Next Article in Journal
Tracking of Systemic Lupus Erythematosus (SLE) Longitudinally Using Biosensor and Patient-Reported Data: A Report on the Fully Decentralized Mobile Study to Measure and Predict Lupus Disease Activity Using Digital Signals—The OASIS Study
Previous Article in Journal
Human TIMP1 Is a Growth Factor That Improves Oocyte Developmental Competence
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Hypothesis

Bacterial Proteases as Potentially Exploitable Modulators of SARS-CoV-2 Infection: Logic from the Literature, Informatics, and Inspiration from the Dog

by
Gerald H. Lushington
1,
Annika Linde
2 and
Tonatiuh Melgarejo
2,*
1
Qnapsyn Biosciences, Inc., Lawrence, KS 66046, USA
2
College of Veterinary Medicine, Western University of Health Sciences, Pomona, CA 91766, USA
*
Author to whom correspondence should be addressed.
Submission received: 11 July 2023 / Revised: 19 August 2023 / Accepted: 18 October 2023 / Published: 30 October 2023

Abstract

:
(1) Background: The COVID-19 pandemic left many intriguing mysteries. Retrospective vulnerability trends tie as strongly to odd demographics as to exposure profiles, genetics, health, or prior medical history. This article documents the importance of nasal microbiome profiles in distinguishing infection rate trends among differentially affected subgroups. (2) Hypothesis: From a detailed literature survey, microbiome profiling experiments, bioinformatics, and molecular simulations, we propose that specific commensal bacterial species in the Pseudomonadales genus confer protection against SARS-CoV-2 infections by expressing proteases that may interfere with the proteolytic priming of the Spike protein. (3) Evidence: Various reports have found elevated Moraxella fractions in the nasal microbiomes of subpopulations with higher resistance to COVID-19 (e.g., adolescents, COVID-19-resistant children, people with strong dietary diversity, and omnivorous canines) and less abundant ones in vulnerable subsets (the elderly, people with narrower diets, carnivorous cats and foxes), along with bioinformatic evidence that Moraxella bacteria express proteases with notable homology to human TMPRSS2. Simulations suggest that these proteases may proteolyze the SARS-CoV-2 spike protein in a manner that interferes with TMPRSS2 priming.
Key Contribution: Why did COVID-19 severely impact some individuals while leaving others largely unaffected? Based on assimilation of multidisciplinary evidence, tied together with informatics and molecular simulations, this manuscript argues for commensal bacterial protease having a significant overlooked influence on host vulnerability to SARS-CoV-2 infections.

1. Introduction

For all its rigor, science is also a discipline of intuition and adventition. Since the days of Friedrich Kekulé correctly perceiving the structure of benzene from his vision of a snake eating its tail, scientists have benefited from unexpected analogies that have suddenly emerged to explain odd observations. In an attempt to rationalize a disparate series of counter-intuitive epidemiological trends emanating from the COVID-19 pandemic, this manuscript develops a hypothesis that may help to resolve questions regarding idiosyncratic variations in human infectious vulnerability based, originally, on our observations on the varying microbiotic profiles of canines and felines.
Among the many ways in which COVID-19 has impacted humanity, the most profound one may actually prove beneficial—a period of prodigious scientific discovery aimed at advancing global health [1,2,3,4,5,6,7,8,9,10,11,12,13]. Adding to the disciplines influenced by pandemic-related research, this hypothesis explores intriguing evidence of microbe–virus interplay, combining verified epidemiological and biochemical reports with new preliminary analyses to argue for the role of microbial proteases in rationalizing coronavirus epidemiology trends, to illuminate potential opportunities for novel therapeutic strategies.
As the first pandemic of the information age and post-genomic era, COVID-19 brought tremendous investigative resources bearing on virology, immunology, and epidemiology, producing a stunning array of new insight that should help to spark new research for years to come. The greatest short-term technological impact of the COVID-19 response was likely the remarkable success of anti-SARS-CoV-2 vaccine programs, but other critical advances in viral testing and surveillance were also achieved with rapid effectiveness [1,2,3,4]. The protocols by which each of these high-impact COVID-19-specific technologies was attained were sophisticated and timely and offer excellent roadmaps for tackling future epidemics, thus bettering the future of world health.
Beyond the aforementioned developments, one must also consider advances in risk-profile analysis, producing important insight into why some subsets of the human population have demonstrated elevated risk of suffering more severe COVID-19 outcomes. Statistical support for many posited risk factors has declined through the pandemic, but some, such as a link between severe COVID-19 and prior medical history of cardiovascular and metabolic disorders (primarily diabetes, hypertension, and obesity) have gained strength according to extensive sustained analysis [5]. Autoimmune risk factors have also been determined to correlate with COVID-19 severity [6,7] and predispose patients to post-acute sequelae [8,9]. Other epidemiologically significant factors include chronic exposure to air pollution [10,11], as well as one metric that assimilates numerous subfactors—socioeconomic status [12,13].
Vaccination, testing, and risk profiling are of tremendous societal value and can substantially reduce overall caseloads and fatalities, but unfortunately, they offer marginal relief to individuals who actually contract dangerous viruses like COVID-19. Further development of safe and reliable treatment strategies for people who fall ill is thus required for full and concerted pandemic response. Much effort has been devoted to designing [14,15,16,17,18] or repurposing [19,20] drugs for SARS-CoV-2 targets, but such pursuits have not yet produced breakthrough success. Such disappointment is not surprising, given the historical challenges of developing antiviral therapeutics [21], but it is imperative to persevere and to innovate. The terrible global toll through a few short years of pandemic, the continuing threat of new coronavirus variants and hitherto undiscovered viral pathogens, and the rhetorical value of devising efficacious antiviral strategies all provide powerful incentives. The most pressing existential threats of COVID-19 may be on the wane, but our best chances to successfully mitigate the next pandemic may emerge from the vast amount of quantitative and qualitative evidence collected during the current one. Conceivably, this body of information may contain crucial mechanistic clues underlying past pharmacological failures and the basis for finding ways to correct them.
One superficially logical strategy for antiviral drug development that rarely tends to achieve lasting therapeutic benefits [22,23,24,25] entails applying standard rational small-molecule design principles toward interfering with viral proteins tasked with host infection or viral replication. One crucial limitation of this approach lies in ignoring long-standing experience with viral resistance, wherein high-specificity targeting is generally overcome by viral genetic evolution, via mutation rates substantially exceeding those of bacteria or cancer cells [26,27,28,29,30]. Alternative host-targeting strategies (e.g., blocking entry-mediating host–protein attachment sites) are less vulnerable to viral resistance, but the strategy has yet to produce major drug candidates [31,32]. Still, these outcomes should be regarded not as abject failures but rather as motivation to formulate new design paradigms. Every recent antiviral disappointment, such as Remdesivir [15], Sofosbuvir [16], and Molnupiravir [17], and controversies over hydroxychloroquine [19] and Ivermectin [20] add new insight into the merits and consequences of modulating viral–host interactions. Such incremental knowledge may eventually inform successful new strategies.
Valuable insights are also emerging from non-traditional hypotheses that may supplant or augment prior pharmacological paradigms. For example, the aforementioned public health risk factor analysis can be cross-purposed to posit circumstances that promote resistance to symptomatic infection or to more tangibly severe manifestations. Indeed, while much of the world is now assumed to have been exposed to SARS-CoV-2, a persistent sub-population exists that, despite not necessarily pursuing rigorous isolation, has experienced few or no symptoms of infection [33,34,35,36,37]. This resistance suggests tangible factors, likely independent of the virus itself, that may enable some individuals to avoid illness. Identifying and characterizing these features might help to inspire novel therapeutic or prophylactic paradigms, thus incentivizing resistivity/vulnerability analysis [33,34,35,36,37].
Genetic factors postulated to protect against COVID-19 infection have been reviewed elsewhere [38], with much attention being paid to aspects of host physiology that may enhance immune-driven clearance of the SARS-CoV-2 virus [38,39,40]. In particular, it is noted that some people display T-cell activity that is measurably more effective in targeting highly conserved features of the virion architecture, leading to rapid viral clearance, even without elevated antibody production, which would normally be indicative of current or prior exposure [37,41]. Other mechanistically vague trends have been noted, such as potential risk reduction among people with pre-existing food allergies [42] and possibly also prior long-term usage of endocrine-modulating drugs like Tamoxifen [43,44]. Ultimately, the breadth and scatter of possible resistance factors has hindered novel drug development, which might have coalesced around any consensus on promising host-related factors.
Given that COVID-19 infections remain difficult to treat [45], especially as new variants diverge from vaccine specificity [46], it is fortunate that the absence of consensus is not a consensus of absence. Rather, if markers from host physiology or medical history fail to support a simple relationship with COVID-19 vulnerability, the difference could implicate other influences as a basis for the relative viral resistance of a given individual. Thus, this hypothesis attains substantial novelty by distancing itself from the host–virus relationship [47] and instead explores evidence that variations in host commensal bacteria, potentially including gut, nasal, and lung microbiota, may differentially facilitate or mitigate viral entry to an extent that affects vulnerability or resistance.
While substantial research has been devoted to co-pathology among various microbes and the SARS-CoV-2 virus [48,49,50,51,52], this hypothesis aims to assess knowledge of the symbiotic host-protective capacity of some bacteria [50,53] while focusing on putative mechanisms at molecular levels of detail, with potential application to therapeutic development. In particular, a novel hypothesis is rationalized, tying key aspects of host protection to the relative capacity of different bacterial serine proteases to interact with the SARS-CoV-2 Spike protein (and other comparable viral Class I fusion proteins present in various other coronaviruses and retroviruses) in a manner that may interfere with the pathological TMPRSS2-mediated process of Spike cleavage and host-membrane permeation.
This hypothesis has emerged from confluent evidence drawn from disparate sources, including the following:
  • Contrast between high-risk versus low-risk segments of the human population;
  • Differential nasal microbiotic profiles of COVID-19 patients versus non-infected people with similar demographics and experiences [54,55,56];
  • Comparative biological assessment of the relative virion-shedding propensity of felines [57] versus various canids [57,58], augmented by microbiotic profiling performed specially for this study across a cohort of client-owned dogs;
  • Genomic and structural biological rationalization through informatics and simulations performed herein.

2. Viral Host Infection: TMPRSS2-Mediated vs. TMPRSS2-Independent Mechanisms

Human TMPRSS2 is a type II trypsin-like transmembrane serine protease that has attracted substantial interest for apparent roles in facilitating host infection by various forms of influenza and coronaviruses [59,60,61,62,63]. Setting aside a persistent lack of consensus over whether TMPRSS2 is primarily expressed by endothelial or epithelial cells [64,65,66,67], it is clear that the protein is abundant on many epithelial surfaces, especially including the lung, stomach, colon, and rectum [68,69,70], but also on the surfaces of sustentacular cells in olfactory mucosa [71]. Although no firm opinion exists on the primary physiological function of TMPRSS2 [72,73], it has gained a great deal of attention for a non-physiological role in priming viral glycoproteins as a precursor to host infection. In particular, research on SARS and MERS coronavirus epidemics spurred a growing recognition of the extent to which TMPRSS2 helps to expedite host-cell fusion and clathrin-mediated endocytosis, as is shown in Figure 1A,B. While various extracellular proteases (furin, ADAM17, and others) are capable of cleaving the SARS-CoV-2 Spike protein along loop regions suitable for infectiously priming the virion, only TMPRSS2 is known to remain associated with both the viral Spike protein and the cellular ACE2 receptor after cleavage. TMPRSS2 thus accomplishes both primary cleavage of Spike and secondary integration with the host-cell membrane, achieving such steps both for coronavirus infections [61,62,74,75] and for various influenza variants [60,61,62,63].
Amid extensive efforts to design COVID-19 therapeutics, attempts have been made to disrupt TMPRSS2-related steps incumbent in SARS-CoV-2 infection [59,76,77]. Little efficacy has been demonstrated to date in such strategies, prompting speculation that TMPRSS2 may facilitate infection without being an absolute requirement for coronavirus and influenza viruses. Indeed, mechanisms for host-cell entry by more recent SARS-CoV-2 variants (especially descendants of the omicron sequence) appear to have reduced reliance on TMPRSS2 [78].
One factor that diminishes the role of TMPRSS2 in promulgating SARS-CoV-2 infections is the apparent capacity of other proteases, such as furin [79,80,81,82], ADAM-10, and ADAM-17 [83,84], to cleave the Spike protein at junctures proximal to the canonical S2′ cleavage sites favored by TMPRSS2, thus achieving comparable release of viral RNA into the host cell. An important advantage that TMPRSS2 offers the virus is a sustained association with ACE2 at the cell surface. This facilitates viral fusion directly with the host-cell plasma membrane.
In the absence of TMPRSS2 (Figure 1C,D), a slower mode of entry may be utilized, whereby host-cell endocytosis ports the virion into an intracellular vesicle, availing an opportunity for subsequent cytosolic release by Cathepsin-B or Cathepsin-L [85,86,87]. The diminished TMPRSS2 reliance of omicron-derived variants of SARS-CoV-2 seems to be due largely to elevated exploitation of this endocytotic cathepsin infection route [78,88,89]. Reduced infectious efficiency of the cathepsin route may explain the altered symptomology exhibited by omicron variants [90,91,92] compared with the TMPRSS2-mediated cell fusion perpetrated by delta (and prior) variants [90]. In particular, the tradeoff for slower individual infection by omicron variants appears to be greater communal transmission enabled by viral preference for transmission-friendly (but TMPRSS2-poor) nasopharyngeal tissue [93,94] while posing less threat to TMPRSS2-rich tissues, such as the deep lung [95,96] and olfactory epithelium [97,98].

3. Role of Protease/Antiprotease Balance in Immunopathology

From a scientific perspective, the rapid, fundamental shift in pathological mechanism achieved by the SARS-CoV-2 omicron variants begs the question of “how and why”. What evolutionary pressure could have favored the rapid genetic shift [99] from an epidemiological successful delta variant to the functionally distinct omicron class, especially when the latter has seemingly offered evolutionary tradeoffs rather than unequivocal mechanistic superiority?
Some answers may tie to the elaborate reliance of various coronaviruses and influenza viruses on proteolytic function, which is regulated within host organisms by a phenomenon known as protease/antiprotease balance (PAB). PAB entails interplay within a superset of proteases, plus an adaptive array of stimulatory and inhibitory proteins and peptides [100,101,102,103]. PAB plays a major role in immunoregulatory function and helps to determine relative resistance or susceptibility to both influenza and COVID-19 infections [104,105], whereas imbalances are implicated in respiratory and gastrointestinal autoimmune conditions [106,107,108,109].
While the relatively static human proteome itself might not have applied evolutionary pressure to spur the complex mutations required to produce omicron, evidence that the omicron variants might have emerged through cross-species rebound (from humans to rodents and back to humans [110,111]) may embody an immunological jolt of intensity sufficient to explain the profound adaptations manifest in the omicron variants. Notably, genomic diversities in rodent proteases and antiproteases are substantially greater than the diversities in those of humans [112,113]. Furthermore, across mammalian proteomes, a disproportionate fraction of protease variations tend to occur in immune-related functions, and in comparison with rodents and some simians, several human immune-relevant protease genes are epigenetically suppressed [114]. However, species-level variations in proteolysis are also not the final determinant, since host microbiomes add substantial diversity to protease/antiprotease balance [108,115], with substantial variation in microbiome constituency being apparent across mammalian species [116,117,118,119], for reasons that include species-level traits and individual genetics, age, sex, diet, and environment.
This hypothesis offers possible insight into the questions raised at the beginning of this section, suggesting further areas of practical inquiry. Relevant questions include the following:
  • Can we propose key players within the proteolytic machinery with major influences on SARS-CoV-2 adaptation and the determination of host vulnerability or resistance to the virus?
  • What are the implications for future SARS-CoV-2 differentiation based on the assumption of PAB as a key evolutionary factor?
  • Can this information be exploited for future health benefits?

