Next Article in Journal
How Interest in Phages Has Bloomed into a Leading Medical Research Activity in Poland
Next Article in Special Issue
Anionic Pulmonary Surfactant Lipid Treatment Inhibits Rhinovirus A Infection of the Human Airway Epithelium
Previous Article in Journal
Novel 3′ Proximal Replication Elements in Umbravirus Genomes
Previous Article in Special Issue
Analysis of Exosomal MicroRNA Dynamics in Response to Rhinovirus Challenge in a Longitudinal Case-Control Study of Asthma
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Rhinovirus Infection and Virus-Induced Asthma

1
Department of Health Science, Gunma Paz University Graduate School of Health Sciences, Takasaki-shi 370-0006, Gunma, Japan
2
Advanced Medical Science Research Center, Gunma Paz University Research Institute, Shibukawa-shi 377-0008, Gunma, Japan
3
Department of Bacteriology, Gunma University Graduate School of Medicine, Maebashi-shi 371-8514, Gunma, Japan
4
Department of Clinical Engineering, Faculty of Medical Technology, Gunma Paz University, Takasaki-shi 370-0006, Gunma, Japan
5
Department of Medical Technology, Faculty of Health Sciences, Kyorin University, 5-4-1 Shimorenjaku, Mitaka-shi 181-8621, Tokyo, Japan
6
Department of Pediatrics, School of Medicine, Sapporo Medical University, Sapporo-shi 060-8543, Hokkaido, Japan
7
Department of Microbiology, School of Medicine, Yokohama City University, Yokohama-shi 236-0004, Kanagawa, Japan
*
Author to whom correspondence should be addressed.
Viruses 2022, 14(12), 2616; https://doi.org/10.3390/v14122616
Submission received: 18 October 2022 / Revised: 21 November 2022 / Accepted: 22 November 2022 / Published: 24 November 2022
(This article belongs to the Special Issue Rhinovirus Infections 2.0)

Abstract

:
While the aetiology of asthma is unclear, the onset and/or exacerbation of asthma may be associated with respiratory infections. Virus-induced asthma is also known as virus-associated/triggered asthma, and the reported main causative agent is rhinovirus (RV). Understanding the relationship between viral infections and asthma may overcome the gaps in deferential immunity between viral infections and allergies. Moreover, understanding the complicated cytokine networks involved in RV infection may be necessary. Therefore, the complexity of RV-induced asthma is not only owing to the response of airway and immune cells against viral infection, but also to allergic immune responses caused by the wide variety of cytokines produced by these cells. To better understand RV-induced asthma, it is necessary to elucidate the nature RV infections and the corresponding host defence mechanisms. In this review, we attempt to organise the complexity of RV-induced asthma to make it easily understandable for readers.

1. Introduction

Although the pathophysiology of asthma associated with viral infections is not fully understood, epidemiological findings have suggested that respiratory infections may be involved in the onset and/or exacerbation of asthma [1,2]. Asthma associated with viral infections is termed virus-induced asthma [3]. Several respiratory viruses can be involved, including human rhinovirus (RV), respiratory syncytial virus (RSV), and human parainfluenza virus; these viruses are causative agents in the onset and/or exacerbation of asthma. In particular, RV has been found to be associated with >60% of cases of asthma exacerbation in children [4].
The airway system consists of airway epithelial and connective tissues and smooth muscles [5]. These tissues contain blood vessels, microlymphoid tissues, and immune cells with various functions [5]. Therefore, the composition of the airway system is complex.
In all studies to date, the maintenance and homeostasis of the airway system have been proposed to be orchestrated by/associated with various immunological mediators, such as cytokines, from various cells in the airway system [6]. Virus-induced asthma has been suggested to disturb maintenance/homeostasis [7]. However, pleiotropy and the redundancy of cytokines are major obstacles in understanding virus-induced asthma [7]. In this review, to better understand the pathophysiology of RV-induced asthma, we describe the disease based on multifaceted findings related to RV virology and biological defence systems.

2. Pathophysiology of Asthma

The pathophysiology of asthma is characterised by airway inflammation and remodelling [8,9]. In asthmatics, airway inflammation is present persistently, not only during attacks [8]. Airway epithelial cells play a pivotal role in airway inflammation in asthma by acting as a barrier to the outside world and producing cytokines and chemokines [10]. Moreover, remodelling, i.e., simultaneous cell death and regeneration with smooth muscle thickening and fibrous tissue, results in hyperplasia. Thus, extensive remodelling may lead to airway stiffening and lumen narrowing, resulting in respiratory disorders [9].

3. Major Processes of Airway Tissue Remodelling in Virus-Induced Asthma

As shown in Figure 1, the major processes of tissue remodelling in virus-induced asthma are as follows: (1) epithelial inflammatory responses, including cytokine and inflammatory factor production, against viral infection; (2) the activation of leukocytes (lymphocytes, neutrophils, eosinophils, mast cells, and monocytes/macrophages) and the production of cytokines/inflammatory factors from them; and (3) the proliferation of fibroblasts and smooth muscle cells triggered by the processes in (1) and (2). Therefore, to better understand airway remodelling induced by an RV infection, understanding RV’s virology and biological defence system, which involves various immunological responders such as chemokines/cytokines and cellular immunity against RV, is necessary [11].

4. RV Virology

RV is a non-enveloped virus that belongs to the family Picornaviridae and the genus Enterovirus and causes various respiratory symptoms and diseases, including rhinitis, sore throat, cough, bronchitis, and pneumonia [12]. RVs are classified into the following species: A (RV-A), B (RV-B), and C (RV-C) [13]. Previous genetic studies have further classified RV-A, RV-B, and RV-C into over 100, 30, and 50 genotypes, respectively [14]. The major viral antigen is viral protein 1 (VP 1), and the amino acid sequences of this protein show wide divergence, which accounts for the wide antigenicity of RVs and other enteroviruses, such as echoviruses [15,16]. The cellular receptors for RV-associated VP1 are intercellular adhesion molecule type-1 (ICAM-I: CD54), cadherin-related family member 3 (CDHR3), or low-density lipoprotein receptor (LDLR) [17]. These receptors are commonly expressed in airway epithelial cells [17]. Notably, a recent study suggested that RV-C induces not only the release of Th1 cytokines but also Th2 cytokines, including interleukin (IL)-4, -5, and -6, in virus-infected airway epithelial cells [18].

5. Obstacles to Understanding Cytokines in Virus-Induced Asthma

For a better understanding of asthma pathophysiology, cytokines have been classified into two major groups: Th1 and Th2. Th1 cytokines, such as IL-1β, tumour necrosis factor (TNF)-α, and interferon (IFN)-γ, are cytokines produced by Th1 lymphocytes. Similarly, cytokines that are produced by Th2 lymphocytes are called Th2 cytokines, which include IL-4, -5, and -13.
Most viral infections induce a dominant production of Th1 cytokines, such as IL-1β and IFNs. These cytokines may induce the production of antiviral antibodies, including IgM and IgG, by activating Th1 and B cells [19,20]. IL-12 and IFN-γ may induce naïve CD4+ T cells’ differentiation into Th1 cells and activate them [21]. Moreover, activated Th1 cells secrete IL-12 and IFN-γ, which activate macrophages and enhance their capacity for digestion and the presentation of antigens [22]. In contrast, allergens, such as food proteins from eggs and meat, may sometimes induce the production of Th2 cytokines [23,24]. These cytokines can activate allergic cells, such as eosinophils and mast cells, and induce IgE (reagin) production [25,26]. IgE binds to allergens, and this allergen–reagin complex activates mast cells and induces histamine release, thereby leading to anaphylaxis [27,28].
To better understand the pathophysiology and phenotypes of asthma, the Th1/Th2 balance paradigm has been widely used [29]. In this paradigm, eosinophilic asthma develops when Th2 cytokines and Th2 lymphocytes are predominant over Th1 cytokines and Th1 lymphocytes. In contrast, it is thought that Th1 predominance causes neutrophilic asthma and autoimmune diseases such as Crohn’s disease and rheumatoid arthritis [30]. However, this paradigm cannot fully explain virus-induced asthma because viral infection leads to Th1 dominance. Recently, it has been discovered that there are cytokines and lymphocytes that do not fit into this classical paradigm.
The function of group 2 innate lymphoid cells (ILC2s) in asthma has attracted much interest. ILC2s are innate immune cells with no antigen specificity that produce large quantities of Th2 cytokines [31]. ILC2s are activated by IL-33 that is released from injured airway epithelial cells and are associated with steroid resistance in asthmatics [31,32]. Thus, ILC2s play an essential role in eosinophilic asthma. Inflammation caused by ILC2s and Th2 lymphocytes is called type 2 inflammation and has been studied intensively [31].
Th17 cells have also attracted attention in recent years and are thought to be a new player in the pathophysiology of various inflammatory diseases [33]. Th17 lymphocytes produce IL-17, which can recruit neutrophils and, like Th1 lymphocytes, is involved in the onset and exacerbation of Crohn’s disease [30]. In the context of the pathophysiology of asthma, an in vivo study suggested that IL-17 may lead to steroid-resistant asthma [34]. Interestingly, Beale et al. reported that IL-25, a family member of IL-17, is necessary for rhinovirus-exacerbated allergic pulmonary inflammation [35].
In general, most cytokines show pleiotropy and redundancy [36]. Moreover, cytokines are aggregates with these characteristics and constitute a network. Therefore, understanding the relationship between cytokines and virus-induced asthma may be difficult. Typical representative examples of such cytokines are IFNs.
IFNs are classified into three groups in humans. Type I IFNs include IFN-α and -β, type II IFNs include IFN-γ, and type III IFNs include IFN-λ [37,38]. Type I IFNs can have an anti-viral effect by suppressing viral replication and activating natural killer cells to eliminate virus-infected cells. Type I IFNs also induce the release of various cytokines [39,40]. CCL5 release is induced by type I IFNs via the Janus kinase/signal transducer and activator of transcription (JAK/STAT) pathways and the activation of interferon-stimulated genes (ISGs) [41]. CCL5 is a crucial chemokine in the migration of CD8 T cells to the lungs and is associated with the typical IFN-γ-dominant Th1 response and anti-viral immunity [42,43]. On the other hand, CCL5 simultaneously induces strong chemotaxis in eosinophils [43,44,45]. Type III IFNs also have an anti-viral effect and regulate various cytokines [46]. When the lung tissue is infected with virus, type III IFNs are released and suppress viral replication without inducing inflammation [47]. Moreover, previous reports suggested that Th2 cytokine levels, including IL-4, IL-5, and IL-33, were decreased by type III IFNs [48,49]. Thus, IFNs activate not only Th1 inflammation but also type 2 inflammation.
In this review, we have tried to avoid this complication by explicating the relationships between the cytokines shown in Table 1 [50,51,52,53,54,55,56,57,58,59,60,61,62,63,64,65,66,67,68,69,70,71,72,73,74,75,76] and their biological activities. Moreover, we simply classify cytokines into Th1, Th2, and others [77,78].

6. Relationships between RV-Infection-Induced Cytokine Responses in Airway Epithelial Cells, Fibroblasts, and Myofibroblasts

The injured airway epithelial cells produce and release various cytokines, chemokines, and damage-associated molecular patterns (DAMPs) that can activate innate immune systems. For example, polyinosinic-polycytidylic acid [poly (I:C)], which is structurally similar to double-stranded RNA and is used instead of an RNA virus infection in vitro and in vivo, can induce the release of IFN-γ, TNF-α, IL-8, and CCL5 in bronchial epithelial cells [79,80,81]. Nakamoto et al. reported that flagellin, derived from Pseudomonas aeruginosa, induces the release of IL-6 and IL-8 in airway epithelial cells [82]. Thus, airway epithelial cells are not only barriers to environmental stimulations but also act as triggers for inflammation and the release of various signalling molecules. Moreover, the cytokines, chemokines, and DAMPs released from airway epithelial cells may induce the release of other cytokines or chemokines from fibroblasts, myofibroblasts, and leukocytes. For example, transforming growth factor β (TGF-β) derived from epithelial cells is important in the development of airway remodelling in refractory asthma and it induces IL-6 production in fibroblasts [83,84]. In this manner, the injury of airway epithelial cells by environmental factors can lead to cascades of cytokine and chemokine production.
Viral infection is one of the major causes of injury to airway epithelial cells. Airway epithelial cells perceive viral infection and virus-induced injury and activate immune responses. One of these responses is the activation of the innate immune system through pattern-recognition receptors (PRRs) [85]. Toll-like receptors (TLRs) are one of the most representative PRRs. TLR3 recognises viral double-stranded RNA, and previous reports have shown that cytokines and chemokines are produced in bronchial epithelial cells via TLR3 activation during viral infection [86,87]. TLR7/8 recognises single-stranded RNA. Several reports have suggested that various cytokines are produced in bronchial epithelial cells via TLR7/8 activation at the time of viral infection [88,89]. Other RNA-sensing PRRs include retinoic acid-inducible gene-I (RIG-I), MDA5 (melanoma differentiation-associated gene 5), and protein kinase R (PKR) [90]. The recognition of viral RNA by these molecules can also induce the release of various cytokines. A response other than cytokine production that can occur as a result of viral infection is the production of reactive oxygen species (ROS) in airway epithelial cells. ROS can induce apoptosis in airway cells [91]. Comstock et al. reported that viral infection and poly (I:C) stimulation promote ROS production in bronchial epithelial cells [92]. Thus, viral infection leads to various immune responses and promotes cascades of cytokine and chemokine production.
Among the viruses that can damage airway epithelial tissue, RV is a major pathogen, and is one that exacerbates asthma, as described above. As shown in Figure 1, RV can infect airway epithelial cells, leading to a cascade of cytokine and chemokine production [93,94,95]. Indeed, RV-infected epithelial cells can produce various T1 cytokines, including granulocyte colony-stimulating factor, IFN-γ, and TNF-α, and type2 cytokines including IL-4, IL-5, and IL-13 [96,97,98]. These cytokines may further induce the production of various cytokines in fibroblasts and myofibroblasts, irrespective of RV infection [99,100]. Moreover, fibroblasts and myofibroblasts infected with RV may also induce the release of cytokines, although this has been confirmed only in an in vitro study [94,98].
As mentioned above, RV infection may induce the production of various cytokines in airway component cells including epithelial cells, fibroblasts, and myofibroblasts [101]. These cytokines also activate leukocytes. Among them, MCP-1 (CCL2), MIP-1α (CCL3), and IP-10 (CXCL10) may activate lymphocytes; MCP-1 (CCL2), MIP-1α (CCL3), and RANTES (CCL5) may activate monocytes/macrophages; GROα (CXCL1), GROγ (CXCL3), ENA-78 (CXCL5), and IL-8 (CXCL8) may activate neutrophils; MIP-1α (CCL3), RANTES (CCL5), eotaxin (CCL11), and eotaxin-2 (CCL24) may activate eosinophils; RANTES (CCL5) and eotaxin (CCL11) may activate mast cells; and MIP-3α (CCL20) activates dendritic cells (DCs) [69,102,103]. Moreover, various chemokines act as chemoattractant proteins that induce the migration and recruitment of neutrophils, eosinophils, and monocytes/macrophages from adjacent vascular tissue to the RV infection foci [104,105].

7. Effector Functions of Leukocytes

Leukocytes are also associated with the pathophysiology of asthma. The effector functions of various leukocytes, such as neutrophils, monocytes/macrophages, eosinophils, and mast cells, are involved in degranulation, the generation of ROS, and phagocytosis [106]. These effector functions are triggered and enhanced by Th1 and/or Th2 cytokines and/or stimulation by various substances, such as platelet-activating factor derived from RV-infected cells [96]. Leukocytes interact with each other through such effector functions and the release of cytokines. Thus, leukocytes form a network among themselves.
DCs are activated by cytokines released from airway epithelial cells, take up allergens, and migrate to local lymph nodes. DCs present antigens via major histocompatibility gene complex class II molecules, and CD40-mediated stimulation can induce CD4 T cells to differentiate into Th1, Th2, or Th17 cells [107].
Th1 lymphocytes produce Th1 cytokines such as IFN-γ and TNF-α and activate monocytes/macrophages and lymphocytes (cytotoxic T cells) in order to kill virus-infected cells [88,89]. Alveolar macrophages play specific roles in the phagocytosis of various particles such as dust, bacteria, and viruses and have strong effector functions including ROS generation and the degranulation of myeloperoxidase [108]. Therefore, in the acute phase of airway infection by RVs, these effector functions of macrophages may be partly responsible for exacerbating virus-induced asthma [109,110]. In addition, Th1 cytokines activate neutrophils. In an in vivo study, activated neutrophils increased the expression of the high-affinity IgE receptor (FcεRI) on lung-conventional DCs [111]. Therefore, Th1 lymphocytes may contribute to accelerating type 2 inflammation.
Moreover, Th2 lymphocytes produce IL-4, IL-5, IL-13, and IL-33 and activate eosinophils and mast cells, leading to allergic reactions, including virus-induced asthma [112,113]. Mast cells release histamine and matrix metalloproteinases (MMPs), and they cause increased vascular permeability and bronchial smooth muscle contractions. Eosinophils contain toxic granules (i.e., major basic protein (MBP), eosinophil cationic protein (ECP), eosinophil-derived neurotoxin (EDN), and eosinophil peroxidase) and efficiently produce ROS upon various stimulations, such as that effected by cytokines, and enhanced effector functions by eosinophils can cause tissue injury and airway remodelling in RV-induced asthma [114,115].
Since the leukocyte network is complicated, the relationship between individual leukocytes is not fully understood. Therefore, it may be necessary to investigate the mechanism by which Th1 lymphocytes accelerate type 2 inflammation, which is unconsidered by the classical Th1/Th2 balance paradigm.

8. Differences between Asthmatics and Non-Asthmatics in Viral Infection

The production of cytokines and chemokines in the airway compartment and by various immune cells may occur in both asthmatics and non-asthmatics. It remains unclear what causes asthmatic airway inflammation. However, several theories have been considered in this respect.
First, a type 2 dominant environment in the airway accelerates type 2 inflammation in airway components. In airway epithelial cells, poly (I:C) stimulation induces greater CCL5 production with IL-4 and IL-13 pre-treatments than without [116]. In addition, sputum IL-13 levels in patients with poorly controlled asthma are higher than those in patients with well-controlled asthma. Thus, a Th2-predominant environment creates type 2 inflammation and may form a spiral that leads to asthma exacerbation in asthmatic patients.
Second, there are low levels of IFN-β in the bronchial epithelial cells of asthmatics [116]. Since IFNs play an essential role in viral elimination, asthmatics with low levels of IFN-β fail to eliminate viruses and develop worse asthma exacerbation. From this perspective, a clinical trial using nebulised IFN-β therapy was conducted and reported some efficacy [117].
Third, there are persistent viral infections in asthmatics. As described above, bronchial epithelial cells and airway macrophages in asthmatics have low levels of IFNs and fail to mount an effective innate immune response in the airway. Thus, viral clearance may be incomplete in the airways of asthmatics. Indeed, Wos et al. reported that RV was detected in the lower airway tissue of asthmatics more often than in non-asthmatics [118].
These points are controversial. The first point is very useful for understanding the pathophysiology of eosinophilic asthma. On the other hand, a multifaceted approach is important to understand the pathophysiology of asthma because it has variously different phenotypes. Thus, the differences between asthma and non-asthma phenotypes need to be more closely scrutinised.

9. Perspective

Although inhaled corticosteroid therapy improves the progression of asthma, it is still a major disease burden globally [119]. An RV infection is a common cause of asthma exacerbation. Thus, understanding the pathophysiology of virus-induced asthma, especially RV-related asthma, is essential for developing new treatment agents. As described in this review, the relationships between viral infection and the production of cytokines in the airway compartment have been clarified to some extent, such as those concerning IFN production and the cytokines induced by IFNs. However, the differences between a normal airway and an asthmatic airway are still unclear. Therefore, further studies may be needed to elucidate the differences between asthmatics and non-asthmatics with respect to viral infection.

Author Contributions

Conceptualisation, H.K.; methodology, Y.H., T.S., M.S., R.K. and H.K.; review and editing, K.O., M.K., T.S., T.T., M.O. and A.R.; visualisation, Y.H. and M.K.; writing and original draft preparation, Y.H., T.S., M.S. and H.K. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Data Availability Statement

Not applicable.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Kusel, M.M.H.; de Klerk, N.H.; Kebadze, T.; Vohma, V.; Holt, P.G.; Johnston, S.L.; Sly, P.D. Early-life respiratory viral infections, atopic sensitization, and risk of subsequent development of persistent asthma. J. Allergy Clin. Immunol. 2007, 119, 1105–1110. [Google Scholar] [CrossRef]
  2. Johnston, N.W.; Sears, M.R. Asthma exacerbations 1: Epidemiology. Thorax 2006, 61, 722–728. [Google Scholar] [CrossRef] [Green Version]
  3. Kurai, D.; Saraya, T.; Ishii, H.; Takizawa, H. Virus-induced exacerbations in asthma and COPD. Front. Microbiol. 2013, 4, 293. [Google Scholar] [CrossRef] [Green Version]
  4. Khetsuriani, N.; Lu, X.; Teague, W.G.; Kazerouni, N.; Anderson, L.J.; Erdman, D.D. Novel human rhinoviruses and exacerbation of asthma in children. Emerg. Infect. Dis. 2008, 14, 1793. [Google Scholar] [CrossRef]
  5. Humberto, E.T.B.; Samuel, A. Histology for Pathologists; Lippincott Williams & Wilkins: Philadelphia, PA, USA, 2019. [Google Scholar]
  6. Toews, G.B. Cytokines and the lung. Eur. Respir. J. 2001, 34, 3s–17s. [Google Scholar] [CrossRef] [Green Version]
  7. Gern, J.E. Mechanisms of virus-induced asthma. J. Pediatr. 2003, 142, S9–S13. [Google Scholar] [CrossRef] [PubMed]
  8. Bousquet, J.; Jeffery, P.K.; Busse, W.W.; Johnson, M.; Vignola, A.M. Asthma: From bronchoconstriction to airways inflammation and remodeling. Am. J. Respir. Crit. Care Med. 2000, 161, 1720–1745. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  9. Al-Muhsen, S.; Johnson, J.R.; Hamid, Q. Remodeling in asthma. J. Allergy Clin. Immunol. 2011, 128, 451–462. [Google Scholar] [CrossRef] [PubMed]
  10. Czarnowicki, T.; Esaki, H.; Gonzalez, J.; Malajian, D.; Shemer, A.; Noda, S.; Talasila, S.; Berry, A.; Gray, J.; Becker, L. Early pediatric atopic dermatitis shows only a cutaneous lymphocyte antigen (CLA)+ TH2/TH1 cell imbalance, whereas adults acquire CLA+ TH22/TC22 cell subsets. J. Allergy Clin. Immunol. 2015, 136, 941–951. [Google Scholar] [CrossRef] [Green Version]
  11. Kimura, H.; Yoshizumi, M.; Ishii, H.; Oishi, K.; Ryo, A. Cytokine production and signaling pathways in respiratory virus infection. Front. Microbiol. 2013, 4, 276. [Google Scholar] [CrossRef]
  12. James, E.; Gern, A.C. Fields Virology, 6th Edition. Clin. Infect. Dis. 2013, 59, 531–549. [Google Scholar] [CrossRef]
  13. Jacobs, S.E.; Lamson, D.M.; St George, K.; Walsh, T.J. Human rhinoviruses. Clin. Microbiol. Rev. 2013, 26, 135–162. [Google Scholar] [CrossRef] [Green Version]
  14. McIntyre, C.L.; Knowles, N.J.; Simmonds, P. Proposals for the classification of human rhinovirus species A, B and C into genotypically assigned types. J. Gen. Virol. 2013, 94, 1791. [Google Scholar] [CrossRef] [PubMed]
  15. Alshrari, A.S.; Hudu, S.A.; Asdaq, S.M.; Ali, A.M.; Kin, C.V.; Omar, A.R.; Pei, C.P.; Sekawi, Z. Bioinformatics analysis of rhinovirus capsid proteins VP1–4 sequences for cross-serotype vaccine development. J. Infect. Public Health 2021, 14, 1603–1611. [Google Scholar] [CrossRef] [PubMed]
  16. McWilliam Leitch, E.C.; Cabrerizo, M.; Cardosa, J.; Harvala, H.; Ivanova, O.E.; Kroes, A.C.; Lukashev, A.; Muir, P.; Odoom, J.; Roivainen, M.; et al. Evolutionary dynamics and temporal/geographical correlates of recombination in the human enterovirus echovirus types 9, 11, and 30. J. Virol. 2010, 84, 9292–9300. [Google Scholar] [CrossRef] [Green Version]
  17. Basnet, S.; Palmenberg, A.C.; Gern, J.E. Rhinoviruses and their receptors. Chest 2019, 155, 1018–1025. [Google Scholar] [CrossRef] [PubMed]
  18. Yuan, X.-H.; Pang, L.-L.; Yang, J.; Jin, Y. Comparison of immune response to human rhinovirus C and respiratory syncytial virus in highly differentiated human airway epithelial cells. Virol. J. 2022, 19, 81. [Google Scholar] [CrossRef]
  19. Romagnani, S. Human TH1 and TH2 subsets: Doubt no more. Immunol. Today 1991, 12, 256–257. [Google Scholar] [CrossRef] [PubMed]
  20. Zuniga, E.I.; Macal, M.; Lewis, G.M.; Harker, J.A. Innate and Adaptive Immune Regulation During Chronic Viral Infections. Annu. Rev. Virol. 2015, 2, 573–597. [Google Scholar] [CrossRef] [Green Version]
  21. Zhang, Y.; Zhang, Y.; Gu, W.; Sun, B. TH1/TH2 cell differentiation and molecular signals. Adv. Exp. Med. Biol. 2014, 841, 15–44. [Google Scholar] [PubMed]
  22. Trinchieri, G. Interleukin-12 and the regulation of innate resistance and adaptive immunity. Nat. Rev. Immunol. 2003, 3, 133–146. [Google Scholar] [CrossRef] [PubMed]
  23. Colombo, P.E.; Quenet, F.; Alric, P.; Mourregot, A.; Neron, M.; Portales, F.; Rouanet, P.; Carrier, G. Distal Pancreatectomy with Celiac Axis Resection (Modified Appleby Procedure) and Arterial Reconstruction for Locally Advanced Pancreatic Adenocarcinoma after FOLFIRINOX Chemotherapy and Chemoradiation Therapy. Ann. Surg. Oncol. 2021, 28, 1106–1108. [Google Scholar] [CrossRef] [PubMed]
  24. Leung, D.Y.; Boguniewicz, M.; Howell, M.D.; Nomura, I.; Hamid, Q.A. New insights into atopic dermatitis. J. Clin. Investig. 2004, 113, 651–657. [Google Scholar] [CrossRef] [PubMed]
  25. Corry, D.B.; Kheradmand, F. Induction and regulation of the IgE response. Nature 1999, 402, B18–B23. [Google Scholar] [CrossRef] [PubMed]
  26. Deo, S.S.; Mistry, K.J.; Kakade, A.M.; Niphadkar, P.V. Role played by Th2 type cytokines in IgE mediated allergy and asthma. Lung India 2010, 27, 66–71. [Google Scholar] [CrossRef]
  27. Hanson, L.A. Atopic allergies and contact allergies. In Immunology; Wigzell, L.A., Ed.; Butterworth-Heinemann: Oxford, UK, 1985; pp. 198–209. [Google Scholar] [CrossRef]
  28. And, K.I.; Ishizaka, T. Mechanisms of reaginic hypersensitivity and IgE antibody response. Immunol. Rev. 1978, 41, 109–148. [Google Scholar] [CrossRef]
  29. Hirahara, K.; Nakayama, T. CD4+ T-cell subsets in inflammatory diseases: Beyond the Th 1/Th 2 paradigm. Int. Immunol. 2016, 28, 163–171. [Google Scholar] [CrossRef] [Green Version]
  30. Brand, S. Crohn’s disease: Th1, Th17 or both? The change of a paradigm: New immunological and genetic insights implicate Th17 cells in the pathogenesis of Crohn’s disease. Gut 2009, 58, 1152–1167. [Google Scholar] [CrossRef] [Green Version]
  31. Caminati, M.; Pham, D.L.; Bagnasco, D.; Canonica, G.W. Type 2 immunity in asthma. World Allergy Organ. J. 2018, 11, 13. [Google Scholar] [CrossRef]
  32. Han, M.; Rajput, C.; Hong, J.Y.; Lei, J.; Hinde, J.L.; Wu, Q.; Bentley, J.K.; Hershenson, M.B. The innate cytokines IL-25, IL-33, and TSLP cooperate in the induction of type 2 innate lymphoid cell expansion and mucous metaplasia in rhinovirus-infected immature mice. J. Immunol. 2017, 199, 1308–1318. [Google Scholar] [CrossRef] [Green Version]
  33. Maddur, M.S.; Miossec, P.; Kaveri, S.V.; Bayry, J. Th17 Cells: Biology, Pathogenesis of Autoimmune and Inflammatory Diseases, and Therapeutic Strategies. Am. J. Pathol. 2012, 181, 8–18. [Google Scholar] [CrossRef]
  34. McKinley, L.; Alcorn, J.F.; Peterson, A.; DuPont, R.B.; Kapadia, S.; Logar, A.; Henry, A.; Irvin, C.G.; Piganelli, J.D.; Ray, A. TH17 cells mediate steroid-resistant airway inflammation and airway hyperresponsiveness in mice. J. Immunol. 2008, 181, 4089–4097. [Google Scholar] [CrossRef] [Green Version]
  35. Makris, S.; Johnston, S. Recent advances in understanding rhinovirus immunity. F1000 Res. 2018, 7, 1537. [Google Scholar] [CrossRef]
  36. Simbirtsev, A.S.; Kozlov, I.G. Cytokine System. In Mechanical Stretch and Cytokines; Kamkin, A., Kiseleva, I., Eds.; Springer: Dordrecht, The Netherlands, 2012; pp. 1–33. [Google Scholar]
  37. Induction and Regulation of IFNs During Viral Infections. J. Interferon Cytokine Res. 2004, 24, 439–454. [CrossRef]
  38. Ank, N.; West, H.; Bartholdy, C.; Eriksson, K.; Thomsen, A.R.; Paludan, S.R. Lambda Interferon (IFN-λ), a Type III IFN, Is Induced by Viruses and IFNs and Displays Potent Antiviral Activity against Select Virus Infections In Vivo. J. Virol. 2006, 80, 4501–4509. [Google Scholar] [CrossRef] [Green Version]
  39. Perry, A.K.; Chen, G.; Zheng, D.; Tang, H.; Cheng, G. The host type I interferon response to viral and bacterial infections. Cell Res. 2005, 15, 407–422. [Google Scholar] [CrossRef] [Green Version]
  40. McNab, F.; Mayer-Barber, K.; Sher, A.; Wack, A.; O’garra, A. Type I interferons in infectious disease. Nat. Rev. Immunol. 2015, 15, 87–103. [Google Scholar] [CrossRef]
  41. Sada, M.; Watanabe, M.; Inui, T.; Nakamoto, K.; Hirata, A.; Nakamura, M.; Honda, K.; Saraya, T.; Kurai, D.; Kimura, H. Ruxolitinib inhibits poly (I:C) and type 2 cytokines-induced CCL5 production in bronchial epithelial cells: A potential therapeutic agent for severe eosinophilic asthma. Immun. Inflamm. Dis. 2021, 9, 363–373. [Google Scholar] [CrossRef]
  42. Galkina, E.; Thatte, J.; Dabak, V.; Williams, M.B.; Ley, K.; Braciale, T.J. Preferential migration of effector CD8+ T cells into the interstitium of the normal lung. J. Clin. Investig. 2005, 115, 3473–3483. [Google Scholar] [CrossRef]
  43. Kohlmeier, J.E.; Miller, S.C.; Smith, J.; Lu, B.; Gerard, C.; Cookenham, T.; Roberts, A.D.; Woodland, D.L. The chemokine receptor CCR5 plays a key role in the early memory CD8+ T cell response to respiratory virus infections. Immunity 2008, 29, 101–113. [Google Scholar] [CrossRef] [Green Version]
  44. Tekkanat, K.K.; Maassab, H.; Miller, A.; Berlin, A.A.; Kunkel, S.L.; Lukacs, N.W. RANTES (CCL5) production during primary respiratory syncytial virus infection exacerbates airway disease. Eur. J. Immunol. 2002, 32, 3276–3284. [Google Scholar] [CrossRef] [PubMed]
  45. Culley, F.J.; Pennycook, A.M.; Tregoning, J.S.; Dodd, J.S.; Walzl, G.; Wells, T.N.; Hussell, T.; Openshaw, P.J. Role of CCL5 (RANTES) in viral lung disease. J. Virol. 2006, 80, 8151–8157. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  46. Krammer, S.; Gutu, C.S.; Grund, J.C.; Chiriac, M.T.; Zirlik, S.; Finotto, S. Regulation and Function of Interferon-Lambda (IFNλ) and Its Receptor in Asthma. Front. Immunol. 2021, 12, 731807. [Google Scholar] [CrossRef] [PubMed]
  47. Galani, I.E.; Triantafyllia, V.; Eleminiadou, E.E.; Koltsida, O.; Stavropoulos, A.; Manioudaki, M.; Thanos, D.; Doyle, S.E.; Kotenko, S.V.; Thanopoulou, K.; et al. Interferon-λ Mediates Non-redundant Front-Line Antiviral Protection against Influenza Virus Infection without Compromising Host Fitness. Immunity 2017, 46, 875–890. [Google Scholar] [CrossRef] [PubMed]
  48. Koch, S.; Finotto, S. Role of Interferon-λ in Allergic Asthma. J. Innate Immun. 2015, 7, 224–230. [Google Scholar] [CrossRef]
  49. Won, J.; Gil, C.H.; Jo, A.; Kim, H.J. Inhaled delivery of Interferon-lambda restricts epithelial-derived Th2 inflammation in allergic asthma. Cytokine 2019, 119, 32–36. [Google Scholar] [CrossRef]
  50. Newcomb, D.C.; Sajjan, U.S.; Nagarkar, D.R.; Goldsmith, A.M.; Bentley, J.K.; Hershenson, M.B. Cooperative effects of rhinovirus and TNF-α on airway epithelial cell chemokine expression. Am. J. Physiol.-Lung Cell. Mol. Physiol. 2007, 293, L1021–L1028. [Google Scholar] [CrossRef]
  51. Liao, W.; Lin, J.-X.; Leonard, W.J. IL-2 family cytokines: New insights into the complex roles of IL-2 as a broad regulator of T helper cell differentiation. Curr. Opin. Immunol. 2011, 23, 598–604. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  52. Hamilton, J.A. GM-CSF in inflammation. J. Exp. Med. 2020, 217, e20190945. [Google Scholar] [CrossRef]
  53. Smiley, S.T.; Grusby, M.J. Interleukin 4. In Encyclopedia of Immunology, 2nd ed.; Delves, P.J., Ed.; Elsevier: Oxford, UK, 1998; pp. 1451–1453. [Google Scholar]
  54. Fulkerson, P.C.; Rothenberg, M.E. Chapter One—Eosinophil Development, Disease Involvement, and Therapeutic Suppression. In Advances in Immunology; Alt, F., Ed.; Academic Press: Cambridge, MA, USA, 2018; Volume 138, pp. 1–34. [Google Scholar]
  55. Griffin, D.E. Cytokines and Chemokines. In Encyclopedia of VirologyI, 3rd ed.; Mahy, B.W.J., Van Regenmortel, M.H.V., Eds.; Academic Press: Oxford, UK, 2008; pp. 620–624. [Google Scholar]
  56. Matsuda, T.; Kishimoto, T. Interleukin 6. In Encyclopedia of Immunology, 2nd ed.; Delves, P.J., Ed.; Elsevier: Oxford, UK, 1998; pp. 1458–1461. [Google Scholar]
  57. Sahiner, U.; Akdis, M.; Akdis, C.A. 1—Introduction to Mechanisms of Allergic Diseases. In Allergy Essentials, 2nd ed.; O’Hehir, R.E., Holgate, S.T., Hershey, G.K.K., Sheikh, A., Eds.; Elsevier: Philadelphia, PA, USA, 2022; pp. 1–24. [Google Scholar]
  58. Mak, T.W.; Saunders, M.E. 17—Cytokines and Cytokine Receptors. In The Immune Response; Mak, T.W., Saunders, M.E., Eds.; Academic Press: Burlington, VT, USA, 2006; pp. 463–516. [Google Scholar]
  59. Ohne, Y.; Silver, J.S.; Thompson-Snipes, L.; Collet, M.A.; Blanck, J.P.; Cantarel, B.L.; Copenhaver, A.M.; Humbles, A.A.; Liu, Y.-J. IL-1 is a critical regulator of group 2 innate lymphoid cell function and plasticity. Nat. Immunol. 2016, 17, 646–655. [Google Scholar] [CrossRef]
  60. Jackson, D.J.; Makrinioti, H.; Rana, B.M.; Shamji, B.W.; Trujillo-Torralbo, M.-B.; Footitt, J.; Del-Rosario, J.; Telcian, A.G.; Nikonova, A.; Zhu, J. IL-33–dependent type 2 inflammation during rhinovirus-induced asthma exacerbations in vivo. Am. J. Respir. Crit. Care Med. 2014, 190, 1373–1382. [Google Scholar] [CrossRef] [Green Version]
  61. Burleson, S.C.M.; Fick, R.B.; Mannie, M.D.; Olmstead, S.G.; Van Scott, M.R. Chapter 35—The Immune Basis of Allergic Lung Disease. In Comparative Biology of the Normal Lung, 2nd ed.; Parent, R.A., Ed.; Academic Press: San Diego, CA, USA, 2015; pp. 683–719. [Google Scholar]
  62. Fujiwara, K.; Matsukawa, A.; Ohkawara, S.; Takagi, K.; Yoshinaga, M. Functional distinction between CXC chemokines, interleukin-8 (IL-8), and growth related oncogene (GRO)alpha in neutrophil infiltration. Lab. Investig. 2002, 82, 15–23. [Google Scholar] [CrossRef] [Green Version]
  63. Al-Alwan, L.A.; Chang, Y.; Mogas, A.; Halayko, A.J.; Baglole, C.J.; Martin, J.G.; Rousseau, S.; Eidelman, D.H.; Hamid, Q. Differential roles of CXCL2 and CXCL3 and their receptors in regulating normal and asthmatic airway smooth muscle cell migration. J. Immunol. 2013, 191, 2731–2741. [Google Scholar] [CrossRef] [Green Version]
  64. Walz, A.; Schmutz, P.; Mueller, C.; Schnyder-Candrian, S. Regulation and function of the CXC chemokine ENA-78 in monocytes and its role in disease. J. Leukoc. Biol. 1997, 62, 604–611. [Google Scholar] [CrossRef]
  65. Mukaida, N. Pathophysiological roles of interleukin-8/CXCL8 in pulmonary diseases. Am. J. Physiol.-Lung Cell. Mol. Physiol. 2003, 284, L566–L577. [Google Scholar] [CrossRef] [Green Version]
  66. Liu, M.; Guo, S.; Hibbert, J.M.; Jain, V.; Singh, N.; Wilson, N.O.; Stiles, J.K. CXCL10/IP-10 in infectious diseases pathogenesis and potential therapeutic implications. Cytokine Growth Factor Rev. 2011, 22, 121–130. [Google Scholar] [CrossRef] [PubMed]
  67. Singh, S.; Anshita, D.; Ravichandiran, V. MCP-1: Function, regulation, and involvement in disease. Int. Immunopharmacol. 2021, 101, 107598. [Google Scholar] [CrossRef]
  68. Maurer, M.; von Stebut, E. Macrophage inflammatory protein-1. Int. J. Biochem. Cell Biol. 2004, 36, 1882–1886. [Google Scholar] [CrossRef] [PubMed]
  69. Rot, A.; Krieger, M.; Brunner, T.; Bischoff, S.C.; Schall, T.; Dahinden, C. RANTES and macrophage inflammatory protein 1 alpha induce the migration and activation of normal human eosinophil granulocytes. J. Exp. Med. 1992, 176, 1489–1495. [Google Scholar] [CrossRef]
  70. Levy, J.A. The unexpected pleiotropic activities of RANTES. J. Immunol. 2009, 182, 3945–3946. [Google Scholar] [CrossRef] [Green Version]
  71. Garcia-Zepeda, E.A.; Rothenberg, M.E.; Ownbey, R.T.; Celestin, J.; Leder, P.; Luster, A.D. Human eotaxin is a specific chemoattractant for eosinophil cells and provides a new mechanism to explain tissue eosinophilia. Nat. Med. 1996, 2, 449–456. [Google Scholar] [CrossRef]
  72. Le Borgne, M.; Etchart, N.; Goubier, A.; Lira, S.A.; Sirard, J.C.; van Rooijen, N.; Caux, C.; Aït-Yahia, S.; Vicari, A.; Kaiserlian, D.; et al. Dendritic cells rapidly recruited into epithelial tissues via CCR6/CCL20 are responsible for CD8+ T cell crosspriming in vivo. Immunity 2006, 24, 191–201. [Google Scholar] [CrossRef]
  73. Roan, F.; Bell, B.D.; Stoklasek, T.A.; Kitajima, M.; Han, H.; Ziegler, S.F. The multiple facets of thymic stromal lymphopoietin (TSLP) during allergic inflammation and beyond. J. Leukoc. Biol. 2012, 91, 877–886. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  74. Zaidi, M.R.; Merlino, G. The Two Faces of Interferon-γ in Cancer. Clin. Cancer Res. 2011, 17, 6118–6124. [Google Scholar] [CrossRef] [Green Version]
  75. Atzeni, F.; Sarzi-Puttini, P. Tumor Necrosis Factor. In Brenner’s Encyclopedia of Genetics, 2nd ed.; Maloy, S., Hughes, K., Eds.; Academic Press: San Diego, CA, USA, 2013; pp. 229–231. [Google Scholar]
  76. Tumor necrosis factor alfa. In Meyler’s Side Effects of Drugs: The International Encyclopedia of Adverse Drug Reactions and Interactions, 15th ed.; Aronson, J.K. (Ed.) Elsevier: Amsterdam, The Netherlands, 2006; pp. 3537–3538. [Google Scholar]
  77. Mosmann, T.R.; Cherwinski, H.; Bond, M.W.; Giedlin, M.A.; Coffman, R.L. Two types of murine helper T cell clone. I. Definition according to profiles of lymphokine activities and secreted proteins. J. Immunol. 1986, 136, 2348–2357. [Google Scholar] [PubMed]
  78. Butcher, M.J.; Zhu, J. Recent advances in understanding the Th1/Th2 effector choice. Fac. Rev. 2021, 10, 30. [Google Scholar] [CrossRef] [PubMed]
  79. Lee, S.M.-Y.; Yip, T.-F.; Yan, S.; Jin, D.-Y.; Wei, H.-L.; Guo, R.-T.; Peiris, J.S.M. Recognition of double-stranded RNA and regulation of interferon pathway by toll-like receptor 10. Front. Immunol. 2018, 9, 516. [Google Scholar] [CrossRef] [Green Version]
  80. Matsumoto, M.; Seya, T. TLR3: Interferon induction by double-stranded RNA including poly(I:C). Adv. Drug Deliv. Rev. 2008, 60, 805–812. [Google Scholar] [CrossRef] [PubMed]
  81. Kumar, A.; Zhang, J.; Yu, F.S. Toll-like receptor 3 agonist poly(I:C)-induced antiviral response in human corneal epithelial cells. Immunology 2006, 117, 11–21. [Google Scholar] [CrossRef] [PubMed]
  82. Nakamoto, K.; Watanabe, M.; Sada, M.; Inui, T.; Nakamura, M.; Honda, K.; Wada, H.; Ishii, H.; Takizawa, H. Pseudomonas aeruginosa-derived flagellin stimulates IL-6 and IL-8 production in human bronchial epithelial cells: A potential mechanism for progression and exacerbation of COPD. Exp. Lung Res. 2019, 45, 255–266. [Google Scholar] [CrossRef]
  83. Michalik, M.; Wójcik-Pszczoła, K.; Paw, M.; Wnuk, D.; Koczurkiewicz, P.; Sanak, M.; Pękala, E.; Madeja, Z. Fibroblast-to-myofibroblast transition in bronchial asthma. Cell. Mol. Life Sci. 2018, 75, 3943–3961. [Google Scholar] [CrossRef] [Green Version]
  84. Gomes, I.; Mathur, S.K.; Espenshade, B.M.; Mori, Y.; Varga, J.; Ackerman, S.J. Eosinophil-fibroblast interactions induce fibroblast IL-6 secretion and extracellular matrix gene expression: Implications in fibrogenesis. J. Allergy Clin. Immunol. 2005, 116, 796–804. [Google Scholar] [CrossRef] [PubMed]
  85. Wang, Q.; Nagarkar, D.R.; Bowman, E.R.; Schneider, D.; Gosangi, B.; Lei, J.; Zhao, Y.; McHenry, C.L.; Burgens, R.V.; Miller, D.J. Role of double-stranded RNA pattern recognition receptors in rhinovirus-induced airway epithelial cell responses. J. Immunol. 2009, 183, 6989–6997. [Google Scholar] [CrossRef] [Green Version]
  86. Kato, A.; Favoreto, S.; Avila, P.C.; Schleimer, R.P. TLR3-and Th2 cytokine-dependent production of thymic stromal lymphopoietin in human airway epithelial cells. J. Immunol. 2007, 179, 1080–1087. [Google Scholar] [CrossRef]
  87. Hewson, C.A.; Jardine, A.; Edwards, M.R.; Laza-Stanca, V.; Johnston, S.L. Toll-like receptor 3 is induced by and mediates antiviral activity against rhinovirus infection of human bronchial epithelial cells. J. Virol. 2005, 79, 12273–12279. [Google Scholar] [CrossRef] [Green Version]
  88. Xagorari, A.; Chlichlia, K. Toll-like receptors and viruses: Induction of innate antiviral immune responses. Open Microbiol. J. 2008, 2, 49. [Google Scholar] [CrossRef] [PubMed]
  89. Caron, G.; Duluc, D.; Frémaux, I.; Jeannin, P.; David, C.; Gascan, H.; Delneste, Y. Direct stimulation of human T cells via TLR5 and TLR7/8: Flagellin and R-848 up-regulate proliferation and IFN-γ production by memory CD4+ T cells. J. Immunol. 2005, 175, 1551–1557. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  90. Matsukura, S.; Kokubu, F.; Kurokawa, M.; Kawaguchi, M.; Ieki, K.; Kuga, H.; Odaka, M.; Suzuki, S.; Watanabe, S.; Homma, T. Role of RIG-I, MDA-5, and PKR on the expression of inflammatory chemokines induced by synthetic dsRNA in airway epithelial cells. Int. Arch. Allergy Immunol. 2007, 143, 80–83. [Google Scholar] [CrossRef]
  91. Cardinale, F.; Giordano, P.; Chinellato, I.; Tesse, R. Respiratory epithelial imbalances in asthma pathophysiology. Allergy Asthma Proc. 2013, 34, 143–149. [Google Scholar] [CrossRef] [PubMed]
  92. Comstock, A.T.; Ganesan, S.; Chattoraj, A.; Faris, A.N.; Margolis, B.L.; Hershenson, M.B.; Sajjan, U.S. Rhinovirus-induced barrier dysfunction in polarized airway epithelial cells is mediated by NADPH oxidase 1. J. Virol. 2011, 85, 6795–6808. [Google Scholar] [CrossRef] [Green Version]
  93. Rajan, D.; McCracken, C.E.; Kopleman, H.B.; Kyu, S.Y.; Lee, F.E.-H.; Lu, X.; Anderson, L.J. Human rhinovirus induced cytokine/chemokine responses in human airway epithelial and immune cells. PLoS ONE 2014, 9, e114322. [Google Scholar] [CrossRef] [Green Version]
  94. 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]
  95. Hayden, F.G. Rhinovirus and the lower respiratory tract. Rev. Med. Virol. 2004, 14, 17–31. [Google Scholar] [CrossRef]
  96. Muehling, L.M.; Heymann, P.W.; Wright, P.W.; Eccles, J.D.; Agrawal, R.; Carper, H.T.; Murphy, D.D.; Workman, L.J.; Word, C.R.; Ratcliffe, S.J.; et al. Human TH1 and TH2 cells targeting rhinovirus and allergen coordinately promote allergic asthma. J. Allergy Clin. Immunol. 2020, 146, 555–570. [Google Scholar] [CrossRef] [PubMed]
  97. Gern, J.E.; Vrtis, R.; Grindle, K.A.; Swenson, C.; Busse, W.W. Relationship of upper and lower airway cytokines to outcome of experimental rhinovirus infection. Am. J. Respir. Crit. Care Med. 2000, 162, 2226–2231. [Google Scholar] [CrossRef]
  98. Thomas, B.J.; Lindsay, M.; Dagher, H.; Freezer, N.J.; Li, D.; Ghildyal, R.; Bardin, P.G. Transforming growth factor-beta enhances rhinovirus infection by diminishing early innate responses. Am. J. Respir. Cell Mol. Biol. 2009, 41, 339–347. [Google Scholar] [CrossRef] [PubMed]
  99. O’Garra, A. Cytokines induce the development of functionally heterogeneous T helper cell subsets. Immunity 1998, 8, 275–283. [Google Scholar] [CrossRef] [Green Version]
  100. Zhang, J.M.; An, J. Cytokines, inflammation, and pain. Int. Anesthesiol. Clin. 2007, 45, 27–37. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  101. Gern, J.E.; Busse, W.W. Association of rhinovirus infections with asthma. Clin. Microbiol. Rev. 1999, 12, 9–18. [Google Scholar] [CrossRef] [PubMed]
  102. Palomino, D.C.; Marti, L.C. Chemokines and immunity. Einstein 2015, 13, 469–473. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  103. Juremalm, M.; Nilsson, G. Chemokine receptor expression by mast cells. Chem. Immunol. Allergy 2005, 87, 130–144. [Google Scholar] [CrossRef]
  104. Sheeran, P.; Jafri, H.; Carubelli, C.; Saavedra, J.; Johnson, C.; Krisher, K.; Sánchez, P.J.; Ramilo, O. Elevated cytokine concentrations in the nasopharyngeal and tracheal secretions of children with respiratory syncytial virus disease. Pediatr. Infect. Dis. J. 1999, 18, 115–122. [Google Scholar] [CrossRef]
  105. Folkerts, G.; Busse, W.W.; Nijkamp, F.P.; Sorkness, R.; Gern, J.E. Virus-induced airway hyperresponsiveness and asthma. Am. J. Respir. Crit. Care Med. 1998, 157, 1708–1720. [Google Scholar] [CrossRef] [PubMed]
  106. van Erp, E.A.; Luytjes, W.; Ferwerda, G.; van Kasteren, P.B. Fc-Mediated Antibody Effector Functions during Respiratory Syncytial Virus Infection and Disease. Front. Immunol. 2019, 10, 548. [Google Scholar] [CrossRef] [Green Version]
  107. Crotty, S. Follicular helper CD4 T cells (Tfh). Annu. Rev. Immunol. 2011, 29, 621–663. [Google Scholar] [CrossRef]
  108. Aderem, A.; Underhill, D.M. Mechanisms of phagocytosis in macrophages. Annu. Rev. Immunol. 1999, 17, 593–623. [Google Scholar] [CrossRef]
  109. Johnston, S.L. Innate immunity in the pathogenesis of virus-induced asthma exacerbations. Proc. Am. Thorac. Soc. 2007, 4, 267–270. [Google Scholar] [CrossRef] [PubMed]
  110. Wark, P.A.; Gibson, P.G. Asthma exacerbations 3: Pathogenesis. Thorax 2006, 61, 909–915. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  111. Mikhail, I.; Grayson, M.H. Asthma and viral infections: An intricate relationship. Ann. Allergy Asthma Immunol. 2019, 123, 352–358. [Google Scholar] [CrossRef]
  112. Barnes, P.J. Th2 cytokines and asthma: An introduction. Respir. Res. 2001, 2, 64. [Google Scholar] [CrossRef] [PubMed]
  113. Brandt, E.B.; Sivaprasad, U. Th2 Cytokines and Atopic Dermatitis. J. Clin. Cell. Immunol. 2011, 2, 1000110. [Google Scholar] [CrossRef] [PubMed]
  114. Kay, A.B.; Phipps, S.; Robinson, D.S. A role for eosinophils in airway remodelling in asthma. Trends Immunol. 2004, 25, 477–482. [Google Scholar] [CrossRef] [PubMed]
  115. Honda, K.; Chihara, J. Eosinophil activation by eotaxin—Eotaxin primes the production of reactive oxygen species from eosinophils. Allergy 1999, 54, 1262–1269. [Google Scholar] [CrossRef] [PubMed]
  116. Herbert, C.; Zeng, Q.X.; Shanmugasundaram, R.; Garthwaite, L.; Oliver, B.G.; Kumar, R.K. Response of airway epithelial cells to double-stranded RNA in an allergic environment. Transl. Respir. Med. 2014, 2, 11. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  117. Djukanović, R.; Harrison, T.; Johnston, S.L.; Gabbay, F.; Wark, P.; Thomson, N.C.; Niven, R.; Singh, D.; Reddel, H.K.; Davies, D.E. The effect of inhaled IFN-β on worsening of asthma symptoms caused by viral infections. A randomized trial. Am. J. Respir. Crit. Care Med. 2014, 190, 145–154. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  118. Wos, M.; Sanak, M.; Soja, J.; Olechnowicz, H.; Busse, W.W.; Szczeklik, A. The presence of rhinovirus in lower airways of patients with bronchial asthma. Am. J. Respir. Crit. Care Med. 2008, 177, 1082–1089. [Google Scholar] [CrossRef] [Green Version]
  119. Dharmage, S.C.; Perret, J.L.; Custovic, A. Epidemiology of asthma in children and adults. Front. Pediatr. 2019, 7, 246. [Google Scholar] [CrossRef]
Figure 1. Schematic illustration of major processes of tissue remodeling in virus-induced asthma. * These data were only shown in an in vitro study.
Figure 1. Schematic illustration of major processes of tissue remodeling in virus-induced asthma. * These data were only shown in an in vitro study.
Viruses 14 02616 g001
Table 1. Relationships among various cytokines, origins, and functions.
Table 1. Relationships among various cytokines, origins, and functions.
CytokinesOriginFunctionRef. No.
Th1IFN-γActivated T cellMacrophage activation[55,74]
IFN-λActivated T cellDendritic cell activation[75]
TNF-αActivated T cell, Monocyte/MacrophagePromotes inflammatory cytokines[50,76]
IL-2Activated T cellT cell/NK cell activation[51]
GM-CSFActivated T cell
Monocyte/Macrophage, Fibroblast
Neutrophil/Macrophage activation[52]
Th2IL-4Activated T cellB cell/T cell activation
IgE isotype switch
[53,58]
IL-5Activated T cell
Eosinophil, Mast cell
Eosinophil proliferation/differentiation[54,55]
IL-6Activated T cellInduce B cells into antibody-producing cells
T-cell differentiation
[56]
IL-13Activated T cell
Mast cell
Macrophage activation
IgE isotype switch
[57,58]
OthersIL-1Monocyte/MΦT cell activation[59]
IL-33Activated T cell
Endothelial/Epithelial cell
Promoting Th2-associated cytokines[60,61]
CXCL1
GROα
Epithelial cells, Macrophage, NeutrophilNeutrophil migration[62]
CXCL3
GROγ
MIP-2β
Monocyte, FibroblastNeutrophil migration/adhesion[63]
CXCL5
ENA-78
EosinophilNeutrophil migration/activation[64]
CXCL8
IL-8
Monocyte/Macrophage, FibroblastNeutrophil migration[62,65]
CXCL10
IP-10
Monocyte, FibroblastT cell/NK cell activation[66]
CCL2
MCP-1
Monocyte/Macrophage, FibroblastMonocyte migration[67]
CCL3
MIP-1α
Monocyte/Macrophage, Activated T cellMonocyte migration/infiltration
Eosinophil/mast cell/Dendritic cell migration
[68]
CCL4
MIP-1β
Monocyte/Macrophage, Activated T cellMonocyte/T cell migration[68]
CCL5
RANTES
Monocyte/Macrophage, FibroblastMonocyte/lymphocyte/Eosinophil/mast cell
migration
[69,70]
CCL11
Eotaxin
Airway Epithelial Cells, FibroblastEosinophil migration/degranulation
Mast cell activation/migration
[71]
CCL20
MIP-3α
Neutrophil, Natural Killer CellDendritic cell activation/migration
T cell/B cell migration
[72]
CCL24
Eotaxin-2
Monocyte/MacrophageEosinophil migration[71]
TSLPEpithelial cellsPromoting Th2-associated cytokines
B cell activation
[73]
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Hayashi, Y.; Sada, M.; Shirai, T.; Okayama, K.; Kimura, R.; Kondo, M.; Okodo, M.; Tsugawa, T.; Ryo, A.; Kimura, H. Rhinovirus Infection and Virus-Induced Asthma. Viruses 2022, 14, 2616. https://doi.org/10.3390/v14122616

AMA Style

Hayashi Y, Sada M, Shirai T, Okayama K, Kimura R, Kondo M, Okodo M, Tsugawa T, Ryo A, Kimura H. Rhinovirus Infection and Virus-Induced Asthma. Viruses. 2022; 14(12):2616. https://doi.org/10.3390/v14122616

Chicago/Turabian Style

Hayashi, Yuriko, Mitsuru Sada, Tatsuya Shirai, Kaori Okayama, Ryusuke Kimura, Mayumi Kondo, Mitsuaki Okodo, Takeshi Tsugawa, Akihide Ryo, and Hirokazu Kimura. 2022. "Rhinovirus Infection and Virus-Induced Asthma" Viruses 14, no. 12: 2616. https://doi.org/10.3390/v14122616

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