key: cord-0056210-kbu1ja7e authors: Aguirre García, María Magdalena; Mancilla-Galindo, Javier; Paredes-Paredes, Mercedes; Tiburcio, Álvaro Zamudio; Ávila-Vanzzini, Nydia title: Mechanisms of infection by SARS-CoV-2, inflammation and potential links with the microbiome date: 2021-01-13 journal: nan DOI: 10.2217/fvl-2020-0310 sha: f82ae034bbf78838f16fe737908bb8028920a58c doc_id: 56210 cord_uid: kbu1ja7e The pandemic SARS coronavirus 2 utilizes efficient mechanisms to establish infection and evade the immune system. Established infection leads to severe inflammation in susceptible patients, the main hallmark of progression to severe coronavirus disease (COVID-19). Knowledge of the mechanisms of disease has expanded rapidly. As inflammation emerges as the central pathophysiological feature in COVID-19, elucidating how the immune system, lungs and gut communicate and interact with microbial components of the ecological niches that conform the human microbiome will shed light on how inflammation and disease progression are promoted. Studying the microbiome in COVID-19 could allow scientists to identify novel approaches to prevent severe inflammation by targeting components of the human microbiome. Innovation in the aforementioned is needed to combat this pandemic. Human coronaviruses (HCoVs) were first isolated from patients with the common cold in the 1960s [1] [2] [3] . Seven HCoVs known to cause disease in humans have since been identified: HCoV-229E, HCoV-NL63, HCoV-OC43, HCoV-HKU1, the SARS coronavirus (SARS-CoV), the Middle East respiratory syndrome coronavirus and the novel SARS-CoV-2 [4] . The latter was identified after a spike in cases of pneumonia of unknown etiology in Wuhan, Hubei Province, China during December 2019 and was initially named novel coronavirus (2019-nCoV) [5, 6] . The virus was renamed SARS-CoV-2 according to the International Committee on Taxonomy of Viruses classification criteria due to its genomic closeness to SARS-CoV; the disease caused by this virus was named coronavirus disease according to the WHO criteria for naming emerging diseases [7] . SARS-CoV-2 belongs to the genera Betacoronavirus and shares a different degree of genomic similarity with the other two epidemic coronaviruses: SARS-CoV (∼79%) and Middle East respiratory syndrome coronavirus (∼50%) [8] . COVID-19 has caused considerable morbidity and mortality worldwide and has become the central phenomenon that is shaping our current societies. Human-to-human transmission is the main route of spread of the virus, mainly through direct contact, respiratory droplets and aerosols [9] [10] [11] [12] . Management of COVID-19 has been extremely challenging due to its high infectivity, lack of effective therapeutics and potentially small groups of individuals (i.e., asymptomatic or mild disease) rapidly spreading the disease [13] [14] [15] [16] [17] . Although research describing COVID-19 and the mechanisms of infection by SARS-CoV-2 and its pathogenesis has expanded rapidly, there is still much to be learnt. Important gaps in knowledge which remain to be elucidated are the dynamic and complex interactions between the virus and the host's immune system, as well as the potential interspecies communications occurring Even though many of the symptoms of COVID-19 could partly be attributed to viral tropism as mentioned earlier, a subset of clinical features is apparently due to immune-mediated mechanisms. Namely, thromboembolic events and multiorgan failure are considered complications of COVID-19 rather than direct effects of the virus, which only patients who progress to severe and critical COVID-19 develop. The delimitation between clinical manifestations caused predominantly by the virus and the complications due to immune dysregulation is evident since most symptoms and pneumonia have their onset in the first 6 days after symptom onset in most patients, whereas complications start to occur between days 7-8, coinciding with the time viral RNA starts to decline ( Figure 1 ) [32] [33] [34] [35] [36] . It has been hypothesized that SARS-CoV-2 could disseminate to other organs via secondary viremia, targeting susceptible organs and causing direct damage [13] after successful replication in cells of the respiratory tract and evasion of innate immune defenses. However, damage to different organs is likely immune mediated as supported by the characteristic inflammatory storm described in patients who progress to severe and critical COVID-19 [37, 38] . Two distinctive features have been noticed in these patients: progressive increases in inflammation and an unusual hypercoagulable state. Determinants of progression to an unregulated inflammatory state have not been completely elucidated yet, although the clinical characteristics of these patients and recent experimental studies have started to uncover some of the key elements. Progression to severe and critical COVID-19 is known to occur more frequently in patients with increasing ages and significant comorbidities, most of which involve some degree of dysregulation of the renin-angiotensin- aldosterone system (RAAS), inflammation or both [39] [40] [41] [42] [43] . An adequate early immune response and optimal regulation of RAAS could be the key early features that prevent progression. Type I and II IFNs induce greater expression of the hACE2 receptor in epithelial surfaces, and patients with inflammatory states involving upregulated IFNs (including COVID-19) are known to express high levels of hACE2 [44] [45] [46] . High basal expression of hACE2 in patients susceptible to COVID-19 could initially put them at increased risk through greater availability of hACE2. However, increased expression of hACE2 following robust early IFN responses in both healthy and diseased individuals would be important to guarantee proper regulation of angiotensin II, which is thought to be an important element in initiating and perpetuating the hyperinflammatory state in COVID-19 through initial hyperactivation of the NF-κB pathway [47] . Early type I and III IFN responses have been related to successful resolution of the infection, while late and sustained type I and III IFN responses are related to disease progression, contributing to the inflammatory storm [29, 48] . Individuals who progress to the immune-mediated injury phase present decreased lymphocyte counts and significant elevation of neutrophils; the neutrophil-to-lymphocyte ratio is an early predictor of progression to severe and critical disease [42, 49] . Coagulation, inflammatory and organ damage markers are significantly elevated in patients with severe and critical COVID-19: C-reactive protein, procalcitonin, ferritin, erythrocyte sedimentation rate, IL-6, IL-2, IL-7, IL-8, IL-9, IL-10, CXCL10, MCP1, MIP1A, TNF-α, bilirubin, aspartate transaminase (AST), alanine transaminase (ALT) and lactate dehydrogenase (LDH) [39, 50] . Increased ferritin, neutrophil-tolymphocyte ratio, IL-6 and D-dimer are associated to increased mortality, while decreases in B cells, T cells and NK cells were characteristically noted in severe COVID-19 at presentation [51, 52] . Longitudinal comparison of lymphocyte subpopulations in patients with mild and severe disease showed marked decreases in CD3 + , CD8 + and CD4 + T cells in severe COVID-19, while no significant differences in the trajectories of B cells and NK cells were observed in the 16 day follow-up [53] . In this same study, IL-2 and IFN-γ peaked and subsequently declined in patients with severe disease, while IL-10 and IL-6 showed a sustained elevation with respect to patients with mild disease. Functional exhaustion of NK and CD8 + T cells with increased expression of NKG2, which were found to recover in convalescent patients, suggests that immune disturbance occurs early in the disease as a combination of both direct and bystander effects [54] . The finding that most patients who undergo mild-to-moderate disease produce neutralizing antibodies and specific T-cell responses [55, 56] , and early evidence of memory cells [57] add to the statement that an adequately mounted early immune response leads to resolution of the infection while generating specific and memory responses to the virus. However, the extent of different memory cells throughout the organs of the immune system and duration of memory remain to be determined. Pathological studies in patients who died after developing critical disease have revealed diffuse alveolar damage, hyaline membrane, alveolar wall thickening and infiltration with macrophages, mild-to-moderate mononuclear response, viral cytopathic effects, significant diffuse hemorrhage, small vessel thrombi with surrounding CD4 + cells, deep venous thrombosis in some cases, and degenerated neutrophils, which could represent neutrophil extracellular traps [52, [58] [59] [60] . These findings are consistent with the important immune dysregulation occurring in critical COVID-19, which together with the high pro-inflammatory cytokine levels and macrophage and monocytic infiltration resemble the macrophage activation syndrome [37] , and reflect the important hypercoagulable state in critical COVID-19, which has been referred to as 'thromboinflammation' due to high correlation between IL-6 levels, fibrinogen and histopathologic findings [61] . The main features of hypercoagulability in critical COVID-19 are normal-to-prolonged prothrombin time and activated partial thromboplastin time, elevated D-dimer, and increased fibrinogen [62] . Tang et al. reported that 71.4% of nonsurvivors and 0.6% of survivors had evidence of overt deep intravascular coagulation; more patients exhibited latent deep intravascular coagulation characterized by a hypercoagulable state, as demonstrated by fibrin thrombus [63] . Pro-inflammatory cytokines are known to contribute to hypercoagulation; IL-6 contributes mainly through increased fibrinogen production, which is known to be an acute phase reactant [64] . Certain fibrinogen polymorphisms are related to greater concentrations in response to IL-6 [65] , which could partially account for the varying frequencies in thrombotic complications encountered in COVID-19 patients. IL-1β and IL-8 promote rapid clot formation through increased fibrin-cross-linking and cellular clot components (more pronounced effect by IL-8), while IL-6 only mildly affects clot conformation [66] . The clinical course of COVID-19 can be divided into three phases (viremia, acute and severe or recovery phase), under two different scenarios (with or without interventions) [13] . The first scenario ( Figure 2A ) reflects the natural course of patients who progress to severe and critical COVID-19. The second scenario represents the timing of interventions (i.e., antivirals, immunoglobulin and low-molecular-weight heparin) which could potentially alter the course of the disease and prevent progression ( Figure 2B ). Patients with significant risk factors could require more interventions than immunocompetent individuals to avoid progression to severe and critical disease. Zhang and colleagues [67] noted that diarrhea accounted for a notable proportion of COVID-19 patients, ranging from 8.0 to 12.9%; high levels of hACE2 mRNA and protein expression were detected in the small intestinal enterocytes. In 2003, postmortem studies after SARS-CoV infection established the important viral tropism for intestinal cells (small and large intestine) where productive viral replication occurred, with subsequent accumulation of virions in the endoplasmic reticulum and shedding of the virus through its apical membrane. SARS-CoV-2 is known to cause less gastrointestinal manifestations than SARS-CoV, which could be partly explained by greater replication kinetics of SARS-CoV in intestinal cell lines [20] . However, SARS-CoV-2 has been shown to infect and productively replicate in human small intestinal organoids, subsequently altering gene expression, increasing cytokine production, promoting IFN-regulated genes and increasing hACE2 expression [46] . Thus, infection of intestinal cells could play an important role in promoting the inflammatory storm in COVID-19, mainly by promoting pro-inflammatory cytokines and through late and prolonged induction of IFNs. The trillions of microbial cells colonizing the human body were long ignored by the scientific community, but in recent years research in the field has rapidly expanded and we have learnt that microbes are a central metabolic hub, promoting in many cases physiological homeostasis and immune functions through a close symbiotic relationship with the host [68] [69] [70] . The gut microbiome has received most attention. Recently, fungi have been recognized as important components of the human microbiome, and their role in health and diseases is increasingly being studied [71, 72] . In addition, during physiological states and after disturbances in the gut microbiome (i.e., single course of antibiotics), fungal species may overtake immune modulation tasks commonly done by bacteria, thereby preventing mucosal damage and vice versa. Viruses are also key components of the human microbiome (human virome) [ Modified with permission from [13] . The gut microbiome spans beyond the GI tract, mediating a variety of intercommunications between the gut, enteric nervous system and the brain [75] ; perturbations in the gut microbiota (dysbiosis) contribute to the initiation of pro-inflammatory signaling. For instance, dysbiosis has been found to increase translocation of bacterial lipopolysaccharide (LPS) due to increased permeability of the intestinal epithelial barrier, leading to obesity and insulin resistance [76] . Novel ways by which microbial microenvironments are shaped in the human body have started to emerge. For instance, small molecules (21-24 nucleotides) of RNA named miRNAs [77] , are known to modify the gut microbiota in patients with colorectal cancer, while also altering gene expression in cancer cells, thus suggesting bilateral communication via these molecules between bacteria and human cells [78, 79] . Intestinal epithelial cells have been shown to produce miRNAs which are delivered through their apical membrane to the intestinal lumen inside extracellular vesicles, and are able to enter bacteria (Fusobacterium nucleatum and Escherichia coli), altering gene expression and promoting their growth [80] . Different miRNA profiles produced by noncolitogenic and colitogenic microbiotas are associated with intestinal inflammation in the latter [81] . Interestingly, miRNAs contained in ginger exosomes after its ingestion have been shown to shape the microbiota (increased Lactobacillaceae and Bacteroidales, and diminished Clostridiaceae) in both humans and mice, while also altering gene expression in Lactobacillus rhamnosus [82] . The miRNAs contained in extracellular vesicles travelling through the bloodstream have been studied as biomarkers of inflammation and prognostic factors in patients with acute myocardial infarction [83, 84] , although their potential as mediators of inflammation and shapers of the microbiota at distant sites has not been studied yet. Aging and comorbidities such as hypertension, cardiovascular disease, diabetes, obesity, chronic respiratory diseases, neurologic diseases and immunosuppression are significant risk factors for severe and critical COVID-19 [40, 85] . The presence of obesity-associated fatty liver disease was associated with an approximately sixfold increased risk of severe COVID-19 (unadjusted odds ratio: 5.77; 95% CI: 1.19-27.91; p = 0.029), even after adjusting for age, sex, smoking, diabetes, hypertension and dyslipidemia (adjusted odds ratio: 6.32; 95% CI: 1.16-34.54; p = 0.033) [86] . Although the precise composition of the healthy gut microbiota is not known, it is evident that microbial diversity adds to the host's health. Obesity, hypertension and diabetes are diseases extensively associated to dysbiosis, in which increased intestinal permeability and chronic inflammation occur [87, 88] . A systemic, low-grade inflammatory state is a hallmark of obesity and the metabolic syndrome. A wide range of inflammatory markers, namely C-reactive protein and pro-inflammatory cytokines are strongly associated with adiposity [89] . Multiple lines of evidence implicate gut dysbiosis as a key modulator of immune signaling in the context of metabolic diseases. LPS binds to toll-like receptors in mucosal and peripheral tissues, thereby initiating proinflammatory signaling [90, 91] . Studies in both human and murine models have linked the obesity phenotype to endotoxemia, characterized by elevated LPS plasma levels [76, 92] . Baseline levels of LPS were 20% higher in patients with obesity or glucose intolerance, whereas those with Type 2 diabetes had levels 125% higher than healthy subjects [93] . On one hand, pathogenic strains that dominate in gut dysbiosis are a rich source of LPS that could enter the circulation and initiate an immune response. On the other hand, there is strong evidence of the essential role of the gut microbiome in maintaining the integrity of the epithelial barrier; its alteration would allow increased intestinal translocation of endotoxins [94] . Microbial ecological niches are mostly limited by the external mucus layer of epithelia in the gut, interacting with the environment in the lumen and having important roles in the metabolism of components of the diet. The inner mucus layer serves as a barrier between epithelial cells and microorganisms, including potential pathogens. Symbiotic bacteria impede the proliferation of exogenous bacteria that could damage the host [95] . The lung microbiome has recently been recognized as a cornerstone in the physiopathology of numerous respiratory diseases [96] . The predominant bacterial phyla in lungs are the same as the ones in the gut, mainly Firmicutes and Bacteroidetes, followed by Proteobacteria and Actinobacteria [97] . From birth and throughout the entire life span, a close correlation between the composition of the gut and lung microbiome exists, suggesting a host-wide network [98] . Conversely, modifications in the lung microbiota after antimicrobial exposure may affect the gut microbiota composition [99] . Interactions between the lung microbiome and immunity are also a two-way process; major inflammatory events in the lungs can morbidly alter the composition of the lung microbiota [100] . As mentioned in the previous section, the long-reaching immunologic impact of the gut microbiome in other organs and systems (i.e., lungs and immune system) is increasingly recognized [101] . The mesenteric lymphatic system shapes the crucial road network allowing intercommunications between the lungs and the intestine. This way, microorganisms, microbial fragments (i.e., LPS) and metabolites (i.e., short chain fatty acids) may cross the intestinal mucosal barrier and reach the lung, thereby modulating the lung immune response [102, 103] . Particularly important players in this long-reaching immune interaction are gut segmented filamentous bacteria, which are commensal bacteria that colonize the ileum in most animals, including humans, and modulate the immune system [104] . Segmented filamentous bacteria regulate CD4 + T-cell differentiation toward the Th17 phenotype, implied in the response to pulmonary fungal infections and manifestations of autoimmune diseases in the lung [105, 106] . Recently, innate lymphoid cells, which are involved in tissue repair, have been shown to be recruited from the gut to the lungs in response to inflammatory signals upon IL-25 [107] . Of note, both Th17 and innate immune cells are major sources of second order cytokines, required for effective early microbial clearance in the lungs [108] . As mentioned earlier, SARS-CoV-2 can infect cells that express high levels of hACE2 and transmembrane serine protease 2, a common feature between the lungs, esophagus, small intestine, and to a lesser extent, the large intestine. Although the specific mechanisms involved in the pathogenesis of diarrhea in COVID-19 are not entirely known, viral infection likely alters intestinal permeability, resulting in enterocyte malabsorption [86] . Intestinal inflammation, as reflected by increased calprotectin levels, correlates with the presence of diarrhea in COVID-19 [109] . Alternatively, SARS-CoV-2 could enter and shape components of the gut microbiome through glycan receptor binding via the S glycoprotein, in a similar fashion to its proven entry to common pathogens of the lungs (Streptococcus pneumoniae and Pseudomonas aeruginosa) [110] . The significance of SARS-CoV-2 entry to microorganisms of the gut and lung microbiomes remains to be studied. Elucidating the basis of diarrhea in COVID-19 is important since patients with diarrhea had a higher need for ventilatory support (26.4 vs 8.2%; p = 0.004) and intensive care (49.0 vs 11.8%; p < 0.001), suggesting increased severity, although no correlation with the mortality rate was found [86] . The small intestine comprises the largest organ with immune functions in humans, and we have discussed how the gut microenvironment is able to affect multiple organs and systems, including the lungs. Researchers from Hong Kong have longitudinally characterized the bacterial [111] and fungal [112] composition of the gut microbiome in hospitalized patients through the entire spectrum of symptomatic COVID-19 (mildto-critical), compared with non-SARS-CoV-2 pneumonia and healthy controls. Higher numbers of opportunistic bacteria (Clostridium hathewayi, Actinomyces viscosus and Bacteroides nordii) and fungi (Candida albicans, C. auris and Aspergillus flavus) were found in patients with COVID-19, while patients who received antibiotics were depleted in bacterial symbionts (Fecalibacterium prausnitzii, Lachnospiraceae bacterium 5 1 63FAA, Eubacterium rectale, Ruminococcus obeum and Dorea formicigenerans). Dysbiosis was found to persist throughout hospitalization, even after resolution of symptoms and a negative throat swab reverse-transcriptase polymerase chain reaction (RT-PCR) for SARS-CoV-2; fungal opportunistic pathogens also persisted (A. flavus and A. niger). Predominance of certain Firmicutes bacteria, which are negatively associated with expression of ACE2 in murine models, was also negatively correlated with severity of COVID-19. The opposite was true for other Firmicutes bacteria that are associated with greater expression of ACE2. Higher expression of hACE2 in normal human tissues has also been noted to occur under the presence of certain microbial and immunological signatures [113] , which further supports the possible link between COVID-19 progression and the microbiome. In a cross-sectional study of hospitalized patients with mild-to-severe COVID-19, gut microbial composition of patients had a low diversity compared with healthy controls, a finding similar to patients with Influenza A H1N1 infection [114] . Relative abundance of Streptococcus and Escherichia/Shigella was higher both in patients with COVID-19 and influenza. Patients with COVID-19 had predominance of Streptococcus, Rothia, Veillonella, Erysipelatoclostridium and Actinomyces. The authors of this study created two prediction models to distinguish COVID-19 from influenza A H1N1 infection and from healthy controls, by using gut microbial components as biomarkers, finding an overall good performance. However, this model was not validated and does not distinguish between mild and severe disease due to similar microbial compositions in both groups at admission. Another prospective study of 15 hospitalized patients with COVID-19 found that viral infection in the GI tract occurs even in the absence of gastrointestinal symptoms and after resolution of all symptoms [115] . Interestingly, active replication of SARS-CoV-2 and transcriptional activity were correlated with shifts in the microbiota with greater abundance of opportunistic bacteria (Collinsella aerofaciens, C. tanakaei, S. infantis and Morganella morganii), while low signatures of SARS-CoV-2 infection in the gut was associated to higher presence of short-chain fatty acid producing bacteria (Parabacteroides merdae, B. stercoris, Alistipes onderdonkii and L. bacterium 1 1 57FAA) . Other studies have implied inflammation as a determinant factor of the intestinal microbiome in COVID-19, which could in turn accentuate dysregulation of the immune function: reductions in Bifidobacterium, Lactobacillus and Eubacterium, while significant increases in Corynebacterium, Actinobacterium and Ruthenibacterium (Firmicutes) were detected; fungi not commonly found in healthy subjects were also found (Aspergillus and Kluyveromyces) [116, 117] . Changes in the intestinal microbiota in the context of patients with severe disease are thought to reflect the prominent inflammatory state. Patients who develop acute respiratory distress syndrome (ARDS) have shifts in the pulmonary microbiota toward a richer composition in usual components of the gastrointestinal microbiome (Enterobacteriaceae and Bacteriodetes) [118] . Even when gastrointestinal pathogens have not been encountered in respiratory samples of patients with ARDS and SARS-CoV-2 infection, patients with COVID-19 have diminished microbial diversity in their lung microbiome, with increased abundance of opportunistic pathogens (Candida and other viruses), and transcriptional patterns that are associated with inflammation pathways [119] . We searched MEDLINE and EMBASE through OVID, PubMed, BioRxiv and MedRxiv for research on COVID-19 published until 30 October 2020. We used the publicly available COVID-19 Living Evidence on COVID-19 dataset [ For studies published prior to 2020, no specific search strategy was conducted and references were chosen at the authors' best judgment to provide educational context to readers, and may thus be biased toward the authors' opinions in this perspective article. Knowledge on the mechanisms at the molecular, cellular and systems levels by which SARS-CoV-2 causes disease in humans have been studied and communicated in an incredibly expedite manner never seen before. SARS-CoV-2 enters a wide variety of human cells in an extraordinarily efficient way which allows the virus to evade the immune system initially, thereby impeding robust type I and III IFN responses much needed to eliminate the virus. Dysregulation of RAAS and late type I and III IFN overactivation contribute to progression of the disease in susceptible individuals, although other mechanisms leading to severe and critical COVID-19 are most likely uncovered at this moment. Potential infection of cells of the immune system and direct damage to these cells by SARS-CoV-2 remains to be studied to understand how the immune response could be directly affected by the virus. We have discussed some ways by which the microbiota could possibly promote inflammation in individuals with COVID-19. We have mentioned bacterial translocation and the presence of LPS in the bloodstream as a potential promoter of inflammation, which could contribute in a similar way to the inflammatory storm which occurs in sepsis. miRNAs are also potential promoters of systemic inflammation which could be acting in different ways. For example, established dysbiosis in patients with chronic diseases could be the set point allocating these patients at increased risk of developing greater inflammation. Studying dietary and lifestyle habits in persons with comorbidities before COVID-19 infection and looking for differences in these groups could be an interesting way of establishing this connection. Cohorts of patients with chronic diseases in whom the microbiota composition has been prospectively studied could serve as the perfect group to study how dysbiosis affects predisposition to COVID-19 severity and outcomes. The gut and lungs are robustly intercommunicated through pathways such as the mesenteric lymphatic system, although the bloodstream could also serve as another highway communicating the lungs and gut through small molecules such as bacterial metabolites or miRNAs, which are likely to be able to travel long distances in the body since they are contained inside extracellular vesicles. What happens in the lung can have consequences in the gut and vice versa, while intercommunication through the immune system is undoubted. Interestingly, SARS-CoV-2 has been shown to enter bacteria through rhamnosylated epitopes which are present in some pathogenic bacteria. Exploring the expression of these epitopes in components of both healthy and dysbiotic microbiomes could be the first step toward understanding if SARS-CoV-2 is able to shape the microbiota by directly interacting with their components. The same could apply to fungal components of the microbiome. To date, only the microbiota (taxonomy of microbial components) has been studied in the context of COVID-19. Studies aiming to describe the metagenomics and transcriptomics of microbial components in the context of COVID-19 will allow to start characterizing the extent to which the microbiome affects or is affected by COVID-19. Finally, answering these questions could allow the scientific community to develop ingenious ways to ameliorate disproportionate inflammation, prevent disease progression or improve patient outcomes. A great number of scientists from multiple backgrounds have united efforts and redirected their attention toward rapid characterization of SARS-CoV-2 and the disease it causes in humans . This is unprecedented, since no biologic phenomena had ever drawn this amount of attention. Discoveries in this topic have occurred in an impressively accelerated way, and the pace at which breakthroughs are occurring suggests that it will not be long until we have precise characterizations of the virus, its pathophysiology and available therapeutics, including vaccines, to combat this pandemic. Characterization of the interplays between the lungs, gut, immune system and the human microbiome will aid to have a greater understanding of how these elements could determine disease progression or modulate responses to infection. This will open new venues toward developing innovative interventions for COVID-19. Perspective Aguirre García, Mancilla-Galindo, Paredes-Paredes, Zamudio Tiburcio &Ávila-Vanzzini Executive summary • Coronavirus disease (COVID-19), the pandemic caused by the SARS coronavirus 2 (SARS-CoV-2), has challenged current societies due to rapid spread and lack of therapeutics. • The mechanisms involved include antigen hiding, pre-activation of its spike protein and avid recognition of its receptor, the human angiotensin-converting enzyme 2. This allows the virus to initially prevent adequate interferon (IFN) responses. Immune-mediated injury is the predominant pathophysiological driver in severe & critical COVID-19 • Early IFN responses have been related to successful resolution of the infection, whereas late and sustained IFN responses are related to disease progression. • Unregulated angiotensin II could promote inflammation pathways, especially in individuals with diseases involving alterations in the renin-angiotensin-aldosterone system. • SARS-CoV-2 productively infects human enterocytes, altering gene expression, cytokine production, IFN pathways and human angiotensin-converting enzyme 2 expression. Gut microbiome • The components of the human microbiome comprise different microorganisms (i.e., bacteria, viruses and fungi). • miRNAs are 21-24 RNA molecules frequently encountered inside extracellular vesicles. miRNAs produced by the intestinal epithelium and from diet are able to shape the gut microbiome, while certain miRNA profiles promote shifting to a microbiome associated with decreased or increased intestinal inflammation. Microbial interactions within the gut-lung axis • The gut-lung axis results from complex interactions between the different microbial components of both the gut and lung niches, and the immune system. Events occurring in the lungs are able to alter the gut and vice versa. • Microorganisms, microbial fragments (i.e., lipopolysaccharide) and metabolites (i.e., short-chain fatty acids) may cross the intestinal mucosal barrier and reach the lung through the mesenteric lymphatic system and bloodstream. • Diarrhea is an important manifestation of COVID-19, which correlates with inflammation and disease severity. This symptom is likely a result of malabsorption, although SARS-CoV-2 is able to enter microorganisms with rhamnosylated epitopes, which leaves an open question for possible direct effects in microbial components of the microbiota. • Few studies have investigated the role of the microbiome in COVID-19. In patients without diarrhea, dysbiosis was related to disease severity. • In acute respiratory distress syndrome, the lung microbiota shifts toward a richer composition in habitual intestinal microorganisms. • Studies with translational approaches are needed to better understand COVID-19 and possible interventions. Cultivation of viruses from a high proportion of patients with colds A new virus isolated from the human respiratory tract Recovery in tracheal organ cultures of novel viruses from patients with respiratory disease Zoonotic origins of human coronaviruses A pneumonia outbreak associated with a new coronavirus of probable bat origin A novel coronavirus from patients with pneumonia in China The species severe acute respiratory syndrome-related coronavirus: classifying 2019-nCoV and naming it SARS-CoV-2 Genomic characterisation and epidemiology of 2019 novel coronavirus: implications for virus origins and receptor binding First known person-to-person transmission of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) in the USA Temporal dynamics in viral shedding and transmissibility of COVID-19 Pathogenesis and transmission of SARS-CoV-2 in golden hamsters Transmission of severe acute respiratory syndrome coronavirus 2 via close contact and respiratory droplets among human angiotensin-converting enzyme 2 mice • Proposes the trajectories of biomarkers in patients with coronavirus disease (COVID-19) with or without interventions Estimating the overdispersion in COVID-19 transmission using outbreak sizes outside China Recent advances, approaches and challenges in targeting pathways for potential COVID-19 vaccines development Potential therapeutic options for COVID-19: current status, challenges, and future perspectives A perspective on emerging therapeutic interventions for COVID-19. Front Structure, function, and antigenicity of the SARS-CoV-2 spike glycoprotein SARS-CoV-2 cell entry depends on ACE2 and TMPRSS2 and is blocked by a clinically proven protease inhibitor Comparative tropism, replication kinetics, and cell damage profiling of SARS-CoV-2 and SARS-CoV with implications for clinical manifestations, transmissibility, and laboratory studies of COVID-19: an observational study Cell entry mechanisms of SARS-CoV-2 SARS-CoV-2 strategically mimics proteolytic activation of human ENaC In situ structural analysis of SARS-CoV-2 spike reveals flexibility mediated by three hinges A multibasic cleavage site in the spike protein of SARS-CoV-2 is essential for infection of human lung cells The four horsemen of a viral apocalypse: the pathogenesis of SARS-CoV-2 infection (COVID-19) Expression of the SARS-CoV-2 cell receptor gene ACE2 in a wide variety of human tissues Comparative replication and immune activation profiles of SARS-CoV-2 and SARS-CoV in human lungs: an ex vivo study with implications for the pathogenesis of COVID-19 Disparate temperature-dependent virus -host dynamics for SARS-CoV-2 and SARS-CoV in the human respiratory epithelium Type I and type III interferons -induction, signaling, evasion, and application to combat COVID-19 • Review of the important role of type I and III interferons in COVID-19 pathogenesis Regional differences in airway epithelial cells reveal tradeoff between defense against oxidative stress and defense against rhinovirus Immunology of COVID-19: current state of the science Symptom progression of COVID-19 Clinical characteristics of coronavirus disease 2019 in China Factors associated with death in critically ill patients with coronavirus disease 2019 in the US Features of anosmia in COVID-19 Duration of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infectivity: when is it safe to discontinue isolation? The role of cytokines including interleukin-6 in COVID-19 induced pneumonia and macrophage activation syndrome-like disease Cytokine release syndrome in severe COVID-19 Clinical features of patients infected with 2019 novel coronavirus in Wuhan Features of 20 133 UK patients in hospital with covid-19 using the ISARIC WHO Clinical Characterisation Protocol: prospective observational cohort study Risk factors associated with acute respiratory distress syndrome and death in patients with coronavirus disease 2019 pneumonia in Wuhan, China Short-term outcomes of COVID-19 and risk factors for progression Factors associated with COVID-19-related death using OpenSAFELY SARS-CoV-2 receptor ACE2 Is an interferon-stimulated gene in human airway epithelial cells and is detected in specific cell subsets across tissues Cigarette smoke exposure and inflammatory signaling increase the expression of the SARS-CoV-2 receptor ACE2 in the respiratory tract • Demonstrates that SARS coronavirus 2 (SARS-CoV-2) infects enterocytes, which could be related to immune dysregulation and gastrointestinal symptoms in a subset of patients COVID-19: a new virus, but a familiar receptor and cytokine release syndrome Impaired type I interferon activity and inflammatory responses in severe COVID-19 patients Neutrophil-to-lymphocyte ratio predicts critical illness patients with 2019 coronavirus disease in the early stage Mild versus severe COVID-19: laboratory markers Clinical course and risk factors for mortality of adult inpatients with COVID-19 in Wuhan, China: a retrospective cohort study Pathological findings of COVID-19 associated with acute respiratory distress syndrome Longitudinal characteristics of lymphocyte responses and cytokine profiles in the peripheral blood of SARS-CoV-2 infected patients Functional exhaustion of antiviral lymphocytes in COVID-19 patients Detection of SARS-CoV-2-specific humoral and cellular immunity in COVID-19 convalescent individuals Targets of T cell responses to SARS-CoV-2 coronavirus in humans with COVID-19 disease and unexposed individuals Breadth of concomitant immune responses prior to patient recovery: a case report of non-severe COVID-19 COVID-19 autopsies Autopsy findings and venous thromboembolism in patients with COVID-19 Pulmonary and cardiac pathology in African American patients with COVID-19: an autopsy series from New Orleans Thromboinflammation and the hypercoagulability of COVID-19 Hypercoagulability of COVID-19 patients in intensive care unit: a report of thromboelastography findings and other parameters of hemostasis Abnormal coagulation parameters are associated with poor prognosis in patients with novel coronavirus pneumonia Fibrinogen as a key regulator of inflammation in disease Fibrinogen and clot-related phenotypes determined by fibrinogen polymorphisms: independent and IL-6-interactive associations Simultaneous presence of hypercoagulation and increased clot lysis time due to IL-1β, IL-6 and IL-8 Specific ACE2 expression in small intestinal enterocytes may cause gastrointestinal symptoms and injury after 2019-nCoV infection Host-bacterial mutualism in the human intestine The ecology of human microbiota: dynamics and diversity in health and disease The microbiota of the respiratory tract: gatekeeper to respiratory health The emerging world of the fungal microbiome The fungal frontier: a comparative analysis of methods used in the study of the human gut mycobiome The virome in host health and disease Virome-host interactions in intestinal health and disease Gut-brain axis in 2016: brain-gut-microbiota axis-mood, metabolism and behaviour Metabolic endotoxemia initiates obesity and insulin resistance Origins and evolution of eukaryotic RNA interference Interactions between the microRNAs and microbiota in cancer development: roles and therapeutic opportunities Human colon mucosal biofilms and murine host communicate via altered mRNA and microRNA expression during cancer The host shapes the gut microbiota via fecal microRNA Host-derived fecal microRNAs can indicate gut microbiota healthiness and ability to induce inflammation Plant-derived exosomal microRNAs shape the gut microbiota Identifying circulating microRNAs as biomarkers of cardiovascular disease: a systematic review Therapeutic potential of miR-21 regulation by human peripheral blood derived-small extracellular vesicles in myocardial infarction Does comorbidity increase the risk of patients with covid-19: evidence from meta-analysis Obesity as a risk factor for greater severity of COVID-19 in patients with metabolic associated fatty liver disease Richness of human gut microbiome correlates with metabolic markers Gut microbiota dysbiosis contributes to the development of hypertension Obesity and C-reactive protein in various populations: a systematic review and meta-analysis Probiotic Escherichia coli Nissle 1917 inhibits leaky gut by enhancing mucosal integrity The interaction of large bowel microflora with the colonic mucus barrier Lipopolysaccharide activates an innate immune system response in human adipose tissue in obesity and Type 2 diabetes High fat intake leads to acute postprandial exposure to circulating endotoxin in Type 2 diabetic subjects Understanding the role of the gut microbiome and microbial metabolites in obesity and obesity-associated metabolic disorders: current evidence and perspectives Gut flora in health and disease Respiratory mycobiome and suggestion of inter-kingdom network during acute pulmonary exacerbation in cystic fibrosis Topographical continuity of bacterial populations in the healthy human respiratory tract Neonatal gut and respiratory microbiota: coordinated development through time and space Collateral effects of antibiotics on mammalian gut microbiomes Outgrowth of the bacterial airway microbiome after rhinovirus exacerbation of chronic obstructive pulmonary disease Protective microbiota: from localized to long-reaching co-immunity Contributions of the intestinal microbiome in lung immunity Gut-dependent microbial translocation induces inflammation and cardiovascular events after ST-elevation myocardial infarction Comparative analysis of the distribution of segmented filamentous bacteria in humans, mice and chickens Pulmonary Th17 antifungal immunity is regulated by the gut microbiome Segmented filamentous bacteria provoke lung autoimmunity by inducing gut-lung axis Th17 cells expressing dual TCRs S1P-dependent interorgan trafficking of group 2 innate lymphoid cells supports host defense Early local immune defences in the respiratory tract Faecal calprotectin indicates intestinal inflammation in COVID-19 Novel ACE2-independent carbohydrate-binding of SARS-CoV-2 spike protein to host lectins and lung microbiota Alterations in gut microbiota of patients with COVID-19 during time of hospitalization One of the first studies to longitudinally characterize the bacterial gut microbiota in patients with varying degrees of COVID-19 severity Alterations in fecal fungal microbiome of patients with COVID-19 during time of hospitalization until discharge •• Longitudinal characterization of the fungal gut microbiome in patients with varying degrees of COVID-19 severity ACE2 and TMPRSS2 expression by clinical, HLA, immune, and microbial correlates across 34 human cancers and matched normal tissues: implications for SARS-CoV-2 COVID-19 Shows that high expression of human angiotensin-converting enzyme 2 relates to specific immunological and microbial profiles, suggesting that a possible link between the microbiome, inflammation and COVID-19 progression could exist Alterations of the gut microbiota in patients with coronavirus disease 2019 or H1N1 influenza Depicting SARS-CoV-2 faecal viral activity in association with gut microbiota composition in patients with COVID-19 Demonstrates that active replication of SARS-CoV-2 and transcriptional activity are correlated with high abundance of opportunistic bacteria, while low signatures of SARS-CoV-2 infection are associated to higher presence of short-chain fatty acid producing bacteria Lung microbiota in the acute respiratory disease: from coronavirus to metabolomics Immunodepletion with hypoxemia: a potential high risk subtype of coronavirus disease