Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
1.
Cell Biol Toxicol ; 39(1): 145-163, 2023 02.
Artigo em Inglês | MEDLINE | ID: mdl-35870039

RESUMO

Human induced pluripotent stem cell derived cardiomyocytes (hiPSC-CMs) hold great potential in the cardiovascular field for human disease modeling, drug development, and regenerative medicine. However, multiple hurdles still exist for the effective utilization of hiPSC-CMs as a human-based experimental platform that can be an alternative to the current animal models. To further expand their potential as a research tool and bridge the translational gap, we have generated a cardiac-specific hiPSC reporter line that differentiates into fluorescent CMs using CRISPR-Cas9 genome editing technology. The CMs illuminated with the mScarlet fluorescence enable their non-invasive continuous tracking and functional cellular phenotyping, offering a real-time 2D/3D imaging platform. Utilizing the reporter CMs, we developed an imaging-based cardiotoxicity screening system that can monitor distinct drug-induced structural toxicity and CM viability in real time. The reporter fluorescence enabled visualization of sarcomeric disarray and displayed a drug dose-dependent decrease in its fluorescence. The study also has demonstrated the reporter CMs as a biomaterial cytocompatibility analysis tool that can monitor dynamic cell behavior and maturity of hiPSC-CMs cultured in various biomaterial scaffolds. This versatile cardiac imaging tool that enables real time tracking and high-resolution imaging of CMs has significant potential in disease modeling, drug screening, and toxicology testing.


Assuntos
Células-Tronco Pluripotentes Induzidas , Miócitos Cardíacos , Animais , Humanos , Miócitos Cardíacos/metabolismo , Cardiotoxicidade/metabolismo , Avaliação Pré-Clínica de Medicamentos/métodos , Cadeias Pesadas de Miosina/genética , Cadeias Pesadas de Miosina/metabolismo , Cadeias Pesadas de Miosina/farmacologia , Miosinas Cardíacas/metabolismo , Miosinas Cardíacas/farmacologia
2.
Biol Open ; 10(2)2021 02 15.
Artigo em Inglês | MEDLINE | ID: mdl-33589442

RESUMO

The fundamental basis of muscle contraction 'the sliding filament model' (Huxley and Niedergerke, 1954; Huxley and Hanson, 1954) and the 'swinging, tilting crossbridge-sliding filament mechanism' (Huxley, 1969; Huxley and Brown, 1967) nucleated a field of research that has unearthed the complex and fascinating role of myosin structure in the regulation of contraction. A recently discovered energy conserving state of myosin termed the super relaxed state (SRX) has been observed in filamentous myosins and is central to modulating force production and energy use within the sarcomere. Modulation of myosin function through SRX is a rapidly developing theme in therapeutic development for both cardiovascular disease and infectious disease. Some 70 years after the first discoveries concerning muscular function, modulation of myosin SRX may bring the first myosin targeted small molecule to the clinic, for treating hypertrophic cardiomyopathy (Olivotto et al., 2020). An often monogenic disease HCM afflicts 1 in 500 individuals, and can cause heart failure and sudden cardiac death. Even as we near therapeutic translation, there remain many questions about the governance of muscle function in human health and disease. With this review, we provide a broad overview of contemporary understanding of myosin SRX, and explore the complexities of targeting this myosin state in human disease.This article has an associated Future Leaders to Watch interview with the authors of the paper.


Assuntos
Miosinas Cardíacas/genética , Miosinas Cardíacas/metabolismo , Contração Miocárdica/fisiologia , Biomarcadores , Miosinas Cardíacas/ultraestrutura , Cardiomiopatia Hipertrófica/tratamento farmacológico , Cardiomiopatia Hipertrófica/etiologia , Cardiomiopatia Hipertrófica/metabolismo , Cardiomiopatia Hipertrófica/patologia , Gerenciamento Clínico , Suscetibilidade a Doenças , Humanos , Modelos Biológicos , Terapia de Alvo Molecular , Mutação , Fosforilação
3.
Phytomedicine ; 68: 153179, 2020 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-32062328

RESUMO

BACKGROUND: Intestinal epithelial barrier dysfunction, which involves myosin light chain kinase (MLCK) activation, contributes to the occurrence and progression of inflammation in inflammatory bowel disease (IBD). Wogonoside helps maintain intestinal homeostasis in mice with dextran sulfate sodium (DSS)-induced colitis, but it is unclear whether it modulates intestinal barrier function. PURPOSE: Here, we demonstrate that wogonoside protects against intestinal barrier dysfunction in colitis via the MLCK/pMLC2 pathway both in vivo and in vitro. METHODS: Caco-2 cell monolayers treated with the proinflammatory cytokine TNF-α showed barrier dysfunction and were assessed in the absence and presence of wogonoside for various physiological, morphological, and biochemical parameters. Colitis was induced by 3% DSS in mice, which were used as an animal model to explore the pharmacodynamics of wogonoside. We detected MLCK/pMLC2 pathway proteins via western blot analysis, assessed the cytokines IL-13 and IFN-γ via ELISA, tested bacterial translocation via fluorescence in situ hybridization (FISH) and a proper sampling of secondary lymphoid organs for bacterial culture. In addition, the docking affinity of wogonoside and MLCK was observed with DS2.5 software. RESULTS: Wogonoside alleviated the disruption of transepithelial electrical resistance (TER) in TNF-α exposured Caco-2 cell; FITC-dextran hyperpermeability; loss of the tight junction (TJ) proteins occludin, ZO-1 and claudin-1 in Caco-2 cell monolayers; and bacterial translocation in colitic mice. Moreover, wogonoside reduced the levels of the proinflammatory cytokines IL-13 and IFN-γ to maintain intestinal immune homeostasis. Transmission electron microscopy (TEM) confirmed that wogonoside ameliorated the destruction of intestinal epithelial TJs. Wogonoside not only inhibited the cytoskeletal F-actin rearrangement induced by TNF-α, stabilized the cytoskeletal structure, suppressed MLCK protein expression, and reduced MLC2 phosphorylation. In addition, the results of molecular docking analysis showed that wogonoside had a high affinity for MLCK and formed hydrogen bonds with the amino acid residue LYS261 and π bonds with LYS229. CONCLUSION: Collectively, our study indicates that wogonoside alleviates colitis by protecting against intestinal barrier dysfunction, and the potential mechanism may involve regulation of TJs via the MLCK/pMLC2 signaling pathway. Meanwhile, our study also explains the success of S. baicalensis in the treatment of ulcerative colitis (UC).


Assuntos
Miosinas Cardíacas/metabolismo , Colite/tratamento farmacológico , Flavanonas/farmacologia , Glucosídeos/farmacologia , Cadeias Leves de Miosina/metabolismo , Quinase de Cadeia Leve de Miosina/metabolismo , Animais , Células CACO-2 , Colite/induzido quimicamente , Colite/metabolismo , Sulfato de Dextrana/toxicidade , Flavanonas/química , Glucosídeos/química , Humanos , Mucosa Intestinal/metabolismo , Masculino , Camundongos Endogâmicos C57BL , Simulação de Acoplamento Molecular , Fosforilação , Proteínas de Junções Íntimas/metabolismo
4.
Zhongguo Zhong Yao Za Zhi ; 44(8): 1642-1647, 2019 Apr.
Artigo em Chinês | MEDLINE | ID: mdl-31090329

RESUMO

This paper was aimed to investigate the inhibitory effect of aconitine(AC) on angiotensin Ⅱ(Ang Ⅱ)-induced H9 c2 cell hypertrophy and explore its mechanism of action. The model of hypertrophy was induced by Ang Ⅱ(1×10-6 mol·L-1),and cardiomyocytes were incubated with different concentrations of AC. Western blot was used to quantify the protein expression levels of atrial natriuretic peptide(ANP),brain natriuretic peptide(BNP),ß-myosin heavy chain(ß-MHC),and α-smooth muscle actin(α-SMA). Real-time quantitative PCR(qRT-PCR) was used to quantify the mRNA expression levels of cardiac hypertrophic markers ANP,BNP and ß-MHC. In addition,the fluorescence intensity of the F-actin marker,an important component of myofibrils,was detected by using laser confocal microscope. AC could significantly reverse the increase of total protein content in H9 c2 cells induced by Ang Ⅱ; qRT-PCR results showed that AC could significantly inhibit the ANP,BNP and ß-MHC mRNA up-regulation induced by AngⅡ. Western blot results showed that AC could significantly inhibit the ANP,BNP and ß-MHC protein up-regulation induced by AngⅡ. In addition,F-actin expression induced by Ang Ⅱ could be inhibited by AC,and multiple indicators of cardiomyocyte hypertrophy induced by Ang Ⅱ could be down-regulated,indicating that AC may inhibit cardiac hypertrophy by inhibiting the expression of hypertrophic factors,providing new clues for exploring the cardiovascular protection of AC.


Assuntos
Aconitina/farmacologia , Angiotensina II , Miócitos Cardíacos/efeitos dos fármacos , Actinas/metabolismo , Fator Natriurético Atrial/metabolismo , Miosinas Cardíacas/metabolismo , Cardiomegalia , Células Cultivadas , Humanos , Hipertrofia , Cadeias Pesadas de Miosina/metabolismo , Peptídeo Natriurético Encefálico/metabolismo
5.
J Muscle Res Cell Motil ; 30(3-4): 111-23, 2009.
Artigo em Inglês | MEDLINE | ID: mdl-19507043

RESUMO

A rat model of low myocardial blood flow was established to test the hypothesis that post-translational changes to proteins of the thin and thick muscle filaments correlate with decreased cardiac contractility. Following 3 days of low blood flow by constriction of the left anterior descending artery, rat hearts demonstrated a reduction in fractional shortening at rest and a relative decline in fractional shortening when challenged with high dose versus low dose dobutamine, reflecting reduced energy reserves. Permeabilized fibers from low blood flow hearts demonstrated a decline in maximum force per cross-section and Ca2+ sensitivity as compared to their sham operated counterparts. An examination of sarcomeric proteins by twodimensional gel electrophoresis, mass spectrometry, and phospho-specific antibodies provided evidence for Ser23/24 and Ser43/45 phosphorylation of troponin I (TnI). Total TnI phosphorylation was not different between the groups, but Ser23/24 phosphorylation declined with low blood flow, implying an accompanying increase in phosphorylation at other sites of TnI. Affinity chromatography demonstrated that TnI from low blood flow myocardium had reduced relative affinity to Ca2+ bound troponin C compared to TnI from sham operated hearts, providing a mechanism for reduced Ca2+ sensitivity of force production in low blood flow fibers. These findings suggest that altered TnI function, due to changes in the distribution of phosphorylated sites, is an early contributor to reduced contractility of the heart.


Assuntos
Citoesqueleto de Actina/metabolismo , Cálcio/metabolismo , Coração/fisiologia , Contração Miocárdica/fisiologia , Troponina I/metabolismo , Animais , Velocidade do Fluxo Sanguíneo , Miosinas Cardíacas/metabolismo , Dobutamina/farmacologia , Coração/efeitos dos fármacos , Contração Miocárdica/efeitos dos fármacos , Cadeias Leves de Miosina/metabolismo , Fosforilação , Ratos , Troponina C/metabolismo , Troponina T/metabolismo
6.
PLoS One ; 4(2): e4461, 2009.
Artigo em Inglês | MEDLINE | ID: mdl-19221596

RESUMO

BACKGROUND: Myocardial infarction (MI) due to coronary artery disease remains one of the leading causes of premature death. Replacement of infarcted heart tissue with regenerating myocardium from endogenous progenitor pools or exogenously introduced stem cells remains a therapeutic ideal. Their impracticality mainly lies in their low efficiency in cardiogenic differentiation (CD). Our recent studies with an acute MI animal model have already demonstrated the therapeutic effect of the MeOH extract of Geum japonicum (EGJ), providing clear evidence of myocardial regeneration. METHODS AND FINDINGS: The present study further isolated the active component contained in EGJ using bioassay-guided isolation and investigated its efficacy in the treatment of infarcted heart in animal MI models. We demonstrated that substantial repair of infarcted heart in animal MI models by EGJ can be mimicked by the isolated candidate compound (cardiogenin) in MI animal models. Clear evidence of newly regenerated endogenous mesenchymal stem cells (MSCs) derived cardiomyocytes was observed throughout the infarct zone, accompanied by significantly improved functional performance of the heart. Transplantation of MSCs pretreated with EGJ or cardiogenin into a MI animal model also resulted in substantial regeneration of functional myocardium, implying that the activated MSCs carry all the necessary blueprints for myocardial regeneration. Signaling pathways specific to cell survival, CD identified in embryonic heart induction and angiogenesis were activated in both cardiogenin-treated MSCs and cardiogenin-induced regenerating myocardium. CONCLUSIONS: This study has demonstrated the therapeutic effects of cardiogenin in infarcted heart repair, and identified the associated signalling pathways for effective cardiogenic differentiation of MSCs, cell survival and angiogenesis. These findings should enable new treatment strategies for MI to be developed immediately.


Assuntos
Diferenciação Celular/efeitos dos fármacos , Transplante de Células-Tronco Mesenquimais/métodos , Células-Tronco Mesenquimais , Infarto do Miocárdio/terapia , Extratos Vegetais , Regeneração/efeitos dos fármacos , Saponinas/farmacologia , Saponinas/uso terapêutico , Animais , Proteína Morfogenética Óssea 4/metabolismo , Miosinas Cardíacas/genética , Miosinas Cardíacas/metabolismo , Ecocardiografia , Perfilação da Expressão Gênica , Geum , Humanos , Masculino , Células-Tronco Mesenquimais/citologia , Células-Tronco Mesenquimais/efeitos dos fármacos , Células-Tronco Mesenquimais/fisiologia , Análise em Microsséries , Estrutura Molecular , Contração Miocárdica/efeitos dos fármacos , Miocárdio/citologia , Miocárdio/metabolismo , Miocárdio/patologia , Cadeias Leves de Miosina/genética , Cadeias Leves de Miosina/metabolismo , Extratos Vegetais/farmacologia , Extratos Vegetais/uso terapêutico , Ratos , Ratos Sprague-Dawley , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo , Transdução de Sinais/fisiologia
7.
DNA Seq ; 14(5): 339-50, 2003 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-14756420

RESUMO

A cDNA clone encoding human fast skeletal myosin regulatory light chain (HSRLC) has been isolated and characterized from a fetal muscle cDNA library. The cDNA contains the coding sequence of 170 amino acids (aa) and 58 and 91 nucleotides in the 5' and 3' untranslated regions (UTRs), respectively. HSRLC is encoded by a single copy gene in the human genome and shows a tissue-specific pattern of expression in skeletal muscle. Comparison of derived amino acid sequence of HSRLC with database sequences reveals highly conserved 12 amino acid residues in a putative calcium-binding region. HSRLC is unique among all RLC sequences in having three consecutive potential phosphorylatable serine residues. The Cys-129 of HSRLC corresponds to the critical Gly-117 of scallop RLC that is essential for its regulatory function. The clusters of hydrophobic residues that are believed to stabilize the binding of NH2-terminal of RLC with myosin heavy chain show high sequence conservation in RLCs. Besides identifying specific targets for functional studies of HSRLC by mutagenesis, the results support the concept of an ancestral gene from which the RLC genes have evolved.


Assuntos
Miosinas Cardíacas/metabolismo , DNA Complementar/isolamento & purificação , Evolução Molecular , Músculo Esquelético/metabolismo , Cadeias Leves de Miosina/metabolismo , Regiões 3' não Traduzidas/fisiologia , Sequência de Aminoácidos , Miosinas Cardíacas/genética , Clonagem Molecular , Sequência Conservada , DNA Complementar/genética , Expressão Gênica , Humanos , Dados de Sequência Molecular , Cadeias Leves de Miosina/genética , Especificidade de Órgãos , Isoformas de Proteínas , Alinhamento de Sequência , Homologia de Sequência de Aminoácidos
8.
J Biol Chem ; 277(25): 22734-42, 2002 Jun 21.
Artigo em Inglês | MEDLINE | ID: mdl-11932254

RESUMO

Activation of G(q)-coupled alpha(1)-adrenergic receptors leads to hypertrophic growth of neonatal rat ventricular cardiomyocytes that is associated with increased expression of hypertrophy-related genes, including atrial natriuretic peptide (ANP) and myosin light chain-2 (MLC), as well as increased ribosome synthesis. The role of inositol phosphates in signaling pathways involved in these changes in gene expression was examined by overexpressing inositol phosphate-metabolizing enzymes and determining effects on ANP, MLC, and 45 S ribosomal gene expression following co-transfection of appropriate reporter gene constructs. Overexpression of enzymes that metabolize inositol 1,4,5-trisphosphate did not reduce ANP or MLC responses, but overexpression of the enzyme primarily responsible for metabolism of inositol 4,5-bisphosphate (Ins(1,4)P(2)), inositol polyphosphate 1-phosphatase (INPP), reduced ANP and MLC responses associated with alpha(1)-adrenergic receptor-mediated hypertrophy. Similarly overexpressed INPP reduced ANP and MLC responses associated with contraction-induced hypertrophy. In addition, overexpression of INPP reduced the increase in ribosomal DNA transcription associated with both hypertrophic models. Hypertrophied cells from both cell models as well as ventricular tissue from mouse hearts hypertrophied by pressure overload in vivo contained heightened levels of Ins(1,4)P(2), suggesting reduced INPP activity in three different models of hypertrophy. These studies provide evidence for an involvement of Ins(1,4)P(2) in hypertrophic signaling pathways in ventricular myocytes.


Assuntos
Hipertrofia/tratamento farmacológico , Monoéster Fosfórico Hidrolases/química , Monoéster Fosfórico Hidrolases/farmacologia , Animais , Fator Natriurético Atrial/metabolismo , Western Blotting , Células CHO , Miosinas Cardíacas/metabolismo , Células Cultivadas , Clonagem Molecular , Cricetinae , DNA Complementar/metabolismo , DNA Ribossômico/metabolismo , Eletroforese em Gel de Poliacrilamida , Biblioteca Gênica , Genes Reporter , Humanos , Fosfatos de Inositol/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Modelos Químicos , Miocárdio/metabolismo , Miocárdio/patologia , Cadeias Leves de Miosina/metabolismo , Regiões Promotoras Genéticas , Ligação Proteica , Proteína Quinase C/metabolismo , Ratos , Transdução de Sinais , Fatores de Tempo , Transcrição Gênica , Transfecção
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA