Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 110
Filtrar
Mais filtros

Base de dados
País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
Proc Natl Acad Sci U S A ; 120(12): e2222005120, 2023 03 21.
Artigo em Inglês | MEDLINE | ID: mdl-36913580

RESUMO

Cardiac myosin binding protein-C (cMyBP-C) is a thick filament-associated regulatory protein frequently found mutated in patients suffering from hypertrophic cardiomyopathy (HCM). Recent in vitro experiments have highlighted the functional significance of its N-terminal region (NcMyBP-C) for heart muscle contraction, reporting regulatory interactions with both thick and thin filaments. To better understand the interactions of cMyBP-C in its native sarcomere environment, in situ Foerster resonance energy transfer-fluorescence lifetime imaging (FRET-FLIM) assays were developed to determine the spatial relationship between the NcMyBP-C and the thick and thin filaments in isolated neonatal rat cardiomyocytes (NRCs). In vitro studies showed that ligation of genetically encoded fluorophores to NcMyBP-C had no or little effect on its binding to thick and thin filament proteins. Using this assay, FRET between mTFP conjugated to NcMyBP-C and Phalloidin-iFluor 514 labeling the actin filaments in NRCs was detected by time-domain FLIM. The measured FRET efficiencies were intermediate between those observed when the donor was attached to the cardiac myosin regulatory light chain in the thick filaments and troponin T in the thin filaments. These results are consistent with the coexistence of multiple conformations of cMyBP-C, some with their N-terminal domains binding to the thin filament and others binding to the thick filament, supporting the hypothesis that the dynamic interchange between these conformations mediates interfilament signaling in the regulation of contractility. Moreover, stimulation of NRCs with ß-adrenergic agonists reduces FRET between NcMyBP-C and actin-bound Phalloidin, suggesting that cMyBP-C phosphorylation reduces its interaction with the thin filament.


Assuntos
Miocárdio , Miócitos Cardíacos , Ratos , Animais , Miócitos Cardíacos/metabolismo , Miocárdio/metabolismo , Transferência Ressonante de Energia de Fluorescência , Faloidina/metabolismo , Cadeias Leves de Miosina/metabolismo
2.
J Biol Chem ; 300(1): 105511, 2024 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-38042491

RESUMO

Myosin binding protein-C (MyBP-C) is a multidomain protein that regulates muscle contraction. Mutations in MYBPC3, the gene encoding for the cardiac variant (henceforth called cMyBP-C), are amongst the most frequent causes of hypertrophic cardiomyopathy. Most mutations lead to a truncated version of cMyBP-C, which is most likely unstable. However, missense mutations have also been reported, which tend to cluster in the central domains of the cMyBP-C molecule. This suggests that these central domains are more than just a passive spacer between the better characterized N- and C-terminal domains. Here, we investigated the potential impact of four different missense mutations, E542Q, G596R, N755K, and R820Q, which are spread over the domains C3 to C6, on the function of MyBP-C on both the isolated protein level and in cardiomyocytes in vitro. Effect on domain stability, interaction with thin filaments, binding to myosin, and subcellular localization behavior were assessed. Our studies show that these missense mutations result in slightly different phenotypes at the molecular level, which are mutation specific. The expected functional readout of each mutation provides a valid explanation for why cMyBP-C fails to work as a brake in the regulation of muscle contraction, which eventually results in a hypertrophic cardiomyopathy phenotype. We conclude that missense mutations in cMyBP-C must be evaluated in context of their domain localization, their effect on interaction with thin filaments and myosin, and their effect on protein stability to explain how they lead to disease.


Assuntos
Cardiomiopatia Hipertrófica , Proteínas de Transporte , Mutação de Sentido Incorreto , Humanos , Cardiomiopatia Hipertrófica/genética , Proteínas de Transporte/genética , Domínios Proteicos/genética , Estabilidade Proteica
3.
J Biol Chem ; 300(5): 107254, 2024 May.
Artigo em Inglês | MEDLINE | ID: mdl-38569934

RESUMO

Nesprins comprise a family of multi-isomeric scaffolding proteins, forming the linker of nucleoskeleton-and-cytoskeleton complex with lamin A/C, emerin and SUN1/2 at the nuclear envelope. Mutations in nesprin-1/-2 are associated with Emery-Dreifuss muscular dystrophy (EDMD) with conduction defects and dilated cardiomyopathy (DCM). We have previously observed sarcomeric staining of nesprin-1/-2 in cardiac and skeletal muscle, but nesprin function in this compartment remains unknown. In this study, we show that specific nesprin-2 isoforms are highly expressed in cardiac muscle and localize to the Z-disc and I band of the sarcomere. Expression of GFP-tagged nesprin-2 giant spectrin repeats 52 to 53, localized to the sarcomere of neonatal rat cardiomyocytes. Yeast two-hybrid screening of a cardiac muscle cDNA library identified telethonin and four-and-half LIM domain (FHL)-2 as potential nesprin-2 binding partners. GST pull-down and immunoprecipitation confirmed the individual interactions between nesprin-2/telethonin and nesprin-2/FHL-2, and showed that nesprin-2 and telethonin binding was dependent on telethonin phosphorylation status. Importantly, the interactions between these binding partners were impaired by mutations in nesprin-2, telethonin, and FHL-2 identified in EDMD with DCM and hypertrophic cardiomyopathy patients. These data suggest that nesprin-2 is a novel sarcomeric scaffold protein that may potentially participate in the maintenance and/or regulation of sarcomeric organization and function.


Assuntos
Conectina , Proteínas com Domínio LIM , Proteínas Musculares , Miócitos Cardíacos , Proteínas do Tecido Nervoso , Proteínas Nucleares , Sarcômeros , Animais , Humanos , Camundongos , Ratos , Conectina/metabolismo , Conectina/genética , Proteínas do Citoesqueleto/metabolismo , Proteínas do Citoesqueleto/genética , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Peptídeos e Proteínas de Sinalização Intracelular/genética , Proteínas com Domínio LIM/metabolismo , Proteínas com Domínio LIM/genética , Proteínas com Homeodomínio LIM , Proteínas dos Microfilamentos/metabolismo , Proteínas dos Microfilamentos/genética , Proteínas Musculares/metabolismo , Proteínas Musculares/genética , Miócitos Cardíacos/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Proteínas do Tecido Nervoso/genética , Proteínas Nucleares/metabolismo , Proteínas Nucleares/genética , Ligação Proteica , Sarcômeros/metabolismo , Fatores de Transcrição
4.
Philos Trans A Math Phys Eng Sci ; 380(2220): 20210193, 2022 Apr 04.
Artigo em Inglês | MEDLINE | ID: mdl-35152759

RESUMO

The need for acquiring at least three images to reconstruct an optical section of a sample limits the acquisition rate in structured illumination microscopy (SIM) for optical sectioning. In polarized illumination coded structured illumination microscopy (picoSIM) the three individual light patterns are encoded in a single polarized illumination light distribution, enabling the acquisition of the complete SIM data in a single exposure. Here, we describe our experimental set-up and show experimental results acquired with sequential and single-shot picoSIM. This article is part of the Theo Murphy meeting issue 'Super-resolution structured illumination microscopy (part 2)'.


Assuntos
Processamento de Imagem Assistida por Computador , Iluminação , Microscopia de Fluorescência
5.
J Biol Chem ; 295(45): 15342-15365, 2020 11 06.
Artigo em Inglês | MEDLINE | ID: mdl-32868295

RESUMO

The contraction and relaxation of the heart is controlled by stimulation of the ß1-adrenoreceptor (AR) signaling cascade, which leads to activation of cAMP-dependent protein kinase (PKA) and subsequent cardiac protein phosphorylation. Phosphorylation is counteracted by the main cardiac protein phosphatases, PP2A and PP1. Both kinase and phosphatases are sensitive to intramolecular disulfide formation in their catalytic subunits that inhibits their activity. Additionally, intermolecular disulfide formation between PKA type I regulatory subunits (PKA-RI) has been described to enhance PKA's affinity for protein kinase A anchoring proteins, which alters its subcellular distribution. Nitroxyl donors have been shown to affect contractility and relaxation, but the mechanistic basis for this effect is unclear. The present study investigates the impact of several nitroxyl donors and the thiol-oxidizing agent diamide on cardiac myocyte protein phosphorylation and oxidation. Although all tested compounds equally induced intermolecular disulfide formation in PKA-RI, only 1-nitrosocyclohexalycetate (NCA) and diamide induced reproducible protein phosphorylation. Phosphorylation occurred independently of ß1-AR activation, but was abolished after pharmacological PKA inhibition and thus potentially attributable to increased PKA activity. NCA treatment of cardiac myocytes induced translocation of PKA and phosphatases to the myofilament compartment as shown by fractionation, immunofluorescence, and proximity ligation assays. Assessment of kinase and phosphatase activity within the myofilament fraction of cardiac myocytes after exposure to NCA revealed activation of PKA and inhibition of phosphatase activity thus explaining the increase in phosphorylation. The data suggest that the NCA-mediated effect on cardiac myocyte protein phosphorylation orchestrates alterations in the kinase/phosphatase balance.


Assuntos
Proteínas Quinases Dependentes de AMP Cíclico/antagonistas & inibidores , Miócitos Cardíacos/efeitos dos fármacos , Oxidantes/farmacologia , Fosfoproteínas Fosfatases/antagonistas & inibidores , Receptores Adrenérgicos beta 1/metabolismo , Transdução de Sinais , Acetatos/farmacologia , Animais , Bovinos , Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , Diamida/farmacologia , Humanos , Masculino , Camundongos , Miócitos Cardíacos/metabolismo , Compostos Nitrosos/farmacologia , Oxirredução , Fosfoproteínas Fosfatases/metabolismo , Fosforilação/efeitos dos fármacos , Coelhos , Ratos , Ratos Wistar , Transdução de Sinais/efeitos dos fármacos
6.
Basic Res Cardiol ; 116(1): 14, 2021 02 26.
Artigo em Inglês | MEDLINE | ID: mdl-33637999

RESUMO

Titin truncating variants are a well-established cause of cardiomyopathy; however, the role of titin missense variants is less well understood. Here we describe the generation of a mouse model to investigate the underlying disease mechanism of a previously reported titin A178D missense variant identified in a family with non-compaction and dilated cardiomyopathy. Heterozygous and homozygous mice carrying the titin A178D missense variant were characterised in vivo by echocardiography. Heterozygous mice had no detectable phenotype at any time point investigated (up to 1 year). By contrast, homozygous mice developed dilated cardiomyopathy from 3 months. Chronic adrenergic stimulation aggravated the phenotype. Targeted transcript profiling revealed induction of the foetal gene programme and hypertrophic signalling pathways in homozygous mice, and these were confirmed at the protein level. Unsupervised proteomics identified downregulation of telethonin and four-and-a-half LIM domain 2, as well as the upregulation of heat shock proteins and myeloid leukaemia factor 1. Loss of telethonin from the cardiac Z-disc was accompanied by proteasomal degradation; however, unfolded telethonin accumulated in the cytoplasm, leading to a proteo-toxic response in the mice.We show that the titin A178D missense variant is pathogenic in homozygous mice, resulting in cardiomyopathy. We also provide evidence of the disease mechanism: because the titin A178D variant abolishes binding of telethonin, this leads to its abnormal cytoplasmic accumulation. Subsequent degradation of telethonin by the proteasome results in proteasomal overload, and activation of a proteo-toxic response. The latter appears to be a driving factor for the cardiomyopathy observed in the mouse model.


Assuntos
Cardiomiopatias/genética , Edição de Genes , Mutação de Sentido Incorreto , Proteínas Quinases/genética , Fatores Etários , Animais , Cardiomiopatias/metabolismo , Cardiomiopatias/fisiopatologia , Conectina/metabolismo , Predisposição Genética para Doença , Heterozigoto , Homozigoto , Camundongos Endogâmicos C57BL , Camundongos Mutantes , Fenótipo , Complexo de Endopeptidases do Proteassoma/metabolismo , Proteínas Quinases/metabolismo , Proteólise , Proteoma , Transcriptoma , Função Ventricular Esquerda
7.
Biochem Biophys Res Commun ; 533(3): 376-382, 2020 12 10.
Artigo em Inglês | MEDLINE | ID: mdl-32962862

RESUMO

Human induced pluripotent stem cell derived cardiomyocytes (hiPSC-CMs) resemble fetal cardiomyocytes and electrical stimulation (ES) has been explored to mature the differentiated cells. Here, we hypothesize that ES applied at the beginning of the differentiation process, triggers both differentiation of the hiPSC-CMs into a specialized conduction system (CS) phenotype and cell maturation. We applied ES for 15 days starting on day 0 of the differentiation process and found an increased expression of transcription factors and proteins associated with the development and function of CS including Irx3, Nkx2.5 and contactin 2, Hcn4 and Scn5a, respectively. We also found activation of intercalated disc proteins (Nrap and ß-catenin). We detected ES-induced CM maturation as indicated by increased Tnni1 and Tnni3 expression. Confocal micrographs showed a shift towards expression of the gap junction protein connexin 40 in ES hiPSC-CM compared to the more dominant expression of connexin 43 in controls. Finally, analysis of functional parameters revealed that ES hiPSC-CMs exhibited faster action potential (AP) depolarization, longer intracellular Ca2+ transients, and slower AP duration at 90% of repolarization, resembling fast conducting fibers. Altogether, we provided evidence that ES during the differentiation of hiPSC to cardiomyocytes lead to development of cardiac conduction-like cells with more mature cytoarchitecture. Thus, hiPSC-CMs exposed to ES during differentiation can be instrumental to develop CS cells for cardiac disease modelling, screening individual drugs on a precison medicine type platform and support the development of novel therapeutics for arrhythmias.


Assuntos
Potenciais de Ação/fisiologia , Cálcio/metabolismo , Células-Tronco Pluripotentes Induzidas/fisiologia , Miócitos Cardíacos/fisiologia , Biomarcadores/metabolismo , Diferenciação Celular , Terapia Baseada em Transplante de Células e Tecidos/métodos , Conexinas/genética , Conexinas/metabolismo , Contactina 2/genética , Contactina 2/metabolismo , Estimulação Elétrica , Expressão Gênica , Sistema de Condução Cardíaco/citologia , Sistema de Condução Cardíaco/fisiologia , Proteína Homeobox Nkx-2.5/genética , Proteína Homeobox Nkx-2.5/metabolismo , Proteínas de Homeodomínio/genética , Proteínas de Homeodomínio/metabolismo , Humanos , Canais Disparados por Nucleotídeos Cíclicos Ativados por Hiperpolarização/genética , Canais Disparados por Nucleotídeos Cíclicos Ativados por Hiperpolarização/metabolismo , Células-Tronco Pluripotentes Induzidas/citologia , Proteínas Musculares/genética , Proteínas Musculares/metabolismo , Miócitos Cardíacos/citologia , Canal de Sódio Disparado por Voltagem NAV1.5/genética , Canal de Sódio Disparado por Voltagem NAV1.5/metabolismo , Canais de Potássio/genética , Canais de Potássio/metabolismo , Cultura Primária de Células , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo , Troponina I/genética , Troponina I/metabolismo , beta Catenina/genética , beta Catenina/metabolismo , Proteína alfa-5 de Junções Comunicantes
8.
PLoS Genet ; 13(10): e1007068, 2017 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-29084269

RESUMO

The coronary vasculature is an essential vessel network providing the blood supply to the heart. Disruptions in coronary blood flow contribute to cardiac disease, a major cause of premature death worldwide. The generation of treatments for cardiovascular disease will be aided by a deeper understanding of the developmental processes that underpin coronary vessel formation. From an ENU mutagenesis screen, we have isolated a mouse mutant displaying embryonic hydrocephalus and cardiac defects (EHC). Positional cloning and candidate gene analysis revealed that the EHC phenotype results from a point mutation in a splice donor site of the Myh10 gene, which encodes NMHC IIB. Complementation testing confirmed that the Myh10 mutation causes the EHC phenotype. Characterisation of the EHC cardiac defects revealed abnormalities in myocardial development, consistent with observations from previously generated NMHC IIB null mouse lines. Analysis of the EHC mutant hearts also identified defects in the formation of the coronary vasculature. We attribute the coronary vessel abnormalities to defective epicardial cell function, as the EHC epicardium displays an abnormal cell morphology, reduced capacity to undergo epithelial-mesenchymal transition (EMT), and impaired migration of epicardial-derived cells (EPDCs) into the myocardium. Our studies on the EHC mutant demonstrate a requirement for NMHC IIB in epicardial function and coronary vessel formation, highlighting the importance of this protein in cardiac development and ultimately, embryonic survival.


Assuntos
Vasos Coronários/crescimento & desenvolvimento , Desenvolvimento Embrionário/genética , Cadeias Pesadas de Miosina/genética , Miosina não Muscular Tipo IIB/genética , Pericárdio/crescimento & desenvolvimento , Animais , Diferenciação Celular/genética , Vasos Coronários/metabolismo , Embrião de Mamíferos , Transição Epitelial-Mesenquimal/genética , Humanos , Hidrocefalia/genética , Hidrocefalia/metabolismo , Hidrocefalia/patologia , Camundongos , Camundongos Knockout , Mutação , Miocárdio/metabolismo , Pericárdio/metabolismo
9.
J Mol Cell Cardiol ; 128: 212-226, 2019 03.
Artigo em Inglês | MEDLINE | ID: mdl-30742812

RESUMO

The protein kinase C (PKC) and closely related protein kinase N (PKN) families of serine/threonine protein kinases play crucial cellular roles. Both kinases belong to the AGC subfamily of protein kinases that also include the cAMP dependent protein kinase (PKA), protein kinase B (PKB/AKT), protein kinase G (PKG) and the ribosomal protein S6 kinase (S6K). Involvement of PKC family members in heart disease has been well documented over the years, as their activity and levels are mis-regulated in several pathological heart conditions, such as ischemia, diabetic cardiomyopathy, as well as hypertrophic or dilated cardiomyopathy. This review focuses on the regulation of PKCs and PKNs in different pathological heart conditions and on the influences that PKC/PKN activation has on several physiological processes. In addition, we discuss mechanisms by which PKCs and the closely related PKNs are activated and turned-off in hearts, how they regulate cardiac specific downstream targets and pathways, and how their inhibition by small molecules is explored as new therapeutic target to treat cardiomyopathies and heart failure.


Assuntos
Cardiopatias/genética , Miocárdio/enzimologia , Proteína Quinase C/genética , Proteínas Quinases Dependentes de AMP Cíclico/genética , Cardiopatias/enzimologia , Cardiopatias/patologia , Humanos , Miocárdio/patologia , Proteínas Proto-Oncogênicas c-akt/genética , Transdução de Sinais/genética
10.
J Mol Cell Cardiol ; 121: 287-296, 2018 08.
Artigo em Inglês | MEDLINE | ID: mdl-30048712

RESUMO

Cysteine and glycine rich protein 3 (CSRP3) encodes Muscle LIM Protein (MLP), a well-established disease gene for Hypertrophic Cardiomyopathy (HCM). MLP, in contrast to the proteins encoded by the other recognised HCM disease genes, is non-sarcomeric, and has important signalling functions in cardiomyocytes. To gain insight into the disease mechanisms involved, we generated a knock-in mouse (KI) model, carrying the well documented HCM-causing CSRP3 mutation C58G. In vivo phenotyping of homozygous KI/KI mice revealed a robust cardiomyopathy phenotype with diastolic and systolic left ventricular dysfunction, which was supported by increased heart weight measurements. Transcriptome analysis by RNA-seq identified activation of pro-fibrotic signalling, induction of the fetal gene programme and activation of markers of hypertrophic signalling in these hearts. Further ex vivo analyses validated the activation of these pathways at transcript and protein level. Intriguingly, the abundance of MLP decreased in KI/KI mice by 80% and in KI/+ mice by 50%. Protein depletion was also observed in cellular studies for two further HCM-causing CSRP3 mutations (L44P and S54R/E55G). We show that MLP depletion is caused by proteasome action. Moreover, MLP C58G interacts with Bag3 and results in a proteotoxic response in the homozygous knock-in mice, as shown by induction of Bag3 and associated heat shock proteins. In conclusion, the newly generated mouse model provides insights into the underlying disease mechanisms of cardiomyopathy caused by mutations in the non-sarcomeric protein MLP. Furthermore, our cellular experiments suggest that protein depletion and proteasomal overload also play a role in other HCM-causing CSPR3 mutations that we investigated, indicating that reduced levels of functional MLP may be a common mechanism for HCM-causing CSPR3 mutations.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/genética , Proteínas Reguladoras de Apoptose/genética , Cardiomiopatia Hipertrófica/genética , Coração/fisiopatologia , Proteínas com Domínio LIM/genética , Proteínas Musculares/genética , Animais , Cardiomiopatia Hipertrófica/fisiopatologia , Modelos Animais de Doenças , Técnicas de Introdução de Genes , Humanos , Camundongos , Mutação , Sarcômeros/genética
11.
Am J Pathol ; 187(12): 2645-2658, 2017 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-28935576

RESUMO

Peripartum cardiomyopathy (PPCM) and dilated cardiomyopathy (DCM) show similarities in clinical presentation. However, although DCM patients do not recover and slowly deteriorate further, PPCM patients show either a fast cardiac deterioration or complete recovery. The aim of this study was to assess if underlying cellular changes can explain the clinical similarities and differences in the two diseases. We, therefore, assessed sarcomeric protein expression, modification, titin isoform shift, and contractile behavior of cardiomyocytes in heart tissue of PPCM and DCM patients and compared these with nonfailing controls. Heart samples from ischemic heart disease (ISHD) patients served as heart failure control samples. Passive force was only increased in PPCM samples compared with controls, whereas PPCM, DCM, and ISHD samples all showed increased myofilament Ca2+ sensitivity. Length-dependent activation was significantly impaired in PPCM compared with controls, no impairment was observed in ISHD samples, and DCM samples showed an intermediate response. Contractile impairments were caused by impaired protein kinase A (PKA)-mediated phosphorylation because exogenous PKA restored all parameters to control levels. Although DCM samples showed reexpression of EH-myomesin, an isoform usually only expressed in the heart before birth, PPCM and ISHD did not. The lack of EH-myomesin, combined with low PKA-mediated phosphorylation of myofilament proteins and increased compliant titin isoform, may explain the increase in passive force and blunted length-dependent activation of myofilaments in PPCM samples.


Assuntos
Cardiomiopatias/fisiopatologia , Cardiomiopatia Dilatada/fisiopatologia , Miócitos Cardíacos/patologia , Miofibrilas/patologia , Período Periparto , Adulto , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Isquemia Miocárdica/fisiopatologia , Miócitos Cardíacos/metabolismo , Miofibrilas/metabolismo , Gravidez
12.
J Mol Cell Cardiol ; 106: 1-13, 2017 05.
Artigo em Inglês | MEDLINE | ID: mdl-28359939

RESUMO

Tropomyosin 1 (TPM1) is an essential sarcomeric component, stabilising the thin filament and facilitating actin's interaction with myosin. A number of sarcomeric proteins, such as alpha myosin heavy chain, play crucial roles in cardiac development. Mutations in these genes have been linked to congenital heart defects (CHDs), occurring in approximately 1 in 145 live births. To date, TPM1 has not been associated with isolated CHDs. Analysis of 380 CHD cases revealed three novel mutations in the TPM1 gene; IVS1+2T>C, I130V, S229F and a polyadenylation signal site variant GATAAA/AATAAA. Analysis of IVS1+2T>C revealed aberrant pre-mRNA splicing. In addition, abnormal structural properties were found in hearts transfected with TPM1 carrying I130V and S229F mutations. Phenotypic analysis of TPM1 morpholino-treated embryos revealed roles for TPM1 in cardiac looping, atrial septation and ventricular trabeculae formation and increased apoptosis was seen within the heart. In addition, sarcomere assembly was affected and altered action potentials were exhibited. This study demonstrated that sarcomeric TPM1 plays vital roles in cardiogenesis and is a suitable candidate gene for screening individuals with isolated CHDs.


Assuntos
Cardiopatias Congênitas/genética , Coração/crescimento & desenvolvimento , Cadeias Pesadas de Miosina/genética , Tropomiosina/genética , Actinas/genética , Feminino , Coração/fisiopatologia , Cardiopatias Congênitas/patologia , Ventrículos do Coração/crescimento & desenvolvimento , Ventrículos do Coração/patologia , Humanos , Masculino , Mutação/genética , Fenótipo , Precursores de RNA/genética , Splicing de RNA/genética , Sarcômeros/genética
13.
Biochim Biophys Acta ; 1863(7 Pt B): 1857-63, 2016 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-26577135

RESUMO

Cells that constitute fully differentiated tissues are characterised by an architecture that makes them perfectly suited for the job they have to do. This is especially obvious for cardiomyocytes, which have an extremely regular shape and display a paracrystalline arrangement of their cytoplasmic components. This article will focus on the two major cytoskeletal multiprotein complexes that are found in cardiomyocytes, the myofibrils, which are responsible for contraction and the intercalated disc, which mediates mechanical and electrochemical contact between individual cardiomyocytes. Recent studies have revealed that these two sites are also crucial in sensing excessive mechanical strain. Signalling processes will be triggered that## lead to changes in gene expression and eventually lead to an altered cardiac cytoarchitecture in the diseased heart, which results in a compromised function. Thus, understanding these changes and the signals that lead to them is crucial to design treatment strategies that can attenuate these processes. This article is part of a Special Issue entitled: Cardiomyocyte Biology: Integration of Developmental and Environmental Cues in the Heart edited by Marcus Schaub and Hughes Abriel.


Assuntos
Comunicação Celular , Coração/anatomia & histologia , Coração/fisiologia , Mecanotransdução Celular , Miocárdio/citologia , Miocárdio/metabolismo , Miócitos Cardíacos/metabolismo , Miofibrilas/metabolismo , Animais , Cardiomiopatia Dilatada/genética , Cardiomiopatia Dilatada/metabolismo , Cardiomiopatia Dilatada/fisiopatologia , Regulação da Expressão Gênica , Humanos , Potenciais da Membrana , Contração Miocárdica , Estresse Mecânico
14.
J Muscle Res Cell Motil ; 37(3): 55-69, 2016 06.
Artigo em Inglês | MEDLINE | ID: mdl-27329158

RESUMO

In cardiomyocytes columns of intermyofibrillar mitochondria run up to the intercalated disc (ID); half are collinear with those in the neighbouring cell, suggesting coordinated addition of sarcomeres and mitochondria both within and between cells during cardiomyocyte growth. Recent evidence for an association between sarcoplasmic reticulum (SR) and mitochondria indicates that the SR may be an intermediary in this coordinated behaviour. For this reason we have investigated the arrangement of SR and t tubules with respect to mitochondria and myofibrils, particularly at the ID. In the body of the cardiomyocyte the mitochondrial columns are frequently intersected by transverse tubules. In addition, we find that a majority of axial tubules are sandwiched between mitochondria and myofibril. No tubules are found at the ID. SR coats mitochondrial columns and fibrils throughout their length and reaches towards the peaks of the ID membrane where it attaches in the form of junctional (j)SR. These peripheral ID couplings are often situated between mitochondria and ID membrane, suggesting an SR connection between the two. In dilated cardiomyopathy (DCM) the mitochondria are somewhat disordered and clumped. In a mouse model for DCM, the muscle LIM protein KO, we find that there is a lack of mitochondria near the ID, suggesting the uncoupling of the myofibril/mitochondria organisation during growth. SR still coats the fibrils and reaches the ID folds in a jSR coupling. Unlike in control tissue, however, loops and long fingers of ID membrane penetrate into the proximal sarcomere suggesting a possible intermediary state in cardiomyocyte growth.


Assuntos
Cardiomiopatia Dilatada/diagnóstico , Mitocôndrias/fisiologia , Miócitos Cardíacos/fisiologia , Miofibrilas/fisiologia , Animais , Cardiomiopatia Dilatada/fisiopatologia , Comunicação Celular , Humanos , Junções Intercelulares , Camundongos , Retículo Sarcoplasmático
15.
Histochem Cell Biol ; 144(1): 21-38, 2015 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-25896210

RESUMO

The important role of unconventional myosin VI (MVI) in skeletal and cardiac muscle has been recently postulated (Karolczak et al. in Histochem Cell Biol 139:873-885, 2013). Here, we addressed for the first time a role for this unique myosin motor in myogenic cells as well as during their differentiation into myotubes. During myoblast differentiation, the isoform expression pattern of MVI and its subcellular localization underwent changes. In undifferentiated myoblasts, MVI-stained puncti were seen throughout the cytoplasm and were in close proximity to actin filaments, Golgi apparatus, vinculin-, and talin-rich focal adhesion as well as endoplasmic reticulum. Colocalization of MVI with endoplasmic reticulum was enhanced during myotube formation, and differentiation-dependent association was also seen in sarcoplasmic reticulum of neonatal rat cardiomyocytes (NRCs). Moreover, we observed enrichment of MVI in myotube regions containing acetylcholine receptor-rich clusters, suggesting its involvement in the organization of the muscle postsynaptic machinery. Overexpression of the H246R MVI mutant (associated with hypertrophic cardiomyopathy) in myoblasts and NRCs caused the formation of abnormally large intracellular vesicles. MVI knockdown caused changes in myoblast morphology and inhibition of their migration. On the subcellular level, MVI-depleted myoblasts exhibited aberrations in the organization of actin cytoskeleton and adhesive structures as well as in integrity of Golgi apparatus and endoplasmic reticulum. Also, MVI depletion or overexpression of H246R mutant caused the formation of significantly wider or aberrant myotubes, respectively, indicative of involvement of MVI in myoblast differentiation. The presented results suggest an important role for MVI in myogenic cells and possibly in myoblast differentiation.


Assuntos
Desenvolvimento Muscular , Fibras Musculares Esqueléticas/citologia , Fibras Musculares Esqueléticas/metabolismo , Mioblastos/fisiologia , Miócitos Cardíacos/metabolismo , Cadeias Pesadas de Miosina/metabolismo , Citoesqueleto de Actina/ultraestrutura , Animais , Adesão Celular , Diferenciação Celular , Linhagem Celular , Movimento Celular , Forma Celular , Citoplasma/metabolismo , Retículo Endoplasmático/ultraestrutura , Complexo de Golgi/ultraestrutura , Camundongos , Mioblastos/ultraestrutura , Miócitos Cardíacos/ultraestrutura , Cadeias Pesadas de Miosina/química , Ratos , Retículo Sarcoplasmático/metabolismo
16.
Cell Mol Life Sci ; 71(1): 165-81, 2014 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-23708682

RESUMO

Cardiomyocytes grow during heart maturation or disease-related cardiac remodeling. We present evidence that the intercalated disc (ID) is integral to both longitudinal and lateral growth: increases in width are accommodated by lateral extension of the plicate tread regions and increases in length by sarcomere insertion within the ID. At the margin between myofibril and the folded membrane of the ID lies a transitional junction through which the thin filaments from the last sarcomere run to the ID membrane and it has been suggested that this junction acts as a proto Z-disc for sarcomere addition. In support of this hypothesis, we have investigated the ultrastructure of the ID in mouse hearts from control and dilated cardiomyopathy (DCM) models, the MLP-null and a cardiac-specific ß-catenin mutant, cΔex3, as well as in human left ventricle from normal and DCM samples. We find that the ID amplitude can vary tenfold from 0.2 µm up to a maximum of ~2 µm allowing gradual expansion during heart growth. At the greatest amplitude, equivalent to a sarcomere length, A-bands and thick filaments are found within the ID membrane loops together with a Z-disc, which develops at the transitional junction position. Here, also, the tops of the membrane folds, which are rich in αII spectrin, become enlarged and associated with junctional sarcoplasmic reticulum. Systematically larger ID amplitudes are found in DCM samples. Other morphological differences between mouse DCM and normal hearts suggest that sarcomere inclusion is compromised in the diseased hearts.


Assuntos
Miócitos Cardíacos/ultraestrutura , Sarcômeros/ultraestrutura , Animais , Cardiomiopatia Dilatada/metabolismo , Cardiomiopatia Dilatada/patologia , Ventrículos do Coração/metabolismo , Humanos , Proteínas com Domínio LIM/genética , Proteínas com Domínio LIM/metabolismo , Camundongos , Camundongos Knockout , Microscopia Eletrônica , Proteínas Musculares/genética , Proteínas Musculares/metabolismo , Miócitos Cardíacos/metabolismo , Retículo Sarcoplasmático/metabolismo , Espectrina/metabolismo , Tropomiosina/metabolismo , beta Catenina/genética , beta Catenina/metabolismo
17.
Biochem J ; 457(3): 451-61, 2014 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-24219103

RESUMO

PKD (protein kinase D) is a serine/threonine kinase implicated in multiple cardiac roles, including the phosphorylation of the class II HDAC5 (histone deacetylase isoform 5) and thereby de-repression of MEF2 (myocyte enhancer factor 2) transcription factor activity. In the present study we identify FHL1 (four-and-a-half LIM domains protein 1) and FHL2 as novel binding partners for PKD in cardiac myocytes. This was confirmed by pull-down assays using recombinant GST-fused proteins and heterologously or endogenously expressed PKD in adult rat ventricular myocytes or NRVMs (neonatal rat ventricular myocytes) respectively, and by co-immunoprecipitation of FHL1 and FHL2 with GFP-PKD1 fusion protein expressed in NRVMs. In vitro kinase assays showed that neither FHL1 nor FHL2 is a PKD1 substrate. Selective knockdown of FHL1 expression in NRVMs significantly inhibited PKD activation and HDAC5 phosphorylation in response to endothelin 1, but not to the α1-adrenoceptor agonist phenylephrine. In contrast, selective knockdown of FHL2 expression caused a significant reduction in PKD activation and HDAC5 phosphorylation in response to both stimuli. Interestingly, neither intervention affected MEF2 activation by endothelin 1 or phenylephrine. We conclude that FHL1 and FHL2 are novel cardiac PKD partners, which differentially facilitate PKD activation and HDAC5 phosphorylation by distinct neurohormonal stimuli, but are unlikely to regulate MEF2-driven transcriptional reprogramming.


Assuntos
Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Proteínas com Domínio LIM/metabolismo , Proteínas com Homeodomínio LIM/metabolismo , Proteínas Musculares/metabolismo , Miócitos Cardíacos/metabolismo , Proteína Quinase C/metabolismo , Fatores de Transcrição/metabolismo , Animais , Animais Recém-Nascidos , Células Cultivadas , Endotelina-1/metabolismo , Ativação Enzimática , Ventrículos do Coração/citologia , Ventrículos do Coração/metabolismo , Histona Desacetilases/metabolismo , Humanos , Peptídeos e Proteínas de Sinalização Intracelular/antagonistas & inibidores , Peptídeos e Proteínas de Sinalização Intracelular/química , Peptídeos e Proteínas de Sinalização Intracelular/genética , Isoenzimas/genética , Isoenzimas/metabolismo , Proteínas com Domínio LIM/antagonistas & inibidores , Proteínas com Domínio LIM/química , Proteínas com Domínio LIM/genética , Proteínas com Homeodomínio LIM/antagonistas & inibidores , Proteínas com Homeodomínio LIM/química , Proteínas com Homeodomínio LIM/genética , Fatores de Transcrição MEF2/metabolismo , Camundongos , Proteínas Musculares/antagonistas & inibidores , Proteínas Musculares/química , Proteínas Musculares/genética , Miócitos Cardíacos/citologia , Fragmentos de Peptídeos/antagonistas & inibidores , Fragmentos de Peptídeos/química , Fragmentos de Peptídeos/genética , Fragmentos de Peptídeos/metabolismo , Fosforilação , Proteína Quinase C/genética , Processamento de Proteína Pós-Traducional , Ratos , Proteínas Recombinantes de Fusão/química , Proteínas Recombinantes de Fusão/metabolismo , Fatores de Transcrição/antagonistas & inibidores , Fatores de Transcrição/química , Fatores de Transcrição/genética
18.
Development ; 138(18): 3955-66, 2011 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-21862559

RESUMO

The expression and function of embryonic myosin heavy chain (eMYH) has not been investigated within the early developing heart. This is despite the knowledge that other structural proteins, such as alpha and beta myosin heavy chains and cardiac alpha actin, play crucial roles in atrial septal development and cardiac function. Most cases of atrial septal defects and cardiomyopathy are not associated with a known causative gene, suggesting that further analysis into candidate genes is required. Expression studies localised eMYH in the developing chick heart. eMYH knockdown was achieved using morpholinos in a temporal manner and functional studies were carried out using electrical and calcium signalling methodologies. Knockdown in the early embryo led to abnormal atrial septal development and heart enlargement. Intriguingly, action potentials of the eMYH knockdown hearts were abnormal in comparison with the alpha and beta myosin heavy chain knockdowns and controls. Although myofibrillogenesis appeared normal, in knockdown hearts the tissue integrity was affected owing to apparent focal points of myocyte loss and an increase in cell death. An expression profile of human skeletal myosin heavy chain genes suggests that human myosin heavy chain 3 is the functional homologue of the chick eMYH gene. These data provide compelling evidence that eMYH plays a crucial role in important processes in the early developing heart and, hence, is a candidate causative gene for atrial septal defects and cardiomyopathy.


Assuntos
Coração/embriologia , Coração/fisiologia , Cadeias Pesadas de Miosina/genética , Cadeias Pesadas de Miosina/fisiologia , Animais , Animais Geneticamente Modificados , Cardiomiopatia Dilatada/genética , Embrião de Galinha , Embrião de Mamíferos , Técnicas de Silenciamento de Genes , Coração/anatomia & histologia , Cardiopatias Congênitas/genética , Humanos , Camundongos , Morfogênese/genética , Miocárdio/metabolismo , Cadeias Pesadas de Miosina/metabolismo , Organogênese/genética , Organogênese/fisiologia , Sobrevida , Estudos de Validação como Assunto
20.
Analyst ; 139(18): 4449-52, 2014 Sep 21.
Artigo em Inglês | MEDLINE | ID: mdl-25058315

RESUMO

Cell electrospinning has tremendous applicability to a wide range of uses within both the laboratory and clinic. This has directly resulted from the technology's unique ability to immobilize multiple cell types with a wide range of molecules simultaneously within a fiber during the scaffold generation process. The technology has been shown to generate many cell laden complex architectures from true three-dimensional sheets to those multi-core vessels. Although those studies have demonstrated the versatility of this platform biotechnology, we show here for the first time the ability to immobilize primary cardiac myocytes within these fibers in our quest to develop this technology for creating three-dimensional cardiac patches which could be used for repairing, replacing and rejuvenating damaged, diseased and/or ageing cardiac tissues. These advances are unrivalled by any other technology currently available in the regenerative medicine toolbox, and have many interesting ramifications for repairing a damaged heart.


Assuntos
Miócitos Cardíacos/citologia , Engenharia Tecidual/instrumentação , Alicerces Teciduais/química , Animais , Sobrevivência Celular , Células Cultivadas , Técnicas Eletroquímicas/instrumentação , Desenho de Equipamento , Ratos , Medicina Regenerativa
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA