Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 78
Filtrar
Mais filtros

Bases de dados
Tipo de documento
Intervalo de ano de publicação
2.
Circulation ; 147(2): 142-153, 2023 01 10.
Artigo em Inglês | MEDLINE | ID: mdl-36382596

RESUMO

BACKGROUND: Identifying genetic variants that affect the level of cell cycle reentry and establishing the degree of cell cycle progression in those variants could help guide development of therapeutic interventions aimed at effecting cardiac regeneration. We observed that C57Bl6/NCR (B6N) mice have a marked increase in cardiomyocyte S-phase activity after permanent coronary artery ligation compared with infarcted DBA/2J (D2J) mice. METHODS: Cardiomyocyte cell cycle activity after infarction was monitored in D2J, (D2J×B6N)-F1, and (D2J×B6N)-F1×D2J backcross mice by means of bromodeoxyuridine or 5-ethynyl-2'-deoxyuridine incorporation using a nuclear-localized transgenic reporter to identify cardiomyocyte nuclei. Genome-wide quantitative trait locus analysis, fine scale genetic mapping, whole exome sequencing, and RNA sequencing analyses of the backcross mice were performed to identify the gene responsible for the elevated cardiomyocyte S-phase phenotype. RESULTS: (D2J×B6N)-F1 mice exhibited a 14-fold increase in cardiomyocyte S-phase activity in ventricular regions remote from infarct scar compared with D2J mice (0.798±0.09% versus 0.056±0.004%; P<0.001). Quantitative trait locus analysis of (D2J×B6N)-F1×D2J backcross mice revealed that the gene responsible for differential S-phase activity was located on the distal arm of chromosome 3 (logarithm of the odds score=6.38; P<0.001). Additional genetic and molecular analyses identified 3 potential candidates. Of these, Tnni3k (troponin I-interacting kinase) is expressed in B6N hearts but not in D2J hearts. Transgenic expression of TNNI3K in a D2J genetic background results in elevated cardiomyocyte S-phase activity after injury. Cardiomyocyte S-phase activity in both Tnni3k-expressing and Tnni3k-nonexpressing mice results in the formation of polyploid nuclei. CONCLUSIONS: These data indicate that Tnni3k expression increases the level of cardiomyocyte S-phase activity after injury.


Assuntos
Miócitos Cardíacos , Troponina I , Camundongos , Animais , Troponina I/metabolismo , Camundongos Endogâmicos DBA , Miócitos Cardíacos/metabolismo , Ciclo Celular , Proliferação de Células , Proteínas Serina-Treonina Quinases/genética , Proteínas Serina-Treonina Quinases/metabolismo
3.
J Mol Cell Cardiol ; 182: 86-91, 2023 09.
Artigo em Inglês | MEDLINE | ID: mdl-37517369

RESUMO

Although the myocardial renewal rate in the adult mammalian heart is quite low, recent studies have identified genetic variants which can impact the degree of cardiomyocyte cell cycle reentry. Here we use the compound interest law to model the level of regenerative growth over time in mice exhibiting different rates of cardiomyocyte cell cycle reentry following myocardial injury. The modeling suggests that the limited ability of S-phase adult cardiomyocytes to progress through cytokinesis, rather than the ability to reenter the cell cycle per se, is a major contributor to the low levels of intrinsic regenerative growth in the adult myocardium.


Assuntos
Traumatismos Cardíacos , Miócitos Cardíacos , Camundongos , Animais , Miócitos Cardíacos/metabolismo , Miocárdio/metabolismo , Coração , Ciclo Celular , Traumatismos Cardíacos/metabolismo , Citocinese , Proliferação de Células , Mamíferos
4.
Circulation ; 142(20): 1937-1955, 2020 11 17.
Artigo em Inglês | MEDLINE | ID: mdl-32929985

RESUMO

BACKGROUND: Calmodulin mutations are associated with arrhythmia syndromes in humans. Exome sequencing previously identified a de novo mutation in CALM1 resulting in a p.N98S substitution in a patient with sinus bradycardia and stress-induced bidirectional ventricular ectopy. The objectives of the present study were to determine if mice carrying the N98S mutation knocked into Calm1 replicate the human arrhythmia phenotype and to examine arrhythmia mechanisms. METHODS: Mouse lines heterozygous for the Calm1N98S allele (Calm1N98S/+) were generated using CRISPR/Cas9 technology. Adult mutant mice and their wildtype littermates (Calm1+/+) underwent electrocardiographic monitoring. Ventricular de- and repolarization was assessed in isolated hearts using optical voltage mapping. Action potentials and whole-cell currents and [Ca2+]i, as well, were measured in single ventricular myocytes using the patch-clamp technique and fluorescence microscopy, respectively. The microelectrode technique was used for in situ membrane voltage monitoring of ventricular conduction fibers. RESULTS: Two biologically independent knock-in mouse lines heterozygous for the Calm1N98S allele were generated. Calm1N98S/+ mice of either sex and line exhibited sinus bradycardia, QTc interval prolongation, and catecholaminergic bidirectional ventricular tachycardia. Male mutant mice also showed QRS widening. Pharmacological blockade and activation of ß-adrenergic receptors rescued and exacerbated, respectively, the long-QT phenotype of Calm1N98S/+ mice. Optical and electric assessment of membrane potential in isolated hearts and single left ventricular myocytes, respectively, revealed ß-adrenergically induced delay of repolarization. ß-Adrenergic stimulation increased peak density, slowed inactivation, and left-shifted the activation curve of ICa.L significantly more in Calm1N98S/+ versus Calm1+/+ ventricular myocytes, increasing late ICa.L in the former. Rapidly paced Calm1N98S/+ ventricular myocytes showed increased propensity to delayed afterdepolarization-induced triggered activity, whereas in situ His-Purkinje fibers exhibited increased susceptibility for pause-dependent early afterdepolarizations. Epicardial mapping of Calm1N98S/+ hearts showed that both reentry and focal mechanisms contribute to arrhythmogenesis. CONCLUSIONS: Heterozygosity for the Calm1N98S mutation is causative of an arrhythmia syndrome characterized by sinus bradycardia, QRS widening, adrenergically mediated QTc interval prolongation, and bidirectional ventricular tachycardia. ß-Adrenergically induced ICa.L dysregulation contributes to the long-QT phenotype. Pause-dependent early afterdepolarizations and tachycardia-induced delayed afterdepolarizations originating in the His-Purkinje network and ventricular myocytes, respectively, constitute potential sources of arrhythmia in Calm1N98S/+ hearts.


Assuntos
Calmodulina , Ventrículos do Coração/metabolismo , Mutação de Sentido Incorreto , Miócitos Cardíacos/metabolismo , Ramos Subendocárdicos/metabolismo , Síndrome do Nó Sinusal/congênito , Substituição de Aminoácidos , Animais , Calmodulina/genética , Calmodulina/metabolismo , Modelos Animais de Doenças , Ventrículos do Coração/fisiopatologia , Humanos , Masculino , Camundongos , Camundongos Transgênicos , Ramos Subendocárdicos/fisiopatologia , Síndrome do Nó Sinusal/genética , Síndrome do Nó Sinusal/metabolismo , Síndrome do Nó Sinusal/fisiopatologia
5.
J Biol Chem ; 293(47): 18218-18229, 2018 11 23.
Artigo em Inglês | MEDLINE | ID: mdl-30262665

RESUMO

Protein phosphatase 5 (PP5), a serine/threonine phosphatase, has a wide range of biological functions and exhibits elevated expression in tumor cells. We previously reported that pp5-deficient mice have altered ataxia-telangiectasia mutated (ATM)-mediated signaling and function. However, this regulation was likely indirect, as ATM is not a known PP5 substrate. In the current study, we found that pp5-deficient mice are hypersensitive to genotoxic stress. This hypersensitivity was associated with the marked up-regulation of the tumor suppressor tumor protein p53 and its downstream targets cyclin-dependent kinase inhibitor 1A (p21), MDM2 proto-oncogene (MDM2), and phosphatase and tensin homolog (PTEN) in pp5-deficient tissues and cells. These observations suggested that PP5 plays a role in regulating p53 stability and function. Experiments conducted with p53+/-pp5+/- or p53+/-pp5-/- mice revealed that complete loss of PP5 reduces tumorigenesis in the p53+/- mice. Biochemical analyses further revealed that PP5 directly interacts with and dephosphorylates p53 at multiple serine/threonine residues, resulting in inhibition of p53-mediated transcriptional activity. Interestingly, PP5 expression was significantly up-regulated in p53-deficient cells, and further analysis of pp5 promoter activity revealed that p53 strongly represses PP5 transcription. Our results suggest a reciprocal regulatory interplay between PP5 and p53, providing an important feedback mechanism for the cellular response to genotoxic stress.


Assuntos
Proteínas Nucleares/metabolismo , Fosfoproteínas Fosfatases/metabolismo , Proteína Supressora de Tumor p53/metabolismo , Motivos de Aminoácidos , Animais , Inibidor de Quinase Dependente de Ciclina p21/genética , Inibidor de Quinase Dependente de Ciclina p21/metabolismo , Dano ao DNA , Regulação para Baixo , Camundongos , Camundongos Endogâmicos C57BL , Proteínas Nucleares/química , Proteínas Nucleares/genética , Fosfoproteínas Fosfatases/química , Fosfoproteínas Fosfatases/genética , Regiões Promotoras Genéticas , Proteínas Proto-Oncogênicas c-mdm2/genética , Proteínas Proto-Oncogênicas c-mdm2/metabolismo , Proteína Supressora de Tumor p53/química , Proteína Supressora de Tumor p53/genética
6.
Curr Cardiol Rep ; 21(10): 131, 2019 09 16.
Artigo em Inglês | MEDLINE | ID: mdl-31529165

RESUMO

PURPOSE OF REVIEW: Many forms of heart disease result in the essentially irreversible loss of cardiomyocytes. The ability to promote cardiomyocyte renewal may be a promising approach to reverse injury in diseased hearts. The purpose of this review is to describe the impact of cardiomyocyte cell cycle activation on cardiac function and structure in several different models of myocardial disease. RECENT FINDINGS: Transgenic mice expressing cyclin D2 (D2 mice) exhibit sustained cardiomyocyte renewal in the adult heart. Earlier studies demonstrated that D2 mice exhibited progressive myocardial regeneration in experimental models of myocardial infarction, and that cardiac function was normalized to values seen in sham-operated litter mates by 180 days post-injury. D2 mice also exhibited markedly improved atrial structure in a genetic model of atrial fibrosis. More recent studies revealed that D2 mice were remarkably resistant to heart failure induced by chronic elevated afterload as compared with their wild type (WT siblings), with a 6-fold increase in median survival as well as retention of relatively normal cardiac function. Finally, D2 mice exhibited a progressive recovery in cardiac function to normal levels and a concomitant reduction in adverse myocardial remodeling in an anthracycline cardiotoxicity model. The studies reviewed here make a strong case for the potential utility of inducing cardiomyocyte renewal as a means to treat injured hearts. Several challenges which must be met to develop a viable therapeutic intervention based on these observations are discussed.


Assuntos
Ciclo Celular/fisiologia , Traumatismos Cardíacos/terapia , Coração , Infarto do Miocárdio/metabolismo , Miocárdio/metabolismo , Miócitos Cardíacos/metabolismo , Miócitos Cardíacos/patologia , Regeneração/fisiologia , Animais , Doenças Cardiovasculares/metabolismo , Doenças Cardiovasculares/patologia , Morte Celular , Diferenciação Celular , Proliferação de Células , Ciclina D2/genética , Ciclina D2/fisiologia , Modelos Animais de Doenças , Traumatismos Cardíacos/metabolismo , Traumatismos Cardíacos/patologia , Camundongos , Infarto do Miocárdio/patologia
7.
Development ; 140(9): 1946-57, 2013 May.
Artigo em Inglês | MEDLINE | ID: mdl-23571217

RESUMO

Trabeculation and compaction of the embryonic myocardium are morphogenetic events crucial for the formation and function of the ventricular walls. Fkbp1a (FKBP12) is a ubiquitously expressed cis-trans peptidyl-prolyl isomerase. Fkbp1a-deficient mice develop ventricular hypertrabeculation and noncompaction. To determine the physiological function of Fkbp1a in regulating the intercellular and intracellular signaling pathways involved in ventricular trabeculation and compaction, we generated a series of Fkbp1a conditional knockouts. Surprisingly, cardiomyocyte-restricted ablation of Fkbp1a did not give rise to the ventricular developmental defect, whereas endothelial cell-restricted ablation of Fkbp1a recapitulated the ventricular hypertrabeculation and noncompaction observed in Fkbp1a systemically deficient mice, suggesting an important contribution of Fkbp1a within the developing endocardia in regulating the morphogenesis of ventricular trabeculation and compaction. Further analysis demonstrated that Fkbp1a is a novel negative modulator of activated Notch1. Activated Notch1 (N1ICD) was significantly upregulated in Fkbp1a-ablated endothelial cells in vivo and in vitro. Overexpression of Fkbp1a significantly reduced the stability of N1ICD and direct inhibition of Notch signaling significantly reduced hypertrabeculation in Fkbp1a-deficient mice. Our findings suggest that Fkbp1a-mediated regulation of Notch1 plays an important role in intercellular communication between endocardium and myocardium, which is crucial in controlling the formation of the ventricular walls.


Assuntos
Endocárdio/metabolismo , Ventrículos do Coração/patologia , Miocárdio/metabolismo , Receptor Notch1/metabolismo , Proteínas de Ligação a Tacrolimo/metabolismo , Animais , Linhagem da Célula , Células Cultivadas , Embrião de Mamíferos/embriologia , Embrião de Mamíferos/metabolismo , Embrião de Mamíferos/patologia , Desenvolvimento Embrionário , Endocárdio/embriologia , Endocárdio/patologia , Células Endoteliais/metabolismo , Células Endoteliais/patologia , Feminino , Regulação da Expressão Gênica no Desenvolvimento , Células HEK293 , Ventrículos do Coração/embriologia , Ventrículos do Coração/metabolismo , Humanos , Imuno-Histoquímica , Masculino , Camundongos , Camundongos Knockout/embriologia , Camundongos Knockout/metabolismo , Miocárdio/patologia , Crista Neural/metabolismo , Crista Neural/patologia , Fenótipo , Receptor Notch1/genética , Transdução de Sinais , Proteínas de Ligação a Tacrolimo/genética , Transfecção
9.
Mol Cell ; 30(4): 403-14, 2008 May 23.
Artigo em Inglês | MEDLINE | ID: mdl-18498745

RESUMO

Recent genetic studies have documented a pivotal growth-regulatory role played by the Cullin 7 (CUL7) E3 ubiquitin ligase complex containing the Fbw8-substrate-targeting subunit, Skp1, and the ROC1 RING finger protein. In this report, we identified insulin receptor substrate 1 (IRS-1), a critical mediator of the insulin/insulin-like growth factor 1 signaling, as a proteolytic target of the CUL7 E3 ligase in a manner that depends on mammalian target of rapamycin and the p70 S6 kinase activities. Interestingly, while embryonic fibroblasts of Cul7-/- mice were found to accumulate IRS-1 and exhibit increased activation of IRS-1's downstream Akt and MEK/ERK pathways, these null cells grew poorly and displayed phenotypes reminiscent of those associated with oncogene-induced senescence. Taken together, our findings demonstrate a key role for the CUL7 E3 in targeting IRS-1 for degradation, a process that may contribute to the regulation of cellular senescence.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Proteínas Culina/metabolismo , Ubiquitina/metabolismo , Proteínas Adaptadoras de Transdução de Sinal/genética , Animais , Linhagem Celular , Senescência Celular , Proteínas Culina/genética , Ativação Enzimática , MAP Quinases Reguladas por Sinal Extracelular/genética , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Proteínas F-Box/genética , Proteínas F-Box/metabolismo , Humanos , Proteínas Substratos do Receptor de Insulina , Camundongos , Camundongos Knockout , Fenótipo , Proteínas Quinases/genética , Proteínas Quinases/metabolismo , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/metabolismo , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo , Transdução de Sinais/fisiologia , Serina-Treonina Quinases TOR
10.
Biochim Biophys Acta ; 1833(4): 799-803, 2013 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-23142641

RESUMO

Interventions to effect therapeutic cardiomyocyte renewal have received considerable interest of late. Such interventions, if successful, could give rise to myocardial regeneration in diseased hearts. Regenerative interventions fall into two broad categories, namely approaches based on promoting renewal of pre-existing cardiomyocytes and approaches based on cardiomyogenic stem cell activity. The latter category can be further subdivided into approaches promoting differentiation of endogenous cardiomyogenic stem cells, approaches wherein cardiomyogenic stem cells are harvested, amplified or enriched ex vivo, and subsequently engrafted into the heart, and approaches wherein an exogenous stem cell is induced to differentiate in vitro, and the resulting cardiomyocytes are engrafted into the heart. There is disagreement in the literature regarding the degree to which cardiomyocyte renewal occurs in the normal and injured heart, the mechanism(s) by which this occurs, and the degree to which therapeutic interventions can enhance regenerative growth. This review discusses several caveats which are encountered when attempting to measure cardiomyocyte renewal in vivo which likely contribute, at least in part, to the disagreement regarding the levels at which this occurs in normal, injured and treated hearts. This article is part of a Special Issue entitled: Cardiomyocyte biology: Cardiac pathways of differentiation, metabolism and contraction.


Assuntos
Miocárdio/citologia , Miócitos Cardíacos/citologia , Células-Tronco/citologia , Animais , Bromodesoxiuridina , Diferenciação Celular , Núcleo Celular/metabolismo , Núcleo Celular/ultraestrutura , Proliferação de Células , Rastreamento de Células , Genes Reporter , Integrases , Camundongos , Camundongos Transgênicos , Miocárdio/metabolismo , Miócitos Cardíacos/metabolismo , Regeneração , Células-Tronco/metabolismo , beta-Galactosidase
12.
Development ; 138(2): 303-15, 2011 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-21177343

RESUMO

Dishevelled-associated activator of morphogenesis 1 (Daam1), a member of the formin protein family, plays an important role in regulating the actin cytoskeleton via mediation of linear actin assembly. Previous functional studies of Daam1 in lower species suggest its essential role in Drosophila trachea formation and Xenopus gastrulation. However, its in vivo physiological function in mammalian systems is largely unknown. We have generated Daam1-deficient mice via gene-trap technology and found that Daam1 is highly expressed in developing murine organs, including the heart. Daam1-deficient mice exhibit embryonic and neonatal lethality and suffer multiple cardiac defects, including ventricular noncompaction, double outlet right ventricles and ventricular septal defects. In vivo genetic rescue experiments further confirm that the lethality of Daam1-deficient mice results from the inherent cardiac abnormalities. In-depth analyses have revealed that Daam1 is important for regulating filamentous actin assembly and organization, and consequently for cytoskeletal function in cardiomyocytes, which contributes to proper heart morphogenesis. Daam1 is also found to be important for proper cytoskeletal architecture and functionalities in embryonic fibroblasts. Biochemical analyses indicate that Daam1 does not regulate cytoskeletal organization through RhoA, Rac1 or Cdc42. Our study highlights a crucial role for Daam1 in regulating the actin cytoskeleton and tissue morphogenesis.


Assuntos
Coração Fetal/embriologia , Proteínas dos Microfilamentos/fisiologia , Proteínas rho de Ligação ao GTP/fisiologia , Actinas/metabolismo , Animais , Apoptose , Sequência de Bases , Adesão Celular , Diferenciação Celular , Proliferação de Células , Células Cultivadas , Primers do DNA/genética , Feminino , Coração Fetal/anormalidades , Coração Fetal/citologia , Coração Fetal/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Cardiopatias Congênitas/embriologia , Cardiopatias Congênitas/genética , Cardiopatias Congênitas/metabolismo , Camundongos , Camundongos Knockout , Camundongos Transgênicos , Proteínas dos Microfilamentos/deficiência , Proteínas dos Microfilamentos/genética , Proteínas Monoméricas de Ligação ao GTP/metabolismo , Morfogênese/genética , Morfogênese/fisiologia , Miócitos Cardíacos/citologia , Miócitos Cardíacos/metabolismo , Fenótipo , Gravidez , Proteínas rho de Ligação ao GTP/deficiência , Proteínas rho de Ligação ao GTP/genética
13.
Nature ; 453(7194): 524-8, 2008 May 22.
Artigo em Inglês | MEDLINE | ID: mdl-18432194

RESUMO

The functional heart is comprised of distinct mesoderm-derived lineages including cardiomyocytes, endothelial cells and vascular smooth muscle cells. Studies in the mouse embryo and the mouse embryonic stem cell differentiation model have provided evidence indicating that these three lineages develop from a common Flk-1(+) (kinase insert domain protein receptor, also known as Kdr) cardiovascular progenitor that represents one of the earliest stages in mesoderm specification to the cardiovascular lineages. To determine whether a comparable progenitor is present during human cardiogenesis, we analysed the development of the cardiovascular lineages in human embryonic stem cell differentiation cultures. Here we show that after induction with combinations of activin A, bone morphogenetic protein 4 (BMP4), basic fibroblast growth factor (bFGF, also known as FGF2), vascular endothelial growth factor (VEGF, also known as VEGFA) and dickkopf homolog 1 (DKK1) in serum-free media, human embryonic-stem-cell-derived embryoid bodies generate a KDR(low)/C-KIT(CD117)(neg) population that displays cardiac, endothelial and vascular smooth muscle potential in vitro and, after transplantation, in vivo. When plated in monolayer cultures, these KDR(low)/C-KIT(neg) cells differentiate to generate populations consisting of greater than 50% contracting cardiomyocytes. Populations derived from the KDR(low)/C-KIT(neg) fraction give rise to colonies that contain all three lineages when plated in methylcellulose cultures. Results from limiting dilution studies and cell-mixing experiments support the interpretation that these colonies are clones, indicating that they develop from a cardiovascular colony-forming cell. Together, these findings identify a human cardiovascular progenitor that defines one of the earliest stages of human cardiac development.


Assuntos
Células-Tronco Embrionárias/citologia , Células-Tronco Embrionárias/metabolismo , Miócitos Cardíacos/citologia , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/metabolismo , Ativinas/farmacologia , Proteína Morfogenética Óssea 4 , Proteínas Morfogenéticas Ósseas/farmacologia , Diferenciação Celular/efeitos dos fármacos , Linhagem Celular , Linhagem da Célula/efeitos dos fármacos , Células-Tronco Embrionárias/efeitos dos fármacos , Células-Tronco Embrionárias/transplante , Fator 2 de Crescimento de Fibroblastos/farmacologia , Humanos , Peptídeos e Proteínas de Sinalização Intercelular/farmacologia , Miócitos Cardíacos/efeitos dos fármacos , Miócitos Cardíacos/metabolismo , Técnicas de Patch-Clamp , Proteínas Proto-Oncogênicas c-kit/genética , Fator A de Crescimento do Endotélio Vascular/farmacologia , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/deficiência , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/genética
15.
Hum Mol Genet ; 20(23): 4582-96, 2011 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-21880664

RESUMO

Arrhythmogenic right ventricular cardiomyopathy (ARVC) is an inheritable myocardial disorder associated with fibrofatty replacement of myocardium and ventricular arrhythmia. A subset of ARVC is categorized as Naxos disease, which is characterized by ARVC and a cutaneous disorder. A homozygous loss-of-function mutation of the Plakoglobin (Jup) gene, which encodes a major component of the desmosome and the adherens junction, had been identified in Naxos patients, although the underlying mechanism remained elusive. We generated Jup mutant mice by ablating Jup in cardiomyocytes. Jup mutant mice largely recapitulated the clinical manifestation of human ARVC: ventricular dilation and aneurysm, cardiac fibrosis, cardiac dysfunction and spontaneous ventricular arrhythmias. Ultra-structural analyses revealed that desmosomes were absent in Jup mutant myocardia, whereas adherens junctions and gap junctions were preserved. We found that ventricular arrhythmias were associated with progressive cardiomyopathy and fibrosis in Jup mutant hearts. Massive cell death contributed to the cardiomyocyte dropout in Jup mutant hearts. Despite the increase of ß-catenin at adherens junctions in Jup mutant cardiomyoicytes, the Wnt/ß-catenin-mediated signaling was not altered. Transforming growth factor-beta-mediated signaling was found significantly elevated in Jup mutant cardiomyocytes at the early stage of cardiomyopathy, suggesting an important pathogenic pathway for Jup-related ARVC. These findings have provided further insights for the pathogenesis of ARVC and potential therapeutic interventions.


Assuntos
Displasia Arritmogênica Ventricular Direita/patologia , Miócitos Cardíacos/metabolismo , Miócitos Cardíacos/patologia , gama Catenina/deficiência , Animais , Arritmias Cardíacas/complicações , Arritmias Cardíacas/fisiopatologia , Displasia Arritmogênica Ventricular Direita/complicações , Displasia Arritmogênica Ventricular Direita/fisiopatologia , Morte Celular , Desmossomos/metabolismo , Desmossomos/ultraestrutura , Fibrose , Deleção de Genes , Sistema de Condução Cardíaco/patologia , Sistema de Condução Cardíaco/fisiopatologia , Humanos , Camundongos , Camundongos Mutantes , Miocárdio/metabolismo , Miocárdio/patologia , Miócitos Cardíacos/ultraestrutura , Especificidade de Órgãos , Sarcômeros/metabolismo , Sarcômeros/ultraestrutura , Via de Sinalização Wnt , gama Catenina/metabolismo
16.
Circ Res ; 108(9): 1042-52, 2011 Apr 29.
Artigo em Inglês | MEDLINE | ID: mdl-21372286

RESUMO

RATIONALE: FK506 binding protein (FKBP)12 is a known cis-trans peptidyl prolyl isomerase and highly expressed in the heart. Its role in regulating postnatal cardiac function remains largely unknown. METHODS AND RESULTS: We generated FKBP12 overexpressing transgenic (αMyHC-FKBP12) mice and cardiomyocyte-restricted FKBP12 conditional knockout (FKBP12(f/f)/αMyHC-Cre) mice and analyzed their cardiac electrophysiology in vivo and in vitro. A high incidence (38%) of sudden death was found in αMyHC-FKBP12 mice. Surface and ambulatory ECGs documented cardiac conduction defects, which were further confirmed by electric measurements and optical mapping in Langendorff-perfused hearts. αMyHC-FKBP12 hearts had slower action potential upstrokes and longer action potential durations. Whole-cell patch-clamp analyses demonstrated an ≈ 80% reduction in peak density of the tetrodotoxin-resistant, voltage-gated sodium current I(Na) in αMyHC-FKBP12 ventricular cardiomyocytes, a slower recovery of I(Na) from inactivation, shifts of steady-state activation and inactivation curves of I(Na) to more depolarized potentials, and augmentation of late I(Na), suggesting that the arrhythmogenic phenotype of αMyHC-FKBP12 mice is attributable to abnormal I(Na). Ventricular cardiomyocytes isolated from FKBP12(f/f)/αMyHC-Cre hearts showed faster action potential upstrokes and a more than 2-fold increase in peak I(Na) density. Dialysis of exogenous recombinant FKBP12 protein into FKBP12-deficient cardiomyocytes promptly recapitulated alterations in I(Na) seen in αMyHC-FKBP12 myocytes. CONCLUSIONS: FKBP12 is a critical regulator of I(Na) and is important for cardiac arrhythmogenic physiology. FKPB12-mediated dysregulation of I(Na) may underlie clinical arrhythmias associated with FK506 administration.


Assuntos
Arritmias Cardíacas/fisiopatologia , Sistema de Condução Cardíaco/fisiologia , Canais de Sódio/fisiologia , Proteína 1A de Ligação a Tacrolimo/genética , Proteína 1A de Ligação a Tacrolimo/metabolismo , Potenciais de Ação/fisiologia , Animais , Arritmias Cardíacas/genética , Cálcio/metabolismo , Canais de Cálcio Tipo L/fisiologia , Técnicas In Vitro , Integrases/genética , Síndrome do QT Longo/genética , Síndrome do QT Longo/fisiopatologia , Camundongos , Camundongos Knockout , Contração Miocárdica/fisiologia , Miócitos Cardíacos/fisiologia , Técnicas de Patch-Clamp , Canais de Potássio/fisiologia
17.
Circ Res ; 109(7): 758-69, 2011 Sep 16.
Artigo em Inglês | MEDLINE | ID: mdl-21799151

RESUMO

RATIONALE: Telethonin (also known as titin-cap or t-cap) is a 19-kDa Z-disk protein with a unique ß-sheet structure, hypothesized to assemble in a palindromic way with the N-terminal portion of titin and to constitute a signalosome participating in the process of cardiomechanosensing. In addition, a variety of telethonin mutations are associated with the development of several different diseases; however, little is known about the underlying molecular mechanisms and telethonin's in vivo function. OBJECTIVE: Here we aim to investigate the role of telethonin in vivo and to identify molecular mechanisms underlying disease as a result of its mutation. METHODS AND RESULTS: By using a variety of different genetically altered animal models and biophysical experiments we show that contrary to previous views, telethonin is not an indispensable component of the titin-anchoring system, nor is deletion of the gene or cardiac specific overexpression associated with a spontaneous cardiac phenotype. Rather, additional titin-anchorage sites, such as actin-titin cross-links via α-actinin, are sufficient to maintain Z-disk stability despite the loss of telethonin. We demonstrate that a main novel function of telethonin is to modulate the turnover of the proapoptotic tumor suppressor p53 after biomechanical stress in the nuclear compartment, thus linking telethonin, a protein well known to be present at the Z-disk, directly to apoptosis ("mechanoptosis"). In addition, loss of telethonin mRNA and nuclear accumulation of this protein is associated with human heart failure, an effect that may contribute to enhanced rates of apoptosis found in these hearts. CONCLUSIONS: Telethonin knockout mice do not reveal defective heart development or heart function under basal conditions, but develop heart failure following biomechanical stress, owing at least in part to apoptosis of cardiomyocytes, an effect that may also play a role in human heart failure.


Assuntos
Insuficiência Cardíaca/metabolismo , Coração/fisiopatologia , Mecanotransdução Celular , Proteínas Musculares/deficiência , Miocárdio/metabolismo , Adaptação Fisiológica , Animais , Animais Geneticamente Modificados , Apoptose , Fenômenos Biomecânicos , Linhagem Celular Tumoral , Conectina , Modelos Animais de Doenças , Ecocardiografia , Fibrose , Genótipo , Insuficiência Cardíaca/genética , Insuficiência Cardíaca/patologia , Insuficiência Cardíaca/fisiopatologia , Humanos , Camundongos , Camundongos Knockout , Proteínas Musculares/genética , Miocárdio/patologia , Fenótipo , Interferência de RNA , Ratos , Sarcômeros/metabolismo , Estresse Mecânico , Transfecção , Proteína Supressora de Tumor p53/metabolismo
19.
Eur Heart J ; 33(1): 129-37, 2012 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-21849352

RESUMO

AIMS: Although pharmacological interventions that mobilize stem cells and enhance their homing to damaged tissue can limit adverse post-myocardial infarction (MI) remodelling, cardiomyocyte renewal with this approach is limited. While experimental cell cycle induction can promote cardiomyocyte renewal following MI, this process must compete with the more rapid processes of scar formation and adverse remodelling. The current study tested the hypothesis that the combination of enhanced stem cell mobilization/homing and cardiomyocyte cell cycle induction would result in increased myocardial renewal in injured hearts. METHODS AND RESULTS: Myocardial infarction was induced by coronary artery ligation in adult MHC-cycD2 transgenic mice (which exhibit constitutive cardiomyocyte cell cycle activity) and their non-transgenic littermates. Mice were then treated with saline or with granulocyte colony-stimulating factor (G-CSF) plus the dipeptidylpeptidase-IV (DPP-IV) inhibitor Diprotin A (DipA) for 7 days. Infarct thickness and cardiomyocyte number/infarct/section were significantly improved in MHC-cycD2 mice with G-CSF plus DipA treatment when compared with MHC-cycD2 transgene expression or G-CSF plus DipA treatment alone. Echocardiographic analyses revealed that stem cell mobilization/homing and cardiomyocyte cell cycle activation had an additive effect on functional recovery. CONCLUSION: These data strongly suggest that G-CSF plus DPP-IV inhibition, combined with cardiomyocyte cell cycle activation, leads to enhanced myocardial regeneration following MI. The data are also consistent with the notion that altering adverse post-injury remodelling renders the myocardium more permissive for cardiomyocyte repopulation.


Assuntos
Inibidores da Dipeptidil Peptidase IV/farmacologia , Fator Estimulador de Colônias de Granulócitos/farmacologia , Coração/fisiologia , Infarto do Miocárdio/fisiopatologia , Regeneração/fisiologia , Animais , Ciclina D2/metabolismo , Combinação de Medicamentos , Mobilização de Células-Tronco Hematopoéticas/métodos , Camundongos , Camundongos Transgênicos , Miócitos Cardíacos/metabolismo , Distribuição Aleatória , Volume Sistólico/fisiologia
20.
J Biol Chem ; 286(42): 36820-9, 2011 Oct 21.
Artigo em Inglês | MEDLINE | ID: mdl-21890625

RESUMO

Bone morphogenetic protein 10 (BMP10) belongs to the TGFß-superfamily. Previously, we had demonstrated that BMP10 is a key regulator for ventricular chamber formation, growth, and maturation. Ablation of BMP10 leads to hypoplastic ventricular wall formation, and elevated levels of BMP10 are associated with abnormal ventricular trabeculation/compaction and wall maturation. However, the molecular mechanism(s) by which BMP10 regulates ventricle wall growth and maturation is still largely unknown. In this study, we sought to identify the specific transcriptional network that is potentially mediated by BMP10. We analyzed and compared the gene expression profiles between α-myosin heavy chain (αMHC)-BMP10 transgenic hearts and nontransgenic littermate controls using Affymetrix mouse exon arrays. T-box 20 (Tbx20), a cardiac transcription factor, was significantly up-regulated in αMHC-BMP10 transgenic hearts, which was validated by quantitative RT-PCR and in situ hybridization. Ablation of BMP10 reduced Tbx20 expression specifically in the BMP10-expressing region of the developing ventricle. In vitro promoter analysis demonstrated that BMP10 was able to induce Tbx20 promoter activity through a conserved Smad binding site in the Tbx20 promoter proximal region. Furthermore, overexpression of Tbx20 in myocardium led to dilated cardiomyopathy that exhibited ventricular hypertrabeculation and an abnormal muscular septum, which phenocopied genetically modified mice with elevated BMP10 levels. Taken together, our findings demonstrate that the BMP10-Tbx20 signaling cascade is important for ventricular wall development and maturation.


Assuntos
Proteínas Morfogenéticas Ósseas/biossíntese , Regulação da Expressão Gênica no Desenvolvimento/fisiologia , Ventrículos do Coração/embriologia , Transdução de Sinais/fisiologia , Proteínas com Domínio T/metabolismo , Animais , Proteínas Morfogenéticas Ósseas/genética , Cardiomiopatia Dilatada/embriologia , Cardiomiopatia Dilatada/genética , Cardiomiopatia Dilatada/patologia , Comunicação Interventricular/embriologia , Comunicação Interventricular/genética , Comunicação Interventricular/patologia , Ventrículos do Coração/patologia , Camundongos , Camundongos Transgênicos , Cadeias Pesadas de Miosina/genética , Cadeias Pesadas de Miosina/metabolismo , Elementos de Resposta/fisiologia , Proteínas com Domínio T/genética , Miosinas Ventriculares/genética , Miosinas Ventriculares/metabolismo , Septo Interventricular/embriologia , Septo Interventricular/patologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA