Next Article in Journal
Chitosan-Dextran-Glycerol Hydrogels Loaded with Iron Oxide Nanoparticles for Wound Dressing Applications
Next Article in Special Issue
Tumor Treating Fields (TTFields) Induce Cell Junction Alterations in a Human 3D In Vitro Model of the Blood-Brain Barrier
Previous Article in Journal
Amorphicity and Aerosolization of Soluplus-Based Inhalable Spray Dried Powders
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Airway Epithelial Cell Junctions as Targets for Pathogens and Antimicrobial Therapy

1
Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic Foundation, Cleveland, OH 44195, USA
2
Center for Pediatric Pulmonary Medicine, Cleveland Clinic Children’s, Cleveland, OH 44195, USA
*
Author to whom correspondence should be addressed.
Pharmaceutics 2022, 14(12), 2619; https://doi.org/10.3390/pharmaceutics14122619
Submission received: 4 November 2022 / Revised: 22 November 2022 / Accepted: 24 November 2022 / Published: 27 November 2022
(This article belongs to the Special Issue Targeting Cell Junctions for Therapy and Delivery)

Abstract

:
Intercellular contacts between epithelial cells are established and maintained by the apical junctional complexes (AJCs). AJCs conserve cell polarity and build epithelial barriers to pathogens, inhaled allergens, and environmental particles in the respiratory tract. AJCs consist of tight junctions (TJs) and adherens junctions (AJs), which play a key role in maintaining the integrity of the airway barrier. Emerging evidence has shown that different microorganisms cause airway barrier dysfunction by targeting TJ and AJ proteins. This review discusses the pathophysiologic mechanisms by which several microorganisms (bacteria and viruses) lead to the disruption of AJCs in airway epithelial cells. We present recent progress in understanding signaling pathways involved in the formation and regulation of cell junctions. We also summarize the potential chemical inhibitors and pharmacological approaches to restore the integrity of the airway epithelial barrier. Understanding the AJCs–pathogen interactions and mechanisms by which microorganisms target the AJC and impair barrier function may further help design therapeutic innovations to treat these infections.

1. Introduction

The airway epithelium is at the front line defending the human respiratory system against inhaled environmental insults such as viruses, bacteria, and allergens [1,2]. From the nasal cavity to the alveoli, specialized epithelial cells line the entire respiratory tract. There are distinct regional differences in the epithelial cell populations as the functions vary across different airway levels. The major epithelial cell types lining the airways are generally ciliated and secretory cells in adaptation to the mucociliary escalator, while the distal alveolar regions are lined with alveolar epithelial type I and type II cells to enable efficient air exchange [3]. As an essential part of the innate immune system, the airway epithelium is pivotal to the barrier function of the airway [4]. The primary contributors to the airway epithelial barrier function include mucociliary clearance, antimicrobial peptides secreted by the airway epithelial cells (AECs), and intercellular apical junctional complexes (AJCs) that regulate barrier integrity [5]. The AJCs are formed between adjacent epithelial cells on the apical lateral membranes and are composed of tight junctions (TJs) and adherens junctions (AJs) (Figure 1). Through interactions between the extracellular domains of the membrane-spanning adhesion molecules, TJs and AJs establish connections between neighboring cells. TJs are located at the apex of the lateral membranes between adjoining cells. TJs encircle the cell and form a “seal” between cells. In addition to this “seal”, TJs maintain the cell polarity by separating apical and basolateral membrane components and can control the paracellular permeability [6,7]. TJs comprise three families of transmembrane proteins: the claudin family (claudin-1 through 27 in mammals) [8], the tight junction-associated marvel protein (TAMPs) family (occludin, tricellulin, and MARVEL domain-containing protein 3), and the immunoglobulin family (junctional adhesion molecules (JAM-A) and coxsackie adenovirus receptor (CAR)) [1]. Inside the cell, TJs are connected to the cytoplasmic scaffolding proteins, such as zonula occludens (ZO-1, ZO-2, and ZO-3) and cingulin [9], and in turn, attached to the actin cytoskeleton [10,11,12,13]. AJs are more basally located than TJs and are crucial for initiating and maintaining cell–cell adhesion [14]. AJs are comprised of two primary transmembrane protein families: the cadherin family (such as E-cadherin) and the nectin family. Intracellularly, AJs bind to a cytoplasmic scaffolding and signaling complexes consisting of p120 catenin, β-catenin, and α-catenin proteins and are hence anchored to the actin cytoskeleton, forming a hub for signaling transduction and transcriptional regulation [1,6,7]. Microorganisms such as bacteria and viruses have evolved to utilize various mechanisms to breach the airway epithelial barrier as an early critical step to establish infection. Emerging evidence has revealed increased barrier permeability as a common characteristic for infected cells during respiratory bacterial and viral infection [15,16,17,18,19,20,21]. Increasing permeability not only allow bacteria and virus to reach tissues underneath the airway epithelium and bloodstream but will also facilitate secondary invasion of other allergens and pathogens [2]. Therefore, strategies to modify barrier permeability bear great potential in developing antimicrobial therapeutics.
Most human bacterial pathogens are capable of asymptomatic colonization in the respiratory tract, which can progress toward mild to severe infections such as sinusitis and pneumonia [22]. Bacteria penetrate the airway epithelium by damaging or crossing the AECs. Bacteria can cause lysis or apoptosis of the host cells by releasing exotoxins and enzymes [23]. On the other hand, bacterial infections could lead to disruption of the epithelial barrier structure and function via targeting TJs and AJs (Table 1). Respiratory viral infections result in diseases from the common cold to bronchiolitis and acute respiratory distress. Airway and alveolar epithelial cells are the primary targets for these inhaled pathogens, and respiratory viruses usually enter the host cells via binding to specific receptors [24]. Interestingly, components of the TJs can be hijacked by some viruses to aid their infectious cycle. For example, CARs are used by adenoviruses and coxsackievirus B as receptors during their entry processes [25]. While some respiratory viruses disrupt barrier dysfunction by causing cytotoxicity in host cells, others cause epithelial barrier disruption without affecting cell viability [2]. Increasing evidence has illustrated mechanisms utilized by respiratory viruses to disassemble TJs and AJs by regulating protein expression or localization of their components (Table 2).
In recent years, studies using airway epithelial cell lines, primary epithelial cells cultured at the air–liquid interface (ALI), and murine models have helped to identify the structural and functional damages of AJCs caused by microbial infection as well as the underlying mechanisms. Characterizing the mechanisms by which pathogens impair the AJCs and damage barrier function will help to improve therapeutic innovations in related disease areas. In this review, we will discuss Staphylococcus aureus (S. aureus), Streptococcus pneumoniae (S. pneumoniae), Pseudomonas aeruginosa (P. aeruginosa), Burkholderia, Haemophilus influenzae (H. influenzae), respiratory syncytial virus (RSV), human rhinovirus (HRV), influenza viruses, human parainfluenza viruses (HPIV), and coronavirus family (Figure 2). We will briefly summarize each microorganism’s clinical manifestation, health burdens, and challenges in infection management. Then, we will focus on the current knowledge regarding their impacts on airway epithelial barrier integrity and the pathophysiologic mechanisms of pathogen-induced disruption of the AJC. Finally, we will reflect on the present and potential approaches to restoring the epithelial barrier during infection.

2. Bacteria

2.1. Staphylococcus aureus

Staphylococcus aureus (S. aureus) is a Gram-positive bacterium that belongs to the Micrococcaceae family [79,80]. Humans are natural carriers of S. aureus [81], and these bacteria are commonly present in healthy individuals’ skin and mucous membranes, with the nasal cavities as the most frequent colonization site in up to 30% of the human population [82,83,84]. Even though S. aureus is commensal in humans, it is a frequent cause of a number of clinical manifestations, such as skin and soft tissue infection, pleuropulmonary infection, infective endocarditis, osteomyelitis, and vascular catheter-related infections [85,86]. Treatment of S. aureus infection often includes antibiotic therapy and drainage of the infection site. However, managing S. aureus infection has become increasingly problematic worldwide due to the emergence of antibiotic resistance in strains such as methicillin-resistant S. aureus (MRSA) [87,88,89]. In 2017, the Centers for Disease Control and Prevention (CDC) estimated that more than 323,700 cases in hospitalized patients and more than 10,600 deaths were caused by MRSA in the United States [90].
Previous studies have revealed that S. aureus infection is established and maintained through an array of virulence factors such as adhesion proteins, toxins, secretory enzymes, and immune-modulatory factors [91]. In the respiratory system, S. aureus infection depends on its ability to breach the airway epithelial barrier by affecting the cell–cell junctions. There is emerging evidence indicating that S. aureus damages the airway epithelial barrier by altering the expression of TJ and AJ proteins. For example, it has been shown that conditioned media from S. aureus impairs the airway epithelium by disrupting the TJs between primary human nasal epithelial cells (HNECs) cultured at an ALI, in which discontinuous expression of ZO-1 was observed after treatment with conditioned media [26]. A similar impairment in barrier integrity and change in the expression of ZO-1 were noted when purified S. aureus V8 protease was added to HNECs in ALI cultures [27]. In a study using ALI cultures of HNECs from patients with severe chronic rhinosinusitis with nasal polyps, S. aureus enterotoxin B stimulation was associated with reduced ZO-1 and occludin localization at cell membranes. Similarly, decreased protein levels of claudin-1 and phosphorylation of occludin were also found in patient cells after exposure to S. aureus enterotoxin B [21]. This study also examined the effects of S. aureus enterotoxin B in mice and found that nasal challenge with S. aureus enterotoxin B significantly increased mucosal permeability and decreased mRNA expression of ZO-1 and occludin compared with saline. Furthermore, the authors showed that S. aureus enterotoxin B damaged nasal polyp epithelial cell integrity by triggering the Toll-like receptor 2 (TLR2) pathway [21]. A study by Kalsi et al. reported that S. aureus infection reduced the expression of ZO-1 and occludin in the monolayer of human airway epithelial cells (H441). Taken together, these in vitro and in vivo data suggest that S. aureus impairs epithelial integrity by decreasing the expression of TJ proteins, especially occludin, through secreted viral factors. Interestingly, the antidiabetic drug metformin, which reduces airway glucose permeability and hyperglycemia-induced S. aureus load [92], was shown to improve epithelial barrier function by promoting the abundance and assembly of occludin at TJ via an AMPK-PKCζ pathway in human AECs [28].
AJ proteins have also been implicated as targets of S. aureus disrupting the epithelial barrier. It has also been reported that S. aureus α-toxin (also known as α-hemolysin) binds and activates the metalloproteinase domain-containing protein 10 (ADAM10), resulting in the cleavage of the AJ protein E-cadherin, which disrupts the lung epithelial barrier in mice. In the ADAM10-knockout mice, S. aureus-induced E-cadherin proteolysis and barrier disruption were attenuated compared with control animals, suggesting the potential of inhibiting ADAM10 in alleviating epithelial barrier dysfunction caused by S. aureus [29]. In a genetic screen sought to identify the host factors that mediate α-toxin cytotoxicity from S. aureus [93], several components of the AJ were discovered, including the junctional protein pleckstrin-homology domain-containing protein 7 (PLEKHA7). It was suggested that PLEKHA7 promotes the ADAM10-mediated toxicity of S. aureus involving PDZ domain-containing protein 11 (PDZD11) and Tspan33, which is a known factor for α-toxin cytotoxicity [94]. Therefore, strategies to preserve AJ proteins might have therapeutic potential to mitigate S. aureus virulence.
Another mechanism that S. aureus possibly utilizes to affect intercellular junction and perturb the epithelial barrier is through compromising the actin cytoskeleton. It has been reported that α-toxin and ADAM10 form a complex to disrupt focal adhesion in alveolar epithelial cells, where focal adhesion kinase (FAK) and steroid receptor coactivator (Src) dephosphorylation were observed [30]. This study showed that the α-toxin-ADAM10 complex was assembled in cholesterol/sphingolipid-rich caveolar rafts on the membrane, which likely provided access to deactivate caveolae-associated proteins FAK and Src. It is well known that the activated FAK/Src complex phosphorylates p130Cas and paxillin, which in turn link integrin receptors to Rho family GTPases, actin cytoskeleton, and focal adhesion [95,96]. Therefore, S. aureus might disturb barrier integrity by compromising the actin network and disrupting focal adhesions. More recently, S. aureus α-toxin-induced actin filament remodeling was revealed in human AECs [31]. In 16HBE14o- human bronchial epithelial (16HBE) cells, S. aureus α-toxin led to the hypophosphorylation of cofilin at Ser3 via inhibiting p21-activated protein kinase and LIM kinase activities. Cofilin is an actin-depolymerizing factor, and the dephosphorylation of pSer3-cofilin results in its activation [97]. Therefore, α-toxin caused a loss of actin stress fibers and destabilization of cell shape and cell–cell connection. The authors also noticed that protein kinase A and small GTPases (Rho, Rac, Cdc42) did not seem to be involved in this response. Interestingly, evidence has shown that a dynamic actin cytoskeleton and activation of Src family protein–tyrosine kinases (PTKs) in 293T cells mediate S. aureus invasion, and Src PTK-deficient cells are resistant to S. aureus infection [98].
Collectively, progress in unraveling mechanisms of S. aureus infection in AECs provided potential approaches to combat the epithelial barrier disruption caused by S. aureus.

2.2. Streptococcus pneumonia

Streptococcus pneumoniae (S. pneumoniae) are lancet-shaped, Gram-positive bacteria that commonly inhabit the healthy upper respiratory tract in humans [99,100]. As an opportunistic pathogen, S. pneumoniae can cause non-invasive and invasive infections, such as otitis media, sinusitis, bacteremia, pneumonia, and meningitis [99,101]. Globally, S. pneumoniae is one of the most common causes of community-acquired pneumonia and the leading cause of pneumonia mortality [102]. Typical treatments of S. pneumoniae include antibiotic therapies, while the more severe cases might need intensive care unit (ICU)-level care and mechanical ventilation. However, the prevalence of antimicrobial resistance in S. pneumoniae has considerably increased since it was first reported in the 1960s [103], posing a growing challenge to the clinical management of S. pneumoniae infection [104,105]. A global antimicrobial surveillance program from 1997 to 2016 reported an overall 65.8% penicillin susceptibility in S. pneumoniae isolated worldwide in 2015–2016 [106], and the U.S. CDC identified the drug-resistant S. pneumoniae as a serious threat with an estimated death toll of 3600 among 900,000 infections in 2014 [90]. Pneumococcal conjugate vaccines (PCVs) against S. pneumoniae have significantly reduced invasive pneumococcal disease caused by vaccine-targeted serotypes [107,108]. However, because of the existence of 100 different serotypes [109], the replacement of vaccine serotypes by non-vaccine serotypes has been reported [110,111,112], raising growing concern about a new challenge and urging for a better understanding of pathogenies of S. pneumoniae infection.
From their colonization in the upper respiratory tract, S. pneumoniae needs to cross the barriers of pulmonary epithelial cells before reaching blood circulation [113]. Previous studies have shown that S. pneumoniae impairs the integrity of the epithelial barriers in mice and cultured epithelial monolayers [32]. Separation of the TJs is also observed in human respiratory tissues after S. pneumoniae infection [33], and S. pneumoniae infection causes dose-dependent AJ disruption in lung epithelial cells in vivo and in vitro [34]. Intercellular junction proteins have been implicated as key targets during the epithelial morphological and functional disruption caused by S. pneumoniae. Peter et al. found that pneumococcal infection reduced alveolar ZO-1, occludin, claudin-5, and VE-cadherin when comparing naïve and S. pneumoniae-infected human lung tissues using spectral confocal microscopy and Western blot [35]. S. pneumoniae-induced cleavage and degradation of E-cadherin [29,36] and reduced claudins 7 and 10 [32] were also reported in lung epithelial cells.
Continuous research endeavors have uncovered possible mechanisms utilized by these bacteria to diminish TJ/AJ components of host cells. In an in vitro model of the pneumococcal invasion process, S. pneumoniae was shown to traverse the A549 epithelial monolayers through the facilitation of bacteria-bound plasmin, which disrupted intercellular junctions of epithelial cells by cleaving and degrading E-cadherin [36]. Pneumolysin (PLY), a pore-forming toxin released by S. pneumoniae, was also reported to trigger E-cadherin cleavage via activating ADAM10 [29]. Activation of the TLR pathway is another mechanism used by S. pneumoniae to get across the epithelium. TLR-dependent decreases of claudins 7 and 10 were observed in a murine model as well as cultured 16HBE monolayers after S. pneumoniae infection [32]. This study also revealed that the downregulation of claudins was mediated by an increase in transcriptional repressor Snail1 following the activation of TLR/p38 mitogen-activated protein kinase (MAPK)/TGF-β cascade caused by S. pneumoniae. On the other hand, treatment with recombinant interferon-β (IFN-β) inhibited bacterial invasion and transmigration in mouse models [20]. Moreover, in this intranasal infection model, increased mRNA levels of Cdh1 (E-cadherin), Tjp1 (ZO-1), Cldn4 (claudin 4), Cldn5 (claudin 5), and Cldn18 (claudin 18) were observed after IFN-β treatment. These data suggest that increased expression levels of TJ and AJ proteins might be of therapeutic interest in combating S. pneumoniae invasion. Interestingly, perijunctional F-actin, a cytoskeletal structure connected to TJ and AJ, was severely distorted during the first 3–4 h after S. pneumoniae infection prior to the detection of deficits in barrier integrity. As an essential structural foundation for the maintenance of barrier integrity, perijunctional F-actin could be another target to mitigate the structural and functional barrier damage at an early stage of S. pneumoniae infection [114].

2.3. Pseudomonas aeruginosa

Pseudomonas aeruginosa (P. aeruginosa) is a Gram-negative, aerobic rod bacterium of the Pseusomonadaceae family and a member of the Gammaproteobacteris [115]. P. aeruginosa is commonly found in the environment as an inhabitant in soil, water, and plants. P. aeruginosa is an emerging opportunistic pathogen that causes recurrent or persistent infections in patients with cystic fibrosis (CF), primary immunodeficiency, diabetes mellitus, severe burn, and those affected by cancer and acquired immunodeficiency syndrome (AIDS) [116,117].
P. aeruginosa causes infections in various organs, such as the respiratory system, urinary tract, gastrointestinal tract, skin, soft tissue, bone, joints, and blood [116]. The epidemiological data show that P. aeruginosa, particularly antibiotic resistance strains, can cause widespread nosocomial spread [116]. Indeed, nosocomial P. aeruginosa is the fourth most commonly isolated pathogen and the most common Gram-negative organism, accounting for 10% of all hospital-acquired infections, especially in ICU patients. In 2017, the CDC estimated 32,600 infections among hospitalized patients and 2700 deaths in the United States [90].
Adhesion to the airway epithelium is a critical step for P. aeruginosa infection. In addition to causing infection and inflammation, there are multiple reports of the effect of P. aeruginosa on the airway epithelial barrier. One study showed that P. aeruginosa infection decreased the transepithelial electric resistance (TEER) and increased the paracellular glucose flux across the Calu-3 airway epithelial cells. Furthermore, P. aeruginosa infection decreased the expression of occludin and claudin-1 and induced the cleavage of occludin but had no effect on E-cadherin expression [37]. These effects of P. aeruginosa on Calu-3 TJ were thought to be through the induction of hyperglycemia since pre-treatment with the antidiabetic drug metformin decreased the P. aeruginosa-induced barrier disruption and inhibited bacterial growth [37]. This is consistent with the data that elevated bronchial glucose is associated with an increased risk of respiratory infection in patients admitted to intensive care patients [118,119]. In CF patients who are at risk of P. aeruginosa colonization and infection, the elevated blood glucose concentrations in airway surface liquid (ASL) associated with cystic fibrosis-related diabetes (CFRD) has been shown to increase bacterial growth such as P. aeruginosa and S. aureus. This phenomenon was thought to be secondary to the impact of hyperglycemia on the cystic fibrosis transmembrane conductance regulator (CFTR), which has an integral role in airway barrier function [120]. Impaired CFTR could also lead to viscous ASL and enhance infection susceptibility [37]. The lack of proper CFTR function, viscous ASL, and impaired epithelial barrier prompts airway colonization with multiple bacteria, including P. aeruginosa. Another study compared the impact of P. aeruginosa and E. coli infection on airway barrier structure and function using in vitro and in vivo models. The in vitro studies showed a decreased TEER, damaging ZO-1 structure, and increased permeability to bacterial infections through the epithelial layer. At the same time, E. coli infection did not affect the epithelial barrier. Similarly, the in vivo studies showed that C57BL/6 mice infected with P. aeruginosa exhibited an increased permeability to fluorescein isothiocyanate (FITC)-labeled polyplexes into the lung parenchyma, indicating disruption of airway epithelium, while E. coli infection did not affect airway barrier function [38]. Nomura et al. applied P. aeruginosa elastase (PE) on HNECs cultures. They observed a transient decrease in TEER associated with a reduction in the expression of claudin-1 and -4, occludin, and tricellulin. Interestingly, PE exposure did not affect ZO-1, ZO-2, E-cadherin, or β-catenin. The effect of PE on TJs was attenuated by chemical inhibitors of PKC, MAPK, phosphoinositide 3-kinase (PI3K), p38 MAPK, JNK, COX-1 and -2, and NF-κB pathways. There was evidence of decreased PAR-2; consequently, the PE-induced TJ disruption was attenuated by a PAR-2 agonist, while the PAR-2 knockdown, even in the absence of PE, downregulated the TJs [39]. Likewise, Clark et al. found that exposure of Calu3 cells to PE induced PKC signaling, disrupted occludin and ZO-1 proteins, and caused actin cytoskeletal reorganization [40].
The endotoxins released by P. aeruginosa, such as rhamnolipids, could also disrupt TJ structure and decrease barrier function, especially in patients with CF who are chronically colonized with these bacteria [41]. This evidence was supported by observations that adding merely the purified rhamnolipids could trigger a reduction in TEER in a dose and time-dependent manner and disrupt the TJ structure [41]. Moreover, to establish their interaction with the host and deliver cytotoxins directly into eukaryotic cells to mediate the pathogenesis, Gram-negative bacterial secrete multiple effector proteins via their Type III Secretion System (T3SS) apparatus. Four effectors, protein Exoenzyme (Exo) S, ExoT, ExoU, and ExoY, are used by P. aeruginosa strains to mediate the pathogenesis [121]. For example, the Exo S and ExoT disrupted the actin cytoskeleton of host AEC and induced barrier disruption by impairing cell-to-cell adhesion, and ExoY disrupted the barrier integrity without cytotoxicity, while another effector protein, ExoU, exerted rapid necrotic cytotoxicity [121,122,123,124,125,126]. Similarly, another study showed that type III toxins, especially ExoS-induced permeability in 16HBE cells, altered the distribution of ZO-1 and occludin [42].

2.4. Burkholderia

Burkholderia, a genus of Pseudomonadota, contains more than 80 Gram-negative coccobacilli species, which are ubiquitous within the environment and pathogenic to plants, insects, animals, and humans [127,128]. Burkholderia cepacia complex (BCC) is a group of at least 24 species. One of them is B. cepacia, which is an opportunistic human pathogen that most often causes pneumonia and fatal infections in individuals with CF, chronic lung disease, sickle cell, and immunodeficiency. These bacteria can also cause severe nosocomial infections, bacteremia, and sepsis [129]. In 20% of patients, the bacteria can trigger cepacia syndrome, which is characterized by fatal necrotizing pneumonia with bacteremia [128,130]. Antibiotic therapy for these bacteria is challenging due to the intrinsic antimicrobial resistance and acquired resistance to antimicrobial agents [131]. Additionally, some CF patients can be infected with B. cepacia and P. aeruginosa together, which causes more severe infection.
It has been shown that B. cepacia can pass through airway epithelium; however, the involved mechanisms still need to be better understood. Mounting evidence suggests that B. cenocepacia can cross the respiratory epithelium by disrupting TJs. Kim et al. utilized polarized 16HBE cells in vitro and found that B. cenocepacia infection decreased TEER and increased the flux of FITC-labeled bovine serum albumin (BSA) across the cell monolayer [43]. They did not observe significant cytotoxicity or cell death in infected cells. Confocal fluorescence microscopy observed a substantial decrease in occludin expression, while ZO-1 remained intact. Furthermore, they saw the dephosphorylation of occludin and concluded that B. cenocepacia-induced dephosphorylation, and occludin dissociation could facilitate the migration of bacteria through the respiratory epithelium, leading to bacteremia [43]. Sajjan et al. used lung explants from CF recipients who underwent lung transplants with and without B. cepacia and autopsy sections of CF patients with B. cepacia infection [44]. They showed evidence that B. cepacia altered the epithelial cell barrier by increasing the permeability, which allows bacteria to migrate through the polarized respiratory epithelium to the lung interstitium and lumen of blood capillaries and cause blood infection and sepsis [44]. Another study used three strains of B. cepacia complex known to cause cepacia syndrome to study the mechanisms of bacteremia caused by these organisms. Using well-differentiated human AEC cultures, they showed that these strains could invade the epithelial barrier via different invasion pathways. For example, the B. cepacia BC-7 formed a biofilm near the apical cell surface, attached to the cell surface, disrupted the superficial epithelial cell layer, and dislocated the actin cytoskeleton. In contrast, the HI2258 strain (genomovar IV) did not form biofilms and passed through the between epithelial cells without disrupting the integrity of the epithelium, suggesting a paracytosis route. However, strain J-1, with both biofilm-dependent and independent ability, invaded the tissue by both formation of biofilm as well as inducing both paracytosis and cell destruction [45].
In a similar study, Duff et al. used three lung epithelial cells to examine mechanisms by which BCC invades the epithelial barrier [46]. Four species of BCC were used in this study, and all caused a decrease in TEER. However, the investigators showed that some of these species could easily traverse the cell monolayer, while others were slower to translocate, which suggests differences in their potential to invade the epithelial barrier. Confocal microscopy and Western blot analysis showed a decreased expression of ZO-1 and E-cadherin in Calu-3 cells [46], which contrasts with undisrupted ZO-1 observed in 16HBE cells by Kim et al. [43], and it might suggest a cell-specific difference or various study time points. Of note, Duff et al. showed data for confocal studies and Western blot analysis of ZO-1 using Calu-3 cells, not 16HBE cells.
Blume et al. infected 16HBE and primary human bronchial epithelial cells (HBECs) with Burkholderia thailandensis (B. thailandensis) and found a dose- and time-dependent decrease in TEER. Additionally, they observed an increase in the bacterial traverse from the apical to the basolateral compartment, which was associated with a dose-dependent increase in TNF-α cytokine. The effect of B. thailandensis on TEER was marginally modified by pre-treatment with a TNF-α neutralizing agent (etanercept). In contrast, pre-treatment with a corticosteroid (fluticasone propionate) significantly prevented the decrease in TEER. These data suggest that TNF-α neutralizing agents or steroids might be a therapeutic option to maintain airway barrier functions and reduce inflammation [47].
One study compared the impact of two environmental strains with a clinical strain of B. cenocepacia on non-CF bronchial epithelial cells, 16HBE, and CF cell line, CFBE41o−. They observed a drop in TEER in cells exposed to environmental strains similar to the clinical strain. Interestingly, in this study, the extent of decrease in TEER was comparable between CF and non-CF cell lines. In contrast, the ZO-1 disruption was more pronounced in cells infected with clinical strain than environmental strains. The ZO-1 distribution seemed to be disrupted at baseline in the CF cell line, with more significant disruption in infected cells [48]. Another study infected the CFBE41o- cell line with B. contaminans strains and saw a time-dependent decrease in TEER and an increase in FITC-labeled BSA flux across the cell monolayers. They also observed reductions in ZO-1, occludin, and claudin-1 expressions [49].
Taken together, these studies show evidence that infection with Burkholderia species decreases airway epithelial barrier integrity and increases proinflammatory cytokines that contribute to bacteremia, infection dissemination, and airway inflammation.

2.5. Haemophilus Influenzae

Haemophilus influenzae (H. influenzae) is a Gram-negative, coccobacillus opportunistic anaerobic pathogenic bacterium of the family Pasteurellaceae [132]. There are two major strains of H. influenzae based on the presence or absence of a distinct capsular polysaccharide antigen [132]. The encapsulated strains are classified based on their specific capsular antigens into six distinct groups designated as serotypes a–f, with type b being the most common strain. The unencapsulated strains are termed non-typeable Haemophilus influenzae (NTHi) since they do not have a capsular serotype [133]. The encapsulated strains usually cause more invasive infections. While the unencapsulated strains are less invasive, they can trigger inflammatory responses in humans, leading to many symptoms. In the pre-vaccine era, H. influenzae type b (Hib) was the leading cause of severe bacterial infections such as pneumonia and meningitis among children. However, since the arrival of an effective Hib conjugate vaccine in 1988, the NTHi strains have been the most common cause of bacterial respiratory tract infection in young children as well as exacerbations of chronic obstructive pulmonary disease (COPD) and bronchiectasis in adults [133]. In addition, the NTHi can often cause more invasive diseases such as meningitis and sepsis. Currently, there are no vaccines to protect against NTHi, since there are many strains of NTHi with significant genetic heterogenicity; thus, the antibody against one particular strain does not protect against other strains [134].
The pathogeneses of NTHi are not well understood, but it is known that these bacteria possess several adhesive factors which promote colonization, leading to infection. Previous studies have shown that NTHi infection of the lower airways could cause chronic airway inflammation and even cause susceptibility to other infections [135,136]. Alveolar epithelium is composed of a mixed monolayer of type I and type II alveolar epithelial cells. Kaufhold et al. used an in vitro model of alveolar epithelial cells type II and A549 cells to study the role of NTHi on airway barrier function [50]. They showed that infection with NTHi decreased E-cadherin mRNA expression and protein levels. Additionally, immunofluorescence staining 24 h post-NTHi infection disrupted E-cadherin and ZO-1 structure. This was thought to be secondary to the upregulation of fibroblast growth factor 2 (FGF2) and activation of the mTOR pathway, since a pharmacological inhibitor of mTOR, rapamycin, prevented the reduction in E-cadherin expression [50].
It has been shown that NTHi activates the NF-κB signaling pathways by either NF-κB translocation-dependent or -independent pathways [137]. NF-κB is a known transcription factor regulating cell responses to inflammation. Studies conducted by Ward et al. showed that inhibiting IκB kinase, an enzyme complex that forms part of the NF-κB signaling pathway, triggered a dose-dependent decrease in TEER in the absence of cell death [138]. In a comprehensive study, the authors used primary rat type II alveolar epithelial cells to generate a model of type I cell monolayers and analyzed the effect of IκB kinase inhibitors on several TJ proteins. For example, immunofluorescence microscopy revealed the disruption of claudin-18, ZO-1, and ZO-2 as well as cortical actin cytoskeletal rearrangements and alteration of β-catenin protein [138]. Studying various TJ proteins, the investigators found an increase in claudin-4 and claudin-5 and a decrease in claudin-18 with altered TJ morphology. An increase in claudin-4 was thought to be a proinflammatory response. In contrast, a claudin-5 increase is associated with an increase in the paracellular leak in alveolar epithelial cells, and a decrease in claudin-18 correlates with an impaired barrier function [138,139,140,141,142]. There is evidence that the AJ protein E-cadherin expression is reduced in COPD patients [143]. A recent study by Glockner et al. analyzed the impact of NTHi on the re-differentiation of epithelial cells and airway remodeling. Using primary human HBECs grown at ALI infected with NTHi, they studied the E-cadherin as an epithelial marker [144]. An upregulation of mesenchymal marker vimentin in HBECs was observed, which suggests airway remodeling. They also found that NTHi infection did not change the expression or stability of E-cadherin. However, this study did not directly analyze the role of E-cadherin changes in the epithelial barrier.
Multiple groups have revealed the importance of airway epithelial barrier and differentiation in asthma [2,145,146]. Asthma was previously recognized as a T helper 2 (Th2) high eosinophilic inflammatory disease responding to glucocorticosteroids. However, a severe neutrophilic phenotype, not responsive to current therapeutic modalities, has been established in a large subgroup of asthmatic individuals [147]. A recent paper reviewed the role of persistent bacterial infection in asthma [148]. It summarized evidence that NTHi can adhere to respiratory tract epithelium, upregulate neutrophil chemokines and proinflammatory cytokines, and induce a persistent infection. Clinical trials have observed the benefits of using antimicrobials such as azithromycin in reducing asthma exacerbation, likely by decreasing the H. influenzae load and reducing inflammatory cytokines [149,150]. However, due to frequent side effects and the risk of antimicrobial resistance development, azithromycin has been considered only in a selected group of asthmatics [151]. This evidence, combined with the impact of NTHi on reducing the expression of TJ proteins, suggests that NTHi infection plays a critical role in inducing chronic neutrophilic asthma by impairing the airway epithelial barrier. Therefore, investigating and targeting chronic NTHi infection and airway dysfunction in severe persistent neutrophilic asthma is critical.

3. Viruses

3.1. Respiratory Syncytial Virus

Human respiratory syncytial virus (RSV) is an enveloped, negative-strand RNA virus of the genus Orthopneumovirus, family Pneumoviridae [152] that transmits through aerosolized droplets [153,154]. RSV infection affects the lungs and respiratory tract and often causes mild, cold-like symptoms in healthy populations. However, RSV can lead to severe infections and cause pneumonia or bronchiolitis in young children and other vulnerable groups, such as premature infants, older adults, and individuals with underlying immune, cardiac or pulmonary conditions [155]. As a common virus, RSV infection poses a significant burden worldwide. RSV is the leading cause of acute lower respiratory tract infection in infants and young children globally and a major cause of hospitalization and mortality in the elderly and immunocompromised population [156,157,158,159,160]. RSV infection has also been associated with persistent wheezing and asthma [161,162,163,164]. There are multiple other factors contributing to RSV pathogenesis and disease severity [165]. For example, the severity of RSV-associated symptoms is influenced by host factors such as Vitamin D level and genetic predispositions [166]. Environmental factors such as exposure to air pollution and tobacco smoke have also been recognized as key players with the potential to increase the risk of RSV infection and exacerbate clinical manifestations related to RSV [167,168,169,170,171].
The current management for RSV is primarily supportive because there are no effective antiviral therapies or vaccines against RSV yet [172,173]. So far, aerosolized ribavirin and palivizumab are the only two antiviral drugs approved by the U.S. FDA for treating or preventing severe respiratory tract infections caused by RSV [174]. Ribavirin is the only licensed drug to treat RSV infection; however, the clinical application of ribavirin is highly limited by its unproven efficacy, non-specificity, and potential toxicity [155,175,176]. Palivizumab is the only product available for preventing RSV infection despite rapid expansion in the development of RSV vaccines [157,173]. This monoclonal antibody (mAb) has been shown to reduce hospitalization, recurrent wheezing, and perhaps nonatopic asthma caused by RSV infection in certain high-risk infants [164,177]. However, the application of palivizumab is restricted to small high-risk populations, leaving the majority of infants unprotected [178,179]. These unmet clinical needs highlight the significance and urgency to understand the pathological mechanism of RSV infection.
The primary targets of RSV are ciliated airway epithelial cells and type I alveolar cells [180,181,182]. Transcription and replication start rapidly after RSV enters the epithelial cells, which is followed by the assembly and release of viral particles from host cells. Over time, the virus will breach the epithelial barrier and cause widespread inflammation and pulmonary damage if not cleared in time [1,15,183]. Using in vitro cultures of primary HBECs and human bronchial cell lines such as 16HBE, multiple groups, including us, showed that RSV infection decreased the TEER and increased the permeability of the monolayers without obvious cytopathology [15,18,51,52,53]. In rodent models, intranasal RSV infection increased the permeability of the airway epithelial barrier in C57BL/6 mice [15] and BALB/c mice [54].
The evident disassembly of AJC was observed during RSV infection and caused the “leaky” barrier in these models. Moreover, emerging evidence indicates that changes in the molecular components of epithelial TJs and AJs occur during RSV infection. Increased claudin-2 (a pore-forming claudin whose overexpression increases paracellular permeability), as well as decreased protein levels of ZO-1, occludin, and claudin-1, were described in lung tissues from RSV-infected C57BL/6 mice, while the levels of claudin-18, β-catenin, and E-cadherin were not significantly altered [15]. Another murine model of RSV infection exhibited decreased mRNA of claudin-1 and occludin in lung samples compared with control animals [54]. Interestingly, unlike in vivo observations, RSV infection did not impose significant impacts on the protein levels of TJ and AJ proteins in bronchial epithelial monolayers, including ZO-1, occludin, E-cadherin, and β-catenin [18]. Others examined RSV infection of cultured nasal epithelial monolayers and reported an increase in claudin-4 but no change in the expression of other AJC proteins [55]. This discrepancy might be caused by differences between in vitro and in vivo models of RSV infection, such as the inflammatory response to viral infection in vivo. For example, IL-4, IL-13, and IFN-γ are known to disrupt TJ integrity and decrease the expression of different junctional proteins [184,185,186]. Furthermore, these studies suggest alternative mechanisms for RSV-induced barrier dysfunction besides dysregulated expressions of TJ and AJ proteins.
Recently discovered evidence indicates that AJC disorganization by RSV could result from TJ/AJ structural rearrangements caused by perijunctional F-actin cytoskeletal remodeling. TJ and AJ are anchored to the actin cytoskeleton network, especially the perijunctional F-actin bundles, and it is well acknowledged that this connection is a critical regulator of AJC structure and epithelial barrier function [7,9,187]. For example, emerging evidence suggests that RSV induces the depolymerization of perijunctional F-actin, and pharmacologically stabilizing F-actin filaments modify RSV-induced barrier dysfunction [53]. Previously, our group showed evidence of protein kinase D (PKD) activation during RSV infection in 16HBE cells, while PKD antagonists protected cells from barrier disruption and AJC disassembly caused by RSV [18]. PKD has been implicated in cell adhesion, cell motility, and invasion via regulating actin remodeling [188,189,190]. Others have shown that PKD controls actin polymerization via phosphorylation of cortactin [191], which is an actin-binding protein and a key player in regulating F-actin dynamics and promoting actin assembly [192,193,194,195]. Indeed, cortactin phosphorylation was observed in association with PKD activation during RSV infection [18]. Therefore, the PKD pathway could be an upstream regulator of F-actin cytoskeletal remodeling whose activation disrupts AJC and airway epithelial barrier during RSV infection. RSV has been revealed to destabilize the perijunctional F-actin network and lead to a disrupted epithelial barrier by inhibiting cyclic adenosine monophosphate (cAMP)/Rap1/Rac1 signaling [53]. This study found that activating Rap1 by cAMP analog 8-pCPT-2-O-Me-cAMP-AM mitigated RSV-induced epithelial barrier disruption, which was in agreement with findings that elevating cAMP levels by adenylyl cyclase activator forskolin or cAMP analogs 8-Bromo-cAMP could protect airway barrier function and reduce RSV replication in cultured epithelial cells [51]. Considering the crucial role of cAMP/Rap1/Rac1 signaling in controlling the dynamics of the F-actin cytoskeleton, the cAMP pathway could be an essential mechanism involved in the RSV-induced reorganization of the F-actin network to interrupt the TJ and AJ. Collectively, these studies point out that RSV-induced AJC dysfunction could be regulated by the cAMP pathway and PKD pathway via perijunctional F-actin cytoskeletal remodeling. Moreover, cortactin expression decreased in RSV-infected cell monolayers and mouse airways and was implicated as the cause for the reduced activity of Rap1 [53], implying possible cross-talks between these two pathways. Accordingly, these studies provide insights into the therapeutic potentials of forskolin, cAMP analogs, and PKD inhibitors in RSV management through junction proteins, especially forskolin, which showed protective effects on the barrier integrity even when added 24 h after RSV inoculation [51].
Other signaling pathways are also implicated to be responsible for RSV-induced airway epithelial barrier disruption. For instance, the cleavage of E-cadherin and accumulation of soluble extracellular fragments of E-cadherin (referred to as soluble E-cadherin) were detected both in vivo and in vitro after RSV infection, and adding recombinant soluble E-cadherin to cell culture caused TJ disassembly in primary HBECs monolayers [15]. It is not clear about the protease(s) responsible for this cleavage, but it would be interesting to explore the impacts of limiting the soluble E-cadherin on RSV-induced barrier deficits. Others report that RSV infection increased bronchial epithelial monolayer permeability in vitro via inducing vascular endothelial growth factor (VEGF) [56] and activating the p38 MAPK pathway [52]. However, whether and how TJs and AJs are involved in these pathways warrant further investigations. Other cell types play an important role in RSV virulence and could be targeted for disease management. RSV induces the production of proinflammatory mediators in AECs and recruits immune cells, including neutrophils [196]. Indeed, significant neutrophil infiltration has been observed in the lungs of infants with severe RSV-induced bronchiolitis [197]. In a co-culture assay system of neutrophils and RSV-infected A549, Deng et al. found that trans-epithelial migration of neutrophils deteriorated RSV-induced epithelial barrier disruption [198].

3.2. Human Rhinovirus

First identified in the 1950s, human rhinovirus (HRV) is a member of the Enterovirus genus in the Picornaviridae family [199,200]. This non-enveloped virus contains a positive-sense, single-stranded RNA [200,201]. As a ubiquitous virus, HRV transmits through direct contact or aerosol [202] and causes infections year-round [203,204,205]. HRVs are the most common cause of upper respiratory tract infections [206,207]. Although HRV is not typically considered to cause high mortality, it has been implicated in lower respiratory diseases including pneumonia, bronchiolitis, asthma, and COPD [208,209,210,211,212,213], indicating its potential to cause acute or chronic respiratory diseases. Currently, HRV infection management is mainly supportive care, because there is no affordable and effective therapy against RVs [214]. Numerous antiviral reagents against HRVs have been tested but showed limited efficiency or severe side effects [207]. At the same time, the development of an HRV vaccine is precluded by the number of different HRV serotypes [214].
The primary targets of HRV infection and replication in humans are AECs in both upper and lower airways [215,216,217]. HRV is reported to infect both ciliated and non-ciliated epithelial cells [218]. HRV has been described to impair barrier integrity by disrupting TJs and AJs. Previous studies in polarized primary human AECs and 16HBE cells showed that HRV infection reduced TEER and increased paracellular permeability without causing detectable cytopathology [57,58,59,60]. However, others observed a cytopathic effect of HRV infection in association with comprised barrier integrity [61,219]. Despite the inconsistent observations in cell toxicity, ample evidence indicates that HRV infection-dependent impairment of the airway epithelial barrier correlates with disruption of the expression and localization of TJ/AJ proteins. Both in vitro and in vivo studies have shown that HRV caused the dissociation of ZO-1 from TJ and consequently led to the loss of epithelial barrier integrity of nasal epithelial cells [58]. In primary HNECs grown at ALI, Yeo and Jang observed decreased mRNA and protein levels of ZO-1, occludin, claudin-1, and E-cadherin after HRV infection [62]. Similarly, another study reported HRV-induced reduction in ZO-1, occludin, claudin, and E-cadherin proteins in primary HNECs [63]. Consistently, the reduced expression of Crumbs 3, ZO-1, occludin, claudins 1 and 4, and E-cadherin was observed after HRV infection in an in vitro model of injured/regenerating airway epithelium [57]. Looi et al. also observed a reduced protein expression of TJ proteins ZO-1, occludin, and claudin-1 following HRV infection in both human airway epithelial cell line NuLi-1 and primary AECs obtained from patients [61]. Interestingly, HRV infection caused a transient dissociation of TJ in primary AECs from non-asthmatic children but a significant reduction in ZO-1 and occludin expression in cells from asthmatic children [64], correlating to the relationship between HRV infection and exacerbated barrier dysfunction in children with asthma. In HRV-infected human precision-cut lung slices (PCLS), the gene expression of claudin-8 was significantly downregulated compared with that in control tissues [65]. This series of evidence implies a critical role of HRV-induced disruption of AJC in the development and progress of HRV-related respiratory diseases.
Understanding the molecular mechanisms of HRV-induced AJC disassembly and downregulation of TJ/AJ protein expressions is important to the discovery of new therapeutic targets. Oxidative stress could lead to the impairment of airway epithelial barrier function [167,220,221], and HRV has been shown to induce oxidative stress in AECs [222]. Previous studies revealed that HRV disrupted the barrier function of polarized 16HBE monolayer by stimulating reactive oxygen species (ROS) generation, possibly through dsRNA generated during HRV replication [66,67]. NADPH oxidase (NOX) mediates the production of ROS and contributes to respiratory viruses-induced barrier disruption through inflammatory pathways [223,224]. Comstock et al. reported that HRV infection induced ROS generation, and inhibiting NOX or NOX1 blocked the disruptive effects of HRV on barrier integrity and the dissociation of ZO-1 and occludin from AJC [66]. In addition to the NOX-1-dependent ROS mechanism, Unger et al. provided evidence that mitochondrial ROS generation stimulated by HRV also contributes to the disruption of airway epithelial barrier function [67]. Consistently, Kim et al. reported that HRV decreased TJ/AJ proteins in primary HNECs by inducing NOX-derived ROS production. They showed that HRV infection induced NOX-derived ROS production, and inhibiting NOX prevented an HRV-induced decrease in TJ/AJ proteins. Furthermore, the ROS-dependent inhibition of phosphatases is closely linked to reduced TJ/AJ proteins caused by HRV [63]. Taken together, diminishing ROS might show protective effects on epithelial cells against HRV and serve as a therapeutic strategy. Indeed, the role of anti-oxidants in HRV-induced barrier disruption has been investigated. Pretreatment with Ginsenoside Re, a ginsenoside found in Panax ginseng with anti-oxidant actions [225], decreased HRV-induced disruption of TJ/AJ proteins through inhibiting ROS-mediated phosphatases inactivation in HNECs [68]. Wogonin is a flavonoid-like chemical with anti-oxidant and anti-inflammatory properties [226]. A recent study revealed that wogonin prevented increases in intracellular ROS following HRV infection and alleviated the decreases of AJ/TJ proteins by suppressing the phosphorylation of ROS-mediated pathways Akt/NF-κB and ERK1/2 in HNECs [69]. In addition to oxidative stress, other mechanisms have also been implicated in mediating HRV-induced AJC changes. The upregulated expression of Snail, a transcriptional repressor of TJ and AJ proteins, was reported after HRV infection of injured/regenerating airway epithelium [57]. Whether Snail contributes to HRV-induced TJ/AJ change in regular AECs still warrants further investigation. The qPCR arrays and pathway analysis were performed on HRV-infected cells to evaluate associations between human airway epithelial TJ proteins and antiviral response. Network analysis suggested that HRV infection decreased TJ protein and increased epithelial permeability potentially via antiviral responses of IL-15 [61]; however, further experiments, including the addition of neutralizing antibodies, are needed to provide additional evidence for therapeutic potentials. Emerging omics approaches have also provided some new perspectives. For example, proteomic and metabolomic analysis of HBECs at ALI characterized altered metabolic pathways during HRV infection. This investigation revealed PGC-1α, a key mitochondrial biogenesis transcriptional coactivator, as a novel antiviral target, as investigators found that several HRV strains reduced PGC-1α expression, and promoted PGC-1α expression could restore barrier function during HRV infection [60].

3.3. Influenza Viruses

Influenza viruses are enveloped, negative-sense RNA viruses with a segmented genome [200,227]. As the major representative of the Orthomyxoviridae family, influenza viruses are grouped into four genera: influenzavirus A, influenzavirus B, influenzavirus C, and influenzavirus D. [200]. Three types of influenza viruses (A, B, and C) infect humans. Type A and B viruses cause significant morbidity and mortality annually worldwide and are referred to as seasonal influenza viruses [228]. Influenza A (IAV) and B viruses are generally believed to be transmitted at a short range (1–2 m) from person to person through large droplets and aerosols [229]. The clinical manifestations of seasonal influenza cover a wide spectrum, from asymptomatic infection, and uncomplicated upper-respiratory-tract symptoms to complications that can result in severe diseases such as lethal pneumonia, acute respiratory distress syndrome (ARDS), and secondary bacterial infection of the lower respiratory tract [227,228,230,231]. IAVs are the only influenza viruses known to cause flu pandemics and have been the focus of investigations. The World Health Organization (WHO) estimated that annual influenza epidemics cause 3–5 million cases of severe illnesses and up to 300,000–650,000 deaths worldwide [174,232]. In the United States alone, the 2019–2020 influenza season was estimated to cause 16 million medical visits and 25,000 deaths [233]. The segmented genome of influenza viruses enables antigenic shift through reassortment, which mediates cross-species transmission and is associated with influenza A pandemics. Influenza virus also evolves through accumulating mutations, which is a process known as antigenic drift caused by RNA polymerase infidelity [227,234]. Due to such genetic diversity and rapid viral evolution, influenza infections remain a severe threat to public health despite effective antiviral treatments and vaccines being available.
Human influenza viruses target epithelial cells of the upper and lower respiratory tract for infection and replication [227,235]. IAV infection has been well-documented to damage epithelial junctions and impair the barrier function of airway epithelium [113]. In well-differentiated normal human bronchial epithelial (NHBE) cells, infection with the 2009 pandemic IAV/H1N1pdm09 strain led to a decline in barrier function, which was marked by a decrease in TEER and an increase in permeability [17]. This study also showed disrupted staining of ZO-1 and F-actin cytoskeleton as well as cell death after IAV infection. In another study, primary human alveolar type II cells were infected by two strains of H1N1pdm09 viruses, and researchers observed a similar decrease in TEER and an increase in barrier permeability [70]. In an in vitro co-culture model of human alveolar epithelial cells and endothelial cells, Short et al. showed that IAV infection damaged the barrier integrity independently of endothelial cells [16]. Although IAV infection of epithelial cells can induce cell death [236,237], this study detected no apparent cell death after IAV infection, indicating that the barrier disruption was not due to cell death. The investigators also reported that significantly decreased TEER and increased barrier permeability caused by IAV infection were associated with the disruption of TJs amongst epithelial cells, specifically with loss of TJ protein claudin-4 [16]. Interestingly, they found that the barrier damage caused by IAV was independent of the activation of proinflammatory cytokines, indicating the direct effect of IAV in reducing claudin-4 expression through unknown mechanisms.
Despite these observations, the molecular mechanisms of IAV-induced damage to the epithelial barrier are far from well understood. Using cultured A549 cells and an in vivo mouse model, Ruan et al. revealed that IAV H1N1 infection activated MAPK and PI3K signaling, which induced the activation of Gli1, a transcription factor in the sonic hedgehog signaling pathway, and subsequent Snail expression [71]. Increased Snail decreased the expression of AJ protein E-cadherin and TJ proteins ZO-1 and occludin and increased paracellular permeability. Intraperitoneal injection of GANT61, a Gli1 Inhibitor, blocked IAV-Induced expression of Gli1 and Snail and restored expression of E-cadherin and occludin in vivo. GANT61 treatment also suppressed the pathological changes in IAV H1N1-infected lungs [71]. Whether Gli1 can be targeted for antiviral therapy for the H1N1 virus is still unknown and warrants further investigation. The H5N1 virus is a newly emerged strain of IAV from animal reservoirs and raised great concerns about a severe pandemic. A recent study reported that H5N1 viruses reduced the expression of ZO-1, occludin, claudin-1, and E-cadherin in A549 cells, NL20 cells (a human noncancerous alveolar epithelial cell line), and the lungs of H5N1 virus-infected mice [72]. The authors showed evidence that H5N1 virus infection impaired the alveolar epithelial barrier by accelerating the turnover of several AJC proteins. This study suggested a mechanism that H5N1 viruses activated TAK1 and its downstream MAPK, p38, and ERK, leading to the increased expression of E3 ubiquitin ligase Itch and ubiquitination of occludin, claudin-1, and E-cadherin proteins. Furthermore, inhibiting the TAK1–Itch pathway restored the epithelial barrier structure and function in vitro and in vivo [72]. However, whether the TAK1–Itch pathway can be a valid antiviral targeted for the H5N1 virus is uncertain.

3.4. Human Parainfluenza Virus

Human parainfluenza viruses (HPIVs) are enveloped, negative-sense, single-stranded RNA viruses and members of the Paramyxoviridae family. HPIVs are classified into four distinct serotypes: HPIV-1, HPIV-2, HPIV-3, and HPIV-4 [238,239]. In addition, another parainfluenza virus 5 (PIV5) infects animals such as dogs, pigs, cats, and sometimes humans [240]. HPIVs cause illnesses such as pharyngitis, croup, tracheobronchitis, bronchiolitis, croup, pneumonia, and respiratory distress syndrome. HPIVs transmission occurs through direct person-to-person contact or large droplets or indirect contact via contaminated surfaces [241].
HPIV infects the ciliated epithelial cells and causes inflammation, which contributes to the disease pathogenesis. A couple of studies have explored the impact of tight HPIV infectivity on AEC barrier integrity. One study showed that the exposure of AECs to HPIV3 and PIV5 triggered a reduction in TEER and increased permeability. The study did not analyze the AJC structure or involved pathways [73]. Another study infected human alveolar epithelial cells (A549 cells) with HPIV2 and found an induction of claudin-1 mRNA expression by HPIV2. However, there were no changes in the expression of other TJs, such as ZO-1, ZO-2, occludin, claudin-3, claudin-4, or claudin-7 [74]. The claudin-1 upregulation was thought to be partly induced by a functional HPIV2 V protein, which impacts viral growth, modulates TJ molecules, and facilitates efficient virus propagation. They used a rPIV2 VC193/197A, which expressed a mutant V protein and observed an enhancement of CLDN1 mRNA upregulation compared to the wild type. To investigate the importance of claudin-1, they used A549 cell lines that stably over-express CLDN1 and found a decrease in HPIV2 growth and protein expression. Furthermore, in Madin–Darby canine kidney (MDCK) cells, CLDN1 knockout provoked a significant increase in HPIV2 growth and protein expression. Similarly, HPIV2 cell-to-cell spread deceased in A549, overexpressing the CLDN1 and increased in MDCK CLDN1-KO cells, emphasizing the role of claudin-1 [74]. At present, there are no licensed vaccines or antiviral drugs for HPIV infection. Therefore, gaining knowledge of the molecular mechanisms, particularly the role of claudin-1, can aid in developing treatment options to dampen HPIVs growth and spread.

3.5. Coronaviruses

Coronaviruses are enveloped, single-stranded, positive-sense RNA viruses of the Coronaviridae family [200]. Seven human coronaviruses (HCoV) have been characterized to date. HCoV-NL63, HCoV-229E, HCoV-OC43, and HCoV-HKU1 commonly cause mild upper respiratory illnesses. In contrast, severe acute respiratory syndrome coronavirus (SARS-CoV), Middle East respiratory syndrome coronavirus (MERS-CoV), and severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) are highly pathogenic and have resulted in outbreaks with high mortality rates [242,243].
SARS-CoV was first reported in southern China in February 2003 [244,245,246] and spread to over 30 countries, leading to over 8000 confirmed cases with a mortality rate of ~9.6% by the end of the epidemic in June 2003 [247,248]. MERS-CoV emerged in Saudi Arabia in 2012 [249,250] and has since caused over 2000 confirmed cases with a mortality rate of ~35% [243,248,251]. SARS-CoV-2 was first identified in China in December 2019 [252] and has caused a massive global pandemic of coronavirus disease 2019 (COVID-19), infecting more than 634 million people and resulting in more than 6.5 million deaths worldwide [253]. The human-to-human transmissibility of MERS-CoV is low [254], whereas SARS-CoV and SARS-CoV-2 readily transmit via the respiratory tract through close contact with infectious droplets and aerosols [255,256]. The clinical manifestations of SARS, MERS, and COVID-19 share many similarities; acute respiratory tract infections are the most common symptoms and could lead to rapid respiratory failure and mortality [243,257]. To date, there is no vaccine or specific antiviral drug available for either SARS-CoV or MERS-CoV [174,248]. The rapid scientific response to COVID-19 enabled the development of highly effective vaccines and drugs at an unparalleled speed [258]. Currently, COVID-19 management includes antivirals such as Molnupiravir and Paxlovid in early stages and immunotherapeutic agents such as IFNs and corticosteroids in more advanced stages [259,260,261]. Despite the significant progress in combating SARS-CoV-2, the emergence of its mutant variants has become a concerning issue and threat to public health.
SARS-CoV utilizes the angiotensin-converting enzyme 2 (ACE2) as a receptor and primarily infects ciliated bronchial epithelial cells and type II alveolar epithelial cells [262,263]. MERS-CoV binds to dipeptidyl peptidase 4 (DPP4; also known as CD26) and exhibits wide tissue and cell tropism [264,265]. SARS-CoV-2 also uses ACE2 for entry [266] but has a broader cell tropism than SARS-CoV. In addition to ciliated epithelial cells and type II alveolar epithelial cells in the lungs, SARS-CoV-2 also infects intestinal epithelial cells and brain cells [267]. Zhu et al. showed that SARS-CoV-2 infection induced cytopathic effects and decreased TEER in organotypic human airway epithelial cultures. However, the effect of the virus on TJ and AJ was not examined in this study [76]. One study used a primary human bronchial epithelium model and showed that SARS-CoV-2 infection caused a transient decrease in TEER and increased paracellular permeability for dextran-FITC, where disrupted ZO-1 staining pattern and apoptosis were observed in infected epithelial cells [77]. The major cause of respiratory failure is damage to the epithelial–endothelial barrier of the alveolus [268,269]. Deinhardt-Emmer et al. elucidated that SARS-CoV-2 damaged the epithelial/endothelial barrier and induced robust immune reactions using a human chip model composed of epithelial, endothelial, and mononuclear cells. In this study, immunostaining of E-cadherin and scanning electron microscopy showed a disruption in epithelial barrier structure after viral infection. The investigators also noted that the epithelial/endothelial barrier permeability to FITC-dextran significantly increased [78].
Efforts to understand the impact of coronaviruses on the airway epithelial barrier have revealed some possible underlying mechanisms. In a recent publication, Rouaud et al. showed that ACE2 is localized and concentrated in the epithelial apical cell junction, which could facilitate the internalization of the virus across the airway epithelial cell barriers [270]. It has been reported that the SARS-CoV envelope (E) protein binds to human Proteins Associated with Lin Seven 1 (PALS1), which is a tight junction-associated protein crucial for the establishment and maintenance of epithelial polarity. The ectopic expression of the SARS-CoV E protein delayed the formation of TJs in kidney epithelial cell monolayers [75]. PALS1 was also shown to facilitate the intracellular traffic of E-cadherin in kidney epithelial cells, and the loss of PALS1 led to severe TJ and AJ disruption [271]. Interestingly, De Maio et al. predicted that the SARS-CoV-2 E protein binds more stably with PALS1 compared to SARS-CoV and could cause enhanced epithelial barrier disruption contributing to the pathogenesis of COVID-19 [272]. Moreover, there is evidence that the C-terminal domain of the SARS-CoV-2 E protein binds to the second PDZ domain of ZO-1 [273]. Although experimental evidence is still lacking, it is possible that SARS-CoV and the SARS-CoV-2 E protein bind to PALS1 and disrupt its trafficking to TJ and the proper trafficking of E-cadherin and ZO-1 to AJC in airway epithelial cells, resulting in the loss of barrier integrity. SARS-CoV-2 infection results in the downregulation of claudin-18a in induced pluripotent stem cell-derived alveolar epithelial type II cells, which might play a role in the loss of alveolar epithelial barrier function and pulmonary edema in COVID-19 patients [274]. Recent bioinformatics analysis identified the integral role of cell junction and cytoskeletal genes in COVID-19 and suggested their therapeutic potential [275]. SARS-CoV-2 infection also activates the release of proinflammatory cytokines, and their potential roles in barrier disruption have been reviewed by others [276]. Compared with the emerging studies on SARS-CoV and SARS-CoV-2, the impact of MERS on the airway epithelial barrier is not very well understood. As the COVID-19 pandemic is still an ongoing global public health problem, further investigation is needed to understand the pathogenesis of coronavirus and provide more information for drug development, especially the impact on epithelial barriers in both cultured cells and animal models.

4. Conclusions

With a large surface in direct exposure to the environment, the human respiratory epithelial barrier plays a crucial part in the host defense mechanisms against inhaled stimulants. Breaking the airway epithelial barrier is the stepping stone for environmental pathogens to establish infection in the respiratory tract. Mounting evidence has shown that impairing the AJC, a key contributing factor to cell–cell contact in the respiratory epithelium, is an important strategy used by bacteria and viruses to disrupt the integrity of the airway epithelial barrier. Here, we reviewed the recent findings regarding the impacts on AJC of a selected group of bacteria and respiratory viruses that impose substantial health and economic burdens. We highlighted the growing knowledge regarding the molecular mechanisms by which these microorganisms cause the dysregulation of AJCs and breach of the airway epithelial barrier. The reduced expression or mislocalization of TJ components ZO-1, occludin, and several claudin family members, as well as AJ proteins E-cadherin and β-catenin, have been highly implicated in pathogen-induced barrier dysfunction. Manipulating some of the underlying molecular pathways showed promising potential in reinstating epithelial barrier function. These advances present unique opportunities for the development of clinically relevant antimicrobial therapies. Targeting the host cells may also help to address the challenges of emerging pathogen resistance and the existence of multiple serological types.
Despite the growing knowledge of the mechanisms involved, their applications in mitigating epithelial barrier dysfunction in disease states require further and thorough examination. So far, most investigations have been conducted with in vitro models of the airway epithelial barrier. Future studies that examine the junctional structure and function in appropriate animal models or using 3D cell culture models such as organoids will be imperative to validate the mechanisms in vivo. Rapid advances in single-cell sequencing and spatial transcriptomics technologies will enable the molecular analysis of changes in the airway epithelial barrier with significantly improved time and spatial resolutions, which we foresee could accelerate the development of AJC-focused antimicrobial treatments. Identifying the molecular pathways that enhance AJC function will also be of earnest interest for its implications in the design of novel antimicrobial strategies.

Author Contributions

Conceptualization, N.G. and F.R.; Writing—Original Draft Preparation, N.G. and F.R.; Writing—Review and Editing, N.G. and F.R.; Visualization, N.G. and F.R.; Supervision, F.R.; Funding Acquisition, N.G. and F.R. All authors have read and agreed to the published version of the manuscript.

Funding

This work was funded by the Mark Lauer Pediatric Research Grant, Cleveland Clinic Children’s 2021–2023, (to N.G.) and National Heart, Lung, and Blood Institute Grant R01-HL-148057 (to F.R.).

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Conflicts of Interest

The authors declare no conflict of interest.

Abbreviations

16HBE16HBE14o- human bronchial epithelial
ADAM10metalloproteinase domain-containing protein 10
AECsairway epithelial cells
ALIair–liquid interface
AJadherens junction
AJCapical junctional complex
BSAbovine serum albumin
cAMPcyclic adenosine monophosphate
CARcoxsackie and adenovirus receptor
CDCCenters for Disease Control and Prevention
CFcystic fibrosis
CFTRcystic fibrosis transmembrane conductance regulator
COPDchronic obstructive pulmonary disease
COVID-19Coronavirus disease 2019
FAKfocal adhesion kinase
FITCfluorescein isothiocyanate
HBECshuman bronchial epithelial cells
HNECshuman nasal epithelial cells
HPIVhuman parainfluenza virus
HRVhuman rhinovirus
IAVinfluenza A virus
ICUintensive care unit
IFNinterferon
JAMjunctional adhesion molecule
MAPKmitogen-activated protein kinase
MDCKMadin–Darby canine kidney
MERS-CoVMiddle East respiratory syndrome coronavirus
MRSAmethicillin-resistant Staphylococcus aureus
NHBEnormal human bronchial epithelial
NOXNADPH oxidase
PALS1Proteins Associated with Lin Seven 1
PI3Kphosphoinositide 3-kinase
PKDprotein kinase D
PTKProtein–tyrosine kinase
ROSreactive oxygen species
RSVrespiratory syncytial virus
SARS-CoVsevere acute respiratory syndrome coronavirus
SARS-CoV-2severe acute respiratory syndrome coronavirus-2
Srcsteroid receptor coactivator
TEERtrans-epithelial electrical resistance
TLRToll-like receptor
TJtight junction
WHOWorld Health Organization
ZOzonula occludens

References

  1. Linfield, D.T.; Raduka, A.; Aghapour, M.; Rezaee, F. Airway tight junctions as targets of viral infections. Tissue Barriers 2021, 9, 1883965. [Google Scholar] [CrossRef] [PubMed]
  2. Rezaee, F.; Georas, S.N. Breaking barriers. New insights into airway epithelial barrier function in health and disease. Am. J. Respir. Cell Mol. Biol. 2014, 50, 857–869. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  3. Davis, J.D.; Wypych, T.P. Cellular and functional heterogeneity of the airway epithelium. Mucosal Immunol. 2021, 14, 978–990. [Google Scholar] [CrossRef] [PubMed]
  4. Hewitt, R.J.; Lloyd, C.M. Regulation of immune responses by the airway epithelial cell landscape. Nat. Rev. Immunol. 2021, 21, 347–362. [Google Scholar] [CrossRef] [PubMed]
  5. Ganesan, S.; Comstock, A.T.; Sajjan, U.S. Barrier function of airway tract epithelium. Tissue Barriers 2013, 1, e24997. [Google Scholar] [CrossRef] [Green Version]
  6. Garcia, M.A.; Nelson, W.J.; Chavez, N. Cell-Cell Junctions Organize Structural and Signaling Networks. Cold Spring Harb. Perspect. Biol. 2018, 10, a029181. [Google Scholar] [CrossRef] [Green Version]
  7. Rusu, A.D.; Georgiou, M. The multifarious regulation of the apical junctional complex. Open Biol. 2020, 10, 190278. [Google Scholar] [CrossRef]
  8. Tsukita, S.; Tanaka, H.; Tamura, A. The Claudins: From Tight Junctions to Biological Systems. Trends Biochem. Sci. 2019, 44, 141–152. [Google Scholar] [CrossRef]
  9. Zihni, C.; Mills, C.; Matter, K.; Balda, M.S. Tight junctions: From simple barriers to multifunctional molecular gates. Nat. Rev. Mol. Cell Biol. 2016, 17, 564–580. [Google Scholar] [CrossRef]
  10. Van Itallie, C.M.; Fanning, A.S.; Bridges, A.; Anderson, J.M. ZO-1 stabilizes the tight junction solute barrier through coupling to the perijunctional cytoskeleton. Mol. Biol. Cell 2009, 20, 3930–3940. [Google Scholar] [CrossRef]
  11. Fanning, A.S.; Van Itallie, C.M.; Anderson, J.M. Zonula occludens-1 and -2 regulate apical cell structure and the zonula adherens cytoskeleton in polarized epithelia. Mol. Biol. Cell 2012, 23, 577–590. [Google Scholar] [CrossRef]
  12. Fanning, A.S.; Jameson, B.J.; Jesaitis, L.A.; Anderson, J.M. The Tight Junction Protein ZO-1 Establishes a Link between the Transmembrane Protein Occludin and the Actin Cytoskeleton*. J. Biol. Chem. 1998, 273, 29745–29753. [Google Scholar] [CrossRef] [Green Version]
  13. Wittchen, E.S.; Haskins, J.; Stevenson, B.R. Protein Interactions at the Tight Junction: Actin has multiple binding partners, and ZO-1 forms independent complexes with ZO-2 AND ZO-3*. J. Biol. Chem. 1999, 274, 35179–35185. [Google Scholar] [CrossRef] [Green Version]
  14. Takeichi, M. Dynamic contacts: Rearranging adherens junctions to drive epithelial remodelling. Nat. Rev. Mol. Cell Biol. 2014, 15, 397–410. [Google Scholar] [CrossRef]
  15. Smallcombe, C.C.; Linfield, D.T.; Harford, T.J.; Bokun, V.; Ivanov, A.I.; Piedimonte, G.; Rezaee, F. Disruption of the airway epithelial barrier in a murine model of respiratory syncytial virus infection. Am. J. Physiol. Lung Cell. Mol. Physiol. 2019, 316, L358–L368. [Google Scholar] [CrossRef]
  16. Short, K.R.; Kasper, J.; van der Aa, S.; Andeweg, A.C.; Zaaraoui-Boutahar, F.; Goeijenbier, M.; Richard, M.; Herold, S.; Becker, C.; Scott, D.P.; et al. Influenza virus damages the alveolar barrier by disrupting epithelial cell tight junctions. Eur. Respir. J. 2016, 47, 954–966. [Google Scholar] [CrossRef] [Green Version]
  17. Pharo, E.A.; Williams, S.M.; Boyd, V.; Sundaramoorthy, V.; Durr, P.A.; Baker, M.L. Host–Pathogen Responses to Pandemic Influenza H1N1pdm09 in a Human Respiratory Airway Model. Viruses 2020, 12, 679. [Google Scholar] [CrossRef]
  18. Rezaee, F.; DeSando, S.A.; Ivanov, A.I.; Chapman, T.J.; Knowlden, S.A.; Beck, L.A.; Georas, S.N. Sustained Protein Kinase D Activation Mediates Respiratory Syncytial Virus-Induced Airway Barrier Disruption. J. Virol. 2013, 87, 11088–11095. [Google Scholar] [CrossRef] [Green Version]
  19. Chiu, C.-C.; Shi, Y.-F.; Yang, J.-J.; Hsiao, Y.-C.; Tzang, B.-S.; Hsu, T.-C. Effects of Human Parvovirus B19 and Bocavirus VP1 Unique Region on Tight Junction of Human Airway Epithelial A549 Cells. PLoS ONE 2014, 9, e107970. [Google Scholar] [CrossRef]
  20. LeMessurier, K.S.; Häcker, H.; Chi, L.; Tuomanen, E.; Redecke, V. Type I Interferon Protects against Pneumococcal Invasive Disease by Inhibiting Bacterial Transmigration across the Lung. PLoS Pathog. 2013, 9, e1003727. [Google Scholar] [CrossRef]
  21. Martens, K.; Seys, S.F.; Alpizar, Y.A.; Schrijvers, R.; Bullens, D.M.A.; Breynaert, C.; Lebeer, S.; Steelant, B. Staphylococcus aureus enterotoxin B disrupts nasal epithelial barrier integrity. Clin. Exp. Allergy 2021, 51, 87–98. [Google Scholar] [CrossRef] [PubMed]
  22. Hakansson, A.P.; Orihuela, C.J.; Bogaert, D. Bacterial-Host Interactions: Physiology and Pathophysiology of Respiratory Infection. Physiol. Rev. 2018, 98, 781–811. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  23. Eisele, N.A.; Anderson, D.M. Host Defense and the Airway Epithelium: Frontline Responses That Protect against Bacterial Invasion and Pneumonia. J. Pathog. 2011, 2011, 249802. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  24. Maginnis, M.S. Virus-Receptor Interactions: The Key to Cellular Invasion. J. Mol. Biol. 2018, 430, 2590–2611. [Google Scholar] [CrossRef] [PubMed]
  25. Torres-Flores, J.M.; Arias, C.F. Tight Junctions Go Viral! Viruses 2015, 7, 5145–5154. [Google Scholar] [CrossRef]
  26. Malik, Z.; Roscioli, E.; Murphy, J.; Ou, J.; Bassiouni, A.; Wormald, P.-J.; Vreugde, S. Staphylococcus aureus impairs the airway epithelial barrier in vitro. Int. Forum Allergy Rhinol. 2015, 5, 551–556. [Google Scholar] [CrossRef]
  27. Murphy, J.; Ramezanpour, M.; Stach, N.; Dubin, G.; Psaltis, A.J.; Wormald, P.J.; Vreugde, S. Staphylococcus aureus V8 protease disrupts the integrity of the airway epithelial barrier and impairs IL-6 production in vitro. Laryngoscope 2018, 128, E8–E15. [Google Scholar] [CrossRef]
  28. Kalsi, K.K.; Garnett, J.P.; Patkee, W.; Weekes, A.; Dockrell, M.E.; Baker, E.H.; Baines, D.L. Metformin attenuates the effect of Staphylococcus aureus on airway tight junctions by increasing PKCζ-mediated phosphorylation of occludin. J. Cell. Mol. Med. 2019, 23, 317–327. [Google Scholar] [CrossRef] [Green Version]
  29. Inoshima, I.; Inoshima, N.; Wilke, G.A.; Powers, M.E.; Frank, K.M.; Wang, Y.; Wardenburg, J.B. A Staphylococcus aureus pore-forming toxin subverts the activity of ADAM10 to cause lethal infection in mice. Nat. Med. 2011, 17, 1310–1314. [Google Scholar] [CrossRef] [Green Version]
  30. Wilke, G.A.; Wardenburg, J.B. Role of a disintegrin and metalloprotease 10 in Staphylococcus aureus alpha-hemolysin mediated cellular injury. Proc. Natl. Acad. Sci. USA 2010, 107, 13473–13478. [Google Scholar] [CrossRef]
  31. Ziesemer, S.; Eiffler, I.; Schönberg, A.; Müller, C.; Hochgräfe, F.; Beule, A.G.; Hildebrandt, J.P. Staphylococcus aureus α-Toxin Induces Actin Filament Remodeling in Human Airway Epithelial Model Cells. Am. J. Respir. Cell Mol. Biol. 2018, 58, 482–491. [Google Scholar] [CrossRef]
  32. Clarke, T.B.; Francella, N.; Huegel, A.; Weiser, J.N. Invasive Bacterial Pathogens Exploit TLR-Mediated Downregulation of Tight Junction Components to Facilitate Translocation across the Epithelium. Cell Host Microbe 2011, 9, 404–414. [Google Scholar] [CrossRef] [Green Version]
  33. Rayner, C.F.; Jackson, A.D.; Rutman, A.; Dewar, A.; Mitchell, T.J.; Andrew, P.W.; Cole, P.J.; Wilson, R. Interaction of pneumolysin-sufficient and -deficient isogenic variants of Streptococcus pneumoniae with human respiratory mucosa. Infect. Immun. 1995, 63, 442–447. [Google Scholar] [CrossRef] [Green Version]
  34. Mo, D.; Xu, S.; Rosa, J.P.; Hasan, S.; Adams, W. Dynamic Python-Based Method Provides Quantitative Analysis of Intercellular Junction Organization During S. pneumoniae Infection of the Respiratory Epithelium. Front. Cell. Infect. Microbiol. 2022, 12, 865528. [Google Scholar] [CrossRef]
  35. Peter, A.; Fatykhova, D.; Kershaw, O.; Gruber, A.D.; Rueckert, J.; Neudecker, J.; Toennies, M.; Bauer, T.T.; Schneider, P.; Schimek, M.; et al. Localization and pneumococcal alteration of junction proteins in the human alveolar-capillary compartment. Histochem. Cell Biol. 2017, 147, 707–719. [Google Scholar] [CrossRef]
  36. Attali, C.; Durmort, C.; Vernet, T.; Di Guilmi, A.M. The interaction of Streptococcus pneumoniae with plasmin mediates transmigration across endothelial and epithelial monolayers by intercellular junction cleavage. Infect. Immun. 2008, 76, 5350–5356. [Google Scholar] [CrossRef] [Green Version]
  37. Patkee, W.R.; Carr, G.; Baker, E.H.; Baines, D.L.; Garnett, J.P. Metformin prevents the effects of Pseudomonas aeruginosa on airway epithelial tight junctions and restricts hyperglycaemia-induced bacterial growth. J. Cell. Mol. Med. 2016, 20, 758–764. [Google Scholar] [CrossRef] [Green Version]
  38. Rejman, J.; Di Gioia, S.; Bragonzi, A.; Conese, M. Pseudomonas aeruginosa infection destroys the barrier function of lung epithelium and enhances polyplex-mediated transfection. Hum. Gene Ther. 2007, 18, 642–652. [Google Scholar] [CrossRef]
  39. Nomura, K.; Obata, K.; Keira, T.; Miyata, R.; Hirakawa, S.; Takano, K.-i.; Kohno, T.; Sawada, N.; Himi, T.; Kojima, T. Pseudomonas aeruginosa elastase causes transient disruption of tight junctions and downregulation of PAR-2 in human nasal epithelial cells. Respir. Res. 2014, 15, 21. [Google Scholar] [CrossRef] [Green Version]
  40. Clark, C.A.; Thomas, L.K.; Azghani, A.O. Inhibition of protein kinase C attenuates Pseudomonas aeruginosa elastase-induced epithelial barrier disruption. Am. J. Respir. Cell Mol. Biol. 2011, 45, 1263–1271. [Google Scholar] [CrossRef]
  41. Zulianello, L.; Canard, C.; Kohler, T.; Caille, D.; Lacroix, J.S.; Meda, P. Rhamnolipids are virulence factors that promote early infiltration of primary human airway epithelia by Pseudomonas aeruginosa. Infect. Immun. 2006, 74, 3134–3147. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  42. Soong, G.; Parker, D.; Magargee, M.; Prince, A.S. The Type III Toxins of Pseudomonas aeruginosa Disrupt Epithelial Barrier Function. J. Bacteriol. 2008, 190, 2814–2821. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  43. Kim, J.Y.; Sajjan, U.S.; Krasan, G.P.; LiPuma, J.J. Disruption of tight junctions during traversal of the respiratory epithelium by Burkholderia cenocepacia. Infect. Immun. 2005, 73, 7107–7112. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  44. Sajjan, U.; Corey, M.; Humar, A.; Tullis, E.; Cutz, E.; Ackerley, C.; Forstner, J. Immunolocalisation of Burkholderia cepacia in the lungs of cystic fibrosis patients. J. Med. Microbiol. 2001, 50, 535–546. [Google Scholar] [CrossRef] [Green Version]
  45. Schwab, U.; Leigh, M.; Ribeiro, C.; Yankaskas, J.; Burns, K.; Gilligan, P.; Sokol, P.; Boucher, R. Patterns of epithelial cell invasion by different species of the Burkholderia cepacia complex in well-differentiated human airway epithelia. Infect. Immun. 2002, 70, 4547–4555. [Google Scholar] [CrossRef] [Green Version]
  46. Duff, C.; Murphy, P.G.; Callaghan, M.; McClean, S. Differences in invasion and translocation of Burkholderia cepacia complex species in polarised lung epithelial cells in vitro. Microb. Pathog. 2006, 41, 183–192. [Google Scholar] [CrossRef]
  47. Blume, C.; David, J.; Bell, R.E.; Laver, J.R.; Read, R.C.; Clark, G.C.; Davies, D.E.; Swindle, E.J. Modulation of Human Airway Barrier Functions during Burkholderia thailandensis and Francisella tularensis Infection Running Title: Airway Barrier Functions during Bacterial Infections. Pathogens 2016, 5, 53. [Google Scholar] [CrossRef]
  48. Bevivino, A.; Pirone, L.; Pilkington, R.; Cifani, N.; Dalmastri, C.; Callaghan, M.; Ascenzioni, F.; McClean, S. Interaction of environmental Burkholderia cenocepacia strains with cystic fibrosis and non-cystic fibrosis bronchial epithelial cells in vitro. Microbiology 2012, 158, 1325–1333. [Google Scholar] [CrossRef] [Green Version]
  49. Ferreira, A.S.; Silva, I.N.; Fernandes, F.; Pilkington, R.; Callaghan, M.; McClean, S.; Moreira, L.M. The tyrosine kinase BceF and the phosphotyrosine phosphatase BceD of Burkholderia contaminans are required for efficient invasion and epithelial disruption of a cystic fibrosis lung epithelial cell line. Infect. Immun. 2015, 83, 812–821. [Google Scholar] [CrossRef] [Green Version]
  50. Kaufhold, I.; Osbahr, S.; Shima, K.; Marwitz, S.; Rohmann, K.; Dromann, D.; Goldmann, T.; Dalhoff, K.; Rupp, J. Nontypeable Haemophilus influenzae (NTHi) directly interfere with the regulation of E-cadherin in lung epithelial cells. Microbes Infect. 2017, 19, 560–566. [Google Scholar] [CrossRef]
  51. Rezaee, F.; Harford, T.J.; Linfield, D.T.; Altawallbeh, G.; Midura, R.J.; Ivanov, A.I.; Piedimonte, G. cAMP-dependent activation of protein kinase A attenuates respiratory syncytial virus-induced human airway epithelial barrier disruption. PLoS ONE 2017, 12, e0181876. [Google Scholar] [CrossRef] [Green Version]
  52. Singh, D.; McCann, K.L.; Imani, F. MAPK and heat shock protein 27 activation are associated with respiratory syncytial virus induction of human bronchial epithelial monolayer disruption. Am. J. Physiol. Lung Cell. Mol. Physiol. 2007, 293, L436–L445. [Google Scholar] [CrossRef] [Green Version]
  53. Gao, N.; Raduka, A.; Rezaee, F. Respiratory syncytial virus disrupts the airway epithelial barrier by decreasing cortactin and destabilizing F-actin. J. Cell Sci. 2022, 135, jcs259871. [Google Scholar] [CrossRef]
  54. Kast, J.I.; McFarlane, A.J.; Głobińska, A.; Sokolowska, M.; Wawrzyniak, P.; Sanak, M.; Schwarze, J.; Akdis, C.A.; Wanke, K. Respiratory syncytial virus infection influences tight junction integrity. Clin. Exp. Immunol. 2017, 190, 351–359. [Google Scholar] [CrossRef] [Green Version]
  55. Masaki, T.; Kojima, T.; Okabayashi, T.; Ogasawara, N.; Ohkuni, T.; Obata, K.; Takasawa, A.; Murata, M.; Tanaka, S.; Hirakawa, S.; et al. A nuclear factor-κB signaling pathway via protein kinase C δ regulates replication of respiratory syncytial virus in polarized normal human nasal epithelial cells. Mol. Biol. Cell 2011, 22, 2144–2156. [Google Scholar] [CrossRef]
  56. Kilani, M.M.; Mohammed, K.A.; Nasreen, N.; Hardwick, J.A.; Kaplan, M.H.; Tepper, R.S.; Antony, V.B. Respiratory syncytial virus causes increased bronchial epithelial permeability. Chest 2004, 126, 186–191. [Google Scholar] [CrossRef] [Green Version]
  57. Faris, A.N.; Ganesan, S.; Chattoraj, A.; Chattoraj, S.S.; Comstock, A.T.; Unger, B.L.; Hershenson, M.B.; Sajjan, U.S. Rhinovirus Delays Cell Repolarization in a Model of Injured/Regenerating Human Airway Epithelium. Am. J. Respir. Cell Mol. Biol. 2016, 55, 487–499. [Google Scholar] [CrossRef] [Green Version]
  58. Sajjan, U.; Wang, Q.; Zhao, Y.; Gruenert, D.C.; Hershenson, M.B. Rhinovirus disrupts the barrier function of polarized airway epithelial cells. Am. J. Respir. Crit. Care Med. 2008, 178, 1271–1281. [Google Scholar] [CrossRef] [Green Version]
  59. Waltl, E.E.; Selb, R.; Eckl-Dorna, J.; Mueller, C.A.; Cabauatan, C.R.; Eiwegger, T.; Resch-Marat, Y.; Niespodziana, K.; Vrtala, S.; Valenta, R.; et al. Betamethasone prevents human rhinovirus- and cigarette smoke- induced loss of respiratory epithelial barrier function. Sci. Rep. 2018, 8, 9688. [Google Scholar] [CrossRef] [Green Version]
  60. Michi, A.N.; Yipp, B.G.; Dufour, A.; Lopes, F.; Proud, D. PGC-1α mediates a metabolic host defense response in human airway epithelium during rhinovirus infections. Nat. Commun. 2021, 12, 3669. [Google Scholar] [CrossRef]
  61. Looi, K.; Troy, N.M.; Garratt, L.W.; Iosifidis, T.; Bosco, A.; Buckley, A.G.; Ling, K.-M.; Martinovich, K.M.; Kicic-Starcevich, E.; Shaw, N.C.; et al. Effect of human rhinovirus infection on airway epithelium tight junction protein disassembly and transepithelial permeability. Exp. Lung Res. 2016, 42, 380–395. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  62. 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]
  63. 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]
  64. 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] [Green Version]
  65. Wronski, S.; Beinke, S.; Obernolte, H.; Belyaev, N.N.; Saunders, K.A.; Lennon, M.G.; Schaudien, D.; Braubach, P.; Jonigk, D.; Warnecke, G.; et al. Rhinovirus-induced Human Lung Tissue Responses Mimic Chronic Obstructive Pulmonary Disease and Asthma Gene Signatures. Am. J. Respir. Cell Mol. Biol. 2021, 65, 544–554. [Google Scholar] [CrossRef]
  66. Comstock, A.T.; Ganesan, S.; Chattoraj, A.; Faris, A.N.; Margolis, B.L.; Hershenson, M.B.; Sajjan, U.S. Rhinovirus-Induced Barrier Dysfunction in Polarized Airway Epithelial Cells Is Mediated by NADPH Oxidase 1. J. Virol. 2011, 85, 6795–6808. [Google Scholar] [CrossRef] [Green Version]
  67. Unger, B.L.; Ganesan, S.; Comstock, A.T.; Faris, A.N.; Hershenson, M.B.; Sajjan, U.S. Nod-Like Receptor X-1 Is Required for Rhinovirus-Induced Barrier Dysfunction in Airway Epithelial Cells. J. Virol. 2014, 88, 3705–3718. [Google Scholar] [CrossRef] [Green Version]
  68. Kim, K.A.; Jung, J.H.; Choi, Y.S.; Kim, S.T. Ginsenoside Re Protects Rhinovirus-Induced Disruption of Tight Junction Through Inhibition Of ROS-Mediated Phosphatases Inactivation In Human Nasal Epithelial Cells. J. Allergy Clin. Immunol. 2018, 141, AB175. [Google Scholar] [CrossRef]
  69. Kim, K.A.; Jung, J.H.; Choi, Y.S.; Kim, S.T. Wogonin inhibits tight junction disruption via suppression of inflammatory response and phosphorylation of AKT/NF-κB and ERK1/2 in rhinovirus-infected human nasal epithelial cells. Inflamm. Res. 2022, 71, 357–368. [Google Scholar] [CrossRef]
  70. Travanty, E.; Zhou, B.; Zhang, H.; Di, Y.P.; Alcorn, J.F.; Wentworth, D.E.; Mason, R.; Wang, J. Differential Susceptibilities of Human Lung Primary Cells to H1N1 Influenza Viruses. J. Virol. 2015, 89, 11935–11944. [Google Scholar] [CrossRef]
  71. Ruan, T.; Sun, J.; Liu, W.; Prinz, R.A.; Peng, D.; Liu, X.; Xu, X. H1N1 Influenza Virus Cross-Activates Gli1 to Disrupt the Intercellular Junctions of Alveolar Epithelial Cells. Cell Rep. 2020, 31, 107801. [Google Scholar] [CrossRef]
  72. Ruan, T.; Sun, Y.; Zhang, J.; Sun, J.; Liu, W.; Prinz, R.A.; Peng, D.; Liu, X.; Xu, X. H5N1 infection impairs the alveolar epithelial barrier through intercellular junction proteins via Itch-mediated proteasomal degradation. Commun. Biol. 2022, 5, 186. [Google Scholar] [CrossRef]
  73. Zhang, L.; Collins, P.L.; Lamb, R.A.; Pickles, R.J. Comparison of differing cytopathic effects in human airway epithelium of parainfluenza virus 5 (W3A), parainfluenza virus type 3, and respiratory syncytial virus. Virology 2011, 421, 67–77. [Google Scholar] [CrossRef] [Green Version]
  74. Yumine, N.; Matsumoto, Y.; Ohta, K.; Fukasawa, M.; Nishio, M. Claudin-1 inhibits human parainfluenza virus type 2 dissemination. Virology 2019, 531, 93–99. [Google Scholar] [CrossRef]
  75. Teoh, K.-T.; Siu, Y.-L.; Chan, W.-L.; Schlüter, M.A.; Liu, C.-J.; Peiris, J.S.M.; Bruzzone, R.; Margolis, B.; Nal, B. The SARS Coronavirus E Protein Interacts with PALS1 and Alters Tight Junction Formation and Epithelial Morphogenesis. Mol. Biol. Cell 2010, 21, 3838–3852. [Google Scholar] [CrossRef] [Green Version]
  76. Zhu, N.; Wang, W.; Liu, Z.; Liang, C.; Wang, W.; Ye, F.; Huang, B.; Zhao, L.; Wang, H.; Zhou, W.; et al. Morphogenesis and cytopathic effect of SARS-CoV-2 infection in human airway epithelial cells. Nat. Commun. 2020, 11, 3910. [Google Scholar] [CrossRef]
  77. 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]
  78. Deinhardt-Emmer, S.; Böttcher, S.; Häring, C.; Giebeler, L.; Henke, A.; Zell, R.; Jungwirth, J.; Jordan, P.M.; Werz, O.; Hornung, F.; et al. SARS-CoV-2 Causes Severe Epithelial Inflammation and Barrier Dysfunction. J. Virol. 2021, 95, e00110–e00121. [Google Scholar] [CrossRef]
  79. Lowy, F.D. Staphylococcus aureus Infections. N. Engl. J. Med. 1998, 339, 520–532. [Google Scholar] [CrossRef]
  80. Gnanamani, A.; Hariharan, P.; Paul-Satyaseela, M. Staphylococcus aureus: Overview of Bacteriology, Clinical Diseases, Epidemiology, Antibiotic Resistance and Therapeutic Approach. In Frontiers in Staphylococcus aureus; Enany, S., Alexander, L., Eds.; IntechOpen: London, UK, 2017. [Google Scholar]
  81. Chmielowiec-Korzeniowska, A.; Tymczyna, L.; Wlazło, Ł.; Nowakowicz-Dębek, B.; Trawińska, B. Staphylococcus aureus carriage state in healthy adult population and phenotypic and genotypic properties of isolated strains. Adv. Dermatol. Allergol. 2020, 37, 184–189. [Google Scholar] [CrossRef]
  82. Sakr, A.; Brégeon, F.; Mège, J.-L.; Rolain, J.-M.; Blin, O. Staphylococcus aureus Nasal Colonization: An Update on Mechanisms, Epidemiology, Risk Factors, and Subsequent Infections. Front. Microbiol. 2018, 9, 2419. [Google Scholar] [CrossRef] [PubMed]
  83. Wertheim, H.F.; Melles, D.C.; Vos, M.C.; van Leeuwen, W.; van Belkum, A.; Verbrugh, H.A.; Nouwen, J.L. The role of nasal carriage in Staphylococcus aureus infections. Lancet Infect. Dis. 2005, 5, 751–762. [Google Scholar] [CrossRef] [PubMed]
  84. Laux, C.; Peschel, A.; Krismer, B. Staphylococcus aureus Colonization of the Human Nose and Interaction with Other Microbiome Members. Microbiol. Spectr. 2019, 7, 34. [Google Scholar] [CrossRef] [PubMed]
  85. Tong, S.Y.; Davis, J.S.; Eichenberger, E.; Holland, T.L.; Fowler, V.G., Jr. Staphylococcus aureus infections: Epidemiology, pathophysiology, clinical manifestations, and management. Clin. Microbiol. Rev. 2015, 28, 603–661. [Google Scholar] [CrossRef] [Green Version]
  86. David, M.Z.; Daum, R.S. Treatment of Staphylococcus aureus Infections. In Staphylococcus aureus: Microbiology, Pathology, Immunology, Therapy and Prophylaxis; Bagnoli, F., Rappuoli, R., Grandi, G., Eds.; Springer International Publishing: Cham, Switzerland, 2017; pp. 325–383. [Google Scholar]
  87. Guo, Y.; Song, G.; Sun, M.; Wang, J.; Wang, Y. Prevalence and Therapies of Antibiotic-Resistance in Staphylococcus aureus. Front. Cell. Infect. Microbiol. 2020, 10, 107. [Google Scholar] [CrossRef] [Green Version]
  88. Lee, A.S.; de Lencastre, H.; Garau, J.; Kluytmans, J.; Malhotra-Kumar, S.; Peschel, A.; Harbarth, S. Methicillin-resistant Staphylococcus aureus. Nat. Rev. Dis. Primers 2018, 4, 18033. [Google Scholar] [CrossRef] [Green Version]
  89. Turner, N.A.; Sharma-Kuinkel, B.K.; Maskarinec, S.A.; Eichenberger, E.M.; Shah, P.P.; Carugati, M.; Holland, T.L.; Fowler, V.G., Jr. Methicillin-resistant Staphylococcus aureus: An overview of basic and clinical research. Nat. Rev. Microbiol. 2019, 17, 203–218. [Google Scholar] [CrossRef]
  90. CDC. 2019 Antibiotic Resistance Threats Report. Available online: https://www.cdc.gov/drugresistance/biggest-threats.html#pse (accessed on 3 November 2022).
  91. Cheung, G.Y.C.; Bae, J.S.; Otto, M. Pathogenicity and virulence of Staphylococcus aureus. Virulence 2021, 12, 547–569. [Google Scholar] [CrossRef]
  92. Garnett, J.P.; Baker, E.H.; Naik, S.; Lindsay, J.A.; Knight, G.M.; Gill, S.; Tregoning, J.S.; Baines, D.L. Metformin reduces airway glucose permeability and hyperglycaemia-induced Staphylococcus aureus load independently of effects on blood glucose. Thorax 2013, 68, 835–845. [Google Scholar] [CrossRef] [Green Version]
  93. Popov, L.M.; Marceau, C.D.; Starkl, P.M.; Lumb, J.H.; Shah, J.; Guerrera, D.; Cooper, R.L.; Merakou, C.; Bouley, D.M.; Meng, W.; et al. The adherens junctions control susceptibility to Staphylococcus aureus α-toxin. Proc. Natl. Acad. Sci. USA 2015, 112, 14337–14342. [Google Scholar] [CrossRef]
  94. Shah, J.; Rouaud, F.; Guerrera, D.; Vasileva, E.; Popov, L.M.; Kelley, W.L.; Rubinstein, E.; Carette, J.E.; Amieva, M.R.; Citi, S. A Dock-and-Lock Mechanism Clusters ADAM10 at Cell-Cell Junctions to Promote α-Toxin Cytotoxicity. Cell Rep. 2018, 25, 2132–2147.e7. [Google Scholar] [CrossRef] [Green Version]
  95. Vuori, K.; Hirai, H.; Aizawa, S.; Ruoslahti, E. Introduction of p130cas signaling complex formation upon integrin-mediated cell adhesion: A role for Src family kinases. Mol. Cell. Biol. 1996, 16, 2606–2613. [Google Scholar] [CrossRef] [Green Version]
  96. Nojima, Y.; Morino, N.; Mimura, T.; Hamasaki, K.; Furuya, H.; Sakai, R.; Sato, T.; Tachibana, K.; Morimoto, C.; Yazaki, Y.; et al. Integrin-mediated cell adhesion promotes tyrosine phosphorylation of p130Cas, a Src homology 3-containing molecule having multiple Src homology 2-binding motifs. J. Biol. Chem. 1995, 270, 15398–15402. [Google Scholar] [CrossRef] [Green Version]
  97. Carlier, M.F.; Ressad, F.; Pantaloni, D. Control of actin dynamics in cell motility. Role of ADF/cofilin. J. Biol. Chem. 1999, 274, 33827–33830. [Google Scholar] [CrossRef] [Green Version]
  98. Agerer, F.; Michel, A.; Ohlsen, K.; Hauck, C.R. Integrin-mediated Invasion of Staphylococcus aureus into Human Cells Requires Src Family Protein-tyrosine Kinases. J. Biol. Chem. 2003, 278, 42524–42531. [Google Scholar] [CrossRef] [Green Version]
  99. Weiser, J.N.; Ferreira, D.M.; Paton, J.C. Streptococcus pneumoniae: Transmission, colonization and invasion. Nat. Rev. Microbiol. 2018, 16, 355–367. [Google Scholar] [CrossRef]
  100. Henriques-Normark, B.; Tuomanen, E.I. The pneumococcus: Epidemiology, microbiology, and pathogenesis. Cold Spring Harb. Perspect. Med. 2013, 3, a010215. [Google Scholar] [CrossRef]
  101. Subramanian, K.; Henriques-Normark, B.; Normark, S. Emerging concepts in the pathogenesis of the Streptococcus pneumoniae: From nasopharyngeal colonizer to intracellular pathogen. Cell. Microbiol. 2019, 21, e13077. [Google Scholar] [CrossRef] [Green Version]
  102. Brooks, L.R.K.; Mias, G.I. Streptococcus pneumoniae’s Virulence and Host Immunity: Aging, Diagnostics, and Prevention. Front. Immunol. 2018, 9, 1366. [Google Scholar] [CrossRef] [Green Version]
  103. Evans, W.; Hansman, D. Tetracycline-Resistant Pneumococcus. Lancet 1963, 281, 451. [Google Scholar] [CrossRef]
  104. Andrejko, K.; Ratnasiri, B.; Hausdorff, W.P.; Laxminarayan, R.; Lewnard, J.A. Antimicrobial resistance in paediatric Streptococcus pneumoniae isolates amid global implementation of pneumococcal conjugate vaccines: A systematic review and meta-regression analysis. Lancet Microbe 2021, 2, e450–e460. [Google Scholar] [CrossRef] [PubMed]
  105. Cherazard, R.; Epstein, M.; Doan, T.-L.; Salim, T.; Bharti, S.; Smith, M.A. Antimicrobial Resistant Streptococcus pneumoniae: Prevalence, Mechanisms, and Clinical Implications. Am. J. Ther. 2017, 24, e361–e369. [Google Scholar] [CrossRef]
  106. Sader, H.S.; Mendes, R.E.; Le, J.; Denys, G.; Flamm, R.K.; Jones, R.N. Antimicrobial Susceptibility of Streptococcus pneumoniae from North America, Europe, Latin America, and the Asia-Pacific Region: Results From 20 Years of the SENTRY Antimicrobial Surveillance Program (1997–2016). Open Forum Infect. Dis. 2019, 6, S14–S23. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  107. Klugman, K.P.; Rodgers, G.L. Time for a third-generation pneumococcal conjugate vaccine. Lancet Infect. Dis. 2021, 21, 14–16. [Google Scholar] [CrossRef] [PubMed]
  108. Suzuki, M.; Dhoubhadel, B.G.; Ishifuji, T.; Yasunami, M.; Yaegashi, M.; Asoh, N.; Ishida, M.; Hamaguchi, S.; Aoshima, M.; Ariyoshi, K.; et al. Serotype-specific effectiveness of 23-valent pneumococcal polysaccharide vaccine against pneumococcal pneumonia in adults aged 65 years or older: A multicentre, prospective, test-negative design study. Lancet Infect. Dis. 2017, 17, 313–321. [Google Scholar] [CrossRef]
  109. Ganaie, F.; Saad, J.S.; McGee, L.; van Tonder, A.J.; Bentley, S.D.; Lo, S.W.; Gladstone, R.A.; Turner, P.; Keenan, J.D.; Breiman, R.F.; et al. A New Pneumococcal Capsule Type, 10D, is the 100th Serotype and Has a Large cps Fragment from an Oral Streptococcus. mBio 2020, 11, 32430472. [Google Scholar] [CrossRef]
  110. Feikin, D.R.; Kagucia, E.W.; Loo, J.D.; Link-Gelles, R.; Puhan, M.A.; Cherian, T.; Levine, O.S.; Whitney, C.G.; O’Brien, K.L.; Moore, M.R. Serotype-specific changes in invasive pneumococcal disease after pneumococcal conjugate vaccine introduction: A pooled analysis of multiple surveillance sites. PLoS Med. 2013, 10, e1001517. [Google Scholar] [CrossRef] [Green Version]
  111. Ladhani, S.N.; Collins, S.; Djennad, A.; Sheppard, C.L.; Borrow, R.; Fry, N.K.; Andrews, N.J.; Miller, E.; Ramsay, M.E. Rapid increase in non-vaccine serotypes causing invasive pneumococcal disease in England and Wales, 2000–2017: A prospective national observational cohort study. Lancet Infect. Dis. 2018, 18, 441–451. [Google Scholar] [CrossRef] [Green Version]
  112. Galanis, I.; Lindstrand, A.; Darenberg, J.; Browall, S.; Nannapaneni, P.; Sjöström, K.; Morfeldt, E.; Naucler, P.; Blennow, M.; Örtqvist, Å.; et al. Effects of PCV7 and PCV13 on invasive pneumococcal disease and carriage in Stockholm, Sweden. Eur. Respir. J. 2016, 47, 1208–1218. [Google Scholar] [CrossRef] [Green Version]
  113. LeMessurier, K.S.; Tiwary, M.; Morin, N.P.; Samarasinghe, A.E. Respiratory Barrier as a Safeguard and Regulator of Defense Against Influenza A Virus and Streptococcus pneumoniae. Front. Immunol. 2020, 11, 3. [Google Scholar] [CrossRef]
  114. Fliegauf, M.; Sonnen, A.F.P.; Kremer, B.; Henneke, P. Mucociliary Clearance Defects in a Murine In Vitro Model of Pneumococcal Airway Infection. PLoS ONE 2013, 8, e59925. [Google Scholar] [CrossRef] [Green Version]
  115. Reynolds, D.; Kollef, M. The Epidemiology and Pathogenesis and Treatment of Pseudomonas aeruginosa Infections: An Update. Drugs 2021, 81, 2117–2131. [Google Scholar] [CrossRef]
  116. Morin, C.D.; Deziel, E.; Gauthier, J.; Levesque, R.C.; Lau, G.W. An Organ System-Based Synopsis of Pseudomonas aeruginosa Virulence. Virulence 2021, 12, 1469–1507. [Google Scholar] [CrossRef]
  117. Norbury, W.; Herndon, D.N.; Tanksley, J.; Jeschke, M.G.; Finnerty, C.C. Infection in Burns. Surg. Infect. (Larchmt) 2016, 17, 250–255. [Google Scholar] [CrossRef] [Green Version]
  118. Philips, B.J.; Meguer, J.X.; Redman, J.; Baker, E.H. Factors determining the appearance of glucose in upper and lower respiratory tract secretions. Intensive Care Med. 2003, 29, 2204–2210. [Google Scholar] [CrossRef]
  119. Baker, E.H.; Janaway, C.H.; Philips, B.J.; Brennan, A.L.; Baines, D.L.; Wood, D.M.; Jones, P.W. Hyperglycaemia is associated with poor outcomes in patients admitted to hospital with acute exacerbations of chronic obstructive pulmonary disease. Thorax 2006, 61, 284–289. [Google Scholar] [CrossRef] [Green Version]
  120. Garnett, J.P.; Gray, M.A.; Tarran, R.; Brodlie, M.; Ward, C.; Baker, E.H.; Baines, D.L. Elevated paracellular glucose flux across cystic fibrosis airway epithelial monolayers is an important factor for Pseudomonas aeruginosa growth. PLoS ONE 2013, 8, e76283. [Google Scholar] [CrossRef] [Green Version]
  121. Horna, G.; Ruiz, J. Type 3 secretion system of Pseudomonas aeruginosa. Microbiol. Res. 2021, 246, 126719. [Google Scholar] [CrossRef]
  122. Morrow, K.A.; Frank, D.W.; Balczon, R.; Stevens, T. The Pseudomonas aeruginosa Exoenzyme Y: A Promiscuous Nucleotidyl Cyclase Edema Factor and Virulence Determinant. Handb. Exp. Pharmacol. 2017, 238, 67–85. [Google Scholar] [CrossRef]
  123. Mancl, J.M.; Suarez, C.; Liang, W.G.; Kovar, D.R.; Tang, W.J. Pseudomonas aeruginosa exoenzyme Y directly bundles actin filaments. J. Biol. Chem. 2020, 295, 3506–3517. [Google Scholar] [CrossRef]
  124. Wagener, B.M.; Anjum, N.; Christiaans, S.C.; Banks, M.E.; Parker, J.C.; Threet, A.T.; Walker, R.R.; Isbell, K.D.; Moser, S.A.; Stevens, T.; et al. Exoenzyme Y Contributes to End-Organ Dysfunction Caused by Pseudomonas aeruginosa Pneumonia in Critically Ill Patients: An Exploratory Study. Toxins 2020, 12, 369. [Google Scholar] [CrossRef] [PubMed]
  125. Wood, S.; Goldufsky, J.; Shafikhani, S.H. Pseudomonas aeruginosa ExoT Induces Atypical Anoikis Apoptosis in Target Host Cells by Transforming Crk Adaptor Protein into a Cytotoxin. PLoS Pathog. 2015, 11, e1004934. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  126. Wood, S.J.; Goldufsky, J.W.; Bello, D.; Masood, S.; Shafikhani, S.H. Pseudomonas aeruginosa ExoT Induces Mitochondrial Apoptosis in Target Host Cells in a Manner That Depends on Its GTPase-activating Protein (GAP) Domain Activity. J. Biol. Chem. 2015, 290, 29063–29073. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  127. Syed, I.; Wooten, R.M. Interactions Between Pathogenic Burkholderia and the Complement System: A Review of Potential Immune Evasion Mechanisms. Front. Cell. Infect. Microbiol. 2021, 11, 701362. [Google Scholar] [CrossRef] [PubMed]
  128. Coenye, T.; Vandamme, P.; Govan, J.R.; LiPuma, J.J. Taxonomy and identification of the Burkholderia cepacia complex. J. Clin. Microbiol. 2001, 39, 3427–3436. [Google Scholar] [CrossRef] [Green Version]
  129. Huse, H.K.; Lee, M.J.; Wootton, M.; Sharp, S.E.; Traczewski, M.; LiPuma, J.J.; Jorth, P. Evaluation of Antimicrobial Susceptibility Testing Methods for Burkholderia cenocepacia and Burkholderia multivorans Isolates from Cystic Fibrosis Patients. J. Clin. Microbiol. 2021, 59, e0144721. [Google Scholar] [CrossRef]
  130. Bressler, A.M.; Kaye, K.S.; LiPuma, J.J.; Alexander, B.D.; Moore, C.M.; Reller, L.B.; Woods, C.W. Risk factors for Burkholderia cepacia complex bacteremia among intensive care unit patients without cystic fibrosis: A case-control study. Infect. Control Hosp. Epidemiol. 2007, 28, 951–958. [Google Scholar] [CrossRef]
  131. Rhodes, K.A.; Schweizer, H.P. Antibiotic resistance in Burkholderia species. Drug Resist. Updates 2016, 28, 82–90. [Google Scholar] [CrossRef] [Green Version]
  132. Aziz, A.; Sarovich, D.S.; Nosworthy, E.; Beissbarth, J.; Chang, A.B.; Smith-Vaughan, H.; Price, E.P.; Harris, T.M. Molecular Signatures of Non-typeable Haemophilus influenzae Lung Adaptation in Pediatric Chronic Lung Disease. Front. Microbiol. 2019, 10, 1622. [Google Scholar] [CrossRef] [Green Version]
  133. Su, Y.C.; Jalalvand, F.; Thegerstrom, J.; Riesbeck, K. The Interplay Between Immune Response and Bacterial Infection in COPD: Focus Upon Non-typeable Haemophilus influenzae. Front. Immunol. 2018, 9, 2530. [Google Scholar] [CrossRef]
  134. Kadry, N.A.; Porsch, E.A.; Shen, H.; St Geme, J.W., 3rd. Immunization with HMW1 and HMW2 adhesins protects against colonization by heterologous strains of nontypeable Haemophilus influenzae. Proc. Natl. Acad. Sci. USA 2021, 118, e2019923118. [Google Scholar] [CrossRef]
  135. Ahearn, C.P.; Gallo, M.C.; Murphy, T.F. Insights on persistent airway infection by non-typeable Haemophilus influenzae in chronic obstructive pulmonary disease. Pathog. Dis. 2017, 75, ftx042. [Google Scholar] [CrossRef] [Green Version]
  136. Sajjan, U.S.; Jia, Y.; Newcomb, D.C.; Bentley, J.K.; Lukacs, N.W.; LiPuma, J.J.; Hershenson, M.B. H. influenzae potentiates airway epithelial cell responses to rhinovirus by increasing ICAM-1 and TLR3 expression. FASEB J. 2006, 20, 2121–2123. [Google Scholar] [CrossRef] [Green Version]
  137. Watanabe, T.; Jono, H.; Han, J.; Lim, D.J.; Li, J.D. Synergistic activation of NF-kappaB by nontypeable Haemophilus influenzae and tumor necrosis factor alpha. Proc. Natl. Acad. Sci. USA 2004, 101, 3563–3568. [Google Scholar] [CrossRef] [Green Version]
  138. Ward, C.; Schlingmann, B.; Stecenko, A.A.; Guidot, D.M.; Koval, M. NF-kappaB inhibitors impair lung epithelial tight junctions in the absence of inflammation. Tissue Barriers 2015, 3, e982424. [Google Scholar] [CrossRef]
  139. Rokkam, D.; Lafemina, M.J.; Lee, J.W.; Matthay, M.A.; Frank, J.A. Claudin-4 levels are associated with intact alveolar fluid clearance in human lungs. Am. J. Pathol. 2011, 179, 1081–1087. [Google Scholar] [CrossRef]
  140. Wang, F.; Daugherty, B.; Keise, L.L.; Wei, Z.; Foley, J.P.; Savani, R.C.; Koval, M. Heterogeneity of claudin expression by alveolar epithelial cells. Am. J. Respir. Cell Mol. Biol. 2003, 29, 62–70. [Google Scholar] [CrossRef] [Green Version]
  141. Koval, M. Claudin heterogeneity and control of lung tight junctions. Annu. Rev. Physiol. 2013, 75, 551–567. [Google Scholar] [CrossRef]
  142. LaFemina, M.J.; Sutherland, K.M.; Bentley, T.; Gonzales, L.W.; Allen, L.; Chapin, C.J.; Rokkam, D.; Sweerus, K.A.; Dobbs, L.G.; Ballard, P.L.; et al. Claudin-18 deficiency results in alveolar barrier dysfunction and impaired alveologenesis in mice. Am. J. Respir. Cell Mol. Biol. 2014, 51, 550–558. [Google Scholar] [CrossRef] [Green Version]
  143. Milara, J.; Peiro, T.; Serrano, A.; Cortijo, J. Epithelial to mesenchymal transition is increased in patients with COPD and induced by cigarette smoke. Thorax 2013, 68, 410–420. [Google Scholar] [CrossRef]
  144. Glockner, M.; Marwitz, S.; Rohmann, K.; Watz, H.; Nitschkowski, D.; Rupp, J.; Dalhoff, K.; Goldmann, T.; Dromann, D. Haemophilus influenzae causes cellular trans-differentiation in human bronchial epithelia. Innate Immun. 2021, 27, 251–259. [Google Scholar] [CrossRef] [PubMed]
  145. Schleimer, R.P.; Berdnikovs, S. Etiology of epithelial barrier dysfunction in patients with type 2 inflammatory diseases. J. Allergy Clin. Immunol. 2017, 139, 1752–1761. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  146. Georas, S.N.; Rezaee, F. Epithelial barrier function: At the front line of asthma immunology and allergic airway inflammation. J. Allergy Clin. Immunol. 2014, 134, 509–520. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  147. Nair, P.; Surette, M.G.; Virchow, J.C. Neutrophilic asthma: Misconception or misnomer? Lancet Respir. Med. 2021, 9, 441–443. [Google Scholar] [CrossRef] [PubMed]
  148. Brown, M.A.; Jabeen, M.; Bharj, G.; Hinks, T.S.C. Non-typeable Haemophilus influenzae airways infection: The next treatable trait in asthma? Eur. Respir. Rev. 2022, 31, 220008. [Google Scholar] [CrossRef]
  149. Gibson, P.G.; Yang, I.A.; Upham, J.W.; Reynolds, P.N.; Hodge, S.; James, A.L.; Jenkins, C.; Peters, M.J.; Marks, G.B.; Baraket, M.; et al. Effect of azithromycin on asthma exacerbations and quality of life in adults with persistent uncontrolled asthma (AMAZES): A randomised, double-blind, placebo-controlled trial. Lancet 2017, 390, 659–668. [Google Scholar] [CrossRef] [Green Version]
  150. Brusselle, G.G.; Vanderstichele, C.; Jordens, P.; Deman, R.; Slabbynck, H.; Ringoet, V.; Verleden, G.; Demedts, I.K.; Verhamme, K.; Delporte, A.; et al. Azithromycin for prevention of exacerbations in severe asthma (AZISAST): A multicentre randomised double-blind placebo-controlled trial. Thorax 2013, 68, 322–329. [Google Scholar] [CrossRef] [Green Version]
  151. Serisier, D.J. Risks of population antimicrobial resistance associated with chronic macrolide use for inflammatory airway diseases. Lancet Respir. Med. 2013, 1, 262–274. [Google Scholar] [CrossRef]
  152. Rima, B.; Collins, P.; Easton, A.; Fouchier, R.; Kurath, G.; Lamb, R.A.; Lee, B.; Maisner, A.; Rota, P.; Wang, L.; et al. ICTV Virus Taxonomy Profile: Pneumoviridae. J. Gen. Virol. 2017, 98, 2912–2913. [Google Scholar] [CrossRef]
  153. Grayson, S.A.; Griffiths, P.S.; Perez, M.K.; Piedimonte, G. Detection of airborne respiratory syncytial virus in a pediatric acute care clinic. Pediatr. Pulmonol. 2017, 52, 684–688. [Google Scholar] [CrossRef]
  154. Hall, C.B. Respiratory syncytial virus: Its transmission in the hospital environment. Yale J. Biol. Med. 1982, 55, 219–223. [Google Scholar]
  155. Griffiths, C.; Drews, S.J.; Marchant, D.J. Respiratory Syncytial Virus: Infection, Detection, and New Options for Prevention and Treatment. Clin. Microbiol. Rev. 2017, 30, 277–319. [Google Scholar] [CrossRef] [Green Version]
  156. Shi, T.; McAllister, D.A.; O’Brien, K.L.; Simoes, E.A.F.; Madhi, S.A.; Gessner, B.D.; Polack, F.P.; Balsells, E.; Acacio, S.; Aguayo, C.; et al. Global, regional, and national disease burden estimates of acute lower respiratory infections due to respiratory syncytial virus in young children in 2015: A systematic review and modelling study. Lancet 2017, 390, 946–958. [Google Scholar] [CrossRef] [Green Version]
  157. Rezaee, F.; Linfield, D.T.; Harford, T.J.; Piedimonte, G. Ongoing developments in RSV prophylaxis: A clinician’s analysis. Curr. Opin. Virol. 2017, 24, 70–78. [Google Scholar] [CrossRef]
  158. Li, Y.; Johnson, E.K.; Shi, T.; Campbell, H.; Chaves, S.S.; Commaille-Chapus, C.; Dighero, I.; James, S.L.; Mahé, C.; Ooi, Y.; et al. National burden estimates of hospitalisations for acute lower respiratory infections due to respiratory syncytial virus in young children in 2019 among 58 countries: A modelling study. Lancet Respir. Med. 2021, 9, 175–185. [Google Scholar] [CrossRef]
  159. Srikantiah, P.; Vora, P.; Klugman, K.P. Assessing the Full Burden of Respiratory Syncytial Virus in Young Infants in Low- and Middle-Income Countries: The Importance of Community Mortality Studies. Clin. Infect. Dis. 2021, 73, S177–S179. [Google Scholar] [CrossRef]
  160. Li, Y.; Wang, X.; Blau, D.M.; Caballero, M.T.; Feikin, D.R.; Gill, C.J.; Madhi, S.A.; Omer, S.B.; Simões, E.A.F.; Campbell, H.; et al. Global, regional, and national disease burden estimates of acute lower respiratory infections due to respiratory syncytial virus in children younger than 5 years in 2019: A systematic analysis. Lancet 2022, 399, 2047–2064. [Google Scholar] [CrossRef]
  161. Carbonell-Estrany, X.; Pérez-Yarza, E.G.; García, L.S.; Guzmán Cabañas, J.M.; Bòria, E.V.; Atienza, B.B.; Group, I.S. Long-Term Burden and Respiratory Effects of Respiratory Syncytial Virus Hospitalization in Preterm Infants—The SPRING Study. PLoS ONE 2015, 10, e0125422. [Google Scholar] [CrossRef] [Green Version]
  162. Wu, P.; Hartert, T.V. Evidence for a causal relationship between respiratory syncytial virus infection and asthma. Expert Rev. Anti Infec. Ther. 2011, 9, 731–745. [Google Scholar] [CrossRef] [Green Version]
  163. Sigurs, N.; Aljassim, F.; Kjellman, B.; Robinson, P.D.; Sigurbergsson, F.; Bjarnason, R.; Gustafsson, P.M. Asthma and allergy patterns over 18 years after severe RSV bronchiolitis in the first year of life. Thorax 2010, 65, 1045–1052. [Google Scholar] [CrossRef] [Green Version]
  164. Jartti, T.; Gern, J.E. Role of viral infections in the development and exacerbation of asthma in children. J. Allergy Clin. Immunol. 2017, 140, 895–906. [Google Scholar] [CrossRef] [PubMed]
  165. Taleb, S.A.; Al Thani, A.A.; Al Ansari, K.; Yassine, H.M. Human respiratory syncytial virus: Pathogenesis, immune responses, and current vaccine approaches. Eur. J. Clin. Microbiol. Infect. Dis. 2018, 37, 1817–1827. [Google Scholar] [CrossRef] [PubMed]
  166. Belderbos, M.E.; Houben, M.L.; Wilbrink, B.; Lentjes, E.; Bloemen, E.M.; Kimpen, J.L.; Rovers, M.; Bont, L. Cord blood vitamin D deficiency is associated with respiratory syncytial virus bronchiolitis. Pediatrics 2011, 127, e1513–e1520. [Google Scholar] [CrossRef] [PubMed]
  167. Smallcombe, C.C.; Harford, T.J.; Linfield, D.T.; Lechuga, S.; Bokun, V.; Piedimonte, G.; Rezaee, F. Titanium dioxide nanoparticles exaggerate respiratory syncytial virus-induced airway epithelial barrier dysfunction. Am. J. Physiol. Lung Cell. Mol. Physiol. 2020, 319, L481–L496. [Google Scholar] [CrossRef]
  168. Nicolai, A.; Frassanito, A.; Nenna, R.; Cangiano, G.; Petrarca, L.; Papoff, P.; Pierangeli, A.; Scagnolari, C.; Moretti, C.; Midulla, F. Risk Factors for Virus-induced Acute Respiratory Tract Infections in Children Younger Than 3 Years and Recurrent Wheezing at 36 Months Follow-Up After Discharge. Pediatr. Infect. Dis. J. 2017, 36, 179–183. [Google Scholar] [CrossRef]
  169. Karr, C.J.; Rudra, C.B.; Miller, K.A.; Gould, T.R.; Larson, T.; Sathyanarayana, S.; Koenig, J.Q. Infant exposure to fine particulate matter and traffic and risk of hospitalization for RSV bronchiolitis in a region with lower ambient air pollution. Environ. Res. 2009, 109, 321–327. [Google Scholar] [CrossRef] [Green Version]
  170. Bradley, J.P.; Bacharier, L.B.; Bonfiglio, J.; Schechtman, K.B.; Strunk, R.; Storch, G.; Castro, M. Severity of respiratory syncytial virus bronchiolitis is affected by cigarette smoke exposure and atopy. Pediatrics 2005, 115, e7–e14. [Google Scholar] [CrossRef] [Green Version]
  171. Hashiguchi, S.; Yoshida, H.; Akashi, T.; Komemoto, K.; Ueda, T.; Ikarashi, Y.; Miyauchi, A.; Konno, K.; Yamanaka, S.; Hirose, A.; et al. Titanium dioxide nanoparticles exacerbate pneumonia in respiratory syncytial virus (RSV)-infected mice. Environ. Toxicol. Pharmacol. 2015, 39, 879–886. [Google Scholar] [CrossRef]
  172. Xing, Y.; Proesmans, M. New therapies for acute RSV infections: Where are we? Eur. J. Pediatr. 2019, 178, 131–138. [Google Scholar] [CrossRef]
  173. Mammas, I.N.; Drysdale, S.B.; Rath, B.; Theodoridou, M.; Papaioannou, G.; Papatheodoropoulou, A.; Koutsounaki, E.; Koutsaftiki, C.; Kozanidou, E.; Achtsidis, V.; et al. Update on current views and advances on RSV infection. Int. J. Mol. Med. 2020, 46, 509–520. [Google Scholar] [CrossRef]
  174. Behzadi, M.A.; Leyva-Grado, V.H. Overview of Current Therapeutics and Novel Candidates Against Influenza, Respiratory Syncytial Virus, and Middle East Respiratory Syndrome Coronavirus Infections. Front. Microbiol. 2019, 10, 1327. [Google Scholar] [CrossRef]
  175. Krilov, L.R. Safety issues related to the administration of ribavirin. Pediatr. Infect. Dis. J. 2002, 21, 479–481. [Google Scholar] [CrossRef]
  176. Nicholson, E.G.; Munoz, F.M. A Review of Therapeutics in Clinical Development for Respiratory Syncytial Virus and Influenza in Children. Clin. Ther. 2018, 40, 1268–1281. [Google Scholar] [CrossRef] [Green Version]
  177. Frogel, M.; Nerwen, C.; Cohen, A.; VanVeldhuisen, P.; Harrington, M.; Boron, M. Prevention of hospitalization due to respiratory syncytial virus: Results from the Palivizumab Outcomes Registry. J. Perinatol. 2008, 28, 511–517. [Google Scholar] [CrossRef] [Green Version]
  178. Shahabi, A.; Peneva, D.; Incerti, D.; McLaurin, K.; Stevens, W. Assessing Variation in the Cost of Palivizumab for Respiratory Syncytial Virus Prevention in Preterm Infants. PharmacoEcon.-Open 2018, 2, 53–61. [Google Scholar] [CrossRef] [Green Version]
  179. Esposito, S.; Abu Raya, B.; Baraldi, E.; Flanagan, K.; Martinon Torres, F.; Tsolia, M.; Zielen, S. RSV Prevention in All Infants: Which Is the Most Preferable Strategy? Front. Immunol. 2022, 13, 880368. [Google Scholar] [CrossRef]
  180. Zhang, L.; Peeples, M.E.; Boucher, R.C.; Collins, P.L.; Pickles, R.J. Respiratory syncytial virus infection of human airway epithelial cells is polarized, specific to ciliated cells, and without obvious cytopathology. J. Virol. 2002, 76, 5654–5666. [Google Scholar] [CrossRef] [Green Version]
  181. Johnson, J.E.; Gonzales, R.A.; Olson, S.J.; Wright, P.F.; Graham, B.S. The histopathology of fatal untreated human respiratory syncytial virus infection. Mod. Pathol. 2007, 20, 108–119. [Google Scholar] [CrossRef] [Green Version]
  182. Battles, M.B.; McLellan, J.S. Respiratory syncytial virus entry and how to block it. Nat. Rev. Microbiol. 2019, 17, 233–245. [Google Scholar] [CrossRef]
  183. Spacova, I.; De Boeck, I.; Bron, P.A.; Delputte, P.; Lebeer, S. Topical Microbial Therapeutics against Respiratory Viral Infections. Trends Mol. Med. 2021, 27, 538–553. [Google Scholar] [CrossRef]
  184. Bruewer, M.; Utech, M.; Ivanov, A.I.; Hopkins, A.M.; Parkos, C.A.; Nusrat, A. Interferon-gamma induces internalization of epithelial tight junction proteins via a macropinocytosis-like process. FASEB J. 2005, 19, 923–933. [Google Scholar] [CrossRef] [PubMed]
  185. Saatian, B.; Rezaee, F.; Desando, S.; Emo, J.; Chapman, T.; Knowlden, S.; Georas, S.N. Interleukin-4 and interleukin-13 cause barrier dysfunction in human airway epithelial cells. Tissue Barriers 2013, 1, e24333. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  186. Utech, M.; Ivanov, A.I.; Samarin, S.N.; Bruewer, M.; Turner, J.R.; Mrsny, R.J.; Parkos, C.A.; Nusrat, A. Mechanism of IFN-gamma-induced endocytosis of tight junction proteins: Myosin II-dependent vacuolarization of the apical plasma membrane. Mol. Biol. Cell 2005, 16, 5040–5052. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  187. Hartsock, A.; Nelson, W.J. Adherens and tight junctions: Structure, function and connections to the actin cytoskeleton. Biochim. Biophys. Acta 2008, 1778, 660–669. [Google Scholar] [CrossRef] [Green Version]
  188. Rozengurt, E. Protein Kinase D Signaling: Multiple Biological Functions in Health and Disease. Physiology 2011, 26, 23–33. [Google Scholar] [CrossRef] [Green Version]
  189. Zhang, X.; Connelly, J.; Chao, Y.; Wang, Q.J. Multifaceted Functions of Protein Kinase D in Pathological Processes and Human Diseases. Biomolecules 2021, 11, 483. [Google Scholar] [CrossRef]
  190. Olayioye, M.A.; Barisic, S.; Hausser, A. Multi-level control of actin dynamics by protein kinase D. Cell. Signal. 2013, 25, 1739–1747. [Google Scholar] [CrossRef]
  191. Eiseler, T.; Hausser, A.; De Kimpe, L.; Van Lint, J.; Pfizenmaier, K. Protein Kinase D Controls Actin Polymerization and Cell Motility through Phosphorylation of Cortactin. J. Biol. Chem. 2010, 285, 18672–18683. [Google Scholar] [CrossRef] [Green Version]
  192. Schnoor, M.; Stradal, T.E.; Rottner, K. Cortactin: Cell Functions of a Multifaceted Actin-Binding Protein. Trends Cell Biol. 2018, 28, 79–98. [Google Scholar] [CrossRef]
  193. Cosen-Binker, L.I.; Kapus, A. Cortactin: The Gray Eminence of the Cytoskeleton. Physiology 2006, 21, 352–361. [Google Scholar] [CrossRef]
  194. Weed, S.A.; Parsons, J.T. Cortactin: Coupling membrane dynamics to cortical actin assembly. Oncogene 2001, 20, 6418–6434. [Google Scholar] [CrossRef]
  195. Bougnères, L.; Girardin, S.p.E.; Weed, S.A.; Karginov, A.V.; Olivo-Marin, J.-C.; Parsons, J.T.; Sansonetti, P.J.; Van Nhieu, G.T. Cortactin and Crk cooperate to trigger actin polymerization during Shigella invasion of epithelial cells. J. Cell Biol. 2004, 166, 225–235. [Google Scholar] [CrossRef] [Green Version]
  196. Vázquez, Y.; González, L.; Noguera, L.; González, P.A.; Riedel, C.A.; Bertrand, P.; Bueno, S.M. Cytokines in the Respiratory Airway as Biomarkers of Severity and Prognosis for Respiratory Syncytial Virus Infection: An Update. Front. Immunol. 2019, 10, 1154. [Google Scholar] [CrossRef]
  197. McNamara, P.S.; Ritson, P.; Selby, A.; Hart, C.A.; Smyth, R.L. Bronchoalveolar lavage cellularity in infants with severe respiratory syncytial virus bronchiolitis. Arch. Dis. Child. 2003, 88, 922–926. [Google Scholar] [CrossRef] [Green Version]
  198. Deng, Y.; Herbert, J.A.; Smith, C.M.; Smyth, R.L. An in vitro transepithelial migration assay to evaluate the role of neutrophils in Respiratory Syncytial Virus (RSV) induced epithelial damage. Sci. Rep. 2018, 8, 6777. [Google Scholar] [CrossRef] [Green Version]
  199. Price, W.H. The Isolation of a New Virus Associated with Respiratory Clinical Disease in Humans. Proc. Natl. Acad. Sci. USA 1956, 42, 892–896. [Google Scholar] [CrossRef] [Green Version]
  200. Nelson, P.P.; Papadopoulos, N.G.; Skevaki, C. Respiratory Viral Pathogens. In Encyclopedia of Respiratory Medicine, 2nd ed.; Janes, S.M., Ed.; Academic Press: Oxford, UK, 2022; pp. 129–137. [Google Scholar]
  201. Stobart, C.C.; Nosek, J.M.; Moore, M.L. Rhinovirus Biology, Antigenic Diversity, and Advancements in the Design of a Human Rhinovirus Vaccine. Front. Microbiol. 2017, 8, 2412. [Google Scholar] [CrossRef]
  202. Kutter, J.S.; Spronken, M.I.; Fraaij, P.L.; Fouchier, R.A.M.; Herfst, S. Transmission routes of respiratory viruses among humans. Curr. Opin. Virol. 2018, 28, 142–151. [Google Scholar] [CrossRef]
  203. Monto, A.S. The seasonality of rhinovirus infections and its implications for clinical recognition. Clin. Ther. 2002, 24, 1987–1997. [Google Scholar] [CrossRef]
  204. Morikawa, S.; Kohdera, U.; Hosaka, T.; Ishii, K.; Akagawa, S.; Hiroi, S.; Kase, T. Seasonal variations of respiratory viruses and etiology of human rhinovirus infection in children. J. Clin. Virol. 2015, 73, 14–19. [Google Scholar] [CrossRef]
  205. Moriyama, M.; Hugentobler, W.J.; Iwasaki, A. Seasonality of Respiratory Viral Infections. Annu. Rev. Virol. 2020, 7, 83–101. [Google Scholar] [CrossRef]
  206. Kennedy, J.L.; Turner, R.B.; Braciale, T.; Heymann, P.W.; Borish, L. Pathogenesis of rhinovirus infection. Curr. Opin. Virol. 2012, 2, 287–293. [Google Scholar] [CrossRef] [Green Version]
  207. Bizot, E.; Bousquet, A.; Charpié, M.; Coquelin, F.; Lefevre, S.; Le Lorier, J.; Patin, M.; Sée, P.; Sarfati, E.; Walle, S.; et al. Rhinovirus: A Narrative Review on Its Genetic Characteristics, Pediatric Clinical Presentations, and Pathogenesis. Front. Pediatr. 2021, 9, 643219. [Google Scholar] [CrossRef]
  208. Jain, S.; Williams, D.J.; Arnold, S.R.; Ampofo, K.; Bramley, A.M.; Reed, C.; Stockmann, C.; Anderson, E.J.; Grijalva, C.G.; Self, W.H.; et al. Community-Acquired Pneumonia Requiring Hospitalization among U.S. Children. N. Engl. J. Med. 2015, 372, 835–845. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  209. Meissner, H.C. Viral Bronchiolitis in Children. N. Engl. J. Med. 2016, 374, 62–72. [Google Scholar] [CrossRef] [PubMed]
  210. Fedele, G.; Schiavoni, I.; Nenna, R.; Pierangeli, A.; Frassanito, A.; Leone, P.; Petrarca, L.; Scagnolari, C.; Midulla, F. Analysis of the immune response in infants hospitalized with viral bronchiolitis shows different Th1/Th2 profiles associated with respiratory syncytial virus and human rhinovirus. Pediatr. Allergy Immunol. 2018, 29, 555–557. [Google Scholar] [CrossRef] [PubMed]
  211. Gern, J.E. The ABCs of rhinoviruses, wheezing, and asthma. J. Virol. 2010, 84, 7418–7426. [Google Scholar] [CrossRef] [Green Version]
  212. Merckx, J.; Ducharme, F.M.; Martineau, C.; Zemek, R.; Gravel, J.; Chalut, D.; Poonai, N.; Quach, C. Respiratory Viruses and Treatment Failure in Children with Asthma Exacerbation. Pediatrics 2018, 142, e20174105. [Google Scholar] [CrossRef] [Green Version]
  213. Seemungal, T.; Harper-Owen, R.; Bhowmik, A.; Moric, I.; Sanderson, G.; Message, S.; Maccallum, P.; Meade, T.W.; Jeffries, D.J.; Johnston, S.L.; et al. Respiratory viruses, symptoms, and inflammatory markers in acute exacerbations and stable chronic obstructive pulmonary disease. Am. J. Respir. Crit. Care Med. 2001, 164, 1618–1623. [Google Scholar] [CrossRef]
  214. Coultas, J.A.; Cafferkey, J.; Mallia, P.; Johnston, S.L. Experimental Antiviral Therapeutic Studies for Human Rhinovirus Infections. J. Exp. Pharmacol. 2021, 13, 645–659. [Google Scholar] [CrossRef]
  215. Warner, S.M.; Wiehler, S.; Michi, A.N.; Proud, D. Rhinovirus replication and innate immunity in highly differentiated human airway epithelial cells. Respir. Res. 2019, 20, 150. [Google Scholar] [CrossRef]
  216. Arruda, E.; Boyle, T.R.; Winther, B.; Pevear, D.C.; Gwaltney, J.M., Jr.; Hayden, F.G. Localization of Human Rhinovirus Replication in the Upper Respiratory Tract by In Situ Hybridization. J. Infec. Dis. 1995, 171, 1329–1333. [Google Scholar] [CrossRef]
  217. Papadopoulos, N.G.; Bates, P.J.; Bardin, P.G.; Papi, A.; Leir, S.H.; Fraenkel, D.J.; Meyer, J.; Lackie, P.M.; Sanderson, G.; Holgate, S.T.; et al. Rhinoviruses Infect the Lower Airways. J. Infec. Dis. 2000, 181, 1875–1884. [Google Scholar] [CrossRef] [Green Version]
  218. Ganjian, H.; Rajput, C.; Elzoheiry, M.; Sajjan, U. Rhinovirus and Innate Immune Function of Airway Epithelium. Front. Cell. Infect. Microbiol. 2020, 10, 277. [Google Scholar] [CrossRef]
  219. Boland, H.; Adrian, E.; Schwarzbach, H.; Burger-Kentischer, A.; Jonigk, D.; Braubach, P.; Rohde, G.; Bellinghausen, C. Protective effect of interferon type I on barrier function of human airway epithelium during rhinovirus infections in vitro. ERJ Open Res. 2022, 8, 92. [Google Scholar] [CrossRef]
  220. Rao, R. Oxidative stress-induced disruption of epithelial and endothelial tight junctions. Front. Biosci. 2008, 13, 7210–7226. [Google Scholar] [CrossRef] [Green Version]
  221. Yamamoto, N.; Kan, O.K.; Tatsuta, M.; Ishii, Y.; Ogawa, T.; Shinozaki, S.; Fukuyama, S.; Nakanishi, Y.; Matsumoto, K. Incense smoke-induced oxidative stress disrupts tight junctions and bronchial epithelial barrier integrity and induces airway hyperresponsiveness in mouse lungs. Sci. Rep. 2021, 11, 7222. [Google Scholar] [CrossRef]
  222. Biagioli, M.C.; Kaul, P.; Singh, I.; Turner, R.B. The role of oxidative stress in rhinovirus induced elaboration of IL-8 by respiratory epithelial cells. Free Radic. Biol. Med. 1999, 26, 454–462. [Google Scholar] [CrossRef]
  223. Panday, A.; Sahoo, M.K.; Osorio, D.; Batra, S. NADPH oxidases: An overview from structure to innate immunity-associated pathologies. Cell. Mol. Immunol. 2015, 12, 5–23. [Google Scholar] [CrossRef] [Green Version]
  224. Khomich, O.A.; Kochetkov, S.N.; Bartosch, B.; Ivanov, A.V. Redox Biology of Respiratory Viral Infections. Viruses 2018, 10, 392. [Google Scholar] [CrossRef] [Green Version]
  225. Gao, X.-Y.; Liu, G.-C.; Zhang, J.-X.; Wang, L.-H.; Xu, C.; Yan, Z.-A.; Wang, A.; Su, Y.-F.; Lee, J.-J.; Piao, G.-C.; et al. Pharmacological Properties of Ginsenoside Re. Front. Pharmacol. 2022, 13, 754191. [Google Scholar] [CrossRef]
  226. Huang, W.H.; Lee, A.R.; Yang, C.H. Antioxidative and anti-inflammatory activities of polyhydroxyflavonoids of Scutellaria baicalensis GEORGI. Biosci. Biotechnol. Biochem. 2006, 70, 2371–2380. [Google Scholar] [CrossRef] [Green Version]
  227. Krammer, F.; Smith, G.J.D.; Fouchier, R.A.M.; Peiris, M.; Kedzierska, K.; Doherty, P.C.; Palese, P.; Shaw, M.L.; Treanor, J.; Webster, R.G.; et al. Influenza. Nat. Rev. Dis. Prim. 2018, 4, 3. [Google Scholar] [CrossRef] [PubMed]
  228. Uyeki, T.M.; Hui, D.S.; Zambon, M.; Wentworth, D.E.; Monto, A.S. Influenza. Lancet 2022, 400, 693–706. [Google Scholar] [CrossRef] [PubMed]
  229. Leung, N.H.L. Transmissibility and transmission of respiratory viruses. Nat. Rev. Microbiol. 2021, 19, 528–545. [Google Scholar] [CrossRef]
  230. Kalil, A.C.; Thomas, P.G. Influenza virus-related critical illness: Pathophysiology and epidemiology. Crit. Care 2019, 23, 258. [Google Scholar] [CrossRef] [Green Version]
  231. Short, K.R.; Kroeze, E.J.B.V.; Fouchier, R.A.M.; Kuiken, T. Pathogenesis of influenza-induced acute respiratory distress syndrome. Lancet Infect. Dis. 2014, 14, 57–69. [Google Scholar] [CrossRef] [PubMed]
  232. WHO. Influenza in Focus: Up to 650,000 People Die of Respiratory Diseases Linked to Seasonal Flu Each Year. Available online: https://www.who.int/news/item/13-12-2017-up-to-650-000-people-die-of-respiratory-diseases-linked-to-seasonal-flu-each-year (accessed on 3 November 2022).
  233. CDC. Estimated Flu-Related Illnesses, Medical Visits, Hospitalizations, and Deaths in the United States—2019–2020 Flu Season. Available online: https://www.cdc.gov/flu/about/burden/2019-2020.html (accessed on 3 November 2022).
  234. Huang, Q.J.; Song, K.; Xu, C.; Bolon, D.N.A.; Wang, J.P.; Finberg, R.W.; Schiffer, C.A.; Somasundaran, M. Quantitative structural analysis of influenza virus by cryo-electron tomography and convolutional neural networks. Structure 2022, 30, 777–786.e3. [Google Scholar] [CrossRef] [PubMed]
  235. Denney, L.; Ho, L.P. The role of respiratory epithelium in host defence against influenza virus infection. Biomed. J. 2018, 41, 218–233. [Google Scholar] [CrossRef] [PubMed]
  236. Mauad, T.; Hajjar, L.A.; Callegari, G.D.; da Silva, L.F.; Schout, D.; Galas, F.R.; Alves, V.A.; Malheiros, D.M.; Auler, J.O., Jr.; Ferreira, A.F.; et al. Lung pathology in fatal novel human influenza A (H1N1) infection. Am. J. Respir. Crit. Care Med. 2010, 181, 72–79. [Google Scholar] [CrossRef]
  237. Atkin-Smith, G.K.; Duan, M.; Chen, W.; Poon, I.K.H. The induction and consequences of Influenza A virus-induced cell death. Cell Death Dis. 2018, 9, 1002. [Google Scholar] [CrossRef] [PubMed]
  238. Branche, A.R.; Falsey, A.R. Parainfluenza Virus Infection. Semin. Respir. Crit. Care Med. 2016, 37, 538–554. [Google Scholar] [CrossRef] [PubMed]
  239. Henrickson, K.J. Parainfluenza viruses. Clin. Microbiol. Rev. 2003, 16, 242–264. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  240. Chen, Z. Parainfluenza virus 5-vectored vaccines against human and animal infectious diseases. Rev. Med. Virol. 2018, 28, e1965. [Google Scholar] [CrossRef]
  241. Burke, C.W.; Bridges, O.; Brown, S.; Rahija, R.; Russell, C.J. Mode of parainfluenza virus transmission determines the dynamics of primary infection and protection from reinfection. PLoS Pathog. 2013, 9, e1003786. [Google Scholar] [CrossRef] [Green Version]
  242. Cui, J.; Li, F.; Shi, Z.-L. Origin and evolution of pathogenic coronaviruses. Nat. Rev. Microbiol. 2019, 17, 181–192. [Google Scholar] [CrossRef] [Green Version]
  243. Schoeman, D.; Gordon, B.; Fielding, B.C. Coronaviruses. In Encyclopedia of Infection and Immunity; Rezaei, N., Ed.; Elsevier: Oxford, UK, 2022; pp. 241–258. [Google Scholar]
  244. Drosten, C.; Günther, S.; Preiser, W.; van der Werf, S.; Brodt, H.R.; Becker, S.; Rabenau, H.; Panning, M.; Kolesnikova, L.; Fouchier, R.A.; et al. Identification of a novel coronavirus in patients with severe acute respiratory syndrome. N. Engl. J. Med. 2003, 348, 1967–1976. [Google Scholar] [CrossRef]
  245. Peiris, J.S.; Lai, S.T.; Poon, L.L.; Guan, Y.; Yam, L.Y.; Lim, W.; Nicholls, J.; Yee, W.K.; Yan, W.W.; Cheung, M.T.; et al. Coronavirus as a possible cause of severe acute respiratory syndrome. Lancet 2003, 361, 1319–1325. [Google Scholar] [CrossRef] [Green Version]
  246. Ksiazek, T.G.; Erdman, D.; Goldsmith, C.S.; Zaki, S.R.; Peret, T.; Emery, S.; Tong, S.; Urbani, C.; Comer, J.A.; Lim, W.; et al. A novel coronavirus associated with severe acute respiratory syndrome. N. Engl. J. Med. 2003, 348, 1953–1966. [Google Scholar] [CrossRef]
  247. Perlman, S.; Netland, J. Coronaviruses post-SARS: Update on replication and pathogenesis. Nat. Rev. Microbiol. 2009, 7, 439–450. [Google Scholar] [CrossRef] [Green Version]
  248. Fung, T.S.; Liu, D.X. Human Coronavirus: Host-Pathogen Interaction. Annu. Rev. Microbiol. 2019, 73, 529–557. [Google Scholar] [CrossRef]
  249. Groot, R.J.d.; Baker, S.C.; Baric, R.S.; Brown, C.S.; Drosten, C.; Enjuanes, L.; Fouchier, R.A.M.; Galiano, M.; Gorbalenya, A.E.; Memish, Z.A.; et al. Commentary: Middle East Respiratory Syndrome Coronavirus (MERS-CoV): Announcement of the Coronavirus Study Group. J. Virol. 2013, 87, 7790–7792. [Google Scholar] [CrossRef] [Green Version]
  250. Zaki, A.M.; van Boheemen, S.; Bestebroer, T.M.; Osterhaus, A.D.; Fouchier, R.A. Isolation of a novel coronavirus from a man with pneumonia in Saudi Arabia. N. Engl. J. Med. 2012, 367, 1814–1820. [Google Scholar] [CrossRef]
  251. Chafekar, A.; Fielding, B.C. MERS-CoV: Understanding the Latest Human Coronavirus Threat. Viruses 2018, 10, 93. [Google Scholar] [CrossRef]
  252. 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] [Green Version]
  253. WHO. WHO Coronavirus (COVID-19) Dashboard. Available online: https://covid19.who.int (accessed on 3 November 2022).
  254. Sharif-Yakan, A.; Kanj, S.S. Emergence of MERS-CoV in the Middle East: Origins, transmission, treatment, and perspectives. PLoS Pathog. 2014, 10, e1004457. [Google Scholar] [CrossRef] [Green Version]
  255. Van Doremalen, N.; Bushmaker, T.; Morris, D.H.; Holbrook, M.G.; Gamble, A.; Williamson, B.N.; Tamin, A.; Harcourt, J.L.; Thornburg, N.J.; Gerber, S.I.; et al. Aerosol and Surface Stability of SARS-CoV-2 as Compared with SARS-CoV-1. N. Engl. J. Med. 2020, 382, 1564–1567. [Google Scholar] [CrossRef]
  256. Meselson, M. Droplets and Aerosols in the Transmission of SARS-CoV-2. N. Engl. J. Med. 2020, 382, 2063. [Google Scholar] [CrossRef]
  257. Mann, R.; Perisetti, A.; Gajendran, M.; Gandhi, Z.; Umapathy, C.; Goyal, H. Clinical Characteristics, Diagnosis, and Treatment of Major Coronavirus Outbreaks. Front. Med. 2020, 7, 581521. [Google Scholar] [CrossRef]
  258. Robinson, P.C.; Liew, D.F.L.; Tanner, H.L.; Grainger, J.R.; Dwek, R.A.; Reisler, R.B.; Steinman, L.; Feldmann, M.; Ho, L.-P.; Hussell, T.; et al. COVID-19 therapeutics: Challenges and directions for the future. Proc. Natl. Acad. Sci. USA 2022, 119, e2119893119. [Google Scholar] [CrossRef]
  259. Van de Veerdonk, F.L.; Giamarellos-Bourboulis, E.; Pickkers, P.; Derde, L.; Leavis, H.; van Crevel, R.; Engel, J.J.; Wiersinga, W.J.; Vlaar, A.P.J.; Shankar-Hari, M.; et al. A guide to immunotherapy for COVID-19. Nat. Med. 2022, 28, 39–50. [Google Scholar] [CrossRef] [PubMed]
  260. Song, L.-G.; Xie, Q.-X.; Lao, H.-L.; Lv, Z.-Y. Human coronaviruses and therapeutic drug discovery. Infect. Dis. Poverty 2021, 10, 28. [Google Scholar] [CrossRef] [PubMed]
  261. Cascella, M.; Rajnik, M.; Aleem, A.; Dulebohn, S.C.; Di Napoli, R. Features, Evaluation, and Treatment of Coronavirus (COVID-19). In StatPearls; StatPearls Publishing LLC: Treasure Island, FL, USA, 2022. [Google Scholar]
  262. Li, W.; Moore, M.J.; Vasilieva, N.; Sui, J.; Wong, S.K.; Berne, M.A.; Somasundaran, M.; Sullivan, J.L.; Luzuriaga, K.; Greenough, T.C.; et al. Angiotensin-converting enzyme 2 is a functional receptor for the SARS coronavirus. Nature 2003, 426, 450–454. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  263. Qian, Z.; Travanty, E.A.; Oko, L.; Edeen, K.; Berglund, A.; Wang, J.; Ito, Y.; Holmes, K.V.; Mason, R.J. Innate immune response of human alveolar type II cells infected with severe acute respiratory syndrome-coronavirus. Am. J. Respir. Cell Mol. Biol. 2013, 48, 742–748. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  264. Lu, G.; Hu, Y.; Wang, Q.; Qi, J.; Gao, F.; Li, Y.; Zhang, Y.; Zhang, W.; Yuan, Y.; Bao, J.; et al. Molecular basis of binding between novel human coronavirus MERS-CoV and its receptor CD26. Nature 2013, 500, 227–231. [Google Scholar] [CrossRef] [Green Version]
  265. Choudhry, H.; Bakhrebah, M.A.; Abdulaal, W.H.; Zamzami, M.A.; Baothman, O.A.; Hassan, M.A.; Zeyadi, M.; Helmi, N.; Alzahrani, F.; Ali, A.; et al. Middle East respiratory syndrome: Pathogenesis and therapeutic developments. Future Virol. 2019, 14, 237–246. [Google Scholar] [CrossRef] [Green Version]
  266. Hoffmann, M.; Kleine-Weber, H.; Schroeder, S.; Krüger, N.; Herrler, T.; Erichsen, S.; Schiergens, T.S.; Herrler, G.; Wu, N.H.; Nitsche, A.; et al. SARS-CoV-2 Cell Entry Depends on ACE2 and TMPRSS2 and Is Blocked by a Clinically Proven Protease Inhibitor. Cell 2020, 181, 271–280.E8. [Google Scholar] [CrossRef]
  267. Zhang, Q.; Xiang, R.; Huo, S.; Zhou, Y.; Jiang, S.; Wang, Q.; Yu, F. Molecular mechanism of interaction between SARS-CoV-2 and host cells and interventional therapy. Signal Transduct. Target. Ther. 2021, 6, 233. [Google Scholar] [CrossRef]
  268. Vassiliou, A.G.; Kotanidou, A.; Dimopoulou, I.; Orfanos, S.E. Endothelial Damage in Acute Respiratory Distress Syndrome. Int. J. Mol. Sci. 2020, 21, 8793. [Google Scholar] [CrossRef]
  269. D’Agnillo, F.; Walters, K.-A.; Xiao, Y.; Sheng, Z.-M.; Scherler, K.; Park, J.; Gygli, S.; Rosas, L.A.; Sadtler, K.; Kalish, H.; et al. Lung epithelial and endothelial damage, loss of tissue repair, inhibition of fibrinolysis, and cellular senescence in fatal COVID-19. Sci. Transl. Med. 2021, 13, eabj7790. [Google Scholar] [CrossRef]
  270. Rouaud, F.; Méan, I.; Citi, S. The ACE2 Receptor for Coronavirus Entry Is Localized at Apical Cell—Cell Junctions of Epithelial Cells. Cells 2022, 11, 627. [Google Scholar] [CrossRef]
  271. Wang, Q.; Chen, X.-W.; Margolis, B. PALS1 Regulates E-Cadherin Trafficking in Mammalian Epithelial Cells. Mol. Biol. Cell 2007, 18, 874–885. [Google Scholar] [CrossRef] [Green Version]
  272. De Maio, F.; Lo Cascio, E.; Babini, G.; Sali, M.; Della Longa, S.; Tilocca, B.; Roncada, P.; Arcovito, A.; Sanguinetti, M.; Scambia, G.; et al. Improved binding of SARS-CoV-2 Envelope protein to tight junction-associated PALS1 could play a key role in COVID-19 pathogenesis. Microbes Infect. 2020, 22, 592–597. [Google Scholar] [CrossRef]
  273. Shepley-McTaggart, A.; Sagum, C.A.; Oliva, I.; Rybakovsky, E.; DiGuilio, K.; Liang, J.; Bedford, M.T.; Cassel, J.; Sudol, M.; Mullin, J.M.; et al. SARS-CoV-2 Envelope (E) protein interacts with PDZ-domain-2 of host tight junction protein ZO1. PLoS ONE 2021, 16, e0251955. [Google Scholar] [CrossRef]
  274. Hekman, R.M.; Hume, A.J.; Goel, R.K.; Abo, K.M.; Huang, J.; Blum, B.C.; Werder, R.B.; Suder, E.L.; Paul, I.; Phanse, S.; et al. Actionable Cytopathogenic Host Responses of Human Alveolar Type 2 Cells to SARS-CoV-2. Mol. Cell 2020, 80, 1104–1122.E9. [Google Scholar] [CrossRef]
  275. Adil, M.S.; Khulood, D.; Narayanan, S.P.; Somanath, P.R. Bioinformatics analyses reveal cell-barrier junction modulations in lung epithelial cells on SARS-CoV-2 infection. Tissue Barriers 2022, 10, 2000300. [Google Scholar] [CrossRef]
  276. Tugizov, S. Virus-associated disruption of mucosal epithelial tight junctions and its role in viral transmission and spread. Tissue Barriers 2021, 9, 1943274. [Google Scholar] [CrossRef]
Figure 1. Schematic diagram illustrating the structure of apical junction complexes in the airway epithelium. In the human airway epithelium, the surface mucus layers and the physical barrier formed by airway epithelial cells serve as the first line of defense against the external environment. The apical junctional complexes (AJCs) between adjacent epithelial cells establish cell polarity and restrict epithelial permeability via maintaining cell–cell contact. AJCs are located on the top lateral membranes between neighboring cells and include tight junctions (TJs) and adherens junctions (AJs). The inset shows an enlarged illustration of several protein components of TJ and AJ. Claudin, occludin, and junctional adhesion molecule (JAM) are examples of TJ transmembrane proteins. E-cadherin and nectin are shown as examples of the AJ transmembrane proteins. These proteins interact with cytoplasmic adapter proteins, such as zonula occludens (ZO-1/2/3) and β-catenin, to connect to the prejunctional actin cytoskeleton. The assembly and remodeling of the filamentous actin (F-actin) network are critical for the epithelial barrier function and are regulated by key organizers such as cortactin and actin-related protein-2/3 (Arp2/3) complex. This image was created with BioRender.com.
Figure 1. Schematic diagram illustrating the structure of apical junction complexes in the airway epithelium. In the human airway epithelium, the surface mucus layers and the physical barrier formed by airway epithelial cells serve as the first line of defense against the external environment. The apical junctional complexes (AJCs) between adjacent epithelial cells establish cell polarity and restrict epithelial permeability via maintaining cell–cell contact. AJCs are located on the top lateral membranes between neighboring cells and include tight junctions (TJs) and adherens junctions (AJs). The inset shows an enlarged illustration of several protein components of TJ and AJ. Claudin, occludin, and junctional adhesion molecule (JAM) are examples of TJ transmembrane proteins. E-cadherin and nectin are shown as examples of the AJ transmembrane proteins. These proteins interact with cytoplasmic adapter proteins, such as zonula occludens (ZO-1/2/3) and β-catenin, to connect to the prejunctional actin cytoskeleton. The assembly and remodeling of the filamentous actin (F-actin) network are critical for the epithelial barrier function and are regulated by key organizers such as cortactin and actin-related protein-2/3 (Arp2/3) complex. This image was created with BioRender.com.
Pharmaceutics 14 02619 g001
Figure 2. Disrupted epithelial barrier induced by invading microorganisms. Bacterial and viral infection can cause AJC disassembly and barrier disruption, the consequent increase in epithelial permeability will allow or exacerbate secondary invasions of allergens and pathogens into the subepithelial space. Multiple molecular pathways have been implicated in mediating or contributing to pathogen-induced AJC disassembly. Some examples are protein kinase C/D (PKC/D) activation, reactive oxygen species (ROS) generation, and rearrangement of the F-actin cytoskeleton. Therapeutics such as agonists and antagonists have been investigated and/or proposed to restore the epithelial barrier function caused in infected epithelial cells. Refer to Table 1; Table 2 for detailed information about the critical pathways and potential therapeutics. cAMP, cyclic adenosine monophosphate; HRV, human rhinovirus; IAV, influenza A virus; MERS-CoV, Middle East respiratory syndrome coronavirus; RSV, respiratory syncytial virus; SARS-CoV, severe acute respiratory syndrome coronavirus; SARS-CoV-2, severe acute respiratory syndrome coronavirus-2. This image was created with BioRender.com.
Figure 2. Disrupted epithelial barrier induced by invading microorganisms. Bacterial and viral infection can cause AJC disassembly and barrier disruption, the consequent increase in epithelial permeability will allow or exacerbate secondary invasions of allergens and pathogens into the subepithelial space. Multiple molecular pathways have been implicated in mediating or contributing to pathogen-induced AJC disassembly. Some examples are protein kinase C/D (PKC/D) activation, reactive oxygen species (ROS) generation, and rearrangement of the F-actin cytoskeleton. Therapeutics such as agonists and antagonists have been investigated and/or proposed to restore the epithelial barrier function caused in infected epithelial cells. Refer to Table 1; Table 2 for detailed information about the critical pathways and potential therapeutics. cAMP, cyclic adenosine monophosphate; HRV, human rhinovirus; IAV, influenza A virus; MERS-CoV, Middle East respiratory syndrome coronavirus; RSV, respiratory syncytial virus; SARS-CoV, severe acute respiratory syndrome coronavirus; SARS-CoV-2, severe acute respiratory syndrome coronavirus-2. This image was created with BioRender.com.
Pharmaceutics 14 02619 g002
Table 1. Bacterial disruption of airway epithelial barrier and AJC.
Table 1. Bacterial disruption of airway epithelial barrier and AJC.
MicroorganismModelImpacts on Airway Epithelial Barrier FunctionImpacts on Apical Junctional Complex (AJC)MechanismsPotential
Therapeutics
Ref
Staphylococcus
aureus (S. aureus)
Primary epithelial cells from CRSwNP patients-ALI;
S. aureus enterotoxin B
↓ TEER,
↑ permeability
↓ ZO-1 (IF),
↓ occludin (IF),
↓ phospho-occludin (WB),
↓ claudin-1(WB)
TLR2 pathwayTLR2 blocking antibody[21]
C57BL/6J mice; S. aureus enterotoxin B↑ Permeability↓ ZO-1 (mRNA),
↓ occludin (mRNA)
Primary HNECs-ALI; S. aureus conditioned media↓ TEERTJ separation (EM),
discontinuous staining of ZO-1
--[26]
Primary HNECs-ALI;
S. aureus extracellular proteases
↓ TEER,
↑ permeability
Discontinuous staining of ZO-1--[27]
Human airway epithelial cells (H441)↑ Permeability↓ ZO-1 (IF),
↓ occludin (IF)
AMPK-PKCζ pathwayMetformin[28]
C57BL/6J mice;
S. aureus α-toxin
↑ Permeability↑ E-cadherin cleavage (WB)ADAM10 activityADAM10 inhibition[29]
A549;
S. aureus α-toxin
↓ Focal adhesion -FAK/Src/F-actin reorganization-[30]
16HBE;
S. aureus α-toxin
↑ Paracellular gaps-PAK/LIMK/cofilin/F-actin reorganization-[31]
Streptococcus
pneumoniae
(S. pneumoniae)
A549, 16HBE, C57BL/6J miceBacterial transmigration, ↑ permeability--IFN-β[20]
A549-↑ E-cadherin cleavage/degradation (WB)ADAM10 activation by pneumolysinADAM10 inhibition[29]
16HBE, C57BL/6J mice↓ TEER,
↑ permeability
↓ Claudin-7,10 (mRNA)TLR/p38/TGF-β/Snail1-[32]
Human respiratory tissues-TJ separation (EM)--[33]
H292,
BALB/c mice
-↓ E-cadherin (IF)--[34]
human lung tissue-(IF, WB)
↓ ZO-1,
↓ occludin,
↓ claudin-5,
↓ VE-cadherin
--[35]
A549 Bacterial transmigration↑VE-cadherin cleavage/degradation (WB)bacteria-bound plasmin-[36]
Pseudomonas
aeruginosa
(P. aeruginosa)
Calu3↓ TEER,
↑ permeability
↓ occludin and claudin-1,
↑ cleavage of occludin (WB)
hyperglycemiaMetformin[37]
16HBE↓ TEER,
↑ permeability
↓ ZO-1 (IF)--[38]
C57BL/6 mice↑ Permeability---
Primary HNECs; P. aeruginosa elastase (PE)↓ TEER
(transient)
Transient ↓ claudin-1 and -4, occludin, and tricellulin (WB)Decreased PAR-2; activation of PKC, MAPK, PI3K, p38 MAPK, JNK, COX-1 and -2, and NF-κB pathwaysPAR-2 agonist; inhibitors for PKC, MAPK, PI3K, p38 MAPK, JNK, COX-1 and -2, and NF-κB pathways[39]
Calu3;
P. aeruginosa elastase (PE)
↓ TEER↓ localization of ZO-1 and occludin on membrane (IF, WB)PKC signaling/ F-actin reorganizationPKC inhibitor[40]
Primary HNECs-ALI↓ TEER,
↑ permeability
-endotoxin rhamnolipids-[41]
16HBE↑ PermeabilityAltered distribution of ZO-1 (IF), ezrin (IF), and occludin (IF, WB)Type III toxins (ExoS, -T, and -Y. ExoS)-[42]
Burkholderia16HBE↓ TEER,
↑ permeability
↓ occludin (IF)occludin dephosphorylation-[43]
Human lung explant↑ Permeability---[44]
Primary Human AECs-ALI; B. cepacia BC-7Invasion and destruction of epithelial cells (EM)-Biofilm-dependent, rearrangements of the actin cytoskeleton-[45]
Primary Human AECs-ALI; B. cepacia HI2258Invasion and destruction of epithelial cells (EM)-Biofilm-independent-
Primary Human AECs-ALI; B. cepacia J-1Invasion and destruction of epithelial cells (EM)-Biofilm-dependent and independent-
Calu-3↓ TEER↓ ZO-1 and E-cadherin (IF)--[46]
Primary HBECs-ALI, 16HBE↓ TEERDisrupted occludin (IF)Increasing TNF-α cytokineTNF-α neutralizing agent and steroids[47]
16HBE, CF cell line (CFBE41o−)↓ TEER↓ ZO-1 (IF, WB)--[48]
16HBE, CF cell line (CFBE41o−)↓ TEER,
↑ permeability
↓ ZO-1, occludin, and claudin-1 (WB)--[49]
Haemophilus influenzae (H. influenzae)Primary human alveolar epithelial cells type II, A549-↓ E-cadherin (IF, WB, mRNA), ↓ ZO-1 (IF)FGF2 upregulation and activation of mTOR pathwayrapamycin[50]
List of abbreviations: 16HBE, 16HBE14o-human bronchial epithelial; ADAM10, metalloproteinase domain-containing protein 10; AECs, Airway epithelial cells; ALI, air–liquid interface; AMPK, AMP-activated protein kinase; CF, cystic fibrosis; COX-1 and -2, cyclooxygenase-1 and -2; CRSwNP, chronic rhinosinusitis with nasal polyps; EM, electron microscopy; F-actin, filamentous actin; FAK, focal adhesion kinase; HBECs, human bronchial epithelial cells; HNECs, human nasal epithelial cells; IF, immunofluorescence; JNK, c-Jun N-terminal kinase; MAPK, mitogen-activated protein kinase; NF-κB, nuclear factor-κB; PAR-2, protease-activated receptors; PI3K, phosphoinositide 3-kinase; PKC, protein kinase C; Src, steroid receptor coactivator; TEER, trans-epithelial electrical resistance; TGF-β, transforming growth factor-β; TLR, Toll-like receptor; TNF-α, tumor necrosis factor-α; TJ, tight junction; WB, Western blot; ZO, zonula occludens. ↓, decreased; ↑, increased; -, not determined.
Table 2. Virial disruption of airway epithelial barrier and AJC.
Table 2. Virial disruption of airway epithelial barrier and AJC.
MicroorganismModelImpacts on Airway Epithelial Barrier FunctionImpacts on Apical Junctional Complex (AJC)MechanismsPotential
Therapeutics
Ref
Respiratory Syncytial Virus (RSV)Primary mTECs -ALI↓ TEER,
↑ permeability
disassembly of ZO-1, occludin, β-catenin (IF),
↑ E-cadherin cleavage (WB)
--[15]
C57BL/6J mice↑ Permeability↓ ZO-1 (IHC, WB),
mislocalization of occludin (IHC),
↓ occludin (WB),
↓ claudin-1 (IHC, WB),
↑ claudin-2 (IHC, WB),
↑ E-cadherin cleavage (WB)
--
16HBE↓ TEER,
↑ permeability
disassembly of ZO-1, occludin, E-cadherin, β-catenin (IF)PKD activation/actin cytoskeletal remodelingPKD inhibitors[18]
Primary NHBE cells-ALI, 16HBE↓ TEER,
↑ permeability
disassembly of ZO-1, occludin, E-cadherin, β-catenin (IF)-Forskolin, cAMP analog[51]
Primary HBECs, A549↓TEER,
↑ paracellular gaps
-p38 MAPK activation/actin cytoskeletal remodelingp38 MAPK inhibitor[52]
16HBE↓ TEER,
↑ permeability
disassembly of ZO-1, occludin, E-cadherin, β-catenin (IF)cortactin decrease/Rap1 inhibition/actin cytoskeletal remodelingF-actin stabilizer, Rap1 activator [53]
BALB/c mice↑ Permeability↓ occludin and claudin-1 (mRNA)--[54]
Primary HNECs↑ TEER↑ claudin-4 and occludin (IF, WB, mRNA)TGF-β1/PKCδ/HIF-1α/NF-κB-[55]
Primary HBECs↓ TEER,
↑ paracellular gaps
-inducing VEGFVEGF antibody[56]
Human Rhinovirus (HRV)Primary human AECs-ALI↓ TEER,
↑ permeability (transient)
↓ occludin, Crumbs3, and E-cadherin (IF, WB, mRNA)EGFR activation and Snail increaseSnail inhibition[57]
Primary human AECs-ALI,
16HBE, calu-3, C57BL/6 mice
↓ TEER,
↑ permeability
dissociation of ZO-1 from TJ (IF, IHC, WB)--[58]
Primary HNECs-ALI↓ TEER--Betamethasone[59]
Primary NHBE cells-ALI↓ TEER,
↑ permeability
disruption of ZO-1 and occludin (IF)PGC-1α decreasePGC-1α activator[60]
Primary human AECs-ALI,
NuLi-1
↑ Permeability↓ membrane ZO-1, occludin, and claudin-1 (In-Cell Western),
↓ ZO-1 (mRNA)
IL-15-[61]
Primary HNECs-ALI↓ TEER↓ ZO-1, occludin, claudin-1, and E-cadherin (IF, WB, mRNA)--[62]
Primary HNECs-ALI-↓ ZO-1, occludin, claudin-1, and E-cadherin (WB)ROS-mediated phosphatases inhibitionNOX inhibitor[63]
Primary human AECs from asthmatic children-ALI
↓ TEER,
↑ permeability
↓ ZO-1 and occludin (In-Cell Western, IF),
↓ claudin-1 (In-Cell Western)
--[64]
Human precision-cut lung slices-↓ claudin-8 (mRNA)--[65]
16HBE↓ TEER,
↑ bacterial transmigration
↓ ZO-1, occludin (IF)ROS generationRac1/NOX/NOX1 inhibitor, quercetin[66]
16HBE↓ TEER,
↑ bacterial transmigration
↓ occludin (IF, WB)mitochondrial ROS generationantioxidant targeted to mitochondria[67]
Primary HNECs-ALI-Disruption of ZO-1, occludin, claudin-1, and E-cadherin (WB)ROS generationGinsenoside Re[68]
Primary HNECs-ALI, RPMI 2650-Disruption of ZO-1, occludin, claudin-1, and E-cadherin (WB)Akt/NF-κB and ERK1/2 activationWogonin[69]
Influenza A Virus (IAV)Coculture of human alveolar epithelial and endothelial cells; H1N1 and H5N1↓ TEER,
↑ permeability
↓ AJC integrity (EM),
↓ JAM (IF),
↓ claudin-4 (IF)
--[16]
Primary NHBE cells-ALI; H1N1↓ TEER,
↑ permeability
Disassembly of ZO-1 (IF)--[17]
Primary human alveolar epithelial cells type II; H1N1↓ TEER,
↑ permeability
---[70]
A549,
C57BL/6J mice;
H1N1
↓ TEER
↑ permeability
↓ ZO-1 (WB, IF)
↓ occludin (IF, WB)
↓ E-cadherin (WB)
MAPK/PI3K/Gli1/Snail activationGli1 Inhibitor[71]
A549, NL20,
C57BL/6J mice;
H5N1
↓ TEER↓ ZO-1(WB),
↓ occludin (IF, WB),
↓ claudin-1 (IF, WB),
↓ E-cadherin (WB)
TAK1/Itch- mediated protein degradationTAK1-Itch inhibition[72]
Human Parainfluenza Virus (HPIV)Primary human AECs-ALI;
HPIV3 and HPIV5
↓ TEER,
↑ permeability
---[73]
A549;
HPIV2
Cell-to-cell spread of HPIV2↑ claudin-1 mRNAHPIV2 V proteinIncreasing claudin-1[74]
Severe Acute Respiratory Syndrome
(SARS-CoV)
MDCKII; SARS-E protein overexpression↓ TEER↓ E-cadherin
and ZO-1(IF),
delayed the formation of TJs (IF)
PALS1 Binding-[75]
Severe Acute Respiratory Syndrome 2
(SARS-CoV-2)
Organotypic human airway epithelial cultures-ALI↓ TEER---[76]
Reconstructed human bronchial epithelium-ALI↓ TEER,
↑ permeability (transient)
Disrupted ZO-1 (IF)--[77]
Chip model of human epithelial, endothelial, and mononuclear cells↑ PermeabilityDisrupted cell–cell contact (SEM), Disrupted E-cadherin (IF)--[78]
List of abbreviations: cAMP, cyclic adenosine monophosphate; EGFR, epidermal growth factor receptor; ERK1/2, extracellular signal-regulated protein kinase 1/2; HIF-1α, hypoxia-inducible factor 1-alpha; IHC, immunohistochemistry; IL, interleukin; mTECs, mouse tracheal epithelial cells; MDCKII, Madin–Darby Canine Kidney II epithelial cells; NHBE, normal human bronchial epithelial; NOX: NADPH oxidase; PGC-1α, peroxisome proliferator-activated receptor-gamma coactivator-1α; PKD, protein kinase D; ROS: reactive oxygen species; SEM, scanning electron microscopy; VEGF, vascular endothelial growth factor. ↓, decreased; ↑, increased; -, not determined.
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Gao, N.; Rezaee, F. Airway Epithelial Cell Junctions as Targets for Pathogens and Antimicrobial Therapy. Pharmaceutics 2022, 14, 2619. https://doi.org/10.3390/pharmaceutics14122619

AMA Style

Gao N, Rezaee F. Airway Epithelial Cell Junctions as Targets for Pathogens and Antimicrobial Therapy. Pharmaceutics. 2022; 14(12):2619. https://doi.org/10.3390/pharmaceutics14122619

Chicago/Turabian Style

Gao, Nannan, and Fariba Rezaee. 2022. "Airway Epithelial Cell Junctions as Targets for Pathogens and Antimicrobial Therapy" Pharmaceutics 14, no. 12: 2619. https://doi.org/10.3390/pharmaceutics14122619

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