Next Article in Journal
Vibrotactile Feedback for a Person with Transradial Amputation and Visual Loss: A Case Report
Previous Article in Journal
A Motorcycle Paramedic Increases the Survival Rate of Patients after OHCA
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

COVID-19 and Gastrointestinal Tract: From Pathophysiology to Clinical Manifestations

1
Gastroenterology Unit, Division of Gastroenterology, Hepatology, and Nutrition, Department of Life, Health and Environmental Sciences, University of L’Aquila, Piazzale Salvatore Tommasi 1, 67100 L’Aquila, Italy
2
Department of Medicine, Gastroenterology Division, Howard University College of Medicine, Washington, DC 20060, USA
*
Author to whom correspondence should be addressed.
Medicina 2023, 59(10), 1709; https://doi.org/10.3390/medicina59101709
Submission received: 3 August 2023 / Revised: 10 September 2023 / Accepted: 21 September 2023 / Published: 24 September 2023
(This article belongs to the Section Gastroenterology & Hepatology)

Abstract

:
Background: Since its first report in Wuhan, China, in December 2019, COVID-19 has become a pandemic, affecting millions of people worldwide. Although the virus primarily affects the respiratory tract, gastrointestinal symptoms are also common. The aim of this narrative review is to provide an overview of the pathophysiology and clinical manifestations of gastrointestinal COVID-19. Methods: We conducted a systematic electronic search of English literature up to January 2023 using Medline, Scopus, and the Cochrane Library, focusing on papers that analyzed the role of SARS-CoV-2 in the gastrointestinal tract. Results: Our review highlights that SARS-CoV-2 directly infects the gastrointestinal tract and can cause symptoms such as diarrhea, nausea/vomiting, abdominal pain, anorexia, loss of taste, and increased liver enzymes. These symptoms result from mucosal barrier damage, inflammation, and changes in the microbiota composition. The exact mechanism of how the virus overcomes the acid gastric environment and leads to the intestinal damage is still being studied. Conclusions: Although vaccination has increased the prevalence of less severe symptoms, the long-term interaction with SARS-CoV-2 remains a concern. Understanding the interplay between SARS-CoV-2 and the gastrointestinal tract is essential for future management of the virus.

1. Introduction

Since the first report in December 2019 in Wuhan, China, coronavirus disease 2019 (COVID-19) spread all over the world, causing more than 758 million cases and 6.85 million deaths [1].
The clinical course of Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2) infection ranges from asymptomatic to a rapidly progressing and life-threatening disease and is associated with a variety of symptoms [2].
Like other coronaviruses, SARS-CoV-2 infects the gastrointestinal tract, inducing nausea, vomiting, abdominal pain, and diarrhea [3,4,5].
Since the COVID-19 occurrence in late 2019, intense research efforts on an unprecedented scale have focused on the study of SARS-CoV-2 entry mechanisms and clinical presentations.
Prior to COVID-19, there were two short-lived pandemics—SARS-CoV-1 in 2002 and MERS in 2012. The first cases of SARS-CoV-1 were detected in China and quickly spread with a high lethality rate of 11%, resulting in 8422 reported cases and 916 deaths. Similarly, MERS emerged in Saudi Arabia with a high mortality rate of approximately 37% and was also traced back to bats. Both viruses caused similar symptoms such as fever, cough, dyspnea, and atypical pneumonia, as well as affecting the gastrointestinal tract with diarrhea being a common symptom. However, MERS had a higher prevalence of GI symptoms, mortality rate, and need for extreme treatment measures such as mechanical ventilation compared to SARS-CoV-1. Despite the potential for fecal–oral transmission, extrapulmonary symptoms were not given much attention due to the short-lived and localized nature of these pandemics [6,7].
The aim of this narrative review is to compile a selection of relevant papers to summarize the current evidence on the pathophysiology of gastrointestinal SARS-CoV-2 infection, fecal–oral route transmission, clinical manifestations, and the outcomes of patients with gastrointestinal symptoms.

2. Materials and Methods

A systematic electronic search of the English literature up to January 2023 was performed using Medline, EMBASE, Scopus, and the Cochrane Library. The search strategy used a combination of Medical Subject Headings (MeSH) and keywords as follows: COVID-19; SARS-CoV-2; gastrointestinal symptoms; intestinal symptoms; gastrointestinal infection; intestinal infection; intestinal replication; long COVID; post-acute COVID syndrome.
Five authors (F.V., N.C., S.M., S.F., E.S.) identified relevant articles by screening the abstracts. Additional studies were selected after a manual review of the reference list of the identified studies and review articles. Any discrepancy was resolved by consensus, referring to the original articles. Out of 4783 citations, 147 relevant articles were selected and included in the present narrative review.

3. Prevalence of Gastrointestinal Symptoms

Several studies reported gastrointestinal symptoms in patients affected by COVID-19. Their prevalence in adults is high, with diarrhea, nausea, and abdominal pain being the most frequent ones (16.5%, 9.7%, 4.5%, respectively) [8]. Anorexia or loss of appetite (1.6%), vomiting (1.5%), and loss of taste (1.3%) are less common. Increased liver enzymes are not rare (5.6%) (Table 1) [8]. Prevalences, however, are hardly comparable as different series do not report all main gastrointestinal symptoms. This likely results from the different weight attributed by authors to mild and/or infrequent symptoms, which were not invariably reported.
In the pediatric population, the incidence of gastrointestinal symptoms is higher compared to adults [9]. A recent meta-analysis reported a higher prevalence of nausea/vomiting (19.7%) and abdominal pain (20.3%) but not diarrhea (19.08%) [9]. However, wide variations have been described in different series [10].
The relationship between gastrointestinal symptoms, mortality, and more severe systemic disease presentation is debatable. Available evidence suggests that the overall presence of gastrointestinal symptoms is not associated with increased mortality rates (OR = 0.88; 95% CI 0.71–1.09; p = 0.23). The same applies to individual symptoms, such as diarrhea (p = 0.96), nausea/vomiting (p = 0.46), and abdominal pain (p = 0.3) [11].
A recent meta-analysis correlating the incidence of gastrointestinal symptoms with severe presentation in adults showed that abdominal pain (OR = 2.70, 95% CI 1.17–6.27, p = 0.02), but not diarrhea, nausea, or vomiting, is associated with aggressive disease [12].
In the pediatric population, the presence of diarrhea significantly correlates with more severe clinical course (OR 3.9, 95% CI: 1.80–8.73; p < 0.01). This is not the case for nausea/vomiting and abdominal pain [9].
The persistence of gastrointestinal symptoms for more than two weeks after discharge is also high, as 3.23% of patients reported nausea, 3.19% vomiting, and 4.12% prolonged diarrhea, while the persistence of abdominal pain was 1.68% [13].
Gastrointestinal bleeding (GIB) was reported in 2% of COVID-19 patients, respectively, 1% for upper GI and 1% for lower GI [14]. The most common causes were ulcers (25.3%), erosive/ulcerative diffuse damage (16.1%), and petechial/hemorrhagic gastropathy (9.2%) in upper gastrointestinal bleeding. Ischemic colitis was reported in one-third of patients with lower gastrointestinal bleeding [15]. Again, some differences have been reported in different cohorts [4]. The presentation with GIB however represents a negative prognostic factor, as reported by a recent systematic review comparing the outcomes of 808 COVID-19 patients with GIB and 18,179 non-GIB COVID-19 patients. The overall incidence of GIB was 0.06%, with GIB patients showing a higher death rate than non-GIB patients (25.4% vs. 16.4%, p < 0.001) [4].

4. Gastrointestinal Infection

Respiratory droplets are the main route of transmission of SARS-CoV-2 [16], but SARS-CoV-2 RNA was also detected in fecal samples [17,18,19,20].
Direct evidence of fecal–oral transmission is still lacking, but emerging evidence supports the hypothesis [21,22]. SARS-CoV-2 RNA has indeed been detected in anal swabs and stool samples in over 50% of infected patients [23,24,25]. RNA levels in the stools ranged from 102 to 105 copies/mL, but in several reports fecal shedding exceeded 107 copies/mL [23,26,27,28]. This is in line with nasopharyngeal fluids concentration (105–1011 copies/mL) [27,28]. SARS-CoV-2 concentration in stool samples peaks 2 to 3 weeks after symptom onset [25,29], and, as reported in a small German cohort, the RNA load in fecal specimens reflects what is found in sputum in 86% of cases (6 of 7 patients) [23]. Live SARS-CoV-2 was also observed in the feces of COVID-19 patients, confirming potential fecal–oral transmission [30]. However, despite being easily detected by electron microscopy [31], SARS-CoV-2 isolation from stools is difficult [32].
Recent evidence suggests that viral variants show different gastrointestinal infectivity. Reduced viral replication of Omicron BA.1 and BA.2 variants compared with the B.1.617.2/Delta variant was reported in studies carried out in organoids [33].
The persistence of the virus in stools is significantly longer (median 22 days, interquartile range 17–31 days) than in respiratory (18 days, 13–29 days; p = 0.02) and serum samples (16 days, 11–21 days; p < 0.001) [25]. Polymerase chain reaction detected viral RNA in fecal samples of patients with no detectable virus in respiratory tract specimens [19,25,34].
Several studies have shown the presence of SARS-CoV-2 in epithelial cells of both the small and large intestines, as demonstrated by intestinal biopsies [17,35,36]. Additionally, the transcription of subgenomic SARS-CoV-2 mRNA (sgmRNA) indicates active viral replication in the intestine, as sgmRNA is only transcribed in infected cells and not packaged into virions [23]. Intestinal organoids primarily secrete SARS-CoV-2 apically [37], which may explain viral excretion in feces.
Although evidence suggests that the gut is an active site of SARS-CoV-2 replication, it is still unclear whether the virus present in feces is directly infectious. While viral RNA may be shed in fecal specimens, the presence of viral ribonucleic acid does not necessarily imply the presence of live transmissible virus [38]. Therefore, while direct and indirect data support the hypothesis that SARS-CoV-2 actively infects human intestinal epithelial cells, further research is needed to determine whether the virus in feces is directly infectious.

5. SARS-CoV-2 Structure and Interaction with the Host

SARS-CoV-2 is a single-stranded β-coronavirus, with 29.9 kb RNA genome and an envelope with spikes on the surface (Figure 1) [39]. It shares up to 80% of the gene sequence with other pathogenic members of the coronavirus family, such as SARS-CoV-1 and Middle East Respiratory Syndrome coronavirus (MERS-CoV) [40].
Two-thirds of viral RNA, in the first open reading frame (ORF 1a/b), translates two polyproteins, encoding for 16 non-structural proteins (NSP). The remaining viral genome expresses accessory proteins, interfering with the host innate immune response, and structural proteins. The four essential structural proteins include the spike (S) glycoprotein, small envelope (E) protein, nucleocapsid (N) protein, and matrix (M) protein [41]. Proteins M, E, and S form the viral envelope [42]. The N protein is structurally bound to the viral RNA and is involved in viral replication [43], while the M protein plays a role in determining the shape of the viral envelope [43].
Open reading frames encoding nine accessory proteins (3a, 3b, 6, 7a, 7b, 8, 9b, 9c, and 10) and two polyproteins (pp1a and pp1ab) are also present in the genome [44].
Polyproteins pp1a and pp1ab, but not accessory proteins, are involved in viral replication [44].
SARS-CoV-2 interacts with target cells through envelope spike glycoprotein which binds the angiotensin-converting enzyme 2 (ACE2) receptor of the host (Figure 2) [41]. The binding affinity for the human ACE2 receptor is 10–20 times stronger for SARS-CoV-2 than for its predecessor SARS-CoV-1 [45]. After binding ACE2 receptors, the transmembrane serine protease 2 (TMPRSS2) mediates the cleavage of the spike glycoprotein, regulating the internalization of the virus into target cells [46]. The two subunits, S1 and S2, favor the binding of the virus to the cell and the fusion between the two cellular membranes [46].
Two proteolytic events are however needed to activate SARS-CoV-2. The first is the cut in the specific cleavage site between the S1 and S2 domains, which is recognized by various proteases, including TMPRSS2 [46] and furin [47,48]. The second cleavage site is within the S2 domain and allows the exposition of the fusion peptide, which enables membrane fusion. This second cleavage can be either performed by TMPRSS2 on the surface of the host cell or by lysosomal proteases, such as cathepsin L in the endolysosomes [49]. Following the entry into the cell, the virus is uncoated and replicates using the host replication system [42].

6. Route of Gastrointestinal Infection

The exact route of infection is still undefined, but the virus likely reaches the gut after being swallowed.
In vitro studies show that SARS-CoV-2, like other enveloped viruses, loses infectivity after 10 min incubation in gastric fluid [50]. Nonetheless, this route of transmission is possible as other viruses, such as influenza virus, despite being vulnerable to digestive juices, retain infectivity when protected by highly viscous mucus [51].
The glycosylation of the S protein is a further mechanism by which other coronaviruses survive the adverse milieu of the stomach and bile-salt-containing duodenal juice [52]. The mechanism may be shared by SARS-CoV-2.
Indirect evidence deriving from in vitro models of other coronaviruses suggests that fasting or fed state modulate infectivity. Indeed, MERS rapidly loses infectivity in simulated fasting state, low pH gastric fluid, but not after 2 h of exposure to fed-state condition [53].
It has also been hypothesized that chronic H. pylori infection leading to atrophic gastritis and intestinal metaplasia could facilitate SARS-CoV-2 intestinal infection by reducing stomach acidity [54,55]. H. pylori also increases the expression of ACE-2 receptors in the GI tract [56]. Clinically, a strong correlation between the occurrence of abdominal pain (19.4% vs. 2.6%, p = 0.007) or diarrhea (32.3% vs. 9.1%, p = 0.006) and H. pylori infection has been reported in COVID-19 patients [57].
Similarly, proton pump inhibitor-induced hypochloridria could favor SARS-CoV-2 intestinal infection [58], but available data are conflicting. Patients on PPI had significantly higher requirement for oxygen therapy, intensive care unit admission, and invasive ventilation than patients not taking PPI (fully adjusted OR (aOR): 2.39; 95% CI: 1.08–5.10) in a post hoc analysis from a nationwide Korean cohort [5]. Conversely, several meta-analyses did not confirm these early reports [59,60].

7. Gastrointestinal Tract–Virus Interaction

Lungs are the primary route of SARS-CoV-2 infection, but ACE2 receptors, an 805 amino acids-, type I cell-surface glycoprotein [61], are highly expressed also on the brush border of the enterocytes [62,63]. The ACE2 receptor is detected by immunofluorescent staining also in the glandular cells of the stomach and colon [17]. Viral RNA has been detected also in the esophageal mucosa, but the lack of viral nucleocapsid protein staining suggests a low viral load [17]. This is in keeping with low ACE2 expression in squamous esophageal epithelial cells. ACE2 receptors are minimally expressed by enteroendocrine cells, Paneth cells, and goblet cells [61,64,65,66].
Under physiological conditions, ACE2 receptors in the gastrointestinal tract are associated with the amino acid carrier B0AT1, which regulates the homeostasis of tryptophan, and thus stimulates the production of mechanistic target of rapamycin (mTOR)-dependent antimicrobial peptides from Paneth cells [67,68].
Besides ACE2 receptors, other molecules may play a role in SARS-CoV-2 infection. It has been demonstrated that TMPRSS2 is highly expressed in the gastrointestinal tract [50], not only in enterocytes [50] but also in intestinal goblet [69] and Paneth cells (Figure 2) [70,71]. Other serine proteases of the same family, such as TMPRSS4, are also highly expressed in mature enterocytes and potentially favor SARS-CoV-2 infection [50]. An additive effect of the two enzymes has been documented, suggesting synergic effect resulting from distinct cellular and subcellular localization of the two proteases [50].
The protease furin, widely present in the stomach, small bowel, and colon [72], also enables the S protein to separate into two pinching structures [73]. Although furin significantly increases the cleavage of the S protein, promoting SARS-CoV-2 infectivity and spread, its presence is not essential [74].
In addition to proteases, other alternative entry molecules, such as neuropilin-1 (NRP1), have been identified [75]. The expression of NRP1 in the small bowel has been reported on both human biopsies and organoids [76,77]. Following protease cleavage of the S protein into S1 and S2, a polybasic Arg-Arg-Ala-Arg carboxyl-terminal sequence on S1 binds NRP1 [75,78]. The role of NRP1 is less clear than ACE2 receptors, but the protein might mediate SARS-CoV-2 infection in ACE2-negative cells [77] and possibly explain different disease behavior in adults and children [76].
Recent studies suggest a possible interaction between the SARS-CoV-2 S protein and the cluster of differentiation (CD) 147 binding site [79], possibly through cyclophilin A-mediated regulation of ACE2 receptors [80], but conflicting results have also been published [81].
A small Italian case series [82] suggested that VEGF through CD147 may trigger gastrointestinal ischemia in SARS-CoV-2, as reported in other conditions [83]. However, validation in larger series is needed.
Protease-mediated membrane fusion represents the usual SARS-CoV-2 entry route in the host cell. An alternative option has recently been advocated, consisting of endocytosis resulting from SARS-CoV-2 binding to ACE2 receptors followed by S protein cleavage by cathepsin L [49] expressed in a variety of tissues including the gastrointestinal tract [84,85].

8. Gastrointestinal Damage, Inflammation, and Symptoms

SARS-CoV-2 is responsible for direct and indirect damage of the gastrointestinal system and symptoms (Figure 3).
SARS-CoV-2 induces syncytia formation in human enteroids [50]. Independently of other viral components, the S protein induces cell fusion in vitro [50]. Syncytia formation is cytopathic, resulting in loss of integrity of the intestinal barrier, and favors cell-to-cell viral spread and immune-response evasion [50]. Epithelial damage is also related to reduced expression of tight junction marker genes, such as ZO-3 and CLDN1 [86].
In vivo intragastric inoculation with SARS-CoV-2 reduces cell proliferation and increases apoptosis of intestinal epithelial and goblet cells in an animal model of SARS-CoV-2 infection [87]. Other in vitro studies on Caco-2 cells confirmed viral replication but did not report cytopathic effects [88]. It should also be considered that data from in vitro models using tumoral cell lines with different protein expression need confirmation in human biopsies.
Duodenal biopsies collected during upper endoscopy in a small case series of COVID-19 patients with GI symptoms were characterized by villous blunting and increased intraepithelial lymphocytes [89]. This supports the view that intestinal damage is mediated by the immune response, more than by direct viral damage, and may explain the lack of cytopathic effect reported in some models.
The enhanced migration of immune cells to the intestinal mucosa during SARS-CoV-2 infection is in line with a study using mass cytometry [36]. Reduced dendritic cells in the lamina propria were also reported [36] but not in studies using mass cytometry in postmortem tissue samples [90].
Infection likely triggers innate immune response in the intestine through the recognition of pathogen-associated molecular patterns (PAMPs), as reported in the lung, leading to recruitment of immune cells and favoring adaptative response [91,92].
SARS-CoV-2 infection increases the expression of proinflammatory genes such as IL-1b, IL-6, CCL2, CCL3, CCL5, and CXCL10 in human small intestinal epithelial cells derived from pluripotent stem cells [86]. The S1 spike protein stimulates IL-6 and IL-8 secretion in CACO-2 human intestinal epithelial cells [93]. Some studies also reported an increased release of intestinal IL-18 [94,95]. Interestingly, elevated IL-18 levels correlate with disease severity [95,96].
In vitro, intestinal epithelial cells were reported to produce type III interferon (IFN) in response to SARS-CoV-2 infection, leading to less effective viral replication [97]. Other studies on IFN-III had opposite findings [37].
SARS-CoV-2 infection induces the inflammatory response in the gut [98], in turn increasing the likelihood of intestinal symptoms. COVID-19 patients with diarrhea have higher concentrations of fecal calprotectin (FC) compared to patients without diarrhea (123.2 ± 58.8 vs. 17.3 ± 4.8, p < 0.001) [98]. Interestingly, FC levels significantly correlate with serum IL-6 (p < 0.001) but not with C-reactive protein [98].
The pathogenic role of SARS-CoV-2 on enterocytes is indirectly supported by plasma citrulline concentrations [99]. The molecule is almost exclusively produced by enterocytes and is not incorporated into proteins, thus representing a biomarker of small bowel enterocyte mass and function [100]. The accepted citrulline cutoff level for the diagnosis of the short bowel syndrome (<20 μmol/L) has been found in 61.5% of patients from a small cohort, and 15.4% had concentrations below 10 μmol/L [99], suggesting severe intestinal damage. Correlation of low citrulline levels with digestive symptoms (62.5% vs. 20%, p = 0.05) and PCR (84.5 vs. 13.5 mg/L, p = 0.03) was also present [99].
High concentrations of serotonin (5-HT) were documented in COVID-19 patients with diarrhea compared to those who did not present with the symptom, or healthy controls [101]. The role of 5-HT in regulating GI motility and inflammation is well known [102], but its involvement in SARS-CoV-2-associated diarrhea is undocumented.
Intestinal inflammation is associated with severe systemic disease as fecal calprotectin levels were increased in patients with abnormal chest X-ray findings [103].
Active intestinal inflammation in vivo in COVID-19 patients is supported by high fecal levels of the pro-inflammatory IL-8 and low fecal levels of anti-inflammatory IL-10 compared to controls [104]. Fecal IL-23 levels were high in patients with severe COVID-19 disease [104].
SARS-CoV-2-specific IgA antibodies were high in patients with severe disease [104]. Their clinical importance is unclear, as a longitudinal study showed that IgA and IgM antibodies rapidly decay, but not IgG antibodies, which are detectable up to 105 days after symptom onset [105].
The role of adaptative immune response in GI symptoms has been reported by several studies. COVID-19 patients with diarrhea have increased CD3+ and higher CD4+ T cell counts compared to patients without diarrhea. As CD8+ T cell counts were lower in the diarrhea group, the CD4/CD8 ratio was increased. High counts of CD19+ B cells and low CD16+CD56+ natural killer (NK) cells were also reported in diarrhea [106].

9. SARS-CoV-2 and Gut Microbiota Alteration

Gut microbiota changes have been linked to gastrointestinal symptoms. Several studies investigated gut microbiota through 16S rRNA gene-amplified sequencing in COVID-19 patients [107]. Antibiotic-naïve patients with COVID-19 showed higher concentrations of opportunistic pathogens, including Actinomyces viscosus, Clostridium hathewayi, and Bacteroides nordii, compared to controls (Figure 4). A recent meta-analysis of 16 studies confirmed a significant reduction in alpha diversity in COVID-19 patients compared to controls (standardized mean difference, SMD = −0.78; 95% CI, −1.25 to −0.31) [107]. Changes persisted after recovery (SMD = −1.14; 95% CI, −1.60 to −0.68) [107]. Depletion of beneficial symbiotic bacterial strains was observed in patients on antibiotics [108].
Interestingly, a small study comparing microbiota dysregulation in patients affected by H1N1 influenza A and COVID-19 identified specific microbial signatures in the two diseases, with H1N1 displaying lower diversity and different overall microbial composition [109].
Gut microbiota alterations influence the metabolism of several compounds and negatively affect infection rates and symptoms. Reduced gut microbiome metabolism of tryptophan was reported in patients with COVID-19 and GI symptoms [110]. Tryptophan is absorbed by the intestinal epithelial cell B0AT1/ACE2 transporter and indirectly regulates the expression of antimicrobial peptides [111]. It may be anticipated that SARS-CoV-2-binding intestinal ACE2 receptors might reduce tryptophan absorption and ultimately modify the microbiota [112].
Analysis of the bacterial function in 66 antibiotics-naïve COVID-19 patients and 70 controls showed that patients with severe disease are characterized by significant (p < 0.001) impairment of short-chain fatty acid (SCFA) and L-isoleucine biosynthesis that persists over 30 days after recovery [113]. The reduction in SCFA and L-isoleucine significantly correlated with disease severity and levels of inflammation-related proteins such as CXCL-10, NT-proB-type natriuretic peptide, and C-reactive protein (p < 0.05). This reflects a functional modification of intestinal microbiome. However, in vitro studies aimed at the evaluation of the anti-inflammatory effects of butyrate led to contrasting results [114,115]. In vivo butyrate supplementation studies to improve symptoms have not been carried out.
Some of the role of microbiota on SARS-CoV-2 outcomes is indirectly supported by the finding that the genus Collinsella, producing ursodeoxycholate and other secondary bile acids, was negatively correlated with mortality in a Japanese cohort [116]. The underlying rationale resides in the reduced farnesoid X receptor signaling with consequent downregulation of ACE2 receptor transcription, which decreases susceptibility to SARS-CoV-2 in vitro, in vivo, and in human lungs and livers perfused ex situ [117].

10. Chronic GI Tract Diseases and SARS-CoV-2

The mechanisms by which SARS-CoV-2 affects the gastrointestinal tract of healthy subjects are still largely unclear, more so considering that the host–virus bidirectional relation may modify both the course of infection and the preexisting disease.
ACE2 and TMPRSS2 are overexpressed in inflammatory bowel disease (IBD) patients [118,119,120]. However, cytoplasmic and membrane ACE2 are significantly higher in ulcerative colitis (UC) compared to healthy controls. In Crohn’s disease (CD), the difference is observed only in the membrane [118]. Moreover, colonic ACE2 expression is lower in CD than in UC (p < 0.0001) [121].
Increased expression of ACE2 and TMPRSS2 in inflamed versus non-inflamed colonic and ileal segments has been reported by some authors [122] but not by others [118], casting doubt on the modulation of receptor expression induced by active disease.
Moreover, circulating components of the alternative renin–angiotensin system, ACE2 and angiotensin (1–7), were increased in a small series of patients with IBD [123], possibly playing a protective role against SARS-CoV-2 infection [124].
Potentially clinically relevant differences have been reported for other proteins involved in the infection. TMPRSS2 is overexpressed in IBD [118,122,125], while furin levels are significantly lower in colonic specimens of UC patients versus controls [126]. The clinical relevance of these findings is at best debatable as clinical databases, including SECURE-IBD [127], Dutch [128], and Danish [129] series, did not report increased COVID-19 severity in IBD patients. Furthermore, the incidence rate of COVID-19 is not increased in IBD patients undergoing immunomodulant therapy [130].
Differences in relevant epithelial protein expression are present in other chronic diseases. ACE2 is overexpressed in the stomach of patients with chronic gastritis [54,55,131] and underexpressed in eosinophilic esophagitis and gastroenteritis [132]. However, to our knowledge, no association with outcome has been reported [133,134,135].
Overall, poor general conditions, resulting from concomitant disease, are by far more relevant in worsening clinical course and outcome than the expression of receptors or the existence of immune-related illnesses [136].

11. Gastrointestinal Symptoms and Long COVID

COVID-19 is associated with long-term gastrointestinal symptoms, persisting in up to 8.4% of patients at 3 months and in 6.6% at 6 months [137]. Schmulson proposed new criteria for defining post-COVID-19 functional gastrointestinal disorders (FGID) as follows. Patients should not meet the criteria for FGID before COVID-19, symptoms should fulfill Rome IV criteria and persist after the resolution of acute SARS-CoV-2 infection [138].
The alterations underlying the development of post-infective FGID are still undefined but likely reside in persistent subclinical inflammation, increased intestinal permeability, and microbiota changes. It is known that irritable bowel syndrome (IBS) may develop after an acute infection (e.g., Campylobacter, Shigella, Salmonella, Giardia, Noroviruses) [139,140,141], and the same might apply to SARS-CoV-2.
A 6-month prospective study in post-acute COVID-19 syndrome reported high levels of Ruminococcus gnavus and Bacteroides vulgatus and low levels of Faecalibacterium prausnitzii. Butyrate-producing bacteria showed significant inverse correlations with occurrence of the syndrome [142], but it is unclear whether modifications of the intestinal flora precede or are caused by long COVID.
As far as gut permeability is concerned, the concentration markers, such as the fatty acid binding protein 2, were higher in COVID-19 patients compared to controls (p = 0.0013) [143]. Zonulins and cadherins are also likely involved [144,145].
Incomplete viral clearance or persistence of viral antigens is an additional mechanism triggering symptoms, and it appears that the S1 protein alone is sufficient to stimulate the production of inflammatory cytokines in vitro [93]. Patients with persistent viral shedding in the stool were more likely to report nausea (aOR = 1.61, CI 95% = 1.09–2.39), vomiting (3.20, 1.11–9.21), and abdominal pain (2.05, 1.09–3.86) but not diarrhea (1.10, 0.63–1.91) [146]. Moreover, the presence of SARS-CoV-2 antigens in the colon was documented 257 days following diagnosis, in the absence of ongoing fecal shedding [147].
Patients who have received the bivalent vaccine for COVID-19, which combines two different vaccine types, may experience different outcomes compared to those who have received the monovalent vaccine, which contains only one type of vaccine. While both vaccines have been shown to be effective in preventing COVID-19 infection, post-bivalent vaccine patients may have a higher level of protection against certain strains of the virus [148]. Additionally, post-bivalent vaccine patients may experience a different side-effect profile than monovalent vaccine patients due to the different types of vaccines used in the combination. However, further research is needed to fully understand the differences between these two vaccine types and their impact on patients’ health. Ultimately, regardless of the type of vaccine received, vaccination remains a critical tool in the fight against the COVID-19 pandemic. In addition to the potential differences in vaccine effectiveness and side-effect profiles, there may also be differences in the gastroenterology symptoms experienced by patients who have received bivalent versus monovalent vaccines. Some studies have suggested that COVID-19 infection can lead to gastrointestinal symptoms such as nausea, vomiting, and diarrhea. While the vaccines themselves do not typically cause these symptoms, it is possible that post-bivalent vaccine patients may experience a different gastrointestinal symptom profile due to the different types of vaccines used in the combination. However, more research is needed to fully understand the potential differences in gastrointestinal symptoms between these two vaccine types. Regardless, it is to know that long-hauler symptoms are less in people infected with Omicron who have received the bivalent vaccine [149,150,151].

12. Conclusions

SARS-CoV-2 infection dramatically changed the worldwide health scenario. Following the initial impact characterized by acute severe infection and high mortality rate, the virus is now becoming endemic due to massive vaccination campaigns and disease-induced antibodies. We will thus be facing a long-term interaction with the SARS-CoV-2 virus, with increased prevalence of less aggressive symptoms.
Epidemiological and clinical data indicate that in the first phase of epidemics, gastrointestinal symptoms were frequent, occurring in up to one-third of patients. The figure will increase as the virus becomes endemic.
The rapid global spread of this new, highly aggressive virus prompted fast sharing of biomedical knowledge. This often resulted in redundant information and the publication of review articles sometimes reporting unconfirmed or questionable data.
The present narrative review provides an updated overview of SARS-CoV-2 gastrointestinal infection and symptoms and the pathophysiological mechanism of intestinal damage. This may prove of some use for physicians treating SARS-CoV-2 patients in clinical practice. However, most hard information derives from preclinical/in vitro studies, whereas clinical and epidemiological studies are mainly retrospective, underpowered, lack reliable protocols, and the length of follow-up is necessarily short.
Thus, major questions regarding the interaction of SARS-CoV-2 and the GI tract are still unanswered. It is unclear how the virus survives in the gastric environment, which cell types are primarily infected in vivo, and to which extent preexisting intestinal disease modifies susceptibility, clinical course, and prognosis. Similarly, the long-term effects of post-acute COVID-19 syndrome are still to be determined, but it is likely that, as after other infections, post-infective functional disorders may develop. The results of well-designed ongoing studies are needed to answer these questions and provide reliable insight into the GI effects of SARS-CoV2 infection. The bivalent COVID-19 vaccine, combining two different vaccine types, offers a higher level of protection against new virus strains compared to the monovalent vaccine. It is presently undefined whether the two vaccine types or Omicron-derived new virus strains are associated with changing prevalence or severity of side effects or differences in the profile of gastrointestinal symptoms.
In conclusion, gastrointestinal symptoms associated with SARS-CoV-2 infection are usually mild and self-limiting. Symptomatic treatment may be needed in case of nausea and vomiting. Rehydration associated to mild diet may prove useful in some patients with diarrhea, while loperamide and other anti-diarrheal drugs are likely best avoided as in other gastrointestinal infections.
Clinical trials using dietary modification, probiotics, prebiotics, and fecal microbiota transplantation are underway to determine their effectiveness in the treatment of acute COVID-19 and possibly enhance the effectiveness of SARS-CoV-2 vaccines [152,153,154,155,156,157,158]. Only long-term clinical data and future research shall provide better insight into the complex bidirectional interaction between the virus and the alimentary tract in healthy patients and those with preexisting gastrointestinal diseases.

Author Contributions

F.V. designed the study and wrote the manuscript; F.V., N.C., S.M., S.F. and E.S. performed the literature review and prepared Tables and Figures; A.V. reviewed the manuscript and provided critical comments; H.A. and G.L. supervised, wrote, and critically reviewed the manuscript. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. World Health Organization. Coronavirus Disease (COVID-19). Available online: https://covid19.who.int/ (accessed on 4 March 2023).
  2. Zhou, F.; Yu, T.; Du, R.; Fan, G.; Liu, Y.; Liu, Z.; Xiang, J.; Wang, Y.; Song, B.; Gu, X.; et al. Clinical course and risk factors for mortality of adult inpatients with COVID-19 in Wuhan, China: A retrospective cohort study. Lancet 2020, 395, 1054–1062. [Google Scholar] [CrossRef] [PubMed]
  3. Huang, C.; Wang, Y.; Li, X.; Ren, L.; Zhao, J.; Hu, Y.; Zhang, L.; Fan, G.; Xu, J.; Gu, X.; et al. Clinical features of patients infected with 2019 novel coronavirus in Wuhan, China. Lancet 2020, 395, 497–506. [Google Scholar] [CrossRef] [PubMed]
  4. Ashktorab, H.; Russo, T.; Oskrochi, G.; Latella, G.; Massironi, S.; Luca, M.; Chirumamilla, L.G.; Laiyemo, A.O.; Brim, H. Clinical and Endoscopic Outcomes in Coronavirus Disease-2019 Patients with Gastrointestinal Bleeding. Gastro Hep. Adv. 2022, 1, 487–499. [Google Scholar] [CrossRef] [PubMed]
  5. Pizuorno, A.; Brim, H.; Ashktorab, H. Gastrointestinal manifestations and SARS-CoV-2 infection. Curr. Opin. Pharmacol. 2021, 61, 114–119. [Google Scholar] [CrossRef] [PubMed]
  6. Dyall, J.; Gross, R.; Kindrachuk, J.; Johnson, R.F.; Olinger, G.G., Jr.; Hensley, L.E.; Frieman, M.B.; Jahrling, P.B. Middle East Respiratory Syndrome and Severe Acute Respiratory Syndrome: Current Therapeutic Options and Potential Targets for Novel Therapies. Drugs 2017, 77, 1935–1966. [Google Scholar] [CrossRef]
  7. de Wit, E.; van Doremalen, N.; Falzarano, D.; Munster, V.J. SARS and MERS: Recent insights into emerging coronaviruses. Nat. Rev. Microbiol. 2016, 14, 523–534. [Google Scholar] [CrossRef]
  8. Shehab, M.; Alrashed, F.; Shuaibi, S.; Alajmi, D.; Barkun, A. Gastroenterological and hepatic manifestations of patients with COVID-19, prevalence, mortality by country, and intensive care admission rate: Systematic review and meta-analysis. BMJ Open Gastroenterol. 2021, 8, e000571. [Google Scholar] [CrossRef]
  9. Bolia, R.; Dhanesh Goel, A.; Badkur, M.; Jain, V. Gastrointestinal Manifestations of Pediatric Coronavirus Disease and Their Relationship with a Severe Clinical Course: A Systematic Review and Meta-analysis. J. Trop. Pediatr. 2021, 67, fmab051. [Google Scholar] [CrossRef]
  10. Russo, T.; Pizuorno, A.; Oskrochi, G.; Latella, G.; Massironi, S.; Schettino, M.; Aghemo, A.; Pugliese, N.; Brim, H.; Ashktorab, H. Gastrointestinal Manifestations, Clinical Characteristics and Outcomes of COVID-19 in Adult and Pediatric Patients. SOJ Microbiol. Infect. Dis. 2021, 8, 109. [Google Scholar] [CrossRef]
  11. Wang, Y.; Li, Y.; Zhang, Y.; Liu, Y.; Liu, Y. Are gastrointestinal symptoms associated with higher risk of Mortality in COVID-19 patients? A systematic review and meta-analysis. BMC Gastroenterol. 2022, 22, 106. [Google Scholar] [CrossRef]
  12. Hayashi, Y.; Wagatsuma, K.; Nojima, M.; Yamakawa, T.; Ichimiya, T.; Yokoyama, Y.; Kazama, T.; Hirayama, D.; Nakas, H. The characteristics of gastrointestinal symptoms in patients with severe COVID-19: A systematic review and meta-analysis. J. Gastroenterol. 2021, 56, 409–420. [Google Scholar] [CrossRef] [PubMed]
  13. Yusuf, F.; Fahriani, M.; Mamada, S.S.; Frediansyah, A.; Abubakar, A.; Maghfirah, D.; Fajar, J.K.; Maliga, H.A.; Ilmawan, M.; Emran, T.B.; et al. Global prevalence of prolonged gastrointestinal symptoms in COVID-19 survivors and potential pathogenesis: A systematic review and meta-analysis. F1000Research 2021, 10, 301. [Google Scholar] [CrossRef]
  14. Marasco, G.; Maida, M.; Morreale, G.C.; Licata, M.; Renzulli, M.; Cremon, C.; Stanghellini, V.; Barbara, G. Gastrointestinal Bleeding in COVID-19 Patients: A Systematic Review with Meta-Analysis. Can. J. Gastroenterol. Hepatol. 2021, 2021, 2534975. [Google Scholar] [PubMed]
  15. Vanella, G.; Capurso, G.; Burti, C.; Fanti, L.; Ricciardiello, L.; Souza Lino, A.; Boskoski, I.; Bronswijk, M.; Tyberg, A.; Krishna Kumar Nair, G.; et al. Gastrointestinal mucosal damage in patients with COVID-19 undergoing endoscopy: An international multicentre study. BMJ Open Gastroenterol. 2021, 8, e000578. [Google Scholar] [CrossRef]
  16. van Doremalen, N.; Bushmaker, T.; Morris, D.H.; Holbrook, M.G.; Gamble, A.; Williamson, B.N.; Tamin, A.; Harcourt, J.L.; Thornburg, N.J.; Gerber, S.I.; et al. Aerosol and surface stability of SARS-CoV-2 as compared with SARS-CoV-1. N. Engl. J. Med. 2020, 382, 1564–1567. [Google Scholar] [CrossRef] [PubMed]
  17. Xiao, F.; Tang, M.; Zheng, X.; Liu, Y.; Li, X.; Shan, H. Evidence for Gastrointestinal Infection of SARS-CoV-2. Gastroenterology 2020, 158, 1831–1833. [Google Scholar] [CrossRef]
  18. Xing, Y.H.; Ni, W.; Wu, Q.; Li, W.J.; Li, G.J.; Wang, W.D.; Tong, J.N.; Song, X.F.; Wing-Kin Wong, G.; Xing, Q.S. Prolonged viral shedding in feces of pediatric patients with coronavirus disease 2019. J. Microbiol. Immunol. Infect. 2020, 53, 473–480. [Google Scholar] [CrossRef]
  19. Wu, Y.; Guo, C.; Tang, L.; Hong, Z.; Zhou, J.; Dong, X.; Yin, H.; Xiao, Q.; Tang, Y.; Qu, X.; et al. Prolonged presence of SARS-CoV-2 viral RNA in faecal samples. Lancet Gastroenterol. Hepatol. 2020, 5, 434–435. [Google Scholar] [CrossRef]
  20. Chen, Y.; Chen, L.; Deng, Q.; Wu, K.; Ni, L.; Yang, Y.; Liu, B.; Wang, W.; Wei, C.; Yang, J.; et al. The presence of SARS-CoV-2 RNA in the feces of COVID-19 patients. J. Med. Virol. 2020, 92, 833–840. [Google Scholar] [CrossRef]
  21. Kang, M.; Wei, J.; Yuan, J.; Guo, J.; Zhang, Y.; Hang, J.; Qu, Y.; Qian, H.; Zhuang, Y.; Chen, X.; et al. Probable Evidence of Fecal Aerosol Transmission of SARS-CoV-2 in a High-Rise Building. Ann. Intern. Med. 2020, 173, 974–980. [Google Scholar] [CrossRef]
  22. Qian, Q.; Fan, L.; Liu, W.; Li, J.; Yue, J.; Wang, M.; Ke, X.; Yin, Y.; Chen, Q.; Jiang, C. Direct evidence of active SARS-CoV-2 replication in the intestine. Clin. Infect. Dis. 2021, 73, 361–366. [Google Scholar] [CrossRef] [PubMed]
  23. Wölfel, R.; Corman, V.M.; Guggemos, W.; Seilmaier, M.; Zange, S.; Müller, M.A.; Niemeyer, D.; Jones, T.C.; Vollmar, P.; Rothe, C. Virological assessment of hospitalized patients with COVID-19. Nature 2020, 581, 465–469. [Google Scholar] [CrossRef]
  24. Xu, Y.; Li, X.; Zhu, B.; Liang, H.; Fang, C.; Gong, Y.; Guo, Q.; Sun, X.; Zhao, D.; Shen, J.; et al. Characteristics of pediatric SARS-CoV-2 infection and potential evidence for persistent fecal viral shedding. Nat. Med. 2020, 26, 502–505. [Google Scholar] [CrossRef]
  25. Zheng, S.; Fan, J.; Yu, F.; Feng, B.; Lou, B.; Zou, Q.; Xie, G.; Lin, S.; Wang, R.; Yang, X.; et al. Viral load dynamics and disease severity in patients infected with SARS-CoV-2 in Zhejiang province, China, January–March 2020: Retrospective cohort study. BMJ 2020, 369, m1443. [Google Scholar] [CrossRef] [PubMed]
  26. Jones, D.L.; Baluja, M.Q.; Graham, D.W.; Corbishley, A.; McDonald, J.E.; Malham, S.K.; Hillary, L.S.; Connor, T.R.; Gaze, W.H.; Moura, I.B.; et al. Shedding of SARS-CoV-2 in feces and urine and its potential role in person-to-person transmission and the environment-based spread of COVID-19. Sci. Total Environ. 2020, 749, 141364. [Google Scholar] [CrossRef] [PubMed]
  27. Han, M.S.; Seong, M.W.; Heo, E.Y.; Park, J.H.; Kim, N.; Shin, S.; Cho, S.I.; Park, S.S.; Choi, E.H. Sequential analysis of viral load in a neonate and her mother infected with SARS-CoV-2. Clin. Infect. Dis. 2020, 71, 2236–2239. [Google Scholar] [CrossRef]
  28. Pan, Y.; Zhang, D.; Yang, P.; Poon, L.M.; Wang, Q. Viral load of SARSCoV-2 in clinical samples. Lancet Infect. Dis. 2020, 20, 411–412. [Google Scholar] [CrossRef]
  29. Walsh, K.A.; Jordan, K.; Clyne, B.; Rohde, D.; Drummond, L.; Byrne, P.; Ahern, S.; Carty, P.G.; O’Brien, K.K.; O’Murchu, E.; et al. SARS-CoV-2 detection, viral load and infectivity over the course of an infection. J. Infect. 2020, 81, 357–371. [Google Scholar] [CrossRef]
  30. Xiao, F.; Sun, J.; Xu, Y.; Li, F.; Huang, X.; Li, H.; Zhao, J.; Huang, J.; Zhao, J. Infectious SARS-CoV-2 in Feces of Patient with Severe COVID-19. Emerg. Infect. Dis. 2020, 26, 1920–1922. [Google Scholar] [CrossRef]
  31. Wang, W.; Xu, Y.; Gao, R.; Lu, R.; Han, K.; Wu, G.; Tan, W. Detection of SARS-CoV-2 in Different Types of Clinical Specimens. JAMA 2020, 323, 1843–1844. [Google Scholar] [CrossRef]
  32. Natarajan, A.; Han, A.; Zlitni, S.; Brooks, E.F.; Vance, S.E.; Wolfe, M.; Singh, U.; Jagannathan, P.; Pinsky, B.A.; Boehm, A.; et al. Standardized preservation, extraction and quantification techniques for detection of fecal SARS-CoV-2 RNA. Nat. Commun. 2021, 12, 5753. [Google Scholar] [CrossRef] [PubMed]
  33. Miyakawa, K.; Machida, M.; Kawasaki, T.; Nishi, M.; Akutsu, H.; Ryo, A. Reduced replication efficacy of severe acute respiratory syndrome coronavirus 2 omicron variant in “mini-gut” organoids. Gastroenterology 2022, 163, 514–516. [Google Scholar] [CrossRef] [PubMed]
  34. Cheung, K.S.; Hung, I.F.N.; Chan, P.P.Y.; Lung, K.C.; Tso, E.; Liu, R.; Ng, Y.Y.; Chu, M.Y.; Chung, T.W.H.; Tam, A.R.; et al. Gastrointestinal manifestations of SARS-CoV-2 infection and virus load in fecal samples from the Hong Kong cohort and systematic review and meta-analysis. Gastroenterology 2020, 159, 81–95. [Google Scholar] [CrossRef] [PubMed]
  35. von Stillfried, S.; Villwock, S.; Bülow, R.D.; Djudjaj, S.; Buhl, E.M.; Maurer, A.; Ortiz-Brüchle, N.; Celec, P.; Klinkhammer, B.M.; Wong, D.W.L.; et al. SARS-CoV-2 RNA screening in routine pathology specimens. Microb. Biotechnol. 2021, 14, 1627–1641. [Google Scholar] [CrossRef] [PubMed]
  36. Livanos, A.E.; Jha, D.; Cossarini, F.; Gonzalez-Reiche, A.S.; Tokuyama, M.; Aydillo, T.; Parigi, T.L.; Ladinsky, M.S.; Ramos, I.; Dunleavy, K.; et al. Intestinal Host Response to SARS-CoV-2 Infection and COVID-19 Outcomes in Patients with Gastrointestinal Symptoms. Gastroenterology 2021, 160, 2435–2450. [Google Scholar] [CrossRef]
  37. Lamers, M.M.; Beumer, J.; van der Vaart, J.; Knoops, K.; Puschhof, J.; Breugem, T.I.; Ravelli, R.B.G.; van Schayck, J.P.; Mykytyn, A.Z.; Duimel, H.Q.; et al. SARS-CoV-2 productively infects human gut enterocytes. Science 2020, 369, 50–54. [Google Scholar] [CrossRef]
  38. Yang, D.; Perbtani, Y.B.; Loeb, J.; Liu, N.; Draganov, P.V.; Estores, D.E.; Lauzardo, M.; Maurelli, A.; Lednicky, J.A.; Morris, J.G. Detection of SARS-CoV-2 in the gastrointestinal tract among patients with negative nasopharyngeal COVID-19 testing prior to endoscopy. Endosc. Int. Open 2021, 9, E1276–E1282. [Google Scholar] [CrossRef]
  39. Fehr, A.R.; Perlman, S. Coronaviruses: An overview of their replication and pathogenesis. In Methods in Molecular Biology; Humana Press: New York, NY, USA, 2015; Volume 1282, pp. 1–23. [Google Scholar]
  40. Lu, R.; Zhao, X.; Li, J.; Niu, P.; Yang, B.; Wu, H.; Wang, W.; Song, H.; Huang, B.; Zhu, N.; et al. Genomic characterisation and epidemiology of 2019 novel coronavirus: Implications for virus origins and receptor binding. Lancet 2020, 395, 565–574. [Google Scholar] [CrossRef]
  41. Guo, Y.R.; Cao, Q.D.; Hong, Z.S.; Tan, Y.Y.; Chen, S.D.; Jin, H.J.; Tan, K.S.; Wang, D.Y.; Yan, Y. The origin, transmission and clinical therapies on coronavirus disease 2019 (COVID-19) outbreak. An update on the status. Mil. Med. Res. 2020, 7, 11. [Google Scholar] [CrossRef]
  42. Arya, R.; Kumari, S.; Pandey, B.; Mistry, H.; Bihani, S.C.; Das, A.; Prashar, V.; Gupta, G.D.; Panicker, L.; Kumar, M. Structural insights into SARS-CoV-2 proteins. J. Mol. Biol. 2021, 433, 166725. [Google Scholar] [CrossRef]
  43. Schoeman, D.; Fielding, B.C. Coronavirus envelope protein: Current knowledge. Virol. J. 2019, 16, e69. [Google Scholar] [CrossRef] [PubMed]
  44. Naqvi, A.A.T.; Fatima, K.; Mohammad, T.; Fatima, U.; Singh, I.K.; Singh, A.; Atif, S.M.; Hariprasad, G.; Hasan, G.M.; Hassan, M.I. Insights into SARS-CoV-2 genome, structure, evolution, pathogenesis and therapies: Structural genomics approach. Biochim. Biophys. Acta Mol. Basis Dis. 2020, 1866, 165878. [Google Scholar] [CrossRef] [PubMed]
  45. Wrapp, D.; Wang, N.; Corbett, K.S.; Goldsmith, J.A.; Hsieh, C.L.; Abiona, O.; Graham, B.S.; McLellan, J.S. Cryo-EM structure of the 2019-nCoV spike in the prefusion conformation. Science 2020, 367, 1260–1263. [Google Scholar] [CrossRef] [PubMed]
  46. Hoffmann, M.; Kleine-Weber, H.; Schroeder, S.; Krüger, N.; Herrler, T.; Erichsen, S.; Schiergens, T.S.; Herrler, G.; Wu, N.H.; Nitsche, A.; et al. SARS-CoV-2 cell entry depends on ACE2 and TMPRSS2 and is blocked by a clinically proven protease inhibitor. Cell 2020, 181, 271–280. [Google Scholar] [CrossRef] [PubMed]
  47. Walls, A.C.; Park, Y.J.; Tortorici, M.A.; Wall, A.; McGuire, A.T.; Veesler, D. Structure, Function, and Antigenicity of the SARS-CoV-2 Spike Glycoprotein. Cell 2020, 181, 281–292.e6. [Google Scholar] [CrossRef]
  48. Shang, J.; Wan, Y.; Luo, C.; Ye, G.; Geng, Q.; Auerbach, A.; Li, F. Cell entry mechanisms of SARS-CoV-2. Proc. Natl. Acad. Sci. USA 2020, 117, 11727–11734. [Google Scholar] [CrossRef]
  49. Ou, X.; Liu, Y.; Lei, X.; Li, P.; Mi, D.; Ren, L.; Guo, L.; Guo, R.; Chen, T.; Hu, J.; et al. Characterization of spike glycoprotein of SARS-CoV-2 on virus entry and its immune cross-reactivity with SARS-CoV. Nat. Commun. 2020, 11, 1620. [Google Scholar] [CrossRef]
  50. Zang, R.; Gomez Castro, M.F.; McCune, B.T.; Zeng, Q.; Rothlauf, P.W.; Sonnek, N.M.; Liu, Z.; Brulois, K.F.; Wang, X.; Greenberg, H.B.; et al. TMPRSS2 and TMPRSS4 promote SARS-CoV-2 infection of human small intestinal enterocytes. Sci. Immunol. 2020, 5, eabc3582. [Google Scholar] [CrossRef]
  51. Hirose, R.; Nakaya, T.; Naito, Y.; Daidoji, T.; Watanabe, Y.; Yasuda, H.; Konishi, H.; Itoh, Y. Mechanism of human influenza virus RNA persistence and virion survival in feces: Mucus protects virions from acid and digestive juices. J. Infect. Dis. 2017, 216, 105–109. [Google Scholar] [CrossRef]
  52. Holmes, K.V. Enteric infections with coronaviruses and toroviruses. In Novartis Foundation Symposium; John Wiley: Chichester, UK, 2001. [Google Scholar]
  53. Zhou, J.; Li, C.; Zhao, G.; Chu, H.; Wang, D.; Yan, H.H.; Poon, V.K.; Wen, L.; Wong, B.H.; Zhao, X.; et al. Human intestinal tract serves as an alternative infection route for Middle East respiratory syndrome coronavirus. Sci. Adv. 2017, 3, eaao4966. [Google Scholar] [CrossRef]
  54. Simsek, C.; Erul, E.; Balaban, H.Y. Role of gastrointestinal system on transmission and pathogenesis of SARS-CoV-2. World J. Clin. Cases 2021, 9, 5427–5434. [Google Scholar] [CrossRef] [PubMed]
  55. Zhang, M.; Feng, C.; Zhang, X.; Hu, S.; Zhang, Y.; Min, M.; Liu, B.; Ying, X.; Liu, Y. Susceptibility Factors of Stomach for SARS-CoV-2 and Treatment Implication of Mucosal Protective Agent in COVID-19. Front. Med. 2021, 7, 597967. [Google Scholar] [CrossRef] [PubMed]
  56. Sugimoto, M.; Yamaoka, Y.; Shirai, N.; Furuta, T. Role of renin-angiotensin system in gastric oncogenesis. J. Gastroenterol. Hepatol. 2012, 27, 442–451. [Google Scholar] [CrossRef] [PubMed]
  57. Balamtekin, N.; Artuk, C.; Arslan, M.; Gülşen, M. The Effect of Helicobacter pylori on the Presentation and Clinical Course of Coronavirus Disease 2019 Infection. J. Pediatr. Gastroenterol. Nutr. 2021, 72, 511–513. [Google Scholar] [CrossRef] [PubMed]
  58. Lee, S.W.; Ha, E.K.; Yeniova, A.Ö.; Moon, S.Y.; Kim, S.Y.; Koh, H.Y.; Yang, J.M.; Jeong, S.J.; Moon, S.J.; Cho, J.Y.; et al. Severe clinical outcomes of COVID-19 associated with proton pump inhibitors: A nationwide cohort study with propensity score matching. Gut 2021, 70, 76–84. [Google Scholar] [CrossRef]
  59. Zippi, M.; Fiorino, S.; Budriesi, R.; Micucci, M.; Corazza, I.; Pica, R.; de Biase, D.; Gallo, C.G.; Hong, W. Paradoxical relationship between proton pump inhibitors and COVID-19: A systematic review and meta-analysis. World J. Clin. Cases 2021, 9, 2763–2777. [Google Scholar] [CrossRef]
  60. Israelsen, S.B.; Ernst, M.T.; Lundh, A.; Lundbo, L.F.; Sandholdt, H.; Hallas, J.; Benfield, T. Proton Pump Inhibitor Use Is Not Strongly Associated With SARS-CoV-2 Related Outcomes: A Nationwide Study and Meta-analysis. Clin. Gastroenterol. Hepatol. 2021, 19, 1845–1854. [Google Scholar] [CrossRef]
  61. Hamming, I.; Timens, W.; Bulthuis, M.; Lely, A.T.; Navis, G.J.; van Goor, H. Tissue distribution of ACE2 protein, the functional receptor for SARS Coronavirus. J. Pathol. 2004, 203, 631–637. [Google Scholar] [CrossRef]
  62. Qi, F.; Qian, S.; Zhang, S.; Zhang, Z. Single cell RNA sequencing of 13 human tissues identify cell types and receptors of human coronaviruses. Biochem. Biophys. Res. Commun. 2020, 526, 135–140. [Google Scholar] [CrossRef]
  63. Zou, X.; Chen, K.; Zou, J.; Han, P.; Hao, J.; Han, Z. Single-cell RNA-seq data analysis on the receptor ACE2 expression reveals the potential risk of different human organs vulnerable to 2019-nCoV infection. Front. Med. 2020, 14, 185–192. [Google Scholar] [CrossRef]
  64. Vuille-dit-Bille, R.N.; Camargo, S.M.; Emmenegger, L.; Sasse, T.; Kummer, E.; Jando, J.; Hamie, Q.M.; Meier, C.F.; Hunziker, S.; Forras-Kaufmann, Z.; et al. Human intestine luminal ACE2 and amino acid transporter expression increased by ACE-inhibitors. Amino Acids 2015, 47, 693–705. [Google Scholar] [CrossRef]
  65. Sungnak, W.; Huang, N.; Bécavin, C.; Berg, M. HCA Lung Biological Network. SARS-CoV-2 Entry Genes Are Most Highly Expressed in Nasal Goblet and Ciliated Cells within Human Airways. arXiv 2020, arXiv:2003.06122v1. [Google Scholar]
  66. Zhang, H.; Li, H.B.; Lyu, J.R.; Lei, X.M.; Li, W.; Wu, G.; Lyu, J.; Dai, Z.M. Specific ACE2 expression in small intestinal enterocytes may cause gastrointestinal symptoms and injury after 2019-nCoV infection. Int. J. Infect. Dis. 2020, 96, 19–24. [Google Scholar] [CrossRef]
  67. Camargo, S.M.; Singer, D.; Makrides, V.; Huggel, K.; Pos, K.M.; Wagner, C.A.; Kuba, K.; Danilczyk, U.; Skovby, F.; Kleta, R.; et al. Tissue-specific amino acid transporter partners ACE2 and collectrin differentially interact with hartnup mutations. Gastroenterology 2009, 136, 872–882. [Google Scholar] [CrossRef]
  68. Gao, N.; Dou, X.; Yin, T.; Yang, Y.; Yan, D.; Ma, Z.; Bi, C.; Shan, A. Tryptophan Promotes Intestinal Immune Defense through Calcium-Sensing Receptor (CaSR)-Dependent Metabolic Pathways. J. Agric. Food Chem. 2021, 69, 13460–13473. [Google Scholar] [CrossRef]
  69. Qi, J.; Zhou, Y.; Hua, J.; Zhang, L.; Bian, J.; Liu, B.; Zhao, Z.; Jin, S. The scRNA-seq Expression Profiling of the Receptor ACE2 and the Cellular Protease TMPRSS2 Reveals Human Organs Susceptible to SARS-CoV-2 Infection. Int. J. Environ. Res. Public Health 2021, 18, 284. [Google Scholar] [CrossRef]
  70. Pearce, S.C.; Suntornsaratoon, P.; Kishida, K.; Al-Jawadi, A.; Guardia, J.; Nadler, I.; Flores, J.; Shiarella, R.; Auvinen, M.; Yu, S.; et al. Expression of SARS-CoV-2 entry factors, electrolyte, and mineral transporters in different mouse intestinal epithelial cell types. Physiol. Rep. 2021, 9, e15061. [Google Scholar] [CrossRef]
  71. Singh, M.; Bansal, V.; Feschotte, C. A Single-Cell RNA Expression Map of Human Coronavirus Entry Factors. Cell Rep. 2020, 32, 108175. [Google Scholar] [CrossRef]
  72. Zhou, L.; Niu, Z.; Jiang, X.; Zhang, Z.; Zheng, Y.; Wang, Z.; Zhu, Y.; Gao, L.; Huang, H.; Wang, X.; et al. SARS-CoV-2 Targets by the pscRNA Profiling of ACE2, TMPRSS2 and Furin Proteases. iScience 2020, 23, 101744. [Google Scholar] [CrossRef]
  73. Johnson, B.A.; Xie, X.; Bailey, A.L.; Kalveram, B.; Lokugamage, K.G.; Muruato, A.; Zou, J.; Zhang, X.; Juelich, T.; Smith, J.K.; et al. Loss of furin cleavage site attenuates SARS-CoV-2 pathogenesis. Nature 2021, 591, 293–299. [Google Scholar] [CrossRef]
  74. Papa, G.; Mallery, D.L.; Albecka, A.; Welch, L.G.; Cattin-Ortolá, J.; Luptak, J.; Paul, D.; McMahon, H.T.; Goodfellow, I.G.; Carteret, 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]
  75. Daly, J.L.; Simonetti, B.; Klein, K.; Chen, K.E.; Williamson, M.K.; Antón-Plágaro, C.; Shoemark, D.K.; Simón-Gracia, L.; Bauer, M.; Hollandi, R.; et al. Neuropilin-1 is a host factor for SARS-CoV-2 infection. Science 2020, 370, 861–865. [Google Scholar] [CrossRef] [PubMed]
  76. Berni Canani, R.; Comegna, M.; Paparo, L.; Cernera, G.; Bruno, C.; Strisciuglio, C.; Zollo, I.; Gravina, A.G.; Miele, E.; Cantone, E.; et al. Age-Related Differences in the Expression of Most Relevant Mediators of SARS-CoV-2 Infection in Human Respiratory and Gastrointestinal Tract. Front. Pediatr. 2021, 9, 697390. [Google Scholar] [CrossRef] [PubMed]
  77. Zhang, F.; Li, W.; Feng, J.; Ramos da Silva, S.; Ju, E.; Zhang, H.; Chang, Y.; Moore, P.S.; Guo, H.; Gao, S.J. SARS-CoV-2 pseudovirus infectivity and expression of viral entry-related factors ACE2, TMPRSS2, Kim-1, and NRP-1 in human cells from the respiratory, urinary, digestive, reproductive, and immune systems. J. Med. Virol. 2021, 93, 6671–6685. [Google Scholar] [CrossRef] [PubMed]
  78. Cantuti-Castelvetri, L.; Ojha, R.; Pedro, L.D.; Djannatian, M.; Franz, J.; Kuivanen, S.; van der Meer, F.; Kallio, K.; Kaya, T.; Anastasina, M.; et al. Neuropilin-1 facilitates SARS-CoV-2 cell entry and infectivity. Science 2020, 370, 856–860. [Google Scholar] [CrossRef]
  79. Helal, M.A.; Shouman, S.; Abdelwaly, A.; Elmehrath, A.O.; Essawy, M.; Sayed, S.M.; Saleh, A.H.; El-Badri, N. Molecular basis of the potential interaction of SARS-CoV-2 spike protein to CD147 in COVID-19 associated-lymphopenia. J. Biomol. Struct. Dyn. 2022, 40, 1109–1119. [Google Scholar] [CrossRef]
  80. Fenizia, C.; Galbiati, S.; Vanetti, C.; Vago, R.; Clerici, M.; Tacchetti, C.; Daniele, T. SARS-CoV-2 entry: At the crossroads of CD147 and ACE2. Cells 2021, 10, 1434. [Google Scholar] [CrossRef]
  81. Shilts, J.; Crozier, T.W.M.; Greenwood, E.J.D.; Lehner, P.J.; Wright, G.J. No evidence for basigin/CD147 as a direct SARS-CoV-2 spike binding receptor. Sci. Rep. 2021, 11, 413. [Google Scholar] [CrossRef]
  82. Bortolotti, D.; Simioni, C.; Neri, L.M.; Rizzo, R.; Semprini, C.M.; Occhionorelli, S.; Laface, I.; Sanz, J.M.; Schiuma, G.; Rizzo, S.; et al. Relevance of VEGF and CD147 in different SARS-CoV-2 positive digestive tracts characterized by thrombotic damage. FASEB J. 2021, 35, e21969. [Google Scholar] [CrossRef]
  83. Wang, H.; Ye, J.; Liu, R.; Chen, G.; Zhao, J.; Huang, L.; Yang, F.; Li, M.; Zhang, S.; Xie, J.; et al. Clinical significance of CD147 in children with inflammatory bowel disease. Biomed. Res. Int. 2020, 2020, 7647181. [Google Scholar] [CrossRef]
  84. Tamhane, T.; Arampatzidou, M.; Gerganova, V.; Tacke, M.; Illukkumbura, R.; Dauth, S.; Schaschke, N.; Peters, C.; Reinheckel, T.; Brix, K. The activity and localization patterns of cathepsins B and X in cells of the mouse gastrointestinal tract differ along its length. Biol. Chem. 2014, 395, 1201–1219. [Google Scholar] [CrossRef]
  85. Tamhane, T.; Lllukkumbura, R.; Lu, S.; Maelandsmo, G.M.; Haugen, M.H.; Brix, K. Nuclear cathepsin L activity is required for cell cycle progression of colorectal carcinoma cells. Biochimie 2016, 122, 208–218. [Google Scholar] [CrossRef] [PubMed]
  86. Yamada, S.; Noda, T.; Okabe, K.; Yanagida, S.; Nishida, M.; Kanda, Y. SARS-CoV-2 induces barrier damage and inflammatory responses in the human iPSC-derived intestinal epithelium. J. Pharmacol. Sci. 2022, 149, 139–146. [Google Scholar] [CrossRef] [PubMed]
  87. Jiao, L.; Li, H.; Xu, J.; Yang, M.; Ma, C.; Li, J.; Zhao, S.; Wang, H.; Yang, Y.; Yu, W.; et al. The Gastrointestinal Tract Is an Alternative Route for SARS-CoV-2 Infection in a Nonhuman Primate Model. Gastroenterology 2021, 160, 1647–1661. [Google Scholar] [CrossRef] [PubMed]
  88. Zupin, L.; Fontana, F.; Clemente, L.; Ruscio, M.; Ricci, G.; Crovella, S. Effect of Short Time of SARS-CoV-2 Infection in Caco-2 Cells. Viruses 2022, 14, 704. [Google Scholar] [CrossRef]
  89. Lehmann, M.; Allers, K.; Heldt, C.; Meinhardt, J.; Schmidt, F.; Rodriguez-Sillke, Y.; Kunkel, D.; Schumann, M.; Böttcher, C.; Stahl-Hennig, C.; et al. Human small intestinal infection by SARS-CoV-2 is characterized by a mucosal infiltration with activated CD8 + T cells. Mucosal Immunol. 2021, 14, 1381–1392. [Google Scholar] [CrossRef]
  90. Wang, C.; Xu, J.; Wang, S.; Pan, S.; Zhang, J.; Han, Y.; Huang, M.; Wu, D.; Yang, Q.; Yang, X.; et al. Imaging Mass Cytometric Analysis of Postmortem Tissues Reveals Dysregulated Immune Cell and Cytokine Responses in Multiple Organs of COVID-19 Patients. Front. Microbiol. 2020, 11, 600989. [Google Scholar] [CrossRef]
  91. Lee, S.; Channappanavar, R.; Kanneganti, T.D. Coronaviruses: Innate Immunity, Inflammasome Activation, Inflammatory Cell Death, and Cytokines. Trends Immunol. 2020, 41, 1083–1099. [Google Scholar] [CrossRef]
  92. Lowery, S.A.; Sariol, A.; Perlman, S. Innate immune and inflammatory responses to SARS-CoV-2: Implications for COVID-19. Cell Host Microbe 2021, 29, 1052–1062. [Google Scholar] [CrossRef]
  93. Forsyth, C.B.; Zhang, L.; Bhushan, A.; Swanson, B.; Zhang, L.; Mamede, J.I.; Voigt, R.M.; Shaikh, M.; Engen, P.A.; Keshavarzian, A. The SARS-CoV-2 S1 Spike Protein Promotes MAPK and NF-kB Activation in Human Lung Cells and Inflammatory Cytokine Production in Human Lung and Intestinal Epithelial Cells. Microorganisms 2022, 10, 1996. [Google Scholar] [CrossRef]
  94. Zhang, L.; Zhang, Y.; Wang, R.; Liu, X.; Zhao, J.; Tsuda, M.; Li, Y. SARSCoV-2 infection of intestinal epithelia cells sensed by RIG-I and DHX-15 evokes innate immune response and immune cross-talk. Front. Cell Infect. Microbiol. 2023, 12, 1035711. [Google Scholar] [CrossRef] [PubMed]
  95. Tao, W.; Zhang, G.; Wang, X.; Guo, M.; Zeng, W.; Xu, Z.; Cao, D.; Pan, A.; Wang, Y.; Zhang, K.; et al. Analysis of the intestinal microbiota in COVID-19 patients and its correlation with the inflammatory factor IL-18. Med. Microecol. 2020, 5, 100023. [Google Scholar] [CrossRef] [PubMed]
  96. Nasser, S.M.T.; Rana, A.A.; Doffinger, R.; Kafizas, A.; Khan, T.A.; Nasser, S. Elevated free interleukin-18 associated with severity and mortality in prospective cohort study of 206 hospitalised COVID-19 patients. Intensive Care Med. Exp. 2023, 11, 9. [Google Scholar] [CrossRef] [PubMed]
  97. Stanifer, M.L.; Kee, C.; Cortese, M.; Zumaran, C.M.; Triana, S.; Mukenhirn, M.; Kraeusslich, H.G.; Alexandrov, T.; Bartenschlager, R.; Boulant, S. Critical Role of Type III Interferon in Controlling SARS-CoV-2 Infection in Human Intestinal Epithelial Cells. Cell Rep. 2020, 32, 107863. [Google Scholar] [CrossRef]
  98. Effenberger, M.; Grabherr, F.; Mayr, L.; Schwaerzler, J.; Nairz, M.; Seifert, M.; Hilbe, R.; Seiwald, S.; Scholl-Buergi, S.; Fritsche, G.; et al. Faecal calprotectin indicates intestinal inflammation in COVID-19. Gut 2020, 69, 1543–1544. [Google Scholar] [CrossRef]
  99. Uzzan, M.; Soudan, D.; Peoc’h, K.; Weiss, E.; Corcos, O.; Treton, X. Patients with COVID-19 present with low plasma citrulline concentrations that associate with systemic inflammation and gastrointestinal symptoms. Dig. Liver Dis. 2020, 52, 1104–1105. [Google Scholar] [CrossRef]
  100. Crenn, P.; Messing, B.; Cynober, L. Citrulline as a biomarker of intestinal failure due to enterocyte mass reduction. Clin. Nutr. 2008, 27, 328–339. [Google Scholar] [CrossRef]
  101. Ha, S.; Jin, B.; Clemmensen, B.; Park, P.; Mahboob, S.; Gladwill, V.; Lovely, F.M.; Gottfried-Blackmore, A.; Habtezion, A.; Verma, S.; et al. Serotonin is elevated in COVID-19-associated diarrhoea. Gut 2021, 70, 2015–2017. [Google Scholar] [CrossRef]
  102. Gershon, M.D.; Tack, J. The serotonin signaling system: From basic understanding to drug development for functional Gi disorders. Gastroenterology 2007, 132, 397–414. [Google Scholar] [CrossRef]
  103. Ojetti, V.; Saviano, A.; Covino, M.; Acampora, N.; Troiani, E.; Franceschi, F.; Gemelli against COVID-19 Group. COVID-19 and intestinal inflammation: Role of fecal calprotectin. Dig. Liver Dis. 2020, 52, 1231–1233. [Google Scholar] [CrossRef]
  104. Britton, G.J.; Chen-Liaw, A.; Cossarini, F.; Livanos, A.E.; Spindler, M.P.; Plitt, T.; Eggers, J.; Mogno, I.; Gonzalez-Reiche, A.S.; Siu, S.; et al. Limited intestinal inflammation despite diarrhea, fecal viral RNA and SARS-CoV-2-specific IgA in patients with acute COVID-19. Sci. Rep. 2021, 11, 13308. [Google Scholar] [CrossRef]
  105. Isho, B.; Abe, K.T.; Zuo, M.; Jamal, A.J.; Rathod, B.; Wang, J.H.; Li, Z.; Chao, G.; Rojas, O.L.; Bang, Y.M.; et al. Persistence of serum and saliva antibody responses to SARS-CoV-2 spike antigens in COVID-19 patients. Sci. Immunol. 2020, 5, eabe5511. [Google Scholar] [CrossRef] [PubMed]
  106. Wang, X.; Wei, J.; Zhu, R.; Chen, L.; Ding, F.; Zhou, R.; Ge, L.; Xiao, J.; Zhao, Q. Contribution of CD4+ T cell-mediated inflammation to diarrhea in patients with COVID-19. Int. J. Infect. Dis. 2022, 120, 1–11. [Google Scholar] [CrossRef] [PubMed]
  107. Cheng, X.; Zhang, Y.; Li, Y.; Wu, Q.; Wu, J.; Park, S.K.; Guo, C.; Lu, J. Meta-analysis of 16S rRNA microbial data identified alterations of the gut microbiota in COVID-19 patients during the acute and recovery phases. BMC Microbiol. 2022, 22, 274. [Google Scholar] [CrossRef]
  108. Zuo, T.; Zhang, F.; Lui, G.C.Y.; Yeoh, Y.K.; Li, A.Y.L.; Zhan, H.; Wan, Y.; Chung, A.C.K.; Cheung, C.P.; Chen, N.; et al. Alterations in Gut Microbiota of Patients with COVID-19 During Time of Hospitalization. Gastroenterology 2020, 159, 944–955.e8. [Google Scholar] [CrossRef] [PubMed]
  109. Gu, S.; Chen, Y.; Wu, Z.; Chen, Y.; Gao, H.; Lv, L.; Guo, F.; Zhang, X.; Luo, R.; Huang, C.; et al. Alterations of the Gut Microbiota in Patients With Coronavirus Disease 2019 or H1N1 Influenza. Clin. Infect. Dis. 2020, 71, 2669–2678. [Google Scholar] [CrossRef]
  110. Blackett, J.W.; Sun, Y.; Purpura, L.; Margolis, K.G.; Elkind, M.S.V.; O’Byrne, S.; Wainberg, M.; Abrams, J.A.; Wang, H.H.; Chang, L.; et al. Decreased Gut Microbiome Tryptophan Metabolism and Serotonergic Signaling in Patients with Persistent Mental Health and Gastrointestinal Symptoms After COVID-19. Clin. Transl. Gastroenterol. 2022, 13, e00524. [Google Scholar] [CrossRef]
  111. Hashimoto, T.; Perlot, T.; Rehman, A.; Trichereau, J.; Ishiguro, H.; Paolino, M.; Sigl, V.; Hanada, T.; Hanada, R.; Lipinski, S.; et al. ACE2 links amino acid malnutrition to microbial ecology and intestinal inflammation. Nature 2012, 487, 477–481. [Google Scholar] [CrossRef]
  112. Ye, Q.; Wang, B.; Zhang, T.; Xu, J.; Shang, S. The mechanism and treatment of gastrointestinal symptoms in patients with COVID-19. Am. J. Physiol. Gastrointest. Liver Physiol. 2020, 319, G245–G252. [Google Scholar] [CrossRef]
  113. Zhang, F.; Wan, Y.; Zuo, T.; Yeoh, Y.K.; Liu, Q.; Zhang, L.; Zhan, H.; Lu, W.; Xu, W.; Lui, G.C.Y.; et al. Prolonged Impairment of Short-Chain Fatty Acid and L-Isoleucine Biosynthesis in Gut Microbiome in Patients with COVID-19. Gastroenterology 2022, 162, 548–561.e4. [Google Scholar] [CrossRef]
  114. Li, J.; Richards, E.M.; Handberg, E.M.; Pepine, C.J.; Raizada, M.K. Butyrate regulates COVID-19-relevant genes in gut epithelial organoids from normotensive rats. Hypertension 2021, 77, e13–e16. [Google Scholar] [CrossRef] [PubMed]
  115. Pascoal, L.B.; Rodrigues, P.B.; Genaro, L.M.; Gomes, A.B.D.S.P.; Toledo-Teixeira, D.A.; Parise, P.L.; Bispo-Dos-Santos, K.; Simeoni, C.L.; Guimarães, P.V.; Buscaratti, L.I.; et al. Microbiota-derived short-chain fatty acids do not interfere with SARS-CoV-2 infection of human colonic samples. Gut Microbes 2021, 13, 1–9. [Google Scholar] [CrossRef]
  116. Hirayama, M.; Nishiwaki, H.; Hamaguchi, T.; Ito, M.; Ueyama, J.; Maeda, T.; Kashihara, K.; Tsuboi, Y.; Ohno, K. Intestinal Collinsella may mitigate infection and exacerbation of COVID-19 by producing ursodeoxycholate. PLoS ONE 2021, 16, e0260451. [Google Scholar] [CrossRef] [PubMed]
  117. Brevini, T.; Maes, M.; Webb, G.J.; John, B.V.; Fuchs, C.D.; Buescher, G.; Wang, L.; Griffiths, C.; Brown, M.L.; Scott, W.E., 3rd; et al. FXR inhibition may protect from SARS-CoV-2 infection by reducing ACE2. Nature 2023, 615, 134–142. [Google Scholar] [CrossRef] [PubMed]
  118. McAllister, M.J.; Kirkwood, K.; Chuah, S.C.; Thompson, E.J.; Cartwright, J.A.; Russell, C.D.; Dorward, D.A.; Lucas, C.D.; Ho, G.T. Intestinal Protein Characterisation of SARS-CoV-2 Entry Molecules ACE2 and TMPRSS2 in Inflammatory Bowel Disease (IBD) and Fatal COVID-19 Infection. Inflammation 2022, 45, 567–572. [Google Scholar] [CrossRef] [PubMed]
  119. Jablaoui, A.; Kriaa, A.; Mkaouar, H.; Akermi, N.; Soussou, S.; Wysocka, M.; Wołoszyn, D.; Amouri, A.; Gargouri, A.; Maguin, E.; et al. Fecal Serine Protease Profiling in Inflammatory Bowel Diseases. Front. Cell Infect. Microbiol. 2020, 10, 21. [Google Scholar] [CrossRef]
  120. Potdar, A.A.; Dube, S.; Naito, T.; Li, K.; Botwin, G.; Haritunians, T.; Li, D.; Casero, D.; Yang, S.; Bilsborough, J.; et al. Altered Intestinal ACE2 Levels Are Associated with Inflammation, Severe Disease, and Response to Anti-Cytokine Therapy in Inflammatory Bowel Disease. Gastroenterology 2021, 160, 809–822. [Google Scholar] [CrossRef]
  121. Potdar, A.A.; Dube, S.; Naito, T.; Botwin, G.; Haritunians, T.; Li, D.; Yang, S.; Bilsborough, J.; Denson, L.A.; Daly, M.; et al. Reduced expression of COVID-19 host receptor, ACE2 is associated with small bowel inflammation, more severe disease, and response to anti-TNF therapy in Crohn’s disease. medRxiv 2020. [Google Scholar]
  122. Nowak, J.K.; Lindstrøm, J.C.; Kalla, R.; Ricanek, P.; Halfvarson, J.; Satsangi, J. Age, Inflammation, and Disease Location Are Critical Determinants of Intestinal Expression of SARS-CoV-2 Receptor ACE2 and TMPRSS2 in Inflammatory Bowel Disease. Gastroenterology 2020, 159, 1151–1154. [Google Scholar] [CrossRef]
  123. Garg, M.; Burrell, L.M.; Velkoska, E.; Griggs, K.; Angus, P.W.; Gibson, P.R.; Lubel, J.S. Upregulation of circulating components of the alternative renin-angiotensin system in inflammatory bowel disease: A pilot study. J. Renin Angiotensin Aldosterone Syst. 2015, 16, 559–569. [Google Scholar] [CrossRef]
  124. Krishnamurthy, S.; Lockey, R.F.; Kolliputi, N. Soluble ACE2 as a potential therapy for COVID-19. Am. J. Physiol. Cell Physiol. 2021, 320, C279–C281. [Google Scholar] [CrossRef] [PubMed]
  125. Bangma, A.; Voskuil, M.D.; Weersma, R.K. TNFα-Antagonist Use and Mucosal Inflammation Are Associated with Increased Intestinal Expression of SARS-CoV-2 Host Protease TMPRSS2 in Patients with Inflammatory Bowel Disease. Gastroenterology 2021, 160, 2621–2622. [Google Scholar] [CrossRef] [PubMed]
  126. Dong, S.; Lu, Y.; Peng, G.; Li, J.; Li, W.; Li, M.; Wang, H.; Liu, L.; Zhao, Q. Furin inhibits epithelial cell injury and alleviates experimental colitis by activating the Nrf2-Gpx4 signaling pathway. Dig. Liver Dis. 2021, 53, 1276–1285. [Google Scholar] [CrossRef] [PubMed]
  127. Queiroz, N.S.F.; Martins, C.A.; Quaresma, A.B.; Hino, A.A.F.; Steinwurz, F.; Ungaro, R.C.; Kotze, P.G. COVID-19 outcomes in patients with inflammatory bowel diseases in Latin America: Results from SECURE-IBD registry. J. Gastroenterol. Hepatol. 2021, 36, 3033–3040. [Google Scholar] [CrossRef]
  128. Derikx, L.A.A.P.; Lantinga, M.A.; de Jong, D.J.; van Dop, W.A.; Creemers, R.H.; Römkens, T.E.H.; Jansen, J.M.; Mahmmod, N.; West, R.L.; Tan, A.C.I.T.L.; et al. Clinical Outcomes of COVID-19 in Patients with Inflammatory Bowel Disease: A Nationwide Cohort Study. J. Crohns Colitis 2021, 15, 529–539. [Google Scholar] [CrossRef]
  129. Attauabi, M.; Poulsen, A.; Theede, K.; Pedersen, N.; Larsen, L.; Jess, T.; Rosager Hansen, M.; Verner-Andersen, M.K.; Haderslev, K.V.; Berg Lødrup, A.; et al. Prevalence and Outcomes of COVID-19 Among Patients with Inflammatory Bowel Disease-A Danish Prospective Population-based Cohort Study. J. Crohns Colitis 2021, 15, 540–550. [Google Scholar] [CrossRef]
  130. Alrashed, F.; Battat, R.; Abdullah, I.; Charabaty, A.; Shehab, M. Impact of medical therapies for inflammatory bowel disease on the severity of COVID-19: A systematic review and meta-analysis. BMJ Open Gastroenterol. 2021, 8, e000774. [Google Scholar] [CrossRef]
  131. Xu, J.; Chu, M.; Zhong, F.; Tan, X.; Tang, G.; Mai, J.; Lai, N.; Guan, C.; Liang, Y.; Liao, G. Digestive symptoms of COVID-19 and expression of ACE2 in digestive tract organs. Cell Death Discov. 2020, 6, 76. [Google Scholar] [CrossRef]
  132. Chiang, A.W.T.; Duong, L.D.; Shoda, T.; Nhu, Q.M.; Ruffner, M.; Hara, T.; Aaron, B.; Joplin, E.; Manresa, M.C.; Abonia, J.P.; et al. Type 2 Immunity and Age Modify Gene Expression of Coronavirus-induced Disease 2019 Receptors in Eosinophilic Gastrointestinal Disorders. J. Pediatr. Gastroenterol. Nutr. 2021, 72, 718–722. [Google Scholar] [CrossRef]
  133. Li, J.; Tian, A.; Yang, D.; Zhang, M.; Chen, L.; Wen, J.; Chen, P. Celiac Disease and the Susceptibility of COVID-19 and the Risk of Severe COVID-19: A Mendelian Randomization Study. Clin. Transl. Gastroenterol. 2022, 13, e00480. [Google Scholar] [CrossRef]
  134. Zevit, N.; Chehade, M.; Leung, J.; Marderfeld, L.; Dellon, E.S. Eosinophilic Esophagitis Patients Are Not at Increased Risk of Severe COVID-19: A Report from a Global Registry. J. Allergy Clin. Immunol. Pract. 2022, 10, 143–149. [Google Scholar] [CrossRef] [PubMed]
  135. Santacroce, G.; Lenti, M.V.; Aronico, N.; Miceli, E.; Lovati, E.; Lucotti, P.C.; Coppola, L.; Gentile, A.; Latorre, M.A.; Di Terlizzi, F.; et al. Impact of COVID-19 in immunosuppressive drug-naïve autoimmune disorders: Autoimmune gastritis, celiac disease, type 1 diabetes, and autoimmune thyroid disease. Pediatr. Allergy Immunol. 2022, 33, 105–107. [Google Scholar] [CrossRef] [PubMed]
  136. Treskova-Schwarzbach, M.; Haas, L.; Reda, S.; Pilic, A.; Borodova, A.; Karimi, K.; Koch, J.; Nygren, T.; Scholz, S.; Schönfeld, V.; et al. Pre-existing health conditions and severe COVID-19 outcomes: An umbrella review approach and meta-analysis of global evidence. BMC Med. 2021, 19, 212. [Google Scholar] [CrossRef] [PubMed]
  137. Golla, R.; Vuyyuru, S.; Kante, B.; Kumar, P.; Thomas, D.M.; Makharia, G.; Kedia, S.; Ahuja, V. Long-term Gastrointestinal Sequelae Following COVID-19: A Prospective Follow-up Cohort Study. Clin. Gastroenterol. Hepatol. 2023, 21, 789–796. [Google Scholar] [CrossRef]
  138. Schmulson, M.; Ghoshal, U.C.; Barbara, G. Managing the inevitable surge of post–COVID-19 functional gastrointestinal disorders. Am. J. Gastroenterol. 2021, 116, 4–7. [Google Scholar] [CrossRef] [PubMed]
  139. Spiller, R.C. Estimating the importance of infection in IBS. Am. J. Gastroenterol. 2003, 98, 238–241. [Google Scholar] [CrossRef]
  140. Porter, C.K.; Gormley, R.; Tribble, D.R.; Cash, B.D.; Riddle, M.S. The Incidence and gastrointestinal infectious risk of functional gastrointestinal disorders in a healthy US adult population. Am. J. Gastroenterol. 2011, 106, 130–138. [Google Scholar] [CrossRef]
  141. Neal, K.R.; Barker, L.; Spiller, R.C. Prognosis in post infective irritable bowel syndrome: A 6 year follow up study. Gut 2002, 51, 410–413. [Google Scholar] [CrossRef]
  142. Liu, Q.; Mak, J.W.Y.; Su, Q.; Yeoh, Y.K.; Lui, G.C.; Ng, S.S.S.; Zhang, F.; Li, A.Y.L.; Lu, W.; Hui, D.S.; et al. Gut microbiota dynamics in a prospective cohort of patients with post-acute COVID-19 syndrome. Gut 2022, 71, 544–552. [Google Scholar] [CrossRef]
  143. Prasad, R.; Patton, M.J.; Floyd, J.L.; Fortmann, S.; DuPont, M.; Harbour, A.; Wright, J.; Lamendella, R.; Stevens, B.R.; Oudit, G.Y.; et al. Plasma Microbiome in COVID-19 Subjects: An Indicator of Gut Barrier Defects and Dysbiosis. Int. J. Mol. Sci. 2022, 23, 9141. [Google Scholar] [CrossRef]
  144. Okuyucu, M.; Yalcin Kehribar, D.; Çapraz, M.; Çapraz, A.; Arslan, M.; Çelik, Z.B.; Usta, B.; Birinci, A.; Ozgen, M. The Relationship Between COVID-19 Disease Severity and Zonulin Levels. Cureus 2022, 14, e28255. [Google Scholar] [CrossRef]
  145. Osman, I.O.; Garrec, C.; de Souza, G.A.P.; Zarubica, A.; Belhaouari, D.B.; Baudoin, J.P.; Lepidi, H.; Mege, J.L.; Malissen, B.; Scola, B.; et al. Control of CDH1/E-Cadherin Gene Expression and Release of a Soluble Form of E-Cadherin in SARS-CoV-2 Infected Caco-2 Intestinal Cells: Physiopathological Consequences for the Intestinal Forms of COVID-19. Front. Cell Infect. Microbiol. 2022, 12, 798767. [Google Scholar] [CrossRef]
  146. Natarajan, A.; Zlitni, S.; Brooks, E.F.; Vance, S.E.; Dahlen, A.; Hedlin, H.; Park, R.M.; Han, A.; Schmidtke, D.T.; Verma, R.; et al. Gastrointestinal symptoms and fecal shedding of SARS-CoV-2 RNA suggest prolonged gastrointestinal infection. Med 2022, 3, 371–387.e9. [Google Scholar] [CrossRef]
  147. Zollner, A.; Koch, R.; Jukic, A.; Pfister, A.; Meyer, M.; Rössler, A.; Kimpel, J.; Adolph, T.E.; Tilg, H. Post-acute COVID19 is characterized by gut viral antigen persistence in inflammatory bowel diseases. Gastroenterology 2022, 163, 495–506. [Google Scholar] [CrossRef]
  148. Chalkias, S.; Harper, C.; Vrbicky, K.; Walsh, S.R.; Essink, B.; Brosz, A.; McGhee, N.; Tomassini, J.E.; Chen, X.; Chang, Y.; et al. A Bivalent Omicron-Containing Booster Vaccine against COVID-19. N. Engl. J. Med. 2022, 387, 1279–1291. [Google Scholar] [CrossRef]
  149. Willan, J.; Agarwal, G.; Bienz, N. Mortality and burden of post-COVID-19 syndrome have reduced with time across SARS-CoV-2 variants in haematology patients. Br. J. Haematol. 2023, 201, 640–644. [Google Scholar] [CrossRef]
  150. Cortellini, A.; Tabernero, J.; Mukherjee, U.; Salazar, R.; Sureda, A.; Maluquer, C.; Ferrante, D.; Bower, M.; Sharkey, R.; Mirallas, O.; et al. SARS-CoV-2 omicron (B.1.1.529)-related COVID-19 sequelae in vaccinated and unvaccinated patients with cancer: Results from the OnCovid registry. Lancet Oncol. 2023, 24, 335–346. [Google Scholar] [CrossRef]
  151. Kahlert, C.R.; Strahm, C.; Güsewell, S.; Cusini, A.; Brucher, A.; Goppel, S.; Möller, E.; Möller, J.C.; Ortner, M.; Ruetti, M.; et al. Post-acute sequelae after SARS-CoV-2 infection by viral variant and vaccination status: A multicenter cross-sectional study. Clin. Infect. Dis. 2023, 77, 194–202. [Google Scholar] [CrossRef]
  152. Bolesławska, I.; Kowalówka, M.; Bolesławska-Król, N.; Przysławski, J. Ketogenic Diet and Ketone Bodies as Clinical Support for the Treatment of SARS-CoV-2-Review of the Evidence. Viruses 2023, 15, 1262. [Google Scholar] [CrossRef]
  153. Sharma, S.; Di Castelnuovo, A.; Costanzo, S.; Persichillo, M.; Panzera, T.; Ruggiero, E.; De Curtis, A.; Storto, M.; Cavallo, P.; Gianfagna, F.; et al. Habitual adherence to a traditional Mediterranean diet and risk of SARS-CoV-2 infection and Coronavirus disease 2019 (COVID-19): A longitudinal analysis. Int. J. Food Sci. Nutr. 2023, 74, 382–394. [Google Scholar] [CrossRef]
  154. Tomkinson, S.; Triscott, C.; Schenk, E.; Foey, A. The Potential of Probiotics as Ingestible Adjuvants and Immune Modulators for Antiviral Immunity and Management of SARS-CoV-2 Infection and COVID-19. Pathogens 2023, 12, 928. [Google Scholar] [CrossRef] [PubMed]
  155. Mardi, A.; Kamran, A.; Pourfarzi, F.; Zare, M.; Hajipour, A.; Doaei, S.; Abediasl, N.; Hackett, D. Potential of macronutrients and probiotics to boost immunity in patients with SARS-CoV-2: A narrative review. Front. Nutr. 2023, 10, 1161894. [Google Scholar] [CrossRef] [PubMed]
  156. Banerjee, A.; Somasundaram, I.; Das, D.; Jain Manoj, S.; Banu, H.; Mitta Suresh, P.; Paul, S.; Bisgin, A.; Zhang, H.; Sun, X.F.; et al. Functional Foods: A Promising Strategy for Restoring Gut Microbiota Diversity Impacted by SARS-CoV-2 Variants. Nutrients 2023, 15, 2631. [Google Scholar] [CrossRef]
  157. Cheong, K.L.; Yu, B.; Teng, B.; Veeraperumal, S.; Xu, B.; Zhong, S.; Tan, K. Post-COVID-19 syndrome management: Utilizing the potential of dietary polysaccharides. Biomed. Pharmacother. 2023, 166, 115320. [Google Scholar] [CrossRef]
  158. Cheong, K.L.; Chen, S.; Teng, B.; Veeraperumal, S.; Zhong, S.; Tan, K. Oligosaccharides as Potential Regulators of Gut Microbiota and Intestinal Health in Post-COVID-19 Management. Pharmaceuticals 2023, 16, 860. [Google Scholar] [CrossRef] [PubMed]
Figure 1. SARS-CoV-2 structure. SARS-CoV-2 is a single-stranded β-coronavirus. Two-thirds of viral RNA, in the first open reading frame (ORF 1a/b), encodes for 16 non-structural proteins (NSP). The remaining viral genome expresses accessory proteins, interfering with the host innate immune response, and four structural proteins (S, E, N, M).
Figure 1. SARS-CoV-2 structure. SARS-CoV-2 is a single-stranded β-coronavirus. Two-thirds of viral RNA, in the first open reading frame (ORF 1a/b), encodes for 16 non-structural proteins (NSP). The remaining viral genome expresses accessory proteins, interfering with the host innate immune response, and four structural proteins (S, E, N, M).
Medicina 59 01709 g001
Figure 2. Main entry route of SARS-CoV-2 in gastrointestinal tract. The spike protein of SARS-CoV-2 binds angiotensin-converting enzyme 2 (ACE2) receptors. Priming by TMPRSS2, TMPRSS4 (or other proteases such as FURIN) leads to endocytosis. The virus is uncoated, genomic RNA is released, and viral proteins are synthesized using the host replication system.
Figure 2. Main entry route of SARS-CoV-2 in gastrointestinal tract. The spike protein of SARS-CoV-2 binds angiotensin-converting enzyme 2 (ACE2) receptors. Priming by TMPRSS2, TMPRSS4 (or other proteases such as FURIN) leads to endocytosis. The virus is uncoated, genomic RNA is released, and viral proteins are synthesized using the host replication system.
Medicina 59 01709 g002
Figure 3. SARS-CoV-2–gastrointestinal tract interactions. SARS-CoV-2 infection induces a disruption of intestinal barrier integrity. Gut microbiota is altered with a reduction in microbial diversity, reduction in beneficial bacterial strains, and an increase in pathogens. The mechanical barrier, consisting of intestinal epithelial cells and tight junctions, is also affected by the infection. SARS-CoV-2 induces immune activation, resulting in increased levels of cytokines and leukocytes.
Figure 3. SARS-CoV-2–gastrointestinal tract interactions. SARS-CoV-2 infection induces a disruption of intestinal barrier integrity. Gut microbiota is altered with a reduction in microbial diversity, reduction in beneficial bacterial strains, and an increase in pathogens. The mechanical barrier, consisting of intestinal epithelial cells and tight junctions, is also affected by the infection. SARS-CoV-2 induces immune activation, resulting in increased levels of cytokines and leukocytes.
Medicina 59 01709 g003
Figure 4. Gut microbiome alterations in COVID-19. In healthy individuals, Faecalibacterium prausnitzii, Eubacterium and Roseburia are the prevalent strains of gut microbiome. These bacteria show anti-inflammatory properties and produce SCFA. In patients affected by COVID-19, a depletion of symbionts and an increase in opportunistic pathogens were described. Gut dysbiosis observed during COVID-19 infection persists after SARS-CoV-2 clearance.
Figure 4. Gut microbiome alterations in COVID-19. In healthy individuals, Faecalibacterium prausnitzii, Eubacterium and Roseburia are the prevalent strains of gut microbiome. These bacteria show anti-inflammatory properties and produce SCFA. In patients affected by COVID-19, a depletion of symbionts and an increase in opportunistic pathogens were described. Gut dysbiosis observed during COVID-19 infection persists after SARS-CoV-2 clearance.
Medicina 59 01709 g004
Table 1. Gastrointestinal symptoms in adult and pediatric patients.
Table 1. Gastrointestinal symptoms in adult and pediatric patients.
Adult PatientsPediatric Patients
Diarrhea16.5% 19.0%
Nausea/vomiting11.2% 19.7%
abdominal pain4.5%20.3%
Anorexia1.6%10%
Data reported in two recent meta-analyses by Shehab [8] and Bolia [9] for adults and children, respectively.
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

Vernia, F.; Ashktorab, H.; Cesaro, N.; Monaco, S.; Faenza, S.; Sgamma, E.; Viscido, A.; Latella, G. COVID-19 and Gastrointestinal Tract: From Pathophysiology to Clinical Manifestations. Medicina 2023, 59, 1709. https://doi.org/10.3390/medicina59101709

AMA Style

Vernia F, Ashktorab H, Cesaro N, Monaco S, Faenza S, Sgamma E, Viscido A, Latella G. COVID-19 and Gastrointestinal Tract: From Pathophysiology to Clinical Manifestations. Medicina. 2023; 59(10):1709. https://doi.org/10.3390/medicina59101709

Chicago/Turabian Style

Vernia, Filippo, Hassan Ashktorab, Nicola Cesaro, Sabrina Monaco, Susanna Faenza, Emanuele Sgamma, Angelo Viscido, and Giovanni Latella. 2023. "COVID-19 and Gastrointestinal Tract: From Pathophysiology to Clinical Manifestations" Medicina 59, no. 10: 1709. https://doi.org/10.3390/medicina59101709

Article Metrics

Back to TopTop