Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 27
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
JCI Insight ; 4(19)2019 09 12.
Artigo em Inglês | MEDLINE | ID: mdl-31513548

RESUMO

Perturbations in biomechanical stimuli during cardiac development contribute to congenital cardiac defects such as hypoplastic left heart syndrome (HLHS). This study sought to identify stretch-responsive pathways involved in cardiac development. miRNA-Seq identified miR-486 as being increased in cardiomyocytes exposed to cyclic stretch in vitro. The right ventricles (RVs) of patients with HLHS experienced increased stretch and had a trend toward higher miR-486 levels. Sheep RVs dilated from excessive pulmonary blood flow had 60% more miR-486 compared with control RVs. The left ventricles of newborn mice treated with miR-486 mimic were 16.9%-24.6% larger and displayed a 2.48-fold increase in cardiomyocyte proliferation. miR-486 treatment decreased FoxO1 and Smad signaling while increasing the protein levels of Stat1. Stat1 associated with Gata-4 and serum response factor (Srf), 2 key cardiac transcription factors with protein levels that increase in response to miR-486. This is the first report to our knowledge of a stretch-responsive miRNA that increases the growth of the ventricle in vivo.


Assuntos
Ventrículos do Coração/crescimento & desenvolvimento , Síndrome do Coração Esquerdo Hipoplásico/genética , MicroRNAs/metabolismo , Animais , Animais Recém-Nascidos , Fenômenos Biomecânicos , Proliferação de Células/fisiologia , Células Cultivadas , Ventrículos do Coração/metabolismo , Humanos , Síndrome do Coração Esquerdo Hipoplásico/patologia , Síndrome do Coração Esquerdo Hipoplásico/fisiopatologia , Mecanotransdução Celular/fisiologia , Camundongos , Miócitos Cardíacos/metabolismo , Miócitos Cardíacos/fisiologia , Fator de Transcrição STAT1/metabolismo , Ovinos
2.
Circ Res ; 122(4): 583-590, 2018 02 16.
Artigo em Inglês | MEDLINE | ID: mdl-29269349

RESUMO

RATIONALE: Myocardial infarction is a major cause of adult mortality worldwide. The origin(s) of cardiac fibroblasts that constitute the postinfarct scar remain controversial, in particular the potential contribution of bone marrow lineages to activated fibroblasts within the scar. OBJECTIVE: The aim of this study was to establish the origin(s) of infarct fibroblasts using lineage tracing and bone marrow transplants and a robust marker for cardiac fibroblasts, the Collagen1a1-green fluorescent protein reporter. METHODS AND RESULTS: Using genetic lineage tracing or bone marrow transplant, we found no evidence for collagen-producing fibroblasts derived from hematopoietic or bone marrow lineages in hearts subjected to permanent left anterior descending coronary artery ligation. In fact, fibroblasts within the infarcted area were largely of epicardial origin. Intriguingly, collagen-producing fibrocytes from hematopoietic lineages were observed attached to the epicardial surface of infarcted and sham-operated hearts in which a suture was placed around the left anterior descending coronary artery. CONCLUSIONS: In this controversial field, our study demonstrated that the vast majority of infarct fibroblasts were of epicardial origin and not derived from bone marrow lineages, endothelial-to-mesenchymal transition, or blood. We also noted the presence of collagen-producing fibrocytes on the epicardial surface that resulted at least in part from the surgical procedure.


Assuntos
Células da Medula Óssea/citologia , Linhagem da Célula , Infarto do Miocárdio/terapia , Miofibroblastos/citologia , Animais , Células da Medula Óssea/metabolismo , Transplante de Medula Óssea/efeitos adversos , Células Cultivadas , Colágeno Tipo I/genética , Colágeno Tipo I/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Infarto do Miocárdio/patologia , Miofibroblastos/metabolismo , Miofibroblastos/patologia , Pericárdio/citologia
3.
Arch Biochem Biophys ; 615: 53-60, 2017 02 01.
Artigo em Inglês | MEDLINE | ID: mdl-28088328

RESUMO

Mutations in the human cardiac motor protein beta-myosin heavy chain (ßMHC) have been long recognized as a cause of familial hypertrophic cardiomyopathy. Recently, mutations (P830L and A1004S) in the less abundant but faster isoform alpha-myosin heavy chain (αMHC) have been linked to dilated cardiomyopathy (DCM). In this study, we sought to determine the cellular contractile phenotype associated with these point mutations. Ventricular myocytes were isolated from 2 month male Sprague Dawley rats. Cells were cultured in M199 media and infected with recombinant adenovirus containing the P830L or the A1004S mutant human αMHC at a MOI of 500 for 18 h. Uninfected cells (UI), human ßMHC (MOI 500, 18 h), and human αMHC (MOI 500, 18 h) were used as controls. Cells were loaded with fura-2 (1 µM, 15 min) after 48 h. Sarcomere shortening and calcium transients were recorded in CO2 buffered M199 media (36°±1 C) with and without 10 nM isoproterenol (Iso). The A1004S mutation resulted in decreased peak sarcomere shortening while P830L demonstrated near normal shortening kinetics at baseline. In the presence of Iso, the A1004S sarcomere shortening was identical to the ßMHC shortening while the P830L was identical to the αMHC control. All experimental groups had identical calcium transients. Despite a shared association with DCM, the P830L and A1004S αMHC mutations alter myocyte contractility in completely different ways while at the same preserving peak intracellular calcium.


Assuntos
Cálcio/metabolismo , Células Musculares/citologia , Cadeias Pesadas de Miosina/genética , Animais , Cardiomiopatia Dilatada , Homeostase , Humanos , Hipertrofia , Isoproterenol/química , Cinética , Masculino , Mutagênese , Contração Miocárdica , Miócitos Cardíacos/metabolismo , Cadeias Pesadas de Miosina/metabolismo , Fenótipo , Mutação Puntual , Ratos , Ratos Sprague-Dawley , Proteínas Recombinantes/metabolismo , Sarcômeros/metabolismo , Miosinas Ventriculares/metabolismo
4.
J Clin Invest ; 125(8): 3256-68, 2015 Aug 03.
Artigo em Inglês | MEDLINE | ID: mdl-26193633

RESUMO

The sinoatrial node (SAN) maintains a rhythmic heartbeat; therefore, a better understanding of factors that drive SAN development and function is crucial to generation of potential therapies, such as biological pacemakers, for sinus arrhythmias. Here, we determined that the LIM homeodomain transcription factor ISL1 plays a key role in survival, proliferation, and function of pacemaker cells throughout development. Analysis of several Isl1 mutant mouse lines, including animals harboring an SAN-specific Isl1 deletion, revealed that ISL1 within SAN is a requirement for early embryonic viability. RNA-sequencing (RNA-seq) analyses of FACS-purified cells from ISL1-deficient SANs revealed that a number of genes critical for SAN function, including those encoding transcription factors and ion channels, were downstream of ISL1. Chromatin immunoprecipitation assays performed with anti-ISL1 antibodies and chromatin extracts from FACS-purified SAN cells demonstrated that ISL1 directly binds genomic regions within several genes required for normal pacemaker function, including subunits of the L-type calcium channel, Ank2, and Tbx3. Other genes implicated in abnormal heart rhythm in humans were also direct ISL1 targets. Together, our results demonstrate that ISL1 regulates approximately one-third of SAN-specific genes, indicate that a combination of ISL1 and other SAN transcription factors could be utilized to generate pacemaker cells, and suggest ISL1 mutations may underlie sick sinus syndrome.


Assuntos
Proliferação de Células/fisiologia , Regulação da Expressão Gênica no Desenvolvimento/fisiologia , Proteínas com Homeodomínio LIM/metabolismo , Contração Miocárdica/fisiologia , Nó Sinoatrial/embriologia , Fatores de Transcrição/metabolismo , Animais , Anquirinas/genética , Anquirinas/metabolismo , Sobrevivência Celular , Cromatina/genética , Cromatina/metabolismo , Deleção de Genes , Proteínas com Homeodomínio LIM/genética , Camundongos , Camundongos Transgênicos , Ligação Proteica , Síndrome do Nó Sinusal/embriologia , Síndrome do Nó Sinusal/genética , Síndrome do Nó Sinusal/patologia , Nó Sinoatrial/citologia , Proteínas com Domínio T/genética , Proteínas com Domínio T/metabolismo , Fatores de Transcrição/genética
5.
Am J Physiol Cell Physiol ; 309(7): C470-9, 2015 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-26157009

RESUMO

Emery-Dreifuss muscular dystrophy (EDMD) is a degenerative disease primarily affecting skeletal muscles in early childhood as well as cardiac muscle at later stages. EDMD is caused by a number of mutations in genes encoding proteins associated with the nuclear envelope (e.g., Emerin, Lamin A/C, and Nesprin). Recently, a novel protein, Lim-domain only 7 (lmo7) has been reported to play a role in the molecular pathogenesis of EDMD. Prior in vitro and in vivo studies suggested the intriguing possibility that Lmo7 plays a role in skeletal or cardiac muscle pathophysiology. To further understand the in vivo role of Lmo7 in striated muscles, we generated a novel Lmo7-null (lmo7(-/-)) mouse line. Using this mouse line, we examined skeletal and cardiac muscle physiology, as well as the role of Lmo7 in a model of muscular dystrophy and regeneration using the dystrophin-deficient mdx mouse model. Our results demonstrated that lmo7(-/-) mice had no abnormalities in skeletal muscle morphology, physiological function, or regeneration. Cardiac function was also unaffected. Moreover, we found that ablation of lmo7 in mdx mice had no effect on the observed myopathy and muscular regeneration exhibited by mdx mice. Molecular analyses also showed no changes in dystrophin complex factors, MAPK pathway components, and Emerin levels in lmo7 knockout mice. Taken together, we conclude that Lmo7 is dispensable for skeletal muscle and cardiac physiology and pathophysiology.


Assuntos
Coração/fisiologia , Proteínas com Domínio LIM/genética , Músculo Esquelético/fisiologia , Distrofia Muscular de Emery-Dreifuss/patologia , Miocárdio/metabolismo , Fatores de Transcrição/genética , Animais , Expressão Gênica/genética , Proteínas com Domínio LIM/metabolismo , Sistema de Sinalização das MAP Quinases/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Distrofia Muscular de Emery-Dreifuss/genética , Fatores de Transcrição/metabolismo
6.
Cell Rep ; 10(9): 1521-1533, 2015 Mar 10.
Artigo em Inglês | MEDLINE | ID: mdl-25753418

RESUMO

Heart failure is characterized by the transition from an initial compensatory response to decompensation, which can be partially mimicked by transverse aortic constriction (TAC) in rodent models. Numerous signaling molecules have been shown to be part of the compensatory program, but relatively little is known about the transition to decompensation that leads to heart failure. Here, we show that TAC potently decreases the RBFox2 protein in the mouse heart, and cardiac ablation of this critical splicing regulator generates many phenotypes resembling those associated with decompensation in the failing heart. Global analysis reveals that RBFox2 regulates splicing of many genes implicated in heart function and disease. A subset of these genes undergoes developmental regulation during postnatal heart remodeling, which is reversed in TAC-treated and RBFox2 knockout mice. These findings suggest that RBFox2 may be a critical stress sensor during pressure overload-induced heart failure.

7.
J Mol Cell Cardiol ; 79: 133-44, 2015 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-25446186

RESUMO

Perturbed biomechanical stimuli are thought to be critical for the pathogenesis of a number of congenital heart defects, including Hypoplastic Left Heart Syndrome (HLHS). While embryonic cardiomyocytes experience biomechanical stretch every heart beat, their molecular responses to biomechanical stimuli during heart development are poorly understood. We hypothesized that biomechanical stimuli activate specific signaling pathways that impact proliferation, gene expression and myocyte contraction. The objective of this study was to expose embryonic mouse cardiomyocytes (EMCM) to cyclic stretch and examine key molecular and phenotypic responses. Analysis of RNA-Sequencing data demonstrated that gene ontology groups associated with myofibril and cardiac development were significantly modulated. Stretch increased EMCM proliferation, size, cardiac gene expression, and myofibril protein levels. Stretch also repressed several components belonging to the Transforming Growth Factor-ß (Tgf-ß) signaling pathway. EMCMs undergoing cyclic stretch had decreased Tgf-ß expression, protein levels, and signaling. Furthermore, treatment of EMCMs with a Tgf-ß inhibitor resulted in increased EMCM size. Functionally, Tgf-ß signaling repressed EMCM proliferation and contractile function, as assayed via dynamic monolayer force microscopy (DMFM). Taken together, these data support the hypothesis that biomechanical stimuli play a vital role in normal cardiac development and for cardiac pathology, including HLHS.


Assuntos
Embrião de Mamíferos/citologia , Miócitos Cardíacos/citologia , Miócitos Cardíacos/metabolismo , Transdução de Sinais , Estresse Mecânico , Fator de Crescimento Transformador beta/metabolismo , Animais , Proliferação de Células/efeitos dos fármacos , Tamanho Celular , Regulação da Expressão Gênica/efeitos dos fármacos , Ontologia Genética , Camundongos , Contração Miocárdica/efeitos dos fármacos , Miócitos Cardíacos/efeitos dos fármacos , Miofibrilas/metabolismo , Análise de Sequência de RNA , Transdução de Sinais/efeitos dos fármacos , Fator de Crescimento Transformador beta/farmacologia
8.
J Clin Invest ; 124(7): 2921-34, 2014 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-24937432

RESUMO

Activation and accumulation of cardiac fibroblasts, which result in excessive extracellular matrix deposition and consequent mechanical stiffness, myocyte uncoupling, and ischemia, are key contributors to heart failure progression. Recently, endothelial-to-mesenchymal transition (EndoMT) and the recruitment of circulating hematopoietic progenitors to the heart have been reported to generate substantial numbers of cardiac fibroblasts in response to pressure overload-induced injury; therefore, these processes are widely considered to be promising therapeutic targets. Here, using multiple independent murine Cre lines and a collagen1a1-GFP fusion reporter, which specifically labels fibroblasts, we found that following pressure overload, fibroblasts were not derived from hematopoietic cells, EndoMT, or epicardial epithelial-to-mesenchymal transition. Instead, pressure overload promoted comparable proliferation and activation of two resident fibroblast lineages, including a previously described epicardial population and a population of endothelial origin. Together, these data present a paradigm for the origins of cardiac fibroblasts during development and in fibrosis. Furthermore, these data indicate that therapeutic strategies for reducing pathogenic cardiac fibroblasts should shift from targeting presumptive EndoMT or infiltrating hematopoietically derived fibroblasts, toward common pathways upregulated in two endogenous fibroblast populations.


Assuntos
Insuficiência Cardíaca/patologia , Miocárdio/patologia , Animais , Biomarcadores/metabolismo , Pressão Sanguínea , Cardiomegalia/metabolismo , Cardiomegalia/patologia , Linhagem da Célula , Colágeno Tipo I/genética , Colágeno Tipo I/metabolismo , Cadeia alfa 1 do Colágeno Tipo I , Endocárdio/metabolismo , Endocárdio/patologia , Transição Epitelial-Mesenquimal , Fibroblastos/metabolismo , Fibroblastos/patologia , Fibrose , Perfilação da Expressão Gênica , Insuficiência Cardíaca/etiologia , Insuficiência Cardíaca/fisiopatologia , Masculino , Camundongos , Camundongos Knockout , Camundongos Transgênicos , Miocárdio/metabolismo , Pericárdio/metabolismo , Pericárdio/patologia , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo
9.
Hum Mol Genet ; 23(22): 5879-92, 2014 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-24943590

RESUMO

Proper localization and anchorage of nuclei within skeletal muscle is critical for cellular function. Alterations in nuclear anchoring proteins modify a number of cellular functions including mechanotransduction, nuclear localization, chromatin positioning/compaction and overall organ function. In skeletal muscle, nesprin 1 and desmin are thought to link the nucleus to the cytoskeletal network. Thus, we hypothesize that both of these factors play a key role in skeletal muscle function. To examine this question, we utilized global ablation murine models of nesprin 1, desmin or both nesprin 1 and desmin. Herein, we have created the nesprin-desmin double-knockout (DKO) mouse, eliminating a major fraction of nuclear-cytoskeletal connections and enabling understanding of the importance of nuclear anchorage in skeletal muscle. Globally, DKO mice are marked by decreased lifespan, body weight and muscle strength. With regard to skeletal muscle, DKO myonuclear anchorage was dramatically decreased compared with wild-type, nesprin 1(-/-) and desmin(-/-) mice. Additionally, nuclear-cytoskeletal strain transmission was decreased in DKO skeletal muscle. Finally, loss of nuclear anchorage in DKO mice coincided with a fibrotic response as indicated by increased collagen and extracellular matrix deposition and increased passive mechanical properties of muscle bundles. Overall, our data demonstrate that nesprin 1 and desmin serve redundant roles in nuclear anchorage and that the loss of nuclear anchorage in skeletal muscle results in a pathological response characterized by increased tissue fibrosis and mechanical stiffness.


Assuntos
Núcleo Celular/metabolismo , Desmina/genética , Músculo Esquelético/metabolismo , Distrofias Musculares/metabolismo , Proteínas do Tecido Nervoso/genética , Proteínas Nucleares/genética , Animais , Núcleo Celular/genética , Proteínas do Citoesqueleto , Desmina/metabolismo , Feminino , Fibrose/genética , Fibrose/metabolismo , Humanos , Masculino , Camundongos , Camundongos Knockout , Músculo Esquelético/patologia , Distrofias Musculares/genética , Distrofias Musculares/patologia , Proteínas do Tecido Nervoso/metabolismo , Proteínas Nucleares/metabolismo , Transporte Proteico
10.
PLoS Genet ; 10(2): e1004114, 2014 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-24586179

RESUMO

Recent interest has focused on the importance of the nucleus and associated nucleoskeleton in regulating changes in cardiac gene expression in response to biomechanical load. Mutations in genes encoding proteins of the inner nuclear membrane and nucleoskeleton, which cause cardiomyopathy, also disrupt expression of a biomechanically responsive gene program. Furthermore, mutations in the outer nuclear membrane protein Nesprin 1 and 2 have been implicated in cardiomyopathy. Here, we identify for the first time a role for the outer nuclear membrane proteins, Nesprin 1 and Nesprin 2, in regulating gene expression in response to biomechanical load. Ablation of both Nesprin 1 and 2 in cardiomyocytes, but neither alone, resulted in early onset cardiomyopathy. Mutant cardiomyocytes exhibited altered nuclear positioning, shape, and chromatin positioning. Loss of Nesprin 1 or 2, or both, led to impairment of gene expression changes in response to biomechanical stimuli. These data suggest a model whereby biomechanical signals are communicated from proteins of the outer nuclear membrane, to the inner nuclear membrane and nucleoskeleton, to result in changes in gene expression required for adaptation of the cardiomyocyte to changes in biomechanical load, and give insights into etiologies underlying cardiomyopathy consequent to mutations in Nesprin 1 and 2.


Assuntos
Cardiomiopatias/genética , Miocárdio/metabolismo , Proteínas do Tecido Nervoso/genética , Proteínas Nucleares/genética , Animais , Fenômenos Biomecânicos , Cardiomiopatias/metabolismo , Cardiomiopatias/patologia , Núcleo Celular/metabolismo , Proteínas do Citoesqueleto , Regulação da Expressão Gênica , Humanos , Camundongos , Mutação , Miócitos Cardíacos/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Membrana Nuclear/genética , Membrana Nuclear/metabolismo , Matriz Nuclear/metabolismo , Proteínas Nucleares/metabolismo
11.
Circ Res ; 114(3): 538-48, 2014 Jan 31.
Artigo em Inglês | MEDLINE | ID: mdl-24481844

RESUMO

The linker of nucleoskeleton and cytoskeleton (LINC) complex, composed of proteins within the inner and the outer nuclear membranes, connects the nuclear lamina to the cytoskeleton. The importance of this complex has been highlighted by the discovery of mutations in genes encoding LINC complex proteins, which cause skeletal or cardiac myopathies. Herein, this review summarizes structure, function, and interactions of major components of the LINC complex, highlights how mutations in these proteins may lead to cardiac disease, and outlines future challenges in the field.


Assuntos
Citoesqueleto/química , Citoesqueleto/fisiologia , Cardiopatias/fisiopatologia , Miócitos Cardíacos/fisiologia , Matriz Nuclear/química , Matriz Nuclear/fisiologia , Plaquinas/química , Plaquinas/fisiologia , Animais , Citoesqueleto/patologia , Cardiopatias/patologia , Humanos , Miócitos Cardíacos/química , Miócitos Cardíacos/patologia , Matriz Nuclear/patologia
12.
Hum Mol Genet ; 23(1): 209-25, 2014 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-23975679

RESUMO

Recent human genetic studies have provided evidences that sporadic or inherited missense mutations in four-and-a-half LIM domain protein 1 (FHL1), resulting in alterations in FHL1 protein expression, are associated with rare congenital myopathies, including reducing body myopathy and Emery-Dreifuss muscular dystrophy. However, it remains to be clarified whether mutations in FHL1 cause skeletal muscle remodeling owing to gain- or loss of FHL1 function. In this study, we used FHL1-null mice lacking global FHL1 expression to evaluate loss-of-function effects on skeletal muscle homeostasis. Histological and functional analyses of soleus, tibialis anterior and sternohyoideus muscles demonstrated that FHL1-null mice develop an age-dependent myopathy associated with myofibrillar and intermyofibrillar (mitochondrial and sarcoplasmic reticulum) disorganization, impaired muscle oxidative capacity and increased autophagic activity. A longitudinal study established decreased survival rates in FHL1-null mice, associated with age-dependent impairment of muscle contractile function and a significantly lower exercise capacity. Analysis of primary myoblasts isolated from FHL1-null muscles demonstrated early muscle fiber differentiation and maturation defects, which could be rescued by re-expression of the FHL1A isoform, highlighting that FHL1A is necessary for proper muscle fiber differentiation and maturation in vitro. Overall, our data show that loss of FHL1 function leads to myopathy in vivo and suggest that loss of function of FHL1 may be one of the mechanisms underlying muscle dystrophy in patients with FHL1 mutations.


Assuntos
Peptídeos e Proteínas de Sinalização Intracelular/genética , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Proteínas com Domínio LIM/genética , Proteínas com Domínio LIM/metabolismo , Proteínas Musculares/genética , Proteínas Musculares/metabolismo , Músculo Esquelético/patologia , Distrofias Musculares/patologia , Miofibrilas/patologia , Fatores Etários , Animais , Diferenciação Celular , Feminino , Humanos , Masculino , Camundongos , Camundongos Transgênicos , Atividade Motora , Músculo Esquelético/metabolismo , Distrofias Musculares/genética , Distrofia Muscular de Emery-Dreifuss/patologia , Mioblastos Esqueléticos/metabolismo , Mioblastos Esqueléticos/patologia , Miofibrilas/metabolismo
13.
J Clin Invest ; 123(10): 4294-308, 2013 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-24091324

RESUMO

Ischemic damage is recognized to cause cardiomyocyte (CM) death and myocardial dysfunction, but the role of cell-matrix interactions and integrins in this process has not been extensively studied. Expression of α7ß1D integrin, the dominant integrin in normal adult CMs, increases during ischemia/reperfusion (I/R), while deficiency of ß1 integrins increases ischemic damage. We hypothesized that the forced overexpression of integrins on the CM would offer protection from I/R injury. Tg mice with CM-specific overexpression of integrin α7ß1D exposed to I/R had a substantial reduction in infarct size compared with that of α5ß1D-overexpressing mice and WT littermate controls. Using isolated CMs, we found that α7ß1D preserved mitochondrial membrane potential during hypoxia/reoxygenation (H/R) injury via inhibition of mitochondrial Ca2+ overload but did not alter H/R effects on oxidative stress. Therefore, we assessed Ca2+ handling proteins in the CM and found that ß1D integrin colocalized with ryanodine receptor 2 (RyR2) in CM T-tubules, complexed with RyR2 in human and rat heart, and specifically bound to RyR2 amino acids 165-175. Integrins stabilized the RyR2 interdomain interaction, and this stabilization required integrin receptor binding to its ECM ligand. These data suggest that α7ß1D integrin modifies Ca2+ regulatory pathways and offers a means to protect the myocardium from ischemic injury.


Assuntos
Integrinas/metabolismo , Isquemia Miocárdica/metabolismo , Traumatismo por Reperfusão Miocárdica/metabolismo , Miócitos Cardíacos/metabolismo , Sequência de Aminoácidos , Animais , Cálcio/metabolismo , Hipóxia Celular , Células Cultivadas , Humanos , Integrinas/química , Masculino , Potencial da Membrana Mitocondrial , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Dados de Sequência Molecular , Isquemia Miocárdica/patologia , Traumatismo por Reperfusão Miocárdica/patologia , Fragmentos de Peptídeos/química , Fragmentos de Peptídeos/metabolismo , Fosforilação , Ligação Proteica , Domínios e Motivos de Interação entre Proteínas , Processamento de Proteína Pós-Traducional , Estabilidade Proteica , Subunidades Proteicas/metabolismo , Ratos , Ratos Sprague-Dawley , Canal de Liberação de Cálcio do Receptor de Rianodina/química , Canal de Liberação de Cálcio do Receptor de Rianodina/metabolismo
14.
Proc Natl Acad Sci U S A ; 110(37): 14855-60, 2013 Sep 10.
Artigo em Inglês | MEDLINE | ID: mdl-23969833

RESUMO

There is immense cellular and molecular heterogeneity in biological systems. Here, we demonstrate the utility of integrating an inverted light microscope with an ambient ionization source, nanospray electrospray desorption ionization, attached to a high-resolution mass spectrometer to characterize the molecular composition of mouse spinal cords. We detected a broad range of molecules, including peptides and proteins, as well as metabolites such as lipids, sugars, and other small molecules, including S-adenosyl methionine and glutathione, through top-down MS. Top-down analysis revealed variation in the expression of Hb, including the transition from fetal to adult Hb and heterogeneity in Hb subunits consistent with the genetic diversity of the mouse models. Similarly, temporal changes to actin-sequestering proteins ß-thymosins during development were observed. These results demonstrate that interfacing microscopy with ambient ionization provides the means to perform targeted in situ ambient top-down mass spectral analysis to study the pattern of proteins, lipids, and sugars in biologically heterogeneous samples.


Assuntos
Microscopia/métodos , Espectrometria de Massas por Ionização por Electrospray/métodos , Medula Espinal/crescimento & desenvolvimento , Medula Espinal/metabolismo , Sequência de Aminoácidos , Animais , Padronização Corporal , Metabolismo dos Carboidratos , Feminino , Hemoglobinas/genética , Hemoglobinas/metabolismo , Metabolismo dos Lipídeos , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos ICR , Camundongos Knockout , Camundongos Transgênicos , Microscopia/instrumentação , Dados de Sequência Molecular , Proteínas do Tecido Nervoso/metabolismo , Gravidez , Espectrometria de Massas por Ionização por Electrospray/instrumentação , Medula Espinal/embriologia , Espectrometria de Massas em Tandem/instrumentação , Espectrometria de Massas em Tandem/métodos , Timosina/genética , Timosina/metabolismo
15.
Circ Res ; 112(3): e25-8, 2013 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-23371905

RESUMO

RATIONALE: Rossdeutsch et al describe a requirement for thymosin ß4 (Tß4) in vascular development. Impaired mural cell migration, differentiation, partial embryonic lethality, and hemorrhaging were observed after analysis of 2 lines of mice, one of which was germline null for Tß4 and another in which Tß4 was knocked down by endothelial-specific expression of Tß4 short hairpin RNA. These data are in direct contrast to our published global and cardiac-specific Tß4-knockout lines. Thus, the role of Tß4 needs to be clarified to understand its importance in cardiovascular development. OBJECTIVE: To investigate and clarify the role of Tß4 in vascular smooth muscle cell development and vessel stability. METHODS AND RESULTS: Examination of Tß4 global knockouts did not demonstrate embryonic hemorrhaging, altered mural cell development, or lethality. Endothelial-specific knockouts also did not exhibit any embryonic lethality and were viable to adulthood. CONCLUSIONS: Analysis of our Tß4 global and cardiac- and endothelial-specific knockout models demonstrated that Tß4 is dispensable for embryonic viability and vascular development.


Assuntos
Células Endoteliais/metabolismo , Músculo Liso Vascular/metabolismo , Miócitos de Músculo Liso/metabolismo , Timosina/metabolismo , Animais , Biomarcadores/metabolismo , Sobrevivência Celular , Embrião de Mamíferos/irrigação sanguínea , Embrião de Mamíferos/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Genótipo , Idade Gestacional , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Músculo Liso Vascular/embriologia , Fenótipo , Timosina/deficiência , Timosina/genética
16.
Am J Pathol ; 181(4): 1226-35, 2012 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-22954422

RESUMO

Cardiac hypertrophy, whether pathological or physiological, induces a variety of additional morphological and physiological changes in the heart, including altered contractility and hemodynamics. Events exacerbating these changes are documented during later stages of hypertrophy (usually termed pathological hypertrophy). Few studies document the morphological and physiological changes during early physiological hypertrophy. We define acute cardiac remodeling events in response to transverse aortic constriction (TAC), including temporal changes in hypertrophy, collagen deposition, capillary density, and the cell populations responsible for these changes. Cardiac hypertrophy induced by TAC in mice was detected 2 days after surgery (as measured by heart weight, myocyte width, and wall thickness) and peaked by day 7. Picrosirius staining revealed increased collagen deposition 7 days after TAC; immunostaining and flow cytometry indicated a concurrent increase in fibroblasts. The findings correlated with angiogenesis in TAC hearts; a decrease in capillary density was observed at day 2, with recovery to sham-surgery levels by day 7. Increased pericyte levels, which were observed 2 days after TAC, may mediate this angiogenic transition. Gene expression suggests a coordinated response in growth, extracellular matrix, and angiogenic factors to mediate the observed morphological changes. Our data demonstrate that morphological changes in response to cardiovascular injury occur rapidly, and the present findings allow correlation of specific events that facilitate these changes.


Assuntos
Cardiomegalia/patologia , Miocárdio/patologia , Animais , Aorta Torácica/metabolismo , Aorta Torácica/patologia , Aorta Torácica/fisiopatologia , Aorta Torácica/cirurgia , Cardiomegalia/fisiopatologia , Proliferação de Células , Colágeno/metabolismo , Constrição Patológica , Modelos Animais de Doenças , Matriz Extracelular/metabolismo , Imunofluorescência , Regulação da Expressão Gênica , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Miocárdio/ultraestrutura , Miócitos Cardíacos/metabolismo , Miócitos Cardíacos/patologia , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Coloração e Rotulagem , Remodelação Ventricular
17.
Am J Physiol Regul Integr Comp Physiol ; 303(7): R689-99, 2012 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-22874425

RESUMO

We examined the role that enzymatic protein O-GlcNAcylation plays in the development of diabetic cardiomyopathy in a mouse model of Type 2 diabetes mellitus (DM2). Mice injected with low-dose streptozotocin and fed a high-fat diet developed mild hyperglycemia and obesity consistent with DM2. Studies were performed from 1 to 6 mo after initiating the DM2 protocol. After 1 mo, DM2 mice showed increased body weight, impaired fasting blood glucose, and hyperinsulinemia. Echocardiographic evaluation revealed left ventricular diastolic dysfunction by 2 mo and O-GlcNAcylation of several cardiac proteins and of nuclear transcription factor Sp1. By 4 mo, systolic dysfunction was observed and sarcoplasmic reticulum Ca(2+) ATPase expression decreased by 50%. Fibrosis was not observed at any timepoint in DM2 mice. Levels of the rate-limiting enzyme of the hexosamine biosynthetic pathway, glutamine:fructose-6-phosphate amidotransferase (GFAT) were increased as early as 2 mo. Fatty acids, which are elevated in DM2 mice, can possibly be linked to excessive protein O-GlcNAcylation levels, as cultured cardiac myocytes in normal glucose treated with oleic acid showed increased O-GlcNAcylation and GFAT levels. These data indicate that the early onset of diastolic dysfunction followed by the loss of systolic function, in the absence of cardiac hypertrophy or fibrosis, is associated with increased cardiac protein O-GlcNAcylation and increased O-GlcNAcylation levels of key calcium-handling proteins. A link between excessive protein O-GlcNAcylation and cardiac dysfunction is further supported by results showing that reducing O-GlcNAcylation by O-GlcNAcase overexpression improved cardiac function in the diabetic mouse. In addition, fatty acids play a role in stimulating excess O-GlcNAcylation. The nature and time course of changes observed in cardiac function suggest that protein O-GlcNAcylation plays a mechanistic role in the triggering of diabetic cardiomyopathy in DM2.


Assuntos
Diabetes Mellitus Experimental/complicações , Diabetes Mellitus Experimental/metabolismo , Cardiomiopatias Diabéticas/metabolismo , Progressão da Doença , Miócitos Cardíacos/metabolismo , Proteínas/metabolismo , Animais , Células Cultivadas , Diabetes Mellitus Experimental/induzido quimicamente , Cardiomiopatias Diabéticas/patologia , Cardiomiopatias Diabéticas/fisiopatologia , Modelos Animais de Doenças , Ecocardiografia , Glutamina-Frutose-6-Fosfato Transaminase (Isomerizante)/metabolismo , Glicosilação , Camundongos , Camundongos Endogâmicos C57BL , N-Acetilglucosaminiltransferases/metabolismo , Estreptozocina/efeitos adversos , Disfunção Ventricular Esquerda , beta-N-Acetil-Hexosaminidases/metabolismo
18.
Arterioscler Thromb Vasc Biol ; 32(5): 1308-19, 2012 May.
Artigo em Inglês | MEDLINE | ID: mdl-22402364

RESUMO

OBJECTIVE: Although significant research has detailed angiogenesis during development and cancer, little is known about cardiac angiogenesis, yet it is critical for survival following pathological insult. The transcription factor c-Myc is a target of anticancer therapies because of its mitogenic and proangiogenic induction. In the current study, we investigate its role in cardiac angiogenesis in a cell-dependent and gene-specific context. METHODS AND RESULTS: Angiogenesis assays using c-Myc-deficient cardiac endothelial cells and fibroblasts demonstrate that c-Myc is essential to vessel formation, and fibroblast-mediated vessel formation is dependent on c-Myc expression in fibroblasts. Gene analyses revealed that c-Myc-mediated gene expression is unique in cardiac angiogenesis and varies in a cell-dependent manner. In vitro 3-dimensional cultures demonstrated c-Myc's role in the expression of secreted angiogenic factors, while also providing evidence for c-Myc-mediated cell-cell interactions. Additional in vivo vascular analyses support c-Myc's critical role in capillary formation and vessel patterning during development and also in response to a pathological stimulus where its expression in myocytes is required for angiogenic remodeling. CONCLUSIONS: These data demonstrate that proper c-Myc expression in cardiac fibroblasts and myocytes is essential to cardiac angiogenesis. These results have the potential for novel therapeutic applications involving the angiogenic response during cardiac remodeling.


Assuntos
Vasos Coronários/citologia , Neovascularização Fisiológica/genética , Proteínas Proto-Oncogênicas c-myc/biossíntese , RNA/genética , Transdução de Sinais , Animais , Comunicação Celular , Células Cultivadas , Vasos Coronários/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Microscopia Confocal , Reação em Cadeia da Polimerase , Proteínas Proto-Oncogênicas c-myc/genética
19.
Circ Res ; 110(3): 456-64, 2012 Feb 03.
Artigo em Inglês | MEDLINE | ID: mdl-22158707

RESUMO

RATIONALE: Thymosin beta 4 (Tß4) is a 43-amino acid factor encoded by an X-linked gene. Recent studies have suggested that Tß4 is a key factor in cardiac development, growth, disease, epicardial integrity, and blood vessel formation. Cardiac-specific short hairpin (sh)RNA knockdown of tß4 has been reported to result in embryonic lethality at E14.5-16.5, with severe cardiac and angiogenic defects. However, this shRNA tß4-knockdown model did not completely abrogate Tß4 expression. To completely ablate Tß4 and to rule out the possibility of off-target effects associated with shRNA gene silencing, further studies of global or cardiac-specific knockouts are critical. OBJECTIVE: We examined the role of Tß4 in developing and adult heart through global and cardiac specific tß4-knockout mouse models. METHODS AND RESULTS: Global tß4-knockout mice were born at mendelian ratios and exhibited normal heart and blood vessel formation. Furthermore, in adult global tß4-knockout mice, cardiac function, capillary density, expression of key cardiac fetal and angiogenic genes, epicardial marker expression, and extracellular matrix deposition were indistinguishable from that of controls. Tissue-specific tß4-deficient mice, generated by crossing tß4-floxed mice to Nkx2.5-Cre and αMHC-Cre, were also found to have no phenotype. CONCLUSIONS: We conclude that Tß4 is dispensable for embryonic viability, heart development, coronary vessel development, and adult myocardial function.


Assuntos
Coração/embriologia , Coração/fisiologia , Timosina/fisiologia , Animais , Vasos Coronários/embriologia , Vasos Coronários/fisiologia , Desenvolvimento Embrionário/efeitos dos fármacos , Feminino , Regulação da Expressão Gênica no Desenvolvimento/efeitos dos fármacos , Masculino , Camundongos , Camundongos Knockout , Modelos Animais , Neovascularização Fisiológica/fisiologia , RNA Interferente Pequeno/farmacologia , Timosina/deficiência , Timosina/genética
20.
J Clin Invest ; 121(8): 3269-76, 2011 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-21747165

RESUMO

The small GTPase RhoA serves as a nodal point for signaling through hormones and mechanical stretch. However, the role of RhoA signaling in cardiac pathophysiology is poorly understood. To address this issue, we generated mice with cardiomyocyte-specific conditional expression of low levels of activated RhoA (CA-RhoA mice) and demonstrated that they exhibited no overt cardiomyopathy. When challenged by in vivo or ex vivo ischemia/reperfusion (I/R), however, the CA-RhoA mice exhibited strikingly increased tolerance to injury, which was manifest as reduced myocardial lactate dehydrogenase (LDH) release and infarct size and improved contractile function. PKD was robustly activated in CA-RhoA hearts. The cardioprotection afforded by RhoA was reversed by PKD inhibition. The hypothesis that activated RhoA and PKD serve protective physiological functions during I/R was supported by several lines of evidence. In WT mice, both RhoA and PKD were rapidly activated during I/R, and blocking PKD augmented I/R injury. In addition, cardiac-specific RhoA-knockout mice showed reduced PKD activation after I/R and strikingly decreased tolerance to I/R injury, as shown by increased infarct size and LDH release. Collectively, our findings provide strong support for the concept that RhoA signaling in adult cardiomyocytes promotes survival. They also reveal unexpected roles for PKD as a downstream mediator of RhoA and in cardioprotection against I/R.


Assuntos
Regulação Enzimológica da Expressão Gênica , Coração/fisiopatologia , Traumatismo por Reperfusão/metabolismo , Proteínas rho de Ligação ao GTP/fisiologia , Animais , L-Lactato Desidrogenase/metabolismo , Masculino , Camundongos , Camundongos Knockout , Camundongos Transgênicos , Contração Miocárdica/fisiologia , Miocárdio/enzimologia , Miocárdio/patologia , Perfusão , Fenótipo , Proteína Quinase C/metabolismo , Proteínas rho de Ligação ao GTP/genética , Proteína rhoA de Ligação ao GTP
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...