Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 39
Filtrar
Mais filtros

Base de dados
País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
Artigo em Inglês | MEDLINE | ID: mdl-38781602

RESUMO

Oral hormonal contraception (OHC) is a widely employed method in females for the prevention of unintended pregnancies, as well as for the treatment of menstrual disorders, endometriosis, and polycystic ovarian syndrome. However, it is believed that with OHCs use, some females may have higher risk of cardiovascular diseases, such as hypertension, diabetes, myocardial infarction, thrombosis, and heart failure. Although such risks are infrequently detected in healthy young females with the use of oral contraceptives, slightly elevated risks of cardiovascular diseases have been observed among reproductive-aged healthy females. However, prolonged use of OHC has also been claimed to have protective cardiac effects and may contribute to reduced risk of cardiovascular disease. In fact, the debate on whether OHC administration increases the risk of cardiovascular diseases has been ongoing with inconsistent and controversial viewpoints. Nevertheless, a great deal of work has been carried out to understand the relationship between OHC use and the occurrence of cardiovascular risk in females who use OHC for preventing the unwanted pregnancy or treatment of other disorders. Therefore, in this review we summarize the most recent available evidence regarding the association between the use of oral hormonal contraceptives and the risk for cardiovascular disease in females who are using OHC to prevent unintended pregnancy.

2.
Artigo em Inglês | MEDLINE | ID: mdl-38781601

RESUMO

Despite major advances in cardiac research over the past three decades, cardiovascular disease (CVD) still remains the leading cause of morbidity and mortality in women and men worldwide. However, a major challenge for health care providers is that the current guidelines for cardiovascular drug therapies do not consider the impact of sex in the development of treatment plan for optimizing therapies for women. Clinical research in recent years suggests significant pharmacological and pharmacokinetic differences between females and males which have been attributed in part to differences in body composition, plasma protein binding capacity, drug metabolism and excretion. Herein, we provide a comprehensive review regarding sex- specific differences and drugs commonly used for cardiovascular diseases in women and men. Understanding how sex related differences influence drug efficacy and cardiovascular disease outcomes is crucial for not only optimizing treatment strategies for women and men but to encourage the implementation of specific guidelines that address sex difference as a consideration for treatment of cardiovascular diseases.

3.
Artigo em Inglês | MEDLINE | ID: mdl-38728749

RESUMO

Retinoic acid-related orphan receptors (RORs) serve as transcription factors that play a pivotal role in a myriad of physiological processes within the body. Their involvement extends to critical biological processes that confer protective effects in the heart, immune system, and nervous system, as well as contributing to the mitigation of several aggressive cancer types. These protective functions are attributed to ROR's regulation of key proteins and the management of various cellular processes, including autophagy, mitophagy, inflammation, oxidative stress, and glucose metabolism, highlighting the emerging need for pharmacological approaches to modulate ROR expression. Thus, the modulation of RORs is a rapidly growing area of research aimed not only at comprehending these receptors, but also at manipulating them to attain the desired physiological response. Despite the presence of natural ROR ligands, the development of synthetic agonists with high selectivity for these receptors holds substantial therapeutic potential. The exploration and advancement of such compounds can effectively target diseases associated with ROR dysregulation, thereby providing avenues for therapeutic interventions. Herein, we provide a comprehensive examination of the multifaceted role of ROR in diverse physiological and pathophysiological conditions, accompanied by an in-depth exploration of a spectrum of ROR agonists, inverse agonists, and antagonists.

4.
Circulation ; 146(12): 934-954, 2022 09 20.
Artigo em Inglês | MEDLINE | ID: mdl-35983756

RESUMO

BACKGROUND: Cytokines such as tumor necrosis factor-α (TNFα) have been implicated in cardiac dysfunction and toxicity associated with doxorubicin (DOX). Although TNFα can elicit different cellular responses, including survival or death, the mechanisms underlying these divergent outcomes in the heart remain cryptic. The E3 ubiquitin ligase TRAF2 (TNF receptor associated factor 2) provides a critical signaling platform for K63-linked polyubiquitination of RIPK1 (receptor interacting protein 1), crucial for nuclear factor-κB (NF-κB) activation by TNFα and survival. Here, we investigate alterations in TNFα-TRAF2-NF-κB signaling in the pathogenesis of DOX cardiotoxicity. METHODS: Using a combination of in vivo (4 weekly injections of DOX 5 mg·kg-1·wk-1) in C57/BL6J mice and in vitro approaches (rat, mouse, and human inducible pluripotent stem cell-derived cardiac myocytes), we monitored TNFα levels, lactate dehydrogenase, cardiac ultrastructure and function, mitochondrial bioenergetics, and cardiac cell viability. RESULTS: In contrast to vehicle-treated mice, ultrastructural defects, including cytoplasmic swelling, mitochondrial perturbations, and elevated TNFα levels, were observed in the hearts of mice treated with DOX. While investigating the involvement of TNFα in DOX cardiotoxicity, we discovered that NF-κB was readily activated by TNFα. However, TNFα-mediated NF-κB activation was impaired in cardiac myocytes treated with DOX. This coincided with loss of K63- linked polyubiquitination of RIPK1 from the proteasomal degradation of TRAF2. Furthermore, TRAF2 protein abundance was markedly reduced in hearts of patients with cancer treated with DOX. We further established that the reciprocal actions of the ubiquitinating and deubiquitinating enzymes cellular inhibitors of apoptosis 1 and USP19 (ubiquitin-specific peptidase 19), respectively, regulated the proteasomal degradation of TRAF2 in DOX-treated cardiac myocytes. An E3-ligase mutant of cellular inhibitors of apoptosis 1 (H588A) or gain of function of USP19 prevented proteasomal degradation of TRAF2 and DOX-induced cell death. Furthermore, wild-type TRAF2, but not a RING finger mutant defective for K63-linked polyubiquitination of RIPK1, restored NF-κB signaling and suppressed DOX-induced cardiac cell death. Last, cardiomyocyte-restricted expression of TRAF2 (cardiac troponin T-adeno-associated virus 9-TRAF2) in vivo protected against mitochondrial defects and cardiac dysfunction induced by DOX. CONCLUSIONS: Our findings reveal a novel signaling axis that functionally connects the cardiotoxic effects of DOX to proteasomal degradation of TRAF2. Disruption of the critical TRAF2 survival pathway by DOX sensitizes cardiac myocytes to TNFα-mediated necrotic cell death and DOX cardiotoxicity.


Assuntos
Cardiomiopatias , NF-kappa B , Fator 2 Associado a Receptor de TNF , Animais , Apoptose , Cardiomiopatias/metabolismo , Cardiotoxicidade , Enzimas Desubiquitinantes/metabolismo , Doxorrubicina/toxicidade , Endopeptidases , Humanos , Lactato Desidrogenases/metabolismo , Camundongos , Mitocôndrias/metabolismo , Miócitos Cardíacos/metabolismo , NF-kappa B/metabolismo , Ratos , Fator 2 Associado a Receptor de TNF/genética , Troponina T/metabolismo , Fator de Necrose Tumoral alfa/metabolismo , Ubiquitina-Proteína Ligases/metabolismo , Proteases Específicas de Ubiquitina/metabolismo , Proteases Específicas de Ubiquitina/farmacologia
5.
Can J Physiol Pharmacol ; 101(1): 1-7, 2023 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-36318826

RESUMO

Cardiovascular disease is the leading cause of morbidity and mortality worldwide. However, sex differences can impact differently the etiology and outcome of cardiovascular disease when comparing men and women. Women have unique genetic and hormonal risk factors that can be associated with the development of cardiovascular diseases. Furthermore, certain phenotypes of cardiovascular diseases are more prevalent to women. Molecular clocks control circadian rhythms of different physiological systems in our body, including the cardiovascular system. Increased evidence in recent years points to a link between cardiovascular disease and regulation by circadian rhythms. However, the difference between circadian regulation of cardiovascular disease in women and men is poorly understood. In this review, we highlight the recent advances in circadian-regulated cardiovascular diseases with a specific focus on the pathogenesis of heart disease in women. Understanding circadian-regulated pathways and sex-specific differences between men and women may contribute to better diagnosis and development of sex-targeted interventions to better treat cardiovascular diseases.


Assuntos
Doenças Cardiovasculares , Sistema Cardiovascular , Relógios Circadianos , Feminino , Masculino , Humanos , Doenças Cardiovasculares/etiologia , Doenças Cardiovasculares/genética , Relógios Circadianos/genética , Ritmo Circadiano/genética , Fatores de Risco
7.
Circulation ; 139(7): 965-980, 2019 02 12.
Artigo em Inglês | MEDLINE | ID: mdl-30742538

RESUMO

BACKGROUND: Over the past several years, a variety of human and animal studies have shown that circadian clocks regulate biological cardiovascular rhythms in both health and disease. For example, heart rate and blood pressure fluctuate over 24-hour daily periods, such that levels are higher in the morning and progressively decline in the evening. METHODS AND RESULTS: It is interesting to note that the timing of the administration of various cardiac treatments can also benefit some cardiovascular outcomes. Circadian rhythms have been implicated in the pathogenesis of a number of cardiovascular diseases, including myocardial infarction, ischemia-reperfusion injury after myocardial infarction, and heart failure. Cell death is a major component of ischemia-reperfusion injury and posited as the central underlying cause of ventricular remodeling and cardiac dysfunction following myocardial infarction. It is notable that the time of day profoundly influences cardiac tolerance and sensitivity to cardiac injury. CONCLUSIONS: Herein, we highlight the novel relationship between circadian rhythms and homeostatic processes that governs cell fate by apoptosis, necrosis, and autophagy. Understanding how these intricate processes interconnect at the cellular level is of paramount clinical importance for optimizing treatment strategies to achieve maximum cardiovascular outcome.


Assuntos
Apoptose , Autofagia , Doenças Cardiovasculares/patologia , Ritmo Circadiano , Miócitos Cardíacos/patologia , Animais , Proteínas Reguladoras de Apoptose/metabolismo , Proteínas Relacionadas à Autofagia/metabolismo , Doenças Cardiovasculares/metabolismo , Doenças Cardiovasculares/fisiopatologia , Doenças Cardiovasculares/terapia , Cronoterapia , Peptídeos e Proteínas de Sinalização do Ritmo Circadiano/metabolismo , Humanos , Miócitos Cardíacos/metabolismo , Necrose , Transdução de Sinais , Fatores de Tempo
11.
J Neuroinflammation ; 13(1): 123, 2016 05 26.
Artigo em Inglês | MEDLINE | ID: mdl-27230771

RESUMO

BACKGROUND: Amyotrophic lateral sclerosis (ALS) is a progressive fatal neurodegenerative disease, involving both upper and lower motor neurons. The disease is induced by multifactorial pathologies, and as such, it requires a multifaceted therapeutic approach. CXCR4, a chemokine receptor widely expressed in neurons and glial cells and its ligand, CXCL12, also known as stromal-cell-derived factor (SDF1), modulate both neuronal function and apoptosis by glutamate release signaling as well as hematopoietic stem and progenitor cells (HSPCs) migration into the blood and their homing towards injured sites. Inhibition approaches towards the CXCR4/CXCL12 signaling may result in preventing neuronal apoptosis and alter the HSPCs migration and homing. Such inhibition can be achieved by means of treatment with AMD3100, an antagonist of the chemokine receptor CXCR4. METHODS: We chronically treated male and female transgenic mice model of ALS, SOD1(G93A) mice, with AMD3100. Mice body weight and motor function, evaluated by Rotarod test, were recorded once a week. The most effective treatment regimen was repeated for biochemical and histological analyses in female mice. RESULTS: We found that chronic administration of AMD3100 to SOD1(G93A) mice led to significant extension in mice lifespan and improved motor function and weight loss. In addition, the treatment significantly improved microglial pathology and decreased proinflammatory cytokines in spinal cords of treated female mice. Furthermore, AMD3100 treatment decreased blood-spinal cord barrier (BSCB) permeability by increasing tight junction proteins levels and increased the motor neurons count in the lamina X area of the spinal cord, where adult stem cells are formed. CONCLUSIONS: These data, relevant to the corresponding disease mechanism in human ALS, suggest that blocking CXCR4 by the small molecule, AMD3100, may provide a novel candidate for ALS therapy with an increased safety.


Assuntos
Esclerose Lateral Amiotrófica/tratamento farmacológico , Esclerose Lateral Amiotrófica/patologia , Compostos Heterocíclicos/uso terapêutico , Receptores CXCR4/antagonistas & inibidores , Esclerose Lateral Amiotrófica/genética , Animais , Benzilaminas , Proteínas de Ligação ao Cálcio/metabolismo , Quimiocina CCL2/metabolismo , Claudina-5/metabolismo , Ciclamos , Citocinas/metabolismo , Modelos Animais de Doenças , Regulação da Expressão Gênica/efeitos dos fármacos , Regulação da Expressão Gênica/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Proteínas dos Microfilamentos/metabolismo , Neurônios Motores/patologia , Força Muscular/efeitos dos fármacos , Força Muscular/genética , Transtornos Psicomotores/tratamento farmacológico , Transtornos Psicomotores/etiologia , Transtornos Psicomotores/genética , Superóxido Dismutase/genética , Superóxido Dismutase/metabolismo , Proteína da Zônula de Oclusão-1/metabolismo
17.
Neurodegener Dis ; 13(2-3): 107-9, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-23942195

RESUMO

BACKGROUND AND METHODS: We previously reported the development of site-directed monoclonal antibodies able to inhibit the initiation of the amyloid precursor protein (APP) processing site, named 'blocking ß site 1' (BBS1). The beneficial effect of intracerebroventricular administration of these antibodies in the triple transgenic mouse model of Alzheimer's disease (AD) showed improvement in cognitive functions and reduction in tau and amyloid pathology. Amyloid-ß may not be the only active component of AD neurotoxicity and may involve other proteolytic APP fragments such as the APP intracellular domain, proposed to work as a transcription factor involved in the regulation of p53 and glycogen synthase kinase 3ß (GSK3ß) as well as affecting several physiological processes contributing to AD pathology. RESULTS: We show that inhibition of the ß-secretase cleavage site via site-directed antibodies resulted in a major reduction in phosphorylated GSK3ß levels, which is the active form of GSK3ß, as well as in p53 levels. CONCLUSION: A therapy that is capable of reducing not only the direct hallmarks of AD but also the components that lead to neuronal apoptosis might have neuroprotective potential in AD treatment.


Assuntos
Doença de Alzheimer/metabolismo , Precursor de Proteína beta-Amiloide/antagonistas & inibidores , Anticorpos Monoclonais/uso terapêutico , Apoptose/efeitos dos fármacos , Doença de Alzheimer/genética , Animais , Apoptose/genética , Modelos Animais de Doenças , Regulação para Baixo , Humanos , Camundongos , Camundongos Transgênicos
18.
Cardiovasc Res ; 120(2): 164-173, 2024 03 13.
Artigo em Inglês | MEDLINE | ID: mdl-38165268

RESUMO

AIMS: The mitochondrial dynamics protein Mitofusin 2 (MFN2) coordinates critical cellular processes including mitochondrial bioenergetics, quality control, and cell viability. The NF-κB kinase IKKß suppresses mitochondrial injury in doxorubicin cardiomyopathy, but the underlying mechanism is undefined. METHODS AND RESULTS: Herein, we identify a novel signalling axis that functionally connects IKKß and doxorubicin cardiomyopathy to a mechanism that impinges upon the proteasomal stabilization of MFN2. In contrast to vehicle-treated cells, MFN2 was highly ubiquitinated and rapidly degraded by the proteasomal-regulated pathway in cardiac myocytes treated with doxorubicin. The loss of MFN2 activity resulted in mitochondrial perturbations, including increased reactive oxygen species (ROS) production, impaired respiration, and necrotic cell death. Interestingly, doxorubicin-induced degradation of MFN2 and mitochondrial-regulated cell death were contingent upon IKKß kinase activity. Notably, immunoprecipitation and proximity ligation assays revealed that IKKß interacted with MFN2 suggesting that MFN2 may be a phosphorylation target of IKKß. To explore this possibility, mass spectrometry analysis identified a novel MFN2 phospho-acceptor site at serine 53 that was phosphorylated by wild-type IKKß but not by a kinase-inactive mutant IKKßK-M. Based on these findings, we reasoned that IKKß-mediated phosphorylation of serine 53 may influence MFN2 protein stability. Consistent with this view, an IKKß-phosphomimetic MFN2 (MFN2S53D) was resistant to proteasomal degradation induced by doxorubicin whereas wild-type MFN2 and IKKß-phosphorylation defective MFN2 mutant (MFNS53A) were readily degraded in cardiac myocytes treated with doxorubicin. Concordantly, gain of function of IKKß or MFN2S53D suppressed doxorubicin-induced mitochondrial injury and cell death. CONCLUSIONS: The findings of this study reveal a novel survival pathway for IKKß that is mutually dependent upon and obligatory linked to the phosphorylation and stabilization of the mitochondrial dynamics protein MFN2.


Assuntos
Cardiomiopatias , Quinase I-kappa B , Humanos , Quinase I-kappa B/metabolismo , Transdução de Sinais , Doxorrubicina , Proteínas Mitocondriais/metabolismo , Serina
20.
Can J Cardiol ; 39(12): 1772-1780, 2023 12.
Artigo em Inglês | MEDLINE | ID: mdl-37652255

RESUMO

Circadian rhythms are 24-hour cycles that regulate physical, mental, and behavioural changes of most living organisms. In the heart, circadian rhythms regulate processes such as heart rate, blood pressure, blood coagulability, and vascular tone. However, in addition to regulating physiologic processes, circadian rhythms regulate pathophysiologic processes in the heart. In this regard, circadian rhythms regulate the onset, severity, and outcome of many cardiovascular diseases (CVDs), including myocardial infarction, diabetic cardiomyopathy, doxorubicin (Dox)-induced cardiotoxicity, and heart failure. Notably, the underlying mechanism of many of these diseases is linked to impaired cellular quality control processes, such as autophagy. Autophagy is a homeostatic cellular process that regulates the removal of damaged cellular components, allowing their degradation and recycling into their basic constituents for production of cellular energy. Many studies from recent years point to a regulatory link between autophagy and circadian machinery in the control of CVDs. In this review, we highlight the recent discoveries in the field of circadian-induced autophagy in the heart and provide the molecular mechanisms and signalling pathways that underlie the crosstalk between autophagy and clock gene control in response to cardiac injury. Understanding the mechanisms that underlie circadian-induced autophagy in response to cardiac stress may prove to be beneficial in developing novel therapeutic approaches to treat cardiac disease.


Assuntos
Doenças Cardiovasculares , Cardiopatias , Humanos , Doenças Cardiovasculares/metabolismo , Ritmo Circadiano/genética , Autofagia/genética , Coração
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA