Next Article in Journal
Skeletal Muscle DNA Methylation and mRNA Responses to a Bout of Higher versus Lower Load Resistance Exercise in Previously Trained Men
Previous Article in Journal
A Novel Window into Angiogenesis—Intravital Microscopy in the AV-Loop-Model
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Pathogenesis and Mechanisms of SARS-CoV-2 Infection in the Intestine, Liver, and Pancreas

1
Department of Pathology, School of Medicine, Louisiana State University Health School of Medicine, New Orleans, LA 70112, USA
2
Louisiana Cancer Research Center, Louisiana State University Health Sciences Center, New Orleans, LA 70112, USA
3
Department of Pathology and Laboratory Medicine, Tulane University School of Medicine, New Orleans, LA 70112, USA
*
Author to whom correspondence should be addressed.
These authors contributed equally to this work.
Cells 2023, 12(2), 262; https://doi.org/10.3390/cells12020262
Submission received: 23 November 2022 / Revised: 30 December 2022 / Accepted: 5 January 2023 / Published: 9 January 2023
(This article belongs to the Section Cellular Pathology)

Abstract

:
The novel coronavirus, SARS-CoV-2, rapidly spread worldwide, causing an ongoing global pandemic. While the respiratory system is the most common site of infection, a significant number of reported cases indicate gastrointestinal (GI) involvement. GI symptoms include anorexia, abdominal pain, nausea, vomiting, and diarrhea. Although the mechanisms of GI pathogenesis are still being examined, viral components isolated from stool samples of infected patients suggest a potential fecal–oral transmission route. In addition, viral RNA has been detected in blood samples of infected patients, making hematologic dissemination of the virus a proposed route for GI involvement. Angiotensin-converting enzyme 2 (ACE2) receptors serve as the cellular entry mechanism for the virus, and these receptors are particularly abundant throughout the GI tract, making the intestine, liver, and pancreas potential extrapulmonary sites for infection and reservoirs sites for developing mutations and new variants that contribute to the uncontrolled spread of the disease and resistance to treatments. This transmission mechanism and the dysregulation of the immune system play a significant role in the profound inflammatory and coagulative cascades that contribute to the increased severity and risk of death in several COVID-19 patients. This article reviews various potential mechanisms of gastrointestinal, liver, and pancreatic injury.

Graphical Abstract

1. Introduction

Coronavirus disease-2019 (COVID-19) was first reported in Wuhan, China, in December 2019, after which it rapidly spread, causing a significant global public health problem. At the time of writing this review, there have been over 632,953,782 confirmed cases and 6,593,715 deaths worldwide; the United States appears to be the most affected country, with 96,752,266 cases reported, albeit this may be due to the lack of proper testing and reporting in other regions [1]. This disease is caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), a single-stranded and enveloped positive RNA coronavirus belonging to the genus Betacoronavirus [2]. This novel virus has a 29.9 kB genome composed of 14 open frames that encode 29 proteins; of these, 16 are non-structural proteins (nsps), 4 are structural proteins, including the nucleocapsid, spike glycoprotein, envelope, and membrane glycoprotein, and the remaining 9 are accessory proteins [3,4,5]. The spike glycoprotein contains a receptor binding domain that enables the virus to bind to human angiotensin-converting enzyme 2 (ACE2) receptors and infect host cells [6,7,8]. Spike protein mutations result in the emergence of new viral variants, which can be more contagious and potentially resistant to vaccines [9,10].
Initially, SARS-CoV-2 infects the upper respiratory tract epithelium, where respiratory epithelial cells containing ACE2 receptors bind with the virally encoded spike glycoprotein. Transmembrane protease serine 2 (TMPRSS2) receptors prime the spike protein and promote viral endocytosis [7]. After primary infection, peak viral shedding occurs during the first week of infection and likely contributes to the high transmissibility of the virus [11]. The severity of COVID-19 ranges from asymptomatic to critical, with more than 80% of cases classified as a self-limiting mild-to-moderate disease presenting with symptoms that may include fever, headache, anosmia, and mild cough [12,13]. Up to 20% of patients will develop severe disease including approximately 5% classified as critical [14], where widespread infection of lung alveolar epithelial cells causes extreme disruption of gas exchange in the alveoli, contributing to the development of severe hypoxemia, edema, and lung fibrosis [9]. Viral dissemination to the lower respiratory tract follows, resulting in the development of viral pneumonia [15], and some patients exhibited severe adverse outcomes complicated by acute respiratory distress syndrome, respiratory failure, and in 1–2% of cases in the US, death [16,17].
Intracellularly, the viral genome is uncoated, released, and translated into various viral components. The replicase gene involves two open reading frames (ORFs) that encode the viral sections of the replication–transcription complex (RTC) [18,19]. Expression of the replicase gene and subsequent encoding of ORF1a and ORF1b leads to the production of replicase polyproteins, pp1a and pp1ab, respectively. The replicase polyproteins are cleaved by viral proteases into 11 (pp1a) or 16 (pp1ab) individual subunits of the RTC that are capable of associating with each other and host cell components to induce proviral changes in the environment. Nsp3, nsp4, and nsp6 have been shown to induce a change in membrane formation resulting in virus-induced double-membrane vesicles (DMVs) [19], most likely derived from the endoplasmic reticulum (ER) [20]. The RTC participates in the replication and subsequent generation of a nested set of subgenomic RNA, which is translated into viral structural proteins (S, M, N, and E). The newly formed virus particles are assembled by exploiting the membranes and other elements of the ER and Golgi complex. Eventually, the mature virions are transported in vesicles and released from the cell via exocytosis into the extracellular compartments [21,22].
Viral modifications in the autophagic and extracellular vesicle pathways contribute to the overactivated and impaired immune responses that ultimately promote viral replication, the release of viral particles into the extracellular compartment [21], and the hypercoagulative states related to disease severity. Research shows that several cell types are affected by SARS-CoV-2 infection [23,24,25], with some having deleterious effects on disease severity and outcome, including epithelial, endothelial, and immune cells. Various immune and inflammatory changes induced by the virus contribute to endothelial cell damage and dysfunction in COVID-19 that may induce hypercoagulative states [26,27,28]. In hypercoagulative states, there is an increased chance of developing lung thrombi and emboli that contribute to the respiratory distress and mortality encountered in COVID-19 patients.
The high expression of ACE2 in the alimentary tract makes the small and large bowels vulnerable to SARS-CoV-2 infection [29,30]. Although fever, cough, and shortness of breath are the most common symptoms, around 17.6% of the patients present with gastrointestinal manifestations [31]. Of the 17.6%, anorexia is the most common presentation in 26.8% of cases, followed by diarrhea (12.5%), nausea and vomiting (10.2%), and abdominal pain (9.2%). Furthermore, liver injury is observed in 15% to 53% of the cases. The real-time reverse-transcription polymerase chain reaction (RT-PCR) assay is the standard test for laboratory diagnosis of SARS-CoV-2 samples collected from infected patients, including stool samples [32]. This review summarizes the pathogenesis and potential mechanisms of gastrointestinal, liver, and pancreatic injuries in patients with COVID-19.

2. Mechanisms of SARS-CoV-2 Infection in the Intestine

2.1. Viral Kinetics and Physiologic Luminal Barriers

The positive detection of SARS-CoV-2 in stool constitutes evidence that the virus exists and replicates in the intestines of infected patients [33,34,35]. More than half of the hospitalized patients with COVID-19 demonstrated positive stool samples, particularly those with diarrhea. Interestingly, some studies have shown that a subset of COVID-19 patients still presents with positive SARS-CoV-2 stool after negative nasopharyngeal tests [36,37]. The exact mechanism for how the virus infects the gastrointestinal tract is unknown. Still, hypotheses include fecal–oral transmission, direct digestion after viral exposure, and mucociliary clearance of respiratory virions that may allow entry into the alimentary tract via the esophagus [38]. SARS-CoV-2 viral shedding has been detected in the stool of COVID-19 patients and, as with most enteric viruses, it could be a potential fecal–oral transmission route [34,39,40,41,42,43]. A study showed that SARS-CoV-2 was detected on toilet surfaces and the bathroom door handle in a patient room with confirmed fecal viral positivity by RT-PCR [44]. For this reason, individuals using poorly sanitized areas, such as public toilets, are potentially more vulnerable to viral transmission after touching the nose, mouth, or eyes with contaminated hands. Furthermore, SARS-CoV-2 has been detected in raw urban sewage as a part of population-wide surveillance and may be particularly beneficial as an early warning system in monitoring for mutated variants [45,46]. The virus’s genetic diversity due to high mutation rates threatens COVID-19 treatment options and the current vaccination programs. A drawback to wastewater surveillance is that samples may reveal artificial genomes that do not correspond to a virus circulating in the population; however, they can still be linked to specific variant clusters and provide information about variants circulating in an area.
Enteric RNA viruses, including rotavirus, norovirus, and astrovirus, are mainly transmitted via the fecal–oral route [47,48,49]. After ingestion, these viruses are carried to the intestine, where they invade the epithelial cells, cause gastrointestinal symptoms, and are excreted with stool. SARS-CoV-2 may exhibit a similar mechanism of transmission and pathogenesis in the gastrointestinal tract [30,39,43,50,51]. Most enveloped viruses would not survive in the harsh alimentary tract conditions; however, a notable exception is enteric coronaviruses. Though the survival mechanisms are unclear, some studies suggest the possibility of SARS-CoV-2 surviving and being transported to the intestines in saliva or secretions [30,52,53]. Gastric acid is an essential physiological barrier that plays a vital role in the inactivation of swallowed pathogens. Studies have shown that the use of a proton pump inhibitor, a medication that causes a reduction in gastric acid production, has been linked with increased mortality in patients with COVID-19 infection [54,55,56,57]. The suppression of gastric acid can facilitate the passage of the virus through the stomach to the intestine [58]. Because of the drug-induced, less acidic, virus-friendly environment in the intestine and the high viral load in the stool, the virus may survive for a longer time in the gastrointestinal tract compared to the respiratory tract [43].

2.2. Possible Viral Entry Mechanisms to the Intestinal Epithelial Cells

ACE2 is an important receptor on the cell membrane of host cells. It is widely distributed in various organs, including the nasopharynx, lung, esophagus, stomach, intestine, liver, and pancreas. Studies have shown that ACE2 is abundantly expressed in the gastrointestinal tract, particularly the colon, at approximately 100 times more than in the lung [59,60,61]. Multiple proteins are anchored on the viral surface, including structural spike, membrane, nucleocapsid, and envelope proteins. The interaction between the virus’s transmembrane spike glycoprotein and the ACE2 receptor on enterocytes promotes the invasion of host brush border cells. The spike protein must be primed by TMPRSS2 which promotes endocytosis and facilitates the invasion of intestinal enterocytes by the SARS-CoV-2 virus [62]. Enterocytes are intestinal absorptive cells that create the brush border of the luminal surface of the intestines. These cells highly express ACE2 and TMPRSS2 receptors [7,62,63]. The brush border has interspersed immune cells called microfold cells (M cells) that initiate localized immune responses by taking up antigens from the lumen of the intestines and delivering them to antigen-presenting cells and other immune cells that migrate to a pocket on the basement membrane surface of M cells. However, in the presence of some viruses, the ingested viral particles are not transported across the M cell to underlying lymphoid follicles but instead hijack the host cells’ machinery and begin actively replicating within the M cells, like the well-studied coronavirus transmissible gastroenteritis virus [64,65]. The abundant viral replication results in destroyed M cells, leading to mucosal inflammation and diarrhea. As M cells are destroyed, SARS-CoV-2 avoids immune detection via antigen presentation to the underlying lymphoid follicles. This is carried out by apical virion exocytosis and by utilizing spike protein fragments, which remain in the cytosol after replication and virion assembly, to invade adjacent uninfected cells basolaterally [66]. Since a highly weakened immune response may be launched from the direct infection of enterocytes by inducing the release of cytokines, viral particles in the lumen can continue to infect uninfected enterocytes, allowing viral proliferation and subsequent damage to the gastrointestinal tract. Figure 1 depicts the entry and viral life cycle mechanisms in intestinal epithelial cells.

2.3. Autophagy and the Replication of SARS-CoV-2

An essential part of the antiviral defense mechanism for cells is autophagy. Autophagy is crucial to maintain cellular homeostasis, remove misfolded proteins, eliminate viral particles, and reduce virion formation [67]. Cells conduct this process in response to cellular stress by using autophagy machinery to deliver materials in the cytosol to the interior of lysosomes for degradation, subsequently recovering metabolic energy sources and targeting damaged or unnecessary organelles to lysosomes for removal. However, autophagy pathways and machinery can be hijacked by certain invading viruses, including SARS-CoV-2, and then used against host cells to aid viral replication, proliferation, and translocation.
The incorporation of viral genes into the host cell genome can alter the expression of autophagy-related genes via serum starvation, pH imbalances, stimulation of T and B cell antigen receptors, and cytokine stimulation. In studies involving HCV, an RNA virus whose viral replication kinetics are similar to SARS-CoV-2, it has been shown that the virus activates autophagy during the acute infection period [68,69]. Several replication intermediates are synthesized in the acute period of the COVID-19 infection. The clearance of intermediate replication products by virus-activated autophagy has been suggested as the mechanism of escape from interferon (IFN) lambda-related immune response [70]. Like other coronaviruses, the formation of pseudo-enveloped vesicles during SARS-CoV-2-induced autophagy relies on ER-derived membranes [71]. These viral-induced DMV membranes lack properly functioning ER chaperones and protein markers typically found in uninfected ER-derived membranes that assist the autophagic machinery with substrate recognition and targeting to lysosomes [67,72,73].
LC3 is a key protein in the autophagy pathway and is involved in substrate selection and the formation of autophagosomes [74,75]. Autophagy induction can be detected by converting LC3B-I, a widely distributed cytosolic protein, to LC3B-II, a conjugated form of LC3B-I concentrated in autophagolysosomal membranes [75,76]. SARS-CoV-2 infected cells contain the viral protein nsp15, which is a uridine-specific endoribonuclease unique to the order Nidovirales, that processes mature viral RNA during replication, and degrades viral sequences that activate host immune sensors by functioning as an RNase and degrading viral dsRNA intermediates, thus preventing immune recognition and delaying type I IFN response [77,78]. In a significant viral load, nsp15 overexpression has been shown to decrease LC3B-II and lead to the accumulation of p62, an autophagic receptor, indicating that this viral protein is involved in blocking autophagy induction, although the precise mechanisms are not clear [78,79,80]. Nsp15 has been considered a target for therapeutic techniques due to studies conducted that revealed that loss of nsp15 function correlates to both decreased replication and pathogenesis [81,82].
Studies show that SARS-CoV-2 inhibits autophagic flux using viral proteins ORF3a and ORF7a by disturbing the interplay between autophagosomes and lysosomes [83]. ORF3a plays a role in inducing autophagy [84]; however, it induces incomplete autophagy by interacting with and deregulating Beclin 1, an essential autophagy protein that regulates downstream autophagosome formation and maturation, and subsequently causes a buildup of immature autophagosomes incapable of phagolysosome fusion [85]. This prevents autophagic cargo degradation and aids in promoting viral RNA replication [86]. ORF7a was found to disrupt and reduce lysosomal acidity, which prevented autophagosomal degradation [78,83]. This leads to the accumulation of lysosomes that are incapable of autophagosomal degradation. Additionally, ORF7a may promote virion release by avoiding degradation [83]. ORF8 is a SARS-CoV-2 open reading frame that encodes a viral protein detected in LC3-labeled autophagosomes [87]. It was shown that this viral protein directly interacts with MHC I leading to MHC I downregulation and impairment in viral antigen presentation, which protects infected cells from being recognized and eradicated by T cells. LC3 usually facilitates lysosomal fusion and degradation, but studies [88,89] have revealed that viral protein disruption impairs LC3 function and allows SARS-CoV-2 virions to use the lysosomal trafficking pathway as a method of release from infected cells.
IRF8 is a transcription factor that promotes macrophage and dendritic cell differentiation and is critical for the expression of autophagy-related genes, stress-induced autophagy regulation, antigen presentation, and clearance of intracellular pathogens [90,91]. Studies show that COVID-19 patients have significantly reduced expression of IRF8, which contributes to the impairment of immune responses mediated by autophagy pathways [89,92,93] and ultimately leads to increased viral proliferation and deregulated inflammatory pathways due to IRF8′s role in MDSC pathology, which is discussed below.
Autophagy flux impairments can lead to ER stress, causing a subsequent increase in unfolded proteins leading to chronic tissue injury and excessive reactive oxygen species (ROS) production due to mitochondrial dysfunction [79,94]. Under normal conditions, autophagy suppresses inflammatory factors, but defective autophagy seen in SARS-CoV-2 reveals that the virus and viral-altered processes may uncontrollably stimulate inflammatory pathways [67,79,95]. One example is that ROS accumulation stimulates various mechanisms of inflammation, including inflammasomes and calpain, to produce IL-1B and IL-1a, respectively [96,97,98]. They also increase RIG-I-like receptors to enhance type-I IFN production. Inflammation in the gastrointestinal system can lead to diarrhea and severe abdominal pain. Autophagy is normally a major defense mechanism used to regulate and suppress levels of proinflammatory cytokines by efficiently removing stimuli, such as defective mitochondria, or pathogens, such as viral components, which activate these inflammatory factors. Dysregulation of autophagy-related genes, such as Beclin 1 and LC3 as seen in SARS-CoV-2 infection, causes a marked increase in cytokine secretion [96,99,100], implicating autophagy dysfunction as a significant contributor to an uncontrolled cytokine storm that is accountable for COVID-19 severity and death. The cytokine storm in turn will cause severe intestinal symptomatology, including diarrhea. In addition, the reduced activation of mTOR, a set of kinase-type proteins directly involved in autophagy induction, results in reduced secretion of antimicrobial agents by Paneth cells allowing uncontrolled viral proliferation in the intestines [67,92,101,102].

2.4. The Role of Extracellular Vesicles/Exosomes in Viral Transmission and Spreading to the GI System

SARS-CoV-2 RNA has been detected in blood samples of infected patients [42,103,104], making the hematologic dissemination of the virus to the digestive system another proposed alternative route of transmission. Extracellular vesicles (EV) are found in various biofluids and serve as carriers that transfer biomolecules to target cells all over the body. EV secretion is a known pathway aiding in the transmission and spread of several RNA viruses using exosomes, the smallest EV subclass; however, the mechanisms that viruses use to accomplish this is still up for debate [105,106,107]. One model proposes that the autophagic pathway is involved as LC3-labeled “autophagosome-like” organelles selectively capture a cluster of viral particles and release them non-cytolytically from an infected cell [108,109,110]. These released particles can subsequently be picked up by exosomes in biofluids and transported to distant organs. An additional proposed mechanism states viral proteins, RNA, or entire virions may be directed into exosomes that are then expelled from the infected cell via exocytosis into the extracellular space [107,111]. Exosomes then bind to target cells and induce susceptibility for viral infection within these healthy cells using membrane receptors, such as ACE2 and CD9, while fusing to the target cell membrane and unloading viral cargo [105,111,112]. It is thought that EVs play an important role in the transformation of COVID-19 infection from a local to a systemic infection contributing to the development of systemic inflammatory response syndrome [113]. This is a potential mechanism demonstrating how SARS-CoV-2 proteins, RNA, and even entire virions may be transported from the respiratory system after primary infection through biofluids and into the GI system to infect intestinal cells and cause GI illness. In a recently published article, it has been shown that inflammatory signals are carried to cells in distant organs through Tenascin C and Fibrinogen Beta carried by exosomes obtained from COVID-19 patients [113]. In another study, it was shown that some of the drugs that inhibit exosomes through different mechanisms inhibit SARS-CoV-2 replication [114]. All these preliminary results show that extracellular vesicles and exosomes have an important role in GI manifestations caused by direct or indirect mechanisms of COVID 19 infection.

2.5. Proinflammatory Effects of COVID-19 Infection and Systemic Inflammatory Response Syndrome

The production and release of large quantities of proinflammatory cytokines are referred to as a “cytokine storm” and have been well-established in the COVID-19 outbreak [115,116,117,118]. It is attributed to a systemic inflammatory reaction and multiple organ dysfunction, including the gastrointestinal system, seen in more severe cases [30,93,119]. Viral infections of intestinal cells cause intestinal epithelial cellular stress and have been implicated in inducing the release of various proinflammatory cytokines and chemokines that normally work to limit viral infection and spread [120,121]. In the presence of SARS-CoV-2, these chemical immune responses result in tissue and organ damage, coagulopathy, and acute respiratory distress syndrome (ARDS), which in turn increase the release of even more proinflammatory cytokines and chemokines, creating a cycle of uncontrolled inflammation and damage [122,123,124].
Various mechanisms have been explored regarding the molecular pathogenesis of SARS-CoV-2 in the intestines. ACE2 plays a role in controlling the absorption of amino acids, including tryptophan, in the small intestine [125]. Tryptophan is involved in regulating the suppression of intestinal inflammation and modulating intestinal microbiota. Infected enterocytes result in altered ACE2 expression, causing impaired tryptophan absorption and subsequent deregulation of tryptophan metabolism [126]. Alterations to tryptophan metabolism deregulate the metabolic ability to suppress intestinal inflammation [125,127]. Another mechanism involved in the pathogenesis of SARS-CoV-2 in intestinal cells involves the significant induction of type-I and type-III IFNs and the significant upregulation of proinflammatory mediators, indicating that the virus massively triggers innate immune responses [128].
Some of the viral nsps, such as nsp14, can block IFN-I responses and antagonize cytokine production by inducing host mRNA degradation and inhibiting the phosphorylation of both signal transducer and activator of transcription (STAT) 1 and interferon regulatory factor 3 (IRF3) [78,129,130,131]. In healthy cells, IRF3 plays a role in the innate antiviral response by inducing type-I IFNs. Nsp15 can inhibit IRF3 translocation by antagonizing IFN production [77], and nsp15-mediated inhibition of IRF3 is an additional method by which SARS-CoV-2 can evade the host’s immune response [132,133].
Infected cells release mediators and chemokines that quickly cause local neutrophil accumulation and the additional secretion of cytokines and chemokines, attracting further immune cells such as T lymphocytes [134]. The activation of T lymphocytes leads to a high expression of various cytokines, including IFN-γ, IL-2, IL-6, IL-7, IL-10, and human granulocyte-macrophage colony-stimulating factor [135,136]. These inflammatory cytokines can cause a unique immune activation, ultimately promoting host cell necrosis [50]. Microscopically, the epithelium of the digestive tract shows patchy plasma-lymphocytic infiltration with interstitial edema [137,138]. Vasculitis, active inflammation, viral inclusions, and submucosal fibrin thrombi have also been reported [139]. As a result, SARS-CoV-2 may cause gastrointestinal symptoms by the cytotoxic effect secondary to direct viral invasion in target cells and/or immune-mediated tissue and end-organ damage [140]. In severe cases, these immune responses fail to clear out the pathogens due to the impaired autophagy pathway mentioned above.

2.6. Myeloid Derived Supressor Cells (MDSCs)

An interesting finding in SARS-CoV-2’s mechanisms of immune evasion concerns MDSCs. MDSCs, or myeloid-derived suppressor cells, are pathologically activated monocytes and granulocytes [141]. These cells have unique metabolic and gene expression profiles that enable them to inhibit immune responses involving T cells, B cells, and natural killer (NK) cells and induce systemic and local immunosuppression while promoting the accumulation of other types of immunosuppressive cells, such as T-regulatory cells [142,143,144]. In the context of cancer, MDSC function worsens the disease, but in autoimmune and infectious diseases, MDSCs can limit severity [141]. Several studies have shown increased MDSCs, and more specifically, monocyte-MDSCs (M-MDSCs), in the blood samples of severe COVID-19 patients [92,145,146]. Multiple studies have demonstrated a significant expansion of M-MDSCs in SARS-CoV-2 infections that may occur via IRF8 and IL-6-mediated mechanisms [92,147,148,149]. IL-6 is a pleiotropic cytokine with both anti- and proinflammatory activities and was shown to be significantly increased in severe COVID-19 cases compared to mild cases [150]. This suggests that IL-6 can serve as a marker to indicate the transition from mild to severe COVID-19. High concentrations of IL-6 may be responsible for maintaining MDSC’s suppressive activity of T cells permitting prolonged viral survival. As mentioned earlier, IRF8, which was shown to be significantly reduced in COVID-19, promotes macrophage and dendritic cell differentiation [151] and has a critical role in autophagy, antigen presentation, and clearing intracellular pathogens [90]. Autophagy controls MHC II antigen presentation and is crucial for boosting tolerance and anti-inflammatory processes by myeloid cells [67,91,101,152]. Still, this process is impaired when IRF8 is downregulated, preventing immune cells from responding to antigens. IL-6 upregulation and IRF8 downregulation allow the maintenance of MDSC-induced immune suppression and subsequent advancement of hyperinflammation activation, contributing to the development of cytokine release syndrome or ‘cytokine storm.’
L-arginine is a semi-essential amino acid and intestinal L-arginine is involved in protein production, urea removal, and nitric oxide (NO), urea, creatine, and polyamine synthesis. It also plays a role in regulating intestinal pathogens to prevent infection. In healthy adults, adequate amounts of L-arginine are synthesized endogenously, but in the context of catabolic stress seen in COVID-19, endogenous synthesis is insufficient [145]. Depletion of free L-arginine has been implicated as a mechanism of T cell dysfunction [153,154,155,156]. This is most likely a result of the significant expansion of MDSCs seen in COVID-19 patients [145,157]. MDSCs highly express arginase I and iNOS that both metabolize free L-arginine, and one study demonstrated that L-arginine consumption by MDSCs induces platelet activation due to the reduced NO production secondary to the depletion of L-arginine. NO plays a role in inhibiting platelet activation [158,159], therefore an L-arginine deficiency is associated with increased severity and thrombotic complications reported in severe COVID-19 cases [145,160,161]. MDSCs also express specific ligands that interact with T cells and lead to T cell exhaustion, further suppressing the T cell response. Therefore, in response to viral invasion, the accumulation of MDSCs in the intestines inhibit the gut mucosal immune response allowing increased viral proliferation and subsequent inflammation.

2.7. The Effects of COVID-19 Infection on the Coagulation System in Gastrointestinal Tract

In critically ill patients, infectious complications are common and may activate multiple systemic inflammatory and coagulation responses that are intended to defend the host, but specific viral genes and proteins may manipulate the response leading to disseminated intravascular coagulation. Coagulopathy occurs when blood’s ability to form clots is impaired and has been seen in COVID-19 more frequently and with greater severity when compared to other diseases caused by respiratory viruses [162,163,164,165]. COVID-19-associated coagulopathy may present as either microthrombi or macrothrombi, and both can cause multiple organ damage, increased disease severity, and death [166]. Immunothrombosis occurs when viral pathogens and their components bind to pattern-recognizing receptors (PRRs) on immune cells and trigger inflammatory responses inducing proinflammatory cytokines that activate coagulation, particularly when damaged vascular endothelium, also known as vasculopathy, and resultant endothelial dysfunction promotes clot formation [27,28,167,168,169,170,171]. The pathogenesis of COVID-19 vasculopathy is still debated, with two prominent theories. The first is that the virus causes direct endothelial injury [6,60,172,173,174,175]; however, arguments have been made that endothelial cells do not express ACE2 or express it at very low levels, making this cell type resistant to infection [176,177]. The second is that the viral infection triggers a cytokine storm, the source of endothelial cell injury [177,178,179,180]. However, with disease progression, it is likely that multiple factors play into the severity of damage and subsequent thrombosis that causes organ damage. SARS-CoV-2 binds to ACE2 on lung epithelial cells resulting in a local ACE2 deficiency. ACE2 regulates the renin-angiotensin-aldosterone system (RAAS), which is involved in blood pressure and vascular resistance regulation, and the kallikrein-kinin system, which regulates vasoactive peptides [8,181]. Therefore, the deregulation of ACE2 can increase kinin activity and result in angioedema [166,182]. A ‘kinin storm’ may occur due to systemic kallikrein-kinin system activation resulting in increased inflammation, vascular permeability, effusion, and ultimately organ damage. Increased vasodilation and permeability allow vascular leakage of various components involved in the hyperactive innate immune response, such as autoantibodies, ROS, cytokines, and chemokines. These leaked substances cause a fibrinolysis imbalance contributing to glycocalyx damage and vascular endothelial injury that results in endothelial prothrombogenic activity and increased levels of von Willebrand factor, fibrin, tissue factor, and P-selectin. These increases contribute to the already overactivated coagulation cascade induced by catastrophic inflammation and increased cytokine production. This intense inflammatory state causes a severe hemostatic system derangement, systemic blood coagulation, and downregulation of the fibrinolytic and anticoagulant pathways, increasing the risk of thrombi formation. RAAS dysregulation is a major contributor to endothelial damage and is a significant component in microthrombi formation and COVID-19’s subsequent ischemic manifestations [183].
Ischemic colitis, a rare presentation of SARS-CoV-2 involvement in the colon, has been reported in some cases [138,183,184,185]. Microscopic examination of these cases showed coagulative mucosal necrosis with crypt apoptosis and small vessel thrombosis, mainly noted in the submucosal vasculature [184]. Reports indicate that patients with COVID-19 have high levels of D-dimer and fibrinogen, which increases the risk of micro- and macro-circulatory thrombosis [28,186,187]. Additionally, hypoxia that occurs in severe cases of COVID-19 can promote thrombosis formation by increasing the viscosity of the blood and activating the hypoxia-inducible transcription factor-dependent signaling pathway [171].

2.8. COVID-19 and Microbiota Changes

Studies of patients with GI-related COVID-19 symptoms lacked beneficial bacteria and showed a significant increase in opportunistic pathogens [188,189,190]. Anti-inflammatory bacteria normally found in healthy individuals were absent from the gut microbiomes of COVID-19 patients, with studies showing that the gastrointestinal virome functionality revealed increased inflammation, stress, and virulence responses and may be due to other viruses taking advantage of the host immune dysfunction [188]. As previously mentioned, ACE2 mediates the absorption of neutral amino acids, such as tyrosine, valine, and tryptophan in the intestine [8] and tryptophan plays a vital role in regulating gastrointestinal flora [126,127,191]. The binding of SARS-CoV-2 to ACE2 receptors in the bowel can reduce the number of free receptors and interfere with tryptophan absorption. This induces malabsorption and causes dysbiosis, an imbalance of the intestinal microbiota. Diarrhea seen in COVID-19 may result as a consequence of decreased ACE2 expression, subsequent reduced tryptophan absorption leading to gut dysbiosis, and dysregulated autophagic pathways. Downregulation of ACE2 in infected enterocytes induces intestinal dysbiosis. Dysbiosis inhibits mechanistic target of rapamycin (mTOR) which causes the activation of intestinal autophagy [101,192]. This causes autophagic degradation of NHE3, an intestinal epithelial brush-border Na+/H+ exchanger protein that regulates the NaCl absorption [193]. Reduced NHE3 levels reduce the intestine’s ability to absorb water which increases intestinal fluids and results in secretory-type diarrhea [194].
Studies have shown altered content of Clostridium, Lactobacillus, and Bifidobacterium in the intestinal flora of patients with SARS-CoV-2 [189,195,196,197]. A portion of intestinal flora are amino acid-fermenting bacteria that utilize arginine to function [198]. Alterations in intestinal flora may lead to an increased presence of L-arginine-fermenting bacteria leading to additional depletion of free L-arginine in the intestines. Depletion of intestinal L-arginine is known to increase MDSCs, suppress local T cell function as described above, as well as increase the production of ROS, thus exacerbating intestinal inflammation [145].
The dysbiosis of the bowel flora promotes viral replication and enhances the release of inflammatory proteins and cytokines, which ultimately contributes to the ‘cytokine storm’ state and subsequent widespread tissue damage, septic shock, and multi-organ failure [189]. Other factors altering gut flora in patients with COVID-19 include diabetes mellitus, old age, the use of antibiotics and antivirals, and steroids which can negatively affect the prognosis in these patients [199].

3. Mechanisms of SARS-CoV-2 Infection in the Liver

COVID-19-associated liver injury has been reported in several studies presenting with abnormal liver function biochemistries [200,201,202,203,204,205], and viral presence was confirmed by detecting SARS-CoV-2 in liver tissues [206,207,208,209]. In COVID-19, liver injury was characterized by elevated total bilirubin levels and increases in alanine transaminase and/or aspartate aminotransferases, and the level of these enzymes directly correlated with the severity of the disease [210,211,212,213].
The exact mechanism for pathogenesis by SARS-CoV-2 of the liver is controversial and most likely involves not one, but several mechanisms. Several studies have reported the involvement of ACE2, the overproduction and release of inflammation mediators, coagulation and thrombi formation, and hypoxia [60,204,209,213,214,215,216,217,218]. ACE2 receptors were found to be expressed in cholangiocytes more than in hepatocytes [170]; however, histopathological analysis revealed abundant virus particles within the cytoplasm of hepatocytes [213]. This could indicate that the presence of SARS-CoV-2 initiates an upregulation of ACE2 in this cell subtype allowing for viral infection [214]. In addition to the presence of viral particles in the cytoplasm of hepatocytes, liver biopsies revealed prominent Kupffer cells, acidophilic bodies, portal tracts with chronic inflammatory infiltrates, and ballooning degeneration of hepatocytes with frequent mitoses. Ultrastructural examination showed mitochondrial swelling, endoplasmic reticulum dilation, and glycogen granule decrease, accompanied by massive hepatic apoptosis, which is a known mechanism utilized by cells to dispose of damaged or virally infected cells. The histopathological changes observed in the livers of patients with acute SARS-CoV-2 infection are depicted in Figure 2.
ACE2 is expressed 20 times higher in the biliary epithelial cells, also known as cholangiocytes, than in hepatocytes indicating liver injury may also contribute to bile duct injury [218]. Several studies also showed a decrease in CD4+ and CD8+ lymphocytes [219,220,221], which can be a direct result of the increased numbers of MDSCs, as discussed above.
Additionally, KI-67 immunohistochemistry has shown an increased number of hepatocytes in the replicative phase with a proliferative index of 11.4% [206]. Prominent bi-nucleated hepatocytes with occasional apoptosis were also identified in some cases [213]. The findings of mitoses, apoptosis, and an elevated KI-67 index indicate potential cell cycle disruption caused by SARS-CoV-2 that warrants further investigation. Figure 2 demonstrates some of the histopathological features seen in the liver after infection with SARS-CoV-2. Figure 3 shows the immunohistochemical detection of viral nucleocapsid and spike proteins in hepatocytes of patients with active SARS-CoV-2 infection.
Liver endothelial cells were also found to abundantly express ACE2 [215] indicating that liver vascular injury induced by SARS-CoV-2 endothelial infection and replication may result in increased vascular permeability and damage allowing leakage of inflammatory cytokines and free ROS into liver tissues [222]. Interestingly, we found expression of both nucleocapsid and spike proteins in the endothelial cells of livers from patients with acute COVID-19 infections, indicating that the virus actively reaches the liver through the vascular system, either through endothelial cells of through macrophages or lymphocytes, which are also robustly positive for SARS-CoV-2 proteins. Endothelial damage exposes underlying collagen, which subsequently attracts platelets and neutrophils to promote NETosis and clot formation. Accumulation of platelets and immature neutrophils in the liver may further initiate endothelial damage and lead to the microvascular thrombi observed in severe and nonsurvivable cases of COVID-19 [170,223,224]. Figure 4 shows the expression of nucleocapsid and spike in portal inflammatory cells, lymphocytes, macrophages and endothelial cells.
Drug-induced liver damage is another contributing factor to hepatocyte injury in patients with COVID-19. Many medications used to treat hospitalized patients have a significant hepatotoxic effect, including acetaminophen, IFN, antibacterial agents, remdesivir, and lopinavir/ritonavir. Ischemia due to vascular coagulopathy, thrombosis, and systematic inflammatory cytokines storm are other contributing factors to significant liver damage in patients with severe COVID-19 infection [15,172].

4. Mechanisms of SARS-CoV-2 Infection in the Pancreas

To date, there is insufficient data to establish a definitive causative mechanism of pancreatic injury related to COVID-19. There have been two prominent theories proposed describing the mechanisms of pancreatic damage seen in SARS-CoV-2 infection, including a direct and an indirect mechanism. The direct mechanism may be explained by studies showing that pancreatic acinar, ductal, and islet cells highly express ACE2 and TMPRSS2 receptors that promote SARS-CoV-2 cell entry [225,226,227]. A study conducted by Szlachcic et al. used in vitro models of human pancreatic cells to show that SARS-CoV-2 is capable of infecting and actively replicating in these cell types [226]. Furthermore, Liu et al. reported that in healthy individuals, the pancreas was found to express higher levels of ACE2 mRNA than in the lungs [225]. Indirect pancreatic injury may be induced by the systemic inflammatory and immune responses induced by SARS-CoV-2 and subsequent endothelial cell damage that mediates the leakage of these proinflammatory substances for distribution to various distant tissues, including the pancreas [228,229]. It is also possible that the virus may initially spread from the duodenal epithelium to ACE2-expressing acinar and islet cells via the pancreatic duct inducing a cytolytic effect that permits the release of pancreatic enzymes into the interstitial space. This may be mediated by infection-induced dysregulation of the gut microbiota and subsequent disruption of the intestinal barrier as described above.
Microscopic examination of COVID-19 pancreatic tissues at autopsy revealed the expression of entry receptors, severe fatty replacement of acinar cell mass, arteriosclerosis, and fibrosis [230,231,232,233]. During pancreatic inflammation, activated stellate cells release inflammatory cytokines and chemokines resulting in pancreatic fibrosis [234]. ACE2 downregulation caused by direct viral infection in pancreatic cells may disrupt ACE2/angiotensin-(1–7) interactions that normally regulate insulin resistance, insulin secretion, and pancreatic B cell survival [235]. Loss of ACE2 functions lead to accumulation of angiotensin II. Increased levels of angiotensin II dysregulates the protective arm of the RAAS, resulting in increased fibrosis, inflammation, and hypertension [236]. Multiple cases of acute pancreatitis in patients with COVID-19 have been reported in the literature [36,237,238,239,240] and altered serum cytokine levels of increased IL-6, IL-8, and IL-10 were found to be similar in cases of severe acute pancreatitis and in cases of COVID-19 [229]. Higher IL-6 levels are a recognized severity marker for both diseases individually [241,242]. A case-series study of 52 COVID-19 patients showed elevated amylase and lipase levels in 17% of the cases [243]. Previous studies reported that leakage of pancreatic triglyceride lipase in visceral adipose tissue, including intrapancreatic adipose tissue, results in increased adipose tissue lipolysis which generates excess nonesterified fatty acids [229,244]. These toxic fatty acids are known to inhibit mitochondrial complexes I and V, and cause pancreatic necrosis, ARDS, and multisystem organ failure [245]. Thus, acute pancreatitis in COVID-19 patients may potentially occur directly due to the cytopathic effect of SARS-CoV-2, secondary to leakage of pancreatic intracellular enzymes that result in autodigestion and cell death, or indirectly by cytotoxic immune responses.

5. Conclusions

While COVID-19 is mainly an upper and lower respiratory disease, increasing cases and reports point to the involvement of extrapulmonary organs and systems by SARS-CoV-2 infection. Gastrointestinal symptoms are increasingly reported in patients with COVID-19, and studies of these patients revealed the presence of the virus in enterocytes, pancreatic epithelial and islet cells, hepatocytes, and cholangiocytes. While the abundance of ACE2 receptors on these cells makes them highly susceptible to SARS-CoV-2 infection, the autophagic, vesicular, and metabolically active machinery creates an ideal microenvironment for viral replication, proliferation, and systemic transmission. The mechanisms of gastrointestinal injuries are largely multifactorial and involve a myriad of alterations to autophagy, immune evasion, and hyperinflammatory responses that contribute to GI symptomology and may compound to create a series of catastrophic events that can progress to an unsustainable disease state. Elevated transaminase and pancreatic enzyme levels following severe COVID-19 infections may be associated with viremia-induced systemic inflammatory response syndrome that can lead to sepsis. Additionally, elevated liver enzymes in severe cases of COVID-19 may indicate the possibility of hepatic and pancreatic damage that further contribute to the severity of the disease. It is critical to monitor these enzymes as they may predict a poorer prognosis. Viral shedding in the stool points to a possible fecal-oral transmission route, so it is essential to implement precautionary measures to prevent future re-infections. Hematologic studies should be conducted regularly in patients with coagulative risk factors, more severe disease states, and immobile patients, as viral presence in the blood has been implicated in alterations involving coagulation cascades, the development of thrombi and emboli, and systemic inflammatory-related burdens. Further studies are needed to determine potential treatments that may inhibit and mitigate the effects of autophagic and immune dysfunction.

Author Contributions

Conceptualization and writing: M.T.B., Z.K., A.R.K., G.L. and L.D.V. All authors have read and agreed to the published version of the manuscript.

Funding

This work was made possible thanks in part to Grant GM121288, Molecular Histopathology & Analytical Microscopy Core of the Center for Translational Viral Oncology.

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. WHO. Who Coronavirus (COVID-19) Dashboard. Available online: https://covid19.who.int (accessed on 17 November 2022).
  2. Coronaviridae Study Group of the International Committee on Taxonomy of Viruses. The species Severe acute respiratory syndrome-related coronavirus: Classifying 2019-nCoV and naming it SARS-CoV-2. Nat. Microbiol. 2020, 5, 536–544. [Google Scholar] [CrossRef] [Green Version]
  3. Aldaais, E.A.; Yegnaswamy, S.; Albahrani, F.; Alsowaiket, F.; Alramadan, S. Sequence and structural analysis of COVID-19 E and M proteins with MERS virus E and M proteins—A comparative study. Biochem. Biophys. Rep. 2021, 26, 101023. [Google Scholar] [CrossRef]
  4. Bai, C.; Zhong, Q.; Gao, G.F. Overview of SARS-CoV-2 genome-encoded proteins. Sci. China Life Sci. 2022, 65, 280–294. [Google Scholar] [CrossRef]
  5. Rauti, R.; Shahoha, M.; Leichtmann-Bardoogo, Y.; Nasser, R.; Paz, E.; Tamir, R.; Miller, V.; Babich, T.; Shaked, K.; Ehrlich, A.; et al. Effect of SARS-CoV-2 proteins on vascular permeability. eLife 2021, 10, e69314. [Google Scholar] [CrossRef]
  6. Scialo, F.; Daniele, A.; Amato, F.; Pastore, L.; Matera, M.G.; Cazzola, M. ACE2: The Major Cell Entry Receptor for SARS-CoV-2. Lung 2020, 198, 867–877. [Google Scholar] [CrossRef]
  7. Hoffmann, M.; Kleine-Weber, H.; Schroeder, S.; Kruger, 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.e278. [Google Scholar] [CrossRef]
  8. Kuba, K.; Imai, Y.; Ohto-Nakanishi, T.; Penninger, J.M. Trilogy of ACE2: A peptidase in the renin-angiotensin system, a SARS receptor, and a partner for amino acid transporters. Pharmacol. Ther. 2010, 128, 119–128. [Google Scholar] [CrossRef]
  9. Mason, R.J. Pathogenesis of COVID-19 from a cell biology perspective. Eur. Respir. J. 2020, 55, 2000607. [Google Scholar] [CrossRef] [Green Version]
  10. Tao, K.; Tzou, P.L.; Nouhin, J. The biological and clinical significance of emerging SARS-CoV-2 variants. Nat. Rev. Genet. 2021, 22, 757–773. [Google Scholar] [CrossRef]
  11. He, X.; Lau, E.H.Y.; Wu, P.; Deng, X.; Wang, J.; Hao, X.; Lau, Y.C.; Wong, J.Y.; Guan, Y.; Tan, X.; et al. Temporal dynamics in viral shedding and transmissibility of COVID-19. Nat. Med. 2020, 26, 672–675. [Google Scholar] [CrossRef]
  12. Chavez, S.; Long, B.; Koyfman, A.; Liang, S.Y. Coronavirus Disease (COVID-19): A primer for emergency physicians. Am. J. Emerg. Med. 2021, 44, 220–229. [Google Scholar] [CrossRef]
  13. Long, B.; Carius, B.M.; Chavez, S.; Liang, S.Y.; Brady, W.J.; Koyfman, A.; Gottlieb, M. Clinical update on COVID-19 for the emergency clinician: Presentation and evaluation. Am. J. Emerg. Med. 2022, 54, 46–57. [Google Scholar] [CrossRef]
  14. Wiersinga, W.J.; Rhodes, A.; Cheng, A.C.; Peacock, S.J.; Prescott, H.C. Pathophysiology, Transmission, Diagnosis, and Treatment of Coronavirus Disease 2019 (COVID-19): A Review. JAMA 2020, 324, 782–793. [Google Scholar] [CrossRef]
  15. Huang, C.; Wang, Y.; Li, X.; Ren, L.; Zhao, J.; Hu, Y.; Zhang, L.; Fan, G.; Xu, J.; Gu, X.; et al. Clinical features of patients infected with 2019 novel coronavirus in Wuhan, China. Lancet 2020, 395, 497–506. [Google Scholar] [CrossRef] [Green Version]
  16. Lentz, S.; Roginski, M.A.; Montrief, T.; Ramzy, M.; Gottlieb, M.; Long, B. Initial emergency department mechanical ventilation strategies for COVID-19 hypoxemic respiratory failure and ARDS. Am. J. Emerg. Med. 2020, 38, 2194–2202. [Google Scholar] [CrossRef]
  17. Dong, E.; Du, H.; Gardener, L. An interactive web-based dashboard to track COVID-19 in real time. Lancet Inf. Dis. 2022, 20, 533–534. [Google Scholar] [CrossRef]
  18. Mishchenko, E.L.; Va, V.A.I. Replication-transcription complex of coronaviruses: Functions of individual viral non-structural subunits, properties and architecture of their complexes. Vavilovskii Zhurnal Genet. Sel. 2022, 26, 121–127. [Google Scholar] [CrossRef]
  19. Angelini, M.M.; Akhlaghpour, M.; Neuman, B.W.; Buchmeier, M.J. Severe acute respiratory syndrome coronavirus nonstructural proteins 3, 4, and 6 induce double-membrane vesicles. mBio 2013, 4, e00524-13. [Google Scholar] [CrossRef] [Green Version]
  20. Wolff, G.; Melia, C.E.; Snijder, E.J.; Barcena, M. Double-Membrane Vesicles as Platforms for Viral Replication. Trends Microbiol. 2020, 28, 1022–1033. [Google Scholar] [CrossRef]
  21. Mohamadian, M.; Chiti, H.; Shoghli, A.; Biglari, S.; Parsamanesh, N.; Esmaeilzadeh, A. COVID-19: Virology, biology and novel laboratory diagnosis. J. Gene Med. 2021, 23, e3303. [Google Scholar] [CrossRef]
  22. Woodby, B.; Arnold, M.M.; Valacchi, G. SARS-CoV-2 infection, COVID-19 pathogenesis, and exposure to air pollution: What is the connection? Ann. N. Y. Acad. Sci. 2021, 1486, 15–38. [Google Scholar] [CrossRef] [PubMed]
  23. Liu, J.; Li, Y.; Liu, Q.; Yao, Q.; Wang, X.; Zhang, H.; Chen, R.; Ren, L.; Min, J.; Deng, F.; et al. SARS-CoV-2 cell tropism and multiorgan infection. Cell Discov. 2021, 7, 17. [Google Scholar] [CrossRef] [PubMed]
  24. Ondruschka, B.; Heinrich, F.; Lindenmeyer, M.; Edler, C.; Möbius, D.; Czogalla, J.; Heinemann, A.; Braun, F.; Aepfelbacher, M.; Lütgehetmann, M.; et al. Multiorgan tropism of SARS-CoV-2 lineage B.1.1.7. Int. J. Leg. Med. 2021, 135, 2347–2349. [Google Scholar] [CrossRef] [PubMed]
  25. Puelles, V.G.; Lutgehetmann, M.; Lindenmeyer, M.T.; Sperhake, J.P.; Wong, M.N.; Allweiss, L.; Chilla, S.; Heinemann, A.; Wanner, N.; Liu, S.; et al. Multiorgan and Renal Tropism of SARS-CoV-2. N. Engl. J. Med. 2020, 383, 590–592. [Google Scholar] [CrossRef] [PubMed]
  26. Carnevale, S.; Beretta, P.; Morbini, P. Direct endothelial damage and vasculitis due to SARS-CoV-2 in small bowel submucosa of COVID-19 patient with diarrhea. J. Med. Virol. 2021, 93, 61–63. [Google Scholar] [CrossRef]
  27. Iba, T.; Connors, J.M.; Levy, J.H. The coagulopathy, endotheliopathy, and vasculitis of COVID-19. Inflamm. Res. 2020, 69, 1181–1189. [Google Scholar] [CrossRef]
  28. Katneni, U.K.; Alexaki, A.; Hunt, R.C.; Schiller, T.; DiCuccio, M.; Buehler, P.W.; Ibla, J.C.; Kimchi-Sarfaty, C. Coagulopathy and Thrombosis as a Result of Severe COVID-19 Infection: A Microvascular Focus. Thromb. Haemost. 2020, 120, 1668–1679. [Google Scholar] [CrossRef]
  29. 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]
  30. Zhang, H.; Kang, Z.; Gong, H.; Xu, D.; Wang, J.; Li, Z.; Li, Z.; Cui, X.; Xiao, J.; Zhan, J.; et al. Digestive system is a potential route of COVID-19: An analysis of single-cell coexpression pattern of key proteins in viral entry process. Gut 2020, 69, 1010–1018. [Google Scholar] [CrossRef] [Green Version]
  31. Aguila, E.J.T.; Cua, I.H.Y.; Dumagpi, J.E.L.; Francisco, C.P.D.; Raymundo, N.T.V.; Sy-Janairo, M.L.L.; Cabral-Prodigalidad, P.A.I.; Lontok, M.A.D. COVID-19 and its effects on the digestive system and endoscopy practice. JGH Open 2020, 4, 324–331. [Google Scholar] [CrossRef]
  32. Munne, K.; Bhanothu, V.; Bhor, V.; Patel, V.; Mahale, S.D.; Pande, S. Detection of SARS-CoV-2 infection by RT-PCR test: Factors influencing interpretation of results. Virusdisease 2021, 32, 187–189. [Google Scholar] [CrossRef] [PubMed]
  33. Lamers, M.M.; Beumer, J.; van der Vaart, J.; Knoops, K.; Puschhof, J.; Breugem, T.I.; Ravelli, R.B.G.; Paul van Schayck, J.; Mykytyn, A.Z.; Duimel, H.Q.; et al. SARS-CoV-2 productively infects human gut enterocytes. Science 2020, 369, 50–54. [Google Scholar] [CrossRef] [PubMed]
  34. 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] [PubMed]
  35. Zhou, P.; Yang, X.L.; Wang, X.G.; Hu, B.; Zhang, L.; Zhang, W.; Si, H.R.; Zhu, Y.; Li, B.; Huang, C.L.; et al. A pneumonia outbreak associated with a new coronavirus of probable bat origin. Nature 2020, 579, 270–273. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  36. Cheung, S.; Delgado Fuentes, A.; Fetterman, A.D. Recurrent Acute Pancreatitis in a Patient with COVID-19 Infection. Am. J. Case Rep. 2020, 21, e927076. [Google Scholar] [CrossRef] [PubMed]
  37. Kipkorir, V.; Cheruiyot, I.; Ngure, B.; Misiani, M.; Munguti, J. Prolonged SARS-CoV-2 RNA detection in anal/rectal swabs and stool specimens in COVID-19 patients after negative conversion in nasopharyngeal RT-PCR test. J. Med. Virol. 2020, 92, 2328–2331. [Google Scholar] [CrossRef] [PubMed]
  38. Chen, T.H.; Hsu, M.T.; Lee, M.Y.; Chou, C.K. Gastrointestinal Involvement in SARS-CoV-2 Infection. Viruses 2022, 14, 1188. [Google Scholar] [CrossRef]
  39. Amirian, E.S. Potential fecal transmission of SARS-CoV-2: Current evidence and implications for public health. Int. J. Infect. Dis. 2020, 95, 363–370. [Google Scholar] [CrossRef]
  40. Chen, Y.; Chen, L.; Deng, Q.; Zhang, G.; Wu, K.; Ni, L.; Yang, Y.; Liu, B.; Wang, W.; Wei, C.; 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] [Green Version]
  41. 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]
  42. Peng, L.; Liu, J.; Xu, W.; Luo, Q.; Chen, D.; Lei, Z.; Huang, Z.; Li, X.; Deng, K.; Lin, B.; et al. SARS-CoV-2 can be detected in urine, blood, anal swabs, and oropharyngeal swabs specimens. J. Med. Virol. 2020, 92, 1676–1680. [Google Scholar] [CrossRef] [PubMed]
  43. Tian, Y.; Rong, L.; Nian, W.; He, Y. Review article: Gastrointestinal features in COVID-19 and the possibility of faecal transmission. Aliment. Pharmacol. Ther. 2020, 51, 843–851. [Google Scholar] [CrossRef] [PubMed]
  44. Ong, S.W.X.; Tan, Y.K.; Chia, P.Y.; Lee, T.H.; Ng, O.T.; Wong, M.S.Y.; Marimuthu, K. Air, Surface Environmental, and Personal Protective Equipment Contamination by Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2) from a Symptomatic Patient. JAMA 2020, 323, 1610–1612. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  45. Avgeris, M.; Adamopoulos, P.G.; Galani, A.; Xagorari, M.; Gourgiotis, D.; Trougakos, I.P.; Voulgaris, N.; Dimopoulos, M.A.; Thomaidis, N.S.; Scorilas, A. Novel Nested-Seq Approach for SARS-CoV-2 Real-Time Epidemiology and In-Depth Mutational Profiling in Wastewater. Int. J. Mol. Sci. 2021, 22, 8498. [Google Scholar] [CrossRef]
  46. Itarte, M.; Bofill-Mas, S.; Martínez-Puchol, S.; Torrell, H.; Ceretó, A.; Carrasco, M.; Forés, E.; Canela, N.; Girones, R.; Rusiñol, M. Looking for a needle in a haystack. SARS-CoV-2 variant characterization in sewage. Curr. Opin. Environ. Sci. Health 2021, 24, 100308. [Google Scholar] [CrossRef]
  47. Anderson, E.J.; Weber, S.G. Rotavirus infection in adults. Lancet Infect. Dis. 2004, 4, 91–99. [Google Scholar] [CrossRef]
  48. Atmar, R.L.; Opekun, A.R.; Gilger, M.A.; Estes, M.K.; Crawford, S.E.; Neill, F.H.; Graham, D.Y. Norwalk virus shedding after experimental human infection. Emerg. Infect. Dis. 2008, 14, 1553–1557. [Google Scholar] [CrossRef]
  49. De Graaf, M.; Beck, R.; Caccio, S.M.; Duim, B.; Fraaij, P.; Le Guyader, F.S.; Lecuit, M.; Le Pendu, J.; de Wit, E.; Schultsz, C. Sustained fecal-oral human-to-human transmission following a zoonotic event. Curr. Opin. Virol. 2017, 22, 1–6. [Google Scholar] [CrossRef]
  50. Ma, C.; Cong, Y.; Zhang, H. COVID-19 and the Digestive System. Am. J. Gastroenterol. 2020, 115, 1003–1006. [Google Scholar] [CrossRef]
  51. Yeo, C.; Kaushal, S.; Yeo, D. Enteric involvement of coronaviruses: Is faecal-oral transmission of SARS-CoV-2 possible? Lancet Gastroenterol. Hepatol. 2020, 5, 335–337. [Google Scholar] [CrossRef]
  52. Huang, N.; Pérez, P.; Kato, T.; Mikami, Y.; Okuda, K.; Gilmore, R.C.; Conde, C.D.; Gasmi, B.; Stein, S.; Beach, M.; et al. SARS-CoV-2 infection of the oral cavity and saliva. Nat. Med. 2021, 27, 892–903. [Google Scholar] [CrossRef] [PubMed]
  53. To, K.K.; Tsang, O.T.; Yip, C.C.; Chan, K.H.; Wu, T.C.; Chan, J.M.; Leung, W.S.; Chik, T.S.; Choi, C.Y.; Kandamby, D.H.; et al. Consistent Detection of 2019 Novel Coronavirus in Saliva. Clin. Infect. Dis. Off. Publ. Infect. Dis. Soc. Am. 2020, 71, 841–843. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  54. Jeang, K.-T. HIV-1: Molecular Biology and Pathogenesis Viral Mechanisms, 2nd ed.; Elsevier: Amsterdam, The Netherlands, 2007; Volume 55. [Google Scholar]
  55. Hariyanto, T.I.; Prasetya, I.B.; Kurniawan, A. Proton pump inhibitor use is associated with increased risk of severity and mortality from coronavirus disease 2019 (COVID-19) infection. Dig. Liver Dis. 2020, 52, 1410–1412. [Google Scholar] [CrossRef] [PubMed]
  56. Liu, J.J.; Sloan, M.E.; Owings, A.H.; Figgins, E.; Gauthier, J.; Gharaibeh, R.; Robinson, T.; Williams, H.; Sindel, C.B.; Backus, F.; et al. Increased ACE2 Levels and Mortality Risk of Patients With COVID-19 on Proton Pump Inhibitor Therapy. Am. J. Gastroenterol. 2021, 116, 1638–1645. [Google Scholar] [CrossRef]
  57. Yozgat, A.; Kasapoğlu, B.; Can, G.; Tanoğlu, A.; Sakin, Y.S.; Yalçın, K.S.; Gürler, M.; Kaplan, M.; Kaban, M.G.; Kırsoy, M.; et al. Long-term proton pump inhibitor use is a risk factor for mortality in patients hospitalized for COVID-19. Turk. J. Med. Sci. 2021, 51, 1675–1681. [Google Scholar] [CrossRef]
  58. Martinsen, T.C.; Fossmark, R.; Waldum, H.L. The Phylogeny and Biological Function of Gastric Juice-Microbiological Consequences of Removing Gastric Acid. Int. J. Mol. Sci. 2019, 20, 6031. [Google Scholar] [CrossRef] [Green Version]
  59. Atlas, H.P. Tissue Expression of ace2—Summary—The Human Protein Atlas. Available online: https://www.proteinatlas.org/ENSG00000130234-ACE2/tissue (accessed on 17 November 2022).
  60. Hamming, I.; Timens, W.; Bulthuis, M.L.; Lely, A.T.; Navis, G.; van Goor, H. Tissue distribution of ACE2 protein, the functional receptor for SARS coronavirus. A first step in understanding SARS pathogenesis. J. Pathol. 2004, 203, 631–637. [Google Scholar] [CrossRef]
  61. Li, M.Y.; Li, L.; Zhang, Y.; Wang, X.S. Expression of the SARS-CoV-2 cell receptor gene ACE2 in a wide variety of human tissues. Infect. Dis. Poverty 2020, 9, 45. [Google Scholar] [CrossRef]
  62. 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]
  63. Lee, J.J.; Kopetz, S.; Vilar, E.; Shen, J.P.; Chen, K.; Maitra, A. Relative Abundance of SARS-CoV-2 Entry Genes in the Enterocytes of the Lower Gastrointestinal Tract. Genes 2020, 11, 645. [Google Scholar] [CrossRef]
  64. Flint, S.J. (Ed.) Principles of Virology, 3rd ed.; ASM Press: Washington, DC, USA, 2009; Volume 2, p. 419. [Google Scholar]
  65. Siebers, A.; Finlay, B.B. M cells and the pathogenesis of mucosal and systemic infections. Trends Microbiol. 1996, 4, 22–29. [Google Scholar] [CrossRef] [PubMed]
  66. Hörnich, B.F.; Großkopf, A.K.; Schlagowski, S.; Tenbusch, M.; Kleine-Weber, H.; Neipel, F.; Stahl-Hennig, C.; Hahn, A.S. SARS-CoV-2 and SARS-CoV Spike-Mediated Cell-Cell Fusion Differ in Their Requirements for Receptor Expression and Proteolytic Activation. J. Virol. 2021, 95, e00002-21. [Google Scholar] [CrossRef] [PubMed]
  67. Mao, J.; Lin, E.; He, L.; Yu, J.; Tan, P.; Zhou, Y. Autophagy and Viral Infection. Adv. Exp. Med. Biol. 2019, 1209, 55–78. [Google Scholar] [CrossRef]
  68. Rautou, P.E.; Cazals-Hatem, D.; Feldmann, G.; Mansouri, A.; Grodet, A.; Barge, S.; Martinot-Peignoux, M.; Duces, A.; Bièche, I.; Lebrec, D.; et al. Changes in autophagic response in patients with chronic hepatitis C virus infection. Am. J. Pathol. 2011, 178, 2708–2715. [Google Scholar] [CrossRef] [Green Version]
  69. Sir, D.; Chen, W.L.; Choi, J.; Wakita, T.; Yen, T.S.; Ou, J.H. Induction of incomplete autophagic response by hepatitis C virus via the unfolded protein response. Hepatology 2008, 48, 1054–1061. [Google Scholar] [CrossRef] [Green Version]
  70. Aydin, Y.; Koksal, A.R.; Reddy, V.; Lin, D.; Osman, H.; Heidari, Z.; Rhadhi, S.M.; Wimley, W.C.; Parsi, M.A.; Dash, S. Extracellular Vesicle Release Promotes Viral Replication during Persistent HCV Infection. Cells 2021, 10, 984. [Google Scholar] [CrossRef] [PubMed]
  71. Shima Fakher, P.P.; Ghavami, S.; Mokarram, P. The Role of Autophagy in Respiratory Complications of COVID-19. Shiraz E-Med. J. 2020, 21, 3. [Google Scholar] [CrossRef] [Green Version]
  72. Foerster, E.G.; Mukherjee, T.; Cabral-Fernandes, L.; Rocha, J.D.B.; Girardin, S.E.; Philpott, D.J. How autophagy controls the intestinal epithelial barrier. Autophagy 2022, 18, 86–103. [Google Scholar] [CrossRef] [PubMed]
  73. Maier, H.J.; Britton, P. Involvement of autophagy in coronavirus replication. Viruses 2012, 4, 3440–3451. [Google Scholar] [CrossRef]
  74. Shang, C.; Zhuang, X.; Zhang, H.; Li, Y.; Zhu, Y.; Lu, J.; Ge, C.; Cong, J.; Li, T.; Li, N.; et al. Inhibition of Autophagy Suppresses SARS-CoV-2 Replication and Ameliorates Pneumonia in hACE2 Transgenic Mice and Xenografted Human Lung Tissues. J. Virol. 2021, 95, e01537-21. [Google Scholar] [CrossRef]
  75. Tanida, I.; Minematsu-Ikeguchi, N.; Ueno, T.; Kominami, E. Lysosomal turnover, but not a cellular level, of endogenous LC3 is a marker for autophagy. Autophagy 2005, 1, 84–91. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  76. Kabeya, Y.; Mizushima, N.; Ueno, T.; Yamamoto, A.; Kirisako, T.; Noda, T.; Kominami, E.; Ohsumi, Y.; Yoshimori, T. LC3, a mammalian homologue of yeast Apg8p, is localized in autophagosome membranes after processing. EMBO J. 2000, 19, 5720–5728. [Google Scholar] [CrossRef] [PubMed]
  77. Saramago, M.; Costa, V.G.; Souza, C.S.; Bárria, C.; Domingues, S.; Viegas, S.C.; Lousa, D.; Soares, C.M.; Arraiano, C.M.; Matos, R.G. The nsp15 Nuclease as a Good Target to Combat SARS-CoV-2: Mechanism of Action and Its Inactivation with FDA-Approved Drugs. Microorganisms 2022, 10, 342. [Google Scholar] [CrossRef] [PubMed]
  78. Hayn, M.; Hirschenberger, M.; Koepke, L.; Nchioua, R.; Straub, J.H.; Klute, S.; Hunszinger, V.; Zech, F.; Prelli Bozzo, C.; Aftab, W.; et al. Systematic functional analysis of SARS-CoV-2 proteins uncovers viral innate immune antagonists and remaining vulnerabilities. Cell Rep. 2021, 35, 109126. [Google Scholar] [CrossRef]
  79. Habibzadeh, P.; Dastsooz, H.; Eshraghi, M.; Łos, M.J.; Klionsky, D.J.; Ghavami, S. Autophagy: The Potential Link between SARS-CoV-2 and Cancer. Cancers 2021, 13, 5721. [Google Scholar] [CrossRef]
  80. Barbati, C.; Celia, A.I.; Colasanti, T.; Vomero, M.; Speziali, M.; Putro, E.; Buoncuore, G.; Savino, F.; Colafrancesco, S.; Ucci, F.M.; et al. Autophagy Hijacking in PBMC From COVID-19 Patients Results in Lymphopenia. Front. Immunol. 2022, 13, 903498. [Google Scholar] [CrossRef]
  81. Nguyen, H.T.; Falzarano, D.; Gerdts, V.; Liu, Q. Construction of a Noninfectious SARS-CoV-2 Replicon for Antiviral-Drug Testing and Gene Function Studies. J. Virol. 2021, 95, e0068721. [Google Scholar] [CrossRef]
  82. Kumar, S.; Kashyap, P.; Chowdhury, S.; Kumar, S.; Panwar, A.; Kumar, A. Identification of phytochemicals as potential therapeutic agents that binds to Nsp15 protein target of coronavirus (SARS-CoV-2) that are capable of inhibiting virus replication. Phytomed. Int. J. Phytother. Phytopharm. 2021, 85, 153317. [Google Scholar] [CrossRef]
  83. Koepke, L.; Hirschenberger, M.; Hayn, M.; Kirchhoff, F.; Sparrer, K.M. Manipulation of autophagy by SARS-CoV-2 proteins. Autophagy 2021, 17, 2659–2661. [Google Scholar] [CrossRef]
  84. Su, W.Q.; Yu, X.J.; Zhou, C.M. SARS-CoV-2 ORF3a Induces Incomplete Autophagy via the Unfolded Protein Response. Viruses 2021, 13, 2467. [Google Scholar] [CrossRef]
  85. Miao, G.; Zhao, H.; Li, Y.; Ji, M.; Chen, Y.; Shi, Y.; Bi, Y.; Wang, P.; Zhang, H. ORF3a of the COVID-19 virus SARS-CoV-2 blocks HOPS complex-mediated assembly of the SNARE complex required for autolysosome formation. Dev. Cell 2021, 56, 427–442.e425. [Google Scholar] [CrossRef] [PubMed]
  86. Qu, Y.; Wang, X.; Zhu, Y.; Wang, W.; Wang, Y.; Hu, G.; Liu, C.; Li, J.; Ren, S.; Xiao, M.Z.X.; et al. ORF3a-Mediated Incomplete Autophagy Facilitates Severe Acute Respiratory Syndrome Coronavirus-2 Replication. Front. Cell Dev. Biol. 2021, 9, 716208. [Google Scholar] [CrossRef] [PubMed]
  87. Zhang, Y.; Chen, Y.; Li, Y.; Huang, F.; Luo, B.; Yuan, Y.; Xia, B.; Ma, X.; Yang, T.; Yu, F.; et al. The ORF8 protein of SARS-CoV-2 mediates immune evasion through down-regulating MHC-Ι. Proc. Natl. Acad. Sci. USA 2021, 118, e2024202118. [Google Scholar] [CrossRef] [PubMed]
  88. Ghosh, S.; Dellibovi-Ragheb, T.A.; Kerviel, A.; Pak, E.; Qiu, Q.; Fisher, M.; Takvorian, P.M.; Bleck, C.; Hsu, V.W.; Fehr, A.R.; et al. β-Coronaviruses Use Lysosomes for Egress Instead of the Biosynthetic Secretory Pathway. Cell 2020, 183, 1520–1535.e1514. [Google Scholar] [CrossRef]
  89. Samimi, N.; Farjam, M.; Klionsky, D.J.; Rezaei, N. The role of autophagy in the pathogenesis of SARS-CoV-2 infection in different cell types. Autophagy 2022, 18, 1728–1731. [Google Scholar] [CrossRef] [PubMed]
  90. Gupta, M.; Shin, D.M.; Ramakrishna, L.; Goussetis, D.J.; Platanias, L.C.; Xiong, H.; Morse, H.C., III; Ozato, K. IRF8 directs stress-induced autophagy in macrophages and promotes clearance of Listeria monocytogenes. Nat. Commun. 2015, 6, 6379. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  91. Moorman, H.R.; Reategui, Y.; Poschel, D.B.; Liu, K. IRF8: Mechanism of Action and Health Implications. Cells 2022, 11, 2630. [Google Scholar] [CrossRef]
  92. Tomić, S.; Đokić, J.; Stevanović, D.; Ilić, N.; Gruden-Movsesijan, A.; Dinić, M.; Radojević, D.; Bekić, M.; Mitrović, N.; Tomašević, R.; et al. Reduced Expression of Autophagy Markers and Expansion of Myeloid-Derived Suppressor Cells Correlate with Poor T Cell Response in Severe COVID-19 Patients. Front. Immunol. 2021, 12, 614599. [Google Scholar] [CrossRef]
  93. 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.e2434. [Google Scholar] [CrossRef]
  94. Gassen, N.C.; Papies, J.; Bajaj, T.; Emanuel, J.; Dethloff, F.; Chua, R.L.; Trimpert, J.; Heinemann, N.; Niemeyer, C.; Weege, F.; et al. SARS-CoV-2-mediated dysregulation of metabolism and autophagy uncovers host-targeting antivirals. Nat. Commun. 2021, 12, 3818. [Google Scholar] [CrossRef]
  95. Li, F.; Li, J.; Wang, P.-H.; Yang, N.; Huang, J.; Ou, J.; Xu, T.; Zhao, X.; Liu, T.; Huang, X.; et al. SARS-CoV-2 spike promotes inflammation and apoptosis through autophagy by ROS-suppressed PI3K/AKT/mTOR signaling. Biochim. Biophys. Acta Mol. Basis Dis. 2021, 1867, 166260. [Google Scholar] [CrossRef]
  96. Dupont, N.; Jiang, S.; Pilli, M.; Ornatowski, W.; Bhattacharya, D.; Deretic, V. Autophagy-based unconventional secretory pathway for extracellular delivery of IL-1β. EMBO J. 2011, 30, 4701–4711. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  97. Juibari, A.D.; Rezadoost, M.H. The key role of Calpain in COVID-19 as a therapeutic strategy. Inflammopharmacology 2022, 30, 1479–1491. [Google Scholar] [CrossRef] [PubMed]
  98. Zhou, R.; Yazdi, A.S.; Menu, P.; Tschopp, J. A role for mitochondria in NLRP3 inflammasome activation. Nature 2011, 469, 221–225. [Google Scholar] [CrossRef] [PubMed]
  99. Boroumand-Noughabi, S.; Khoshnegah, Z.; Amel Jamehdar, S.; Ayatollahi, H.; Sheikhi, M.; Rostami, M.; Keramati, M.R. Deregulation of the Expression of Beclin1 and Light Chain 3(LC3), Autophagy-Related Genes, in COVID-19 Patients. Med. J. Islam. Repub. Iran 2022, 36, 99. [Google Scholar] [CrossRef]
  100. Li, K.; Hao, Z.; Zhao, X.; Du, J.; Zhou, Y. SARS-CoV-2 infection-induced immune responses: Friends or foes? Scand. J. Immunol. 2020, 92, e12895. [Google Scholar] [CrossRef]
  101. De Oliveira, A.P.; Lopes, A.L.F.; Pacheco, G.; de Sá Guimarães Nolêto, I.R.; Nicolau, L.A.D.; Medeiros, J.V.R. Premises among SARS-CoV-2, dysbiosis and diarrhea: Walking through the ACE2/mTOR/autophagy route. Med. Hypotheses 2020, 144, 110243. [Google Scholar] [CrossRef]
  102. Mutsafi, Y.; Altan-Bonnet, N. Enterovirus Transmission by Secretory Autophagy. Viruses 2018, 10, 139. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  103. Moustafa, A.; Khalel, R.S.; Aziz, R.K. Traces of SARS-CoV-2 RNA in Peripheral Blood Cells of Patients with COVID-19. OMICS J. Integr. Biol. 2021, 25, 475–483. [Google Scholar] [CrossRef]
  104. Richter, E.; Al Arashi, D.; Schulte, B.; Bode, C.; Marx, B.; Aldabbagh, S.; Schlüter, C.; Kümmerer, B.M.; Oldenburg, J.; Funk, M.B.; et al. Detectable SARS-CoV-2 RNAemia in Critically Ill Patients, but Not in Mild and Asymptomatic Infections. Transfus. Med. Hemother. 2021, 48, 154–160. [Google Scholar] [CrossRef]
  105. Sbarigia, C.; Vardanyan, D.; Buccini, L.; Tacconi, S.; Dini, L. SARS-CoV-2 and extracellular vesicles: An intricate interplay in pathogenesis, diagnosis and treatment. Front. Nanotechnol. 2022, 4, 987034. [Google Scholar] [CrossRef]
  106. Altan-Bonnet, N.; Perales, C.; Domingo, E. Extracellular vesicles: Vehicles of en bloc viral transmission. Virus Res. 2019, 265, 143–149. [Google Scholar] [CrossRef] [PubMed]
  107. Santiana, M.; Ghosh, S.; Ho, B.A.; Rajasekaran, V.; Du, W.-L.; Mutsafi, Y.; De Jésus-Diaz, D.A.; Sosnovtsev, S.V.; Levenson, E.A.; Parra, G.I.; et al. Vesicle-Cloaked Virus Clusters Are Optimal Units for Inter-organismal Viral Transmission. Cell Host Microbe 2018, 24, 208–220.e208. [Google Scholar] [CrossRef] [Green Version]
  108. Chen, Y.H.; Du, W.; Hagemeijer, M.C.; Takvorian, P.M.; Pau, C.; Cali, A.; Brantner, C.A.; Stempinski, E.S.; Connelly, P.S.; Ma, H.C.; et al. Phosphatidylserine vesicles enable efficient en bloc transmission of enteroviruses. Cell 2015, 160, 619–630. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  109. Bird, S.W.; Maynard, N.D.; Covert, M.W.; Kirkegaard, K. Nonlytic viral spread enhanced by autophagy components. Proc. Natl. Acad. Sci. USA 2014, 111, 13081–13086. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  110. Jackson, W.T.; Giddings, T.H., Jr.; Taylor, M.P.; Mulinyawe, S.; Rabinovitch, M.; Kopito, R.R.; Kirkegaard, K. Subversion of cellular autophagosomal machinery by RNA viruses. PLoS Biol. 2005, 3, e156. [Google Scholar] [CrossRef] [Green Version]
  111. Fleming, A.; Sampey, G.; Chung, M.C.; Bailey, C.; van Hoek, M.L.; Kashanchi, F.; Hakami, R.M. The carrying pigeons of the cell: Exosomes and their role in infectious diseases caused by human pathogens. Pathog. Dis. 2014, 71, 109–120. [Google Scholar] [CrossRef] [Green Version]
  112. Wang, J.; Chen, S.; Bihl, J. Exosome-Mediated Transfer of ACE2 (Angiotensin-Converting Enzyme 2) from Endothelial Progenitor Cells Promotes Survival and Function of Endothelial Cell. Oxid. Med. Cell Longev. 2020, 2020, 4213541. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  113. Sur, S.; Khatun, M.; Steele, R.; Isbell, T.S.; Ray, R.; Ray, R.B. Exosomes from COVID-19 Patients Carry Tenascin-C and Fibrinogen-beta in Triggering Inflammatory Signals in Cells of Distant Organ. Int. J. Mol. Sci. 2021, 22, 3184. [Google Scholar] [CrossRef]
  114. Kongsomros, S.; Suksatu, A.; Kanjanasirirat, P.; Manopwisedjaroen, S.; Prasongtanakij, S.; Jearawuttanakul, K.; Borwornpinyo, S.; Hongeng, S.; Thitithanyanont, A.; Chutipongtanate, S. Anti-SARS-CoV-2 Activity of Extracellular Vesicle Inhibitors: Screening, Validation, and Combination with Remdesivir. Biomedicines 2021, 9, 1230. [Google Scholar] [CrossRef]
  115. Feng, Y.; Ling, Y.; Bai, T.; Xie, Y.; Huang, J.; Li, J.; Xiong, W.; Yang, D.; Chen, R.; Lu, F.; et al. COVID-19 with Different Severities: A Multicenter Study of Clinical Features. Am. J. Respir. Crit. Care Med. 2020, 201, 1380–1388. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  116. Huang, K.J.; Su, I.J.; Theron, M.; Wu, Y.C.; Lai, S.K.; Liu, C.C.; Lei, H.Y. An interferon-gamma-related cytokine storm in SARS patients. J. Med. Virol. 2005, 75, 185–194. [Google Scholar] [CrossRef] [PubMed]
  117. Karki, R.; Sharma, B.R.; Tuladhar, S.; Williams, E.P.; Zalduondo, L.; Samir, P.; Zheng, M.; Sundaram, B.; Banoth, B.; Malireddi, R.K.S.; et al. Synergism of TNF-α and IFN-γ Triggers Inflammatory Cell Death, Tissue Damage, and Mortality in SARS-CoV-2 Infection and Cytokine Shock Syndromes. Cell 2021, 184, 149–168.e117. [Google Scholar] [CrossRef] [PubMed]
  118. Ruan, Q.; Yang, K.; Wang, W.; Jiang, L.; Song, J. Clinical predictors of mortality due to COVID-19 based on an analysis of data of 150 patients from Wuhan, China. Intensive Care Med. 2020, 46, 846–848. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  119. Zhang, H.; Shao, B.; Dang, Q.; Chen, Z.; Zhou, Q.; Luo, H.; Yuan, W.; Sun, Z. Pathogenesis and Mechanism of Gastrointestinal Infection with COVID-19. Front. Immunol. 2021, 12, 674074. [Google Scholar] [CrossRef]
  120. Ntyonga-Pono, M.P. COVID-19 infection and oxidative stress: An under-explored approach for prevention and treatment? Pan Afr. Med. J. 2020, 35, 12. [Google Scholar] [CrossRef]
  121. Zhou, T.; Wu, J.; Zeng, Y.; Li, J.; Yan, J.; Meng, W.; Han, H.; Feng, F.; He, J.; Zhao, S.; et al. SARS-CoV-2 triggered oxidative stress and abnormal energy metabolism in gut microbiota. MedComm 2022, 3, e112. [Google Scholar] [CrossRef]
  122. Zhang, C.; Wu, Z.; Li, J.W.; Zhao, H.; Wang, G.Q. Cytokine release syndrome in severe COVID-19: Interleukin-6 receptor antagonist tocilizumab may be the key to reduce mortality. Int. J. Antimicrob. Agents 2020, 55, 105954. [Google Scholar] [CrossRef]
  123. Liu, J.; Li, S.; Liu, J.; Liang, B.; Wang, X.; Wang, H.; Li, W.; Tong, Q.; Yi, J.; Zhao, L.; et al. Longitudinal characteristics of lymphocyte responses and cytokine profiles in the peripheral blood of SARS-CoV-2 infected patients. EBioMedicine 2020, 55, 102763. [Google Scholar] [CrossRef]
  124. Kuppalli, K.; Rasmussen, A.L. A glimpse into the eye of the COVID-19 cytokine storm. EBioMedicine 2020, 55, 102789. [Google Scholar] [CrossRef]
  125. 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] [PubMed]
  126. Yokoyama, Y.; Ichiki, T.; Yamakawa, T.; Tsuji, Y.; Kuronuma, K.; Takahashi, S.; Narimatsu, E.; Nakase, H. Impaired tryptophan metabolism in the gastrointestinal tract of patients with critical coronavirus disease 2019. Front. Med. 2022, 9, 941422. [Google Scholar] [CrossRef] [PubMed]
  127. Sorgdrager, F.J.H.; Naudé, P.J.W.; Kema, I.P.; Nollen, E.A.; Deyn, P.P. Tryptophan Metabolism in Inflammaging: From Biomarker to Therapeutic Target. Front. Immunol. 2019, 10, 2565. [Google Scholar] [CrossRef] [PubMed]
  128. Stanifer, M.L.; Guo, C.; Doldan, P.; Boulant, S. Importance of Type I and III Interferons at Respiratory and Intestinal Barrier Surfaces. Front. Immunol. 2020, 11, 608645. [Google Scholar] [CrossRef]
  129. Lee, J.-H.; Koepke, L.; Kirchhoff, F.; Sparrer, K.M.J. Interferon antagonists encoded by SARS-CoV-2 at a glance. Med. Microbiol. Immunol. 2022, 1–7. [Google Scholar] [CrossRef]
  130. Lei, X.; Dong, X.; Ma, R.; Wang, W.; Xiao, X.; Tian, Z.; Wang, C.; Wang, Y.; Li, L.; Ren, L.; et al. Activation and evasion of type I interferon responses by SARS-CoV-2. Nat. Commun. 2020, 11, 3810. [Google Scholar] [CrossRef]
  131. Hsu, J.C.; Laurent-Rolle, M.; Pawlak, J.B.; Wilen, C.B.; Cresswell, P. Translational shutdown and evasion of the innate immune response by SARS-CoV-2 NSP14 protein. Proc. Natl. Acad. Sci. USA 2021, 118, e2101161118. [Google Scholar] [CrossRef] [PubMed]
  132. Low, Z.Y.; Zabidi, N.Z.; Yip, A.J.W.; Puniyamurti, A.; Chow, V.T.K.; Lal, S.K. SARS-CoV-2 Non-Structural Proteins and Their Roles in Host Immune Evasion. Viruses 2022, 14, 1991. [Google Scholar] [CrossRef]
  133. Oh, S.J.; Shin, O.S. SARS-CoV-2-mediated evasion strategies for antiviral interferon pathways. J. Microbiol. 2022, 60, 290–299. [Google Scholar] [CrossRef]
  134. Neurath, M.F. COVID-19 and immunomodulation in IBD. Gut 2020, 69, 1335–1342. [Google Scholar] [CrossRef]
  135. Chi, Y.; Ge, Y.; Wu, B.; Zhang, W.; Wu, T.; Wen, T.; Liu, J.; Guo, X.; Huang, C.; Jiao, Y.; et al. Serum Cytokine and Chemokine Profile in Relation to the Severity of Coronavirus Disease 2019 in China. J. Infect. Dis. 2020, 222, 746–754. [Google Scholar] [CrossRef] [PubMed]
  136. Liu, Y.; Zhang, C.; Huang, F.; Yang, Y.; Wang, F.; Yuan, J.; Zhang, Z.; Qin, Y.; Li, X.; Zhao, D.; et al. Elevated plasma levels of selective cytokines in COVID-19 patients reflect viral load and lung injury. Natl. Sci. Rev. 2020, 7, 1003–1011. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  137. 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] [PubMed]
  138. Zhang, M.L.; Jacobsen, F.; Pepe-Mooney, B.J.; Mino-Kenudson, M.; Deshpande, V.; Shih, A.R.; Mattia, A.R.; Goessling, W.; Hwabejire, J.O.; Velmahos, G.C.; et al. Clinicopathological findings in patients with COVID-19-associated ischaemic enterocolitis. Histopathology 2021, 79, 1004–1017. [Google Scholar] [CrossRef] [PubMed]
  139. Xu, Z.; Shi, L.; Wang, Y.; Zhang, J.; Huang, L.; Zhang, C.; Liu, S.; Zhao, P.; Liu, H.; Zhu, L.; et al. Pathological findings of COVID-19 associated with acute respiratory distress syndrome. Lancet Respir. Med. 2020, 8, 420–422. [Google Scholar] [CrossRef]
  140. Galanopoulos, M.; Gkeros, F.; Doukatas, A.; Karianakis, G.; Pontas, C.; Tsoukalas, N.; Viazis, N.; Liatsos, C.; Mantzaris, G.J. COVID-19 pandemic: Pathophysiology and manifestations from the gastrointestinal tract. World J. Gastroenterol. 2020, 26, 4579–4588. [Google Scholar] [CrossRef]
  141. Veglia, F.; Sanseviero, E.; Gabrilovich, D.I. Myeloid-derived suppressor cells in the era of increasing myeloid cell diversity. Nat. Rev. Immunol. 2021, 21, 485–498. [Google Scholar] [CrossRef]
  142. Delano, M.J.; Scumpia, P.O.; Weinstein, J.S.; Coco, D.; Nagaraj, S.; Kelly-Scumpia, K.M.; O’Malley, K.A.; Wynn, J.L.; Antonenko, S.; Al-Quran, S.Z.; et al. MyD88-dependent expansion of an immature GR-1(+)CD11b(+) population induces T cell suppression and Th2 polarization in sepsis. J. Exp. Med. 2007, 204, 1463–1474. [Google Scholar] [CrossRef]
  143. Sinha, P.; Clements, V.K.; Bunt, S.K.; Albelda, S.M.; Ostrand-Rosenberg, S. Cross-talk between myeloid-derived suppressor cells and macrophages subverts tumor immunity toward a type 2 response. J. Immunol. 2007, 179, 977–983. [Google Scholar] [CrossRef] [Green Version]
  144. Zhao, F.; Gong, W.; Song, J.; Shen, Z.; Cui, D. The paradoxical role of MDSCs in inflammatory bowel diseases: From bench to bedside. Front. Immunol. 2022, 13, 1021634. [Google Scholar] [CrossRef]
  145. Dean, M.J.; Ochoa, J.B.; Sanchez-Pino, M.D.; Zabaleta, J.; Garai, J.; Del Valle, L.; Wyczechowska, D.; Baiamonte, L.B.; Philbrook, P.; Majumder, R.; et al. Severe COVID-19 Is Characterized by an Impaired Type I Interferon Response and Elevated Levels of Arginase Producing Granulocytic Myeloid Derived Suppressor Cells. Front. Immunol. 2021, 12, 695972. [Google Scholar] [CrossRef] [PubMed]
  146. Rowlands, M.; Segal, F.; Hartl, D. Myeloid-Derived Suppressor Cells as a Potential Biomarker and Therapeutic Target in COVID-19. Front. Immunol. 2021, 12, 697405. [Google Scholar] [CrossRef] [PubMed]
  147. Bedoya, S.A.; Ye, F.; Best, V.; Ramilo, O.; Ramilo, O.; Mejias, A. 2783. Expansion of Monocytic Myeloid-Derived Suppressor Cells in Infants with Severe Respiratory Syncytial Virus (RSV) Infection. Open Forum Infect. Dis. 2019, 6, S983. [Google Scholar] [CrossRef] [Green Version]
  148. Beliakova-Bethell, N.; Maruthai, K.; Xu, R.; Salvador, L.C.M.; Garg, A. Monocytic-Myeloid Derived Suppressor Cells Suppress T-Cell Responses in Recovered SARS-CoV-2-Infected Individuals. Front. Immunol. 2022, 13, 894543. [Google Scholar] [CrossRef] [PubMed]
  149. McDaniel, M.M.; Kottyan, L.C.; Singh, H.; Pasare, C. Suppression of Inflammasome Activation by IRF8 and IRF4 in cDCs Is Critical for T Cell Priming. Cell Rep. 2020, 31, 107604. [Google Scholar] [CrossRef] [PubMed]
  150. Vatansever, H.S.; Becer, E. Relationship between IL-6 and COVID-19: To be considered during treatment. Future Virol. 2020, 15, 817–822. [Google Scholar] [CrossRef]
  151. Waight, J.D.; Netherby, C.; Hensen, M.L.; Miller, A.; Hu, Q.; Liu, S.; Bogner, P.N.; Farren, M.R.; Lee, K.P.; Liu, K.; et al. Myeloid-derived suppressor cell development is regulated by a STAT/IRF-8 axis. J. Clin. Investig. 2013, 123, 4464–4478. [Google Scholar] [CrossRef] [PubMed]
  152. Lee, H.K.; Lund, J.M.; Ramanathan, B.; Mizushima, N.; Iwasaki, A. Autophagy-dependent viral recognition by plasmacytoid dendritic cells. Science 2007, 315, 1398–1401. [Google Scholar] [CrossRef] [Green Version]
  153. Jiménez-Cortegana, C.; Liró, J.; Palazón-Carrión, N.; Salamanca, E.; Sojo-Dorado, J.; de la Cruz-Merino, L.; Pascual, Á.; Rodríguez-Baño, J.; Sánchez-Margalet, V. Increased Blood Monocytic Myeloid Derived Suppressor Cells but Low Regulatory T Lymphocytes in Patients with Mild COVID-19. Viral Immunol. 2021, 34, 639–645. [Google Scholar] [CrossRef]
  154. Reizine, F.; Lesouhaitier, M.; Gregoire, M.; Pinceaux, K.; Gacouin, A.; Maamar, A.; Painvin, B.; Camus, C.; Le Tulzo, Y.; Tattevin, P.; et al. SARS-CoV-2-Induced ARDS Associates with MDSC Expansion, Lymphocyte Dysfunction, and Arginine Shortage. J. Clin. Immunol. 2021, 41, 515–525. [Google Scholar] [CrossRef]
  155. Rodriguez, P.C.; Hernandez, C.P.; Morrow, K.; Sierra, R.; Zabaleta, J.; Wyczechowska, D.D.; Ochoa, A.C. L-arginine deprivation regulates cyclin D3 mRNA stability in human T cells by controlling HuR expression. J. Immunol. 2010, 185, 5198–5204. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  156. Zhu, X.; Pribis, J.P.; Rodriguez, P.C.; Morris, S.M., Jr.; Vodovotz, Y.; Billiar, T.R.; Ochoa, J.B. The central role of arginine catabolism in T-cell dysfunction and increased susceptibility to infection after physical injury. Ann. Surg. 2014, 259, 171–178. [Google Scholar] [CrossRef] [PubMed]
  157. Agrati, C.; Sacchi, A.; Bordoni, V.; Cimini, E.; Notari, S.; Grassi, G.; Casetti, R.; Tartaglia, E.; Lalle, E.; D’Abramo, A.; et al. Expansion of myeloid-derived suppressor cells in patients with severe coronavirus disease (COVID-19). Cell Death Differ. 2020, 27, 3196–3207. [Google Scholar] [CrossRef]
  158. Adebayo, A.; Varzideh, F.; Wilson, S.; Gambardella, J.; Eacobacci, M.; Jankauskas, S.; Donkor, K.; Kansakar, U.; Trimarco, V.; Mone, P.; et al. L-arginine and COVID-19: An update. Nutrients 2021, 13, 3951. [Google Scholar] [CrossRef] [PubMed]
  159. Grassi, G.; Notari, S.; Gili, S.; Bordoni, V.; Casetti, R.; Cimini, E.; Tartaglia, E.; Mariotti, D.; Agrati, C.; Sacchi, A. Myeloid-Derived Suppressor Cells in COVID-19: The Paradox of Good. Front. Immunol. 2022, 13, 842949. [Google Scholar] [CrossRef]
  160. Hottz, E.D.; Azevedo-Quintanilha, I.G.; Palhinha, L.; Teixeira, L.; Barreto, E.A.; Pão, C.R.R.; Righy, C.; Franco, S.; Souza, T.M.L.; Kurtz, P.; et al. Platelet activation and platelet-monocyte aggregate formation trigger tissue factor expression in patients with severe COVID-19. Blood 2020, 136, 1330–1341. [Google Scholar] [CrossRef]
  161. Sacchi, A.; Grassi, G.; Notari, S.; Gili, S.; Bordoni, V.; Tartaglia, E.; Casetti, R.; Cimini, E.; Mariotti, D.; Garotto, G.; et al. Expansion of Myeloid Derived Suppressor Cells Contributes to Platelet Activation by L-Arginine Deprivation during SARS-CoV-2 Infection. Cells 2021, 10, 2111. [Google Scholar] [CrossRef]
  162. Cui, S.; Chen, S.; Li, X.; Liu, S.; Wang, F. Prevalence of venous thromboembolism in patients with severe novel coronavirus pneumonia. J. Thromb. Haemost. 2020, 18, 1421–1424. [Google Scholar] [CrossRef] [Green Version]
  163. Klok, F.A.; Kruip, M.; van der Meer, N.J.M.; Arbous, M.S.; Gommers, D.; Kant, K.M.; Kaptein, F.H.J.; van Paassen, J.; Stals, M.A.M.; Huisman, M.V.; et al. Incidence of thrombotic complications in critically ill ICU patients with COVID-19. Thromb. Res. 2020, 191, 145–147. [Google Scholar] [CrossRef]
  164. Llitjos, J.F.; Leclerc, M.; Chochois, C.; Monsallier, J.M.; Ramakers, M.; Auvray, M.; Merouani, K. High incidence of venous thromboembolic events in anticoagulated severe COVID-19 patients. J. Thromb. Haemost. 2020, 18, 1743–1746. [Google Scholar] [CrossRef]
  165. Tan, B.K.; Mainbourg, S.; Friggeri, A.; Bertoletti, L.; Douplat, M.; Dargaud, Y.; Grange, C.; Lobbes, H.; Provencher, S.; Lega, J.C. Arterial and venous thromboembolism in COVID-19: A study-level meta-analysis. Thorax 2021, 76, 970–979. [Google Scholar] [CrossRef]
  166. Conway, E.M.; Mackman, N.; Warren, R.Q.; Wolberg, A.S.; Mosnier, L.O.; Campbell, R.A.; Gralinski, L.E.; Rondina, M.T.; van de Veerdonk, F.L.; Hoffmeister, K.M.; et al. Understanding COVID-19-associated coagulopathy. Nat. Rev. Immunol. 2022, 22, 639–649. [Google Scholar] [CrossRef]
  167. Lazzaroni, M.G.; Piantoni, S.; Masneri, S.; Garrafa, E.; Martini, G.; Tincani, A.; Andreoli, L.; Franceschini, F. Coagulation dysfunction in COVID-19: The interplay between inflammation, viral infection and the coagulation system. Blood Rev. 2021, 46, 100745. [Google Scholar] [CrossRef]
  168. Bunch, C.M.; Moore, E.E.; Moore, H.B.; Neal, M.D.; Thomas, A.V.; Zackariya, N.; Zhao, J.; Zackariya, S.; Brenner, T.J.; Berquist, M.; et al. Immuno-Thrombotic Complications of COVID-19: Implications for Timing of Surgery and Anticoagulation. Front. Surg. 2022, 9, 889999. [Google Scholar] [CrossRef]
  169. Nicolai, L.; Leunig, A.; Brambs, S.; Kaiser, R.; Weinberger, T.; Weigand, M.; Muenchhoff, M.; Hellmuth, J.C.; Ledderose, S.; Schulz, H.; et al. Immunothrombotic Dysregulation in COVID-19 Pneumonia Is Associated with Respiratory Failure and Coagulopathy. Circulation 2020, 142, 1176–1189. [Google Scholar] [CrossRef]
  170. Ackermann, M.; Verleden, S.E.; Kuehnel, M.; Haverich, A.; Welte, T.; Laenger, F.; Vanstapel, A.; Werlein, C.; Stark, H.; Tzankov, A.; et al. Pulmonary Vascular Endothelialitis, Thrombosis, and Angiogenesis in COVID-19. N. Engl. J. Med. 2020, 383, 120–128. [Google Scholar] [CrossRef]
  171. Gomez-Mesa, J.E.; Galindo-Coral, S.; Montes, M.C.; Munoz Martin, A.J. Thrombosis and Coagulopathy in COVID-19. Curr. Probl. Cardiol. 2021, 46, 100742. [Google Scholar] [CrossRef]
  172. Varga, Z.; Flammer, A.J.; Steiger, P.; Haberecker, M.; Andermatt, R.; Zinkernagel, A.S.; Mehra, M.R.; Schuepbach, R.A.; Ruschitzka, F.; Moch, H. Endothelial cell infection and endotheliitis in COVID-19. Lancet 2020, 395, 1417–1418. [Google Scholar] [CrossRef]
  173. 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]
  174. Daly, J.L.; Simonetti, B.; Klein, K.; Chen, K.E.; Williamson, M.K.; Anton-Plagaro, C.; Shoemark, D.K.; Simon-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]
  175. Liu, F.; Han, K.; Blair, R.; Kenst, K.; Qin, Z.; Upcin, B.; Wörsdörfer, P.; Midkiff, C.C.; Mudd, J.; Belyaeva, E.; et al. SARS-CoV-2 Infects Endothelial Cells In Vivo and In Vitro. Front. Cell. Infect. Microbiol. 2021, 11, 701278. [Google Scholar] [CrossRef] [PubMed]
  176. Schimmel, L.; Chew, K.Y.; Stocks, C.J.; Yordanov, T.E.; Essebier, P.; Kulasinghe, A.; Monkman, J.; dos Santos Miggiolaro, A.F.R.; Cooper, C.; de Noronha, L.; et al. Endothelial cells are not productively infected by SARS-CoV-2. Clin. Transl. Immunol. 2021, 10, e1350. [Google Scholar] [CrossRef] [PubMed]
  177. McCracken, I.R.; Saginc, G.; He, L.; Huseynov, A.; Daniels, A.; Fletcher, S.; Peghaire, C.; Kalna, V.; Andaloussi-Mäe, M.; Muhl, L.; et al. Lack of Evidence of Angiotensin-Converting Enzyme 2 Expression and Replicative Infection by SARS-CoV-2 in Human Endothelial Cells. Circulation 2021, 143, 865–868. [Google Scholar] [CrossRef] [PubMed]
  178. Nagashima, S.; Mendes, M.C.; Camargo Martins, A.P.; Borges, N.H.; Godoy, T.M.; Miggiolaro, A.; da Silva Dezidério, F.; Machado-Souza, C.; de Noronha, L. Endothelial Dysfunction and Thrombosis in Patients with COVID-19-Brief Report. Arterioscler. Thromb. Vasc. Biol. 2020, 40, 2404–2407. [Google Scholar] [CrossRef]
  179. Nicosia, R.F.; Ligresti, G.; Caporarello, N.; Akilesh, S.; Ribatti, D. COVID-19 Vasculopathy: Mounting Evidence for an Indirect Mechanism of Endothelial Injury. Am. J. Pathol. 2021, 191, 1374–1384. [Google Scholar] [CrossRef]
  180. Teuwen, L.A.; Geldhof, V.; Pasut, A.; Carmeliet, P. COVID-19: The vasculature unleashed. Nat. Rev. Immunol. 2020, 20, 389–391. [Google Scholar] [CrossRef]
  181. Cooper, S.L.; Boyle, E.; Jefferson, S.R.; Heslop, C.R.A. Role of the Renin-Angiotensin-Aldosterone and Kinin-Kallikrein Systems in the Cardiovascular Complications of COVID-19 and Long COVID. Int. J. Mol. Sci. 2021, 22, 8255. [Google Scholar] [CrossRef]
  182. Martens, C.P.; Van Mol, P.; Wauters, J.; Wauters, E.; Gangnus, T.; Noppen, B.; Callewaert, H.; Feyen, J.H.M.; Liesenborghs, L.; Heylen, E.; et al. Dysregulation of the kallikrein-kinin system in bronchoalveolar lavage fluid of patients with severe COVID-19. EBioMedicine 2022, 83, 104195. [Google Scholar] [CrossRef]
  183. Bonaffini, P.A.; Franco, P.N.; Bonanomi, A.; Giaccherini, C.; Valle, C.; Marra, P.; Norsa, L.; Marchetti, M.; Falanga, A.; Sironi, S. Ischemic and hemorrhagic abdominal complications in COVID-19 patients: Experience from the first Italian wave. Eur. J. Med. Res. 2022, 27, 165. [Google Scholar] [CrossRef]
  184. Paul, T.; Joy, A.R.; Alsoub, H.; Parambil, J.V. Case Report: Ischemic Colitis in Severe COVID-19 Pneumonia: An Unforeseen Gastrointestinal Complication. Am. J. Trop. Med. Hyg. 2021, 104, 63–65. [Google Scholar] [CrossRef]
  185. Wu, X.; Jing, H.; Wang, C.; Wang, Y.; Zuo, N.; Jiang, T.; Novakovic, V.A.; Shi, J. Intestinal Damage in COVID-19: SARS-CoV-2 Infection and Intestinal Thrombosis. Front. Microbiol. 2022, 13, 860931. [Google Scholar] [CrossRef] [PubMed]
  186. Yao, Y.; Cao, J.; Wang, Q.; Shi, Q.; Liu, K.; Luo, Z.; Chen, X.; Chen, S.; Yu, K.; Huang, Z.; et al. D-dimer as a biomarker for disease severity and mortality in COVID-19 patients: A case control study. J. Intensive Care 2020, 8, 49. [Google Scholar] [CrossRef] [PubMed]
  187. Poudel, A.; Poudel, Y.; Adhikari, A.; Aryal, B.B.; Dangol, D.; Bajracharya, T.; Maharjan, A.; Gautam, R. D-dimer as a biomarker for assessment of COVID-19 prognosis: D-dimer levels on admission and its role in predicting disease outcome in hospitalized patients with COVID-19. PLoS ONE 2021, 16, e0256744. [Google Scholar] [CrossRef] [PubMed]
  188. Wang, B.; Zhang, L.; Wang, Y.; Dai, T.; Qin, Z.; Zhou, F.; Zhang, L. Alterations in microbiota of patients with COVID-19: Potential mechanisms and therapeutic interventions. Signal Transduct. Target. Ther. 2022, 7, 143. [Google Scholar] [CrossRef]
  189. Yeoh, Y.K.; Zuo, T.; Lui, G.C.; Zhang, F.; Liu, Q.; Li, A.Y.; Chung, A.C.; Cheung, C.P.; Tso, E.Y.; Fung, K.S.; et al. Gut microbiota composition reflects disease severity and dysfunctional immune responses in patients with COVID-19. Gut 2021, 70, 698–706. [Google Scholar] [CrossRef]
  190. Sun, Z.; Song, Z.-G.; Liu, C.; Tan, S.; Lin, S.; Zhu, J.; Dai, F.-H.; Gao, J.; She, J.-L.; Mei, Z.; et al. Gut microbiome alterations and gut barrier dysfunction are associated with host immune homeostasis in COVID-19 patients. BMC Med. 2022, 20, 24. [Google Scholar] [CrossRef]
  191. Eroğlu, İ.; Eroğlu, B.; Güven, G.S. Altered tryptophan absorption and metabolism could underlie long-term symptoms in survivors of coronavirus disease 2019 (COVID-19). Nutrition 2021, 90, 111308. [Google Scholar] [CrossRef]
  192. Yang, J.; Zhao, X.; Patel, A.; Potru, R.; Azizi-Ghannad, S.; Dolinger, M.; Cao, J.; Bartholomew, C.; Mazurkiewicz, J.; Conti, D.; et al. Rapamycin Inhibition of mTOR Reduces Levels of the Na+/H+ Exchanger 3 in Intestines of Mice and Humans, Leading to Diarrhea. Gastroenterology 2015, 149, 151–162. [Google Scholar] [CrossRef] [Green Version]
  193. Cumhur Cure, M.; Cure, E. Effects of the Na(+)/H(+) Ion Exchanger on Susceptibility to COVID-19 and the Course of the Disease. J. Renin-Angiotensin-Aldosterone Syst. 2021, 2021, 4754440. [Google Scholar] [CrossRef]
  194. He, P.; Yun, C.C. Mechanisms of the regulation of the intestinal Na+/H+ exchanger NHE3. J. Biomed. Biotechnol. 2010, 2010, 238080. [Google Scholar] [CrossRef]
  195. Dhar, D.; Mohanty, A. Gut microbiota and COVID-19-possible link and implications. Virus Res. 2020, 285, 198018. [Google Scholar] [CrossRef] [PubMed]
  196. 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]
  197. 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.e948. [Google Scholar] [CrossRef] [PubMed]
  198. Yang, Y.; Bin, P.; Tao, S.; Zhu, G.; Wu, Z.; Cheng, W.; Ren, W.; Wei, H. Evaluation of the Mechanisms Underlying Amino Acid and Microbiota Interactions in Intestinal Infections Using Germ-Free Animals. Infect. Microbes Dis. 2021, 3, 79–86. [Google Scholar] [CrossRef]
  199. Viana, S.D.; Nunes, S.; Reis, F. ACE2 imbalance as a key player for the poor outcomes in COVID-19 patients with age-related comorbidities—Role of gut microbiota dysbiosis. Ageing Res. Rev. 2020, 62, 101123. [Google Scholar] [CrossRef]
  200. Hundt, M.A.; Deng, Y.; Ciarleglio, M.M.; Nathanson, M.H.; Lim, J.K. Abnormal Liver Tests in COVID-19: A Retrospective Observational Cohort Study of 1,827 Patients in a Major U.S. Hospital Network. Hepatology 2020, 72, 1169–1176. [Google Scholar] [CrossRef]
  201. Lei, P.; Zhang, L.; Han, P.; Zheng, C.; Tong, Q.; Shang, H.; Yang, F.; Hu, Y.; Li, X.; Song, Y. Liver injury in patients with COVID-19: Clinical profiles, CT findings, the correlation of the severity with liver injury. Hepatol. Int. 2020, 14, 733–742. [Google Scholar] [CrossRef]
  202. Parohan, M.; Yaghoubi, S.; Seraji, A. Liver injury is associated with severe coronavirus disease 2019 (COVID-19) infection: A systematic review and meta-analysis of retrospective studies. Hepatol. Res. 2020, 50, 924–935. [Google Scholar] [CrossRef]
  203. Phipps, M.M.; Barraza, L.H.; LaSota, E.D.; Sobieszczyk, M.E.; Pereira, M.R.; Zheng, E.X.; Fox, A.N.; Zucker, J.; Verna, E.C. Acute Liver Injury in COVID-19: Prevalence and Association with Clinical Outcomes in a Large U.S. Cohort. Hepatology 2020, 72, 807–817. [Google Scholar] [CrossRef]
  204. Fan, Z.; Chen, L.; Li, J.; Tian, C.; Zhang, Y.; Huang, S.; Liu, Z.; Cheng, J. Clinical Features of COVID-19-Related Liver Damage. medRxiv 2020. [Google Scholar] [CrossRef]
  205. Kulkarni, A.V.; Kumar, P.; Tevethia, H.V.; Premkumar, M.; Arab, J.P.; Candia, R.; Talukdar, R.; Sharma, M.; Qi, X.; Rao, P.N.; et al. Systematic review with meta-analysis: Liver manifestations and outcomes in COVID-19. Aliment. Pharmacol. Ther. 2020, 52, 584–599. [Google Scholar] [CrossRef] [PubMed]
  206. Chau, T.N.; Lee, K.C.; Yao, H.; Tsang, T.Y.; Chow, T.C.; Yeung, Y.C.; Choi, K.W.; Tso, Y.K.; Lau, T.; Lai, S.T.; et al. SARS-associated viral hepatitis caused by a novel coronavirus: Report of three cases. Hepatology 2004, 39, 302–310. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  207. Zhang, B.; Zhou, X.; Qiu, Y.; Feng, F.; Feng, J.; Jia, Y.; Zhu, H.; Hu, K.; Liu, J.; Liu, Z.; et al. Clinical characteristics of 82 death cases with COVID-19. medRxiv 2020. [Google Scholar] [CrossRef] [Green Version]
  208. Lagana, S.M.; Kudose, S.; Iuga, A.C.; Lee, M.J.; Fazlollahi, L.; Remotti, H.E.; Del Portillo, A.; De Michele, S.; de Gonzalez, A.K.; Saqi, A.; et al. Hepatic pathology in patients dying of COVID-19: A series of 40 cases including clinical, histologic, and virologic data. Mod. Pathol. 2020, 33, 2147–2155. [Google Scholar] [CrossRef] [PubMed]
  209. Wanner, N.; Andrieux, G.; Badia-i-Mompel, P.; Edler, C.; Pfefferle, S.; Lindenmeyer, M.T.; Schmidt-Lauber, C.; Czogalla, J.; Wong, M.N.; Okabayashi, Y.; et al. Molecular consequences of SARS-CoV-2 liver tropism. Nat. Metab. 2022, 4, 310–319. [Google Scholar] [CrossRef]
  210. Cai, Q.; Huang, D.; Yu, H.; Zhu, Z.; Xia, Z.; Su, Y.; Li, Z.; Zhou, G.; Gou, J.; Qu, J.; et al. COVID-19: Abnormal liver function tests. J. Hepatol. 2020, 73, 566–574. [Google Scholar] [CrossRef] [PubMed]
  211. Chen, L.Y.; Chu, H.K.; Bai, T. Liver damage at admission is an independent prognostic factor for COVID-19. J. Dig. Dis. 2020, 21, 512–518. [Google Scholar] [CrossRef]
  212. Guan, W.J.; Ni, Z.Y.; Hu, Y.; Liang, W.H.; Ou, C.Q.; He, J.X.; Liu, L.; Shan, H.; Lei, C.L.; Hui, D.S.C.; et al. Clinical Characteristics of Coronavirus Disease 2019 in China. N. Engl. J. Med. 2020, 382, 1708–1720. [Google Scholar] [CrossRef]
  213. Wang, Y.; Liu, S.; Liu, H.; Li, W.; Lin, F.; Jiang, L.; Li, X.; Xu, P.; Zhang, L.; Zhao, L.; et al. SARS-CoV-2 infection of the liver directly contributes to hepatic impairment in patients with COVID-19. J. Hepatol. 2020, 73, 807–816. [Google Scholar] [CrossRef]
  214. Paizis, G.; Tikellis, C.; Cooper, M.E.; Schembri, J.M.; Lew, R.A.; Smith, A.I.; Shaw, T.; Warner, F.J.; Zuilli, A.; Burrell, L.M.; et al. Chronic liver injury in rats and humans upregulates the novel enzyme angiotensin converting enzyme 2. Gut 2005, 54, 1790–1796. [Google Scholar] [CrossRef]
  215. McConnell, M.J.; Kondo, R.; Kawaguchi, N.; Iwakiri, Y. COVID-19 and Liver Injury: Role of Inflammatory Endotheliopathy, Platelet Dysfunction, and Thrombosis. Hepatol. Commun. 2022, 6, 255–269. [Google Scholar] [CrossRef]
  216. Dawood, D.R.M.; Salum, G.M.; El-Meguid, M.A. The Impact of COVID-19 on Liver Injury. Am. J. Med. Sci. 2022, 363, 94–103. [Google Scholar] [CrossRef] [PubMed]
  217. Pirola, C.J.; Sookoian, S. SARS-CoV-2 virus and liver expression of host receptors: Putative mechanisms of liver involvement in COVID-19. Liver Int. 2020, 40, 2038–2040. [Google Scholar] [CrossRef] [PubMed]
  218. Chai, X.H.L.; Zhang, Y.; Han, W.; Lu, Z.; Ke, A.; Zhou, J.; Shi, G.; Fang, N.; Fan, J.; Cai, J.; et al. Specific ACE2 Expression in Cholangiocytes May Cause Liver Damage After 2019-nCoV Infection. bioRxiv 2020. [Google Scholar] [CrossRef] [Green Version]
  219. Lax, S.F.; Skok, K.; Zechner, P.; Kessler, H.H.; Kaufmann, N.; Koelblinger, C.; Vander, K.; Bargfrieder, U.; Trauner, M. Pulmonary Arterial Thrombosis in COVID-19 with Fatal Outcome. Ann. Intern. Med. 2020, 173, 350–361. [Google Scholar] [CrossRef] [PubMed]
  220. Shaveisi-Zadeh, F.; Nikkho, B.; Khadem Erfan, M.B.; Amiri, A.; Azizi, A.; Mansouri, N.; Tarlan, M.; Rostami-Far, Z. Changes in liver enzymes and association with prognosis in patients with COVID-19: A retrospective case-control study. J. Int. Med. Res. 2022, 50, 3000605221110067. [Google Scholar] [CrossRef]
  221. Diaz-Louzao, C.; Barrera-Lopez, L.; Lopez-Rodriguez, M.; Casar, C.; Vazquez-Agra, N.; Pernas-Pardavila, H.; Marques-Afonso, A.; Vidal-Vazquez, M.; Montoya, J.G.; Andrade, A.H.; et al. Longitudinal relationship of liver injury with inflammation biomarkers in COVID-19 hospitalized patients using a joint modeling approach. Sci. Rep. 2022, 12, 5547. [Google Scholar] [CrossRef]
  222. Sonzogni, A.; Previtali, G. Liver histopathology in severe COVID 19 respiratory failure is suggestive of vascular alterations. Liver Int. 2020, 40, 2110–2116. [Google Scholar] [CrossRef] [PubMed]
  223. Alkattan, W.; Yaqinuddin, A.; Shafqat, A.; Kashir, J. NET-Mediated Pathogenesis of COVID-19: The Role of NETs in Hepatic Manifestations. J. Health Allied Sci. NU 2022, 12, 235–242. [Google Scholar] [CrossRef]
  224. Zhu, Y.; Chen, X.; Liu, X. NETosis and Neutrophil Extracellular Traps in COVID-19: Immunothrombosis and Beyond. Front. Immunol. 2022, 13, 838011. [Google Scholar] [CrossRef]
  225. Liu, F.; Long, X.; Zhang, B.; Zhang, W.; Chen, X.; Zhang, Z. ACE2 Expression in Pancreas May Cause Pancreatic Damage After SARS-CoV-2 Infection. Clin. Gastroenterol. Hepatol. Off. Clin. Pract. J. Am. Gastroenterol. Assoc. 2020, 18, 2128–2130.e2122. [Google Scholar] [CrossRef] [PubMed]
  226. Szlachcic, W.J.; Dabrowska, A.; Milewska, A.; Ziojla, N.; Blaszczyk, K.; Barreto-Duran, E.; Sanak, M.; Surmiak, M.; Owczarek, K.; Grzanka, D.; et al. SARS-CoV-2 infects an in vitro model of the human developing pancreas through endocytosis. iScience 2022, 25, 104594. [Google Scholar] [CrossRef] [PubMed]
  227. Wu, C.T.; Lidsky, P.V.; Xiao, Y.; Lee, I.T.; Cheng, R.; Nakayama, T.; Jiang, S.; Demeter, J.; Bevacqua, R.J.; Chang, C.A.; et al. SARS-CoV-2 infects human pancreatic β cells and elicits β cell impairment. Cell Metab. 2021, 33, 1565–1576.e1565. [Google Scholar] [CrossRef]
  228. Correia de Sá, T.; Soares, C.; Rocha, M. Acute pancreatitis and COVID-19: A literature review. World J. Gastrointest. Surg. 2021, 13, 574–584. [Google Scholar] [CrossRef] [PubMed]
  229. Hegyi, P.; Szakacs, Z.; Sahin-Toth, M. Lipotoxicity and Cytokine Storm in Severe Acute Pancreatitis and COVID-19. Gastroenterology 2020, 159, 824–827. [Google Scholar] [CrossRef] [PubMed]
  230. Kusmartseva, I.; Wu, W.; Syed, F.; Van Der Heide, V.; Jorgensen, M.; Joseph, P.; Tang, X.; Candelario-Jalil, E.; Yang, C.; Nick, H.; et al. Expression of SARS-CoV-2 Entry Factors in the Pancreas of Normal Organ Donors and Individuals with COVID-19. Cell Metab. 2020, 32, 1041–1051.e1046. [Google Scholar] [CrossRef]
  231. Müller, J.A.; Groß, R.; Conzelmann, C.; Krüger, J.; Merle, U.; Steinhart, J.; Weil, T.; Koepke, L.; Bozzo, C.P.; Read, C.; et al. SARS-CoV-2 infects and replicates in cells of the human endocrine and exocrine pancreas. Nat. Metab. 2021, 3, 149–165. [Google Scholar] [CrossRef]
  232. Shaharuddin, S.H.; Wang, V.; Santos, R.S.; Gross, A.; Wang, Y.; Jawanda, H.; Zhang, Y.; Hasan, W.; Garcia, G.; Arumugaswami, V.; et al. Deleterious Effects of SARS-CoV-2 Infection on Human Pancreatic Cells. Front. Cell. Infect. Microbiol. 2021, 11, 678482. [Google Scholar] [CrossRef]
  233. Steenblock, C.; Richter, S.; Berger, I.; Barovic, M.; Schmid, J.; Schubert, U.; Jarzebska, N.; von Mässenhausen, A.; Linkermann, A.; Schürmann, A.; et al. Viral infiltration of pancreatic islets in patients with COVID-19. Nat. Commun. 2021, 12, 3534. [Google Scholar] [CrossRef]
  234. Qadir, M.M.F.; Bhondeley, M.; Beatty, W.; Gaupp, D.D.; Doyle-Meyers, L.A.; Fischer, T.; Bandyopadhyay, I.; Blair, R.V.; Bohm, R.; Rappaport, J.; et al. SARS-CoV-2 infection of the pancreas promotes thrombofibrosis and is associated with new-onset diabetes. JCI Insight 2021, 6, e151551. [Google Scholar] [CrossRef] [PubMed]
  235. Santos, R.A.S.; Sampaio, W.O.; Alzamora, A.C.; Motta-Santos, D.; Alenina, N.; Bader, M.; Campagnole-Santos, M.J. The ACE2/Angiotensin-(1-7)/MAS Axis of the Renin-Angiotensin System: Focus on Angiotensin-(1-7). Physiol. Rev. 2018, 98, 505–553. [Google Scholar] [CrossRef] [Green Version]
  236. Memon, B.; Abdelalim, E.M. ACE2 function in the pancreatic islet: Implications for relationship between SARS-CoV-2 and diabetes. Acta Physiol. 2021, 233, e13733. [Google Scholar] [CrossRef] [PubMed]
  237. Aloysius, M.M.; Thatti, A.; Gupta, A.; Sharma, N.; Bansal, P.; Goyal, H. COVID-19 presenting as acute pancreatitis. Pancreatology 2020, 20, 1026–1027. [Google Scholar] [CrossRef] [PubMed]
  238. Anand, E.R.; Major, C.; Pickering, O.; Nelson, M. Acute pancreatitis in a COVID-19 patient. Br. J. Surg. 2020, 107, e182. [Google Scholar] [CrossRef] [PubMed]
  239. Hadi, A.; Werge, M.; Kristiansen, K.T.; Pedersen, U.G.; Karstensen, J.G.; Novovic, S.; Gluud, L.L. Coronavirus Disease-19 (COVID-19) associated with severe acute pancreatitis: Case report on three family members. Pancreatology 2020, 20, 665–667. [Google Scholar] [CrossRef]
  240. Kataria, S.; Sharif, A.; Ur Rehman, A.; Ahmed, Z.; Hanan, A. COVID-19 Induced Acute Pancreatitis: A Case Report and Literature Review. Cureus 2020, 12, e9169. [Google Scholar] [CrossRef] [PubMed]
  241. Hojyo, S.; Uchida, M.; Tanaka, K.; Hasebe, R.; Tanaka, Y.; Murakami, M.; Hirano, T. How COVID-19 induces cytokine storm with high mortality. Inflamm. Regen. 2020, 40, 37. [Google Scholar] [CrossRef]
  242. Rao, S.A.; Kunte, A.R. Interleukin-6: An Early Predictive Marker for Severity of Acute Pancreatitis. Indian J. Crit. Care Med. Peer-Rev. Off. Publ. Indian Soc. Crit. Care Med. 2017, 21, 424–428. [Google Scholar] [CrossRef]
  243. Wang, F.; Wang, H.; Fan, J.; Zhang, Y.; Wang, H.; Zhao, Q. Pancreatic Injury Patterns in Patients With Coronavirus Disease 19 Pneumonia. Gastroenterology 2020, 159, 367–370. [Google Scholar] [CrossRef]
  244. De Oliveira, C.; Khatua, B.; Noel, P.; Kostenko, S.; Bag, A.; Balakrishnan, B.; Patel, K.S.; Guerra, A.A.; Martinez, M.N.; Trivedi, S.; et al. Pancreatic triglyceride lipase mediates lipotoxic systemic inflammation. J. Clin. Investig. 2020, 130, 1931–1947. [Google Scholar] [CrossRef]
  245. El-Kurdi, B.; Khatua, B.; Rood, C.; Snozek, C.; Cartin-Ceba, R.; Singh, V.P. Mortality From Coronavirus Disease 2019 Increases with Unsaturated Fat and May Be Reduced by Early Calcium and Albumin Supplementation. Gastroenterology 2020, 159, 1015–1018.e4. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Mechanisms of infection and pathogenesis of COVID-19 in the intestine. The molecular pathogenesis of SARS-CoV-2 infection of epithelial gastrointestinal cells is depicted, including the viral replication cycle.
Figure 1. Mechanisms of infection and pathogenesis of COVID-19 in the intestine. The molecular pathogenesis of SARS-CoV-2 infection of epithelial gastrointestinal cells is depicted, including the viral replication cycle.
Cells 12 00262 g001
Figure 2. Liver damage after SARS-CoV-2 infection. Severe chronic portal inflammation is observed in livers of patients with acute COVID-19 (Panels (A,B), Hematoxylin and Eosin, original magnification 200× and 400×, respectively). At higher magnification the fragmented nuclei of apoptotic hepatocytes are present (Panel (C), arrow), as well as Binucleation of hepatocytes (Panel (D), arrow). In severe cases, extensive areas of necrosis were observed (Panel (E)) Panels (CE), Hematoxylin & Eosin, original magnification 400×.
Figure 2. Liver damage after SARS-CoV-2 infection. Severe chronic portal inflammation is observed in livers of patients with acute COVID-19 (Panels (A,B), Hematoxylin and Eosin, original magnification 200× and 400×, respectively). At higher magnification the fragmented nuclei of apoptotic hepatocytes are present (Panel (C), arrow), as well as Binucleation of hepatocytes (Panel (D), arrow). In severe cases, extensive areas of necrosis were observed (Panel (E)) Panels (CE), Hematoxylin & Eosin, original magnification 400×.
Cells 12 00262 g002
Figure 3. Detection of SARS-CoV-2 proteins in liver samples. Immunohistochemistry for SARS-CoV-2 Nucleocapsid shows robust immunoreactivity in hepatocytes, in areas of liver damage (Panel (A) original magnification 100× and Panel (C) original magnification 400×). Spike protein is also present in the cytoplasm of hepatocytes in areas of necrosis (Panel (B) original magnification 100×, and steatosis (Panel (D) original magnification 400×).
Figure 3. Detection of SARS-CoV-2 proteins in liver samples. Immunohistochemistry for SARS-CoV-2 Nucleocapsid shows robust immunoreactivity in hepatocytes, in areas of liver damage (Panel (A) original magnification 100× and Panel (C) original magnification 400×). Spike protein is also present in the cytoplasm of hepatocytes in areas of necrosis (Panel (B) original magnification 100×, and steatosis (Panel (D) original magnification 400×).
Cells 12 00262 g003
Figure 4. Expression of SARS-CoV-2 proteins in liver inflammatory and endothelial cells from cases of acute fatal COVID-19. Nucleocapsid is detected in portal inflammatory cells (Panel (A), original magnification 200×). Spike is also robustly expressed in portal lymphocytes (Panel (B), original magnification 600×). Nucleocapsid is also present in macrophages (Panel (C), original magnification 400×) and endothelial cells (Panel (D), original magnification 400×) in liver samples.
Figure 4. Expression of SARS-CoV-2 proteins in liver inflammatory and endothelial cells from cases of acute fatal COVID-19. Nucleocapsid is detected in portal inflammatory cells (Panel (A), original magnification 200×). Spike is also robustly expressed in portal lymphocytes (Panel (B), original magnification 600×). Nucleocapsid is also present in macrophages (Panel (C), original magnification 400×) and endothelial cells (Panel (D), original magnification 400×) in liver samples.
Cells 12 00262 g004
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

Khreefa, Z.; Barbier, M.T.; Koksal, A.R.; Love, G.; Del Valle, L. Pathogenesis and Mechanisms of SARS-CoV-2 Infection in the Intestine, Liver, and Pancreas. Cells 2023, 12, 262. https://doi.org/10.3390/cells12020262

AMA Style

Khreefa Z, Barbier MT, Koksal AR, Love G, Del Valle L. Pathogenesis and Mechanisms of SARS-CoV-2 Infection in the Intestine, Liver, and Pancreas. Cells. 2023; 12(2):262. https://doi.org/10.3390/cells12020262

Chicago/Turabian Style

Khreefa, Zaid, Mallory T. Barbier, Ali Riza Koksal, Gordon Love, and Luis Del Valle. 2023. "Pathogenesis and Mechanisms of SARS-CoV-2 Infection in the Intestine, Liver, and Pancreas" Cells 12, no. 2: 262. https://doi.org/10.3390/cells12020262

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop