Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 85
Filter
Add more filters










Publication year range
1.
J Virol ; 98(4): e0006424, 2024 Apr 16.
Article in English | MEDLINE | ID: mdl-38488360

ABSTRACT

As one of the most important causative agents of severe gastroenteritis in children, piglets, and other young animals, species A rotaviruses have adversely impacted both human health and the global swine industry. Vaccines against rotaviruses (RVs) are insufficiently effective, and no specific treatment is available. To understand the relationships between porcine RV (PoRV) infection and enterocytes in terms of the cellular lipid metabolism, we performed an untargeted liquid chromatography mass spectrometry (LC-MS) lipidomics analysis of PoRV-infected IPEC-J2 cells. Herein, a total of 451 lipids (263 upregulated lipids and 188 downregulated lipids), spanning sphingolipid, glycerolipid, and glycerophospholipids, were significantly altered compared with the mock-infected group. Interestingly, almost all the ceramides among these lipids were upregulated during PoRV infection. LC-MS analysis was used to validated the lipidomics data and demonstrated that PoRV replication increased the levels of long-chain ceramides (C16-ceramide, C18-ceramide, and C24-ceramide) in cells. Furthermore, we found that these long-chain ceramides markedly inhibited PoRV infection and that their antiviral actions were exerted in the replication stage of PoRV infection. Moreover, downregulation of endogenous ceramides with the ceramide metabolic inhibitors enhanced PoRV propagation. Increasing the levels of ceramides by the addition of C6-ceramide strikingly suppressed the replication of diverse RV strains. We further found that the treatment with an apoptotic inhibitor could reverse the antiviral activity of ceramide against PoRV replication, demonstrating that ceramide restricted RV infection by inducing apoptosis. Altogether, this study revealed that ceramides played an antiviral role against RV infection, providing potential approaches for the development of antiviral therapies.IMPORTANCERotaviruses (RVs) are among the most important zoonosis viruses, which mainly infected enterocytes of the intestinal epithelium causing diarrhea in children and the young of many mammalian and avian species. Lipids play an essential role in viral infection. A comprehensive understanding of the interaction between RV and lipid metabolism in the enterocytes will be helpful to control RV infection. Here, we mapped changes in enterocyte lipids following porcine RV (PoRV) infection using an untargeted lipidomics approach. We found that PoRV infection altered the metabolism of various lipid species, especially ceramides (derivatives of the sphingosine). We further demonstrated that PoRV infection increased the accumulation of ceramides and that ceramides exerted antiviral effects on RV replication by inducing apoptosis. Our findings fill a gap in understanding the alterations of lipid metabolism in RV-infected enterocytes and highlight the antiviral effects of ceramides on RV infection, suggesting potential approaches to control RV infection.


Subject(s)
Ceramides , Rotavirus Infections , Rotavirus , Animals , Ceramides/metabolism , Lipid Metabolism , Lipidomics , Rotavirus/physiology , Swine , Enterocytes/metabolism , Enterocytes/virology , Rotavirus Infections/metabolism , Cell Line
2.
Nat Commun ; 13(1): 17, 2022 01 10.
Article in English | MEDLINE | ID: mdl-35013162

ABSTRACT

Defense against intracellular infection has been extensively studied in vertebrate hosts, but less is known about invertebrate hosts; specifically, the transcription factors that induce defense against intracellular intestinal infection in the model nematode Caenorhabditis elegans remain understudied. Two different types of intracellular pathogens that naturally infect the C. elegans intestine are the Orsay virus, which is an RNA virus, and microsporidia, which comprise a phylum of fungal pathogens. Despite their molecular differences, these pathogens induce a common host transcriptional response called the intracellular pathogen response (IPR). Here we show that zip-1 is an IPR regulator that functions downstream of all known IPR-activating and regulatory pathways. zip-1 encodes a putative bZIP transcription factor, and we show that zip-1 controls induction of a subset of genes upon IPR activation. ZIP-1 protein is expressed in the nuclei of intestinal cells, and is at least partially required in the intestine to upregulate IPR gene expression. Importantly, zip-1 promotes resistance to infection by the Orsay virus and by microsporidia in intestinal cells. Altogether, our results indicate that zip-1 represents a central hub for triggers of the IPR, and that this transcription factor has a protective function against intracellular pathogen infection in C. elegans.


Subject(s)
Basic-Leucine Zipper Transcription Factors , Caenorhabditis elegans , Enterocytes , Host-Pathogen Interactions/physiology , Animals , Basic-Leucine Zipper Transcription Factors/immunology , Basic-Leucine Zipper Transcription Factors/metabolism , Caenorhabditis elegans/immunology , Caenorhabditis elegans/microbiology , Caenorhabditis elegans/virology , Caenorhabditis elegans Proteins/immunology , Caenorhabditis elegans Proteins/metabolism , Enterocytes/immunology , Enterocytes/microbiology , Enterocytes/virology , Immunity, Innate/physiology , Intestines/microbiology , Intestines/virology , Invertebrates/immunology , Microsporidia/pathogenicity , RNA Viruses/pathogenicity
3.
Cell Rep ; 37(13): 110150, 2021 12 28.
Article in English | MEDLINE | ID: mdl-34965418

ABSTRACT

Enteric pathogens overcome barrier immunity within the intestinal environment that includes the endogenous flora. The microbiota produces diverse ligands, and the full spectrum of microbial products that are sensed by the epithelium and prime protective immunity is unknown. Using Drosophila, we find that the gut presents a high barrier to infection, which is partially due to signals from the microbiota, as loss of the microbiota enhances oral viral infection. We report cyclic dinucleotide (CDN) feeding is sufficient to protect microbiota-deficient flies from enhanced oral infection, suggesting that bacterial-derived CDNs induce immunity. Mechanistically, we find CDN protection is dSTING- and dTBK1-dependent, leading to NF-kB-dependent gene expression. Furthermore, we identify the apical nucleoside transporter, CNT2, as required for oral CDN protection. Altogether, our studies define a role for bacterial products in priming immune defenses in the gut.


Subject(s)
Alphavirus Infections/immunology , Antiviral Agents/pharmacology , Drosophila melanogaster/immunology , Enterocytes/immunology , Membrane Proteins/metabolism , Membrane Transport Proteins/metabolism , Nucleotides, Cyclic/administration & dosage , Alphavirus Infections/drug therapy , Alphavirus Infections/virology , Animals , Drosophila melanogaster/drug effects , Drosophila melanogaster/genetics , Drosophila melanogaster/metabolism , Enterocytes/drug effects , Enterocytes/virology , Female , Immunity, Innate , Membrane Proteins/genetics , Membrane Transport Proteins/genetics , NF-kappa B/genetics , NF-kappa B/metabolism , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Sindbis Virus/immunology
4.
Int J Infect Dis ; 113: 82-86, 2021 Dec.
Article in English | MEDLINE | ID: mdl-34597762

ABSTRACT

OBJECTIVES: SARS-CoV-2 exhibits tropism for the gastrointestinal tract; however, lesions in enterocytes and their correlation with disease severity and patient prognosis are still unknown. METHODS: SARS-CoV-2 patients were enrolled in 5 medical centres in São Paulo, Brazil and their clinical characteristics and laboratory findings recorded. At admission, day 7 and day 14 of hospitalisation, plasma and urine samples were collected, and cytokine levels and intestinal fatty acid-binding protein (I-FABP) concentrations measured. RESULTS: COVID-19 patients displayed ≈48-, 74- and 125-fold increased urinary I-FABP levels at admission (n=283; P<0.001), day 7 (n=142; P<0.01) and day 14 (n=75; P<0.01) of hospitalisation. Critically ill patients and nonsurvivors showed higher I-FABP concentrations compared with patients with less severe illness. At admission, infected patients demonstrated enhanced production of plasma interferon (IFN)-γ and interleukin (IL)-6. The receiver operating characteristic curve suggested I-FABP as a biomarker for COVID-19 disease severity at admission (P<0.0001; Youden index=6.89; area under the curve=0.699). Patients with I-FABP ≥6.89 showed higher IL-6 and C-reactive protein levels (P<0.001) at admission and had a prolonged length of hospital stay. CONCLUSIONS: Our findings revealed damage to enterocytes in SARS-CoV-2 infection, which is associated with illness severity, poor prognosis and exacerbated inflammatory response.


Subject(s)
COVID-19 , Fatty Acid-Binding Proteins/analysis , Biomarkers , Brazil , C-Reactive Protein , COVID-19/diagnosis , Enterocytes/virology , Humans , Interferon-gamma , Interleukin-6 , Prospective Studies
5.
Cells ; 10(9)2021 08 30.
Article in English | MEDLINE | ID: mdl-34571900

ABSTRACT

Iron is crucial to the regulation of the host innate immune system and the outcome of many infections. Hepatitis C virus (HCV), one of the major viral human pathogens that depends on iron to complete its life cycle, is highly skilled in evading the immune system. This study presents the construction and validation of a physiologically relevant triple-cell co-culture model that was used to investigate the input of iron in HCV infection and the interplay between HCV, iron, and determinants of host innate immunity. We recorded the expression patterns of key proteins of iron homeostasis involved in iron import, export and storage and examined their relation to the iron regulatory hormone hepcidin in hepatocytes, enterocytes and macrophages in the presence and absence of HCV. We then assessed the transcriptional profiles of pro-inflammatory cytokines Interleukin-6 (IL-6) and interleukin-15 (IL-15) and anti-inflammatory interleukin-10 (IL-10) under normal or iron-depleted conditions and determined how these were affected by infection. Our data suggest the presence of a link between iron homeostasis and innate immunity unfolding among liver, intestine, and macrophages, which could participate in the deregulation of innate immune responses observed in early HCV infection. Coupled with iron-assisted enhanced viral propagation, such a mechanism may be important for the establishment of viral persistence and the ensuing chronic liver disease.


Subject(s)
Enterocytes/pathology , Hepatitis C/pathology , Hepatocytes/pathology , Homeostasis , Immunity, Innate , Iron/metabolism , Macrophages/pathology , Coculture Techniques , Cytokines/metabolism , Enterocytes/immunology , Enterocytes/metabolism , Enterocytes/virology , Hepacivirus/immunology , Hepacivirus/metabolism , Hepatitis C/immunology , Hepatitis C/metabolism , Hepatitis C/virology , Hepatocytes/immunology , Hepatocytes/metabolism , Hepatocytes/virology , Humans , Macrophages/immunology , Macrophages/metabolism , Macrophages/virology
6.
Int J Mol Sci ; 22(15)2021 Jul 29.
Article in English | MEDLINE | ID: mdl-34360898

ABSTRACT

Previous studies have shown that microRNAs (miRNAs) are closely related to many viral infections. However, the molecular mechanism of how miRNAs regulate porcine epidemic diarrhea virus (PEDV) infection remains unclear. In this study, we first constructed a PEDV-infected IPEC-J2 cytopathic model to validate the relationship between miR-129a-3p expression levels and PEDV resistance. Secondly, we explored the effect of miR-129a-3p on PEDV infection by targeting the 3'UTR region of the ligand ectodysplasin (EDA) gene. Finally, transcriptome sequencing was used to analyze the downstream regulatory mechanism of EDA. The results showed that after 48 h of PEDV infection, IPEC-J2 cells showed obvious pathological changes, and miR-129a-3p expression was significantly downregulated (p < 0.01). Overexpression of miR-129a-3p mimics inhibited PEDV replication in IPEC-J2 cells; silencing endogenous miR-129a-3p can promote viral replication. A dual luciferase assay showed that miR-129a-3p could bind to the 3'UTR region of the EDA gene, which significantly reduced the expression level of EDA (p < 0.01). Functional verification showed that upregulation of EDA gene expression significantly promoted PEDV replication in IPEC-J2 cells. Overexpression of miR-129a-3p can activate the caspase activation and recruitment domain 11 (CARD11) mediated NF-κB pathway, thus inhibiting PEDV replication. The above results suggest that miR-129a-3p inhibits PEDV replication in IPEC-J2 cells by activating the NF-κB pathway by binding to the EDA 3'UTR region. Our results have laid the foundation for in-depth study of the mechanism of miR-129a-3p resistance and its application in porcine epidemic diarrhea disease-resistance breeding.


Subject(s)
Coronavirus Infections/metabolism , Ectodysplasins/metabolism , Enterocytes/metabolism , MicroRNAs/metabolism , NF-kappa B/metabolism , Porcine epidemic diarrhea virus/physiology , Signal Transduction/genetics , Virus Replication/genetics , 3' Untranslated Regions , Animals , Chlorocebus aethiops , Coronavirus Infections/virology , Down-Regulation/genetics , Ectodysplasins/genetics , Enterocytes/virology , HEK293 Cells , Humans , MicroRNAs/genetics , Swine , Transfection , Up-Regulation/genetics , Vero Cells , Exome Sequencing/methods
7.
Mol Biol Rep ; 48(5): 4667-4675, 2021 May.
Article in English | MEDLINE | ID: mdl-34023987

ABSTRACT

The transmembrane protease serine 2 (TMPRSS2) is a membrane anchored protease that primarily expressed by epithelial cells of respiratory and gastrointestinal systems and has been linked to multiple pathological processes in humans including tumor growth, metastasis and viral infections. Recent studies have shown that TMPRSS2 expressed on cell surface of host cells could play a crucial role in activation of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) spike protein which facilitates the rapid early entry of the virus into host cells. In addition, direct suppression of TMPRSS2 using small drug inhibitors has been demonstrated to be effective in decreasing SARS-CoV-2 infection in vitro, which presents TMPRSS2 protease as a potential therapeutic strategy for SARS-CoV-2 infection. Recently, SARS-CoV-2 has been shown to be capable of infecting gastrointestinal enterocytes and to provoke gastrointestinal disorders in patients with COVID-19 disease, which is considered as a new transmission route and target organ of SARS-CoV-2. In this review, we highlight the biochemical properties of TMPRSS2 protease and discuss the potential targeting of TMPRSS2 by inhibitors to prevent the SARS-CoV-2 spreading through gastro-intestinal tract system as well as the hurdles that need to be overcome.


Subject(s)
COVID-19/metabolism , Enterocytes/drug effects , SARS-CoV-2/physiology , Serine Endopeptidases/metabolism , Serine Proteinase Inhibitors/pharmacology , Antiviral Agents/pharmacology , Drug Evaluation, Preclinical , Enterocytes/metabolism , Enterocytes/virology , Humans , SARS-CoV-2/drug effects , Small Molecule Libraries/pharmacology , Spike Glycoprotein, Coronavirus/metabolism , Virus Internalization/drug effects , COVID-19 Drug Treatment
8.
Infect Genet Evol ; 92: 104892, 2021 08.
Article in English | MEDLINE | ID: mdl-33957338

ABSTRACT

Novel coronavirus SARS-CoV-2 was recently outbreak worldwide causes severe acute respiratory syndrome along with gastrointestinal symptoms for some infected patients. Information on detail pathogenesis, host immune responses and responsible biological pathways are limited. Therefore, infection specific host gut responses and dietary supplements to neutralize immune inflammation demand extensive research. This study aimed to find differences in global co-expression protein-protein interaction sub-network and enriched biological processes in SARS-CoV and SARS-CoV-2 infected gut enterocytes cell line. Attempts have also been made to predict some dietary supplements to boost human health. The SARS-CoV and SARS-CoV-2 infected differential express proteins were integrated with the human protein interaction network and co-expression subnetworks were constructed. Common hubs of these sub-networks reshape central cellular pathways of metabolic processes, lipid localization, hypoxia response to decrease oxygen level and transport of bio-molecules. The major biological process enriched in the unique hub of SARS-CoV-2 significantly differ from SARS-CoV, related to interferon signaling, regulation of viral process and influenza-A enzymatic pathway. Predicted dietary supplements can improve SARS-CoV-2 infected person''s health by boosting the host immunity/reducing inflammation. To the best of our knowledge this is the first report on co-expression network mediated biological process in human gut enterocytes to predict dietary supplements/compounds.


Subject(s)
COVID-19/virology , Enterocytes/metabolism , Enterocytes/virology , SARS-CoV-2/metabolism , Severe acute respiratory syndrome-related coronavirus/metabolism , Dietary Supplements , Gene Expression Regulation , Humans , Protein Interaction Maps , RNA-Seq , Severe acute respiratory syndrome-related coronavirus/genetics , SARS-CoV-2/genetics
10.
Biol Open ; 10(3)2021 03 23.
Article in English | MEDLINE | ID: mdl-33757938

ABSTRACT

People with underlying conditions, including hypertension, obesity, and diabetes, are especially susceptible to negative outcomes after infection with coronavirus SARS-CoV-2, which causes COVID-19. Hypertension and respiratory inflammation are exacerbated by the Renin-Angiotensin-Aldosterone System (RAAS), which normally protects from rapidly dropping blood pressure via Angiotensin II (Ang II) produced by the enzyme Ace. The Ace paralog Ace2 degrades Ang II, counteracting its chronic effects, and serves as the SARS-CoV-2 receptor. Ace, the coronavirus, and COVID-19 comorbidities all regulate Ace2, but we do not yet understand how. To exploit zebrafish (Danio rerio) to help understand the relationship of the RAAS to COVID-19, we must identify zebrafish orthologs and co-orthologs of human RAAS genes and understand their expression patterns. To achieve these goals, we conducted genomic and phylogenetic analyses and investigated single cell transcriptomes. Results showed that most human RAAS genes have one or more zebrafish orthologs or co-orthologs. Results identified a specific type of enterocyte as the specific site of expression of zebrafish orthologs of key RAAS components, including Ace, Ace2, Slc6a19 (SARS-CoV-2 co-receptor), and the Angiotensin-related peptide cleaving enzymes Anpep (receptor for the common cold coronavirus HCoV-229E), and Dpp4 (receptor for the Middle East Respiratory Syndrome virus, MERS-CoV). Results identified specific vascular cell subtypes expressing Ang II receptors, apelin, and apelin receptor genes. These results identify genes and cell types to exploit zebrafish as a disease model for understanding mechanisms of COVID-19.


Subject(s)
Enterocytes , Gene Expression Regulation , Renin-Angiotensin System/genetics , SARS-CoV-2 , Zebrafish Proteins , Zebrafish , Animals , COVID-19/genetics , COVID-19/metabolism , Disease Models, Animal , Enterocytes/metabolism , Enterocytes/virology , Humans , SARS-CoV-2/genetics , SARS-CoV-2/metabolism , Zebrafish/genetics , Zebrafish/metabolism , Zebrafish/virology , Zebrafish Proteins/biosynthesis , Zebrafish Proteins/genetics
11.
Mucosal Immunol ; 14(3): 751-761, 2021 05.
Article in English | MEDLINE | ID: mdl-33674763

ABSTRACT

Although they globally cause viral gastroenteritis in children, astroviruses are understudied due to the lack of well-defined animal models. While murine astroviruses (muAstVs) chronically infect immunodeficient mice, a culture system and understanding of their pathogenesis is lacking. Here, we describe a platform to cultivate muAstV using air-liquid interface (ALI) cultures derived from mouse enteroids, which support apical infection and release. Chronic muAstV infection occurs predominantly in the small intestine and correlates with higher interferon-lambda (IFN-λ) expression. MuAstV stimulates IFN-λ production in ALI, recapitulating our in vivo findings. We demonstrate that goblet cells and enterocytes are targets for chronic muAstV infection in vivo, and that infection is enhanced by parasite co-infection or type 2 cytokine signaling. Depletion of goblet cells from ALI limits muAstV infection in vitro. During chronic infection, muAstV stimulates IFN-λ production in infected cells and induces ISGs throughout the intestinal epithelium in an IFN-λ-receptor-dependent manner. Collectively, our study provides insights into the cellular tropism and innate immune responses to muAstV and establishes an enteroid-based culture system to propagate muAstV in vitro.


Subject(s)
Astroviridae Infections/immunology , Astroviridae/physiology , Cytokines/metabolism , Enterocytes/virology , Gastroenteritis/immunology , Goblet Cells/virology , Th2 Cells/immunology , Animals , Cells, Cultured , Coinfection , Enterocytes/immunology , Goblet Cells/immunology , Humans , Immunity, Innate , Mice , Mice, Inbred C57BL , Mice, Knockout , Organ Culture Techniques , Viral Tropism
12.
EMBO Mol Med ; 13(4): e13191, 2021 04 09.
Article in English | MEDLINE | ID: mdl-33544398

ABSTRACT

SARS-CoV-2, the agent that causes COVID-19, invades epithelial cells, including those of the respiratory and gastrointestinal mucosa, using angiotensin-converting enzyme-2 (ACE2) as a receptor. Subsequent inflammation can promote rapid virus clearance, but severe cases of COVID-19 are characterized by an inefficient immune response that fails to clear the infection. Using primary epithelial organoids from human colon, we explored how the central antiviral mediator IFN-γ, which is elevated in COVID-19, affects epithelial cell differentiation, ACE2 expression, and susceptibility to infection with SARS-CoV-2. In mouse and human colon, ACE2 is mainly expressed by surface enterocytes. Inducing enterocyte differentiation in organoid culture resulted in increased ACE2 production. IFN-γ treatment promoted differentiation into mature KRT20+ enterocytes expressing high levels of ACE2, increased susceptibility to SARS-CoV-2 infection, and resulted in enhanced virus production in infected cells. Similarly, infection-induced epithelial interferon signaling promoted enterocyte maturation and enhanced ACE2 expression. We here reveal a mechanism by which IFN-γ-driven inflammatory responses induce a vulnerable epithelial state with robust replication of SARS-CoV-2, which may have an impact on disease outcome and virus transmission.


Subject(s)
COVID-19/etiology , Interferon-gamma/immunology , Models, Immunological , SARS-CoV-2 , Angiotensin-Converting Enzyme 2/genetics , Angiotensin-Converting Enzyme 2/metabolism , Animals , COVID-19/immunology , COVID-19/pathology , Cell Differentiation/immunology , Colon/immunology , Colon/pathology , Colon/virology , Disease Susceptibility , Enterocytes/metabolism , Enterocytes/pathology , Enterocytes/virology , Gene Expression , Host Microbial Interactions/immunology , Humans , Interferon-gamma/administration & dosage , Intestinal Mucosa/immunology , Intestinal Mucosa/pathology , Intestinal Mucosa/virology , Mice , Organoids/immunology , Organoids/pathology , Organoids/virology , SARS-CoV-2/genetics , SARS-CoV-2/immunology , SARS-CoV-2/pathogenicity , Virus Replication/immunology
13.
RNA Biol ; 18(5): 796-808, 2021 05.
Article in English | MEDLINE | ID: mdl-33406999

ABSTRACT

The pathogenic human enterovirus EV-A71 has raised serious public health concerns. A hallmark of EV-A71 infection is the distortion of host transcriptomes in favour of viral replication. While high-throughput approaches have been exploited to dissect these gene dysregulations, they do not fully capture molecular perturbations at the single-cell level and in a physiologically relevant context. In this study, we applied a single-cell RNA sequencing approach on infected differentiated enterocyte cells (C2BBe1), which model the gastrointestinal epithelium targeted initially by EV-A71. Our single-cell analysis of EV-A71-infected culture provided several lines of illuminating observations: 1) This systems approach demonstrated extensive cell-to-cell variation in a single culture upon viral infection and delineated transcriptomic differences between the EV-A71-infected and bystander cells. 2) By analysing expression profiles of known EV-A71 receptors and entry facilitation factors, we found that ANXA2 was closely correlated in expression with the viral RNA in the infected population, supporting its role in EV-A71 entry in the enteric cells. 3) We further catalogued dysregulated lncRNAs elicited by EV-A71 infection and demonstrated the functional implication of lncRNA CYTOR in promoting EV-A71 replication. Viewed together, our single-cell transcriptomic analysis illustrated at the single-cell resolution the heterogeneity of host susceptibility to EV-A71 and revealed the involvement of lncRNAs in host antiviral response.


Subject(s)
Enterovirus A, Human/pathogenicity , Host-Pathogen Interactions/genetics , Transcriptome , Cells, Cultured , Enterocytes/metabolism , Enterocytes/pathology , Enterocytes/virology , Enterovirus A, Human/genetics , Enterovirus A, Human/immunology , Enterovirus Infections/genetics , Enterovirus Infections/immunology , Enterovirus Infections/pathology , Enterovirus Infections/virology , Gene Expression Profiling , Gene Expression Regulation/genetics , Gene Expression Regulation/immunology , Host-Pathogen Interactions/immunology , Humans , Intestinal Mucosa/metabolism , Intestinal Mucosa/pathology , Intestinal Mucosa/virology , RNA, Long Noncoding/genetics , Single-Cell Analysis , Virus Replication/genetics
14.
Am J Infect Control ; 49(3): 385-386, 2021 03.
Article in English | MEDLINE | ID: mdl-32791258

ABSTRACT

COVD-19 disease is characteristically respiratory in nature; however, some patients have gastrointestinal symptoms. These include changes in taste, nausea/vomiting, abdominal pain, and diarrhea. A report has been published of a young patient who repeatedly tested positive in stool samples while nasopharyngeal tests remained negative. This raises doubts about our understanding of the dynamics of COVID-19 disease. The current report describes a need for selective stool testing to explore fecal shedding of viral RNA and presents a hypothesis for direct infection of enterocytes in cases of hypochlorhydria.


Subject(s)
Achlorhydria/virology , COVID-19/complications , Diarrhea/virology , Enterocytes/virology , SARS-CoV-2 , COVID-19/virology , Feces/virology , Gastrointestinal Tract/virology , Humans , RNA, Viral/metabolism
15.
Front Cell Infect Microbiol ; 10: 575559, 2020.
Article in English | MEDLINE | ID: mdl-33363049

ABSTRACT

The current COVID-19 pandemic is a great challenge for worldwide researchers in the human microbiota area because the mechanisms and long-term effects of the infection at the GI level are not yet deeply understood. In the current review, scientific literature including original research articles, clinical studies, epidemiological reports, and review-type articles concerning human intestinal infection with SARS-CoV-2 and the possible consequences on the microbiota were reviewed. Moreover, the following aspects pertaining to COVID-19 have also been discussed: transmission, resistance in the human body, the impact of nutritional status in relation to the intestinal microbiota, and the impact of comorbid metabolic disorders such as inflammatory bowel disease (IBS), obesity, and type two diabetes (T2D). The articles investigated show that health, age, and nutritional status are associated with specific communities of bacterial species in the gut, which could influence the clinical course of COVID-19 infection. Fecal microbiota alterations were associated with fecal concentrations of SARS-CoV-2 and COVID-19 severity. Patients suffering from metabolic and gastrointestinal (GI) disorders are thought to be at a moderate-to-high risk of infection with SARS-CoV-2, indicating the direct implication of gut dysbiosis in COVID-19 severity. However, additional efforts are required to identify the initial GI symptoms of COVID-19 for possible early intervention.


Subject(s)
COVID-19/microbiology , Dysbiosis/etiology , Gastrointestinal Microbiome , Pandemics , SARS-CoV-2/physiology , Animals , COVID-19/complications , COVID-19/epidemiology , COVID-19/transmission , Comorbidity , Diabetes Mellitus, Type 2/epidemiology , Diabetes Mellitus, Type 2/microbiology , Disease Reservoirs/virology , Enterocytes/pathology , Enterocytes/virology , Feces/microbiology , Feces/virology , Gastrointestinal Diseases/etiology , Gastrointestinal Diseases/microbiology , Humans , Irritable Bowel Syndrome/epidemiology , Irritable Bowel Syndrome/microbiology , Metabolic Syndrome/epidemiology , Metabolic Syndrome/microbiology , Obesity/epidemiology , Obesity/microbiology , Risk Factors , SARS-CoV-2/isolation & purification , SARS-CoV-2/pathogenicity
16.
Med Hypotheses ; 144: 110243, 2020 Nov.
Article in English | MEDLINE | ID: mdl-33254549

ABSTRACT

Recently, a new coronavirus (SARS-CoV-2) was discovered in China. Due to its high level of contagion, it has already reached most countries, quickly becoming a pandemic. Although the most common symptoms are related to breathing problems, SARS-CoV-2 infections also affect the gastrointestinal tract culminating in inflammation and diarrhea. However, the mechanisms related to these enteric manifestations are still not well understood. Evidence shows that the SARS-CoV-2 binds to the angiotensin-converting enzyme receptor 2 (ACE2) in host cells as a viral invasion mechanism and can infect the lungs and the gut. Other viruses have already been linked to intestinal symptoms through binding to ACE2. In turn, this medical hypothesis article conjectures that the ACE2 downregulation caused by the SARS-CoV-2 internalization could lead to decreased activation of the mechanistic target of mTOR with increased autophagy and lead to intestinal dysbiosis, resulting in diarrhea. Besides that, dysbiosis can directly affect the respiratory system through the lungs. Although there are clues to other viruses that modulate the ACE2/gut/lungs axis, including the participation of autophagy and dysbiosis in the development of gastrointestinal symptoms, there is still no evidence of the ACE2/mTOR/autophagy pathway in SARS-CoV-2 infections. Thus, we propose that the new coronavirus causes a change in the intestinal microbiota, which culminates in a diarrheal process through the ACE2/mTOR/autophagy pathway into enterocytes. Our assumption is supported by premises that unregulated intestinal microbiota increases the susceptibility to other diseases and extra-intestinal manifestations, which can even cause remote damage in lungs. These putative connections lead us to suggest and encourage future studies aiming at assessing the aforementioned hypothesis and regulating dysbiosis caused by SARS-CoV-2 infection, in order to confirm the decrease in lung injuries and the improvement in the prognosis of the disease.


Subject(s)
Angiotensin-Converting Enzyme 2/metabolism , Autophagy , COVID-19/metabolism , Diarrhea/complications , Dysbiosis/complications , SARS-CoV-2 , TOR Serine-Threonine Kinases/metabolism , COVID-19/complications , Enterocytes/virology , Gastrointestinal Microbiome , Gastrointestinal Tract/virology , Humans , Intestines/virology , Models, Theoretical , Pandemics , Renin-Angiotensin System
17.
Sci Rep ; 10(1): 12636, 2020 07 28.
Article in English | MEDLINE | ID: mdl-32724066

ABSTRACT

Rotavirus is the most common cause of acute gastroenteritis (AGE) in young children. Bacillus clausii (B. clausii) is a spore-forming probiotic that is able to colonize the gut. A mixture of four B. clausii strains (O/C, T, SIN and N/R) is commonly used for the treatment of AGE, and it has been demonstrated that it can reduce the duration and severity of diarrhea in children with AGE. Few studies have sought to characterize the mechanisms responsible for such beneficial effects. Intestinal effects of probiotics are likely to be strain-specific. We conducted a series of in vitro experiments investigating the activities of this mixture of B. clausii strains on biomarkers of mucosal barrier integrity and immune function in a cellular model of Rotavirus infection. B. clausii protected enterocytes against Rotavirus-induced decrease in trans-epithelial electrical resistance, and up-regulated expression of mucin 5AC and tight junction proteins (occludin and zonula occludens-1), all of which are important for effective mucosal barrier function. B. clausii also inhibited reactive oxygen species production and release of pro-inflammatory cytokines (interleukin-8 and interferon-ß) in Rotavirus-infected cells, and down-regulated pro-inflammatory Toll-like receptor 3 pathway gene expression. Such mechanisms likely contributed to the observed protective effects of B. clausii against reduced cell proliferation and increased apoptosis in Rotavirus-infected enterocytes.


Subject(s)
Bacillus clausii/growth & development , Enterocytes/drug effects , Erythrocytes/drug effects , Probiotics/administration & dosage , Rotavirus Infections/prevention & control , Rotavirus/drug effects , Apoptosis , Cell Cycle , Cell Proliferation , Enterocytes/virology , Erythrocytes/virology , Humans , In Vitro Techniques , Interferon-beta/metabolism , Interleukin-8/metabolism , Mucin 5AC/genetics , Mucin 5AC/metabolism , Occludin/genetics , Occludin/metabolism , Protective Agents , Rotavirus/isolation & purification , Rotavirus Infections/virology , Zonula Occludens-1 Protein/genetics , Zonula Occludens-1 Protein/metabolism
18.
Genes (Basel) ; 11(6)2020 06 11.
Article in English | MEDLINE | ID: mdl-32545271

ABSTRACT

There is increasing evidence of gastrointestinal (GI) infection by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). We surveyed the co-expression of SARS-CoV-2 entry genes ACE2 and TMPRSS2 throughout the GI tract to assess potential sites of infection. Publicly available and in-house single-cell RNA-sequencing datasets from the GI tract were queried. Enterocytes from the small intestine and colonocytes showed the highest proportions of cells co-expressing ACE2 and TMPRSS2. Therefore, the lower GI tract represents the most likely site of SARS-CoV-2 entry leading to GI infection.


Subject(s)
Betacoronavirus/metabolism , Enterocytes/metabolism , Lower Gastrointestinal Tract/metabolism , Peptidyl-Dipeptidase A/genetics , Serine Endopeptidases/genetics , Angiotensin-Converting Enzyme 2 , Base Sequence , COVID-19 , Cells, Cultured , Coronavirus Infections/pathology , Enterocytes/virology , Gastrointestinal Diseases/virology , Humans , Lower Gastrointestinal Tract/virology , Membrane Proteins/genetics , Membrane Proteins/metabolism , Pandemics , Peptidyl-Dipeptidase A/metabolism , Pneumonia, Viral/pathology , SARS-CoV-2 , Sequence Analysis , Serine Endopeptidases/metabolism , Virus Internalization
20.
Nat Med ; 26(7): 1077-1083, 2020 07.
Article in English | MEDLINE | ID: mdl-32405028

ABSTRACT

A novel coronavirus-severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2)-emerged in humans in Wuhan, China, in December 2019 and has since disseminated globally1,2. As of April 16, 2020, the confirmed case count of coronavirus disease 2019 (COVID-19) had surpassed 2 million. Based on full-genome sequence analysis, SARS-CoV-2 shows high homology to SARS-related coronaviruses identified in horseshoe bats1,2. Here we show the establishment and characterization of expandable intestinal organoids derived from horseshoe bats of the Rhinolophus sinicus species that can recapitulate bat intestinal epithelium. These bat enteroids are fully susceptible to SARS-CoV-2 infection and sustain robust viral replication. Development of gastrointestinal symptoms in some patients with COVID-19 and detection of viral RNA in fecal specimens suggest that SARS-CoV-2 might cause enteric, in addition to respiratory, infection3,4. Here we demonstrate active replication of SARS-CoV-2 in human intestinal organoids and isolation of infectious virus from the stool specimen of a patient with diarrheal COVID-19. Collectively, we established the first expandable organoid culture system of bat intestinal epithelium and present evidence that SARS-CoV-2 can infect bat intestinal cells. The robust SARS-CoV-2 replication in human intestinal organoids suggests that the human intestinal tract might be a transmission route of SARS-CoV-2.


Subject(s)
Betacoronavirus/pathogenicity , Coronavirus Infections/pathology , Coronavirus Infections/transmission , Intestines/virology , Organoids/virology , Pneumonia, Viral/pathology , Pneumonia, Viral/transmission , Animals , COVID-19 , Cell Differentiation , Cells, Cultured , Child, Preschool , Chiroptera/virology , Chlorocebus aethiops , Coronavirus Infections/virology , Enterocytes/pathology , Enterocytes/physiology , Enterocytes/virology , Female , Humans , Infant , Intestinal Mucosa/pathology , Intestinal Mucosa/virology , Intestines/pathology , Male , Organoids/pathology , Pandemics , Pneumonia, Viral/virology , Reverse Transcriptase Polymerase Chain Reaction , SARS-CoV-2 , Vero Cells , Viral Load/genetics , Viral Load/methods , Viral Tropism/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...