Your browser doesn't support javascript.
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
1.
Blood Adv ; 2023 Mar 15.
Article in English | MEDLINE | ID: covidwho-2259452

ABSTRACT

Several independent lines of evidence suggest that megakaryocytes are dysfunctional in severe COVID-19. Herein, we characterized peripheral circulating megakaryocytes in a large cohort of COVID-19 inpatients and correlated subpopulation frequencies with clinical outcomes. Using peripheral blood, we show that megakaryocytes are increased in the systemic circulation in COVID-19, and we identify and validate S100A8/A9 as a defining marker of megakaryocyte dysfunction. We further reveal a subpopulation of S100A8/A9+ megakaryocytes that contain SARS-CoV-2 protein and RNA. Using flow cytometry of peripheral blood and in vitro studies on SARS-CoV-2 infected primary human megakaryocytes, we demonstrate that megakaryocytes can transfer viral antigens to emerging platelets. Mechanistically, we show that SARS-CoV-2 containing megakaryocytes are NFκB-activated, via p65 and p52, express NFκB-mediated cytokines, IL-6 and IL-1ß, and display high surface expression of TLR2 and TLR4, canonical drivers of NFκB. In a cohort of 218 COVID-19 inpatients, we correlate frequencies of megakaryocyte subpopulations with clinical outcomes and show that SARS-CoV-2 containing megakaryocytes are a strong risk factor for mortality and multi-organ injury, including respiratory failure, mechanical ventilation, acute kidney injury, thrombotic events, and ICU admission. Further, we show that SARS-CoV-2+ megakaryocytes are present in lung and brain autopsy tissues from deceased COVID-19 donors. This study offers the first evidence implicating SARS-CoV-2+ peripheral megakaryocytes in severe disease and suggests that circulating megakaryocytes warrant investigation in inflammatory disorders beyond COVID-19.

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
5.
Cells ; 9(11)2020 11 23.
Article in English | MEDLINE | ID: covidwho-945726

ABSTRACT

The novel severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has caused the ongoing coronavirus disease 2019 (COVID-19) pandemic, with more than 50 million cases reported globally. Findings have consistently identified an increased severity of SARS-CoV-2 infection in individuals with diabetes. Osteopontin, a cytokine-like matrix-associated phosphoglycoprotein, is elevated in diabetes and drives the expression of furin, a proprotein convertase implicated in the proteolytic processing and activation of several precursors, including chemokines, growth factors, hormones, adhesion molecules, and receptors. Elevated serum furin is a signature of diabetes mellitus progression and is associated with a dysmetabolic phenotype and increased risk of diabetes-linked premature mortality. Additionally, furin plays an important role in enhancing the infectivity of SARS-CoV-2 by promoting its entry and replication in the host cell. Here, we hypothesize that diabetes-induced osteopontin and furin protein upregulation results in worse outcomes in diabetic patients with SARS-CoV-2 infection owing to the roles of these protein in promoting viral infection and increasing metabolic dysfunction. Thus, targeting the osteopontin-furin axis may be a plausible strategy for reducing mortality in SARS-CoV-2 patients with diabetes.


Subject(s)
COVID-19/epidemiology , Diabetes Mellitus/blood , Diabetes Mellitus/epidemiology , Furin/blood , Osteopontin/blood , SARS-CoV-2/pathogenicity , Severity of Illness Index , Adolescent , Adult , Aged , Aged, 80 and over , COVID-19/mortality , COVID-19/virology , Child , Child, Preschool , Comorbidity , Female , Humans , Infant , Infant, Newborn , Male , Middle Aged , Renin-Angiotensin System , SARS-CoV-2/metabolism , Up-Regulation , Virulence , Young Adult
6.
Gastroenterology ; 160(1): 39-46, 2021 01.
Article in English | MEDLINE | ID: covidwho-936157

ABSTRACT

The role of angiotensin converting enzyme 2 has expanded from regulating the renin angiotensin system to regulating intestinal amino acid homeostasis and the gut microbiome. Recently, angiotensin converting enzyme 2 was identified as a primary receptor for severe acute respiratory syndrome coronaviruses 1 and 2 being expressed in multiple tissues including the luminal surface of the gut. In this brief perspective, we examine the role of angiotensin converting enzyme 2 as the receptor for severe acute respiratory syndrome coronavirus 2 and the impact of coronavirus disease 19 infection on the gut microbiome and on the gut epithelium.


Subject(s)
Angiotensin-Converting Enzyme 2/metabolism , COVID-19/enzymology , Gastroenteritis/enzymology , Gastrointestinal Microbiome , Intestinal Mucosa/enzymology , Receptors, Virus/metabolism , SARS-CoV-2/pathogenicity , Angiotensin-Converting Enzyme 2/therapeutic use , Animals , Anti-Inflammatory Agents/therapeutic use , Antiviral Agents/therapeutic use , COVID-19/microbiology , COVID-19/virology , Feces/microbiology , Feces/virology , Gastroenteritis/drug therapy , Gastroenteritis/microbiology , Gastroenteritis/virology , Gastrointestinal Microbiome/drug effects , Host-Pathogen Interactions , Humans , Intestinal Mucosa/drug effects , Intestinal Mucosa/microbiology , Intestinal Mucosa/virology , Renin-Angiotensin System , SARS-CoV-2/drug effects , Virus Internalization , COVID-19 Drug Treatment
7.
Clin Sci (Lond) ; 134(19): 2581-2595, 2020 10 16.
Article in English | MEDLINE | ID: covidwho-872745

ABSTRACT

The angiotensin-converting enzyme 2 (ACE2) has emerged as a critical regulator of the renin-angiotensin system (RAS), which plays important roles in cardiovascular homeostasis by regulating vascular tone, fluid and electrolyte balance. ACE2 functions as a carboxymonopeptidase hydrolyzing the cleavage of a single C-terminal residue from Angiotensin-II (Ang-II), the key peptide hormone of RAS, to form Angiotensin-(1-7) (Ang-(1-7)), which binds to the G-protein-coupled Mas receptor and activates signaling pathways that counteract the pathways activated by Ang-II. ACE2 is expressed in a variety of tissues and overwhelming evidence substantiates the beneficial effects of enhancing ACE2/Ang-(1-7)/Mas axis under many pathological conditions in these tissues in experimental models. This review will provide a succinct overview on current strategies to enhance ACE2 as therapeutic agent, and discuss limitations and future challenges. ACE2 also has other functions, such as acting as a co-factor for amino acid transport and being exploited by the severe acute respiratory syndrome coronaviruses (SARS-CoVs) as cellular entry receptor, the implications of these functions in development of ACE2-based therapeutics will also be discussed.


Subject(s)
Angiotensin-Converting Enzyme 2/therapeutic use , Animals , Disease Models, Animal , Gene Transfer Techniques , Humans , Models, Biological , Organ Specificity , Renin-Angiotensin System
8.
Ophthalmol Ther ; 9(4): 853-875, 2020 Dec.
Article in English | MEDLINE | ID: covidwho-871596

ABSTRACT

The coronavirus disease 2019 (COVID-19) pandemic has instigated severe global turmoil both medically and socioeconomically. Research continues to rapidly develop in order to fully comprehend the new severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). This study focuses on the rare ophthalmologic manifestations of the SARS-CoV-2 disease process in both adults and children. There is evidence to suggest that viral transmission can occur via tears and conjunctival secretions, although it is not a predominant finding. This review considers all the published studies describing ocular findings and SARS-CoV-2 viral transmission through the eye. The review addresses the ongoing debate over the importance of ocular manifestations during this pandemic. The most updated safety guidelines, protocols, timelines of ocular manifestations during the disease course, and treatment recommendations are discussed. The majority of patients with COVID-19 with eye symptoms presented with them initially. It is possible that the virus becomes inoculated at the site of the eye and spreads via the nasolacrimal duct to the respiratory system. There are also some reports which show that ocular findings present later in the disease course, suggestive of a correlation between ocular manifestation and increased disease severity as the infection becomes systemic. We highlight the importance of recognizing conjunctivitis as an early finding of COVID-19, and that testing or appropriate follow-up could be beneficial in both the pediatric and adult populations.

9.
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
10.
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
12.
Circ Res ; 126(10): 1456-1474, 2020 05 08.
Article in English | MEDLINE | ID: covidwho-217630

ABSTRACT

ACE2 (angiotensin-converting enzyme 2) has a multiplicity of physiological roles that revolve around its trivalent function: a negative regulator of the renin-angiotensin system, facilitator of amino acid transport, and the severe acute respiratory syndrome-coronavirus (SARS-CoV) and SARS-CoV-2 receptor. ACE2 is widely expressed, including, in the lungs, cardiovascular system, gut, kidneys, central nervous system, and adipose tissue. ACE2 has recently been identified as the SARS-CoV-2 receptor, the infective agent responsible for coronavirus disease 2019, providing a critical link between immunity, inflammation, ACE2, and cardiovascular disease. Although sharing a close evolutionary relationship with SARS-CoV, the receptor-binding domain of SARS-CoV-2 differs in several key amino acid residues, allowing for stronger binding affinity with the human ACE2 receptor, which may account for the greater pathogenicity of SARS-CoV-2. The loss of ACE2 function following binding by SARS-CoV-2 is driven by endocytosis and activation of proteolytic cleavage and processing. The ACE2 system is a critical protective pathway against heart failure with reduced and preserved ejection fraction including, myocardial infarction and hypertension, and against lung disease and diabetes mellitus. The control of gut dysbiosis and vascular permeability by ACE2 has emerged as an essential mechanism of pulmonary hypertension and diabetic cardiovascular complications. Recombinant ACE2, gene-delivery of Ace2, Ang 1-7 analogs, and Mas receptor agonists enhance ACE2 action and serve as potential therapies for disease conditions associated with an activated renin-angiotensin system. rhACE2 (recombinant human ACE2) has completed clinical trials and efficiently lowered or increased plasma angiotensin II and angiotensin 1-7 levels, respectively. Our review summarizes the progress over the past 20 years, highlighting the critical role of ACE2 as the novel SARS-CoV-2 receptor and as the negative regulator of the renin-angiotensin system, together with implications for the coronavirus disease 2019 pandemic and associated cardiovascular diseases.


Subject(s)
Betacoronavirus/physiology , Cardiovascular Diseases , Coronavirus Infections , Pandemics , Peptidyl-Dipeptidase A/physiology , Pneumonia, Viral , Renin-Angiotensin System/physiology , ADAM17 Protein/physiology , Angiotensin-Converting Enzyme 2 , Animals , COVID-19 , Cardiovascular Diseases/etiology , Cardiovascular Diseases/physiopathology , Coronavirus Infections/complications , Coronavirus Infections/drug therapy , Coronavirus Infections/metabolism , Coronavirus Infections/physiopathology , Coronavirus Infections/virology , Diabetes Complications/metabolism , Diabetes Complications/physiopathology , Humans , Molecular Targeted Therapy , Pneumonia, Viral/complications , Pneumonia, Viral/metabolism , Pneumonia, Viral/physiopathology , Pneumonia, Viral/virology , Receptors, Virus/physiology , SARS-CoV-2 , Virus Attachment , COVID-19 Drug Treatment
SELECTION OF CITATIONS
SEARCH DETAIL