Your browser doesn't support javascript.
Show: 20 | 50 | 100
Results 1 - 20 de 20
Filter
2.
Infect Genet Evol ; 95: 105092, 2021 11.
Article in English | MEDLINE | ID: covidwho-1433676

ABSTRACT

OBJECTIVES: To compare the demographics, clinical characteristics and severity of patients infected with nine different SARS-CoV-2 variants, during three phases of the COVID-19 epidemic in Marseille. METHODS: A single centre retrospective cohort study was conducted in 1760 patients infected with SARS-CoV-2 of Nextstrain clades 20A, 20B, and 20C (first phase, February-May 2020), Pangolin lineages B.1.177 (we named Marseille-2) and B.1.160 (Marseille-4) variants (second phase, June-December 2020), and B.1.1.7 (alpha), B.1.351 (beta), P.1 (gamma) and A.27 (Marseille-501) variants (third phase, January 2021-today). Outcomes were the occurrence of clinical failures, including hospitalisation, transfer to the intensive-care unit, and death. RESULTS: During each phase, no major differences were observed with regards to age and gender distribution, the prevalence of chronic diseases, and clinical symptoms between variants circulating in a given phase. The B.1.177 and B.1.160 variants were associated with more severe outcomes. Infections occurring during the second phase were associated with a higher rate of death as compared to infections during the first and third phases. Patients in the second phase were more likely to be hospitalised than those in the third phase. Patients infected during the third phase were more frequently obese than others. CONCLUSION: A large cohort study is recommended to evaluate the transmissibility and to better characterise the clinical severity of emerging variants.


Subject(s)
COVID-19/pathology , Diabetes Mellitus/pathology , Genome, Viral , Hypertension/pathology , Obesity/pathology , SARS-CoV-2/pathogenicity , Adult , Aged , COVID-19/epidemiology , COVID-19/mortality , COVID-19/virology , Comorbidity , Diabetes Mellitus/epidemiology , Diabetes Mellitus/mortality , Diabetes Mellitus/virology , Female , France/epidemiology , Genotype , Heart Diseases/epidemiology , Heart Diseases/mortality , Heart Diseases/pathology , Heart Diseases/virology , Hospitalization/statistics & numerical data , Hospitals , Humans , Hypertension/epidemiology , Hypertension/mortality , Hypertension/virology , Intensive Care Units , Male , Middle Aged , Neoplasms/epidemiology , Neoplasms/mortality , Neoplasms/pathology , Neoplasms/virology , Obesity/epidemiology , Obesity/mortality , Obesity/virology , Phylogeny , Retrospective Studies , SARS-CoV-2/classification , SARS-CoV-2/genetics , SARS-CoV-2/isolation & purification , Sequence Analysis, RNA , Severity of Illness Index , Survival Analysis
3.
Front Endocrinol (Lausanne) ; 12: 726967, 2021.
Article in English | MEDLINE | ID: covidwho-1394754

ABSTRACT

In March 2020, the WHO declared coronavirus disease 2019 (COVID-19), caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), a global pandemic. Obesity was soon identified as a risk factor for poor prognosis, with an increased risk of intensive care admissions and mechanical ventilation, but also of adverse cardiovascular events. Obesity is associated with adipose tissue, chronic low-grade inflammation, and immune dysregulation with hypertrophy and hyperplasia of adipocytes and overexpression of pro-inflammatory cytokines. However, to implement appropriate therapeutic strategies, exact mechanisms must be clarified. The role of white visceral adipose tissue, increased in individuals with obesity, seems important, as a viral reservoir for SARS-CoV-2 via angiotensin-converting enzyme 2 (ACE2) receptors. After infection of host cells, the activation of pro-inflammatory cytokines creates a setting conducive to the "cytokine storm" and macrophage activation syndrome associated with progression to acute respiratory distress syndrome. In obesity, systemic viral spread, entry, and prolonged viral shedding in already inflamed adipose tissue may spur immune responses and subsequent amplification of a cytokine cascade, causing worse outcomes. More precisely, visceral adipose tissue, more than subcutaneous fat, could predict intensive care admission; and lower density of epicardial adipose tissue (EAT) could be associated with worse outcome. EAT, an ectopic adipose tissue that surrounds the myocardium, could fuel COVID-19-induced cardiac injury and myocarditis, and extensive pneumopathy, by strong expression of inflammatory mediators that could diffuse paracrinally through the vascular wall. The purpose of this review is to ascertain what mechanisms may be involved in unfavorable prognosis among COVID-19 patients with obesity, especially cardiovascular events, emphasizing the harmful role of excess ectopic adipose tissue, particularly EAT.


Subject(s)
COVID-19/metabolism , Cardiomyopathies/metabolism , Intra-Abdominal Fat/metabolism , Obesity/metabolism , Adipose Tissue/metabolism , Adipose Tissue/pathology , Angiotensin-Converting Enzyme 2/metabolism , COVID-19/complications , COVID-19/immunology , Cardiomyopathies/immunology , Cardiomyopathies/pathology , Heart Diseases/immunology , Heart Diseases/metabolism , Heart Diseases/pathology , Humans , Inflammation , Intra-Abdominal Fat/pathology , Obesity/complications , Obesity/immunology , Obesity/pathology , Pericardium , Prognosis , SARS-CoV-2/metabolism , Serine Endopeptidases/metabolism
4.
Cardiovasc Pathol ; 54: 107370, 2021.
Article in English | MEDLINE | ID: covidwho-1309178

ABSTRACT

BACKGROUND: Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is commonly associated with myocardial injury and heart failure. The pathophysiology behind this phenomenon remains unclear, with many diverse and multifaceted hypotheses. To contribute to this understanding, we describe the underlying cardiac findings in fifty patients who died with coronavirus disease 2019 (COVID-19). METHODS: Included were autopsies performed on patients with a positive SARS-CoV-2 reverse-transcriptase-polymerase-chain reaction test from the index hospitalization. In the case of out-of-hospital death, patients were included if post-mortem testing was positive. Complete autopsies were performed according to a COVID-19 safety protocol, and all patients underwent both macroscopic and microscopic examination. If available, laboratory findings and echocardiograms were reported. RESULTS: The median age of the decedents was 63.5 years. The most common comorbidities included hypertension (90.0%), diabetes (56.0%) and obesity (50.0%). Lymphocytic inflammatory infiltrates in the heart were present in eight (16.0%) patients, with focal myocarditis present in two (4.0%) patients. Acute myocardial ischemia was observed in eight (16.0%) patients. The most common findings were myocardial fibrosis (80.0%), hypertrophy (72.0%), and microthrombi (66.0%). The most common causes of death were COVID-19 pneumonia in 18 (36.0%), COVID-19 pneumonia with bacterial superinfection in 12 (24.0%), and COVID-19 pneumonia with pulmonary embolism in 10 (20.0%) patients. CONCLUSIONS: Cardiovascular comorbidities were prevalent, and pathologic changes associated with hypertensive and atherosclerotic cardiovascular disease were the most common findings. Despite markedly elevated inflammatory markers and cardiac enzymes, few patients exhibited inflammatory infiltrates or necrosis within cardiac myocytes. A unifying pathophysiologic mechanism behind myocardial injury in COVID-19 remains elusive, and additional autopsy studies are needed.


Subject(s)
COVID-19/pathology , Heart Diseases/pathology , Myocardium/pathology , SARS-CoV-2/pathogenicity , Adult , Aged , Aged, 80 and over , Atherosclerosis/mortality , Atherosclerosis/pathology , Autopsy , COVID-19/immunology , COVID-19/mortality , COVID-19/virology , Comorbidity , Female , Heart Diseases/immunology , Heart Diseases/mortality , Heart Diseases/virology , Host-Pathogen Interactions , Humans , Hypertension/mortality , Hypertension/pathology , Inflammation Mediators/analysis , Male , Middle Aged , Myocardium/immunology , Necrosis , SARS-CoV-2/immunology , Up-Regulation
5.
Life Sci ; 281: 119718, 2021 Sep 15.
Article in English | MEDLINE | ID: covidwho-1271709

ABSTRACT

AIMS: Hypoxia, a pathophysiological condition, is profound in several cardiopulmonary diseases (CPD). Every individual's lethality to a hypoxia state differs in terms of hypoxia exposure time, dosage units and dependent on the individual's genetic makeup. Most of the proposed markers for CPD were generally aim to distinguish disease samples from normal samples. Although, as per the 2018 GOLD guidelines, clinically useful biomarkers for several cardio pulmonary disease patients in stable condition have yet to be identified. We attempt to address these key issues through the identification of Dynamic Network Biomarkers (DNB) to detect hypoxia induced early warning signals of CPD before the catastrophic deterioration. MATERIALS AND METHODS: The human microvascular endothelial tissues microarray datasets (GSE11341) of lung and cardiac expose to hypoxia (1% O2) for 3, 24 and 48 h were retrieved from the public repository. The time dependent differentially expressed genes were subjected to tissue specificity and promoter analysis to filtrate the noise levels in the networks and to dissect the tissue specific hypoxia induced genes. These filtered out genes were used to construct the dynamic segmentation networks. The hypoxia induced dynamic differentially expressed genes were validated in the lung and heart tissues of male rats. These rats were exposed to hypobaric hypoxia (simulated altitude of 25,000 or PO2 - 282 mm of Hg) progressively for 3, 24 and 48 h. KEY FINDINGS: To identify the temporal key genes regulated in hypoxia, we ranked the dominant genes based on their consolidated topological features from tissue specific networks, time dependent networks and dynamic networks. Overall topological ranking described VEGFA as a single node dynamic hub and strongly communicated with tissue specific genes to carry forward their tissue specific information. We named this type of VEGFAcentric dynamic networks as "V-DNBs". As a proof of principle, our methodology helped us to identify the V-DNBs specific for lung and cardiac tissues namely V-DNBL and V-DNBC respectively. SIGNIFICANCE: Our experimental studies identified VEGFA, SLC2A3, ADM and ENO2 as the minimum and sufficient candidates of V-DNBL. The dynamic expression patterns could be readily exploited to capture the pre disease state of hypoxia induced pulmonary vascular remodelling. Whereas in V-DNBC the minimum and sufficient candidates are VEGFA, SCL2A3, ADM, NDRG1, ENO2 and BHLHE40. The time dependent single node expansion indicates V-DNBC could also be the pre disease state pathological hallmark for hypoxia-associated cardiovascular remodelling. The network cross-talk and expression pattern between V-DNBL and V-DNBC are completely distinct. On the other hand, the great clinical advantage of V-DNBs for pre disease predictions, a set of samples during the healthy condition should suffice. Future clinical studies might further shed light on the predictive power of V-DNBs as prognostic and diagnostic biomarkers for CPD.


Subject(s)
Heart Diseases/metabolism , Hypoxia/metabolism , Lung Diseases/metabolism , Vascular Endothelial Growth Factor A/metabolism , Animals , Biomarkers/metabolism , Clinical Deterioration , Gene Expression Regulation , Heart Diseases/etiology , Heart Diseases/pathology , Humans , Hypoxia/complications , Hypoxia/genetics , Lung Diseases/etiology , Lung Diseases/pathology , Male , Rats , Rats, Sprague-Dawley
6.
Cardiovasc Res ; 117(10): 2148-2160, 2021 08 29.
Article in English | MEDLINE | ID: covidwho-1266112

ABSTRACT

The pandemic of coronavirus disease (COVID)-19 is a global threat, causing high mortality, especially in the elderly. The main symptoms and the primary cause of death are related to interstitial pneumonia. Viral entry also into myocardial cells mainly via the angiotensin converting enzyme type 2 (ACE2) receptor and excessive production of pro-inflammatory cytokines, however, also make the heart susceptible to injury. In addition to the immediate damage caused by the acute inflammatory response, the heart may also suffer from long-term consequences of COVID-19, potentially causing a post-pandemic increase in cardiac complications. Although the main cause of cardiac damage in COVID-19 remains coagulopathy with micro- (and to a lesser extent macro-) vascular occlusion, open questions remain about other possible modalities of cardiac dysfunction, such as direct infection of myocardial cells, effects of cytokines storm, and mechanisms related to enhanced coagulopathy. In this opinion paper, we focus on these lesser appreciated possibilities and propose experimental approaches that could provide a more comprehensive understanding of the cellular and molecular bases of cardiac injury in COVID-19 patients. We first discuss approaches to characterize cardiac damage caused by possible direct viral infection of cardiac cells, followed by formulating hypotheses on how to reproduce and investigate the hyperinflammatory and pro-thrombotic conditions observed in the heart of COVID-19 patients using experimental in vitro systems. Finally, we elaborate on strategies to discover novel pathology biomarkers using omics platforms.


Subject(s)
COVID-19/virology , Heart Diseases/virology , Heart/virology , Myocytes, Cardiac/virology , SARS-CoV-2/pathogenicity , Animals , Biomarkers/metabolism , Blood Coagulation , COVID-19/complications , Fibrosis , Heart/physiopathology , Heart Diseases/metabolism , Heart Diseases/pathology , Heart Diseases/physiopathology , Host-Pathogen Interactions , Humans , Inflammation Mediators/metabolism , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/pathology , Ventricular Remodeling
7.
Pediatr Ann ; 50(3): e128-e135, 2021 Mar.
Article in English | MEDLINE | ID: covidwho-1196056

ABSTRACT

Coronavirus disease 2019 (COVID-19) has a predilection to cardiac involvement. The early clinical phase, during viremia, may manifest as pericarditis, acute myocarditis, and sepsis-related cardiomyopathy. Delayed presentations, such as multisystem inflammatory syndrome in children, coronary artery dilation/aneurysms, and late myocarditis, may occur in the weeks after the acute infection. These delayed presentations commonly test negative for severe acute respiratory syndrome coronavirus 2 via polymerase chain reaction testing and are thought to be primarily postviral hyperinflammatory sequelae. The long-term consequences of cardiac involvement in COVID-19 are unknown. Most recommendations for cardiac management are based on known conditions that are similar. For example, coronary aneurysms can be managed under Kawasaki disease guidelines. Similarly, for patients with COVID-19 myocarditis, they can be cleared for sports under protocols for other types of myocarditis. There is concern for cardiac involvement as a subclinical entity even in more minor presentations. Several expert algorithms have been developed for clearing competition athletes to return to exercise. Sports clearance should be individualized considering the severity of disease, age of patient, and performance level of the sport. [Pediatr Ann. 2021;50(3):e128-e135.].


Subject(s)
COVID-19/complications , Heart Diseases/diagnosis , Heart Diseases/etiology , Adolescent , Age Factors , COVID-19/pathology , Child , Heart Diseases/pathology , Humans , SARS-CoV-2 , Young Adult
8.
Cardiovasc Res ; 117(6): 1557-1566, 2021 05 25.
Article in English | MEDLINE | ID: covidwho-1127334

ABSTRACT

AIMS: Patients with severe respiratory syndrome caused by SARS-CoV-2 undergo cardiac complications due to hyper-inflammatory conditions. Although the presence of the virus has been detected in the myocardium of infected patients, and infection of induced pluripotent cell-derived cardiomyocytes has been demonstrated, the reported expression of Angiotensin-Converting Enzyme-2 (ACE2) in cardiac stromal cells suggests that SARS-CoV-2 may determine cardiac injury by sustaining productive infection and increasing inflammation. METHODS AND RESULTS: We analysed expression of ACE2 receptor in primary human cardiac stromal cells derived from cardiospheres, using proteomics and transcriptomics before exposing them to SARS-CoV-2 in vitro. Using conventional and high sensitivity PCR methods, we measured virus release in the cellular supernatants and monitored the intracellular viral bioprocessing. We performed high-resolution imaging to show the sites of intracellular viral production and demonstrated the presence of viral particles in the cells with electron microscopy. We finally used RT-qPCR assays to detect genes linked to innate immunity and fibrotic pathways coherently regulated in cells after exposure to the virus. CONCLUSIONS: Our findings indicate that cardiac stromal cells are susceptible to SARS-CoV-2 infection and produce variable viral yields depending on the extent of cellular ACE2 receptor expression. Interestingly, these cells also evolved towards hyper-inflammatory/pro-fibrotic phenotypes independently of ACE2 levels. Thus, SARS-CoV-2 infection of myocardial stromal cells could be involved in cardiac injury and explain the high number of complications observed in severe cases of COVID-19.


Subject(s)
Angiotensin-Converting Enzyme 2/metabolism , COVID-19/virology , Heart Diseases/virology , Myocardium/enzymology , Receptors, Virus/metabolism , SARS-CoV-2/pathogenicity , Stromal Cells/virology , Virion/pathogenicity , Aged , Aged, 80 and over , Angiotensin-Converting Enzyme 2/genetics , Animals , COVID-19/complications , Chlorocebus aethiops , Female , Fibrosis , Heart Diseases/enzymology , Heart Diseases/pathology , Host-Pathogen Interactions , Humans , Inflammation Mediators/metabolism , Male , Middle Aged , Myocardium/ultrastructure , Phenotype , Receptors, Virus/genetics , SARS-CoV-2/ultrastructure , Spheroids, Cellular , Stromal Cells/enzymology , Stromal Cells/ultrastructure , Vero Cells , Virion/ultrastructure
10.
Trends Mol Med ; 27(1): 7-10, 2021 01.
Article in English | MEDLINE | ID: covidwho-1060237

ABSTRACT

Recently, the US FDA has authorized a drug repurposing trial with calcitonin gene-related peptide (CGRP) receptor antagonists to reduce lung inflammation in coronavirus 2019 (COVID-19). However, the well-established cardiopulmonary protective effects of CGRP raise concerns about the safety of antagonizing CGRP in COVID-19. Awareness regarding potential cardiopulmonary adverse effects may enable their early detection and prevent illness from worsening.


Subject(s)
COVID-19 Drug Treatment , COVID-19 , Calcitonin Gene-Related Peptide Receptor Antagonists , Heart Diseases , Receptors, Calcitonin Gene-Related Peptide/metabolism , SARS-CoV-2/metabolism , Animals , COVID-19/metabolism , COVID-19/pathology , Calcitonin Gene-Related Peptide Receptor Antagonists/adverse effects , Calcitonin Gene-Related Peptide Receptor Antagonists/therapeutic use , Heart Diseases/chemically induced , Heart Diseases/metabolism , Heart Diseases/pathology , Humans
11.
Cardiovasc Res ; 116(14): 2207-2215, 2020 12 01.
Article in English | MEDLINE | ID: covidwho-1048209

ABSTRACT

AIMS: Coronavirus disease 2019 is caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) and has emerged as a global pandemic. SARS-CoV-2 infection can lead to elevated markers of cardiac injury associated with higher risk of mortality. It is unclear whether cardiac injury is caused by direct infection of cardiomyocytes or is mainly secondary to lung injury and inflammation. Here, we investigate whether cardiomyocytes are permissive for SARS-CoV-2 infection. METHODS AND RESULTS: Two strains of SARS-CoV-2 infected human induced pluripotent stem cell-derived cardiomyocytes as demonstrated by detection of intracellular double-stranded viral RNA and viral spike glycoprotein expression. Increasing concentrations of viral RNA are detected in supernatants of infected cardiomyocytes, which induced infections in Caco-2 cell lines, documenting productive infections. SARS-CoV-2 infection and induced cytotoxic and proapoptotic effects associated with it abolished cardiomyocyte beating. RNA sequencing confirmed a transcriptional response to viral infection as demonstrated by the up-regulation of genes associated with pathways related to viral response and interferon signalling, apoptosis, and reactive oxygen stress. SARS-CoV-2 infection and cardiotoxicity was confirmed in a 3D cardiosphere tissue model. Importantly, viral spike protein and viral particles were detected in living human heart slices after infection with SARS-CoV-2. Coronavirus particles were further observed in cardiomyocytes of a patient with coronavirus disease 2019. Infection of induced pluripotent stem cell-derived cardiomyocytes was dependent on cathepsins and angiotensin-converting enzyme 2, and was blocked by remdesivir. CONCLUSION: This study demonstrates that SARS-CoV-2 infects cardiomyocytes in vitro in an angiotensin-converting enzyme 2- and cathepsin-dependent manner. SARS-CoV-2 infection of cardiomyocytes is inhibited by the antiviral drug remdesivir.


Subject(s)
Apoptosis , COVID-19/virology , Heart Diseases/virology , Myocytes, Cardiac/virology , SARS-CoV-2/pathogenicity , Adenosine Monophosphate/analogs & derivatives , Adenosine Monophosphate/pharmacology , Alanine/analogs & derivatives , Alanine/pharmacology , Angiotensin-Converting Enzyme 2/metabolism , Antiviral Agents/pharmacology , Apoptosis/drug effects , COVID-19/metabolism , COVID-19/pathology , Caco-2 Cells , Cathepsins/metabolism , Heart Diseases/drug therapy , Heart Diseases/metabolism , Heart Diseases/pathology , Host-Pathogen Interactions , Humans , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/pathology , Reactive Oxygen Species/metabolism , SARS-CoV-2/drug effects , Signal Transduction , COVID-19 Drug Treatment
12.
J Neuroimmune Pharmacol ; 16(1): 59-70, 2021 03.
Article in English | MEDLINE | ID: covidwho-1018438

ABSTRACT

COVID-19 is an infectious respiratory illness caused by the virus strain severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) and until now, there is no effective therapy against COVID-19. Since SARS-CoV-2 binds to angiotensin-converting enzyme 2 (ACE2) for entering into host cells, to target COVID-19 from therapeutic angle, we engineered a hexapeptide corresponding to the ACE2-interacting domain of SARS-CoV-2 (AIDS) that inhibits the association between receptor-binding domain-containing spike S1 and ACE-2. Accordingly, wild type (wt), but not mutated (m), AIDS peptide inhibited SARS-CoV-2 spike S1-induced activation of NF-κB and expression of IL-6 in human lungs cells. Interestingly, intranasal intoxication of C57/BL6 mice with recombinant SARS-CoV-2 spike S1 led to fever, increase in IL-6 in lungs, infiltration of neutrophils into the lungs, arrhythmias, and impairment in locomotor activities, mimicking some of the important symptoms of COVID-19. However, intranasal treatment with wtAIDS, but not mAIDS, peptide reduced fever, protected lungs, improved heart function, and enhanced locomotor activities in SARS-CoV-2 spike S1-intoxicated mice. Therefore, selective targeting of ACE2-to-SARS-CoV-2 interaction by wtAIDS may be beneficial for COVID-19.


Subject(s)
Angiotensin-Converting Enzyme 2/therapeutic use , COVID-19 Drug Treatment , COVID-19/complications , Fever/drug therapy , Fever/etiology , Heart Diseases/etiology , Heart Diseases/prevention & control , Inflammation/drug therapy , Inflammation/etiology , Lung Diseases/etiology , Lung Diseases/prevention & control , Peptide Fragments/therapeutic use , Administration, Intranasal , Animals , Arrhythmias, Cardiac/etiology , Arrhythmias, Cardiac/prevention & control , COVID-19/pathology , Female , Heart Diseases/pathology , Interleukin-6/metabolism , Lung Diseases/pathology , Male , Mice , Mice, Inbred C57BL , Motor Activity/drug effects , Neutrophil Infiltration/drug effects , Spike Glycoprotein, Coronavirus/toxicity
13.
ACS Appl Mater Interfaces ; 13(1): 312-323, 2021 Jan 13.
Article in English | MEDLINE | ID: covidwho-997781

ABSTRACT

In this study, we present a modulated synthesis nanocrystalline defective UiO-66 metal-organic framework as a potential chloroquine diphosphate (CQ) delivery system. Increasing the concentration of hydrochloric acid during the modulated synthesis resulted in a considerable increase of pore volume, which enhanced the CQ loading in CQ@UiO-66 composites. Drug release tests for CQ@UiO-66 composites have confirmed prolonged CQ release in comparison with pure CQ. In vivo tests on a Danio reiro model organism have revealed that CQ released from CQ@UiO-66 25% showed lower toxicity and fewer cardiotoxic effects manifested by cardiac malformations and arrhythmia in comparison to analogous doses of CQ. Cytotoxicity tests proved that the CQ loaded on the defective UiO-66 cargo resulted in increased viability of cardiac cells (H9C2) as compared to incubation with pure CQ. The experimental results presented here may be a step forward in the context of reducing the cardiotoxicity CQ.


Subject(s)
Chloroquine/analogs & derivatives , Heart Diseases/drug therapy , Metal-Organic Frameworks/pharmacology , Nanoparticles/chemistry , Animals , Chloroquine/adverse effects , Chloroquine/chemistry , Chloroquine/pharmacology , Disease Models, Animal , Drug Delivery Systems/adverse effects , Drug Liberation/drug effects , HEK293 Cells , Heart Diseases/chemically induced , Heart Diseases/pathology , Humans , Hydrochloric Acid/pharmacology , Metal-Organic Frameworks/chemistry , Organometallic Compounds/chemistry , Organometallic Compounds/pharmacology , Phthalic Acids/chemistry , Phthalic Acids/pharmacology , Zebrafish/genetics
14.
Int J Cardiovasc Imaging ; 37(4): 1349-1360, 2021 Apr.
Article in English | MEDLINE | ID: covidwho-932570

ABSTRACT

Severe acute respiratory syndrome coronavirus 2 (SARS- CoV-2), previously named "2019 novel coronavirus" (2019-nCoV) is an emerging disease and a major public health issue. At the moment, little is known, except that its spread is on a steady upward trend. That is the reason why it was declared pandemic since March 11th, 2020. Respiratory symptoms dominate the clinical manifestations of the virus, but in a few patients also other organs are involved, such as their heart. This review article provides an overview of the existing literature regarding imaging of heart injury during COVID-19 acute infection and follow-up.


Subject(s)
COVID-19/complications , Cardiac Imaging Techniques , Heart Diseases/diagnostic imaging , Myocardium/pathology , COVID-19/diagnosis , COVID-19/virology , Heart Diseases/pathology , Heart Diseases/virology , Host-Pathogen Interactions , Humans , Predictive Value of Tests , Prognosis , Risk Factors , SARS-CoV-2/pathogenicity
15.
Epidemiol Infect ; 148: e266, 2020 10 23.
Article in English | MEDLINE | ID: covidwho-889103

ABSTRACT

OBJECTIVES: Cardiac injury is associated with poor prognosis of 2019 novel coronavirus disease 2019 (COVID-19), but the risk factors for cardiac injury have not been fully studied. In this study, we carried out a systematic analysis of clinical characteristics in COVID-19 patients to determine potential risk factors for cardiac injury complicated COVID-19 virus infection. METHODS: We systematically searched relevant literature published in Pubmed, Embase, Europe PMC, CNKI and other databases. All statistical analyses were performed using STATA 16.0. RESULTS: We analysed 5726 confirmed cases from 17 studies. The results indicated that compared with non-cardiac-injured patients, patients with cardiac injury are older, with a greater proportion of male patients, with higher possibilities of existing comorbidities, with higher risks of clinical complications, need for mechanical ventilation, ICU transfer and mortality. Moreover, C-reactive protein, procalcitonin, D-dimer, NT-proBNP and blood creatinine in patients with cardiac injury are also higher while lymphocyte counts and platelet counts decreased. However, we fortuitously found that patients with cardiac injury did not present higher clinical specificity for chest distress (P = 0.304), chest pain (P = 0.334), palpitations (P = 0.793) and smoking (P = 0.234). Similarly, the risk of concomitant arrhythmia (P = 0.103) did not increase observably either. CONCLUSION: Age, male gender and comorbidities are risk factors for cardiac injury complicated COVID-19 infection. Such patients are susceptible to complications and usually have abnormal results of laboratory tests, leading to poor outcomes. Contrary to common cardiac diseases, cardiac injury complicated COVID-19 infection did not significantly induce chest distress, chest pain, palpitations or arrhythmias. Our study indicates that early prevention should be applied to COVID-19 patients with cardiac injury to reduce adverse outcomes.


Subject(s)
Coronavirus Infections/complications , Heart Diseases/complications , Pneumonia, Viral/complications , Age Factors , Betacoronavirus , COVID-19 , Comorbidity , Coronavirus Infections/pathology , Heart Diseases/pathology , Humans , Pandemics , Pneumonia, Viral/pathology , Risk Factors , SARS-CoV-2 , Sex Factors
16.
J Mol Cell Cardiol ; 143: 145-158, 2020 06.
Article in English | MEDLINE | ID: covidwho-831617

ABSTRACT

Cardiac troponin I (cTnI), the inhibitory-unit, and cardiac troponin T (cTnT), the tropomyosin-binding unit together with the Ca-binding unit (cTnC) of the hetero-trimeric troponin complex signal activation of the sarcomeres of the adult cardiac myocyte. The unique structure and heart myocyte restricted expression of cTnI and cTnT led to their worldwide use as biomarkers for acute myocardial infarction (AMI) beginning more than 30 years ago. Over these years, high sensitivity antibodies (hs-cTnI and hs-cTnT) have been developed. Together with careful determination of history, physical examination, and EKG, determination of serum levels using hs-cTnI and hs-cTnT permits risk stratification of patients presenting in the Emergency Department (ED) with chest pain. With the ability to determine serum levels of these troponins with high sensitivity came the question of whether such measurements may be of diagnostic and prognostic value in conditions beyond AMI. Moreover, the finding of elevated serum troponins in physiological states such as exercise and pathological states where cardiac myocytes may be affected requires understanding of how troponins may be released into the blood and whether such release may be benign. We consider these questions by relating membrane stability to the complex biology of troponin with emphasis on its sensitivity to the chemo-mechanical and micro-environment of the cardiac myocyte. We also consider the role determinations of serum troponins play in the precise phenotyping in personalized and precision medicine approaches to promote cardiac health.


Subject(s)
Cellular Microenvironment , Heart Diseases/metabolism , Myocytes, Cardiac/metabolism , Sarcomeres/metabolism , Troponin/metabolism , Aged , Animals , Biomarkers/blood , Cytoskeleton , Disease Susceptibility , Epitopes , Heart Diseases/etiology , Heart Diseases/pathology , Humans , Precision Medicine/methods , Protein Interaction Domains and Motifs , Proteolysis , Sarcomeres/genetics , Stress, Physiological , Translational Research, Biomedical , Troponin/blood
17.
J Mol Histol ; 51(6): 613-628, 2020 Dec.
Article in English | MEDLINE | ID: covidwho-813346

ABSTRACT

The outbreak of severe acute respiratory syndrome coronavirus 2 (SARS-CoV2) in December 2019 form Wuhan, China leads to coronavirus disease 2019 (COVID-19) pandemic. While the common cold symptoms are observed in mild cases, COVID-19 is accompanied by multiorgan failure in severe patients. The involvement of different organs in severe patients results in lengthening the hospitalization duration and increasing the mortality rate. In this review, we aimed to investigate the involvement of different organs in COVID-19 patients, particularly in severe cases. Also, we tried to define the potential underlying mechanisms of SARS-CoV2 induced multiorgan failure. The multi-organ dysfunction is characterized by acute lung failure, acute liver failure, acute kidney injury, cardiovascular disease, and as well as a wide spectrum of hematological abnormalities and neurological disorders. The most important mechanisms are related to the direct and indirect pathogenic features of SARS-CoV2. Although the presence of angiotensin-converting enzyme 2, a receptor of SARS-CoV2 in the lung, heart, kidney, testis, liver, lymphocytes, and nervous system was confirmed, there are controversial findings to about the observation of SARS-CoV2 RNA in these organs. Moreover, the organ failure may be induced by the cytokine storm, a result of increased levels of inflammatory mediators, endothelial dysfunction, coagulation abnormalities, and infiltration of inflammatory cells into the organs. Therefore, further investigations are needed to detect the exact mechanisms of pathogenesis. Since the involvement of several organs in COVID-19 patients is important for clinicians, increasing their knowledge may help to improve the outcomes and decrease the rate of mortality and morbidity.


Subject(s)
Coronavirus Infections/pathology , Heart Diseases/pathology , Kidney Diseases/pathology , Liver Diseases/pathology , Multiple Organ Failure/pathology , Pneumonia, Viral/pathology , Angiotensin-Converting Enzyme 2 , Betacoronavirus , COVID-19 , Cytokine Release Syndrome/pathology , Heart Diseases/virology , Humans , Kidney/pathology , Kidney Diseases/virology , Liver/pathology , Liver Diseases/virology , Lung/pathology , Multiple Organ Failure/virology , Myocardium/pathology , Pandemics , Peptidyl-Dipeptidase A/metabolism , SARS-CoV-2
20.
Basic Res Cardiol ; 115(3): 31, 2020 04 09.
Article in English | MEDLINE | ID: covidwho-46686

ABSTRACT

From January 2020, coronavirus disease (COVID-19) originated in China has spread around the world. The disease is caused by the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). The presence of myocarditis, cardiac arrest, and acute heart failure in COVID-19 patients suggests the existence of a relationship between SARS-CoV-2 infection and cardiac disease. The Notch signalling is a major regulator of cardiovascular function and it is also implicated in several biological processes mediating viral infections. In this report we discuss the possibility to target Notch signalling to prevent SARS-CoV-2 infection and interfere with the progression of COVID-19- associated heart and lungs disease.


Subject(s)
Betacoronavirus/pathogenicity , Coronavirus Infections/drug therapy , Coronavirus Infections/physiopathology , Heart Diseases/drug therapy , Heart Diseases/etiology , Lung Diseases/drug therapy , Lung Diseases/etiology , Pneumonia, Viral/drug therapy , Pneumonia, Viral/physiopathology , Receptors, Notch/antagonists & inhibitors , ADAM17 Protein/antagonists & inhibitors , Angiotensin-Converting Enzyme 2 , Betacoronavirus/drug effects , COVID-19 , China , Coronavirus Infections/pathology , Coronavirus Infections/virology , Disease Progression , Furin/metabolism , Heart Arrest/etiology , Heart Arrest/pathology , Heart Diseases/pathology , Heart Diseases/physiopathology , Heart Failure/etiology , Heart Failure/pathology , Humans , Interleukin-6/immunology , Lung Diseases/pathology , Lung Diseases/physiopathology , Myocarditis/etiology , Myocarditis/pathology , Pandemics , Peptidyl-Dipeptidase A/deficiency , Peptidyl-Dipeptidase A/metabolism , Pneumonia, Viral/pathology , Pneumonia, Viral/virology , Receptors, Notch/metabolism , SARS-CoV-2 , Signal Transduction/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL