism can exert
rapid effects, such as those observed after intravenous
injection of recombinant cytokine. Furthermore, it is improbable that transport of IL-1 across the BBB significantly accounts for the effects of peripherally administered IL-1 on the HPA axis, since the pattern of neuropeptide gene and cIEG expression and the temporal profile
of ACTH secretion differs markedly between peripheral
and central administration (see sects. IIA and IVA3). It
nevertheless seems plausible that such transport mechanisms across the BBB may play a significant role when
peripheral blood levels of endogenous cytokines are elevated for longer periods of time. This would seem particularly pertinent when considering chronic inflammation
where plasma IL-6 levels can remain elevated for prolonged periods (hours to weeks).
C. Role of Readily Diffusible Intermediates
The apparent lack of IL-1 receptors within neuronal
elements in the pPVN and the limited entry of IL-1 into
11-25-98 11:16:36
pra
APS-Phys Rev
Downloaded from on April 23, 2014
Loss of BBB integrity may occur during inflammatory
insults to the brain such as those accompanying CNS disease
(e.g., multiple sclerosis, meningitis, brain tumors, AIDS dementia), brain trauma, cerebrovascular lesions, or seizures
(368). Furthermore, administration of large doses of LPS
can increase BBB permeability (91, 199, 483, 496, 781, 878).
Such disruption of the BBB enables not only the passage of
large peptides such as cytokines, but also augments the rate
of entry of cells, such as macrophages, monocytes, lymphocytes, and neutrophils, which are capable of cytokine synthesis and secretion, but whose passage into the normal,
healthy brain is very limited.
The association between peripheral inflammatory
events and the CNS production of cytokines has led to a
number of studies investigating the possible influence of
cytokines on BBB permeability. In monolayer cultures of
cerebral endothelial cells, either LPS or IL-1b, IL-6 or TNFa produces a decline in transendothelial electrical resistance data supportive of an elevation in BBB permeability
(199, 200). In vivo studies have demonstrated that intracerebroventricular TNF-a increases BBB permeability in the
rat (413) and pig (535), and enhanced brain TNF-a production has been linked to the increased BBB permeability
associated with a number of CNS inflammatory conditions
(768, 769, 780). However, the ability of systemic administration of cytokines (IL-1a, IL-1b, IL-2, IL-6, TNF-a) to
influence BBB integrity has been debated (33, 229, 413,
719). Furthermore, experiments demonstrating increased
BBB after peripheral administration of LPS have either
used exceedingly high doses (483) or have observed increased BBB permeability only over a protracted time
course (496). Enhanced BBB permeability as a result of
either CNS or severe peripheral infection may therefore
permit the entry of cytokines themselves or cytokine-producing cells into the CNS, and cytokines so derived may
contribute to CNS-mediated acute phase responses. However, it is clear that the initial neuroendocrine effects of
peripherally administered cytokines or LPS can be observed more quickly, and at lower doses, than can be
accounted for by damage to the BBB.
33
34
ANDREW V. TURNBULL AND CATHERINE L. RIVIER
the CNS have led to the hypothesis that IL-1 stimulates
the HPA axis via enhancing the production of intermediates that directly interact with hypothalamic neurosecretory processes. These intermediates include classical neurotransmitters, such as catecholamines (see sect. VF), serotonin (285, 477, 478), histamine (293, 421, 638), and
more readily diffusible agents such as the lipid autacoids,
eicosanoids, and the gaseous mediator nitric oxide (NO)
(see sect. VC2).
1. Eicosanoids
/ 9j0c$$oc11
P13-8
plasma ACTH concentrations from the pituitary, the direct
effect of PGE2 on pituitary ACTH secretion in vitro appears to be inhibitory (921). Collectively, these data suggest that the stimulatory actions of PG on ACTH secretion
in vivo are exerted at the level of the hypothalamus or
elsewhere within the CNS.
The majority of studies investigating the effects of
IL-1 or LPS on PG formation have focused on PGE2 , which
is elevated in the systemic blood of either rats or rabbits
injected with LPS (712, 935). Prostaglandin E2 can readily
cross the BBB (183, 224), raising the possibility that PGE2
may enter the brain and stimulate neurosecretory activity
in the pPVN. However, although the onset of the increase
in blood PGE2 levels was reported to occur simultaneously with the onset of fever after either LPS or IL-1 in
the rabbit (712), it is not significantly elevated in the rat
until after the peak of the ACTH response to IL-1b has
been reached (935). Furthermore, administration of doses
of PGE2 that result in blood levels 100- to 400-fold greater
than those observed in blood after IL-1b, has a much
smaller effect on plasma ACTH concentrations than induced by IL-1b itself (935). Collectively, these data indicate that increased entry of PGE2 into the brain from the
systemic circulation is unlikely to be a major intermediary
step by which peripheral IL-1 stimulates HPA axis secretory activity.
In addition to increased circulating levels of PGE2 ,
the brain itself represents a potential source of PGE2 in
response to peripheral injection of IL-1 or LPS. Both isoforms of COX are present in the brain of normal rats or
mice and are expressed predominantly in neurons (100,
104). The patterns of immunostaining for COX-1 and COX2 are distinct and are most prominent in regions of the
brain subserving the processing and integration of visceral
and special sensory inputs, and in the elaboration of autonomic, endocrine, and behavioral responses (100, 104,
400, 877). In the rat, the PGE2 concentrations in the OVLT,
the POAH, PVN, hippocampus, and CSF of the lateral ventricle are all increased within 20 min of intravenous injection of IL-1b (429, 940). All these neuronal structures contain both COX-1 and COX-2 immunoreactivities (100, 104).
Ex vivo measurements indicate that mouse hippocampal
slices produce more PGE2 after either intraperitoneal or
intracerebroventricular treatment with either IL-1a or IL1b (950), whereas in vitro studies have shown that IL-1b
and IL-6 increase the release of PGE2 , but not PGF2a ,
TxB2 , or 6-keto-PGF1a from rat hypothalamic explants
(582). Similarly, peripheral administration of LPS in vivo
induces increased hypothalamic PGE2 production (789,
799), whereas IL-6 or acute local inflammation increase
CSF levels of PGE2 (169, 207). The cellular source of PGE2
in the CNS in response to LPS, turpentine-induced local
inflammation, or IL-1 is most likely the cerebrovasculature
and/or associated perivascular, structures in which COX2 mRNA is markedly induced by these treatments (140–
11-25-98 11:16:36
pra
APS-Phys Rev
Downloaded from on April 23, 2014
Eicosanoids are formed by the metabolism of arachidonic acid. The rate-limiting step in the production of
eicosanoids is the conversion of arachidonic acid to PGH2
by the enzyme cyclooxygenase (COX; also known as PGH
synthase/cyclooxygenase and PG endoperoxide synthase). Two forms of COX have been characterized: a
ubiquitously expressed form (COX-1) and a more recently
described second form (COX-2) that is induced by various
factors including mitogens, hormones, serum, and, of
most relevance to this discussion, cytokines (374, 406,
609, 980, 985). Conversion of the resulting PGH2 by specific synthases results in the generation of three eicosanoid families: the prostaglandins (PG), the thromboxanes
(Tx), and prostacyclins. The role of PG in the activation
of the HPA axis by IL-1 has been particularly well studied
largely because of the recognition that PG are critical
mediators of IL-1 actions within most peripheral tissues
studied (343).
Prostaglandins exert effects on HPA axis secretory
activity at multiple levels. In the rat, elevated plasma
ACTH concentrations are observed within 10 min of intravenous injection of PGE1 , PGE2 , or PGF2a , but not PGD2
(560, 580, 928, 935). Similarly, increases in plasma ACTH
concentration are produced by administration of PGE2
directly into the several brain sites, including the preoptic
anterior hypothalamus (POAH) (397, 931), the OVLT
(397), and the ME (530). Intracerebroventricular administration of PGE2 elicits c-fos mRNA in CRF-containing neurons within the pPVN as well as increased pPVN CRF
hnRNA (445). Indeed, PG receptors of the EP-1 subtype
(45), but not EP-3 subtype (236, 826), have been demonstrated within the PVN. However, the majority of interest
in potential sites of PG action within the CNS has focused
on the POAH. Prostaglandin E2 injected into the POAH
induces Fos expression in the pPVN (742), and ACTH
secretion induced by PGE2 delivered into the POAH can
be prevented by prior treatment with an anti-CRF antiserum (931), indicating the importance of activation of hypothalamic CRF secretion. However, in vitro studies investigating the release of CRF from hypothalamic explants have disagreed, with PGE2 being reported to either
stimulate or have no effect on CRF secretion (57, 135,
652). Despite the fact that intravenous PGE2 elevates
Volume 79
January 1999
REGULATION OF HPA AXIS BY CYTOKINES
/ 9j0c$$oc11
P13-8
generally accepted to be a critical step in their stimulatory
actions on HPA axis secretory activity, the anatomic sites
and cell types responsible for PG synthesis are less clear.
Two possible sites, the cerebrovasculature and CVO, for
which there is substantial evidence that PG play a role in
the transduction of a peripheral IL-1 signal into a CNS
response are discussed in sections VD and VE.
2. Nitric oxide
Nitric oxide is now considered a putative neuromodulator within the mammalian CNS (107, 186, 555). The
localization of NO synthase (NOS), the enzyme responsible for NO formation, within the cerebrovasculature and
the neuroendocrine hypothalamus (885), the readily diffusible nature of NO, and the proposed role of NO in the
mediation of the effects of cytokines (e.g., Refs. 284, 509,
948) provided good evidence for the hypothesis that NO
may play a role in the regulation of the HPA axis by cytokines, in a manner akin, or as an adjunct, to PG. However,
it appears that during activation of the HPA axis by IL-1
or other inflammatory stimuli, the primary effect of NO
is in restraining the HPA axis response (172, 458, 687, 692,
884, 885, 887, 993). The following paragraphs describe the
distribution and regulation of NOS within the HPA axis
and also the experiments investigating the effects of manipulation of NOS on the HPA axis response to IL-1.
Nitric oxide synthase is the enzyme that catalyzes the
conversion of L-arginine to L-citrulline and the gaseous
mediator NO. Several isoforms of NOS have been identified (265). Nitric oxide synthase I is expressed in the central and peripheral nervous systems and is otherwise
known as brain or neuronal NOS. Nitric oxide synthase
II is found in many cell types, such as hepatocytes, macrophages, smooth muscle cells, and glia. Finally, NOS III is
synonymous with endotheial NOS. These three isoforms
are distinct gene products, differ in terms of their pattern
of expression (NOS I and III are constitutively expressed,
whereas NOS II is present only after induction by cytokines or endotoxin), are either calcium/calmodulin dependent (NOS I and III) or independent (NOS II), and exhibit
different kinetic properties. That NO acts as a neuromodulator was first indicated by the demonstration that inhibitors of NOS block the stimulation of cGMP synthesis in
brain slices by glutamate acting at NMDA receptors (276).
However, NO clearly does not behave like a conventional
neurotransmitter, since it is neither stored in nerve terminals nor does it influence its target cell via interaction
with a cell-surface receptor. Rather, it diffuses from nerve
terminals and forms covalent linkages with several potential postsynaptic intracellular targets (e.g., guanylyl cyclase).
Nitric oxide synthase-like activity, NOS immunoreactivity, and NOS mRNA are present within the PVN and
SON of the hypothalamus (19, 107, 127, 129, 147, 456, 917–
11-25-98 11:16:36
pra
APS-Phys Rev
Downloaded from on April 23, 2014
142, 231, 444, 903, 904) (see sect. VD). Microglia (231),
neurons (140, 903), meningeal macrophages (231), and
astrocytes (652) represent other possible sources of PG
in the brain.
Inhibitors of COX activity, such as indomethacin or
ibuprofen, have been the most commonly used method
of investigating the role of PG in HPA axis responses to
IL-1. As such, these studies indicate the importance of
arachidonic acid metabolites but do not specifically identify PG as the principal players. Peripheral administration
of indomethacin abrogates the rise in plasma ACTH concentrations due to intravenous administration of IL-1a,
IL-1b, or TNF-a (560, 569, 688, 694, 771, 880, 930). The
inhibition observed is not always complete and, in particular, appears to be short-lived, even when indomethacin or
ibuprofen is administered several times (C. Rivier, unpublished data). Similar abrogation of IL-1-induced ACTH secretion is produced by indomethacin when IL-1 is injected
intracerebroventricular (399, 688, 930) or locally into the
ME (530), but results have been inconsistent when IL-1b
has been injected intraperitoneally (215, 685). In response
to peripheral LPS or local inflammation, administration
of COX inhibitors reduces ACTH secretion, an effect that
cannot be accounted for by effects of COX inhibition on
the inflammation per se (880, 884). Peripheral administration of indomethacin also inhibits the expression of Fos
in the pPVN of rats injected intravenously, intraperitoneally, or intracerebroventricularly with either IL-1b or LPS
(443, 588, 718, 925) or of rats infused continuously with
IL-6 (intravenous) (589).
Because indomethacin or ibuprofen can elevate basal
GC secretion, it has been suggested that the inhibitory
effects of COX inhibitors on cytokine- or inflammationinduced ACTH secretion may be due to enhanced GC
feedback. However, a number of lines of evidence indicate
that this is probably not the case. For example, elevations
of plasma ACTH concentrations produced by either IL-1b
(694) or TNF-a (771) in adrenalectomized rats are also
abrogated by indomethacin inhibitors, whereas ACTH secretion due to a variety of other stimuli, including intravenous CRF (771, 880), electrofootshock (880), immobilization (399), or insulin-induced hypoglycemia (771), is unaffected by COX inhibition. In vitro studies have also
demonstrated that CRF release from hypothalamic explants produced by either IL-1a, IL-1b, or IL-6 is prevented
by indomethacin (56, 135, 500, 583). Finally, in addition to
the abrogating effects of COX inhibitors on IL-1-induced
ACTH secretion, prior intracerebroventricular passive immunoneutralization with antibodies to either PGE1 , PGE2 ,
or PGF2a also produces a significant reduction in IL-1induced elevations in plasma ACTH (937). Collectively,
these data argue for a direct role of PG in the mediation
of IL-1 induced activation of the HPA axis.
Although the generation of PG in response to IL-1,
and a number of other cytokines (e.g., TNF-a, IL-6), is
35
36
ANDREW V. TURNBULL AND CATHERINE L. RIVIER
/ 9j0c$$oc11
P13-8
tocin (692). These latter findings raise the possibility that
increased sensitivity of corticotropes to AVP and/or oxytocin after L-NAME accounts for its potentiation of IL1b-induced ACTH secretion. However, administration of
neutralizing antisera to AVP does not influence the exacerbation of IL-1-induced ACTH secretion produced by LNAME (687). Recent studies (887) with partially selective
NOS inhibitors identify endothelial (type III) NOS as the
most likely source of NO that inhibits the HPA axis response to IL-1b, raising the possibility of functional interactions between NO and IL-1 signaling pathways in cerebrovascular elements (see sect. VD). On the other hand,
administration of combined adrenergic antagonists, propanolol and prazosin, partially reverses the effects of LNAME on IL-1-, but not AVP-, induced ACTH secretion
(687), suggesting that the inhibitory influence of NO on
HPA activity may be due to a suppressive effect on catecholaminergic pathways (see sect. VF).
It is interesting to note that the suppressive effect
of NO on the HPA axis response to systemic IL-1 has
proinflammatory consequences because of the diminution
of endogenous GC levels. This proinflammatory neuroendocrine action of NO is in agreement with its local actions
as an inflammatory mediator at the site of tissue damage
or infection (449). Therefore, NO appears to represent a
new class of mediator that belongs to the large families
of cytokines, neurotransmitters, and hormones that constitute the common chemical language of the immune and
neuroendocrine systems.
D. Induction of Intermediates at Blood-Brain
Barrier Interface
Although there has been much debate as to the precise location of IL-1 receptors within neuronal elements
of the brain (see sect. IVA1A), all studies agree that the
cerebrovasculature and perivascular elements are the
most prominent loci of IL-1R1 mRNA expression in rat or
mouse brain (178, 238, 969, 970, 972, 982). Intense signal
for IL-1R1 mRNA expression has been described over endothelial cells of the postcapillary venules throughout the
entire brain (178, 238, 972, 982), and a number of lines of
evidence support a role for such endothelial IL-1 receptors
in the transmission of IL-1 signal from blood to brain.
Immunoreactive IL-1b is found on the luminal side of
endothelial cells lining cerebral venules in rats treated
peripherally with LPS (901), whereas increased PGE2 immunostaining has been detected in the cerebral microvasculature after peripheral injection of either IL-1b (904) or
LPS (903). Indeed, perivascular elements (probably perivascular microglia) are the major site of COX-2 mRNA
induction after systemic treatment with LPS, turpentine,
or IL-1 (103, 140–142, 231, 444). Perivascular cells were
identified as the primary cell group within the CNS that
11-25-98 11:16:36
pra
APS-Phys Rev
Downloaded from on April 23, 2014
919). In the pPVN, NOS is found within a subpopulation
of CRF-expressing neurons (782, 870). Circumstances
known to alter the activity of the pPVN, e.g., immobilization stress (129), intracerebroventricular CRF injection
(459), intracerebroventricular IL-1b (459), or systemic
LPS treatment (456), upregulate NOS expression in this
nucleus, suggesting a role for NO in the regulation of HPA
axis. In addition, like the components of the IL-1 system,
NOS mRNA is expressed in cerebrovascular elements
(969, 974). With regard to localization within the pituitary
gland, NOS is found in both the posterior (106, 107) and
anterior (148) pituitary. In the anterior pituitary, NOS
mRNA and protein are expressed at only low levels in
normal animals but are markedly induced by systemic
LPS (974). Nitric oxide synthase is also present in the
hypophysial portal vasculature (149), which delivers hypothalamic releasing factors to the anterior pituitary.
Possible roles of NO in biological processes have
been determined using NO donors (L-arginine, nitroprusside), inhibitors of NOS activity (arginine derivatives), and
NO scavengers (hemoglobin). Inhibitors of NOS activity
are generally low molecular mass (150–500 Da), and the
BBB does not appear to hinder their diffusion. These inhibitors have therefore been studied after either systemic
or central injection, the latter being chosen when effects
on systemic parameters, in particular, blood pressure,
were to be avoided.
Intravenous pretreatment of rats with the NOS substrate L-arginine blunts the rise in plasma ACTH due to
intravenous IL-1b (692). Conversely, the NOS inhibitor
NG-nitro-L-arginine methyl ester (L-NAME) (intravenous)
exaggerates and prolongs the ACTH and corticosterone
responses to IL-1b, an action which is specific to the Lisomer of NAME (the isomer active at NOS), can be reversed by competition with L-arginine, and is unrelated to
its hypertensive effects (687, 692, 885, 887). Thus endogenous NO appears to inhibit the ACTH response to systemic IL-1b. Potentiation of ACTH secretion by L-NAME
is also observed during responses to turpentine-induced
local inflammation and systemic LPS treatment, but interestingly, not to IL-1b administered directly into the brain
(intracerebroventricular) (692, 884).
The mechanisms by which NO influences the HPA
response to IL-1b, inflammation, and endotoxemia are far
from clear, but a number of findings have shed some light
on likely candidates. Because CRF is an obligatory mediator of the ACTH response to IL-1, a number of in vitro
studies have investigated whether NO influences hypothalamic CRF/AVP secretion. However, in hypothalamic explants, NOS inhibitors have been reported to either blunt
(116, 724) or exacerbate (172, 993) the increase in CRF/
AVP secretion produced by IL-1. In vivo, L-NAME does
not markedly influence ACTH secretion produced by
treatment with CRF but potentiates the ACTH response
to the two less potent ACTH secretagogues, AVP and oxy-
Volume 79
January 1999
REGULATION OF HPA AXIS BY CYTOKINES
E. Actions at Circumventricular Organs
In addition to cytokine entry into the CNS by way of
carrier-mediated transport or diffusion through a disrupted BBB, a number of regions of CNS relatively devoid
of a BBB permit cytokine interaction with neuronal elements. These CVO include structures lining the anteroventral border of the third ventricle (AV3V) (namely, the
/ 9j0c$$oc11
P13-8
OVLT and SFO), the ME, and the AP, posterior lobe of
the pituitary, subcommisural organ, and the pineal gland.
Their capillaries do not form tight junctions and are thus
far more readily penetrable via the paracellular route
(305). Circumventricular organs not only contain capillaries with far greater permeability than the rest of the CNS,
but the capillary density in these regions is extraordinarily
high (369). Penetration of cytokines into brain parenchyma within the CVO does not imply that they may diffuse to interact with deeper brain structures, however,
because tight junctions between modified ependymal cells
in these regions form a diffusion barrier between CVO
and the rest of the brain (369). But these ‘‘leaky’’ sites do
provide a means with which cytokines such as IL-1 can
influence neuronal activity in these regions of the CNS.
The CVO that have been proposed as potential sites of
IL-1 action are the structures lining the AV3V region (in
particular the OVLT), the ME, and the area postrema, and
these are considered further in sections VE1–3.
1. Structures lining the anteroventral border of the
third ventricle
Substantial evidence has accumulated that the OVLT
may serve as an interface between the blood and the brain
during the process of an inflammatory response (568,
821). In particular, the role of the OVLT during fever induced by various cytokines or inflammatory stimuli has
been well documented, and the OVLT has been proposed
as a ‘‘pathway’’ for circulating IL-1 to influence neuronal
activity (86, 568, 820, 822).
Efferent projections from both the OVLT and the SFO
to the PVN provide good anatomic evidence indicating
an influence of these structures on the activity of CRF
neurosecretory neurons (369, 474, 783). A number of anatomic studies have therefore investigated whether the
AV3V region may be a site responsive to the systemic
administration of IL-1b. Induction of c-fos mRNA in the
OVLT and SFO has been noted within 30–60 min of intraperitoneal injection of IL-1b (98, 187), and Fos protein
expression is apparent at 3 h after intravenous IL-1b (237).
Similarly, c-fos mRNA has been detected in the OVLT and
SFO after intravenous injection of IL-6 (899) and after
peripheral injection of LPS (232, 679, 718). The SFO is
one of the select regions of the rat brain that displays
changes in protein synthesis within 1 h after subcutaneous
injection of IL-1b (962), whereas AV3V neurons identified
electrophysiologically as projecting to the PVN are responsive to IL-1b within 15 min of its intra-arterial injection (610). Experiments injecting colloidal gold-labeled IL1b (intravenous) into rabbits demonstrated that within 10
min of its intravenous injection, IL-1b is bound to the
luminal surface of the endothelium within the OVLT (330).
This is followed by internalization of IL-1b and transport
to the basal membrane, although IL-1b was not observed
11-25-98 11:16:36
pra
APS-Phys Rev
Downloaded from on April 23, 2014
express both IL-1R1 and IL-1b-induced cIEG expression
1 h after intravenous injection of IL-1b (238). Finally, IL1R1 expression by perivascular cells, but not neuronal
elements, exhibits the expected ligand-induced downregulation after intravenous IL-1b (238).
The microvasculature has long been recognized as a
target and source of cytokines, with a number of studies
focusing specifically on the cerebrovasculature. Such
studies have shown that cerebral endothelial cells express
cytokine ligands (e.g., IL-1a, IL-1b, IL-6), cytokine-related
enzymes (ICE), or receptors (e.g., TNF, IL-1) and are sites
of cytokine action (49, 51, 171, 190, 242, 380, 713, 970).
Cytokine induction of readily diffusible intermediates,
such as NO (412, 969) or PG (77, 78, 201, 904), represents
a possible means by which cytokines within the bloodstream can influence cerebral function.
A number of in vitro paradigms have been developed
to investigate PG formation in the cerebrovasculature. Addition of LPS to isolated feline cerebral microvessels (consisting mainly of capillaries) has been shown to increase
their secretion of PGE2 but not 6-keto-PGF1a (77, 78).
Studies by Van Dam et al. (904) have demonstrated that
cultured rat cerebral endothelial cells (RCEC) bind rat IL1b specifically and express IL-1R1, but not IL-1R2, mRNA.
Prostaglandin E2 is the main arachidonic acid metabolite
found in rat cerebral endothelial cells (201), and IL-1b
increases their secretion of PGE2 and 6-keto-PGF1a , but
not TxB2, within 3 h of incubation. As with IL-1b, IL-6
stimulates PGE2 and 6-keto-PGF1a secretion within a period of 3 h (201).
Induction of PGE2 secretion through the serosal surface of endothelial cells by luminally confined IL-1 (or
possibly other cytokines) represents an extremely plausible means by which IL-1 in the circulation could influence
the neuroendocrine hypothalamus. Prostaglandin formation in endothelial/perivascular cells expressing IL-1 receptors within the PVN (178, 982) may provide a relatively
local and direct mechanism of HPA axis activation in response to blood-borne IL-1. However, other lines of evidence also indicate that CVO and/or medullary catecholaminergic cell groups are important anatomic structures
in mediating the effects of IL-1 on the HPA axis, and there
is considerable evidence that PG formation (possibly in
perivascular elements) occurs in these regions in response to IL-1.
37
38
ANDREW V. TURNBULL AND CATHERINE L. RIVIER
2. Median eminence
The ME contains the terminal projections from CRF
secretory neurons arising from the pPVN. Being an area
of the hypothalamus that is richly supplied by vasculature
relatively devoid of a BBB, the ME is a potential site by
which circulating factors can enhance CRF secretion by
interacting with pPVN nerve terminals, and stimulating
CRF release without directly stimulating CRF cell bodies
in the pPVN. Evidence supporting the concept that the
/ 9j0c$$oc11
P13-8
ME is a primary site of action of circulating IL-1 in the
stimulation of CRF secretion derives from a number of
experimental findings. For example, microinjection of IL1a, IL-1b, or IL-6 into the ME rapidly stimulates ACTH
secretion (525–527, 530, 772). The doses of IL-1 or IL-6
required to elicit increases in plasma ACTH concentration
after microinjection into the ME are significantly, although not markedly (2- to 5-fold), less than those required to elicit ACTH after intravenous administration of
cytokine. As demonstrated for intravenous or intracerebroventricular injection of IL-1, the plasma ACTH response to intra-ME IL-1b is markedly reduced by indomethacin (530), suggesting the importance of PG formation as an intermediate step. However, in vitro studies
attempting to determine whether the enhanced CRF release observed after incubation of hypothalamic explants
(which include the ME) can be accounted for by actions
on the ME have yielded conflicting data (583, 813, 863).
Although intravenous IL-1b induces c-fos mRNA and
Fos protein in the PVN at 60–180 min, the observation
that intracerebroventricular and intravenous injection of
doses of IL-1b elicit differential c-fos mRNA expression
in the PVN at 30 min suggested that the ME may be the
primary target after intravenous IL-1b (684). This conclusion was based on the fact that both routes of IL-1b administration rapidly stimulate ACTH secretion, yet c-fos
mRNA in the pPVN 30 min after IL-1b was observed only
after intracerebroventricular IL-1b. This suggests that
pPVN cell bodies represent an early target of IL-1b after
its intracerebroventricular, but not intravenous, administration. Because both routes of IL-1b administration elicit
rapid (within minutes) increases in CRF secretion, it
seemed likely that IL-1b administered intravenously acts
at the level of the ME (684). However, subsequent neuroanatomic data supporting this hypothesis are not strong.
In situ histochemical studies have described the expression of IL-1R1 mRNA in the rat ME to be either weak
(982) or absent (238, 972), whereas in the mouse, only
labeling on ‘‘scattered cells’’ is apparent (178). Radioligand binding studies have similarly shown either low
(251) or no (516) signal within the rat ME and no specific
signal in the mouse ME (30, 848). Indeed, although LPS
induces c-fos mRNA in several layers of the ME (679), cfos mRNA or Fos protein induction has not been observed
in the ME in response to systemic IL-1b (98, 187, 237,
588).
3. Area postrema
Recent studies implicated the AP in the mediation of
HPA activation in response to IL-1b administered intravenously (238). Whereas other CVO do not contain IL-1
receptors to any appreciable extent (at least in neuronal
elements), the AP was shown to contain positive hybridization signal for IL-1R1 mRNA, although the cell type
11-25-98 11:16:36
pra
APS-Phys Rev
Downloaded from on April 23, 2014
in the neighboring POAH (330). In addition to being responsive to IL-1b, the AV3V regions have been demonstrated to be a site of production of IL-1b in response to
systemic treatment with LPS (see sect. IVA2B).
Consistent with a role for the AV3V in mediating the
effects of systemic IL-1b action on the HPA axis, placement of either radiofrequency or kainic acid-induced lesions in the OVLT markedly reduces ACTH secretion in
response to intravenous IL-1b, but not immobilization
stress (397). However, there is little or no evidence for
the existence of IL-1 receptors within neuronal elements
within the AV3V. With the consideration of the density of
vasculature in this region and the demonstration of IL-1
receptors on endothelial and perivascular cells, these may
well represent the cellular targets of IL-1b within the
AV3V. Consistent with the idea of PG formation being the
principal action of IL-1b in the AV3V, enhanced PGE2
formation in this region has been noted after systemic
treatment with IL-1b or LPS (429, 903, 940). Furthermore,
microinjection of indomethacin into the OVLT markedly
attenuates the rise in plasma ACTH concentrations due
to IL-1b (397). That adjacent neuroanatomic structures
such as the POAH represent the target of PG action is
supported by a number of observations. The POAH (but
not the OVLT itself) contains a high density of PGE2 binding sites (523, 524, 933, 934), mRNA for the PG receptor
EP-3 (236, 826), and the plasma ACTH response to intravenous IL-1b is inhibited by either electrolytic or kainic acidinduced lesions of the POAH or by microinjection of a
PG antagonist into this brain region (397).
Although there is substantial evidence that the AV3V,
and in particular the OVLT, may serve as an neuroanatomic site of action of circulating IL-1b, none of the experiments investigating the effects of either surgical or pharmacological manipulation of the OVLT showed complete
inhibition of the plasma ACTH response to IL-1b (397).
Furthermore, the dose of IL-1b used in these studies was
extremely high (10 mg/kg iv). In more recent dose-response studies, the minimum dose of IL-1b required to
induce Fos protein in the OVLT (3.58 mg/kg) was found
to be Ç10-fold higher than that required to stimulate Fos
expression in the pPVN (237), suggesting that the involvement of the OVLT in the HPA axis response to systemic
IL-1b may be restricted to high circulating levels of IL-1b.
Volume 79
January 1999
REGULATION OF HPA AXIS BY CYTOKINES
expressing IL-1R1 could not be identified. Most importantly, the AP is the only CVO that reliably displays intravenous IL-1b-induced cIEG expression (237, 238). However, ablation of the AP does not influence IL-1b-induced
CRF mRNA or Fos expression within the PVN (235), suggesting that it is not a critical relay site in the activation
of the hypothalamic PVN in response to IL-1b.
F. Potential Role of Catecholamines and Evidence
of Activation of Medullary Cell Groups
/ 9j0c$$oc11
P13-8
medullary catecholaminergic fibers (using 6-hydroxydopamine) consistently produce a marked depletion of hypothalamic NE concentrations and inhibit the plasma ACTH
or corticosterone response to intracerebroventricular or
intraperitoneal IL-1b (160, 949). A similar inhibitory effect
has been noted when 6-hydroxydopamine is infused into
the lateral ventricle (526, 949) or directly into the PVN
(160). Discrete lesions of ascending catecholaminergic inputs to the hypothalamus (affecting either dorsal, ventral
or intermediate axons) showed that the plasma ACTH
responses to either intra-arterial IL-1b or LPS, or intraPVN IL-1b, is dictated by brain stem catecholaminergic
input to the hypothalamus and that the effect of lesion
was dependent on the route of cytokine administration
and precise location of the lesion (39, 291).
Recent studies have provided even more substantial
evidence for the critical role of medullary catecholaminergic innervation of the hypothalamus in the activation of
the HPA axis in response to systemic IL-1. Administration
of IL-1b, either intravenously or intraperitoneally, results
in marked expression of c-fos mRNA or Fos protein in
the NTS of the rat (98, 187, 237). Similarly, all studies
investigating the CNS distribution of either Fos protein
of c-fos mRNA in response to systemic LPS have noted
induction of this early-immediate gene expression in
(likely catecholaminergic) medullary cell groups within
1–3 h of LPS injection (230, 232, 678, 679, 718, 924). Ericsson et al. (237) described Fos protein expression throughout the rostrocaudal extent of the medial and commisural
nuclei of the NTS, as well as in the rostral ventrolateral
medulla and in the dorsal motor nucleus of the vagus
within 3 h of intravenous IL-1b. These authors demonstrated that Fos expression in the NTS is very sensitive
to IL-1b and is induced at doses lower than those required
to elicit a specific signal within the pPVN. Retrograde
tracing studies indicated that the NTS and rostral ventrolateral medulla are the sources of projections to the PVN
that most consistently express IL-1b-induced Fos protein
(237). The majority of these projections are catecholaminergic (237). Furthermore, unilateral surgical disruption of
ascending catecholaminergic projections from the medulla to the PVN prevents intravenous IL-1b-induced expression of Fos protein and CRF mRNA in the ipsilateral,
but not contralateral, pPVN (237, 468). Finally, although
such a surgical disruption of medullary-PVN fibers does
not prove a catecholaminergic involvement, a recent
study demonstrated that 6-hydroxydopamine lesions also
substantially reduce intraperitoneal IL-1b-induced Fos expression in the PVN of the mouse (831).
The activation of medullary catecholaminergic cell
groups seems unlikely to be because of a direct activation
by IL-1, since these cell groups do not express IL-1R1
mRNA (238). Given the demonstrated critical role of PG
in the HPA axis response to IL-1 (see sects. VC1 and VD),
it is not surprising that there has been considerable inter-
11-25-98 11:16:36
pra
APS-Phys Rev
Downloaded from on April 23, 2014
In addition to PG, there is a second class of intermediates for which there exists substantial evidence in the
mediation of IL-1-induced activation of the HPA axis,
namely, monoamines. Monoamine utilization and/or content in the hypothalamus and other regions of the CNS is
altered by immunologic challenges, such as those produced by inoculation with sheep red blood cells (69, 767,
997), NDV (220, 221), Poly I:C (221), and influenza virus
(219) and by peripheral administration of LPS (193, 212,
222, 450, 478). Intraperitoneal administration of either IL1a or IL-1b to mice also elevates cerebral metabolism of
norepinephrine (NE) and serotonin, with effects being
most marked in the hypothalamus (211, 222). Similarly,
intravenous or intraperitoneal IL-1b increases NE turnover in the hypothalamus (including PVN) as well as other
regions of the rat brain (386, 553, 790). Intrahypothalamic
or intracerebroventricular infusion of IL-1b also modulates hypothalamic monoamine levels (554, 773, 776, 860).
As for other cytokines, intraperitoneal injection of IL-2
increases hypothalamic NE turnover in mice (996), and
peripheral administration of IL-6 produces either small
(222) or no increases (213, 858, 996) in hypothalamic NE
and serotonin turnover. Intraperitoneal TNF-a was found
to have no effect on hypothalamic NE turnover in mice
(213) or rats (858) but was reported to inhibit NE release
from the isolated ME in vitro (226).
There is extensive evidence that noradrenergic innervation of the hypothalamus influences the secretion of
CRF from the hypothalamus (reviewed in Refs. 9, 649,
727, 739, 833). The PVN and SON represent two of the
most prominent terminal fields of catecholaminergic neurons. These projections derive from medullary cell groups,
in particular the NTS. Noradrenergic and adrenergic neurons from the rostral and caudal regions of the NTS project preferentially to the pPVN, as does the major projection field of the NTS, the C1 adrenergic cell group (739).
Lesion of pathways from medullary catecholaminergic
cell groups to the PVN has been reported to disrupt HPA
axis responses to diverse stimuli (181, 832).
The use of adrenergic receptor antagonists to explore
the role of catecholamines in IL-1-induced activation of
the HPA axis has produced inconsistent data (134, 695,
949). On the other hand, neurotoxic lesions of ascending
39
40
ANDREW V. TURNBULL AND CATHERINE L. RIVIER
G. Activation of Vagal Afferent Fibers
Studies over the last few years have indicated a novel
route through which peripheral cytokines may influence
the CNS without gaining access to brain parenchyma, the
BBB interface, or even the systemic circulation. In 1994,
Nance and co-workers (925) demonstrated that the induction of Fos protein in the PVN and SON of the hypothalamus produced by an intraperitoneal injection of LPS is
prevented by surgical subdiaphragmatic transection of the
vagus (SDVX) several days before LPS administration
(925). At a similar time, Dantzer and co-workers (90)
showed that SDVX reduces sickness behavior after intraperitoneal LPS, whereas Watkins et al. (944) reported that
hyperalgesia produced by intraperitoneal LPS is also reduced by prior sectioning of the vagus (944). These data
suggested that the vagus is a neural afferent route by
which inflammation within the peritoneal cavity can influence the brain. A number of independent laboratories
have now confirmed the existence of such a mechanism in
rats, mice, and guinea pigs. The SDVX procedure reduces
fever (297, 389, 607, 701, 761), sleep (389, 607), NE turnover (359), food-motivated sickness behavior (90, 108,
/ 9j0c$$oc11
P13-8
451), and the elevation of hypothalamic IL-1b mRNA (451)
induced by the administration of LPS into the abdominal/
peritoneal cavity.
Dantzer and co-workers (90, 451) demonstrated that
the inhibitory effect of SDVX on CNS-mediated responses
to intraperitoneal LPS was not due to a reduced peripheral
inflammatory response, since LPS-induced expression of
cytokines in tissues other than the brain is unaffected by
SDVX (90, 451). This suggests that it is the transduction
of a cytokine signal to the brain that is impaired in SDVX
animals. Indeed, it has now been shown that the fever
(607, 942), increased sleep (319), NE turnover (261), hyperalgesia (945), behavioral effects (88, 89, 294), and induction of IL-1b mRNA in the CNS (320) produced by
intraperitoneal IL-1 are all diminished or absent in SDVX
animals. Similarly, SDVX inhibits hyperalgesia and conditioned taste aversion induced by intraperitoneal TNF-a
(294, 941). Although SDVX inhibits CNS-mediated effects
of intraperitoneal TNF-a (e.g., hyperalgesia, activation of
the HPA axis), ‘‘peripheral organ’’ effects (e.g., reduction
of glucocorticoid binding globulin) are unaffected by this
surgical procedure (262), suggesting that the role of the
vagus is specific to CNS-mediated acute phase responses.
The vagus also appears to play a role in the activation
of the HPA axis in response to peripheral LPS or cytokine.
In rats, SDVX attenuates the rise in plasma ACTH and
corticosterone concentrations produced by intraperitoneal IL-1b (261, 390). The number of CRF containing neurons in the pPVN that stain positive for Fos and the rise
in plasma ACTH concentration produced by LPS are likewise attenuated in SDVX animals (279), indicating that
vagotomy interferes with the activating signal to the neuroendocrine hypothalamus. Similarly, SDVX blocks the
rise in plasma corticosterone caused by intraperitoneal
TNF-a (262).
A number of studies have addressed the question of
how SDVX interferes with the generation of CNS-mediated acute phase responses to either LPS or IL-1. The
SDVX procedure is a major surgical intervention resulting
in SDVX animals being less ‘‘healthy’’ than sham-operated
or control rats. It has been suggested that the inability of
SDVX rats to respond normally to inflammatory stimuli
may relate to their overall health status rather than to a
specific effect of SDVX on ‘‘immune-to-brain’’ communication. However, even in SDVX rats receiving special perioperative care to prevent malnutrition, the febrile response
to intraperitoneal LPS is still impaired (699). In addition,
SDVX animals respond with elevations in body temperature (543, 700), hyperalgesia (941), and activation of the
HPA axis (390, 943) in a manner similar to sham-operated
animals when the stimulus is not an inflammatory one
(e.g., insulin- or electrofootshock-induced elevations in
plasma ACTH concentrations).
Although the importance of vagal afferents in the mediation of CNS-mediated acute phase responses to abdom-
11-25-98 11:16:36
pra
APS-Phys Rev
Downloaded from on April 23, 2014
est in the possibility that PG are an important intermediary between IL-1 action and activation of medullary catecholaminergic neurons. Accordingly, the induction of
medullary Fos protein (235) and the changes in hypothalamic monoamine turnover (534, 857, 858, 860) produced
by IL-1 have been shown to be prevented by COX inhibitors. Indeed, the PG receptor subtype EP-3 is present in
the NTS and ventrolateral medulla, in which IL-1-sensitive
catecholamine neurons reside (236). Furthermore, the administration of PGE2 into the rostral ventrolateral medulla
provokes a pattern of Fos expression in hypothalmic nuclei (including the pPVN) that closely resembles that produced by intravenous IL-1b (235). These observations
have led to the conclusion that PGE2 released from perivascular cells in the medulla as a consequence of IL-1
stimulation activates local catecholaminergic neurons
that project to the PVN (235).
In addition to a role in the activation of the HPA
axis produced by IL-1 interacting with neuronal and/or
cerebrovascular elements within the brain, a further importance of medullary catecholaminergic cell groups in
the mediation of cerebral influence of peripheral IL-1 has
recently become apparent. Medullary cell groups play an
important role in the processing of visceral sensory information carried by sensory components of the vagus and
glossopharyngeal nerves (739). Recent evidence that an
intact vagus is critical to elaboration of CNS responses to
abdominal/peritoneal inflammation (see sect. VG) suggests that medullary cell groups may receive information
regarding the localized peripheral activity of IL-1 (238).
Volume 79
January 1999
REGULATION OF HPA AXIS BY CYTOKINES
H. Cytokine Synthesis Within Brain
The previous discussions focussed on answering the
question of how IL-1 generated within the periphery may
signal the brain to elicit HPA activation. Recent studies
have demonstrated that IL-1, as well as other cytokines,
are also generated within the brain (see sect. IVA2), thus
raising the question of whether such brain-derived IL-1
may influence HPA axis secretory activity. The present
data regarding the distribution of IL-1 receptors within
the brain would seem to argue against a role for cerebral
/ 9j0c$$oc11
P13-8
IL-1 in regulating the activity of the neuroendocrine hypothalamus, since IL-1 receptors are located predominantly
in barrier-related regions (e.g., perivascular elements)
suited to transducing an IL-1 signal from blood to the
brain, but not to mediating effects of IL-1 generated within
brain. The only hypothalamic nucleus that expresses IL1 receptors on neuronal elements is the arcuate nucleus,
which is known to influence HPA axis secretory activity
(238), but whose role in mediating the effects of IL-1 has
not been determined. Despite a lack of evidence for an
action of cerebral IL-1 on HPA axis secretory activity
based on the distribution of IL-1 receptors within the brain
parenchyma, IL-1 administered directly into the brain induces Fos expression in the pPVN, elevates PVN CRF
mRNA, enhances CRF secretion from the ME, and increases plasma concentrations of ACTH and corticosterone (see sect. IVA3). Furthermore, the doses of IL-1
needed to stimulate HPA axis secretory activity are lower
than those required when the cytokine is administered
peripherally, clearly indicating a CNS site of action of
cerebrally administered IL-1. Finally, incubation of the
hypothalamus with IL-1 in vitro elicits CRF secretion (see
sect. IVA3). Collectively, these data argue strongly that
when CNS production of IL-1 is increased it is likely to
stimulate HPA axis secretory activity.
Pathologies comprising direct cellular insults (infection, trauma, ischemia, and disease) to the CNS clearly
induce the synthesis of IL-1 and other cytokines within the
brain. However, only very few studies have investigated
cytokine regulation of the HPA axis during such pathologies, although the work performed has implicated brainderived IL-1 in the activation of the HPA axis in response
to CNS viral disease. For example, intracerebral infusion
or transgenic expression of gp120, the envelope protein
of the human immunodeficiency virus, causes an increase
in brain IL-1 biological activity and mRNA and elevates
plasma ACTH and corticosterone concentrations (112,
355, 657, 661, 827). Coinfusion of a-melanocyte-stimulating hormone (which inhibits IL-1 actions, but not synthesis) completely prevents the elevation in plasma corticosterone concentrations, suggesting that IL-1 generated in
brain as a consequence of gp120 infusion produces activation of the HPA axis (827). Similarly, induction of IL-1b
within the brain has been proposed as the mechanism
responsible for increased adrenocortical activity in rats
inoculated with the neurotropic herpes simplex virus-1
(50).
Many studies have begun to address a question of
much broader significance to the concept to neuroimmune regulation, i.e., whether events unrelated to direct
cellular insults to the CNS can induce the synthesis within
the brain of cytokines, which subsequently regulate CNS
activity, and play a physiological role in orchestrating
adaptive responses. In particular, work has focused on
the possibility that an immune/inflammatory response
11-25-98 11:16:36
pra
APS-Phys Rev
Downloaded from on April 23, 2014
inal inflammation is not undisputed (e.g., Ref. 755), the
weight of evidence supporting this hypothesis is now substantial, and a number of studies have begun to unravel
how cytokines might interact with the vagus. Administration of IL-1 into the hepatic portal vein produces increased
electrical activity of the hepatic branch of the vagus (586,
587), suggesting the possible importance of this vagal
branch. However, in contrast to SDVX, selective sectioning of the hepatic vagus does not inhibit the rise in
either plasma corticosterone or hypothalamic NE turnover in response to intraperitoneal IL-1 (261), or the suppression of food intake in response to LPS (447). Similarly,
the hypothesis that C-fiber afferents are the principal vagal fiber type important in the vagal signal to the brain
has been disproven (109). What does seem clear at the
present time is that while abdominal vagal afferents themselves do not appear to contain functional IL-1 receptors,
abdominal paraganglia, which are in close proximity to
and synapse with vagal fibers, bind biotinylated IL-1ra
specifically (296). Furthermore, macrophages are found
close to vagal fibers and paraganglia after treatment with
LPS (295), suggesting a close association between IL-1producing cell types, IL-1 receptors, and abdominal vagal
afferents.
The vagus appears to be important in signaling the
brain specifically during intra-abdominal/peritoneal inflammation. This is evidenced by the lack of effect of
SDVX on CNS-mediated acute phase responses, such as
activation of the HPA axis (235, 399), when either IL-1 or
LPS is administered via routes other than into the abdomen/peritoneum (88, 89, 235, 297, 399, 645). However,
global depletion of C-fiber afferents (by repeated systemic
treatment with capsaicin during adulthood) inhibits the
plasma ACTH response to intravenous IL-1b (932). Furthermore, the initial (within 45 min) rise in plasma ACTH
concentrations produced by intramuscular turpentine is
reduced in rats treated neonatally with capsaicin (879).
Therefore, it seems possible that sensory fibers, other
than vagal abdominal/peritoneal afferents (e.g., from skin
or muscle), are capable of signaling the brain of the occurrence of high local concentrations of cytokines in the
periphery.
41
42
ANDREW V. TURNBULL AND CATHERINE L. RIVIER
/ 9j0c$$oc11
P13-8
LPS (2.5 mg ip). These authors simultaneously measured
the quantities of human IL-1ra that escaped into the general circulation and in separate experiments demonstrated that these levels in plasma had no effect on the
increase in CRF mRNA in response to LPS. Therefore, it
would appear that IL-1 acts within the brain to stimulate
the increase in CRF mRNA in response to peripheral LPS,
although concomitant plasma ACTH levels were not measured. However, whether or not the IL-1 responsible for
elevating CRF mRNA was of CNS rather than peripheral
origin can still not be assumed.
We also found evidence indicating that TNF-a may
act within the brain to activate the HPA axis in response to
local inflammation. The intracerebroventricular, but not
intravenous, administration of either 5 ml anti-TNF-a antiserum or 1–50 mg of a soluble TNF receptor construct
inhibits the second rise in plasma ACTH concentrations
produced by intramuscular turpentine (881). Because no
increases in plasma TNF-a are apparent during discrete,
localized inflammation, we assumed that TNF-a not only
acted within the brain but that the CNS was the major
source of TNF-a. However, we found no evidence of elevated TNF-a, IL-1, or IL-6 synthesis within the rat brain,
as assessed by in situ hybridization histochemistry or
semiquantitative PCR. Whether increased release of prestored TNF-a (105) or synergistic actions of TNF-a with
PG (whose levels in the brain are elevated) (256) accounts
for these observations is not known.
Surprisingly, more convincing evidence that IL-1 generated within the CNS plays a role in stimulating HPA
axis secretory activity comes from studies investigating
the neuroendocrine responses to immobilization stress
(546, 777, 778). Minami et al. (546) reported that IL-1b
mRNA expression was induced in the rat hypothalamus
within 30 min of the commencement of immobilization
stress. Interleukin-1b mRNA peaked at 60 min and was
still elevated at 120 min. These results are particularly
surprising given that these authors detected IL-1b mRNA
using Northern blot hybridization, whereas the more sensitive technique of RT-PCR has been required to reliably
demonstrate increases CNS expression of IL-1b mRNA in
response to systemic LPS. Nonetheless, this group subsequently reported increases in hypothalamic IL-1 biological
activity within 30–60 min of immobilization stress and
furthermore demonstrated that microinjection of IL-1ra
(2 mg) into the hypothalamus inhibits the stress-induced
rise in plasma ACTH concentrations by as much as 50%
(778). On the other hand, we have found no significant
effects of either IL-1ra (100 mg icv) or anti-TNF-a antiserum on the rise in plasma ACTH concentrations produced
by electrofootshock in rats (Ref. 881 and unpublished
data), whereas others (716) have found that IL-6 deficiency does not influence the HPA axis response to immobilization in mice. These data would seem to indicate that
the participation of cytokines in psychological/physical
11-25-98 11:16:36
pra
APS-Phys Rev
Downloaded from on April 23, 2014
within the periphery is paralleled by, or even initiates,
cytokine production within the CNS. As detailed in section
IVA2B, the peripheral administration of LPS has been
shown to induce cytokine and, in particular, IL-1 synthesis
within the brain. However, by far the majority of studies
which investigated cellular localization of cytokines demonstrated that IL-1 mRNA and protein are only induced
within brain parenchyma at doses (hundreds of micrograms to milligrams) well above those required to observe
physiological responses such as activation of the HPA
axis (õ1 mg/kg). Furthermore, cytokine induction within
brain parenchyma is observed only several hours after
LPS administration (compare increases in ACTH secretion at 30–45 min). Indeed, the location of IL-1 mRNA
and protein that is induced fastest and at lowest doses
appears to be in barrier-related regions such as the CVO
and perivascular sites, suggesting a site of action on the
blood side of the BBB. Therefore, cytokine synthesis
within brain parenchyma would appear not to be an absolute requirement for at least those responses that occur
rapidly after LPS administration.
Nevertheless, studies have demonstrated that inhibition of IL-1 action within the brain by intracerebral administration of anti-IL-1b antibodies can inhibit acute phase
responses to peripheral LPS (418, 709). However, such
experiments failed to demonstrate that the IL-1b inhibited
is generated within the brain and does not enter from the
periphery (see section VB) and also have not necessarily
indicated that the site of action of the injected antibodies
is within the brain. We have recently found that pretreatment of rats intracerebroventricularly with an anti-TNFa antiserum delays the onset of the rise in plasma ACTH
concentrations produced by intravenous injection of 5 mg/
kg LPS, suggesting an action of TNF-a within the brain
(889). However, intravenous administration of the same
volume of antiserum produced qualitatively and quantitatively the same effect as that observed with intracerebroventricular anti-TNF-a (889). Subsequent studies indicated that antisera rapidly dissipate to the periphery after
intracerebroventricular injection and can produce immunoneutralizing concentrations within the periphery within
only a very short time period (Ç1–4 h) (890). Experiments
using injections of antisera/antibodies or receptor antagonists to inhibit the action of cytokines within the brain
clearly need to demonstrate that effects observed cannot
be accounted for by actions of the inhibitors in the periphery, particularly given the high concentrations of cytokines that can occur in blood and peripheral tissues during
inflammation/infection.
One tightly controlled study has indicated that IL-1
may well act within the brain to stimulate HPA axis activation in response to peripheral administration of LPS (387).
Kakucska et al. (387) demonstrated that continuous intracerebroventricular infusion of human IL-1ra completely
prevents the rise in pPVN CRF mRNA observed 8 h after
Volume 79
January 1999
REGULATION OF HPA AXIS BY CYTOKINES
43
stress-induced HPA activation is probably limited to specific cytokines and specific stressors.
Overall, although there is substantial evidence that
IL-1 can be produced in the brain during CNS insults,
severe endotoxemia, and possibly also acute physical/psychological stress, there is only limited data implicating
CNS-derived IL-1 in the regulation of the associated HPA
axis secretory responses. Direct testing of the physiological role of CNS-derived cytokines in the regulation of the
HPA axis awaits the development of appropriate genetargeting strategies that selectively mutate cytokine synthesis in a region- or cell type-specific manner (875).
/ 9j0c$$oc11
P13-8
11-25-98 11:16:36
pra
APS-Phys Rev
Downloaded from on April 23, 2014
tions can be observed within 5–10 min of IL-1 administration. However, there is evidence that IL-1 administered intraperitoneally stimulates ACTH secretion in the mouse
in an IL-6-dependent fashion (584, 640). It is unfortunate,
therefore, that given the much higher incidence of high
levels of IL-6 rather than IL-1 during pathophysiological
states, there is a relative paucity of information regarding
the mechanisms of activation of the HPA axis in response
to exogenous IL-6 than there is in response to exogenous
IL-1 (see sects. VA– VH). At least part of the reason why
this is the case is because of the relative impotency of
IL-6 compared with IL-1 when these cytokines have been
administered to animals (66, 527, 909, 1003). Whether this
reflects a true difference in the potencies of the two cytoI. Local Interleukin-1 Induction of Circulating
kines to interact with components of the HPA axis, or
Interleukin-6
reflects the fact that IL-1 also induces prolonged high circulating levels of IL-6, is not presently known. Nevertheless, it
Finally, it seems pertinent to consider the possibility is clearly important to determine whether the mechanisms
that IL-1 induced by peripheral infection/inflammation can described above for IL-1 are also relevant for, or are in fact
activate the HPA axis by a mechanism that does not in- mediated by, IL-6. Such future studies would clearly benefit
volve a ‘‘direct’’ interaction of IL-1 with any of the above from the use of infusions of IL-6 to mimic the levels of
mechanisms. There is substantial evidence that the effect IL-6 actually observed during pathological conditions.
of IL-1 on the HPA axis during peripheral infection/inflammation is mediated via the induction of high circulating levels of IL-6 and that it is IL-6 that actually interacts VI. CONCLUSIONS
with components of the HPA axis. Indeed, IL-1 potently
stimulates the synthesis and secretion of IL-6 (e.g., Refs.
The landmark discoveries of Besedovsky’s and Bla470, 585, 766, 871). Studies of the HPA axis response to lock’s groups culminated with their demonstrations that
viral infection (MCMV; see sect. IIIA3) clearly indicate IL-1 is an important endogenous regulator of the HPA
that systemic administration of IL-1ra inhibits the rise in axis. Since that time, a tremendous growth in the study
plasma corticosterone levels (716). Because IL-1b was of cytokine biology has provided several major advanceundetectable in the general circulation, plasma IL-1a con- ments in the understanding of immune-neuroendocrine
centrations were not elevated during MCMV, and the interactions. First, we now know that, in addition to ILmarked rise in plasma IL-6 levels was reduced by ú75% 1, numerous other cytokines are capable of influencing
by treatment with IL-1ra, the authors concluded that IL-6 HPA secretory axis activity, with most having a stimulais a circulating mediator of IL-1 actions (716). Indeed, they tory action. Cytokine receptors have been cloned, characshowed that IL-6-deficient mice also display a markedly terized, and localized to many neuroendocrine (among
reduced corticosterone response to MCMV (716). Simi- other) tissues. Furthermore, it is now recognized that the
larly, turpentine-induced local inflammation induces ele- HPA axis exposure to cytokines is not restricted to those
vated levels of IL-1b locally but not systemically, IL-1 is carried within the vascular supply, since the CNS, pituresponsible for inducing high circulating levels of IL-6 in itary, and adrenal are all capable of synthesizing a variety
plasma, and IL-6-deficient mice display a markedly re- of cytokines, whose levels are increased during endotoxeduced HPA axis response (see sect. IIIC). Such studies mia. Furthermore, neural afferents, such as those carried
strongly suggest that circulating IL-6 may mediate the ef- within the vagus nerve, may be a target of cytokine action
fects on the HPA axis of locally increased levels of IL-1. that conveys information from inflammatory sites to the
Indeed, IL-6 has been suggested to mediate a number of brain. Finally, recent studies suggesting that cytokine regother IL-1 actions, including inhibition of proteoglycan ulation of the HPA axis may occur not only during infecsynthesis in cartilage, induction of thymocyte prolifera- tion, inflammation, and trauma, but also during periods
tion, and the production of fever (97, 150, 333, 585, 602). of psychological and/or physical stress unrelated to the
Therefore, there is substantial evidence that during patho- presence of tissue disease or damage, have given a new
physiological circumstances IL-6 may well be a major me- perspective on the nature of immune-neuroendocrine indiator of IL-1 action.
teractions.
It seems unlikely that intravenous administration of
Clearly, the pituitary and adrenal glands represent
exogenous IL-1 results in an IL-6-dependent activation of potential targets of cytokine action on the HPA axis when
the HPA axis, since elevations of plasma ACTH concentra- these organs are exposed to prolonged elevated cytokine
44
ANDREW V. TURNBULL AND CATHERINE L. RIVIER
We thank Dr. Steve Hopkins for helpful discussions during
the preparation of this manuscript.
Research in the authors’ laboratories was supported by
National Institutes of Health Grants DK-26741 and MH-51774,
the Foundation for Research Inc., Aaron and Amoco Fellowships, and the British Medical Research Council.
2.
3.
4.
5.
6.
7.
8.
9.
10.
11.
12.
13.
14.
15.
16.
17.
18.
REFERENCES
1. ABRAHAM, E. J., AND J. E. MINTON. Cytokines in the hypophysis:
a comparative look at interleukin-6 in the porcine anterior pitu-
/ 9j0c$$oc11
P13-8
19.
11-25-98 11:16:36
itary gland. Comp. Biochem. Physiol. A Physiol. 116: 203–207,
1997.
ADASHI, E. Y. The potential role of IL-1 in the ovulatory process:
an evolving hypothesis. J. Reprod. Immunol. 35: 1–9, 1997.
ADDISON, T. On the Constitutional and Local Effects of the Suprarenal Capsules. London: Highley, 1855.
AGUADO, F., J. RODRIGO, L. CACICEDO, AND B. MELLSTROM.
Distribution of insulin like growth factor-I receptor mRNA in rat
brain. Regulation in the hypothalamo-neurohypophysial system.
J. Mol. Endocrinol. 11: 231–239, 1993.
AKITA, S., P. M. CONN, AND S. MELMED. Leukemia inhibitory
factor (LIF) induces acute adrenocorticotrophic hormone (ACTH)
secretion in fetal rhesus macaque primates: a novel dynamic test
of pituitary function. J. Clin. Endocrinol. Metab. 81: 4170–4173,
1996.
AKITA, S., J. MALKIN, AND S. MELMED. Disrupted murine leukemia inhibitory factor (LIF) gene attenuates adrenocorticotropic
hormone (ACTH) secretion. Endocrinology 137: 3140–3143, 1996.
AKITA, S., J. WEBSTER, S. G. REN, H. TAKINO, J. SAID, O. ZAND,
AND S. MELMED. Human and murine pituitary expression of leukemia inhibitory factor. Novel intrapituitary regulation of adrenocorticotropin hormone synthesis and secretion. J. Clin. Invest. 95:
1288–1298, 1995.
AKIYAMA, H., T. NISHIMURA, H. KONDO, K. IKEDA, Y. HAYASHI, AND P. L. MCGEER. Expression of the receptor for macrophage colony stimulating factor by brain microglia and its upreguation in brains of patients with Alzheimer’s disease and amyotropic lateral sclerosis. Brain Res. 639: 171–174, 1994.
AL-DAMLUGI, S. Adrenergic mechanisms in the control of corticotrophin secretion. J. Endocrinol. 119: 5–14, 1988.
ALHEIM, K., Z. CHAI, G. FANTUZZI, H. HASANVAN, D. MALINOWSKY, E. DI SANTO, P. GHEZZI, C. A. DINARELLO, AND T.
BARTFAI. Hyperresponsive febrile reactions to interleukin (IL)
1a and IL-1b and altered brain cytokine mRNA and serum cytokine levels, in IL-1b-deficient mice. Proc. Natl. Acad. Sci. USA 94:
2681–2686, 1997.
ALLAERTS, W., P. H. M. JEUCEN, R. DEBETS, S. HOEFAKKER,
AND E. CLAASSEN. Heterogeneity of pituitary folliculo-stellate
cells: implications for interleukin-6 production and accessory
function in vitro. J. Neuroendocrinol. 9: 43–53, 1997.
ALOISI, F., A. CARE, G. BORSELLINO, P. GALLO, S. ROSA, A.
BASSANI, A. CABIBBO, U. TESTA, G. LEVI, AND C. PESCHLE.
Production of hemolymphopoietic cytokines (IL-6, IL-8, colony
stimulating factors) by normal human astrocytes in response to
IL-1b and tumor necrosis factor-a. J. Immunol. 149: 2358–2366,
1992.
AMANO, O., Y. YOSHITAKE, K. NISHIKAWA, AND S. ISEKI. Immunocytochemical localization of basic fibroblast growth factor in
the rat pituitary gland. Arch. Histol. Cytol. 56: 269–276, 1993.
ANDREIS, P. G., G. NERI, A. S. BELLONI, G. MAZZAOCHI, A.
KASPRZAK, AND G. NUSSDORFER. Interleukin-1b enhances corticosterone secretion by acting directly on the rat adrenal gland.
Endocrinology 129: 53–57, 1991.
ANTONI, F. A., G. FINK, AND W. J. SHERWOOD. Corticotropinreleasing peptides in rat hypophysial portal blood after paraventricular nucleus lesions: a marked reduction in the concentration
of corticotropin-releasing factor-41, but no change in vasopressin.
J. Endocrinol. 125: 175–183, 1990.
APPEL, K., M. BUTTINI, A. SAUTER, AND P. J. GEBICKEHAERTER. Cloning of rat interleukin-3 receptor beta-subunit from
cultured microglia and its mRNA expression in vivo. J. Neurosci.
15: 5800–5809, 1995.
ARANGUEZ, I., C. TORRES, AND N. RUBIO. The receptor for tumor necrosis factor on murine astrocytes: characterization, intracellular degradation, and regulation by cytokines and Theiler’s
murine encephalomyelitis virus. Glia 13: 185–194, 1995.
ARAUJO, D. M., P. A. LAPCHAK, B. COLLIER, AND R. QUIRION.
Localization of interleukin-2 immunoreactivty and interleukin-2
receptors in the rat brain: interaction with the cholinergic system.
Brain Res. 498: 257–266, 1989.
AREVALO, R., F. SANCHEZ, J. R. ALONSO, J. CARRETERO, R.
VAZQUEZ, AND J. AIJON. NADPH-diaphorase activity in the hypo-
pra
APS-Phys Rev
Downloaded from on April 23, 2014
levels. However, the majority of evidence indicates that
either direct or indirect stimulation of hypothalamic CRF
secretion is the primary means by which cytokines (at
least IL-1, IL-6, and TNF-a) activate the HPA axis. The
neuroanatomic and neuropharmacological pathways by
which IL-1 has been proposed to influence the neuroendocrine hypothalamus are numerous and diverse. Accurate
models of the mechanisms by which IL-1 activates the
HPA axis have to take into account the consistently reported inhibitory effects of either inhibitors of PG synthesis or disruption of catecholaminergic input into the hypothalamus.
In the situation where IL-1 levels are elevated endogeneously (rather than by exogenous administration), the
mechanism by which IL-1 activates the HPA axis is determined largely by the anatomic location of the tissues and
bodily fluids that possess markedly increased IL-1 concentrations. In this respect, distinct proposed mechanisms
seem most appropriate to the situations where IL-1 is
increased in blood, peripheral tissue, or the brain. In reality, however, the extent to which elevations in cytokine
concentrations can be compartmentalized on an anatomic
basis is unclear. For example, it might be assumed that
during a local inflammatory insult when IL-1 levels are
elevated only at the local site of tissue damage, a neural
afferent mechanism seems the most likely candidate pathway for activation of the HPA axis. However, because IL1 induces the secretion of IL-6, which subsequently gains
access to the systemic circulation, it seems likely that IL6 is, at least partly, responsible for activation of the HPA
axis. An additional complication is that none of the proposed mechanisms of activation of the HPA axis by cytokines takes into account the fact that elaboration of a
single cytokine in response to a homeostatic threat is
an unlikely event. Furthermore, there are no systematic
studies of the mechanisms by which multiple cytokines
(e.g., IL-1 plus IL-6) may induce HPA axis activation. Perhaps the most striking feature of the numerous studies
investigating the mechanisms of IL-1 induced activation
of the HPA axis is the tremendous diversity of plausible
immune-neuroendocrine interactions. This diversity ensures that the occurrence of challenges to cellular, tissue,
or system homeostasis is reliably conveyed to the neuroendocrine hypothalamus.
Volume 79
January 1999
20.
21.
22.
23.
24.
25.
26.
27.
29.
30.
31.
32.
33.
34.
35.
36.
37.
38.
39.
thalamic magnocellular neurosecretory nuclei of the rat. Brain
Res. Bull. 28: 599–603, 1992.
ARZT, E., R. BURIC, G. STELZER, J. STALLA, J. SAUER, U. RENNER, AND G. K. STALLA. Interleukin involvement in anterior pituitary cell growth regulation: effects of IL-2 and IL-6. Endocrinology
132: 459–467, 1993.
ARZT, E., AND G. K. STALLA. Cytokines: autocrine and paracrine
roles in the anterior pituitary. Neuroimmunomodulation 3: 28–
34, 1996.
ARZT, E., G. STEIZER, U. RENNER, M. LANGE, O. A. MULLER,
AND G. K. STALLA. Interleukin-2 and interleukin-2 receptor expression in human corticotrophic adenoma and murine pituitary
cell cultures. J. Clin. Invest. 90: 1944–1951, 1992.
ATKINS, E. Pathogenesis of fever. Physiol. Rev. 40: 580–646, 1960.
ATKINS, M. B., J. A. GOULD, M. ALLEGRETTA, J. J. LI, R. A.
DEMPSEY, R. A. RUDDERS, D. R. PARKINSON, S. REICHLIN,
AND J. W. MIER. Phase I evaluation of recombinant interleukin-2
in patients with advanced malignant disease. J. Clin. Oncol. 4:
1380–1391, 1986.
AUBRY, J.-M., A. V. TURNBULL, G. P. POZZOLI, C. RIVIER, AND
W. VALE. Endotoxin and interleukin-1b decrease corticotropinreleasing factor receptor type I mRNA levels in the rat pituitary.
Endocrinology 138: 1621–1626, 1997.
BAES, M., W. ALLAERTS, AND C. DENEF. Evidence for functional
communication between folliculo-stellate cells and hormone-secreting cells in perifused anterior pituitary cell aggregates. Endocrinology 120: 685–691, 1987.
BALLMER, P. E., M. A. MCNURLAN, B. G. SOUTHORN, I. GRANT,
AND P. J. GARLICK. Effects of human interleukin-1 beta on protein
synthesis in rat tissues compared with a classical acute-phase
response reaction induced by turpentine. Rapid response of muscle to interleukin-1 beta. Biochem. J. 279: 683–688, 1991.
BAN, E. M., F. HAOUR, AND R. LENSTRA. Brain interleukin-1 gene
expression induced by peripheral lipopolysaccharide administration. Cytokine 4: 48–54, 1992.
BAN, E., C. MARQUETTE, A. SARRIEAU, F. FITZPATRICK, G.
FILLION, G. MILON, W. ROSTENE, AND F. HAOUR. Regulation
of interleukin-1 receptor expression in mouse brain and pituitary
by lipopolysaccharide and glucocorticoids. Neuroendocrinology
58: 581–587, 1993.
BAN, E. M., G. MILON, N. PRUDHOMME, G. FILLION, AND F.
HAOUR. Receptors for interleukin-1 (a and b) in mouse brain:
mapping and neuronal localization in hippocampus. Neuroscience
43: 21–30, 1991.
BAN, E., L. SARLIEVE, AND F. HAOUR. Interleukin-1 binding sites
on astrocytes. Neuroscience 52: 725–733, 1993.
BANDTLOW, C. E., M. MEYER, D. LINDHOLM, M. SPRANGER,
R. HEUMANN, AND H. THOENEN. Regional and cellular codistribution of interleukin 1b and nerve growth factor mRNA in the
adult rat brain: possible relationship to the regulation of nerve
growth factor synthesis. J. Cell Biol. 111: 1701–1711, 1990.
BANKS, W. A., AND A. J. KASTIN. The interleukin-1a, -1b, and
-2 do not acutely disrupt the murine blood-brain barrier. Int. J.
Immunopharmacol. 14: 629–636, 1992.
BANKS, W. A., A. J. KASTIN, AND R. D. BROADWELL. Passage of
cytokines across the blood-brain barrier. Neuroimmunomodulation 2: 241–248, 1995.
BANKS, W. A., A. J. KASTIN, AND D. A. DURHAM. Bidirectional
transport of interleukin-1 alpha across the blood-brain-barrier.
Brain Res. Bull. 23: 433–437, 1989.
BANKS, W. A., A. J. KASTIN, AND E. G. GUTIERREZ. Penetration
of interleukin-6 across the murine blood-brain-barrier. Neurosci.
Lett. 179: 53–56, 1994.
BANKS, W. A., L. ORTIZ, S. R. PLOTKIN, AND A. J. KASTIN. Human interleukin (IL) 1a, murine IL-1a and murine IL-1b are transported from blood to brain in the mouse by a shared saturable
transport mechanism. J. Pharmacol. Exp. Ther. 259: 988–996,
1991.
BARBACID, M. The Trk family of neurotrophin receptors. J. Neurobiol. 25: 1386–1403, 1994.
BARBANEL, G., S. GAILLET, M. MEKAOUCHE, L. GIVALOIS, G.
IXART, P. SIAUD, A. SZAFARCZYK, F. MALAVAL, AND I. ASSENMACHER. Complex catecholaminergic modulation of the stimula-
/ 9j0c$$oc11
P13-8
40.
41.
42.
43.
44.
45.
46.
47.
48.
49.
50.
51.
52.
53.
54.
55.
56.
11-25-98 11:16:36
45
tory effect of interleukin-1b on the corticotropic axis. Brain Res.
626: 31–36, 1993.
BARBANEL, G., G. IXART, A. SZAFARCZYK, F. MALAVAL, AND
I. ASSENMACHER. Intrahypothalamic infusion of interleukin-1b
increases the release of corticotropin-releasing hormone (CRH
41) and adrenocorticotropic hormone (ACTH) in free-moving rats
bearing a push-pull cannula in the median eminence. Brain Res.
516: 31–36, 1990.
BARBARINO, A., S. COLASANTI, S. M. CORSELLO, M. A. SATTA,
S. D. CASA, C. A. ROTA, R. TARTAGLIONE, AND A. BAINI. Dexamethasone inhibition of interferon-g2-induced stimulation of cortisol and growth hormone secretion in chronic myeloproliferative
syndrome. J. Clin. Endocrinol. Metab. 80: 1329–1332, 1995.
BARTFAI, T., C. ANDERSSON, J. BRISTULF, M. SCHULTZBERG,
AND S. SVENSON. Interleukin-1 in the noradrenergic chromaffin
cells in the rat adrenal medulla. Ann. NY Acad. Sci. 207–213,
1990.
BARTLETT, W. P., X. S. LI, M. WILLIAMS, AND S. BENKOVIC.
Localization of insulin-like growth factor-1 mRNA in murine central nervous system during postnatal development. Dev. Biol. 147:
239–250, 1991.
BASILE, D. P., AND M. A. HOLZWARTH. Basic fibroblast growth
factor may mediate proliferation in the compensatory adrenal
growth response. Am. J. Physiol. 265 (Regulatory Integrative
Comp. Physiol. 34): R1253–R1261, 1993.
BATSHAKE, B., C. NILSSON, AND J. SUNDELIN. Molecular characterization of the mouse prostanoid EP1 receptor gene. Eur. J.
Biochem. 231: 809–814, 1995.
BAZAN, J. F., J. C. TIMANS, AND R. A. KASTELEIN. A newly identified interleukin-1? Nature 379: 591, 1996.
BAZZONI, F., AND B. BEUTLER. The tumor necrosis factor ligand
and receptor families. N. Engl. J. Med. 334: 1717–1725, 1996.
BEACH, J. E., R. C. SMALLRIDGE, C. A. KINZER, E. W. BERNTON, J. W. HOLADAY, AND H. G. FEIN. Rapid release of multiple
hormones from rat anterior pituitaries perifused with recombinant
interleukin-1. Life Sci. 44: 1–7, 1989.
BEBO, B. F., AND D. S. LINTHICUM. Expression of mRNA for 55kDa and 75-kDa tumor necrosis factor (TNF) receptors in mouse
cerebrovascular endothelium: effects of interleukin-1 beta, interferon-gamma and TNF-alpha on cultured cells. J. Neuroimmunol.
62: 161–167, 1995.
BEN HUR, T., J. ROSENTHAL, A. ITZIK, AND J. WEIDENFELD.
Adrenocortical activation by herpes virus: involvement of IL-1
beta and central noradrenergic system. Neuroreport 7: 927–931,
1996.
BENIGNI, F., R. FAGGIONI, M. SIRONI, G. FANTUZZI, P. VANDENABEELE, N. TAKAHASHI, S. SACCO, W. FIERS, W. A. BUURMAN, AND P. GHEZZI. TNF receptor p55 plays a major role in
centrally mediated increases of serum IL-6 and corticosterone
after intracerebroventricular injection of TNF. J. Immunol. 157:
5563–5568, 1996.
BENIGNI, F., G. FANTUZZI, S. SACCO, M. SIRONI, P. POZZI, C. A.
DINARELLO, J. D. SIPE, V. POLI, M. CAPPELLETTI, G. PAONESSA, D. PENNICA, N. PANAYOTATOS, AND P. GHEZZI. Six different cytokines that share gp130 as a receptor subunit, induce serum
amyloid A and potentiate the induction of interleukin-6 and the
activation of the hypothalamo-pituitary-adrenal axis by interleukin-1. Blood 87: 1851–1854, 1996.
BERGERS, G., A. REIKERSTORFER, S. BRASELMANN, P. GRANINGER, AND M. BUSSLINGER. Alternative promoter usage of the
Fos-responsive gene Fit-1 generates mRNA isoforms coding for
either secreted or membrane-bound proteins related to the IL-1
receptor. EMBO J. 13: 1176–1188, 1994.
BERKENBOSCH, F., D. E. C. DEGOEIJ, A. DEL REY, AND H. O.
BESEDOVSKY. Neuroendocrine, sympathetic and metabolic responses induced by interleukin-1. Neuroendocrinology 50: 570–
576, 1989.
BERKENBOSCH, F., J. VAN OERS, A. DEL REY, F. TILDERS,
AND H. BESEDOVSKY. Corticotropin-releasing factor-producing
neurons in the rat activated by interleukin-1. Science 238: 524–
526, 1987.
BERNARDINI, R., A. E. CALOGERO, G. MAUCERI, AND G. P.
CHROUSOS. Rat hypothalamic corticotropin-releasing hormone
pra
APS-Phys Rev
Downloaded from on April 23, 2014
28.
REGULATION OF HPA AXIS BY CYTOKINES
46
57.
58.
59.
60.
61.
62.
63.
65.
66.
67.
68.
69.
70.
71.
72.
73.
74.
75.
76.
secretion in vitro is stimulated by interleukin-1 in an eicosanoid
dependent manner. Life Sci. 47: 1601–1607, 1990.
BERNARDINI, R., A. CHIARENZA, A. E. CALOGERO, P. W.
GOLD, AND G. P. CHROUSOS. Arachidonic acid metabolites modulate rat hypothalamic corticotropin-releasing hormone secretion
in vitro. Neuroendocrinology 50: 708–715, 1989.
BERNARDINI, R., T. C. KAMILARIS, A. E. CALOGERO, E. O.
JOHNSON, M. T. GOMEZ, P. W. GOLD, AND G. P. CHROUSOS.
Interactions between tumor necrosis factor-a, hypothalamic corticotropin-releasing hormone, and adrenocorticotropin secretion in
the rat. Endocrinology 126: 2876–2881, 1990.
BERNARDINI, R., G. MAUCERI, M. P. IURATO, A. CHIARENZA,
L. LEMPEREUR, AND U. SCAPAGNINI. Response of the hypothalamic-pituitary-adrenal axis to interleukin 1 in the aging rat. Prog.
Neuroendocrinol. Immunol. 5: 166–171, 1992.
BERNHAGEN, J., T. CALANDRA, R. A. MITCHELL, S. B. MARTIN,
K. J. TRACEY, W. VOELTER, K. R. MANOGUE, A. CERAMI, AND
R. BUCALA. Macrohpage migration inhibitory factor (MIF) is a
pituitary-derived cytokine that potentiates lethal endotoxemia.
Nature 365: 756–759, 1993.
BERNHAGEN, J., R. A. MITCHELL, T. CALANDRA, W. VOELTER,
A. CERAMI, AND R. BUCALA. Purification, bioactivity and secondary structure analysis of mouse and human macrophage migration
inhibitory factor (MIF). Biochemistry 33: 14144–14155, 1994.
BERNTON, E. W., J. E. BEACH, J. W. HOLADAY, R. C.
SMALLRIDGE, AND H. G. FEIN. Release of multiple hormones by
a direct action of interleukin-1 on pituitary cells. Science 238: 519–
521, 1987.
BESEDOVSKY, H., AND A. DEL REY. Neuroendocrine and metabolic responses induced by interleukin-1. J. Neurosci. Res. 18:
172–178, 1987.
BESEDOVSKY, H. O., AND A. DEL REY. Mechanism of virus-induced stimulation of the hypothalamus-pituitary-adrenal axis. J.
Steroid Biochem. Mol. Biol. 34: 235–239, 1989.
BESEDOVSKY, H. O., AND A. DEL REY. Immune-neuroendocrine
interactions: facts and hypotheses. Endocr. Rev. 17: 64–102, 1996.
BESEDOVSKY, H. O., A. DEL REY, I. KLUSMAN, H. FURUKAWA,
G. M. ARDITI, AND A. KABIERSCH. Cytokines as modulators of
the hypothalamus-pituitary-adrenal axis. J. Steroid Biochem. Mol.
Biol. 40: 613–618, 1991.
BESEDOVSKY, H. O., A. DEL REY, AND E. SORKIN. Antigenic
competition between horse and sheep red blood cells as hormonedependent phenomenon. Clin. Exp. Immunol. 37: 106–113, 1979.
BESEDOVSKY, H. O., A. DEL REY, AND E. SORKIN. Lymphokinecontaining supernatants from Con A-stimulated cells increase corticosterone blood levels. J. Immunol. 126: 385–387, 1981.
BESEDOVSKY, H., A. DEL REY, E. SORKIN, M. DA PRADA, R.
BURRI, AND C. G. HONEGGER. The immune response evokes
changes in brain noradrenergic neurons. Science 221: 564–566,
1983.
BESEDOVSKY, H., A. DEL REY, E. SORKIN, AND C. A. DINARELLO. Immunoregulatory feedback between interleukin-1 and glucocorticoid hormones. Science 233: 652–654, 1986.
BESEDOVSKY, H. O., A. DEL REY, E. SORKIN, W. LOTZ, AND
U. SCHWULERA. Lymphoid cells produce an immunoregulatory
glucocorticoid increasing factor (GIF) acting through the pituitary
gland. Clin. Exp. Immunol. 59: 622–628, 1985.
BESEDOVSKY, H., E. SORKIN, D. FELIX, AND H. HAAS. Hypothalamic changes during the immune response. Eur. J. Immunol. 7:
323–325, 1977.
BESEDOVSKY, H., E. SORKIN, M. KELLER, AND J. MULLER.
Changes in blood hormone levels during the immune response.
Proc. Soc. Exp. Biol. Med. 150: 466–470, 1975.
BILEZIKJIAN, L. M., A. L. BLOUNT, C. A. CAMPEN, C. GONZALEZ-MANCHON, AND W. VALE. Activin-A inhibits proopiomelanocortin messenger RNA accumulation and adrenocorticotropin secretion of AtT20 cells. Mol. Endocrinol. 5: 1389–1395, 1991.
BILEZIKJIAN, L. M., A. Z. CORRIGAN, AND W. W. VALE. ActivinB, inhibin-B and follistatin as autocrine/paracrine factors of the
rat anterior pituitary. Front. Neuroendocrinol. 3: 1–19, 1994.
BINDONI, M., V. PERCIAVALLE, S. BERRETTA, N. BELLUARDO,
AND T. DIAMANTSTEIN. Interleukin 2 modifies the bioelectric
/ 9j0c$$oc11
P13-8
77.
78.
79.
80.
81.
82.
83.
84.
85.
86.
87.
88.
89.
90.
91.
92.
93.
94.
95.
96.
97.
98.
11-25-98 11:16:36
Volume 79
activity of some neurosecretory nuclei in the rat hypothalamus.
Brain Res. 462: 10–14, 1988.
BISHAI, I., AND F. COCEANI. Differential effects of endotoxin and
cytokines on prostaglandin E2 formation in cerebral microvessels
and brain parenchyma: implications for the pathogenesis of fever.
Cytokine 8: 371–376, 1996.
BISHAI, I., C. DINARELLO, AND F. COCEANI. Prostaglandin formation in feline cerebral microvessels: effects of endotoxin and
interleukin-1. Can. J. Physiol. Pharmacol. 65: 2225–2230, 1987.
BLALOCK, J. E. Proopiomelanocortin-derived peptides in the immune system. Clin. Endocrinol. 22: 823–827, 1985.
BLALOCK, J. E. A molecular basis for bidirectional communication between the immune and neuroendocrine systems. Physiol.
Rev. 69: 1–32, 1989.
BLALOCK, J. E. The syntax of immune-neuroendocrine communication. Immunol. Today 15: 504–511, 1994.
BLALOCK, J. E., D. HARBUR-MCMENAMIN, AND E. M. SMITH.
Peptide hormones shared by the neuroendocrine and immunologic systems. J. Immunol. 135, Suppl.: 858s–861s, 1985.
BLALOCK, J. E., H. M. JOHNSON, E. M. SMITH, AND B. A. TORRES. Enhancement of the in vitro antibody response by thyrotropin. Biochem. Biophys. Res. Commun. 125: 30–34, 1984.
BLALOCK, J. E., AND J. D. STANTON. Common pathways of interferon and hormonal action. Nature 283: 406–408, 1980.
BLATTEIS, C. M. Neuromodulative actions of cytokines. Yale J.
Biol. Med. 63: 133–146, 1990.
BLATTEIS, C. M. The OVLT: the interface between the brain and
circulating pyrogens. In: Neuroimmunology of Fever, edited by
T. Bartfai and D. Ottoson. Oxford, UK: Pergamon, 1992, p. 167–
176.
BLUETHMANN, H., J. ROTHE, N. SCHULTZE, M. TKACHUK, AND
P. KOEBEL. Establishment of the role of IL-6 and TNF receptor
1 using gene knockout mice. J. Leukoc. Biol. 56: 565–570, 1994.
BLUTHE, R.-M., B. MICHAUD, K. W. KELLEY, AND R. DANTZER.
Vagotomy attenuates behavioral effects of interleukin-1 injected
peripherally but not centrally. Neuroreport 7: 1485–1488, 1996.
BLUTHE, R. M., B. MICHAUD, K. W. KELLEY, AND R. DANTZER.
Vagotomy blocks behavioural effects of interleukin-1 injected via
the intraperitoneal route but not via other systemic routes. Neuroreport 7: 2823–2827, 1996.
BLUTHE, R. M., V. WALTER, P. PARNET, S. LAYE, J. LESTAGE,
D. VERRIER, S. POOLE, B. E. STENNING, K. W. KELLEY, AND R.
DANTZER. Lipopolysaccharide induces sickness behavior in rats
by a vagal mediated mechanism. C. R. Acad. Sci. III 317: 499–
503, 1994.
BOJE, K. M. Cerebrovascular permeability changes during experimental meningitis in the rat. J. Pharmacol. Exp. Ther. 274: 1199–
1203, 1995.
BOKA, G., P. ANGLADE, D. WALLACH, F. JAVOY-AGID, Y. AGID,
AND E. C. HIRSCH. Immunocytochemical analysis of tumor necrosis factor and its receptors in Parkinson’s disease. Neurosci. Lett.
172: 151–154, 1995.
BONDY, C., H. WERNER, C. T. ROBERTS, AND D. LEROITH. Cellular pattern of type-I insulin-like growth factor receptor gene expression during maturation of the rat brain: comparison with insulin-like growth factors I and II. Neuroscience 46: 909–923, 1992.
BONNERT, T. P., K. E. GARKA, P. PARNET, G. SONODA, J. R.
TESTA, AND J. E. SIMS. The cloning and characterization of human
MyD88: a member of an IL-1 receptor related family. FEBS Lett.
402: 81–84, 1997.
BOUSQUET, C., D. W. RAY, AND S. MELMED. A common proopiomelanocortin-binding element mediates leukemia inhibitory
factor and corticotropin-releasing hormone transcriptional synergy. J. Biol. Chem. 272: 10551–10557, 1997.
BOYLE, M., G. YAMAMOTO, M. CHEN, J. RIVIER, AND W. VALE.
Interleukin 1 prevents loss of corticotropic responsiveness to
beta-adrenergic stimulation in vitro. Proc. Natl. Acad. Sci. USA
85: 5556–5560, 1988.
BRACH, M. A., B. LOWENBERG, L. MONTOVANI, U. SCHWULERA, R. MERTELSMANN, AND F. HERRMAN. Interleukin-6 (IL-6)
is an intermediate in IL-1-induced proliferation of leukemic human
megakaryocytes. Blood 76: 1972–1979, 1990.
BRADY, L. S., A. B. LYNN, M. HERKENHAM, AND Z. GOTTES-
pra
APS-Phys Rev
Downloaded from on April 23, 2014
64.
ANDREW V. TURNBULL AND CATHERINE L. RIVIER
January 1999
99.
100.
101.
102.
103.
104.
105.
106.
108.
109.
110.
111.
112.
113.
114.
115.
116.
117.
FELD. Systemic interleukin-1 induces early and late patterns of
c-fos mRNA expression in brain. J. Neurosci. 14: 4951–4964, 1994.
BRANDT, E. R., I. R. MACKAY, P. J. HERTZOG, B. F. CHEETHAM,
M. SHERRITT, AND C. C. BERNARD. Molecular detection of interferon-alpha expression in multiple sclerosis brain. J. Neuroimmunol. 44: 1–5, 1993.
BREDER, C. D., D. DEWITT, AND R. P. KRAIG. Characterization
of inducible cyclooxygenase in rat brain. J. Comp. Neurol. 355:
296–315, 1995.
BREDER, C. D., C. A. DINARELLO, AND C. B. SAPER. Interleukin1 immunoreactive innervation of the human hypothalamus. Science 240: 321–323, 1988.
BREDER, C. D., C. HAZUKA, T. GHAYUR, C. KLUG, M. HUGININ,
K. YASUDA, M. TENG, AND C. B. SAPER. Regional distribution of
tumor necrosis factor a expression in the mouse brain after systemic lipopolysaccharide administration. Proc. Natl. Acad. Sci.
USA 91: 11393–11397, 1994.
BREDER, C. D., AND C. B. SAPER. Expression of inducible
cyclooxygenase mRNA in the mouse brain after systemic administration of bacterial lipopolysaccharide. Brain Res. 713: 64–69,
1996.
BREDER, C. D., W. L. SMITH, A. RAZ, J. MASFERRER, K. SEIBERT, P. NEEDLEMAN, AND C. B. SAPER. Distribution and characterization of cyclooxygenase immunoreactivity in the ovine brain.
J. Comp. Neurol. 322: 409–438, 1992.
BREDER, C. D., M. TSUJIMOTO, Y. TERANO, D. W. SCOTT, AND
C. B. SAPER. Distribution and characterization of tumor necrosis
factor-a-like immunoreactivity in the murine central nervous system. J. Comp. Neurol. 337: 543–567, 1993.
BREDT, D. S., G. E. GLATT, P. M. HWANG, M. FOTUHI, T. M.
DAWSON, AND S. H. SNYDER. Nitric oxide synthase protein and
mRNA are discretely localized in neuronal populations of the
mammalian CNS together with NADPH diaphorase. Neuron 7:
615–624, 1991.
BREDT, D. S., P. M. HWANG, AND S. H. SNYDER. Localization of
nitric oxide synthase indicating a neural role for nitric oxide.
Nature 347: 768–770, 1990.
BRET-DIBAT, J.-L., R.-M. BLUTHE, S. KENT, K. W. KELLEY, AND
R. DANTZER. Lipopolysaccharide and interleukin-1 depress foodmotivated behavior in mice by a vagal-mediated mechanism.
Brain Behav. Immun. 9: 242–246, 1995.
BRET-DIBAT, J. L., C. CREMINON, J. Y. COURAUD, K. W. KELLEY, R. DANTZER, AND S. KENT. Systemic capsaicin pretreatment
fails to block the decrease in food-motivated behavior induced by
lipopolysaccharide and interleukin-1 beta. Brain Res. Bull. 42:
443–449, 1997.
BRISTULF, J., AND T. BARTFAI. Interleukin-1b and tumor necrosis
factor-a stimulate the mRNA expression of interleukin-1 receptors
in mouse anterior pituitary AtT-20 cells. Neurosci. Lett. 187: 53–
56, 1995.
BRISTULF, J., A. SIMONCSITS, AND T. BARTFAI. Characterization
of a neuronal interleukin-1 receptor and the corresponding mRNA
in the mouse anterior pituitary cell line AtT-20. Neurosci. Lett.
128: 173–176, 1991.
BRODISH, A. Extra-CNS corticotropin-releasing factors. Ann. NY
Acad. Sci. 297: 420–435, 1977.
BRODISH, A. Tissue corticotropin releasing factors. Federation
Proc. 36: 2088–2093, 1977.
BROSH, N., D. STERNBERG, J. HONIGWACHS-SHA’ANANI, B.C. LEE, Y. SHAV-TAL, E. TZEHOVAL, L. M. SHULMAN, J. TOLEDO, Y. HACHMAN, P. CARMI, W. JIANG, J. SASSE, F. HORN,
Y. BURSTEIN, AND D. ZIPORI. The plasmacytoma growth inhibitor
restrictin-P is an antagonist of interleukin-6 and interleukin-11. J.
Biol. Chem. 270: 29594–29600, 1995.
BROWN, S. L., L. R. SMITH, AND J. E. BLALOCK. Interleukin-1 and
interleukin-2 enhance proopiomelanocortin gene expression in pituitary cells. J. Immunol. 139: 3181–3183, 1987.
BRUNETTI, L., P. PREZIOSI, E. RAGAZZONI, AND M. VACCA.
Involvement of nitric oxide in basal and interleukin-1 beta-induced
CRF and ACTH release in vitro. Life Sci. 53: 219–222, 1993.
BRUNETTI, L., P. PREZIOSI, E. RAGAZZONI, AND M. VACCA.
Effects of lipopolysaccharide on hypothalamic-pituitary-adrenal
axis in vitro. Life Sci. 54: 165–171, 1994.
/ 9j0c$$oc11
P13-8
47
118. BUCALA, R. Identification of MIF as a new pituitary hormone and
macrophage cytokine and its role in endotoxic shock. Immunol.
Lett. 43: 23–26, 1994.
119. BUCALA, R. MIF, a previously unrecognized pituitary hormone
and macrophage cytokine, is a pivotal mediator in endotoxic
shock. Circ. Shock 44: 35–39, 1994.
120. BUCALA, R. MIF rediscovered: cytokine, pituitary hormone, and
glucocorticoid-induced regulator of the immune response. FASEB
J. 10: 1607–1613, 1996.
121. BUCKINGHAM, J. C., H. D. LOXLEY, H. C. CHRISTIAN, AND J. G.
PHILIP. Activation of the HPA axis by immune insults: roles and
interactions of cytokines, eicosanoids, and glucocorticoids. Pharmacol. Biochem. Behav. 54: 285–298, 1996.
122. BURGER, D., AND J.-M. DAYER. Inhibitory cytokines and cytokine
inhibitors. Neurology 45, Suppl.: S39–S43, 1995.
123. BURRELL, R. Human responses to bacterial endotoxin. Circ.
Shock 43: 137–153, 1994.
124. BUTTINI, M., AND H. BODDEKE. Peripheral lipopolysaccharide
stimulation induces interleukin-1b messenger RNA in rat brain
microglial cells. Neuroscience 65: 523–530, 1995.
125. CAHAO, M. V. Neurotrophin receptors: a window into neuronal
differentiation. Neuron 9: 583–593, 1992.
126. CALANDRA, T., J. BERHAGEN, C. N. METZ, L. A. SPIEGEL, M.
BACHER, T. DONNELLY, A. CERAMI, AND R. BUCALA. MIF as a
glucocorticoid-induced modulator of cytokine production. Nature
377: 68–71, 1995.
127. CALKA, J., AND C. H. BLOCK. Relationship of vasopressin with
NADPH-diaphorase in the hypothalamo-neurohypophysial system.
Brain Res. Bull. 32: 207–210, 1993.
128. CALLAHAN, T. A., AND D. T. PIEKUT. Differential Fos expression
induced by IL-1 beta and IL-6 in rat hypothalamus and pituitary
gland. J. Neuroimmunol. 73: 207–211, 1997.
129. CALZA, L., L. GIARDINO, AND S. CECCATELLI. NOS mRNA in
the paraventricular nucleus of young and aged rats after immobilization stress. Neuroreport 4: 627–630, 1993.
130. CAMBRONERO, J. C., J. BORRELL, AND C. GUAZA. Glucocorticoids modulate rat hypothalamic corticotropin-releasing factor release induced by interleukin-1. J. Neurosci. Res. 24: 470–476, 1989.
131. CAMBRONERO, J. C., F. J. RIVAS, J. BORREL, AND C. GUAZA.
Interleukin-2 induces corticotropin-releasing hormone release
from superfused rat hypothalami: influence of glucocorticoids.
Endocrinology 131: 677–683, 1992.
132. CAMBRONERO, J. C., F. J. RIVAS, J. BORREL, AND C. GUAZZA.
Interleukin-1-beta induces pituitary adrenocorticotropin secretion: evidence for glucocorticoid modulation. Neuroendocrinology 55: 648–654, 1992.
133. CAMBRONERO, J. C., F. J. RIVAS, J. BORRELL, AND C. GUAZA.
Adrenalectomy does not change CRF secretion induced by interleukin-1 from rat perifused hypothalami. Regul. Peptides 41:
237–247, 1992.
134. CAMBRONERO, J. C., F. J. RIVAS, J. BORRELL, AND C. GUAZA.
Release of corticotropin-releasing factor from superfused hypothalami induced by interleukin-1 is not dependent on adrenergic
mechanism. Eur. J. Pharmacol. 219: 75–80, 1992.
135. CAMBRONERO, J. C., J. RIVAS, J. BORRELL, AND C. GUAZA. Role
of arachidonic acid metabolism on corticotropin-releasing factor
(CRF)-release induced by interleukin-1 from superfused rat hypothalami. J. Neuroimmunol. 39: 57–66, 1992.
136. CAMERON, V. A., E. NISHIMURA, L. S. MATHEWS, K. A. LEWIS,
P. E. SAWCHENKO, AND W. W. VALE. Hybridization histochemical localization of activin preceptor subtypes in rat brain, pituitary,
ovary, and testis. Endocrinology 134: 799–808, 1994.
137. CANNON, J. G., AND C. A. DINARELLO. Increased plasma interleukin-1 activity in women after ovulation. Science 227: 1247–1249,
1985.
138. CANNON, J. G., R. A. FIELDING, M. A. FIATARONE, S. F. ORENCOLE, C. A. DINARELLO, AND W. J. EVANS. Increased interleukin-1 beta in human skeletal muscle after exercise. Am. J. Physiol.
257 (Regulatory Integrative Comp. Physiol. 26): R451–R455,
1989.
139. CANNON, J. G., AND M. J. KLUGER. Endogenous pyrogen activity
in human plasma after exercise. Science 220: 617–619, 1983.
140. CAO, C., K. MATSUMURA, K. YAMAGATA, AND Y. WATANABE.
11-25-98 11:16:36
pra
APS-Phys Rev
Downloaded from on April 23, 2014
107.
REGULATION OF HPA AXIS BY CYTOKINES
48
141.
142.
143.
144.
145.
146.
147.
149.
150.
151.
152.
153.
154.
155.
156.
157.
158.
Induction by lipopolysaccharide of cyclooxygenase-2 mRNA in rat
brain: its possible role in the febrile response. Brain Res. 697:
187–196, 1995.
CAO, C., K. MATSUMURA, K. YAMAGATA, AND Y. WATANABE.
Endothelial cells of the rat brain vasculature express cyclooxygenase-2 mRNA in response to systemic interleukin-1 beta: a possible
site of prostaglandin synthesis responsible for fever. Brain Res.
733: 263–272, 1996.
CAO, C., K. MATSUMURA, K. YAMAGATA, AND Y. WATANABE.
Involvememnt of cyclooxygenase-2 in LPS-induced fever and regulation of its mRNA by LPS in the rat brain. Am. J. Physiol. 272
(Regulatory Integrative Comp. Physiol. 41): R1712–R1725, 1997.
CARDOSO, E., E. ARZT, M. COUMROGLON, E. C. ANDRADA,
AND J. A. ANDRADA. Alpha-interferon induces cortisol release by
human adrenals in vitro. Int. Arch. Allergy Appl. Immunol. 93:
263–266, 1990.
CARLSON, D. E. Adrenocorticotropin correlates strongly with endotoxemia after intravenous but not after intraperitoneal inoculations of E. coli. Shock 7: 65–69, 1997.
CARMELIET, P., H. VANKELECOM, J. VAN DAMME, A. BILLIAU,
AND C. DENEF. Release of interleukin-6 from anterior pituitary
cell aggregates: developmental pattern and modulation by glucocorticoids and forskolin. Neuroendocrinology 53: 29–34, 1991.
CASTELL, J. V., T. GEIGER, V. GROSS, T. ANDUS, E. WALTER, T.
HIRANO, T. KISHIMOTO, AND P. C. HEINRICH. Plasma clearance,
organ distribution and target cells of interleukin-6/hepatocytestimulating factor in the rat. Eur. J. Biochem. 177: 357–361, 1988.
CECCATELLI, S., AND M. ERIKSSON. The effect of lactation on
nitric oxide synthase gene expression. Brain Res. 625: 177–179,
1993.
CECCATELLI, S., A. L. HULTING, X. ZHANG, L. GUSTAFSSON,
M. VILLAR, AND T. HOKFELT. Nitric oxide synthase in the rat
anterior pituitary gland and the role of nitric oxide in regulation
of luteinizing hormone secretion. Proc. Natl. Acad. Sci. USA 90:
11292–11296, 1993.
CECCATELLI, S., J. M. LUNDBERG, J. FAHRENKRUG, D. S.
BREDT, S. H. SNYDER, AND T. HOEKFELT. Evidence for the
involvement of nitric oxide in the regulation of hypothalamic portal blood flow. Neuroscience 51: 769–772, 1992.
CHAI, Z., S. GATTI, C. TONIATTI, V. POLI, AND T. BARTFAI.
Interleukin (IL)-6 gene expression in the central nervous system
is necessary for fever response to lipopolysaccharide of IL-1b: a
study on IL-6 deficient mice. J. Exp. Med. 183: 311–316, 1996.
CHAIDARUN, S. S., M. C. EGGO, P. M. STEWART, AND M. C.
SHEPPARD. Modulation of epidermal growth factor binding and
receptor gene expression by hormones and growth factors in
sheep pituitary cells. J. Endocrinol. 143: 489–496, 1994.
CHAN, R. K. W., E. R. BROWN, A. ERICSSON, K. J. KOVACS, AND
P. E. SAWCHENKO. A comparison of two immediate-early genes,
c-fos and NGFI-B, as markers for functional activation in stressrelated neuroendocrine circuitry. J. Neurosci. 13: 5126–5138,
1993.
CHANG, S. L., T. REN, AND J. E. ZADINA. Interleukin-1 activation
of FOS proto-oncogene protein in the rat hypothalamus. Brain
Res. 617: 123–130, 1993.
CHANG, Y., S. ALBRIGHT, AND F. LEE. Cytokines in the central
nervous system: expression of macrophage colony stimulating factor and its receptor during development. J. Neuroimmunol. 52:
9–17, 1994.
CHEIFETZ, S., N. LING, R. GUILLEMAN, AND J. MASSAGUE. A
surface component on GH3 pituitary cells that recognizes transforming growth factor-b, activin and inhibin. J. Biol. Chem. 263:
17225–17228, 1988.
CHILDS, G. V., J. PATTERSON, G. UNABIA, D. ROUGEAU, AND
P. WU. Epidermal growth factor enhances ACTH secretion and
expression of POMC mRNA by corticotropes in mixed and enriched cultures. Mol. Cell. Neurosci. 2: 235–243, 1991.
CHILDS, G. V., D. ROUGEAU, AND G. UNABIA. Corticotropin-releasing hormone and epidermal growth factor: mitogens for anterior pituitary corticotropes. Endocrinology 136: 1595–1602, 1995.
CHOVER-GONZALEZ, A. J., S. L. LIGHTMAN, AND M. S. HARBUZ.
An investigation of the effects of interleukin-1b on plasma arginine
/ 9j0c$$oc11
P13-8
159.
160.
161.
162.
163.
164.
165.
166.
167.
168.
169.
170.
171.
172.
173.
174.
175.
176.
177.
11-25-98 11:16:36
Volume 79
vasopressin in the rat: role of adrenal steroids. J. Endocrinol. 142:
361–366, 1994.
CHRISTENSEN, J. D., E. W. HANSEN, AND B. FJALLAND. Influence of interleukin-1b on the secretion of oxytocin and vasopressin from the isolated rat neurohypophysis. Pharmacol. Toxicol.
67: 81–83, 1990.
CHULUYAN, H. E., D. SAPHIER, W. M. ROHN, AND A. J. DUNN.
Noradrenergic innervation of the hypothalamus participates in
adrenocortical responses to interleukin-1. Neuroendocrinology
56: 106–111, 1992.
COCEANI, F., J. LEES, AND C. A. DINARELLO. Occurrence of
interleukin-1 in cerebrospinal fluid in the conscious cat. Brain
Res. 446: 245–250, 1988.
COHEN, M. C., AND S. COHEN. Cytokine function. A study in
biological diversity. Am. J. Clin. Pathol. 105: 589–598, 1996.
COLOTTA, F., F. RE, M. MUZIO, R. BERTINI, N. POLENTARUTTI,
M. SIRONI, J. G. GIRI, S. K. DOWER, J. E. SIMS, AND A. MANTOVANI. Interleukin-1 type II receptor: a decoy target for IL-1 that
is regulated by IL-4. Science 261: 472–475, 1993.
CONNER, J. M., AND S. VARON. Nerve growth factor immunoreactivity in the anterior pituitary of the rat. Neuroreport 4: 395–398,
1993.
CONNOR, T. J., AND B. E. LEONARD. Depression, stress and immunological activation: the role of cytokines in depressive disorders. Life Sci. 62: 583–606, 1997.
CONTI, B., J. W. JAHNG, C. TINTI, J. H. SON, AND T. H. JOH.
Induction of interferon-gamma inducing factor in the adrenal cortex. J. Biol. Chem. 272: 2035–2037, 1997.
COOPER, A. L., S. BROUWER, A. V. TURNBULL, G. N. LUHESHI,
S. J. HOPKINS, S. L. KUNKEL, AND N. J. ROTHWELL. Tumor necrosis factor-alpha and fever after peripheral inflammation in the
rat. Am. J. Physiol. 267 (Regulatory Integrative Comp. Physiol.
36): R1431–R1436, 1994.
COOPER, A. L., AND N. J. ROTHWELL. Mechanisms of early and
late hypermetabolism and fever after localized tissue injury in
rats. Am. J. Physiol. 261 (Endocrinol. Metab. 24): E698–E708,
1991.
COOPER, A. L., A. V. TURNBULL, S. J. HOPKINS, AND N. J. ROTHWELL. Dietary n-3 fatty acids inhibit fever induced by inflammation in the rat. Mediators Inflammation 3: 353–357, 1994.
CORNFIELD, L. J., AND M. A. SILLS. High affinity interleukin-6
binding sites in bovine hypothalamus. Eur. J. Pharmacol. 202:
113–115, 1991.
CORSINI, E., A. DUFOUR, E. CIUSANI, M. GELATI, S. FRIGERIO,
A. GRITTI, L. CAJOLA, G. L. MANCARDI, G. MASSA, AND A. SALMAGGI. Human brain endothelial cells and astrocytes produce IL1 beta but not IL-10. Scand. J. Immunol. 44: 506–511, 1996.
COSTA, A., P. TRAINER, M. BESSER, AND A. GROSSMAN. Nitric
oxide modulates the release of corticotropin-releasing hormone
from the rat hypothalamus in vitro. Brain Res. 605: 187–192, 1993.
COULIE, P. G., M. STEVENS, AND J. VAN SNICK. High- and lowaffinity receptors for murine IL-6. Distinct distribution on B and
T cells. Eur. J. Immunol. 19: 2107–2114, 1989.
CREASEY, A. A., P. STEVENS, J. KENNEY, A. C. ALISON, K. WARREN, R. CATLETT, L. HINSHAW, AND F. B. TAYLOR. Endotoxin
and cytokine profile in plasma of baboons challenged with lethal
and sublethal Escherichia coli. Circ. Shock 33: 84–91, 1991.
CROFFORD, L. J., K. T. KALOGERAS, G. MASTORAKOS, M. A.
MAGIAKOU, J. WELLS, K. S. KANIK, P. W. GOLD, G. P.
CHROUSOS, AND R. L. WILDER. Circadian relationship between
interleukin (IL)-6 and hypothalamic-pituitary-adrenal axis hormones: failure of IL-6 to cause sustained hypercortisolism in patients with early untreated rheumatoid arthritis. J. Clin. Endocrinol. Metab. 82: 1279–1283, 1997.
CROWN, J., A. JAKUBOWSKI, N. KEMENY, M. GORDON, C.
GASPARETTO, G. WONG, C. SHERIDAN, G. TONER, AND J. BOTET. A phase I trial of recombinant human interleukin-1 beta
alone and in combination with myelosuppressive doses of 5-fluorouracil in patients with gastrointestinal cancer. Blood 78: 1420–
1427, 1991.
CUNNINGHAM, E. T., JR., E. WADA, D. B. CARTER, D. E. TRACEY, J. F. BATTEY, AND E. B. DE SOUZA. Localization of interleu-
pra
APS-Phys Rev
Downloaded from on April 23, 2014
148.
ANDREW V. TURNBULL AND CATHERINE L. RIVIER
January 1999
178.
179.
180.
181.
182.
183.
184.
185.
187.
188.
189.
190.
191.
192.
193.
194.
195.
196.
kin-1 receptor messenger RNA in murine hippocampus. Endocrinology 128: 2666–2668, 1991.
CUNNINGHAM, E. T., JR., E. WADA, D. B. CARTER, D. E. TRACEY, J. F. BATTEY, AND E. B. DE SOUZA. In situ histochemical
localization of type I interleukin-1 receptor messenger RNA in the
central nervous system, pituitary, and adrenal gland of the mouse.
J. Neurosci. 12: 1101–1114, 1992.
CURTI, B. D., W. J. URBA, D. L. LONGO, J. E. JANIK, W. H.
SHARFMAN, L. L. MILLER, G. CIZZA, M. SHIMIZU, J. J. OPPENHEIM, W. G. ALVORD, AND J. W. SMITH. Endocrine effects of IL1 alpha and beta administered in a phase I trial to patients with
advanced cancer. J. Immunother. Emphasis Tumor Immunol.
19: 142–148, 1996.
DARLING, G., D. S. GOLDSTEIN, R. STULL, C. M. GORTHSCHBOTH, AND J. A. NORTON. Tumor necrosis factor: immune endocrine interaction. Surgery 106: 1155–1160, 1989.
DARLINGTON, D. N., J. SHINSAKO, AND M. F. DALLMAN. Medullary lesions eliminate ACTH responses to hypotensive hemorrhage. Am. J. Physiol. 251 (Regulatory Integrative Comp. Physiol.
20): R106–R115, 1986.
DARNAY, B. G., AND B. B. AGGARWAL. Early events in TNF signaling: a story of associations and dissociations. J. Leukoc. Biol.
61: 559–566, 1997.
DASCOMBE, M. J., AND A. S. MILTON. Study on the possible entry
of bacterial endotoxin and prostaglandin E2 into the central nervous system from the blood. Br. J. Pharmacol. 66: 465–472, 1979.
DAVIES, A. M. The role of neurotrophins in the developing nervous system. J. Neurobiol. 25: 1334–1348, 1994.
DAVIES, A. M. Tracking neurotrophin function. Nature 368: 193–
194, 1994.
DAWSON, T. M., AND S. H. SNYDER. Gases as biological messengers: nitric oxide and carbon monoxide in the brain. J. Neurosci.
14: 5147–5159, 1994.
DAY, H. E. W., AND H. AKIL. Differential pattern of c-fos mRNA
in rat brain following central and peripheral administration of
interleukin-1-beta: implications for mechanism of action. Neuroendocrinology 63: 207–218, 1996.
DE, A., T. E. MORGAN, R. C. SPETH, N. BOYADJIEVA, AND D. K.
SARKAR. Pituitary lactotrope expresses transforming growth factor b (TGFb) type II receptor mRNA and protein and contains
125
I-TGFb1 binding sites. J. Endocrinol. 149: 19–27, 1996.
DEFORGE, L. E., AND D. G. REMICK. Kinetics of TNF, IL-6 and
IL-8 gene expression in LPS-stimulated human whole blood. Biochem. Biophys. Res. Commun. 174: 18–24, 1991.
DELI, M. A., L. DESCAMPS, M. P. DEHOUCK, R. CECCHELLI,
F. JOO, C. S. ABRAHAM, AND G. TORPIER. Exposure of tumor
necrosis factor-alpha to luminal membrane of bovine capillary
endothelial cells cocultured with astrocytes induces a delayed
increase in permeability and cytoplasmic stress fiber formation.
J. Neurosci. Res. 41: 717–726, 1995.
DEL REY, A., H. BESEDOVSKY, AND E. SORKIN. Endogenous
blood levels of corticosterone control the immunologic cell mass
and B cell activity in mice. J. Immunol. 133: 572–575, 1984.
DEL REY, A., H. FURUKAWA, G. MONGE-ARDITI, A. KABIERSCH, K.-H. VOIGT, AND H. O. BESEDOVSKY. Alterations in
the pituitary-adrenal axis of adult mice following neonatal exposure to interleukin-1. Brain Behav. Immun. 10: 235–248, 1996.
DELRUE, C., B. DELEPLANQUE, F. ROUGE-PONT, S. VITIELLO,
AND P. J. NEVEU. Brain monoaminergic, neuroendocrine, and immune responses to an immune challenge in relation to brain and
behavioral lateralization. Brain Behav. Immun. 8: 137–152, 1994.
DENICOFF, K. D., T. M. DURKIN, M. T. LOTZE, P. E. QUINLAN,
C. L. DAVIS, S. J. LISTWAK, S. A. ROSENBERG, AND D. R. RUBINOW. The neuroendocrine effects of interleukin-2 treatment. J.
Clin. Endocrinol. Metab. 69: 402–410, 1989.
DE RIJK, R., N. VAN ROOJEN, H. O. BESEDOVSKY, A. DEL REY,
AND F. BERKENBOSCH. Selective depletion of macrophages prevents pituitary adrenal activation in response to subpyrogenic, but
not pyrogenic, doses of bacterial endotoxin in rats. Endocrinology
128: 330–338, 1991.
DE RIJK, R. H., A. BOELEN, F. J. H. TILDER, AND F. BERKENBOSCH. Induction of interleukin-6 by circulating adrenaline in the
rat. Psychoneuroendocrinology 19: 155–163, 1994.
/ 9j0c$$oc11
P13-8
49
197. DE SIMONI, M. G., R. DEL BO, A. DE LUIGI, S. SIMARD, AND G.
FORLONI. Central endotoxin induces different patterns of interleukin (IL)-1b and IL-6 messenger ribonucleic acid expression
and IL-6 secretion in the brain and periphery. Endocrinology 136:
897–902, 1995.
198. DE SOUZA, E. B., E. L. WEBSTER, D. E. GRIGORIADIS, AND D. E.
TRACEY. Corticotropin-releasing factor (CRF) and interleukin-1
receptors in the brain-pituitary-immune axis. Psychopharmacol.
Bull. 25: 299–305, 1989.
199. DE VRIES, H. E., M. C. M. BLOM-POOSEMALEN, A. G. DE BOER,
T. J. C. VAN BERKEL, D. D. BREIMER, AND J. K. KUIPER. Effect
of endotoxin on permeability of bovine cerebral endothelial cell
layers in vitro. J. Pharmacol. Exp. Ther. 277: 1418–1423, 1996.
200. DE VRIES, H. E., M. C. M. BLOM-POOSEMALEN, M. VAN OOSTEN, A. G. DE BOER, T. J. C. VAN BERKEL, D. D. BREIMER, AND
J. K. KUIPER. The influence of cytokines on the integrity of the
blood-brain-barrier in vitro. J. Neuroimmunol. 64: 37–43, 1996.
201. DE VRIES, H. E., K. H. HOOGENDOORN, J. VAN DIJK, F. J. ZIJLSTRA, A.-M. VAN DAM, D. D. BREIMER, T. J. C. VAN BERKEL,
A. G. DE BOER, AND J. KUIPER. Eicosanoid production by rat
cerebral endothelial cells: stimulation by lipopolysaccharide, interleukin-1 and interleukin-6. J. Neuroimmunol. 59: 1–8, 1995.
202. DEYERLE, K. L., J. E. SIMS, S. K. DOWER, AND M. A. BOTHWELL.
Pattern of IL-1 receptor gene expression suggests role in noninflammatory processes. J. Immunol. 149: 1657–1665, 1992.
203. DINAERELLO, C. A., AND R. C. THOMPSON. Blocking IL-1: interleukin-1 receptor antagonist in vivo and in vitro. Immunol.
Today 12: 404–410, 1991.
204. DINAERELLO, C. A., P. WEINER, AND S. M. WOLFF. Radiolabelling and disposition in rabbits of purified human leukocytic
pyrogen (Abstract). Clin. Res. 26: 522A, 1978.
205. DINARELLO, C. A. Interleukin-1 and interleukin-1 antagonism.
Blood 77: 1627–1652, 1991.
206. DINARELLO, C. A. The proinflammatory cytokines interleukin-1
and tumor necrosis factor and treatment of the septic shock syndrome. J. Infect. Dis. 163: 1177–1184, 1991.
207. DINARELLO, C. A., J. G. CANNON, J. MANCILLA, I. BISHAI, J.
LEES, AND F. COCEANI. Interleukin-6 as an endogenous pyrogen:
induction of prostaglandin E2 in brain but not in peripheral blood
mononuclear cells. Brain Res. 562: 199–206, 1991.
208. DOBREA, G. M., J. R. UNNERSTALL, AND M. S. RAO. The expression of CNTF message and immunoreactivity in the central and
peripheral nervous system of the rat. Brain Res. Dev. Brain Res.
66: 209–219, 1992.
209. DOPP, J. M., A. MACKENZIE-GRAHAM, G. C. OTERO, AND J. E.
MERRILL. Differential expression, cytokine modulation, and specific functions of type-1 and type-2 tumor necrosis factor receptors
in rat glia. J. Neuroimmunol. 75: 104–112, 1997.
210. DU, X., E. T. EVERETT, G. WANG, W. H. LEE, Z. YANG, AND
D. A. WILLIAMS. Murine interleukin-11 (IL-11) is expressed at high
levels in the hippocampus and expression is developmentally regulated in the testis. J. Cell. Physiol. 168: 362–372, 1996.
211. DUNN, A. J. Systemic interleukin-1 administration stimulates hypothalamic norepinephrine metabolism paralleling the increased
plasma corticosterone. Life Sci. 43: 429–435, 1988.
212. DUNN, A. J. Endotoxin-induced activation of cerebral catecholamine and serotonin metabolism: comparison with interleukin-1.
J. Pharmacol. Exp. Ther. 261: 964–969, 1992.
213. DUNN, A. J. The role of interleukin-1 and tumor necrosis factor a
in the neurochemical and neuroendocrine responses to endotoxin.
Brain Res. Bull. 29: 807–812, 1992.
214. DUNN, A. J. Role of cytokines in infection-induced stress. Ann.
NY Acad. Sci. 697: 189–202, 1993.
215. DUNN, A. J., AND H. E. CHULUYAN. The role of cyclooxygenase
and lipoxygenase in the interleukin-1-induced activation of the
HPA axis: dependence on the route of injection. Life Sci. 51: 219–
225, 1992.
216. DUNN, A. J., AND H. E. CHULUYAN. Endotoxin elicits normal tryptophan and indolamine responses but impaired catecholamine and
pituitary-adrenal responses in endotoxin-resistant mice. Life Sci.
54: 847–853, 1994.
218. DUNN, A. J., M. L. POWELL, AND J. M. GASKIN. Virus-induced
increases in plasma corticosterone. Science 238: 1423–1424, 1987.
11-25-98 11:16:36
pra
APS-Phys Rev
Downloaded from on April 23, 2014
186.
REGULATION OF HPA AXIS BY CYTOKINES
50
ANDREW V. TURNBULL AND CATHERINE L. RIVIER
/ 9j0c$$oc11
P13-8
236.
237.
238.
239.
240.
241.
242.
243.
244.
245.
246.
247.
248.
249.
250.
251.
252.
253.
11-25-98 11:16:36
vation of stress-related neuroendocrine circuitry by intravenous
interleukin-1. J. Neurosci. 17: 7166–7179, 1997.
ERICSSON, A., M. EK, AND N. LINDEFORS. Distribution of prostaglandin E2 receptor (EP3 subtype) mRNA containing cells in the
rat central nervous system. Soc. Neurosci. Abstr. 21: 45.9, 1995.
ERICSSON, A., K. J. KOVACS, AND P. E. SAWCHENKO. A functional anatomical analysis of central pathways subserving the effects of interleukin-1 on stress-related neuroendocrine neurons.
J. Neurosci. 14: 897–913, 1994.
ERICSSON, A., C. LIU, R. P. HART, AND P. E. SAWCHENKO. Type
I interleukin-1 receptor in the rat brain: distribution, regulation
and relationship to sites of IL-1-induced cellular activation. J.
Comp. Neurol. 361: 681–698, 1995.
ERKUT, Z. A., M. A. HOFMAN, R. RAVID, AND D. F. SWAAB. Increased activity of hypothalamic corticotropin-releasing hormone
neurons in multiple sclerosis. J. Neuroimmunol. 62: 27–33, 1995.
ERNFORS, P., C. WETMORE, L. OLSON, AND H. PERSSON. Identification of cells in rat brain and peripheral tissues expressing
mRNA for members of the nerve growth factor family. Neuron 5:
511–526, 1990.
ESPEVICK, T., M. BROCKHAUS, H. LOETSCHER, U. NONSTAD,
AND R. SHALABY. Characterization of binding and biological effects of monoclonal antibodies against a human tumor necrosis
factor receptor. J. Exp. Med. 171: 415–426, 1990.
FABRY, Z., K. M. FITZSIMMONS, J. A. HERLEIN, T. O. MONINGER, M. B. DOBBS, AND M. N. HART. Production of the cytokines interleukin-1 and 6 by murine brain microvessel endothelium and smooth muscle pericytes. J. Neuroimmunol. 47: 23–34,
1993.
FAGARSON, M., R. ESKAY, AND J. AXELROD. Interleukin-1 potentiates the secretion of b endorphin induced by secretagogues
in mouse pituitary cell line (AtT20). Proc. Natl. Acad. Sci. USA
86: 2070–2073, 1989.
FAN, X., AND G. V. CHILDS. Epidermal growth factor and transforming growth factor-alpha messenger ribonucleic acids and
their receptors in the rat anterior pituitary: localization and regulation. Endocrinology 136: 2284–2293, 1995.
FAN, X., G. T. NAGLE, T. J. COLLINS, AND G. V. CHILDS. Differential regulation of epidermal growth factor and transforming
growth factor-alpha messenger ribonucleic acid in the rat anterior
pituitary and hypothalamus induced by stresses. Endocrinology
136: 873–880, 1995.
FANTUZZI, G., F. BENIGNI, M. SIRONI, M. CONNI, M. CARELLI,
L. CANTONNI, L. SHAPIRO, C. A. DINARELLO, J. D. SIPE, AND
P. GHEZZI. Ciliary neurotrophic factor (CNTF) induces serum
amyloid A, hypoglycemia and anorexia, and potentiates IL-1 induced corticosterone and IL-6 production in mice. Cytokine 7:
150–156, 1995.
FANTUZZI, G., AND C. A. DINARELLO. The inflammatory response
in interleukin-1b-deficient mice: comparison with other cytokinerelated knockout mice. J. Leukoc. Biol. 59: 489–493, 1996.
FANTUZZI, G., G. KU, M. W. HARDING, D. J. LIVINGSTON, J. D.
SIPE, K. KUIDA, R. A. FLAVELL, AND C. A. DINARELLO. Response
to local inflammation of IL-1b-converting enzyme deficient mice.
J. Immunol. 158: 1818–1824, 1997.
FANTUZZI, G., A. J. PUREN, M. W. HARDING, D. J. LIVINGSTON,
AND C. A. DINARELLO. Interleukin-18 regulation of interferon
gamma production and cell proliferation as shown in interleukin1 beta converting enzyme (caspase-1) deficient mice. Blood 91:
2118–2125, 1998.
FANTUZZI, G., H. ZHENG, R. FAGGIONI, F. BENIGNI, P. GHEZZI,
J. D. SIPE, A. R. SHAW, AND C. A. DINARELLO. Effect of endotoxin in IL-1 beta-deficient mice. J. Immunol. 157: 291–296, 1996.
FARRAR, W. L., P. L. KILIAN, M. R. RUFF, J. M. HILL, AND C. B.
PERT. Visualization and characterization of interleukin 1 receptors in brain. J. Immunol. 139: 459–463, 1987.
FARRAR, W. L., M. VINCOUR, AND J. M. HILL. In situ hybridization
histochemistry localization of interleukin-3 mRNA in mouse brain.
Blood 73: 137–140, 1989.
FASSBENDER, K., R. SCHMIDT, R. MOSSNER, M. DAFFERTSHOFER, AND M. HENNERICI. Pattern of activation of the hypothalamic-pituitary-adrenal axis in acute stroke. Relation to acute con-
pra
APS-Phys Rev
Downloaded from on April 23, 2014
219. DUNN, A. J., M. L. POWELL, C. MEITIN, AND P. A. SMALL. Virus
infection as a stressor: influenza virus elevates plasma concentrations of corticosterone, and brain concentrations of MHPG and
tryptophan. Physiol. Behav. 45: 591–594, 1989.
220. DUNN, A. J., M. L. POWELL, W. V. MORSEHEAD, J. M. GASKIN,
AND N. R. HALL. Effects of Newcastle disease virus administration
to mice on the metabolism of cerebral biogenic amines, plasma
corticosterone, and lymphocyte proliferation. Brain Behav. Immun. 1: 216–230, 1987.
221. DUNN, A. J., AND S. L. VICKERS. Neurochemical and neuroendocrine responses to Newcastle disease virus administration in mice.
Brain Res. 645: 103–112, 1994.
222. DUNN, A. J., AND J. WANG. Cytokine effects on CNS biogenic
amines. Neuroimmunomodulation 2: 319–328, 1995.
223. EBISUI, O., J. FUKATA, N. MURAKAMI, H. KOBAYASHI, H. SEGAWA, S. MURO, I. HANAOKA, Y. NAITO, Y. MASUI, Y. OHMOTO,
H. IMURA, AND K. NAKAO. Effect of IL-1 receptor antagonist and
antiserum to TNF-a on LPS-induced plasma ACTH and corticosterone in rats. Am. J. Physiol. 266 (Endocrinol. Metab. 29): E986–
E992, 1994.
224. EGUCHI, N., H. HAYASHI, Y. URADE, S. ITO, AND O. HAYAISHI.
Central action of prostaglandin E2 and its methyl ester in the
induction of hyperthermia after their systemic administration in
urethane anesthetized rats. J. Pharmacol. Exp. Ther. 247: 671–
679, 1988.
225. EISENBERG, S. P., R. J. EVANS, W. P. AREND, E. VERDERBER,
M. T. BREWER, C. H. HANNUM, AND R. C. THOMPSON. Primary
structure and functional expression from complimentary DNA of
a known interleukin-1 receptor antagonist. Nature 343: 341–346,
1990.
226. ELENKOV, I. J., K. KOVACS, E. DUDA, E. STARK, AND E. S. VIZI.
Presynaptic inhibitory effect of TNF-a on the release of noradrenaline in isolated median eminence. J. Neuroimmunol. 43: 117–120,
1992.
227. ELENKOV, I. J., K. KOVACS, J. KISS, L. BERTOK, AND E. S. VIZI.
Lipopolysaccharide is able to bypass corticotrophin-releasing factor in affecting plasma ACTH and corticosterone levels: evidence
from rats with lesions of the paraventricular nucleus. J. Endocrinol. 133: 231–236, 1992.
228. EL-HUSSEINI, A., J. A. PATTERSON, AND R. P. SHIU. Basic fibroblast growth factor (bFGF) and two of its receptors, FGFR1 and
FGFR2: gene expression in the rat brain during postnatal development as determined by quantitative RT-PCR. Mol. Cell. Endocrinol. 104: 191–200, 1994.
229. ELLISON, M. D., J. T. POVLISHOCK, AND R. E. MERCHANT.
Blood-brain barrier dysfunction in cats following recombinant interleukin-2 infusion. Cancer Res. 47: 5765–5770, 1987.
230. ELMQUIST, J. K., M. R. ACKERMANN, K. B. REGISTER, R. B.
RIMLER, L. R. ROSS, AND C. D. JACOBSON. Induction of fos-like
immunoreactivity in the rat brain following Pasteurella multocida
endotoxin administration. Endocrinology 133: 3054–3057, 1993.
231. ELMQUIST, J. K., C. D. BREDER, J. E. SHERIN, T. E. SCAMMELL,
W. F. HICKEY, D. DEWITT, AND C. B. SAPER. Intravenous lipopolysaccharide induces cyclooygenase-like immunoreactivity in
rat brain perivascular microglia and meningeal macrophages. J.
Comp. Neurol. 381: 119–129, 1997.
232. ELMQUIST, J. K., T. E. SCAMMELL, C. D. JACOBSON, AND C. B.
SAPER. Distribution of Fos-like immunoreactivity in the rat brain
following intravenous lipopolysaccharide administration. J.
Comp. Neurol. 371: 85–103, 1996.
233. ENGELMANN, H., H. HOLTMANN, C. BRALEBUSCH, Y. S. AVNIL,
I. SAROV, Y. NOPHAR, E. HADAS, O. LEITNER, AND D. WALLACH. Antibodies to a soluble form of a tumor necrosis factor
(TNF) receptor have TNF-like activity. J. Biol. Chem. 265: 14497–
14504, 1990.
234. ERICKSON, S. L., F. J. DE SAUVAGE, K. KIKLY, K. CARVERMOORE, S. PITTS-MEEK, N. GILLETT, K. C. F. SHEEHAN, R. D.
SCHREIBER, D. V. GOEDDEL, AND M. W. MOORE. Decreased
sensitivity to tumor necrosis factor but normal T-cell development
in TNF receptor-2 deficient mice. Nature 372: 560–563, 1994.
235. ERICSSON, A., C. ARIAS, AND P. E. SAWCHENKO. Evidence for
an intramedullary prostaglandin-dependent mechanism in the acti-
Volume 79
January 1999
254.
255.
256.
257.
258.
259.
260.
262.
263.
264.
265.
266.
267.
268.
269.
270.
fusional state, extent of brain damage, and clinical outcome.
Stroke 25: 1105–1108, 1994.
FATTORI, E., M. CAPPALLETTI, P. COASTA, C. SELLITTO, L.
CANTONI, M. CARELLI, R. FAGGIONI, G. FANTUZZI, P. GHEZZI,
AND V. POLI. Defective inflammatory response in interleukin-6
deficient mice. J. Exp. Med. 180: 1243–1250, 1994.
FEIGE, J. J., C. COCHET, C. SAVONA, D. L. SHI, M. KERAMIDAS,
G. DEFAYE, AND E. M. CHAMBAZ. Transforming growth factor
beta 1: an autocrine regulator of adrenocortical steroidogenesis.
Endocr. Res. 17: 267–279, 1991.
FERNANDEZ-ALONSO, A., K. BENAMAR, M. SANCHIBRIAN,
F. J. LOPEZ-VALUPUESTA, AND F. J. MINANO. Role of interleukin-1b, interleukin-6 and macrophage inflammatory protein-1b in
prostaglandin E2-induced hyperthermia in rats. Life Sci. 59: 185–
190, 1996.
FERNANDEZ-BOTRAN, R. Soluble cytokine receptors: their role
in immunoregulation. FASEB J. 5: 2567–2571, 1991.
FERRARA, N., J. WINER, AND W. J. HENZEL. Pituitary follicular
cells secrete an inhibitor of aortic endothelial cell growth: identification as leukemia inhibitory factor. Proc. Natl. Acad. Sci. USA
89: 698–702, 1992.
FERRER, I., S. ALCANTARA, J. BALLABRIGA, M. OLIVE, R.
BLANCO, R. RIVERA, M. CARMONA, M. BERRUEZO, S. PITARCH, AND A. M. PLANAS. Transforming growth factor-alpha
(TGF-alpha) and epidermal growth factor-receptor (EGF-R) immunoreactivity in normal and pathologic brain. Prog. Neurobiol.
49: 99–123, 1996.
FERRETTI, M., V. CASINI-RAGGI, T. T. PIZARRO, S. P. EISENBERG, C. C. NAST, AND F. COMINELLI. Neutralization of endogenous IL-1 receptor antagonist exacerbates and prolongs inflammation in rabbit immune colitis. J. Clin. Invest. 94: 449–453, 1994.
FLESHNER, M., L. E. GOEHLER, J. HERMAN, J. K. RELTON, S. F.
MAIER, AND L. R. WATKINS. Interleukin-1 beta induced corticosterone elevation and hypothalamic NE depletion is vagally mediated. Brain Res. Bull. 37: 605–610, 1995.
FLESHNER, M., L. SLIBERT, T. DEAK, L. E. GOEHLER, D. MARTIN, L. R. WATKINS, AND S. F. MAIER. TNF-alpha-induced corticosterone elevation but not serum protein or corticosteroid binding globulin reduction is vagally mediated. Brain Res. Bull. 44:
701–706, 1997.
FONG, Y., K. J. TRACEY, L. L. MOLDAWER, D. G. HESSE, K. B.
MANOGUE, J. S. KENNEY, A. T. LEE, G. C. KUO, A. C. ALLISON,
S. F. LOWRY, AND A. CERAMI. Antibodies to cachectin/tumor necrosis factor reduce interleukin-1 beta and interleukin-6 appearance during lethal bacteremia. J. Exp. Med. 170: 1627–1633, 1989.
FONTANA, A., E. WEBER, AND J.-M. DAYER. Synthesis of interleukin-1/endogenous pyrogen in the brain of endotoxin-treated mice:
a step in fever induction? J. Immunol. 133: 1696–1698, 1984.
FORSTERMANN, U., I. GATH, P. SCHWARZ, E. I. CLOSS, AND H.
KLEINHART. Isoforms of nitric oxide synthase. Properties, cellular distribution and expressional control. Biochem. Pharmacol.
50: 1321–1332, 1995.
FREI, K., K. NOHAVA, U. V. MALIPIERO, C. SCHWARDEL, AND
A. FONTANA. Production of macrophage colony-stimulating factor by astrocytes and brain macrophages. J. Neuroimmunol. 40:
189–195, 1992.
FRENCH, R. A., J. F. ZACHARY, R. DANTZER, L. S. FRAWLEY,
R. CHIZZONITE, P. PARNET, AND K. W. KELLEY. Dual expression
of p80 type I and p68 type II interleukin-1 receptors on anterior
pituitary cells synthesizing growth hormone. Endocrinology 137:
4027–4036, 1996.
FUKATA, J., T. USUI, Y. NAITOH, Y. NAKAI, AND H. IMURA.
Effects of recombinant interleukin-1a, -1b, 2 and 6 on ACTH synthesis and release in mouse pituitary tumor cell line AtT20. J.
Endocrinol. 122: 33–39, 1989.
GABELLEC, M.-M., R. GRIFFAIS, G. FILLION, AND F. HAOUR.
Expression of interleukin 1a, interleukin 1b and interleukin 1
receptor antagonist mRNA in mouse brain: regulation by bacterial
lipopolysaccharide (LPS) treatment. Brain Res. Mol. Brain Res.
31: 122–130, 1995.
GABELLEC, M.-M., R. GRIFFAIS, G. FILLION, AND F. HAOUR.
Interleukin-1 receptors type I and type II in the mouse brain:
/ 9j0c$$oc11
P13-8
271.
272.
273.
274.
275.
276.
277.
278.
279.
280.
281.
282.
283.
284.
285.
286.
287.
288.
289.
11-25-98 11:16:36
51
kinetics of mRNA expressions after peripheral administration of
bacterial lipopolysaccharide. J. Neuroimmunol. 66: 65–70, 1996.
GABELLEC, M.-M., M. JAFARIAN-TEHRANI, R. GRIFFAIS, AND
F. HAOUR. Interleukin-1 receptor accessory protein transcripts
in the brain and spleen: kinetics after peripheral administration
of bacterial lipopolysaccharide in mice. Neuroimmunomodulation 3: 304–309, 1996.
GADIENT, R. A., A. LACHMUND, K. UNSICKER, AND U. OTTEN.
Expression of interleukin-6 (IL-6) and IL-6 receptor mRNAs in rat
adrenal medulla. Neurosci. Lett. 194: 17–20, 1995.
GADIENT, R. A., AND U. OTTEN. Differential expression of interleukin-6 (IL-6) and interleukin-6 receptor (IL-6R) mRNAs in rat
hypothalamus. Neurosci. Lett. 153: 13–16, 1993.
GADIENT, R. A., AND U. OTTEN. Expression of interleukin-6 (IL6) and interleukin-6 receptor (IL-6R) mRNAs in rat brain during
postnatal development. Brain Res. 637: 10–14, 1994.
GAILLARD, R. C., D. TURNILL, P. SAPPINO, AND A. F. MULLER.
Tumor necrosis factor alpha inhibits the hormonal response of
the pituitary gland to hypothalamic releasing factors. Endocrinology 127: 101–106, 1990.
GARTHWAITE, J., G. GARTHAITE, R. M. PALMER, AND S. MONCADA. NMDA receptor activation induces nitric oxide synthesis
from arginine in rat brain slices. Eur. J. Pharmacol. 172: 413–
416, 1989.
GATTI, S., AND T. BARTFAI. Induction of tumor necrosis factoralpha mRNA in the brain after peripheral endotoxin treatment:
comparison with interleukin-1 family and interleukin-6. Brain Res.
624: 291–294, 1993.
GAY, N. J., AND F. J. KEITH. Drosophila and IL-1 receptor. Nature
351: 355–356, 1991.
GAYKEMA, R. P. A., I. DIJKSTRA, AND F. J. H. TILDERS. Subdiaphragmatic vagotomy suppresses endotoxin-induced activation
of hypothalamic corticotropin-releasing hormone neurons and
ACTH secretion. Endocrinology 136: 4717–4720, 1995.
GAYLE, D., S. E. ILYIN, AND C. R. PLATA-SALAMAN. Interleukin1 receptor type I mRNA levels in brain regions from male and
female rats. Brain Res. Bull. 42: 463–467, 1997.
GAYLE, D., S. E. ILYIN, AND C. R. PLATA-SALAMAN. Central nervous system IL-1 beta system and neuropeptide Y mRNAs during
IL-1 beta-induced anorexia in rats. Brain Res. Bull. 44: 311–317,
1997.
GAYLE, M. A., J. E. SIMS, S. K. DOWER, AND J. L. SLACK. Monoclonal antibody 1994–01 (also known as ALVA 42) reported to
recognize type II IL-1 receptor is specific for HLA-DR alpha and
beta chains. Cytokine 6: 83–86, 1994.
GEHR, F., R. GENTZ, M. BROCKHAUS, H. LOETSCHER, AND W.
LESSIAUER. Both tumor necrosis factor receptor types mediate
proliferative signals in human mononuclear cell activation. J. Immunol. 149: 911–917, 1992.
GELLER, D. A., A. K. NUSSLER, M. DI SILVINO, C. J. LOWENSTEIN, R. A. SHAPIRO, S. C. WANG, R. L. SIMMONS, AND
T. R. BILLIAR. Cytokines, endotoxin, and glucocorticoids regulate
the expression of inducible nitric oxide synthase in hepatocytes.
Proc. Natl. Acad. Sci. USA 90: 522–526, 1993.
GEMMA, C., P. GHEZZI, AND M. G. DE SIMONI. Activation of
the hypothalamic serotoninergic system by interleukin-1. Eur. J.
Pharmacol. 209: 139–140, 1991.
GERSHENWALD, J. E., Y. FONG, T. J. FAHEY, S. E. CALVANO, R.
CHIZZONITE, P. L. KILIAN, S. F. LOWRY, AND L. L. MOLDAWER.
Interleukin-1 receptor blockade attenuates the host inflammatory
response. Proc. Natl. Acad. Sci. USA 87: 4966–4970, 1990.
GHAYUR, T., S. BANERJEE, M. HUGUNIN, D. BUTLER, L. HERZOG, A. CARTER, L. QUINTAL, L. SEKUT, R. TALNIAN, M.
PASKIND, W. WONG, R. KAMEN, D. TRACEY, AND H. ALLEN.
Caspase-1 processes IFN-gamma-inducing factor and regulates
LPS-induced IFN-gamma production. Nature 386: 619–623, 1997.
GIBBS, R. B., J. T. MCCABE, C. R. BUCK, M. V. CHAO, AND D. W.
PFAFF. Expression of NGF receptor in the rat forebrain detected
with in situ hybridization and immunohistochemistry. Brain Res.
Mol. Brain Res. 6: 275–287, 1989.
GISSLINGER, H., T. SVOBODA, M. CLODI, B. GILLY, H. LUDWIG,
L. HAVELEC, AND A. LUGER. Interferon-alpha stimulates the hy-
pra
APS-Phys Rev
Downloaded from on April 23, 2014
261.
REGULATION OF HPA AXIS BY CYTOKINES
52
290.
291.
292.
293.
294.
296.
297.
298.
299.
300.
301.
302.
303.
304.
pothalamo-pituitary axis in vivo and in vitro. Neuroendocrinology
57: 489–495, 1993.
GIVALOIS, L., J. DORNAND, M. MEKAOUCHE, M. D. SOLIER,
A. F. BRISTOW, G. IXART, I. SIAUD, I. ASSENMACHER, AND G.
BARBANEL. Temporal cascade of plasma level surges in ACTH,
corticosterone, and cytokines in endotoxin-challenged rats. Am.
J. Physiol. 267 (Regulatory Integrative Comp. Physiol. 36): R164–
R170, 1994.
GIVALOIS, L., S. GAILLET, M. MEKAOUCHE, G. IXART, A. F.
BRISTOW, P. SIAUD, A. SZAFARCZYK, F. MALAVAL, I. ASSENMACHER, AND G. BARBANEL. Deletion of the ventral noradrenergic bundle obliterates the early ACTH response after systemic
LPS, independently from the plasma IL-1b surge. Endocrine 3:
481–485, 1995.
GIVALOIS, L., P. SIAUD, M. MEKAOUCHE, G. IXART, F. MALAVAL, I. ASSENMACHER, AND G. BARBANEL. Early hypothalamic
activation of combined Fos and CRH41 immunoreactivity and of
CRH41 release in push-pull cannulated rats after systemic endotoxin challenge. Mol. Chem. Pathol. 26: 171–186, 1995.
GIVALOIS, L., P. SIAUD, M. MEKAOUCHE, G. IXART, F. MALAVAL, I. ASSENMACHER, AND G. BARBANEL. Involvement of central histamine in the early phase of ACTH and corticosterone
responses to endotoxin in rats. Neuroendocrinology 63: 219–226,
1996.
GOEHLER, L. E., C. R. BUSCH, N. TARTAGLIA, J. RELTON, D.
SISK, S. F. MAIER, AND L. R. WATKINS. Blockade of cytokine
induced conditioned taste aversion by subdiaphragmatic vagotomy: further evidence for vagal mediation of immune-brain communication. Neurosci. Lett. 185: 163–166, 1995.
GOEHLER, L. E., R. P. A. GAYKEMA, F. J. H. TILDERS, S. F.
MAIER, AND L. R. WATKINS. Localization of immune and inflammatory markers in the rat vagus nerve and liver hilus (Abstract).
Proc. Annu. Meet. Soc. Neurosci. 26th Washington DC 1996, p.
43.9.
GOEHLER, L. E., J. K. RELTON, D. DRIPPS, R. KIECHLE, N. TARTAGLIA, S. F. MAIER, AND L. R. WATKINS. Vagal paraganglia bind
biotinylated interleukin-1 receptor antagonist: a possible mechanism for immune-to-brain communication. Brain Res. Bull. 43:
357–364, 1997.
GOLDBACH, J.-M., J. ROTH, AND E. ZEISBERGER. Fever suppression by subdiaphragmatic vagotomy in guinea pigs depends on the
route of pyrogen administration. Am. J. Physiol. 272 (Regulatory
Integrative Comp. Physiol. 41): R675–R681, 1997.
GONZALEZ, A. M., A. LOGAN, W. YING, D. A. LAPPI, M. BERRY,
AND A. BAIRD. Fibroblast growth factor in the hypothalamo-pituitary axis: differential expression of fibroblast growth factor-2 and
a high affinity receptor. Endocrinology 134: 2289–2297, 1994.
GONZALEZ-HERNANDEZ, J. A., S. R. BORNSTEIN, M. EHRHART-BORNSTEIN, E. SPATH-SCHWALBE, G. JIRIKOWSKI, AND
W. A. SCHERBAUM. Interleukin-6 messenger ribonucleic acid expression in human adrenal gland in vivo: new clue to a paracrine
or autocrine regulation of adrenal function. J. Clin. Endocrinol.
Metab. 79: 1492–1497, 1994.
GONZALEZ-HERNANDEZ, J. A., M. EHRHART-BORNSTEIN, E.
SPATH-SCHWALBE, W. A. SCHERBAUM, AND S. R. BORNSTEIN.
Human adrenal cells express tumor necrosis factor-alpha messenger ribonucleic acid: evidence for paracrine control of adrenal
function. J. Clin. Endocrinol. Metab. 81: 807–813, 1996.
GOSPODAROWICZ, D., A. BAIRD, J. CHENG, G. M. LUI, F. ESCH,
AND P. BOHLEN. Isolation of fibroblast growth factor from bovine
adrenal gland: physiochemical and biological characterization.
Endocrinology 118: 82–90, 1986.
GOTTFIRES, C. G., J. BALLDIN, K. BLENNOW, G. BRANE, I.
KARLSSON, B. REGLAND, AND A. WALLIN. Regulation of the
hypothalamo-pituitary-adrenal axis in dementia disorders. Ann.
NY Acad. Sci. 746: 336–343, 1994.
GOTTSCHALL, P. E., K. KOVES, K. MIZUNO, I. TATSUNO, AND A.
ARIMURA. Glucocorticoid upregulation of interleukin 1 receptor
expression in a glioblastoma cell line. Am. J. Physiol. 261 (Endocrinol. Metab. 24): E362–E368, 1991.
GREENFEDER, S. A., P. NUNES, L. KWEE, M. LABOW, R. A.
CHIZZONITE, AND G. JU. Molecular cloning and characterization
/ 9j0c$$oc11
P13-8
305.
306.
307.
308.
309.
310.
311.
312.
313.
314.
315.
316.
317.
318.
319.
320.
321.
322.
11-25-98 11:16:36
Volume 79
of a second subunit of the interleukin 1 receptor complex. J. Biol.
Chem. 270: 13757–13765, 1995.
GROSS, P. M., AND A. WEINDL. Peering through the windows of
the brain. J. Cereb. Blood Flow Metab. 7: 663–672, 1987.
GROTHE, C., AND K. UNSICKER. Immunocytochemical mapping
of basic fibroblast growth factor in the developing and adult rat
adrenal gland. Histochemistry 94: 141–147, 1990.
GU, Y., K. KUIDA, H. TSUTSUI, G. KU, K. HSIAO, M. A. FLEMING,
N. HAYASHI, K. HIGASHINO, H. OKAMURA, K. NAKANISHI, M.
KURIMOTO, T. TANIMOTO, R. A. FLAVELL, V. SATO, M. W. HARDING, D. J. LIVINGSTON, AND M. S. SU. Activation of interferongamma inducing factor mediated by interleukin-1 beta converting
enzyme. Science 275: 206–209, 1997.
GUTIERREZ, E. G., W. A. BANKS, AND A. J. KASTIN. Murine tumor necrosis factor alpha is transported from blood to brain in
the mouse. J. Neuroimmunol. 47: 169–176, 1993.
GUTIERREZ, E. G., W. A. BANKS, AND A. J. KASTIN. Blood-borne
interleukin-1 receptor antagonist crosses the blood-brain barrier.
J. Neuroimmunol. 55: 153–160, 1994.
GWOSDOW, A. R., M. S. A. KUMAR, AND H. H. BODE. Interleukin1 stimulation of the hypothalamo-pituitary-adrenal axis. Am. J.
Physiol. 258 (Endocrinol. Metab. 21): E65–E70, 1990.
GWOSDOW, A. R., N. A. O’CONNELL, J. A. SPENCER, M. S. KUMAR, R. K. AGARWAL, H. H. BODE, AND A. B. ABOU-SAMRA.
Interleukin-1 induced corticosterone release occurs by an adrenergic mechanism from rat adrenal gland. Am. J. Physiol. 263 (Endocrinol. Metab. 26): E461–E466, 1992.
GWOSDOW, A. R., J. A. SPENCER, N. A. O’CONNELL, AND A. B.
ABOU-SAMRA. Interleukin-1 activates protein kinase A and stimulates adrenocorticotropic hormone release from AtT20 cells. Endocrinology 132: 710–714, 1993.
HACHMAN, M., N. CRISTAL, R. M. WHITE, S. SEGAL, AND R. N.
APTE. Complementary organ expression of IL-1 vs. IL-6 and CSF1 activities in normal and LPS-injected mice. Cytokine 8: 21–31,
1996.
HAGAN, P., S. POOLE, AND A. F. BRISTOW. Endotoxin-stimulated
production of rat hypothalamic interleukin-1b in vivo and in vitro,
measured by specific immunoradiometric assay. J. Mol. Endocrinol. 11: 31–36, 1993.
HAGAN, P., F. TILDERS, S. POOLE, AND A. F. BRISTOW. Development of a two-site immunoradiometric assay for rat/human corticotropin-releasing factor. Application to the measurement of interleukin-1b-induced production of hypothalamic CRF in vitro. J.
Immunol. Methods 160: 11–18, 1993.
HANISCH, U.-K., S. LYONS, F. KIRCHOFF, C. NOLTE, AND H.
KETTENMANN. Expression of a functional interleukin-15 receptor complex by microglial cells in culture (Abstract). Proc. Annu.
Meet. Soc. Neurosci. 26th Washington DC 1996, p. 336.15.
HANISCH, U. K., W. ROWE, S. SHARMA, M. J. MEANEY, AND R.
QUIRION. Hypothalamic-pituitary-adrenal activity during chronic
central administration of interleukin-2. Endocrinology 135: 2465–
2472, 1994.
HANNUM, C. H., C. J. WILCOX, W. P. AREND, F. G. JOSLIN, D. J.
DRIPPS, P. M. HEIMDAL, L. G. ARMES, A. SOMMER, S. P. EISENBERG, AND R. C. THOMPSON. Interleukin-1 receptor antagonist
activity of a human interleukin-1 inhibitor. Nature 343: 336–340,
1990.
HANSEN, M. K., AND J. M. KRUEGER. Subdiaphragmatic vagotomy blocks the sleep- and fever-promoting effects of interleukin1 beta. Am. J. Physiol. 273 (Regulatory Integrative Comp. Physiol. 42): R1246–R1253, 1997.
HANSEN, M. K., P. TAISHI, Z. CHEN, AND J. M. KRUEGER. Vagotomy blocks the induction of interleukin-1 beta (IL-1beta) mRNA
in the brain of rats in response to sytemic IL-1beta. J. Neurosci.
18: 2247–2253, 1998.
HAOUR, F., E. BAN, G. MILON, D. BAMAN, AND G. FILLION.
Brain interleukin-1 receptors: characterization and modulation
after lipopolysaccharide injection. Prog. Neuroendocrinol. Immunol. 3: 201–208, 1990.
HARBUZ, M. S., A. J. CHOVER-GONZALEZ, S. BISWAS, S. L.
LIGHTMAN, AND H. S. CHOWDREY. Role of central catecholamines in the modulation of corticotrophin-releasing factor mRNA
pra
APS-Phys Rev
Downloaded from on April 23, 2014
295.
ANDREW V. TURNBULL AND CATHERINE L. RIVIER
January 1999
323.
324.
325.
326.
327.
328.
329.
331.
332.
333.
334.
335.
336.
337.
338.
339.
340.
341.
during adjuvant-induced arthritis. Br. J. Rheumatol. 33: 205–209,
1994.
HARBUZ, M. S., G. L. CONDE, O. MARTI, S. L. LIGHTMAN, AND
D. S. JESSOP. The hypothalamic-pituitary-adrenal axis in autoimmunity. Ann. NY Acad. Sci. 823: 214–224, 1997.
HARBUZ, M. S., J. P. LEONARD, S. L. LIGHTMAN, AND M. L.
CUZNER. Changes in hypothalamic corticotrophin-releasing factor and anterior pituitary pro-opiomelanocortin mRNA during the
course of experimental allergic encephalomyelitis. J. Neuroimmunol. 45: 127–132, 1993.
HARBUZ, M. S., R. G. REES, D. ECKLAND, D. S. JESSOP, D.
BREWERTON, AND S. L. LIGHTMAN. Paradoxical responses of
hypothalamic corticotropin-releasing factor (CRF) messenger ribonucleic acid (mRNA) and CRF-41 peptide and adenohypophysial proopiomelanocortin mRNA during chronic inflammatory
stress. Endocrinology 130: 1394–1400, 1992.
HARBUZ, M. S., A. STEPHANOU, R. A. KNIGHT, A. J. CHOVERGONZALEZ, AND S. L. LIGHTMAN. Action of interleukin-2 and
interleukin-4 on CRF mRNA in the hypothalamus POMC mRNA
in the anterior pituitary. Brain Behav. Immun. 6: 214–222, 1992.
HARBUZ, M. S., A. STEPHANOU, N. SARLIS, AND S. L.
LIGHTMAN. The effects of recombinant human interleukin (IL)1a, IL-1b or IL-6 on hypothalamo-pituitary-adrenal axis activation.
J. Endocrinol. 133: 349–355, 1992.
HARLIN, C. A., AND C. R. PARKER. Investigation of the effect of
interleukin-1 beta on steroidogenesis in the human fetal adrenal
gland. Steroids 56: 72–76, 1991.
HASHIMOTO, K., T. NISHIOKA, C. TOJO, AND T. TAKAO. Nitric
oxide plays no role in ACTH release induced by interleukin-1b,
corticotropin-releasing hormone, arginine vasopressin and phorbol myristate acetate in rat pituitary cell cultures. Endocr. J. 42:
435–439, 1995.
HASHIMOTO, M., Y. ISHIKAWA, S. YOKOTA, F. GOTO, F.
BANDO, Y. SAKAKIBARA, AND M. IRIKI. Action of circulating
interleukin-1 on the rabbit brain. Brain Res. 540: 217–223, 1991.
HEALY, D. L., G. D. HODGEN, H. M. SCHULTE, G. P. CHROUSOS,
D. L. LORIAUX, N. R. HALL, AND A. L. GOLDSTEIN. The thymusadrenal connection: thymosin has corticotropin-releasing activity
in primates. Science 222: 1353–1355, 1983.
HEANEY, M. L., AND D. W. GOLDE. Soluble cytokine receptors.
Blood 87: 847–857, 1996.
HELLE, M., J. P. BRAKENHOFF, E. R. DE GROOT, AND L. A.
AARDEN. Interleukin-6 is involved in interleukin 1-induced activities. Eur. J. Pharmacol. 18: 957–959, 1988.
HELLER, R. A., K. SONG, N. FAN, AND D. J. CHANG. The p70
tumor necrosis factor receptor mediates cytotoxicity. Cell 70: 47–
56, 1992.
HENCH, P. S., E. C. KENDALL, C. H. SLOCUMB, AND H. F. POLLEY. The effect of a hormone of the adrenal cortex (17-hydroxycorticosterone: compound E) and of pituitary adrenocorticotropic
hormone on rheumatoid arthritis. Mayo Clinic Proc. 24: 181–197,
1949.
HERMUS, R. M., G. C. SWEEP, M. J. VAN DER MEER, H. A. ROSS,
A. G. SMALS, T. J. BENRAAD, AND P. W. KLOPPENBORG. Continuous infusion of interleukin-1 beta induces a nonthyroidal illness
syndrome in the rat. Endocrinology 131: 2139–2146, 1992.
HERSEY, P., A. COATES, M. RALLINGS, C. HALL, M. MACDONALD, A. SPURLING, A. EDWARDS, W. H. MCCARTHY, AND G. W.
MILTON. Comparative study on the effects of recombinant alpha2 interferon on immune function in patients with disseminated
melanomas. J. Biol. Response Modif. 5: 236–249, 1986.
HIGGINS, G. A., AND J. A. OLSCHOWKA. Induction of interleukin1b mRNA in adult rat brain. Brain Res. Mol. Brain Res. 9: 143–
148, 1991.
HILLHOUSE, E. W. Interleukin-2 stimulates the secretion of arginine vasopressin but not corticotropin-releasing hormone from
rat hypothalamic cells in vitro. Brain Res. 650: 323–325, 1994.
HILLHOUSE, E. W., AND K. MOSLEY. Peripheral endotoxin induces hypothalamic immunoreactive interleukin-1 beta in the rat.
Br. J. Pharmacol. 109: 289–290, 1993.
HO, M. M., AND G. P. VINSON. Endocrine control of the distribution of basic fibroblast growth factor, insulin-like growth factor
and transforming growth factor-b1 mRNAs in adult rat adrenals
/ 9j0c$$oc11
P13-8
342.
343.
344.
345.
346.
347.
348.
349.
350.
351.
352.
353.
354.
355.
356.
357.
358.
359.
360.
11-25-98 11:16:36
53
using non-radioactive in situ hybridization. J. Endocrinol. 144:
379–387, 1995.
HOLSBOER, F., G. K. STALLA, U. VON BARDELEBEN, K. HAMMANN, H. MULLER, AND O. A. MULLER. Acute adrenocortical
stimulation by recombinant gamma interferon in human controls.
Life Sci. 42: 1–5, 1988.
HOPKINS, S. J. Cytokines and their significance in rheumatic disease. In: Anti-rheumatic Drugs, edited by M. C. L. E. Orme. New
York: Pergamon, 1990, p. 49–118.
HOPKINS, S. J., AND N. J. ROTHWELL. Cytokines and the nervous
system I: expression and regulation. Trends Neurosci. 18: 83–88,
1995.
HORAN, M. A., R. A. LITTLE, N. J. ROTHWELL, AND P. J. L. M.
STRIJBOS. Comparison of the effects of several endotoxin preparations on body temperature and metabolic rate in the rat. Can.
J. Physiol. Pharmacol. 67: 1011–1014, 1989.
HU, S.-B., L. A. TANNAHILL, AND S. L. LIGHTMAN. Interleukin1b induces corticotropin-releasing factor-41 release from cultured
hypothalamic cells through protein kinase C and cAMP-dependent
protein kinase pathways. J. Neuroimmunol. 40: 49–56, 1992.
HU, Y., H. DIETRICH, M. HEROLD, P. C. HEINRICH, AND G. WICK.
Disturbed immuno-endocrine communication via the hypothalamo-pituitary-adrenal axis in autoimmune disease. Int. Arch. Allergy Immunol. 102: 232–241, 1993.
HUANG, J., X. GAO, S. LI, AND Z. CAO. Recruitment of IRAK to
the interleukin-1 receptor complex requires interleukin 1 receptor
accessory protein. Proc. Natl. Acad. Sci. USA 94: 12829–12832,
1997.
HUETTNER, C., W. PAULUS, AND W. ROGGENDORF. Messenger
RNA expression of the immunosuppressive cytokine IL-10 in human gliomas. Am. J. Pathol. 146: 317–322, 1995.
HUGHES, T. K., P. CADET, P. L. RADY, S. K. TYRING, R. CHIN,
AND E. M. SMITH. Evidence for the production and action of interleukin-10 in pituitary cells. Cell. Mol. Neurobiol. 14: 59–69,
1994.
HUNTER, C. A., J. TIMANS, P. PISACANE, S. MENON, G. CAI,
W. WALKER, M. ASTE-AMEZAGA, R. CHIZZONITE, J. F. BAZAN,
AND R. A. KASTELEIN. Comparison of the effects of interleukin1 alpha, interleukin-1 beta and interferon-gamma-inducing factor
on the production of interferon-gamma by natural killer. Eur. J.
Immunol. 27: 2787–2792, 1997.
ILYIN, S. E., AND C. R. PLATA-SALAMAN. An approach to study
molecular mechanisms involved in cytokine-induced anorexia. J.
Neurosci. Methods 70: 33–38, 1996.
ILYIN, S. E., AND C. R. PLATA-SALAMAN. In vivo regulation of
the IL-1b system (ligand, receptors I and II, receptor accessory
protein and receptor antagonist) and TNF-a mRNAs in specific
brain regions. Biochem. Biophys. Res. Commun. 227: 861–867,
1996.
ILYIN, S. E., AND C. R. PLATA-SALAMAN. Molecular regulation of
the brain interleukin-1 beta system in obese (fa/fa) and lean (Fa/
Fa) Zucker rats. Brain Res. Mol. Brain Res. 43: 209–218, 1996.
ILYIN, S. E., AND C. R. PLATA-SALAMAN. HIV-1 envelope glycoprotein gp120 regulates brain IL-1 beta system and TNF-alpha
mRNAs in vivo. Brain Res. Bull. 44: 67–73, 1997.
ILYIN, S. E., AND C. R. PLATA-SALAMAN. HIV-1 gp120 glycoprotein modulates cytokine mRNAs in vivo: implications to cytokine
feedback systems. Biochem. Biophys. Res. Commun. 231: 514–
518, 1997.
ILYIN, S. E., G. SONTI, D. GAYLE, AND C. R. PLATA-SALAMAN.
Regulation of brain interleukin-1b (IL-1b) system mRNAs in response to pathophysiological concentrations of IL-1b in the cerebrospinal fluid. J. Mol. Neurosci. 7: 169–181, 1996.
IMURA, H., J.-I. FUKATA, AND T. MORI. Cytokines and endocrine
function: an interaction between the immune and neuroendocrine
systems. Clin. Endocrinol. 35: 107–115, 1991.
ISHIZUKA, Y., Y. ISHIDA, T. KUNITAKE, K. KATO, T. HANAMORI,
Y. MITSUYAMA, AND H. KANNAN. Effects of area postrema lesion
and abdominal vagotomy on interleukin-1b-induced norepinephrine release in the hypothalamic paraventricular nucleus region
in the rat. Neurosci. Lett. 223: 57–60, 1997.
IWATA, H., A. MATSUYAMA, N. OKUMURA, S. YOSHIDA, Y. LEE,
K. IMAIZUMI, AND S. SHIOSAKA. Localization of basic FGF-like
pra
APS-Phys Rev
Downloaded from on April 23, 2014
330.
REGULATION OF HPA AXIS BY CYTOKINES
54
361.
362.
363.
364.
365.
366.
367.
368.
370.
371.
372.
373.
374.
375.
376.
377.
378.
379.
380.
immunoreactivity in the hypothalamo-hypophyseal neuroendocrine axis. Brain Res. 550: 329–332, 1991.
JAATTELA, M., O. CARPEN, U. H. STENMAN, AND E. SAKSELA.
Regulation of ACTH-induced steroidogenesis in human fetal adrenal glands by rTNF-alpha. Mol. Cell. Endocrinol. 68: R31–R36,
1990.
JAATTELA, M., V. ILVESMAKI, R. VOUTILAINEN, U.-H. STENMAN, AND E. SAKSELA. Tumor necrosis factor as a potent inhibitor of adrenocorticotropin-induced cortisol production and steroidogenic P450 enzyme gene expression in cultured human fetal
adrenal cells. Endocrinology 128: 623–629, 1991.
JAFFE, H. L. The influence of the suprarenal gland on the thymus.
III. Stimulation of the growth of the thymus gland following double
suprarenalectomy in young rats. J. Exp. Med. 40: 753–760, 1924.
JANICKI, P. K. Binding of human alpha-interferon in the brain
tissue membranes of rat. Res. Commun. Chem. Pathol. Pharmacol. 75: 117–120, 1992.
JANSKY, L., S. VYBIRAL, D. POPOSOLOVA, J. ROTH, J. DORNAND, E. ZEISBERGER, AND J. KAMINKOVA. Production of systemic and hypothalamic cytokines during the early phase of endotoxin fever. Neuroendocrinology 62: 55–61, 1995.
JESSOP, D. S., AND S. L. LIGHTMAN. Priming of the anterior pituitary with corticotropin-releasing hormone in vitro does not facilitate an ACTH response to interleukin-1b. Immunol. Lett. 41: 225–
228, 1994.
JOHANSSON, A., T. OLSSON, B. CALBERG, K. KARLSSON, AND
M. FAGERLUND. Hypercortisolism after stroke: partly cytokine
mediated? J. Neurol. Sci. 147: 43–47, 1997.
JOHANSSON, B. B. The blood-brain barrier and perivascular cells.
In: Immune Responses in the Nervous System, edited by N. J.
Rothwell. Oxford, UK: Bios Scientific, 1995, p. 1–26.
JOHNSON, A. K., AND P. M. GROSS. Sensory circumventricular
organs and brain homeostatic pathways. FASEB J. 7: 678–686,
1993.
JOHNSON, H. M., E. M. SMITH, B. A. TORRES, AND J. E. BLALOCK. Regulation of the in vitro antibody response by neuroendocrine hormones. Proc. Natl. Acad. Sci. USA 79: 4171–4174, 1982.
JOHNSON, H. M., B. A. TORRES, E. M. SMITH, L. D. DION, AND
J. E. BLALOCK. Regulation of lymphokine (gamma-interferon)
production by corticotropin. J. Immunol. 132: 246–250, 1984.
JOHNSON, R. W., S. ARKINS, R. DANTZER, AND K. W. KELLEY.
Hormones, lymphohemopoietic cytokines and the neuroimmune
axis. Comp. Biochem. Physiol. A Physiol. 116: 183–201, 1997.
JOHNSON, R. W., G. GHEUSI, S. SEGRETI, R. DANTZER, AND
K. W. KELLEY. C3H/HeJ mice are refractory to lipopolysaccharide
in the brain. Brain Res. 752: 219–226, 1997.
JONES, D. A., D. P. CARLTON, T. M. MCINTYRE, G. A. ZIMMERMAN, AND S. M. PRESCOTT. Molecular cloning of human prostaglandin endoperoxide synthase type II and demonstration of expression in response to cytokines. J. Biol. Chem. 268: 9049–9054,
1993.
JONES, T. H., M. DANIELS, R. A. JAMES, S. K. JUSTICE, R.
MCCORKLE, A. PRICE, P. KENDALL-TAYLOR, AND A. P. WEETMAN. Production of bioactive and immunoactive IL-6 and expression of IL-6 mRNA by human pituitary adenomas. J. Clin. Endocrinol. Metab. 78: 180–187, 1994.
JONES, T. H., S. K. JUSTICE, A. PRICE, AND K. CHAPMAN. Interleukin-6 secreting human pituitary adenomas in vitro. J. Clin.
Endocrinol. Metab. 73: 207–209, 1991.
JONES, T. H., R. L. KENNEDY, S. K. JUSTICE, AND A. PRICE. Interleukin-1 stimulates the release of interleukin-6 from cultured
human pituitary adenoma cells. Acta Endocrinol. 128: 405–410,
1993.
JONES, T. H., R. L. KENNEDY, S. K. JUSTICE, AND A. PRICE. Pituitary adenomas with high and low basal inositol phospholipid
turnover; the stimulatory effect of kinins and an association with
interleukin-6 secretion. Clin. Endocrinol. 39: 433–439, 1993.
JORDAN, M., I. G. OTTERNESS, R. NG, A. GESSNER, M. ROLLINGHOFF, AND H. U. BEUSCHER. Neutralization of endogenous
IL-6 supresses induction of IL-1 receptor antagonist. J. Immunol.
154: 4081–4090, 1995.
JOSEPH, J., J. L. GRUN, F. D. LUBLIN, AND R. L. KNOBLER. Cytokine induction in vitro in mouse brain endothelial cells and astro-
/ 9j0c$$oc11
P13-8
381.
382.
383.
384.
385.
386.
387.
388.
389.
390.
391.
392.
393.
394.
395.
396.
397.
398.
399.
11-25-98 11:16:36
Volume 79
cytes by exposure to mouse hepatitis virus (MHV-4, JHM). Adv.
Exp. Med. Biol. 342: 443–448, 1993.
JU, G., X. ZHANG, B.-Q. JIN, AND C.-S. HUANG. Activation of
corticotropin-releasing factor-containing neurons in the paraventricular nucleus of the hypothalamus by interleukin-1 in the rat.
Neurosci. Lett. 132: 151–154, 1991.
JUDD, A. M., AND R. M. MACLEOD. Angiotensin II increases interleukin-6 release from rat adrenal glomerulosa cells. Prog. Neuroendocrinol. Immunol. 4: 240–247, 1991.
JUDD, A. M., AND R. M. MACLEOD. Adrenocorticotropin increases
interleukin-6 release from rat adrenal zona glomerulosa cells. Endocrinology 130: 1245–1254, 1992.
JUDD, A. M., AND R. M. MACLEOD. Differential release of tumor
necrosis factor and IL-6 from adrenal zona glomerulosa cells in
vitro. Am. J. Physiol. 268 (Endocrinol. Metab. 31): E114–E120,
1995.
JUDD, A. M., B. L. SPANGELO, AND R. M. MACLEOD. Rat adrenal
zone glomerulosa cells produce interleukin-6. Prog. Neuroendocrinol. Immunol. 3: 282–292, 1990.
KABIERSCH, A., A. DEL REY, C. G. HONEGGER, AND H. O.
BESEDOVSKY. Interleukin-1 induces changes in norepinepherine
metabolism in the rat brain. Brain Behav. Immun. 2: 267–274,
1988.
KAKUCSKA, I., Y. QI, B. D. CLARK, AND R. M. LECHAN. Endotoxin-induced corticotropin-releasing hormone gene expression
in the hypothalamic paraventricular nucleus is mediated centrally
by interleukin-1. Endocrinology 133: 815–821, 1993.
KANEKO, M., K. KANEKO, J. SHINSAKO, AND M. F. DALLMAN.
Adrenal sensitivity to adrenocorticotropin varies diurnally. Endocrinology 109: 70–75, 1981.
KAPAS, L., M. K. HANSEN, H. Y. CHANG, AND J. M. KRUEGER.
Vagotomy attenuates but does not prevent the somnogenic and
febrile effects of lipopolysaccharide in rats. Am. J. Physiol. 274
(Regulatory Integrative Comp. Physiol. 42): R406–R411, 1998.
KAPCALA, L. P., J. R. HE, Y. GAO, J. O. PIEPER, AND L. J. DETOLLA. Subdiaphragmatic vagotomy inhibits intra-abdominal interleukin-1b stimulation of adrenocorticotropin secretion. Brain
Res. 728: 247–254, 1996.
KAR, S., J. G. CHABOT, AND R. QUIRION. Quantitative autoradiographic localization of [125I]insulin-like growth factor I, [125I]insulin-like growth factor II, and [125I]insulin receptor binding sites
in developing and adult rat brain. J. Comp. Neurol. 333: 375–397,
1993.
KARANTH, S., K. LYSON, M. C. AGUILA, AND S. M. MCCANN.
Effects of luteinizing-hormone-releasing hormone, a-melanocytestimulating hormone, naloxone, dexamethasone and indomethacin on interleukin-2-induced corticotropin-releasing factor release. Neuroimmunomodulation 2: 166–173, 1995.
KARANTH, S., K. LYSON, AND S. M. MCCANN. Role of nitric oxide
in interleukin 2-induced corticotropin-releasing factor release
from incubated hypothalami. Proc. Natl. Acad. Sci. USA 90: 3383–
3387, 1993.
KARANTH, S., K. LYSON, AND S. M. MCCANN. Cyclosporin A inhibits interleukin-2-induced release of corticotropin-releasing hormone. Neuroimmunomodulation 1: 82–85, 1994.
KARANTH, S., AND S. M. MCCANN. Anterior pituitary hormone
control by interleukin-2. Proc. Natl. Acad. Sci. USA 88: 2961–2965,
1991.
KATOH-SEMBA, R., Y. KAISHO, A. SHINTANI, M. NAGAHAMA,
AND K. KATO. Tissue distribution and immunocytochemical localization of neurotrophin-3 in the brain and peripheral tissues of
rats. J. Neurochem. 66: 330–337, 1996.
KATSUURA, G., A. ARIMURA, K. KOVES, AND P. E.
GOTTSCHALL. Involvement of organum vasculosum of lamina
terminalis and pre-optic area in interleukin-1 beta-induced ACTH
release. Am. J. Physiol. 258 (Endocrinol. Metab. 21): E163–E171,
1990.
KATSUURA, G., P. E. GOTTSCHALL, AND A. ARIMURA. Identification of a high affinity receptor for interleukin-1 beta in rat brain.
Biochem. Biophys. Res. Commun. 156: 61–67, 1988.
KATSUURA, G., P. E. GOTTSCHALL, R. R. DAHL, AND A. ARIMURA. Adrenocorticotropin release induced by intracerebroventricular injection of recombinant human interleukin-1 in rats: pos-
pra
APS-Phys Rev
Downloaded from on April 23, 2014
369.
ANDREW V. TURNBULL AND CATHERINE L. RIVIER
January 1999
400.
401.
402.
403.
404.
405.
406.
407.
409.
410.
411.
412.
413.
414.
415.
416.
417.
418.
419.
sible involvement of prostaglandin. Endocrinology 122: 1773–
1779, 1988.
KAWASAKI, M., Y. YOSHIHARA, M. YAMAJI, AND Y. WATANABE.
Expression of prostaglandin endoperoxide synthase in rat brain.
Brain Res. Mol. Brain Res. 19: 39–46, 1993.
KEANE, K. M., D. A. GIEGEL, W. J. LIPINSKI, M. J. CALLAHAN,
AND B. D. SHIVERS. Cloning, tissue expression and regulation of
rat interleukin 1b converting enzyme. Cytokine 7: 105–110, 1995.
KEHRER, P., D. TURNILL, J.-M. DAYER, A. F. MULLER, AND R. C.
GAILLARD. Human recombinant interleukin-1a and -b, but not
recombinant tumor necrosis factor a stimulate ACTH release from
rat anterior pituitary cells in vitro in a prostaglandin E2 and cAMP
independent manner. Neuroendocrinology 48: 160–166, 1988.
KEIFER, R., AND G. W. KREUTZBERG. Gamma interferon-like immunoreactivity in the rat nervous system. Neuroscience 37: 725–
734, 1990.
KELLER-WOOD, M. E., AND M. F. DALLMAN. Corticosteroid inhibition of ACTH secretion. Endocr. Rev. 5: 1–24, 1984.
KELLER-WOOD, M. E., J. SHINSAKO, AND M. F. DALLMAN. Integral as well as proportional adrenal responses to ACTH. Am. J.
Physiol. 245 (Regulatory Integrative Comp. Physiol. 14): R53–
R59, 1983.
KENNEDY, B. P., C.-C. CHAN, S. A. CULP, AND W. A. CROMLISH.
Cloning and expression of rat prostaglandin endoperoxide synthase (cyclooxygenase)-2 cDNA. Biochem. Biophys. Res. Commun. 197: 494–500, 1993.
KENNEDY, M. K., D. S. TORRANCE, K. S. PICHA, AND K. M.
MOHLER. Analysis of cytokine mRNA expression in the central
nervous sytem of mice with experimental autoimmune encephalomyelitis reveals that IL-10 mRNA expression correlates with recovery. J. Immunol. 149: 2496–2505, 1992.
KENT, S., R. M. BLUTHE, K. W. KELLEY, AND R. DANTZER. Sickness behavior as a new target for drug development. Trends Pharmacol. Sci. 13: 24–28, 1992.
KENT, S., J. L. BRET-DIBAT, K. W. KELLEY, AND R. DANTZER.
Mechanisms of sickness-induced decreases in food-motivated behavior. Neurosci. Biobehav. Rev. 20: 171–175, 1996.
KERAMIDAS, M., J. J. BOURGARIAT, E. TABONE, P. CORTICELLI, E. M. CHAMBAZ, AND J. J. FEIGE. Immunolocalization of
transforming growth factor-beta 1 in the bovine adrenal cortex
using antipeptide antibodies. Endocrinology 129: 517–526, 1991.
KESHET, E., S. D. LYMAN, D. E. WILLIAMS, D. M. ANDERSON,
N. A. JENKINS, N. G. COPELAND, AND L. F. PARADA. Embryonic
RNA expression patterns of the c-kit receptor and its cognate
ligand suggest multiple functional roles in mouse development.
EMBO J. 10: 2425–2435, 1991.
KILBOURN, R. G., AND P. BELLONI. Endothelial cell production
of nitrogen oxides in response to interferon gamma in combination with tumor necrosis factor, interleukin-1, or endotoxin. J.
Natl. Cancer Inst. 82: 772–776, 1990.
KIM, K. S., C. A. WASS, A. S. CROSS, AND S. OPAL. Modulation
of blood-brain-barrier permeability by tumor necrosis factor an
antibody to tumor necrosis factor in the rat. Lymphokine Cytokine Res. 11: 293–298, 1992.
KINOUCHI, K., G. BROWN, G. PASTERNAK, AND B. DONNER.
Identification and characterization of receptors for tumor necrosis
factor-alpha in the brain. Biochem. Biophys. Res. Commun. 18:
1532–1538, 1991.
KISHIMOTO, T., S. AKIRA, M. NARAZAKI, AND T. TAGA. Interleukin-6 family of cytokines and gp130. Blood 86: 1243–1254, 1995.
KISHIMOTO, T., T. TAGA, AND S. AKIRA. Cytokine signal transduction. Cell 76: 253–262, 1994.
KITAMURA, H., A. KONNO, M. MORIMATSU, B. D. JUNG, K. KIMURA, AND M. SAITO. Immobilization stress increases hepatic IL6 expression in mice. Biochem. Biophys. Res. Commun. 238: 707–
711, 1997.
KLIR, J. J., J. L. MCCLELLAN, AND M. J. KLUGER. Interleukin-1b
causes the increase in anterior hypothalamic interleukin-6 during
LPS-induced fever in rats. Am. J. Physiol. 266 (Regulatory Integrative Comp. Physiol. 35): R1845–R1848, 1994.
KLIR, J. J., J. ROTH, Z. SZELENYI, J. L. MCCLELLAN, AND M. J.
KLUGER. Role of hypothalamic interleukin-6 and tumor necrosis
/ 9j0c$$oc11
P13-8
420.
421.
422.
423.
424.
425.
426.
427.
428.
429.
430.
431.
432.
433.
434.
435.
436.
437.
11-25-98 11:16:36
55
factor-b in LPS fever in rat. Am. J. Physiol. 265 (Regulatory Integrative Comp. Physiol. 34): R512–R517, 1993.
KLUGER, M. J. Fever: role of pyrogens and cryogens. Physiol.
Rev. 71: 93–127, 1991.
KNIGGE, U., A. KJAER, H. JORGENSEN, M. GARBARG, C. ROSS,
A. ROULEAU, AND J. WARBERG. Role of hypothalamic histaminergic neurons in mediation of ACTH and beta-endorphin responses to LPS endotoxin in vivo. Neuroendocrinology 60: 243–
251, 1994.
KOBAYASHI, H., J. FUKATA, N. MURAKAMI, T. USUI, O. EBISUI,
S. MURO, I. HANAOKA, K. INOUE, H. IMURA, AND K. NAKAO.
Tumor necrosis factor receptors in the pituitary cells. Brain Res.
758: 45–50, 1997.
KOBAYASHI, H., J. FUKATA, T. TOMINAGA, N. MURAKAMI, M.
FUKUSHIMA, O. EBISUI, H. SEGAWA, Y. NAKAO, AND H. IMURA.
Regulation of interleukin-1 receptors on AtT-20 mouse pituitary
tumour cells. FEBS Lett. 298: 100–104, 1992.
KOENIG, J. I., K. SNOW, B. D. CLARK, R. TONI, J. G. CANNO,
A. R. SHAW, C. A. DINARELLO, S. REICHLEIN, S. L. LEE, AND
R. M. LECHAN. Intrinsic pituitary interleukin-1b is induced by
bacterial lipopolysaccharide. Endocrinology 126: 3053–3058,
1990.
KOH, S., G. A. OYLER, AND G. A. HIGGINS. Localization of nerve
growth factor receptor messenger RNA and protein in the adult
rat brain. Exp. Neurol. 106: 209–221, 1989.
KOIKE, K., Y. SAKAMOTO, T. SAWADA, M. OHMICHI, Y. KANDA,
A. NOHARA, K. HIROTA, H. KIYAMA, AND A. MIYAKE. The production of CINC/gro, a member of the interleukin-8 family, in rat
anterior pituitary gland. Biochem. Biophys. Res. Commun. 202:
161–167, 1994.
KOIV, L., E. MERISALU, K. ZILMER, T. TOMBERG, AND A. E.
KAASIK. Changes of sympatho-adrenal and hypothalamo-pituitary-adrenocortical system in patients with head injury. Acta Neurol. Scand. 96: 52–58, 1997.
KOJIMA, H., M. TAKEUCHI, T. OHTA, Y. NISHIDA, N. ARAI, M.
IKEDA, H. IKEGAMI, AND M. KURIMOTO. Interleukin-18 activates
the IRAK-TRAF6 pathway in mouse EL-4 cells. Biochem. Biophys.
Res. Commun. 244: 183–186, 1998.
KOMAKI, G., A. ARIMURA, AND K. KOVES. Effect of intravenous
injection of IL-1b on PGE2 levels in several brain areas as determined by microdialysis. Am. J. Physiol. 262 (Endocrinol. Metab.
25): E246–E251, 1992.
KONISHI, Y., D. H. CHIU, T. KUNISHITA, T. YAMAMURA, Y. HIGASHI, AND T. TABIRA. Demonstration of interleukin-3 receptorassociated antigen in the central nervous system. J. Neurosci.
Res. 41: 572–582, 1995.
KONONEN, J., S. SOINILA, H. PERSSON, J. HONANIEMI, T. KOKFELT, AND M. PELTO-HUIKKO. Neurotrophins and their receptors
in the rat pituitary gland: regulation of BDNF and trkB mRNA
levels by adrenal hormones. Brain Res. Mol. Brain Res. 27: 347–
354, 1994.
KOPF, M., H. BAUMANN, G. FREER, M. FREUDENBERG, M.
LAMERS, T. KISHIMOTO, R. ZINKERNAGEL, H. BLUETHMANN,
AND G. KOHLER. Impaired immune and acute-phase responses in
interleukin-6 deficient mice. Nature 368: 339–342, 1994.
KORHERR, C., R. HOFMEISTER, H. WESCH, AND W. FALK. A
critical role for interleukin-1 receptor accessory protein in interleukin-1 signaling. Eur. J. Immunol. 27: 262–267, 1997.
KOVACS, K. J., AND I. J. ELENKOV. Differential dependence of
ACTH secretion induced by various cytokines on the integrity of
the paraventricular nucleus. J. Neuroendocrinol. 7: 15–23, 1995.
KOVACS, K. J., A. FOLDES, AND E. S. VIZI. C-kit ligand (Stem Cell
Factor) affects neuronal activity, stimulates pituitary-adrenal axis
and prolactin secretion. J. Neuroimmunol. 65: 133–141, 1996.
KOVACS, K. J., AND P. E. SAWCHENKO. Sequence of stress-induced alterations in indices of synaptic and transcriptional activation in parvocellular neurosecretory neurons. J. Neurosci. 16:
262–273, 1996.
KOZAK, W., V. POLI, D. SOSZYNSKI, C. A. CONN, L. R. LEON, AND
M. J. KLUGER. Sickness behavior in mice deficient in interleukin6 during turpentine abscess and influenza pneumonitis. Am. J.
Physiol. 272 (Regulatory Integrative Comp. Physiol. 41): R621–
R630, 1997.
pra
APS-Phys Rev
Downloaded from on April 23, 2014
408.
REGULATION OF HPA AXIS BY CYTOKINES
56
ANDREW V. TURNBULL AND CATHERINE L. RIVIER
/ 9j0c$$oc11
P13-8
458.
459.
460.
461.
462.
463.
464.
465.
466.
467.
468.
469.
470.
471.
472.
473.
474.
475.
11-25-98 11:16:36
in rats injected with interleukin-1 systemically or into the brain
ventricles. J. Neuroendocrinol. 6: 217–224, 1994.
LEE, S., AND C. RIVIER. Prenatal alcohol exposure alters the hypothalamic-pituitary-adrenal axis response of immature offspring to
interleukin-1: is nitric oxide involved? Alcohol. Clin. Exp. Res. 18:
1242–1247, 1994.
LEE, S., AND C. RIVIER. The intracerebroventricular injection of
corticotropin-releasing factor or interleukin-1b increases mRNA
levels of constitutive nitric oxide synthase in the rat hypothalamus
(Abstract). Proc. Annu. Meet. Soc. Neurosci. 26th Washington
DC 1996, p. 336.
LEE, Y. B., J. SATOH, D. G. WALKER, AND S. U. KIM. Interleukin15 gene expression in human astrocytes and microglia in culture.
Neuroreport 7: 1062–1066, 1996.
LEMAY, L. G., I. G. OTTERNESS, A. J. VANDER, AND M. J.
KLUGER. In vivo evidence that the rise in plasma IL-6 following
injection of a fever-inducing dose of LPS is mediated by IL-1b.
Cytokine 2: 199–204, 1990.
LEMAY, L. G., A. J. VANDER, AND M. J. KLUGER. The effects of
psychological stress on plasma interleukin-6 activity in rats. Physiol. Behav. 47: 957–961, 1990.
LEON, L. R., M. GLACCUM, AND M. J. KLUGER. The IL-1 type I
receptor mediates the acute phase response to turpentine, but not
LPS, in mice. Am. J. Physiol. 271 (Regulatory Integrative Comp.
Physiol. 40): R1668–R1674, 1996.
LEON, S., R. S. CARROL, K. DASHNER, D. GLOWACKA, AND P. M.
BLACK. Messenger ribonucleic acid expression of platelet-derived
growth factor subunits and receptors in pituitary adenomas. J.
Clin. Endocrinol. Metab. 79: 51–55, 1994.
LESNIAK, M. A., J. M. HILL, W. KEIUSS, M. ROJESKI, C. B. PERT,
AND J. ROTH. Receptors for insulin-like growth factors I and II:
autoradiographic localization in rat brain and comparison to receptors for insulin. Endocrinology 123: 2089–2099, 1988.
LEVINE, S., F. BERKENBOSCH, D. SUCHECKI, AND F. J. H. TILDERS. Pituitary-adrenal and interleukin-6 responses to recombinant
interleukin-1 in neonatal rats. Psychoneuroendocrinology 19: 143–
153, 1994.
LEWIS, M., L. A. TARTAGLIA, A. LEE, G. L. BENNET, G. C. RICE,
G. H. W. WONG, E. Y. CHEN, AND D. V. GOEDDEL. Cloning and
expression of cDNAs for two distinct murine tumor necrosis factor receptors demonstrate one receptor is species specific. Proc.
Natl. Acad. Sci. USA 88: 2830–2834, 1991.
LI, H.-Y., A. ERICSSON, AND P. E. SAWCHENKO. Distinct mechanisms underlie activation of hypothalamic neurosecretory neurons and their medullary catecholaminergic afferents in categorically different stress paradigms. Proc. Natl. Acad. Sci. USA 93:
2359–2364, 1996.
LI, P., H. ALLEN, S. BANERJEE, S. FRANKLIN, L. HERZOG, C.
JOHNSTON, J. MCDOWELL, M. PASKIND, L. RODMAN, J. SALFELD, E. TOWNE, D. TRACEY, S. WARDELL, F. Y. WEI, W.
WONG, R. KAMEN, AND T. SESHADRI. Mice deficient in IL-1bconverting enzyme are defective in production of mature IL-1b
and resistant to endotoxic shock. Cell 80: 401–411, 1995.
LIBERT, C., P. BROUCKAERT, A. SHAW, AND W. FIERS. Induction
of interleukin-6 by human and murine recombinant interleukin-1
in mice. Eur. J. Pharmacol. 20: 691–694, 1990.
LICINIO, J., AND M.-L. WONG. Interleukin 1 receptor antagonist
gene expression in rat pituitary in the systemic inflammatory response syndrome: pathological implications. Mol. Psychiatry 2:
99–103, 1997.
LICINIO, J., M.-L. WONG, AND P. W. GOLD. Localization of interleukin-1 receptor antagonist mRNA in rat brain. Endocrinology
1991: 562–564, 1991.
LICINIO, J., M.-L. WONG, AND P. W. GOLD. Neutrophil-activating
pepide-1/interleukin-8 mRNA is localized in rat hypothalamus and
hippocampus. Neuroreport 3: 753–756, 1992.
LIND, R. W., G. W. VAN HOESEN, AND A. K. JOHNSON. An HRP
study of the connections of the subfornical organ of the rat. J.
Comp. Neurol. 210: 265–277, 1982.
LINDOLM, D., E. CASTREN, M. BERZAGHI, A. BLOCHL, AND H.
THOENEN. Activity-dependent and hormonal regulation of neurotrophin mRNA levels in the brain: implications for neuronal plasticity. J. Neurobiol. 25: 1362–1372, 1994.
pra
APS-Phys Rev
Downloaded from on April 23, 2014
438. KOZAK, W., H. ZHENG, C. A. CONN, D. SOSZYNSKI, L. H. VAN
DER PLOEG, AND M. J. KLUGER. Thermal and behavioral effects
of lipopolysaccharide and influenza in interleukin-1 beta-deficient
mice. Am. J. Physiol. 269 (Regulatory Integrative Comp. Physiol.
38): R969–R977, 1995.
439. KRUEGER, J. M. Somnogenic activity of immune response modifiers. Trends Pharmacol. Sci. 11: 122–126, 1990.
440. KRUEGER, J. M., AND J. A. MAJDE. Cytokines and sleep. Int.
Arch. Allergy Immunol. 106: 97–100, 1995.
441. KUSHNER, I. The acute phase response: an overview. Methods
Enzymol. 163: 373–383, 1988.
442. LABOW, M., D. SHUSTER, M. ZETTERSTOM, P. NUNES, R.
TERRY, E. B. CULLINAN, T. BARTFAI, C. SOLORZANO, L. L.
MOLDAWER, R. CHIZZONITE, AND K. W. MCINTYRE. Absence of
IL-1 signalling and reduced inflammatory response in IL-1 type I
receptor-deficient mice. J. Immunol. 159: 2452–2461, 1997.
443. LACROIX, S., AND S. RIVEST. Functional circuitry in the brain of
immune-challenged rats: partial involvement of prostaglandins. J.
Comp. Neurol. 387: 307–324, 1997.
444. LACROIX, S., AND S. RIVEST. Effect of acute systemic inflammatory response and cytokines on the transcription of the genes
encoding cyclooxygenase enzymes (COX-1 and COX-2) in the rat
brain. J. Neurochem. 70: 452–466, 1998.
445. LACROIX, S., L. VALLIERES, AND S. RIVEST. c-Fos mRNA pattern
and CRF neuronal activity throughout the brain of rats injected
centrally with a prostaglandin of E2 type. J. Neuroimmunol. 70:
163–179, 1996.
446. LANDGRAF, R., I. NEUMANN, F. HOLSBOER, AND Q. J. PITTMAN. Interleukin-1b stimulates both central and peripheral release of vasopressin and oxytocin in the rat. Eur. J. Neurosci. 7:
592–598, 1995.
447. LANGHANS, W., R. HARLACHER, G. BALKOWSKI, AND E.
SCHARRER. Comparison of the effects of bacterial lipopolysaccharide and muramyl dipeptide on food intake. Physiol. Behav.
47: 805–813, 1990.
448. LAPCHAK, P. A., AND D. M. ARAUJO. Interleukin-2 regulates
monoamine and opioid peptide release from the hypothalamus.
Neuroreport 4: 303–306, 1993.
449. LASKIN, J. D., D. E. HECK, AND D. L. LASKIN. Multifunctional role
of nitric oxide in inflammation. Trends Endocrinol. Metab. 5: 377–
382, 1994.
450. LAVICKY, J., AND A. J. DUNN. Endotoxin administration stimulates cerebral catecholamine release in freely moving rats as assessed by microdialysis. J. Neurosci. Res. 40: 407–413, 1995.
451. LAYE, S., R. M. BLUTHE, S. KENT, C. COMBE, C. MEDINA, P.
PARNET, K. KELLEY, AND R. DANTZER. Subdiaphragmatic vagotomy blocks induction of IL-1 beta mRNA in mice brain in response
to peripheral LPS. Am. J. Physiol. 268 (Regulatory Integrative
Comp. Physiol. 37): R1327–R1331, 1995.
452. LAYE, S., E. GOUJON, C. COMBE, R. VANHOY, K. W. KELLY, P.
PARNET, AND R. DANTZER. Effects of lipopolysaccharide and
glucocorticoids on expression of interleukin-1 beta converting enzyme in the pituitary and brain of mice. J. Neuroimmunol. 68:
61–66, 1996.
453. LAYE, S., P. PARNET, E. GOUJON, AND R. DANTZER. Peripheral
administration of lipopolysaccharide induces the expression of
cytokine transcripts in the brain and pituitaries of mice. Brain
Res. Mol. Brain Res. 27: 157–162, 1994.
454. LECHAN, R. M., R. TONI, B. D. CLARK, J. G. CANNON, A. R.
SHAW, C. A. DINARELLO, AND S. REICHLIN. Immunoreactive interleukin-1b localization in the rat forebrain. Brain Res. 514: 135–
140, 1990.
455. LECHNER, O., Y. HU, M. JAFARIAN-TEHRANI, H. DIETRICH, S.
SCHWARZ, M. HEROLD, F. HAOUR, AND G. WICK. Disturbed
immunoendocrine communication via the hypothalamo-pituitaryadrenal axis in murine lupus. Brain Behav. Immun. 10: 337–350,
1996.
456. LEE, S., G. BARBANEL, AND C. RIVIER. Systemic endotoxin increases steady-state gene expression of hypothalamic nitric oxide
synthase: comparison with corticotropin-releasing factor and vasopressin gene transcripts. Brain Res. 705: 136–148, 1995.
457. LEE, S., AND C. RIVIER. Hypophysiotropic role and hypothalamic
gene expression of corticotropin-releasing factor and vasopressin
Volume 79
January 1999
REGULATION OF HPA AXIS BY CYTOKINES
/ 9j0c$$oc11
P13-8
494.
495.
496.
497.
498.
499.
500.
501.
502.
503.
504.
505.
506.
507.
508.
509.
510.
511.
11-25-98 11:16:36
COMBE, P. GHIARA, AND N. J. ROTHWELL. Importance of brain
IL-1 type II receptors in fever and thermogenesis in the rat. Am.
J. Physiol. 265 (Endocrinol. Metab. 28): E585–E591, 1993.
LUHESHI, G., A. J. MILLER, S. BROUWER, M. J. DASCOMBE,
N. J. ROTHWELL, AND S. J. HOPKINS. Interleukin-1 receptor antagonist inhibits endotoxin fever and systemic interleukin-6 induction in the rat. Am. J. Physiol. 270 (Endocrinol. Metab. 33): E91–
E95, 1995.
LUHESHI, G. N., A. STEFFERL, A. V. TURNBULL, M. J. DASCOMBE, S. BROUWER, S. J. HOPKINS, AND N. J. ROTHWELL.
Febrile response to tissue inflammation involves both peripheral
and brain IL-1 and TNF-a in the rat. Am. J. Physiol. 272 (Regulatory Integrative Comp. Physiol. 41): R862–R868, 1997.
LUSTIG, S., H. D. DANENBERG, Y. KAFRI, D. KOBILER, AND D.
BEN-NATHAN. Viral neuroinvasion and encephalitis induced by
lipopolysaccharide and its mediators. J. Exp. Med. 176: 707–712,
1992.
LYMANGROVER, J. R., AND A. BRODISH. Tissue CRF: an extrahypothalamic corticotropihin releasing factor (CRF) in peripheral
blood of stressed rats. Neuroendocrinology 12: 225–235, 1973.
LYSON, K., AND S. M. MCCANN. The effect of interleukin-6 on
pituitary hormone release in vivo and in vitro. Neuroendocrinology 54: 262–266, 1991.
LYSON, K., AND S. M. MCCANN. Induction of adrenocorticotropic
hormone release by interleukin-6 in vivo and in vitro. Ann. NY
Acad. Sci. 650: 182–185, 1992.
LYSON, K., AND S. M. MCCANN. Involvement of arachidonic cascade pathways in interleukin-6-stimulated corticotropin-releasing
factor release in vitro. Neuroendocrinology 55: 708–713, 1992.
LYSON, K., AND S. M. MCCANN. Alpha melanocyte-stimulating hormone abolishes IL-1 and IL-6-induced corticotropin-releasing factor release from the hypothalamus in vitro. Neuroendocrinology
58: 191–195, 1993.
MA, Y. J., K. BERG-VON DER EMDE, M. MOHOLT-SIEBERT, D. F.
HILL, AND S. R. OJEDA. Region-specific regulation of transforming
growth factor a (TGFa) gene expression in astrocytes of the neuroendocrine brain. J. Neurosci. 14: 5644–5651, 1994.
MA, Y. J., M. E. COSTA, AND S. R. OJEDA. Developmental expression of the genes encoding transforming growth factor alpha and
its receptor in the hypothalamus of female rhesus macaques. Neuroendocrinology 60: 346–359, 1994.
MA, Y. J., D. F. HILL, M. P. JUNIER, M. E. COSTA, S. E. FELDER,
AND S. R. OJEDA. Expression of epidermal growth factor receptor
changes in the hypothalamus during the onset of female puberty.
Mol. Cell. Neurosci. 5: 246–262, 1994.
MACKAY, F., J. ROTHE, H. BLUETHMANN, H. LOETSCHER, AND
W. LESSLAUER. Differential responses of fibroblasts from wildtype and TNF-R55-deficient mice to mouse and human TNF-a
activation. J. Immunol. 153: 5274–5284, 1994.
MACKIEWICZ, A., M. WIZNEROWICZ, E. ROEB, A. KARCZEWSKA, J. NOWAK, P. C. HEINRICH, AND S. ROSE-JOHN. Soluble
interleukin-6 receptor is biologically active in vivo. Cytokine 7:
142–149, 1995.
MACKIEWICZ, M., P. J. SOLLARS, M. D. OGILVIE, AND A. J. PACK.
Modulation of IL-1b gene expression in the rat CNS during sleep
deprivation. Neuroreport 7: 529–533, 1996.
MACLEOD, R. M., F. M. HUGHES, W. C. GOROSPE, AND B. L.
SPANGELO. Synthesis, release, and actions of interleukin 6 in
neuroendocrine tissues: methods and overview. Methods Neurosci. 17: 3–15, 1993.
MACMICKING, J. D., C. NATHAN, G. HOM, N. CHARTRAIN, D. S.
FLETCHER, M. TRUMBAUER, K. STEVENS, Q. W. XIE, K. SOKOL, AND N. HUTCHINSON. Altered response to bacterial infection and endotoxic shock in mice lacking inducible nitric oxide
synthase. Cell 81: 641–650, 1995.
MAES, M., E. BOSMANS, H. Y. MELTZER, S. SCHARPE, AND E.
SUY. Interleukin-1 beta: a putatuve mediator of HPA axis hyperactivity in major depression? Am. J. Psychiatry 150: 1189–1193,
1993.
MAIER, S. F., AND L. R. WATKINS. Intracerebroventricular interleukin-1 receptor antagonist blocks the enhancement of fear
conditioning and interference with escape produced by inescapable shock. Brain Res. 695: 279–282, 1995.
pra
APS-Phys Rev
Downloaded from on April 23, 2014
476. LING, C. R., AND M. A. FOSTER. Changes in NMR relaxation time
associated with local inflammatory response. Phys. Med. Biol. 27:
853–859, 1982.
477. LINTHORST, A. C. E., C. FLACHSKAMM, F. HOLSBOER, AND
J. M. H. M. REUL. Local administration of recombinant human interleukin-1b in the rat hippocampus increases serotonergic neurotransmission, hypothalamic-pituitary-adrenocortical axis activity
and body temperature. Endocrinology 135: 520–533, 1994.
478. LINTHORST, A. C. E., C. FLACHSKAMM, F. HOLSBOER, AND
J. M. H. M. REUL. Activation of serotonergic and adrenergic neurotransmission in the rat hippocampus after peripheral administration of bacterial endotoxin: involvement of cyclo-oxygenase
pathway. Neuroscience 72: 989–997, 1996.
479. LISSONI, P., P. BARNI, F. ROVELLI, S. CRISPINO, G. FUMAGALLI, S. PESCIA, M. VAGHI, G. CAMESASCA, AND G. TANCINI.
Neuroendocrine effects of subcutaneous interleukin-2 injection in
cancer patients. Tumor 77: 212–215, 1991.
480. LISSONI, P., S. BARNI, E. TISI, F. ROVELLI, S. PITTALIS, R. RESCALDANI, L. VIGORE, A. BIONDI, A. ARDIZZOIA, AND G. TANCINI. In vivo biological results of the association between interleukin-2 and interleukin-3 in the immunotherapy of cancer. Eur. J.
Cancer 29: 1127–1132, 1993.
481. LISSONI, P., F. ROVELLI, G. TANCINI, E. TISI, M. R. RIVOLTA,
A. ARDIZZOIA, AND F. BRIVIO. Inhibitory effect on interleukin-3
on interleukin-2-induced cortisol release in the immunotherapy of
cancer. J. Biol. Regul. Homeostasis Agents 6: 113–115, 1992.
482. LIU, C., D. CHALMERS, R. MAKI, AND E. B. DE SOUZA. Rat homolog of mouse interleukin-1 receptor accessory protein: cloning,
localization and modulation studies. J. Neuroimmunol. 66: 41–
48, 1996.
483. LIU, L., T. KITA, N. TANAKA, AND Y. KINOSHITA. The expression
of tumor necrosis factor in the hypothalamus after treatment with
lipopolysaccharide. Int. J. Exp. Pathol. 77: 37–44, 1996.
484. LJUNGDAHL, A., T. OLSSON, P. H. VAN DER MEIDE, R. HOLMDAHL, L. KLARESKOG, AND B. HOJEBERG. Interferon-gammalike immunoreactivity in certain neurons of the central and peripheral nervous system. J. Neurosci. Res. 24: 451–456, 1989.
485. LODDICK, S. A., M.-L. WONG, P. B. BONGIORNO, P. W. GOLD, J.
LICINIO, AND N. J. ROTHWELL. Endogenous interleukin-1 receptor antagonist is neuroprotective. Biochem. Biophys. Res. Commun. 234: 211–215, 1997.
486. LOGAN, A., AND M. BERRY. Transforming growth factor-b1 and
basic fibroblast growth factor in the injured CNS. Trends Pharmacol. Sci. 14: 337–343, 1993.
487. LONG, N. C., I. OTTERNESS, S. L. KUNKEL, A. J. VANDER, AND
M. J. KLUGER. The roles of interleukin-1b and tumor necrosis
factor in lipopolysaccharide fever in rats. Am. J. Physiol. 259
(Regulatory Integrative Comp. Physiol. 28): R724–R728, 1990.
488. LORD, K. A., B. HOFFMAN-LIEBERMANN, AND D. A. LIEBERMANN. Nucleotide sequence and expression of a cDNA encoding
MyD88, a novel myeloid differentiation primary response gene
induced by IL6. Oncogene 5: 1095–1097, 1990.
489. LOVENBERG, T. W., P. D. CROWE, C. LIU, D. T. CHALMERS,
X.-J. LIU, C. LIAW, W. CLEVENGER, T. OLTSERDORF, E. B.
DE SOUZA, AND R. A. MAKI. Cloning of a cDNA encoding a novel
interleukin-1 receptor related protein (IL-1R-rp2). J. Neuroimmunol. 70: 113–122, 1996.
490. LOWENTHAL, J. W., B. E. CASTLE, J. CHRISTIANSEN, J.
SCHREURS, D. RENNICK, N. ARAI, P. HOY, Y. TAKEBE, AND
M. HOWARD. Expression of high affinity receptors for murine
interleukin-4 (BSF-1) on hemopoietic and nonhemopoietic cells.
J. Immunol. 140: 456–464, 1988.
491. LOXLEY, H. D., A.-M. COWELL, R. J. FLOWER, AND J. C. BUCKINGHAM. Modulation of the hypothalamo-pituitary-adrenocortical
responses to cytokines in the rat by lipocortin 1 and glucocorticoids: a role for lipocortin 1 in the feedback inhibition of CRF-41
release? Neuroendocrinology 57: 810–814, 1993.
492. LUGER, A., A. E. CALOGERO, K. KALOGERAS, W. T. GALLUCCI,
P. W. GOLD, D. L. LORIAUX, AND G. P. CHROUSOS. Interaction of
epidermal growth factor with the hypothalamo-pituitary-adrenal
axis: potential physiologic relevance. J. Clin. Endocrinol. Metab.
66: 334–337, 1988.
493. LUHESHI, G., S. J. HOPKINS, R. A. LEFEUVRE, M. J. DAS-
57
58
ANDREW V. TURNBULL AND CATHERINE L. RIVIER
/ 9j0c$$oc11
P13-8
530.
531.
532.
533.
534.
535.
536.
537.
538.
539.
540.
541.
542.
543.
544.
545.
11-25-98 11:16:36
M. GIMENO, AND V. RETTORI. Induction by cytokines of the pattern of pituitary hormone secretion in infection. Neuroimmunomodulation 1: 2–13, 1994.
MCCOY, J. G., S. G. MATTA, AND B. M. SHARP. Prostaglandins
mediate the ACTH response to interleukin-1-beta instilled into
the hypothalamic median eminence. Neuroendocrinology 60: 426–
435, 1994.
MCEWAN, B. S., C. A. BIRON, K. W. BRUNSON, K. BULLOCH,
W. H. CHAMBERS, F. S. DHABHAR, R. H. GOLDFARB, R. P. KITSON, A. H. MILLER, R. L. SPENCER, AND J. M. WEISS. The role
of adrenocorticoids as modulators of immune function in health
and disease: neural, endocrine and immune interactions. Brain
Res. Rev. 23: 79–133, 1997.
MCGILLIS, J. P., N. R. HALL, G. V. VAHOUNY, AND A. L.
GOLDSTEIN. Thymosin fraction 5 causes increased serum corticosterone in rodents in vivo. J. Immunol. 134: 3952–3955, 1985.
MCKELVIE, P. A., K. M. ROSEN, H. C. KINNEY, AND L. VILLAKOMAROFF. Insulin-like growth factor II expression in the developing human brain. J. Neuropathol. Exp. Neurol. 51: 464–471,
1992.
MEFFORD, I. N., AND M. P. HEYES. Increased biogenic amine release in mouse hypothalamus following immunological challenge:
antagonism by indomethacin. J. Neuroimmunol. 27: 55–61, 1990.
MEGYERI, P., C. S. ABRAHA, P. TEMESVARI, J. KOVACS, T. VAS,
AND C. P. SPEER. Recombinant tumor necrosis factor a constricts
pial arterioles and increases blood-brain barrier permeability in
newborn piglets. Neurosci. Lett. 148: 137–140, 1992.
MEKAOUCHE, M., L. GIVALOIS, G. BARBANEL, P. SIAUD, D.
MAUREL, A. F. BRISTOW, J. BOISSIN, I. ASSENMACHER, AND
G. IXART. Chronic restraint enhances interleukin-1-beta release
in the basal state and after an endotoxin challenge, independently
of adrenocorticotropin and corticosterone release. Neuroimmunomodulation 1: 292–299, 1994.
MESIANO, S., S. H. MELLON, D. GOSPODAROWICZ, A. M. DI
BLASIO, AND R. B. JAFFE. Basic fibroblast growth factor expression is regulated by corticotropin in the human fetal adrenal: a
model for adrenal growth regulation. Proc. Natl. Acad. Sci. USA
88: 5428–5432, 1991.
MICHAELSON, M. D., M. F. MEHLER, H. XU, R. E. GROSS, AND
J. A. KESSLER. Interleukin-7 is trophic for embryonic neurons
and is expressed in developing brain. Dev. Biol. 179: 251–263,
1996.
MICHELSON, D., L. STONE, E. GALLIVEN, M. A. MAGIAKOU,
G. P. CHROUSOS, E. M. STERNBERG, AND P. W. GOLD. Multiple
sclerosis is associated with alterations in hypothalamic-pituitaryadrenal axis function. J. Clin. Endocrinol. Metab. 79: 848–853,
1994.
MILKENKOVIC, L., V. RETTORI, G. D. SNYDER, B. BEUTLER,
AND S. M. MCCANN. Cachectin alters anterior pituitary hormone
release by a direct action in vitro. Proc. Natl. Acad. Sci. USA 86:
2418–2422, 1989.
MILLER, A. J., S. J. HOPKINS, AND G. N. LUHESHI. Sites of action
of IL-1 in the development of fever and cytokine responses to
tissue inflammation in the rat. Br. J. Pharmacol. 120: 1274–1279,
1997.
MILLER, A. J., G. N. LUHESHI, N. J. ROTHWELL, AND S. J. HOPKINS. Local cytokine induction by LPS in the rat air pouch and
its relationship to the febrile response. Am. J. Physiol. 272 (Regulatory Integrative Comp. Physiol. 41): R857–R861, 1997.
MILLIGAN, E. D., M. M. MCGORRY, M. FLESHNER, R. P. GAYEKEMA, L. E. GOEHLER, L. R. WATKINS, AND S. F. MAIER. Subdiaphragmatic does not prevent fever following intracerebroventricular prostaglandin E2: further evidence for the importance of vagal
afferents in immune-to-brain communication. Brain Res. 766:
240–243, 1997.
MILTON, N. G., E. W. HILLHOUSE, AND A. S. MILTON. Activation
of the hypothalamo-pituitary-adrenocortical axis in the conscious
rabbit by the pyrogen polyinosinic: polycytidylic acid is dependent
on corticotropihin-releasing factor-41. J. Endocrinol. 135: 69–75,
1992.
MILTON, N. G., C. H. SELF, AND E. W. HILLHOUSE. Effects of
pyrogenic immunomodulators on the release of corticotropin-re-
pra
APS-Phys Rev
Downloaded from on April 23, 2014
512. MALARKEY, W. B., AND B. J. ZVARA. Interleukin-1b and other
cytokines stimulate adrenocorticotropin release from cultured pituitary cells of patients with Cushing’s disease. J. Clin. Endocrinol. Metab. 69: 196–199, 1989.
513. MALIPIERO, U. V., K. FREI, AND A. FONTANA. Production of
hemopoietic colony-stimulating factors by astrocytes. J. Immunol. 144: 3816–3821, 1990.
514. MANOVA, K., R. F. BACHVAROVA, E. J. HUANG, S. SANCHEZ,
S. M. PRONOVOST, E. VELAZQUEZ, B. MCGURIE, AND P. BESMER. c-kit receptor and ligand expression in postantal development of the mouse cerebellum suggests a function for c-kit in
inhibitory interneuons. J. Neurosci. 12: 4663–4676, 1992.
515. MARKS, J. L., D. PORTE, AND D. G. BASKIN. Localization of type
I insulin-like growth factor receptor messenger RNA in the adult
rat brain by in situ hybridization. Mol. Endocrinol. 5: 1158–1168,
1991.
516. MARQUETEE, C., A.-M. VAN DAM, E. CAN, P. LANIECE, M. CRUMEYROLLE-ARIAS, G. FILLION, F. BERKENBOSCH, AND F.
HAOUR. Rat interleukin-1b binding sites in rat hypothalamus and
pituitary gland. Neuroendocrinology 62: 362–369, 1995.
517. MARQUETTE, C., A.-M. VAN DAM, N. VAN ROOIJEN, F. BERKENBOSCH, AND F. HAOUR. Peripheral macrophage depletion
prevents downregulation of central interleukin-1 receptors in mice
after endotoxin administration. Psychoneuroendocrinology 19:
189–196, 1994.
518. MARTIN, F., AND J. BOYA. Immunocytochemical localization of
basic fibroblast growth factor in the human pituitary gland. Neuroendocrinology 62: 523–529, 1995.
519. MASTORAKOS, G., G. P. CHROUSOS, AND J. S. WEBER. Recombinant interleukin-6 activates the hypothalamic-pituitary-adrenal
axis in humans. J. Clin. Endocrinol. Metab. 77: 1690–1694, 1993.
520. MASTORAKOS, G., J. S. WEBER, M.-A. MAGIAKOU, H. GUNN,
AND G. P. CHROUSOS. Hypothalamic-pituitary-adrenal axis activation and stimulation of systemic vasopressin secretion by recombinant interleukin-6 in humans: potential implications for the syndrome of inappropriate vasopressin secretion. J. Clin. Endocrinol. Metab. 79: 934–939, 1994.
521. MATHEWS, L. S., AND W. W. VALE. Expression cloning of an activin receptor, a predicted transmembrane serine kinase. Cell 65:
973–982, 1991.
522. MATSUMOTO, H., C. KOYAMA, T. SAWADA, K. KOIKE, A. MIYAKE, A. ARIMURA, AND K. INOUE. Pituitary folliculo-stellate-like
cell line (TtT/GF) responds to novel hypophysiotropic peptide
(pituitary adenylate cyclase activating peptide), showing increased 3*,5*-monophosphate and interleukin-6 secretion and cell
proliferation. Endocrinology 133: 2150–2155, 1993.
523. MATSUMURA, K., Y. WATANABE, K. IMAI-MATSUMURA, M.
CONNOLLY, Y. KOYAMA, H. ONOE, AND Y. WATANABE. Mapping
of prostaglandin E2 binding sites in rat brain using quantitative
autoradiography. Brain Res. 581: 292–298, 1992.
524. MATSUMURA, K., Y. WATANABE, H. ONOE, Y. WATANABE, AND
O. HAYAISHI. High density of prostaglandin E2 binding sites in
the anterior wall of the 3rd ventricle: a possible site of its hyperthermic action. Brain Res. 533: 147–151, 1990.
525. MATTA, S. G., K. M. LINNER, AND B. M. SHARP. Interleukin-1b
and interleukin-1b stimulate adrenocorticotropin secretion in the
rat through a similar hypothalamic receptor(s): effects of interleukin-1 receptor antagonist protein. Neuroendocrinology 57: 14–22,
1993.
526. MATTA, S. G., J. SINGH, R. NEWTON, AND B. M. SHARP. The
adrenocorticotropin response to interleukin-1b instilled into the
rat median eminence depends on the local release of catecholamines. Endocrinology 127: 2175–2182, 1990.
527. MATTA, S. G., J. WEATHERBEE, AND B. M. SHARP. A central
mechanism is involved in the secretion of ACTH in response to
IL-6 in rats: comparison to and interaction with IL-1b. Neuroendocrinology 56: 516–525, 1992.
528. MAZZOCCHI, G., F. G. MUSAJO, L. K. MALENDOWICZ, P. G. ANDREIS, AND G. G. NUSSDORFER. Interleukin-1b stimulates corticotropin-releasing hormone (CRH) and adrenocorticotropin
(ACTH) release by rat adrenal gland in vitro. Mol. Cell. Neurosci.
79: 470–473, 1993.
529. MCCANN, S. M., S. KARANTH, A. KAMAT, W. L. DEES, K. LYSON,
Volume 79
January 1999
546.
547.
548.
549.
550.
551.
552.
554.
555.
556.
557.
558.
559.
560.
561.
562.
563.
leasing factor-41 and prostaglandin E2 from intact rat hypothalamus in vitro. Br. J. Pharmacol. 109: 88–93, 1993.
MINAMI, M., Y. KURAISHI, T. YAMAGUCHI, S. NAKAI, Y. HIRAI,
AND M. SATOH. Immobilization stress induces interleukin-1b
mRNA in the rat hypothalamus. Neurosci. Lett. 123: 254–256,
1991.
MINANO, F. J., A. FERNANDEZ-ALONSO, K. BENAMAR, R. K.
MYERS, M. SANCIBRIAN, R. M. RUIZ, AND J. A. ARMENGOL.
Macrophage inflammatory protein-1 beta (MIP-1 beta) produced
endogenously in brain during E. coli fever in rats. Eur. J. Neurosci. 8: 424–428, 1996.
MIRTELLA, A., G. TRINGALI, G. GUERRIERO, P. GHIARA, L.
PARENTE, P. PREZIOSI, AND P. NAVARRA. Evidence that the
interleukin-1b-induced prostaglandin E2 release from rat hypothalamus is mediated by type I and type II interleukin-1 receptors. J.
Neuroimmunol. 61: 171–177, 1995.
MISSALE, C., F. BORONI, S. SIGALA, A. BURIANI, M. FABRIS,
A. LEON, R. DAL TOSO, AND P. SPANO. Nerve growth factor in
the anterior pituitary: localization in mammotroph cells and cosecretion with prolactin by a dopamine-regulated mechanism.
Proc. Natl. Acad. Sci. USA 93: 4240–4245, 1996.
MITCHAM, J. L., P. PARNET, T. P. BONNERT, K. E. GARKA, M. J.
GERHART, J. L. SLACK, M. A. GAYLE, S. K. DOWER, AND J. E.
SIMS. T1/ST2 signaling establishes it as a member of an expanding
interleukin-1 receptor family. J. Biol. Chem. 271: 5777–5783, 1996.
MIYABO, S., E. OOYA, K. MIYANAGA, N. AOYAGI, M. HIRAI, S.
KISHIDA, AND T. NAKAI. Stimulation of the hypothalamo-pituitary-adrenal axis by epidermal growth factor. Regul. Pept. 31: 65–
74, 1990.
MIYAJIMA, A., T. KITAMURA, N. HARADA, T. YOKOTA, AND
K.-I. ARAI. Cytokine receptors and signal transduction. Annu. Rev.
Immunol. 10: 295–331, 1992.
MOHANKUMAR, P. S., AND S. K. QUADRI. Systemic administration of interleukin-1 stimulates norepinephrine release in the paraventricular nucleus. Life Sci. 52: 1961–1967, 1993.
MOHANKUMAR, P. S., S. THYAGARAJAN, AND S. KALEEM AUADRI. Interleukin-1 stimulates the release of dopamine and dihydroxyphenylacetic acid from the hypothalamus in vivo. Life Sci.
48: 925–930, 1991.
MONCADA, S., R. M. J. PALMER, AND E. A. HIGGS. Nitric oxide:
physiology, pathophysiology and pharmacology. Pharmacol. Rev.
43: 109–142, 1991.
MONTGOMERY, D. W., J. A. LEFEVRE, E. D. ULRICH, C. R. ADAMSON, AND C. F. ZUKOSKI. Identification of prolactin-like proteins synthesized by normal murine lymphocytes. Endocrinology
127: 2601–2603, 1990.
MONTGOMERY, D. W., C. F. ZUKOSKI, G. N. SHAH, A. R. BUCKLEY, T. PACHOLCZYK, AND D. H. RUSSEL. Concanavalin-A-stimulated murine splenocytes produce a factor with prolactin-like bioactivity and immunoreactivity. Biochem. Biophys. Res. Commun.
145: 692–698, 1987.
MORGAN, J. I., AND T. CURRAN. Stimulus-transcription coupling
in the nervous system: involvement of the inducible proto-oncogenes fos and jun. Annu. Rev. Neurosci. 14: 421–451, 1991.
MORGANTI-KOSSMAN, M. C., T. KOSSMAN, AND S. M. WAHL.
Cytokines and neuropathology. Trends Pharmacol. Sci. 13: 286–
291, 1992.
MORIMOTO, A., N. MURAKAMI, T. NAKAMORI, Y. SAKATA, AND
T. WATANABE. Possible involvement of prostaglandin E in development of ACTH response in rats induced by human recombinant
interleukin-1. J. Physiol. (Lond.) 411: 245–256, 1989.
MORROW, L. E., J. L. MCCLELLAN, C. A. CONN, AND M. J.
KLUGER. Glucocorticoids alter fever and IL-6 responses to psychological stress and to lipopolysaccharide. Am. J. Physiol. 264
(Regulatory Integrative Comp. Physiol. 33): R1010–R1016, 1993.
MOUIHATE, A., AND J. LESTAGE. Epidermal growth factor: a
potential paracrine and autocrine system within the pituitary.
Neuroreport 6: 1401–1404, 1995.
MUCHAMUEL, T., S. MENON, P. PISACANE, M. C. HOWARD, AND
D. A. COCKAYNE. IL-13 protects mice from lipopolysaccharideinduced lethal endotoxemia. Correlation with down-modulation
of TNF-a, IFN-gamma and IL-12 production. J. Immunol. 158:
2898–2903, 1997.
/ 9j0c$$oc11
P13-8
59
564. MUELLER, S. G., AND J. E. KUDLOW. Transforming growth factorb (TGFb) inhibits TGFb expression in bovine anterior pituitaryderived cells. Mol. Endocrinol. 5: 1439–1446, 1991.
565. MULLER, H., E. HAMMES, C. HIEMKE, AND G. HESS. Interferonalpha-2-induced stimulation of ACTH and cortisol secretion in
man. Neuroendocrinology 54: 499–503, 1991.
566. MULLER, H., C. HIEMKE, E. HAMMES, AND G. HESS. Sub-acute
effects of interferon-alpha 2 on adrenocorticotrophic hormone,
cortisol, growth hormone and prolactin in humans. Psychoneuroendocrinology 17: 459–465, 1992.
567. MUNCK, A., P. M. GUYRE, AND N. J. HOLBROOK. Physiological
functions of glucocorticoids in stress and their relation to pharmacological actions. Endocr. Rev. 5: 25–44, 1984.
568. MURAKAMI, N. Function of the OVLT as an entrance into the
brain for endogenous pyrogens. In: Neuroimmunology of Fever,
edited by T. Bartfai and D. Ottoson. Oxford, UK: Pergamon, 1992,
p. 107–114.
569. MURAKAMI, N., AND T. WATANABE. Activation of ACTH release
is mediated by the same molecule as the final mediator, PGE2, of
the febrile response in rats. Brain Res. 478: 171–174, 1989.
570. MURAMAMI, N., J. FUKATA, T. TSUKADA, H. KOBAYASHI, O.
EBISUI, H. SEGAWA, S. MURO, H. IMURA, AND K. NAKAO. Bacterial lipopolysaccharide-induced expression of interleukin-6 messenger ribonucleic acid in the rat hypothalamus, pituitary, adrenal
gland, and spleen. Endocrinology 133: 2574–2578, 1993.
571. MURROS, K., R. FOGELHOLM, S. KETTUNEN, AND A. L. VUORELA. Serum cortisol and outcome of ischemic brain infarction. J.
Neurol. Sci. 116: 12–17, 1993.
572. NAITO, Y., J. FUKATA, Y. MASUI, Y. HIRAI, N. MURAKAMI, T.
TOMINAGA, Y. NAKAI, S. TAMAI, K. MORI, AND H. IMURA. interleukin-1b analogues with markedly reduced pyrogenic activity
can stimulate secretion of adrenocorticotropic hormone in rats.
Biochem. Biophys. Res. Commun. 167: 103–109, 1990.
573. NAITO, Y., J. FUKATA, S. NAKAISHI, Y. NAKAI, Y. HIRAI, S.
TAMAI, K. MORI, AND H. IMURA. Chronic effects of interleukin1 on hypothalamus, pituitary and adrenal glands in rat. Neuroendocrinology 51: 637–641, 1990.
574. NAITO, Y., J. FUKATA, T. TOMINAGA, Y. MASUI, Y. HIRAI, N.
MURAKAMI, S. TAMAI, AND H. IMURA. Adrenocorticotropic hormone releasing activities of interleukins in a homologous in vivo
system. Biochem. Biophys. Res. Commun. 164: 1262–1267, 1989.
575. NAITOH, Y., J. FUKATA, T. TOMINAGA, Y. NAKAI, S. TAMAI, K.
MORI, AND H. IMURA. Interleukin-6 stimulates the secretion of
adrenocorticotropic hormone in conscious, freely moving rats.
Biochem. Biophys. Res. Commun. 155: 1459–1463, 1988.
576. NAKAMORI, T., A. MORIMOTO, K. YAMAGUCHI, T. WATANABE,
N. C. LONG, AND N. MURAKAMI. Organosum vasculosum laminae
terminalis (OVLT) is a brain site to produce interleukin-1b during
fever. Brain Res. 618: 155–159, 1993.
577. NAKAMORI, T., A. MORIMOTO, K. YAMAGUCHI, T. WATANABE,
AND N. MURAKAMI. Interleukin-1 beta production in the rabbit
brain during endotoxin-induced fever. J. Physiol. (Lond.) 476:
177–186, 1994.
578. NAKAMORI, T., Y. SAKATA, T. WATANABE, A. MORIMOTO, S.
NAKAMURA, AND N. MURAKAMI. Suppression of interleukin-1
beta production in the circumventricular organs in endotoxintolerant rabbits. J. Physiol. (Lond.) 675: 103–109, 1995.
579. NASH, A. D., M. R. BRANDON, AND P. A. BELLO. Effects of tumor
necrosis factor-a on growth hormone and interleukin-6 mRNA in
ovine pituitary cells. Mol. Cell. Endocrinol. 84: R31–R37, 1992.
580. NASUSHITA, R., H. WATANOBE, AND K. TAKEBE. A comparative
study of adrenocorticotropin-releasing activity of prostaglandins
E1, E2, F2a and D2 in the rat. Prostaglandins Leukotrienes Essent.
Fatty Acids 56: 165–168, 1997.
581. NAVARRA, P. The effects of endotoxin on the neuroendocrine
axis. Curr. Opin. Endocrinol. Diabetes 2: 127–133, 1995.
582. NAVARRA, P., G. POZZOLI, L. BRUNETTI, E. RAGAZZONI, M.
BESSER, AND A. GROSSMAN. Interleukin-1b and interleukin-6
specifically increase the release of prostaglandin E2 from rat hypothalamic explants in vitro. Neuroendocrinology 56: 61–68, 1992.
583. NAVARRA, P., S. TSAGARAKIS, M. FARIA, L. H. REES, G. M. BESSER, AND A. B. GROSSMAN. Interleukins-1 and -6 stimulate the
release of corticotropin-releasing hormone-41 from rat hypothala-
11-25-98 11:16:36
pra
APS-Phys Rev
Downloaded from on April 23, 2014
553.
REGULATION OF HPA AXIS BY CYTOKINES
60
584.
585.
586.
587.
588.
589.
590.
592.
593.
594.
595.
596.
597.
598.
599.
600.
mus in vitro via the eicosanoid cyclooxygenase pathway. Endocrinology 128: 37–44, 1991.
NETA, R., R. PERLSTEIN, S. N. VOGEL, G. D. LEDNEY, AND J.
ABRAHAMS. Role of interleukin-6 (IL-6) in the protection from
lethal irradiation and in endocrine responses to IL-1 and tumor
necrosis factor. J. Exp. Med. 175: 689–694, 1992.
NIETFELD, J. J., B. WILLBRINK, M. HELLE, J. L. VAN ROY, W.
DEN OTTER, A. J. SWAAK, AND O. HUBER-BRUNING. Interleukin-1-induced interleukin-6 is required for the inhibition of proteoglycan synthesis by interleukin-1 in human articular carilage. Arthritis Rheum. 33: 1695–1701, 1990.
NIIJIMA, A. The afferent discharges from sensors for interleukin1b in the hepato-portal system in the rat (Abstract). J. Physiol.
(Lond.) 446: 236P, 1992.
NIIJIMA, A. The afferent discharges from sensors for interleukin
1 beta in the hepatoportal system in the anesthetized rat. J. Auton.
Nerv. Syst. 61: 287–291, 1996.
NIIMI, M., M. SAT, Y. WADA, J. TAKAHARA, AND K. KAWANISHI.
Effect of central and continuous intravenous injection of interleukin-1b on brain c-fos expression in the rat: involvement of prostaglandins. Neuroimmunomodulation 3: 87–92, 1996.
NIIMI, M., Y. WADA, M. SATO, J. TAKAHARA, AND K. KAWANISHI. Effect of continuous intravenous injection of interleukin-6
and pretreatment with cyclooxygenase inhibitor on brain c-fos
expression in the rat. Neuroendocrinology 66: 47–53, 1997.
NISHIBORI, M., N. NAKAYA, A. TAHARA, M. KAWABATA, S.
MORI, AND K. SAEKI. Presence of macrophage migration factor
(MIF) in ependyma, astrocytes and neurons in the bovine brain.
Neurosci. Lett. 213: 193–196, 1996.
NISHINO, T., J. BERNHAGEN, H. SHIIKI, T. CALANDRA, K. DOHI,
AND R. BUCALA. Localization of macrophage migration inhibitory
factor (MIF) to secretory granules within the corticotrophic and
thyrotrophic cells of the pituitary gland. Mol. Med. 1: 781–788,
1995.
NISHIYORI, A., M. MINAMI, S. TAKAMI, AND M. SATOH. Type 2
interleukin-1 receptor mRNA is induced by kainic acid in the rat
brain. Brain Res. 50: 237–245, 1997.
NITTA, T., K. SATO, M. ALLEGRETTA, S. BROCKE, M. LIM, D. J.
MITCHELL, AND L. STEINMAN. Expression of granulocyte colony
stimulating factor and granulocyte-macrophage colony stimulating factor genes in human astrocytoma cell lines and in glioma
specimens. Brain Res. 571: 19–25, 1992.
NOBEL, C. S., AND M. SCHULTZBERG. Induction of interleukin1b mRNA and enkephalin mRNA in the rat adrenal gland by lipopolysaccharides studied by in situ hybridization histochemistry.
Neuroimmunomodulation 2: 61–73, 1995.
NOHAVA, K., U. MALIPIERO, K. FREI, AND A. FONTANA. Neurons
and neuroblastoma as a source of macrophage colony-stimulating
factor. Eur. J. Immunol. 22: 2539–2545, 1992.
NOLTEN, W. E., D. GOLDSTEIN, M. LINDSTROM, M. V. MCKENNA, I. H. CARLSON, D. L. TRUMP, J. SCHILLER, E. C. BORDEN, AND E. N. EHRLICH. Effects of cytokines on the pituitaryadrenal axis in cancer patients. J. Interferon Res. 13: 349–357,
1993.
O’CONNELL, N. A., A. KUMAR, K. CHATZIPANTELI, A. MOHAN,
R. K. AGARWAL, C. HEAD, S. R. BORNSTEIN, A. B. ABOU-SAMARA, AND A. R. GWOSDOW. Interleukin-1 regulates corticosterone secretion from the rat adrenal gland through a catecholamine-dependent and prostaglandin E2-independent mechanism.
Endocrinology 135: 460–467, 1994.
O’GRADY, M. P., N. R. S. HALL, AND R. A. MENZIES. Interleukin1b stimulates adrenocorticotropin and corticosterone release in
10-day-old rat pups. Psychoneuroendocrinology 18: 241–247,
1993.
OHGO, S., K. NAKATSURU, E. ISHIKAWA, AND S. MATSUKURA.
Interleukin-1 (IL-1) stimulates the release of corticotropin-releasing factor (CRF) from superfused rat hypothalamo-neurohypophyseal complexes (HNC) independently of the histaminergic mechanism. Brain Res. 558: 217–223, 1991.
OHGO, S., K. NAKATSURU, Y. OKI, E. ISHIKAWA, AND S. MATSUKURA. Stimulation by interleukin-1 (IL-1) of the release of rat
corticotropin-releasing factor (CRF), which is independent of the
/ 9j0c$$oc11
P13-8
601.
602.
603.
604.
605.
606.
607.
608.
609.
610.
611.
612.
613.
614.
615.
616.
617.
618.
619.
11-25-98 11:16:36
Volume 79
cholinergic mechanism, from superfused rat hypothalamo-neurohypophysial complexes. Brain Res. 550: 213–219, 1991.
OHMICHI, M., K. HIROTA, K. KOIKE, H. KURACHI, S. OHTSUKA,
N. MATSUZAKI, M. YAMAGUCHI, A. MIYAKE, AND O. TANIZAWA.
Binding sites for interleukin-6 in the anterior pituitary gland. Neuroendocrinology 55: 199–203, 1992.
OHZATO, H., M. MONDEN, K. YOSHIZAKI, A. OGATA, N. NISHIMOTO, M. GOTOH, T. KISHIMOTO, AND T. MORI. Systemic production of interleukin-6 following acute inflammation in the rat.
Biochem. Biophys. Res. Commun. 197: 1556–1562, 1993.
OLDENBURG, H. S. A., M. A. ROGY, D. D. LAZARUS, K. J. VAN
ZEE, B. P. KEELER, R. A. CHIZZONITE, S. F. LOWRY, AND L. L.
MOLDAWER. Cachexia and the acute-phase protein response in
inflammation are regulated by interleukin-6. Eur. J. Immunol. 23:
1889–1894, 1993.
OLSEN, N. J., W. E. NICHOLSON, C. R. DEBOLD, AND D. N.
ORTH. Lymphocyte-derived adrenocorticotropin is insufficient to
stimulate adrenal steroidogenesis in hypophysectomized rats. Endocrinology 130: 2113–2119, 1992.
OPP, M. R., AND J. M. KRUEGER. Anti-interleukin-1b reduces
sleep and sleep rebound after sleep deprivation in rats. Am. J.
Physiol. 266 (Regulatory Integrative Comp. Physiol. 35): R688–
R695, 1994.
OPP, M. R., AND J. M. KRUEGER. Interleukin-1 is involved in responses to sleep deprivation in the rabbit. Brain Res. 639: 57–65,
1994.
OPP, M. R., AND L. A. TOTH. Somnogenic and pyrogenic effects
of interleukin-1beta and lipopolysaccharide in intact and vagotomized rats. Life Sci. 62: 923–936, 1998.
ORANGE, J. S., T. P. SALAZAR-MATHER, S. M. OPAL, R. L. SPENCER, A. H. MILLER, B. S. MCEWAN, AND C. A. BIRON. Mechanism
of interleukin-12-mediated toxicities during experimental viral infections: role of tumor necrosis factor and glucocorticoids. J. Exp.
Med. 181: 901–914, 1995.
O’SULLIVAN, M. G., F. H. CHILTON, E. M. HUGGINS, AND C. E.
MCCALL. Lipopolysaccharide priming of alveolar macrophages for
enhanced synthesis of prostanoids involves induction of a novel
prostaglandin H synthase. J. Biol. Chem. 267: 14547–14550, 1992.
OTA, K., T. KATAFUCHI, A. TAKAKI, AND T. HORI. AV3V neurons
that send axons to hypothalamic nuclei respond to the systemic
injection of IL-1b. Am. J. Physiol. 272 (Regulatory Integrative
Comp. Physiol. 41): R532–R540, 1997.
OTT, L., B. YOUNG, AND C. MCCLAIN. The metabolic response to
brain injury. J. Parenterol. Enteral Nutr. 11: 488–493, 1987.
OTTAVIANI, E., E. CASELGRANDI, AND C. FRANCESCHI. Cytokines and evolution: in vitro effects of IL-1 alpha, IL-1 beta, TNFalpha and TNF-beta on the ancestral type of stress response. Biochem. Biophys. Res. Commun. 207: 288–292, 1995.
OTTAVIANI, E., E. CASELGRANDI, F. PETRAGLIA, AND C. FRANCESCHI. Stress response in the freshwater snail Planobarius corneus (L.) (gastropoda, pulmonata): interaction between CRF,
ACTH, and biogenic amines. Gen. Comp. Endocrinol. 87: 354–
360, 1992.
OTTAVIANI, E., A. COSSARIZZA, C. ORTOLANI, D. MONTI, AND
C. FRANCESCHI. ACTH-like molecules in gastropod molluscs: a
possible role in ancestral immune response and stress. Proc. R.
Soc. Lond. B Biol. Sci. 245: 215–218, 1991.
OTTAVIANI, E., AND C. FRANCHESCHI. The neuroimmunology
of stress from invertebrates to man. Prog. Neurobiol. 48: 421–440,
1996.
OTTAVIANI, E., AND C. FRANCHESCHI. The invertebrate phagocytic immunocyte: clues to a common evolution of immune and
neuroendocrine systems. Immunol. Today 18: 169–174, 1997.
OTTAVIANI, E., A. FRANCHINI, E. CASELGRANDI, A. COSSARIZZA, AND C. FRANCESCHI. Relationship between corticotropinreleasing factor and interleukin-2: evolutionary evidence. FEBS
Lett. 351: 19–21, 1994.
OTTEN, U., J. B. BAUMANN, AND J. GIRARD. Stimulation of the
pituitary-adrenocortical axis by nerve growth factor. Nature 282:
412–414, 1979.
OVADIA, H., O. ABRAMSKY, V. BARAK, N. CONFORTI, D. SAPHIER, AND J. WEIDENFELD. Effect of interleukin-1 on adreno-
pra
APS-Phys Rev
Downloaded from on April 23, 2014
591.
ANDREW V. TURNBULL AND CATHERINE L. RIVIER
January 1999
620.
621.
622.
623.
624.
625.
626.
627.
629.
630.
631.
632.
633.
634.
635.
636.
637.
638.
639.
cortical activity in intact and hypothalamic deafferented rats. Exp.
Brain Res. 76: 246–249, 1989.
PAAJANEN, H., W. GRODD, D. REVEL, B. ENGELSTAD, AND R. C.
BRASCH. Gadolium-DTPA enhanced MR imaging of intramuscular
abscesses. Magn. Reson. Imaging 5: 109–115, 1987.
PANERI, A. E., AND P. SACERDOTE. b-Endorphin in the immune
system: a role at last? Immunol. Today 18: 317–319, 1997.
PAPANICOLAOU, D. A., J. S. PETRIDES, C. TSIGOS, S. BINA,
K. T. KALOGERAS, R. WILDER, P. W. GOLD, P. A. DEUTSER, AND
G. P. CHROUSOS. Exercise stimulates interleukin-6 secretion: inhibition by glucocorticoids and correlation with catecholamines.
Am. J. Physiol. 271 (Endocrinol. Metab. 40): E601–E625, 1996.
PARDY, K., D. MURPHY, D. CARTER, AND K. M. HUI. The influence of interleukin-2 on vasopressin and oxytocin gene expression
in the rodent hypothalamus. J. Neuroimmunol. 42: 131–138, 1993.
PARK, J. H., AND S. H. SHIN. Induction of interleukin-12 gene expression in the brain in septic shock. Biochem. Biophys. Res.
Commun. 224: 391–396, 1996.
PARNET, P., S. AMINDARI, C. WU, D. BRUNKE-REESE, E. GOUJON, J. A. WEYHENMEYER, R. DANTZER, AND K. W. KELLEY.
Expression of type I and type II interleukin-1 receptors in mouse
brain. Brain Res. 27: 63–70, 1994.
PARNET, P., D. BRUNKE, E. GOUJON, J. DESMOTTES-MAINARD, A. BIRAGYN, S. ARKINS, R. DANTZER, AND K. W. KELLEY.
Molecular identification of two types of IL-1 receptors in the murine pituitary gland. J. Neuroendocrinol. 5: 213–218, 1993.
PARNET, P., K. E. GARKA, T. P. BONNERT, S. K. DOWER, AND
J. E. SIMS. IL-1Rrp is a novel receptor-like molecule similar to the
type I interleukin-1 receptor and its homolgues T1/ST2 and IL-1R
AcP. J. Biol. Chem. 271: 3967–3970, 1996.
PARSADAIANTZ, S. M., N. LEVIN, V. LENOIR, J. L. ROBERTS,
AND B. KERDELHUE. Human interleukin-1b: corticotropin releasing factor and ACTH release and gene expression in the male rat:
in vivo and in vitro studies. J. Neurosci. Res. 37: 675–682, 1994.
PARSADANIANTZ, S. M., V. LENOIR, B. TERLAIN, AND B. KERDELHUE. Lack of effect of interleukins 1a and 1b, during in vitro
perifusion, on anterior pituitary release of adrenocorticotropic
hormone and b endorphin in the male rat. J. Neurosci. Res. 34:
315–323, 1993.
PATH, G., AND S. R. BORNSTEIN. Interleukin-6 and interleukin-6
receptor in the human adrenal gland: expression and effects on
steroidogenesis. J. Clin. Endocrinol. Metab. 82: 2343–2349, 1997.
PATTERSON, J. C., AND G. V. CHILDS. Nerve growth factor and
its receptor in the anterior pituitary. Endocrinology 135: 1689–
1696, 1994.
PATTERSON, J. C., AND G. V. CHILDS. Nerve growth factor in
the anterior pituitary: regulation of secretion. Endocrinology 135:
1697–1704, 1994.
PAUL, W. E. Pleiotropy and redundancy: T-cell derived lymphokines in the immune response. Cell 57: 521–524, 1989.
PAYNE, L. C., F. OBAL, M. R. OPP, AND J. M. KRUEGER. Stimulation and inhibition of growth hormone secretion by interleukin1b: the involvement of growth hormone-releasing hormone. Neuroendocrinology 56: 118–123, 1992.
PAYNE, L. C., D. A. WEIGENT, AND J. E. BLALOCK. Induction of
pituitary sensitivity to interleukin-1: a new function for corticotropin-releasing hormone. Biochem. Biophys. Res. Commun. 198:
480–484, 1994.
PEQUEGNAT, W., N. A. GARRICK, AND E. STOVER. Neuroscience
findings in AIDS: a review of research sponsored by the National
Institute of Mental Health. Prog. Neuropsychopharmacol. Biol.
Psychiatry 16: 145–170, 1992.
PEREDA, M. P., V. GOLDBERG, A. CHERVIN, G. CARRIZO, A.
MOLINA, J. ANDRADA, J. SAUER, U. RENNER, G. K. STALLA,
AND E. ARZT. Interleukin-2 (IL-2) and IL-6 regulate c-fos protooncogene expression in human pituitary adenoma explants. Mol.
Cell. Endocrinol. 124: 33–42, 1996.
PERLSTEIN, R. S., N. R. MEHTA, E. H. MOUGEY, R. NETA, AND
M. H. WHITNALL. Systemically administered H1 and H2 receptor
antagonists do not block the ACTH response to bacterial lipopolysaccharide and interleukin-1. Neuroendocrinology 60: 418–425,
1994.
PERLSTEIN, R. S., E. H. MOUGEY, W. E. JACKSON, AND R. NETA.
/ 9j0c$$oc11
P13-8
640.
641.
642.
643.
644.
645.
646.
647.
648.
649.
650.
651.
652.
653.
654.
655.
11-25-98 11:16:36
61
Interleukin-1 and interleukin-6 act synergistically to stimulate the
release of adrenocorticotropic hormone in vivo. Lymphokine Cytokine Res. 10: 141–146, 1991.
PERLSTEIN, R. S., M. H. WHITNALL, J. S. ABRAMS, E. H.
MOUGHEY, AND R. NETA. Synergistic roles of interleukin-6, interleukin-1, and tumor necrosis factor in the adrenocorticotropin
response to bacterial lipopolysaccharide in vivo. Endocrinology
132: 946–952, 1993.
PETERS, M., S. JACOBS, M. EHLERS, P. VOLLMER, J. MULLBERG, E. WOLF, G. BREM, K.-H. MEYER ZUM BUSCHENFELDE,
AND S. ROSE-JOHN. The function of the soluble interleukin-6 (IL6) receptor in vivo: sensitization of human soluble IL-6 receptor
transgenic mice towards IL-6 and prolongation of the plasma halflife of IL-6. J. Exp. Med. 183: 1399–1406, 1996.
PFEFFER, K., T. MATSUYAMA, T. M. KUNDIG, A. WAKEMAN, K.
KISHIHARA, A. SHAHINIAN, K. WIEGMANN, P. S. OHASHI, M.
KRONKE, AND T. W. MAK. Mice deficient for the 55 kD tumor
necrosis factor receptor are resistant to endotoxic shock, yet succumb to L. monocytogenes infection. Cell 73: 7698–7702, 1993.
PITOSSI, F. J., AND H. O. BESEDOVSKY. A multispecific internal
control (pRat6) for the analysis of rat cytokine mRNA levels by
quantitative RT-PCR. Eur. Cytokine Netw. 7: 377–379, 1996.
PITOSSI, F., A. DEL REY, A. KABIERSCH, AND H. BESEDOVSKY.
Induction of cytokine transcripts in the CNS and pituitary following peripheral administration of endotoxin to mice. J. Neurosci.
Res. 48: 287–298, 1997.
PITTERMAN, A. B., G. FIEDLANDER, G. KELLY, T. G. ROPCHACK, D. J. GOLDSTEIN, AND H. R. KEISER. Abdominal vagotomy does not modify endotoxic shock in rats. Life Sci. 33: 1033–
1037, 1983.
PLATA-SALAMAN, C. R., AND S. E. ILYIN. Interleukin-1 beta (IL1 beta)-induced modulation of the hypothalamic IL-1 beta system,
tumor necrosis factor-alpha, and transforming growth factor beta
1 mRNAs in obese (fa/fa) and lean (Fa/Fa) Zucker rats: implications to IL-1 beta feedback systems and cytokine-cytokine interactions. J. Neurosci. Res. 49: 541–550, 1997.
PLOTKIN, S. R., W. A. BANKS, AND A. J. KASTIN. Comparison of
saturable transport and extracellular pathways in the passage of
interleukin-1b across the blood-brain barrier. J. Neuroimmunol.
67: 41–47, 1996.
PLOTSKY, P. M. Pathways to the secretion of adrenocorticotropin:
a view from the portal. J. Neuroendocrinol. 3: 1–9, 1991.
PLOTSKY, P. M., E. T. J. CUNNINGHAM, AND E. P. WIDMAIER.
Catecholaminergic modulation of corticotropin-releasing factor
and adrenocorticotropin secretion. Endocr. Rev. 10: 437–458,
1989.
PLOTSKY, P. M., A. KJAER, S. W. SUTTON, P. E. SAWCHENKO,
AND W. VALE. Central activin administration modulates corticotropin-releasing hormone and adrenocorticotropin secretion. Endocrinology 128: 2520–2525, 1991.
PLOTSKY, P. M., AND P. E. SAWCHENKO. Hypophysial portal
plasma levels, median eminence content and immunohistochemical staining of corticotropin-releasing factor, arginine vasopressin
and oxytocin following pharmacological adrenalectomy. Endocrinology 120: 1361–1369, 1987.
POZZOLI, G., A. COSTA, M. GRIMALDI, G. SCHETTINI, P. PREZIOSI, A. GROSSMAN, AND P. NAVARRA. Lipopolysaccharide modulation of eicosanoid and corticotropin-releasing hormone release
from rat hypothalamic explants and astrocyte cultures in vitro:
evidence for the involvement of prostaglandin E2 but not prostaglandin F2 alpha and lack of effect of nerve growth factor. J. Endocrinol. 140: 103–109, 1994.
POZZOLI, G., C. MANCUSO, A. MIRTELLA, P. PREZIOSI, A. B.
GROSSMAN, AND P. NAVARRA. Carbon monoxide as a novel neuroendocrine modulator: inhibition of stimulated corticotropin-releasing hormone release from acute rat hypothalamic explants.
Endocrinology 135: 2314–2317, 1994.
PURBA, J. S., F. C. RAADSHEER, M. A. HOFMAN, R. RAVID, C. H.
POLMAN, W. KAMPHORST, AND D. F. SWAAB. Increased number
of corticotropin-releasing hormone expressing neurons in the hypothalamic paraventricular nucleus of patients with multiple sclerosis. Neuroendocrinology 62: 62–70, 1995.
QUAN, N., S. K. SUNDAR, AND J. M. WEISS. Induction of interleu-
pra
APS-Phys Rev
Downloaded from on April 23, 2014
628.
REGULATION OF HPA AXIS BY CYTOKINES
62
656.
657.
658.
659.
660.
661.
662.
664.
665.
666.
667.
668.
669.
670.
671.
672.
673.
674.
kin-1 in various brain regions after peripheral and central injections of lipopolysaccharide. J. Neuroimmunol. 49: 125–134, 1994.
QUAN, N., Z. ZHANG, M. EMERY, R. BONSALL, AND J. M. WEISS.
Detection of interleukin-1 bioactivity in various brain regions of
normal healthy rats. Neuroimmunomodulation 3: 47–55, 1996.
QUAN, N., Z. ZHANG, M. EMERY, E. LAI, R. BONSALL, V. S.
KALYANARAMAN, AND J. M. WEISS. In vivo induction of interleukin-1 bioactivity in brain tissue after intracerebral infusion of native gp120 and gp160. Neuroimmunomodulation 3: 56–61, 1996.
RABER, J., AND F. E. BLOOM. IL-2 induces vasopressin release
from the hypothalamus and the amygdala: role of nitric oxidemediating signal. J. Neurosci. 14: 6187–6195, 1994.
RABER, J., G. F. KOOB, AND F. E. BLOOM. Interleukin-2 (IL-2)
induces corticotropin-releasing factor (CRF) release from the
amygdala and involves a nitric oxide-mediated signaling: comparison with the hypothalamic response. J. Pharmacol. Exp. Ther.
272: 815–824, 1995.
RABER, J., G. F. KOOB, AND F. E. BLOOM. Interferon-g and transforming growth factor-b1 regulate corticotropin-releasing factor
release from the amygdala: comparison with the hypothalamic
response. Neurochem. Int. 30: 455–463, 1997.
RABER, J., S. M. TOGGAS, S. LEE, F. E. BLOOM, C. J. EPSTEIN,
AND L. MUCKE. Central nervous system expression of HIV-1
Gp120 activates the hypothalamic-pituitary-adrenal axis: evidence
for involvement of NMDA receptors and nitric oxide synthase.
Virology 226: 362–373, 1996.
RADY, P. L., E. M. SMITH, P. CADET, M. R. OPP, S. K. TYRING,
AND T. K. HUGHES. Presence of interleukin-10 transcripts in human pituitary and hypothalamus. Cell. Mol. Neurobiol. 15: 289–
296, 1995.
RAIVICH, G., J. GEHRMANN, AND G. W. KREUTZBERG. Increase
of macrophage colony-stimulating factor and granulocyte-macrophage colony-stimulating factor receptors in the regenerating rat
facial nucleus. J. Neurosci. Res. 30: 682–686, 1991.
RAPOPORT, S. I. Blood-Brain Barrier in Physiology and Medicine. New York: Raven, 1976.
RASMUSSEN, A. K., U. FELDT-RASMUSSEN, AND K. BENDTZEN.
The effect of interleukin-1 on the thyroid gland. Autoimmunity
16: 141–148, 1993.
RAY, D., AND S. MELMED. Pituitary cytokine and growth factor
expression and action. Endocr. Rev. 18: 206–228, 1997.
RAY, D. W., S. G. REN, AND S. MELMED. Leukemia inhibitory factor (LIF) stimulates proopiomelanocortin (POMC) expression in
a corticotroph cell line. Role of Stat pathway. J. Clin. Invest. 97:
1852–1859, 1996.
REINISCH, N., M. WOLKERSDORFER, C. M. KAHLER, K. YE,
C. A. DINARELLO, AND C. J. WIEDERMANN. Interleukin-1 receptor type I mRNA in mouse brain as affected by peripheral administration of bacterial lipopolysaccharide. Neurosci. Lett. 166: 165–
167, 1994.
RENNER, U., C. J. NEWTON, U. PAGOTTO, J. SAUER, E. ARZT,
AND G. STALLA. Involvement of interleukin-1 and interleukin-1
receptor antagonist in rat anterior pituitary cell growth regulation.
Endocrinology 136: 3186–3193, 1995.
RENNERT, P. D., AND G. HEINRICH. Nerve growth factor mRNA
in brain: localization by in situ hybridization. Biochem. Biophys.
Res. Commun. 138: 813–818, 1986.
RETTORI, V., N. BELOVA, M. GIMENO, AND S. M. MCCANN. Inhibition of nitric oxide synthase in the hypothalamus blocks the
increase in plasma prolactin induced by intraventricular injection
of interleukin-1b in the rat. Neuroimmunomodulation 1: 116–
120, 1994.
RETTORI, V., W. L. DEES, J. K. HINEY, K. LYSON, AND S. M.
MCCANN. An interleukin-1-alpha-like neuronal system in the preoptic-hypothalamic region and its induction by bacterial lipopolysaccharide in concentrations which alter pituitary hormone release. Neuroimmunomodulation 1: 251–258, 1994.
RETTORI, V., L. MILENKOVIC, B. A. BEUTLER, AND S. M.
MCCANN. Hypothalamic action of cachectin to alter pituitary hormone release. Brain Res. Bull. 23: 471–475, 1989.
REYES, T. M., AND C. L. COE. Interleukin-1b differentially affects
interleukin-6 and soluble interleukin-6 receptor in the blood and
/ 9j0c$$oc11
P13-8
675.
676.
677.
678.
679.
680.
681.
682.
683.
684.
685.
686.
687.
688.
689.
690.
691.
692.
693.
694.
11-25-98 11:16:36
Volume 79
central nervous system of the monkey. J. Neuroimmunol. 66:
135–141, 1996.
REZAI, A. R., A. REZAI, O. MARTINEZ-MAZA, M. VANDERMEYDEN, AND M. H. WEISS. Interleukin-6 and interleukin-6 receptor gene expression in pituitary tumors. J. Neuro-oncol. 19: 131–
135, 1994.
RITCHIE, P. K., H. H. KNOGHT, M. ASHBY, AND A. M. JUDD. Serotonin increases interleukin-6 release and decreases tumor necrosis
factor release from rat adrenal zona glomerulosa cells in vitro.
Endocrine 5: 291–297, 1996.
RITCHIE, P. K., B. L. SPANGELO, D. K. KRZYMOWSKI, T. B. ROSSITER, E. KURTH, AND A. M. JUDD. Adenosine increases interleukin 6 release and decreases tumor necrosis factor release from
rat adrenal zona glomerulosa cells, ovarian cells, and peritoneal
macrophages. Cytokine 9: 187–198, 1997.
RIVEST, S. Molecular mechanism and neural pathways mediating
the influence of interleukin-1 on the activity of neuroendocrine
CRF motoneurons in the rat. Int. J. Dev. Neurosci. 13: 135–146,
1995.
RIVEST, S., AND N. LAFLAMME. Neuronal activity and neuropeptide gene expression transcription in the brains of immune-challenged rats. J. Neuroendocrinol. 7: 501–525, 1995.
RIVEST, S., N. LAFLAMME, AND R. E. NAPPI. Immune challenge
and immobilization stress induce transcription of the gene encoding the CRF receptor in selective nuclei of the rat hypothalamus.
J. Neurosci. 15: 2680–2695, 1995.
RIVEST, S., AND C. RIVIER. Influence of the paraventricular nucleus of the hypothalamus in the alteration of neuroendocrine
functions induced by intermittent footshock or interleukin. Endocrinology 129: 2049–2057, 1991.
RIVEST, S., AND C. RIVIER. Stress and interleukin-1b-induced activation of c-fos, NGFI-B and CRF gene expression in the hypothalamic PVN: comparison between Sprague-Dawley, Fisher-344 and
Lewis rats. J. Neuroendocrinol. 6: 101–117, 1994.
RIVEST, S., AND C. RIVIER. The role of corticotropin-releasing
factor and interleukin-1 in the regulation of neurons controlling
reproductive functions. Endocr. Rev. 16: 177–199, 1995.
RIVEST, S., G. TORRES, AND C. RIVIER. Differential effects of
central and peripheral injection of interleukin-1b on brain c-fos
expression and neuroendocrine functions. Brain Res. 587: 13–23,
1992.
RIVIER, C. Effect of peripheral and central cytokines on the hypothalamo-pituitary-adrenal axis of the rat. Ann. NY Acad. Sci. 697:
97–105, 1993.
RIVIER, C. Stimulatory effect of interleukin-1 beta on the hypothalamic-pitutary-adrenal axis of the rat: influence of age, gender and
circulating sex steroids. J. Endocrinol. 140: 365–372, 1994.
RIVIER, C. Blockade of nitric oxide formation augments ACTH
released by blood-borne interleukin-1b: role of vasopressin, prostaglandins and a-1 adrenergic receptors. Endocrinology 136:
3597–3603, 1995.
RIVIER, C. Influence of immune signals on the hypothalamic-pituitary axis of the rodent. Front. Neuroendocrinol. 16: 151–182,
1995.
RIVIER, C. Mechanisms of altered prolactin secretion due to the
administration of interleukin-1b into the brain ventricles of the
rat. Neuroendocrinology 62: 198–206, 1995.
RIVIER, C., R. CHIZZONITE, AND W. VALE. In the mouse, the
activation of the hypothalamic-pituitary-adrenal axis by a lipopolysaccharide (endotoxin) is mediated through interleukin-1. Endocrinology 125: 2800–2805, 1989.
RIVIER, C., AND P. M. PLOTSKY. Mediation by corticotropin-releasing factor (CRF) of adenohypophysial hormone secretion.
Annu. Rev. Physiol. 48: 475–494, 1986.
RIVIER, C., AND G. H. SHEN. In the rat, endogenous nitric oxide
modulates the response of the hypothalamo-pituitary-adrenal axis
to interleukin-1b, vasopressin, and oxytocin. J. Neurosci. 14:
1985–1993, 1994.
RIVIER, C., AND W. VALE. Interaction of corticotropin-releasing
factor (CRF) and arginine vasopressin (AVP) on ACTH secretion
in vivo. Endocrinology 113: 939–942, 1983.
RIVIER, C., AND W. VALE. Stimulatory effect of interleukin-1 on
pra
APS-Phys Rev
Downloaded from on April 23, 2014
663.
ANDREW V. TURNBULL AND CATHERINE L. RIVIER
January 1999
695.
696.
697.
698.
699.
700.
701.
702.
704.
705.
706.
707.
708.
709.
710.
711.
712.
713.
714.
ACTH secretion in the rat: is it modulated by prostaglandins?
Endocrinology 129: 384–388, 1991.
RIVIER, C., W. VALE, AND M. BROWN. In the rat, interleukin-1a
and -b stimulate adrenocorticotropin and catecholamine release.
Endocrinology 125: 3090–3102, 1989.
RIVIER, J., A. V. TURNBULL, AND C. RIVIER. Acute local inflammation induces temporally distinct patterns of c-fos expression in
the rat brain (Abstract). Proc. Annu. Meet. Soc. Neurosci. 27th
New Orleans LA 1997 p. 593-2.
ROBERTS, V. J., S. L. BARTH, H. MEUNIER, AND W. VALE. Hybridization histochemical and immunohistochemical localization
of inhibin/activin subunits and messenger ribonucleic acids in the
rat brain. J. Comp. Neurol. 364: 473–493, 1996.
ROH, M. S., K. A. DRAZENOVICH, J. J. BARBOSE, C. A. DINARELLO, AND C. F. COBB. Direct stimulation of the adrenal cortex by
interleukin-1. Surgery 102: 140–146, 1987.
ROMANOVSKY, A. A., V. A. KULCHITSKY, C. T. SIMONS, N. SUGIMOTO, AND M. SZEKELY. Febrile responsiveness of vagotomized
rats is suppressed even in the absence of malnutrition. Am. J.
Physiol. 273 (Regulatory Integrative Comp. Physiol. 42): R777–
R783, 1997.
ROMANOVSKY, A. A., V. A. KULCHITSKY, C. T. SIMONS, N. SUGIMOTO, AND M. SZEKELY. Cold defense mechanisms in vagotomized rats. Am. J. Physiol. 273 (Regulatory Integrative Comp.
Physiol. 42): R784–R789, 1997.
ROMANOVSKY, A. A., C. T. SIMONS, M. SZEKELY, AND V. A. KULCHITSKY. The vagus nerve in the thermoregulatory response to
systemic inflammation. Am. J. Physiol. 273 (Regulatory Integrative Comp. Physiol. 42): R407–R413, 1997.
ROOSTH, J., R. B. POLLARD, S. L. BROWN, AND W. J. MEYER.
Cortisol stimulation by recombinant interferon-a2. J. Neuroimmunol. 12: 311–316, 1986.
ROSE-JOHN, S., AND P. C. HEINRICH. Soluble receptors for cytokines and growth factors: generation and biological function. Biochem. J. 300: 281–290, 1994.
ROTEIN, P., S. K. BURGESS, J. D. MILDBRANDT, AND J. E.
KRAUSE. Differential expression of insulin-like growth factor
genes in rat central nervous system. Proc. Natl. Acad. Sci. USA
85: 265–269, 1988.
ROTH, J., C. A. CONN, M. J. KLUGER, AND E. ZEISBERGER. Kinetics of systemic and intrahypothalamic IL-6 and tumor necrosis
factor during endotoxin fever in guinea pigs. Am. J. Physiol. 265
(Regulatory Integrative Comp. Physiol. 34): R653–R658, 1993.
ROTH, P., A. BARTOCCI, AND E. R. STANLEY. Lipopolysaccharide
induces synthesis of mouse colony-stimulating factor-1 in vivo. J.
Immunol. 158: 3874–3880, 1997.
ROTHE, J., W. LESSLAUER, H. LOETSCHER, Y. LANG, P.
LOEBEL, F. KOENTGEN, A. ALTHAGE, R. ZINKERNAGEL, M.
STEINMETZ, AND H. BLUETHMANN. Mice lacking the tumor necrosis receptor 1 are resistant to TNF-mediated toxicity but highly
susceptible to infection by Listeria monocytokgens. Nature 364:
798–802, 1993.
ROTHWELL, N. J. Functions and mechanisms of interleukin-1 in
the brain. Trends Pharmacol. Sci. 12: 430–436, 1991.
ROTHWELL, N. J., N. J. BUSBRIDGE, H. HUMPHRAY, AND P. HISSEY. Central actions of interleukin-1b on fever and thermogenesis.
In: The Physiological and Pathological Effects of Cytokines, edited by C. A. Dinarello, M. J. Kluger, J. J., Powanda, and J. J. Oppenheim. New York: Wiley-Liss, 1990, p. 307–311.
ROTHWELL, N. J., AND S. J. HOPKINS. Cytokines and the central
nervous system II: actions and mechanism of action. Trends Neurosci. 18: 130–136, 1995.
ROTHWELL, N. J., G. LUHESHI, AND S. TOULMOND. Cytokines
and their receptors in the central nervous system: physiology,
pharmacology, and pathology. Pharmacol. Ther. 69: 85–95, 1996.
ROTONDO, D., H. T. ABUL, A. S. MILTON, AND J. DAVIDSON.
Pyrogenic immunomodulators increase the level of prostaglandin
E2 in the blood simultaneously with the onset of fever. Eur. J.
Pharmacol. 154: 145–152, 1988.
ROTT, O., U. TONTSCH, B. FLEISCHER, AND E. CASH. Interleukin-6 production in ‘‘normal’’ and HTLV-1 tax-expressing brainspecific endothelial cells. Eur. J. Immunol. 23: 1987–1991, 1993.
RUBIO, N. Demonstration of the presence of an interleukin-1 re-
/ 9j0c$$oc11
P13-8
715.
716.
717.
718.
719.
721.
722.
723.
724.
725.
726.
727.
728.
729.
730.
731.
732.
733.
734.
735.
736.
11-25-98 11:16:36
63
ceptor on the surface of murine astrocytes and its regulation by
cytokines and Theilers virus. Immunology 82: 178–183, 1994.
RUTANEN, E.-M. Cytokines in reproduction. Trends Mol. Med.
25: 343–347, 1993.
RUZEK, M. C., A. H. MILLER, S. M. OPAL, B. D. PEARCE, AND
C. A. BIRON. Characterization of early cytokine responses and an
interleukin (IL)-6-dependent pathway of endogenous glucocorticoid induction during murine cytomegalovirus infection. J. Exp.
Med. 185: 1185–1192, 1997.
RUZICKA, B. B., AND H. AKIL. Differential cellular regulation of
pro-opiomelanocortin by interleukin-1-beta and corticotropin-releasing hormone. Neuroendocrinology 61: 136–151, 1995.
SAGAR, S. M., K. J. PRICE, N. W. KASTING, AND F. R. SHARP.
Anatomic patterns of FOS immunostaining in rat brain following
systemic endotoxin administration. Brain Res. Bull. 36: 381–392,
1995.
SAIJA, A., P. PRINCI, M. LANZA, M. SCALESSE, E. ARAMNEJAD,
AND A. DE SARRO. Systemic administration can affect blood-brain
barrier permeability. Life Sci. 56: 775–784, 1995.
SAKAMOTO, Y., K. KOIJE, H. KIYAMA, K. KONISHI, K. WATANABE, Y. OSAKO, K. HIROTA, AND A. MIYAKE. Endotoxin activates a chemokinergic neuronal pathway in the hypothalamo-pituitary system. Endocrinology 137: 4503–4506, 1996.
SALAS, M. A., S. W. EVANS, M. J. LEVELL, AND J. T. WHICHER.
Interleukin-6 and ACTH act synergistically to stimulate the release
of corticosterone from adrenal gland cells. Clin. Exp. Immunol.
79: 470–473, 1990.
SANDBERG, A. C., C. ENGBERG, M. LAKE, H. VON HOLST, AND
V. R. SARA. The expression of insulin-like growth factor I and
insulin-like growth factor II genes in the human fetal and adult
brain and in glioma. Neurosci. Lett. 93: 114–119, 1988.
SANDI, C., AND C. GUAZA. Evidence for a role of nitric oxide in
the corticotropin-releasing factor release induced by interleukin1b. Eur. J. Pharmacol. 274: 17–23, 1995.
SAPHIER, D. Neurophysiological and endocrine consequences of
immune activity. Psychoneuroendocrinology 14: 63–87, 1989.
SAPHIER, D. Neuroendocrine effects of interferon-alpha in the
rat. Adv. Exp. Med. Biol. 373: 209–218, 1995.
SAPHIER, D., AND S. FELDMAN. Adrenoceptor specificity in the
central regulation of adrenocortical secretion. Neuropharmacology 28: 1231–1237, 1989.
SAPHIER, D., AND H. OVADIA. Selective facilitation of putative
corticotropin-releasing factor-secreting neurons by interleukin-1.
Neurosci. Lett. 114: 283–288, 1990.
SAPHIER, D., S. C. ROERIG, C. ITO, W. R. VLASAK, G. E. FARRAR, J. E. BROYLES, AND J. E. WELCH. Inhibition of neural and
neuroendocrine activity by alpha-interferon: neuroendocrine,
electrophysiological, and biochemical studies in the rat. Brain
Behav. Immun. 8: 37–56, 1994.
SAPOLSKY, R., C. RIVIER, G. YAMAMOTO, P. PLOTSKY, AND
W. VALE. Interleukin-1 stimulates the secretion of hypothalamic
corticotropin-releasing factor. Science 238: 522–524, 1987.
SARKAR, D. K., K. H. KIM, AND S. MINAMI. Transforming growth
factor-b1 messenger RNA and protein expression in the pituitary
gland: its action on prolactin secretion and lactotropic growth.
Mol. Endocrinol. 6: 1825–1833, 1992.
SARLIS, N. J., A. STEPHANOU, R. A. KNIGHT, S. L. LIGHTMAN,
AND H. S. CHOWDREY. Effects of glucocorticoids and chronic
inflammatory stress upon anterior pituitary interleukin-6 mRNA
in the rat. Br. J. Rheumatol. 32: 653–657, 1993.
SATO, N., AND A. MIYAJIMA. Multimeric cytokine receptors: common versus specific functions. Curr. Opin. Cell Biol. 6: 174–179,
1994.
SAUER, J., E. ARZT, H. GUMPRECHT, U. HOPFNER, AND G. K.
STALLA. Expression of interleukin-1 receptor antagonist in human pituitary adenomas in vitro. J. Clin. Endocrinol. Metab. 79:
1857–1863, 1994.
SAWADA, M., Y. ITOH, A. SUZUMURA, AND T. MARUNOUCHI.
Expression of cytokine receptors in cultured neuronal and glial
cell lines. Neurosci. Lett. 160: 131–134, 1993.
SAWADA, M., A. SUZUMURA, Y. ITOH, AND T. MARUNOUCHI.
Production of interleukin-5 by mouse astrocytes and microglia in
culture. Neurosci. Lett. 155: 175–178, 1993.
pra
APS-Phys Rev
Downloaded from on April 23, 2014
703.
REGULATION OF HPA AXIS BY CYTOKINES
64
ANDREW V. TURNBULL AND CATHERINE L. RIVIER
/ 9j0c$$oc11
P13-8
755.
756.
757.
758.
759.
760.
761.
762.
763.
764.
765.
766.
767.
768.
769.
770.
771.
772.
773.
774.
11-25-98 11:16:36
receptor antagonist protein and mRNA in the rat adrenal gland.
J. Interferon Cytokine Res. 15: 721–729, 1995.
SCHWARTZ, G. J., C. R. PLATA-SALAMAN, AND W. LANGHANS.
Subdiaphragmatic vagal deafferentation fails to block feeding suppressive effects of LPS and IL-1 beta in rats. Am. J. Physiol. 273
(Regulatory Integrative Comp. Physiol. 42): R1193–R1198, 1997.
SCHWARTZ, J., AND D. W. RAY. Leukemia inhibitory factor as an
intrapituitary mediator of ACTH secretion in normal sheep anterior pituitary. Proc. Annu. Meet. Endocr. Soc. 79th Minneapolis
MN 1997, p. 2–161.
SCHWEIGERER, L., G. NEUFELD, J. FRIEDMAN, J. A. ABRAHAM, J. C. FIDDES, AND D. GOSPODAROWICZ. Basic fibroblast
growth factor: production and growth stimulation in cultured adrenal cortex cells. Endocrinology 120: 796–800, 1987.
SCOTT, G. M., R. J. WARD, D. J. WRIGHT, J. A. ROBINSON, J. K.
ONWUBALILI, AND C. L. GAUCI. Effects of cloned interferon a2
in normal volunteers: febrile reactions and changes in circulating
corticosteroids and trace metals. Antimicrob. Agents Chemother.
23: 589–592, 1983.
SEGRETI, J., G. GHEUSI, R. DANTZER, K. W. KELLEY, AND R. W.
JOHNSON. Defect in interleukin-1beta secretion prevents sickness behavior in C3H/HeJ mice. Physiol. Behav. 61: 873–878, 1997.
SEHGAL, P. B. Interleukin-6-type cytokines in vivo: regulated bioavailability. Proc. Soc. Exp. Biol. Med. 213: 238–247, 1996.
SEHIC, E., AND C. M. BLATTEIS. Blockade of lipopolysaccharideinduced fever by subdiaphragmatic vagotomy in guinea pigs.
Brain Res. 726: 160–166, 1996.
SEI, Y., L. VITKOVIC, AND M. M. YOKOYAMA. Cytokines in the
central nervous system: regulatory roles in neuronal function, cell
death and repair. Neuroimmunomodulation 2: 121–133, 1995.
SELLMEYER, D. E., AND C. GRUNFIELD. Endocrine and metabolic disturbances in human immunodeficiency virus infection and
the acquired immune deficiency syndrome. Endocr. Rev. 17: 518–
532, 1996.
SELYE, H. A syndrome produced by diverse nocuous agents. Nature 138: 32, 1936.
SELYE, H. Thymus and adrenals in the response of the organism
to injuries and intoxications. Br. J. Exp. Pathol. 17: 234–248, 1936.
SHALABY, M. R., A. WAAGE, L. A. AARDEN, AND T. ESPEVICK.
Endotoxin, tumor necrosis factor a and interleukin-1 induce interleukin-6 production in vivo. Clin. Immunol. Immunopathol.
53: 3131–3133, 1989.
SHANKS, N., D. FRANCIS, S. ZALCMAN, M. J. MEANEY, AND H.
ANISMAN. Alterations in central catecholamines associated with
immune responding in adult and aged mice. Brain Res. 666: 77–
87, 1994.
SHARIEF, M. K., M. CIARDI, AND E. J. THOMPSON. Blood-brain
barrier damage in patients with bacterial meningitis: association
with tumor necrosis factor-a but not interleukin-1b. J. Infect. Dis.
166: 350–358, 1992.
SHARIEF, M. K., AND E. J. THOMPSON. In vivo relationship of
tumor necrosis factor-a to blood-brain barrier damage in patients
with active multiple sclerosis. J. Neuroimmunol. 38: 27–34, 1992.
SHARIF, S. F., R. J. HAIRI, V. A. CHANG, P. S. BARIE, R. S.
WANG, AND J. B. GHAJAR. Human astrocyte production of tumor
necrosis factor-alpha, interleukin-1 beta, and interleukin-6 following exposure to lipopolysaccharide endotoxin. Neurol. Res. 15:
109–112, 1993.
SHARP, B. M., AND S. G. MATTA. Prostaglandins mediate the adrenocorticotropin response to tumor necrosis factor in rats. Endocrinology 132: 269–274, 1993.
SHARP, B. M., S. G. MATTA, P. K. PETERSON, R. NEWTON, C.
CHAO, AND K. MCALLEN. Tumor necrosis factor-a is a potent
ACTH secretagogue: comparison with interleukin-1b. Endocrinology 124: 3131–3133, 1989.
SHIMIZU, H., Y. UEHARA, Y. SHIMOMURA, M. NEGISHI, S. TAKAHASHI, I. KOBAYASHI, AND S. KOBAYASHI. Effects of recombinant human interleukin-1 beta on hypothalamic monoamine metabolism of rats. Biogenic Amines 7: 517–523, 1990.
SHIMON, I., D. W. RAY, AND S. MELMED. Cytokine-dependent
gp130 receptor subunit regulates human fetal pituitary adrenocorticotropin hormone and growth hormone secretion. J. Clin. Invest. 100: 357–363, 1997.
pra
APS-Phys Rev
Downloaded from on April 23, 2014
737. SAWADA, T., K. KOIKE, Y. KANDA, Y. SAKAMOTO, A. NOHARA,
M. OHMICHI, K. HIROTA, AND A. MIYAKE. In vitro effects of cinc/
gro, a member of the interleukin-8 family, on hormone secretion
by rat anterior pituitary cells. Biochem. Biophys. Res. Commun.
202: 155–160, 1994.
738. SAWCHENKO, P. E., E. R. BROWN, R. K. W. CHAN, A. ERICSSON, H.-Y. LI, B. L. ROLAND, AND K. J. KOVACS. The paraventricular nucleus of the hypothalamus and functional neuroanatomy of
visceromotor responses to stress. Prog. Brain Res. 107: 201–222,
1996.
739. SAWCHENKO, P. E., E. T. J. CUNNINGHAM, J. C. BITTENCOURT, AND R. K. W. CHAN. Aminergic and peptidergic pathways
subserving the stress response. In: Stress: Neuroendocrine and
Molecular Approaches, edited by R. Kvetnansky, R. McCarty, and
J. Axelrod. New York: Gordon & Breach, 1992, p. 15–27.
740. SAWCHENKO, P. E., L. W. SWANSON, AND W. W. VALE. Co-expression of CRF- and vasopressin-immunoreactivity in parvocellular neurosecretory neurons in the adrenalectomized rat. Proc.
Natl. Acad. Sci. USA 81: 1883–1887, 1984.
741. SCACCIANOCE, S., G. CIGLIANA, R. NICOLAI, L. A. MUSCULO,
A. PORCU, D. NAVARRA, J. R. PEREZ-POLO, AND L. ANGELUCCI.
Hypothalamic involvement in the activation of the pituitary-adrenocortical axis by nerve growth factor. Neuroendocrinology 58:
202–209, 1993.
742. SCAMMELL, T. E., J. K. ELMQUIST, J. D. GRIFFIN, AND C. B.
SAPER. Ventromedial preoptic prostaglandin E2 activates feverproducing autonomic pathways. J. Neurosci. 16: 6246–6254, 1996.
743. SCHINDLER, R., J. MANCILLA, S. ENDRES, R. GHORBANI, S. C.
CLARK, AND C. A. DINARELLO. Correlations and interactions in
the production of interleukin-6 (IL-6), IL-1, and tumor necrosis
factor in human blood mononuclear cells: IL-6 supresses IL-1 and
TNF. Blood 75: 40–47, 1990.
744. SCHMIDT, E. D., A. W. J. W. JANSZEN, F. G. WOUTERLOOD, AND
F. J. H. TILDERS. Interleukin-1-induced long-lasting changes in
hypothalamic corticotropin-releasing hormone (CRH)-neurons
and hyperresponsiveness of the hypothalamus-pituitary-adrenal
axis. J. Neurosci. 15: 7417–7426, 1995.
745. SCHOBITZ, B., E. R. DE KLOET, AND F. HOLSBOER. Gene expression and function of interleukin 1, interleukin 6 and tumor necrosis factor in the brain. Prog. Neurobiol. 44: 397–432, 1994.
746. SCHOBITZ, B., E. R. DE KLOET, W. SUTANTO, AND F. HOLSBOER. Cellular localization of interleukin-6 mRNA and interleukin-6 receptor mRNA in rat brain. Eur. J. Neurosci. 5: 1426–1435,
1993.
747. SCHOBITZ, B., G. PEZESHKI, T. POHL, U. HEMMANN, P. C. HEINRICH, F. HOLSBOER, AND M. H. M. REUL. Soluble interleukin6 (IL-6) receptor augments central effects of IL-6 in vivo. FASEB
J. 9: 659–664, 1995.
748. SCHOBITZ, B., M. VAN DEN DOBBELSTEEN, F. HOLSBOER, W.
SUTANTO, AND E. R. DE KLOET. Regulation of interleukin-6 gene
expression in rat. Endocrinology 132: 1569–1576, 1993.
749. SCHOBITZ, B., D. A. M. VOORHUIS, AND E. R. DE KLOET. Localization of interleukin-6 mRNA and interleukin 6 receptor mRNA
in rat brain. Neurosci. Lett. 136: 189–192, 1992.
750. SCHOTANUS, K., G. B. MAKARA, F. J. H. TILDERS, AND F. BERKENBOSCH. ACTH response to a low dose but not a high dose
of bacterial endotoxin in rats is completely mediated by corticotropin-releasing hormone. Neuroimmunomodulation 1: 300–307,
1994.
751. SCHOTANUS, K., R. H. MELOEN, W. C. PUJIK, F. BERKENOBOSCH, R. BINNEKADE, AND F. J. H. TILDERS. Effects of monoclonal antibodies to specific epitopes of rat interleukin-1 beta (IL1b) on IL-1b-induced ACTH corticosterone and IL-6 responses in
rats. J. Neuroendocrinol. 7: 255–262, 1995.
752. SCHOTANUS, K., F. J. TILDERS, AND F. BERKENBOSCH. Human
recombinant interleukin-1 receptor antagonist prevents adrenocorticotropin, but not interleukin-6 responses to bacterial endotoxin. Endocrinology 133: 2461–2468, 1993.
753. SCHULTZBERG, M., C. ANDERSSON, A. UNDEN, M. TROYEBLOMBERG, S. B. SVENSON, AND T. BARTFAI. Interleukin-1 in
adrenal chromaffin cells. Neuroscience 30: 805–810, 1989.
754. SCHULTZBERG, M., S. TINGSBORG, S. NOBEL, J. LUNDKVIST,
S. SVENSON, A. SIMONCSITS, AND T. BARTFAI. Interleukin-1
Volume 79
January 1999
REGULATION OF HPA AXIS BY CYTOKINES
/ 9j0c$$oc11
P13-8
794.
795.
796.
797.
798.
799.
800.
801.
802.
803.
804.
805.
806.
807.
808.
809.
810.
811.
11-25-98 11:16:36
corticosterone in hypophysectomized mice: a possible lymphoid
adrenal axis. Science 218: 1311–1312, 1982.
SMITH, E. M., M. PHAN, T. E. KRUGER, D. H. COPPENHAVER,
AND J. E. BLALOCK. Human lymphocyte production of immunoreactive thyrotropin. Proc. Natl. Acad. Sci. USA 80: 6010–6013, 1983.
SMITH, J. W., W. J. URBA, B. D. CURTI, L. J. ELWOOD, R. G.
STEIS, J. E. JANIK, W. H. SHARFMAN, L. L. MILLER, R. G. FENTON, K. C. CONLON, M. SZNOL, S. P. CREEKMORE, N. F. WELLS,
F. W. RUSCETTI, J. R. KELLER, K. HESTDAL, M. SHIMUZU, J.
ROSSIO, W. G. ALVORD, J. J. OPPENHEIM, AND D. L. LONGO.
The toxic and hematologic effects of interleukin-1 alpha administered in a phase I trial to patients with advanced malignancies. J.
Clin. Oncol. 10: 1141–1152, 1992.
SMITH, L. R., S. L. BROWN, AND J. E. BLALOCK. Interleukin-2 induction of ACTH secretion: presence of an interleukin-2 receptor
alpha-chain-like molecule on pituitary cells. J. Neuroimmunol.
21: 249–254, 1989.
SMITH, M. A., S. MAKINO, S. Y. KIM, AND R. KVETNANSKY. Stress
increases brain-derived neurotropic factor messenger ribonucleic
acid in the hypothalamus and pituitary. Endocrinology 136: 3743–
3750, 1995.
SMITH, M. A., S. MAKINO, R. KVETNANSKY, AND R. M. POST.
Stress and glucocorticoids affect the expression of brain-derived
neurotrophic and neurotrophin-3 mRNAs in the hippocampus. J.
Neurosci. 15: 1768–1777, 1995.
SMITH, T., A. K. HEWSON, L. QUARRIE, J. P. LEONARD, AND
M. L. CUZNER. Hypothalamic PGE2 and cAMP production and
adrenocortical activation following intraperitoneal endotoxin injection: in vivo microdialysis studies in Lewis and Fischer rats.
Neuroendocrinology 59: 396–405, 1994.
SOSZYNSKI, D., W. KOZAK, C. A. CONN, K. RUDOLPH, AND M. J.
KLUGER. Beta-adrenoceptor antagonists suppress elevation in
body temperature and increase in plasma IL-6 in rats exposed to
open field. Neuroendocrinology 63: 459–467, 1996.
SPANGELO, B. L., P. D. DEHOLL, L. KALABAY, B. R. BOND, AND
P. ARNAUD. Neurointermediate pituitary lobe cells synthesize and
release interleukin-6 in vitro: effects of lipopolysaccharide and
interleukin-1b. Endocrinology 135: 556–563, 1994.
SPANGELO, B. L., AND W. C. GOROSPE. Role of cytokines in the
neuroendocrine-immune system axis. Front. Neuroendocrinol. 16:
1–22, 1995.
SPANGELO, B. L., P. C. ISAKSON, AND R. M. MACLEOD. Production of interleukin-6 by anterior pituitary cells is stimulated by
increased intracellular adenosine 3*,5*-monophosphate and vasoactive intestinal peptide. Endocrinology 127: 2886–2894, 1990.
SPANGELO, B. L., AND W. D. JARVIS. Lysophosphatidylcholine
stimulates interleukin-6 release from rat anterior pituitary cells in
vitro. Endocrinology 137: 4419–4426, 1996.
SPANGELO, B. L., W. D. JARVIS, A. M. JUDD, AND R. M. MACLEOD. Induction of interleukin-6 release by interleukin-1 in rat
anterior pituitary cells in vitro: evidence for an eicosanoid-dependent mechanism. Endocrinology 129: 2886–2894, 1991.
SPANGELO, B. L., A. M. JUDD, P. C. ISAKSON, AND R. M. MACLEOD. Interleukin-1 stimulates interleukin-6 release from rat anterior pitutary cells in vitro. Endocrinology 128: 2685–2692, 1991.
SPANGELO, B. L., R. M. MACLEOD, AND P. C. ISAKSON. Production of interleukin-6 by anterior pituitary cells in vitro. Endocrinology 126: 582–586, 1990.
SPANGELO, B. L., AND R. M. WRIGHT. Arachidonic acid stimulates interleukin-6 release from rat anterior pituitary cells in vitro:
comparison with peritoneal macrophages. Prog. Neuroendocrinol.
Immunol. 5: 235–243, 1992.
SPATH-SCHWALBE, E., J. BORN, H. SCHREZENMEIER, S. R.
BORNSTEIN, P. STROMEYER, S. DRESCHSLER, AND H.-L.
FEHM. Interleukin-6 stimulates the hypothalamo-pituitary-adrenocortical axis in man. J. Clin. Endocrinol. Metab. 79: 1212–1214,
1994.
SPATH-SCWALBE, E., F. PORZOLT, W. DIGEL, J. BORN, B.
KLOSS, AND H. L. FEHM. Elevated plasma cortisol levels during
interferon-gamma treatment. Immunopharmacology 17: 141–145,
1989.
SPILLANTINI, M. G., L. ALOE, E. ALLEVA, R. DE SIMONE, M.
GOEDERT, AND R. LEVI-MONTALCINI. Nerve growth factor
pra
APS-Phys Rev
Downloaded from on April 23, 2014
775. SHIMON, I., X. YAN, D. W. RAY, AND S. MELMED. Human fetal
pituitary cells express functional gp130-related cytokine-specific
receptors: regulation of ACTH and growth hormone secretion.
Proc. Annu. Meet. Endocr. Soc. 79th Minneapolis MN 1997, p.
OR42–5.
776. SHINTANI, F., S. KANBA, T. NAKAKI, M. NIBUYA, N. KINOSHITA,
E. SUZUKI, G. YAGI, R. KATO, AND M. ASAI. Interleukin-1b augments release of norepinephrine, dopamine, and serotonin in the
rat anterior hypothalamus. J. Neurosci. 13: 3574–3581, 1993.
777. SHINTANI, F., T. NAKAI, S. KANBA, R. KATO, AND M. ASAI. Role
of interleukin-1 in stress responses. A putative neurotransmitter.
Mol. Neurobiol. 10: 47–71, 1995.
778. SHINTANI, F., T. NAKAI, S. KANBA, S. KOICHI, G. YAGI, M.
SHIOZAWA, S. ALSO, R. KATO, AND M. ASAI. Involvement of
interleukin-1 in immobilization stress-induced increase in plasma
adrenocorticotropic hormone and in release of hypothalamic catecholamines in the rat. J. Neurosci. 15: 1961–1970, 1995.
779. SHIZUYA, K., T. KOMORI, R. FUJIWARA, S. MIYAHARA, M. OHMORI, AND J. NOMURA. The influence of restraint stress on the
exposure of mRNAs for IL-6 and the IL-6 receptor in the hypothalamus and midbrain of the rat. Life Sci. 61: 135–140, 1997.
780. SHOHAMI, E., R. BASS, D. WALLACH, A. YAMIN, AND R. GALLILY.
Inhibition of tumor necrosis factor alpha (TNFa) activity in rat
brain is associated with cerebroprotection after closed head injury. J. Cereb. Blood Flow Metab. 16: 378–384, 1996.
781. SHUKLA, A., M. DIKSHIT, AND R. C. SRIMAL. Nitric oxide modulates blood-brain barrier permeability during infections with an
activated bacterium. Neuroreport 6: 1629–1632, 1995.
782. SIAUD, P., M. MEKAOUCHE, G. IXART, M. BALMEFREZOL, L.
GIVALOIS, G. BARBANEL, AND I. ASSENMACHER. A subpopulation of corticotropin-releasing hormone neurosecretory cells in
the paraventricular nucleus of the hypothalamus also contain
NADPH-diaphorase. Neurosci. Lett. 170: 51–54, 1994.
783. SILVERMAN, A. J., D. L. HOFFMAN, AND E. A. ZIMMERMAN. The
descending afferent connections of the paraventricular nucleus
of the hypothalamus (PVN). Brain Res. Bull. 6: 47–61, 1981.
784. SIMS, J. E., AND S. K. DOWER. Interleukin-1 receptors. Eur. Cytokine Netw. 5: 539–546, 1994.
785. SIMS, J. E., M. A. GAYLE, J. L. SLACK, M. R. ALDERSON, T. A.
BIRD, J. G. GIRI, F. COLOTTA, F. RE, A. MANTOVANI, K. SHANEBECK, K. H. GRABSTEIN, AND S. K. DOWER. Interleukin-1 signaling occurs exclusively via the type I receptor. Proc. Natl. Acad.
Sci. USA 90: 6155–6159, 1993.
786. SIMS, J. E., J. G. GIRI, AND S. K. DOWER. The two interleukin1 receptors play different roles in IL-1 actions. Clin. Immunol.
Immunopathol. 72: 9–14, 1994.
787. SIPE, K. J., D. SRISAWASDI, R. DANTZER, K. W. KELLEY, AND
J. A. WEYHENMEYER. An endogenous 55 kDa TNF receptor mediates cell death in a neural cell line. Brain Res. 38: 222–232,
1996.
788. SIPPY, B. D., F. M. HOFMAN, D. WALLACH, AND D. R. HINTON.
Increased expression of tumor necrosis factor-a receptors in the
brains of patients with AIDS. J. Acquired Immune Deficiency
Syndrome Hum. Retrovirol. 10: 511–521, 1995.
789. SIRKO, S., I. BISHAI, AND F. COCEANI. Prostaglandin formation
in the hypothalamus in vivo: effect of pyrogens. Am. J. Physiol.
263 (Regulatory Integrative Comp. Physiol. 32): R616–R624,
1989.
790. SMAGIN, G. N., A. H. SWIERGIEL, AND A. J. DUNN. Peripheral
administration of interleukin-1 increases extracellular concentrations of norepinephrine in rat hypothalamus: comparison with
plasma corticosterone. Psychoneuroendocrinology 21: 83–93,
1996.
791. SMITH, B. K., AND M. J. KLUGER. Human IL-1 receptor antagonist
partially suppresses LPS fever but not plasma levels of IL-6 in
Fisher rats. Am. J. Physiol. 263 (Regulatory Integrative Comp.
Physiol. 32): R653–R655, 1992.
792. SMITH, E. M., AND J. E. BLALOCK. Human lymphocyte production
of corticotropin and endorphin-like substances: association with
leukocyte interferon. Proc. Natl. Acad. Sci. USA 78: 7530–7534,
1981.
793. SMITH, E. M., W. J. MEYER, AND J. E. BLALOCK. Virus-induced
65
66
ANDREW V. TURNBULL AND CATHERINE L. RIVIER
/ 9j0c$$oc11
P13-8
828. SUZUKI, E., F. SHINTANI, S. KANBA, M. ASAI, AND T. NAKAKI.
Immobilization stress increases levels of interleukin-1 receptor
antagonist in various rat brain regions. Cell. Mol. Neurobiol. 17:
557–562, 1997.
829. SVANES, K. Diphasic increase in vascular permeability in turpentine induced inflammation in skin and musculature of mice. Acta
Pathol. Microbiol. Scand. A79: 335–344, 1971.
830. SWEEP, C. G. J. F., M. J. M. VAN DER MEER, A. R. M. M. HERMUS, A. G. H. SMALS, J. W. M. VAN DER MEER, G. J. PESMAN,
S. J. WILLEMSEN, T. J. BENRAAD, AND P. W. C. KLOPPENBORG.
Chronic stimulation of the pituitary-adrenal axis in rats by interleukin-1b infusion: in vivo and in vitro studies. Endocrinology
130: 1153–1164, 1992.
831. SWIERGIEL, A. H., A. J. DUNN, AND E. A. STONE. The role of
cerebral noradrenergic systems in the fos response to interleukin1. Brain Res. Bull. 41: 61–64, 1996.
832. SZAFARCZYK, A., G. ALONSO, G. IXART, F. MALAVAL, AND I.
ASSENMACHER. Diurnal-stimulated and stress induced ACTH release is mediated by ventral noradrenergic bundle. Am. J. Physiol.
249 (Endocrinol. Metab. 12): E219–E226, 1985.
833. SZAFARCZYK, A., V. GUILLAUME, B. CONTE-DEVOLX, G.
ALONSO, F. MALAVAL, N. PARES-HERBUTE, C. OLIVER, AND
I. ASSENMACHER. Central catecholaminergic system stimulates
secretion of CRH at different sites. Am. J. Physiol. 255 (Endocrinol. Metab. 18): E463–E468, 1988.
834. TADA, M., A.-C. DISERENS, I. DESBAILLETS, AND N. DE
TRIBOLET. Analysis of cytokine receptor messenger RNA expression in human glioblastoma cells and normal astrocytes by reverse
transcription polymerase chain reaction. J. Neurosurg. 80: 1063–
1073, 1994.
835. TAGA, T., K. KAWANISHI, R. R. HARDY, T. HIRANO, AND T. KISHIMOTO. Receptors for B cell stimulatory factor. 2. Quantification, specificity, distribution and regulation of their expression. J.
Exp. Med. 166: 967–981, 1987.
836. TAGLIALATELA, G., L. ANGELUCCI, S. SCACCIANOCE, P. J.
FOREMAN, AND J. R. PEREZ-POLO. Nerve growth factor modulates the activation of the hypothalamo-pituitary-adrenocortical
axis during the stress response. Endocrinology 129: 2212–2218,
1991.
837. TAGOH, H., H. NISHIJO, T. UWANO, H. KISHI, T. ONO, AND A.
MURAGUCHI. Reciprocal IL-1b gene expression in medial and
lateral hypothalamic areas in SART-stressed mice. Neurosci. Lett.
184: 17–20, 1995.
838. TAISHI, P., S. BREDOW, N. GUHA-THAKURTA, F. O. OBAL, AND
J. M. KRUEGER. Diurnal variations of interleukin-1b mRNA and
b-actin mRNA in rat brain. J. Neuroimmunol. 75: 69–74, 1997.
839. TAKAHASHI, S., L. KAPAS, J. FANG, AND J. M. KRUEGER. An
anti-tumor necrosis factor antibody suppresses sleep in rats and
rabbits. Brain Res. 690: 241–244, 1995.
840. TAKAO, T., S. G. CULP, AND E. B. DE SOUZA. Reciprocal modulation of interleukin-1b (IL-1b) and IL-1 receptors by lipopolysaccharide (endotoxin) treatment in the mouse brain-endocrine-immune axis. Endocrinology 132: 1497–1504, 1993.
841. TAKAO, T., S. G. CULP, R. C. NEWTON, AND E. B. DE SOUZA.
Type I interleukin-1 receptors in the mouse brain-endocrine-immune axis labeled with [125I]recombinant human interleukin-1 receptor antagonist. J. Neuroimmunol. 41: 51–60, 1992.
842. TAKAO, T., W. M. MITCHEL, AND E. B. DE SOUZA. Interleukin-1
receptors in mouse kidney: identification, localization, and modulation by lipopolysaccharide treatment. Endocrinology 128: 2618–
2624, 1991.
843. TAKAO, T., H. NAKATA, C. TOJO, H. KUROKAWA, T. NISHIOKA,
K. HASHIMOTO, AND E. B. DE SOUZA. Regulation of interleukin1 receptors and hypothalamic-pituitary-adrenal axis by lipopolysaccharide treatment in the mouse. Brain Res. 649: 265–270, 1994.
844. TAKAO, T., W. NANAMIYA, T. TAKEMURE, M. NISHIYAMA, K.
ASABA, S. MAKINO, K. HASHIMOTO, AND E. B. DE SOUZA. Endotoxin induced increases in rat plasma pituitary-adrenocortical hormones are better reflected by alterations in tumor necrosis factora than interleukin-1b. Life Sci. 61: 263–268, 1997.
845. TAKAO, T., R. C. NEWTON, AND E. B. DE SOUZA. Species differences [125I]interleukin-1 binding in brain, endocrine and immune
tissues. Brain Res. 623: 172–176, 1993.
11-25-98 11:16:36
pra
APS-Phys Rev
Downloaded from on April 23, 2014
mRNA and protein increase in hypothalamus in a mouse model
of aggression. Proc. Natl. Acad. Sci. USA 86: 8555–8559, 1989.
812. SPINAZZE, S., S. VIVIANI, P. BIDOLI, F. ROVELLI, P. PALMER,
C. R. FRANKS, F. ARIENTI, L. RIVOLTINI, AND G. PARMIANI.
Effect of prolonged subcutaneous administration of interleukin-2
on the circadian rhythms of cortisol and beta-endorphin in advanced small cell lung cancer patients. Tumor 77: 496–499, 1991.
813. SPINEDI, E., R. HADID, T. DANEVA, AND R. GAILLARD. Cytokines stimulate the CRH but not the vasopressin neuronal system:
evidence for a median eminence site of interleukin-6 action. Neuroendocrinology 56: 46–53, 1992.
814. SPIVAK, B., B. SHOHAT, R. MESTER, S. AVRAHAM, I. GIL-AD,
A. BLEICH, A. VALEVSKI, AND A. WEIZMAN. Elevated levels of
serum interleukin-1 beta in combat related posttraumatic stress
disorder. Biol. Psychiatry 42: 345–348, 1997.
815. SPRENGER, H., C. JACOBS, M. NAIN, A. M. GRESSNER, H.
PRINZ, W. WESEMAN, AND S. GERNSA. Enhanced release of cytokines, interleukin-2 receptors, and neopterin after long-distance
running. Clin. Immunol. Immunopathol. 63: 188–195, 1992.
816. STEFANA, B., D. W. RAY, AND S. MELMED. Leukemia inhibitory
factor induces differentiation of pituitary corticotroph function:
an immuno-neuroendocrine phenotypic switch. Proc. Natl. Acad.
Sci. USA 93: 12502–12506, 1996.
817. STEFFERL, A., S. J. HOPKINS, N. J. ROTHWELL, AND G. N.
LUHESHI. The role of TNF-a in fever: opposing actions of human
and murine TNF-a and interactions with IL-1b in the rat. Br. J.
Pharmacol. 118: 1919–1924, 1996.
818. STEPHANOU, A., N. J. SARLIS, R. A. KNIGHT, S. L. LIGHTMAN,
AND H. S. CHOWDREY. Effects of cyclosporin A on the hypothalamo-pituitary-adrenal axis and anterior pituitary interleukin-6
mRNA expression during chronic inflammatory stress in the rat.
J. Neuroimmunol. 41: 215–222, 1992.
819. STEPIEN, H., G. ZEREK-MEFEN, S. MUCHA, K. WINCZYK, AND
J. FRYCZAK. Interleukin-1b stimulates cell proliferation in the
intermediate lobe of the rat pituitary gland. J. Endocrinol. 140:
337–341, 1994.
820. STITT, J. T. Evidence for the involvement of the organum vasculosum laminae terminalis in the febrile response of rabbits and rats.
J. Physiol. (Lond.) 368: 501–511, 1985.
821. STITT, J. T. Passage of immunomodulators across the blood-brain
barrier. Yale J. Biol. Med. 63: 121–131, 1990.
822. STITT, J. T. Prostaglandin, the OVLT and fever. In: Neuroimmunology of Fever, edited by T. Bartfai and D. Ottoson. Oxford, UK:
Pergamon, 1992, p. 155–165.
823. STOCKLI, K. A., L. E. LILLIEN, M. NAHER-NOE, G. BREITFELD,
R. A. HUGHES, M. C. RAFF, H. THOENEN, AND M. SENDTNER.
Regional distribution, developmental changes, and cellular localization of CNTF-mRNA and protein in the rat brain. J. Cell Biol.
115: 447–459, 1991.
823a. ST. PIERRE, B. A., D. A. GRANGER, J. L. WONG, AND J. E. MERRIL. A study of tumor necrosis factor, tumor necrosis factor receptors, and nitric oxide in human fetal glial cells. Adv. Pharmacol.
34: 415–438, 1995.
824. SUDA, T., F. TOZAWA, T. USHIYAMA, T. SUMITOMO, M. YAMADA, AND H. DEMURA. Interleukin-1 stimulates corticotropinreleasing factor gene expression in rat hypothalamus. Endocrinology 126: 1223–1228, 1990.
825. SUDA, T., F. TOZAWA, T. USHIYAMA, N. TOMORI, T. SUMITOMO, Y. NAKAGAMI, M. YAMADA, H. DEMUA, AND K. SHIZUME. Effects of protein kinase-C-related adrenocorticotropin secretagogues and interleukin-1 on proopiomelanocortin gene expression in rat anterior pituitary cells. Endocrinology 124: 1444–
1449, 1989.
826. SUGIMOTO, Y., R. SHIGEMOTO, T. NAMBA, M. NEGISHI, N. MIZUNO, S. NARUMIYA, AND A. ICHIKAWA. Distribution of the messenger RNA for the prostaglandin E receptor subtype EP3 in the
mouse central nervous system. Neuroscience 62: 919–928, 1994.
827. SUNDAR, S. K., M. A. CIERPIAL, L. S. KAMARAJU, S. LONG, S.
HSIEH, C. LORENZ, M. AARON, J. C. RITCHIE, AND J. M. WEISS.
Human immunodeficiency virus glycoprotein (gp120) infused into
rat brain induces interleukin-1 to elevate pituitary-adrenal activity
and decrease peripheral cellular immune responses. Proc. Natl.
Acad. Sci. USA 88: 11246–11250, 1991.
Volume 79
January 1999
REGULATION OF HPA AXIS BY CYTOKINES
/ 9j0c$$oc11
P13-8
865.
866.
867.
868.
869.
870.
871.
872.
873.
874.
875.
876.
877.
878.
879.
880.
881.
882.
11-25-98 11:16:36
typic plasticity of CRF neurons during stress. Ann. NY Acad. Sci.
697: 39–52, 1993.
TINGSBORG, S., M. ZETTERSTROM, K. ALHEIM, H. HASANVAN,
M. SCHULTZBERG, AND T. BARTFAI. Regionally specific induction of ICE mRNA and enzyme activity in the rat brain and adrenal
gland by LPS. Brain Res. 712: 153–158, 1996.
TINGSBORG, S., M. ZIOLKOWSKA, M. ZETTERSTROM, H. HASANVAN, AND T. BARTFAI. Regulation of ICE activity and ICE
isoforms by LPS. Mol. Psychiatry 2: 122–124, 1997.
TOMINAGA, T., J. FUKATA, Y. NAITO, T. USUI, N. MURAKAMI,
M. FUKUSHIMA, Y. NAKAI, Y. HIRAI, AND H. IMURA. Prostaglandin-dependent in vitro stimulation of adrenocortical steroidogenesis by interleukins. Endocrinology 128: 526–531, 1991.
TOMOZAWA, Y., T. INOUE, AND M. SATOH. Expression of type I
interleukin-1 receptor mRNA and its regulation in cultured astrocytes. Neurosci. Lett. 195: 57–60, 1995.
TORIGUE, K., S. USHIO, T. OKURA, S. S. KOBAYASHI, M. TANIAI,
T. KUNIKATA, T. MURAKAMI, O. SANOU, H. KOJIMA, M. FUJII,
T. OHTA, M. IKEDA, H. IKEGAMI, AND M. KURIMOTO. Purification and characterization of the human interleukin-18 receptor. J.
Biol. Chem. 272: 25737–25742, 1997.
TORRES, G., S. LEE, AND C. RIVIER. Ontogeny of the rat hypothalamic nitric oxide synthase and colocalization with neuropeptides.
Mol. Cell. Neurosci. 4: 155–163, 1993.
TOSATO, G., AND K. D. JONES. Interleukin-1 induces interleukin6 production in peripheral blood monocytes. Blood 75: 1305–1310,
1990.
TRACEY, D. E., AND E. B. DE SOUZA. Identification of interleukin1 receptors in mouse pituitary cell membranes and AtT-20 pituitary tumor cells. Soc. Neurosci. Abstr. 14: 1052, 1988.
TSAGARAKIS, S., G. GILLIES, L. H. REES, M. BESSER, AND A.
GROSSMAN. Interleukin-1 directly stimulates the release of corticotrophin releasing factor from rat hypothalamus. Neuroendocrinology 49: 98–101, 1989.
TSAGARAKIS, S., G. KONTOGEORGOS, P. GIANNOU, N. THALASSINOS, J. WOOLLEY, G. M. BESSER, AND A. GROSSMAN. Interleukin-6, a growth promoting cytokine, is present in human
adenomas: an immunocytochemical study. Clin. Endocrinol. 37:
163–167, 1992.
TSIEN, J. Z., D. F. CHEN, D. GERBER, C. TOM, E. H. MERCER,
D. J. ANDERSON, M. MAYFORD, E. R. KANDEL, AND S. TONEGAWA. Subregion- and cell type-restricted gene knockout in mouse
brain. Cell 87: 1317–1326, 1996.
TSIGOS, C., D. A. PAPANICOLAOU, R. DEFENSOR, C. S. MITSIADIS, I. KYROU, AND G. P. CHROUSOS. Dose effects of recombinant interleukin-6 on pituitary hormone secretion and energy expenditure. Neuroendocrinology 66: 54–62, 1997.
TSUBOKURA, S., Y. WATANABE, H. EHARA, K. IMAMURA, O.
SUGIMOTO, H. KAGAMIYAMA, S. YAMAMOTO, AND O. HAYAISHI. Localization of prostaglandin endoperoxide synthase in
neurons and glia in monkey brain. Brain Res. 543: 15–24, 1991.
TUNKEL, A. R., S. W. ROSSER, E. J. HANSEN, AND W. M.
SCHELD. Blood-brain barrier alterations in bacterial meningitis:
development of an in vitro model and observations on the effects
of lipopolysaccharide. In Vitro Cell. Dev. Biol. 27: 113–120, 1991.
TURNBULL, A. V., R. C. DOW, S. J. HOPKINS, A. WHITE, G. FINK,
AND N. J. ROTHWELL. Mechanism of the activation of the pituitary-adrenal axis by tissue injury in the rat. Psychoneuroendocrinology 19: 165–178, 1994.
TURNBULL, A. V., S. LEE, AND C. RIVIER. Mechanisms of hypothalamic pituitary-adrenal axis stimulation by immune signals in
the adult rat. Ann. NY Acad. Sci. 840: 434–443, 1998.
TURNBULL, A. V., F. J. PITOSSI, J.-J. LEBRUN, S. LEE, J. C.
MELTZER, D. M. NANCE, A. DEL REY, H. O. BESEDOVSKY, AND
C. RIVIER. Inhibition of tumor necrosis factor-a (TNF-a) action
within the central nervous system markedly reduces the plasma
adrenocorticotropin response to peripheral local inflammation in
rats. J. Neurosci. 17: 3262–3273, 1997.
TURNBULL, A. V., AND C. RIVIER. Brain-periphery connections:
do they play a role in mediating the effect of centrally injected
interleukin-1b on gonadal function? Neuroimmunomodulation 2:
224–235, 1995.
pra
APS-Phys Rev
Downloaded from on April 23, 2014
846. TAKAO, T., C. TOJO, T. NISHIOKA, H. KUROKAWA, T. TAKEMURA, K. HASHIMOTO, AND E. B. DE SOUZA. Reciprocal modulation of corticotropin-releasing factor and interleukin-1 receptors
following ether-laparotomy stress in the mouse. Brain Res. 660:
170–174, 1994.
847. TAKAO, T., C. TOJO, T. NISHIOKA, S. MAKINO, K. HASHIMOTO,
AND E. B. DE SOUZA. Stress-induced upregulation of pituitary
interleukin-1 receptors is mediated by corticotropin-releasing factor. Life Sci. 59: 165–168, 1996.
848. TAKAO, T., D. E. TRACEY, W. M. MITCHELL, AND E. B. DE
SOUZA. Interleukin-1 receptors in mouse brain: characterization
and neuronal localization. Endocrinology 127: 3070–3078, 1990.
849. TAKE, S., Y. KANEMITSU, T. YASAKA, T. KATAFUCHI, T. HORI,
AND F. ECKENSTEIN. Immobilization stress modulates the expression of interferon-g mRNA in the mouse (Abstract). Proc.
Annu. Meet. Soc. Neurosci. 26th Washington DC 1996, p. 336.18.
850. TAKEDA, K., H. TSUTSUI, T. YOSHIMOTO, O. ADACHI, N. YOSHIDA, T. KISHIMOTO, H. OKAMURA, K. NAKANISHI, AND S.
AKIRA. Defective NK cell activity and Th1 response in IL-18-deficient mice. Immunity 8: 383–390, 1998.
851. TARTAGLIA, L. A., R. F. WEBER, I. S. FIGARI, C. REYNOLDS,
M. A. PALLADINO, AND D. V. GOEDDEL. The two different receptors for tumor necrosis factor mediate distinct cellular responses.
Proc. Natl. Acad. Sci. USA 88: 9292–9296, 1991.
852. TATAKI, A., Q. H. HUANG, A. SOMOGYVARI-VIGH, AND A. ARIMURA. Immobilization stress may increase plasma interleukin-6
via central and peripheral catecholamines. Neuroimmunomodulation 1: 335–342, 1994.
853. TATSUNO, I., A. SOMOGYVARI-VIGH, K. MIZUNO, P. E.
GOTTSCHALL, H. HIDAKA, AND A. ARIMURA. Neuropeptide regulation of interleukin-6 production from the pituitary: stimulation
by adenylate cyclase activating polypeptide and calcitonin generelated peptide. Endocrinology 129: 1797–1804, 1991.
854. TAYLOR, A. D., H. D. LOXLEY, R. J. FLOWER, AND J. C. BUCKINGHAM. Immunoneutralization of lipocortin 1 reverses the acute
inhibitory effects of dexamethasone on the hypothalamo-pituitaryadrenocortical responses to cytokines in the rat in vitro and in
vivo. Neuroendocrinology 62: 19–31, 1995.
855. TCHELINGERIAN, J.-L., M. MONGUE, F. LE SAUX, B. ZALC, AND
C. JACQUE. Differential oligodendrocyte expression of the tumor
necrosis factor receptors in vivo and in vitro. J. Neurochem. 65:
2377–2380, 1995.
856. TERADA, Y., H. SINOMIYA, AND M. NAKANO. Defect of calmodulin-binding protein expression of interleukin-1 gene by LPS-nonresponder C3H/HeJ mouse macrophages. Biochem. Biophys. Res.
Commun. 158: 723–729, 1989.
857. TERAO, A., H. KITAMURA, A. ASANO, M. KOBAYASHI, AND M.
SAITO. Roles of prostaglandins D2 and E2 in interleukin-1-induced
activation of norepinephrine turnover in the brain and peripheral
organs of rats. J. Neurochem. 65: 2742–2747, 1995.
858. TERAO, A., M. OIKAWA, AND M. SAITO. Cytokine-induced change
in hypothalamic norepinephrine turnover: involvement of corticotropin-releasing hormone and prostaglandins. Brain Res. 622:
257–261, 1993.
859. TERAO, A., M. OIKAWA, AND M. SAITO. Tissue-specific increase
in norepinephrine turnover by central interleukin-1, but not by
interleukin-6, in rats. Am. J. Physiol. 266 (Regulatory Integrative
Comp. Physiol. 35): R400–R404, 1994.
860. TERRAZZINO, S., C. PEREGO, AND M. G. DE SIMONI. Noradrenaline release in hypothalamus and ACTH secretion induced by central interleukin-1b. Neuroreport 6: 2465–2468, 1995.
861. THOMPSON, N. L., K. C. FLANDERS, J. M. SMITH, L. R. ELLINGSWORTH, A. B. ROBERTS, AND M. B. SPORN. Expression of transforming growth factor beta 1 in specific cells and tissues of adult
and neonatal mice. J. Cell Biol. 108: 661–669, 1989.
862. THOMSON, A. The Cytokine Handbook. London: Academic, 1991.
863. TILDERS, F. J. H., R. H. DE RIJK, A.-M. VAN DAM, V. A. VINCENT,
K. SCHOTANUS, AND J. H. A. PERSOONS. Activation of the hypothalamus-pituitary-adrenal axis by bacterial endotoxins: routes
and intermediate signals. Psychoneuroendocrinology 19: 209–232,
1994.
864. TILDERS, F. J. H., E. D. SCHMIDT, AND D. C. E. DE GOEIJ. Pheno-
67
68
ANDREW V. TURNBULL AND CATHERINE L. RIVIER
/ 9j0c$$oc11
P13-8
902.
903.
904.
905.
906.
907.
908.
909.
910.
911.
912.
913.
914.
915.
916.
917.
11-25-98 11:16:36
terleukin-1b in ramified microglia in rat brain: a light and electron
microscopic study. Neuroscience 65: 815–826, 1995.
VAN DAM, A.-M., M. BROUNS, S. LOUISSE, AND F. BERKENBOSCH. Appearance of interleukin-1 in macrophages and in ramified microglia in the brain of endotoxin-treated rats: a pathway
for the induction of non-specific symptoms of sickness? Brain
Res. 588: 291–296, 1992.
VAN DAM, A. M., M. BROUNS, W. MAN-A-HING, AND F. BERKENBOSCH. Immunocytochemical detection of prostaglandin E2 in
microvasculature and in neurons of rat brain after administration
of bacterial endotoxin. Brain Res. 613: 331–336, 1993.
VAN DAM, A.-M., H. E. DE VRIES, J. KUIPER, F. J. ZIJLSTRA,
E. G. DE BOER, F. J. TILDERS, AND F. BERKENBOSCH. Interleukin-1 receptors on rat brain endothelial cells: a role in neuroimmune interaction? FASEB J. 10: 351–356, 1996.
VAN DAM, M., J. MULLBERG, H. SCHOOLTINK, T. STOYAN,
J. P. J. BRAKENHOFF, L. GRAEVE, P. C. HEINRICH, AND S.
ROSE-JOHN. Structure-function analysis of interleukin-6 utilizing
human/murine chimeric molecules. J. Biol. Chem. 268: 15285–
15290, 1993.
VAN DER MEER, M. J. M., A. R. M. N. HERMUS, G. J. PESMAN,
AND C. G. J. SWEEP. Effects of cytokines on pituitary b-endorphin
and adrenal corticosterone release in vitro. Cytokine 8: 238–247,
1996.
VAN DER MEER, M. J. M., C. G. J. F. SWEEP, G. J. PESMAN, G. F.
BORM, AND A. R. M. N. HERMUS. Synergism between IL-1b and
TNF-a on the activity of the pituitary-adrenal axis and on food
intake of rats. Am. J. Physiol. 268 (Endocrinol. Metab. 31): E551–
E557, 1995.
VAN DER MEER, M. J. M., C. G. J. F. SWEEP, G. J. PESMAN,
F. J. H. TILDERS, AND A. R. M. M. HERMUS. Chronic stimulation
of the hypothalamus-pituitary-adrenal axis in rats by interleukin
1b: central and peripheral mechanisms. Cytokine 8: 910–919,
1996.
VAN DER MEER, M. J. M., C. G. J. SWEEP, C. E. M. RIJNKELS,
G. J. PESMAN, F. J. H. TILDERS, P. W. C. KLOPPENBORG, AND
A. R. M. M. HERMUS. Acute stimulation of the hypothalamic-pituitary-adrenal axis by IL-1b, TNFa and IL-6: a dose response study.
J. Endocrinol. Invest. 19: 175–182, 1996.
VAN GOOL, J., H. VAN VUGT, M. HELLE, AND L. A. AARDEN. The
relation among stress, adrenalin, interleukin-6 and acute phase
proteins in the rat. Clin. Immunol. Immunopathol. 57: 200–210,
1990.
VANKELOM, H., P. CARMELIAT, H. HEREMANS, J. VAN DAMME,
R. DIJKMANS, A. BILLIAU, AND C. DENEF. Interferon gamma
inhibits stimulated adrenocorticotropin, prolactin, and growth
hormone secretion in normal rat anterior pituitary cell cultures.
Endocrinology 126: 2919–2926, 1990.
VANKELECOM, H., P. CARMELIET, J. VAN DAMME, A. BILLIAU,
AND C. DENEF. Production of interleukin-6 by folliculo-stellate
cells of the anterior pituitary gland in a histiotypic cell aggregate
culture system. Neuroendocrinology 49: 102–106, 1989.
VANKELECOM, H., P. MATTHYS, J. VAN DAMME, H. HEREMANS, A. BILLIAU, AND C. DENEF. Immunocytochemical evidence that S-100-positive cells of the mouse anterior pituitary
contain interleukin-6 immunoreactivity. J. Histochem. Cytochem.
41: 151–156, 1993.
VEENING, J. G., M. J. M. VAN DER MEER, H. JOOSTEN,
A. R. M. N. HERMUS, C. E. M. RIJNNKELS, L. M. GEERAEDTS,
AND C. G. J. SWEEP. Intravenous administration of interleukin-1
beta induces Fos-like immunoreactivity in corticotropin-releasing
hormone neurones in the paraventricular hypothalamic nucleus
of the rat. J. Chem. Neuroanat. 6: 391–397, 1993.
VELKENIERS, B., P. VERGANI, J. TROUILLAS, J. D’HAENS, R. J.
HOOGHE, AND E. L. HOOGHE-PETERS. Expression of IL-6 mRNA
in normal rat and human pituitaries and in human pituitary adenomas. J. Histochem. Cytochem. 42: 67–76, 1994.
VELLUCCI, S. V., R. F. PARROT, R. F. DA COSTA, S. OHKURA,
AND K. M. KENDRICK. Increased body temperature, cortisol secretion, and hypothalamic expression of c-fos, corticotropin releasing
hormone and interleukin-1b, following central administration of
interleukin-1b in the sheep. Brain Res. 29: 64–70, 1995.
VILLAR, M. J., S. CECCATELLI, K. BEDECS, T. BARTFAI, D.
pra
APS-Phys Rev
Downloaded from on April 23, 2014
883. TURNBULL, A. V., AND C. RIVIER. Regulation of the HPA axis by
cytokines. Brain Behav. Immun. 9: 253–275, 1995.
884. TURNBULL, A. V., AND C. RIVIER. Corticotropin-releasing factor,
vasopressin and prostaglandins mediate, and nitric oxide restrains, the HPA axis response to acute local inflammation in the
rat. Endocrinology 137: 455–463, 1996.
885. TURNBULL, A. V., AND C. RIVIER. Cytokine effects on neuroendocrine axes: influence of nitric oxide and carbon monoxide. In:
Cytokines in the Nervous System (Neuroscience Intelligence
Unit), edited by N. J. Rothwell. Heidelberg, Germany: SpringerVerlag, 1996, p. 93–116.
886. TURNBULL, A. V., AND C. RIVIER. Cytokines within the neuroendocrine system. Curr. Opin. Endocrinol. Diabetes 3: 149–156,
1996.
887. TURNBULL, A. V., AND C. RIVIER. Selective inhibitors of nitric
oxide synthase (NOS) implicate a constitutive isoform of NOS in
the regulation of interleukin-1-induced ACTH secretion in rats.
Endocrine 5: 135–145, 1996.
888. TURNBULL, A. V., AND C. RIVIER. Corticotropin-releasing factor
(CRF) and endocrine responses to stress: CRF receptors, binding
protein and related peptides. Proc. Soc. Exp. Biol. Med. 215: 1–
10, 1997.
889. TURNBULL, A. V., AND C. RIVIER. Intracereboventricular (icv)
passive immunization I: the effect of icv administration of an antisera to tumor necrosis factor-a on the plasma ACTH response to
lipopolysaccharide in rats. Endocrinology 139: 119–127, 1998.
890. TURNBULL, A. V., AND C. RIVIER. Intracereboventricular passive
immunization. II. Intracerebroventricular infusion of neuropeptide
antisera can inhibit neuropeptide signaling in peripheral tissues.
Endocrinology 139: 128–136, 1998.
891. TURNBULL, A. V., W. VALE, AND C. RIVIER. Urocortin, a corticotropin-releasing factor-related mammalian peptide, inhibits edema
due to thermal injury in rats. Eur. J. Pharmacol. 303: 213–216,
1996.
892. TURNBULL, A. V., J. VAUGHAN, J. RIVIER, W. VALE, AND C.
RIVIER. Regulation of adrenocorticotropin secretion in the rat by
the novel mammalian neuropeptide, urocortin. Proc. Int. Congr.
Endocrinol. 10th San Francisco CA 1996, P. 3–534.
893. UEHARA, A., S. GILLIS, AND A. ARIMURA. Effects of interleukin1 on hormone release from rat pituitary cells in primary culture.
Neuroendocrinology 45: 343–347, 1987.
894. UEHARA, A., P. E. GOTTSCHALL, R. R. DAHL, AND A. ARIMURA.
Interleukin-1 stimulates ACTH release by an indirect action which
requires endogenous corticotropin-releasing factor. Endocrinology 121: 1580–1582, 1987.
895. UETA, Y., A. LEVY, H. S. CHOWDREY, AND S. L. LIGHTMAN. S100 antigen positive folliculostellate cells are not the major source
of IL-6 gene expression in human pituitary adenomas. J. Neuroendocrinol. 7: 467–474, 1995.
896. UR, E., P. D. WHITE, AND A. GROSSMAN. Hypothesis: cytokines
may be activated to cause depressive illness and chronic fatigue
syndrome. Eur. Arch. Psychiatry Clin. Neurosci. 241: 317–322,
1992.
897. VALE, W., J. RIVIER, J. VAUGHAN, R. MCCLINTOCK, A. CORRIGAN, W. WOO, D. KARR, AND J. SPIESS. Purification and characterization of an FSH releasing protein from porcine ovarian follicular fluid. Nature 321: 776–779, 1986.
898. VALE, W., J. SPEISS, C. RIVIER, AND J. RIVIER. Characterization
of a 41-amino acid residue ovine hypothalamic peptide that stimulates the secretion of corticotropin and b-endorphin. Science 213:
1394–1397, 1981.
899. VALLIERES, L., S. LACROIX, AND S. RIVEST. Influence of interleukin-6 on neural activity and transcription of the gene encoding
corticotropin-releasing factor in the rat brain: an effect depending
upon the route of administration. Eur. J. Neurosci. 9: 1461–1472,
1997.
900. VALLIERES, L., AND L. RIVEST. Regulation of the genes encoding
interleukin-6, its receptor, and gp130 in the rat brain in response to
the immune activator lipopolysaccharide and the proinflammatory
cytokine interleukin-1b. J. Neurochem. 69: 1668–1683, 1997.
901. VAN DAM, A.-M., J. BAUER, F. J. H. TILDERS, AND F. BERKENBOSCH. Endotoxin-induced appearance of immunoreactive in-
Volume 79
January 1999
918.
919.
920.
921.
922.
923.
924.
926.
927.
928.
929.
930.
931.
932.
933.
934.
935.
BREDT, S. H. SNYDER, AND T. HOKFELT. Upregulation of nitric
oxide synthase and galanin message-associated peptide in hypothalamic magnocellular neurons after hypophysectomy. Immunohistochemical and in situ hybridization studies. Brain Res. 650:
219–228, 1994.
VILLAR, M. J., S. CECCATELLI, M. RONNQVIST, AND T. HOKFELT. Nitric oxide synthase increases in hypothalamic magnocellular neurones after salt loading in the rat. An immunohistochemical and in situ hybridization study. Brain Res. 644: 273–281, 1994.
VINCENT, S. R., AND H. KIMURA. Histochemical mapping of the
nitric oxide synthase in the rat brain. Neuroscience 46: 755–784,
1992.
VITI, A., M. MUSCETLOLA, L. PAULESU, V. BOCCI, AND A. ALMI.
Effect of exercise on plasma interferon levels. J. Appl. Physiol.
59: 426–428, 1985.
VLASKOVSKA, M., G. HERRTING, AND W. KNEPEL. Adrenocorticotropin and b-endorphin release from rat adenohypophysis in
vitro: inhibition by prostaglandin E2 formed locally in response to
vasopressin and corticotropin releasing factor. Endocrinology
115: 895–903, 1984.
WADA, Y., M. SATO, M. NIIMI, M. TAMAKI, T. ISHIDA, AND J.
TAKAHARA. Inhibitory effect of interleukin-1 on growth hormone
secretion in conscious male rats. Endocrinology 136: 3936–3941,
1995.
WAGUESPACK, P. J., W. A. BANKS, AND A. J. KASTIN. Interleukin-2 does not cross the blood-brain barrier by a saturable transport system. Brain Res. Bull. 34: 103–109, 1994.
WAN, W., L. JANZ, C. Y. VRIEND, C. M. SORENSON, A. H.
GREENBERG, AND D. M. NANCE. Differential induction of c-fos
immunoreactivity in hypothalamus and brain stem nuclei following central and peripheral administration of endotoxin. Brain Res.
Bull. 32: 581–587, 1993.
WAN, W., L. WETMORE, C. M. SORENSON, A. H. GREENBERG,
AND D. M. NANCE. Neural and biochemical mediators of endotoxin and stress-induced c-fos expression in the rat brain. Brain
Res. Bull. 34: 7–14, 1994.
WANG, Z., S. G. REN, AND S. MELMED. Hypothalamic and pituitary leukemia inhibitory factor gene expression in vivo: a novel
endotoxin-inducible neuro-endocrine interface. Endocrinology
137: 2947–2953, 1996.
WATANABE, D., R. YOSHIMURA, M. KHALIL, K. YOSHIDA, T.
KISHIMOTO, T. TAGA, AND H. KIYAMA. Characteristic localization of gp130 (the signal transducing receptor component used
in common for IL-6/IL-11/CNTF/OSM) in the rat brain. Eur. J.
Neurosci. 8: 1630–1640, 1996.
WATANABE, T., A. MORIMOTO, K. MORIMOTO, T. NAKAMORI,
AND N. MURAKAMI. ACTH release induced in rats by noradrenaline is mediated by prostaglandin E2. J. Physiol. (Lond.) 443: 431–
439, 1991.
WATANABE, T., A. MORIMOTO, AND N. MURAKAMI. Threshold
dose of interleukin-1b for induction of an ACTH response is higher
than a febrile response. Pflu¨gers Arch. 419: 629–631, 1990.
WATANABE, T., A. MORIMOTO, AND N. MURAKAMI. ACTH response in rats during biphasic fever induced by interleukin-1. Am.
J. Physiol. 261 (Regulatory Integrative Comp. Physiol. 30):
R1104–R1108, 1991.
WATANABE, T., A. MORIMOTO, Y. SAKATA, AND N. MURAKAMI.
ACTH response induced by interleukin-1 is mediated by CRF secretion stimulated by hypothalamic PGE. Experientia 46: 481–
484, 1990.
WATANABE, T., A. MORIMOTO, N. TAN, T. MAKISUMI, S. SHIMADA, T. NAKAMORI, AND N. MURAKMI. ACTH response induced in capsaicin-desensitized rats by intravenous injection of
interleukin-1 or prostaglandin E. J. Physiol. (Lond.) 475: 139–
145, 1994.
WATANABE, Y., Y. WATANABE, K. HAMADA, M.-C. BOMMELARBAYT, F. DRAY, T. KANEKO, N. YUMOTO, AND O. HAYAISHI.
Distinct localization of prostaglandin D2, E2 and F2a binding sites
in the monkey brain. Brain Res. 478: 143–148, 1989.
WATANABE, Y., Y. WATANABE, AND O. HAYAISHI. Quantitative
autoradiographic localization of prostaglandin E2 binding sites in
monkey diencephalon. J. Neurosci. 8: 2003–2010, 1988.
WATANOBE, H., R. NASUSHITA, AND K. TAKEBE. A study of the
/ 9j0c$$oc11
P13-8
936.
937.
938.
939.
940.
941.
942.
943.
944.
945.
946.
947.
948.
949.
950.
951.
952.
11-25-98 11:16:36
69
role of circulating prostaglandin E2 in the adrenocorticotropin
response to intravenous administration of interleukin-1b in the
rat. Neuroendocrinology 62: 596–600, 1995.
WATANOBE, H., S. SASAKI, AND K. TAKEBE. Evidence that intravenous administration of interleukin-1 stimulates corticotropin releasing hormone secretion in the median eminence of freely moving rats: estimation by push pull perfusion. Neurosci. Lett. 133:
7–10, 1991.
WATANOBE, H., S. SASAKI, AND K. TAKEBE. Role of prostaglandins E1, E2 and F2 alpha in the brain in interleukin-1 beta-induced
adrenocorticotropin secretion in the rat. Cytokine 7: 710–712,
1995.
WATANOBE, H., AND K. TAKEBE. Intravenous administration of
tumor necrosis factor-a stimulates corticotropin-releasing hormone secretion in the push-pull cannulated median eminence of
freely moving rats. Neuropeptides 22: 81–84, 1992.
WATANOBE, H., AND K. TAKEBE. Intrahypothalamic perfusion
with interleukin-1-beta stimulates the local release of corticotropin-releasing hormone and arginine vasopressin and plasma adrenocorticotropin in freely moving rats: a comparative perfusion of
the paraventricular nucleus and the median eminence. Neuroendocrinology 57: 593–599, 1993.
WATANOBE, H., AND K. TAKEBE. Effects of intravenous administration of interleukin-1 beta on the release of prostaglandin E2,
corticotropin-releasing factor, and arginine vasopressin in several
hypothalamic areas of freely moving rats: estimation by push-pull
perfusion. Neuroendocrinology 60: 8–15, 1994.
WATKINS, L. R., L. E. GOEHLER, J. RELTON, M. T. BREWER,
AND S. F. MAIER. Mechanisms of tumor necrosis factor-a (TNFa) hyperalgesia. Brain Res. 692: 244–250, 1995.
WATKINS, L. R., L. E. GOEHLER, J. K. RELTON, N. TARTAGLIA,
L. SILBERT, D. MARTIN, AND S. F. MAIER. Blockade of interleukin-1 induced hyperthermia by subdiaphragmatic vagotomy: evidence for vagal mediation of immune brain communication. Neurosci. Lett. 183: 27–31, 1995.
WATKINS, L. R., S. F. MAIER, AND L. E. GOEHLER. Cytokine-tobrain communication: a review and analysis of alternative mechanisms. Life Sci. 57: 1011–1026, 1995.
WATKINS, L. R., E. P. WIERTELAK, L. E. GOEHELER, K. MOONEY-HEIBERGER, J. MARTINEZ, L. FURNESS, K. P. SMITH, AND
S. F. MAIER. Neurocircuitry of illness-induced hyperalgesia.
Brain Res. 639: 283–299, 1994.
WATKINS, L. R., E. P. WIERTELAK, L. E. GOEHLER, K. P. SMITH,
D. MARTIN, AND S. F. MAIER. Characterization of cytokine-induced hyperalgesia. Brain Res. 654: 15–26, 1994.
WEBER, M. M., P. MICHL, C. J. AUERNHAMMER, AND D. ENGELHARDT. Interleukin-3 and interleukin-6 stimulate cortisol secretion from adult human adrenocortical cells. Endocrinology
138: 2207–2210, 1997.
WEBSTER, E. L., D. E. TRACEY, AND E. B. DE SOUZA. Upregulation of interleukin-1 receptors in mouse AtT-20 pituitary cells following treatment with corticotropin-releasing factor. Endocrinology 129: 2796–2798, 1991.
WEI, X.-Q., I. G. CHARLES, A. SMITH, J. URE, G. J. FENG, F. P.
HUANG, D. XU, W. MULLER, S. MONCADA, AND F. Y. LIEW. Altered immune responses in mice lacking inducible nitric oxide
synthase. Nature 375: 408–411, 1995.
WEIDENFELD, J., O. ABRAMSKY, AND H. OVADIA. Evidence for
the involvement of the central nervous adrenergic system in interleukin 1-induced adrenocortical response. Neuropharmacology
28: 1411–1414, 1989.
WEIDENFELD, J., M. CRUMEYROLLE-ARIAS, AND F. HAOUR.
Effect of bacterial endotoxin and interleukin-1 on prostaglandin
biosynthesis by the hippocampus of mouse brain: role of interleukin-1 receptors and glucocorticoids. Neuroendocrinology 62: 39–
46, 1995.
WEIGENT, D. A., J. B. BAXTER, W. E. WEAR, L. R. SMITH, K. L.
BOST, AND J. E. BLALOCK. Production of immunoreactive growth
hormone by mononuclear leukocytes. FASEB J. 2: 2812–2818,
1988.
WEIGENT, D. A., AND J. E. BLALOCK. Interaction beteen neuroendocrine and immune systems: common hormones and receptors.
Immunol. Rev. 100: 79–108, 1987.
pra
APS-Phys Rev
Downloaded from on April 23, 2014
925.
REGULATION OF HPA AXIS BY CYTOKINES
70
ANDREW V. TURNBULL AND CATHERINE L. RIVIER
/ 9j0c$$oc11
P13-8
973.
974.
975.
976.
977.
978.
979.
980.
981.
982.
983.
984.
985.
986.
987.
988.
989.
990.
991.
992.
11-25-98 11:16:36
I receptor mRNA in rat brain. Neuroimmunomodulation 1: 110–
115, 1994.
WONG, M. L., AND J. LICINO. Localization of stem cell factor
mRNA in adult rat hippocampus. Neuroimmunomodulation 1:
181–187, 1994.
WONG, M. L., V. RETTORI, A. AL-SHEKHLEE, P. B. BONGIORNO,
G. CANTEROS, S. M. MCCANN, P. W. GOLD, AND J. LICINIO. Inducible nitric oxide synthase gene expression in the brain during
systemic inflammation. Nature Med. 2: 581–584, 1996.
WOOD, C. E., J. SHINSAKO, AND M. F. DALLMAN. Comparison of
canine corticosteroid responses to mean and phasic increases in
ACTH. Am. J. Physiol. 242 (Endocrinol. Metab. 5): E102–E108,
1982.
WOOLF, P. D., C. COX, M. KELLY, D. NICHOLS, J. V. MCDONALD,
AND R. W. HAMILL. The adrenocortical response to brain injury:
correlation with the severity of neurological dysfunction, effects
of intoxication, and patient outcome. Alcohol. Clin. Exp. Res. 14:
917–921, 1990.
WUCHERPFENNIG, K. W., J. NEWCOMBE, H. LI, C. KEDDY, M. L.
CUZNER, AND D. A. HAFLER. T cell receptor Valpha-Vbeta repertoire and cytokine gene expression in active multiple sclerosis
lesions. J. Exp. Med. 175: 993–1002, 1992.
WUSTEMAN, M., D. G. D. WIGHT, AND M. ELIA. Protein metabolism after injury with turpentine: a rat model for clinical trauma.
Am. J. Physiol. 259 (Endocrinol. Metab. 22): E763–E769, 1990.
XIA, M., S. QIN, M. MCNAMARA, C. MACKAY, AND B. T. HYMAN.
Interleukin-8 receptor B immunoreactivity in brain and neuritic
placques of Alzheimer’s disease. Am. J. Pathol. 150: 1267–1274,
1997.
XIE, W., J. G. CHIPMAN, D. L. ROBERTSON, R. L. ERIKSON, AND
D. L. SIMMONS. Expression of a mitogen-responsive gene encoding prostaglandin synthase is regulated by mRNA splicing. Proc.
Natl. Acad. Sci. USA 88: 2692–2696, 1991.
YABUUCHI, K., E. MARUTA, M. MINAMI, AND M. SATOH. Induction of interleukin-1b mRNA in the hypothalamus following subcutaneous injections of Formalin into the rat hind paws. Neurosci.
Lett. 207: 109–112, 1996.
YABUUCHI, K., M. MINAMI, S. KATSUMATA, AND M. SATOH.
Localization of type I interleukin-1 receptor mRNA in the rat brain.
Brain Res. 27: 27–36, 1994.
YAMADA, T., M. A. HORISBERGER, N. KAWAGUCHI, I. MOROO,
AND T. TOYODA. Immunohistochemistry using antibodies to
alpha-interferon and its induced protein, MxA, in Alzheimer’s and
Parkinson’s disease brain tissue. Neurosci. Lett. 181: 61–64, 1994.
YAMADA, T., AND I. YAMANAKA. Microglial localization of alphainterferon receptor in human brain tissues. Neurosci. Lett. 189:
73–76, 1995.
YAMAGATA, K., K. I. ANDREASSON, W. E. KAUFMANN, C. A.
BARNES, AND P. F. WORLEY. Expression of a mitogen-inducible
cyclooxygenase in brain neurons: regulation by synaptic activity
and glucocorticoids. Neuron 11: 371–386, 1993.
YAMAGUCHI, M., K. KOIKE, N. MATSUZAKI, Y. YOSHIMOTO, T.
TAMIGUSHI, A. MIYAKE, AND O. TANIZAWA. The interferon family stimulates the secretion of prolactin and interleukin-6 by the
pituitary gland in vitro. J. Endocrinol. Invest. 14: 457–461, 1991.
YAMAMORI, T. Localization of cholinergic differentiation factor/
leukemia inhibitory factor mRNA in the rat brain and peripheral
tissues. Proc. Natl. Acad. Sci. USA 88: 7298–7302, 1991.
YAN, H. Q., M. A. BANOS, P. HERREGODTS, R. HOOGHE, AND
E. L. HOOGHE-PETERS. Expression of interleukin (IL)-1b, IL-6
and their respective receptors in the normal rat brain and after
injury. Eur. J. Immunol. 22: 2963–2971, 1992.
YANAGISAWA, K., T. TAKAGI, T. TSUKAMOTO, T. TETSUKA,
AND S. TOMINAGA. Presence of a novel primary response gene
ST2L, encoding a product highly similar to the interleukin-1 receptor type 1. FEBS Lett. 318: 83–87, 1993.
YAO, J., AND R. W. JOHNSON. Induction of interleukin-1 beta converting enzyme (ICE) in murine microglia by lipopolysacchairde.
Brain Res. 51: 170–178, 1997.
YASIN, S. A., A. COSTA, M. L. FORSLING, AND A. GROSSMAN.
Interleukin-1b and interleukin-6 stimulate neurohypophysial hormone release in vitro. J. Neuroendocrinol. 6: 179–184, 1994.
YASIN, S. A., A. COSTA, D. HUCKS, M. L. FORSLING, AND A.
pra
APS-Phys Rev
Downloaded from on April 23, 2014
953. WEIGENT, D. A., AND J. E. BLALOCK. Production of peptide hormones and neurotransmitters by the immune system. Chem. Immunol. 69: 1–30, 1997.
954. WEISS, J. M., AND S. K. SUNDAR. Interleukin-1: effects in the central nervous sytem and relevance to AIDS. Clin. Neuropharmacol.
15, Suppl. 1: 661–662, 1992.
955. WESTERMANN, R., M. JOHANNSEN, K. UNSICKER, AND C.
GROTHE. Basic fibroblast growth factor (bFGF) immunoreactivity is present in chromaffin cells. J. Neurochem. 55: 285–292, 1990.
956. WETHER, G. A., M. ABATE, A. HOGG, H. CHEESMAN, B. OLDFIELD, D. HARDS, P. HUDSON, B. POWER, K. FREED, AND A. C.
HERINGTON. Localization of insulin-like growth factor-I mRNA
in rat brain by in situ hybridization: relationship to IGF-I receptors.
Mol. Endocrinol. 4: 773–778, 1990.
957. WHITCOMB, R. W., W. M. LINEHAM, L. M. WAHL, AND R. A. KNAZEK. Monocytes stimulate cortisol production by cultured human
adrenocortical cells. J. Clin. Endocrinol. Metab. 66: 33–38, 1988.
958. WHITNALL, M. H., R. S. PERLSTEIN, E. H. MOUGEY, AND R.
NETA. Effects of interleukin-1 on the stress-responsive and -nonresponsive subtypes of corticotropin-releasing hormone neurosecretory axons. Endocrinology 131: 37–44, 1992.
959. WICK, G. W., Y. HU, S. SCHWARZ, AND G. KROEMER. Immunoendocrine communication via the hypothalamo-pituitary-adrenal
axis in autoimmune diseases. Endocr. Rev. 14: 539–563, 1993.
960. WILCOX, J. N., AND R. DERYNCK. Localization of cells synthesizing transforming growth factor-alpha mRNA in the mouse brain.
J. Neurosci. 8: 1901–1904, 1988.
961. WILLIAMS, C. A., AND B. L. ALLEN-HOFFMAN. Transforming
growth factor-beta 1 stimulates fibronectin production in bovine
adrenocortical cells in culture. J. Biol. Chem. 265: 6467–6472,
1990.
962. WILLIAMS, L. M., P. E. BALLMER, L. T. HANNAH, I. GRANT, AND
P. J. GARLICK. Changes in regional protein synthesis in rat brain
and pituitary after systemic interleukin-1b administration. Am. J.
Physiol. 267 (Endocrinol. Metab. 30): E915–E920, 1994.
963. WILT, S., E. MILWARD, J. ZHOU, K. NAGASATO, H. PATTON, R.
RUSTEN, D. GRIFFIN, M. O’CONNOR, AND M. DUBOIS-DALCQ.
In vitro evidence for a dual role of tumor necrosis factor-a in
human immunodeficiency virus type I encephalopathy. Ann. Neurol. 37: 382–394, 1995.
964. WINTER, J. S. D., K. W. GOW, Y. S. PERRY, AND A. H.
GREENBERG. A stimulatory effect of interleukin-1 on adrenocortical cortisol secretion mediated by prostaglandins. Endocrinology 127: 1904–1990, 1990.
965. WOLOSKI, B. M. R. N. J., AND J. C. JAMIESON. Rat corticotropin,
insulin and thyroid hormone levels during the acute phase response to inflammation. Comp. Biochem. Physiol. 86: 15–19, 1987.
966. WOLOSKI, B. M. R. N. J., E. M. SMITH, W. J. MEYER III, G. M.
FULLER, AND J. E. BLALOCK. Corticotropin-releasing activity of
monokines. Science 230: 1035–1037, 1985.
967. WOLVERS, D. A., C. MARQUETTE, F. BERKENBOSCH, AND F.
HAOUR. Tumor necrosis factor-alpha: specific binding sites in
rodent brain and pituitary gland. Eur. Cytokine Netw. 4: 377–381,
1993.
968. WONG, G. H. W., L. A. TARTAGLIA, M. S. LEE, AND D. V. GOEDDEL. Antiviral activity of tumor necrosis is signalled through the
55-kDa type 1 receptor. J. Immunol. 149: 3550–3553, 1992.
969. WONG, M. L., P. B. BONGIORNO, A. AL-SHEKHLEE, A. ESPOSITO, P. KHATRI, AND J. LICINIO. IL-1 beta, IL-1 receptor type I
receptor and iNOS gene expression in rat brain vasculature and
perivascular areas. Neuroreport 7: 2445–2448, 1996.
970. WONG, M.-L., P. B. BONGIORNO, P. W. GOLD, AND J. LICINIO.
Localization of interleukin-1b converting enzyme mRNA in rat
brain vasculature: evidence that the genes encoding the interleukin-1 system are constitutively expressed in brain blood vessels.
Neuroimmunomodulation 2: 141–148, 1995.
971. WONG, M.-L., P. B. BONGIORNO, V. RETTORI, S. M. MCCANN,
AND J. LICINIO. Interleukin- (IL) 1b, IL-1 receptor antagonist, IL10, and IL-13 gene expression in the central nervous system and
anterior pituitary during systemic inflammation: pathophysiological implications. Proc. Natl. Acad. Sci. USA 94: 227–232, 1997.
972. WONG, M.-L., AND J. LICINIO. Localization of interleukin 1 type
Volume 79
January 1999
993.
994.
995.
996.
997.
998.
999.
REGULATION OF HPA AXIS BY CYTOKINES
GROSSMAN. Interleukin-induced vasopressin release is inhibited
by L-arginine. Ann. NY Acad. Sci. 689: 693–695, 1993.
YASIN, S., A. COSTA, P. TRAINER, R. WINDLE, M. L. FORSLING,
AND A. GROSSMAN. Nitric oxide modulates the release of vasopressin from rat hypothalamic explants. Endocrinology 133: 1466–
1469, 1993.
YOON, D. Y., AND C. A. DINARELLO. Antibodies to domains II and
III of the IL-1 receptor accessory protein inhibit IL-1 beta activity
but not binding: regulation of IL-1 responses is via type I receptor,
not the accessory protein. J. Immunol. 160: 3170–3179, 1998.
YOUNG, W. S., III, E. MEZEY, AND R. E. SIEGEL. Quantitative
in situ hybridization histochemistry reveals increased levels of
corticotropin-releasing factor mRNA after adrenalectomy in rats.
Neurosci. Lett. 70: 198–203, 1986.
ZALCMAN, S., J. M. GREEN-JOHNSON, L. MURRAY, D. M.
NANCE, D. DYCK, H. ANISMAN, AND A. H. GREENBURG. Cytokine-specific central monoamine alterations induced by interleukin-1, -2 and -6. Brain Res. 643: 40–49, 1994.
ZALCMAN, S., N. SHANKS, AND H. ANISMAN. Time-dependent
variations of central norepinephrine and dopamine following antigen administration. Brain Res. 557: 69–76, 1991.
ZANETTI, G., D. HEUMANN, J. GERAIN, J. KOHLER, P. ABBET,
C. BARRAS, R. LUCAS, M.-P. GLAUSER, AND J.-D. BAUMGARTNER. Cytokine production after intravenous or peritoneal Gramnegative bacterial challenge in mice. J. Immunol. 148: 1890–1897,
1992.
ZELAZOWSKI, P., V. K. PATCHEV, E. B. ZELAZOWSKA, G. P.
1000.
1001.
1002.
1003.
1004.
71
CHROUSOS, P. W. GOLD, AND E. M. STERNBERG. Release of hypothalamic corticotropin-releasing hormone and arginine vasopressin by interleukin-1 beta and alpha MSH: studies in rats with
different susceptibility to inflammatory disease. Brain Res. 631:
22–26, 1993.
ZHANG, S. C., AND S. FEDEROFF. Cellular localization of stem
cell factor and c-kit receptor in the mouse central nervous system.
J. Neurosci. Res. 47: 1–15, 1997.
ZHENG, H., D. FLETCHER, W. KOZAK, M. JIANG, K. J. HOFMAN,
C. A. CONN, D. SOSZYNSKI, C. GRABIEC, M. E. TRUMBAUER,
A. SHAW, M. J. KOSTURA, K. STEVENS, H. ROSEN, R. J. NORTH,
H. Y. CHEN, M. J. TOCCI, M. J. KLUGER, AND L. H. T. VAN DER
PLOEG. Resistance to fever induction and impaired acute-phase
response in interleukin-1b-deficient mice. Immunity 3: 9–19,
1995.
ZHOU, D., A. W. KUSNECOV, M. R. SHURIN, M. DEPAOLI, AND B.
RABIN. Exposure to physical and psychological stressors elevates
plasma interleukin-6: relationship to the activation of the hypothalamo-pituitary-adrenal axis. Endocrinology 133: 2523–2530, 1993.
ZHOU, D., N. SHANKS, S. E. RIECHMAN, R. LIANG, A. W. KUSNECOV, AND B. S. RABIN. Interleukin-6 modulates interleukin-1and stress-induced activation of the hypothalamic-pituitary-adrenal axis in male rats. Neuroendocrinology 63: 227–236, 1996.
ZIELENIEWSKI, W., J. ZIELENIEWSKI, AND H. STEPIEN. Effect
of interleukin-1a, IL-1b and IL-1 receptor antibody on the proliferation and steroidogenesis of regenerating rat adrenal cortex. Exp.
Clin. Endocrinol. Diabetes 103: 373–377, 1995.
Downloaded from on April 23, 2014
/ 9j0c$$oc11
P13-8
11-25-98 11:16:36
pra
APS-Phys Rev
Regulation of the Hypothalamic-Pituitary-Adrenal Axis
by Cytokines: Actions and Mechanisms of Action
ANDREW V. TURNBULL and CATHERINE L. RIVIER
Physiol Rev 79:1-71, 1999.
You might find this additional info useful...
This article has been cited by 96 other HighWire hosted articles, the first 5 are:
Intestinal microbiota and immune function in the pathogenesis of irritable bowel syndrome
Yehuda Ringel and Nitsan Maharshak
Am J Physiol Gastrointest Liver Physiol, October 15, 2013; 305 (8): G529-G541.
[Abstract] [Full Text] [PDF]
The Stress Response to Surgery and Postoperative Delirium: Evidence of Hypothalamic−
Pituitary−Adrenal Axis Hyperresponsiveness and Decreased Suppression of the GH/IGF-1
Axis
Joaquim Cerejeira, Pedro Batista, Vasco Nogueira, Adriano Vaz-Serra and Elizabeta B.
Mukaetova-Ladinska
J Geriatr Psychiatry Neurol, September , 2013; 26 (3): 185-194.
[Abstract] [Full Text] [PDF]
Duress without stress: Cryptobia infection results in HPI axis dysfunction in rainbow trout
Barry N Madison, Patrick T K Woo and Nicholas J Bernier
J Endocrinol, September , 2013; 218 (3): 287-297.
[Abstract] [Full Text] [PDF]
The Stress Response to Surgery and Postoperative Delirium: Evidence of Hypothalamic−
Pituitary−Adrenal Axis Hyperresponsiveness and Decreased Suppression of the GH/IGF-1
Axis
Joaquim Cerejeira, Pedro Batista, Vasco Nogueira, Adriano Vaz-Serra and Elizabeta B.
Mukaetova-Ladinska
J Geriatr Psychiatry Neurol, July 17, 2013; .
[Abstract] [Full Text] [PDF]
Updated information and services including high resolution figures, can be found at:
/content/79/1/1.full.html
Additional material and information about Physiological Reviews can be found at:
http://www.the-aps.org/publications/prv
This information is current as of April 23, 2014.
Physiological Reviews provides state of the art coverage of timely issues in the physiological and biomedical sciences. It is published
quarterly in January, April, July, and October by the American Physiological Society, 9650 Rockville Pike, Bethesda MD
20814-3991. Copyright © 1999 by the American Physiological Society. ISSN: 0031-9333, ESSN: 1522-1210. Visit our website at
http://www.the-aps.org/.
Downloaded from on April 23, 2014
The interface between neurology and psychiatry: the case of multiple sclerosis
Eugene K. O. Wong, Rajeev Krishnadas and Jonathan Cavanagh
APT, September , 2013; 19 (5): 370-377.
[Abstract] [Full Text] [PDF]