4. Host Serpins and Proteases as Modulators of Virus Infection

Among a handful of surprising trends that have emerged regarding COVID-19 vulnerability is growing evidence that people with prior diagnoses for allergic asthma are less likely to exhibit reduced risk of severe SARS-CoV-2 infections and hospitalizations compared with the population as a whole [120,121,122,123,124]. There are various possible factors that might underlie such an observation, but it is worth noting that the most widely considered non-pathocentric hypothesis—a protective role of regular corticosteroid use by asthmatics—does not fare particularly well in view of a conflicted and generally unfavorable relationship between general corticosteroid usage and COVID-19 outcomes [125,126,127,128]. The answer, therefore, may hinge on fundamental facets in molecular physiology and in proteolytic divergence in the pathology of the two disorders.
Protease/antiprotease imbalance is a hot topic in respiratory pathology research. While COPD and bronchiectasis are more extensively studied as examples of respiratory proteolytic disorders [106,107,129,130], allergic asthma pathology is generally also considered in this category [131,132]. To relate PAB knowledge to COVID-19 pathology, we may begin by considering antiproteases, whose effects on respiratory disorders have been widely studied [104]. The most widely reported antiprotease marker candidates for allergic asthma are SERPINB10, SERPINE1, and SERPINA3. SERPINB10 appears to offer a straightforward, correlative asthmatic relationship, presenting substantial overexpression in epithelial eosinophilia for various non-infectious respiratory pathologies [133,134,135]. In terms of anti-correlators, people with SERPINA3 (also known as alpha-1 antichymotrysin/A1AC) deficiencies tend to suffer disproportionately from non-bronchial allergic asthma [136,137]. Little is known about the manner in which SERPINA3/A1AC mitigates asthmatic response, other than a general assumption that the protein has immunoregulatory and anti-inflammatory effects, perhaps through regulation of neutrophil serine proteases [138]. The respiratory epithelial presence of specific isoforms of SERPINE1 (also called plasminogen activator inhibitor-1/PAI-1) has been examined as a possible asthma correlator [139], but the full body of literature yields a complex picture, with some research suggesting that that SERPINE1/PAI-1 dysregulation is of primary interest in cases reporting very specific polymorphisms of the protein (in particular the 4G/5G form) rather than as a general factor [140,141], while other studies consign its relevance to simple prediction of adverse corticosteroid side-effects [142,143].
In terms of relevance to COVID-19 vulnerability, SERPINB10, SERPINE1, and SERPINA3 paint very different pictures. There is minimal evidence that SERPINB10 plays any differential role, whether exacerbatory or mitigative, in determining COVID-19 outcomes [144]. Patient abundance of SERPINA3/A1AC, by contrast, is proposed to be very important for COVID-19 outcomes, but the relationship is directly analogous to trends for asthmatics, such that A1AC deficiency could be considered a major risk factor for both allergic asthma and severe viral infections [145,146,147]. Finally, as a converse example, SERPINE1/PAI-1 overabundance seems to correlate strongly with both severe non-infectious respiratory pathology [141,148,149,150] and a propensity to foster coagulopathic outcomes in severe COVID-19 cases, including interrelated acute respiratory distress syndrome (ARDS) and thrombotic manifestations [151,152,153], through apparent impairment of fibrinolytic activity [151]. From this complexity, it may be reasonable to conclude that some SERPINs have significant effects on both infectious and autoimmune respiratory pathologies, but that the effects are independent of any net relationship between cases of allergic asthma and trends of diminished COVID-19 severity.
Turning the attention to prospective protease markers, one finds multiple recent studies reporting correlations between elevated levels of proteases and allergic asthmatic response [107,154]. Of particular interest is Human Airway Trypsin-like Protease (HAT; also called TMPRSS11D) having unusual abundance in samples associated with asthmatic pathology [155,156]. From the perspective of a possible role in viral infections, it is notable that although HAT/TMPRSS11D had been implicated as a possible facilitator for the original SARS-CoV pathogen [155,157], its capacity to serve in a manner analogous to TMPRSS2 in the cleavage of the SARS-CoV-2 Spike protein and subsequent host-cell induction is measurably diminished by a factor of roughly 19-fold [158]. Thus, unlike antiprotease markers shared between allergic asthma and COVID-19 analytics, HAT/TMPRSS11D represents a unique example whereby relative abundance of the protein produces an opposite effect on pathological trends in allergic asthma versus COVID-19. This fact offers a tantalizing prospect that HAT/TMPRSS11D, while behaving as an approximate analog of TMPRSS2 and other proteases in exacerbating autoimmune conditions, may exhibit functional inefficiency that kinetically interferes with the process of SARS-CoV-2 infection and replication.

5. Microbiotic Mimicry and Amplification of Protease/Antiprotease Dynamics

While the host PAB machinery alone provides factors that may influence trends in COVID-19 vulnerability and resistance, it is important to recall that human genetics are only one determinant of our pathophysiologically relevant proteome. A crucial component of human biochemical complexity and diversity emerges from microbial complementarity, with distinct microbiomes in upper respiratory [159,160,161,162,163], lower respiratory [115,164,165,166,167], and gastrointestinal tracts [166,167,168,169,170] all having tangibly different effects on viral outcomes. Among the various biochemical effects associated with microbial proteases are known modulations of the innate immune system including IFNγ, several interleukins, and others [171], plus proteolytic stabilization of viral capsids in a manner that may bolster virion persistence outside of hosts in moist environments [172].
A detailed survey of viral modulation by microbial proteases and antiproteases, although potentially impactful, would be a huge undertaking, far in excess of this modest hypothesis article. It is, nonetheless, interesting to identify bacterial homologs to interesting proteases, such as HAT, A1AC, and PAI-1, within the proteomes of bacterial species classes that appear to exert influence on SAR-CoV-2 infectivity. These bacterial classes include both potential exacerbators of COVID-19 (e.g., Staphylococcus [173] and Pseudomonas [174,175]) and proposed mitigators (e.g., Dolosigranulum [173], Corynebacterium [173], and Moraxella [52,176]). Relevant proteases and antiproteases from these species families are reported in Table 1, according to homology (% identity) and alignment coverage (% coverage) relative to HAT, A1Ac, and PAI-1.
In this table, a trivial observation is that Dolosigranulum bacteria do not seem to produce close homologs to the COVID-19-relevant SERPINs or to the TMPRSS family of serine proteases. This would suggest that if Dolosigranulum bacteria do help to mitigate the threats posed by SARS-CoV-2, the mechanism is not analogous to prominent host proteolytic biochemistry and may actually not involve PAB at all. Beyond this simple observation, it may be helpful to establish context for the reported numbers. In general, larger values for sequence alignment coverage are indicative of conservation of multiple functional domains, which increases the likelihood that one protein shares not only similar primary function but possibly also comparable secondary (or even tertiary) functions and may also exhibit preferences for similar subcellular milieu. This would thus imply, for example, substantial analogies between Staphylococcus and Pseudomonas homologs to human PAI-1 and an even greater analogy for the Moraxella homolog to SERPINa3. The fraction of aligned sequences in the various other pairings suggests effective conservation of at least one functional domain in all cases, except for the case of Pseudomonas, which likely has no functional homolog to human SERPINa3 (i.e., the 16% sequence coverage and no overlap with the characteristic SERPIN center inhibitory loop imply poor candidacy).
By comparison, sequence identity is a stronger guarantee of the conservation of local structural and functional attributes. While high sequence identities offer proportionally higher probability of structural and functional conservation, there is an empirically very useful lower bound of 30% identity, above which a pair of proteins may be considered homologous. This boundary, while semi-arbitrary, has withstood decades of practical application, with the assessment that roughly 90% of all protein pairs with at least 30% sequence identity across known functional domains exhibit similar three dimensional structures and comparable functions for those domains [177]. This assumption amplifies the prospective significance of the Corynebacterium homolog relative to human SERPINe1; the Pseudomonas homolog versus human SERPINa3; and the Staphylococcus, Pseudomonas, and Moraxella homologs to human HAT. It should also be mentioned that functional analogies frequently exist between protein pairs with sequence identities slightly less than 30% [178], thus implying that tangible homology is possible among all putative pairings in Table 1, other than SERPINa3/PNB35482.1 (Pseudomonas).
From weighing levels of both sequence coverage and identity, it is the opinion of the authors that the most interesting proteins listed in Table 1 are the Staphylococcus and Pseudomonas homologs to SERPINe1, the Staphylococcus and Moraxella homologs to SERPINa3, and the Pseudomonas and Moraxella homologs to HAT. All of these bacterial homologs to human SERPINs are known to have protease inhibitory function, while both of the bacterial HAT homologs are known proteases. Admittedly, the marginal sequence homologies cannot be used to definitively argue that the antiprotease specificities of bacterial SERPIN homologs closely mirror those of the asthma-relevant and COVID-19-relevant SERPINe1 and SERPINa3, or whether the bacterial proteases might hasten or hinder the SARS-CoV-2 spike mechanism of host-cell entry. Nonetheless, the levels of sequence identity and structural conservation of functionally crucial domains (i.e., the catalytic domain on proteases and the inhibitory domain on SERPINs) imply that these bacterial homologs should produce a functional shift in PAB, quite possibly in a manner that influences the host response to invading viruses.
Given the exceptional proteomic diversity present within the pan-species protease/antiprotease spectrum, the best actionable evidence might emerge from a confluence of corroborating evidence, such as exacerbatory or inhibitory roles of bacterial species that also encode proteases or antiproteases similar to COVID-19-related host proteins. As evidence of this, it should be noted that experimental mutagenetic manipulation of human SERPINb3 transformed an antiprotease with minimal capacity for TMPRSS2 inhibition into a TMPRSS2 blocker more potent than SERPINa3 [179]. Since evolutionary pressures can foster rapid bacterial mutations, it is possible that some forms of bacteria may have adapted their proteolytic capacity for the purpose of modifying human TMPRSS2 activity, as suggested in studies relating to the synergistic interplay among TMPRSS2, androgen control, and microbiome composition [180,181,182,183].

6. Vulnerability Trends in Comparative Biology and Human Population

Throughout the COVID-19 pandemic, valuable research was conducted on viral transmission among animal vectors, including common companion animals such as dogs and cats. Notably, although both cats and dogs proved susceptible to contracting the SARS-COV-2 pathogen and produced antibodies, feline infections often progressed to contagious levels of viral shedding, whereas dogs, although vulnerable to acquiring a viral load, rarely displayed tangible shedding and spread [54]. There are comparably stark variations in SARS-COV-2 transmission within the broader canid families, such that coyotes are found to resemble dogs (i.e., largely impervious to SARS-CoV-2 shedding and COVID-19 symptoms), while foxes exhibit SARS-CoV-2 susceptibility comparable to that of house cats [55]. The boundaries of COVID-19 vulnerabilities among these animals thus do not clearly respect either genetics (i.e., canids versus felids) or domestication status (companion animals versus closely related wild species). A more plausible delineation may emerge from dietary considerations—dogs and coyotes are omnivorous, whereas house cats and foxes are classified as carnivores (cats are strict meat eaters; foxes are also primarily carnivorous, although food insecurity may prompt their consumption of berries and other carbohydrates). This putative factor aligns with corroborative evidence of relationships between dietary diversity and human COVID-19 vulnerability, whereby plant-predominant diets with higher consumption of vegetables, whole grains, nuts, and pulses confer statistically significant protection against moderate or severe SARS-CoV-2 infections relative to a standard western diet with more red meat, simple carbohydrates, and processed foods [184,185,186,187].
Unsurprisingly, other COVID-19 trends appear to be independent of obvious dietary influences. In particular, infected children tend to be statistically less likely to produce communicable levels of virus compared with adults, as per a 2021 study that found that children fostered contagion at a rate of about 63% that of adults [188]. Interestingly, adolescents (ages 14–17) were found, statistically, to have the lowest rates of shedding [189].
While immunological arguments may be devised to independently frame each such vulnerability trend as a distinct theme, there is evidence that the vulnerabilities may, at least in part, share common origins. Namely, it is our hypothesis that vulnerability to SARS-CoV-2 is influenced by PAB shifts due to variations in the nasopharyngeal mucosal microbiome. To further specify this hypothesis, our analysis points to evidence that some species within the genus Moraxellaceae may express specific proteases that interfere with the infectious capacity of SARS-CoV-2, to the point where gross symptoms and shedding may be significantly affected. In terms of the age-related fluctuations in COVID-19 vulnerability, it is noted that the genetic diversity of human respiratory microbiota increases throughout childhood, reaching a peak in late adolescence or early adulthood, before declining through the adult years [190]. It may be further noted that children tend to have a substantial fraction of Moraxella catarrhalis in their respiratory microbiomes, while adults have greater Streptococcal fractions [51,191]. Separate from the simple age-dependent microbiome argument, there is a pronounced relationship between a healthy, diversified human diet and the immunological benefits of commensal bacteria [166,192,193,194]. Namely, strong evidence suggests that greater dietary diversity tends to correspond to greater diversity in host commensal bacterial populations [195,196,197,198,199].
Regardless of the circumstances under which microbiotic variations emerge, numerous links may be drawn between host commensal bacterial profiles and resistance to severe or symptomatic COVID-19 infections, including a simple but statistically significant correlation between COVID-19 vulnerability and pre-existing autoimmune conditions associated with microbiotic deficiencies [200]. The microbiotic argument is substantially strengthened by clinical observations of elevated abundance of nasal Moraxella catarrhalis in children who have resisted COVID-19 infection relative to those with symptomatic cases [52,201]. These trends are further augmented by comparative pathology arguments, such as recognition that the relative resistance of domesticated dogs to symptomatic or shedding manifestations of COVID-19 may correspond to the widely elevated prevalence of Moraxella species in the nasal microbiome of individual members of the Canis lupus familiaris species. Given the importance of nasal mucosa in both contracting and potentially treating COVID-19 (in particular as a target for active vaccine RNA antigens [202]), we quantitatively profiled the nasal microbiome of a cohort of forty client-owned domesticated dogs (see Table 2), across which Moraxella sp. was the only bacterial class to be universally detected. Furthermore, Moraxella fractions dominated the nasal microbiome in most individual dogs, leading to an overall mean abundance roughly four times greater than the second most prevalent class (Pseudomonas aeruginosa).
Despite the overall disparity between Moraxella and Pseudomonas abundances within dog nasal microbiomes, it is interesting to compare the host viral effects arising from the two bacterial genera. In particular, various Pseudomonas species seem to influence the capacity of respiratory viruses to infect airway epithelial cells, but the net effect may be opposite to that by Moraxella. Specifically, P. aeruginosa was observed to suppress normal airway epithelial antiviral response in a manner that exacerbates the rate and spread of viral infections [203]. Furthermore, of particular relevance to COVID-19, one notes that P. aeruginosa may facilitate SARS-CoV-2 entry into airway epithelial cells by amplifying local abundance of the COVID-19-promoting host protease TMPRSS2 [175,204,205,206].
In addition to this possible effect relating to TMPRSS2 function, it is worth mentioning that other studies have proposed bacterial down-regulation of ACE2 receptors as a means for diminishing the extent of viral host-cell compromise. ACE2 expression is modulated by exposure to various antigens [207,208], as well as part of the interferon response to viral [209,210] and oncological [211,212] pathogens; thus, a microbiotic capacity for modifying or obfuscating the pathogenic up-regulatory influence may affect the host’s vulnerability to viral invasion. Indeed, host levels of P. aeruginosa do appear to influence ACE2 abundance, with measurable impact on ACE2-related pathological mechanisms [205,213,214].
These proposed mechanisms for an apparent capacity of Pseudomonas bacteria to exacerbate SARS-CoV-2 infection, in concert with evidence that composition of the nasal microbiome may play a role in determining COVID-19 vulnerability in certain subsets of the human (and broader mammalian) populations, prompt the question of whether Pseudomonas bacteria directly or indirectly influence viral infection of airway epithelial mucosa. Such insight may provide contra-indicative insight into how commensal microbiota might protect the host. Moraxella, whose presence in epithelial mucosa tends to correspond with significantly diminished COVID-19 vulnerability [53,215,216], seems like a natural point of comparison, especially given the close genetic similarity between the Pseudomonas and Moraxella genera.

7. Prospective Biochemical Rationalizations

Among the two prospective mechanisms for comparing Pseudomonas and Moraxella influence, the ACE2 model may explain why a greater Pseudomonas fraction may promote SARS-CoV-2 infections [205,213,214] but does not provide a clear rationale for why Moraxella might have the opposite effect, as current evidence is lacking for comparable Moraxella-triggered ACE2 flux. TMPRSS2, on the other hand, does provide a basis to rationalize a Moraxella effect. Specifically, there is substantial evidence that Moraxella counteracts the capacity of TMPRSS2 proteins to achieve the viral priming required for cell invasion [53,216]. Unlike the Pseudomonas/ACE2 case, however, it seems unlikely that the capacity of Moraxella bacteria to modulate TMPRSS2 action arises from direct influence on expression. Rather, the most readily measurable TMPRSS2-mediated influence of commensal bacteria seems to entail suppression of the enzyme’s activity, not abundance, as evinced by changes in TMPRSS2-triggered pathways, including the immunological interferon antiviral response [175,217], and downstream androgen-mediated angiogenic effects [181,182,218,219]. This implies that any prospective TMPRSS2-related benefit arising from an elevated Moraxella fraction may be attributed to effects that diminish the rate of viral Spike protein cleavage relative to basal TMPRSS2 action. This further suggests a competitive dynamic, perhaps achieved through interactions between Spike and some Moraxella protein that mimics and interferes with a key aspect of TMPRSS2-mediated Spike cleavage or membrane induction inherent in SARS-CoV-2 infection.
The BLAST-P analysis (Table 1) indicated the Moraxella Serine Protease (MSP) protein (GenBank ID MBC7754020.1; 37.82% identical to TMPRSS2, covering 56% of the sequence alignment) as a possible candidate for TMPRSS2 interference. In comparison, the analogous enzyme from Pseudomonas sp. (GenBank ID WP_082643865.1; PSP for short) is slightly more phylogenetically distant from TMPRSS2 (36.21% sequence conservation; 54% coverage), but we see, in Figure 2, that PSP may exhibit a tertiary structure that is more analogous to the human protein. Specifically, MSP and PSP both show substantial sequence conservation relative to TMPRSS2 within the active catalytic triad (marked by the rose-colored “C” boxes in Figure 2), but MSP has a pair of proximal sequence deletions that are not observed for PSP.
The largest deletion (TMPRSS2 residues 353-364; marked with a green “P” in Figure 2) lies in a peripheral loop where MSP sequence diverges from TMPRSS2 and PSP. The authors have not found clear literature evidence assigning a specific function to this loop, but it is notable that one of the MSP deletion positions in the sequence alignment corresponds to a TMPRSS2 residue (V361) within a tetrad (N358-D359-L360-V361) that has been resolved in the 7MEQ crystal structure of TMPRSS2 [220] as a distinct, H-bond-stabilized β-turn, independent of any larger β-sheet tertiary structure. Definitive evidence does not exist for whether this β-turn (present in TMPRSS2 and PSP but absent in MSP) plays a key role in determining functions and behaviors of these proteins, but it should be noted that many surface β-turns serve as recognition motifs for protein–protein interactions [221], with the most prevalent class of β-turn-binding proteins being GPCRs [221,222]. Again, acknowledging that the absence of experimental confirmation of specific binding renders any etiological assertion largely speculative, it is worth noting prior reports of TMPRSS2 activating a variety of GCPRs [223,224]. Furthermore, various COVID-19 symptoms (e.g., anosmia, congestion, neurological effects, etc.) are believed to involve GPCR dysregulation [225,226,227]. The speculative conclusion one might draw from the β-turn deletion in MSP, therefore, is that TMPRSS2 and PSP might support a pathologically important GCPR interaction on the host-cell surface, while the Moraxella homolog, MSP, lacks this attribute.
A smaller but more intuitively impactful deletion, marked “del” in Figure 2, is in close proximity to the proteolytic active site, corresponding to TMPRSS2 residues W306 and H307. This deletion dictates a sterically smaller MSP active site (see protruding feature marked with “B1” in Figure 3B). In combination with a steric pucker (MSP residues E229 and K236 form a protruding salt bridge marked with “B2” in Figure 3B), this may alter the MSP enzymatic selectivity relative to PSP and TMPRSS2, both of which likely have broader catalytic channels.
To simulate how altered catalytic site topology might impact possible cleavage of the SARS-CoV-2 Spike protein, we performed rigorous Gaussian accelerated molecular dynamics (GAMD) calculations (see Supplementary Information) to predict the relative binding free energies and enzymatic viability of TMPRSS2, MSP, and PSP toward a 27 amino acid peptide (P809-K835) spanning the S2′ cleavage site (R815-S816). From a representative set of different initial ligand approach modes, the simulations tended to preferentially converge to one of two possible bound conformations, where the first is depicted in Figure 3A,C (mode 1: quadrant A shows conformation; quadrant C shows relevant interatomic distances), while the second is shown in Figure 3B,D (mode 2: conformation and interatomic distances). These two binding modes are accommodated by a conformational shift in the catalytic serine, as can be noted by comparing quadrants Figure 3C vs. Figure 3D, as well as the Figure S1 schematic in Supplementary Information.
To assess these prospects, binding-free-energy predictions were performed (see Table 3) to evaluate hypothetical trends associated with prospective proteolytic cleavage of Spike. The collective evidence from Figure 2 and Table 3 suggests a key distinction between the binding of Spike S2′ to TMPRSS2 versus MSP. TMPRSS2 is predicted to favor mode 1, which places the target R815 carbonyl carbon in plausibly reactive proximity to catalytic serine S401 (the separation is 4.6 Å in the instantaneous structure in Figure 3A,C but averages 4.94 ± 0.04 Å over the duration of the simulation), whereas in the context of the MSP cavity, an analogous Spike conformation encounters steric hindrance, preventing a close R815 approach to the catalytic S206 serine. Conversely, mode 2 supports a theoretically viable catalytic approach for S2′ binding to MSP, but the close approach is for K184 (4.9 Å instantaneous approach in Figure 3B,D, time-averaged to 5.11 ± 0.06 Å). PSP, meanwhile, presents yet a third scenario, whereby Table 3 reflects a preference for Spike complexation via mode 1 but places K814 (not R815) in possibly reactive proximity (time-averaged separation of 4.93 ± 0.04 Å).
The binding-free-energy predictions in Table 3 add further insight into hypothetical trends associated with prospective proteolytic cleavage of Spike. Considering that TMPRSS2, MSP, and PSP all favor trypsin-like cleavage between a cationic amino acid and an adjacent polar residue, it is not surprising that those binding modes predicted to be most energetically favorable (mode 1 for TMPRSS2 and PSP; mode 2 for MSP) also correspond to those that place cationic targets in the closest proximity to the reactive serine. The magnitudes of their predicted binding energies are also instructive. Discounting the unfavorable complexes (MSP conformation 1 and PSP conformation 2), we are left with at least one conformation for each protease whose associative free energy significantly exceeds the predicted activation barrier for TMPRSS2 proteolytic cleavage, which ranges from 15.8 to 17.1 kcal/mol for a variety of cleavable peptides [228].
From these energy predictions, it is reasonable to infer that the exothermic energy gained from Spike binding to TMPRSS2, MSP, and PSP may, in all three cases, provide ample energetic impetus for the enzyme to overcome the reactive barrier and cleave the peptide. While the precise reaction kinetics have not been reported for proteolytic cleavage by MSP and PSP, it is worth noting that widespread conservation of protease catalytic triad structures imposes homogeneity on reactive proficiency across the broad manifold of serine proteases [229,230,231]. Known proteolysis barriers for HIV protease (15.1–17.9 kcal/mol) [232,233], for example, bear a strong resemblance to TMPRSS2 reactive barriers. Such functional similarity across otherwise rather different enzymes implies a reasonable expectation that MSP and PSP could also support cleavage reactions within this approximate range.
There is, however, one additional enzymological factor that may help to rationalize distinctions in how TMPRSS2, MSP, and PSP may comport toward a cleavable viral Spike protein—the capacity of each enzyme to release its products. For this, very stark differences can be observed in 5 ns GAMD simulations (see Supplementary Information for computational details) of the egress of Spike N-terminal (Figure 4A) and C-terminal (Figure 4B) fragments from our three enzymes of interest. TMPRSS2 achieves rapid release of both fragments, whereas both of the bacterial enzymes tend to persistently sustain complexes with post-cleavage Spike fragments. Interestingly, differences manifest between PSP and MSP in adhering to the N-terminal fragment that connects to the Spike ACE2-binding domain (i.e., PSP is predicted to sustain strong binding without release, while MSP is predicted to potentially loosen the complex but then permit the rebinding of the fragment in a slightly altered conformation), but both PSP and MSP hold onto the C-terminal fragment with comparable consistency. Since free cleavage of the C-terminal portion of the SARS-CoV-2 spike protein is crucial for host infection, given its role as a molecular switch in the process of syncytia formation [234,235], our preliminary analysis suggests varied TMPRSS2, MSP, and PSP proteolytic dynamics that could translate to significantly different influences on the molecular kinetics underlying viral infection. Although neither MSP nor PSP is likely to achieve the enzymatic processing of SARS-CoV-2 with the efficiency of the TMPRSS2 interaction, this inefficiency may actually be the source of a tangible host-protective effect, such that sustained complexation with the pathologically critical Spike C-terminal cleavage product may impair the capacity of SARS-CoV-2 virions to inflict infection and compromise host lymphocyte response [235].
Ultimately, the preliminary GAMD simulations were pursued for the purposes of rationalizing a prospective role of Moraxella and/or Pseudomonas in altering the PAB dynamics required for SARS-CoV-2 proteolytic virion priming. Our results suggest that the MSP (from Moraxella) and PSP (Pseudomonas) proteases may persistently bind the viral Spike protein and perhaps achieve a form of proteolytic cleavage, but with a product clearance rate substantially lower than that for human TMPRSS2. This scenario may, via inefficiency, imply a tangible host-protective effect. Based on this prediction, combined with empirical evidence arising from a diverse set of previously published observations and trends, this hypothesis hypothesizes that given sufficient expression of Moraxella serine protease within the host mucosal epithelia, such catalytically inefficient MSP–spike complexes may compete with the normal pathological virion invasion of host cells to an extent that impairs the capacity of the virus to infect the host at a rate that compromises lymphocyte response [235], thus rationalizing the apparent antiviral effects of Moraxella commensal bacteria.

8. Future Directions

Even when accounting for distinct pathological trends associated with different viral variants, the outcomes of COVID-19 cases vary in ways that are difficult to explain comprehensively using host attributes alone and should be further examined. Among humans, statistical divergence manifests with varying social geography, sex, and age. Racial heritage seems to play a role, but fine-grained analysis suggests that sex, age, and socioeconomic status are stronger predictors within a given racial community [236], though no single host metric confers strong predictivity. From the perspective of comparative biology, the relative vulnerabilities of companion animals, in particular cats versus dogs, suggest that dietary diversity may be an important factor—an argument that strengthens when one explores the relative vulnerabilities of other canids, such as foxes and coyotes, whereby dogs and coyotes (both omnivorous) display reduced vulnerability to symptomatic SARS-CoV-2 infection, whereas house cats and foxes (primarily carnivores) have markedly higher susceptibility, despite foxes being genetically much closer to dogs and coyotes.
Numerous superficially plausible hypotheses may be formulated to seek unification across the many different vulnerability markers, but diet and social geography (in particular as it relates to diet and exposure to pollution and pathogens) have documented effects on host microbiome profiles, which, in turn, have attracted growing attention as possible influences on COVID-19 vulnerabilities [50,52,160,161,162,165,170,173,183,190,191,192,193,200,217]. Reasonable interpretations for how such microbe–virus interplay may arise must recognize not only the observations that bacteria use proteases to modulate host physiology in ways that benefit bacterial persistence and replication [171,237,238,239] and inter-bacterial competition [239,240,241] but also the evidence that bacterial proteolytic action can tangibly modulate viral efficacy [172].
The diversity of host, viral, and bacterial proteases, and their associated substrate specificities could support numerous theoretical models to account for bacterial influences on COVID-19 vulnerability. To prioritize models, there is value in finding independent observations that point toward an internally consistent paradigm. For example, a combination of prior published COVID-19 vulnerability studies (on humans and other mammals, plus our own studies on dogs) suggest that the presence of Moraxella in host nasal microbiomes may coincide with reduced rates of severe COVID-19 infection, in concert with documented presence of a TMPRSS2-like protease in the Moraxella proteome. This confluence of factors motivates the primary hypothesis of this article, which is that commensal Moraxella may afford a degree of host protection against SARS-CoV-2 by supplying a competitive binder for the S2′ motif on the SARS-CoV-2 Spike protein. Molecular simulations predict that while TMPRSS2 should have far greater capacity to process S2′ as a cleavage substrate, Moraxella serine protease (MSP) may be capable of exothermically complexing with S2′ and MSP may persistently bind the resulting cleaved fragments in a manner that could delay or abrogate subsequent viral infection steps, such as syncytia formation.
As a caveat to these observations, it should be noted that a comparably homologous protein, the serine protease from Pseudomonas bacteria, may exhibit similarly competitive prospects for circumventing TMPRSS2 action on SARS-CoV-2 Spike proteins, even though there is little evidence for Pseudomonas strains exerting a net protective effect against COVID-19. It is possible that unlike Moraxella, Pseudomonas may exert multiple opposing influences, including the fact that Pseudomonas presence in human nasal microbiomes is far more widely considered to have pathological effects [242,243,244], and such microbial infections may perturb host immunity in a manner that incites co-infections with SARS-CoV-2 [174,245,246]. Nonetheless, the relative prospects for viral modulation by both Moraxella and Pseudomonas strains should merit careful consideration in further exploration of this hypothesis, leading to directly testable assertions, including our suggestion that co-incubation of Vero E6 cells with (a) Moraxella and with (b) Pseudomonas should lead to diminished SARS-CoV-2 infectivity relative to sterile controls, while more immuno-realistic models will show divergence, such that Moraxella proves to be a more effective mitigator.

9. Synopsis

It is finally worth reiterating a key motivation for pursuing the evaluation of such hypotheses. Despite remarkable advances in medical and pharmaceutical technologies, viruses continue to largely evade our therapeutic arsenals. Bacteria and viruses, however, have interacted with each other for hundreds of millions of years, often attaining a degree of detente that may benefit the health of host organisms. Theoretically, such viral–bacterial dynamics may support development of an in vitro environment within which therapeutically practicable commensal microbes might be trained as adjuvant allies in our battles with known or emerging viral threats. More broadly, it is reasonable to believe that identification and characterization of microbial/viral balances might help to explain how some individuals attain tangible protection against dangerous viral pathogens, thus providing a basis for acquiring molecular mechanistic insight that could inform new strategies to mitigate future pandemics. While much of these prospects remain distinctly hypothetical, however, humanity should expect to confront other deadly viruses. In order to persevere and thrive, we must continue to mine data and anomalies in order to intuit, analyze, and potentially exploit new tools for the challenges ahead.

Supplementary Materials

The following supporting information can be downloaded at: https://0-www-mdpi-com.brum.beds.ac.uk/article/10.3390/biotech12040061/s1, Figure S1: FigS1.tif (Orientations of S2’ Coil Bound to TMPRSS-like Proteases), Protocol: BioTech_supplement_TM.pdf. References [220,247,248,249,250,251,252,253,254,255,256,257,258] have been cited in Supplementary Materials.

Author Contributions

Conceptualization, G.H.L., A.L. and T.M.; methodology, G.H.L., A.L. and T.M.; software, G.H.L.; validation, G.H.L., A.L. and T.M.; formal analysis, G.H.L., A.L. and T.M.; investigation, G.H.L., A.L. and T.M.; resources, G.H.L., A.L. and T.M.; data curation, G.H.L., A.L. and T.M.; writing—original draft preparation, G.H.L.; writing—review and editing, G.H.L., A.L. and T.M.; funding acquisition, T.M. All authors have read and agreed to the published version of the manuscript.

Funding

The study was made possible by funding from the Western U Office for Research and Biotechnology, the College of Veterinary Medicine Office for Research (2021; intramural fund), and the True One Medicine Initiative (2021; award number WU 28119V).

Data Availability Statement

Data are available from the authors upon request.

