Skip to main content

The airway neuro-immune axis as a therapeutic target in allergic airway diseases

Abstract

Recent evidence has increasingly underscored the importance of the neuro-immune axis in mediating allergic airway diseases, such as allergic asthma and allergic rhinitis. The intimate spatial relationship between neurons and immune cells suggests that their interactions play a pivotal role in regulating allergic airway inflammation. Upon direct activation by allergens, neurons and immune cells engage in interactions, during which neurotransmitters and neuropeptides released by neurons modulate immune cell activity. Meanwhile, immune cells release inflammatory mediators such as histamine and cytokines, stimulating neurons and amplifying neuropeptide production, thereby exacerbating allergic inflammation. The dynamic interplay between the nervous and immune systems suggests that targeting the neuro-immune axis in the airway could represent a novel approach to treating allergic airway diseases. This review summarized recent evidence on the nervous system’s regulatory mechanisms in immune responses and identified potential therapeutic targets along the peripheral nerve-immune axis for allergic asthma and allergic rhinitis. The findings will provide novel perspectives on the management of allergic airway diseases in the future.

Introduction

Allergic airway diseases, such as allergic asthma and allergic rhinitis, are characterized by the prevalence of type 2 helper T (Th2) cells and increased eosinophil infiltration in the airway. While allergic asthma manifests as bronchospasm and airway obstruction, allergic rhinitis presents symptoms such as nasal congestion, rhinorrhea, itching, and sneezing [1, 2]. The immunopathogenic mechanisms underlying allergic airway diseases have been previously elucidated [3, 4]. However, there is emerging evidence indicating the involvement of neurogenic mechanisms in the pathogenesis of allergic airway diseases. Dixon’s research has indicated that the onset of bronchospasm in individuals with asthma is triggered by the activation of neurons in the nasal mucosa, reinforcing the idea that neural mechanisms contribute to the onset of asthma [5]. Moreover, an increase in peptidergic nerves and neuropeptides has been observed in cases of allergic rhinitis [6, 7]. Pharmaceutical compounds, including alpha- and beta-adrenergic agonists, can elicit symptoms of allergic rhinitis. The surgical severing of specific nasal nerves has been demonstrated to alleviate symptoms associated with allergic rhinitis [8, 9]. The findings of these studies suggest that the innervation of peptidergic classes plays a significant role in the neurogenic mechanism of allergic rhinitis.

Recent analysis of the spatial distribution of immune cells and nerve fibers has revealed that the interaction between the immune system and the neurological system regulates allergic airway inflammation, rather than operating independently. Upon activation by cytokines in response to allergens, peripheral neurons convey signals to the central nervous system. This activation induces the release of neuropeptides and neurotransmitters, stimulating neurons through axonal reflexes. Subsequently, neuropeptides and neurotransmitters impact various immune cells, resulting in the generation of inflammatory mediators, including cytokines, lipid mediators, and histamines. Inflammatory mediators, in turn, enhance neuronal excitability, reducing the threshold for neuronal activation in response to stimulation. This establishes a positive feedback loop, activating neurons even in response to subthreshold or non-noxious stimuli, intensifying neuronal activity at the inflammation site [10].

This review summarizes recent findings elucidating the intricate mechanisms by which the nervous system modulates immune responses and identifies potential therapeutic targets for allergic asthma and allergic rhinitis through the airway neuro-immune axis. The insights presented herein provide a novel perspective on the prospective management of allergic airway diseases.

The distribution and expression of peripheral neurons and immune cells in the airway contribute to the intricate crosstalk between these two systems

The intricate crosstalk between peripheral neurons and immune cells in the airway is closely linked to their distribution and expression patterns. The peripheral nervous system, a crucial component of the broader nervous system, consists of the autonomic and somatic nervous systems. Traditionally, the autonomic nervous system comprises sympathetic and parasympathetic nerves, with the vagus nerve holding a pivotal role in parasympathetic regulation, while the enteric nervous system forms part of the autonomic network [11]. Within the respiratory tract, these nerves play a pivotal role, including sympathetic, parasympathetic, and sensory nerves, with primary distribution in the nasal mucosa. They are instrumental in regulating vasomotor activity, blood flow within the nasal cavity, and the transmission of sensory signals from the nose [12]. In both human and murine lungs, the innervation profile exhibits a dense distribution of parasympathetic nerves, with sympathetic nerves being relatively sparse, constituting approximately 20% of the overall lung innervation [13]. These nerves intricately traverse the trachea, bronchi, blood vessels, and provide support to submucosal glands [14, 15].

In a non-inflammatory state, the airway epithelium and smooth muscle layer harbor a limited number of dendritic cells (DCs) and nerve fibers. Most T cells and DCs are located beneath the smooth muscle, with about 10% of DCs and 4% of T cells forming clusters with sensory nerve fibers. Mast cells, essential components of the immune response, are strategically positioned around microvessels, visceral tissue mucosa, submucosa, and smooth muscle, establishing robust connections with nerves through the expression of cell adhesion molecules or the release of neurotrophic factors and cytokines [16,17,18]. Furthermore, the presence of neuronal vascular cell adhesion protein 1 and intercellular adhesion molecule 1 suggests that eosinophils may also be attracted to parasympathetic nerves. This extensive distribution profile underscores the potential for intricate interactions between neurons and immune cells in the airway microenvironment [19]. The interplay between their spatial disposition and functional roles provides a foundation for comprehending the nuanced crosstalk within the peripheral neurons-immune cells axis in the airway.

Peripheral neurons orchestrate airway immune responses during inflammation

The intricate interaction between neurons and immune cells in the airways is orchestrated by primary mediators, encompassing inflammatory mediators, neurotrophins, and neuropeptides. Previous research has predominantly focused on the role of inflammatory mediators in facilitating these interactions. Upon allergen stimulation, epithelial cells release alarmins, such as interleukin (IL)-25, IL-33, and thymic stromal lymphopoietin, directly impacting type 2 innate lymphoid cells (ILC2s). This cascade induces the production of IL-4 and IL-5, activating eosinophils, promoting mast cell proliferation, and stimulating goblet cells, ultimately resulting in airway hyperresponsiveness and increased mucus production. Additionally, IL-4 facilitates DCs in allergen uptake, activating Th2 cells and prompting B lymphocytes to produce allergen-specific IgE antibodies [20, 21].

Cross-linking high-affinity IgE receptors on mast cells and basophils triggers the release of inflammatory mediators, including histamine and prostaglandins. These mediators not only regulate neuronal sensitization by acting on specific membrane receptors but also modulate sodium currents, increasing neuronal excitability. Elevated excitability reduces the stimulation threshold, enabling neurons to be activated by subthreshold or non-injurious stimuli, consequently leading to the release of neuropeptides [22]. Furthermore, small-molecule mediators from other immune cells not only exert pro-inflammatory effects but also directly activate peripheral neurons by binding to receptors expressed on their surface [23, 24].

Studies have highlighted the pivotal role of neurotrophins, including nerve growth factor (NGF), brain-derived neurotrophic factor (BDNF), neurotrophic factor 3 (NT-3), and neurotrophic factor 4/5 (NT-4/5), in mediating interactions between peripheral neurons and immune cells. While higher levels of BDNF have been observed in cases of allergic rhinitis and allergic asthma, further investigations are ongoing to elucidate its impact on nerve fibers. Conversely, NGF has been previously shown to stimulate the formation of nerve fibers and facilitate the infiltration of various immune cells, such as eosinophils and lymphocytes. These immune cells, in turn, trigger Th2 cell-mediated allergic airway inflammation [25, 26].

Neuropeptides play a pivotal role in facilitating communication between peripheral neurons and immune cells. Protease allergens, such as those from house dust mites, directly stimulate TRPV1 + sensory neurons, eliciting the release of neuropeptide SP. This neuropeptide subsequently stimulates DCs and mast cells via Mas-related G-protein coupled receptor members A1 and Mrgprb2, respectively, leading to the generation of pro-inflammatory mediators such as histamine and cytokines. This intricate process triggers additional neuropeptide release from neurons [27, 28]. Consequently, the interaction between neurotransmitters or neuropeptides released by neurons and immune cells in regulating allergic airway inflammation is emerging as a prominent and intriguing area of research.

Sympathetic nerve-ILC2 crosstalk modulates Th2 responses in allergic asthma

Sympathetic nerves play a crucial role in regulating immune responses by releasing the neurotransmitter norepinephrine (NE), which binds to the β2 adrenergic receptor (β2AR). This interaction acts on airway smooth muscle cells to induce bronchodilation. Consequently, β2 adrenergic receptor agonists are commonly employed as bronchodilators in combination with glucocorticoids to alleviate asthma symptoms and reduce airway inflammation [29, 30]. Recent 3D studies of innervation and immunological responses in lung tissue have unveiled that sympathetic innervation inhibits immune responses mediated by LPS or IL-33 in approximately 20% of total axons in the lung. Further investigations have illuminated that sympathetic nerves mediate the suppression of immune responses through signals binding to β2AR [11]. Parallel studies have identified that β2AR expression on mouse ILC2s co-localizes with adrenergic neurons in the intestine, and the absence of β2AR may lead to excessive activation of ILC2s in lung tissue, resulting in the production of type 2 cytokines and inducing lung inflammation. Conversely, the administration of β2AR agonists resulted in a reduction in inflammation [31]. These findings suggest that norepinephrine released from sympathetic nerves, combined with β2AR expressed on the surface of ILC2s, inhibits immune responses by negatively regulating signaling pathways.

Parasympathetic nerves-receptors crosstalk regulates type 2 inflammation in allergic asthma

In the pathophysiology of asthma, acetylcholine (ACh) serves as a neurogenic neurotransmitter released by parasympathetic postganglionic neurons. ACh not only induces smooth muscle contraction and mucus secretion but also fosters airway inflammation, contributing to airway remodeling [32]. Within the airway, various types of acetylcholine receptors, including nicotinic and muscarinic receptors, bind to ACh and are expressed on different cells, such as epithelial cells, macrophages, and T cells [33]. Studies have indicated that M3-type muscarinic receptors exacerbate asthma symptoms, primarily located in the airway smooth muscle and submucosal glands, where they regulate smooth muscle contraction and mucus secretion [34]. In contrast, M2 receptors typically inhibit the secretion of ACh, but under stimuli like allergens and viral infections, dysfunctional M2 receptors may increase ACh release, leading to bronchoconstriction and mucus secretion [35]. However, because the ACh released from the vagus nerve binds to different receptors than the parasympathetic-derived acetylcholine, the effects are opposite. When the vagus nerve is stimulated, it suppresses the inflammatory response [36]. After the vagus nerve is deactivated, the pro-inflammatory cytokines increase, exacerbating lung inflammation. These effects are induced by acetylcholine through the α7 nicotinic acetylcholine receptor (α7nAChR) [37]. Furthermore, studies suggest that α7 nicotinic receptor agonists may directly inhibit ILC2s, thereby reducing type 2 inflammatory responses [38]. Therefore, ACh constricts airways via M3 muscarinic receptors while mitigating type 2 inflammation in asthma through nicotinic receptors.

Parasympathetic nerves also release vasoactive intestinal peptide (VIP) in addition to ACh [39]. VIP plays multiple biological roles in mediating immune function by interacting with specific receptors expressed on various immune cells including chemoattractant receptor-homologous molecule expressed on Th2 cells (CRTH2), vasoactive intestinal peptide receptor type 1 (VPAC-1), vasoactive intestinal peptide receptor type 2 (VPAC-2), and pituitary adenylate cyclase activating polypeptide receptors (PAC1) [40, 41]. VIP acts as an effective anti-inflammatory agent to dilate the bronchial tubes [42]. Mice lacking the VIP gene showed airway inflammation and hyperresponsiveness. Asthma symptoms were relieved after intraperitoneal injection of VIP, indicating that VIP plays an anti-inflammatory role in the development of asthma [43, 44]. Conversely, VIP binding to the VPAC2 receptor upregulates IL-5 and IL-13, promotes the accumulation of eosinophils, and induces the conversion of the Th2 phenotype, altering the immune response to allergy and inflammation [45, 46]. Therefore, immune responses mediated by VIP also vary depending on the interacting receptors.

Sensory nerves and neuroendocrine cells contribute to the pathophysiology of allergic asthma

Upon exposure to allergens, neuropeptides such as substance P (SP), neurokinin A (NKA), and calcitonin gene-related peptide (CGRP) are released from sensory nerves, significantly contributing to the pathophysiology of asthma. Elevated levels of SP are observed in cases of allergic asthma, and its activity is mediated by key receptors, including neurokinin-1 receptors (NK1R) and NK2R. Apart from mediating bronchoconstriction, vasodilation, and increased mucus secretion, SP activates various immune cells, including dendritic cells, macrophages, eosinophils, neutrophils, monocytes, and lymphocytes [28, 47,48,49]. SP induces inflammation in neutrophils by mediating NK1 receptor-induced synthesis of chemokines such as CCL4 and CXCL8 [50], consistent with findings showing a positive correlation between SP expression and asthma severity [51]. Additionally, SP interacting with the Mrgprb2 receptor/MrgprX2 (human) on mast cells leads to the release of pro-inflammatory cytokines, chemokines, and the recruitment of immune cells like neutrophils, monocytes, and macrophages [27]. NKA, also known as tachykinin, collaborates with SP to promote bronchoconstriction and enhance the antigen presentation function of DCs when combined with the NK2R signal, supporting the immune response [52]. So far, SP and NKA have played roles in promoting inflammation and exacerbating asthma.

CGRP is synthesized by neuroendocrine cells and stored in sensory nerve terminals. CGRP directly interacts with ILC2s to release IL-5 and IL-13, leading to the recruitment of eosinophils and triggering the Th2 response [53]. Furthermore, CGRP regulates Th9 responses, and stimulates the production of GATA3 and IL-9, amplifying airway inflammation [42]. However, studies demonstrate that CGRP has anti-inflammatory properties affecting the antigen presentation of DCs, inhibiting DC maturation in mouse lungs, subsequently reducing the activation and proliferation of antigen-specific T cells, and increasing the number of regulatory T cells [54]. Research on the multimodal regulation of DC function by nociceptors in specialized sensory nerve endings suggests that CGRP enhances the sentinel function of antigen capture and recognition by DCs in the absence of immune stimulation. In contrast, with simultaneous itching or pain and pathogen presence, activated sensory nerves release the chemokine CCL2, attracting DCs to interact and upregulating the expression of IL-12 p40 and IL-6, shared by IL-12 and IL-23, thereby increasing DC cytokine effects [55]. Therefore, further research on CGRP’s function is necessary (Fig. 1; Table 1).

Sympathetic nerves influence airway immunity in allergic rhinitis

In addition to norepinephrine, secreted by sympathetic nerves in the nasal mucosa and acting as a potent vasoconstrictor, sympathetic nerves also release neuropeptide Y (NPY). NPY-secreting nerve fibers are predominantly located in arteries and veins, with a smaller number in the epithelium and glands. This distribution suggests that NPY plays a crucial role in regulating blood flow [56]. For example, in the human nasal mucosa, NPY may constrict arteries and regulate vasomotion [57]. Additionally, NPY enhances the vasoconstrictive effects of NE [58]. While sympathetic nerves predominantly maintain vascular tone under normal circumstances, NPY counters the proinflammatory and vasodilatory effects of neurotransmitters and neuropeptides released from parasympathetic and sensory nerves in allergic conditions [59, 60].

Parasympathetic nerves recruit eosinophils to regulate airway immunity in allergic rhinitis

Parasympathetic fibers intricately innervate the blood vessels and eccrine glands within the nasal mucosa, with a predominant presence in the glands [61]. A comprehensive examination of the neural atlas of the nasal mucosa reveals a significant increase in nerve fibers containing neuropeptides, particularly VIPergic fibers. This emphasizes the significance of VIP innervation in allergic rhinitis [62]. Previous research has elucidated that VIP exerts its biological functions through two receptors, VPAC-1 and VPAC-2 [63]. Recent studies underscore a novel neuroimmune axis, the VIP-CRTH2 axis, which recruits eosinophils. In patients with allergic rhinitis (AR), nasal cavity stimulation leads to a significant increase in neurocrine VIP content, and the secreted VIP recruits eosinophils through the CRTH2 receptor [64]. Furthermore, eosinophils secrete prostaglandin D2 (PGD2), which competes with VIP for CRTH2 receptor signaling, contributing to eosinophil attraction. Leukocytes isolated from nasal secretions of AR patients reveal that CRTH2 is upregulated specifically during the late phase of the response, indicating a crucial role for CRTH2 in the significant recruitment of eosinophils to the inflamed allergic nose [64]. VIP may also modulate the release of acetylcholine, promote vasodilation, and enhance the glandular secretory response, potentially leading to nasal congestion [65].

Sensory nerves and neuroendocrine cells influence the pathophysiology of allergic rhinitis

Neuropeptides, including SP, NKA, and CGRP, play a significant role in the regulation of allergic rhinitis. Studies have demonstrated a notable increase in the expression of neuropeptides such as CGPG and SP in the nasal cavity of individuals with allergic rhinitis (AR) compared to control participants [66]. SP and NKA act as chemoattractants and activators of various immune cells in AR, leading to cell infiltration and aggravation of inflammation. Additionally, they induce vasodilation in the nasal mucosa, plasma extravasation, and glandular secretion [66, 67]. The application of SP to the nasal mucosa in humans results in elevated mRNA expression of pro-inflammatory cytokines IL-1, IL-3, IL-5, IL-6, TNFα, and IFN-γ. This suggestes that SP may regulate allergic responses by increasing the production of specific regulatory cytokines [68].

In allergic rhinitis, the binding sites of CGRP are mainly located in small arteries, consistent with CGRP serving as a sustained and enduring arterial dilator. Consequently, CGRP may induce prolonged dilation of arterial vessels, leading to increased blood flow in the nasal cavity and subsequently leakage of albumin and plasma water into the nasal cavity [69] (Fig. 1; Table 1).

Fig. 1
figure 1

Schematic diagram illustrating the interaction between airway nerve and allergic airway cells

Table 1 Summarizing the impact of airway nerves on targeted cells in allergic airway diseases

The clinical application of the airway neuro-immune axis as a therapeutic target in allergic airway diseases

A potential starting point for treating allergic airway diseases lies in critical aspects of clinical therapy. Emerging research on the interaction between the nervous system and the immune system provides novel insights and approaches for allergic respiratory diseases. Generally, drugs for treating allergic airway diseases can be categorized into three main groups:

Drugs targeting inflammatory mediators and receptors released by immune cells

In the management of allergic airway diseases, medications targeting inflammatory mediators released by immune cells include TSLP mAb, IL-5 mAb, IL-13 mAb, and IL-33 mAb. Drugs that target pro-inflammatory mediator receptors encompass histamine Type 1/2 receptor antagonists and IL-4 receptor alpha (IL4-Ra) mAb. Recently, the development of drugs targeting chemokine receptors acting on CCR3 has shown promise. Studies have introduced a novel peptide nanoparticle CCR3 inhibitor effectively preventing eosinophil recruitment to the lungs and airways, along with mitigating airway hyperresponsiveness, thereby reducing inflammation [70].

Drugs targeting ion channels that activate neurons

Drugs targeting ion channels activating neurons are also under investigation. The Transient Receptor Potential Cation Channel Subfamily V Member 1 (TRPV1) is a crucial ion channel receptor in sensory neurons, and its overexpression induced by eosinophils can be mitigated by capsaicin binding to TRPV1 [71]. Conversely, treatment with a TRPV1 antagonist reduces airway hyperresponsiveness and inflammation [63].

Drugs targeting neurotransmitters and neuropeptides released by neurons and their receptors

Drugs that target neurotransmitters and neuropeptides released from neurons and their receptors have broad applications in influencing neuroimmune interactions. Clinically, the standard treatment drugs for asthma include short-acting or long-acting β2-receptor agonists (such as albuterol and formoterol) because β2 receptor agonists have been traditionally recognized as bronchodilators. Recent studies have revealed that the signaling of norepinephrine released from pulmonary sympathetic nerves to β2 receptors can not only dilate airways but also reduce lung inflammation through favorable immunomodulatory effects [13, 72]. The mechanism of action may be that the signaling of β2-adrenergic receptors inhibits the function of ILC2s [31]. In addition, medications that target acetylcholine receptors, such as inhaled selective M3 receptor antagonists used as bronchodilators, are currently employed in the treatment of asthma and COPD [73, 74]. In cases of allergic rhinitis and allergic asthma, where eosinophil inflammation is predominant, the use of anti-CRTH2 antibodies is beneficial. This is because eosinophils express CRTH2 at high levels, but not VPAC1 or VPAC2, leading to a significant decrease in VIP binding [40].

Recently, neuropeptides have been discovered as a new therapeutic target. The exogenous administration of α-CGRP reduces the level of eosinophils and increases the production of regulatory T cells, suggesting that CGRP may serve as an anti-inflammatory mediator and a new target for therapy [54, 75]. Because VIP serves a variety of biological purposes, VIP analogs and/or antagonists may offer improved therapeutic alternatives for treating allergic diseases [76, 77]. The role of SP receptor antagonists and NK1R antagonists in human clinical research is unclear [78]. However, studies have demonstrated that NK2 receptor antagonists (such as tachykinin) partially inhibit NKA-induced bronchoconstriction in asthmatic patients. As a result, tachykinin receptor antagonists may be used to treat asthma [35]. All of these medications influence neurotransmitters and neuropeptides, either directly or indirectly, inhibiting the inflammatory response and relieving allergy symptoms.

Moreover, alternative treatments such as psychotherapy and neurorehabilitation may contribute to managing allergic airway diseases. Studies have demonstrated a direct relationship between asthma onset/severity and the patient’s mood and stress. Negative emotions’ impact on the brain can increase systemic inflammation and pro-inflammatory cytokines [79]. Psychotherapy or neurorehabilitation methods, such as meditation, exercise, and social interaction, can reduce inflammation by modulating neural circuit activity and the function of brain centers involved in asthma [80].

Conclusions

As our scientific exploration into allergic airway diseases advances, we transcend the conventional view of attributing them solely to the autonomous functions of the immune or nervous system. The revelation of the airway neuro-immune axis brings forth novel perspectives on allergic airway diseases, emphasizing the crucial role of the nervous system in regulating immune responses, thus influencing drug treatments. Despite significant progress in immune-targeted therapy for allergic airway inflammation, drug resistance poses a substantial challenge. Therefore, future research aims to delve deeper into this field, seeking a more profound comprehension of the cellular and molecular mechanisms through which the nervous system regulates immune responses via the airway neuro-immune axis. This exploration holds the potential to unveil more effective therapeutic options for managing allergic airway diseases.

Data availability

No datasets were generated or analysed during the current study.

Abbreviations

ACh:

Acetylcholine

α7nAChR:

α7 nicotinic acetylcholine receptor

AR:

Allergic rhinitis

BDNF:

Brain-derived neurotrophic factor

β2AR:

β2 adrenergic receptor

CGRP:

Calcitonin gene-related peptide

CRTH2:

Chemoattractant receptor-homologous molecule expressed on Th2 cells

DCs:

Dendritic cells

ILC2s:

Type 2 innate lymphoid cells

IL-25:

Interleukin (IL)-25

IL4-Ra:

IL-4 receptor alpha

NE:

Norepinephrine

NGF:

Nerve growth factor

NKA:

Neurokinin A

NK1R:

Neurokinin-1 receptors

NPY:

Neuropeptide Y

NT:

Nneurotrophic factor

PAC1:

Pituitary adenylate cyclase activating polypeptide receptors

PGD2:

Prostaglandin D2

SP:

Substance P

Th2:

Type 2 helper T

TRPV1:

Transient Receptor Potential Cation Channel Subfamily V member 1

VIP:

Vasoactive intestinal peptide

VPAC-1/2:

Vasoactive intestinal peptide receptor type 1/2

References

  1. Cai S, Lou H. [Neuroimmunomodulation in allergic rhinitis]. Lin Chung Er Bi Yan Hou Tou Jing Wai Ke Za Zhi. 2021;35:859–64.

    Google Scholar 

  2. Komlósi ZI, Van De Veen W, Kovács N, Szűcs G, Sokolowska M, O’Mahony L, et al. Cellular and molecular mechanisms of allergic asthma. Mol Aspects Med. 2022;85:100995.

    Article  PubMed  Google Scholar 

  3. Ouyang S, Liu C, Xiao J, Chen X, Lui AC, Li X. Targeting IL-17A/glucocorticoid synergy to CSF3 expression in neutrophilic airway diseases. JCI Insight. 2020;5:e132836.

    Article  PubMed  PubMed Central  Google Scholar 

  4. Shamji MH, Sharif H, Layhadi JA, Zhu R, Kishore U, Renz H. Diverse immune mechanisms of allergen immunotherapy for allergic rhinitis with and without asthma. J Allergy Clin Immunol. 2022;149:791–801.

    Article  CAS  PubMed  Google Scholar 

  5. Dixon WE, Brodie TG. Contributions to the physiology of the lungs. J Physiol. 1903;29:97–173.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. Heppt W, Peiser C, Cryer A, Dinh QT, Zweng M, Witt C, et al. Innervation of human nasal mucosa in environmentally triggered hyperreflectoric rhinitis. J Occup Environ Med. 2002;44:924–9.

    Article  CAS  PubMed  Google Scholar 

  7. Lacroix JS, Kurt AM, Pochon N, Bretton C, Lundberg JM, Deshusses J. Neutral endopeptidase activity and concentration of sensory neuropeptide in the human nasal mucosa. Eur Arch Otorhinolaryngol. 1995;252:465–8.

    Article  CAS  PubMed  Google Scholar 

  8. Fowler J, Chin CJ, Massoud E. Rhinitis medicamentosa: a nationwide survey of Canadian otolaryngologists. J of Otolaryngol - Head & Neck Surg. 2019;48:70.

    Article  Google Scholar 

  9. Qi Y, Liu J, Peng S, Hou S, Zhang M, Wang Z. Efficacy of selective vidian neurectomy for allergic rhinitis combined with chronic rhinosinusitis. ORL. 2021;83:327–34.

    Article  PubMed  Google Scholar 

  10. Kabata H, Artis D. Neuro-immune crosstalk and allergic inflammation. J Clin Invest. 2019;129:1475–82.

    Article  PubMed  PubMed Central  Google Scholar 

  11. Murtazina A, Adameyko I. The peripheral nervous system. Development. 2023;150:dev201164.

    Article  CAS  PubMed  Google Scholar 

  12. Klimov V, Cherevko N, Klimov A, Novikov P. Neuronal-immune cell units in allergic inflammation in the nose. IJMS. 2022;23:6938.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Liu T, Yang L, Han X, Ding X, Li J, Yang J. Local sympathetic innervations modulate the lung innate immune responses. Sci Adv. 2020;6:eaay1497.

    Article  CAS  PubMed  PubMed Central  ADS  Google Scholar 

  14. Barnes PJ. Neural control of human airways in health and disease. Am Rev Respir Dis. 1986;134(6):1289–314.

    CAS  PubMed  Google Scholar 

  15. Kummer W, Lips KS, Pfeil U. The epithelial cholinergic system of the airways. Histochem Cell Biol. 2008;130:219–34.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Veres TZ, Shevchenko M, Krasteva G, Spies E, Prenzler F, Rochlitzer S, et al. Dendritic cell-nerve clusters are sites of T cell proliferation in allergic airway inflammation. Am J Pathol. 2009;174:808–17.

    Article  PubMed  PubMed Central  Google Scholar 

  17. Undem BJ, Taylor-Clark T. Mechanisms underlying the neuronal-based symptoms of allergy. J Allergy Clin Immunol. 2014;133:1521–34.

    Article  PubMed  PubMed Central  Google Scholar 

  18. Furuno T, Hagiyama M, Sekimura M, Okamoto K, Suzuki R, Ito A, et al. Cell adhesion molecule 1 (CADM1) on mast cells promotes interaction with dorsal root ganglion neurites by heterophilic binding to nectin-3. J Neuroimmunol. 2012;250:50–8.

    Article  CAS  PubMed  Google Scholar 

  19. Drake MG, Lebold KM, Roth-Carter QR, Pincus AB, Blum ED, Proskocil BJ, et al. Eosinophil and airway nerve interactions in asthma. J Leukoc Biol. 2018;104:61–7.

    Article  CAS  PubMed  Google Scholar 

  20. Morianos I, Semitekolou M. Dendritic cells: critical regulators of allergic asthma. IJMS. 2020;21:7930.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Duchesne M, Okoye I, Lacy P. Epithelial cell alarmin cytokines: Frontline mediators of the asthma inflammatory response. Front Immunol. 2022;13:975914.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Michaelis M, Vogel C, Blenk K-H, Arnarson A, Jänig W. Inflammatory mediators sensitize acutely axotomized nerve fibers to mechanical stimulation in the rat. J Neurosci. 1998;18:7581–7.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Oetjen LK, Kim BS. Interactions of the immune and sensory nervous systems in atopy. FEBS J. 2018;285:3138–51.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Campion M, Smith L, Gatault S, Métais C, Buddenkotte J, Steinhoff M. Interleukin-4 and interleukin-13 evoke scratching behaviour in mice. Exp Dermatol. 2019;28:1501–4.

    Article  CAS  PubMed  Google Scholar 

  25. Weihrauch T, Limberg MM, Gray N, Schmelz M, Raap U. Neurotrophins: neuroimmune interactions in human atopic diseases. IJMS. 2023;24:6105.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Samario-Román J, Larqué C, Pánico P, Ortiz-Huidobro RI, Velasco M, Escalona R, et al. NGF and its role in immunoendocrine communication during metabolic syndrome. IJMS. 2023;24:1957.

    Article  PubMed  PubMed Central  Google Scholar 

  27. Green DP, Limjunyawong N, Gour N, Pundir P, Dong X. A mast-cell-specific receptor mediates neurogenic inflammation and pain. Neuron. 2019;101:412–420e3.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Perner C, Flayer CH, Zhu X, Aderhold PA, Dewan ZNA, Voisin T, et al. Substance P release by sensory neurons triggers dendritic cell migration and initiates the Type-2 immune response to allergens. Immunity. 2020;53:1063–1077e7.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Pelaia C, Crimi C, Crimi N, Ricciardi L, Scichilone N, Valenti G, et al. Indacaterol/glycopyrronium/mometasone fixed dose combination for uncontrolled asthma. Expert Rev Respir Med. 2022;16(2):183–95.

    Article  CAS  PubMed  Google Scholar 

  30. Beasley R, Harrison T, Peterson S, Gustafson P, Hamblin A, Bengtsson T, et al. Evaluation of budesonide-formoterol for maintenance and reliever therapy among patients with poorly controlled asthma: a systematic review and meta-analysis. JAMA Netw Open. 2022;5:e220615.

    Article  PubMed  PubMed Central  Google Scholar 

  31. Moriyama S, Brestoff JR, Flamar A-L, Moeller JB, Klose CSN, Rankin LC, et al. b2-adrenergic receptor–mediated negative regulation of group 2 innate lymphoid cell responses. Science. 2018;359(6379):1056–61.

    Article  CAS  PubMed  ADS  Google Scholar 

  32. Kistemaker LEM, Gosens R. Acetylcholine beyond bronchoconstriction: roles in inflammation and remodeling. Trends Pharmacol Sci. 2015;36:164–71.

    Article  CAS  PubMed  Google Scholar 

  33. Wessler I, Kirkpatrick CJ. Cholinergic signaling controls immune functions and promotes homeostasis. Int Immunopharmacol. 2020;83:106345.

    Article  CAS  PubMed  Google Scholar 

  34. Joos GF. The role of neuroeffector mechanisms in the pathogenesis of asthma. Curr Allergy Asthma Rep. 2001;1:134–43.

    Article  CAS  PubMed  Google Scholar 

  35. Joos, Germonpré P. Neural mechanisms in asthma: neural mechanisms in asthma. Clin Experimental Allergy. 2000;30:60–5.

    Article  Google Scholar 

  36. Norcliffe-Kaufmann L. The Vagus and glossopharyngeal nerves in two autonomic disorders. J Clin Neurophysiol. 2019;36:443–51.

    Article  PubMed  Google Scholar 

  37. Pinheiro NM, Banzato R, Tibério I, Prado MAM, Prado VF, Hamouda AK, et al. Acute lung injury in cholinergic-deficient mice supports anti-inflammatory role of α7 nicotinic acetylcholine receptor. IJMS. 2021;22:7552.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. Yuan F, Jiang L, Li Q, Sokulsky L, Wanyan Y, Wang L, et al. A selective α7 nicotinic acetylcholine receptor agonist, PNU-282987, attenuates ILC2s activation and alternaria-induced airway inflammation. Front Immunol. 2021;11:598165.

    Article  PubMed  PubMed Central  Google Scholar 

  39. Kawashima M, Yajima T, Tachiya D, Kokubun S, Ichikawa H, Sato T. Parasympathetic neurons in the human submandibular ganglion. Tissue Cell. 2021;70:101496.

    Article  CAS  PubMed  Google Scholar 

  40. El-Shazly AE, Begon DY, Kustermans G, Arafa M, Dortu E, Henket M, et al. Novel association between vasoactive intestinal peptide and CRTH2 receptor in recruiting eosinophils. J Biol Chem. 2013;288:1374–84.

    Article  CAS  PubMed  Google Scholar 

  41. Langer I, Jeandriens J, Couvineau A, Sanmukh S, Latek D. Signal transduction by VIP and PACAP receptors. Biomedicines. 2022;10:406.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Pavón-Romero GF, Serrano-Pérez NH, García-Sánchez L, Ramírez-Jiménez F, Terán LM. Neuroimmune pathophysiology in asthma. Front Cell Dev Biol. 2021;9:663535.

    Article  PubMed  PubMed Central  Google Scholar 

  43. Szema AM, Hamidi SA, Lyubsky S, Dickman KG, Mathew S, Abdel-Razek T, et al. Mice lacking the VIP gene show airway hyperresponsiveness and airway inflammation, partially reversible by VIP. Am J Physiol Lung Cell Mol Physiol. 2006;291:L880–886.

    Article  CAS  PubMed  Google Scholar 

  44. Hamidi SA, Szema AM, Lyubsky S, Dickman KG, Degene A, Mathew SM, et al. Clues to VIP function from knockout mice. Ann N Y Acad Sci. 2006;1070:5–9.

    Article  CAS  PubMed  ADS  Google Scholar 

  45. Nussbaum JC, Van Dyken SJ, von Moltke J, Cheng LE, Mohapatra A, Molofsky AB, et al. Type 2 innate lymphoid cells control eosinophil homeostasis. Nature. 2013;502:245–8.

    Article  CAS  PubMed  PubMed Central  ADS  Google Scholar 

  46. Villanueva-Romero R, Gutiérrez-Cañas I, Carrión M, González-Álvaro I, Rodríguez-Frade JM, Mellado M, et al. Activation of th lymphocytes alters pattern expression and cellular location of VIP receptors in healthy donors and early arthritis patients. Sci Rep. 2019;9:7383.

    Article  CAS  PubMed  PubMed Central  ADS  Google Scholar 

  47. Jean EE, Good O, Rico JMI, Rossi HL, Herbert DR. Neuroimmune regulatory networks of the airway mucosa in allergic inflammatory disease. J Leukoc Biol. 2022;111:209–21.

    Article  CAS  PubMed  Google Scholar 

  48. O’Connor TM, O’Connell J, O’Brien DI, Goode T, Bredin CP, Shanahan F. The role of substance P in inflammatory disease. J Cell Physiol. 2004;201:167–80.

    Article  PubMed  Google Scholar 

  49. Wang N, Wang J, Zhang Y, Hu S, Zhang T, Wu Y, et al. Substance P-induced lung inflammation in mice is mast cell dependent. Clin Experimental Allergy. 2022;52:46–58.

    Article  CAS  Google Scholar 

  50. Spitsin S, Meshki J, Winters A, Tuluc F, Benton TD, Douglas SD. Substance P-mediated chemokine production promotes monocyte migration. J Leukoc Biol. 2017;101:967–73.

    Article  CAS  PubMed  Google Scholar 

  51. Drake MG, Scott GD, Blum ED, Lebold KM, Nie Z, Lee JJ, et al. Eosinophils increase airway sensory nerve density in mice and in human asthma. Sci Transl Med. 2018;10:eaar8477.

    Article  PubMed  PubMed Central  Google Scholar 

  52. Ohtake J, Kaneumi S, Tanino M, Kishikawa T, Terada S, Sumida K, et al. Neuropeptide signaling through neurokinin-1 and neurokinin-2 receptors augments antigen presentation by human dendritic cells. J Allergy Clin Immunol. 2015;136:1690–4.

    Article  CAS  PubMed  Google Scholar 

  53. Sui P, Wiesner DL, Xu J, Zhang Y, Lee J, Dyken SV. Pulmonary neuroendocrine cells amplify allergic asthma responses. Science. 2018;360(6393):eaan8546.

    Article  PubMed  PubMed Central  Google Scholar 

  54. Peng L-H, Qin X-Q, Tan R-R, Liu C, Liu H-J, Qu X. Calcitonin gene-related peptide regulates the potential antigen uptake ability of human bronchial epithelial cells. J Interferon Cytokine Res. 2018;38:463–8.

    Article  CAS  PubMed  Google Scholar 

  55. Hanč P, Gonzalez RJ, Mazo IB, Wang Y, Lambert T, Ortiz G, et al. Multimodal control of dendritic cell functions by nociceptors. Science. 2023;379:eabm5658.

    Article  PubMed  PubMed Central  Google Scholar 

  56. Shende P, Desai D. Physiological and therapeutic roles of Neuropeptide Y on Biological functions. Adv Exp Med Biol. 2020;1237:37–47.

    Article  CAS  PubMed  Google Scholar 

  57. Baraniuk JN, Castellino S, Dilling Lundgren J, Goff J, Mullol J, Merida M, et al. Neuropeptide Y (NPY) in human nasal mucosa. Am J Respir Cell Mol Biol. 1990;3:165–73.

    Article  CAS  PubMed  Google Scholar 

  58. Potter EK. Neuropeptide Y as an autonomic neurotransmitter. Pharmacol Ther. 1988;37:251–73.

    Article  CAS  PubMed  Google Scholar 

  59. Holzer P. Local effector functions of capsaicin-sensitive sensory nerve endings: involvement of tachykinins, calcitonin gene-related peptide and other neuropeptides. Neuroscience. 1988;24:739–68.

    Article  CAS  PubMed  Google Scholar 

  60. Barnes PJ. Neuropeptides in human airways: function and clinical implications. Am Rev Respir Dis. 1987;136:77–83.

    Article  ADS  Google Scholar 

  61. Van Gerven L, Boeckxstaens G, Hellings P. Up-date on neuro-immune mechanisms involved in allergic and non-allergic rhinitis. Rhin. 2012;50:227–35.

    Article  Google Scholar 

  62. Fischer A, Wussow A, Cryer A, Schmeck B, Noga O, Zweng M, et al. Neuronal plasticity in persistent perennial allergic rhinitis. J Occup Environ Med. 2005;47:20–5.

    Article  CAS  PubMed  Google Scholar 

  63. Kim J-H. The emerging role of TRPV1 in airway inflammation. Allergy Asthma Immunol Res. 2018;10:187.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  64. El-Shazly A, Roncarati P, Lejeune M, Lefebvre P, Delvenne P. Tyrosine kinase inhibition is an important factor for gene expression of CRTH2 in human eosinophils and lymphocytes: a novel mechanism for explaining eosinophils recruitment by the neuro-immune axis in allergic rhinitis. Int Immunopharmacol. 2017;45:180–6.

    Article  CAS  PubMed  Google Scholar 

  65. Baraniuk JN, Lundgren JD, Okayama M, Mullol J, Merida M, Shelhamer JH, et al. Vasoactive intestinal peptide in human nasal mucosa. J Clin Invest. 1990;86:825–31.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  66. Meng Y, Lu H, Wang C, Wang Y, Meng N, Yang K, et al. Naso-ocular neuropeptide interactions in allergic rhinoconjunctivitis, rhinitis, and conjunctivitis. World Allergy Organization Journal. 2021;14:100540.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  67. Nabe T, Tsuzuike N, Ohtani Y, Mizutani N, Watanabe S, Fujii M, et al. Important roles of tachykinins in the development of allergic nasal hyperresponsiveness in guinea-pigs. Clin Experimental Allergy. 2009;39:138–46.

    Article  CAS  Google Scholar 

  68. Okamoto Y, Shirotori K, Kudo K, Ishikawa K, Ito E, Togawa K, et al. Cytokine expression after the topical administration of substance P to human nasal mucosa. The role of substance P in nasal allergy. J Immunol. 1993;151:4391–8.

    Article  CAS  PubMed  Google Scholar 

  69. Baraniuk JN, Lundgren JD, Goff J, Mullol J, Castellino S, Merida M, et al. Calcitonin gene-related peptide in human nasal mucosa. Am J Physiol. 1990;258:L81–88.

    CAS  PubMed  Google Scholar 

  70. Grozdanovic M, Laffey KG, Abdelkarim H, Hitchinson B, Harijith A, Moon H-G, et al. Novel peptide nanoparticle–biased antagonist of CCR3 blocks eosinophil recruitment and airway hyperresponsiveness. J Allergy Clin Immunol. 2019;143:669–680e12.

    Article  CAS  PubMed  Google Scholar 

  71. Fokkens W, Hellings P, Segboer C. Capsaicin for rhinitis. Curr Allergy Asthma Rep. 2016;16:60.

    Article  PubMed  PubMed Central  Google Scholar 

  72. Bosmann M, Grailer JJ, Zhu K, Matthay MA, Sarma JV, Zetoune FS, et al. Anti-inflammatory effects of β2 adrenergic receptor agonists in experimental acute lung injury. FASEB J. 2012;26:2137–44.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  73. Maarsingh H, Oldenburger A, Han B, Zuidhof AB, Elzinga CRS, Timens W, et al. Effects of (a combination of) the Beta2-adrenoceptor agonist indacaterol and the muscarinic receptor antagonist glycopyrrolate on intrapulmonary airway constriction. Cells. 2021;10:1237.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  74. McIvor ER, McIvor RA. The evolving role of tiotropium in asthma. J Asthma Allergy. 2017;10:231–6.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  75. Rochlitzer S, Veres TZ, Kühne K, Prenzler F, Pilzner C, Knothe S, et al. The neuropeptide calcitonin gene-related peptide affects allergic airway inflammation by modulating dendritic cell function: CGRP modulates dendritic cell function and allergic airway inflammation. Clin Experimental Allergy. 2011;41:1609–21.

    Article  CAS  Google Scholar 

  76. Abad C, Gomariz R, Waschek J. Neuropeptide mimetics and antagonists in the treatment of inflammatory disease: focus on VIP and PACAP. CTMC. 2006;6:151–63.

    Article  CAS  Google Scholar 

  77. Linden A. Bronchodilation by an inhaled VPAC2 receptor agonist in patients with stable asthma. Thorax. 2003;58:217–21.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  78. Voisin T, Bouvier A, Chiu IM. Neuro-immune interactions in allergic diseases: novel targets for therapeutics. Int Immunol. 2017;29:247–61.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  79. Denlinger LC, Heymann P, Lutter R, Gern JE. Exacerbation-prone asthma. J Allergy Clin Immunology: Pract. 2020;8:474–82.

    Google Scholar 

  80. Vafaee F, Shirzad S, Shamsi F, Boskabady MH. Neuroscience and treatment of asthma, new therapeutic strategies and future aspects. Life Sci. 2022;292:120175.

    Article  CAS  PubMed  Google Scholar 

Download references

Funding

This work was supported by the National Natural Science Foundation of China (82370039, 82001702), Guangdong Basic and Applied Basic Research Foundation (2021A1515011045, 2022A1515140075), Scientific Research Project of Traditional Chinese Medicine Bureau of Guangdong Province (20221412), Medical Research Foundation of Guangdong Province (B2023056), the Science Foundation of Dongguan Science and Technology Bureau (20231800940452), The Two Universities’ Paired and Cooperated Research Team Project of Guangdong Medical University (4SG22261G), the Innovation Project for College Students (S202210571065, S202310571086).

Author information

Authors and Affiliations

Authors

Contributions

WW and JL wrote the manuscript; SC and SO revised the manuscript. All the authors reviewed the manuscript.

Corresponding author

Correspondence to Suidong Ouyang.

Ethics declarations

Ethical approval

Not applicable.

Competing interests

The authors declare no competing interests.

Additional information

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Rights and permissions

Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article’s Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article’s Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/. The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated in a credit line to the data.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Wu, W., Li, J., Chen, S. et al. The airway neuro-immune axis as a therapeutic target in allergic airway diseases. Respir Res 25, 83 (2024). https://doi.org/10.1186/s12931-024-02702-8

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s12931-024-02702-8

Keywords