Next Article in Journal
The Role of Mycoplasma bovirhinis in the Development of Singular and Concomitant Respiratory Infections in Dairy Calves from Southern Brazil
Previous Article in Journal
Detection of Felis catus Gammaherpesvirus 1 in Domestic Cat Saliva: Prevalence, Risk Factors, and Attempted Virus Isolation
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Mucosal Immunity against SARS-CoV-2 in the Respiratory Tract

1
Department of Otorhinolaryngology, Yonsei University College of Medicine, Seoul 03722, Republic of Korea
2
Department of Biomedical Sciences, Yonsei University College of Medicine, Seoul 03722, Republic of Korea
*
Author to whom correspondence should be addressed.
Pathogens 2024, 13(2), 113; https://doi.org/10.3390/pathogens13020113
Submission received: 11 December 2023 / Revised: 20 January 2024 / Accepted: 22 January 2024 / Published: 26 January 2024
(This article belongs to the Section Immunological Responses and Immune Defense Mechanisms)

Abstract

:
The respiratory tract, the first-line defense, is constantly exposed to inhaled allergens, pollutants, and pathogens such as respiratory viruses. Emerging evidence has demonstrated that the coordination of innate and adaptive immune responses in the respiratory tract plays a crucial role in the protection against invading respiratory pathogens. Therefore, a better understanding of mucosal immunity in the airways is critical for the development of novel therapeutics and next-generation vaccines against severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) and other respiratory viruses. Since the coronavirus disease 2019 pandemic, our knowledge of mucosal immune responses in the airways has expanded. In this review, we describe the latest knowledge regarding the key components of the mucosal immune system in the respiratory tract. In addition, we summarize the host immune responses in the upper and lower airways following SARS-CoV-2 infection and vaccination, and discuss the impact of allergic airway inflammation on mucosal immune responses against SARS-CoV-2.

1. Introduction

Since the emergence of the initial case of pneumonia with an unknown cause in Wuhan, China, in December 2019 [1], the novel severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has rapidly spread worldwide. SARS-CoV-2 infection causes coronavirus disease 2019 (COVID-19), which manifests a broad spectrum of clinical presentations, ranging from asymptomatic infection to severe disease [2]. COVID-19 has caused global morbidity and devastating disruptions in daily life. Prophylactic vaccines using various platforms were developed during the COVID-19 pandemic, and their administration started in populations worldwide in December 2020. Despite improved protection against SARS-CoV-2 infection through the development of vaccines, a deeper understanding of the immune response to SARS-CoV-2 will help guide the development of next-generation vaccines or therapeutics against respiratory viral infections. This knowledge will contribute to improved preparedness for future pandemics.
The COVID-19 outbreak and global efforts to overcome the pandemic have substantially increased our knowledge of immune responses against respiratory viruses, particularly in the human system. However, although blood cells are not the primary site of SARS-CoV-2 infection, the majority of the analysis of the immune response has been limited to peripheral blood (PB). The nasal cavity is the site of both viral entry and the initial replication of SARS-CoV-2 [3]. SARS-CoV-2 spreads to the epithelial cells of the lower respiratory tract and causes pneumonia, particularly in cases of severe COVID-19 [4], whereas limited levels of viral replication can occur in other tissues [5,6]. Respiratory viruses, such as the coronavirus and influenza virus, are potential causes of future pandemics and primarily infect the respiratory mucosa; therefore, it is important to understand and measure local immune responses in the airway.
Several reviews have discussed the various aspects of the systemic immune response to SARS-CoV-2 following infection and vaccination [7,8]. In the current review, we focus on the local immune response to SARS-CoV-2 in the airway. We summarize the current knowledge of the characteristics of the mucosal immune system in the respiratory tract, and discuss airway immune responses in the context of COVID-19. Furthermore, we describe alterations in mucosal immune responses against SARS-CoV-2 under allergic airway inflammation conditions.

2. An Overview of the Characteristics of Respiratory Mucosal Immunity

2.1. Epithelial Barrier

The epithelial barrier serves as the first line of defense against invasion by respiratory pathogens and allergens in the airways. It includes cell–cell junctions that function as a physical barrier, mucociliary clearance for mechanically removing inhaled pathogens and allergens the commensal microbiota, immunoglobulins (Igs), and defense molecules [9]. These are crucial for trapping and cleansing risk factors and modulating the immune response. Cell–cell junctions comprise various structures, including tight junctions (TJs), adherens junctions (AJs), gap junctions, desmosomes, and hemidesmosomes. In particular, apical junction complexes (AJCs) play an important role in epithelial barrier function. The AJC consists of the most apically located TJs and underlying AJs [10]. TJs regulate the movement of ions and molecules, as well as paracellular transport, whereas AJs are essential for initiating and maintaining cell–cell adhesion [11]. TJs are composed of proteins, such as occludin (OCLN), claudin, junctional adhesion molecules, and zonula occludens (ZO), whereas AJs contain complexes such as cadherin/catenin and nectin/afadin [12]. The expression of these genes and proteins in the AJC serves as an indicator of epithelial barrier function and transepithelial electrical resistance (TER) [13]. In addition, commensal microbiota plays a defensive role against viral infections in the upper respiratory tract by modulating-type I and type III interferon (IFN)-mediated immune mechanisms [14,15,16]. Nasal commensal Staphylococcus epidermidis was reported to provide initial antiviral protection against influenza A virus infection by enhancing the IFN-λ-dependent innate immune response in the nasal mucosa [16].
Epithelial barrier dysfunction can be caused by several factors, including mucociliary dysfunction and microbial dysbiosis. In particular, the disruption of epithelial barrier integrity leads to a decrease in TER and increased barrier permeability. This disruption can occur owing to various pathogens, allergens, or other pathological conditions. The infection of human rhinovirus in human nasal epithelial cells in vitro leads to a decreased expression of membrane proteins, including ZO-1, E-cadherin, claudin-1, and OCLN [17,18]. Similar alterations were observed after the infection of bronchial epithelial cells (BECs), where a reduced expression of TJ proteins at the epithelial barrier, decreased TER, and the dissociation of ZO-1 from the TJs was observed [19]. Several studies have reported defective epithelial barriers in patients with allergic rhinitis (AR), chronic rhinosinusitis, and asthma [20,21,22]. The expression of CLND-1, OCLN, and ZO-1 proteins was found to be significantly reduced in the BECs of asthmatic children compared to that in non-asthmatic controls [19]. House dust mites (HDMs) are an AR allergen and exhibit proteolytic activity that cleaves TJ proteins [23,24]. Steelant et al. reported a decrease in OCLN and ZO-1 expression along with increased epithelial permeability in the nasal epithelium of patients with HDM-induced AR [22].

2.2. Innate Immunity

Airway surface fluid is composed of mucus, antimicrobial peptides (AMPs), and enzymes, playing a crucial role as the first line of host defense in nature. Mucus is produced by mucous glands and goblet cells, capturing inhaled debris and pathogens and facilitating mucociliary clearance through ciliary beating. The primary components of airway mucus are O-glycosylated mucin glycoproteins, categorized as gel-forming and transmembrane mucins. In gel-forming mucins, MUC5AC and MUC5B play a crucial role in effectively clearing pathogens through mucociliary clearance. Among the transmembrane mucins, MUC1, MUC4, and MUC16 are the major ones, with MUC1 being the most abundant. While MUC1 and MUC4 are present in both the upper and lower respiratory tracts, MUC16 is exclusively expressed in the lower tract. These mucins create a barrier in the respiratory epithelium, acting as decoy receptors against pathogens. Additionally, they contribute to mucociliary clearance by shedding the extracellular domain, which binds to pathogens and releases them into the lumen, from the cell surface [25]. AMPs include cationic defensins like human β-defensins (HBDs) 1, 2, 3, 4, human cathelicidins (LL-37), and secretory leukocyte protease inhibitor (SLPI). They exhibit synergistic activity with host defense molecules like lysozymes and lactoferrin. AMPs are upregulated during infections, demonstrating microbiocidal effects and immune modulation activities such as chemotactically attracting immune cells and modulating cytokine production [26].
The immune response to viral infections is initiated by the innate immune system, which recognizes pathogens and induces the production of pro-inflammatory cytokines and chemokines. Innate responses are triggered when immune cells with pattern recognition receptors (PRRs) recognize pathogen-associated molecular patterns (PAMPs) which include viral molecules, such as viral RNA or oxidized phospholipids, or damage-associated molecular patterns (DAMPs), such as endogenous host molecules released from damaged and dying cells. PRRs, such as Toll-like receptors (TLRs), retinoic acid-inducible gene (RIG-I)-like receptors (RLRs), nucleotide-binding oligomerization domain (NOD)-like receptors (NLRs), and other cytosolic virus sensors, initiate the immune response to viral infections [27].
After the recognition of PAMPs and DAMPs, antiviral responses are activated through type I or III interferon (IFN) signaling pathways and the production of pro-inflammatory cytokines and chemokines, ultimately suppressing viral replication, eliminating infected cells, and initiating an adaptive immune response [28,29]. Viral recognition leads to IFN induction via three major pathways: cyclic guanosine monophosphate-adenosine monophosphate synthase/stimulator of IFN genes (cGAS/STING), TLR/TIR-domain-containing adapter-inducing IFN-β (TRIF)/Myeloid differentiation factor 88 (MyD88), and RLR/mammalian mitochondrial antiviral signaling protein (MAVS) pathways. All pathways activate the kinase TBK1 which phosphorylates the transcription factors IFN regulatory factor (IRF)3 and IRF7, thereby stimulating IFN production. They also activate the nuclear factor-κB (NF-κB) family of transcription factors and the subsequent production of pro-inflammatory cytokines [30].
Type I IFNs (IFN-α, IFN-β) bind to IFNAR (IFN-α/β receptor), and type III IFNs (IFN-λ 1,2,3,4) bind to IFNLR (IFN-λ receptor). Type I and III IFNs are key players in the antiviral innate immune response and activate several signaling cascades, including the JAK/STAT pathway. Subsequently, they form IFN-stimulated gene (ISG) factor 3 (ISGF3) complexes, driving the expression of hundreds of ISGs that primarily elicit an effective antiviral response [28,31,32,33]. IFN-λ acts in a more targeted fashion, as the expression of its receptor IFNLR1 is limited to mucosal epithelial cells of the respiratory, gastrointestinal, and reproductive tracts and specific immune cells, whereas IFNAR is expressed in most cells throughout the body [34,35]. Respiratory epithelial cells respond to both IFN-λ and IFN-α/β. In general, during the early stage of infection, an immune response based on IFN-β and IFN-λ is produced by infected cells, which is probably local. However, as the infection progresses, plasmacytoid dendritic cells become major cellular sources of IFN-α, leading to a more systemic response [36]. In some respiratory virus infections, respiratory epithelial cells primarily produce IFN-λ rather than IFN-α and IFN-β, implying a key role of IFN-λ in mediating antiviral immunity in the respiratory tract [37,38]. These results suggest a significant potential for type III IFN to be used as a therapeutic target for respiratory viral infections, considering its higher tissue specificity compared to type I IFN.
In addition, dendritic cells (DCs), as one of the most potent types of antigen-presenting cells, play a crucial role in innate immunity and contribute significantly to antiviral responses. They consist of two main functional subtypes: conventional or myeloid DCs (cDCs) and plasmacytoid DCs (pDCs). cDCs include CD1c+, CD16+, and CD141+ cDC subsets and also monocyte-derived DCs generated by circulating monocytes in the presence of inflammation [39]. In human tissues, CD141-expressing cDC1s in the vascular wall and mucosa stimulate Th1 responses, while CD1c-expressing cDC2s in the lamina propria produce inflammatory chemokines, facilitating immune cell aggregation, and may contribute to immune tolerance [40]. pDCs are present throughout the lung tissue including the airway. They play a vital role in innate immunity by producing a substantial amount of type I IFN during early antiviral responses through the stimulation of TLR 7/9 [39].

2.3. Tissue-Resident Memory T Cells in the Respiratory Tract

Following the innate immune response, the adaptive immune system is activated to control and eradicate infections. Despite the importance of humoral immune responses, coordinated cellular immunity is also essential for disease control [41,42]. Sterilizing immunity is primarily mediated by antibodies; however, insufficient antibody levels or mutations in antibody-binding sites may cause reinfection. In such scenarios, memory T cell responses are crucial for the prevention of viral dissemination and progression into severe disease, particularly in the context of infections with variants. Previous studies have shown that virus-specific memory T cells are long-lasting after recovery from viral infection [43,44]. Robust T-cell responses in the respiratory tract may play a crucial role in impeding disease progression by rapidly exerting effector functions, given that re-exposure to respiratory viruses primarily occurs in the airway [45].
Tissue-resident memory T (TRM) cells are a subset of memory T cells characterized by long-term residency in non-lymphoid tissues [46,47]. TRM cells are found in almost all peripheral tissues, including the skin [48], lung [49,50], gut [51], brain [52,53], and genital tract [54]. TRM cells from various tissues exhibit a common gene expression profile, in addition to some tissue-specific differences in gene expression. In animal models, the tissue-residency of T cells can be determined by various experimental methods, including parabiotic surgery, in vivo antibody labeling, T cell depletion, and tissue transplantation [47]. While these techniques rigorously determine TRM cells, their application in humans has clear limitations. Thus, phenotypic or transcriptional profiling is the primary method for the identification of TRM cells in humans. TRM cells are frequently defined by the expression of surface markers associated with tissue retention [46,47]. As CD69 promotes tissue retention by downregulating sphingosine-1-phosphate receptor-1 (S1PR1) that is required for tissue egress [55], CD69 is the canonical marker of TRM cells. In addition to CD69, other surface markers, including adhesion molecules and tissue-homing chemokine receptors, characterize TRM phenotypes. However, the expression of these surface markers on TRM cells varies across tissues. CD103 (αE integrin), which binds to E-cadherin on epithelial cells, is highly expressed on TRM cells in epithelial tissues such as the skin, intestines, and lungs [47]. In contrast, most CD8+ TRM cells in the liver and secondary lymphoid organs lack CD103 expression [56,57,58]. Several studies have reported that CD103+CD4+ TRM cells are also present in barrier tissues [59,60]. Collectively, the combination of CD69 and CD103 may serve as a reliable marker for identifying human TRM cells, especially in epithelial tissues. CD49a (integrin α1), which forms very late antigen-1 with CD29 (integrin β1), is expressed on a subset of CD4+ and CD8+ TRM cells in diverse tissues [57,60,61,62,63]. A recent study in mice demonstrated that CD49a facilitates the locomotion of virus-specific CD8+ TRM cells within the lungs [64]. In addition, the tissue retention of CD8+ TRM cells is impaired in the absence of very late antigen-1 [62]. Furthermore, TRM cells express a variety of tissue-homing chemokine receptors required for localization in tissues. CXCR6, the chemokine receptor for CXCL16, is highly expressed in human TRM cells in the lungs [65], liver [57], and lymphoid tissues [60]. Studies in mice have demonstrated that CXCR6 is required for the recruitment of CD8+ TRM cells to the airway epithelium [65]. CXCR3, the receptor for the chemokines CXCL9, CXCL10, and CXCL11, is also expressed in a proportion of lung TRM cells [49,66].
TRM cells rapidly exert effector functions upon a secondary pathogen encounter and restrict disease progression. Accumulating evidence has shown a protective role for airway TRM cells against respiratory viral infections. An experimental respiratory syncytial virus (RSV) challenge in healthy adult volunteers showed that the abundance of pre-existing virus-specific CD8+ TRM cells in bronchoalveolar lavage fluid (BALF) before infection correlated with reduced symptoms and viral loads [67]. In a mouse model, the transfer of airway TRM cells from previously infected animals led to the protection against RSV infection [68]. In the mouse model of coronavirus infections, protection from disease was primarily mediated by virus-specific CD4+ TRM cells in the upper airway to promote the secretion of IFN-γ and the recruitment of virus-specific CD8+ T cells [69]. Several studies have also demonstrated that lung CD4+ and CD8+ TRM cells confer protective immunity against influenza virus infection [66,70]. Virus-specific TRM cells are also critically required for optimal protection from infection with heterosubtypic influenza viruses [71]. Furthermore, a seminal study has shown that nasal CD8+ TRM cells limit viral spread to the lungs and reduce the severity of pulmonary diseases [72].

2.4. Tissue-Resident Memory B Cells and Antibody-Secreting Cells in the Respiratory Tract

Similar to memory T cells, memory B cells persist long-term and rapidly differentiate into antibody-secreting cells upon antigen re-encounter [73]. Memory B cells can re-enter the germinal center upon antigen re-exposure and undergo further affinity maturation [74]. Antibodies produced by plasma cells that are terminally differentiated cells contribute to the defense against pathogen reinfection [75]. The cooperation between memory B cells and plasma cells confers robust protection against pathogens [74]. The proximity of antigen-presenting cells, memory follicular helper T (TFH) cells, and memory B cells facilitates robust recall responses.
Analogous to TRM cells, recent data have revealed the existence of a specific subset of memory B cells residing in peripheral tissues, known as tissue-resident memory B (BRM) cells [76,77,78,79,80]. Allie et al. conducted a parabiosis surgery and intravenous antibody labeling and demonstrated that pulmonary influenza infection elicited lung BRM cells with distinct phenotypes compared to their systemic counterparts [76]. Similarly, another study showed that intranasal immunization induced IgA-producing BRM cells in the lungs [77]. Specific markers for the identification of BRM cells are required to investigate their characteristics. To identify specific markers for BRM cells, the transcriptional profiles of both murine and human BRM cells have been analyzed [79,81]. The results showed that lung memory B cells exhibited a higher expression of CXCR3 and CD69 than their counterparts in the mediastinal lymph nodes and spleen. The downregulation of CCR7, SELL, S1PR1, and KLF2, and the upregulation of CXCR3, CCR6, and CD69 were observed in lung BRM cells [79]. Consistent with these results, the upregulation of CD69, CXCR3, and CCR6, the chemokine receptor for CCL20, has been observed in memory B cells of the human lungs [78,79]. Most CD27+ memory B cells from the human gut also express CD69 [80]. These results suggest that BRM cells share common signatures underlying tissue residency with TRM cells in non-lymphoid organs. However, the transcriptional program determining the differentiation and fate of memory B cells remains unclear.
Humoral responses to pathogen re-encounters have been investigated in peripheral tissues. BRM cells rapidly differentiate into antibody-secreting cells during secondary infections and subsequently secrete antibodies against pathogens [76]. In influenza infections, reinfection induces the formation of inducible bronchus-associated lymphoid tissue, which supports the maturation and selection of B cells, thereby generating BRM cells as well as resident memory TFH cells [82]. In addition, mice with IgA-producing lung BRM cells following local immunization show superior protection against secondary challenges with both homologous and heterologous strains of the influenza virus, supporting the cross-reactivity of local humoral immunity [77]. Intriguingly, both CXCR3+CCR6+ virus-specific B cells and CXCR3CCR6+ bystander B cells are generated in the lung after infections with the influenza virus and SARS-CoV-2 [83]. However, the potential benefits of bystander B cells remains to be elucidated. In the case of influenza reinfection, alveolar macrophages are key initiators of humoral recall responses through the secretion of IFN-γ and CXCR3 ligands, which in turn activate the recruitment of CXCR3+ BRM cells to the infection site [84]. Although it remains unclear whether memory TFH cells are critically required for the initiation of these recall responses, it can be assumed that BRM cells as well as resident memory TFH cells are recruited close to alveolar macrophages, thereby exerting robust protective immunity.

3. SARS-CoV-2 Entry into the Respiratory Tract

The coronavirus virion contains spike (S), envelope (E), and membrane (M) proteins, with an RNA genome complexed with nucleocapsid (N) proteins to form a helical capsid [85]. The S protein is anchored to the viral envelope and is composed of S1 and S2 subunits. During infection, two cleavage events occur for cellular entry: the cleavage at the junction of the S1 and S2 subunits and at the S2′ site, located upstream from the fusion peptide within the S2 subunit [86]. In SARS-CoV-2, the S1–S2 boundary is cleaved by furin during virus maturation [87], and when the S protein binds to the angiotensin-converting enzyme 2 (ACE2) entry receptor of the host target cell, the S2′ cleavage site is exposed [88]. The exposed S2′ site is cleaved by different host proteases depending on the entry route. If the transmembrane protease serine subtype 2 (TMPRSS2) is sufficiently expressed near ACE2 on the target cell, the virus-ACE2 complex encounters TMPRSS2 at the cell surface, leading to S2′ cleavage followed by membrane fusion to release viral RNA into the cell cytoplasm [89,90]. This step must occur after ACE2 binding to ensure viral S protein activation [91,92]. The S protein cleavage by TMPRSS2 facilitates early entry into the cell membrane as opposed to late entry through the endosome. Although TMPRSS2 can be replaced by other proteases, its binding to the ACE2 receptor is essential for cell entry. If TMPRSS2 expression is insufficient and the complex does not encounter TMPRSS2, the virus-ACE2 complex is internalized into endosomes. Cleavage at the S2′ site is then performed by low pH-triggered cathepsin in the endosomes, leading to the fusion with the endosomal membrane and the release of viral RNA into the cell cytoplasm [88,93].
Following cellular entry, genomic RNA is translated into two large polyproteins, pp1a, corresponding to NSP1 to NSP11 from open reading frame (ORF)1a, and pp1ab, corresponding to NSP12 to NSP16 from ORF1b. These encode 16 nonstructural proteins (NSPs) that facilitate the formation of the viral replication–transcription complex [94]. The SARS-CoV-2 genome encodes structural proteins, including S, E, M, and N, as well as accessory proteins, including ORF3a, 3b, 6, 7a, 7b, 8, 9b, 9c, and 10. These ORFs and NSPs play crucial roles in viral replication and the evasion of the host immune response [95]. Replicated genomic RNA and structural proteins are assembled in the endoplasmic reticulum–Golgi intermediate compartment and the fully formed virions are exocytosed [96].
Numerous studies have focused on the expression levels of ACE2 rather than TMPRSS2 to understand the differences in SARS-CoV-2 infection risk and clinical outcomes [97,98]. The co-expression of ACE2 and TMPRSS2 within cells is considered important for the entry of SARS-CoV-2. Notably, these viral entry-associated genes of SARS-CoV-2 are known to be highly expressed in the nasal epithelial cells [3,99], and their expression in the upper respiratory epithelium is believed to have a positive correlation with viral susceptibility and transmissibility. An average three-fold increase in ACE2 expression has been observed in secretory cells from patients with COVID-19 compared to control individuals [4]. Notably, SARS-CoV-2 causes a decrease in epithelial barrier function and the disruption of TJs. It also perturbs ciliogenesis and downregulates Foxj1 expression, leading to the loss of ciliated epithelial cells and the impairment of mucociliary clearance [100]. These results suggest a pathogenic mechanism that underlies SARS-CoV-2 spread in the respiratory tract.

4. Innate Immunity to SARS-CoV-2 Infection in the Airway

Innate immune defense plays a key role in controlling SARS-CoV-2 infections, and deficiencies in the innate immune system can trigger severe COVID-19 [101,102]. Zhang et al. reported that patients with inborn defects in TLR3- and IRF7-dependent type I IFN immunity are prone to life-threatening COVID-19 pneumonia [102]. Bastard et al. reported that the presence of autoantibodies against type I IFN-α2 and IFN-ω is associated with a high risk of severe COVID-19 [101].
Accumulating evidence supports that both soluble and transmembrane mucins play important roles in SARS-CoV-2 infection, but whether they are protective or pathogenic still remains controversial. Mucus hypersecretion may exhibit a negative impact on disease development or progression due to reduced mucocililary clearance. Indeed, the protein levels of MUC1 and MUC5AC were elevated in airway mucus of patients with COVID-19 compared to control individuals [103], and a high production of MUC5AC was observed in SARS-CoV-2-infected primary respiratory [104]. Other researchers also showed that SARS-CoV-2 infection is associated with a high prevalence of MUC5B-dominated mucus plugging in the distal lung, and MUC5B expression was increased in airway regions of COVID-19 autopsy lungs [105]. These data suggest that mucolytic agents may be therapeutics for COVID-19. In contrast, a recent study reported in vitro evidence of protective functions of the glycosylated extracellular domains of transmembrane mucins in different respiratory cell types by preventing SARS-CoV-2 binding and entry [106]. A recent study conducting genome-wide CRISPR screens showed that the overexpression of transmembrane mucins MUC1, MUC4, or MUC21 reduced SARS-CoV-2 infection compared to cells with a non-targeting guide [107]. Further in vivo studies need to address the precise role of each mucin in the pathophysiology of COVID-19.
AMPs, such as defensins, may also play a role in protection against SARS-CoV-2 infection. A previous study showed that β-defensin transcripts were increased in the nasopharyngeal swab samples from patients with SARS-CoV-2 infection compared to those from control individuals [108]. Recent research also suggests that human defensins may inhibit SARS-CoV-2 infection by blocking viral entry [109]. The precise role of AMPs needs to be further elucidated.
Similar to other respiratory viruses, SARS-CoV-2 RNAs are mainly recognized by TLRs and RLRs, including RIG-I and MDA5 [110,111,112]. TLRs activate the TRIF/MyD88 signaling pathway, and RLRs (RIG-I and MDA5) activate the MAVS pathway, leading to the production of various cytokines, including pro-inflammatory cytokines such as TNF-α, IL-1β, IL-6, and type I and III IFNs [113]. NLRs, such as NLRP3 inflammasome, and the cGAS–STING signaling pathway, which is activated upon the detection of cytosolic DNA from damaged host mitochondria, have also been reported to sense SARS-CoV-2 infection and induce the production of type I IFNs and pro-inflammatory cytokines [96,114,115,116]. A decreased expression of PRRs implies a weaker innate antiviral response. Loske et al. reported that adults, who exhibit higher rates of SARS-CoV-2 infection and an increased risk of severe COVID-19 compared to children, demonstrated lower levels of PRRs, such as MDA5 and RIG-I, in the upper airway epithelial cells and innate immune cells compared to children [117].
SARS-CoV-2 infection is characterized by a significant lack of IFN production and secretion [118,119]. A notable difference between SARS-CoV-2 and influenza A virus infection is the poor induction of type I IFN response in COVID-19 [120,121]. Hadjadj et al. reported that patients with severe COVID-19 demonstrated a low or no IFN-α response and an absence of circulating IFN-β [121]. However, the mechanisms underlying these delayed and inefficient type I IFN responses in SARS-CoV-2 infection remain unclear. Several SARS-CoV-2 proteins have recently been reported as antagonists of the type 1 IFN response. NSPs, structural proteins M and N, and accessory proteins (ORF3a, 3b, 6, 7a, and 9b) interfere with type I IFN signaling directly or indirectly [122,123,124,125]. Some of them also degrade factors related to the IFN pathway via autophagy. ORF9b antagonizes type I and III IFN by targeting multiple signaling pathways, including RIG-I/MDA-5-MAVS, TLR3-TRIF, and cGAS–STING [126]. NSP15 suppresses type I IFN production by inhibiting phosphorylation and the nuclear translocation of IRF3 [127]. NSP13 inhibits type I IFN production by degrading TBK1 via p62-dependent selective autophagy [128].
pDCs act as the primary source of type I IFN upon detecting SARS-CoV-2-infected cells in the nasal mucosa. The sensing mechanism of SARS-CoV-2-infected cells by pDCs requires integrin-mediated cell adhesion (αLβ2 integrin and ICAM-1), allowing them to efficiently inhibit viral replication through a local response at the contact site with infected cells [129]. According to recent studies, the loss of pDC response was observed in severe COVID-19 [129,130].
Following poor IFN I and III responses, patients with severe COVID-19 fail to suppress viral replication in the early phase of the infection, leading to an exaggerated inflammatory response in the late phase. The dysregulated release of pro-inflammatory cytokines may contribute to life-threatening immune responses in COVID-19, such as a cytokine storm mediated by inflammatory cell death (PANoptosis). PANoptosis is primarily induced by the synergism of TNF-α and IFN-γ and is dependent on the activation of the JAK/STAT1/IRF1 axis and the subsequent activation of caspase-8 to drive cell death [131].
Analyses of human airway specimens have supported the essential role of the local innate immune response. A single-cell RNA sequencing analysis of nasopharyngeal swab samples from patients with COVID-19 showed that epithelial cells from severe cases exhibited the blunted expression of IFN-responsive or antiviral genes, suggesting that impaired antiviral immune responses in the nasal epithelium may underlie severe disease [132]. Additionally, IFN-α2 levels in endotracheal aspirate from patients with COVID-19 negatively correlated with the duration of hospital stay [133]. In contrast, an exaggerated innate immune response and the augmented recruitment of immune cells may contribute to tissue injury in patients with severe COVID-19. Patients with critical COVID-19 have been shown to exhibit stronger interactions between epithelial and immune cells and a higher activation status of inflammatory macrophages expressing CCL2, CCL3, CCL20, CXCL1, CXCL3, CXCL10, IL8, IL1B, and TNF than moderate cases [4].
Of note, the innate cellular response in the airways of pediatric patients differs from that observed in adults. In the nasal airways, the local innate IFN response to SARS-CoV-2 is stronger in pediatric immune cells compared with adult immune cells. A previous study reported that the airway epithelium showed a higher steady-state expression of IFN-response genes in children [134]. Pre-stimulation with IFNs may restrict viral spread and underlie mild diseases in children. Many types of innate immune cells also had elevated IFN response signatures in children compared to adults, particularly CD56lo natural killer cells, natural killer T cells, neutrophils, and CXCL10+ monocytes [134].

5. Adaptive Immunity to SARS-CoV-2 Infection in the Airway

5.1. T-Cell Responses against SARS-CoV-2 in the Airway

As with infections with other respiratory viruses, TRM cells have been suggested to play a critical role in rapid protection against SARS-CoV-2 infection (Figure 1). After the resolution of the natural infection, SARS-CoV-2-specific T cells have been found in various tissues, including the bone marrow, spleen, lungs, and lymph nodes [135]. These SARS-CoV-2-specific T cells persist for at least six months after infection, and their frequency correlates with that of circulating T cells. In addition, a previous T-cell receptor sequencing analysis showed that CD8+ T cells carrying SARS-CoV-2-specific T-cell receptors were observed in nasal tissue after the resolution of SARS-CoV-2 infection [136]. However, the longevity of SARS-CoV-2-specific airway TRM cells following natural infection remains enigmatic.
Intriguingly, previous studies using stimulation-based functional assays have shown that SARS-CoV-2-reactive T cells expressing canonical TRM markers, including CD69 and CD103, are detected in the tonsils and BALF samples of individuals unexposed to SARS-CoV-2 [137,138]. These SARS-CoV-2-reactive T cells were thought to be induced by previous infection with other common cold coronaviruses. Although these tissue-resident cross-reactive T cells were shown to recognize multiple SARS-CoV-2 epitopes present in structural and nonstructural proteins, the protective role of these cells needs to be addressed.
Mucosal-associated invariant T (MAIT) cells are a subset of innate T cells that are involved in mucosal immunity and protection against viral infections. Several studies showed reduced frequencies and activated phenotypes of circulating MAIT cells in patients with COVID-19. In contrast, a significant enrichment of MAIT cells with activated phenotypes was observed in the airways, suggesting a potential contribution to the regulation of local infection and inflammation in patients with COVID-19 [139]. Additionally, others suggested that altered MAIT cell functions due to IFN-α–IL-18 imbalance may contribute to the disease severity of COVID-19 [140]. Remarkably, the frequency of circulating MAIT cells was restored in convalescent patients, indicating dynamic recruitment to the tissues during the acute phase and subsequent release back into the circulation after the resolution of the disease [141]. These findings collectively indicate that MAIT cells are involved in the immune response against SARS-CoV-2 and suggest their possible contribution to COVID-19 immuno-pathogenesis.
Since the administration of COVID-19 vaccines, whether intramuscular vaccination induces SARS-CoV-2 S-specific TRM cells in the respiratory tract has been a topic of scientific interest. Several studies have performed stimulation-based assays to investigate the presence of SARS-CoV-2-reactive T cells in airway samples of vaccinees. One study identified S-specific T cells in nasopharyngeal swab samples from vaccinees [142], consistent with recent findings that mRNA vaccination induces TRM cell generation in a mouse model [143]. Another study also reported that S-specific CD4+ T cells were detected in the lung tissue of vaccinated patients, although polyfunctional CD107a+IFN-γ+ TRM cells were virtually absent in vaccinated individuals [144]. These results indicate that mRNA vaccination induces SARS-CoV-2 S-specific T cell responses in the lungs, although to a limited extent. In contrast, a study analyzing paired PB and BALF samples from vaccinated individuals without breakthrough infections showed that the frequency of S-reactive T cells was significantly lower in BALF than in PB [145]. In another study, SARS-CoV-2-reactive T cells were not detected in the nasal secretions of vaccinees without infection [146]. Furthermore, a recent analysis of BALF samples reported that vaccination alone did not elicit S-specific T cell responses that were significantly greater than those in pre-pandemic samples [147].
Apart from the controversy surrounding whether intramuscular vaccination induces SARS-CoV-2 S-specific TRM cells in the airways, a growing body of evidence from animal models has demonstrated the superior ability of intranasal vaccination to induce airway TRM cells [143,148,149,150]. Therefore, innovative vaccines with different routes of administration are being developed, with a focus on inducing greater mucosal immunity that can provide durable protection at the site of infection.

5.2. Humoral Immune Responses to SARS-CoV-2 in the Airway

Although circulating antibodies in PB contribute to viral clearance, the successful generation of mucosal memory B cells allows for a rapid increase in the local antibody titer, which could mediate efficient viral clearance and the prevention of viral spread upon reinfection (Figure 1) [151]. As described above, memory B cells have been identified in multiple mucosal tissues, including the lungs of mice, following influenza virus infection [76]. Similarly, a previous study showed that following a natural infection, SARS-CoV-2-specific memory B cells were induced in multiple human tissues, including the bone marrow, spleen, lungs, and lymph nodes [135]. Furthermore, IgA is a key component of the mucosal immune response to SARS-CoV-2. Virus-specific IgA antibodies are produced by antibody-secreting cells in the mucosal-associated lymphoid tissues, such as the palatine tonsils, and are secreted into the respiratory tract, where they play several important roles in mucosal defense, such as preventing the entry of the virus and reducing transmission. SARS-CoV-2-specific IgA antibodies were detected in the saliva and BALF of patients with COVID-19, and SARS-CoV-2 neutralization was more closely correlated with IgA than with IgM or IgG in the first weeks after symptom onset [152]. Recent studies also showed that wild-type SARS-CoV-2 spike-specific nasal IgA antibodies are associated with protection against infections with SARS-CoV-2 variants, including Omicron BA.1, BA.2, and BA.5 [153,154]. However, another study reported waning nasal SARS-CoV-2-specific IgA antibodies nine months after COVID-19 [155], and these antibodies were not induced by intramuscular COVID-19 vaccination [156]. These data raise the need to develop novel vaccine strategies.
Interestingly, adults and children exhibit different IgA mucosal antibody responses to SARS-CoV-2. Some studies, particularly in children, have reported that mild or low antigen exposure might promote mucosal SARS-CoV-2-specific IgA responses, showing an earlier IgA mucosal immune response to SARS-CoV-2 [157,158]. The levels of SARS-CoV-2 S protein-specific IgA in nasal fluid inversely correlated with age [157]. In addition, a longitudinal study by Chan et al. reported that early and robust nasal S1-specific IgA levels are linked to a rapid decline in viral load, as evidenced by analyses of SARS-CoV-2 S1-specific IgA levels and viral titers in nasal epithelial lining fluid [158]. In the study, pediatric patients, especially those who were asymptomatic, exhibited a rapid induction of IgA within the initial four days post diagnosis, while a noteworthy increase in IgA was detected only between 12 and 16 days post diagnosis in adult patients [158]. These findings may also support why children showed a lower risk of SARS-CoV-2 infection and milder disease status.
The palatine tonsils and adenoids are secondary lymphoid structures that are located at the nasopharynx and oropharynx, where antigen-specific T and B cell responses in the upper respiratory tract can be generated. It was reported that tonsils and adenoids are sites of the persistence of SARS-CoV-2 in children even without symptoms [159]. Therefore, memory B cells in the palatine tonsils and adenoids may play a pivotal role in immune defense against SARS-CoV-2. A recent study analyzing tonsil and adenoid tissues from pediatric patients with COVID-19 showed that the majority of SARS-CoV-2 S1-specific B cells from tonsils and adenoids exhibited distinct phenotypes, characterized by a higher expression of CXCR3 and HOPX and a lower expression of several inhibitory receptors, including FCGR2B, FCRL2, FCRL3, and TNFRSF13B [160]. In BCR sequencing analyses, S1+ B cells were primarily IgG1 and IgA1 class-switched cells, with high frequencies of somatic hypermutation (SHM) in VH genes and a low clonal diversity compared to S1− B cells, indicating their antigen-driven clonal expansion and GC origin [160]. In that study, the frequencies of S1+RBD+ B cells in adenoids significantly correlated with serum neutralization titers for B.1.351 (Beta), B.1.526 (Iota), B.1.617.2 (Delta), and B.1.1.529 (Omicron) variants [160], suggesting a critical role for adenoid B cells in generating immune responses to SARS-CoV-2. Furthermore, GC B cells are expanded in adenoids after COVID-19. These results provide evidence for persistent SARS-CoV-2-specific B cell responses in pharyngeal lymphoid tissues following infection.
Tertiary lymphoid structures (TLSs) often develop at sites of inflammation and are key sites in which memory B cells are reactivated, giving rise to antibodies that are capable of mediating rapid viral clearance. However, it remains unclear whether virus-specific BRM cells arise from germinal center responses in TLSs, such as inducible bronchus-associated lymphoid tissue, located in mucosal tissues, or whether they migrate to the mucosal tissue after development in the draining lymph node. Additional studies are required to elucidate the presence and role of TLSs in the airways regarding humoral immune responses to SARS-CoV-2 and other respiratory viruses.
Similar to mucosal cellular immunity, the development of novel vaccination approaches that induce mucosal memory B cell responses is required. Studies have shown that an intranasal adenovirus-based COVID-19 vaccine induced mucosal B cells and antibodies in mice [161] and rhesus macaques [162] and was protective against upper and lower airway infections. In a hamster model, the intranasal delivery of an adenovirus-based vaccine generated a robust neutralizing antibody response and provided better protection than intramuscular delivery [163]. Additionally, Diallo et al. demonstrated that an intranasally delivered S protein trimer with adjuvant potently elicited S-specific IgG and IgA antibodies in the nasal cavity and lungs [150]. Collectively, these results indicate that vaccines capable of eliciting humoral responses in mucosal tissues may be effective strategies for inducing protective immunity.

6. Alterations in Mucosal Immune Responses against SARS-CoV-2 in the Allergic Airway Diseases

6.1. Differential Expression of Viral Entry-Related Receptors in the Allergic Airway

Factors involved in viral entry into respiratory epithelial cells can be influenced by environmental stimuli within the airway. Distinct expression patterns of these entry-related genes have been reported in allergic airway inflammation, primarily mediated by type 2 cytokines such as IL-4, 5, and 13. Several studies of SARS-CoV-2 have indicated a significant decrease in ACE2 expression and an increase in TMPRSS2 expression in airway epithelial cells in response to type 2 inflammation [164,165,166,167,168,169]. Kimura et al. found that IL-13 significantly reduced ACE2 expression and increased TMPRSS2 expression in primary BECs. Similar expression patterns for ACE2 and TMPRSS2 were observed in nasal epithelial cells and BECs in type 2 asthma and AR, which were mainly mediated by type 2 inflammation [167]. Coden et al. also reported that ACE2 was downregulated and TMPRSS2 was upregulated in BECs from patients with Th2-high asthma compared with those with Th2-low asthma [168]. In addition, pre-stimulation with IL-13 before SARS-CoV-2 infection led to a reduction in viral replication and ACE2 protein levels in BECs from children with allergic asthma compared with those from control subjects [170]. This finding suggests a protective effect of type 2 inflammation against SARS-CoV-2 entry. Consistent with these results, a previous study analyzing 900 patients infected with SARS-CoV-2 showed that patients with type 2 inflammation had lower viral loads upon entry into the study [171]. Conversely, several studies have reported that patients with COVID-19 have a lower prevalence of asthma and AR than the general population [172,173,174].
In contrast, ACE2 expression in airway epithelial cells increases in the presence of type 1 inflammation or a low Th2 profile. Saheb et al. found that an increase in IL-17, a cytokine associated with type 2-low asthma, was significantly associated with a higher ACE2 expression in BECs [175]. In an in vitro study using airway epithelial cells, Ziegler et al. discovered that ACE2 is a human IFN-stimulated gene (ISG), demonstrating that it was elevated in response to type I IFNs and, to a lesser extent, type II IFNs [166].
Collectively, these results show that the expression of SARS-CoV-2 entry-related receptors may undergo distinctive changes in the allergic airway.

6.2. Innate and Adaptive Immune Responses

In allergic airways, impaired innate immune responses to viral infections have been reported; these are associated with a biased immune response towards Th2 polarity and are primarily mediated by type 2 inflammation [176]. Type 2 inflammation is characterized by elevated eosinophils in the airways or PB and an abundance of type 2 cytokines. Gilles et al. reported that the type 2 cytokines IL-4 and IL-13 could impair the immune response to rhinovirus 16 infection by inhibiting TLR3 expression and IRF3 signaling [177]. Additionally, allergic inflammation and eosinophilia induced by IL-5 suppresses TLR7 expression in the lungs [178]. Eosinophils may suppress TLR7 responses and weaken antiviral responses; however, further investigations are required to understand the impact of TLR7 downregulation on SARS-CoV-2 infection.
The innate immune response can be modulated by allergens such as pollen and HDM. Akbarshahi et al. reported that in human BECs and a mouse model of asthma, exposure to HDM before viral infection decreased the IFN-β and IFN-λ response by affecting the TLR3 signaling pathway [179]. Pollen exposure can also affect the immune responses to viral infections. Hajighasemi et al. proposed that pollen exposure directly affects antiviral defense within the airway and enhances susceptibility to SARS-CoV-2 infection [180]. Another study reported that exposure to pollen during viral infections reduced the production of proinflammatory chemokines and type I/III IFNs, and increased viral replication [177]. Additionally, several proteases within the pollen may disrupt epithelial barrier integrity and function, similar to HDM, resulting in the increased permeability of the subepithelial layers by respiratory viruses [181].
Contrary to the previously described impaired immune response in allergic airways, some studies have suggested a protective role of allergic airway inflammation in antiviral defense. One study investigated the potential influence of AR on the immune response following vaccination with a two-dose inactivated SARS-CoV-2 vaccine. The results indicated that patients with pre-existing AR demonstrated a Tfh2 cell-associated enhanced humoral immune response to the inactivated SARS-CoV-2 vaccine compared with the control subjects. Furthermore, elevated levels of neutralizing antibodies were observed at 10–12 months post infection in the AR group compared to those in the control group [182]. Consistent with these results, Chen et al. investigated the levels of SARS-CoV-2-specific humoral and cellular immunity in patients with and without asthma, and found that the level of SARS-CoV-2-specific neutralizing antibodies was higher in COVID-19 survivors with asthma than in those without asthma at the eight-month follow-up, whereas no significant differences were noted in the cellular immunity levels between the two groups. This implies that patients with asthma may benefit from augmented humoral immunity during the recovery period from COVID-19 [183]. Additionally, a positive correlation between the levels of SARS-CoV-2-specific T cell memory responses and the blood eosinophil and regulatory T cell percentages was observed, suggesting a potential protective role of eosinophils in antiviral host defense [183]. Ferastraoaru et al. found that patients with SARS-CoV-2 infection and pre-existing eosinophilia (absolute eosinophil count ≥ 150 cells/μL) also showed a decreased likelihood of hospitalization and mortality [184]. Similarly, a retrospective cohort study on patients with SARS-CoV-2 infection reported that patients with a blood eosinophil count greater than or equal to 200 cells/μL demonstrated lower mortality regardless of the presence of asthma [185].

6.3. Susceptibility to Infection and Clinical Outcomes of COVID-19

The effect of allergic airway inflammation on susceptibility to SARS-CoV-2 infection and clinical outcomes is complicated and controversial. A study of 70,557 patients who underwent SARS-CoV-2 testing found that AR was associated with a lower risk of SARS-CoV-2 infection. Asthma exhibited a similar protective effect against SARS-CoV-2 infection in patients aged under 65 years, despite its higher risk of hospitalization than that in healthy controls [186]. Furthermore, a large retrospective cohort study of patients with SARS-CoV-2-induced pneumonia found a significant association between atopic status and mild COVID-19 [187]. In contrast, in a nationwide Korean cohort, asthma and AR were associated with an increased likelihood of SARS-CoV-2 infection and worse clinical outcomes; however, patients with non-allergic asthma had a greater risk of SARS-CoV-2 test positivity and more severe clinical outcomes than those with allergic asthma [188]. Similarly, in a propensity-score-matched nationwide cohort study, patients with AR had a higher risk of developing SARS-CoV-2 infection [189]. Certainly, it is necessary to consider the severity of comorbid diseases. A national cohort study in the UK reported an increased risk of hospitalization owing to COVID-19 among patients with severe or poorly controlled asthma compared to those without asthma. However, patients with mild or well-controlled asthma did not show a significantly increased risk of hospitalization and mortality from COVID-19 compared with those without asthma [190].

7. Concluding Remarks and Perspectives

An extensive body of research conducted during the COVID-19 pandemic has substantially advanced our understanding of immune responses against SARS-CoV-2 and respiratory viruses. However, most studies have relied on easily accessible PB. Currently, there is a growing need for more detailed investigations of the features and protective roles of mucosal immune responses. Mounting evidence from animal studies has shown that the establishment of neutralizing antibodies and immune memory in the respiratory tract leads to superior protection against invading respiratory viruses. Table 1 summarizes the key components of mucosal immune responses against SARS-CoV-2. However, the regulatory mechanisms governing the generation and maintenance of immune memory in the respiratory tract are not fully understood. In addition, the exact impact of respiratory diseases, such as allergic airway inflammation, on mucosal antiviral defense needs to be further addressed. Further mechanistic investigations coupled with data from human cohort studies will provide novel insights into the development of effective vaccines that induce greater mucosal immunity.

Author Contributions

H.-E.N. and M.-S.R. conceived and designed the study. H.-E.N. and M.-S.R. collected and analyzed the relevant reports. H.-E.N. and M.-S.R. wrote the manuscript. All authors have read and agreed to the published version of the manuscript.

Funding

This work was supported by a grant (RS-2023-00222762) from the National Research Foundation of Korea, funded by the Ministry of Science and ICT.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

No new data were created or analyzed in this study. Data sharing is not applicable to this article.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Zhou, P.; Yang, X.L.; Wang, X.G.; Hu, B.; Zhang, L.; Zhang, W.; Si, H.R.; Zhu, Y.; Li, B.; Huang, C.L.; et al. A pneumonia outbreak associated with a new coronavirus of probable bat origin. Nature 2020, 579, 270–273. [Google Scholar] [CrossRef]
  2. Huang, C.; Wang, Y.; Li, X.; Ren, L.; Zhao, J.; Hu, Y.; Zhang, L.; Fan, G.; Xu, J.; Gu, X.; et al. Clinical features of patients infected with 2019 novel coronavirus in Wuhan, China. Lancet 2020, 395, 497–506. [Google Scholar] [CrossRef]
  3. Ahn, J.H.; Kim, J.; Hong, S.P.; Choi, S.Y.; Yang, M.J.; Ju, Y.S.; Kim, Y.T.; Kim, H.M.; Rahman, M.D.T.; Chung, M.K.; et al. Nasal ciliated cells are primary targets for SARS-CoV-2 replication in the early stage of COVID-19. J. Clin. Investig. 2021, 131, e148517. [Google Scholar] [CrossRef]
  4. Chua, R.L.; Lukassen, S.; Trump, S.; Hennig, B.P.; Wendisch, D.; Pott, F.; Debnath, O.; Thurmann, L.; Kurth, F.; Volker, M.T.; et al. COVID-19 severity correlates with airway epithelium-immune cell interactions identified by single-cell analysis. Nat Biotechnol 2020, 38, 970–979. [Google Scholar] [CrossRef]
  5. Puelles, V.G.; Lutgehetmann, M.; Lindenmeyer, M.T.; Sperhake, J.P.; Wong, M.N.; Allweiss, L.; Chilla, S.; Heinemann, A.; Wanner, N.; Liu, S.; et al. Multiorgan and Renal Tropism of SARS-CoV-2. N. Engl. J. Med. 2020, 383, 590–592. [Google Scholar] [CrossRef]
  6. Wanner, N.; Andrieux, G.; Badia, I.M.P.; Edler, C.; Pfefferle, S.; Lindenmeyer, M.T.; Schmidt-Lauber, C.; Czogalla, J.; Wong, M.N.; Okabayashi, Y.; et al. Molecular consequences of SARS-CoV-2 liver tropism. Nat. Metab. 2022, 4, 310–319. [Google Scholar] [CrossRef] [PubMed]
  7. Sette, A.; Sidney, J.; Crotty, S. T Cell Responses to SARS-CoV-2. Annu. Rev. Immunol. 2023, 41, 343–373. [Google Scholar] [CrossRef]
  8. Sette, A.; Crotty, S. Adaptive immunity to SARS-CoV-2 and COVID-19. Cell 2021, 184, 861–880. [Google Scholar] [CrossRef]
  9. Hill, D.B.; Button, B.; Rubinstein, M.; Boucher, R.C. Physiology and pathophysiology of human airway mucus. Physiol. Rev. 2022, 102, 1757–1836. [Google Scholar] [CrossRef]
  10. Tsukita, S.; Tanaka, H.; Tamura, A. The Claudins: From Tight Junctions to Biological Systems. Trends Biochem. Sci. 2019, 44, 141–152. [Google Scholar] [CrossRef] [PubMed]
  11. Tsukita, S.; Furuse, M.; Itoh, M. Multifunctional strands in tight junctions. Nat. Rev. Mol. Cell Biol. 2001, 2, 285–293. [Google Scholar] [CrossRef]
  12. Rezaee, F.; Georas, S.N. Breaking Barriers New Insights into Airway Epithelial Barrier Function in Health and Disease. Am. J. Resp. Cell Mol. 2014, 50, 857–869. [Google Scholar] [CrossRef]
  13. Carlier, F.M.; de Fays, C.; Pilette, C. Epithelial Barrier Dysfunction in Chronic Respiratory Diseases. Front. Physiol. 2021, 12, 691227. [Google Scholar] [CrossRef]
  14. Chen, H.W.; Liu, P.F.; Liu, Y.T.; Kuo, S.; Zhang, X.Q.; Schooley, R.T.; Rohde, H.; Gallo, R.L.; Huang, C.M. Nasal commensal Staphylococcus epidermidis counteracts influenza virus. Sci. Rep. 2016, 6, 27870. [Google Scholar] [CrossRef]
  15. Antunes, K.H.; Fachi, J.L.; de Paula, R.; da Silva, E.F.; Pral, L.P.; dos Santos, A.A.; Dias, G.B.M.; Vargas, J.E.; Puga, R.; Mayer, F.Q.; et al. Microbiota-derived acetate protects against respiratory syncytial virus infection through a GPR43-type 1 interferon response. Nat. Commun. 2019, 10, 3273. [Google Scholar] [CrossRef]
  16. Kim, H.J.; Jo, A.; Jeon, Y.J.; An, S.; Lee, K.M.; Yoon, S.S.; Choi, J.Y. Nasal commensal Staphylococcus epidermidis enhances interferon-lambda-dependent immunity against influenza virus. Microbiome 2019, 7, 80. [Google Scholar] [CrossRef]
  17. Yeo, N.K.; Jang, Y.J. Rhinovirus infection-induced alteration of tight junction and adherens junction components in human nasal epithelial cells. Laryngoscope 2010, 120, 346–352. [Google Scholar] [CrossRef] [PubMed]
  18. Kim, K.A.; Jung, J.H.; Kang, I.G.; Choi, Y.S.; Kim, S.T. ROS Is Involved in Disruption of Tight Junctions of Human Nasal Epithelial Cells Induced by HRV16. Laryngoscope 2018, 128, E393–E401. [Google Scholar] [CrossRef] [PubMed]
  19. Looi, K.; Buckley, A.G.; Rigby, P.J.; Garratt, L.W.; Iosifidis, T.; Zosky, G.R.; Larcombe, A.N.; Lannigan, F.J.; Ling, K.M.; Martinovich, K.M.; et al. Effects of human rhinovirus on epithelial barrier integrity and function in children with asthma. Clin. Exp. Allergy 2018, 48, 513–524. [Google Scholar] [CrossRef]
  20. Xiao, C.; Puddicombe, S.M.; Field, S.; Haywood, J.; Broughton-Head, V.; Puxeddu, I.; Haitchi, H.M.; Vernon-Wilson, E.; Sammut, D.; Bedke, N.; et al. Defective epithelial barrier function in asthma. J. Allergy Clin. Immunol. 2011, 128, 549–556.e12. [Google Scholar] [CrossRef] [PubMed]
  21. Murphy, J.; Ramezanpour, M.; Roscioli, E.; Psaltis, A.J.; Wormald, P.J.; Vreugde, S. Mucosal zinc deficiency in chronic rhinosinusitis with nasal polyposis contributes to barrier disruption and decreases ZO-1. Allergy 2018, 73, 2095–2097. [Google Scholar] [CrossRef]
  22. Steelant, B.; Farré, R.; Wawrzyniak, P.; Belmans, J.; Dekimpe, E.; Vanheel, H.; Van Gerven, L.; Krohn, I.K.; Bullens, D.M.A.; Ceuppens, J.L.; et al. Impaired barrier function in patients with house dust mite-induced allergic rhinitis is accompanied by decreased occludin and zonula occludens-1 expression. J. Allergy Clin. Immun. 2016, 137, 1043–1053.e5. [Google Scholar] [CrossRef]
  23. Wang, J.; Kang, X.; Huang, Z.Q.; Shen, L.; Luo, Q.; Li, M.Y.; Luo, L.P.; Tu, J.H.; Han, M.; Ye, J. Protease-Activated Receptor-2 Decreased Zonula Occlidens-1 and Claudin-1 Expression and Induced Epithelial Barrier Dysfunction in Allergic Rhinitis. Am. J. Rhinol. Allergy 2021, 35, 26–35. [Google Scholar] [CrossRef]
  24. Zheng, J.; Zeng, M.; Nian, J.B.; Zeng, L.Y.; Fu, Z.; Huang, Q.J.; Wei, X. The CXCR4/miR-125b/FoxP3 axis regulates the function of the epithelial barrier via autophagy in allergic rhinitis. Am. J. Transl. Res. 2020, 12, 2570–2584. [Google Scholar]
  25. Chatterjee, M.; van Putten, J.P.M.; Strijbis, K. Defensive Properties of Mucin Glycoproteins during Respiratory Infections-Relevance for SARS-CoV-2. mBio 2020, 11, e02374-20. [Google Scholar] [CrossRef]
  26. Baindara, P.; Ganguli, S.; Chakraborty, R.; Mandal, S.M. Preventing Respiratory Viral Diseases with Antimicrobial Peptide Master Regulators in the Lung Airway Habitat. Clin. Pract. 2023, 13, 125–147. [Google Scholar] [CrossRef] [PubMed]
  27. Medzhitov, R.; Janeway, C.A., Jr. Innate immunity: The virtues of a nonclonal system of recognition. Cell 1997, 91, 295–298. [Google Scholar] [CrossRef] [PubMed]
  28. Durbin, R.K.; Kotenko, S.V.; Durbin, J.E. Interferon induction and function at the mucosal surface. Immunol. Rev. 2013, 255, 25–39. [Google Scholar] [CrossRef] [PubMed]
  29. Newton, A.H.; Cardani, A.; Braciale, T.J. The host immune response in respiratory virus infection: Balancing virus clearance and immunopathology. Semin. Immunopathol. 2016, 38, 471–482. [Google Scholar] [CrossRef] [PubMed]
  30. Dalskov, L.; Gad, H.H.; Hartmann, R. Viral recognition and the antiviral interferon response. EMBO J. 2023, 42, e112907. [Google Scholar] [CrossRef] [PubMed]
  31. Platanias, L.C. Mechanisms of type-I- and type-II-interferon-mediated signalling. Nat. Rev. Immunol. 2005, 5, 375–386. [Google Scholar] [CrossRef] [PubMed]
  32. Schoggins, J.W.; Wilson, S.J.; Panis, M.; Murphy, M.Y.; Jones, C.T.; Bieniasz, P.; Rice, C.M. A diverse range of gene products are effectors of the type I interferon antiviral response. Nature 2011, 472, 481–485. [Google Scholar] [CrossRef] [PubMed]
  33. Stanifer, M.L.; Guo, C.C.; Doldan, P.; Boulant, S. Importance of Type I and III Interferons at Respiratory and Intestinal Barrier Surfaces. Front. Immunol. 2020, 11, 608645. [Google Scholar] [CrossRef] [PubMed]
  34. Mordstein, M.; Neugebauer, E.; Ditt, V.; Jessen, B.; Rieger, T.; Falcone, V.; Sorgeloos, F.; Ehl, S.; Mayer, D.; Kochs, G.; et al. Lambda Interferon Renders Epithelial Cells of the Respiratory and Gastrointestinal Tracts Resistant to Viral Infections. J. Virol. 2010, 84, 5670–5677. [Google Scholar] [CrossRef]
  35. Sommereyns, C.; Paul, S.; Staeheli, P.; Michiels, T. IFN-lambda (IFN-λ) is expressed in a tissue-dependent fashion and primarily acts on epithelial cells in vivo. PLoS Pathog. 2008, 4, e1000017. [Google Scholar] [CrossRef] [PubMed]
  36. Barchet, W.; Cella, M.; Odermatt, B.; Asselin-Paturel, C.; Colonna, M.; Kalinke, U. Virus-induced interferon alpha production by a dendritic cell subset in the absence of feedback signaling in vivo. J. Exp. Med. 2002, 195, 507–516. [Google Scholar] [CrossRef]
  37. Jewell, N.A.; Cline, T.; Mertz, S.E.; Smirnov, S.V.; Flaño, E.; Schindler, C.; Grieves, J.L.; Durbin, R.K.; Kotenko, S.V.; Durbin, J.E. Lambda Interferon Is the Predominant Interferon Induced by Influenza A Virus Infection. J. Virol. 2011, 85, 650. [Google Scholar] [CrossRef]
  38. Fox, J.M.; Crabtree, J.M.; Sage, L.K.; Tompkins, S.M.; Tripp, R.A. Interferon Lambda Upregulates IDO1 Expression in Respiratory Epithelial Cells After Influenza Virus Infection. J. Interferon. Cytokine Res. 2015, 35, 554–562. [Google Scholar] [CrossRef]
  39. Wang, X.; Guan, F.; Miller, H.; Byazrova, M.G.; Cndotti, F.; Benlagha, K.; Camara, N.O.S.; Lei, J.; Filatov, A.; Liu, C. The role of dendritic cells in COVID-19 infection. Emerg. Microbes Infect. 2023, 12, 2195019. [Google Scholar] [CrossRef]
  40. Collin, M.; Bigley, V. Human dendritic cell subsets: An update. Immunology 2018, 154, 3–20. [Google Scholar] [CrossRef]
  41. Rydyznski Moderbacher, C.; Ramirez, S.I.; Dan, J.M.; Grifoni, A.; Hastie, K.M.; Weiskopf, D.; Belanger, S.; Abbott, R.K.; Kim, C.; Choi, J.; et al. Antigen-Specific Adaptive Immunity to SARS-CoV-2 in Acute COVID-19 and Associations with Age and Disease Severity. Cell 2020, 183, 996–1012.e19. [Google Scholar] [CrossRef]
  42. Rha, M.S.; Kim, A.R.; Shin, E.C. SARS-CoV-2-Specific T Cell Responses in Patients with COVID-19 and Unexposed Individuals. Immune Netw. 2021, 21, e2. [Google Scholar] [CrossRef]
  43. Dan, J.M.; Mateus, J.; Kato, Y.; Hastie, K.M.; Yu, E.D.; Faliti, C.E.; Grifoni, A.; Ramirez, S.I.; Haupt, S.; Frazier, A.; et al. Immunological memory to SARS-CoV-2 assessed for up to 8 months after infection. Science 2021, 371, eabf4063. [Google Scholar] [CrossRef]
  44. Jung, J.H.; Rha, M.S.; Sa, M.; Choi, H.K.; Jeon, J.H.; Seok, H.; Park, D.W.; Park, S.H.; Jeong, H.W.; Choi, W.S.; et al. SARS-CoV-2-specific T cell memory is sustained in COVID-19 convalescent patients for 10 months with successful development of stem cell-like memory T cells. Nat. Commun. 2021, 12, 4043. [Google Scholar] [CrossRef] [PubMed]
  45. Wherry, E.J.; Barouch, D.H. T cell immunity to COVID-19 vaccines. Science 2022, 377, 821–822. [Google Scholar] [CrossRef] [PubMed]
  46. Mueller, S.N.; Mackay, L.K. Tissue-resident memory T cells: Local specialists in immune defence. Nat. Rev. Immunol. 2016, 16, 79–89. [Google Scholar] [CrossRef] [PubMed]
  47. Szabo, P.A.; Miron, M.; Farber, D.L. Location, location, location: Tissue resident memory T cells in mice and humans. Sci. Immunol. 2019, 4, eaas9673. [Google Scholar] [CrossRef] [PubMed]
  48. Mackay, L.K.; Rahimpour, A.; Ma, J.Z.; Collins, N.; Stock, A.T.; Hafon, M.L.; Vega-Ramos, J.; Lauzurica, P.; Mueller, S.N.; Stefanovic, T.; et al. The developmental pathway for CD103(+)CD8+ tissue-resident memory T cells of skin. Nat. Immunol. 2013, 14, 1294–1301. [Google Scholar] [CrossRef] [PubMed]
  49. Hombrink, P.; Helbig, C.; Backer, R.A.; Piet, B.; Oja, A.E.; Stark, R.; Brasser, G.; Jongejan, A.; Jonkers, R.E.; Nota, B.; et al. Programs for the persistence, vigilance and control of human CD8(+) lung-resident memory T cells. Nat. Immunol. 2016, 17, 1467–1478. [Google Scholar] [CrossRef] [PubMed]
  50. Pizzolla, A.; Nguyen, T.H.; Sant, S.; Jaffar, J.; Loudovaris, T.; Mannering, S.I.; Thomas, P.G.; Westall, G.P.; Kedzierska, K.; Wakim, L.M. Influenza-specific lung-resident memory T cells are proliferative and polyfunctional and maintain diverse TCR profiles. J. Clin. Invest. 2018, 128, 721–733. [Google Scholar] [CrossRef] [PubMed]
  51. Bartolome-Casado, R.; Landsverk, O.J.B.; Chauhan, S.K.; Richter, L.; Phung, D.; Greiff, V.; Risnes, L.F.; Yao, Y.; Neumann, R.S.; Yaqub, S.; et al. Resident memory CD8 T cells persist for years in human small intestine. J. Exp. Med. 2019, 216, 2412–2426. [Google Scholar] [CrossRef]
  52. Smolders, J.; Heutinck, K.M.; Fransen, N.L.; Remmerswaal, E.B.M.; Hombrink, P.; Ten Berge, I.J.M.; van Lier, R.A.W.; Huitinga, I.; Hamann, J. Tissue-resident memory T cells populate the human brain. Nat. Commun. 2018, 9, 4593. [Google Scholar] [CrossRef]
  53. Urban, S.L.; Jensen, I.J.; Shan, Q.; Pewe, L.L.; Xue, H.H.; Badovinac, V.P.; Harty, J.T. Peripherally induced brain tissue-resident memory CD8(+) T cells mediate protection against CNS infection. Nat. Immunol. 2020, 21, 938–949. [Google Scholar] [CrossRef]
  54. Woodward Davis, A.S.; Vick, S.C.; Pattacini, L.; Voillet, V.; Hughes, S.M.; Lentz, G.M.; Kirby, A.C.; Fialkow, M.F.; Gottardo, R.; Hladik, F.; et al. The human memory T cell compartment changes across tissues of the female reproductive tract. Mucosal. Immunol. 2021, 14, 862–872. [Google Scholar] [CrossRef] [PubMed]
  55. Shiow, L.R.; Rosen, D.B.; Brdickova, N.; Xu, Y.; An, J.; Lanier, L.L.; Cyster, J.G.; Matloubian, M. CD69 acts downstream of interferon-alpha/beta to inhibit S1P1 and lymphocyte egress from lymphoid organs. Nature 2006, 440, 540–544. [Google Scholar] [CrossRef]
  56. Pallett, L.J.; Burton, A.R.; Amin, O.E.; Rodriguez-Tajes, S.; Patel, A.A.; Zakeri, N.; Jeffery-Smith, A.; Swadling, L.; Schmidt, N.M.; Baiges, A.; et al. Longevity and replenishment of human liver-resident memory T cells and mononuclear phagocytes. J. Exp. Med. 2020, 217, e20200050. [Google Scholar] [CrossRef]
  57. Kim, J.H.; Han, J.W.; Choi, Y.J.; Rha, M.S.; Koh, J.Y.; Kim, K.H.; Kim, C.G.; Lee, Y.J.; Kim, A.R.; Park, J.; et al. Functions of human liver CD69(+)CD103(−)CD8(+) T cells depend on HIF-2alpha activity in healthy and pathologic livers. J. Hepatol. 2020, 72, 1170–1181. [Google Scholar] [CrossRef]
  58. Buggert, M.; Nguyen, S.; Salgado-Montes de Oca, G.; Bengsch, B.; Darko, S.; Ransier, A.; Roberts, E.R.; Del Alcazar, D.; Brody, I.B.; Vella, L.A.; et al. Identification and characterization of HIV-specific resident memory CD8(+) T cells in human lymphoid tissue. Sci. Immunol. 2018, 3, eaar4526. [Google Scholar] [CrossRef] [PubMed]
  59. Oja, A.E.; Piet, B.; Helbig, C.; Stark, R.; van der Zwan, D.; Blaauwgeers, H.; Remmerswaal, E.B.M.; Amsen, D.; Jonkers, R.E.; Moerland, P.D.; et al. Trigger-happy resident memory CD4(+) T cells inhabit the human lungs. Mucosal. Immunol. 2018, 11, 654–667. [Google Scholar] [CrossRef]
  60. Kumar, B.V.; Ma, W.; Miron, M.; Granot, T.; Guyer, R.S.; Carpenter, D.J.; Senda, T.; Sun, X.; Ho, S.H.; Lerner, H.; et al. Human Tissue-Resident Memory T Cells Are Defined by Core Transcriptional and Functional Signatures in Lymphoid and Mucosal Sites. Cell Rep. 2017, 20, 2921–2934. [Google Scholar] [CrossRef] [PubMed]
  61. Zitti, B.; Hoffer, E.; Zheng, W.; Pandey, R.V.; Schlums, H.; Perinetti Casoni, G.; Fusi, I.; Nguyen, L.; Karner, J.; Kokkinou, E.; et al. Human skin-resident CD8(+) T cells require RUNX2 and RUNX3 for induction of cytotoxicity and expression of the integrin CD49a. Immunity 2023, 56, 1285–1302.e7. [Google Scholar] [CrossRef] [PubMed]
  62. Ray, S.J.; Franki, S.N.; Pierce, R.H.; Dimitrova, S.; Koteliansky, V.; Sprague, A.G.; Doherty, P.C.; de Fougerolles, A.R.; Topham, D.J. The collagen binding alpha1beta1 integrin VLA-1 regulates CD8 T cell-mediated immune protection against heterologous influenza infection. Immunity 2004, 20, 167–179. [Google Scholar] [CrossRef]
  63. Snyder, M.E.; Finlayson, M.O.; Connors, T.J.; Dogra, P.; Senda, T.; Bush, E.; Carpenter, D.; Marboe, C.; Benvenuto, L.; Shah, L.; et al. Generation and persistence of human tissue-resident memory T cells in lung transplantation. Sci. Immunol. 2019, 4, eaav5581. [Google Scholar] [CrossRef]
  64. Reilly, E.C.; Lambert Emo, K.; Buckley, P.M.; Reilly, N.S.; Smith, I.; Chaves, F.A.; Yang, H.; Oakes, P.W.; Topham, D.J. T(RM) integrins CD103 and CD49a differentially support adherence and motility after resolution of influenza virus infection. Proc. Natl. Acad. Sci. USA 2020, 117, 12306–12314. [Google Scholar] [CrossRef]
  65. Wein, A.N.; McMaster, S.R.; Takamura, S.; Dunbar, P.R.; Cartwright, E.K.; Hayward, S.L.; McManus, D.T.; Shimaoka, T.; Ueha, S.; Tsukui, T.; et al. CXCR6 regulates localization of tissue-resident memory CD8 T cells to the airways. J. Exp. Med. 2019, 216, 2748–2762. [Google Scholar] [CrossRef]
  66. Slutter, B.; Pewe, L.L.; Kaech, S.M.; Harty, J.T. Lung airway-surveilling CXCR3(hi) memory CD8(+) T cells are critical for protection against influenza A virus. Immunity 2013, 39, 939–948. [Google Scholar] [CrossRef]
  67. Jozwik, A.; Habibi, M.S.; Paras, A.; Zhu, J.; Guvenel, A.; Dhariwal, J.; Almond, M.; Wong, E.H.C.; Sykes, A.; Maybeno, M.; et al. RSV-specific airway resident memory CD8+ T cells and differential disease severity after experimental human infection. Nat. Commun. 2015, 6, 10224. [Google Scholar] [CrossRef]
  68. Kinnear, E.; Lambert, L.; McDonald, J.U.; Cheeseman, H.M.; Caproni, L.J.; Tregoning, J.S. Airway T cells protect against RSV infection in the absence of antibody. Mucosal. Immunol. 2018, 11, 249–256. [Google Scholar] [CrossRef]
  69. Zhao, J.; Zhao, J.; Mangalam, A.K.; Channappanavar, R.; Fett, C.; Meyerholz, D.K.; Agnihothram, S.; Baric, R.S.; David, C.S.; Perlman, S. Airway Memory CD4(+) T Cells Mediate Protective Immunity against Emerging Respiratory Coronaviruses. Immunity 2016, 44, 1379–1391. [Google Scholar] [CrossRef]
  70. Teijaro, J.R.; Turner, D.; Pham, Q.; Wherry, E.J.; Lefrancois, L.; Farber, D.L. Cutting edge: Tissue-retentive lung memory CD4 T cells mediate optimal protection to respiratory virus infection. J. Immunol. 2011, 187, 5510–5514. [Google Scholar] [CrossRef]
  71. Wu, T.; Hu, Y.; Lee, Y.T.; Bouchard, K.R.; Benechet, A.; Khanna, K.; Cauley, L.S. Lung-resident memory CD8 T cells (TRM) are indispensable for optimal cross-protection against pulmonary virus infection. J. Leukoc. Biol. 2014, 95, 215–224. [Google Scholar] [CrossRef]
  72. Pizzolla, A.; Nguyen, T.H.O.; Smith, J.M.; Brooks, A.G.; Kedzieska, K.; Heath, W.R.; Reading, P.C.; Wakim, L.M. Resident memory CD8(+) T cells in the upper respiratory tract prevent pulmonary influenza virus infection. Sci. Immunol. 2017, 2, eaam6970. [Google Scholar] [CrossRef]
  73. Laidlaw, B.J.; Ellebedy, A.H. The germinal centre B cell response to SARS-CoV-2. Nat. Rev. Immunol. 2022, 22, 7–18. [Google Scholar] [CrossRef] [PubMed]
  74. Inoue, T.; Kurosaki, T. Memory B cells. Nat. Rev. Immunol. 2023, 24, 5–17. [Google Scholar] [CrossRef]
  75. Nutt, S.L.; Hodgkin, P.D.; Tarlinton, D.M.; Corcoran, L.M. The generation of antibody-secreting plasma cells. Nat. Rev. Immunol. 2015, 15, 160–171. [Google Scholar] [CrossRef] [PubMed]
  76. Allie, S.R.; Bradley, J.E.; Mudunuru, U.; Schultz, M.D.; Graf, B.A.; Lund, F.E.; Randall, T.D. The establishment of resident memory B cells in the lung requires local antigen encounter. Nat. Immunol. 2019, 20, 97–108. [Google Scholar] [CrossRef] [PubMed]
  77. Oh, J.E.; Song, E.; Moriyama, M.; Wong, P.; Zhang, S.; Jiang, R.; Strohmeier, S.; Kleinstein, S.H.; Krammer, F.; Iwasaki, A. Intranasal priming induces local lung-resident B cell populations that secrete protective mucosal antiviral IgA. Sci. Immunol. 2021, 6, eabj5129. [Google Scholar] [CrossRef] [PubMed]
  78. Barker, K.A.; Etesami, N.S.; Shenoy, A.T.; Arafa, E.I.; Lyon de Ana, C.; Smith, N.M.; Martin, I.M.; Goltry, W.N.; Barron, A.M.; Browning, J.L.; et al. Lung-resident memory B cells protect against bacterial pneumonia. J. Clin. Investig. 2021, 131, e141810. [Google Scholar] [CrossRef]
  79. Tan, H.X.; Juno, J.A.; Esterbauer, R.; Kelly, H.G.; Wragg, K.M.; Konstandopoulos, P.; Alcantara, S.; Alvarado, C.; Jones, R.; Starkey, G.; et al. Lung-resident memory B cells established after pulmonary influenza infection display distinct transcriptional and phenotypic profiles. Sci. Immunol. 2022, 7, eabf5314. [Google Scholar] [CrossRef]
  80. Weisel, N.M.; Weisel, F.J.; Farber, D.L.; Borghesi, L.A.; Shen, Y.; Ma, W.; Luning Prak, E.T.; Shlomchik, M.J. Comprehensive analyses of B-cell compartments across the human body reveal novel subsets and a gut-resident memory phenotype. Blood 2020, 136, 2774–2785. [Google Scholar] [CrossRef]
  81. Mathew, N.R.; Jayanthan, J.K.; Smirnov, I.V.; Robinson, J.L.; Axelsson, H.; Nakka, S.S.; Emmanouilidi, A.; Czarnewski, P.; Yewdell, W.T.; Schon, K.; et al. Single-cell BCR and transcriptome analysis after influenza infection reveals spatiotemporal dynamics of antigen-specific B cells. Cell Rep. 2021, 35, 109286. [Google Scholar] [CrossRef]
  82. Tan, H.X.; Esterbauer, R.; Vanderven, H.A.; Juno, J.A.; Kent, S.J.; Wheatley, A.K. Inducible Bronchus-Associated Lymphoid Tissues (iBALT) Serve as Sites of B Cell Selection and Maturation Following Influenza Infection in Mice. Front. Immunol. 2019, 10, 611. [Google Scholar] [CrossRef]
  83. Gregoire, C.; Spinelli, L.; Villazala-Merino, S.; Gil, L.; Holgado, M.P.; Moussa, M.; Dong, C.; Zarubica, A.; Fallet, M.; Navarro, J.M.; et al. Viral infection engenders bona fide and bystander subsets of lung-resident memory B cells through a permissive mechanism. Immunity 2022, 55, 1216–1233.e19. [Google Scholar] [CrossRef]
  84. MacLean, A.J.; Richmond, N.; Koneva, L.; Attar, M.; Medina, C.A.P.; Thornton, E.E.; Gomes, A.C.; El-Turabi, A.; Bachmann, M.F.; Rijal, P.; et al. Secondary influenza challenge triggers resident memory B cell migration and rapid relocation to boost antibody secretion at infected sites. Immunity 2022, 55, 718–733.e8. [Google Scholar] [CrossRef]
  85. Wang, M.Y.; Zhao, R.; Gao, L.J.; Gao, X.F.; Wang, D.P.; Cao, J.M. SARS-CoV-2: Structure, Biology, and Structure-Based Therapeutics Development. Front. Cell Infect. Mi. 2020, 10, 587269. [Google Scholar] [CrossRef]
  86. Belouzard, S.; Chu, V.C.; Whittaker, G.R. Activation of the SARS coronavirus spike protein via sequential proteolytic cleavage at two distinct sites. Proc. Natl. Acad. Sci. USA 2009, 106, 5871–5876. [Google Scholar] [CrossRef]
  87. Shang, J.; Wan, Y.; Luo, C.; Ye, G.; Geng, Q.; Auerbach, A.; Li, F. Cell entry mechanisms of SARS-CoV-2. Proc. Natl. Acad. Sci. USA 2020, 117, 11727–11734. [Google Scholar] [CrossRef] [PubMed]
  88. Jackson, C.B.; Farzan, M.; Chen, B.; Choe, H. Mechanisms of SARS-CoV-2 entry into cells. Nat. Rev. Mol. Cell Bio. 2022, 23, 3–20. [Google Scholar] [CrossRef]
  89. Matsuyama, S.; Nagata, N.; Shirato, K.; Kawase, M.; Takeda, M.; Taguchi, F. Efficient Activation of the Severe Acute Respiratory Syndrome Coronavirus Spike Protein by the Transmembrane Protease TMPRSS2. J. Virol. 2010, 84, 12658–12664. [Google Scholar] [CrossRef] [PubMed]
  90. Iwata-Yoshikawa, N.; Okamura, T.; Shimizu, Y.; Hasegawa, H.; Takeda, M.; Nagata, N. TMPRSS2 Contributes to Virus Spread and Immunopathology in the Airways of Murine Models after Coronavirus Infection. J. Virol. 2019, 93, e01815-18. [Google Scholar] [CrossRef] [PubMed]
  91. Takeda, M. Protease-dependent virus tropism and pathogenicity: The role for TMPRSS2. Uirusu 2019, 69, 61–72. [Google Scholar] [CrossRef] [PubMed]
  92. Matsuyama, S.; Ujike, M.; Morikawa, S.; Tashiro, M.; Taguchi, F. Protease-mediated enhancement of severe acute respiratory syndrome coronavirus infection. Proc. Natl. Acad. Sci. USA 2005, 102, 12543–12547. [Google Scholar] [CrossRef] [PubMed]
  93. Zhao, M.M.; Yang, W.L.; Yang, F.Y.; Zhang, L.; Huang, W.J.; Hou, W.; Fan, C.F.; Jin, R.H.; Feng, Y.M.; Wang, Y.C.; et al. Cathepsin L plays a key role in SARS-CoV-2 infection in humans and humanized mice and is a promising target for new drug development. Signal Transduct. Target. Ther. 2021, 6, 134. [Google Scholar] [CrossRef] [PubMed]
  94. Thiel, V.; Ivanov, K.A.; Putics, A.; Hertzig, T.; Schelle, B.; Bayer, S.; Weissbrich, B.; Snijder, E.J.; Rabenau, H.; Doerr, H.W.; et al. Mechanisms and enzymes involved in SARS coronavirus genome expression. J. Gen. Virol. 2003, 84, 2305–2315. [Google Scholar] [CrossRef] [PubMed]
  95. Yadav, R.; Chaudhary, J.K.; Jain, N.; Chaudhary, P.K.; Khanra, S.; Dhamija, P.; Sharma, A.; Kumar, A.; Handu, S. Role of Structural and Non-Structural Proteins and Therapeutic Targets of SARS-CoV-2 for COVID-19. Cells 2021, 10, 821. [Google Scholar] [CrossRef]
  96. Diamond, M.S.; Kanneganti, T.D. Innate immunity: The first line of defense against SARS-CoV-2. Nat. Immunol. 2022, 23, 165–176. [Google Scholar] [CrossRef]
  97. Camiolo, M.; Gauthier, M.; Kaminski, N.; Ray, A.; Wenzel, S.E. Expression of SARS-CoV-2 receptor ACE2 and coincident host response signature varies by asthma inflammatory phenotype. J. Allergy Clin. Immunol. 2020, 146, 315–324.e7. [Google Scholar] [CrossRef]
  98. Gheware, A.; Ray, A.; Rana, D.; Bajpai, P.; Nambirajan, A.; Arulselvi, S.; Mathur, P.; Trikha, A.; Arava, S.; Das, P.; et al. ACE2 protein expression in lung tissues of severe COVID-19 infection. Sci. Rep. 2022, 12, 4058. [Google Scholar] [CrossRef]
  99. Sungnak, W.; Huang, N.; Bécavin, C.; Berg, M.; Queen, R.; Litvinukova, M.; Talavera-López, C.; Maatz, H.; Reichart, D.; Sampaziotis, F.; et al. SARS-CoV-2 entry factors are highly expressed in nasal epithelial cells together with innate immune genes. Nat. Med. 2020, 26, 681–687. [Google Scholar] [CrossRef] [PubMed]
  100. Robinot, R.; Hubert, M.; de Melo, G.D.; Lazarini, F.; Bruel, T.; Smith, N.; Levallois, S.; Larrous, F.; Fernandes, J.; Gellenoncourt, S.; et al. SARS-CoV-2 infection induces the dedifferentiation of multiciliated cells and impairs mucociliary clearance. Nat. Commun. 2021, 12, 4354. [Google Scholar] [CrossRef] [PubMed]
  101. Bastard, P.; Rosen, L.B.; Zhang, Q.; Michailidis, E.; Hoffmann, H.H.; Zhang, Y.; Dorgham, K.; Philippot, Q.; Rosain, J.; Beziat, V.; et al. Autoantibodies against type I IFNs in patients with life-threatening COVID-19. Science 2020, 370, eabd4585. [Google Scholar] [CrossRef] [PubMed]
  102. Zhang, Q.; Bastard, P.; Liu, Z.; Le Pen, J.; Moncada-Velez, M.; Chen, J.; Ogishi, M.; Sabli, I.K.D.; Hodeib, S.; Korol, C.; et al. Inborn errors of type I IFN immunity in patients with life-threatening COVID-19. Science 2020, 370, eabd4570. [Google Scholar] [CrossRef] [PubMed]
  103. Lu, W.J.; Liu, X.Q.; Wang, T.; Liu, F.; Zhu, A.R.; Lin, Y.P.; Luo, J.P.; Ye, F.; He, J.X.; Zhao, J.C.; et al. Elevated MUC1 and MUC5AC mucin protein levels in airway mucus of critical ill COVID-19 patients. J. Med. Virol. 2021, 93, 582–584. [Google Scholar] [CrossRef] [PubMed]
  104. Morrison, C.B.; Edwards, C.E.; Shaffer, K.M.; Araba, K.C.; Wykoff, J.A.; Williams, D.R.; Asakura, T.; Dang, H.; Morton, L.C.; Gilmore, R.C.; et al. SARS-CoV-2 infection of airway cells causes intense viral and cell shedding, two spreading mechanisms affected by IL-13. Proc. Natl. Acad. Sci. USA 2022, 119, e2119680119. [Google Scholar] [CrossRef] [PubMed]
  105. Kato, T.; Asakura, T.; Edwards, C.E.; Dang, H.; Mikami, Y.; Okuda, K.; Chen, G.; Sun, L.; Gilmore, R.C.; Hawkins, P.; et al. Prevalence and Mechanisms of Mucus Accumulation in COVID-19 Lung Disease. Am. J. Resp. Crit Care 2022, 206, 1336–1352. [Google Scholar] [CrossRef] [PubMed]
  106. Chatterjee, M.; Huang, L.Z.X.; Mykytyn, A.Z.; Wang, C.Y.; Lamers, M.M.; Westendorp, B.; Wubbolts, R.W.; van Putten, J.P.M.; Bosch, B.J.; Haagmans, B.L.; et al. Glycosylated extracellular mucin domains protect against SARS-CoV-2 infection at the respiratory surface. PLoS Pathog. 2023, 19, e1011571. [Google Scholar] [CrossRef]
  107. Biering, S.B.; Sarnik, S.A.; Wang, E.; Zengel, J.R.; Leist, S.R.; Schäfer, A.; Sathyan, V.; Hawkins, P.; Okuda, K.; Tau, C.; et al. Genome-wide bidirectional CRISPR screens identify mucins as host factors modulating SARS-CoV-2 infection. Nat. Genet. 2022, 54, 1078–1089. [Google Scholar] [CrossRef]
  108. Gilbert, C.; Lefeuvre, C.; Preisser, L.; Pivert, A.; Soleti, R.; Blanchard, S.; Delneste, Y.; Ducancelle, A.; Couez, D.; Jeannin, P. Age-Related Expression of IFN-lambda1 Versus IFN-I and Beta-Defensins in the Nasopharynx of SARS-CoV-2-Infected Individuals. Front. Immunol. 2021, 12, 750279. [Google Scholar] [CrossRef]
  109. Xu, C.; Wang, A.; Marin, M.; Honnen, W.; Ramasamy, S.; Porter, E.; Subbian, S.; Pinter, A.; Melikyan, G.B.; Lu, W.; et al. Human Defensins Inhibit SARS-CoV-2 Infection by Blocking Viral Entry. Viruses 2021, 13, 1246. [Google Scholar] [CrossRef]
  110. Bortolotti, D.; Gentili, V.; Rizzo, S.; Schiuma, G.; Beltrami, S.; Strazzabosco, G.; Fernandez, M.; Caccuri, F.; Caruso, A.; Rizzo, R. TLR3 and TLR7 RNA Sensor Activation during SARS-CoV-2 Infection. Microorganisms 2021, 9, 1820. [Google Scholar] [CrossRef] [PubMed]
  111. Yamada, T.; Sato, S.; Sotoyama, Y.; Orba, Y.; Sawa, H.; Yamauchi, H.; Sasaki, M.; Takaoka, A. RIG-I triggers a signaling-abortive anti-SARS-CoV-2 defense in human lung cells. Nat. Immunol. 2021, 22, 820–828. [Google Scholar] [CrossRef]
  112. Thorne, L.G.; Reuschl, A.K.; Zuliani-Alvarez, L.; Whelan, M.V.X.; Turner, J.; Noursadeghi, M.; Jolly, C.; Towers, G.J. SARS-CoV-2 sensing by RIG-I and MDA5 links epithelial infection to macrophage inflammation. Embo J. 2021, 40, e107826. [Google Scholar] [CrossRef]
  113. Lowery, S.A.; Sariol, A.; Perlman, S. Innate immune and inflammatory responses to SARS-CoV-2: Implications for COVID-19. Cell Host. Microbe 2021, 29, 1052–1062. [Google Scholar] [CrossRef]
  114. Siu, K.L.; Yuen, K.S.; Castano-Rodriguez, C.; Ye, Z.W.; Yeung, M.L.; Fung, S.Y.; Yuan, S.; Chan, C.P.; Yuen, K.Y.; Enjuanes, L.; et al. Severe acute respiratory syndrome coronavirus ORF3a protein activates the NLRP3 inflammasome by promoting TRAF3-dependent ubiquitination of ASC. FASEB J. 2019, 33, 8865–8877. [Google Scholar] [CrossRef] [PubMed]
  115. Xu, H.; Akinyemi, I.A.; Chitre, S.A.; Loeb, J.C.; Lednicky, J.A.; McIntosh, M.T.; Bhaduri-McIntosh, S. SARS-CoV-2 viroporin encoded by ORF3a triggers the NLRP3 inflammatory pathway. Virology 2022, 568, 13–22. [Google Scholar] [CrossRef]
  116. Han, L.; Zheng, Y.; Deng, J.; Nan, M.L.; Xiao, Y.; Zhuang, M.W.; Zhang, J.; Wang, W.; Gao, C.; Wang, P.H. SARS-CoV-2 ORF10 antagonizes STING-dependent interferon activation and autophagy. J. Med. Virol. 2022, 94, 5174–5188. [Google Scholar] [CrossRef]
  117. Loske, J.; Rohmel, J.; Lukassen, S.; Stricker, S.; Magalhaes, V.G.; Liebig, J.; Chua, R.L.; Thurmann, L.; Messingschlager, M.; Seegebarth, A.; et al. Pre-activated antiviral innate immunity in the upper airways controls early SARS-CoV-2 infection in children. Nat. Biotechnol. 2022, 40, 319–324. [Google Scholar] [CrossRef] [PubMed]
  118. Xia, H.; Cao, Z.; Xie, X.; Zhang, X.; Chen, J.Y.; Wang, H.; Menachery, V.D.; Rajsbaum, R.; Shi, P.Y. Evasion of Type I Interferon by SARS-CoV-2. Cell Rep. 2020, 33, 108234. [Google Scholar] [CrossRef] [PubMed]
  119. Lei, X.; Dong, X.; Ma, R.; Wang, W.; Xiao, X.; Tian, Z.; Wang, C.; Wang, Y.; Li, L.; Ren, L.; et al. Activation and evasion of type I interferon responses by SARS-CoV-2. Nat. Commun. 2020, 11, 3810. [Google Scholar] [CrossRef] [PubMed]
  120. Blanco-Melo, D.; Nilsson-Payant, B.E.; Liu, W.C.; Uhl, S.; Hoagland, D.; Moller, R.; Jordan, T.X.; Oishi, K.; Panis, M.; Sachs, D.; et al. Imbalanced Host Response to SARS-CoV-2 Drives Development of COVID-19. Cell 2020, 181, 1036–1045.e9. [Google Scholar] [CrossRef]
  121. Hadjadj, J.; Yatim, N.; Barnabei, L.; Corneau, A.; Boussier, J.; Smith, N.; Pere, H.; Charbit, B.; Bondet, V.; Chenevier-Gobeaux, C.; et al. Impaired type I interferon activity and inflammatory responses in severe COVID-19 patients. Science 2020, 369, 718–724. [Google Scholar] [CrossRef]
  122. Thoms, M.; Buschauer, R.; Ameismeier, M.; Koepke, L.; Denk, T.; Hirschenberger, M.; Kratzat, H.; Hayn, M.; Mackens-Kiani, T.; Cheng, J.; et al. Structural basis for translational shutdown and immune evasion by the Nsp1 protein of SARS-CoV-2. Science 2020, 369, 1249–1255. [Google Scholar] [CrossRef] [PubMed]
  123. Schubert, K.; Karousis, E.D.; Jomaa, A.; Scaiola, A.; Echeverria, B.; Gurzeler, L.A.; Leibundgut, M.; Thiel, V.; Muhlemann, O.; Ban, N. SARS-CoV-2 Nsp1 binds the ribosomal mRNA channel to inhibit translation. Nat. Struct. Mol. Biol. 2020, 27, 959–966. [Google Scholar] [CrossRef] [PubMed]
  124. Konno, Y.; Kimura, I.; Uriu, K.; Fukushi, M.; Irie, T.; Koyanagi, Y.; Sauter, D.; Gifford, R.J.; Nakagawa, S.; Sato, K.; et al. SARS-CoV-2 ORF3b Is a Potent Interferon Antagonist Whose Activity Is Increased by a Naturally Occurring Elongation Variant. Cell Rep. 2020, 32, 108185. [Google Scholar] [CrossRef] [PubMed]
  125. Chen, K.; Xiao, F.; Hu, D.; Ge, W.; Tian, M.; Wang, W.; Pan, P.; Wu, K.; Wu, J. SARS-CoV-2 Nucleocapsid Protein Interacts with RIG-I and Represses RIG-Mediated IFN-beta Production. Viruses 2020, 13, 47. [Google Scholar] [CrossRef] [PubMed]
  126. Han, L.; Zhuang, M.W.; Deng, J.; Zheng, Y.; Zhang, J.; Nan, M.L.; Zhang, X.J.; Gao, C.; Wang, P.H. SARS-CoV-2 ORF9b antagonizes type I and III interferons by targeting multiple components of the RIG-I/MDA-5-MAVS, TLR3-TRIF, and cGAS-STING signaling pathways. J. Med. Virol. 2021, 93, 5376–5389. [Google Scholar] [CrossRef] [PubMed]
  127. Zhang, D.Q.; Ji, L.K.; Chen, X.; He, Y.M.; Sun, Y.J.; Ji, L.; Zhang, T.C.; Shen, Q.; Wang, X.C.; Wang, Y.; et al. SARS-CoV-2 Nsp15 suppresses type I interferon production by inhibiting IRF3 phosphorylation and nuclear translocation. Iscience 2023, 26, 107705. [Google Scholar] [CrossRef]
  128. Sui, C.; Xiao, T.Y.; Zhang, S.Y.; Zeng, H.X.; Zheng, Y.; Liu, B.Y.; Xu, G.; Gao, C.J.; Zhang, Z. SARS-CoV-2 NSP13 Inhibits Type I IFN Production by Degradation of TBK1 via p62-Dependent Selective Autophagy. J. Immunol. 2022, 208, 753–761. [Google Scholar] [CrossRef] [PubMed]
  129. Venet, M.; Ribeiro, M.S.; Décembre, E.; Bellomo, A.; Joshi, G.; Nuovo, C.; Villard, M.; Cluet, D.; Perret, M.; Pescamona, R.; et al. Severe COVID-19 patients have impaired plasmacytoid dendritic cell-mediated control of SARS-CoV-2. Nat. Commun. 2023, 14, 694. [Google Scholar] [CrossRef]
  130. Laing, A.G.; Lorenc, A.; Del Molino Del Barrio, I.; Das, A.; Fish, M.; Monin, L.; Munoz-Ruiz, M.; McKenzie, D.R.; Hayday, T.S.; Francos-Quijorna, I.; et al. A dynamic COVID-19 immune signature includes associations with poor prognosis. Nat. Med. 2020, 26, 1623–1635. [Google Scholar] [CrossRef]
  131. Karki, R.; Sharma, B.R.; Tuladhar, S.; Williams, E.P.; Zalduondo, L.; Samir, P.; Zheng, M.; Sundaram, B.; Banoth, B.; Malireddi, R.K.S.; et al. Synergism of TNF-α and IFN-γ Triggers Inflammatory Cell Death, Tissue Damage, and Mortality in SARS-CoV-2 Infection and Cytokine Shock Syndromes. Cell 2021, 184, 149–168.e17. [Google Scholar] [CrossRef] [PubMed]
  132. Ziegler, C.G.K.; Miao, V.N.; Owings, A.H.; Navia, A.W.; Tang, Y.; Bromley, J.D.; Lotfy, P.; Sloan, M.; Laird, H.; Williams, H.B.; et al. Impaired local intrinsic immunity to SARS-CoV-2 infection in severe COVID-19. Cell 2021, 184, 4713–4733.e22. [Google Scholar] [CrossRef]
  133. Zhang, W.; Chua, B.Y.; Selva, K.J.; Kedzierski, L.; Ashhurst, T.M.; Haycroft, E.R.; Shoffner-Beck, S.K.; Hensen, L.; Boyd, D.F.; James, F.; et al. SARS-CoV-2 infection results in immune responses in the respiratory tract and peripheral blood that suggest mechanisms of disease severity. Nat. Commun. 2022, 13, 2774. [Google Scholar] [CrossRef] [PubMed]
  134. Yoshida, M.; Worlock, K.B.; Huang, N.; Lindeboom, R.G.H.; Butler, C.R.; Kumasaka, N.; Dominguez Conde, C.; Mamanova, L.; Bolt, L.; Richardson, L.; et al. Local and systemic responses to SARS-CoV-2 infection in children and adults. Nature 2022, 602, 321–327. [Google Scholar] [CrossRef] [PubMed]
  135. Poon, M.M.L.; Rybkina, K.; Kato, Y.; Kubota, M.; Matsumoto, R.; Bloom, N.I.; Zhang, Z.; Hastie, K.M.; Grifoni, A.; Weiskopf, D.; et al. SARS-CoV-2 infection generates tissue-localized immunological memory in humans. Sci. Immunol. 2021, 6, eabl9105. [Google Scholar] [CrossRef]
  136. Roukens, A.H.E.; Pothast, C.R.; Konig, M.; Huisman, W.; Dalebout, T.; Tak, T.; Azimi, S.; Kruize, Y.; Hagedoorn, R.S.; Zlei, M.; et al. Prolonged activation of nasal immune cell populations and development of tissue-resident SARS-CoV-2-specific CD8(+) T cell responses following COVID-19. Nat. Immunol. 2022, 23, 23–32. [Google Scholar] [CrossRef]
  137. Diniz, M.O.; Mitsi, E.; Swadling, L.; Rylance, J.; Johnson, M.; Goldblatt, D.; Ferreira, D.; Maini, M.K. Airway-resident T cells from unexposed individuals cross-recognize SARS-CoV-2. Nat. Immunol. 2022, 23, 1324–1329. [Google Scholar] [CrossRef]
  138. Niessl, J.; Sekine, T.; Lange, J.; Konya, V.; Forkel, M.; Maric, J.; Rao, A.; Mazzurana, L.; Kokkinou, E.; Weigel, W.; et al. Identification of resident memory CD8(+) T cells with functional specificity for SARS-CoV-2 in unexposed oropharyngeal lymphoid tissue. Sci. Immunol. 2021, 6, eabk0894. [Google Scholar] [CrossRef]
  139. Jouan, Y.; Guillon, A.; Gonzalez, L.; Perez, Y.; Boisseau, C.; Ehrmann, S.; Ferreira, M.; Daix, T.; Jeannet, R.; Francois, B.; et al. Phenotypical and functional alteration of unconventional T cells in severe COVID-19 patients. J. Exp. Med. 2020, 217, e20200872. [Google Scholar] [CrossRef]
  140. Flament, H.; Rouland, M.; Beaudoin, L.; Toubal, A.; Bertrand, L.; Lebourgeois, S.; Rousseau, C.; Soulard, P.; Gouda, Z.; Cagninacci, L.; et al. Outcome of SARS-CoV-2 infection is linked to MAIT cell activation and cytotoxicity. Nat. Immunol. 2021, 22, 322–335. [Google Scholar] [CrossRef]
  141. Parrot, T.; Gorin, J.B.; Ponzetta, A.; Maleki, K.T.; Kammann, T.; Emgard, J.; Perez-Potti, A.; Sekine, T.; Rivera-Ballesteros, O.; Karolinska, C.-S.G.; et al. MAIT cell activation and dynamics associated with COVID-19 disease severity. Sci. Immunol. 2020, 5, eabe1670. [Google Scholar] [CrossRef]
  142. Ssemaganda, A.; Nguyen, H.M.; Nuhu, F.; Jahan, N.; Card, C.M.; Kiazyk, S.; Severini, G.; Keynan, Y.; Su, R.C.; Ji, H.; et al. Expansion of cytotoxic tissue-resident CD8(+) T cells and CCR6(+)CD161(+) CD4(+) T cells in the nasal mucosa following mRNA COVID-19 vaccination. Nat. Commun. 2022, 13, 3357. [Google Scholar] [CrossRef]
  143. Kunzli, M.; O’Flanagan, S.D.; LaRue, M.; Talukder, P.; Dileepan, T.; Stolley, J.M.; Soerens, A.G.; Quarnstrom, C.F.; Wijeyesinghe, S.; Ye, Y.; et al. Route of self-amplifying mRNA vaccination modulates the establishment of pulmonary resident memory CD8 and CD4 T cells. Sci. Immunol. 2022, 7, eadd3075. [Google Scholar] [CrossRef]
  144. Pieren, D.K.J.; Kuguel, S.G.; Rosado, J.; Robles, A.G.; Rey-Cano, J.; Mancebo, C.; Esperalba, J.; Falco, V.; Buzon, M.J.; Genesca, M. Limited induction of polyfunctional lung-resident memory T cells against SARS-CoV-2 by mRNA vaccination compared to infection. Nat. Commun. 2023, 14, 1887. [Google Scholar] [CrossRef] [PubMed]
  145. Tang, J.; Zeng, C.; Cox, T.M.; Li, C.; Son, Y.M.; Cheon, I.S.; Wu, Y.; Behl, S.; Taylor, J.J.; Chakaraborty, R.; et al. Respiratory mucosal immunity against SARS-CoV-2 after mRNA vaccination. Sci. Immunol. 2022, 7, eadd4853. [Google Scholar] [CrossRef] [PubMed]
  146. Lim, J.M.E.; Tan, A.T.; Le Bert, N.; Hang, S.K.; Low, J.G.H.; Bertoletti, A. SARS-CoV-2 breakthrough infection in vaccinees induces virus-specific nasal-resident CD8+ and CD4+ T cells of broad specificity. J. Exp. Med. 2022, 219, e20220780. [Google Scholar] [CrossRef] [PubMed]
  147. Mitsi, E.; Diniz, M.O.; Reine, J.; Collins, A.M.; Robinson, R.E.; Hyder-Wright, A.; Farrar, M.; Liatsikos, K.; Hamilton, J.; Onyema, O.; et al. Respiratory mucosal immune memory to SARS-CoV-2 after infection and vaccination. Nat. Commun. 2023, 14, 6815. [Google Scholar] [CrossRef]
  148. Afkhami, S.; D’Agostino, M.R.; Zhang, A.; Stacey, H.D.; Marzok, A.; Kang, A.; Singh, R.; Bavananthasivam, J.; Ye, G.; Luo, X.Q.; et al. Respiratory mucosal delivery of next-generation COVID-19 vaccine provides robust protection against both ancestral and variant strains of SARS-CoV-2. Cell 2022, 185, 896–915.e19. [Google Scholar] [CrossRef] [PubMed]
  149. Mao, T.; Israelow, B.; Pena-Hernandez, M.A.; Suberi, A.; Zhou, L.; Luyten, S.; Reschke, M.; Dong, H.; Homer, R.J.; Saltzman, W.M.; et al. Unadjuvanted intranasal spike vaccine elicits protective mucosal immunity against sarbecoviruses. Science 2022, 378, eabo2523. [Google Scholar] [CrossRef]
  150. Diallo, B.K.; Ni Chasaide, C.; Wong, T.Y.; Schmitt, P.; Lee, K.S.; Weaver, K.; Miller, O.; Cooper, M.; Jazayeri, S.D.; Damron, F.H.; et al. Intranasal COVID-19 vaccine induces respiratory memory T cells and protects K18-hACE mice against SARS-CoV-2 infection. NPJ Vaccines 2023, 8, 68. [Google Scholar] [CrossRef]
  151. Onodera, T.; Takahashi, Y.; Yokoi, Y.; Ato, M.; Kodama, Y.; Hachimura, S.; Kurosaki, T.; Kobayashi, K. Memory B cells in the lung participate in protective humoral immune responses to pulmonary influenza virus reinfection. Proc. Natl. Acad. Sci. USA 2012, 109, 2485–2490. [Google Scholar] [CrossRef] [PubMed]
  152. Sterlin, D.; Mathian, A.; Miyara, M.; Mohr, A.; Anna, F.; Claer, L.; Quentric, P.; Fadlallah, J.; Devilliers, H.; Ghillani, P.; et al. IgA dominates the early neutralizing antibody response to SARS-CoV-2. Sci. Transl. Med. 2021, 13, eabd2223. [Google Scholar] [CrossRef]
  153. Marking, U.; Bladh, O.; Havervall, S.; Svensson, J.; Greilert-Norin, N.; Aguilera, K.; Kihlgren, M.; Salomonsson, A.C.; Mansson, M.; Gallini, R.; et al. 7-month duration of SARS-CoV-2 mucosal immunoglobulin-A responses and protection. Lancet Infect. Dis. 2023, 23, 150–152. [Google Scholar] [CrossRef] [PubMed]
  154. Havervall, S.; Marking, U.; Svensson, J.; Greilert-Norin, N.; Bacchus, P.; Nilsson, P.; Hober, S.; Gordon, M.; Blom, K.; Klingstrom, J.; et al. Anti-Spike Mucosal IgA Protection against SARS-CoV-2 Omicron Infection. N. Engl. J. Med. 2022, 387, 1333–1336. [Google Scholar] [CrossRef] [PubMed]
  155. Liew, F.; Talwar, S.; Cross, A.; Willett, B.J.; Scott, S.; Logan, N.; Siggins, M.K.; Swieboda, D.; Sidhu, J.K.; Efstathiou, C.; et al. SARS-CoV-2-specific nasal IgA wanes 9 months after hospitalisation with COVID-19 and is not induced by subsequent vaccination. EBioMedicine 2023, 87, 104402. [Google Scholar] [CrossRef]
  156. Azzi, L.; Dalla Gasperina, D.; Veronesi, G.; Shallak, M.; Ietto, G.; Iovino, D.; Baj, A.; Gianfagna, F.; Maurino, V.; Focosi, D.; et al. Mucosal immune response in BNT162b2 COVID-19 vaccine recipients. EBioMedicine 2022, 75, 103788. [Google Scholar] [CrossRef]
  157. Cervia, C.; Nilsson, J.; Zurbuchen, Y.; Valaperti, A.; Schreiner, J.; Wolfensberger, A.; Raeber, M.E.; Adamo, S.; Weigang, S.; Emmenegger, M.; et al. Systemic and mucosal antibody responses specific to SARS-CoV-2 during mild versus severe COVID-19. J. Allergy Clin. Immunol. 2021, 147, 545–557.e9. [Google Scholar] [CrossRef]
  158. Chan, R.W.Y.; Chan, K.C.C.; Lui, G.C.Y.; Tsun, J.G.S.; Chan, K.Y.Y.; Yip, J.S.K.; Liu, S.; Yu, M.W.L.; Ng, R.W.Y.; Chong, K.K.L.; et al. Mucosal Antibody Response to SARS-CoV-2 in Paediatric and Adult Patients: A Longitudinal Study. Pathogens 2022, 11, 397. [Google Scholar] [CrossRef]
  159. Lima, T.M.; Martins, R.B.; Miura, C.S.; Souza, M.V.O.; Cassiano, M.H.A.; Rodrigues, T.S.; Veras, F.P.; Sousa, J.F.; Gomes, R.; Almeida, G.M.; et al. Tonsils are major sites of persistence of SARS-CoV-2 in children. Microbiol. Spectr. 2023, 11, e0134723. [Google Scholar] [CrossRef]
  160. Xu, Q.; Milanez-Almeida, P.; Martins, A.J.; Radtke, A.J.; Hoehn, K.B.; Oguz, C.; Chen, J.; Liu, C.; Tang, J.; Grubbs, G.; et al. Adaptive immune responses to SARS-CoV-2 persist in the pharyngeal lymphoid tissue of children. Nat. Immunol. 2023, 24, 186–199. [Google Scholar] [CrossRef]
  161. Hassan, A.O.; Kafai, N.M.; Dmitriev, I.P.; Fox, J.M.; Smith, B.K.; Harvey, I.B.; Chen, R.E.; Winkler, E.S.; Wessel, A.W.; Case, J.B.; et al. A Single-Dose Intranasal ChAd Vaccine Protects Upper and Lower Respiratory Tracts against SARS-CoV-2. Cell 2020, 183, 169–184.e13. [Google Scholar] [CrossRef]
  162. van Doremalen, N.; Purushotham, J.N.; Schulz, J.E.; Holbrook, M.G.; Bushmaker, T.; Carmody, A.; Port, J.R.; Yinda, C.K.; Okumura, A.; Saturday, G.; et al. Intranasal ChAdOx1 nCoV-19/AZD1222 vaccination reduces viral shedding after SARS-CoV-2 D614G challenge in preclinical models. Sci. Transl. Med. 2021, 13, eabh0755. [Google Scholar] [CrossRef]
  163. Bricker, T.L.; Darling, T.L.; Hassan, A.O.; Harastani, H.H.; Soung, A.; Jiang, X.; Dai, Y.N.; Zhao, H.; Adams, L.J.; Holtzman, M.J.; et al. A single intranasal or intramuscular immunization with chimpanzee adenovirus-vectored SARS-CoV-2 vaccine protects against pneumonia in hamsters. Cell Rep. 2021, 36, 109400. [Google Scholar] [CrossRef]
  164. Wang, H.; Song, J.; Yao, Y.; Deng, Y.K.; Wang, Z.C.; Liao, B.; Ma, J.; He, C.; Pan, L.; Liu, Y.; et al. Angiotensin-converting enzyme II expression and its implication in the association between COVID-19 and allergic rhinitis. Allergy 2021, 76, 906–910. [Google Scholar] [CrossRef]
  165. Wang, M.; Bu, X.; Fang, G.; Luan, G.; Huang, Y.; Akdis, C.A.; Wang, C.; Zhang, L. Distinct expression of SARS-CoV-2 receptor ACE2 correlates with endotypes of chronic rhinosinusitis with nasal polyps. Allergy 2021, 76, 789–803. [Google Scholar] [CrossRef]
  166. Ziegler, C.G.K.; Allon, S.J.; Nyquist, S.K.; Mbano, I.M.; Miao, V.N.; Tzouanas, C.N.; Cao, Y.M.; Yousif, A.S.; Bals, J.; Hauser, B.M.; et al. SARS-CoV-2 Receptor ACE2 Is an Interferon-Stimulated Gene in Human Airway Epithelial Cells and Is Detected in Specific Cell Subsets across Tissues. Cell 2020, 181, 1016–1035.e19. [Google Scholar] [CrossRef]
  167. Kimura, H.; Francisco, D.; Conway, M.; Martinez, F.D.; Vercelli, D.; Polverino, F.; Billheimer, D.; Kraft, M. Type 2 inflammation modulates ACE2 and TMPRSS2 in airway epithelial cells. J. Allergy Clin. Immunol. 2020, 146, 80–88.e8. [Google Scholar] [CrossRef] [PubMed]
  168. Coden, M.E.; Loffredo, L.F.; Abdala-Valencia, H.; Berdnikovs, S. Comparative Study of SARS-CoV-2, SARS-CoV-1, MERS-CoV, HCoV-229E and Influenza Host Gene Expression in Asthma: Importance of Sex, Disease Severity, and Epithelial Heterogeneity. Viruses 2021, 13, 1081. [Google Scholar] [CrossRef] [PubMed]
  169. Jackson, D.J.; Busse, W.W.; Bacharier, L.B.; Kattan, M.; O’Connor, G.T.; Wood, R.A.; Visness, C.M.; Durham, S.R.; Larson, D.; Esnault, S.; et al. Association of respiratory allergy, asthma, and expression of the SARS-CoV-2 receptor ACE2. J. Allergy Clin. Immunol. 2020, 146, 203–206.e3. [Google Scholar] [CrossRef] [PubMed]
  170. Jayavelu, N.D.; Altman, M.C.; Benson, B.; Dufort, M.J.; Vanderwall, E.R.; Rich, L.M.; White, M.P.; Becker, P.M.; Togias, A.; Jackson, D.J.; et al. Type 2 inflammation reduces SARS-CoV-2 replication in the airway epithelium in allergic asthma through functional alteration of ciliated epithelial cells. J. Allergy Clin. Immun. 2023, 152, 56–67. [Google Scholar] [CrossRef] [PubMed]
  171. Rouphael, N.; Maecker, H.; Montgomery, R.R.; Diray-Arce, J.; Kleinstein, S.H.; Altman, M.C.; Bosinger, S.E.; Eckalbar, W.; Guan, L.Y.; Hough, C.L.; et al. Immunophenotyping assessment in a COVID-19 cohort (IMPACC): A prospective longitudinal study. Sci. Immunol. 2021, 6, eabf3733. [Google Scholar] [CrossRef]
  172. Zhang, J.J.; Dong, X.; Cao, Y.Y.; Yuan, Y.D.; Yang, Y.B.; Yan, Y.Q.; Akdis, C.A.; Gao, Y.D. Clinical characteristics of 140 patients infected with SARS-CoV-2 in Wuhan, China. Allergy 2020, 75, 1730–1741. [Google Scholar] [CrossRef]
  173. Jian, L.; Yi, W.; Zhang, N.; Wen, W.P.; Krysko, O.; Song, W.J.; Bachert, C. Perspective: COVID-19, implications of nasal diseases and consequences for their management. J. Allergy Clin. Immun. 2020, 146, 67–69. [Google Scholar] [CrossRef]
  174. Caminati, M.; Lombardi, C.; Micheletto, C.; Roca, E.; Bigni, B.; Furci, F.; Girelli, D.; Senna, G.; Crisafulli, E. Asthmatic patients in COVID-19 outbreak: Few cases despite many cases. J. Allergy Clin. Immunol. 2020, 146, 541–542. [Google Scholar] [CrossRef] [PubMed]
  175. Saheb Sharif-Askari, F.; Goel, S.; Saheb Sharif-Askari, N.; Hafezi, S.; Al Heialy, S.; Hachim, M.Y.; Hachim, I.Y.; Mahboub, B.; Salameh, L.; Abdelrazig, M.; et al. Asthma Associated Cytokines Regulate the Expression of SARS-CoV-2 Receptor ACE2 in the Lung Tissue of Asthmatic Patients. Front. Immunol. 2021, 12, 796094. [Google Scholar] [CrossRef] [PubMed]
  176. Globinska, A.; Pawelczyk, M.; Piechota-Polanczyk, A.; Olszewska-Ziaber, A.; Moskwa, S.; Mikolajczyk, A.; Jablonska, A.; Zakrzewski, P.K.; Brauncajs, M.; Jarzebska, M.; et al. Impaired virus replication and decreased innate immune responses to viral infections in nasal epithelial cells from patients with allergic rhinitis. Clin. Exp. Immunol. 2017, 187, 100–112. [Google Scholar] [CrossRef]
  177. Gilles, S.; Blume, C.; Wimmer, M.; Damialis, A.; Meulenbroek, L.; Gokkaya, M.; Bergougnan, C.; Eisenbart, S.; Sundell, N.; Lindh, M.; et al. Pollen exposure weakens innate defense against respiratory viruses. Allergy 2020, 75, 576–587. [Google Scholar] [CrossRef]
  178. Hatchwell, L.; Collison, A.; Girkin, J.; Parsons, K.; Li, J.Y.; Zhang, J.; Phipps, S.; Knight, D.; Bartlett, N.W.; Johnston, S.L.; et al. Toll-like receptor 7 governs interferon and inflammatory responses to rhinovirus and is suppressed by IL-5-induced lung eosinophilia. Thorax 2015, 70, 854–861. [Google Scholar] [CrossRef]
  179. Akbarshahi, H.; Menzel, M.; Ramu, S.; Persson, I.M.; Bjermer, L.; Uller, L. House dust mite impairs antiviral response in asthma exacerbation models through its effects on TLR3. Allergy 2018, 73, 1053–1063. [Google Scholar] [CrossRef]
  180. Hajighasemi, S.; Spann, K.; Davies, J.M. Does pollen exposure influence innate immunity to SARS-CoV-2 in allergy or asthma? J. Allergy Clin. Immun. 2023, 152, 374–377. [Google Scholar] [CrossRef]
  181. Hosoki, K.; Boldogh, I.; Sur, S. Innate responses to pollen allergens. Curr. Opin. Allergy Clin. Immunol. 2015, 15, 79–88. [Google Scholar] [CrossRef]
  182. Yao, Y.; Wang, Z.Z.; Huang, A.; Liu, Y.; Wang, N.; Wang, Z.C.; Yang, L.; Li, H.J.; Xie, J.G.; Zhu, R.F.; et al. T2 cells associate with enhanced humoral immunity to SARS-CoV-2 inactivated vaccine in patients with allergic rhinitis. Clin. Transl. Med. 2022, 12, e717. [Google Scholar] [CrossRef]
  183. Chen, L.; Yue, J.; Zhang, S.; Bai, W.; Qin, L.; Zhang, C.; Wu, B.; Li, M.; Xu, S.; Jiang, Q.; et al. SARS-CoV-2-Specific Adaptive Immunity in COVID-19 Survivors With Asthma. Front. Immunol. 2022, 13, 947724. [Google Scholar] [CrossRef]
  184. Ferastraoaru, D.; Hudes, G.; Jerschow, E.; Jariwala, S.; Karagic, M.; de Vos, G.; Rosenstreich, D.; Ramesh, M. Eosinophilia in Asthma Patients Is Protective Against Severe COVID-19 Illness. J. Allergy Clin. Immunol. Pract. 2021, 9, 1152–1162.e3. [Google Scholar] [CrossRef]
  185. Ho, K.S.; Howell, D.; Rogers, L.; Narasimhan, B.; Verma, H.; Steiger, D. The relationship between asthma, eosinophilia, and outcomes in coronavirus disease 2019 infection. Ann. Allergy Asthma Immunol. 2021, 127, 42–48. [Google Scholar] [CrossRef] [PubMed]
  186. Ren, J.J.; Pang, W.D.; Luo, Y.X.; Cheng, D.N.; Qiu, K.; Rao, Y.F.; Zheng, Y.B.; Dong, Y.J.; Peng, J.J.; Hu, Y.; et al. Impact of Allergic Rhinitis and Asthma on COVID-19 Infection, Hospitalization, and Mortality. J. Allergy Clin. Immunol. Pract. 2022, 10, 124–133. [Google Scholar] [CrossRef] [PubMed]
  187. Scala, E.; Abeni, D.; Tedeschi, A.; Manzotti, G.; Yang, B.R.; Borrelli, P.; Marra, A.; Giani, M.; Sgadari, A.; Saltalamacchia, F.; et al. Atopic status protects from severe complications of COVID-19. Allergy 2021, 76, 899–902. [Google Scholar] [CrossRef] [PubMed]
  188. Yang, J.M.; Koh, H.Y.; Moon, S.Y.; Yoo, I.K.; Ha, E.K.; You, S.; Kim, S.Y.; Yon, D.K.; Lee, S.W. Allergic disorders and susceptibility to and severity of COVID-19: A nationwide cohort study. J. Allergy Clin. Immunol. 2020, 146, 790–798. [Google Scholar] [CrossRef] [PubMed]
  189. Yang, X.Z.; Zhao, J.M.; Sima, Y.; Zhao, Y.; Zhang, J.; Wang, X.D.; Zhang, L. The association between allergic rhinitis and the risk of coronavirus disease 2019 (COVID-19): A national survey in China. Allergy 2023, 78, 2783–2786. [Google Scholar] [CrossRef] [PubMed]
  190. Dolby, T.; Nafilyan, V.; Morgan, A.; Kallis, C.; Sheikh, A.; Quint, J.K. Relationship between asthma and severe COVID-19: A national cohort study. Thorax 2022, 78, 120–127. [Google Scholar] [CrossRef]
Figure 1. Importance of immune memory in the respiratory mucosa for protection against SARS-CoV-2. When there are high titers of neutralizing antibodies (NAbs) and a substantial number of tissue-resident memory T (TRM) and tissue-resident memory B (BRM) cells in the mucosa of the respiratory tract, infection with severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is blocked, or it only results in mild disease. However, the progression to severe disease occurs when there are low titers of NAbs and weak memory B/T cell responses in the respiratory mucosa.
Figure 1. Importance of immune memory in the respiratory mucosa for protection against SARS-CoV-2. When there are high titers of neutralizing antibodies (NAbs) and a substantial number of tissue-resident memory T (TRM) and tissue-resident memory B (BRM) cells in the mucosa of the respiratory tract, infection with severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is blocked, or it only results in mild disease. However, the progression to severe disease occurs when there are low titers of NAbs and weak memory B/T cell responses in the respiratory mucosa.
Pathogens 13 00113 g001
Table 1. A summary of the key components of mucosal immune responses against SARS-CoV-2.
Table 1. A summary of the key components of mucosal immune responses against SARS-CoV-2.
ImmunityComponentRoles and/or FindingsRefs.
Innate immunityMucins (soluble and transmembrane mucins)Controversial roles: elevated levels of MUC1 and MUC5AC (soluble mucins) may impact disease progression, while transmembrane mucins show in vitro evidence of protective functions by preventing SARS-CoV-2 binding and entry.[103,104,105,106,107]
Antimicrobial peptides
(AMPs)
Increased β-defensin transcripts are observed in SARS-CoV-2 patients; potential role in inhibiting viral entry. [108,109]
Type I IFN response Delayed and inefficient induction in COVID-19 contributes to severe disease.
Inborn defects or autoantibodies against type I IFNs; higher risk of severe COVID-19.
[96,101,102,113,114,115,116,117,118,119,120,121,122,123,124,125,126,127,128]
Plasmacytoid DCs (pDCs)Primary source of type I IFNs; loss of response observed in severe COVID-19.[129,130]
Cytokine storm
(PANoptosis)
Life-threatening immune response mediated by inflammatory cell death; the synergism of TNF-α and IFN-γ.[131]
Adaptive immunityTissue-resident memory
(TRM) cells
Critical role in rapid protection; longevity remains unclear.
Increasing in vivo evidence supporting superior ability of intranasal vaccination to induce airway TRM cells.
[135,136,146]
Mucosal-associated invariant T (MAIT) cellsEnrichment of MAIT cells and activated phenotypes in the airway of patients with COVID-19; potential role in local immune response.[139,140,141]
Mucosal memory B cellsRapid increase in local antibody titer for efficient viral clearance.
SARS-CoV-2-specific B cells in tonsils and adenoids exhibit distinct phenotypes and play potential pivotal role in immune defense, especially in pediatric patients.
[135,151,159,160]
IgA Key component of mucosal immune response; more closely correlated with SARS-CoV-2 neutralization than IgM or IgG.
Children exhibited earlier and more robust mucosal IgA response to SARS-CoV-2; linked to a rapid decline in viral load.
[152,153,154,157,158]
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Noh, H.-E.; Rha, M.-S. Mucosal Immunity against SARS-CoV-2 in the Respiratory Tract. Pathogens 2024, 13, 113. https://doi.org/10.3390/pathogens13020113

AMA Style

Noh H-E, Rha M-S. Mucosal Immunity against SARS-CoV-2 in the Respiratory Tract. Pathogens. 2024; 13(2):113. https://doi.org/10.3390/pathogens13020113

Chicago/Turabian Style

Noh, Hae-Eun, and Min-Seok Rha. 2024. "Mucosal Immunity against SARS-CoV-2 in the Respiratory Tract" Pathogens 13, no. 2: 113. https://doi.org/10.3390/pathogens13020113

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