A novel regulator of lung inflammation and immunity: pulmonary

Q J Med 2014; 107:789–792
doi:10.1093/qjmed/hcu005 Advance Access Publication 18 January 2014
Review
A novel regulator of lung inflammation and immunity:
pulmonary parasympathetic inflammatory reflex
X. YANG, C. ZHAO, Z. GAO and X. SU
From the Unit of Respiratory Infection and Immunity, Institut Pasteur of Shanghai, Chinese Academy
of Sciences, Shanghai 200031, China
Address correspondence to X. Su, MD, PhD, Unit of Respiratory Infection and Immunity, Institut Pasteur of
Shanghai, Chinese Academy of Sciences, B104, Life Science Research Building, 320 Yueyang Road, Shanghai
200031, China. email: [email protected]
Summary
information integrating center. This modulatory circuit might loop the lungs, immune and nervous systems and play a very important role in regulating
lung infection, inflammation and immunity through
the neural innervations and signals when the lungs
encounter pathogenic challenges.
Introduction
macrophages express a7 nAChR that are commonly expressed by neurons.2 Moreover, electric
stimulation of vagus nerve can trigger release of
acetylcholine (ACh), which can activate a7
nAChR in the macrophages and therefore suppress
activation of NF-kB and dampen proinflammatory
cytokine production.3 Studies further revealed that
the spleen plays an important role in mediating
anti-inflammatory activity of vagus nerve.4,5 More
importantly, Tracey group set forth a new working
model for CAP regarding the role of spleen.6 The
main conception is that vagal outflow at splenic
nerve terminus releases adrenergic agonist-norepinephrine that could activate b2 adrenergic receptors
on splenic CD4+CD44highCD62Llow T lymphocytes
that contain choline acetyltransferase (CHAT).
These unique T lymphocytes synthesize ACh that
could act on a7 nAChR-expressing splenic macrophages and suppress NF-kB activity.6 As such, this
theory can be used to interpret the modulatory effects of CAP on proinflammatory responses at
Since the cholinergic anti-inflammatory pathway
(CAP, also called inflammatory reflex) was termed
by Kevin Tracey a decade ago,1 much work has
been done regarding how the parasympathetic,
especially vagus nerve-mediated inflammatory
reflex regulates innate immune responses to injury,
pathogens and tissue ischemia. The neuroimmune
interplay provides the host with a fast, discrete and
localized means of controlling the immune response.1 This review will briefly introduce the
basic formation of pulmonary parasympathetic inflammatory reflex and its regulatory function on
lung infection, inflammation and immunity.
The current status of cholinergic
anti-inflammatory pathway
The remarkable discovery regarding inflammatory
reflex is that proinflammatory cells, especially
! The Author 2014. Published by Oxford University Press on behalf of the Association of Physicians.
All rights reserved. For Permissions, please email: [email protected]
Downloaded from by guest on February 5, 2015
In this review, we first analyzed the current status of
cholinergic anti-inflammatory pathway and then put
forward a novel regulatory machinery—pulmonary
parasympathetic inflammatory reflex, which is composed by lung vagal sensors at afferent arm, a7
nAChR (a7 nicotinic acetylcholine receptors)—
expressing cells at efferent arm and the brain
790
X. Yang et al.
systemic levels. We reported that dysfunction of
cholinergic signaling in the splenocytes might be
associated with postoperative cognitive decline
under metabolic syndrome.7 However, this theory
is challenged by a recent study that vagal efferent
neurons in a rat model neither synapse with splenic
sympathetic neurons nor drive their ongoing activity.8 Thus, it is imperative to rethink the current
CAP working model,9 especially regulatory effects
of pulmonary parasympathetic (vagal) inflammatory
reflex during lung infection, inflammation and
immunity.
Possibility of existence of pulmonary
parasympathetic inflammatory
reflex
The afferent arm of pulmonary
parasympathetic inflammatory
reflex
The vagal nerve endings innervate the distal airways
or alveolar epithelia11,12 and express variety of receptors that can sense mechanical, chemical, biological and other stimuli and convey the signaling
through the afferent vagal nerve to the center of
The efferent arm of pulmonary
parasympathetic inflammatory
reflex
Chemical, mechanical or biological stimuli can directly cause damage of lung alveolar epithelial cells.
Proinflammatory cytokines that are secreted by alveolar macrophages and infiltrated proinflammatory
cells (e.g. monocytes and neutrophils) also contribute to proinflammatory cascades and further lung
injury. Alveolar macrophages, epithelial cells and
inflammatory infiltrated neutrophils express a7
nAChR and could be the players at efferent arm of
pulmonary parasympathetic inflammatory reflex.13,14 Pulmonary vagal nerve endings innervate
lungs and secrete ACh that could activate a7
nAChR-expressing proinflammatory cells at the efferent arm of lung vagal inflammatory reflex, suppress NF-kB activation and secretion of
proinflammatory cytokines therefore lessen the
extent of lung inflammation and injury. This notion
has been strongly supported by the studies that in
acid, LPS and Escherichia Coli-induced acute lung
injury mouse models (by sensing chemical and biological stimuli), activation of a7 nAChR by its agonists could attenuate, on the contrary, vagotomy and
deficiency of a7 nAChR deteriorate the degree of
lung injury.13,14 In a ventilator-induced lung injury
mouse model (by sensing mechanical stretch), vagotomy worsens proinflammatory responses in the
lung.23
Downloaded from by guest on February 5, 2015
The vagus nerve is the 10th cranial nerve and contains sensory (afferent) and motor (efferent) fibers.10
Both right and left vagus nerves descend from the
brain in the carotid sheath, lateral to the carotid artery and then innervate the lungs. Electrical microscopy has shown that vagus nerve fibers were present
in the human lung, especially in the alveoli.11,12
Interestingly, inflammatory responses of the lungs
vary when the pathogenic challenges were stimulated systemically or locally. In an endotoxemia
mouse model (systemic challenge), activation of
a7 nAChR by vagus nerve stimulation reduced the
levels of TNF-a in the plasma, liver and spleen, but
not in the lung.4 However, in an acute lung injury
model (local challenge), expression of a7 nAChR in
alveolar macrophages and neutrophils was
increased. Administration of a7 nAChR agonists
could inhibit NF-kB activity in the BAL (bronchoalveolar lavage) proinflammatory cells and reduce
both TNF-a and MIP-2 levels in the airspaces of
the lung. Vagotomy and deficiency of a7 nAChR
reversed the above mentioned pathological
processes.13,14 These findings indicate a vagal inflammatory circuit may exist between the lung and
nervous system and exert modulatory effects on lung
infection and immunity.
brain (e.g. NTS, nucleus tractus solitorius).15 For
example, vagal C-fiber receptors can sense mechanical and chemical irritant stimuli.16 These vagal
nociceptors can also be activated by proinflammatory cytokines and therefore transmit immune
signals from the lung to the brain.17 Vagal afferent
nerve endings express ion channel-transient
potential channels,18 which can detect the pungent
odor, hot sensations and ion changes. Toll-like receptors 2, 3, 4 and 7 are expressed in the nerve
endings or airway epithelial cells,19 that can respond to bacterial and viral pathogens. In addition,
pulmonary sensory neurons and epithelial cells
are reported to express inflammatory cytokine
receptors (e.g. TNF-a receptor)20,21 which may be
the components of vagal afferent arm of pulmonary
vagal inflammatory reflex. Furthermore, bacteria
and their products could activate sensory neurons
and modulate inflammation that supports the
role of nervous system in host–pathogen
interactions.22
Pulmonary parasympathetic inflammatory reflex
Brain center of pulmonary
parasympathetic inflammatory
reflex
It is assumed that vagal sensors recognize local
proinflammatory cytokines or other mediators and
then transmit the information to brain center. The
information is further processed in the brain, and reflexively acts through the vagal efferent to suppress
proinflammatory
cytokine
production
of
inflammatory cells through release of ACh and activation of a7 nAChR.24 Anatomically, vagus afferent
fibers reside in the nodose ganglion and terminate
primarily within the dorsal vagal complex (DVC) of
the medulla oblongata. The DVC consists of the NTS,
the dorsal motor nucleus of the vagus (DMN) and the
area postrema.25 Stimulation of capsaicin-sensitive
vagal afferents could alter c-fos expression in the
NTS in a mouse model if inoculated with bacteria,26
suggesting that NTS might be the center at brain that
could integrate the information from the vagal sensory fibers, but this hypothesis requires further study.
Airway epithelia and periepithelial tissue are vagally
innervated.11,12 Classically, vagal nerve endings can
directly release ACh upon inflammatory stimulation.
During lung viral infection and inflammation, CHAT
mRNA is upregulated in the lung that could promote
alternative synthesis of ACh in an a7 nAChR-dependent manner (Su X, et al. unpublished data).
These results indicate that vagal nerve endingsreleased ACh might trigger synthesis and secretion
of ACh by the lung cells (possibly epithelial cells or
neuroendocrine cells). This positive feedback between ACh and its receptor maintains a constant
generation of ACh in the alveoli, which can inhibit
NF-kB activity in the a7 nAChR-expressing alveolar
macrophages and infiltrated proinflammatory cells
and therefore dampen lung inflammation and
injury.27
Due to the rapid hydrolysis of ACh (half-life,
2 min), measurement of the concentration of ACh
in the BAL requires prevention of ACh hydrolysis
and advanced techniques (e.g. high performance
liquid chromatography). Indirect way to evaluate
ACh is through measuring AChE activity and choline
(a metabolite of ACh) levels. Study has shown that
AChE activity in proinflammatory cells and BAL choline concentration were markedly increased in the
E. coli pneumonia mice compared with normal
mice.13 Taken together, vagus nerve, lung epithelial
cells and BAL proinflammatory cells may collaboratively participate in the synthesis and hydrolysis of
ACh during lung infection and inflammation.
Responses of pulmonary
parasympathetic inflammatory
reflex to different pathogens
The regulatory effects of pulmonary parasympathetic inflammatory reflex on lung infection and immunity are pattern recognition receptors dependent.
Gram-positive bacterial infection can be recognized
by TLR2. Activation of a7 nAChR by nicotine
increased Streptococcus Pneumoniae-induced lung
infection and lung inflammation.28 TLR4 is the receptor of endotoxin, a component of Gram-negative
bacteria. Activation of a7 nAChR by nicotine reduces mortality of E. coli pneumonia; on the contrary, cervical vagotomy and a7 nAChR deficiency
increases mortality of E. coli pneumonia.14 In addition, TLR3, TLR7 and MAVS-RIG-I systems play key
roles in mediating influenza viral lung infection and
inflammation.29 In vitro experiments have shown
that in the lung epithelial cells, activation of a7
nAChR promotes influenza viral replication.
Deficiency of a7 nAChR protects mice from influenza virus-induced lung infection and inflammation
(Su X, et al. unpublished data). Thus, pulmonary
parasympathetic (vagal) inflammatory reflex is functionally versatile in response to the challenges of
different pathogens.
Perspectives and conclusion
The interaction between nervous and immune systems has been studied for many years, but there are
many key scientific questions we cannot answer.
Particularly, we need to understand how vagus
nerve terminals sense the invading pathogens, how
the infection signals transmit to the brain center and
how the brain responds to infection and regulates the
immune cells to eliminate pathogens. There is more
work to do; nevertheless, this study supports the view
that pulmonary parasympathetic inflammatory reflex
might be present and function as a requisite regulator
for lung infection, inflammation and immunity.
Funding
The National Natural Science Foundation of China
(Grant No.81270139 to X.S.); The Key Project of
Science and Technology of Shanghai (Grant No.
12JC1408900 to X.S.); One Hundred Person
Downloaded from by guest on February 5, 2015
Sources and metabolism of ACh in
the lung
791
792
X. Yang et al.
Project of the Chinese Academy of Sciences
(Grant No. Y316P21209 to X.S.); The Knowledge
Innovation Program of the Chinese Academy of
Sciences (Grant No. Y114P11209 to X.S.).
Conflict of interest: None declared.
References
1. Tracey KJ. The inflammatory reflex. Nature 2002; 420:853–9.
2. Wang H, Yu M, Ochani M, Amella CA, Tanovic M, Susarla S,
et al. Nicotinic acetylcholine receptor alpha7 subunit is
an essential regulator of inflammation. Nature 2003;
421:384–8.
3. Borovikova LV, Ivanova S, Zhang M, Yang H, Botchkina GI,
Watkins LR, et al. Vagus nerve stimulation attenuates the
systemic inflammatory response to endotoxin. Nature 2000;
405:458–62.
4. Huston JM, Ochani M, Rosas-Ballina M, Liao H, Ochani K,
Pavlov VA, et al. Splenectomy inactivates the cholinergic
antiinflammatory pathway during lethal endotoxemia and
polymicrobial sepsis. J Exp Med 2006; 203:1623–8.
6. Rosas-Ballina M, Olofsson PS, Ochani M, Valdes-Ferrer SI,
Levine YA, Reardon C, et al. Acetylcholine-synthesizing T
cells relay neural signals in a vagus nerve circuit. Science
2011; 334:98–101.
7. Su X, Feng X, Terrando N, Yan Y, Chawla A, Koch LG, et al.
Dysfunction of inflammation-resolving pathways is associated with exaggerated postoperative cognitive decline in
a rat model of the metabolic syndrome. Mol Med 2012;
18:1481–90.
8. Bratton BO, Martelli D, McKinley MJ, Trevaks D,
Anderson CR, McAllen RM. Neural regulation of inflammation: no neural connection from the vagus to splenic sympathetic neurons. Exper Physiol 2012; 97:1180–5.
9. Romanovsky AA. The inflammatory reflex: the current model
should be revised. Exper Physiol 2012; 97:1178–9.
15. Ganchrow D, Ganchrow JR, Cicchini V, Bartel DL,
Kaufman D, Girard D, et al. The nucleus of the solitary
tract in the C57BL/6J mouse: Subnuclear parcellation,
chorda tympani nerve projections and brain stem connections. J Comp Neurol 2013; doi: 10.1002/cne.23484.
16. Sant’Ambrogio G, Widdicombe J. Reflexes from airway rapidly adapting receptors. Respir Physiol 2001; 125:33–45.
17. Yu J, Lin S, Zhang J, Otmishi P, Guardiola JJ. Airway nociceptors activated by pro-inflammatory cytokines. Respir
Physiol Neurobiol 2007; 156:116–9.
18. Nassenstein C, Kwong K, Taylor-Clark T, Kollarik M,
Macglashan DM, Braun A, et al. Expression and function of
the ion channel TRPA1 in vagal afferent nerves innervating
mouse lungs. J Physiol 2008; 586:1595–604.
19. Barajon I, Serrao G, Arnaboldi F, Opizzi E, Ripamonti G,
Balsari A, et al. Toll-like receptors 3, 4, and 7 are expressed
in the enteric nervous system and dorsal root ganglia. J
Histochem Cytochem 2009; 57:1013–23.
20. van Houwelingen AH, Kool M, de Jager
van Heuven-Nolsen D, Kraneveld AD,
derived TNF-alpha primes sensory nerve
monary hypersensitivity reaction. J
168:5297–302.
SC, Redegeld FA,
et al. Mast cellendings in a pulImmunol 2002;
21. Saperstein S, Chen L, Oakes D, Pryhuber G, Finkelstein J.
IL-1beta augments TNF-alpha-mediated inflammatory responses from lung epithelial cells. J Interferon Cytokine Res
2009; 29:273–84.
22. Chiu IM, Heesters BA, Ghasemlou N, Von Hehn CA, Zhao F,
Tran J, et al. Bacteria activate sensory neurons that modulate
pain and inflammation. Nature 2013; 501:52–7.
23. dos Santos CC, Shan Y, Akram A, Slutsky AS, Haitsma JJ.
Neuroimmune regulation of ventilator-induced lung injury.
Am J Respir Crit Care Med 2011; 183:471–82.
24. Pavlov VA, Wang H, Czura CJ, Friedman SG, Tracey KJ. The
cholinergic anti-inflammatory pathway: a missing link in
neuroimmunomodulation. Mol Med 2003; 9:125–34.
25. Berthoud HR, Neuhuber WL. Functional and chemical anatomy of the afferent vagal system. Auton Neurosci 2000;
85:1–17.
10. Gallowitsch-Puerta M, Pavlov VA. Neuro-immune interactions via the cholinergic anti-inflammatory pathway. Life
Sci 2007; 80:2325–9.
26. Riley TP, Neal-McKinney JM, Buelow DR, Konkel ME,
Simasko SM. Capsaicin-sensitive vagal afferent neurons contribute to the detection of pathogenic bacterial colonization
in the gut. J Neuroimmunol 2013; 257:36–45.
11. Fox B, Bull TB, Guz A. Innervation of alveolar walls in the
human lung: an electron microscopic study. J Anatomy
1980; 131:683–92.
27. Su X. Leading neutrophils to the alveoli: who is the guider?
Am J Respir Crit Care Med 2012; 186:472–3.
12. Hertweck MS, Hung KS. Ultrastructural evidence for the innervation of human pulmonary alveoli. Experientia 1980;
36:112–3.
28. Giebelen IA, Leendertse M, Florquin S, van der Poll T.
Stimulation of acetylcholine receptors impairs host defence
during pneumococcal pneumonia. Eur Respir J 2009;
33:375–81.
13. Su X, Lee JW, Matthay ZA, Mednick G, Uchida T, Fang X,
et al. Activation of the alpha7 nAChR reduces acid-induced
acute lung injury in mice and rats. Am J Respir Cell Mol Biol
2007; 37:186–92.
29. Pothlichet J, Meunier I, Davis BK, Ting JP, Skamene E, von
Messling V, et al. Type I IFN triggers RIG-I/TLR3/NLRP3dependent inflammasome activation in influenza A virus infected cells. PLoS Pathog 2013; 9:e1003256.
Downloaded from by guest on February 5, 2015
5. Rosas-Ballina M, Ochani M, Parrish WR, Ochani K,
Harris YT, Huston JM, et al. Splenic nerve is required for
cholinergic antiinflammatory pathway control of TNF
in endotoxemia. Proc Natl Acad Sci USA 2008;
105:11008–13.
14. Su X, Matthay MA, Malik AB. Requisite role of the cholinergic alpha7 nicotinic acetylcholine receptor pathway in suppressing Gram-negative sepsis-induced acute lung
inflammatory injury. J Immunol 2010; 184:401–10.