Your browser doesn't support javascript.
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 84
Filtrar
1.
Endocrine ; 80(3): 477-490, 2023 Jun.
Artículo en Inglés | MEDLINE | ID: covidwho-2291939

RESUMEN

Evidence has shown that cardiometabolic disorders (CMDs) are amongst the top contributors to COVID-19 infection morbidity and mortality. The reciprocal impact of COVID-19 infection and the most common CMDs, the risk factors for poor composite outcome among patients with one or several underlying diseases, the effect of common medical management on CMDs and their safety in the context of acute COVID-19 infection are reviewed. Later on, the changes brought by the COVID-19 pandemic quarantine on the general population's lifestyle (diet, exercise patterns) and metabolic health, acute cardiac complications of different COVID-19 vaccines and the effect of CMDs on the vaccine efficacy are discussed. Our review identified that the incidence of COVID-19 infection is higher among patients with underlying CMDs such as hypertension, diabetes, obesity and cardiovascular disease. Also, CMDs increase the risk of COVID-19 infection progression to severe disease phenotypes (e.g. hospital and/or ICU admission, use of mechanical ventilation). Lifestyle modification during COVID-19 era had a great impact on inducing and worsening of CMDs. Finally, the lower efficacy of COVID-19 vaccines was found in patients with metabolic disease.


Asunto(s)
COVID-19 , Enfermedades Cardiovasculares , Humanos , COVID-19/epidemiología , Vacunas contra la COVID-19 , Pandemias , Factores de Riesgo , Obesidad/complicaciones , Obesidad/epidemiología , Enfermedades Cardiovasculares/epidemiología , Enfermedades Cardiovasculares/metabolismo
2.
Arterioscler Thromb Vasc Biol ; 43(5): 628-636, 2023 05.
Artículo en Inglés | MEDLINE | ID: covidwho-2266992

RESUMEN

Recent studies have demonstrated a novel function of red blood cells (RBCs) beyond their classical role as gas transporters, that is, RBCs undergo functional alterations in cardiovascular and metabolic disease, and RBC dysfunction is associated with hypertension and the development of cardiovascular injury in type 2 diabetes, heart failure, preeclampsia, familial hypercholesterolemia/dyslipidemia, and COVID-19. The underlying mechanisms include decreased nitric oxide bioavailability, increased arginase activity, and reactive oxygen species formation. Of interest, RBCs contain diverse and abundant micro (mi)RNAs. miRNA expression pattern in RBCs reflects the expression in the whole blood, serum, and plasma. miRNA levels in RBCs have been found to be altered in various cardiovascular and metabolic diseases, which contributes to the development of cardiovascular complications. Evidence has shown that RBC-derived miRNAs interact with the cardiovascular system via extracellular vesicles and argonaute RISC catalytic component 2 as carriers. Alteration of RBC-to-vascular communication via miRNAs may serve as potential disease mechanism for vascular complications. The present review summarizes RBCs and their released miRNAs as potential mediators of cardiovascular injury. We further focus on the possible mechanisms by which RBC-derived miRNAs regulate cardiovascular function. A better understanding of the function of RBC-derived miRNAs will increase insights into the disease mechanism and potential targets for the treatment of cardiovascular complications.


Asunto(s)
COVID-19 , Enfermedades Cardiovasculares , Diabetes Mellitus Tipo 2 , MicroARNs , Femenino , Embarazo , Humanos , MicroARNs/genética , MicroARNs/metabolismo , Diabetes Mellitus Tipo 2/metabolismo , COVID-19/metabolismo , Eritrocitos/metabolismo , Corazón , Enfermedades Cardiovasculares/genética , Enfermedades Cardiovasculares/metabolismo
3.
Int J Mol Sci ; 24(3)2023 Jan 19.
Artículo en Inglés | MEDLINE | ID: covidwho-2243576

RESUMEN

Cardiovascular and renal diseases are among the leading causes of death worldwide, and regardless of current efforts, there is a demanding need for therapeutic alternatives to reduce their progression to advanced stages. The stress caused by diseases leads to the activation of protective mechanisms in the cell, including chaperone proteins. The Sigma-1 receptor (Sig-1R) is a ligand-operated chaperone protein that modulates signal transduction during cellular stress processes. Sig-1R interacts with various ligands and proteins to elicit distinct cellular responses, thus, making it a potential target for pharmacological modulation. Furthermore, Sig-1R ligands activate signaling pathways that promote cardioprotection, ameliorate ischemic injury, and drive myofibroblast activation and fibrosis. The role of Sig-1R in diseases has also made it a point of interest in developing clinical trials for pain, neurodegeneration, ischemic stroke, depression in patients with heart failure, and COVID-19. Sig-1R ligands in preclinical models have significantly beneficial effects associated with improved cardiac function, ventricular remodeling, hypertrophy reduction, and, in the kidney, reduced ischemic damage. These basic discoveries could inform clinical trials for heart failure (HF), myocardial hypertrophy, acute kidney injury (AKI), and chronic kidney disease (CKD). Here, we review Sig-1R signaling pathways and the evidence of Sig-1R modulation in preclinical cardiac and renal injury models to support the potential therapeutic use of Sig-1R agonists and antagonists in these diseases.


Asunto(s)
Enfermedades Cardiovasculares , Enfermedades Renales , Receptores sigma , Humanos , Cardiomegalia , COVID-19/complicaciones , Insuficiencia Cardíaca/complicaciones , Ligandos , Receptores sigma/agonistas , Receptores sigma/antagonistas & inhibidores , Receptores sigma/genética , Receptores sigma/metabolismo , Transducción de Señal/fisiología , Enfermedades Cardiovasculares/complicaciones , Enfermedades Cardiovasculares/genética , Enfermedades Cardiovasculares/metabolismo , Enfermedades Renales/complicaciones , Enfermedades Renales/genética , Enfermedades Renales/metabolismo
4.
Curr Hypertens Rep ; 24(12): 627-637, 2022 Dec.
Artículo en Inglés | MEDLINE | ID: covidwho-2230226

RESUMEN

PURPOSE OF REVIEW: In this review, we focus on immune cell activation in obesity and cardiovascular disease, highlighting specific immune cell microenvironments present in individuals with atherosclerosis, non-ischemic heart disease, hypertension, and infectious diseases. RECENT FINDINGS: Obesity and cardiovascular disease are intimately linked and often characterized by inflammation and a cluster of metabolic complications. Compelling evidence from single-cell analysis suggests that obese adipose tissue is inflammatory and infiltrated by almost all immune cell populations. How this inflammatory tissue state contributes to more systemic conditions such as cardiovascular and infectious disease is less well understood. However, current research suggests that changes in the adipose tissue immune environment impact an individual's ability to combat illnesses such as influenza and SARS-CoV2. Obesity is becoming increasingly prevalent globally and is often associated with type 2 diabetes and heart disease. An increased inflammatory state is a major contributor to this association. Widespread chronic inflammation in these disease states is accompanied by an increase in both innate and adaptive immune cell activation. Acutely, these immune cell changes are beneficial as they sustain homeostasis as inflammation increases. However, persistent inflammation subsequently damages tissues and organs throughout the body. Future studies aimed at understanding the unique immune cell populations in each tissue compartment impacted by obesity may hold potential for therapeutic applications.


Asunto(s)
COVID-19 , Enfermedades Cardiovasculares , Diabetes Mellitus Tipo 2 , Cardiopatías , Hipertensión , Humanos , Enfermedades Cardiovasculares/metabolismo , Diabetes Mellitus Tipo 2/complicaciones , ARN Viral/metabolismo , Hipertensión/complicaciones , SARS-CoV-2 , Obesidad/complicaciones , Obesidad/metabolismo , Tejido Adiposo/metabolismo , Inflamación , Cardiopatías/metabolismo
6.
Dokl Biol Sci ; 505(1): 95-99, 2022 Aug.
Artículo en Inglés | MEDLINE | ID: covidwho-2008770

RESUMEN

The article focuses on the pathogenetic mechanisms of posttraumatic stress disorder (PTSD), which is associated with psychological stress because of the coronavirus pandemic. The molecular mechanisms responsible for disease susceptibility in some individuals and stress resistance in others are amongst crucial research interests of experimental and clinical medicine. Priority data were obtained to indicate that distortions of synthesis and metabolism and, most significantly, a switch between two energy transport forms, glucose and lipids, underlie myocardial dysfunction in young and old stress-sensitive Wistar rats in a PTSD model. Histochemistry and polarization microscopy showed energy deficit in cardiomyocytes and signs of ischemic and hypoxic areas emerging in the myocardium as a result of an accumulation of NADH and NADPH, which initiate excessive production of reactive oxygen species.


Asunto(s)
Enfermedades Cardiovasculares , Trastornos por Estrés Postraumático , Animales , Enfermedades Cardiovasculares/complicaciones , Enfermedades Cardiovasculares/metabolismo , Enfermedades Cardiovasculares/patología , Miocardio/patología , Ratas , Ratas Wistar , Factores de Riesgo
7.
Biochem Biophys Res Commun ; 626: 66-71, 2022 10 20.
Artículo en Inglés | MEDLINE | ID: covidwho-1966379

RESUMEN

Increasing evidence suggests incomplete recovery of COVID-19 patients, who continue to suffer from cardiovascular diseases, including cerebral vascular disorders (CVD) and neurological symptoms. Recent findings indicate that some of the damaging effects of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection, especially in the brain, may be induced by the spike protein, leading to the disruption of the initial blood-brain barrier (BBB). SARS-CoV-2-infected cells and animals exhibit age-dependent pathogenesis. In this study, we identified endothelial BACE1 as a critical mediator of BBB disruption and cellular senescence induced by the SARS-CoV-2 spike S1 subunit protein. Increased BACE1 in human brain microvascular endothelial cells (HBMVEC) decreases the levels of tight junction proteins, including ZO-1, occludin, and claudins. Moreover, BACE1 overexpression leads to the accumulation of p16 and p21, typical hallmarks of cellular senescence. Our findings show that the SARS-CoV-2 spike S1 subunit protein upregulated BACE1 expression in HBMVECs, causing endothelial leakage. In addition, the SARS-CoV-2 spike S1 subunit protein induced p16 and p21 expression, indicating BACE1-mediated cellular senescence, confirmed by ß-Gal staining in HBMVECs. In conclusion, this study demonstrated that BACE1-mediated endothelial cell damage and senescence may be linked to CVD after COVID-19 infection.


Asunto(s)
COVID-19 , Enfermedades Cardiovasculares , Secretasas de la Proteína Precursora del Amiloide/metabolismo , Animales , Ácido Aspártico Endopeptidasas/metabolismo , Encéfalo/metabolismo , Enfermedades Cardiovasculares/metabolismo , Células Endoteliales/metabolismo , Humanos , SARS-CoV-2 , Glicoproteína de la Espiga del Coronavirus/metabolismo
8.
Pharmacol Rev ; 74(3): 462-505, 2022 07.
Artículo en Inglés | MEDLINE | ID: covidwho-1901904

RESUMEN

The concept of local formation of angiotensin II in the kidney has changed over the last 10-15 years. Local synthesis of angiotensinogen in the proximal tubule has been proposed, combined with prorenin synthesis in the collecting duct. Binding of prorenin via the so-called (pro)renin receptor has been introduced, as well as megalin-mediated uptake of filtered plasma-derived renin-angiotensin system (RAS) components. Moreover, angiotensin metabolites other than angiotensin II [notably angiotensin-(1-7)] exist, and angiotensins exert their effects via three different receptors, of which angiotensin II type 2 and Mas receptors are considered renoprotective, possibly in a sex-specific manner, whereas angiotensin II type 1 (AT1) receptors are believed to be deleterious. Additionally, internalized angiotensin II may stimulate intracellular receptors. Angiotensin-converting enzyme 2 (ACE2) not only generates angiotensin-(1-7) but also acts as coronavirus receptor. Multiple, if not all, cardiovascular diseases involve the kidney RAS, with renal AT1 receptors often being claimed to exert a crucial role. Urinary RAS component levels, depending on filtration, reabsorption, and local release, are believed to reflect renal RAS activity. Finally, both existing drugs (RAS inhibitors, cyclooxygenase inhibitors) and novel drugs (angiotensin receptor/neprilysin inhibitors, sodium-glucose cotransporter-2 inhibitors, soluble ACE2) affect renal angiotensin formation, thereby displaying cardiovascular efficacy. Particular in the case of the latter three, an important question is to what degree they induce renoprotection (e.g., in a renal RAS-dependent manner). This review provides a unifying view, explaining not only how kidney angiotensin formation occurs and how it is affected by drugs but also why drugs are renoprotective when altering the renal RAS. SIGNIFICANCE STATEMENT: Angiotensin formation in the kidney is widely accepted but little understood, and multiple, often contrasting concepts have been put forward over the last two decades. This paper offers a unifying view, simultaneously explaining how existing and novel drugs exert renoprotection by interfering with kidney angiotensin formation.


Asunto(s)
Enfermedades Cardiovasculares , Diabetes Mellitus Tipo 2 , Inhibidores del Cotransportador de Sodio-Glucosa 2 , Angiotensina II/metabolismo , Enzima Convertidora de Angiotensina 2 , Angiotensinógeno/metabolismo , Enfermedades Cardiovasculares/metabolismo , Diabetes Mellitus Tipo 2/metabolismo , Sistemas de Liberación de Medicamentos , Femenino , Humanos , Riñón/irrigación sanguínea , Riñón/metabolismo , Masculino , Renina/metabolismo , Sistema Renina-Angiotensina , Inhibidores del Cotransportador de Sodio-Glucosa 2/metabolismo
9.
WIREs Mech Dis ; 14(5): e1560, 2022 09.
Artículo en Inglés | MEDLINE | ID: covidwho-1898962

RESUMEN

We review the current understanding of formation and development of the coronary microvasculature which supplies oxygen and nutrients to the heart myocardium and removes waste. We emphasize the close relationship, mutual development, and communication between microvasculature endothelial cells and surrounding cardiomyocytes. The first part of the review is focused on formation of microvasculature during embryonic development. We summarize knowledge about establishing the heart microvasculature density based on diffusion distance. Then signaling mechanisms which are involved in forming the microvasculature are discussed. This includes details of cardiomyocyte-endothelial cell interactions involving hypoxia, VEGF, NOTCH, angiopoietin, PDGF, and other signaling factors. The microvasculature is understudied due to difficulties in its visualization. Therefore, currently available imaging methods to delineate the coronary microvasculature in development and in adults are discussed. The second part of the review is dedicated to the importance of the coronary vasculature in disease. Coronary microvasculature pathologies are present in many congenital heart diseases (CHD), especially in pulmonary atresia, and worsen outcomes. In CHDs, where the development of the myocardium is impaired, microvasculature is also affected. In adult patients coronary microvascular disease is one of the main causes of sudden cardiac death, especially in women. Coronary microvasculature pathologies affect myocardial ischemia and vice versa; myocardial pathologies such as cardiomyopathies are closely connected with coronary microvasculature dysfunction. Microvasculature inflammation also worsens the outcomes of COVID-19 disease. Our review stresses the importance of coronary microvasculature and provides an overview of its formation and signaling mechanisms and the importance of coronary vasculature pathologies in CHDs and adult diseases. This article is categorized under: Cardiovascular Diseases > Stem Cells and Development Congenital Diseases > Molecular and Cellular Physiology Cardiovascular Diseases > Molecular and Cellular Physiology.


Asunto(s)
Enfermedades Cardiovasculares , Miocardio , Miocitos Cardíacos , Adulto , COVID-19 , Enfermedades Cardiovasculares/metabolismo , Células Endoteliales , Femenino , Humanos , Microvasos/diagnóstico por imagen , Miocardio/metabolismo , Miocitos Cardíacos/metabolismo
10.
Int J Mol Sci ; 23(3)2022 Jan 18.
Artículo en Inglés | MEDLINE | ID: covidwho-1887210

RESUMEN

The propensity towards platelet-rich thrombus formation increases substantially during normal ageing, and this trend is mediated by decreases in platelet responsiveness to the anti-aggregatory nitric oxide (NO) and prostacyclin (PGI2) pathways. The impairment of soluble guanylate cyclase and adenylate cyclase-based signalling that is associated with oxidative stress represents the major mechanism of this loss of anti-aggregatory reactivity. Platelet desensitization to these autacoids represents an adverse prognostic marker in patients with ischemic heart disease and may contribute to increased thrombo-embolic risk in patients with heart failure. Patients with platelet resistance to PGI2 also are unresponsive to ADP receptor antagonist therapy. Apart from ischemia, diabetes and aortic valve disease are also associated with impaired anti-aggregatory homeostasis. This review examines the association of impaired platelet cyclic nucleotide (i.e., cGMP and cAMP) signalling with the emerging evidence of thromboembolic risk in cardiovascular diseases, and discusses the potential therapeutic strategies targeting this abnormality.


Asunto(s)
Enfermedades Cardiovasculares/complicaciones , Epoprostenol/metabolismo , Óxido Nítrico/metabolismo , Tromboembolia/metabolismo , Adenilil Ciclasas/metabolismo , Enfermedades Cardiovasculares/metabolismo , Resistencia a Medicamentos , Humanos , Estrés Oxidativo , Transducción de Señal , Guanilil Ciclasa Soluble/metabolismo , Tromboembolia/etiología
12.
Cells ; 11(8)2022 04 13.
Artículo en Inglés | MEDLINE | ID: covidwho-1785544

RESUMEN

Cardiovascular disease (CVD) is the leading cause of death worldwide. Current data suggest that patients with cardiovascular diseases experience more serious complications with coronavirus disease-19 (COVID-19) than those without CVD. In addition, severe COVID-19 appears to cause acute cardiac injury, as well as long-term adverse remodeling of heart tissue. Cardiac fibroblasts and myofibroblasts, being crucial in response to injury, may play a pivotal role in both contributing to and healing COVID-19-induced cardiac injury. The role of cardiac myofibroblasts in cardiac fibrosis has been well-established in the literature for decades. However, with the emergence of the novel coronavirus SARS-CoV-2, new cardiac complications are arising. Bursts of inflammatory cytokines and upregulation of TGF-ß1 and angiotensin (AngII) are common in severe COVID-19 patients. Cytokines, TGF-ß1, and Ang II can induce cardiac fibroblast differentiation, potentially leading to fibrosis. This review details the key information concerning the role of cardiac myofibroblasts in CVD and COVID-19 complications. Additionally, new factors including controlling ACE2 expression and microRNA regulation are explored as promising treatments for both COVID-19 and CVD. Further understanding of this topic may provide insight into the long-term cardiac manifestations of the COVID-19 pandemic and ways to mitigate its negative effects.


Asunto(s)
COVID-19 , Enfermedades Cardiovasculares , COVID-19/complicaciones , Enfermedades Cardiovasculares/metabolismo , Fibroblastos/metabolismo , Humanos , Miocardio/metabolismo , Miofibroblastos/metabolismo , Pandemias , SARS-CoV-2 , Factor de Crecimiento Transformador beta1/metabolismo
13.
Mol Omics ; 18(5): 408-416, 2022 06 13.
Artículo en Inglés | MEDLINE | ID: covidwho-1740493

RESUMEN

A predominant source of complication in SARS-CoV-2 patients arises from a severe systemic inflammation that can lead to tissue damage and organ failure. The high inflammatory burden of this viral infection often results in cardiovascular comorbidities. A better understanding of the interaction between immune pathways and cardiovascular proteins might inform medical decisions and therapeutic approaches. In this study we hypothesized that helper T-cell inflammatory pathways (Th1, Th2 and Th17) synergistically correlate with cardiometabolic proteins in serum of COVID-19 patients. We found that Th1, Th2 and Th17 cytokines and chemokines are able to predict expression of 186 cardiometabolic proteins profiled by Olink proteomics.


Asunto(s)
COVID-19 , Enfermedades Cardiovasculares , Enfermedades Cardiovasculares/metabolismo , Humanos , Proteómica , SARS-CoV-2 , Células TH1/metabolismo , Células Th17/metabolismo , Células Th2/metabolismo
14.
Am J Physiol Cell Physiol ; 322(2): C218-C230, 2022 02 01.
Artículo en Inglés | MEDLINE | ID: covidwho-1673516

RESUMEN

Selective autophagy of mitochondria, known as mitophagy, is a major quality control pathway in the heart that is involved in removing unwanted or dysfunctional mitochondria from the cell. Baseline mitophagy is critical for maintaining fitness of the mitochondrial network by continuous turnover of aged and less-functional mitochondria. Mitophagy is also critical in adapting to stress associated with mitochondrial damage or dysfunction. The removal of damaged mitochondria prevents reactive oxygen species-mediated damage to proteins and DNA and suppresses activation of inflammation and cell death. Impairments in mitophagy are associated with the pathogenesis of many diseases, including cancers, inflammatory diseases, neurodegeneration, and cardiovascular disease. Mitophagy is a highly regulated and complex process that requires the coordination of labeling dysfunctional mitochondria for degradation while simultaneously promoting de novo autophagosome biogenesis adjacent to the cargo. In this review, we provide an update on our current understanding of these steps in mitophagy induction and discuss the physiological and pathophysiological consequences of altered mitophagy in the heart.


Asunto(s)
COVID-19/metabolismo , Enfermedades Cardiovasculares/metabolismo , Sistema Cardiovascular/metabolismo , Mitocondrias/metabolismo , Mitofagia/fisiología , Especies Reactivas de Oxígeno/metabolismo , Animales , COVID-19/patología , Enfermedades Cardiovasculares/patología , Sistema Cardiovascular/patología , Humanos , Mitocondrias/patología , Fagocitosis/fisiología
15.
Ann N Y Acad Sci ; 1507(1): 70-83, 2022 01.
Artículo en Inglés | MEDLINE | ID: covidwho-1673249

RESUMEN

For many years, it was believed that the aging process was inevitable and that age-related diseases could not be prevented or reversed. The geroscience hypothesis, however, posits that aging is, in fact, malleable and, by targeting the hallmarks of biological aging, it is indeed possible to alleviate age-related diseases and dysfunction and extend longevity. This field of geroscience thus aims to prevent the development of multiple disorders with age, thereby extending healthspan, with the reduction of morbidity toward the end of life. Experts in the field have made remarkable advancements in understanding the mechanisms underlying biological aging and identified ways to target aging pathways using both novel agents and repurposed therapies. While geroscience researchers currently face significant barriers in bringing therapies through clinical development, proof-of-concept studies, as well as early-stage clinical trials, are underway to assess the feasibility of drug evaluation and lay a regulatory foundation for future FDA approvals in the future.


Asunto(s)
Envejecimiento/genética , Envejecimiento/metabolismo , Congresos como Asunto/tendencias , Gerociencia/tendencias , Longevidad/fisiología , Informe de Investigación , Autofagia/fisiología , COVID-19/genética , COVID-19/metabolismo , COVID-19/mortalidad , Enfermedades Cardiovasculares/genética , Enfermedades Cardiovasculares/metabolismo , Enfermedades Cardiovasculares/terapia , Gerociencia/métodos , Humanos , Metabolómica/métodos , Metabolómica/tendencias , Enfermedades del Sistema Nervioso/genética , Enfermedades del Sistema Nervioso/metabolismo , Enfermedades del Sistema Nervioso/terapia , Trasplante de Células Madre/métodos , Trasplante de Células Madre/tendencias
16.
Int J Mol Sci ; 22(16)2021 Aug 23.
Artículo en Inglés | MEDLINE | ID: covidwho-1662696

RESUMEN

Magnesium (Mg) is a pivotal and very complex component of healthy aging in the cardiovascular-muscle-bone triad. Low Mg levels and low Mg intake are common in the general aging population and are associated with poorer outcomes than higher levels, including vascular calcification, endothelial dysfunction, osteoporosis, or muscle dysfunction/sarcopenia. While Mg supplementation appears to reverse these processes and benefit the triad, more randomized clinical trials are needed. These will allow improvement of preventive and curative strategies and propose guidelines regarding the pharmaceutical forms and the dosages and durations of treatment in order to optimize and adapt Mg prescription for healthy aging and for older vulnerable persons with comorbidities.


Asunto(s)
Enfermedades Cardiovasculares/metabolismo , Magnesio/metabolismo , Osteoporosis/metabolismo , Sarcopenia/metabolismo , Envejecimiento/metabolismo , Animales , Huesos/metabolismo , Envejecimiento Saludable/metabolismo , Humanos , Fuerza Muscular/fisiología , Músculo Esquelético/metabolismo
17.
Int J Mol Sci ; 23(2)2022 Jan 15.
Artículo en Inglés | MEDLINE | ID: covidwho-1631216

RESUMEN

Angiotensin II receptor type 1 (AT1R) and endothelin-1 receptor type A (ETAR) are G-protein-coupled receptors (GPCRs) expressed on the surface of a great variety of cells: immune cells, vascular smooth cells, endothelial cells, and fibroblasts express ETAR and AT1R, which are activated by endothelin 1 (ET1) and angiotensin II (AngII), respectively. Certain autoantibodies are specific for these receptors and can regulate their function, thus being known as functional autoantibodies. The function of these antibodies is similar to that of natural ligands, and it involves not only vasoconstriction, but also the secretion of proinflammatory cytokines (such as interleukin-6 (IL6), IL8 and TNF-α), collagen production by fibroblasts, and reactive oxygen species (ROS) release by fibroblasts and neutrophils. The role of autoantibodies against AT1R and ETAR (AT1R-AAs and ETAR-AAs, respectively) is well described in the pathogenesis of many medical conditions (e.g., systemic sclerosis (SSc) and SSc-associated pulmonary hypertension, cystic fibrosis, and allograft dysfunction), but their implications in cardiovascular diseases are still unclear. This review summarizes the current evidence regarding the effects of AT1R-AAs and ETAR-AAs in cardiovascular pathologies, highlighting their roles in heart transplantation and mechanical circulatory support, preeclampsia, and acute coronary syndromes.


Asunto(s)
Autoanticuerpos/metabolismo , Enfermedades Cardiovasculares/inmunología , Receptor de Angiotensina Tipo 1/inmunología , Receptor de Endotelina A/inmunología , Enfermedades Cardiovasculares/metabolismo , Enfermedades Cardiovasculares/patología , Colágeno/metabolismo , Humanos , Interleucina-6/metabolismo , Interleucina-8/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Receptor de Angiotensina Tipo 1/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo
18.
Bull Exp Biol Med ; 172(3): 283-287, 2022 Jan.
Artículo en Inglés | MEDLINE | ID: covidwho-1611428

RESUMEN

We studied laboratory parameters of patients with COVID-19 against the background of chronic pathologies (cardiovascular pathologies, obesity, type 2 diabetes melitus, and cardiovascular pathologies with allergy to statins). A decrease in pH and a shift in the electrolyte balance of blood plasma were revealed in all studied groups and were most pronounced in patients with cardiovascular pathologies with allergy to statin. It was found that low pH promotes destruction of lipid components of the erythrocyte membranes in patients with chronic pathologies, which was seen from a decrease in Na+/K+-ATPase activity and significant hyponatrenemia. In patients with cardiovascular pathologies and allergy to statins, erythrocyte membranes were most sensitive to a decrease in pH, while erythrocyte membranes of obese patients showed the greatest resistance to low pH and oxidative stress.


Asunto(s)
COVID-19/complicaciones , Hiponatremia/etiología , Hipoxia/complicaciones , ATPasa Intercambiadora de Sodio-Potasio/fisiología , Anciano , COVID-19/metabolismo , Enfermedades Cardiovasculares/complicaciones , Enfermedades Cardiovasculares/metabolismo , Enfermedades Cardiovasculares/virología , Estudios de Casos y Controles , Enfermedad Crónica , Diabetes Mellitus Tipo 2/complicaciones , Diabetes Mellitus Tipo 2/metabolismo , Diabetes Mellitus Tipo 2/virología , Hipersensibilidad a las Drogas/complicaciones , Hipersensibilidad a las Drogas/metabolismo , Hipersensibilidad a las Drogas/virología , Membrana Eritrocítica/metabolismo , Eritrocitos/metabolismo , Femenino , Transferencias de Fluidos Corporales/fisiología , Humanos , Concentración de Iones de Hidrógeno , Inhibidores de Hidroximetilglutaril-CoA Reductasas/efectos adversos , Hiponatremia/metabolismo , Hiponatremia/virología , Hipoxia/metabolismo , Peroxidación de Lípido/fisiología , Masculino , Persona de Mediana Edad , Obesidad/complicaciones , Obesidad/metabolismo , Obesidad/virología , Estrés Oxidativo/fisiología , SARS-CoV-2/fisiología , Sodio/metabolismo , Estrés Fisiológico/fisiología
19.
Biomolecules ; 12(1)2021 12 24.
Artículo en Inglés | MEDLINE | ID: covidwho-1581037

RESUMEN

Coronavirus disease 2019 (COVID-19), the pandemic infection caused by the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), presents with an extremely heterogeneous spectrum of symptoms and signs. The clinical manifestations seem to be correlated with disease severity. COVID-19 susceptibility and mortality show a significant sex imbalance, with men being more prone to infection and showing a higher rate of hospitalization and mortality compared to women. Such variability can be ascribed to both sex-related biological factors and gender-related behavioral cues. This review will discuss the potential mechanisms accounting for sex/gender influence in vulnerability to COVID-19. Cardiovascular diseases play a central role in determining COVID-19 outcome, whether they are pre-existent or arose upon infection. We will pay particular attention to the impact of sex and gender on cardiovascular manifestations related to COVID-19. Finally, we will discuss the sex-dependent variability in some biomarkers for the evaluation of COVID-19 infection and prognosis. The aim of this work is to highlight the significance of gendered medicine in setting up personalized programs for COVID-19 prevention, clinical evaluation and treatment.


Asunto(s)
COVID-19 , Enfermedades Cardiovasculares , Pandemias , SARS-CoV-2/metabolismo , Caracteres Sexuales , COVID-19/complicaciones , COVID-19/epidemiología , COVID-19/metabolismo , Enfermedades Cardiovasculares/epidemiología , Enfermedades Cardiovasculares/etiología , Enfermedades Cardiovasculares/metabolismo , Femenino , Humanos , Masculino , Factores de Riesgo , Índice de Severidad de la Enfermedad , Factores Sexuales
20.
Int J Mol Sci ; 22(24)2021 Dec 17.
Artículo en Inglés | MEDLINE | ID: covidwho-1580691

RESUMEN

Although blood-heart-barrier (BHB) leakage is the hallmark of congestive (cardio-pulmonary) heart failure (CHF), the primary cause of death in elderly, and during viral myocarditis resulting from the novel coronavirus variants such as the severe acute respiratory syndrome novel corona virus 2 (SARS-CoV-2) known as COVID-19, the mechanism is unclear. The goal of this project is to determine the mechanism of the BHB in CHF. Endocardial endothelium (EE) is the BHB against leakage of blood from endocardium to the interstitium; however, this BHB is broken during CHF. Previous studies from our laboratory, and others have shown a robust activation of matrix metalloproteinase-9 (MMP-9) during CHF. MMP-9 degrades the connexins leading to EE dysfunction. We demonstrated juxtacrine coupling of EE with myocyte and mitochondria (Mito) but how it works still remains at large. To test whether activation of MMP-9 causes EE barrier dysfunction, we hypothesized that if that were the case then treatment with hydroxychloroquine (HCQ) could, in fact, inhibit MMP-9, and thus preserve the EE barrier/juxtacrine signaling, and synchronous endothelial-myocyte coupling. To determine this, CHF was created by aorta-vena cava fistula (AVF) employing the mouse as a model system. The sham, and AVF mice were treated with HCQ. Cardiac hypertrophy, tissue remodeling-induced mitochondrial-myocyte, and endothelial-myocyte contractions were measured. Microvascular leakage was measured using FITC-albumin conjugate. The cardiac function was measured by echocardiography (Echo). Results suggest that MMP-9 activation, endocardial endothelial leakage, endothelial-myocyte (E-M) uncoupling, dyssynchronous mitochondrial fusion-fission (Mfn2/Drp1 ratio), and mito-myocyte uncoupling in the AVF heart failure were found to be rampant; however, treatment with HCQ successfully mitigated some of the deleterious cardiac alterations during CHF. The findings have direct relevance to the gamut of cardiac manifestations, and the resultant phenotypes arising from the ongoing complications of COVID-19 in human subjects.


Asunto(s)
COVID-19/complicaciones , Insuficiencia Cardíaca/metabolismo , Corazón/virología , Animales , Sangre/virología , Fenómenos Fisiológicos Sanguíneos/inmunología , COVID-19/fisiopatología , Cardiomegalia/metabolismo , Enfermedades Cardiovasculares/metabolismo , Fenómenos Fisiológicos Cardiovasculares/inmunología , Modelos Animales de Enfermedad , Endotelio/metabolismo , Corazón/fisiopatología , Insuficiencia Cardíaca/virología , Hidroxicloroquina/farmacología , Masculino , Metaloproteinasa 9 de la Matriz/efectos de los fármacos , Metaloproteinasa 9 de la Matriz/metabolismo , Ratones , Ratones Endogámicos C57BL , Células Musculares/metabolismo , Miocardio/metabolismo , SARS-CoV-2/metabolismo , SARS-CoV-2/patogenicidad , Remodelación Ventricular/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA