Your browser doesn't support javascript.
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
2.
Eur J Med Chem ; 228: 114030, 2022 Jan 15.
Article in English | MEDLINE | ID: covidwho-1768048

ABSTRACT

The epidemic coronavirus disease 2019 (COVID-19) caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has now spread worldwide and efficacious therapeutics are urgently needed. 3-Chymotrypsin-like cysteine protease (3CLpro) is an indispensable protein in viral replication and represents an attractive drug target for fighting COVID-19. Herein, we report the discovery of 9,10-dihydrophenanthrene derivatives as non-peptidomimetic and non-covalent inhibitors of the SARS-CoV-2 3CLpro. The structure-activity relationships of 9,10-dihydrophenanthrenes as SARS-CoV-2 3CLpro inhibitors have carefully been investigated and discussed in this study. Among all tested 9,10-dihydrophenanthrene derivatives, C1 and C2 display the most potent SARS-CoV-2 3CLpro inhibition activity, with IC50 values of 1.55 ± 0.21 µM and 1.81 ± 0.17 µM, respectively. Further enzyme kinetics assays show that these two compounds dose-dependently inhibit SARS-CoV-2 3CLprovia a mixed-inhibition manner. Molecular docking simulations reveal the binding modes of C1 in the dimer interface and substrate-binding pocket of the target. In addition, C1 shows outstanding metabolic stability in the gastrointestinal tract, human plasma, and human liver microsome, suggesting that this agent has the potential to be developed as an orally administrated SARS-CoV-2 3CLpro inhibitor.


Subject(s)
Antiviral Agents/pharmacology , COVID-19 Drug Treatment , Coronavirus 3C Proteases/antagonists & inhibitors , Drug Discovery/methods , Antiviral Agents/chemistry , Antiviral Agents/therapeutic use , Dose-Response Relationship, Drug , Drug Evaluation, Preclinical , Gastrointestinal Tract/metabolism , Humans , Kinetics , Microsomes, Liver/metabolism , Molecular Docking Simulation , Protein Binding , Structure-Activity Relationship , Viral Nonstructural Proteins/antagonists & inhibitors
3.
Eur J Pharm Sci ; 172: 106100, 2022 May 01.
Article in English | MEDLINE | ID: covidwho-1587878

ABSTRACT

This collection of contributions from the European Network on Understanding Gastrointestinal Absorption-related Processes (UNGAP) community assembly aims to provide information on some of the current and newer methods employed to study the behaviour of medicines. It is the product of interactions in the immediate pre-Covid period when UNGAP members were able to meet and set up workshops and to discuss progress across the disciplines. UNGAP activities are divided into work packages that cover special treatment populations, absorption processes in different regions of the gut, the development of advanced formulations and the integration of food and pharmaceutical scientists in the food-drug interface. This involves both new and established technical approaches in which we have attempted to define best practice and highlight areas where further research is needed. Over the last months we have been able to reflect on some of the key innovative approaches which we were tasked with mapping, including theoretical, in silico, in vitro, in vivo and ex vivo, preclinical and clinical approaches. This is the product of some of us in a snapshot of where UNGAP has travelled and what aspects of innovative technologies are important. It is not a comprehensive review of all methods used in research to study drug dissolution and absorption, but provides an ample panorama of current and advanced methods generally and potentially useful in this area. This collection starts from a consideration of advances in a priori approaches: an understanding of the molecular properties of the compound to predict biological characteristics relevant to absorption. The next four sections discuss a major activity in the UNGAP initiative, the pursuit of more representative conditions to study lumenal dissolution of drug formulations developed independently by academic teams. They are important because they illustrate examples of in vitro simulation systems that have begun to provide a useful understanding of formulation behaviour in the upper GI tract for industry. The Leuven team highlights the importance of the physiology of the digestive tract, as they describe the relevance of gastric and intestinal fluids on the behaviour of drugs along the tract. This provides the introduction to microdosing as an early tool to study drug disposition. Microdosing in oncology is starting to use gamma-emitting tracers, which provides a link through SPECT to the next section on nuclear medicine. The last two papers link the modelling approaches used by the pharmaceutical industry, in silico to Pop-PK linking to Darwich and Aarons, who provide discussion on pharmacometric modelling, completing the loop of molecule to man.


Subject(s)
COVID-19 , Gastrointestinal Tract , Administration, Oral , Computer Simulation , Gastrointestinal Absorption/physiology , Gastrointestinal Tract/metabolism , Humans , Intestinal Absorption , Male , Models, Biological , Pharmaceutical Preparations/metabolism , Solubility
4.
Molecules ; 26(16)2021 Aug 22.
Article in English | MEDLINE | ID: covidwho-1376916

ABSTRACT

Alcohol consumption is associated with gut dysbiosis, increased intestinal permeability, endotoxemia, and a cascade that leads to persistent systemic inflammation, alcoholic liver disease, and other ailments. Craving for alcohol and its consequences depends, among other things, on the endocannabinoid system. We have analyzed the relative role of central vs. peripheral cannabinoid CB1 receptors (CB1R) using a "two-bottle" as well as a "drinking in the dark" paradigm in mice. The globally acting CB1R antagonist rimonabant and the non-brain penetrant CB1R antagonist JD5037 inhibited voluntary alcohol intake upon systemic but not upon intracerebroventricular administration in doses that elicited anxiogenic-like behavior and blocked CB1R-induced hypothermia and catalepsy. The peripherally restricted hybrid CB1R antagonist/iNOS inhibitor S-MRI-1867 was also effective in reducing alcohol consumption after oral gavage, while its R enantiomer (CB1R inactive/iNOS inhibitor) was not. The two MRI-1867 enantiomers were equally effective in inhibiting an alcohol-induced increase in portal blood endotoxin concentration that was caused by increased gut permeability. We conclude that (i) activation of peripheral CB1R plays a dominant role in promoting alcohol intake and (ii) the iNOS inhibitory function of MRI-1867 helps in mitigating the alcohol-induced increase in endotoxemia.


Subject(s)
Alcohol Drinking/pathology , Cannabinoid Receptor Antagonists/pharmacology , Endotoxemia/pathology , Ethanol/adverse effects , Nitric Oxide Synthase Type II/antagonists & inhibitors , Receptor, Cannabinoid, CB1/antagonists & inhibitors , Alcohol Drinking/blood , Animals , Anxiety/blood , Anxiety/complications , Behavior, Animal/drug effects , Catalepsy/chemically induced , Catalepsy/complications , Cyclohexanols/administration & dosage , Elevated Plus Maze Test , Endotoxemia/blood , Endotoxemia/complications , Endotoxins/blood , Gastrointestinal Tract/drug effects , Gastrointestinal Tract/metabolism , Hypothermia, Induced , Mice, Inbred C57BL , Nitric Oxide Synthase Type II/metabolism , Pyrazoles/administration & dosage , Receptor, Cannabinoid, CB1/metabolism , Rimonabant/administration & dosage , Rimonabant/pharmacology , Stereoisomerism , Sulfonamides/administration & dosage
5.
Front Cell Infect Microbiol ; 11: 590874, 2021.
Article in English | MEDLINE | ID: covidwho-1158345

ABSTRACT

Gut microbiome alterations may play a paramount role in determining the clinical outcome of clinical COVID-19 with underlying comorbid conditions like T2D, cardiovascular disorders, obesity, etc. Research is warranted to manipulate the profile of gut microbiota in COVID-19 by employing combinatorial approaches such as the use of prebiotics, probiotics and symbiotics. Prediction of gut microbiome alterations in SARS-CoV-2 infection may likely permit the development of effective therapeutic strategies. Novel and targeted interventions by manipulating gut microbiota indeed represent a promising therapeutic approach against COVID-19 immunopathogenesis and associated co-morbidities. The impact of SARS-CoV-2 on host innate immune responses associated with gut microbiome profiling is likely to contribute to the development of key strategies for application and has seldom been attempted, especially in the context of symptomatic as well as asymptomatic COVID-19 disease.


Subject(s)
COVID-19/pathology , Dysbiosis/microbiology , Gastrointestinal Microbiome/immunology , Gastrointestinal Tract/microbiology , Immunity, Innate/immunology , Angiotensin-Converting Enzyme 2/biosynthesis , Angiotensin-Converting Enzyme 2/genetics , Angiotensin-Converting Enzyme 2/metabolism , Bacteria/metabolism , COVID-19/therapy , Cardiovascular Diseases/pathology , Diabetes Mellitus, Type 2/pathology , Gastrointestinal Tract/immunology , Gastrointestinal Tract/metabolism , Gene Expression/genetics , Humans , Leukocyte L1 Antigen Complex/biosynthesis , Obesity/pathology , Probiotics/pharmacology , SARS-CoV-2/immunology , Severity of Illness Index
6.
Endocr Rev ; 41(3)2020 06 01.
Article in English | MEDLINE | ID: covidwho-1110054

ABSTRACT

Individuals with diabetes are at increased risk for bacterial, mycotic, parasitic, and viral infections. The severe acute respiratory syndrome (SARS)-CoV-2 (also referred to as COVID-19) coronavirus pandemic highlights the importance of understanding shared disease pathophysiology potentially informing therapeutic choices in individuals with type 2 diabetes (T2D). Two coronavirus receptor proteins, angiotensin-converting enzyme 2 (ACE2) and dipeptidyl peptidase-4 (DPP4) are also established transducers of metabolic signals and pathways regulating inflammation, renal and cardiovascular physiology, and glucose homeostasis. Moreover, glucose-lowering agents such as the DPP4 inhibitors, widely used in subjects with T2D, are known to modify the biological activities of multiple immunomodulatory substrates. Here, we review the basic and clinical science spanning the intersections of diabetes, coronavirus infections, ACE2, and DPP4 biology, highlighting clinical relevance and evolving areas of uncertainty underlying the pathophysiology and treatment of T2D in the context of coronavirus infection.


Subject(s)
Betacoronavirus , Coronavirus Infections/complications , Diabetes Mellitus, Type 2/complications , Diabetes Mellitus, Type 2/drug therapy , Pneumonia, Viral/complications , Angiotensin-Converting Enzyme 2 , Animals , COVID-19 , Coronavirus Infections/metabolism , Diabetes Mellitus, Type 1/complications , Diabetes Mellitus, Type 1/metabolism , Diabetes Mellitus, Type 2/metabolism , Dipeptidyl Peptidase 4/metabolism , Dipeptidyl-Peptidase IV Inhibitors/pharmacology , Dipeptidyl-Peptidase IV Inhibitors/therapeutic use , Gastrointestinal Tract/metabolism , Humans , Insulin/therapeutic use , Lung/metabolism , Obesity/complications , Obesity/metabolism , Pandemics , Peptidyl-Dipeptidase A/metabolism , Pneumonia, Viral/metabolism , Receptors, Coronavirus , Receptors, Virus/metabolism , Risk Factors , SARS-CoV-2 , Serine Endopeptidases/metabolism
7.
Gastroenterology ; 160(5): 1647-1661, 2021 04.
Article in English | MEDLINE | ID: covidwho-1065985

ABSTRACT

BACKGROUND & AIMS: Gastrointestinal (GI) manifestations have been increasingly reported in patients with coronavirus disease 2019 (COVID-19). However, the roles of the GI tract in severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection are not fully understood. We investigated how the GI tract is involved in SARS-CoV-2 infection to elucidate the pathogenesis of COVID-19. METHODS: Our previously established nonhuman primate (NHP) model of COVID-19 was modified in this study to test our hypothesis. Rhesus monkeys were infected with an intragastric or intranasal challenge with SARS-CoV-2. Clinical signs were recorded after infection. Viral genomic RNA was quantified by quantitative reverse transcription polymerase chain reaction. Host responses to SARS-CoV-2 infection were evaluated by examining inflammatory cytokines, macrophages, histopathology, and mucin barrier integrity. RESULTS: Intranasal inoculation with SARS-CoV-2 led to infections and pathologic changes not only in respiratory tissues but also in digestive tissues. Expectedly, intragastric inoculation with SARS-CoV-2 resulted in the productive infection of digestive tissues and inflammation in both the lung and digestive tissues. Inflammatory cytokines were induced by both types of inoculation with SARS-CoV-2, consistent with the increased expression of CD68. Immunohistochemistry and Alcian blue/periodic acid-Schiff staining showed decreased Ki67, increased cleaved caspase 3, and decreased numbers of mucin-containing goblet cells, suggesting that the inflammation induced by these 2 types of inoculation with SARS-CoV-2 impaired the GI barrier and caused severe infections. CONCLUSIONS: Both intranasal and intragastric inoculation with SARS-CoV-2 caused pneumonia and GI dysfunction in our rhesus monkey model. Inflammatory cytokines are possible connections for the pathogenesis of SARS-CoV-2 between the respiratory and digestive systems.


Subject(s)
COVID-19/transmission , Gastroenteritis/pathology , Gastrointestinal Tract/pathology , Lung/pathology , Animals , Bronchi/metabolism , Bronchi/pathology , COVID-19/immunology , COVID-19/metabolism , COVID-19/pathology , COVID-19 Nucleic Acid Testing , Caspase 3/metabolism , Cytokines/immunology , Disease Models, Animal , Gastric Mucosa , Gastroenteritis/metabolism , Gastroenteritis/virology , Gastrointestinal Tract/immunology , Gastrointestinal Tract/metabolism , Goblet Cells/pathology , Intestine, Small/metabolism , Intestine, Small/pathology , Ki-67 Antigen/metabolism , Lung/diagnostic imaging , Lung/immunology , Lung/metabolism , Macaca mulatta , Nasal Mucosa , RNA, Viral/isolation & purification , Random Allocation , Rectum/metabolism , Rectum/pathology , SARS-CoV-2 , Trachea/metabolism , Trachea/pathology
8.
Biomed Pharmacother ; 133: 111064, 2021 Jan.
Article in English | MEDLINE | ID: covidwho-1059802

ABSTRACT

COVID-19 is a pandemic caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). Early reported symptoms include fever, cough, and respiratory symptoms. There were few reports of digestive symptoms. However, with COVID-19 spreading worldwide, symptoms such as vomiting, diarrhoea, and abdominal pain have gained increasing attention. Research has found that angiotensin-converting enzyme 2 (ACE2), the SARS-CoV-2 receptor, is strongly expressed in the gastrointestinal tract and liver. Whether theoretically or clinically, many studies have suggested a close connection between COVID-19 and the digestive system. In this review, we summarize the digestive symptoms reported in existing research, discuss the impact of SARS-CoV-2 on the gastrointestinal tract and liver, and determine the possible mechanisms and aetiology, such as cytokine storm. In-depth exploration of the relationship between COVID-19 and the digestive system is urgently needed.


Subject(s)
COVID-19/complications , Gastrointestinal Diseases/etiology , Liver Diseases/etiology , Pandemics , SARS-CoV-2/pathogenicity , Angiotensin-Converting Enzyme 2/metabolism , Anorexia/etiology , Antiviral Agents/adverse effects , Bile Ducts/metabolism , Bile Ducts/virology , COVID-19/epidemiology , COVID-19/immunology , COVID-19/pathology , Chemical and Drug Induced Liver Injury/etiology , Comorbidity , Cytokine Release Syndrome/etiology , Cytopathogenic Effect, Viral , Gastrointestinal Diseases/epidemiology , Gastrointestinal Microbiome , Gastrointestinal Tract/metabolism , Gastrointestinal Tract/pathology , Gastrointestinal Tract/virology , Humans , Immunosuppressive Agents/adverse effects , Liver/metabolism , Liver/pathology , Liver/virology , Liver Diseases/epidemiology , Liver Transplantation , Non-alcoholic Fatty Liver Disease/etiology , Non-alcoholic Fatty Liver Disease/pathology , Non-alcoholic Fatty Liver Disease/virology , Postoperative Complications , Receptors, Virus/metabolism
9.
Eur Rev Med Pharmacol Sci ; 25(1): 527-540, 2021 Jan.
Article in English | MEDLINE | ID: covidwho-1052580

ABSTRACT

OBJECTIVE: The pathogenesis of coronavirus disease 2019 (COVID-19) remains clear, and no effective treatment exists. SARS-CoV-2 is the virus that causes COVID-19 and uses ACE2 as a cell receptor to invade human cells. Therefore, ACE2 is a key factor to analyze the SARS-CoV-2 infection mechanism. MATERIALS AND METHODS: We included 9,783 sequencing results of different organs, analyzed the effects of different ACE2 expression patterns in organs and immune regulation. RESULTS: We found that ACE2 expression was significantly increased in the lungs and digestive tract. The cellular immunity of individuals with elevated ACE2 expression is activated, whereas humoral immunity is dampened, leading to the release of many inflammatory factors dominated by IL6. Furthermore, by studying the sequencing results of SARS-CoV-2-infected and uninfected cells, IL6 was found to be an indicator of a significant increase in the number of infected cells. However, although patients with high expression of ACE2 will release many inflammatory factors dominated by IL6, cellular immunity in the colorectum is significantly activated. This effect may explain why individuals with SARS-CoV-2 infection have severe lung symptoms and digestion issues, which are important causes of milder symptoms. CONCLUSIONS: This finding indicates that ACE2 and IL6 inhibitors have important value in COVID-19.


Subject(s)
Angiotensin-Converting Enzyme 2/genetics , COVID-19/immunology , Immunity, Cellular , Interleukin-6/immunology , Lung/metabolism , SARS-CoV-2 , COVID-19/genetics , COVID-19/metabolism , Gastrointestinal Tract/immunology , Gastrointestinal Tract/metabolism , Gene Expression Profiling , Gene Ontology , Humans , Immunity, Cellular/genetics , Immunity, Humoral/genetics , Lung/immunology , Organ Specificity , Transcriptome
10.
World J Gastroenterol ; 27(1): 37-54, 2021 Jan 07.
Article in English | MEDLINE | ID: covidwho-1052513

ABSTRACT

The term lipidome is mentioned to the total amount of the lipids inside the biological cells. The lipid enters the human gastrointestinal tract through external source and internal source. The absorption pathway of lipids in the gastrointestinal tract has many ways; the 1st way, the lipid molecules are digested in the lumen before go through the enterocytes, digested products are re-esterified into complex lipid molecules. The 2nd way, the intracellular lipids are accumulated into lipoproteins (chylomicrons) which transport lipids throughout the whole body. The lipids are re-synthesis again inside the human body where the gastrointestinal lipids are: (1) Transferred into the endoplasmic reticulum; (2) Collected as lipoproteins such as chylomicrons; or (3) Stored as lipid droplets in the cytosol. The lipids play an important role in many stages of the viral replication cycle. The specific lipid change occurs during viral infection in advanced viral replication cycle. There are 47 lipids within 11 lipid classes were significantly disturbed after viral infection. The virus connects with blood-borne lipoproteins and apolipoprotein E to change viral infectivity. The viral interest is cholesterol- and lipid raft-dependent molecules. In conclusion, lipidome is important in gastrointestinal fat absorption and coronavirus disease 2019 (COVID-19) infection so lipidome is basic in gut metabolism and in COVID-19 infection success.


Subject(s)
COVID-19/metabolism , Gastrointestinal Absorption/physiology , Gastrointestinal Tract/physiopathology , Lipid Metabolism/physiology , SARS-CoV-2/metabolism , COVID-19/blood , COVID-19/physiopathology , COVID-19/virology , Cholesterol/blood , Cholesterol/metabolism , Gastrointestinal Tract/metabolism , Humans , Lipidomics , Lipoproteins/blood , Lipoproteins/metabolism , SARS-CoV-2/pathogenicity
11.
Eur J Pharm Biopharm ; 155: 103-111, 2020 Oct.
Article in English | MEDLINE | ID: covidwho-837756

ABSTRACT

The harsh conditions of the gastro-intestinal (GI) milieu pose a major barrier to the oral delivery of protein nanocages. Here we studied the stability of Nudaurelia capensis omega virus (NωV) virus-like particles (VLPs) in simulated GI fluids. NωV VLPs capsids and procapsids were transiently expressed in plants, the VLPs were incubated in various simulated GI fluids and their stability was determined by gel electrophoresis, density gradient ultracentrifugation and transmission electron microscopy (TEM). The results showed that the capsids were highly resistant to simulated gastric fluids at pH ≥ 3. Even under the harshest conditions, which consisted of a pepsin solution at pH 1.2, NωV capsids remained assembled as VLPs, though some digestion of the coat protein occurred. Moreover, 80.8% (±10.2%) stability was measured for NωV capsids upon 4 h incubation in simulated intestinal fluids. The high resistance of this protein cage to digestion and denaturation can be attributed to its distinctively compact structure. The more porous form of the VLPs, the procapsid, was less stable under all conditions. Our results suggest that NωV VLPs capsids are likely to endure transit through the GI tract, designating them as promising candidate protein nanocages for oral drug delivery.


Subject(s)
Capsid/metabolism , Insect Viruses , Nanoparticles , Plants/metabolism , RNA Viruses , Animals , Body Fluids , Capsid Proteins/biosynthesis , Centrifugation, Density Gradient , Drug Delivery Systems , Gastrointestinal Tract/metabolism , Humans , Hydrogen-Ion Concentration , Microscopy, Electron, Transmission , Pepsin A/chemistry
12.
Emerg Microbes Infect ; 9(1): 2169-2179, 2020 Dec.
Article in English | MEDLINE | ID: covidwho-792636

ABSTRACT

Studies on patients with the coronavirus disease-2019 (COVID-19) have implicated that the gastrointestinal (GI) tract is a major site of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection. We established a human GI tract cell line model highly permissive to SARS-CoV-2. These cells, C2BBe1 intestinal cells with a brush border having high levels of transmembrane serine protease 2 (TMPRSS2), showed robust viral propagation, and could be persistently infected with SARS-CoV-2, supporting the clinical observations of persistent GI infection in COVID-19 patients. Ectopic expression of viral receptors revealed that the levels of angiotensin-converting enzyme 2 (ACE2) expression confer permissiveness to SARS-CoV-2 infection, and TMPRSS2 greatly facilitates ACE2-mediated SARS-CoV-2 dissemination. Interestingly, ACE2 but not TMPRSS2 expression was significantly promoted by enterocytic differentiation, suggesting that the state of enterocytic differentiation may serve as a determining factor for viral propagation. Thus, our study sheds light on the pathogenesis of SARS-CoV-2 in the GI tract.


Subject(s)
Betacoronavirus/physiology , Coronavirus Infections/virology , Intestinal Mucosa/virology , Pneumonia, Viral/virology , Angiotensin-Converting Enzyme 2 , Betacoronavirus/genetics , COVID-19 , Cell Line , Coronavirus Infections/genetics , Coronavirus Infections/metabolism , Gastrointestinal Tract/metabolism , Gastrointestinal Tract/virology , Humans , Intestinal Mucosa/metabolism , Pandemics , Peptidyl-Dipeptidase A/genetics , Peptidyl-Dipeptidase A/metabolism , Pneumonia, Viral/genetics , Pneumonia, Viral/metabolism , Receptors, Virus/genetics , Receptors, Virus/metabolism , SARS-CoV-2 , Serine Endopeptidases/genetics , Serine Endopeptidases/metabolism
13.
Respir Physiol Neurobiol ; 283: 103548, 2021 01.
Article in English | MEDLINE | ID: covidwho-779595

ABSTRACT

BACKGROUND: Globally, the current medical emergency for novel coronavirus 2019 (COVID-19) leads to respiratory distress syndrome and death. PURPOSE: This review highlighted the effect of COVID-19 on systemic multiple organ failure syndromes. This review is intended to fill a gap in information about human physiological response to COVID-19 infections. This review may shed some light on other potential mechanisms and approaches in COVID -19 infections towards systemic multiorgan failure syndromes. FINDING: SARS-CoV-2 intervened mainly in the lung with progression to pneumonia and acute respiratory distress syndrome (ARDS) via the angiotensin-converting enzyme 2(ACE2) receptor. Depending on the viral load, infection spread through the ACE2 receptor further to various organs such as heart, liver, kidney, brain, endothelium, GIT, immune cell, and RBC (thromboembolism). This may be aggravated by cytokine storm with the extensive release of proinflammatory cytokines from the deregulating immune system. CONCLUSION: The widespread and vicious combinations of cytokines with organ crosstalk contribute to systemic hyper inflammation and ultimately lead to multiple organ dysfunction (Fig. 1). This comprehensive study comprises various manifestations of different organs in COVID-19 and may assist the clinicians and scientists pertaining to a broad approach to fight COVID 19.


Subject(s)
Coronavirus Infections/immunology , Cytokine Release Syndrome/immunology , Multiple Organ Failure/immunology , Peptidyl-Dipeptidase A/metabolism , Pneumonia, Viral/immunology , Respiratory Distress Syndrome/immunology , Spike Glycoprotein, Coronavirus/metabolism , Acute Kidney Injury/immunology , Acute Kidney Injury/physiopathology , Angiotensin-Converting Enzyme 2 , Arrhythmias, Cardiac/immunology , Arrhythmias, Cardiac/physiopathology , Betacoronavirus/metabolism , COVID-19 , Coronavirus Infections/physiopathology , Cytokine Release Syndrome/physiopathology , Cytokines/immunology , Endothelium, Vascular/metabolism , Erythrocytes/metabolism , Gastrointestinal Diseases/immunology , Gastrointestinal Diseases/physiopathology , Gastrointestinal Tract/metabolism , Heart Failure/immunology , Heart Failure/physiopathology , Humans , Inflammation/immunology , Kidney/metabolism , Liver/metabolism , Liver Diseases/immunology , Liver Diseases/physiopathology , Lung/metabolism , Multiple Organ Failure/physiopathology , Myocardium/metabolism , Pandemics , Pneumonia, Viral/physiopathology , Respiratory Distress Syndrome/physiopathology , SARS-CoV-2 , Thromboembolism/immunology , Thromboembolism/physiopathology , Viral Load
14.
Med Hypotheses ; 144: 110271, 2020 Nov.
Article in English | MEDLINE | ID: covidwho-753081

ABSTRACT

COVID-19 is caused by a new strain of coronavirus called SARS-coronavirus-2 (SARS-CoV-2), which is a positive sense single strand RNA virus. In humans, it binds to angiotensin converting enzyme 2 (ACE2) with the help a structural protein on its surface called the S-spike. Further, cleavage of the viral spike protein (S) by the proteases like transmembrane serine protease 2 (TMPRSS2) or Cathepsin L (CTSL) is essential to effectuate host cell membrane fusion and virus infectivity. COVID-19 poses intriguing issues with imperative relevance to clinicians. The pathogenesis of GI symptoms, diabetes-associated mortality, and disease recurrence in COVID-19 are of particular relevance because they cannot be sufficiently explained from the existing knowledge of the viral diseases. Tissue specific variations of SARS-CoV-2 cell entry related receptors expression in healthy individuals can help in understanding the pathophysiological basis the aforementioned collection of symptoms. ACE2 mediated dysregulation of sodium dependent glucose transporter (SGLT1 or SLC5A1) in the intestinal epithelium also links it to the pathogenesis of diabetes mellitus which can be a possible reason for the associated mortality in COVID-19 patients with diabetes. High expression of ACE2 in mucosal cells of the intestine and GB make these organs potential sites for the virus entry and replication. Continued replication of the virus at these ACE2 enriched sites may be a basis for the disease recurrence reported in some, thought to be cured, patients. Based on the human tissue specific distribution of SARS-CoV-2 cell entry factors ACE2 and TMPRSS2 and other supportive evidence from the literature, we hypothesize that SARS-CoV-2 host cell entry receptor-ACE2 based mechanism in GI tissue may be involved in COVID-19 (i) in the pathogenesis of digestive symptoms, (ii) in increased diabetic complications, (iii) in disease recurrence.


Subject(s)
Angiotensin-Converting Enzyme 2/metabolism , COVID-19/physiopathology , Diabetes Complications/metabolism , Diabetes Complications/mortality , Gastrointestinal Tract/virology , Serine Endopeptidases/metabolism , COVID-19/metabolism , Gastrointestinal Diseases/complications , Gastrointestinal Tract/metabolism , Gene Expression Regulation , Gene Expression Regulation, Viral , Humans , Incidence , Intestinal Mucosa/virology , Models, Theoretical , Protein Binding , Proteome , Recurrence , SARS-CoV-2 , Transcriptome , Treatment Outcome
15.
Korean J Gastroenterol ; 76(1): 4-8, 2020 07 25.
Article in English, Korean | MEDLINE | ID: covidwho-675920

ABSTRACT

The World Health Organization (WHO) declared the worldwide pandemic of Coronavirus disease-2019 (COVID-19) On March 11, 2020, just three months after the first outbreak of COVID-19 caused by Severe Acute Respiratory Syndrome Coronavirus 2 in China in December 2019. COVID-19 is a contagious disease that can affect anyone, anytime, anywhere, and has had a huge impact on our lives, including social, economic, educational, and cultural life. In this paper, I would like to explore the issues related to COVID-19 in the gastroenterology and share the experiences of domestic and overseas gastroenterologists, and ultimately to seek ways to effectively prepare for and cope with the pandemic era of COVID-19.


Subject(s)
Betacoronavirus/pathogenicity , Coronavirus Infections/pathology , Gastroenterologists/psychology , Pneumonia, Viral/pathology , Angiotensin-Converting Enzyme 2 , Betacoronavirus/isolation & purification , Betacoronavirus/metabolism , COVID-19 , Coronavirus Infections/transmission , Coronavirus Infections/virology , Endoscopy, Digestive System , Gastrointestinal Tract/metabolism , Humans , Pandemics , Peptidyl-Dipeptidase A/metabolism , Pneumonia, Viral/transmission , Pneumonia, Viral/virology , SARS-CoV-2
16.
Am J Physiol Gastrointest Liver Physiol ; 319(2): G245-G252, 2020 08 01.
Article in English | MEDLINE | ID: covidwho-637284

ABSTRACT

In addition to the typical respiratory response, new coronavirus disease 2019 (COVID-19) is also associated with very common gastrointestinal symptoms. Cases with gastrointestinal symptoms are more likely to be complicated by liver injury and acute respiratory distress syndrome (ARDS). If not treated in time, coma and circulatory failure may ensue. As severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infects the human body through the combination of angiotensin-converting enzyme 2 (ACE2) in the gastrointestinal tract, the mechanism underlying the gastrointestinal symptoms may involve damage to the intestinal mucosal barrier and promotion of the production of inflammatory factors. Indeed, after cells in the lungs become infected by SARS-CoV-2, effector CD4+ T cells reach the small intestine through the gut-lung axis, causing intestinal immune damage and diarrhea; early extensive use of antibacterial and antiviral drugs can also lead to diarrhea in patients. Thus, treatment options for COVID-19 patients should be promptly adjusted when they have gastrointestinal symptoms. As SARS-CoV-2 has been detected in the feces of COVID-19 patients, future prevention and control efforts must consider the possibility of fecal-oral transmission of the virus.


Subject(s)
Betacoronavirus/physiology , Coronavirus Infections , Gastrointestinal Diseases , Gastrointestinal Tract , Pandemics , Pneumonia, Viral , Angiotensin-Converting Enzyme 2 , Antiviral Agents/pharmacology , COVID-19 , Coronavirus Infections/metabolism , Coronavirus Infections/physiopathology , Coronavirus Infections/prevention & control , Coronavirus Infections/transmission , Gastrointestinal Diseases/epidemiology , Gastrointestinal Diseases/physiopathology , Gastrointestinal Diseases/therapy , Gastrointestinal Diseases/virology , Gastrointestinal Tract/metabolism , Gastrointestinal Tract/physiopathology , Gastrointestinal Tract/virology , Humans , Incidence , Infection Control/methods , Pandemics/prevention & control , Patient Selection , Peptidyl-Dipeptidase A/metabolism , Pneumonia, Viral/metabolism , Pneumonia, Viral/physiopathology , Pneumonia, Viral/prevention & control , Pneumonia, Viral/transmission , SARS-CoV-2
SELECTION OF CITATIONS
SEARCH DETAIL