Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 25
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Circ Arrhythm Electrophysiol ; 17(4): e012022, 2024 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-38415356

RESUMO

BACKGROUND: Germline HRAS gain-of-function pathogenic variants cause Costello syndrome (CS). During early childhood, 50% of patients develop multifocal atrial tachycardia, a treatment-resistant tachyarrhythmia of unknown pathogenesis. This study investigated how overactive HRAS activity triggers arrhythmogenesis in atrial-like cardiomyocytes (ACMs) derived from human-induced pluripotent stem cells bearing CS-associated HRAS variants. METHODS: HRAS Gly12 mutations were introduced into a human-induced pluripotent stem cells-ACM reporter line. Human-induced pluripotent stem cells were generated from patients with CS exhibiting tachyarrhythmia. Calcium transients and action potentials were assessed in induced pluripotent stem cell-derived ACMs. Automated patch clamping assessed funny currents. HCN inhibitors targeted pacemaker-like activity in mutant ACMs. Transcriptomic data were analyzed via differential gene expression and gene ontology. Immunoblotting evaluated protein expression associated with calcium handling and pacemaker-nodal expression. RESULTS: ACMs harboring HRAS variants displayed higher beating rates compared with healthy controls. The hyperpolarization activated cyclic nucleotide gated potassium channel inhibitor ivabradine and the Nav1.5 blocker flecainide significantly decreased beating rates in mutant ACMs, whereas voltage-gated calcium channel 1.2 blocker verapamil attenuated their irregularity. Electrophysiological assessment revealed an increased number of pacemaker-like cells with elevated funny current densities among mutant ACMs. Mutant ACMs demonstrated elevated gene expression (ie, ISL1, TBX3, TBX18) related to intracellular calcium homeostasis, heart rate, RAS signaling, and induction of pacemaker-nodal-like transcriptional programming. Immunoblotting confirmed increased protein levels for genes of interest and suppressed MAPK (mitogen-activated protein kinase) activity in mutant ACMs. CONCLUSIONS: CS-associated gain-of-function HRASG12 mutations in induced pluripotent stem cells-derived ACMs trigger transcriptional changes associated with enhanced automaticity and arrhythmic activity consistent with multifocal atrial tachycardia. This is the first human-induced pluripotent stem cell model establishing the mechanistic basis for multifocal atrial tachycardia in CS.


Assuntos
Células-Tronco Pluripotentes Induzidas , Miócitos Cardíacos , Humanos , Pré-Escolar , Miócitos Cardíacos/metabolismo , Cálcio/metabolismo , Átrios do Coração/metabolismo , Taquicardia , Canais de Cálcio/metabolismo , Células-Tronco Pluripotentes Induzidas/metabolismo , Potenciais de Ação/fisiologia , Diferenciação Celular , Proteínas Proto-Oncogênicas p21(ras)/genética , Proteínas Proto-Oncogênicas p21(ras)/metabolismo
2.
Matrix Biol ; 121: 41-55, 2023 08.
Artigo em Inglês | MEDLINE | ID: mdl-37217119

RESUMO

To assess the contribution of individual TGF-ß isoforms to aortopathy in Marfan syndrome (MFS), we quantified the survival and phenotypes of mice with a combined fibrillin1 (the gene defective in MFS) hypomorphic mutation and a TGF-ß1, 2, or 3 heterozygous null mutation. The loss of TGF-ß2, and only TGF-ß2, resulted in 80% of the double mutant animals dying earlier, by postnatal day 20, than MFS only mice. Death was not from thoracic aortic rupture, as observed in MFS mice, but was associated with hyperplastic aortic valve leaflets, aortic regurgitation, enlarged aortic root, increased heart weight, and impaired lung alveolar septation. Thus, there appears to be a relationship between loss of fibrillin1 and TGF-ß2 in the postnatal development of the heart, aorta and lungs.


Assuntos
Haploinsuficiência , Síndrome de Marfan , Animais , Camundongos , Aorta , Fibrilina-1/genética , Síndrome de Marfan/genética , Fenótipo , Fator de Crescimento Transformador beta2/genética
3.
J Cardiovasc Dev Dis ; 10(4)2023 Apr 07.
Artigo em Inglês | MEDLINE | ID: mdl-37103040

RESUMO

Adult hearts are characterized by inefficient regeneration after injury, thus, the features that support or prevent cardiomyocyte (CM) proliferation are important to clarify. Diploid CMs are a candidate cell type that may have unique proliferative and regenerative competence, but no molecular markers are yet known that selectively identify all or subpopulations of diploid CMs. Here, using the conduction system expression marker Cntn2-GFP and the conduction system lineage marker Etv1CreERT2, we demonstrate that Purkinje CMs that comprise the adult ventricular conduction system are disproportionately diploid (33%, vs. 4% of bulk ventricular CMs). These, however, represent only a small proportion (3%) of the total diploid CM population. Using EdU incorporation during the first postnatal week, we demonstrate that bulk diploid CMs found in the later heart enter and complete the cell cycle during the neonatal period. In contrast, a significant fraction of conduction CMs persist as diploid cells from fetal life and avoid neonatal cell cycle activity. Despite their high degree of diploidy, the Purkinje lineage had no enhanced competence to support regeneration after adult heart infarction.

4.
J Mol Cell Cardiol ; 169: 28-40, 2022 08.
Artigo em Inglês | MEDLINE | ID: mdl-35533732

RESUMO

A premature truncation of MYBPHL in humans and a loss of Mybphl in mice is associated with dilated cardiomyopathy, atrial and ventricular arrhythmias, and atrial enlargement. MYBPHL encodes myosin binding protein H-like (MyBP-HL). Prior work in mice indirectly identified Mybphl expression in the atria and in small puncta throughout the ventricle. Because of its genetic association with human and mouse cardiac conduction system disease, we evaluated the anatomical localization of MyBP-HL and the consequences of loss of MyBP-HL on conduction system function. Immunofluorescence microscopy of normal adult mouse ventricles identified MyBP-HL-positive ventricular cardiomyocytes that co-localized with the ventricular conduction system marker contactin-2 near the atrioventricular node and in a subset of Purkinje fibers. Mybphl heterozygous ventricles had a marked reduction of MyBP-HL-positive cells compared to controls. Lightsheet microscopy of normal perinatal day 5 mouse hearts showed enrichment of MyBP-HL-positive cells within and immediately adjacent to the contactin-2-positive ventricular conduction system, but this association was not apparent in Mybphl heterozygous hearts. Surface telemetry of Mybphl-null mice revealed atrioventricular block and atrial bigeminy, while intracardiac pacing revealed a shorter atrial relative refractory period and atrial tachycardia. Calcium transient analysis of isolated Mybphl-null atrial cardiomyocytes demonstrated an increased heterogeneity of calcium release and faster rates of calcium release compared to wild type controls. Super-resolution microscopy of Mybphl heterozygous and homozygous null atrial cardiomyocytes showed ryanodine receptor disorganization compared to wild type controls. Abnormal calcium release, shorter atrial refractory period, and atrial dilation seen in Mybphl null, but not wild type control hearts, agree with the observed atrial arrhythmias, bigeminy, and atrial tachycardia, whereas the proximity of MyBP-HL-positive cells with the ventricular conduction system provides insight into how a predominantly atrial expressed gene contributes to ventricular arrhythmias and ventricular dysfunction.


Assuntos
Arritmias Cardíacas , Cálcio , Doença do Sistema de Condução Cardíaco , Proteínas do Citoesqueleto , Animais , Humanos , Camundongos , Arritmias Cardíacas/genética , Cálcio/metabolismo , Doença do Sistema de Condução Cardíaco/genética , Contactinas/metabolismo , Proteínas do Citoesqueleto/genética , Átrios do Coração/metabolismo , Miosinas/metabolismo , Ramos Subendocárdicos , Taquicardia
5.
Epilepsia ; 62(7): 1546-1558, 2021 07.
Artigo em Inglês | MEDLINE | ID: mdl-33982289

RESUMO

OBJECTIVE: Fibroblast growth factor homologous factors (FHFs) are brain and cardiac sodium channel-binding proteins that modulate channel density and inactivation gating. A recurrent de novo gain-of-function missense mutation in the FHF1(FGF12) gene (p.Arg52His) is associated with early infantile epileptic encephalopathy 47 (EIEE47; Online Mendelian Inheritance in Man database 617166). To determine whether the FHF1 missense mutation is sufficient to cause EIEE and to establish an animal model for EIEE47, we sought to engineer this mutation into mice. METHODS: The Arg52His mutation was introduced into fertilized eggs by CRISPR (clustered regularly interspaced short palindromic repeats) editing to generate Fhf1R52H/F+ mice. Spontaneous epileptiform events in Fhf1R52H/+ mice were assessed by cortical electroencephalography (EEG) and video monitoring. Basal heart rhythm and seizure-induced arrhythmia were recorded by electrocardiography. Modulation of cardiac sodium channel inactivation by FHF1BR52H protein was assayed by voltage-clamp recordings of FHF-deficient mouse cardiomyocytes infected with adenoviruses expressing wild-type FHF1B or FHF1BR52H protein. RESULTS: All Fhf1R52H/+ mice experienced seizure or seizurelike episodes with lethal ending between 12 and 26 days of age. EEG recordings in 19-20-day-old mice confirmed sudden unexpected death in epilepsy (SUDEP) as severe tonic seizures immediately preceding loss of brain activity and death. Within 2-53 s after lethal seizure onset, heart rate abruptly declined from 572 ± 16 bpm to 108 ± 15 bpm, suggesting a parasympathetic surge accompanying seizures that may have contributed to SUDEP. Although ectopic overexpression of FHF1BR52H in cardiomyocytes induced a 15-mV depolarizing shift in voltage of steady-state sodium channel inactivation and slowed the rate of channel inactivation, heart rhythm was normal in Fhf1R52H/+ mice prior to seizure. SIGNIFICANCE: The Fhf1 missense mutation p.Arg52His induces epileptic encephalopathy with full penetrance in mice. Both Fhf1 (p.Arg52His) and Scn8a (p.Asn1768Asp) missense mutations enhance sodium channel Nav 1.6 currents and induce SUDEP with bradycardia in mice, suggesting an FHF1/Nav 1.6 functional axis underlying altered brain sodium channel gating in epileptic encephalopathy.


Assuntos
Arritmias Cardíacas/genética , Fatores de Crescimento de Fibroblastos/genética , Espasmos Infantis/genética , Morte Súbita Inesperada na Epilepsia , Idade de Início , Animais , Animais Recém-Nascidos , Arritmias Cardíacas/etiologia , Sistemas CRISPR-Cas , Eletrocardiografia , Eletroencefalografia , Epilepsia Tônico-Clônica/genética , Genótipo , Humanos , Camundongos , Camundongos Transgênicos , Mutação de Sentido Incorreto/genética , Oligonucleotídeos , Convulsões/etiologia , Convulsões/genética , Canais de Sódio Disparados por Voltagem/metabolismo
6.
J Clin Invest ; 130(4): 1713-1727, 2020 04 01.
Artigo em Inglês | MEDLINE | ID: mdl-31910160

RESUMO

Aberrant expression of the cardiac gap junction protein connexin-43 (Cx43) has been suggested as playing a role in the development of cardiac disease in the mdx mouse model of Duchenne muscular dystrophy (DMD); however, a mechanistic understanding of this association is lacking. Here, we identified a reduction of phosphorylation of Cx43 serines S325/S328/S330 in human and mouse DMD hearts. We hypothesized that hypophosphorylation of Cx43 serine-triplet triggers pathological Cx43 redistribution to the lateral sides of cardiomyocytes (remodeling). Therefore, we generated knockin mdx mice in which the Cx43 serine-triplet was replaced with either phospho-mimicking glutamic acids (mdxS3E) or nonphosphorylatable alanines (mdxS3A). The mdxS3E, but not mdxS3A, mice were resistant to Cx43 remodeling, with a corresponding reduction of Cx43 hemichannel activity. MdxS3E cardiomyocytes displayed improved intracellular Ca2+ signaling and a reduction of NADPH oxidase 2 (NOX2)/ROS production. Furthermore, mdxS3E mice were protected against inducible arrhythmias, related lethality, and the development of cardiomyopathy. Inhibition of microtubule polymerization by colchicine reduced both NOX2/ROS and oxidized CaMKII, increased S325/S328/S330 phosphorylation, and prevented Cx43 remodeling in mdx hearts. Together, these results demonstrate a mechanism of dystrophic Cx43 remodeling and suggest that targeting Cx43 may be a therapeutic strategy for preventing heart dysfunction and arrhythmias in DMD patients.


Assuntos
Sinalização do Cálcio , Cardiomiopatias/metabolismo , Conexina 43/metabolismo , Distrofia Muscular de Duchenne/metabolismo , Miocárdio/metabolismo , Miócitos Cardíacos/metabolismo , Animais , Cardiomiopatias/genética , Cardiomiopatias/patologia , Conexina 43/genética , Humanos , Camundongos , Camundongos Endogâmicos mdx , Camundongos Transgênicos , Microtúbulos/genética , Microtúbulos/metabolismo , Distrofia Muscular de Duchenne/genética , Distrofia Muscular de Duchenne/patologia , Miocárdio/patologia , Miócitos Cardíacos/patologia , NADPH Oxidase 2/genética , NADPH Oxidase 2/metabolismo
7.
Stem Cell Reports ; 4(6): 1089-102, 2015 Jun 09.
Artigo em Inglês | MEDLINE | ID: mdl-26028533

RESUMO

Dysfunction of the specialized cardiac conduction system (CCS) is associated with life-threatening arrhythmias. Strategies to derive CCS cells, including rare Purkinje cells (PCs), would facilitate models for mechanistic studies and drug discovery and also provide new cellular materials for regenerative therapies. A high-throughput chemical screen using CCS:lacz and Contactin2:egfp (Cntn2:egfp) reporter embryonic stem cell (ESC) lines was used to discover a small molecule, sodium nitroprusside (SN), that efficiently promotes the generation of cardiac cells that express gene profiles and generate action potentials of PC-like cells. Imaging and mechanistic studies suggest that SN promotes the generation of PCs from cardiac progenitors initially expressing cardiac myosin heavy chain and that it does so by activating cyclic AMP signaling. These findings provide a strategy to derive scalable PCs, along with insight into the ontogeny of CCS development.


Assuntos
AMP Cíclico/metabolismo , Células-Tronco Embrionárias/citologia , Miócitos Cardíacos/metabolismo , Células de Purkinje/metabolismo , Potenciais de Ação/efeitos dos fármacos , Catequina/farmacologia , Diferenciação Celular/efeitos dos fármacos , Linhagem Celular , Humanos , Miócitos Cardíacos/citologia , Cadeias Pesadas de Miosina/metabolismo , Nitroprussiato/farmacologia , Ácido Oleico/farmacologia , Fenótipo , Células de Purkinje/citologia , Reação em Cadeia da Polimerase em Tempo Real , Transdução de Sinais/efeitos dos fármacos , Transcriptoma/efeitos dos fármacos
8.
Biochem J ; 457(3): 441-9, 2014 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-24228978

RESUMO

To date, over 65 mutations in the gene encoding Cx43 (connexin43) have been linked to the autosomal-dominant disease ODDD (oculodentodigital dysplasia). A subset of these patients experience bladder incontinence which could be due to underlying neurogenic deterioration or aberrant myogenic regulation. BSMCs (bladder smooth muscle cells) from wild-type and two Cx43 mutant lines (Cx43(G60S) and Cx43(I130T)) that mimic ODDD exhibit a significant reduction in total Cx43. Dye transfer studies revealed that the G60S mutant was a potent dominant-negative inhibitor of co-expressed Cx43, a property not equally shared by the I130T mutant. BSMCs from both mutant mouse strains were defective in their ability to contract, which is indicative of phenotype changes due to harbouring the Cx43 mutants. Upon stretching, Cx43 levels were significantly elevated in controls and mutants containing BSMCs, but the non-muscle myosin heavy chain A levels were only reduced in cells from control mice. Although the Cx43(G60S) mutant mice showed no difference in voided urine volume or frequency, the Cx43(I130T) mice voided less frequently. Thus, similar to the diversity of morbidities seen in ODDD patients, genetically modified mice also display mutation-specific changes in bladder function. Furthermore, although mutant mice have compromised smooth muscle contraction and response to stretch, overriding bladder defects in Cx43(I130T) mice are likely to be complemented by neurogenic changes.


Assuntos
Conexina 43/metabolismo , Anormalidades Craniofaciais/fisiopatologia , Modelos Animais de Doenças , Anormalidades do Olho/fisiopatologia , Deformidades Congênitas do Pé/fisiopatologia , Músculo Liso/fisiopatologia , Doenças Musculares/etiologia , Sindactilia/fisiopatologia , Anormalidades Dentárias/fisiopatologia , Bexiga Urinaria Neurogênica/etiologia , Bexiga Urinária/fisiopatologia , Substituição de Aminoácidos , Animais , Comunicação Celular , Células Cultivadas , Conexina 43/antagonistas & inibidores , Conexina 43/genética , Anormalidades Craniofaciais/metabolismo , Anormalidades Craniofaciais/patologia , Anormalidades do Olho/metabolismo , Anormalidades do Olho/patologia , Deformidades Congênitas do Pé/metabolismo , Deformidades Congênitas do Pé/patologia , Junções Comunicantes/metabolismo , Masculino , Camundongos , Camundongos Mutantes , Contração Muscular , Músculo Liso/química , Músculo Liso/metabolismo , Músculo Liso/patologia , Doenças Musculares/fisiopatologia , Proteínas Mutantes/antagonistas & inibidores , Proteínas Mutantes/metabolismo , Cadeias Pesadas de Miosina/metabolismo , Sindactilia/metabolismo , Sindactilia/patologia , Anormalidades Dentárias/metabolismo , Anormalidades Dentárias/patologia , Bexiga Urinária/química , Bexiga Urinária/metabolismo , Bexiga Urinária/patologia , Bexiga Urinaria Neurogênica/fisiopatologia , Incontinência Urinária/etiologia
9.
FASEB J ; 27(4): 1460-7, 2013 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-23271053

RESUMO

MicroRNA cluster miR-17-92 has been implicated in cardiovascular development and function, yet its precise mechanisms of action in these contexts are uncertain. This study aimed to investigate the role of miR-17-92 in morphogenesis and function of cardiac and smooth muscle tissues. To do so, a mouse model of conditional overexpression of miR-17-92 in cardiac and smooth muscle tissues was generated. Extensive cardiac functional studies identified a dose-dependent induction of dilated, hypertrophic cardiomyopathy, and arrhythmia inducibility in transgenic animals, which correlated with premature mortality (98.3 ± 42.5 d, P<0.0001). Expression analyses revealed the abundance of Pten transcript, a known miR-17-92 target, to be inversely correlated with miR-17-92 expression levels and heart size. In addition, we demonstrated through 3'-UTR luciferase assays and expression analyses that Connexin43 (Cx43) is a novel direct target of miR-19a/b and its expression is suppressed in transgenic hearts. Taken together, these data demonstrate that dysregulated expression of miR-17-92 during cardiovascular morphogenesis results in a lethal cardiomyopathy, possibly in part through direct repression of Pten and Cx43. This study highlights the importance of miR-17-92 in both normal and pathological functions of the heart, and provides a model that may serve as a useful platform to test novel antiarrhythmic therapeutics.


Assuntos
Arritmias Cardíacas/genética , Cardiomiopatia Hipertrófica/genética , MicroRNAs/genética , Animais , Arritmias Cardíacas/fisiopatologia , Cardiomiopatia Hipertrófica/mortalidade , Cardiomiopatia Hipertrófica/patologia , Cardiomiopatia Hipertrófica/fisiopatologia , Conexina 43/genética , Conexina 43/metabolismo , Modelos Animais de Doenças , Cardiopatias Congênitas/genética , Camundongos , Camundongos Knockout , PTEN Fosfo-Hidrolase/genética , PTEN Fosfo-Hidrolase/metabolismo
10.
Biochem J ; 449(2): 401-13, 2013 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-23075222

RESUMO

Genetically modified mice mimicking ODDD (oculodentodigital dysplasia), a disease characterized by reduced Cx43 (connexin 43)-mediated gap junctional intercellular communication, represent an in vivo model to assess the role of Cx43 in mammary gland development and function. We previously reported that severely compromised Cx43 function delayed mammary gland development and impaired milk ejection in mice that harboured a G60S Cx43 mutant, yet there are no reports of lactation defects in ODDD patients. To address this further, we obtained a second mouse model of ODDD expressing an I130T Cx43 mutant to assess whether a mutant with partial gap junction channel activity would be sufficient to retain mammary gland development and function. The results of the present study show that virgin Cx43I130T/+ mice exhibited a temporary delay in ductal elongation at 4 weeks. In addition, Cx43I130T/+ mice develop smaller mammary glands at parturition due to reduced cell proliferation despite similar overall gland architecture. Distinct from Cx43G60S/+ mice, Cx43I130T/+ mice adequately produce and deliver milk to pups, suggesting that milk ejection is unaffected. Thus the present study suggests that a loss-of-function mutant of Cx43 with partial gap junction channel coupling conductance results in a less severe mammary gland phenotype, which may partially explain the lack of reported lactation defects associated with ODDD patients.


Assuntos
Conexina 43/genética , Glândulas Mamárias Animais/anormalidades , Glândulas Mamárias Animais/metabolismo , Mutação Puntual , Animais , Western Blotting , Linhagem Celular Tumoral , Células Cultivadas , Conexina 43/metabolismo , Anormalidades Craniofaciais/genética , Anormalidades Craniofaciais/metabolismo , Anormalidades Craniofaciais/patologia , Modelos Animais de Doenças , Células Epiteliais/metabolismo , Anormalidades do Olho/genética , Anormalidades do Olho/metabolismo , Anormalidades do Olho/patologia , Feminino , Deformidades Congênitas do Pé/genética , Deformidades Congênitas do Pé/metabolismo , Deformidades Congênitas do Pé/patologia , Junções Comunicantes/metabolismo , Junções Comunicantes/patologia , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Humanos , Lactação/efeitos dos fármacos , Lactação/genética , Masculino , Camundongos , Camundongos Endogâmicos C3H , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Microscopia de Fluorescência , Ocitocina/farmacologia , Gravidez , Índice de Gravidade de Doença , Sindactilia/genética , Sindactilia/metabolismo , Sindactilia/patologia , Anormalidades Dentárias/genética , Anormalidades Dentárias/metabolismo , Anormalidades Dentárias/patologia
11.
Circulation ; 126(9): 1058-66, 2012 Aug 28.
Artigo em Inglês | MEDLINE | ID: mdl-22837163

RESUMO

BACKGROUND: Notch signaling has previously been shown to play an essential role in regulating cell fate decisions and differentiation during cardiogenesis in many systems including Drosophila, Xenopus, and mammals. We hypothesized that Notch may also be involved in directing the progressive lineage restriction of cardiomyocytes into specialized conduction cells. METHODS AND RESULTS: In hearts where Notch signaling is activated within the myocardium from early development onward, Notch promotes a conduction-like phenotype based on ectopic expression of conduction system-specific genes and cell autonomous changes in electrophysiology. With the use of an in vitro assay to activate Notch in newborn cardiomyocytes, we observed global changes in the transcriptome, and in action potential characteristics, consistent with reprogramming to a conduction-like phenotype. CONCLUSIONS: Notch can instruct the differentiation of chamber cardiac progenitors into specialized conduction-like cells. Plasticity remains in late-stage cardiomyocytes, which has potential implications for engineering of specialized cardiovascular tissues.


Assuntos
Nó Atrioventricular/citologia , Regulação da Expressão Gênica no Desenvolvimento , Miocárdio/metabolismo , Miócitos Cardíacos/metabolismo , Receptor Notch1/fisiologia , Potenciais de Ação , Adenoviridae/genética , Animais , Animais Recém-Nascidos , Fatores de Transcrição Hélice-Alça-Hélice Básicos/biossíntese , Fatores de Transcrição Hélice-Alça-Hélice Básicos/genética , Linhagem da Célula , Contactina 2/biossíntese , Contactina 2/genética , Proteína Homeobox Nkx-2.5 , Proteínas de Homeodomínio/biossíntese , Proteínas de Homeodomínio/genética , Camundongos , Miócitos Cardíacos/ultraestrutura , Canal de Sódio Disparado por Voltagem NAV1.5 , Plasticidade Neuronal , Técnicas de Patch-Clamp , Fenótipo , Ramos Subendocárdicos/citologia , Receptor Notch1/genética , Proteínas Recombinantes de Fusão/fisiologia , Transdução de Sinais/fisiologia , Canais de Sódio/biossíntese , Canais de Sódio/genética , Proteínas com Domínio T/biossíntese , Proteínas com Domínio T/genética , Fatores de Transcrição HES-1 , Fatores de Transcrição/biossíntese , Fatores de Transcrição/genética
12.
Proc Natl Acad Sci U S A ; 109(23): 9071-6, 2012 Jun 05.
Artigo em Inglês | MEDLINE | ID: mdl-22611193

RESUMO

Hematopoietic stem cell (HSC) aging has become a concern in chemotherapy of older patients. Humoral and paracrine signals from the bone marrow (BM) hematopoietic microenvironment (HM) control HSC activity during regenerative hematopoiesis. Connexin-43 (Cx43), a connexin constituent of gap junctions (GJs) is expressed in HSCs, down-regulated during differentiation, and postulated to be a self-renewal gene. Our studies, however, reveal that hematopoietic-specific Cx43 deficiency does not result in significant long-term competitive repopulation deficiency. Instead, hematopoietic Cx43 (H-Cx43) deficiency delays hematopoietic recovery after myeloablation with 5-fluorouracil (5-FU). 5-FU-treated H-Cx43-deficient HSC and progenitors (HSC/P) cells display decreased survival and fail to enter the cell cycle to proliferate. Cell cycle quiescence is associated with down-regulation of cyclin D1, up-regulation of the cyclin-dependent kinase inhibitors, p21(cip1.) and p16(INK4a), and Forkhead transcriptional factor 1 (Foxo1), and activation of p38 mitogen-activated protein kinase (MAPK), indicating that H-Cx43-deficient HSCs are prone to senescence. The mechanism of increased senescence in H-Cx43-deficient HSC/P cells depends on their inability to transfer reactive oxygen species (ROS) to the HM, leading to accumulation of ROS within HSCs. In vivo antioxidant administration prevents the defective hematopoietic regeneration, as well as exogenous expression of Cx43 in HSC/P cells. Furthermore, ROS transfer from HSC/P cells to BM stromal cells is also rescued by reexpression of Cx43 in HSC/P. Finally, the deficiency of Cx43 in the HM phenocopies the hematopoietic defect in vivo. These results indicate that Cx43 exerts a protective role and regulates the HSC/P ROS content through ROS transfer to the HM, resulting in HSC protection during stress hematopoietic regeneration.


Assuntos
Senescência Celular/fisiologia , Conexina 43/metabolismo , Regulação da Expressão Gênica/fisiologia , Células-Tronco Hematopoéticas/fisiologia , Espécies Reativas de Oxigênio/metabolismo , Células Estromais/metabolismo , Animais , Conexina 43/deficiência , Citometria de Fluxo , Fluoruracila/farmacologia , Células-Tronco Hematopoéticas/metabolismo , Lentivirus , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Análise em Microsséries , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transdução Genética
13.
Blood ; 119(22): 5144-54, 2012 May 31.
Artigo em Inglês | MEDLINE | ID: mdl-22498741

RESUMO

Connexin-43 (Cx43), a gap junction protein involved in control of cell proliferation, differentiation and migration, has been suggested to have a role in hematopoiesis. Cx43 is highly expressed in osteoblasts and osteogenic progenitors (OB/P). To elucidate the biologic function of Cx43 in the hematopoietic microenvironment (HM) and its influence in hematopoietic stem cell (HSC) activity, we studied the hematopoietic function in an in vivo model of constitutive deficiency of Cx43 in OB/P. The deficiency of Cx43 in OB/P cells does not impair the steady state hematopoiesis, but disrupts the directional trafficking of HSC/progenitors (Ps) between the bone marrow (BM) and peripheral blood (PB). OB/P Cx43 is a crucial positive regulator of transstromal migration and homing of both HSCs and progenitors in an irradiated microenvironment. However, OB/P Cx43 deficiency in nonmyeloablated animals does not result in a homing defect but induces increased endosteal lodging and decreased mobilization of HSC/Ps associated with proliferation and expansion of Cxcl12-secreting mesenchymal/osteolineage cells in the BM HM in vivo. Cx43 controls the cellular content of the BM osteogenic microenvironment and is required for homing of HSC/Ps in myeloablated animals.


Assuntos
Movimento Celular/fisiologia , Conexina 43/metabolismo , Células-Tronco Hematopoéticas/metabolismo , Osteoblastos/metabolismo , Nicho de Células-Tronco/fisiologia , Animais , Quimiocina CXCL12/genética , Quimiocina CXCL12/metabolismo , Conexina 43/genética , Células-Tronco Hematopoéticas/citologia , Camundongos , Camundongos Mutantes , Osteoblastos/citologia
14.
Circ Arrhythm Electrophysiol ; 4(6): 926-35, 2011 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-21984445

RESUMO

Background- The specialized cardiac conduction system (CCS) expresses a unique complement of ion channels that confer a specific electrophysiological profile. ATP-sensitive potassium (K(ATP)) channels in these myocytes have not been systemically investigated. Methods and Results- We recorded K(ATP) channels in isolated CCS myocytes using Cntn2-EGFP reporter mice. The CCS K(ATP) channels were less sensitive to inhibitory cytosolic ATP compared with ventricular channels and more strongly activated by MgADP. They also had a smaller slope conductance. The 2 types of channels had similar intraburst open and closed times, but the CCS K(ATP) channel had a prolonged interburst closed time. CCS K(ATP) channels were strongly activated by diazoxide and less by levcromakalim, whereas the ventricular K(ATP) channel had a reverse pharmacological profile. CCS myocytes express elevated levels of Kir6.1 but reduced Kir6.2 and SUR2A mRNA compared with ventricular myocytes (SUR1 expression was negligible). SUR2B mRNA expression was higher in CCS myocytes relative to SUR2A. Canine Purkinje fibers expressed higher levels of Kir6.1 and SUR2B protein relative to the ventricle. Numeric simulation predicts a high sensitivity of the Purkinje action potential to changes in ATP:ADP ratio. Cardiac conduction time was prolonged by low-flow ischemia in isolated, perfused mouse hearts, which was prevented by glibenclamide. Conclusions- These data imply a differential electrophysiological response (and possible contribution to arrhythmias) of the ventricular CCS to K(ATP) channel opening during periods of ischemia.


Assuntos
Arritmias Cardíacas/metabolismo , Ventrículos do Coração/metabolismo , Canais KATP/metabolismo , Isquemia Miocárdica/metabolismo , Miócitos Cardíacos/metabolismo , Ramos Subendocárdicos/metabolismo , Transportadores de Cassetes de Ligação de ATP/metabolismo , Potenciais de Ação , Trifosfato de Adenosina/metabolismo , Animais , Antiarrítmicos/farmacologia , Arritmias Cardíacas/fisiopatologia , Arritmias Cardíacas/prevenção & controle , Simulação por Computador , Contactina 2/genética , Modelos Animais de Doenças , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Ventrículos do Coração/efeitos dos fármacos , Ventrículos do Coração/fisiopatologia , Canais KATP/efeitos dos fármacos , Canais KATP/genética , Cinética , Camundongos , Camundongos Transgênicos , Modelos Cardiovasculares , Isquemia Miocárdica/fisiopatologia , Miócitos Cardíacos/efeitos dos fármacos , Análise Numérica Assistida por Computador , Técnicas de Patch-Clamp , Perfusão , Bloqueadores dos Canais de Potássio/farmacologia , Canais de Potássio Corretores do Fluxo de Internalização/metabolismo , Ramos Subendocárdicos/efeitos dos fármacos , Ramos Subendocárdicos/fisiopatologia , RNA Mensageiro/metabolismo , Ratos , Ratos Sprague-Dawley , Receptores de Droga/metabolismo , Receptores de Sulfonilureias
15.
Heart Rhythm ; 8(12): 1923-30, 2011 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-21767519

RESUMO

BACKGROUND: Sodium channel α-subunits in ventricular myocytes (VMs) segregate either to the intercalated disc or to lateral membranes, where they associate with region-specific molecules. OBJECTIVE: To determine the functional properties of sodium channels as a function of their location in the cell. METHODS: Local sodium currents were recorded from adult rodent VMs and Purkinje cells by using the cell-attached macropatch configuration. Electrodes were placed either in the cell midsection (M) or at the cell end (area originally occupied by the intercalated disc [ID]). Channels were identified as tetrodotoxin (TTX)-sensitive (TTX-S) or TTX-resistant (TTX-R) by application of 100 nM of TTX. RESULTS: Average peak current amplitude was larger in ID than in M and largest at the site of contact between attached cells. TTX-S channels were found only in the M region of VMs and not in Purkinje myocytes. TTX-R channels were found in both M and ID regions, but their biophysical properties differed depending on recording location. Sodium current in rat VMs was upregulated by tumor necrosis factor-alpha. The magnitude of current increase was largest in the M region, but this difference was abolished by application of 100 nM of TTX. CONCLUSIONS: Our data suggest that (a) a large fraction of TTX-R (likely Na(v)1.5) channels in the M region of VMs are inactivated at normal resting potential, leaving most of the burden of excitation to TTX-R channels in the ID region; (b) cell-cell adhesion increases functional channel density at the ID; and (c) TTX-S (likely non-Na(v)1.5) channels make a minimal contribution to sodium current under control conditions, but they represent a functional reserve that can be upregulated by exogenous factors.


Assuntos
Comunicação Celular , Miócitos Cardíacos/metabolismo , Canais de Sódio/metabolismo , Animais , Camundongos , Miócitos Cardíacos/efeitos dos fármacos , Ratos , Bloqueadores dos Canais de Sódio/farmacologia , Canais de Sódio/efeitos dos fármacos , Tetrodotoxina/farmacologia , Fator de Necrose Tumoral alfa/farmacologia
16.
FASEB J ; 25(8): 2583-91, 2011 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-21507899

RESUMO

Two related ER oxidation 1 (ERO1) proteins, ERO1α and ERO1ß, dynamically regulate the redox environment in the mammalian endoplasmic reticulum (ER). Redox changes in cysteine residues on intralumenal loops of calcium release and reuptake channels have been implicated in altered calcium release and reuptake. These findings led us to hypothesize that altered ERO1 activity may affect cardiac functions that are dependent on intracellular calcium flux. We established mouse lines with loss of function insertion mutations in Ero1l and Ero1lb encoding ERO1α and ERO1ß. The peak amplitude of calcium transients in homozygous Ero1α mutant adult cardiomyocytes was reduced to 42.0 ± 2.2% (n=10, P ≤ 0.01) of values recorded in wild-type cardiomyocytes. Decreased ERO1 activity blunted cardiomyocyte inotropic response to adrenergic stimulation and sensitized mice to adrenergic blockade. Whereas all 12 wild-type mice survived challenge with 4 mg/kg esmolol, 6 of 8 compound Ero1l and Ero1lb mutant mice succumbed to this level of ß adrenergic blockade (P ≤ 0.01). In addition, mice lacking ERO1α were partially protected against progressive heart failure in a transaortic constriction model [at 10 wk postprocedure, fractional shortening was 0.31 ± 0.02 in the mutant (n=20) vs. 0.23 ± 0.03 in the wild type (n=18); P ≤ 0.01]. These findings establish a role for ERO1 in calcium homeostasis and suggest that modifying the lumenal redox environment may affect the progression of heart failure.


Assuntos
Glicoproteínas/metabolismo , Miócitos Cardíacos/fisiologia , Retículo Sarcoplasmático/metabolismo , Antagonistas de Receptores Adrenérgicos beta 1/farmacologia , Animais , Sinalização do Cálcio , Hipóxia Celular , Retículo Endoplasmático/metabolismo , Acoplamento Excitação-Contração/efeitos dos fármacos , Insuficiência Cardíaca/etiologia , Insuficiência Cardíaca/fisiopatologia , Insuficiência Cardíaca/prevenção & controle , Hemodinâmica , Masculino , Camundongos , Camundongos Knockout , Camundongos Mutantes , Mutagênese Insercional , Miócitos Cardíacos/efeitos dos fármacos , Oxirredução , Oxirredutases , Oxirredutases atuantes sobre Doadores de Grupo Enxofre/metabolismo , Propanolaminas/farmacologia
18.
Proc Natl Acad Sci U S A ; 104(51): 20512-6, 2007 Dec 18.
Artigo em Inglês | MEDLINE | ID: mdl-18077386

RESUMO

Gap junction channels are required for normal cardiac impulse propagation, and gap junction remodeling is associated with enhanced arrhythmic risk. Oculodentodigital dysplasia (ODDD) is a multisystem syndrome due to mutations in the connexin43 (Cx43) gap junction channel gene. To determine the effects of a human connexin channelopathy on cardiac electrophysiology and arrhythmogenesis, we generated a murine model of ODDD by introducing the disease-causing I130T mutant allele into the mouse genome. Cx43 abundance was markedly reduced in mutant hearts with preferential loss of phosphorylated forms that interfered with trafficking and assembly of gap junctions in the junctional membrane. Dual whole-cell patch-clamp studies showed significantly lower junctional conductance between neonatal cell pairs from mutant hearts, and optical mapping of isolated-perfused hearts with voltage-sensitive dyes demonstrated significant slowing of conduction velocity. Programmed electrical stimulation revealed a markedly increased susceptibility to spontaneous and inducible ventricular tachyarrhythmias. In summary, our data demonstrate that the I130T mutation interferes with Cx43 posttranslational processing, resulting in diminished cell-cell coupling, slowing of impulse propagation, and a proarrhythmic substrate.


Assuntos
Anormalidades Múltiplas/genética , Arritmias Cardíacas/genética , Conexina 43/genética , Junções Comunicantes/metabolismo , Sistema de Condução Cardíaco/fisiopatologia , Animais , Arritmias Cardíacas/metabolismo , Eletrofisiologia Cardíaca , Conexina 43/deficiência , Anormalidades Craniofaciais/genética , Modelos Animais de Doenças , Junções Comunicantes/genética , Coração/fisiopatologia , Isoquinolinas/análise , Isoquinolinas/metabolismo , Deformidades Congênitas das Extremidades Inferiores/genética , Camundongos , Camundongos Knockout , Mutação , Miócitos Cardíacos/patologia , Síndrome
19.
Biol Reprod ; 76(5): 804-12, 2007 May.
Artigo em Inglês | MEDLINE | ID: mdl-17229929

RESUMO

GJA1 (also known and referred to here as connexin 43 and abbreviated CX43) is the predominant testicular gap junction protein, and CX43 may regulate Sertoli cell maturation and spermatogenesis. We hypothesized that lack of CX43 would inhibit Sertoli cell differentiation and extend proliferation. To test this, a Sertoli cell-specific Cx43 knockout (SC-Cx43 KO) mouse was generated using Cre-lox technology. Immunohistochemistry indicated that CX43 was not expressed in the Sertoli cells of SC-Cx43 KO mice, but was normal in organs such as the heart. Testicular weight was reduced by 41% and 76% in SC-Cx43 KO mice at 20 and 60 days, respectively, vs. wild-type (wt) mice. Seminiferous tubules of SC-Cx43 KO mice contained only Sertoli cells and actively proliferating early spermatogonia. Sertoli cells normally cease proliferation at 2 wk of age in mice and become terminally differentiated. However, proliferating Sertoli cells were present in SC-Cx43 KO but not wt mice at 20 and 60 days of age. Thyroid hormone receptor alpha (THRA) is high in proliferating Sertoli cells, then declines sharply in adulthood. Thra mRNA expression was increased in 20-day SC-Cx43 KO vs. wt mice, and it showed a trend toward an increase in 60-day mice, indicating that loss of CX43 in Sertoli cells inhibited their maturation. In conclusion, we have generated mice lacking CX43 in Sertoli cells but not other tissues. Our data indicate that CX43 in Sertoli cells is essential for spermatogenesis but not spermatogonial maintenance/proliferation. SC-Cx43 KO mice showed continued Sertoli cell proliferation and delayed maturation in adulthood, indicating that CX43 plays key roles in Sertoli cell development.


Assuntos
Conexina 43/biossíntese , Células de Sertoli/fisiologia , Animais , Peso Corporal/fisiologia , Proliferação de Células , Conexina 43/genética , Genes do Tumor de Wilms/fisiologia , Genótipo , Imuno-Histoquímica , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Miocárdio/metabolismo , Tamanho do Órgão/fisiologia , RNA/biossíntese , RNA/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Testículo/crescimento & desenvolvimento
20.
Nat Protoc ; 1(4): 1799-809, 2006.
Artigo em Inglês | MEDLINE | ID: mdl-17487162

RESUMO

Vertebrate gap junction channels are formed by a family of more than 20 connexin proteins. These gap junction proteins are expressed with overlapping cellular and tissue specificity, and coding region mutations can cause human hereditary diseases. Here we present a summary of what has been learned from voltage clamp studies performed on cell pairs either endogenously expressing gap junctions or in which connexins are exogenously expressed. General protocols presented here are currently used to transfect mammalian cells with connexins and to study the biophysical properties of the heterologously expressed connexin channels. Transient transfection is accomplished overnight with maximal expression occurring at about 36 h; stable transfectants normally can be generated within three or four weeks through colony selection. Electrophysiological protocols are presented for analysis of voltage dependence and single-channel conductance of gap junction channels as well as for studies of chemical gating of these channels.


Assuntos
Conexinas/fisiologia , Junções Comunicantes/fisiologia , Técnicas de Patch-Clamp , Transfecção/métodos , Animais , Linhagem Celular Tumoral , Conexinas/genética , Junções Comunicantes/genética , Humanos , Camundongos , Ratos , Xenopus laevis
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA