Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 367
Filtrar
Mais filtros

Medicinas Complementares
Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Biochem Biophys Res Commun ; 632: 181-188, 2022 12 03.
Artigo em Inglês | MEDLINE | ID: mdl-36215905

RESUMO

The number of patients with heart failure and related deaths is rapidly increasing worldwide, making it a major problem. Cardiac hypertrophy is a crucial preliminary step in heart failure, but its treatment has not yet been fully successful. In this study, we established a system to evaluate cardiomyocyte hypertrophy using a deep learning-based high-throughput screening system and identified drugs that inhibit it. First, primary cultured cardiomyocytes from neonatal rats were stimulated by both angiotensin II and endothelin-1, and cellular images were captured using a phase-contrast microscope. Subsequently, we used a deep learning model for instance segmentation and established a system to automatically and unbiasedly evaluate the cardiomyocyte size and perimeter. Using this system, we screened 100 FDA-approved drugs library and identified 12 drugs that inhibited cardiomyocyte hypertrophy. We focused on ezetimibe, a cholesterol absorption inhibitor, that inhibited cardiomyocyte hypertrophy in a dose-dependent manner in vitro. Additionally, ezetimibe improved the cardiac dysfunction induced by pressure overload in mice. These results suggest that the deep learning-based system is useful for the evaluation of cardiomyocyte hypertrophy and drug screening, leading to the development of new treatments for heart failure.


Assuntos
Cardiomegalia , Aprendizado Profundo , Avaliação Pré-Clínica de Medicamentos , Insuficiência Cardíaca , Animais , Camundongos , Ratos , Angiotensina II/farmacologia , Cardiomegalia/diagnóstico por imagem , Cardiomegalia/tratamento farmacológico , Células Cultivadas , Colesterol , Avaliação Pré-Clínica de Medicamentos/métodos , Endotelina-1 , Ezetimiba , Insuficiência Cardíaca/tratamento farmacológico , Miócitos Cardíacos/citologia , Miócitos Cardíacos/efeitos dos fármacos
2.
Bioengineered ; 13(1): 280-290, 2022 01.
Artigo em Inglês | MEDLINE | ID: mdl-34967264

RESUMO

Long noncoding RNAs (lncRNAs) exert essential effects in regulating myocardial ischemia/reperfusion (MI/R)-induced injury. This work intended to explore the functions of lncRNA SOX2-OT and its regulatory mechanism within MI/R-induced injury. In this study, gene expression was determined by RT-qPCR. Western blotting was applied for the detection of protein levels. Pro-inflammatory cytokine concentrations, cardiomyocyte viability, and apoptosis were detected via ELISA, CCK-8 and flow cytometry. In the in vitro model, SOX2-OT and YY1 were both upregulated, while miR-186-5p was downregulated. SOX2-OT knockdown attenuated oxygen-glucose deprivation/reoxygenation (OGD/R)-induced cardiomyocyte dysregulation through relieving inflammation, promoting proliferation, and reducing apoptosis in OGD/R-treated H2C9 cells. SOX2-OT positively regulated YY1 expression via miR-186-5p. Moreover, miR-186-5p inhibition or YY1 upregulation abolished the effects of SOX2-OT blocking on the inflammatory responses, proliferation, and apoptosis of OGD/R-challenged H2C9 cells. In conclusion, our results, for the first time, demonstrated that SOX2-OT inhibition attenuated MI/R injury in vitro via regulating the miR-186-5p/YY1 axis, offering potential therapeutic targets for MI/R injury treatment.


Assuntos
MicroRNAs/genética , Traumatismo por Reperfusão Miocárdica/genética , Miócitos Cardíacos/citologia , RNA Longo não Codificante/genética , Fator de Transcrição YY1/genética , Animais , Linhagem Celular , Regulação para Baixo , Modelos Biológicos , Traumatismo por Reperfusão Miocárdica/etiologia , Traumatismo por Reperfusão Miocárdica/metabolismo , Miócitos Cardíacos/química , Ratos , Transdução de Sinais , Regulação para Cima , Fator de Transcrição YY1/metabolismo
3.
J Cell Mol Med ; 25(24): 11257-11263, 2021 12.
Artigo em Inglês | MEDLINE | ID: mdl-34761519

RESUMO

Cardiovascular disease is a leading cause of death and disability worldwide. Although genetically modified mouse models offer great potential for robust research in vivo, in vitro studies using isolated cardiomyocytes also provide an important approach for investigating the mechanisms underlying cardiovascular disease pathogenesis and drug actions. Currently, isolation of mouse adult cardiomyocytes often relies on aortic retrograde intubation under a stereoscopic microscope, which poses considerable technical barriers and requires extensive training. Although a simplified, Langendorff-free method has been used to isolate viable cardiomyocytes from the adult mouse heart, the system requires enzymatic digestions and continuous manual technical operation. This study established an optimized approach that allows isolation of adult mouse cardiomyocytes and epicardial activation mapping of mouse hearts using a Langendorff device. We used retrograde puncture through the abdominal aorta in vivo and enzymatic digestion on the Langendorff perfusion device to isolate adult mouse cardiomyocytes without using a microscope. The yields of isolated cardiomyocytes were amenable to patch clamp techniques. Furthermore, this approach allowed epicardial activation mapping. We used a novel, simplified method to isolate viable cardiomyocytes from adult mouse hearts and to map epicardial activation. This novel approach could be beneficial in more extensive research in the cardiac field.


Assuntos
Separação Celular , Mapeamento Epicárdico , Miócitos Cardíacos/citologia , Miócitos Cardíacos/fisiologia , Potenciais de Ação , Animais , Técnicas de Cultura de Células , Separação Celular/métodos , Avaliação Pré-Clínica de Medicamentos , Técnicas Eletrofisiológicas Cardíacas , Mapeamento Epicárdico/métodos , Camundongos , Miócitos Cardíacos/efeitos dos fármacos , Técnicas de Patch-Clamp
4.
Oxid Med Cell Longev ; 2021: 5876841, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34603599

RESUMO

Myocardial fibrosis represents the primary pathological change associated with diabetic cardiomyopathy and heart failure, and it leads to decreased myocardial compliance with impaired cardiac diastolic and systolic function. Quercetin, an active ingredient in various medicinal plants, exerts therapeutic effects against cardiovascular diseases. Here, we investigate whether SIRT5- and IDH2-related desuccinylation is involved in the underlying mechanism of myocardial fibrosis in heart failure while exploring related therapeutic drugs for mitochondrial quality surveillance. Mouse models of myocardial fibrosis and heart failure, established by transverse aortic constriction (TAC), were administered with quercetin (50 mg/kg) daily for 4 weeks. HL-1 cells were pretreated with quercetin and treated with high glucose (30 mM) in vitro. Cardiac function, western blotting, quantitative PCR, enzyme-linked immunosorbent assay, and immunofluorescence analysis were employed to analyze mitochondrial quality surveillance, oxidative stress, and inflammatory response in myocardial cells, whereas IDH2 succinylation levels were detected using immunoprecipitation. Myocardial fibrosis and heart failure incidence increased after TAC, with abnormal cardiac ejection function. Following high-glucose treatment, HL-1 cell activity was inhibited, causing excess production of reactive oxygen species and inhibition of mitochondrial respiratory complex I/III activity and mitochondrial antioxidant enzyme activity, as well as increased oxidative stress and inflammatory response, imbalanced mitochondrial quality surveillance and homeostasis, and increased apoptosis. Quercetin inhibited myocardial fibrosis and improved cardiac function by increasing mitochondrial energy metabolism and regulating mitochondrial fusion/fission and mitochondrial biosynthesis while inhibiting the inflammatory response and oxidative stress injury. Additionally, TAC inhibited SIRT5 expression at the mitochondrial level and increased IDH2 succinylation. However, quercetin promoted the desuccinylation of IDH2 by increasing SIRT5 expression. Moreover, treatment with si-SIRT5 abolished the protective effect of quercetin on cell viability. Hence, quercetin may promote the desuccinylation of IDH2 through SIRT5, maintain mitochondrial homeostasis, protect mouse cardiomyocytes under inflammatory conditions, and improve myocardial fibrosis, thereby reducing the incidence of heart failure.


Assuntos
Glucose/farmacologia , Mitocôndrias Cardíacas/efeitos dos fármacos , Substâncias Protetoras/farmacologia , Quercetina/farmacologia , Sirtuínas/metabolismo , Animais , Sobrevivência Celular/efeitos dos fármacos , Modelos Animais de Doenças , Metabolismo Energético/efeitos dos fármacos , Insuficiência Cardíaca/tratamento farmacológico , Insuficiência Cardíaca/metabolismo , Insuficiência Cardíaca/patologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Mitocôndrias Cardíacas/metabolismo , Miócitos Cardíacos/citologia , Miócitos Cardíacos/efeitos dos fármacos , Miócitos Cardíacos/metabolismo , Proteína 3 que Contém Domínio de Pirina da Família NLR/metabolismo , Estresse Oxidativo/efeitos dos fármacos , Substâncias Protetoras/uso terapêutico , Quercetina/uso terapêutico , Espécies Reativas de Oxigênio/metabolismo
5.
Adv Sci (Weinh) ; 8(21): e2100627, 2021 11.
Artigo em Inglês | MEDLINE | ID: mdl-34486241

RESUMO

Optical stimulation technologies are gaining great consideration in cardiology, neuroscience studies, and drug discovery pathways by providing control over cell activity with high spatio-temporal resolution. However, this high precision requires manipulation of biological processes at genetic level concealing its development from broad scale application. Therefore, translating these technologies into tools for medical or pharmacological applications remains a challenge. Here, an all-optical nongenetic method for the modulation of electrogenic cells is introduced. It is demonstrated that plasmonic metamaterials can be used to elicit action potentials by converting near infrared laser pulses into stimulatory currents. The suggested approach allows for the stimulation of cardiomyocytes and neurons directly on commercial complementary metal-oxide semiconductor microelectrode arrays coupled with ultrafast pulsed laser, providing both stimulation and network-level recordings on the same device.


Assuntos
Potenciais de Ação/efeitos dos fármacos , Raios Infravermelhos , Miócitos Cardíacos/fisiologia , Nanoestruturas/toxicidade , Neurônios/fisiologia , Potenciais de Ação/efeitos da radiação , Animais , Linhagem Celular , Humanos , Metais/química , Camundongos , Microeletrodos , Miócitos Cardíacos/citologia , Nanoestruturas/química , Neurônios/citologia , Porosidade , Ratos , Semicondutores , Dióxido de Silício/química
6.
Commun Biol ; 4(1): 926, 2021 07 29.
Artigo em Inglês | MEDLINE | ID: mdl-34326460

RESUMO

Patients with cardiovascular comorbidities are more susceptible to severe infection with SARS-CoV-2, known to directly cause pathological damage to cardiovascular tissue. We outline a screening platform using human embryonic stem cell-derived cardiomyocytes, confirmed to express the protein machinery critical for SARS-CoV-2 infection, and a SARS-CoV-2 spike-pseudotyped virus system. The method has allowed us to identify benztropine and DX600 as novel inhibitors of SARS-CoV-2 infection in a clinically relevant stem cell-derived cardiomyocyte line. Discovery of new medicines will be critical for protecting the heart in patients with SARS-CoV-2, and for individuals where vaccination is contraindicated.


Assuntos
Antivirais/farmacologia , Avaliação Pré-Clínica de Medicamentos/métodos , Células-Tronco Embrionárias Humanas/citologia , Miócitos Cardíacos/efeitos dos fármacos , Miócitos Cardíacos/virologia , SARS-CoV-2/fisiologia , Benzotropina/farmacologia , Humanos , Miócitos Cardíacos/citologia , Peptídeos/farmacologia
7.
J Mol Cell Cardiol ; 160: 97-110, 2021 11.
Artigo em Inglês | MEDLINE | ID: mdl-34216608

RESUMO

Angiotensin II (Ang II) presents a critical mediator in various pathological conditions such as non-genetic cardiomyopathy. Osmotic pump infusion in rodents is a commonly used approach to model cardiomyopathy associated with Ang II. However, profound differences in electrophysiology and pharmacokinetics between rodent and human cardiomyocytes may limit predictability of animal-based experiments. This study investigates the application of an Organ-on-a-chip (OOC) system in modeling Ang II-induced progressive cardiomyopathy. The disease model is constructed to recapitulate myocardial response to Ang II in a temporal manner. The long-term tissue cultivation and non-invasive functional readouts enable monitoring of both acute and chronic cardiac responses to Ang II stimulation. Along with mapping of cytokine secretion and proteomic profiles, this model presents an opportunity to quantitatively measure the dynamic pathological changes that could not be otherwise identified in animals. Further, we present this model as a testbed to evaluate compounds that target Ang II-induced cardiac remodeling. Through assessing the effects of losartan, relaxin, and saracatinib, the drug screening data implicated multifaceted cardioprotective effects of relaxin in restoring contractile function and reducing fibrotic remodeling. Overall, this study provides a controllable platform where cardiac activities can be explicitly observed and tested over the pathological process. The facile and high-content screening can facilitate the evaluation of potential drug candidates in the pre-clinical stage.


Assuntos
Angiotensina II/efeitos adversos , Cardiomiopatias/induzido quimicamente , Cardiomiopatias/metabolismo , Miócitos Cardíacos/efeitos dos fármacos , Miócitos Cardíacos/patologia , Animais , Cardiomiopatias/patologia , Cardiotônicos/farmacologia , Linhagem Celular , Sobrevivência Celular/efeitos dos fármacos , Técnicas de Cocultura , Avaliação Pré-Clínica de Medicamentos/métodos , Fibroblastos/metabolismo , Fibrose , Humanos , Células-Tronco Pluripotentes Induzidas/citologia , Dispositivos Lab-On-A-Chip , Losartan/farmacologia , Camundongos , Miócitos Cardíacos/citologia , Miócitos Cardíacos/metabolismo , Projetos Piloto , Proteoma , Proteômica/métodos , Proteínas Recombinantes/farmacologia , Relaxina/farmacologia , Remodelação Ventricular/efeitos dos fármacos
8.
Acta Histochem ; 123(5): 151739, 2021 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-34107386

RESUMO

Resveratrol is a kind of iPolyphenols widely existing in herbal medicine. Here we aim to investigate whether resveratrol can reduce the degree of myocardial ischemia/reperfusion (IR) injury and inhibit the development of oxidative stress, and elucidate the molecular mechanism of resveratrol in protecting myocardial cells. The primary rat cardiomyocytes were used to establish an ischemia/reperfusion model in vitro, and a series of routine biochemical experiments were conducted to explore the antioxidant and anti-apoptotic effects of resveratrol in myocardial ischemia-reperfusion injury. Compared with that of the simulated ischemia-refusion (SIR) group, cell viability in the SIR and resveratrol co-treatment groups increased significantly (P < 0.001), the release of lactate dehydrogenase (LDH) and creatine kinase MB (CKMB) decreased, the positive rate of reactive oxygen species (ROS) in cardiomyocytes decreased, and the concentration of catalase and glutathione peroxidase increased significantly (P < 0.001). Besides, resveratrol can activate PI3K/AKT signaling pathway. PI3K siRNA can inhibit the PI3K/AKT signaling mediated by resveratrol. The addition of resveratrol can significantly increase the activity of mitochondrial superoxide dismutase (SOD) and reduce the malondialdehyde (MDA), which indicates that the oxidative damage of mitochondria induced by resveratrol was significantly weakened. The mitochondrial functional changes induced by resveratrol can be reversed by PI3K siRNA. In conclusion, our study shows that resveratrol can reduce ROS in cardiomyocytes by PI3K/AKT signaling pathway activation, and effectively inhibit the apoptosis of cardiomyocytes, thus having a direct protective effect on cardiomyocytes under SR.


Assuntos
Apoptose , Mitocôndrias/metabolismo , Miocárdio/patologia , Fosfatidilinositol 3-Quinases/metabolismo , Polifenóis/química , Proteínas Proto-Oncogênicas c-akt/metabolismo , Traumatismo por Reperfusão/patologia , Animais , Animais Recém-Nascidos , Sobrevivência Celular , Creatina Quinase Forma MB/biossíntese , L-Lactato Desidrogenase/antagonistas & inibidores , Masculino , Malondialdeído/química , Miócitos Cardíacos/citologia , RNA Interferente Pequeno/metabolismo , Ratos , Espécies Reativas de Oxigênio , Resveratrol/farmacologia , Transdução de Sinais
9.
Sci Rep ; 11(1): 11970, 2021 06 07.
Artigo em Inglês | MEDLINE | ID: mdl-34099836

RESUMO

The NADPH oxidase enzymes Nox2 and 4, are important generators of Reactive oxygen species (ROS). These enzymes are abundantly expressed in cardiomyocytes and have been implicated in ischemia-reperfusion injury. Previous attempts with full inhibition of their activity using genetically modified animals have shown variable results, suggesting that a selective and graded inhibition could be a more relevant approach. We have, using chemical library screening, identified a new compound (GLX481304) which inhibits Nox 2 and 4 (with IC50 values of 1.25 µM) without general antioxidant effects or inhibitory effects on Nox 1. The compound inhibits ROS production in isolated mouse cardiomyocytes and improves cardiomyocyte contractility and contraction of whole retrogradely (Langendorff) perfused hearts after a global ischemia period. We conclude that a pharmacological and partial inhibition of ROS production by inhibition of Nox 2 and 4 is beneficial for recovery after ischemia reperfusion and might be a promising venue for treatment of ischemic injury to the heart.


Assuntos
Antioxidantes/química , Inibidores Enzimáticos/química , Traumatismo por Reperfusão Miocárdica/tratamento farmacológico , NADPH Oxidase 2/metabolismo , NADPH Oxidase 4/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Animais , Antioxidantes/farmacologia , Avaliação Pré-Clínica de Medicamentos , Inibidores Enzimáticos/farmacologia , Feminino , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Miócitos Cardíacos/citologia , Miócitos Cardíacos/metabolismo , Oxirredução , Estresse Oxidativo , RNA Mensageiro/metabolismo , Bibliotecas de Moléculas Pequenas/química , Bibliotecas de Moléculas Pequenas/farmacologia
10.
Bioelectrochemistry ; 140: 107811, 2021 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-33862549

RESUMO

Exposures to short-duration, strong electric field pulses have been utilized for stimulation, ablation, and the delivery of molecules into cells. Ultrashort, nanosecond duration pulses have shown unique benefits, but they require higher field strengths. One way to overcome this requirement is to use trains of nanosecond pulses with high repetition rates, up to the MHz range. Here we present a theoretical model to describe the effects of pulse trains on the plasma membrane and intracellular membranes modeled as resistively charged capacitors. We derive the induced membrane potential and the stimulation threshold as functions of pulse number, pulse duration, and repetition rate. This derivation provides a straightforward method to calculate the membrane charging time constant from experimental data. The derived excitation threshold agrees with nerve stimulation experiments, indicating that nanosecond pulses are not more effective than longer pulses in charging nerve fibers. The derived excitation threshold does not, however, correctly predict the nanosecond stimulation of cardiomyocytes. We show that a better agreement is possible if multiple charging time constants are considered. Finally, we expand the model to intracellular membranes and show that pulse trains do not lead to charge buildup, but can create significant oscillations of the intracellular membrane potential.


Assuntos
Estimulação Elétrica , Eletroporação , Animais , Cálcio/metabolismo , Membrana Celular/metabolismo , Permeabilidade da Membrana Celular , Modelos Biológicos , Miócitos Cardíacos/citologia , Miócitos Cardíacos/metabolismo
11.
Int J Mol Sci ; 22(5)2021 Feb 27.
Artigo em Inglês | MEDLINE | ID: mdl-33673574

RESUMO

Offspring born to diabetic or obese mothers have a higher lifetime risk of heart disease. Previously, we found that rat offspring exposed to late-gestational diabetes mellitus (LGDM) and maternal high-fat (HF) diet develop mitochondrial dysfunction, impaired cardiomyocyte bioenergetics, and cardiac dysfunction at birth and again during aging. Here, we compared echocardiography, cardiomyocyte bioenergetics, oxidative damage, and mitochondria-mediated cell death among control, pregestational diabetes mellitus (PGDM)-exposed, HF-diet-exposed, and combination-exposed newborn offspring. We hypothesized that PGDM exposure, similar to LGDM, causes mitochondrial dysfunction to play a central, pathogenic role in neonatal cardiomyopathy. We found that PGDM-exposed offspring, similar to LGDM-exposed offspring, have cardiac dysfunction at birth, but their isolated cardiomyocytes have seemingly less bioenergetics impairment. This finding was due to confounding by impaired viability related to poorer ATP generation, more lipid peroxidation, and faster apoptosis under metabolic stress. To mechanistically isolate and test the role of mitochondria, we transferred mitochondria from normal rat myocardium to control and exposed neonatal rat cardiomyocytes. As expected, transfer provides a respiratory boost to cardiomyocytes from all groups. They also reduce apoptosis in PGDM-exposed males, but not in females. Findings highlight sex-specific differences in mitochondria-mediated mechanisms of developmentally programmed heart disease and underscore potential caveats of therapeutic mitochondrial transfer.


Assuntos
Diabetes Mellitus Experimental/complicações , Diabetes Gestacional/fisiopatologia , Metabolismo Energético , Cardiopatias/prevenção & controle , Mitocôndrias/transplante , Miócitos Cardíacos/citologia , Miócitos Cardíacos/fisiologia , Animais , Dieta Hiperlipídica/efeitos adversos , Feminino , Cardiopatias/etiologia , Cardiopatias/patologia , Masculino , Fenômenos Fisiológicos da Nutrição Materna , Gravidez , Efeitos Tardios da Exposição Pré-Natal/etiologia , Efeitos Tardios da Exposição Pré-Natal/patologia , Efeitos Tardios da Exposição Pré-Natal/prevenção & controle , Ratos , Ratos Sprague-Dawley , Fatores Sexuais
12.
J Tradit Chin Med ; 41(1): 51-58, 2021 02.
Artigo em Inglês | MEDLINE | ID: mdl-33522197

RESUMO

OBJECTIVE: To investigate the efficacy of water fraction from Dioscorea cirrhosa (WF) on oxidative damage and apoptosis of cardiomyocytes induced by H2O2, and to study its mechanism. METHODS: Cell viability was measured by the MST assay kit. The content of malondialdehyde (MDA), release of lactate dehydrogenase (LDH) and activity of catalase (CAT) and superoxide dismutase (SOD) were detected by biochemical kit. The content of reactive oxygen species (ROS) was assessed by nonfluorescent probe 2' ,7'-dichlorofluorescin diacetate (DCFH-DA). JC-1 was used to analyze the mitochondrial membrane potential (mtΔΨ) and Annexin-V-FITC/PI staining was applied to assess apoptosis of H9c2 by flow cytometry. Moreover, the expression of B-cell lymphoma-2 (Bcl-2), Bcl-2-associated X (Bax), caspase-3, caspase-9, cleaved-caspase-3 and cleaved-caspase -9 proteins was determined by western blot analysis. RESULTS: WF increased cell viability and decreased LDH leakage in H9c2 cells exposed to H2O2. WF treatment decreased ROS and MDA level, enhanced SOD and CAT activities, improved mtΔΨ and inhibited apoptosis. Western blot analysis demonstrated that the ratio of Bcl-2/Bax was increased and the expression cleaved-caspase-3, caspase-3, cleaved-caspase-9 and caspase-9 were decreased in group treated with WF. CONCLUSION: WF protects H9c2 myocardial cells on H2O2-induced oxidative stress and apoptosis by scavenging ROS, improving antioxidant capacity, protecting mitochondrial and regulating the proteins expression related to apoptosis.


Assuntos
Apoptose/efeitos dos fármacos , Dioscorea/química , Miócitos Cardíacos/efeitos dos fármacos , Estresse Oxidativo/efeitos dos fármacos , Extratos Vegetais/farmacologia , Animais , Linhagem Celular , Sobrevivência Celular/efeitos dos fármacos , Peróxido de Hidrogênio/toxicidade , Malondialdeído/metabolismo , Potencial da Membrana Mitocondrial/efeitos dos fármacos , Miócitos Cardíacos/citologia , Miócitos Cardíacos/metabolismo , Substâncias Protetoras/farmacologia , Ratos , Espécies Reativas de Oxigênio/metabolismo , Transdução de Sinais/efeitos dos fármacos , Superóxido Dismutase/metabolismo
13.
Phytomedicine ; 82: 153406, 2021 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-33422954

RESUMO

BACKGROUND: Diabetic cardiomyopathy is characterized by both systolic and diastolic dysfunction due to decreased contractility, as well as reduced compliance of the myocardium. Oxidative stress plays a significant role in diabetes mellitus and its cardiovascular complications. Salidroside, a glucoside of the phenylpropanoid tyrosol, reportedly increases the levels of the antioxidative enzymes, nuclear factor erythroid 2-related factor 2, and heme oxygenase-1 (HO-1) to counteract oxidative stress; however, the underlying mechanisms are poorly understood. PURPOSE: Here we investigate the potential cardio-protective effects of salidroside and its mechanism in a diabetic animal model. METHODS: Male db/m, db/db, and age-matched wild-type mice were treated with salidroside at low dose (0.025 mg/kg) or high dose (0.05 mg/kg) by gavage every day for 12 weeks. Cardiac function and structure were assessed by echocardiography and histopathological examination. H9C2 cardiomyocytes were exposed in vitro to advanced glycosylation end products (400 µg/ml) and treated with salidroside (0.1, 1, or 10 µM). The expression of signaling-related genes were explored by western blotting and real-time PCR. RESULTS: Salidroside treatment significantly improved diabetes-induced cardiac dysfunction, hypertrophy, and fibrosis in vivo. Mechanistically, salidroside markedly up-regulates HO-1 expression by activation of the AKT signaling pathway. CONCLUSION: Salidroside protects against cardiomyocyte apoptosis and ventricular remodeling in diabetic mice. This cardio-protective effect of salidroside is dependent on AKT signaling activation.


Assuntos
Apoptose/efeitos dos fármacos , Cardiomiopatias Diabéticas/metabolismo , Glucosídeos/farmacologia , Heme Oxigenase-1/metabolismo , Miócitos Cardíacos/efeitos dos fármacos , Fenóis/farmacologia , Proteínas Proto-Oncogênicas c-akt/metabolismo , Transdução de Sinais/efeitos dos fármacos , Remodelação Ventricular/efeitos dos fármacos , Animais , Modelos Animais de Doenças , Relação Dose-Resposta a Droga , Glucosídeos/administração & dosagem , Masculino , Camundongos , Miócitos Cardíacos/citologia , Fenóis/administração & dosagem
14.
Nutrients ; 13(1)2021 Jan 19.
Artigo em Inglês | MEDLINE | ID: mdl-33477916

RESUMO

Cardiovascular diseases (CVDs), which include congenital heart disease, rhythm disorders, subclinical atherosclerosis, coronary heart disease, and many other cardiac disorders, cause about 30% of deaths globally; representing one of the main health problems worldwide. Among CVDs, ischemic heart diseases (IHDs) are one of the major causes of morbidity and mortality in the world. The onset of IHDs is essentially due to an unbalance between the metabolic demands of the myocardium and its supply of oxygen and nutrients, coupled with a low regenerative capacity of the heart, which leads to great cardiomyocyte (CM) loss; promoting heart failure (HF) and myocardial infarction (MI). To date, the first strategy recommended to avoid IHDs is prevention in order to reduce the underlying risk factors. In the management of IHDs, traditional therapeutic options are widely used to improve symptoms, attenuate adverse cardiac remodeling, and reduce early mortality rate. However, there are no available treatments that aim to improve cardiac performance by replacing the irreversible damaged cardiomyocytes (CMs). Currently, heart transplantation is the only treatment being carried out for irreversibly damaged CMs. Hence, the discovery of new therapeutic options seems to be necessary. Interestingly, recent experimental evidence suggests that regenerative stem cell medicine could be a useful therapeutic approach to counteract cardiac damage and promote tissue regeneration. To this end, researchers are tasked with answering one main question: how can myocardial regeneration be stimulated? In this regard, natural compounds from plant extracts seem to play a particularly promising role. The present review will summarize the recent advances in our knowledge of stem cell therapy in the management of CVDs; focusing on the main properties and potential mechanisms of natural compounds in stimulating and activating stem cells for myocardial regeneration.


Assuntos
Doenças Cardiovasculares/terapia , Miócitos Cardíacos/fisiologia , Extratos Vegetais/farmacologia , Transplante de Células-Tronco , Células-Tronco/fisiologia , Animais , Doenças Cardiovasculares/patologia , Doenças Cardiovasculares/fisiopatologia , Diferenciação Celular , Suplementos Nutricionais , Humanos , Miócitos Cardíacos/citologia , Regeneração , Células-Tronco/citologia
15.
J Mol Histol ; 52(1): 87-99, 2021 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-33179120

RESUMO

Human pluripotent stem cell (hPSC)-derived cardiomyocytes (CMs) hold great promise for the repair of the injured heart, but optimal cell production in a fully chemically defined and cost-effective system is essential for the efficacy and safety of cell transplantation therapies. In this study, we provided a simple and efficient strategy for cardiac differentiation from hPSCs and performed functional evaluation in a rat model of myocardial infarction. Using a chemically defined medium including four components, recombinant human albumin, ascorbic acid, human transferrin, and RPMI 1640, we developed a manageable and cost-effective protocol for robust generation of CMs from hPSCs. Interestingly, the addition of transferrin helped hPSCs to transit from TeSR-E8 medium to the simple cardiac differentiation medium and successfully initiated mesoderm differentiation without significant cell death. The CM generation efficiency was up to 85% based on cTnT expression. We performed transcriptome profiling from differentiation day 0 to 35, and characterized interesting dynamic change of cardiac genes. CMs derived from transferrin-supplemented simple medium have similar transcriptome and the maturation level compared to those generated in B27 minus insulin medium as well as their in vivo counterparts. Importantly, after transplantation, hPSC-derived CMs survived in the infarcted rat heart, significantly improved the physiological function and reduced fibrosis. Our study offers an easy-to-use and cost-effective method for cardiac differentiation and facilitates the translational application of hPSC-derived CMs for heart repair.


Assuntos
Infarto do Miocárdio/patologia , Infarto do Miocárdio/terapia , Miócitos Cardíacos/citologia , Células-Tronco Pluripotentes/citologia , Regeneração , Transferrina/farmacologia , Animais , Morte Celular/efeitos dos fármacos , Diferenciação Celular/efeitos dos fármacos , Perfilação da Expressão Gênica , Regulação da Expressão Gênica/efeitos dos fármacos , Glicogênio Sintase Quinase 3 beta/antagonistas & inibidores , Glicogênio Sintase Quinase 3 beta/metabolismo , Humanos , Infarto do Miocárdio/fisiopatologia , Miócitos Cardíacos/transplante , Inibidores de Proteínas Quinases/farmacologia , Ratos , Transcriptoma/genética
16.
Stem Cell Reports ; 16(1): 168-181, 2021 01 12.
Artigo em Inglês | MEDLINE | ID: mdl-33338435

RESUMO

Burgeoning applications of human induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs) in disease modeling, regenerative medicine, and drug screening have broadened the usage of hiPSC-CMs and entailed their long-term storage. Cryopreservation is the most common approach to store hiPSC-CMs. However, the effects of cryopreservation and recovery on hiPSC-CMs remain poorly understood. Here, we characterized the transcriptome, electro-mechanical function, and drug response of fresh hiPSC-CMs without cryopreservation and recovered hiPSC-CMs from cryopreservation. We found that recovered hiPSC-CMs showed upregulation of cell cycle genes, similar or reduced contractility, Ca2+ transients, and field potential duration. When subjected to treatment of drugs that affect electrophysiological properties, recovered hiPSC-CMs showed an altered drug response and enhanced propensity for drug-induced cardiac arrhythmic events. In conclusion, fresh and recovered hiPSC-CMs do not always show comparable molecular and physiological properties. When cryopreserved hiPSC-CMs are used for assessing drug-induced cardiac liabilities, the altered drug sensitivity needs to be considered.


Assuntos
Criopreservação , Miócitos Cardíacos/metabolismo , Potenciais de Ação , Antiarrítmicos/farmacologia , Cálcio/metabolismo , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Diferenciação Celular , Sobrevivência Celular/efeitos dos fármacos , Avaliação Pré-Clínica de Medicamentos/métodos , Humanos , Células-Tronco Pluripotentes Induzidas/citologia , Células-Tronco Pluripotentes Induzidas/metabolismo , Miócitos Cardíacos/citologia , Miócitos Cardíacos/fisiologia , Nifedipino/farmacologia , Regulação para Cima/efeitos dos fármacos
17.
Methods Mol Biol ; 2239: 47-59, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33226612

RESUMO

There are currently no effective treatments to regenerate the heart after cardiac injury. Following cardiac injury, heart muscle cells, also known as cardiomyocytes, die in large numbers. The adult mammalian heart does not have the ability to replace these dead cardiomyocytes. In their place, fibroblasts invade the injury zone and generate a scar. The scar impairs cardiac function. An important approach to cardiac regeneration is direct cardiac reprogramming, whereby cardiac fibroblasts within the scar are directly converted into functional cardiomyocytes. Several laboratories have achieved direct cardiac reprogramming via overexpression of the cardiac transcription factors. In contrast, we utilize a combination of four miRNAs (miR-1, miR-133, miR-208, miR-499) that we call miR Combo. One common issue regarding direct cardiac reprogramming strategies is low efficiency. Recently, we have demonstrated that the efficiency of direct cardiac reprogramming is enhanced in the chemically defined reprogramming media.


Assuntos
Reprogramação Celular/genética , Meios de Cultura/química , Fibroblastos/citologia , MicroRNAs/metabolismo , Miócitos Cardíacos/citologia , Animais , Ácido Ascórbico/química , Técnicas de Cultura de Células/métodos , Células Cultivadas , Fibroblastos/metabolismo , Citometria de Fluxo , Imunofluorescência , Camundongos , MicroRNAs/genética , Miócitos Cardíacos/metabolismo , Reação em Cadeia da Polimerase em Tempo Real , Regeneração , Selênio/química , Transfecção/métodos
18.
Int J Biochem Cell Biol ; 131: 105902, 2021 02.
Artigo em Inglês | MEDLINE | ID: mdl-33309621

RESUMO

Fibrotic diseases account for more than 8 million deaths worldwide annually. Reactive oxygen species (ROS) has been shown to activate pyroptosis and promote the production of interleukin (IL)-1ß and IL-18, leading to fibrosis development. However, the role of dual oxidase 1 (DUOX1)-induced ROS production and pyroptosis in cardiac fibrosis remains largely unknown. Activin A was used to induce ROS and pyroptosis in cardiomyocytes. ROS level, pyroptosis, and cytokine production were detected using Active Oxygen Detection Kit, flow cytometry, and enzyme-linked immunosorbent assay, respectively. Western blotting analysis was used to measure expression changes of proteins. DUOX1 was silenced or overexpressed to investigate its role in fibrosis. We found that activin A induced ROS production and pyroptosis in cardiomyocytes, which was blocked by the ROS scavenger, N-acetyl-L-cysteine (NAC). Knockdown of DUOX1 reversed activin A-induced ROS production, pyroptosis, cytokine release, and the upregulation of proinflammatory proteins. Overexpression of DUOX1 resulted in opposite effects of knockdown DUOX1. Administration of an ROS scavenger blocked the effect of DUOX1 overexpression. Supplementation of IL-1ß and IL-18 caused significant fibrosis in human cardiac fibroblasts (hCFs). The knockdown of DUOX1 protected cardiomyocytes against activin A-induced fibrosis via the inhibition of ROS, cytokine release, and pyroptosis.


Assuntos
Ativinas/farmacologia , Oxidases Duais/genética , Miócitos Cardíacos/efeitos dos fármacos , Piroptose/efeitos dos fármacos , Espécies Reativas de Oxigênio/metabolismo , Acetilcisteína/farmacologia , Ativinas/antagonistas & inibidores , Caspase 1/genética , Caspase 1/metabolismo , Coenzima A Ligases/genética , Coenzima A Ligases/metabolismo , Colágeno Tipo I/genética , Colágeno Tipo I/metabolismo , Colágeno Tipo III/genética , Colágeno Tipo III/metabolismo , Oxidases Duais/antagonistas & inibidores , Oxidases Duais/metabolismo , Sequestradores de Radicais Livres/farmacologia , Regulação da Expressão Gênica , Humanos , Interleucina-18/genética , Interleucina-18/metabolismo , Interleucina-1beta/genética , Interleucina-1beta/metabolismo , Miócitos Cardíacos/citologia , Miócitos Cardíacos/metabolismo , Proteína 3 que Contém Domínio de Pirina da Família NLR/genética , Proteína 3 que Contém Domínio de Pirina da Família NLR/metabolismo , Estresse Oxidativo/efeitos dos fármacos , Cultura Primária de Células , Piroptose/genética , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/metabolismo , Espécies Reativas de Oxigênio/agonistas , Espécies Reativas de Oxigênio/antagonistas & inibidores , Transdução de Sinais , Proteína Smad2/genética , Proteína Smad2/metabolismo , Proteína Smad3/genética , Proteína Smad3/metabolismo
19.
Int J Mol Sci ; 21(23)2020 Nov 24.
Artigo em Inglês | MEDLINE | ID: mdl-33255277

RESUMO

Relevant, predictive normal, or disease model systems are of vital importance for drug development. The difference between nonhuman models and humans could contribute to clinical trial failures despite ideal nonhuman results. As a potential substitute for animal models, human induced pluripotent stem cell (hiPSC)-derived cardiomyocytes (CMs) provide a powerful tool for drug toxicity screening, modeling cardiovascular diseases, and drug discovery. Here, we review recent hiPSC-CM disease models and discuss the features of hiPSC-CMs, including subtype and maturation and the tissue engineering technologies for drug assessment. Updates from the international multisite collaborators/administrations for development of novel drug discovery paradigms are also summarized.


Assuntos
Diferenciação Celular/efeitos dos fármacos , Descoberta de Drogas , Células-Tronco Pluripotentes Induzidas/efeitos dos fármacos , Miócitos Cardíacos/efeitos dos fármacos , Avaliação Pré-Clínica de Medicamentos/métodos , Humanos , Células-Tronco Pluripotentes Induzidas/citologia , Contração Miocárdica/efeitos dos fármacos , Contração Miocárdica/fisiologia , Miócitos Cardíacos/citologia , Engenharia Tecidual
20.
Metallomics ; 12(12): 1965-1978, 2020 12 23.
Artigo em Inglês | MEDLINE | ID: mdl-33237045

RESUMO

Oxidative stress significantly contributes to heart disease, and thus might be a promising target for ameliorating heart failure. Mounting evidence suggests that selenium has chemotherapeutic potential for treating heart disease due to its regulation of selenoproteins, which play antioxidant regulatory roles. Oxidative stress-induced cardiomyocyte cell cycle arrest contributes to the loss of cardiomyocytes during heart failure. The protective effects and mechanism of selenium against oxidative stress-induced cell cycle arrest in cardiomyocytes warrant further study. H9c2 rat cardiomyoblast cells were treated with hydrogen peroxide in the presence or absence of selenium supplementation. Na2SeO3 pretreatment alleviated H2O2-induced oxidative stress, increased thioredoxin reductase (TXNRD) activity and glutathione peroxidase (GPx) activity and counteracted the H2O2-induced cell cycle arrest at the S phase. These effects were accompanied by attenuation of the H2O2-induced strengthening of the G2/M-phase inhibitory system, including increased mRNA and protein levels of cyclin-dependent kinase 1 (CDK1) and decreased p21 mRNA levels. Notably, Na2SeO3 pretreatment activated the PI3K/AKT signaling pathway, and inhibition of PI3K counteracted the protective effects of selenium on H2O2-induced cell cycle arrest. We corroborated our findings in vivo by inducing oxidative stress in pig heart by feeding a selenium deficient diet, which decreased the TXNRD activity, inactivated PI3K/AKT signaling and strengthened the G2/M-phase inhibitory system. We concluded that the cardioprotective effects of selenium supplementation against oxidative stress-induced cell cycle arrest in cardiomyocytes might be mediated by the selenoprotein-associated (GPx and TXNRD) antioxidant capacity, thereby activating redox status-associated PI3K/AKT pathways, which promote cell cycle progression by targeting the G2/M phase inhibitory system. This study provides new insight into the underlying mechanisms of cardioprotection effects of selenium at the cellular level.


Assuntos
Antioxidantes/farmacologia , Pontos de Checagem do Ciclo Celular/efeitos dos fármacos , Miócitos Cardíacos/efeitos dos fármacos , Estresse Oxidativo/efeitos dos fármacos , Selênio/farmacologia , Animais , Linhagem Celular , Miócitos Cardíacos/citologia , Miócitos Cardíacos/metabolismo , Fosfatidilinositol 3-Quinases/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Ratos , Transdução de Sinais/efeitos dos fármacos , Suínos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA