Next Article in Journal
mRNA Expression of SMPD1 Encoding Acid Sphingomyelinase Decreases upon Antidepressant Treatment
Next Article in Special Issue
The Interplay between Autonomic Nervous System and Inflammation across Systemic Autoimmune Diseases
Previous Article in Journal
Application of Nano-Drug Delivery System Based on Cascade Technology in Cancer Treatment
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Infection-Associated Mechanisms of Neuro-Inflammation and Neuro-Immune Crosstalk in Chronic Respiratory Diseases

by
Belinda Camp
,
Sabine Stegemann-Koniszewski
*,† and
Jens Schreiber
Experimental Pneumonology, Department of Pneumonology, University Hospital Magdeburg, Health Campus Immunology, Infectiology and Inflammation (GC-I³), Otto-von-Guericke University Magdeburg, 39120 Magdeburg, Germany
*
Author to whom correspondence should be addressed.
These authors contributed equally to this work.
Int. J. Mol. Sci. 2021, 22(11), 5699; https://doi.org/10.3390/ijms22115699
Submission received: 19 April 2021 / Revised: 21 May 2021 / Accepted: 24 May 2021 / Published: 27 May 2021
(This article belongs to the Special Issue Neuroinflammatory Processes in Organ Homeostasis and Disease)

Abstract

:
Chronic obstructive airway diseases are characterized by airflow obstruction and airflow limitation as well as chronic airway inflammation. Especially bronchial asthma and chronic obstructive pulmonary disease (COPD) cause considerable morbidity and mortality worldwide, can be difficult to treat, and ultimately lack cures. While there are substantial knowledge gaps with respect to disease pathophysiology, our awareness of the role of neurological and neuro-immunological processes in the development of symptoms, the progression, and the outcome of these chronic obstructive respiratory diseases, is growing. Likewise, the role of pathogenic and colonizing microorganisms of the respiratory tract in the development and manifestation of asthma and COPD is increasingly appreciated. However, their role remains poorly understood with respect to the underlying mechanisms. Common bacteria and viruses causing respiratory infections and exacerbations of chronic obstructive respiratory diseases have also been implicated to affect the local neuro-immune crosstalk. In this review, we provide an overview of previously described neuro-immune interactions in asthma, COPD, and respiratory infections that support the hypothesis of a neuro-immunological component in the interplay between chronic obstructive respiratory diseases, respiratory infections, and respiratory microbial colonization.

1. Introduction

Chronic obstructive airway diseases such as chronic obstructive pulmonary disease (COPD) and bronchial asthma are generally characterized by airway inflammation leading to symptoms such as coughing, wheezing, and shortness of breath, and greatly affect the patient’s quality of life [1,2]. Depending on the disease, acute exacerbations occur due to the presence of allergens; pollutants; smoke; cold or dry air; and pathogenic bacteria, viruses or both [3,4]. Among these agents, viral infection is one of the major drivers of asthma exacerbations [5,6], and viral involvement in COPD exacerbations is equally high [7]. Rhinovirus (RV) and respiratory syncytial virus (RSV) are the predominant viruses linked to the development and exacerbation of chronic airway inflammatory diseases [8], but also other viruses like influenza A virus (IAV) have been implicated in acute exacerbations [9]. Respiratory viruses primarily infect and replicate within airway epithelial cells [10]. In chronically inflamed airways, antiviral responses can be impaired leading to sustained inflammation and erroneous infiltration of immune cells and cytokines, in turn resulting in the exacerbation of the underlying disease [11]. A variety of factors such as host genetic predisposition, age, and comorbidities contribute to a patient’s individual risk for exacerbations and altogether define the progression of chronic respiratory diseases. A role for infections and recently also for microbial colonization [12] of the respiratory tract in the onset and progression of chronic respiratory disease has also been appreciated [13], but the underlying mechanisms remain elusive in many points.
There is increasing evidence for neuro-immune crosstalk in inflammation; nevertheless, the role of infections orchestrating this crosstalk is still elusive. While our knowledge regarding neuro-inflammation in chronic respiratory diseases is increasing, also here there still are substantial gaps. These include the contribution to the initiation of disease as well as to the manifestation with respect to inflammatory phenotypes and the risk for exacerbations.
The respiratory tract receives somatosensory afferent innervation from neurons that reside within the dorsal root ganglion and vagal sensory innervation from neurons of the nodose/jugular ganglia [14]. Different neuropeptides secreted by neurons act on the vasculature and immune system, leading to immune cell recruitment and activation [15,16]. In allergic diseases, there is a direct correlation between the number of mast cells and neurons [17,18]. Furthermore, dendritic cells (DCs) are found closely associated to sensory neurons in the airways [19,20], and eosinophils, a key effector cell type in allergic reactions such as in allergic asthma, have been found to co-localize with cholinergic nerves [21].
In this review, we provide an overview of previously described neuro-immune interactions in the two main chronic respiratory diseases, asthma and chronic obstructive pulmonary disease (COPD). Furthermore, we discuss interactions along the neuro-immune axis as a potential mechanism of respiratory infections to contribute to the pathophysiology of chronic respiratory diseases.

2. Chronic Respiratory Diseases

2.1. Bronchial Asthma

Bronchial asthma is one of the most common chronic respiratory diseases and is estimated to affect more than 300 million patients worldwide [22]. The symptoms of asthma vary over time as well as in intensity, and include wheezing, shortness of breath, chest tightness, and coughing [23]. The disease is characterized by bronchial hyper-reactivity (BHR), mucus overproduction, airway narrowing, and airway wall remodeling [24]. Remodeling processes involve an increase in airway smooth muscles, thickening of the subepithelial basement membrane, angiogenesis, neuronal proliferation, and metaplasia of goblet cells [25]. Asthma is an extremely heterogeneous disease and can be divided into non-allergic (intrinsic) and allergic (exogenous) asthma. Depending on the inflammatory endotype, the disease is divided into type 2-low and type-2-high asthma, the latter being the most common endotype [26]. Initial exposure to an allergen induces its uptake by antigen-presenting cells and finally results in an allergen-specific T helper type 2 (Th2)-response mainly mediated by TH2 cells, group 2 innate lymphoid cells (ILC2s), as well as IgE-producing B cells. This response typically includes the secretion of cytokines such as interleukin-4 (IL-4), IL-5, and IL-13 [22]. Re-exposure to the allergen results in degranulation of mast cells through binding to surface-bound IgE, leading to the release of histamine and other pro-inflammatory mediators that drive allergic inflammation [27,28]. Elevated IgE titers and eosinophilia characterize the disease [29,30,31]. Aiming at tailored measures for prevention and therapy, there is a large interest in defining the factors and mechanisms driving onset and chronic inflammation in allergic asthma and bronchial asthma in general. Over the last decades, there has been an increasing recognition that next to respiratory viral infections, also the presence of colonizing microorganisms in the upper airways has an impact [32]. Nevertheless, the interactions between microorganisms, chronic airway inflammation, and the involvement of neuro-inflammation are poorly understood.

2.2. Chronic Obstructive Pulmonary Disease

COPD is a major cause of death worldwide. It is characterized by not fully reversible airflow limitation, deregulated chronic inflammation, and emphysematous destruction of the lungs [33,34]. COPD generally manifests at an older age and the main risk factors are inhaled particles and gases from cigarette smoking and biomass fuel [35]. Nevertheless, there is also increasing evidence that early-life events have an impact on adult lung function [36]. Chronic inflammation in COPD involves the infiltration of inflammatory cells including neutrophils, macrophages/monocytes, lymphocytes, and in some patients also eosinophils into the airways and lung tissue. These infiltrations result in an excessive expression of proteases and inflammatory mediators that promote further accumulation of inflammatory cells. These processes also lead to remodeling of the airway structure [37]. As in asthma, there are highly heterogeneous inflammatory phenotypes in COPD. Also CD8+ Tc2 cells have been shown to be significantly increased in the lungs of COPD patients where they mainly produce IL-4 and IL-5 [38] and asthma/COPD overlap syndromes are being discussed [39]. Comorbidities are ubiquitous among COPD patients and include cardiovascular diseases, osteoporosis, gastrointestinal diseases, and respiratory conditions such as asthma and lung cancer [40,41,42]. Due to its high heterogeneity and the progressive nature of the disease, effective therapy of COPD remains challenging and there is a high need for a better understanding of the different pathomechanisms at play.

3. Neuro-Immune Interactions in Allergic Asthma and COPD

3.1. Effects of Allergen Challenge on Sensory Nerves and Neuropeptide Release in Asthma

Research regarding neuro-immune crosstalk has focused increasingly on neuro-immune interactions in lung tissue and consequences for respiratory diseases like asthma and COPD, thereby highlighting its importance in this context. A number of immune cells share anatomical localization with peripheral neurons and crosstalk may have effects on inflammation [43]. Especially ILC2s were recently identified to play a key role in the induction of type 2 inflammation in the context of asthma [29]. The central nervous system (CNS) is the processing center of the human body and communication with peripheral tissues occurs via the peripheral nervous system (PNS). The brainstem can communicate with the lung via vagal sensory neurons, which are the fundamental units activated by sensory input from the environment [44]. Also, breathing is tightly regulated by the nervous system and several classes of central and peripheral sensory neurons regulate the respiratory cycle [45,46]. Sensory neurons of the vagus nerve are the major source of nerve fibers that innervate the lung and airways [45,47,48]. Lung nociceptors—a specialized subset of sensory neurons—can be stimulated by inhaled allergens such as house dust mites, but also viruses or bacteria, resulting in the calcium-mediated release of neuropeptides [49]. Neuropeptides such as substance P (SP) and calcitonin gene-related peptide (CGRP) generate neurogenic inflammation and modulate innate immune activation during infection [50]. Patients with asthma are hyper-responsive to SP, and SP causes bronchoconstriction, increases mucus secretion, and facilitates cholinergic neurotransmission and plasma leakage [51]. Previous investigations in mice showed that silencing nociceptor neurons reduced airway inflammation and bronchial hyper-responsiveness [49], possibly displaying a future therapeutic approach in allergic airway inflammation by interrupting sensory neuron-immune signaling.
During allergic inflammation, sensory neuron-derived neuropeptides, including SP, vasoactive intestinal peptide (VIP), and neuromedin U (NMU), are released and directly affect immune cells. Vice versa, especially ILC2- and CD4+ T cell-derived IL-5 stimulates nociceptors to release VIP, which further enhances type 2 inflammation [49]. Further, it has been shown that the activation of ILC2s via NMU and the co-administration of NMU with IL-25 amplified allergic inflammation [52].
Mast cells often reside close to peripheral nerve endings where they release a broad range of pro-inflammatory cytokines and chemokines upon activation [53,54]. The neuropeptide SP facilitates immune cell migration and SP-mediated activation of human mast cells leads to the release of multiple pro-inflammatory cytokines via the homologous Mas-related G protein coupled receptor X2 (MRGPRX2) [53]. Recent advances have shown an interaction between this receptor with sensory neurons in the skin [55]. Additionally, its potential role in modulating allergic asthma is being increasingly recognized [56,57] and it now appears that human lung mast cells express MRGPRX2 [58]. Thus, it is possible that upon allergen challenge, SP is released from sensory nerves and activates MRGPRX2-expressing lung mast cells to mediate bronchoconstriction and Th2-mediated immune responses, resulting in mild allergic asthma. MRGPRX2 is upregulated in asthmatic lungs [57]. Therefore, it is likely that hemokinin-1 (HK-1) produced from lung macrophages, bronchial cells and mast cells [59,60], as well as ligands generated from eosinophils [61,62], neutrophils [63], and epithelial cells [64], further activate mast cells via MRGPRX2 to release inflammatory mediators and thereby amplify airway hyper-responsiveness, resulting in allergic asthma exacerbation [65]. Furthermore, Perner et al. revealed that allergen-induced SP release by transient receptor potential vanilloid (TRPV)+ sensory neurons triggers Mas-related G-protein coupled receptor member A1 (MRGPRA-1)-dependent DC migration to the lymph node, initiating the allergic immune response [66].
IL-31 regulates cell proliferation and is involved in tissue remodeling [67]. It also induces the production of inflammatory cytokines and chemokines in human keratinocytes, monocytes, and bronchial epithelial cells [68,69], suggesting that it may positively regulate inflammation in T2 diseases. Lai et al. show that serum and bronchoalveolar lavage (BAL) IL-31 levels were significantly elevated in patients with asthma, and IL-31 receptor alpha (IL-31 RA) mRNA was upregulated in OVA-challenged mice [70], indicating that IL-31 modulates the development of airway inflammation in asthma. Stimulation of dorsal root ganglia with IL-31 led to increased neuronal growth and neurite elongation [71], and IL-31 has been found to be significantly associated with total serum IgE levels, indicating a possible role in the development of asthmatic symptoms and their severity [72]. Currently, the anti-IL-31 receptor antibody NemolizumabTM is being clinically evaluated for the therapy of atopic dermatitis [73].
Regarding neuro-immune communication in the lung, also pulmonary neuroendocrine cells (PNECs) appear to influence T2 inflammation. PNECs are rare endoderm-derived airway epithelial cells that contain dense core vesicles filled with neuropeptides, amines, and neurotransmitters [74]. Their release of CGRP stimulates ILC2s and elicits downstream immune responses. They also induce goblet-cell hyperplasia via the neurotransmitter gamma-aminobutyric acid (GABA), and lungs from human asthmatics show increased PNECs [74].
Taken together, there are multiple interactions between neuronal regulation and T2 inflammation in allergic asthma. Their exact role and contribution to the disease need to be further clarified, possibly providing promising targets for future therapeutic strategies.

3.2. Neuro-Immune Crosstalk in COPD

While there is substantial knowledge reading neuro-immune interactions in allergic asthma, much less is known about the crosstalk between the nervous and the immune system in COPD. The disease is associated with shortness of breath, coughing, and bronchial obstruction. The majority of symptoms cannot only be attributed to immune cell infiltration, and there is evidence for a significant contribution of abnormal activity in bronchopulmonary vagal sensory neurons [75]. Nociceptive nerves—the majority of sensory nerves in the lung—are adept at sensing the type of tissue injury and inflammation that occurs in the lungs in COPD. These fibers are sensitive to decreases in pH and indeed the pH is likely to be decreased in COPD lungs at sites of tissue inflammation [76,77]. It also could be shown that patients with COPD show a reduction in sympathetic and parasympathetic activity [78]. The sympathetic nervous system controls bronchodilation and mucus production, whereas the parasympathetic nervous system controls bronchoconstriction [79].
VIP is one of the most abundantly expressed molecules in the human airways [80]; it relaxes pulmonary and coronary artery smooth muscles and has anti-inflammatory effects in animal models [81]. VIP also acts on bronchial mucus secretion [80]. Furthermore, studies show that VIP reduces cigarette smoke-induced damage of alveolar cells and apoptosis [81]. Two types of VIP receptors have been characterized, including vasoactive intestinal peptide receptors type I (VPAC1R) and vasoactive intestinal peptide receptors type II (VPAC2R). The expression of the VIP receptors VPAC1R and VPAC2R is increased in the central airways of smokers with chronic bronchitis [82]. This finding might suggest that regulation of mucus secretion by VIP is altered in COPD.
During airway inflammation, epithelial damage promotes reflective mechanisms that signal to both adaptive and innate immune cells by exposing vagal nerve endings in the submucosa to the airway lumen, leading to the release of acetylcholine (Ach) [14,83]. The release of Ach results in the activation of muscarinic 1 receptors (M1R) and muscarinic 3 receptors (M3R), which play a pro-inflammatory role in the lung [83,84].
Studies suggest a direct and indirect effect of SP in regulating neutrophil function and influx into inflamed tissues. Neutrophils may express SP, and SP enhances their phagocytic activity and regulates their influx into inflamed tissues [85]. Furthermore, the increase of neutrophil adhesion to bronchial epithelial cells mediated by SP [86] suggests an important role for SP in modulating neutrophilic airway inflammation. As COPD is predominantly characterized by neutrophilic inflammation [87], regulation of neutrophils by SP within this disease is likely. Neutrophils also play a prominent role in a subset of asthmatics. These neutrophilic asthma patients generally show increased disease severity, functional residual capacity, and exacerbations [88]. The regulation of neutrophils by SP could therefore potentially also play a key role in asthma types dominated by neutrophilic inflammation.
In patients with COPD, an over-activation of PNECs compared to healthy subjects has been reported [89]. PNEC chemoreceptors and their secretion of peptides can cause increased chemoresponsiveness to odorants [89] and, therefore, also PNECS could display promising future therapeutic targets for managing COPD. Furthermore, in COPD patients, increased sputum CGRP levels [90] and increased numbers of CGRP-positive cells in the airways [89] have been reported.
While there are numerous implications for neurological involvements in COPD, especially neuro-immune crosstalk and its contribution to the disease need to be clarified in experimental and clinical studies in the future.

4. Neuro-Immune Crosstalk in Respiratory Infection and Microbial Colonization

There is evidence that viruses and bacteria that enter the respiratory tract or are part of the respiratory microbiome can directly activate nociceptors and therefore may have an impact on neuro-immune communication in chronic respiratory diseases [91]. They cause lung inflammation, irritation, and airway hyperreactivity (AHR), and also neuro-immune interactions occur during infection [79].
In general, infections cause the development and exacerbation of chronic airway inflammatory diseases [8,92]. It has long been recognized that respiratory viral infections trigger exacerbations of asthma in both children and adults, whereas for many years bacterial infection was thought to be the main trigger of exacerbations in COPD [93]. However, viral infections cause an increase in neutrophil activation, worsening symptoms and airway remodeling also in COPD [94]. The viruses and bacteria discussed below play a particularly important role in the context of asthma and COPD.

4.1. Infections with Human Rhinovirus

RVs belong to the most frequent viral pathogens in humans and are transmitted from person to person. RVs directly trigger the synthesis of inflammatory cytokines from epithelial cells, smooth muscle cells, and macrophages [95,96,97,98]. Little is known about how rhinoviruses affect the neuro-immune crosstalk in the respiratory tract, but there are studies showing RV infects neuronal cells [99]. RV infection causes upregulation of transient receptor potential (TRP) channels on sensory nerves while SP and toll-like receptor 4 (TLR4) RNA expression were reduced in infected children [99,100]. Subjects with RV infection showed an association between the expression of IL-1β, IL-6, and IL-8, and illness expression [101].
In the CNS, IL-1β is synthesized and released during injury, infection and disease, mediating inflammatory responses. However, IL-1β is also present in the brain under physiological conditions and can influence hippocampal neuronal function [102]. IL-6 is an essential regulatory factor for neuro-immunomodulation in the CNS, protecting neurons and promoting nerve regeneration [103,104]. The changes in IL-6 levels are strictly associated with neuropathological and physiological changes caused by inflammation or infection [105].
A key part of the response to viral infections is the production of interferons, which then activate their specific receptors. Type I interferons act directly on nociceptors [106]. Due to the potential of the cytokines released during infection to affect neurons, RV-infection possibly acts on the local nervous system with implications for any underlying chronic inflammatory respiratory disease.

4.2. Infections with Influenza A Virus

IAV is a highly contagious respiratory pathogen that remains a cause for morbidity and mortality throughout the world. There are several animal reservoirs for IAV, which, together with its genetic instability, constitutes its epidemic and pandemic potential [107,108]. It has been shown in a mouse model that in IAV infection the sympathetic nervous system increases pro-inflammatory cytokines and immune cell recruitment and thereby exacerbates morbidity and mortality [109]. Furthermore, persisting neuro-inflammation has been observed following infection with neurotropic as well as non-neurotropic IAV strains in mice [110]. Although lacking experimental or clinical proof, these findings imply IAV to potentially affect chronic respiratory diseases via modulation of the neuro-immune axis.
Nerve- and airway-associated macrophages (NAMs) are a tissue-resident, self-renewing macrophage population in the lungs and are primarily localized around airways. They are also found close to the sympathetic nerves in the bronchovascular bundle. Ural et al. demonstrated that these cells are critical for regulating many aspects of the immune response and tissue homeostasis following inflammatory stimuli in the lung. During IAV infection in a mouse model, NAMs regulated virus-induced inflammation. They proliferated robustly following IAV infection, and in their absence, the inflammatory response was augmented, resulting in excessive production of inflammatory cytokines and innate immune cell infiltration [111]. Very recently, Verzele et al. showed that pulmonary IAV infection profoundly impacts the vagal sensory ganglia. Differential gene expression was accompanied by infiltration of MHC II+ cells into the vagal sensory ganglia of IAV-infected mice. Furthermore, the total number of CGRP immune-reactive sensory neurons and the number of inflammatory cellular infiltrates in the vagal sensory ganglia were significantly increased [112].
Furthermore, IAV infections are associated with strongly increased inflammatory gene expression in the respiratory tract [113]. The cytokines mainly released during IAV infection include type I and III interferons as well as pro-inflammatory cytokines such as tumor necrosis factor (TNF)-α, IL-1, and IL-6 [114]. While these cytokines on the one hand play a role in asthma and COPD, they potentially also lead to alterations in the local neuronal network. In this context, we have recently observed that IAV infection leads to sustained changes in lung homeostasis and significantly modulates subsequently induced allergic airway inflammation [115].
Altogether, these findings strongly support an influence of IAV infection along the neuro-immune axis, thereby most likely also affecting chronic respiratory diseases.

4.3. Infections with Respiratory Syncytial Virus

RSV is one of the most important respiratory viral pathogens during childhood and is associated with significant mortality. Most children will experience RSV infection by the age of 2 years. The major characteristics and symptoms are acute bronchiolitis, mucosal and submucosal edema, and luminal occlusion [116]. There is evidence that infection with RSV has effects on the local neuro-immune crosstalk and thereby may affect other respiratory diseases. SP is known to have immunomodulatory potential and specifically regulates the functions of T and B lymphocytes, monocytes, and macrophages [117,118]. Experiments in trachea, main stem bronchi, and lungs of rats showed the presence of RSV antigens on the surface and in the cytoplasm of bronchiolar epithelial cells. No virus was detected in the epithelium of more proximal airways, including the trachea and large bronchi [119]. At the same time, SP receptor (neurokinin 1 receptor) expression was upregulated by RSV [119] and such upregulation has also been associated to chronic inflammatory conditions [118]. Furthermore, it could be shown that non-neuronal cells such as lymphocytes and monocytes express SP and its receptors [120,121]. The activation of these cells that are associated with the release of cytokines like TNF-α, IL-1, IL-6, IL-8, and IL-10 [122,123], are of high interest because they can at the same time promote airway inflammation.
Altogether, next to these cytokines, especially SP may be an important contributor to the inflammatory response during RSV infection, as it is produced in the lungs of infected mice and inhibition of SP alters the inflammatory cell infiltrate and cytokine expression.

4.4. Infections and Colonization with Staphylococcus aureus

Staphylococcus aureus (S. aureus) is being recognized as a major co-factor in atopic diseases like allergic asthma as it manipulates skin and mucosal immune responses at different levels [124]. Various studies suggest that exposure to S. aureus and its associated enterotoxins is a risk factor for the development of allergic asthma [125]. Experimentally, we recently showed that intranasally administered S. aureus enterotoxin B harbors a versatile potential to modulate allergic airway inflammation [126]. During S. aureus lung infection, nociceptors regulate protective immunity through CGRP release via the transient receptor potential ion channel 1 (TRPV1). CGRP decreases the recruitment of neutrophils that mediate killing of pathogens. Therefore, nociceptor ablation increases survival and bacterial clearance in the lung [127]. Sensory neurons that express the TRPV1 ion channel are known to mediate AHR in asthma [48], and activators of TRPV1 induce an increased cough response in COPD patients by activating neuronal TRPV1.
In infections with S. aureus, SP can play a likewise significant role, as it has been shown to stimulate the virulence of S. aureus in a reconstructed human epidermis model [128]. Since SP is also found in the lungs, one can speculate an equal or similar effect of SP upon respiratory S. aureus infection. Sio et al. showed that the enhanced release of SP in the lung could be a critical factor in leading to the development of systemic inflammation and pathogenesis of lung organ damage [129].
Future studies will need to elucidate to what extent neuro-immune interactions potentially contribute to the interactions between S. aureus and especially allergic asthma. In this context, also the neuro-immunological consequences of persistent respiratory colonization, as opposed to acute infection, will be of special interest.

5. Conclusions and Future Directions

We believe that we are only beginning to understand an until now underestimated role of neuro-inflammation and neuro-immune crosstalk in the interplay between respiratory infections, respiratory microbial colonization, and chronic respiratory diseases. A number of neuronal effectors such as SP and CGRP have been shown to be affected in asthma, COPD, and respiratory infection, possibly making them relevant targets for intervention. For others, there are substantial knowledge gaps, especially with respect to infection (Table 1).
Based on the findings summarized in this review, we propose a neuro-immunological component to the interactions between chronic respiratory diseases and respiratory infections, as well as colonization (Figure 1). Aiming at the identification and verification of novel therapeutic targets, future clinical as well as experimental studies in combined models will have to identify the relevant disease-associated as well as pathogen-associated pathways that modulate the susceptibility to as well as the progression and exacerbation of chronic respiratory diseases. Specifically, these will on the one hand include the identification of neurological consequences of chronic respiratory diseases that affect immune responses to acute respiratory infections, exacerbating the underlying disease. On the other hand, also neuronal system-associated processes in response to infection or persistent colonization are likely to affect inflammatory pathways in chronic respiratory diseases and need to be defined.

Author Contributions

Conceptualization, B.C., S.S.-K. and J.S.; writing—original draft preparation, B.C. and S.S.-K.; writing—review and editing, B.C., S.S.-K. and J.S.; All authors have read and agreed to the published version of the manuscript.

Funding

Our research is supported by the Mitteldeutsche Gesellschaft für Pneumologie und Thoraxchirurgie (file number 2019-MDGP-PA-001).

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Acknowledgments

We would like to thank Lina Scheithauer (TU Braunschweig, Germany) for support in creating the figure in this article.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Bao, Z.; Xiong, J.; Li, W.; Chen, Z.; Shen, H.; Ying, S. Genomic instability in chronic airway inflammatory diseases. Biomed. J. 2015, 38, 117–124. [Google Scholar]
  2. Calus, L.; Van Zele, T.; Derycke, L.; Krysko, O.; Dutre, T.; Tomassen, P.; Dullaers, M.; Bachert, C.; Gevaert, P. Local inflammation in chronic upper airway disease. Curr. Pharm. Des. 2012, 18, 2336–2346. [Google Scholar] [CrossRef]
  3. Gautier, C.; Charpin, D. Environmental triggers and avoidance in the management of asthma. J. Asthma Allergy 2017, 10, 47–56. [Google Scholar] [CrossRef] [Green Version]
  4. Viniol, C.; Vogelmeier, C.F. Exacerbations of copd. Eur. Respir. Rev. 2018, 27, 170103. [Google Scholar] [CrossRef] [Green Version]
  5. Adeli, M.; El-Shareif, T.; Hendaus, M.A. Asthma exacerbation related to viral infections: An up to date summary. J. Fam. Med. Prim. Care 2019, 8, 2753–2759. [Google Scholar] [CrossRef]
  6. Grissell, T.V.; Powell, H.; Shafren, D.R.; Boyle, M.J.; Hensley, M.J.; Jones, P.D.; Whitehead, B.F.; Gibson, P.G. Interleukin-10 gene expression in acute virus-induced asthma. Am. J. Respir. Crit. Care Med. 2005, 172, 433–439. [Google Scholar] [CrossRef]
  7. Stolz, D.; Papakonstantinou, E.; Grize, L.; Schilter, D.; Strobel, W.; Louis, R.; Schindler, C.; Hirsch, H.H.; Tamm, M. Time-course of upper respiratory tract viral infection and copd exacerbation. Eur. Respir. J. 2019, 54, 1900407. [Google Scholar] [CrossRef]
  8. Jartti, T.; Gern, J.E. Role of viral infections in the development and exacerbation of asthma in children. J. Allergy Clin. Immunol. 2017, 140, 895–906. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  9. Oliver, B.G.; Robinson, P.; Peters, M.; Black, J. Viral infections and asthma: An inflammatory interface? Eur. Respir. J. 2014, 44, 1666–1681. [Google Scholar] [CrossRef] [PubMed]
  10. Costa, L.D.; Costa, P.S.; Camargos, P.A. Exacerbation of asthma and airway infection: Is the virus the villain? J. Pediatr. 2014, 90, 542–555. [Google Scholar] [CrossRef] [Green Version]
  11. Tan, K.S.; Lim, R.L.; Liu, J.; Ong, H.H.; Tan, V.J.; Lim, H.F.; Chung, K.F.; Adcock, I.M.; Chow, V.T.; Wang, Y. Respiratory viral infections in exacerbation of chronic airway inflammatory diseases: Novel mechanisms and insights from the upper airway epithelium. Front. Cell Dev. Biol. 2020, 8, 99. [Google Scholar] [CrossRef] [Green Version]
  12. Diver, S.; Richardson, M.; Haldar, K.; Ghebre, M.A.; Ramsheh, M.Y.; Bafadhel, M.; Desai, D.; Cohen, E.S.; Newbold, P.; Rapley, L.; et al. Sputum microbiomic clustering in asthma and chronic obstructive pulmonary disease reveals a haemophilus-predominant subgroup. Allergy 2020, 75, 808–817. [Google Scholar] [CrossRef] [Green Version]
  13. Huang, Y.J.; Lynch, S.V. The emerging relationship between the airway microbiota and chronic respiratory disease: Clinical implications. Expert Rev. Respir. Med. 2011, 5, 809–821. [Google Scholar] [CrossRef] [Green Version]
  14. Voisin, T.; Bouvier, A.; Chiu, I.M. Neuro-immune interactions in allergic diseases: Novel targets for therapeutics. Int. Immunol. 2017, 29, 247–261. [Google Scholar] [CrossRef] [Green Version]
  15. Brain, S.D.; Williams, T.J. Interactions between the tachykinins and calcitonin gene-related peptide lead to the modulation of oedema formation and blood flow in rat skin. Br. J. Pharmacol. 1989, 97, 77–82. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  16. Maggi, C.A. Tachykinins and calcitonin gene-related peptide (cgrp) as co-transmitters released from peripheral endings of sensory nerves. Prog. Neurobiol. 1995, 45, 1–98. [Google Scholar] [CrossRef]
  17. Hagiyama, M.; Inoue, T.; Furuno, T.; Iino, T.; Itami, S.; Nakanishi, M.; Asada, H.; Hosokawa, Y.; Ito, A. Increased expression of cell adhesion molecule 1 by mast cells as a cause of enhanced nerve-mast cell interaction in a hapten-induced mouse model of atopic dermatitis. Br. J. Dermatol. 2013, 168, 771–778. [Google Scholar] [CrossRef]
  18. Le, D.D.; Schmit, D.; Heck, S.; Omlor, A.J.; Sester, M.; Herr, C.; Schick, B.; Daubeuf, F.; Fähndrich, S.; Bals, R.; et al. Increase of mast cell-nerve association and neuropeptide receptor expression on mast cells in perennial allergic rhinitis. Neuroimmunomodulation 2016, 23, 261–270. [Google Scholar] [CrossRef] [PubMed]
  19. Le, D.D.; Rochlitzer, S.; Fischer, A.; Heck, S.; Tschernig, T.; Sester, M.; Bals, R.; Welte, T.; Braun, A.; Dinh, Q.T. Allergic airway inflammation induces the migration of dendritic cells into airway sensory ganglia. Respir. Res. 2014, 15, 73. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  20. Veres, T.Z.; Rochlitzer, S.; Shevchenko, M.; Fuchs, B.; Prenzler, F.; Nassenstein, C.; Fischer, A.; Welker, L.; Holz, O.; Müller, M.; et al. Spatial interactions between dendritic cells and sensory nerves in allergic airway inflammation. Am. J. Respir. Cell Mol. Biol. 2007, 37, 553–561. [Google Scholar] [CrossRef]
  21. Sawatzky, D.A.; Kingham, P.J.; Court, E.; Kumaravel, B.; Fryer, A.D.; Jacoby, D.B.; McLean, W.G.; Costello, R.W. Eosinophil adhesion to cholinergic nerves via icam-1 and vcam-1 and associated eosinophil degranulation. Am. J. Physiol. Lung Cell. Mol. Physiol. 2002, 282, L1279–L1288. [Google Scholar] [CrossRef] [Green Version]
  22. Lambrecht, B.N.; Hammad, H. The immunology of asthma. Nat. Immunol. 2015, 16, 45–56. [Google Scholar] [CrossRef]
  23. Holgate, S.T.; Wenzel, S.; Postma, D.S.; Weiss, S.T.; Renz, H.; Sly, P.D. Asthma. Nat. Rev. Dis Primers 2015, 1, 15025. [Google Scholar] [CrossRef]
  24. Papi, A.; Brightling, C.; Pedersen, S.E.; Reddel, H.K. Asthma. Lancet 2018, 391, 783–800. [Google Scholar] [CrossRef]
  25. Mims, J.W. Asthma: Definitions and pathophysiology. Int. Forum Allergy Rhinol. 2015, 5 (Suppl. S1), S2–S6. [Google Scholar] [CrossRef]
  26. Matucci, A.; Vultaggio, A.; Maggi, E.; Kasujee, I. Is IgE or eosinophils the key player in allergic asthma pathogenesis? Are we asking the right question? Respir. Res. 2018, 19, 113. [Google Scholar] [CrossRef] [Green Version]
  27. Amin, K. The role of mast cells in allergic inflammation. Respir. Med. 2012, 106, 9–14. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  28. Barnes, P.J. Pathophysiology of allergic inflammation. Immunol. Rev. 2011, 242, 31–50. [Google Scholar] [CrossRef] [PubMed]
  29. Fahy, J.V. Type 2 inflammation in asthma—Present in most, absent in many. Nat. Rev. Immunol. 2015, 15, 57–65. [Google Scholar] [CrossRef] [PubMed]
  30. Foster, P.S.; Maltby, S.; Rosenberg, H.F.; Tay, H.L.; Hogan, S.P.; Collison, A.M.; Yang, M.; Kaiko, G.E.; Hansbro, P.M.; Kumar, R.K.; et al. Modeling t(h) 2 responses and airway inflammation to understand fundamental mechanisms regulating the pathogenesis of asthma. Immunol. Rev. 2017, 278, 20–40. [Google Scholar] [CrossRef] [PubMed]
  31. Voehringer, D.; Reese, T.A.; Huang, X.; Shinkai, K.; Locksley, R.M. Type 2 immunity is controlled by il-4/il-13 expression in hematopoietic non-eosinophil cells of the innate immune system. J. Exp. Med. 2006, 203, 1435–1446. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  32. Holt, P.G.; Strickland, D.H.; Hales, B.J.; Sly, P.D. Defective respiratory tract immune surveillance in asthma: A primary causal factor in disease onset and progression. Chest 2014, 145, 370–378. [Google Scholar] [CrossRef]
  33. Bagdonas, E.; Raudoniute, J.; Bruzauskaite, I.; Aldonyte, R. Novel aspects of pathogenesis and regeneration mechanisms in copd. Int. J. Chron. Obstr. Pulmon. Dis. 2015, 10, 995–1013. [Google Scholar]
  34. Wang, Y.; Xu, J.; Meng, Y.; Adcock, I.M.; Yao, X. Role of inflammatory cells in airway remodeling in copd. Int. J. Chron. Obstr. Pulmon. Dis. 2018, 13, 3341–3348. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  35. Rabe, K.F.; Watz, H. Chronic obstructive pulmonary disease. Lancet 2017, 389, 1931–1940. [Google Scholar] [CrossRef]
  36. Martinez, F.D. Early-life origins of chronic obstructive pulmonary disease. N. Engl. J. Med. 2016, 375, 871–878. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  37. Hogg, J.C.; Timens, W. The pathology of chronic obstructive pulmonary disease. Annu. Rev. Pathol. 2009, 4, 435–459. [Google Scholar] [CrossRef]
  38. Barczyk, A.; Pierzchała, W.; Kon, O.M.; Cosio, B.; Adcock, I.M.; Barnes, P.J. Cytokine production by bronchoalveolar lavage t lymphocytes in chronic obstructive pulmonary disease. J. Allergy Clin. Immunol. 2006, 117, 1484–1492. [Google Scholar] [CrossRef]
  39. Postma, D.S.; Rabe, K.F. The asthma-copd overlap syndrome. N. Engl. J. Med. 2015, 373, 1241–1249. [Google Scholar] [CrossRef] [Green Version]
  40. Decramer, M.; Janssens, W. Chronic obstructive pulmonary disease and comorbidities. Lancet Respir. Med. 2013, 1, 73–83. [Google Scholar] [CrossRef]
  41. Divo, M.; Cote, C.; de Torres, J.P.; Casanova, C.; Marin, J.M.; Pinto-Plata, V.; Zulueta, J.; Cabrera, C.; Zagaceta, J.; Hunninghake, G.; et al. Comorbidities and risk of mortality in patients with chronic obstructive pulmonary disease. Am. J. Respir. Crit. Care Med. 2012, 186, 155–161. [Google Scholar] [CrossRef] [Green Version]
  42. Patel, A.R.; Hurst, J.R. Extrapulmonary comorbidities in chronic obstructive pulmonary disease: State of the art. Expert Rev. Respir. Med. 2011, 5, 647–662. [Google Scholar] [CrossRef]
  43. Kabata, H.; Artis, D. Neuro-immune crosstalk and allergic inflammation. J. Clin. Investig. 2019, 129, 1475–1482. [Google Scholar] [CrossRef] [Green Version]
  44. Chang, R.B.; Strochlic, D.E.; Williams, E.K.; Umans, B.D.; Liberles, S.D. Vagal sensory neuron subtypes that differentially control breathing. Cell 2015, 161, 622–633. [Google Scholar] [CrossRef] [Green Version]
  45. Carr, M.J.; Undem, B.J. Bronchopulmonary afferent nerves. Respirology 2003, 8, 291–301. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  46. Guyenet, P.G.; Stornetta, R.L.; Bayliss, D.A. Central respiratory chemoreception. J. Comp. Neurol. 2010, 518, 3883–3906. [Google Scholar] [CrossRef] [Green Version]
  47. Canning, B.J.; Mori, N.; Mazzone, S.B. Vagal afferent nerves regulating the cough reflex. Respir. Physiol. Neurobiol. 2006, 152, 223–242. [Google Scholar] [CrossRef]
  48. Tränkner, D.; Hahne, N.; Sugino, K.; Hoon, M.A.; Zuker, C. Population of sensory neurons essential for asthmatic hyperreactivity of inflamed airways. Proc. Natl. Acad. Sci. USA 2014, 111, 11515–11520. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  49. Talbot, S.; Abdulnour, R.E.; Burkett, P.R.; Lee, S.; Cronin, S.J.; Pascal, M.A.; Laedermann, C.; Foster, S.L.; Tran, J.V.; Lai, N.; et al. Silencing nociceptor neurons reduces allergic airway inflammation. Neuron 2015, 87, 341–354. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  50. Chiu, I.M.; Heesters, B.A.; Ghasemlou, N.; Von Hehn, C.A.; Zhao, F.; Tran, J.; Wainger, B.; Strominger, A.; Muralidharan, S.; Horswill, A.R.; et al. Bacteria activate sensory neurons that modulate pain and inflammation. Nature 2013, 501, 52–57. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  51. O’Connor, T.M.; O’Connell, J.; O’Brien, D.I.; Goode, T.; Bredin, C.P.; Shanahan, F. The role of substance p in inflammatory disease. J. Cell Physiol. 2004, 201, 167–180. [Google Scholar] [CrossRef]
  52. Wallrapp, A.; Riesenfeld, S.J.; Burkett, P.R.; Abdulnour, R.E.; Nyman, J.; Dionne, D.; Hofree, M.; Cuoco, M.S.; Rodman, C.; Farouq, D.; et al. The neuropeptide nmu amplifies ilc2-driven allergic lung inflammation. Nature 2017, 549, 351–356. [Google Scholar] [CrossRef] [PubMed]
  53. Green, D.P.; Limjunyawong, N.; Gour, N.; Pundir, P.; Dong, X. A mast-cell-specific receptor mediates neurogenic inflammation and pain. Neuron 2019, 101, 412–420.e3. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  54. Héron, A.; Dubayle, D. A focus on mast cells and pain. J. Neuroimmunol. 2013, 264, 1–7. [Google Scholar] [CrossRef] [PubMed]
  55. Wang, Z.; Babina, M. Mrgprx2 signals its importance in cutaneous mast cell biology: Does mrgprx2 connect mast cells and atopic dermatitis? Exp. Dermatol. 2020, 29, 1104–1111. [Google Scholar] [CrossRef] [PubMed]
  56. An, J.; Lee, J.H.; Won, H.K.; Kang, Y.; Song, W.J.; Kwon, H.S.; Cho, Y.S.; Moon, H.B.; Kim, T.B. Clinical significance of serum mrgprx2 as a new biomarker in allergic asthma. Allergy 2020, 75, 959–962. [Google Scholar] [CrossRef] [PubMed]
  57. Manorak, W.; Idahosa, C.; Gupta, K.; Roy, S.; Panettieri, R., Jr.; Ali, H. Upregulation of mas-related g protein coupled receptor x2 in asthmatic lung mast cells and its activation by the novel neuropeptide hemokinin-1. Respir. Res. 2018, 19, 1. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  58. Plum, T.; Wang, X.; Rettel, M.; Krijgsveld, J.; Feyerabend, T.B.; Rodewald, H.R. Human mast cell proteome reveals unique lineage, putative functions, and structural basis for cell ablation. Immunity 2020, 52, 404–416.e5. [Google Scholar] [CrossRef]
  59. Grassin-Delyle, S.; Naline, E.; Buenestado, A.; Risse, P.A.; Sage, E.; Advenier, C.; Devillier, P. Expression and function of human hemokinin-1 in human and guinea pig airways. Respir. Res. 2010, 11, 139. [Google Scholar] [CrossRef] [Green Version]
  60. Sumpter, T.L.; Ho, C.H.; Pleet, A.R.; Tkacheva, O.A.; Shufesky, W.J.; Rojas-Canales, D.M.; Morelli, A.E.; Larregina, A.T. Autocrine hemokinin-1 functions as an endogenous adjuvant for IgE-mediated mast cell inflammatory responses. J. Allergy Clin. Immunol. 2015, 135, 1019–1030.e8. [Google Scholar] [CrossRef] [Green Version]
  61. Amin, K.; Janson, C.; Bystrom, J. Role of eosinophil granulocytes in allergic airway inflammation endotypes. Scand. J. Immunol. 2016, 84, 75–85. [Google Scholar] [CrossRef] [PubMed]
  62. Yasukawa, A.; Hosoki, K.; Toda, M.; Miyake, Y.; Matsushima, Y.; Matsumoto, T.; Boveda-Ruiz, D.; Gil-Bernabe, P.; Nagao, M.; Sugimoto, M.; et al. Eosinophils promote epithelial to mesenchymal transition of bronchial epithelial cells. PLoS ONE 2013, 8, e64281. [Google Scholar] [CrossRef] [PubMed]
  63. Subramanian, H.; Gupta, K.; Guo, Q.; Price, R.; Ali, H. Mas-related gene x2 (mrgx2) is a novel g protein-coupled receptor for the antimicrobial peptide ll-37 in human mast cells: Resistance to receptor phosphorylation, desensitization, and internalization. J. Biol. Chem. 2011, 286, 44739–44749. [Google Scholar] [CrossRef] [Green Version]
  64. Duits, L.A.; Nibbering, P.H.; van Strijen, E.; Vos, J.B.; Mannesse-Lazeroms, S.P.; van Sterkenburg, M.A.; Hiemstra, P.S. Rhinovirus increases human beta-defensin-2 and -3 mrna expression in cultured bronchial epithelial cells. FEMS Immunol. Med. Microbiol. 2003, 38, 59–64. [Google Scholar] [CrossRef] [Green Version]
  65. Thapaliya, M.; Chompunud Na Ayudhya, C.; Amponnawarat, A.; Roy, S.; Ali, H. Mast cell-specific mrgprx2: A key modulator of neuro-immune interaction in allergic diseases. Curr. Allergy Asthma Rep. 2021, 21, 3. [Google Scholar] [CrossRef]
  66. Perner, C.; Flayer, C.H.; Zhu, X.; Aderhold, P.A.; Dewan, Z.N.A.; Voisin, T.; Camire, R.B.; Chow, O.A.; Chiu, I.M.; Sokol, C.L. Substance p release by sensory neurons triggers dendritic cell migration and initiates the type-2 immune response to allergens. Immunity 2020, 53, 1063–1077.e7. [Google Scholar] [CrossRef] [PubMed]
  67. Murdaca, G.; Greco, M.; Tonacci, A.; Negrini, S.; Borro, M.; Puppo, F.; Gangemi, S. Il-33/il-31 axis in immune-mediated and allergic diseases. Int. J. Mol. Sci. 2019, 20, 5856. [Google Scholar] [CrossRef] [Green Version]
  68. Ip, W.K.; Wong, C.K.; Li, M.L.; Li, P.W.; Cheung, P.F.; Lam, C.W. Interleukin-31 induces cytokine and chemokine production from human bronchial epithelial cells through activation of mitogen-activated protein kinase signalling pathways: Implications for the allergic response. Immunology 2007, 122, 532–541. [Google Scholar] [CrossRef]
  69. Kasraie, S.; Niebuhr, M.; Baumert, K.; Werfel, T. Functional effects of interleukin 31 in human primary keratinocytes. Allergy 2011, 66, 845–852. [Google Scholar] [CrossRef]
  70. Huang, J.; Yue, H.; Jiang, T.; Gao, J.; Shi, Y.; Shi, B.; Wu, X.; Gou, X. Il-31 plays dual roles in lung inflammation in an ova-induced murine asthma model. Biol. Open 2019, 8, bio036244. [Google Scholar] [CrossRef] [Green Version]
  71. Feld, M.; Garcia, R.; Buddenkotte, J.; Katayama, S.; Lewis, K.; Muirhead, G.; Hevezi, P.; Plesser, K.; Schrumpf, H.; Krjutskov, K.; et al. The pruritus- and th2-associated cytokine il-31 promotes growth of sensory nerves. J. Allergy Clin. Immunol. 2016, 138, 500–508.e4. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  72. Yu, J.I.; Han, W.C.; Yun, K.J.; Moon, H.B.; Oh, G.J.; Chae, S.C. Identifying polymorphisms in il-31 and their association with susceptibility to asthma. Korean J. Pathol. 2012, 46, 162–168. [Google Scholar] [CrossRef] [PubMed]
  73. Kabashima, K.; Irie, H. Interleukin-31 as a clinical target for pruritus treatment. Front. Med. 2021, 8, 638325. [Google Scholar] [CrossRef]
  74. Sui, P.; Wiesner, D.L.; Xu, J.; Zhang, Y.; Lee, J.; Van Dyken, S.; Lashua, A.; Yu, C.; Klein, B.S.; Locksley, R.M.; et al. Pulmonary neuroendocrine cells amplify allergic asthma responses. Science 2018, 360, eaan8546. [Google Scholar] [CrossRef] [Green Version]
  75. Undem, B.J.; Kollarik, M. The role of vagal afferent nerves in chronic obstructive pulmonary disease. Proc. Am. Thorac. Soc. 2005, 2, 355–360. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  76. Coleridge, J.C.; Coleridge, H.M. Afferent vagal c fibre innervation of the lungs and airways and its functional significance. Rev. Physiol. Biochem. Pharmacol. 1984, 99, 1–110. [Google Scholar]
  77. Kollarik, M.; Undem, B.J. Mechanisms of acid-induced activation of airway afferent nerve fibres in guinea-pig. J. Physiol. 2002, 543, 591–600. [Google Scholar] [CrossRef] [PubMed]
  78. Vanzella, L.M.; Bernardo, A.F.B.; Carvalho, T.D.; Vanderlei, F.M.; Silva, A.; Vanderlei, L.C.M. Complexity of autonomic nervous system function in individuals with copd. J. Bras. Pneumol. 2018, 44, 24–30. [Google Scholar] [CrossRef]
  79. Blake, K.J.; Jiang, X.R.; Chiu, I.M. Neuronal regulation of immunity in the skin and lungs. Trends Neurosci. 2019, 42, 537–551. [Google Scholar] [CrossRef]
  80. Burian, B.; Storka, A.; Marzluf, B.A.; Yen, Y.C.; Lambers, C.; Robibaro, B.; Vonbank, K.; Mosgoeller, W.; Petkov, V. Vasoactive intestinal peptide (vip) receptor expression in monocyte-derived macrophages from copd patients. Peptides 2010, 31, 603–608. [Google Scholar] [CrossRef]
  81. Mandal, J.; Roth, M.; Costa, L.; Boeck, L.; Rakic, J.; Scherr, A.; Tamm, M.; Stolz, D. Vasoactive intestinal peptide for diagnosing exacerbation in chronic obstructive pulmonary disease. Respiration 2015, 90, 357–368. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  82. Miotto, D.; Boschetto, P.; Bononi, I.; Zeni, E.; Cavallesco, G.; Fabbri, L.M.; Mapp, C.E. Vasoactive intestinal peptide receptors in the airways of smokers with chronic bronchitis. Eur. Respir. J. 2004, 24, 958–963. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  83. Kolahian, S.; Gosens, R. Cholinergic regulation of airway inflammation and remodelling. J. Allergy 2012, 2012, 681258. [Google Scholar] [CrossRef] [Green Version]
  84. Kistemaker, L.E.; van Os, R.P.; Dethmers-Ausema, A.; Bos, I.S.; Hylkema, M.N.; van den Berge, M.; Hiemstra, P.S.; Wess, J.; Meurs, H.; Kerstjens, H.A.; et al. Muscarinic M3 receptors on structural cells regulate cigarette smoke-induced neutrophilic airway inflammation in mice. Am. J. Physiol. Lung Cell. Mol. Physiol. 2015, 308, L96–L103. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  85. Suvas, S. Role of substance p neuropeptide in inflammation, wound healing, and tissue homeostasis. J. Immunol. 2017, 199, 1543–1552. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  86. DeRose, V.; Robbins, R.A.; Snider, R.M.; Spurzem, J.R.; Thiele, G.M.; Rennard, S.I.; Rubinstein, I. Substance p increases neutrophil adhesion to bronchial epithelial cells. J. Immunol. 1994, 152, 1339–1346. [Google Scholar]
  87. Barnes, P.J. Inflammatory mechanisms in patients with chronic obstructive pulmonary disease. J. Allergy Clin. Immunol. 2016, 138, 16–27. [Google Scholar] [CrossRef] [Green Version]
  88. Bullone, M.; Carriero, V.; Bertolini, F.; Folino, A.; Mannelli, A.; Di Stefano, A.; Gnemmi, I.; Torchio, R.; Ricciardolo, F.L.M. Elevated serum IgE, oral corticosteroid dependence and il-17/22 expression in highly neutrophilic asthma. Eur. Respir. J. 2019, 54. [Google Scholar] [CrossRef]
  89. Gu, X.; Karp, P.H.; Brody, S.L.; Pierce, R.A.; Welsh, M.J.; Holtzman, M.J.; Ben-Shahar, Y. Chemosensory functions for pulmonary neuroendocrine cells. Am. J. Respir. Cell. Mol. Biol. 2014, 50, 637–646. [Google Scholar] [CrossRef] [Green Version]
  90. Xu, H.; Zhao, M.; Wang, X. Changes of calcitonin gene-related peptide content in induced sputum from patients with copd and asthma. Zhonghua Jie He He Hu Xi Za Zhi 1999, 22, 558–561. [Google Scholar]
  91. Lagomarsino, V.N.; Kostic, A.D.; Chiu, I.M. Mechanisms of microbial-neuronal interactions in pain and nociception. Neurobiol. Pain 2021, 9, 100056. [Google Scholar] [CrossRef]
  92. Borson, D.B.; Brokaw, J.J.; Sekizawa, K.; McDonald, D.M.; Nadel, J.A. Neutral endopeptidase and neurogenic inflammation in rats with respiratory infections. J. Appl. Physiol. 1989, 66, 2653–2658. [Google Scholar] [CrossRef]
  93. Hewitt, R.; Farne, H.; Ritchie, A.; Luke, E.; Johnston, S.L.; Mallia, P. The role of viral infections in exacerbations of chronic obstructive pulmonary disease and asthma. Ther. Adv. Respir. Dis. 2016, 10, 158–174. [Google Scholar] [CrossRef] [Green Version]
  94. Linden, D.; Guo-Parke, H.; Coyle, P.V.; Fairley, D.; McAuley, D.F.; Taggart, C.C.; Kidney, J. Respiratory viral infection: A potential “missing link” in the pathogenesis of copd. Eur. Respir. Rev. 2019, 28, 180063. [Google Scholar] [CrossRef] [Green Version]
  95. Ghildyal, R.; Dagher, H.; Donninger, H.; de Silva, D.; Li, X.; Freezer, N.J.; Wilson, J.W.; Bardin, P.G. Rhinovirus infects primary human airway fibroblasts and induces a neutrophil chemokine and a permeability factor. J. Med. Virol. 2005, 75, 608–615. [Google Scholar] [CrossRef] [PubMed]
  96. Griego, S.D.; Weston, C.B.; Adams, J.L.; Tal-Singer, R.; Dillon, S.B. Role of p38 mitogen-activated protein kinase in rhinovirus-induced cytokine production by bronchial epithelial cells. J. Immunol. 2000, 165, 5211–5220. [Google Scholar] [CrossRef]
  97. Message, S.D.; Laza-Stanca, V.; Mallia, P.; Parker, H.L.; Zhu, J.; Kebadze, T.; Contoli, M.; Sanderson, G.; Kon, O.M.; Papi, A.; et al. Rhinovirus-induced lower respiratory illness is increased in asthma and related to virus load and th1/2 cytokine and il-10 production. Proc. Natl. Acad. Sci. USA 2008, 105, 13562–13567. [Google Scholar] [CrossRef] [Green Version]
  98. Yamaya, M.; Sekizawa, K.; Suzuki, T.; Yamada, N.; Furukawa, M.; Ishizuka, S.; Nakayama, K.; Terajima, M.; Numazaki, Y.; Sasaki, H. Infection of human respiratory submucosal glands with rhinovirus: Effects on cytokine and icam-1 production. Am. J. Physiol. 1999, 277, L362–L371. [Google Scholar] [CrossRef] [PubMed]
  99. Abdullah, H.; Heaney, L.G.; Cosby, S.L.; McGarvey, L.P. Rhinovirus upregulates transient receptor potential channels in a human neuronal cell line: Implications for respiratory virus-induced cough reflex sensitivity. Thorax 2014, 69, 46–54. [Google Scholar] [CrossRef] [Green Version]
  100. Grissell, T.V.; Chang, A.B.; Gibson, P.G. Reduced toll-like receptor 4 and substance p gene expression is associated with airway bacterial colonization in children. Pediatr. Pulmonol. 2007, 42, 380–385. [Google Scholar] [CrossRef] [PubMed]
  101. Doyle, W.J.; Gentile, D.A.; Cohen, S. Emotional style, nasal cytokines, and illness expression after experimental rhinovirus exposure. Brain Behav. Immun. 2006, 20, 175–181. [Google Scholar] [CrossRef]
  102. Huang, Y.; Smith, D.E.; Ibáñez-Sandoval, O.; Sims, J.E.; Friedman, W.J. Neuron-specific effects of interleukin-1β are mediated by a novel isoform of the il-1 receptor accessory protein. J. Neurosci. 2011, 31, 18048–18059. [Google Scholar] [CrossRef] [Green Version]
  103. Herrmann, O.; Tarabin, V.; Suzuki, S.; Attigah, N.; Coserea, I.; Schneider, A.; Vogel, J.; Prinz, S.; Schwab, S.; Monyer, H.; et al. Regulation of body temperature and neuroprotection by endogenous interleukin-6 in cerebral ischemia. J. Cereb. Blood Flow Metab. 2003, 23, 406–415. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  104. Penkowa, M.; Giralt, M.; Carrasco, J.; Hadberg, H.; Hidalgo, J. Impaired inflammatory response and increased oxidative stress and neurodegeneration after brain injury in interleukin-6-deficient mice. Glia 2000, 32, 271–285. [Google Scholar] [CrossRef]
  105. Hedrich, C.M.; Bruck, N.; Paul, D.; Hahn, G.; Gahr, M.; Rösen-Wolff, A. “Mutation negative” familial cold autoinflammatory syndrome (fcas) in an 8-year-old boy: Clinical course and functional studies. Rheumatol. Int. 2012, 32, 2629–2636. [Google Scholar] [CrossRef]
  106. Barragán-Iglesias, P.; Franco-Enzástiga, Ú.; Jeevakumar, V.; Shiers, S.; Wangzhou, A.; Granados-Soto, V.; Campbell, Z.T.; Dussor, G.; Price, T.J. Type I interferons act directly on nociceptors to produce pain sensitization: Implications for viral infection-induced pain. J. Neurosci. 2020, 40, 3517–3532. [Google Scholar] [CrossRef] [PubMed]
  107. Kormuth, K.A.; Lin, K.; Qian, Z.; Myerburg, M.M.; Marr, L.C.; Lakdawala, S.S. Environmental persistence of influenza viruses is dependent upon virus type and host origin. mSphere 2019, 4, e00552-19. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  108. Parrish, C.R.; Murcia, P.R.; Holmes, E.C. Influenza virus reservoirs and intermediate hosts: Dogs, horses, and new possibilities for influenza virus exposure of humans. J. Virol. 2015, 89, 2990–2994. [Google Scholar] [CrossRef] [Green Version]
  109. Grebe, K.M.; Takeda, K.; Hickman, H.D.; Bailey, A.L.; Embry, A.C.; Bennink, J.R.; Yewdell, J.W. Cutting edge: Sympathetic nervous system increases proinflammatory cytokines and exacerbates influenza a virus pathogenesis. J. Immunol. 2010, 184, 540–544. [Google Scholar] [CrossRef] [Green Version]
  110. Hosseini, S.; Wilk, E.; Michaelsen-Preusse, K.; Gerhauser, I.; Baumgärtner, W.; Geffers, R.; Schughart, K.; Korte, M. Long-term neuroinflammation induced by influenza a virus infection and the impact on hippocampal neuron morphology and function. J. Neurosci. 2018, 38, 3060–3080. [Google Scholar] [CrossRef] [Green Version]
  111. Ural, B.B.; Yeung, S.T.; Damani-Yokota, P.; Devlin, J.C.; de Vries, M.; Vera-Licona, P.; Samji, T.; Sawai, C.M.; Jang, G.; Perez, O.A.; et al. Identification of a nerve-associated, lung-resident interstitial macrophage subset with distinct localization and immunoregulatory properties. Sci. Immunol. 2020, 5, eaax8756. [Google Scholar] [CrossRef]
  112. Verzele, N.A.J.; Chua, B.Y.; Law, C.W.; Zhang, A.; Ritchie, M.E.; Wightman, O.; Edwards, I.N.; Hulme, K.D.; Bloxham, C.J.; Bielefeldt-Ohmann, H.; et al. The impact of influenza pulmonary infection and inflammation on vagal bronchopulmonary sensory neurons. FASEB J. 2021, 35, e21320. [Google Scholar] [CrossRef]
  113. Forbester, J.L.; Humphreys, I.R. Genetic influences on viral-induced cytokine responses in the lung. Mucosal Immunol. 2021, 14, 14–25. [Google Scholar] [CrossRef] [PubMed]
  114. Riazi, K.; Galic, M.A.; Kentner, A.C.; Reid, A.Y.; Sharkey, K.A.; Pittman, Q.J. Microglia-dependent alteration of glutamatergic synaptic transmission and plasticity in the hippocampus during peripheral inflammation. J. Neurosci. 2015, 35, 4942–4952. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  115. Wu, Q.; Jorde, I.; Kershaw, O.; Jeron, A.; Bruder, D.; Schreiber, J.; Stegemann-Koniszewski, S. Resolved influenza a virus infection has extended effects on lung homeostasis and attenuates allergic airway inflammation in a mouse model. Microorganisms 2020, 8, 1878. [Google Scholar] [CrossRef] [PubMed]
  116. Borchers, A.T.; Chang, C.; Gershwin, M.E.; Gershwin, L.J. Respiratory syncytial virus--a comprehensive review. Clin. Rev. Allergy Immunol. 2013, 45, 331–379. [Google Scholar] [CrossRef]
  117. Goetzl, E.J.; Xia, M.; Ingram, D.A.; Kishiyama, J.L.; Kaltreider, H.B.; Byrd, P.K.; Ichikawa, S.; Sreedharan, S.P. Neuropeptide signaling of lymphocytes in immunological responses. Int. Arch. Allergy Immunol. 1995, 107, 202–204. [Google Scholar] [CrossRef]
  118. Maggi, C.A. The effects of tachykinins on inflammatory and immune cells. Regul. Pept. 1997, 70, 75–90. [Google Scholar] [CrossRef]
  119. Piedimonte, G.; Rodriguez, M.M.; King, K.A.; McLean, S.; Jiang, X. Respiratory syncytial virus upregulates expression of the substance p receptor in rat lungs. Am. J. Physiol. 1999, 277, L831–L840. [Google Scholar] [CrossRef]
  120. Ho, W.Z.; Lai, J.P.; Zhu, X.H.; Uvaydova, M.; Douglas, S.D. Human monocytes and macrophages express substance p and neurokinin-1 receptor. J. Immunol. 1997, 159, 5654–5660. [Google Scholar]
  121. Lai, J.P.; Douglas, S.D.; Ho, W.Z. Human lymphocytes express substance p and its receptor. J. Neuroimmunol. 1998, 86, 80–86. [Google Scholar] [CrossRef]
  122. Panuska, J.R.; Merolla, R.; Rebert, N.A.; Hoffmann, S.P.; Tsivitse, P.; Cirino, N.M.; Silverman, R.H.; Rankin, J.A. Respiratory syncytial virus induces interleukin-10 by human alveolar macrophages. Suppression of early cytokine production and implications for incomplete immunity. J. Clin. Investig. 1995, 96, 2445–2453. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  123. Panuska, J.R.; Midulla, F.; Cirino, N.M.; Villani, A.; Gilbert, I.A.; McFadden, E.R., Jr.; Huang, Y.T. Virus-induced alterations in macrophage production of tumor necrosis factor and prostaglandin e2. Am. J. Physiol. 1990, 259, L396–L402. [Google Scholar] [CrossRef] [PubMed]
  124. Bachert, C.; Humbert, M.; Hanania, N.A.; Zhang, N.; Holgate, S.; Buhl, R.; Bröker, B.M. Staphylococcus aureus and its IgE-inducing enterotoxins in asthma: Current knowledge. Eur. Respir. J. 2020, 55, 1901592. [Google Scholar] [CrossRef] [PubMed]
  125. Redinbo, M.R. The microbiota, chemical symbiosis, and human disease. J. Mol. Biol. 2014, 426, 3877–3891. [Google Scholar] [CrossRef] [Green Version]
  126. Jorde, I.; Hildebrand, C.B.; Kershaw, O.; Lücke, E.; Stegemann-Koniszewski, S.; Schreiber, J. Modulation of allergic sensitization and allergic inflammation by Staphylococcus aureus enterotoxin b in an ovalbumin mouse model. Front. Immunol. 2020, 11, 592186. [Google Scholar] [CrossRef]
  127. Baral, P.; Umans, B.D.; Li, L.; Wallrapp, A.; Bist, M.; Kirschbaum, T.; Wei, Y.; Zhou, Y.; Kuchroo, V.K.; Burkett, P.R.; et al. Nociceptor sensory neurons suppress neutrophil and γδ t cell responses in bacterial lung infections and lethal pneumonia. Nat. Med. 2018, 24, 417–426. [Google Scholar] [CrossRef]
  128. N’Diaye, A.; Mijouin, L.; Hillion, M.; Diaz, S.; Konto-Ghiorghi, Y.; Percoco, G.; Chevalier, S.; Lefeuvre, L.; Harmer, N.J.; Lesouhaitier, O.; et al. Effect of substance p in Staphylococcus aureus and Staphylococcus epidermidis virulence: Implication for skin homeostasis. Front. Microbiol. 2016, 7, 506. [Google Scholar] [CrossRef]
  129. Sio, S.W.; Puthia, M.K.; Lu, J.; Moochhala, S.; Bhatia, M. The neuropeptide substance p is a critical mediator of burn-induced acute lung injury. J. Immunol. 2008, 180, 8333–8341. [Google Scholar] [CrossRef] [PubMed] [Green Version]
Figure 1. Proposed interactions between chronic respiratory diseases and respiratory pathogens along the neuro-immune axis. Schematic illustration of a proposed neuro-immunological network including cells and mediators of the neuronal and immune system that contribute to the interplay between chronic respiratory diseases and respiratory infections by direct and indirect interaction. COPD = chronic obstructive pulmonary disease, PNECs = pulmonary neuroendocrine cells, IL = interleukin, IFN = interferon, SP = substance P, VIP = vasoactive intestinal peptide, CGRP = calcitonin gene related peptide, NMU = neuromedin U, ILC2s = group 2 innate lymphoid cells, NAMs = nerve- and airway associated macrophages, TH2 = T helper type 2. Created with BioRender.com.
Figure 1. Proposed interactions between chronic respiratory diseases and respiratory pathogens along the neuro-immune axis. Schematic illustration of a proposed neuro-immunological network including cells and mediators of the neuronal and immune system that contribute to the interplay between chronic respiratory diseases and respiratory infections by direct and indirect interaction. COPD = chronic obstructive pulmonary disease, PNECs = pulmonary neuroendocrine cells, IL = interleukin, IFN = interferon, SP = substance P, VIP = vasoactive intestinal peptide, CGRP = calcitonin gene related peptide, NMU = neuromedin U, ILC2s = group 2 innate lymphoid cells, NAMs = nerve- and airway associated macrophages, TH2 = T helper type 2. Created with BioRender.com.
Ijms 22 05699 g001
Table 1. Effects in asthma, COPD, and respiratory infections for selected players in neuro-immune crosstalk.
Table 1. Effects in asthma, COPD, and respiratory infections for selected players in neuro-immune crosstalk.
AsthmaCOPDRespiratory Infections
SP
  • patients are hyperresponsive to SP; SP causes bronchoconstriction and increases mucus secretion [51].
  • SP facilitates immune cell migration [53].
  • predominantly neutrophilic inflammation in COPD [87].
  • SP expression by neutrophils; SP enhances neutrophil phagocytic activity and regulates influx [85].
  • SP increases neutrophil adhesion to bronchial epithelial cells [86].
  • SP RNA expression is reduced in RV-infected children [99,100].
  • SP receptor expression is upregulated by RSV [119].
  • SP stimulates virulence of S. aureus [128].
VIP
  • VIP directly affects immune cells; IL-5 stimulates nociceptors to release VIP [49].
  • VIP reduced cigarette smoke-induced damage of alveolar cells [81].
  • VIP receptors are increased in the central airways of smokers with chronic bronchitis [82].
PNECs
  • release of CGRP by PNECs stimulates ILC2s [74].
  • induce goblet-cell hyperplasia via GABA [74].
  • lungs from human asthmatics show increased PNECs [74].
  • over-activation of PNECs in COPD patients have been reported [89].
CGRP
  • CGRP generates neurogenic inflammation [50].
  • release of CGRP stimulates ILC2s and elicits downstream responses [74].
  • increased sputum CGRP [89].
  • increased numbers of CGRP-positive cells in the airways [90].
  • the number of CGRP immune-reactive sensory neurons is significantly increased during IAV infection [112].
  • nociceptors regulate protective immunity to S. aureus through CGRP release [127].
COPD = chronic obstructive pulmonary disease, SP = substance P, RV = rhinovirus, RSV = respiratory syncytial virus, S. aureus = Staphylococcus aureus, VIP = vasoactive intestinal peptide, IL = interleukin, PNECs = pulmonary neuroendocrine cells, CGRP = calcitonin gene related peptide, ILC2s = group 2 innate lymphoid cells, GABA = gamma-aminobutyric acid, IAV = influenza A virus.
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Camp, B.; Stegemann-Koniszewski, S.; Schreiber, J. Infection-Associated Mechanisms of Neuro-Inflammation and Neuro-Immune Crosstalk in Chronic Respiratory Diseases. Int. J. Mol. Sci. 2021, 22, 5699. https://doi.org/10.3390/ijms22115699

AMA Style

Camp B, Stegemann-Koniszewski S, Schreiber J. Infection-Associated Mechanisms of Neuro-Inflammation and Neuro-Immune Crosstalk in Chronic Respiratory Diseases. International Journal of Molecular Sciences. 2021; 22(11):5699. https://doi.org/10.3390/ijms22115699

Chicago/Turabian Style

Camp, Belinda, Sabine Stegemann-Koniszewski, and Jens Schreiber. 2021. "Infection-Associated Mechanisms of Neuro-Inflammation and Neuro-Immune Crosstalk in Chronic Respiratory Diseases" International Journal of Molecular Sciences 22, no. 11: 5699. https://doi.org/10.3390/ijms22115699

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop