Your browser doesn't support javascript.
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
1.
PLoS One ; 16(8): e0255976, 2021.
Article in English | MEDLINE | ID: covidwho-1365424

ABSTRACT

BACKGROUND: Cardiac injury associated with cytokine release frequently occurs in SARS-CoV-2 mediated coronavirus disease (COVID19) and mortality is particularly high in these patients. The mechanistic role of the COVID19 associated cytokine-storm for the concomitant cardiac dysfunction and associated arrhythmias is unclear. Moreover, the role of anti-inflammatory therapy to mitigate cardiac dysfunction remains elusive. AIMS AND METHODS: We investigated the effects of COVID19-associated inflammatory response on cardiac cellular function as well as its cardiac arrhythmogenic potential in rat and induced pluripotent stem cell derived cardiomyocytes (iPS-CM). In addition, we evaluated the therapeutic potential of the IL-1ß antagonist Canakinumab using state of the art in-vitro confocal and ratiometric high-throughput microscopy. RESULTS: Isolated rat ventricular cardiomyocytes were exposed to control or COVID19 serum from intensive care unit (ICU) patients with severe ARDS and impaired cardiac function (LVEF 41±5%; 1/3 of patients on veno-venous extracorporeal membrane oxygenation; CK 154±43 U/l). Rat cardiomyocytes showed an early increase of myofilament sensitivity, a decrease of Ca2+ transient amplitudes and altered baseline [Ca2+] upon exposure to patient serum. In addition, we used iPS-CM to explore the long-term effect of patient serum on cardiac electrical and mechanical function. In iPS-CM, spontaneous Ca2+ release events were more likely to occur upon incubation with COVID19 serum and nuclear as well as cytosolic Ca2+ release were altered. Co-incubation with Canakinumab had no effect on pro-arrhythmogenic Ca2+ release or Ca2+ signaling during excitation-contraction coupling, nor significantly influenced cellular automaticity. CONCLUSION: Serum derived from COVID19 patients exerts acute cardio-depressant and chronic pro-arrhythmogenic effects in rat and iPS-derived cardiomyocytes. Canakinumab had no beneficial effect on cellular Ca2+ signaling during excitation-contraction coupling. The presented method utilizing iPS-CM and in-vitro Ca2+ imaging might serve as a novel tool for precision medicine. It allows to investigate cytokine related cardiac dysfunction and pharmacological approaches useful therein.


Subject(s)
Antibodies, Monoclonal, Humanized/pharmacology , Arrhythmias, Cardiac , COVID-19 Drug Treatment , COVID-19 , Calcium Signaling/drug effects , Myocytes, Cardiac , SARS-CoV-2/metabolism , Adult , Aged , Animals , Arrhythmias, Cardiac/etiology , Arrhythmias, Cardiac/metabolism , Arrhythmias, Cardiac/pathology , COVID-19/complications , COVID-19/metabolism , COVID-19/pathology , Calcium/metabolism , Drug Evaluation, Preclinical , Female , Humans , Induced Pluripotent Stem Cells/metabolism , Induced Pluripotent Stem Cells/pathology , Male , Middle Aged , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/pathology , Rats , Rats, Sprague-Dawley , Ventricular Dysfunction, Left/drug therapy , Ventricular Dysfunction, Left/etiology , Ventricular Dysfunction, Left/metabolism , Ventricular Dysfunction, Left/pathology
2.
Indian J Med Microbiol ; 39(3): 380-383, 2021 07.
Article in English | MEDLINE | ID: covidwho-1243016

ABSTRACT

Coronavirus disease 2019 (COVID-19), may present with a myriad of clinical manifestations and complications. Patients with COVID-19 are at increased risk of pulmonary thromboembolism, acute cardiac injury, arrhythmias, acute stroke, and secondary infections. Mucormycosis is a catastrophic fungal infection characterized by vascular invasion, thrombosis, and necrosis of tissues. We report five cases of COVID-19 infection, who developed rhino-orbital mucormycosis, during the course of treatment. Early recognition of this life-threatening infection is the key to allow for optimal treatment and improved outcomes.


Subject(s)
Mucormycosis/diagnosis , Stroke/microbiology , Aged , Arrhythmias, Cardiac/epidemiology , Arrhythmias, Cardiac/pathology , COVID-19/diagnosis , COVID-19/therapy , Female , Humans , India/epidemiology , Male , Middle Aged , Mucormycosis/epidemiology , Mucormycosis/therapy , Pulmonary Embolism , Stroke/epidemiology , Stroke/therapy
3.
PLoS One ; 16(5): e0251918, 2021.
Article in English | MEDLINE | ID: covidwho-1238773

ABSTRACT

OBJECTIVES: Hydroxychloroquine is widely used to treat certain viral and rheumatic diseases including systemic lupus erythematosus. Cardiac arrhythmia is an important safety issue with hydroxychloroquine. The aim of this study was to investigate whether hydroxychloroquine increases new-onset arrhythmia among patients with systemic lupus erythematosus. METHODS: This was a nested case-control study using data from the Longitudinal Health Insurance Database of Taiwan. A conditional logistic regression model was used to analyse differences in the risk of arrhythmia between systemic lupus erythematosus patients with and without hydroxychloroquine treatment after controlling for related variables. RESULTS: A total of 2499 patients with newly diagnosed systemic lupus erythematosus were identified (81% females), of whom 251 were enrolled in the new-onset arrhythmia group (mean age 50.4 years) and 251 in the non-arrhythmia group (mean age 49.1 years). There was no significantly increased risk of cardiac arrhythmia (adjusted odds ratio = 1.49, 95% confidence interval: 0.98-2.25) or ventricular arrhythmia (adjusted odds ratio = 1.02, 95% confidence interval: 0.19-5.41) between the patients with and without hydroxychloroquine treatment. In addition, there were no significant differences in the risk of arrhythmia between those receiving hydroxychloroquine treatment for <180 days or ≥180 days, with a drug adherence rate of <50% or ≥50%, and receiving a daily dose of <400 mg or ≥400 mg. CONCLUSION: In patients with systemic lupus erythematosus, hydroxychloroquine treatment did not significantly increase the risk of cardiac arrhythmia or life-threatening ventricular arrhythmia regardless of the different hydroxychloroquine treatment duration, drug adherence rate, or daily dose.


Subject(s)
Antirheumatic Agents/administration & dosage , Arrhythmias, Cardiac/epidemiology , Hydroxychloroquine/administration & dosage , Lupus Erythematosus, Systemic/drug therapy , Arrhythmias, Cardiac/chemically induced , Arrhythmias, Cardiac/pathology , Female , Humans , Hydroxychloroquine/adverse effects , Lupus Erythematosus, Systemic/complications , Lupus Erythematosus, Systemic/epidemiology , Male , Medication Adherence , Middle Aged , Risk Factors , Taiwan/epidemiology
4.
J Mol Cell Cardiol ; 153: 106-110, 2021 04.
Article in English | MEDLINE | ID: covidwho-989436

ABSTRACT

The coronavirus disease 2019 (COVID-19) outbreak caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has become a global pandemic as declared by World Health Organization (WHO). In the absence of an effective treatment, different drugs with unknown effectiveness, including antimalarial hydroxychloroquine (HCQ), with or without concurrent administration with azithromycin (AZM), have been tested for treating COVID-19 patients with developed pneumonia. However, the efficacy and safety of HCQ and/or AZM have been questioned by recent clinical reports. Direct effects of these drugs on the human heart remain very poorly defined. To better understand the mechanisms of action of HCQ +/- AZM, we employed bioengineered human ventricular cardiac tissue strip (hvCTS) and anisotropic sheet (hvCAS) assays, made with human pluripotent stem cell (hPSC)-derived ventricular cardiomyocytes (hvCMs), which have been designed for measuring cardiac contractility and electrophysiology, respectively. Our hvCTS experiments showed that AZM induced a dose-dependent negative inotropic effect which could be aggravated by HCQ; electrophysiologically, as revealed by the hvCAS platform, AZM prolonged action potentials and induced spiral wave formations. Collectively, our data were consistent with reported clinical risks of HCQ and AZM on QTc prolongation/ventricular arrhythmias and development of heart failure. In conclusion, our study exposed the risks of HCQ/AZM administration while providing mechanistic insights for their toxicity. Our bioengineered human cardiac tissue constructs therefore provide a useful platform for screening cardiac safety and efficacy when developing therapeutics against COVID-19.


Subject(s)
Arrhythmias, Cardiac/pathology , Azithromycin/adverse effects , Chloroquine/adverse effects , Drug-Related Side Effects and Adverse Reactions/pathology , Myocardial Contraction , Myocytes, Cardiac/pathology , Ventricular Function/drug effects , Anti-Bacterial Agents/adverse effects , Antimalarials/adverse effects , Arrhythmias, Cardiac/chemically induced , Drug-Related Side Effects and Adverse Reactions/etiology , Humans , Myocytes, Cardiac/drug effects , Pluripotent Stem Cells/drug effects , Pluripotent Stem Cells/pathology , Tissue Engineering/methods , COVID-19 Drug Treatment
5.
Am J Emerg Med ; 38(7): 1504-1507, 2020 07.
Article in English | MEDLINE | ID: covidwho-912008

ABSTRACT

BACKGROUND: The coronavirus disease of 2019 (COVID-19) is caused by the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). While systemic inflammation and pulmonary complications can result in significant morbidity and mortality, cardiovascular complications may also occur. OBJECTIVE: This brief report evaluates cardiovascular complications in the setting of COVID-19 infection. DISCUSSION: The current COVID-19 pandemic has resulted in over one million infected worldwide and thousands of death. The virus binds and enters through angiotensin-converting enzyme 2 (ACE2). COVID-19 can result in systemic inflammation, multiorgan dysfunction, and critical illness. The cardiovascular system is also affected, with complications including myocardial injury, myocarditis, acute myocardial infarction, heart failure, dysrhythmias, and venous thromboembolic events. Current therapies for COVID-19 may interact with cardiovascular medications. CONCLUSIONS: Emergency clinicians should be aware of these cardiovascular complications when evaluating and managing the patient with COVID-19.


Subject(s)
Cardiovascular Diseases/pathology , Arrhythmias, Cardiac/pathology , Arrhythmias, Cardiac/virology , Betacoronavirus , COVID-19 , Cardiovascular Diseases/virology , Coronavirus Infections/immunology , Heart Failure/pathology , Heart Failure/virology , Humans , Myocarditis/pathology , Myocarditis/virology , Pandemics , Pneumonia, Viral/immunology , SARS-CoV-2
6.
Rev Med Virol ; 31(3): e2172, 2021 05.
Article in English | MEDLINE | ID: covidwho-777664

ABSTRACT

Severe acute respiratory syndrome coronavirus-2 causes the clinical syndrome of coronavirus disease of 2019 (COVID-19) which has become a global pandemic resulting in significant morbidity and mortality. While the virus primarily affects the respiratory system, it also causes a wide variety of complex cardiac manifestations such as acute myopericarditis, acute coronary syndrome, congested heart failure, cardiogenic shock and cardiac arrhythmias. There are numerous proposed mechanisms of cardiac injury, including direct cellular injury, pro-inflammatory cytokine storm, myocardial oxygen-demand mismatch, and systemic inflammation causing multi-organ failure. Additionally, medications commonly used to treat COVID-19 patients have various cardiovascular side effects. We aim to provide a succinct review about the pathophysiology and cardiac manifestations of COVID-19, as well as treatment considerations and the various adaptations made to the current healthcare structure as a result of the pandemic.


Subject(s)
Acute Coronary Syndrome/therapy , Arrhythmias, Cardiac/therapy , COVID-19/therapy , Heart Failure/therapy , Pandemics , Pericarditis/therapy , Shock, Cardiogenic/therapy , Acute Coronary Syndrome/epidemiology , Acute Coronary Syndrome/pathology , Acute Coronary Syndrome/virology , Antiviral Agents/administration & dosage , Antiviral Agents/adverse effects , Arrhythmias, Cardiac/epidemiology , Arrhythmias, Cardiac/pathology , Arrhythmias, Cardiac/virology , Biomarkers/analysis , COVID-19/epidemiology , COVID-19/pathology , COVID-19/virology , Cardiac Catheterization/methods , Comorbidity , Disease Management , Glucocorticoids/administration & dosage , Glucocorticoids/adverse effects , Heart Failure/epidemiology , Heart Failure/pathology , Heart Failure/virology , Hospitalization , Humans , Immunologic Factors/administration & dosage , Immunologic Factors/adverse effects , Pericarditis/epidemiology , Pericarditis/pathology , Pericarditis/virology , Risk Factors , SARS-CoV-2/pathogenicity , Severity of Illness Index , Shock, Cardiogenic/epidemiology , Shock, Cardiogenic/pathology , Shock, Cardiogenic/virology , Texas/epidemiology
SELECTION OF CITATIONS
SEARCH DETAIL