Your browser doesn't support javascript.
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
1.
Function (Oxf) ; 2(4): zqab027, 2021.
Article in English | MEDLINE | ID: covidwho-2188722

ABSTRACT

The SARS-CoV-2 receptor, angiotensin-converting enzyme-2 (ACE2), is expressed at levels of greatest magnitude in the small intestine as compared with all other human tissues. Enterocyte ACE2 is coexpressed as the apical membrane trafficking partner obligatory for expression and activity of the B0AT1 sodium-dependent neutral amino acid transporter. These components are assembled as an [ACE2:B0AT1]2 dimer-of-heterodimers quaternary complex that putatively steers SARS-CoV-2 tropism in the gastrointestinal (GI) tract. GI clinical symptomology is reported in about half of COVID-19 patients, and can be accompanied by gut shedding of virion particles. We hypothesized that within this 4-mer structural complex, each [ACE2:B0AT1] heterodimer pair constitutes a physiological "functional unit." This was confirmed experimentally by employing purified lyophilized enterocyte brush border membrane vesicles exposed to increasing doses of high-energy electron radiation from a 16 MeV linear accelerator. Based on radiation target theory, the results indicated the presence of Na+-dependent neutral amino acid influx transport activity functional unit with target size molecular weight 183.7 ± 16.8 kDa in situ in intact apical membranes. Each thermodynamically stabilized [ACE2:B0AT1] heterodimer functional unit manifests the transport activity within the whole ∼345 kDa [ACE2:B0AT1]2 dimer-of-heterodimers quaternary structural complex. The results are consistent with our prior molecular docking modeling and gut-lung axis approaches to understanding COVID-19. These findings advance understanding the physiology of B0AT1 interaction with ACE2 in the gut, and thereby contribute to translational developments designed to treat or mitigate COVID-19 variant outbreaks and/or GI symptom persistence in long-haul postacute sequelae of SARS-CoV-2.

2.
Int J Mol Sci ; 23(16)2022 Aug 15.
Article in English | MEDLINE | ID: covidwho-1987835

ABSTRACT

The gut is a well-established route of infection and target for viral damage by SARS-CoV-2. This is supported by the clinical observation that about half of COVID-19 patients exhibit gastrointestinal (GI) complications. We aimed to investigate whether the analysis of plasma could provide insight into gut barrier dysfunction in patients with COVID-19 infection. Plasma samples of COVID-19 patients (n = 146) and healthy individuals (n = 47) were collected during hospitalization and routine visits. Plasma microbiome was analyzed using 16S rRNA sequencing and gut permeability markers including fatty acid binding protein 2 (FABP2), peptidoglycan (PGN), and lipopolysaccharide (LPS) in both patient cohorts. Plasma samples of both cohorts contained predominately Proteobacteria, Firmicutes, Bacteroides, and Actinobacteria. COVID-19 subjects exhibit significant dysbiosis (p = 0.001) of the plasma microbiome with increased abundance of Actinobacteria spp. (p = 0.0332), decreased abundance of Bacteroides spp. (p = 0.0003), and an increased Firmicutes:Bacteroidetes ratio (p = 0.0003) compared to healthy subjects. The concentration of the plasma gut permeability marker FABP2 (p = 0.0013) and the gut microbial antigens PGN (p < 0.0001) and LPS (p = 0.0049) were significantly elevated in COVID-19 patients compared to healthy subjects. These findings support the notion that the intestine may represent a source for bacteremia and contribute to worsening COVID-19 outcomes. Therapies targeting the gut and prevention of gut barrier defects may represent a strategy to improve outcomes in COVID-19 patients.


Subject(s)
Actinobacteria , COVID-19 , Gastrointestinal Microbiome , Microbiota , Actinobacteria/genetics , Bacteria/genetics , Dysbiosis/microbiology , Feces/microbiology , Firmicutes/genetics , Gastrointestinal Microbiome/genetics , Humans , Lipopolysaccharides , Peptidoglycan , RNA, Ribosomal, 16S/genetics , SARS-CoV-2
3.
American Heart Journal Plus: Cardiology Research and Practice ; : 100050, 2021.
Article in English | ScienceDirect | ID: covidwho-1458822

ABSTRACT

Unexpected insights and practical advances in cardiovascular disease (CVD) are being discovered by rapidly advancing developments in supercomputers and machine learning (ML) software algorithms. These have been accelerated during the COVID-19 pandemic, and the resulting CVD translational implications of ML are steering new measures of prevention and treatment, new tools for objective clinical diagnosis, and even opportunities for rethinking basic foundations of CVD nosology. As the usual cardiovascular specialist may not be familiar with these tools, the editor has invited this brief overview.

5.
Hypertension ; 76(3): 651-661, 2020 09.
Article in English | MEDLINE | ID: covidwho-714225

ABSTRACT

Discovery of ACE2 (angiotensin-converting enzyme 2) revealed that the renin-angiotensin system has 2 counterbalancing arms. ACE2 is a major player in the protective arm, highly expressed in lungs and gut with the ability to mitigate cardiopulmonary diseases such as inflammatory lung disease. ACE2 also exhibits activities involving gut microbiome, nutrition, and as a chaperone stabilizing the neutral amino acid transporter, B0AT1, in gut. But the current interest in ACE2 arises because it is the cell surface receptor for the novel coronavirus, severe acute respiratory syndrome coronavirus-2, to infect host cells, similar to severe acute respiratory syndrome coronavirus-2. This suggests that ACE2 be considered harmful, however, because of its important other roles, it is paradoxically a potential therapeutic target for cardiopulmonary diseases, including coronavirus disease 2019 (COVID-19), caused by severe acute respiratory syndrome coronavirus-2. This review describes the discovery of ACE2, its physiological functions, and its place in the renin-angiotensin system. It illustrates new analyses of the structure of ACE2 that provides better understanding of its actions particularly in lung and gut, shedding of ACE2 by ADAM17 (a disintegrin and metallopeptidase domain 17 protein), and role of TMPRSS2 (transmembrane serine proteases 2) in severe acute respiratory syndrome coronavirus-2 entry into host cells. Cardiopulmonary diseases are associated with decreased ACE2 activity and the mitigation by increasing ACE2 activity along with its therapeutic relevance are addressed. Finally, the potential use of ACE2 as a treatment target in COVID-19, despite its role to allow viral entry into host cells, is suggested.


Subject(s)
Coronavirus Infections , Hypertension, Pulmonary , Pandemics , Peptidyl-Dipeptidase A/physiology , Pneumonia, Viral , Angiotensin-Converting Enzyme 2 , Betacoronavirus/physiology , COVID-19 , Coronavirus Infections/metabolism , Coronavirus Infections/physiopathology , Coronavirus Infections/therapy , Disease Management , Humans , Hypertension, Pulmonary/metabolism , Hypertension, Pulmonary/virology , Pneumonia, Viral/metabolism , Pneumonia, Viral/physiopathology , Pneumonia, Viral/therapy , Renin-Angiotensin System/physiology , SARS-CoV-2
6.
Diabetes ; 69(9): 1875-1886, 2020 09.
Article in English | MEDLINE | ID: covidwho-646761

ABSTRACT

Individuals with diabetes suffering from coronavirus disease 2019 (COVID-19) exhibit increased morbidity and mortality compared with individuals without diabetes. In this Perspective, we critically evaluate and argue that this is due to a dysregulated renin-angiotensin system (RAS). Previously, we have shown that loss of angiotensin-I converting enzyme 2 (ACE2) promotes the ACE/angiotensin-II (Ang-II)/angiotensin type 1 receptor (AT1R) axis, a deleterious arm of RAS, unleashing its detrimental effects in diabetes. As suggested by the recent reports regarding the pathogenesis of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), upon entry into the host, this virus binds to the extracellular domain of ACE2 in nasal, lung, and gut epithelial cells through its spike glycoprotein subunit S1. We put forth the hypothesis that during this process, reduced ACE2 could result in clinical deterioration in COVID-19 patients with diabetes via aggravating Ang-II-dependent pathways and partly driving not only lung but also bone marrow and gastrointestinal pathology. In addition to systemic RAS, the pathophysiological response of the local RAS within the intestinal epithelium involves mechanisms distinct from that of RAS in the lung; however, both lung and gut are impacted by diabetes-induced bone marrow dysfunction. Careful targeting of the systemic and tissue RAS may optimize clinical outcomes in subjects with diabetes infected with SARS-CoV-2.


Subject(s)
Angiotensin II/metabolism , Betacoronavirus/metabolism , Coronavirus Infections/metabolism , Diabetes Mellitus/metabolism , Peptidyl-Dipeptidase A/metabolism , Pneumonia, Viral/metabolism , Receptor, Angiotensin, Type 1/metabolism , Renin-Angiotensin System , Angiotensin-Converting Enzyme 2 , Bone Marrow/metabolism , COVID-19 , Coronavirus Infections/mortality , Coronavirus Infections/physiopathology , Humans , Intestinal Mucosa/metabolism , Pandemics , Pneumonia, Viral/mortality , Pneumonia, Viral/physiopathology , SARS-CoV-2 , Severity of Illness Index
SELECTION OF CITATIONS
SEARCH DETAIL