Acknowledgments

The authors would like to thank the clinicians involved in the nasal swab collection from client-owned dogs, specifically Wayne Tsang, Nathan Sharp, Young J. Kim, and David Forster, as well as the technical support staff at WesternU Pet Health Center and East Valley Center. We thank the participating clients and their dogs without whom the study would not have been possible (IACUC ref. No. R21IACUC022). We also acknowledge Tracey McNamara for her advocacy in favor of surveying client-owned dogs in the local community for SARS-CoV-2, which inspired the computational modeling and literature review. The authors are grateful for the support of Bradley Henson, John Tegzes, Dominique Griffon and David Clark.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Riley, S.; Ainslie, K.E.C.; Eales, O.; Walters, C.E.; Wang, H.; Atchison, C.; Fronterre, C.; Diggle, P.J.; Ashby, D.; Donnelly, C.A.; et al. Resurgence of SARS-CoV-2: Detection by community viral surveillance. Science 2021, 372, 990–995. [Google Scholar] [CrossRef]
  2. Fernandes, R.S.; de Oliveira Silva, J.; Gomes, K.B.; Azevedo, R.B.; Townsend, D.M.; de Paula Sabino, A.; Branco de Barros, A.L. Recent advances in point of care testing for COVID-19 detection. Biomed. Pharmacother. 2022, 153, 113538. [Google Scholar] [CrossRef]
  3. Kwok, H.F. The significance of advanced COVID-19 diagnostic testing in pandemic control measures. Int. J. Biol. Sci. 2022, 18, 4610–4617. [Google Scholar] [CrossRef] [PubMed]
  4. Alhama, J.; Maestre, J.P.; Martín, M.Á.; Michán, C. Monitoring COVID-19 through SARS-CoV-2 quantification in wastewater: Progress, challenges and prospects. Microb. Biotechnol. 2022, 15, 1719–1728. [Google Scholar] [CrossRef] [PubMed]
  5. Pepera, G.; Tribali, M.-S.; Batalik, L.; Petrov, I.; Papathanasiou, J. Epidemiology, risk factors and prognosis of cardiovascular disease in the Coronavirus Disease 2019 (COVID-19) pandemic era: A systematic review. Rev. Cardiovasc. Med. 2022, 23, 28. [Google Scholar] [CrossRef] [PubMed]
  6. Bahmer, T.; Borzikowsky, C.; Lieb, W.; Horn, A.; Krist, L.; Fricke, J.; Scheibenbogen, C.; Rabe, K.F.; Maetzler, W.; Maetzler, C.; et al. Severity, predictors and clinical correlates of Post-COVID syndrome (PCS) in Germany: A prospective, multi-centre, population-based cohort study. eClinicalMedicine 2022, 51, 101549. [Google Scholar] [CrossRef] [PubMed]
  7. Abdelwahab, N.; Ingraham, N.E.; Nguyen, N.; Siegel, L.; Silverman, G.; Sahoo, H.S.; Pakhomov, S.; Morse, L.R.; Billings, J.; Usher, M.G.; et al. Predictors of Postacute Sequelae of COVID-19 Development and Rehabilitation: A Retrospective Study. Arch. Phys. Med. Rehabil. 2022, 103, 2001–2008. [Google Scholar] [CrossRef] [PubMed]
  8. Lushington, G. Preface to CCHTS Volume 24: The imperative of host-directed pharmacology in the aftermath of the COVID-19 pandemic. Comb. Chem. High. Throughput Screen. 2021, 24, 1–2. [Google Scholar] [CrossRef]
  9. Su, Y.; Yuan, D.; Chen, D.G.; Ng, R.H.; Wang, K.; Choi, J.; Li, S.; Hong, S.; Zhang, R.; Xie, J.; et al. Multiple early factors anticipate post-acute COVID-19 sequelae. Cell 2022, 185, 881–895.e20. [Google Scholar] [CrossRef]
  10. Di Ciaula, A.; Bonfrate, L.; Portincasa, P.; Appice, C.; Belfiore, A.; Binetti, M.; Cafagna, G.; Campanale, G.; Carrieri, A.; Cascella, G.; et al. Nitrogen dioxide pollution increases vulnerability to COVID-19 through altered immune function. Environ. Sci. Pollut. Res. 2022, 29, 44404–44412. [Google Scholar] [CrossRef]
  11. Ravindra, K.; Singh, T.; Vardhan, S.; Shrivastava, A.; Singh, S.; Kumar, P.; Mor, S. COVID-19 pandemic: What can we learn for better air quality and human health? J. Infect Public Health 2022, 15, 187–198. [Google Scholar] [CrossRef] [PubMed]
  12. Al Rifai, M.; Jain, V.; Khan, S.U.; Bk, A.; Mahar, J.H.; Krittanawong, C.; Mishra, S.R.; Dani, S.S.; Petersen, L.A.; Virani, S.S. State-Level Social Vulnerability Index and Healthcare Access: The Behavioral Risk Factor Surveillance System Survey. Am. J. Prev. Med. 2022, 63, 403–409. [Google Scholar] [CrossRef] [PubMed]
  13. Bruce, C.; Gearing, M.E.; DeMatteis, J.; Levin, K.; Mulcahy, T.; Newsome, J.; Wivagg, J. Financial vulnerability and the impact of COVID-19 on American households. PLoS ONE 2022, 17, e0262301. [Google Scholar] [CrossRef] [PubMed]
  14. Kozlov, M. Merck’s COVID Pill Loses Its Lustre: What That Means for the Pandemic. Nature News, 2021. Available online: https://0-www-nature-com.brum.beds.ac.uk/articles/d41586-021-03667-0(accessed on 12 March 2023).
  15. Cohen, J.; Kupferschmidt, K. The ‘Very, Very Bad Look’ of Remdesivir, the First FDA-Approved COVID-19 Drug. Available online: https://www.science.org/content/article/very-very-bad-look-remdesivir-first-fda-approved-covid-19-drug (accessed on 28 October 2020).
  16. Kow, C.S.; Javed, A.; Ramachandram, D.; Hasan, S.S. Clinical outcomes of sofosbuvir-based antivirals in patients with COVID-19: A systematic review and meta-analysis of randomized trials. Expert. Rev. Anti Infect. Ther. 2022, 20, 567–575. [Google Scholar] [CrossRef] [PubMed]
  17. Al-Taie, A.; Denkdemir, F.R.; Sharief, Z.; Buyuk, A.S.; Sardas, S. The Long View on COVID-19 Theranostics and Oral Antivirals: Living with Endemic Disease and Lessons from Molnupiravir. OMICS J. Integr. Biol. 2022, 26, 324–328. [Google Scholar] [CrossRef] [PubMed]
  18. Vegivinti, C.T.R.; Evanson, K.W.; Lyons, H.; Akosman, I.; Barrett, A.; Hardy, N.; Kane, B.; Keesari, P.R.; Pulakurthi, Y.S.; Sheffels, E.; et al. Efficacy of antiviral therapies for COVID-19: A systematic review of randomized controlled trials. BMC Infect. Dis. 2022, 22, 107. [Google Scholar] [CrossRef] [PubMed]
  19. Self, W.H.; Semler, M.W.; Leither, L.M.; Casey, J.D.; Angus, D.C.; Brower, R.G.; Chang, S.Y.; Collins, S.P.; Eppensteiner, J.C.; Filbin, M.R.; et al. Effect of Hydroxychloroquine on Clinical Status at 14 Days in Hospitalized Patients With COVID-19: A Randomized Clinical Trial. JAMA 2020, 324, 2165–2176. [Google Scholar] [CrossRef] [PubMed]
  20. Lim, S.C.L.; Hor, C.P.; Tay, K.H.; Mat Jelani, A.; Tan, W.H.; Ker, H.B.; Chow, T.S.; Zaid, M.; Cheah, W.K.; Lim, H.H.; et al. Efficacy of Ivermectin Treatment on Disease Progression Among Adults with Mild to Moderate COVID-19 and Comorbidities: The I-TECH Randomized Clinical Trial. JAMA Intern. Med. 2022, 182, 426–435. [Google Scholar] [CrossRef]
  21. Relman, D.A.; Hamburg, M.A.; Choffnes, E.R.; Mack, A. Infectious Disease Emergence: Past, Present, and Future; National Academies Press: Cambridge, MA, USA, 2009. [Google Scholar]
  22. Anston, F.; Bayfield, O.; Bardy, P. Developing Antiviral Drugs Is Not Easy—Here’s Why. The Conversation. Available online: https://theconversation.com/developing-antiviral-drugs-is-not-easy-heres-why-159512 (accessed on 12 March 2023).
  23. Howes, L. Why Are Antivirals so Hard to Develop? Chem. Eng. News 2021, 99, 32–35. Available online: https://0-cen-acs-org.brum.beds.ac.uk/pharmaceuticals/drug-discovery/antiviral-drug-development-covid-19/99/i19 (accessed on 12 March 2023). [CrossRef]
  24. Center for Disease Control. Influenza Antiviral Drug Resistance. 2021. Available online: https://www.cdc.gov/flu/treatment/antiviralresistance.htm (accessed on 12 March 2023).
  25. Dance, A. The Challenges of Antiviral Treatments. 2021. Available online: https://knowablemagazine.org/article/health-disease/2021/challenges-antiviral-treatments (accessed on 12 March 2023).
  26. Irwin, K.K.; Renzette, N.; Kowalik, T.F.; Jensen, J.D. Antiviral drug resistance as an adaptive process. Virus Evol. 2016, 2, vew014. [Google Scholar] [CrossRef]
  27. Hiscox, J.A.; Khoo, S.H.; Stewart, J.P.; Owen, A. Shutting the gate before the horse has bolted: Is it time for a conversation about SARS-CoV-2 and antiviral drug resistance? J. Antimicrob. Chemother. 2021, 76, 2230–2233. [Google Scholar] [CrossRef]
  28. Kumar, M.; Mazumder, P.; Mohapatra, S.; Kumar Thakur, A.; Dhangar, K.; Taki, K.; Mukherjee, S.; Kumar Patel, A.; Bhattacharya, P.; Mohapatra, P.; et al. A chronicle of SARS-CoV-2: Seasonality, environmental fate, transport, inactivation, and antiviral drug resistance. J. Hazard. Mater. 2021, 405, 124043. [Google Scholar] [CrossRef] [PubMed]
  29. Vitiello, A. Sars-Cov-2 and risk of antiviral drug resistance. Ir. J. Med. Sci. 2022, 191, 2367–2368. [Google Scholar] [CrossRef] [PubMed]
  30. Mari, A.; Roloff, T.; Stange, M.; Søgaard, K.K.; Asllanaj, E.; Tauriello, G.; Alexander, L.T.; Schweitzer, M.; Leuzinger, K.; Gensch, A.; et al. Global Genomic Analysis of SARS-CoV-2 RNA Dependent RNA Polymerase Evolution and Antiviral Drug Resistance. Microorganisms 2021, 9, 1094. [Google Scholar] [CrossRef] [PubMed]
  31. Mazzon, M.; Marsh, M. Targeting viral entry as a strategy for broad-spectrum antivirals. F1000Research 2019, 8, F1000 Faculty Rev-1628. [Google Scholar] [CrossRef] [PubMed]
  32. Li, Y.; Liu, D.; Wang, Y.; Su, W.; Liu, G.; Dong, W. The Importance of Glycans of Viral and Host Proteins in Enveloped Virus Infection. Front. Immunol. 2021, 12, 638573. [Google Scholar] [CrossRef] [PubMed]
  33. Kalaichandran, A. A Lucky Few Seem “Resistant” to COVID-19. Scientists Want to Know Why. STAT. 2021. Available online: https://www.statnews.com/2021/08/23/lucky-few-seem-resistant-to-covid19-scientists-want-to-know-why-2/ (accessed on 12 March 2023).
  34. Broadbent, L. Haven’t Had COVID Yet? It Could Be More Than Just Luck. Available online: https://economictimes.indiatimes.com/news/how-to/havent-had-covid-yet-it-could-be-more-than-just-luck/articleshow/91665022.cms (accessed on 12 March 2023).
  35. Scientists Are Narrowing in on Why Some People Keep Avoiding COVID. BA.5 Could End that Luck. NBC News. Available online: https://theconversation.com/havent-had-covid-yet-it-could-be-more-than-just-luck-181708 (accessed on 12 March 2023).
  36. Browne, G. (Seot. 12, 2022) The Mystery of Why Some People Don’t Get COVID. Wired. Available online: https://www.wired.com/story/the-mystery-of-why-some-people-dont-get-covid/ (accessed on 12 March 2023).
  37. Muller, M. (March 30, 2022) Are Some People ‘Super-Immune’ to COVID? Bloomberg.com. Available online: https://www.bloomberg.com/news/newsletters/2022-03-30/are-some-people-super-immune-to-covid (accessed on 12 March 2023).
  38. Andreakos, E.; Abel, L.; Vinh, D.C.; Kaja, E.; Drolet, B.A.; Zhang, Q.; O’Farrelly, C.; Novelli, G.; Rodríguez-Gallego, C.; Haerynck, F.; et al. A global effort to dissect the human genetic basis of resistance to SARS-CoV-2 infection. Nat. Immunol. 2022, 23, 159–164. [Google Scholar] [CrossRef] [PubMed]
  39. Daniloski, Z.; Jordan, T.X.; Wessels, H.-H.; Hoagland, D.A.; Kasela, S.; Legut, M.; Maniatis, S.; Mimitou, E.P.; Lu, L.; Geller, E.; et al. Identification of Required Host Factors for SARS-CoV-2 Infection in Human Cells. Cell 2021, 184, 92–105.e16. [Google Scholar] [CrossRef]
  40. Tripathi, D.; Sodani, M.; Gupta, P.K.; Kulkarni, S. Host directed therapies: COVID-19 and beyond. Curr. Res. Pharmacol. Drug Discov. 2021, 2, 100058. [Google Scholar] [CrossRef]
  41. Swadling, L.; Diniz, M.O.; Schmidt, N.M.; Amin, O.E.; Chandran, A.; Shaw, E.; Pade, C.; Gibbons, J.M.; Le Bert, N.; Tan, A.T.; et al. Pre-existing polymerase-specific T cells expand in abortive seronegative SARS-CoV-2. Nature 2022, 601, 110–117. [Google Scholar] [CrossRef]
  42. Fulkerson, P.C.; Lussier, S.J.; Bendixsen, C.G.; Castina, S.M.; Gebretsadik, T.; Marlin, J.S.; Russell, P.B.; Seibold, M.A.; Everman, J.L.; Moore, C.M.; et al. Human Epidemiology and RespOnse to SARS-CoV-2 (HEROS): Objectives, Design and Enrollment Results of a 12-City Remote Observational Surveillance Study of Households with Children using Direct-to-Participant Methods. medRxiv, 2022; preprint. [Google Scholar] [CrossRef]
  43. Bravaccini, S.; Nicolini, F.; Balzi, W.; Azzali, I.; Calistri, A.; Parolin, C.; Vitiello, A.; Biasolo, M.A.; Mazzotti, L.; Gaimari, A.; et al. Tamoxifen Protects Breast Cancer Patients from COVID-19: First Evidence from Real World Data. 2021. preprint. Available online: https://www.researchsquare.com/article/rs-598923/v1 (accessed on 12 March 2023).
  44. Bravaccini, S.; Fonzi, E.; Tebaldi, M.; Angeli, D.; Martinelli, G.; Nicolini, F.; Parrella, P.; Mazza, M. Estrogen and Androgen Receptor Inhibitors: Unexpected Allies in the Fight Against COVID-19. Cell Transpl. 2021, 30, 0963689721991477. [Google Scholar] [CrossRef] [PubMed]
  45. The COVID-19 pandemic in 2023: Far from over. Lancet 2023, 401, 14–20.
  46. Lorenz, J. Study Confirms Benefits of COVID-19 Vaccines, Shows Effectiveness Wanes Over Time. Available online: https://www.contagionlive.com/view/study-confirms-benefits-of-covid-19-vaccines-shows-effectiveness-wanes-over-time (accessed on 12 March 2023).
  47. von Delft, A.; Hall, M.D.; Kwong, A.D.; Purcell, L.A.; Saikatendu, K.S.; Schmitz, U.; Tallarico, J.A.; Lee, A.A. Accelerating antiviral drug discovery: Lessons from COVID-19. Nat. Rev. Drug Discov. 2023, 22, 585–603. [Google Scholar] [CrossRef] [PubMed]
  48. Mirzaei, R.; Goodarzi, P.; Asadi, M.; Soltani, A.; Aljanabi, H.A.A.; Jeda, A.S.; Dashtbin, S.; Jalalifar, S.; Mohammadzadeh, R.; Teimoori, A.; et al. Bacterial co-infections with SARS-CoV-2. IUBMB Life 2020, 72, 2097–2111. [Google Scholar] [CrossRef] [PubMed]
  49. Hughes, S.; Troise, O.; Donaldson, H.; Mughal, N.; Moore, L.S.P. Bacterial and fungal coinfection among hospitalized patients with COVID-19: A retrospective cohort study in a UK secondary-care setting. Clin. Microbiol. Infect. Off. Publ. Eur. Soc. Clin. Microbiol. Infect. Dis. 2020, 26, 1395–1399. [Google Scholar] [CrossRef] [PubMed]
  50. Zurabov, F.M.; Chernevskaya, E.A.; Beloborodova, N.V.; Zurabov, A.Y.; Petrova, M.V.; Yadgarov, M.Y.; Popova, V.M.; Fatuev, O.E.; Zakharchenko, V.E.; Gurkova, M.M.; et al. Bacteriophage Cocktails in the Post-COVID Rehabilitation. Viruses 2022, 14, 2614. [Google Scholar] [CrossRef] [PubMed]
  51. Chen, X.; Liao, B.; Cheng, L.; Peng, X.; Xu, X.; Li, Y.; Hu, T.; Li, J.; Zhou, X.; Ren, B. The microbial coinfection in COVID-19. Appl. Microbiol. Biotechnol. 2020, 104, 7777–7785. [Google Scholar] [CrossRef]
  52. Lai, C.-C.; Yu, W.-L. COVID-19 associated with pulmonary aspergillosis: A literature review. J. Microbiol. Immunol. Infect. Wei Mian Yu Gan Ran Za Zhi 2021, 54, 46–53. [Google Scholar] [CrossRef]
  53. Yamamoto, S.; Saito, M.; Tamura, A.; Prawisuda, D.; Mizutani, T.; Yotsuyanagi, H. The human microbiome and COVID-19: A systematic review. PLoS ONE 2021, 16, e0253293. [Google Scholar] [CrossRef]
  54. Bomar, L.; Brugger, S.D.; Lemon, K.P. Bacterial Microbiota of the Nasal Passages Across the Span of Human Life. Curr. Opin. Microbiol. 2018, 41, 8–14. [Google Scholar] [CrossRef]
  55. Yu, X.; Wang, L.; Zheng, X.; Wen, Y.; Zhang, Z.; Fan, L.; Zhou, Q.; Yang, X.; Xue, B.; Lin, Y. Moraxella occupied the largest proportion in the nasal microbiome in healthy children, which potential protect them from COVID-19. Microb. Pathog. 2022, 170, 105685. [Google Scholar] [CrossRef] [PubMed]
  56. Koch, C.M.; Prigge, A.D.; Anekalla, K.R.; Shukla, A.; Do-Umehara, H.C.; Setar, L.; Chavez, J.; Abdala-Valencia, H.; Politanska, Y.; Markov, N.S.; et al. Immune response to SARS-CoV-2 in the nasal mucosa in children and adults. medRxiv, 2021; preprint. [Google Scholar]
  57. Bosco-Lauth, A.M.; Hartwig, A.E.; Porter, S.M.; Gordy, P.W.; Nehring, M.; Byas, A.D.; VandeWoude, S.; Ragan, I.K.; Maison, R.M.; Bowen, R.A. Experimental infection of domestic dogs and cats with SARS-CoV-2: Pathogenesis, transmission, and response to reexposure in cats. Proc. Natl. Acad. Sci. USA 2020, 117, 26382–26388. [Google Scholar] [CrossRef]
  58. Porter, S.M.; Hartwig, A.E.; Bielefeldt-Ohmann, H.; Bosco-Lauth, A.M.; Root, J.J. Susceptibility of Wild Canids to SARS-CoV-2. Emerg. Infect Dis. 2022, 28, 1852–1855. [Google Scholar] [CrossRef] [PubMed]
  59. Mahoney, M.; Damalanka, V.C.; Tartell, M.A.; Chung, D.; hee Lourenço, A.L.; Pwee, D.; Mayer Bridwell, A.E.; Hoffmann, M.; Voss, J.; Karmakar, P.; et al. A novel class of TMPRSS2 inhibitors potently block SARS-CoV-2 and MERS-CoV viral entry and protect human epithelial lung cells. Proc. Natl. Acad. Sci. USA 2021, 118, e2108728118. [Google Scholar] [CrossRef] [PubMed]
  60. Hatesuer, B.; Bertram, S.; Mehnert, N.; Bahgat, M.M.; Nelson, P.S.; Pöhlman, S.; Schughart, K. Tmprss2 Is Essential for Influenza H1N1 Virus Pathogenesis in Mice. PLoS Pathog. 2013, 9, e1003774. [Google Scholar] [CrossRef] [PubMed]
  61. Bertram, S.; Heurich, A.; Lavender, H.; Gierer, S.; Danisch, S.; Perin, P.; Lucas, J.M.; Nelson, P.S.; Pöhlmann, S.; Soilleux, E.J. Influenza and SARS-Coronavirus Activating Proteases TMPRSS2 and HAT Are Expressed at Multiple Sites in Human Respiratory and Gastrointestinal Tracts. PLoS ONE 2012, 7, e35876. [Google Scholar] [CrossRef] [PubMed]
  62. Shen, L.W.; Mao, H.J.; Wu, Y.L.; Tanaka, Y.; Zhang, W. TMPRSS2: A potential target for treatment of influenza virus and coronavirus infections. Biochimie 2017, 142, 1–10. [Google Scholar] [CrossRef] [PubMed]
  63. Limburg, H.; Harbig, A.; Bestle, D.; Stein, D.A.; Moulton, H.M.; Jaeger, J.; Janga, H.; Hardes, K.; Koepke, J.; Schulte, L.; et al. TMPRSS2 Is the Major Activating Protease of Influenza A Virus in Primary Human Airway Cells and Influenza B Virus in Human Type II Pneumocytes. J. Virol. 2019, 93, e00649-19. [Google Scholar] [CrossRef]
  64. Matarese, A.; Gambardella, J.; Sardu, C.; Santulli, G. miR-98 Regulates TMPRSS2 Expression in Human Endothelial Cells: Key Implications for COVID-19. Biomedicines 2020, 8, 462. [Google Scholar] [CrossRef]
  65. Mahmoud, I.S.; Jarrar, Y.B. Targeting the intestinal TMPRSS2 protease to prevent SARS-CoV-2 entry into enterocytes-prospects and challenges. Mol. Biol. Rep. 2021, 48, 4667–4675. [Google Scholar] [CrossRef]
  66. Sakamoto, A.; Kawakami, R.; Kawai, K.; Gianatti, A.; Pellegrini, D.; Kutys, R.; Guo, L.; Mori, M.; Cornelissen, A.; Sato, Y.; et al. ACE2 (Angiotensin-Converting Enzyme 2) and TMPRSS2 (Transmembrane Serine Protease 2) Expression and Localization of SARS-CoV-2 Infection in the Human Heart. Arterioscler. Thromb. Vasc. Biol. 2021, 41, 542–544. [Google Scholar] [CrossRef] [PubMed]
  67. Schimmel, L.; Chew, K.Y.; Stocks, C.J.; Yordanov, T.E.; Essebier, P.; Kulasinghe, A.; Monkman, J.; dos Santos Miggiolaro, A.F.R.; Cooper, C.; de Noronha, L.; et al. Endothelial cells are not productively infected by SARS-CoV-2. Clin. Transl. Immunol. 2021, 10, e1350. [Google Scholar] [CrossRef] [PubMed]
  68. Uhlén, M.; Fagerberg, L.; Hallström, B.M.; Lindskog, C.; Oksvold, P.; Mardinoglu, A.; Sivertsson, Å.; Kampf, C.; Sjöstedt, E.; Asplund, A.; et al. Tissue-based map of the human proteome. Science 2015, 347, 1260419. [Google Scholar] [CrossRef] [PubMed]
  69. Thul, P.J.; Åkesson, L.; Wiking, M.; Mahdessian, D.; Geladaki, A.; Ait Blal, H.; Alm, T.; Asplund, A.; Björk, L.; Breckels, L.M.; et al. A subcellular map of the human proteome. Science 2017, 356, eaal3321. [Google Scholar] [CrossRef] [PubMed]
  70. Uhlén, M.; Karlsson, M.J.; Hober, A.; Svensson, A.-S.; Scheffel, J.; Kotol, D.; Zhong, W.; Tebani, A.; Strandberg, L.; Edfors, F.; et al. The human secretome. Sci. Signal 2019, 12, eaaz0274. [Google Scholar] [CrossRef] [PubMed]
  71. Bilinska, K.; Jakubowska, P.; Von Bartheld, C.S.; Butowt, R. Expression of the SARS-CoV-2 Entry Proteins, ACE2 and TMPRSS2, in Cells of the Olfactory Epithelium: Identification of Cell Types and Trends with Age. ACS Chem. Neurosci. 2020, 11, 1555–1562. [Google Scholar] [CrossRef] [PubMed]
  72. Thunders, M.; Delahunt, B. Gene of the month: TMPRSS2 (transmembrane serine protease 2). J. Clin. Pathol. 2020, 73, 773–776. [Google Scholar] [CrossRef] [PubMed]
  73. Wettstein, L.; Kirchhoff, F.; Münch, J. The Transmembrane Protease TMPRSS2 as a Therapeutic Target for COVID-19 Treatment. Int. J. Mol. Sci. 2022, 23, 1351. [Google Scholar] [CrossRef]
  74. Simmons, G.; Zmora, P.; Gierer, S.; Heurich, A.; Pöhlmann, S. Proteolytic activation of the SARS-coronavirus spike protein: Cutting enzymes at the cutting edge of antiviral research. Antivir. Res. 2013, 100, 605–614. [Google Scholar] [CrossRef]
  75. Rabaan, A.A.; Alahmed, S.H.; Bazzi, A.M.; Alhani, H.M. A review of candidate therapies for Middle East respiratory syndrome from a molecular perspective. J. Med. Microbiol. 2017, 66, 1261–1274. [Google Scholar] [CrossRef]
  76. Shapira, T.; Monreal, I.A.; Dion, S.P.; Buchholz, D.W.; Imbiakha, B.; Olmstead, A.D.; Jager, M.; Désilets, A.; Gao, G.; Martins, M.; et al. A TMPRSS2 inhibitor acts as a pan-SARS-CoV-2 prophylactic and therapeutic. Nature 2022, 605, 340–348. [Google Scholar] [CrossRef] [PubMed]
  77. Wettstein, L.; Knaff, P.M.; Kersten, C.; Müller, P.; Weil, T.; Conzelmann, C.; Müller, J.A.; Brückner, M.; Hoffmann, M.; Pöhlmann, S.; et al. Peptidomimetic inhibitors of TMPRSS2 block SARS-CoV-2 infection in cell culture. Commun. Biol. 2022, 5, 681. [Google Scholar] [CrossRef] [PubMed]
  78. Meng, B.; Abdullahi, A.; Ferreira, I.A.T.M.; Goonawardane, N.; Saito, A.; Kimura, I.; Yamasoba, D.; Gerber, P.P.; Fatihi, S.; Rathore, S.; et al. Altered TMPRSS2 usage by SARS-CoV-2 Omicron impacts infectivity and fusogenicity. Nature 2022, 603, 706–714. [Google Scholar] [CrossRef] [PubMed]
  79. Whittaker, G.R. SARS-CoV-2 spike and its adaptable furin cleavage site. Lancet Microbe 2021, 2, e488–e489. [Google Scholar] [CrossRef] [PubMed]
  80. Zhang, L.; Mann, M.; Syed, Z.A.; Reynolds, H.M.; Tian, E.; Samara, N.L.; Zeldin, D.C.; Tabak, L.A.; Ten Hagen, K.G. Furin cleavage of the SARS-CoV-2 spike is modulated by O-glycosylation. Proc. Natl. Acad. Sci. USA 2021, 118, e2109905118. [Google Scholar] [CrossRef] [PubMed]
  81. Papa, G.; Mallery, D.L.; Albecka, A.; Welch, L.G.; Cattin-Ortolá, J.; Luptak, J.; Paul, D.; McMahon, H.T.; Goodfellow, I.G.; Carter, A.; et al. Furin cleavage of SARS-CoV-2 Spike promotes but is not essential for infection and cell-cell fusion. PLoS Pathog. 2021, 17, e1009246. [Google Scholar] [CrossRef] [PubMed]
  82. Chan, Y.A.; Zhan, S.H. The Emergence of the Spike Furin Cleavage Site in SARS-CoV-2. Mol. Biol. Evol. 2022, 39, msab327. [Google Scholar] [CrossRef] [PubMed]
  83. Jocher, G.; Grass, V.; Tschirner, S.K.; Riepler, L.; Breimann, S.; Kaya, T.; Oelsner, M.; Hamad, M.S.; Hofmann, L.I.; Blobel, C.P.; et al. ADAM10 and ADAM17 promote SARS-CoV-2 cell entry and spike protein-mediated lung cell fusion. EMBO Rep. 2022, 23, e54305. [Google Scholar] [CrossRef]
  84. Yamamoto, M.; Gohda, J.; Kobayashi, A.; Tomita, K.; Hirayama, Y.; Koshikawa, N.; Seiki, M.; Semba, K.; Akiyama, T.; Kawaguchi, Y.; et al. Metalloproteinase-dependent and TMPRSS2-independnt cell surface entry pathway of SARS-CoV-2 requires the furin-cleavage site and the S2 domain of spike protein. Mbio 2022, 13, e00519-22. [Google Scholar] [CrossRef]
  85. Bosch, B.J.; Bartelink, W.; Rottier, P.J.M. Cathepsin L Functionally Cleaves the Severe Acute Respiratory Syndrome Coronavirus Class I Fusion Protein Upstream of Rather than Adjacent to the Fusion Peptide. J. Virol. 2008, 82, 8887–8890. [Google Scholar] [CrossRef]
  86. Scarcella, M.; d’Angelo, D.; Ciampa, M.; Tafuri, S.; Avallone, L.; Pavone, L.M.; De Pasquale, V. The Key Role of Lysosomal Protease Cathepsins in Viral Infections. Int. J. Mol. Sci. 2022, 23, 9089. [Google Scholar] [CrossRef] [PubMed]
  87. Meyer, B.; Chiaravalli, J.; Gellenoncourt, S.; Brownridge, P.; Bryne, D.P.; Daly, L.A.; Grauslys, A.; Walter, M.; Agou, F.; Chakrabarti, L.A.; et al. Characterising proteolysis during SARS-CoV-2 infection identifies viral cleavage sites and cellular targets with therapeutic potential. Nat. Commun. 2021, 12, 5553. [Google Scholar] [CrossRef] [PubMed]
  88. Du, X.; Tang, H.; Gao, L.; Wu, Z.; Meng, F.; Yan, R.; Qiao, S.; An, J.; Wang, C.; Qin, F.X.-F. Omicron adopts a different strategy from Delta and other variants to adapt to host. Signal Transduct. Target Ther. 2022, 7, 1–3. [Google Scholar] [CrossRef] [PubMed]
  89. Hu, B.; Chan, J.F.-W.; Liu, H.; Liu, Y.; Chai, Y.; Shi, J.; Shuai, H.; Hou, Y.; Huang, X.; Yuen, T.T.-T.; et al. Spike mutations contributing to the altered entry preference of SARS-CoV-2 omicron BA.1 and BA.2. Emerg. Microbes Infect. 2022, 11, 2275–2287. [Google Scholar] [CrossRef] [PubMed]
  90. Zhao, H.; Lu, L.; Peng, Z.; Chen, L.-L.; Meng, X.; Zhang, C.; Ip, J.D.; Chan, W.-M.; Chu, A.W.-H.; Chan, K.-H.; et al. SARS-CoV-2 Omicron variant shows less efficient replication and fusion activity when compared with Delta variant in TMPRSS2-expressed cells. Emerg. Microbes Infect. 2022, 11, 277–283. [Google Scholar] [CrossRef] [PubMed]
  91. Padmanabhan, P.; Dixit, N. Modelling how increased Cathepsin B/L and decreased TMPRSS2 usage for cell entry by the SARS-CoV-2 Omicron variant may affect the efficacy and synergy of TMPRSS2 and Cathepsin B/L inhibitors. J. Theor. Biol. 2023, 572, 111568. [Google Scholar] [CrossRef] [PubMed]
  92. Berdowska, I.; Matusiewicz, M. Cathepsin L, transmembrane peptidase/serine subfamily member 2/4, and other host proteases in COVID-19 pathogenesis—With impact on gastrointestinal tract. World J. Gastroenterol. 2021, 27, 6590–6600. [Google Scholar] [CrossRef]
  93. Migueres, M.; Dimeglio, C.; Mansuy, J.M.; Abravanel, F.; Raymond, S.; Latour, J.; Jeanne, N.; Ranger, N.; Lhomme, S.; Saune, K.; et al. Influence of Nasopharyngeal Viral Load on the Spread of the Omicron BA. 2 Variant. Clin. Infect. Dis. 2022, 76, ciac563. [Google Scholar] [CrossRef]
  94. Kozlov, M. How does Omicron spread so fast? A high viral load isn’t the answer. Nature, 2022; ahead of print. [Google Scholar] [CrossRef]
  95. Lapid, N. Omicron Thrives in Airways, Not Lungs; New Data on Asymptomatic Cases. Reuters. 2021. Available online: https://www.reuters.com/business/healthcare-pharmaceuticals/omicron-thrives-airways-not-lungs-new-data-asymptomatic-cases-2021-12-15/ (accessed on 12 March 2023).
  96. Dance, A. Omicron’s lasting mysteries: Four questions scientists are racing to answer. Nature 2022, 603, 22–24. [Google Scholar] [CrossRef]
  97. Karimian, A.; Behjati, M.; Karimian, M. Molecular mechanisms involved in anosmia induced by SARS-CoV-2, with a focus on the transmembrane serine protease TMPRSS2. Arch Virol. 2022, 167, 1931–1946. [Google Scholar] [CrossRef]
  98. Butowt, R.; Bilińska, K.; von Bartheld, C. Why Does the Omicron Variant Largely Spare Olfactory Function? Implications for the Pathogenesis of Anosmia in Coronavirus Disease 2019. J. Infect Dis. 2022, 226, 1304–1308. [Google Scholar] [CrossRef] [PubMed]
  99. Chakraborty, C.; Bhattacharya, M.; Sharma, A.R.; Mallik, B. Omicron (B.1.1.529)—A new heavily mutated variant: Mapped location and probable properties of its mutations with an emphasis on S-glycoprotein. Int. J. Biol. Macromol. 2022, 219, 980–997. [Google Scholar] [CrossRef] [PubMed]
  100. Stockley, R.A. Chronic bronchitis: The antiproteinase/proteinase balance and the effect of infection and corticosteroids. Clin. Chest Med. 1988, 9, 643–656. [Google Scholar] [CrossRef] [PubMed]
  101. Gadek, J.E.; Pacht, E.R. The protease-antiprotease balance within the human lung: Implications for the pathogenesis of emphysema. Lung 1990, 168, 552–564. [Google Scholar] [CrossRef] [PubMed]
  102. Hutchison, D.C. The rôle of proteases and antiproteases in bronchial secretions. Eur. J. Respir. Dis. Suppl. 1987, 153, 78–85. [Google Scholar] [PubMed]
  103. Misz, M.; Fülep, E. Pathophysiological relations and clinical significance of serine protease inhibitors (serpins). Orv. Hetil. 1990, 131, 1851–1852, 1855–1859. [Google Scholar]
  104. Meyer, M.; Jaspers, I. Respiratory protease/antiprotease balance determines susceptibility to viral infection and can be modified by nutritional antioxidants. Am. J. Physiol. Lung Cell Mol. Physiol. 2015, 308, L1189–L1201. [Google Scholar] [CrossRef]
  105. Goel, K.; Serban, K.A. The multifaceted protease-antiprotease imbalance in COVID-19. EBioMedicine 2022, 78, 103973. [Google Scholar] [CrossRef]
  106. Oriano, M.; Amati, F.; Gramegna, A.; De Soyza, A.; Mantero, M.; Sibila, O.; Chotirmall, S.H.; Voza, A.; Marchisio, P.; Blasi, F.; et al. Protease–Antiprotease Imbalance in Bronchiectasis. Int. J. Mol. Sci. 2021, 22, 5996. [Google Scholar] [CrossRef]
  107. Chakraborti, S.; Sarkar, J.; Pramanik, P.K.; Chakraborti, T. Role of Proteases in Lung Disease: A Brief Overview. In Proteases in Human Diseases; Chakraborti, S., Chakraborti, T., Dhalla, N.S., Eds.; Springer: Singapore, 2017; pp. 333–374. [Google Scholar]
  108. Vergnolle, N. Protease inhibition as new therapeutic strategy for GI diseases. Gut 2016, 65, 1215–1224. [Google Scholar] [CrossRef]
  109. Wang, C.; Zhou, J.; Wang, J.; Li, S.; Fukunaga, A.; Yodoi, J.; Tian, H. Progress in the mechanism targeted drug therapy for, C.O.P.D. Signal Transduct Target. Ther. 2020, 5, 201. [Google Scholar]
  110. Li, X.; Liu, H.; Tong, Y. Concerns on cross-species transmission of SARS-CoV-2 between pets and humans. Front. Microbiol. 2022, 13, 985528. [Google Scholar] [CrossRef] [PubMed]
  111. Wei, C.; Shan, K.-J.; Wang, W.; Zhang, S.; Huan, Q.; Qian, W. Evidence for a mouse origin of the SARS-CoV-2 Omicron variant. J. Genet. Genom. 2021, 48, 1111–1121. [Google Scholar] [CrossRef] [PubMed]
  112. Puente, X.S.; López-Otín, C. A Genomic Analysis of Rat Proteases and Protease Inhibitors. Genome Res. 2004, 14, 609. [Google Scholar] [CrossRef] [PubMed]
  113. Puente, X.S.; Sánchez, L.M.; Overall, C.M.; López-Otín, C. Human and mouse proteases: A comparative genomic approach. Nat. Rev. Genet. 2003, 4, 544–558. [Google Scholar] [CrossRef] [PubMed]
  114. Puente, X.S.; Sánchez, L.M.; Gutiérrez-Fernández, A.; Velasco, G.; López-Otín, C. A genomic view of the complexity of mammalian proteolytic systems. Biochem. Soc. Trans. 2005, 33, 331–334. [Google Scholar] [CrossRef] [PubMed]
  115. Natalini, J.G.; Singh, S.; Segal, L.N. The dynamic lung microbiome in health and disease. Nat. Rev. Microbiol. 2022, 21, 222–235. [Google Scholar] [CrossRef] [PubMed]
  116. Blekhman, R.; Goodrich, J.K.; Huang, K.; Sun, Q.; Bukowski, R.; Bell, J.T.; Spector, T.D.; Keinan, A.; Ley, R.E.; Gevers, D.; et al. Host genetic variation impacts microbiome composition across human body sites. Genome Biol. 2015, 16, 191. [Google Scholar] [CrossRef]
  117. Garrigues, Q.; Apper, E.; Chastant, S.; Mila, H. Gut microbiota development in the growing dog: A dynamic process influenced by maternal, environmental and host factors. Front. Vet. Sci. 2022, 9, 964649. [Google Scholar] [CrossRef]
  118. Hasan, N.; Yang, H. Factors affecting the composition of the gut microbiota, and its modulation. PeerJ 2019, 7, e7502. [Google Scholar] [CrossRef]
  119. Weinstein, S.B.; Martínez-Mota, R.; Stapleton, T.E.; Klure, D.M.; Greenhalgh, R.; Orr, T.J.; Dale, C.; Kohl, K.D.; Dearing, M.D. Microbiome stability and structure is governed by host phylogeny over diet and geography in woodrats (Neotoma spp.). Proc. Natl. Acad. Sci. USA 2021, 118, e2108787118. [Google Scholar] [CrossRef]
  120. Carli, G.; Cecchi, L.; Stebbing, J.; Parronchi, P.; Farsi, A. Is asthma protective against COVID-19? Allergy 2021, 76, 866–868. [Google Scholar] [CrossRef] [PubMed]
  121. Farne, H.; Singanayagam, A. Why asthma might surprisingly protect against poor outcomes in COVID-19. Eur. Respir J. 2020, 56, 2003045. [Google Scholar] [CrossRef] [PubMed]
  122. Hou, H.; Xu, J.; Li, Y.; Wang, Y.; Yang, H. The Association of Asthma With COVID-19 Mortality: An Updated Meta-Analysis Based on Adjusted Effect Estimates. J. Allergy Clin. Immunol. Pract. 2021, 9, 3944–3968.e5. [Google Scholar] [CrossRef] [PubMed]
  123. Murphy, T.R.; Busse, W.; Holweg, C.T.J.; Rajput, Y.; Raimundo, K.; Meyer, C.S.; Seetasith, A.; Gupta, S.; Iqbal, A.; Kaner, R.J. Patients with allergic asthma have lower risk of severe COVID-19 outcomes than patients with nonallergic asthma. BMC Pulm. Med. 2022, 22, 418. [Google Scholar] [CrossRef] [PubMed]
  124. Gao, Y.; Agache, I.; Akdis, M.; Nadeau, K.; Klimek, L.; Jutel, M.; Akdis, C.A. The effect of allergy and asthma as a comorbidity on the susceptibility and outcomes of COVID-19. Int. Immunol. 2021, 34, dxab107. [Google Scholar] [CrossRef] [PubMed]
  125. CDC: Corticosteroids, Antibiotics Generally not Recommended for COVID-19 Outpatients|AHA News. Available online: https://www.aha.org/news/headline/2022-04-26-cdc-corticosteroids-antibiotics-generally-not-recommended-covid-19 (accessed on 12 March 2023).
  126. Singh, A. COVID-19 and Corticosteroids News-Medicalnet. 2022. Available online: https://www.news-medical.net/health/COVID-19-and-Corticosteroids.aspx (accessed on 12 March 2023).
  127. Ebrahimi Chaharom, F.; Pourafkari, L.; Ebrahimi Chaharom, A.A.; Nader, N.D. Effects of corticosteroids on Covid-19 patients: A systematic review and meta-analysis on clinical outcomes. Pulm. Pharmacol. Ther. 2022, 72, 102107. [Google Scholar] [CrossRef]
  128. Bradley, M.C.; Perez-Vilar, S.; Chillarige, Y.; Dong, D.; Martinez, A.I.; Weckstein, A.R.; Dal Pan, G.J. Systemic Corticosteroid Use for COVID-19 in US Outpatient Settings from April 2020 to August 2021. JAMA 2022, 327, 2015–2018. [Google Scholar] [CrossRef]
  129. Pandey, K.C.; De, S.; Mishra, P.K. Role of Proteases in Chronic Obstructive Pulmonary Disease. Front. Pharmacol. 2017, 8, 512. [Google Scholar] [CrossRef]
  130. Dey, T.; Kalita, J.; Weldon, S.; Taggart, C.C. Proteases and Their Inhibitors in Chronic Obstructive Pulmonary Disease. J. Clin. Med. 2018, 7, 244. [Google Scholar] [CrossRef]
  131. Kesic, M.J.; Hernandez, M.; Jaspers, I. Airway protease/antiprotease imbalance in atopic asthmatics contributes to increased Influenza A virus cleavage and replication. Respir. Res. 2012, 13, 82. [Google Scholar] [CrossRef] [PubMed]
  132. Balenga, N.A.; Klichinsky, M.; Xie, Z.; Chan, E.C.; Zhao, M.; Jude, J.; Laviolette, M.; Panettieri, R.A.; Druey, K.M. A fungal protease allergen provokes airway hyperresponsiveness in asthma. Nat. Commun. 2015, 6, 6763. [Google Scholar] [CrossRef] [PubMed]
  133. Deng, Z.; Li, Z.; She, Y.; Xie, B. Increased Expression of SERPINB10 Associated with Postoperative Recurrence in Chronic Rhinosinusitis with Nasal Polyps. Dis. Markers 2022, 2022, e7164318. [Google Scholar] [CrossRef] [PubMed]
  134. Mo, Y.; Zhang, K.; Feng, Y.; Yi, L.; Liang, Y.; Wu, W.; Zhao, J.; Zhang, Z.; Xu, Y.; Hu, Q.; et al. Epithelial SERPINB10, a novel marker of airway eosinophilia in asthma, contributes to allergic airway inflammation. Am. J. Physiol. Lung Cell Mol. Physiol. 2019, 316, L245–L254. [Google Scholar] [CrossRef] [PubMed]
  135. Mo, Y.; Ye, L.; Cai, H.; Zhu, G.; Wang, J.; Zhu, M.; Song, X.; Yang, C.; Jin, M. SERPINB10 contributes to asthma by inhibiting the apoptosis of allergenic Th2 cells. Respir Res. 2021, 22, 178. [Google Scholar] [CrossRef]
  136. Eden, E. Asthma and COPD in alpha-1 antitrypsin deficiency. Evidence for the Dutch hypothesis. COPD 2010, 7, 366–374. [Google Scholar] [CrossRef] [PubMed]
  137. Pini, L.; Paoletti, G.; Heffler, E.; Tantucci, C.; Puggioni, F.; Asthma and Alpha1-Antitrypsin Research Group. Alpha1-antitrypsin deficiency and asthma. Curr. Opin. Allergy Clin. Immunol. 2021, 21, 46. [Google Scholar] [CrossRef]
  138. Benarafa, C. Regulation of Neutrophil Serine Proteases by Intracellular Serpins. Serpin Fam. 2015, 13, 59–76. [Google Scholar]
  139. Chen, T.; Zhou, M.; Lin, M.; Liang, S.; Yan, Y.; Wang, S.; Fang, C.; Li, D.; Ruan, Y. Research Progress on the SERPINE1 Protein and Chronic Inflammatory Diseases of the Upper Respiratory Tract: A Literature Review. Int. Arch. Allergy Immunol. 2021, 182, 1097–1102. [Google Scholar] [CrossRef]
  140. Bucková, D.; Izakovicová Hollá, L.; Vácha, J. Polymorphism 4G/5G in the plasminogen activator inhibitor-1 (PAI-1) gene is associated with IgE-mediated allergic diseases and asthma in the Czech population. Allergy 2002, 57, 446–448. [Google Scholar]
  141. Kowal, K.; Bodzenta-Lukaszyk, A.; Pampuch, A.; Szmitkowski, M.; Donati, M.B.; Iacoviello, L. Plasminogen Activator Inhibitor-1 Plasma Concentration in Allergic Asthma Patients during Allergen Challenge. Int. Arch. Allergy Immunol. 2007, 144, 240–246. [Google Scholar] [CrossRef] [PubMed]
  142. Dijkstra, A.; Postma, D.S.; Bruinenberg, M.; Diemen CC van Boezen, H.M.; Koppelman, G.H.; Timens, W.; Vonk, J.M. SERPINE1-675 4G/5G polymorphism is associated with asthma severity and inhaled corticosteroid response. Eur. Respir. J. 2011, 38, 1036–1043. [Google Scholar] [CrossRef] [PubMed]
  143. Zhang, Y.; Wang, R.; Li, S.; Kong, X.; Wang, Z.; Chen, W.; Lin, N. Genetic polymorphisms in plasminogen activator inhibitor-1 predict susceptibility to steroid-induced osteonecrosis of the femoral head in Chinese population. Diagn. Pathol. 2013, 8, 169. [Google Scholar] [CrossRef] [PubMed]
  144. Park, J.; Kim, H.; Kim, S.Y.; Kim, Y.; Lee, J.-S.; Dan, K.; Seong, M.-W.; Han, D. In-depth blood proteome profiling analysis revealed distinct functional characteristics of plasma proteins between severe and non-severe COVID-19 patients. Sci. Rep. 2020, 10, 22418. [Google Scholar] [CrossRef] [PubMed]
  145. Yang, C.; Zhao, H.; Tebbutt, S.J. Leave no one behind: Inclusion of alpha-1 antitrypsin deficiency patients in COVID-19 vaccine trials. Eur. J. Hum. Genet. 2022, 30, 872–874. [Google Scholar] [CrossRef] [PubMed]
  146. Ritzmann, F.; Chitirala, P.; Krüger, N.; Hoffmann, M.; Zuo, W.; Lammert, F.; Smola, S.; Tov, N.; Alagem, N.; Lepper, P.M.; et al. Therapeutic Application of Alpha-1 Antitrypsin in COVID-19. Am. J. Respir. Crit. Care Med. 2021, 204, 224–227. [Google Scholar] [CrossRef]
  147. Yang, C.; Keshavjee, S.; Liu, M. Alpha-1 Antitrypsin for COVID-19 Treatment: Dual Role in Antiviral Infection and Anti-Inflammation. Front. Pharmacol. 2020, 11, 615398. [Google Scholar] [CrossRef]
  148. Ma, Z.; Paek, D.; Oh, C.K. Plasminogen activator inhibitor-1 and asthma: Role in the pathogenesis and molecular regulation. Clin. Exp. Allergy J. Br. Soc. Allergy Clin. Immunol. 2009, 39, 1136–1144. [Google Scholar] [CrossRef]
  149. Rajesh, K.; Cho, S.H.; Min, J.Y.; Kang, J.; Chan, W.; Kim, D.-Y.; Oh, S.; Torgerson, D.; del Del-Pino-Yanes, M.M.; Hu, D.; et al. PAI-1, Early Life Infections and Asthma Risk, Exacerbations, and Reduced Lung Function. J. Allergy Clin. Immunol. 2016, 137, AB178. [Google Scholar] [CrossRef]
  150. Cho, S.H.; Jo, A.; Casale, T.; Jeong, S.J.; Hong, S.-J.; Cho, J.K.; Holbrook, J.T.; Kumar, R.; Smith, L.J. Soy isoflavones reduce asthma exacerbation in asthmatic patients with high PAI-1–producing genotypes. J. Allergy Clin. Immunol. 2019, 144, 109–117.e4. [Google Scholar] [CrossRef]
  151. Zuo, Y.; Warnock, M.; Harbaugh, A.; Yalavarthi, S.; Gockman, K.; Zuo, M.; Madison, J.A.; Knight, J.S.; Kanthi, Y.; Lawrence, D.A. Plasma tissue plasminogen activator and plasminogen activator inhibitor-1 in hospitalized COVID-19 patients. Sci. Rep. 2021, 11, 1580. [Google Scholar] [CrossRef]
  152. Huang, P.; Zuo, Q.; Li, Y.; Oduro, P.K.; Tan, F.; Wang, Y.; Liu, X.; Li, J.; Wang, Q.; Guo, F.; et al. A Vicious Cycle: In Severe and Critically Ill COVID-19 Patients. Front. Immunol. 2022, 13, 930673. [Google Scholar] [CrossRef] [PubMed]
  153. Khan, S.S. The Central Role of PAI-1 in COVID-19: Thrombosis and beyond. Am. J. Respir Cell Mol. Biol. 2021, 65, 238–240. [Google Scholar] [CrossRef]
  154. Lin, C.-C.; Lin, L.-J.; Wang, S.-D.; Chiang, C.-J.; Chao, Y.-P.; Lin, J.; Kao, S.-T. The Effect of Serine Protease Inhibitors on Airway Inflammation in a Chronic Allergen-Induced Asthma Mouse Model. Mediat. Inflamm. 2014, 2014, e879326. [Google Scholar] [CrossRef]
  155. Menou, A.; Duitman, J.; Flajolet, P.; Sallenave, J.-M.; Mailleux, A.A.; Crestani, B. Human airway trypsin-like protease, a serine protease involved in respiratory diseases. Am. J. Physiol. Lung Cell Mol. Physiol. 2017, 312, L657–L668. [Google Scholar] [CrossRef] [PubMed]
  156. Miki, M.; Yasuoka, S.; Tsutsumi, R.; Nakamura, Y.; Hajime, M.; Takeuchi, Y.; Miki, K.; Kitada, S.; Maekura, R. Human airway trypsin-like protease enhances interleukin-8 synthesis in bronchial epithelial cells by activating protease-activated receptor 2. Arch. Biochem. Biophys. 2019, 664, 167–173. [Google Scholar] [CrossRef] [PubMed]
  157. Bertram, S.; Glowacka, I.; Müller, M.A.; Lavender, H.; Gnirss, K.; Nehlmeier, I.; Niemeyer, D.; He, Y.; Simmons, G.; Drosten, C.; et al. Cleavage and activation of the severe acute respiratory syndrome coronavirus spike protein by human airway trypsin-like protease. J. Virol. 2011, 85, 13363–13372. [Google Scholar] [CrossRef] [PubMed]
  158. Laporte, M.; Raeymaekers, V.; Van Berwaer, R.; Vandeput, J.; Marchand-Casas, I.; Thibaut, H.-J.; Van Looveren, D.; Martens, K.; Hoffmann, M.; Maes, P.; et al. The SARS-CoV-2 and other human coronavirus spike proteins are fine-tuned towards temperature and proteases of the human airways. PLoS Pathog. 2021, 17, e1009500. [Google Scholar] [CrossRef]
  159. Deo, P.N.; Deshmukh, R. Oral microbiome: Unveiling the fundamentals. J. Oral. Maxillofac. Pathol. 2019, 23, 122–128. [Google Scholar] [CrossRef]
  160. Fan, C.; Guo, L.; Gu, H.; Huo, Y.; Lin, H. Alterations in Oral–Nasal–Pharyngeal Microbiota and Salivary Proteins in Mouth-Breathing Children. Front. Microbiol. 2020, 11, 575550. [Google Scholar] [CrossRef]
  161. Gupta, A.; Karyakarte, R.; Joshi, S.; Das, R.; Jani, K.; Shouche, Y.; Sharma, A. Nasopharyngeal microbiome reveals the prevalence of opportunistic pathogens in SARS-CoV-2 infected individuals and their association with host types. Microbes Infect. 2022, 24, 104880. [Google Scholar] [CrossRef] [PubMed]
  162. Kang, H.M.; Kang, J.H. Effects of nasopharyngeal microbiota in respiratory infections and allergies. Clin. Exp. Pediatr. 2021, 64, 543–551. [Google Scholar] [CrossRef] [PubMed]
  163. Li, Y.; Houten CB van Boers, S.A.; Jansen, R.; Cohen, A.; Engelhard, D.; Kraaij, R.; Hiltemann, S.D.; Ju, J.; Fernández, D.; Mankoc, C.; et al. The diagnostic value of nasal microbiota and clinical parameters in a multi-parametric prediction model to differentiate bacterial versus viral infections in lower respiratory tract infections. PLoS ONE 2022, 17, e0267140. [Google Scholar] [CrossRef] [PubMed]
  164. O’Dwyer, D.N.; Dickson, R.P.; Moore, B.B. The Lung Microbiome, Immunity and the Pathogenesis of Chronic Lung Disease. J. Immunol. Baltim. Md. 1950 2016, 196, 4839–4847. [Google Scholar] [CrossRef] [PubMed]
  165. Yagi, K.; Huffnagle, G.B.; Lukacs, N.W.; Asai, N. The Lung Microbiome during Health and Disease. Int. J. Mol. Sci. 2021, 22, 10872. [Google Scholar] [CrossRef] [PubMed]
  166. Espírito Santo, C.; Caseiro, C.; Martins, M.J.; Monteiro, R.; Brandão, I. Gut Microbiota, in the Halfway between Nutrition and Lung Function. Nutrients 2021, 13, 1716. [Google Scholar] [CrossRef] [PubMed]
  167. Sencio, V.; Machado, M.G.; Trottein, F. The lung–gut axis during viral respiratory infections: The impact of gut dysbiosis on secondary disease outcomes. Mucosal Immunol. 2021, 14, 296–304. [Google Scholar] [CrossRef] [PubMed]
  168. Shreiner, A.B.; Kao, J.Y.; Young, V.B. The gut microbiome in health and in disease. Curr. Opin. Gastroenterol. 2015, 31, 69–75. [Google Scholar] [CrossRef]
  169. Brody, H. The gut microbiome. Nature 2020, 577, S5. [Google Scholar] [CrossRef]
  170. Harper, A.; Vijayakumar, V.; Ouwehand, A.C.; ter Haar, J.; Obis, D.; Espadaler, J.; Binda, S.; Desiraju, S.; Day, R. Viral Infections, the Microbiome, and Probiotics. Front. Cell Infect. Microbiol. 2021, 10, 596166. [Google Scholar] [CrossRef]
  171. Potempa, J.; Pike, R.N. Corruption of innate immunity by bacterial proteases. J. Innate Immun. 2009, 1, 70–87. [Google Scholar] [CrossRef] [PubMed]
  172. Corre, M.H.; Bachmann, V.; Kohn, T. Bacterial matrix metalloproteases and serine proteases contribute to the extra-host inactivation of enteroviruses in lake water. ISME J. 2022, 16, 1970–1979. [Google Scholar] [CrossRef] [PubMed]
  173. Andrade, B.G.N.; Cuadrat, R.R.C.; Tonetti, F.R.; Kitazawa, H.; Villena, J. The role of respiratory microbiota in the protection against viral diseases: Respiratory commensal bacteria as next-generation probiotics for COVID-19. Biosci. Microbiota Food Health 2022, 41, 94. [Google Scholar] [CrossRef] [PubMed]
  174. Qu, J.; Cai, Z.; Duan, X.; Zhang, H.; Cheng, H.; Han, S.; Yu, K.; Jiang, Z.; Zhang, Y.; Liu, Y.; et al. Pseudomonas aeruginosa modulates alginate biosynthesis and type VI secretion system in two critically ill COVID-19 patients. Cell Biosci. 2022, 12, 14. [Google Scholar] [CrossRef] [PubMed]
  175. Rhoades, N.S.; Pinski, A.N.; Monsibais, A.N.; Jankeel, A.; Doratt, B.M.; Cinco, I.R.; Ibraim, I.; Messaoudi, I. Acute SARS-CoV-2 infection is associated with an increased abundance of bacterial pathogens, including Pseudomonas aeruginosa in the nose. Cell Rep. 2021, 36, 109637. [Google Scholar] [CrossRef] [PubMed]
  176. Liang, Y.; Qin, X.; Hou, G.; Zhang, X.; Zhang, W. Changes of Moraxella catarrhalis infection in children before and after the COVID-19 pandemic, Zhengzhou, China. J. Infect. 2023, 86, 154–225. [Google Scholar] [CrossRef] [PubMed]
  177. Pearson, W.R. An Introduction to Sequence Similarity (“Homology”) Searching. Curr. Protoc. Bioinform. 2013, 42, Unit3.1. [Google Scholar] [CrossRef]
  178. Hark Gan, H.; Perlow, R.A.; Roy, S.; Ko, J.; Wu, M.; Huang, J.; Yan, S.; Nicoletta, A.; Vafai, J.; Sun, D.; et al. Analysis of Protein Sequence/Structure Similarity Relationships. Biophys. J. 2002, 83, 2781–2791. [Google Scholar] [CrossRef]
  179. Singh, S.; O’Reilly, S.; Gewaid, H.; Bowie, A.G.; Gautier, V.; Worrall, D.M. Reactive Centre Loop Mutagenesis of SerpinB3 to Target TMPRSS2 and Furin: Inhibition of SARS-CoV-2 Cell Entry and Replication. Int. J. Mol. Sci. 2022, 23, 12522. [Google Scholar] [CrossRef]
  180. He, M.; Gao, J.; Wu, J.; Zhou, Y.; Fu, H.; Ke, S.; Yang, H.; Chen, C.; Huang, L. Host Gender and Androgen Levels Regulate Gut Bacterial Taxa in Pigs Leading to Sex-Biased Serum Metabolite Profiles. Front. Microbiol. 2019, 10, 1359. [Google Scholar] [CrossRef]
  181. Baratchian, M.; McManus, J.M.; Berk, M.; Nakamura, F.; Mukhopadhyay, S.; Xu, W.; Erzurum, S.; Drazba, J.; Peterson, J.; Klein, E.A.; et al. Sex, androgens and regulation of pulmonary AR, TMPRSS2 and ACE2. BioRxiv, 2020; preprint. [Google Scholar]
  182. Baratchian, M.; McManus, J.M.; Berk, M.P.; Nakamura, F.; Mukhopadhyay, S.; Xu, W.; Erzurum, S.; Drazba, J.; Peterson, J.; Klein, E.A.; et al. Androgen regulation of pulmonary AR, TMPRSS2 and ACE2 with implications for sex-discordant COVID-19 outcomes. Sci. Rep. 2021, 11, 11130. [Google Scholar] [CrossRef] [PubMed]
  183. Colldén, H.; Landin, A.; Wallenius, V.; Elebring, E.; Fändriks, L.; Nilsson, M.E.; Ryberg, H.; Poutanen, M.; Sjögren, K.; Vandenput, L.; et al. The gut microbiota is a major regulator of androgen metabolism in intestinal contents. Am. J. Physiol. Endocrinol. Metab. 2019, 317, E1182–E1192. [Google Scholar] [CrossRef] [PubMed]
  184. Aman, F.; Masood, S. How Nutrition can help to fight against COVID-19 Pandemic. Pak. J. Med. Sci. 2020, 36, S121–S123. [Google Scholar] [CrossRef] [PubMed]
  185. Calder, P.C. Nutrition and immunity: Lessons for COVID-19. Eur. J. Clin. Nutr. 2021, 75, 1309–1318. [Google Scholar] [CrossRef] [PubMed]
  186. Kahleova, H.; Barnard, N.D. Can a plant-based diet help mitigate COVID-19? Eur. J. Clin. Nutr. 2022, 76, 911–912. [Google Scholar] [CrossRef] [PubMed]
  187. Kim, H.; Rebholz, C.M.; Hegde, S.; LaFiura, C.; Raghavan, M.; Lloyd, J.F.; Cheng, S.; Seidelmann, S.B. Plant-based diets, pescatarian diets and COVID-19 severity: A population-based case–control study in six countries. BMJ Nutr. Prev. Health 2021, 4, 257. [Google Scholar] [CrossRef] [PubMed]
  188. Dattner, I.; Goldberg, Y.; Katriel, G.; Yaari, R.; Gal, N.; Miron, Y.; Ziv, A.; Sheffer, R.; Hamo, Y.; Huppert, A. The role of children in the spread of COVID-19: Using household data from Bnei Brak, Israel, to estimate the relative susceptibility and infectivity of children. PLOS Comput. Biol. 2021, 17, e1008559. [Google Scholar] [CrossRef]
  189. Paul, L.A.; Daneman, N.; Schwartz, K.L.; Science, M.; Brown, K.A.; Whelan, M.; Chan, E.; Buchan, S.A. Association of Age and Pediatric Household Transmission of SARS-CoV-2 Infection. JAMA Pediatr. 2021, 175, 1151–1158. [Google Scholar] [CrossRef]
  190. Tracy, M.; Cogen, J.; Hoffman, L.R. The Pediatric Microbiome and the Lung. Curr. Opin. Pediatr. 2015, 27, 348–355. [Google Scholar] [CrossRef]
  191. Liu, T.; Lin, C.-H.; Chen, Y.-L.; Jeng, S.-L.; Tsai, H.-J.; Ho, C.-L.; Kuo, W.-S.; Hsieh, M.-H.; Chen, P.-C.; Wu, L.S.-H.; et al. Nasal Microbiome Change During and After Exacerbation in Asthmatic Children. Front. Microbiol. 2022, 12, 833726. [Google Scholar] [CrossRef]
  192. Anand, S.; Mande, S.S. Diet, Microbiota and Gut-Lung Connection. Front. Microbiol. 2018, 9, 2147. [Google Scholar] [CrossRef] [PubMed]
  193. Strain, R.; Stanton, C.; Ross, R.P. Effect of diet on pathogen performance in the microbiome. Microbiome Res. Rep. 2022, 1, 13. [Google Scholar] [CrossRef]
  194. Clark, S.E. Commensal bacteria in the upper respiratory tract regulate susceptibility to infection. Curr. Opin. Immunol. 2020, 66, 42–49. [Google Scholar] [CrossRef] [PubMed]
  195. Guo, X.; Lei, H.; Zhang, K.; Ke, F.; Song, C. Diversification of animal gut microbes and NRPS gene clusters in some carnivores, herbivores and omnivores. Biotechnol. Biotechnol. Equip. 2020, 34, 1280–1287. [Google Scholar] [CrossRef]
  196. Lau, S.K.P.; Teng, J.L.L.; Chiu, T.H.; Chan, E.; Tsang, A.K.L.; Panagiotou, G.; Zhai, S.-L.; Woo, P.C.Y. Differential Microbial Communities of Omnivorous and Herbivorous Cattle in Southern China. Comput. Struct. Biotechnol. J. 2018, 16, 54–60. [Google Scholar] [CrossRef]
  197. Senghor, B.; Sokhna, C.; Ruimy, R.; Lagier, J.-C. Gut microbiota diversity according to dietary habits and geographical provenance. Hum. Microbiome J. 2018, 7–8, 1–9. [Google Scholar] [CrossRef]
  198. Nishida, A.H.; Ochman, H. Rates of Gut Microbiome Divergence in Mammals. Mol. Ecol. 2018, 27, 1884. [Google Scholar] [CrossRef]
  199. Zoelzer, F.; Burger, A.L.; Dierkes, P.W. Unraveling differences in fecal microbiota stability in mammals: From high variable carnivores and consistently stable herbivores. Anim. Microbiome 2021, 3, 77. [Google Scholar] [CrossRef]
  200. Finlay, B.B.; Amato, K.R.; Azad, M.; Blaser, M.J.; Bosch, T.C.G.; Chu, H.; Dominguez-Bello, M.G.; Ehrlich, S.D.; Elinav, E.; Geva-Zatorsky, N.; et al. The hygiene hypothesis, the COVID pandemic, and consequences for the human microbiome. Proc. Natl. Acad. Sci. USA 2021, 118, e2010217118. [Google Scholar] [CrossRef]
  201. Xu, X.-W.; Wu, X.-X.; Jiang, X.-G.; Xu, K.-J.; Ying, L.-J.; Ma, C.-L.; Li, S.-B.; Wang, H.-Y.; Zhang, S.; Gao, H.-N.; et al. Clinical findings in a group of patients infected with the 2019 novel coronavirus (SARS-CoV-2) outside of Wuhan, China: Retrospective case series. BMJ 2020, 368, m606. [Google Scholar] [CrossRef]
  202. Yeo, W.S.; Ng, Q.X. Passive inhaled mRNA vaccination for SARS-Cov-2. Med. Hypotheses 2021, 146, 110417. [Google Scholar] [CrossRef] [PubMed]
  203. Sörensen, M.; Kantorek, J.; Byrnes, L.; Boutin, S.; Mall, M.A.; Lasitschka, F.; Zabeck, H.; Nguyen, D.; Dalpke, A.H. Pseudomonas aeruginosa Modulates the Antiviral Response of Bronchial Epithelial Cells. Front. Immunol. 2020, 11, 96. [Google Scholar] [CrossRef] [PubMed]
  204. Ruffin, M.; Bigot, J.; Calmel, C.; Mercier, J.; Pizzorno, A.; Rosa-Calatrava, M.; Corvol, H.; Balloy, V.; Terrier, O.; Guillot, L. 410: Pseudomonas aeruginosa modulates SARS-CoV-2 infectivity in CF airway epithelial cells by increasing expression of the host protease TMPRSS2. J. Cyst. Fibros 2021, 20, S193. [Google Scholar] [CrossRef]
  205. Ruffin, M.; Bigot, J.; Calmel, C.; Mercier, J.; Givelet, M.; Oliva, J.; Pizzorno, A.; Rosa-Calatrava, M.; Corvol, H.; Balloy, V.; et al. Flagellin from Pseudomonas aeruginosa Modulates SARS-CoV-2 Infectivity in Cystic Fibrosis Airway Epithelial Cells by Increasing TMPRSS2 Expression. Front. Immunol. 2021, 12, 714027. [Google Scholar] [CrossRef] [PubMed]
  206. Stanton, B.A.; Hampton, T.H.; Ashare, A. SARS-CoV-2 (COVID-19) and cystic fibrosis. Am. J. Physiol. Lung Cell Mol. Physiol. 2020, 319, L408–L415. [Google Scholar] [CrossRef] [PubMed]
  207. Camiolo, M.; Gauthier, M.; Kaminski, N.; Ray, A.; Wenzel, S.E. Expression of SARS-CoV-2 receptor ACE2 and coincident host response signature varies by asthma inflammatory phenotype. J. Allergy Clin. Immunol. 2020, 146, 315–324.e7. [Google Scholar] [CrossRef]
  208. Zhang, Y.-H.; Zhang, Y.; Dong, X.-F.; Hao, Q.-Q.; Zhou, X.-M.; Yu, Q.-T.; Li, S.-Y.; Chen, X.; Tengbeh, A.F.; Dong, B.; et al. ACE2 and Ang-(1-7) protect endothelial cell function and prevent early atherosclerosis by inhibiting inflammatory response. Inflamm. Res. Off. J. Eur. Histamine Res. Soc. Al 2015, 64, 253–260. [Google Scholar] [CrossRef] [PubMed]
  209. Salka, K.; Abutaleb, K.; Chorvinsky, E.; Thiruvengadam, G.; Arroyo, M.; Gomez, J.L.; Gutierrez, M.J.; Pillai, D.K.; Jaiswal, J.K.; Nino, G. IFN Stimulates ACE2 Expression in Pediatric Airway Epithelial Cells. Am. J. Respir. Cell Mol. Biol. 2021, 64, 515–518. [Google Scholar] [CrossRef]
  210. Katze, M.G.; He, Y.; Gale, M. Viruses and interferon: A fight for supremacy. Nat. Rev. Immunol. 2002, 2, 675–687. [Google Scholar] [CrossRef]
  211. Zhang, Q.; Lu, S.; Li, T.; Yu, L.; Zhang, Y.; Zeng, H.; Qian, X.; Bi, J.; Lin, Y. ACE2 inhibits breast cancer angiogenesis via suppressing the VEGFa/VEGFR2/ERK pathway. J. Exp. Clin. Cancer Res. CR 2019, 38, 173. [Google Scholar] [CrossRef]
  212. de Paula Gonzaga, A.L.A.C.; Palmeira, V.A.; Ribeiro, T.F.S.; Costa, L.B.; de Sá Rodrigues, K.E.; Simões-E-Silva, A.C. ACE2/Angiotensin-(1-7)/Mas Receptor Axis in Human Cancer: Potential Role for Pediatric Tumors. Curr. Drug Targets 2020, 21, 892–901. [Google Scholar] [CrossRef] [PubMed]
  213. Sodhi, C.P.; Nguyen, J.; Yamaguchi, Y.; Werts, A.D.; Lu, P.; Ladd, M.R.; Fulton, W.B.; Kovler, M.L.; Wang, S.; Prindle, T., Jr.; et al. A Dynamic Variation of Pulmonary ACE2 Is Required to Modulate Neutrophilic Inflammation in Response to Pseudomonas aeruginosa Lung Infection in Mice. J. Immunol. 2019, 203, 3000–3012. [Google Scholar] [CrossRef] [PubMed]
  214. Jia, H.; Yue, X.; Lazartigues, E. ACE2 mouse models: A toolbox for cardiovascular and pulmonary research. Nat. Commun. 2020, 11, 5165. [Google Scholar] [CrossRef] [PubMed]
  215. Heinrich, A.; Haarmann, H.; Zahradnik, S.; Frenzel, K.; Schreiber, F.; Klassert, T.E.; Heyl, K.A.; Endres, A.-S.; Schmidtke, M.; Hofmann, J.; et al. Moraxella catarrhalis decreases antiviral innate immune responses by down-regulation of TLR3 via inhibition of p53 in human bronchial epithelial cells. FASEB J. Off. Publ. Fed. Am. Soc. Exp. Biol. 2016, 30, 2426–2434. [Google Scholar] [CrossRef] [PubMed]
  216. Dioguardi, M.; Cazzolla, A.P.; Arena, C.; Sovereto, D.; Caloro, G.A.; Dioguardi, A.; Crincoli, V.; Laino, L.; Troiano, G.; Lo Muzio, L. Innate Immunity in Children and the Role of ACE2 Expression in SARS-CoV-2 Infection. Pediatr. Rep. 2021, 13, 363–382. [Google Scholar] [CrossRef] [PubMed]
  217. Wang, B.; Zhang, L.; Wang, Y.; Dai, T.; Qin, Z.; Zhou, F.; Zhang, L. Alterations in microbiota of patients with COVID-19: Potential mechanisms and therapeutic interventions. Signal Transduct. Target. Ther. 2022, 7, 1–15. [Google Scholar] [CrossRef] [PubMed]
  218. Dwivedi, S.; Purohit, P.; Misra, R.; Pareek, P.; Goel, A.; Khattri, S.; Pant, K.K.; Misra, S.; Sharma, P. Diseases and Molecular Diagnostics: A Step Closer to Precision Medicine. Indian J. Clin. Biochem. 2017, 32, 374–398. [Google Scholar] [CrossRef] [PubMed]
  219. Wang, X.; Ha, D.; Yoshitake, R.; Chen, S. White button mushroom interrupts tissue AR-mediated TMPRSS2 expression and attenuates pro-inflammatory cytokines in C57BL/6 mice. Npj Sci. Food 2021, 5, 20. [Google Scholar] [CrossRef]
  220. Fraser, B.J.; Beldar, S.; Seitova, A.; Hutchinson, A.; Mannar, D.; Li, Y.; Kwon, D.; Tan, R.; Wilson, R.P.; Leopold, K.; et al. Structure and activity of human TMPRSS2 protease implicated in SARS-CoV-2 activation. Nat. Chem. Biol. 2022, 18, 963–971. [Google Scholar] [CrossRef]
  221. Pelay-Gimeno, M.; Glas, A.; Koch, O.; Grossmann, T.N. Structure-Based Design of Inhibitors of Protein–Protein Interactions: Mimicking Peptide Binding Epitopes. Angew. Chem. Int. Ed. Engl. 2015, 54, 8896–8927. [Google Scholar] [CrossRef]
  222. Ruiz-Gómez, G.; Tyndall, J.D.A.; Pfeiffer, B.; Abbenante, G.; Fairlie, D.P. Update 1 of: Over one hundred peptide-activated G protein-coupled receptors recognize ligands with turn structure. Chem. Rev. 2010, 110, PR1-41. [Google Scholar] [CrossRef] [PubMed]
  223. Fuentes-Prior, P. Priming of SARS-CoV-2 S protein by several membrane-bound serine proteinases could explain enhanced viral infectivity and systemic COVID-19 infection. J. Biol. Chem. 2021, 296, 100135. [Google Scholar] [CrossRef] [PubMed]
  224. Pawar, N.R.; Buzza, M.S.; Antalis, T.M. Membrane-Anchored Serine Proteases and Protease-Activated Receptor-2-Mediated Signaling: Co-Conspirators in Cancer Progression. Cancer Res. 2019, 79, 301–310. [Google Scholar] [CrossRef] [PubMed]
  225. Butowt, R.; von Bartheld, C.S. Anosmia in COVID-19: Underlying Mechanisms and Assessment of an Olfactory Route to Brain Infection. Neuroscientist 2021, 27, 582–603. [Google Scholar] [CrossRef] [PubMed]
  226. Abdel Hameid, R.; Cormet-Boyaka, E.; Kuebler, W.M.; Uddin, M.; Berdiev, B.K. SARS-CoV-2 may hijack GPCR signaling pathways to dysregulate lung ion and fluid transport. Am. J. Physiol. Lung Cell Mol. Physiol. 2021, 320, L430–L435. [Google Scholar] [CrossRef] [PubMed]
  227. Szewczykowski, C.; Mardin, C.; Lucio, M.; Wallukat, G.; Hoffmanns, J.; Schröder, T.; Raith, F.; Rogge, L.; Heltmann, F.; Moritz, M.; et al. Long COVID: Association of Functional Autoantibodies against G-Protein-Coupled Receptors with an Impaired Retinal Microcirculation. Int. J. Mol. Sci. 2022, 23, 7209. [Google Scholar] [CrossRef] [PubMed]
  228. Teixeira, L.M.C.; Coimbra, J.T.S.; Ramos, M.J.; Fernandes, P.A. Transmembrane Protease Serine 2 Proteolytic Cleavage of the SARS-CoV-2 Spike Protein: A Mechanistic Quantum Mechanics/Molecular Mechanics Study to Inspire the Design of New Drugs to Fight the COVID-19 Pandemic. J. Chem. Inf. Model. 2022, 62, 2510–2521. [Google Scholar] [CrossRef] [PubMed]
  229. Roy, A.; Lushington, G.H.; McGee, J.; Chaguturu, R. Assay Technologies for Proteases. In Enzyme Technologies; John Wiley & Sons, Ltd.: Hoboken, NJ, USA, 2013; pp. 1–53. [Google Scholar]
  230. Patel, S. A critical review on serine protease: Key immune manipulator and pathology mediator. Allergol. Immunopathol. 2017, 45, 579–591. [Google Scholar] [CrossRef]
  231. Hedstrom, L. Serine Protease Mechanism and Specificity. Chem. Rev. 2002, 102, 4501–4524. [Google Scholar] [CrossRef]
  232. Krzemińska, A.; Moliner, V.; Swiderek, K. Dynamic and Electrostatic Effects on the Reaction Catalyzed by HIV-1 Protease. J. Am. Chem. Soc. 2016, 138, 16283–16298. [Google Scholar] [CrossRef]
  233. Hyland, L.J.; Tomaszek, T.A.; Roberts, G.D.; Carr, S.A.; Magaard, V.W.; Bryan, H.L.; Fakhoury, S.A.; Moore, M.L.; Minnich, M.D.; Culp, J.S. Human immunodeficiency virus-1 protease. 1. Initial velocity studies and kinetic characterization of reaction intermediates by 18O isotope exchange. Biochemistry 1991, 30, 8441–8453. [Google Scholar] [CrossRef] [PubMed]
  234. Yu, S.; Zheng, X.; Zhou, B.; Li, J.; Chen, M.; Deng, R.; Wong, G.; Lavillette, D.; Meng, G. SARS-CoV-2 spike engagement of ACE2 primes S2′ site cleavage and fusion initiation. Proc. Natl. Acad. Sci. USA 2022, 119, e2111199119. [Google Scholar] [CrossRef] [PubMed]
  235. Lin, L.; Li, Q.; Wang, Y.; Shi, Y. Syncytia formation during SARS-CoV-2 lung infection: A disastrous unity to eliminate lymphocytes. Cell Death Differ. 2021, 28, 2019–2021. [Google Scholar] [CrossRef]
  236. Magesh, S.; John, D.; Li, W.T.; Li, Y.; Mattingly-app, A.; Jain, S.; Chang, E.Y.; Ongkeko, W.M. Disparities in COVID-19 Outcomes by Race, Ethnicity, and Socioeconomic Status: A Systematic Review and Meta-analysis. JAMA Netw Open 2021, 4, e2134147. [Google Scholar] [CrossRef] [PubMed]
  237. McQuillen, K. Protein metabolism of bacteria. In Der Stickstoffumsatz/Nitrogen Metabolism; Allen, E.K., Allen, O.N., Böttger, I., Caspersson, T., Dillemann, G., Engel, H., Fischer, H., Guggenheim, M., Haas, P., et al., Eds.; Springer: Berlin/Heidelberg, Germany, 1958; pp. 659–673. [Google Scholar]
  238. Marshall, N.C.; Finlay, B.B.; Overall, C.M. Sharpening Host Defenses during Infection: Proteases Cut to the Chase. Mol. Cell Proteom. MCP 2017, 16, S161–S171. [Google Scholar] [CrossRef] [PubMed]
  239. Potempa, M.; Potempa, J. Protease-dependent mechanisms of complement evasion by bacterial pathogens. Biol. Chem. 2012, 393, 873–888. [Google Scholar] [CrossRef] [PubMed]
  240. Hibbing, M.E.; Fuqua, C.; Parsek, M.R.; Peterson, S.B. Bacterial competition: Surviving and thriving in the microbial jungle. Nat. Rev. Microbiol. 2010, 8, 15–25. [Google Scholar] [CrossRef] [PubMed]
  241. Ouertani, A.; Chaabouni, I.; Mosbah, A.; Long, J.; Barakat, M.; Mansuelle, P.; Mghirbi, O.; Najjari, A.; Ouzari, H.-I.; Masmoudi, A.S.; et al. Two New Secreted Proteases Generate a Casein-Derived Antimicrobial Peptide in Bacillus cereus Food Born Isolate Leading to Bacterial Competition in Milk. Front. Microbiol. 2018, 9, 1148. [Google Scholar] [CrossRef]
  242. Fong, S.A.; Drilling, A.; Morales, S.; Cornet, M.E.; Woodworth, B.A.; Fokkens, W.J.; Psaltis, A.J.; Vreugde, S.; Wormald, P.-J. Activity of Bacteriophages in Removing Biofilms of Pseudomonas aeruginosa Isolates from Chronic Rhinosinusitis Patients. Front. Cell Infect. Microbiol. 2017, 7, 418. [Google Scholar] [CrossRef]
  243. Roach, K. Pseudomonas sinus Infection Tricky to Eradicate. Ariz Dly Star. 2017. Available online: https://tucson.com/lifestyles/Pseudomonas-sinus-infection-tricky-to-eradicate/article_f470ccda-25f5-5bac-abc2-86f2528e1c87.html (accessed on 12 March 2023).
  244. Fothergill, J.L.; Neill, D.R.; Loman, N.; Winstanley, C.; Kadioglu, A. Pseudomonas aeruginosa adaptation in the nasopharyngeal reservoir leads to migration and persistence in the lungs. Nat. Commun. 2014, 5, 4780. [Google Scholar] [CrossRef]
  245. Kusaka, Y.; Ogawa, T.; Yamada, T.; Minami, K.; Umegaki, O.; Ukimura, A.; Kusaka, Y.; Ogawa, T.; Yamada, T.; Minami, K.; et al. Young Healthy Patient with Severe COVID-19 and Fulminant Community-Acquired Pseudomonas aeruginosa Pneumonia: A Case Report. Cureus 2022, 14, e32617. [Google Scholar] [CrossRef]
  246. Qu, J.; Cai, Z.; Liu, Y.; Duan, X.; Han, S.; Liu, J.; Zhu, Y.; Jiang, Z.; Zhang, Y.; Zhuo, C.; et al. Persistent Bacterial Coinfection of a COVID-19 Patient Caused by a Genetically Adapted Pseudomonas aeruginosa Chronic Colonizer. Front. Cell Infect. Microbiol. 2021, 11, 641920. [Google Scholar] [CrossRef]
  247. Miao, Y.; Feher, V.A.; McCammon, J.A. Gaussian Accelerated Molecular Dynamics: Unconstrained Enhanced Sampling and Free Energy Calculation. J. Chem. Theory Comput. 2015, 11, 3584–3595. [Google Scholar] [CrossRef]
  248. Pang, Y.T.; Miao, Y.; Wang, Y.; McCammon, J.A. Gaussian Accelerated Molecular Dynamics in NAMD. J. Chem. Theory Comput. 2017, 13, 9–19. [Google Scholar] [CrossRef]
  249. Phillips, J.C.; Hardy, D.J.; Maia, J.D.C.; Stone, J.E.; Ribeiro, J.V.; Bernardi, R.C.; Buch, R.; Fiorin, G.; Hénin, J.; Jiang, W.; et al. Scalable molecular dynamics on CPU and GPU architectures with NAMD. J. Chem. Phys. 2020, 153, 044130. [Google Scholar] [CrossRef]
  250. Wang, K.; Jia, Z.; Bao, L.; Wang, L.; Cao, L.; Chi, H.; Hu, Y.; Li, Q.; Zhou, Y.; Jiang, Y. Memory B cell repertoire from triple vaccinees against diverse SARS-CoV-2 variants. Nature 2022, 603, 919–925. [Google Scholar] [CrossRef]
  251. Huang, Y.; Yang, C.; Xu, X.; Xu, W.; Liu, S. Structural and functional properties of SARS-CoV-2 spike protein: Potential antivirus drug development for COVID-19. Acta Pharmacol. Sin. 2020, 41, 1141–1149. [Google Scholar] [CrossRef]
  252. Kelley, L.A.; Mezulis, S.; Yates, C.M.; Wass, M.N.; Sternberg, M.J.E. The Phyre2 web portal for protein modeling, prediction and analysis. Nat. Protoc. 2015, 10, 845–858. [Google Scholar] [CrossRef]
  253. Delano, W. Pymol [Software]. Schrodinger, Inc.: New York, NY, USA, 2022. Available online: https://pymol.org/2/(accessed on 1 January 2022).
  254. Huang, J.; MacKerell, A.D. CHARMM36 all-atom additive protein force field: Validation based on comparison to NMR data. J. Comput. Chem. 2013, 34, 2135–2145. [Google Scholar] [CrossRef]
  255. Im, W.; Feig, M.; Brooks, C.L. An Implicit Membrane Generalized Born Theory for the Study of Structure, Stability, and Interactions of Membrane Proteins. Biophys. J. 2003, 85, 2900–2918. [Google Scholar] [CrossRef]
  256. Onufriev, A.; Case, D.A.; Bashford, D. Effective Born radii in the generalized Born approximation: The importance of being perfect. J. Comput. Chem. 2002, 23, 1297–1304. [Google Scholar] [CrossRef]
  257. Koehl, P. Electrostatics calculations: Latest methodological advances. Curr. Opin. Struct. Biol. 2006, 16, 142–151. [Google Scholar] [CrossRef]
  258. Chipot, C.; Pohorille, A. Free Energy Calculations: Theory and Applications in Chemistry and Biology; Springer: Berlin/Heidelberg, Germany, 2007. [Google Scholar]
Figure 1. Host-cell infection, beginning with (A) virion binding to ACE2 receptor (blue) within a clathrin pit. (B) In the presence of TMPRSS2 (purple), extracellular cleavage of Spike (red) by TMPRSS2 enables membrane fusion and direct release of viral RNA into the cell. Otherwise, (C) clathrin (yellow cla) may mediate endocytosis, after which (D) various cathepsins (green) are able to release RNA from the vesicle.
Figure 1. Host-cell infection, beginning with (A) virion binding to ACE2 receptor (blue) within a clathrin pit. (B) In the presence of TMPRSS2 (purple), extracellular cleavage of Spike (red) by TMPRSS2 enables membrane fusion and direct release of viral RNA into the cell. Otherwise, (C) clathrin (yellow cla) may mediate endocytosis, after which (D) various cathepsins (green) are able to release RNA from the vesicle.
Biotech 12 00061 g001
Figure 2. Sequence alignment for human TMPRSS2 (sp|015393), Moraxella serine protease (MBC7754020.1), and Pseudomonas serine protease (MCL6711730.1), showing catalytic triad residues (rose-colored “C”), an important channel-altering deletion (cyan-colored “del”), and a loop conferring differential protein interactions (green “P”). For sequence positions, “*” represents full conservation, “:” is partial, and “.” is weakly conserved.
Figure 2. Sequence alignment for human TMPRSS2 (sp|015393), Moraxella serine protease (MBC7754020.1), and Pseudomonas serine protease (MCL6711730.1), showing catalytic triad residues (rose-colored “C”), an important channel-altering deletion (cyan-colored “del”), and a loop conferring differential protein interactions (green “P”). For sequence positions, “*” represents full conservation, “:” is partial, and “.” is weakly conserved.
Biotech 12 00061 g002
Figure 3. Two bound models of the SARS-CoV-2 Spike S2′ cleavable hairpin. (A) shows Conformation 1, bound to human TMPRSS2 (green), while (B) implies an alternative binding mode to the homologous Moraxella serine protease (blue). In (C), conformation 1 achieves a stable pre-reactive distance of 4.6 A relative to the TMPRSS2 catalytic serine (S441) for cleavage at R815 but does not support close approach to MSP. In (D), however, we see that conformation 2 supports close pre-reactive approach of cleavable K814 to MSP/S206, which is unavailable for approach to TMPRSS2/S441.
Figure 3. Two bound models of the SARS-CoV-2 Spike S2′ cleavable hairpin. (A) shows Conformation 1, bound to human TMPRSS2 (green), while (B) implies an alternative binding mode to the homologous Moraxella serine protease (blue). In (C), conformation 1 achieves a stable pre-reactive distance of 4.6 A relative to the TMPRSS2 catalytic serine (S441) for cleavage at R815 but does not support close approach to MSP. In (D), however, we see that conformation 2 supports close pre-reactive approach of cleavable K814 to MSP/S206, which is unavailable for approach to TMPRSS2/S441.
Biotech 12 00061 g003
Figure 4. Simulated leaving group clearance for SARS-CoV-2 Spike N-terminal (A) and C-terminal (B) fragments from the proteolytic active sites of human TMPRSS2 (blue), Moraxella serine protease (red), and Pseudomonas M6 peptidase (yellow). Lines reflect the distance from proteolytic serine to the N-terminal carbonyl (A) or the C-terminal amine (B), as derived from a 5 nanosecond molecular dynamics simulation.
Figure 4. Simulated leaving group clearance for SARS-CoV-2 Spike N-terminal (A) and C-terminal (B) fragments from the proteolytic active sites of human TMPRSS2 (blue), Moraxella serine protease (red), and Pseudomonas M6 peptidase (yellow). Lines reflect the distance from proteolytic serine to the N-terminal carbonyl (A) or the C-terminal amine (B), as derived from a 5 nanosecond molecular dynamics simulation.
Biotech 12 00061 g004
Table 1. Selected bacterial proteases and antiproteases of potential relevance to microbial modulation of SARS-CoV-2, as prioritized by sequence homology and alignment coverage relative to human proteins PAI-1, A1AC, and HAT. Microbes are listed by species family name. Microbial proteins are referenced in parentheses with GenBank accession numbers.
Table 1. Selected bacterial proteases and antiproteases of potential relevance to microbial modulation of SARS-CoV-2, as prioritized by sequence homology and alignment coverage relative to human proteins PAI-1, A1AC, and HAT. Microbes are listed by species family name. Microbial proteins are referenced in parentheses with GenBank accession numbers.
PAI-1/SERPINe1A1AC/SERPINa3HAT/TMPRSS11D
% Identity% Coverage% Identity% Coverage% Identity% Coverage
Staphylococcus28.76%90%25.87%87%30.30%31%
(MCM1356220.1)(MCM1356220.1)(PAL08087.1)
Pseudomonas27.34%93%32.86%16%36.21%54%
(MAK72596.1)(PNB35482.1)(MCL6711730.1)
Dolosigranulumn/an/an/an/an/an/a
(none)(none)(none)
Corynebacterium32.27%60%29.27%57%27.73%51%
(EEG25322.1)(WP_232022389.1)(WP_003858612.1)
Moraxella28.41%85%28.17%97%37.82%56%
(MBC7753780.1)(WP_219332546.1)(MBC7754020.1)
Table 2. Abundance statistics for the five most prevalent bacterial phyla or families in nasal mucosa swabs from our microbiotic profiling of 40 client-owned companion dogs.
Table 2. Abundance statistics for the five most prevalent bacterial phyla or families in nasal mucosa swabs from our microbiotic profiling of 40 client-owned companion dogs.
Phylum FamilyMean(St. Dev.)CountMaximumMinimum
Moraxella sp.63.52(25.97)4097.1813.47
Gammaproteobacteria sp.4.02(10.48)1361.380.00
Pseudomonas aeruginosa15.89(22.05)2981.370.00
Microbacteriacae sp.2.28(4.45)1521.300.00
Candidatus Gracilibacteria1.70(4.69)1026.130.00
Table 3. Quantitative assessment of prospective modes for SARS-CoV-2 Spike S2’ binding to TMPRSS2, Moraxella serine protease (MSP), and Pseudomonas serine protease (PSP). Binding free energies from our molecular dynamics simulations are given in kcal/mol. Pre-catalytic distances correspond to mean and minimum approaches between the protease active serine and a proteolytically cleavable carbonyl, as identified in Figure 3C (for TMPRSS2) or Figure 3D (MSP).
Table 3. Quantitative assessment of prospective modes for SARS-CoV-2 Spike S2’ binding to TMPRSS2, Moraxella serine protease (MSP), and Pseudomonas serine protease (PSP). Binding free energies from our molecular dynamics simulations are given in kcal/mol. Pre-catalytic distances correspond to mean and minimum approaches between the protease active serine and a proteolytically cleavable carbonyl, as identified in Figure 3C (for TMPRSS2) or Figure 3D (MSP).
TMPRSS2MSPPSP
Conf. 1Conf. 2Conf. 1Conf. 2Conf. 1Conf. 2
Binding energy−77.45−30.985.33−19.25−55.528.78
Std. err.4.384.774.464.363.263.42
Catalytic approach4.94 Å
to R815 (conf. 1)
5.11 Å
to K814 (conf. 2)
4.93 Å
to K814 (conf. 1)
Std. err.0.04 Å0.06 Å0.04 Å
Minimum distance4.42 Å4.43 Å4.39 Å
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Lushington, G.H.; Linde, A.; Melgarejo, T. Bacterial Proteases as Potentially Exploitable Modulators of SARS-CoV-2 Infection: Logic from the Literature, Informatics, and Inspiration from the Dog. BioTech 2023, 12, 61. https://0-doi-org.brum.beds.ac.uk/10.3390/biotech12040061

AMA Style

Lushington GH, Linde A, Melgarejo T. Bacterial Proteases as Potentially Exploitable Modulators of SARS-CoV-2 Infection: Logic from the Literature, Informatics, and Inspiration from the Dog. BioTech. 2023; 12(4):61. https://0-doi-org.brum.beds.ac.uk/10.3390/biotech12040061

Chicago/Turabian Style

Lushington, Gerald H., Annika Linde, and Tonatiuh Melgarejo. 2023. "Bacterial Proteases as Potentially Exploitable Modulators of SARS-CoV-2 Infection: Logic from the Literature, Informatics, and Inspiration from the Dog" BioTech 12, no. 4: 61. https://0-doi-org.brum.beds.ac.uk/10.3390/biotech12040061

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