Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 26
Filtrar
1.
Am J Physiol Heart Circ Physiol ; 322(2): H234-H245, 2022 02 01.
Artigo em Inglês | MEDLINE | ID: mdl-34919456

RESUMO

Mouse models are used to model human diseases and perform pharmacological efficacy testing to advance therapies to humans; most of these studies are conducted in room temperature conditions. At room temperature (22°C), mice are cold-stressed and must use brown adipose tissue (BAT) to maintain body temperature. This cold stress increases catecholamine tone to maintain adipocyte lipid release via lipolysis, which will fuel adaptive thermogenesis. Maintaining rodents at thermoneutral temperatures (28°C) ameliorates the need for adaptive thermogenesis, thus reducing catecholamine tone and BAT activity. Cardiovascular tone is also determined by catecholamine levels in rodents, as ß-adrenergic stimuli are primary drivers of not only lipolytic but also ionotropic and chronotropic responses. As mice have increased catecholamine tone at room temperature, we investigated how thermoneutral housing conditions would impact cardiometabolic function. Here, we show a rapid and reversible effect of thermoneutrality on both heart rate and blood pressure in chow-fed animals, which was blunted in animals fed a high-fat diet. Animals subjected to transverse aortic constriction displayed compensated hypertrophy at room temperature, whereas animals displayed less hypertrophy and a trend toward worse systolic function at thermoneutrality. Despite these dramatic changes in blood pressure and heart rate at thermoneutral housing conditions, enalapril effectively improved cardiac hypertrophy and gene expression alterations. There were surprisingly few differences in cardiac parameters in high-fat-fed animals at thermoneutrality. Overall, these data suggest that thermoneutral housing may alter some aspects of cardiac remodeling in preclinical mouse models of heart failure.NEW & NOTEWORTHY Thermoneutral housing conditions cause rapid and reversible changes in mouse heart rate and blood pressure. Despite dramatic reductions in heart rate and blood pressure, thermoneutrality reduced the compensatory hypertrophic response in a pressure overload heart failure model compared with room temperature housing, and ACE inhibitors were still efficacious to prevent pressure overload-induced cardiac remodeling. The effects of thermoneutrality on heart rate and blood pressure are abrogated in the context of diet-induced obesity.


Assuntos
Regulação da Temperatura Corporal , Doenças Cardiovasculares/fisiopatologia , Modelos Animais de Doenças , Abrigo para Animais/normas , Animais , Doenças Cardiovasculares/metabolismo , Frequência Cardíaca , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Temperatura
2.
Circ Res ; 127(5): 631-646, 2020 08 14.
Artigo em Inglês | MEDLINE | ID: mdl-32418505

RESUMO

RATIONALE: Cardiac CITED4 (CBP/p300-interacting transactivators with E [glutamic acid]/D [aspartic acid]-rich-carboxylterminal domain4) is induced by exercise and is sufficient to cause physiological hypertrophy and mitigate adverse ventricular remodeling after ischemic injury. However, the role of endogenous CITED4 in response to physiological or pathological stress is unknown. OBJECTIVE: To investigate the role of CITED4 in murine models of exercise and pressure overload. METHODS AND RESULTS: We generated cardiomyocyte-specific CITED4 knockout mice (C4KO) and subjected them to an intensive swim exercise protocol as well as transverse aortic constriction (TAC). Echocardiography, Western blotting, qPCR, immunohistochemistry, immunofluorescence, and transcriptional profiling for mRNA and miRNA (microRNA) expression were performed. Cellular crosstalk was investigated in vitro. CITED4 deletion in cardiomyocytes did not affect baseline cardiac size or function in young adult mice. C4KO mice developed modest cardiac dysfunction and dilation in response to exercise. After TAC, C4KOs developed severe heart failure with left ventricular dilation, impaired cardiomyocyte growth accompanied by reduced mTOR (mammalian target of rapamycin) activity and maladaptive cardiac remodeling with increased apoptosis, autophagy, and impaired mitochondrial signaling. Interstitial fibrosis was markedly increased in C4KO hearts after TAC. RNAseq revealed induction of a profibrotic miRNA network. miR30d was decreased in C4KO hearts after TAC and mediated crosstalk between cardiomyocytes and fibroblasts to modulate fibrosis. miR30d inhibition was sufficient to increase cardiac dysfunction and fibrosis after TAC. CONCLUSIONS: CITED4 protects against pathological cardiac remodeling by regulating mTOR activity and a network of miRNAs mediating cardiomyocyte to fibroblast crosstalk. Our findings highlight the importance of CITED4 in response to both physiological and pathological stimuli.


Assuntos
Cardiomegalia Induzida por Exercícios , Hipertrofia Ventricular Esquerda/metabolismo , Miócitos Cardíacos/metabolismo , Fatores de Transcrição/metabolismo , Função Ventricular Esquerda , Remodelação Ventricular , Animais , Comunicação Celular , Células Cultivadas , Modelos Animais de Doenças , Fibroblastos/metabolismo , Fibroblastos/patologia , Fibrose , Regulação da Expressão Gênica , Insuficiência Cardíaca/genética , Insuficiência Cardíaca/patologia , Insuficiência Cardíaca/fisiopatologia , Hipertrofia Ventricular Esquerda/genética , Hipertrofia Ventricular Esquerda/patologia , Hipertrofia Ventricular Esquerda/fisiopatologia , Masculino , Camundongos Knockout , MicroRNAs/genética , MicroRNAs/metabolismo , Miócitos Cardíacos/patologia , Ratos , Transdução de Sinais , Serina-Treonina Quinases TOR/genética , Serina-Treonina Quinases TOR/metabolismo , Fatores de Transcrição/deficiência , Fatores de Transcrição/genética , Transcriptoma
4.
Circ Res ; 121(12): 1370-1378, 2017 Dec 08.
Artigo em Inglês | MEDLINE | ID: mdl-28928113

RESUMO

RATIONALE: Pregnancy profoundly alters maternal physiology. The heart hypertrophies during pregnancy, but its metabolic adaptations, are not well understood. OBJECTIVE: To determine the mechanisms underlying cardiac substrate use during pregnancy. METHODS AND RESULTS: We use here 13C glucose, 13C lactate, and 13C fatty acid tracing analyses to show that hearts in late pregnant mice increase fatty acid uptake and oxidation into the tricarboxylic acid cycle, while reducing glucose and lactate oxidation. Mitochondrial quantity, morphology, and function do not seem altered. Insulin signaling seems intact, and the abundance and localization of the major fatty acid and glucose transporters, CD36 (cluster of differentiation 36) and GLUT4 (glucose transporter type 4), are also unchanged. Rather, we find that the pregnancy hormone progesterone induces PDK4 (pyruvate dehydrogenase kinase 4) in cardiomyocytes and that elevated PDK4 levels in late pregnancy lead to inhibition of PDH (pyruvate dehydrogenase) and pyruvate flux into the tricarboxylic acid cycle. Blocking PDK4 reverses the metabolic changes seen in hearts in late pregnancy. CONCLUSIONS: Taken together, these data indicate that the hormonal environment of late pregnancy promotes metabolic remodeling in the heart at the level of PDH, rather than at the level of insulin signaling.


Assuntos
Miocárdio/metabolismo , Gravidez/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Ácido Pirúvico/metabolismo , Animais , Ciclo do Ácido Cítrico , Ácidos Graxos/metabolismo , Feminino , Glucose/metabolismo , Ácido Láctico/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Progesterona/metabolismo , Piruvato Desidrogenase Quinase de Transferência de Acetil
5.
Mol Cell ; 42(1): 84-95, 2011 Apr 08.
Artigo em Inglês | MEDLINE | ID: mdl-21474070

RESUMO

Adrenergic stimulation of the heart engages cAMP and phosphoinositide second messenger signaling cascades. Cardiac phosphoinositide 3-kinase p110γ participates in these processes by sustaining ß-adrenergic receptor internalization through its catalytic function and by controlling phosphodiesterase 3B (PDE3B) activity via an unknown kinase-independent mechanism. We have discovered that p110γ anchors protein kinase A (PKA) through a site in its N-terminal region. Anchored PKA activates PDE3B to enhance cAMP degradation and phosphorylates p110γ to inhibit PIP(3) production. This provides local feedback control of PIP(3) and cAMP signaling events. In congestive heart failure, p110γ is upregulated and escapes PKA-mediated inhibition, contributing to a reduction in ß-adrenergic receptor density. Pharmacological inhibition of p110γ normalizes ß-adrenergic receptor density and improves contractility in failing hearts.


Assuntos
Proteínas de Ancoragem à Quinase A/metabolismo , Classe Ib de Fosfatidilinositol 3-Quinase/metabolismo , Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , AMP Cíclico/metabolismo , Miócitos Cardíacos/metabolismo , Fosfatos de Fosfatidilinositol/metabolismo , Sequência de Aminoácidos , Animais , Sequência de Bases , Linhagem Celular , Classe Ib de Fosfatidilinositol 3-Quinase/química , Classe Ib de Fosfatidilinositol 3-Quinase/deficiência , Classe Ib de Fosfatidilinositol 3-Quinase/genética , Subunidade RIIalfa da Proteína Quinase Dependente de AMP Cíclico/metabolismo , Nucleotídeo Cíclico Fosfodiesterase do Tipo 3/metabolismo , DNA/genética , Ativação Enzimática , Inibidores Enzimáticos/farmacologia , Insuficiência Cardíaca/tratamento farmacológico , Insuficiência Cardíaca/metabolismo , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Dados de Sequência Molecular , Fragmentos de Peptídeos/química , Fragmentos de Peptídeos/genética , Fragmentos de Peptídeos/metabolismo , Inibidores de Fosfoinositídeo-3 Quinase , Fosforilação , Mapeamento de Interação de Proteínas , Quinoxalinas/farmacologia , Receptores Adrenérgicos beta/metabolismo , Sistemas do Segundo Mensageiro , Homologia de Sequência de Aminoácidos , Tiazolidinedionas/farmacologia
6.
Arterioscler Thromb Vasc Biol ; 36(9): 1854-67, 2016 09.
Artigo em Inglês | MEDLINE | ID: mdl-27386938

RESUMO

OBJECTIVE: S100A6, a member of the S100 protein family, has been described as relevant for cell cycle entry and progression in endothelial cells. The molecular mechanism conferring S100A6's proliferative actions, however, remained elusive. APPROACH AND RESULTS: Originating from the clinically relevant observation of enhanced S100A6 protein expression in proliferating endothelial cells in remodeling coronary and carotid arteries, our study unveiled S100A6 as a suppressor of antiproliferative signal transducers and activators of transcription 1 signaling. Discovery of the molecular liaison was enabled by combining gene expression time series analysis with bioinformatic pathway modeling in S100A6-silenced human endothelial cells stimulated with vascular endothelial growth factor A. This unbiased approach led to successful identification and experimental validation of interferon-inducible transmembrane protein 1 and protein inhibitors of activated signal transducers and activators of transcription as key components of the link between S100A6 and signal transducers and activators of transcription 1. CONCLUSIONS: Given the important role of coordinated endothelial cell cycle activity for integrity and reconstitution of the inner lining of arterial blood vessels in health and disease, signal transducers and activators of transcription 1 suppression by S100A6 may represent a promising therapeutic target to facilitate reendothelialization in damaged vessels.


Assuntos
Proteínas de Ciclo Celular/metabolismo , Ciclo Celular , Proliferação de Células , Células Endoteliais/metabolismo , Proteínas S100/metabolismo , Fator de Transcrição STAT1/metabolismo , Animais , Antígenos de Diferenciação/genética , Antígenos de Diferenciação/metabolismo , Ciclo Celular/efeitos dos fármacos , Proteínas de Ciclo Celular/genética , Proliferação de Células/efeitos dos fármacos , Células Cultivadas , Biologia Computacional , Modelos Animais de Doenças , Células Endoteliais/efeitos dos fármacos , Perfilação da Expressão Gênica/métodos , Redes Reguladoras de Genes , Inativação Gênica , Humanos , Masculino , Proteínas Inibidoras de STAT Ativados/genética , Proteínas Inibidoras de STAT Ativados/metabolismo , Interferência de RNA , Ratos Sprague-Dawley , Reepitelização , Proteína A6 Ligante de Cálcio S100 , Proteínas S100/genética , Fator de Transcrição STAT1/genética , Transdução de Sinais , Proteínas Modificadoras Pequenas Relacionadas à Ubiquitina/genética , Proteínas Modificadoras Pequenas Relacionadas à Ubiquitina/metabolismo , Sus scrofa , Fatores de Tempo , Transcriptoma , Transfecção , Fator A de Crescimento do Endotélio Vascular/farmacologia , Lesões do Sistema Vascular/genética , Lesões do Sistema Vascular/metabolismo , Lesões do Sistema Vascular/patologia
7.
FASEB J ; 28(10): 4408-19, 2014 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-25005176

RESUMO

Mitochondrial dysfunction in adipose tissue occurs in obesity, type 2 diabetes, and some forms of lipodystrophy, but whether this dysfunction contributes to or is the result of these disorders is unknown. To investigate the physiological consequences of severe mitochondrial impairment in adipose tissue, we generated mice deficient in mitochondrial transcription factor A (TFAM) in adipocytes by using mice carrying adiponectin-Cre and TFAM floxed alleles. These adiponectin TFAM-knockout (adipo-TFAM-KO) mice had a 75-81% reduction in TFAM in the subcutaneous and intra-abdominal white adipose tissue (WAT) and interscapular brown adipose tissue (BAT), causing decreased expression and enzymatic activity of proteins in complexes I, III, and IV of the electron transport chain (ETC). This mitochondrial dysfunction led to adipocyte death and inflammation in WAT and a whitening of BAT. As a result, adipo-TFAM-KO mice were resistant to weight gain, but exhibited insulin resistance on both normal chow and high-fat diets. These lipodystrophic mice also developed hypertension, cardiac hypertrophy, and cardiac dysfunction. Thus, isolated mitochondrial dysfunction in adipose tissue can lead a syndrome of lipodystrophy with metabolic syndrome and cardiovascular complications.


Assuntos
Tecido Adiposo Marrom/metabolismo , Tecido Adiposo Branco/metabolismo , Proteínas de Ligação a DNA/metabolismo , Proteínas de Grupo de Alta Mobilidade/metabolismo , Resistência à Insulina , Lipodistrofia/metabolismo , Mitocôndrias/metabolismo , Adiponectina/genética , Adiponectina/metabolismo , Tecido Adiposo Marrom/patologia , Tecido Adiposo Branco/patologia , Animais , Cardiomegalia/genética , Cardiomegalia/metabolismo , Proteínas de Ligação a DNA/genética , Complexo de Proteínas da Cadeia de Transporte de Elétrons/metabolismo , Fígado Gorduroso/genética , Fígado Gorduroso/metabolismo , Fígado Gorduroso/patologia , Proteínas de Grupo de Alta Mobilidade/genética , Hipertensão/genética , Hipertensão/metabolismo , Lipodistrofia/genética , Lipodistrofia/fisiopatologia , Masculino , Camundongos , Aumento de Peso
8.
Cardiovasc Res ; 120(3): 262-272, 2024 03 14.
Artigo em Inglês | MEDLINE | ID: mdl-38084908

RESUMO

AIMS: Physiological cardiac hypertrophy occurs in response to exercise and can protect against pathological stress. In contrast, pathological hypertrophy occurs in disease and often precedes heart failure. The cardiac pathways activated in physiological and pathological hypertrophy are largely distinct. Our prior work demonstrated that miR-222 increases in exercised hearts and is required for exercise-induced cardiac hypertrophy and cardiomyogenesis. Here, we sought to define the role of miR-222 in pathological hypertrophy. METHODS AND RESULTS: We found that miR-222 also increased in pathological hypertrophy induced by pressure overload. To assess its functional significance in this setting, we generated a miR-222 gain-of-function model through cardiac-specific constitutive transgenic miR-222 expression (TgC-miR-222) and used locked nucleic acid anti-miR specific for miR-222 to inhibit its effects. Both gain- and loss-of-function models manifested normal cardiac structure and function at baseline. However, after transverse aortic constriction (TAC), miR-222 inhibition accelerated the development of pathological hypertrophy, cardiac dysfunction, and heart failure. Conversely, miR-222-overexpressing mice had less pathological hypertrophy after TAC, as well as better cardiac function and survival. We identified p53-up-regulated modulator of apoptosis, a pro-apoptotic Bcl-2 family member, and the transcription factors, Hmbox1 and nuclear factor of activated T-cells 3, as direct miR-222 targets contributing to its roles in this context. CONCLUSION: While miR-222 is necessary for physiological cardiac growth, it inhibits cardiac growth in response to pressure overload and reduces adverse remodelling and cardiac dysfunction. These findings support the model that physiological and pathological hypertrophy are fundamentally different. Further, they suggest that miR-222 may hold promise as a therapeutic target in pathological cardiac hypertrophy and heart failure.


Assuntos
Cardiopatias , Insuficiência Cardíaca , MicroRNAs , Camundongos , Animais , MicroRNAs/genética , Cardiomegalia/metabolismo , Insuficiência Cardíaca/metabolismo , Coração , Cardiopatias/patologia , Miócitos Cardíacos/metabolismo , Modelos Animais de Doenças , Proteínas de Homeodomínio/metabolismo
9.
Circulation ; 126(17): 2073-83, 2012 Oct 23.
Artigo em Inglês | MEDLINE | ID: mdl-23008439

RESUMO

BACKGROUND: Phosphoinositide 3-kinase γ (PI3Kγ) signaling engaged by ß-adrenergic receptors is pivotal in the regulation of myocardial contractility and remodeling. However, the role of PI3Kγ in catecholamine-induced arrhythmia is currently unknown. METHODS AND RESULTS: Mice lacking PI3Kγ (PI3Kγ(-/-)) showed runs of premature ventricular contractions on adrenergic stimulation that could be rescued by a selective ß(2)-adrenergic receptor blocker and developed sustained ventricular tachycardia after transverse aortic constriction. Consistently, fluorescence resonance energy transfer probes revealed abnormal cAMP accumulation after ß(2)-adrenergic receptor activation in PI3Kγ(-/-) cardiomyocytes that depended on the loss of the scaffold but not of the catalytic activity of PI3Kγ. Downstream from ß-adrenergic receptors, PI3Kγ was found to participate in multiprotein complexes linking protein kinase A to the activation of phosphodiesterase (PDE) 3A, PDE4A, and PDE4B but not of PDE4D. These PI3Kγ-regulated PDEs lowered cAMP and limited protein kinase A-mediated phosphorylation of L-type calcium channel (Ca(v)1.2) and phospholamban. In PI3Kγ(-/-) cardiomyocytes, Ca(v)1.2 and phospholamban were hyperphosphorylated, leading to increased Ca(2+) spark occurrence and amplitude on adrenergic stimulation. Furthermore, PI3Kγ(-/-) cardiomyocytes showed spontaneous Ca(2+) release events and developed arrhythmic calcium transients. CONCLUSIONS: PI3Kγ coordinates the coincident signaling of the major cardiac PDE3 and PDE4 isoforms, thus orchestrating a feedback loop that prevents calcium-dependent ventricular arrhythmia.


Assuntos
Catecolaminas/toxicidade , Classe Ib de Fosfatidilinositol 3-Quinase/fisiologia , Proteínas Quinases Dependentes de AMP Cíclico/fisiologia , Nucleotídeo Cíclico Fosfodiesterase do Tipo 3/metabolismo , Nucleotídeo Cíclico Fosfodiesterase do Tipo 4/metabolismo , Taquicardia Ventricular/enzimologia , Taquicardia Ventricular/prevenção & controle , Animais , Animais Recém-Nascidos , Biorretroalimentação Psicológica/fisiologia , Sinalização do Cálcio/genética , Classe Ib de Fosfatidilinositol 3-Quinase/deficiência , Classe Ib de Fosfatidilinositol 3-Quinase/genética , Técnicas de Introdução de Genes , Isoenzimas/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Miócitos Cardíacos/enzimologia
10.
Circulation ; 123(4): 391-9, 2011 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-21242482

RESUMO

BACKGROUND: Signaling from phosphoinositide 3-kinase γ (PI3Kγ) is crucial for leukocyte recruitment and inflammation but also contributes to cardiac maladaptive remodeling. To better understand the translational potential of these findings, this study investigates the role of PI3Kγ activity in pressure overload-induced heart failure, addressing the distinct contributions of bone marrow-derived and cardiac cells. METHODS AND RESULTS: After transverse aortic constriction, mice knock-in for a catalytically inactive PI3Kγ (PI3Kγ KD) showed reduced fibrosis and normalized cardiac function up to 16 weeks. Accordingly, treatment with a selective PI3Kγ inhibitor prevented transverse aortic constriction-induced fibrosis. To define the cell types involved in this protection, bone marrow chimeras, lacking kinase activity in the immune system or the heart, were studied after transverse aortic constriction. Bone marrow-derived cells from PI3Kγ KD mice were not recruited to wild-type hearts, thus preventing fibrosis and preserving diastolic function. After prolonged pressure overload, chimeras with PI3Kγ KD bone marrow-derived cells showed slower development of left ventricular dilation and higher fractional shortening than controls. Conversely, in the presence of a wild-type immune system, KD hearts displayed bone marrow-derived cell infiltration and fibrosis at early stages but reduced left ventricular dilation and preserved contractile function at later time points. CONCLUSIONS: Together, these data demonstrate that, in response to transverse aortic constriction, PI3Kγ contributes to maladaptive remodeling at multiple levels by modulating both cardiac and immune cell functions.


Assuntos
Classe Ib de Fosfatidilinositol 3-Quinase/metabolismo , Insuficiência Cardíaca/enzimologia , Leucócitos/enzimologia , Miocárdio/enzimologia , Animais , Classe Ib de Fosfatidilinositol 3-Quinase/genética , Fibrose/genética , Técnicas de Introdução de Genes , Coração/fisiopatologia , Insuficiência Cardíaca/genética , Camundongos , Camundongos Endogâmicos C57BL , Remodelação Ventricular/genética
11.
Circ Res ; 106(4): 757-68, 2010 Mar 05.
Artigo em Inglês | MEDLINE | ID: mdl-20056919

RESUMO

RATIONALE: Phosphoinositide 3-kinase (PI3K)gamma is expressed in hematopoietic cells, endothelial cells (ECs), and cardiomyocytes and regulates different cellular functions relevant to inflammation, tissue remodeling and cicatrization. Recently, PI3Kgamma inhibitors have been indicated for the treatment of chronic inflammatory/autoimmune diseases and atherosclerosis. OBJECTIVE: We aimed to determine PI3Kgamma contribution to the angiogenic capacity of ECs and the effect of PI3Kgamma inhibition on healing of myocardial infarction (MI). METHODS AND RESULTS: Human umbilical ECs were treated with a selective PI3Kgamma inhibitor, AS605240, or a pan-phosphoinositide 3-kinases inhibitor, LY294002. Both inhibitory treatments and small interfering RNA-mediated PI3Kgamma knockdown strongly impaired ECs angiogenic capacity, because of suppression of the PI3K/Akt and mitogen-activated protein kinase pathways. Constitutive activation of Akt rescued the angiogenic defect. Reparative angiogenesis was studied in vivo in a model of MI. AS605240 did not affect MI-induced PI3Kgamma upregulation, whereas it suppressed Akt activation and downstream signaling. AS605240 strongly reduced inflammation, enhanced cardiomyocyte apoptosis, and impaired survival and proliferation of ECs in peri-infarct zone, which resulted in defective reparative neovascularization. As a consequence, AS605240-treated MI hearts showed increased infarct size and impaired recovery of left ventricular function. Similarly, PI3Kgamma-deficient mice showed impaired reparative neovascularization, enhanced cardiomyocyte apoptosis and marked deterioration of cardiac function following MI. Mice expressing catalytically inactive PI3Kgamma also failed to mount a proper neovascularization, although cardiac dysfunction was similar to wild-type controls. CONCLUSIONS: PI3Kgamma expression and catalytic activity are involved at different levels in reparative neovascularization and healing of MI.


Assuntos
Células Endoteliais/enzimologia , Infarto do Miocárdio/enzimologia , Miocárdio/enzimologia , Neovascularização Fisiológica , Fosfatidilinositol 3-Quinases/metabolismo , Regeneração , Animais , Apoptose , Movimento Celular , Proliferação de Células , Sobrevivência Celular , Células Cultivadas , Cromonas/farmacologia , Modelos Animais de Doenças , Relação Dose-Resposta a Droga , Células Endoteliais/efeitos dos fármacos , Células Endoteliais/patologia , Humanos , Inflamação/enzimologia , Inflamação/patologia , Inflamação/fisiopatologia , Leucócitos/enzimologia , Masculino , Camundongos , Morfolinas/farmacologia , Infarto do Miocárdio/tratamento farmacológico , Infarto do Miocárdio/patologia , Infarto do Miocárdio/fisiopatologia , Miocárdio/patologia , Neovascularização Fisiológica/efeitos dos fármacos , Fosfatidilinositol 3-Quinases/genética , Inibidores de Fosfoinositídeo-3 Quinase , Inibidores de Proteínas Quinases/farmacologia , Proteínas Proto-Oncogênicas c-akt/metabolismo , Quinoxalinas/farmacologia , Interferência de RNA , Recuperação de Função Fisiológica , Regeneração/efeitos dos fármacos , Transdução de Sinais , Tiazolidinedionas/farmacologia , Fatores de Tempo , Transfecção , Função Ventricular Esquerda
12.
Circ Res ; 107(10): 1275-89, 2010 Nov 12.
Artigo em Inglês | MEDLINE | ID: mdl-20847309

RESUMO

RATIONALE: Mechanotransduction and the response to biomechanical stress is a fundamental response in heart disease. Loss of phosphoinositide 3-kinase (PI3K)γ, the isoform linked to G protein-coupled receptor signaling, results in increased myocardial contractility, but the response to pressure overload is controversial. OBJECTIVE: To characterize molecular and cellular responses of the PI3Kγ knockout (KO) mice to biomechanical stress. METHODS AND RESULTS: In response to pressure overload, PI3KγKO mice deteriorated at an accelerated rate compared with wild-type mice despite increased basal myocardial contractility. These functional responses were associated with compromised phosphorylation of Akt and GSK-3α. In contrast, isolated single cardiomyocytes from banded PI3KγKO mice maintained their hypercontractility, suggesting compromised interaction with the extracellular matrix as the primary defect in the banded PI3KγKO mice. ß-Adrenergic stimulation increased cAMP levels with increased phosphorylation of CREB, leading to increased expression of cAMP-responsive matrix metalloproteinases (MMPs), MMP2, MT1-MMP, and MMP13 in cardiomyocytes and cardiofibroblasts. Loss of PI3Kγ resulted in increased cAMP levels with increased expression of MMP2, MT1-MMP, and MMP13 and increased MMP2 activation and collagenase activity in response to biomechanical stress. Selective loss of N-cadherin from the adhesion complexes in the PI3KγKO mice resulted in reduced cell adhesion. The ß-blocker propranolol prevented the upregulation of MMPs, whereas MMP inhibition prevented the adverse remodeling with both therapies, preventing the functional deterioration in banded PI3KγKO mice. In banded wild-type mice, long-term propranolol prevented the adverse remodeling and systolic dysfunction with preservation of the N-cadherin levels. CONCLUSIONS: The enhanced propensity to develop heart failure in the PI3KγKO mice is attributable to a cAMP-dependent upregulation of MMP expression and activity and disorganization of the N-cadherin/ß-catenin cell adhesion complex. ß-Blocker therapy prevents these changes thereby providing a novel mechanism of action for these drugs.


Assuntos
Antígenos CD/metabolismo , Caderinas/metabolismo , Cardiomegalia/enzimologia , Classe Ib de Fosfatidilinositol 3-Quinase/metabolismo , AMP Cíclico/metabolismo , Matriz Extracelular/metabolismo , Metaloproteinases da Matriz/metabolismo , Mecanotransdução Celular , Miocárdio/enzimologia , Remodelação Ventricular , Antagonistas Adrenérgicos beta/administração & dosagem , Animais , Fenômenos Biomecânicos , Cardiomegalia/tratamento farmacológico , Cardiomegalia/fisiopatologia , Adesão Celular , Células Cultivadas , Classe Ib de Fosfatidilinositol 3-Quinase/deficiência , Classe Ib de Fosfatidilinositol 3-Quinase/genética , Proteína de Ligação ao Elemento de Resposta ao AMP Cíclico/metabolismo , Modelos Animais de Doenças , Fibroblastos/enzimologia , Quinase 3 da Glicogênio Sintase/metabolismo , Insuficiência Cardíaca/enzimologia , Insuficiência Cardíaca/fisiopatologia , Insuficiência Cardíaca/prevenção & controle , Masculino , Metaloproteinase 13 da Matriz/metabolismo , Metaloproteinase 14 da Matriz/metabolismo , Metaloproteinase 2 da Matriz/metabolismo , Mecanotransdução Celular/efeitos dos fármacos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Contração Miocárdica , Miocárdio/patologia , Miócitos Cardíacos/enzimologia , Fosforilação , Propranolol/administração & dosagem , Proteínas Proto-Oncogênicas c-akt/metabolismo , Estresse Mecânico , Fatores de Tempo , Remodelação Ventricular/efeitos dos fármacos , beta Catenina/metabolismo
13.
Bioessays ; 32(3): 185-196, 2010 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-20162662

RESUMO

In the past decade, the availability of genetically modified animals has enabled the discovery of interesting roles for phosphatidylinositol 3-kinase-gamma (PI3Kgamma) and -delta (PI3Kdelta) in different cell types orchestrating innate and adaptive immune responses. Therefore, these PI3K isoforms appear to be attractive drug targets for the treatment of diseases caused by unrestrained immune reactions. Currently, pharmacological targeting of PI3Kgamma and/or PI3Kdelta represents one of the most promising challenges for companies interested in the development of novel safe treatments for inflammatory diseases. In this review we provide a general outline of PI3Kgamma- and PI3Kdelta-specific functions in distinct subsets of inflammatory cells. We also discuss the therapeutic impact of novel compounds targeting PI3Kgamma, PI3Kdelta or both, in mouse models of autoimmune disorders (systemic lupus erythematosus (SLE) and rheumatoid arthritis), respiratory diseases (allergic asthma and chronic obstructive pulmonary disease) and cardiovascular dysfunctions (atherosclerosis and myocardial infarction).


Assuntos
Inflamação , Inibidores de Fosfoinositídeo-3 Quinase , Subunidades Proteicas/antagonistas & inibidores , Animais , Asma/tratamento farmacológico , Doenças Autoimunes/tratamento farmacológico , Linfócitos B/imunologia , Doenças Cardiovasculares/tratamento farmacológico , Quimiotaxia de Leucócito/fisiologia , Eosinófilos/imunologia , Humanos , Sistema Imunitário/enzimologia , Inflamação/tratamento farmacológico , Inflamação/enzimologia , Leucócitos/imunologia , Mastócitos/imunologia , Fosfatidilinositol 3-Quinases/metabolismo , Subunidades Proteicas/metabolismo , Doença Pulmonar Obstrutiva Crônica/tratamento farmacológico , Transdução de Sinais/imunologia , Linfócitos T/imunologia
14.
Curr Heart Fail Rep ; 8(3): 168-75, 2011 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-21519914

RESUMO

Cardiac pathophysiology heavily relies on receptor-mediated signal transduction, and pharmacologic control of such biological processes has proven successful in preventing and treating multiple heart diseases. Recent progress in the study of receptor-mediated signal transduction events in the heart highlighted the role of a family of lipid kinases known as phosphoinositide 3-kinases (PI3Ks). These enzymes are involved downstream different receptors in the production of a lipid second messenger molecule (namely phosphatidylinositol (3,4,5)-trisphosphate [PIP(3)]), which mediates a large number of biological responses critical for the heart, including cardiomyocyte growth, survival, and contractility as well as cardiovascular inflammation. This review focuses on the recent advances in the understanding of PI3K function in cardiac pathophysiology obtained by studying mouse mutants for different PI3K genes and by validating the effects of PI3K pharmacologic inhibition in preclinical models of critical cardiac diseases like heart failure.


Assuntos
Insuficiência Cardíaca/fisiopatologia , Fosfatidilinositol 3-Quinases/fisiologia , Transdução de Sinais/fisiologia , Animais , Sobrevivência Celular/fisiologia , Modelos Animais de Doenças , Insuficiência Cardíaca/enzimologia , Inflamação/fisiopatologia , Camundongos , Camundongos Transgênicos , Contração Miocárdica/fisiologia , Receptores Adrenérgicos beta/fisiologia , Sistemas do Segundo Mensageiro/fisiologia
15.
J Cardiovasc Pharmacol ; 56(6): 651-8, 2010 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-20881611

RESUMO

BACKGROUND: Phosphoinositide 3-kinase gamma is upregulated in the heart during acute myocardial infarction (AMI) potentially contributing to the development and maintenance of heart failure. METHODS: CD-1 male mice were randomly assigned to pharmacologic inhibition of phosphoinositide 3-kinase gamma using AS-605240 (10 mg/kg/day intraperitoneally) or vehicle (NaCl 0.9% + DMSO 25% solution) for 14 days after experimental AMI induced by surgical coronary artery ligation. Echocardiography was performed at baseline and 1, 7, 14, and 28 days after surgery to measure left ventricular dimensions and function. Infarct size was also measured at weekly intervals to evaluate for infarct resorption. RESULTS: When compared with vehicle-treated mice over the 4-week period, animals treated with AS-605240 showed a smaller increase in left ventricular cavitary dimensions, a smaller decrease in left ventricular systolic function (P < 0.05), and a significant increase in posterior wall diastolic and systolic thickness reflective of compensatory hypertrophy (P < 0.05). Initial infarct size (measured at 24 hours) was not different comparing AS-605240 (29% ± 4%) and vehicle-treated mice (31% ± 1%, P = nonsignificant). At 4 weeks after AMI, infarct size was significantly smaller in the AS-605240-treated mice (14% ± 2%) compared with vehicle-treated mice (28% ± 3%, P < 0.001), reflecting greater infarct resorption. CONCLUSIONS: Phosphoinositide 3-kinase gamma inhibition with AS-605240 after AMI leads to enhanced infarct resorption, greater compensatory hypertrophy of the nonischemic myocardium, and more favorable cardiac remodeling and function.


Assuntos
Infarto do Miocárdio/tratamento farmacológico , Infarto do Miocárdio/enzimologia , Inibidores de Fosfoinositídeo-3 Quinase , Quinoxalinas/farmacologia , Quinoxalinas/uso terapêutico , Tiazolidinedionas/farmacologia , Tiazolidinedionas/uso terapêutico , Remodelação Ventricular/efeitos dos fármacos , Animais , Classe Ib de Fosfatidilinositol 3-Quinase/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos ICR , Remodelação Ventricular/fisiologia
16.
Aging Cell ; 19(6): e13159, 2020 06.
Artigo em Inglês | MEDLINE | ID: mdl-32441410

RESUMO

Heart failure with preserved ejection fraction (HFpEF) is the most common type of HF in older adults. Although no pharmacological therapy has yet improved survival in HFpEF, exercise training (ExT) has emerged as the most effective intervention to improving functional outcomes in this age-related disease. The molecular mechanisms by which ExT induces its beneficial effects in HFpEF, however, remain largely unknown. Given the strong association between aging and HFpEF, we hypothesized that ExT might reverse cardiac aging phenotypes that contribute to HFpEF pathophysiology and additionally provide a platform for novel mechanistic and therapeutic discovery. Here, we show that aged (24-30 months) C57BL/6 male mice recapitulate many of the hallmark features of HFpEF, including preserved left ventricular ejection fraction, subclinical systolic dysfunction, diastolic dysfunction, impaired cardiac reserves, exercise intolerance, and pathologic cardiac hypertrophy. Similar to older humans, ExT in old mice improved exercise capacity, diastolic function, and contractile reserves, while reducing pulmonary congestion. Interestingly, RNAseq of explanted hearts showed that ExT did not significantly modulate biological pathways targeted by conventional HF medications. However, it reversed multiple age-related pathways, including the global downregulation of cell cycle pathways seen in aged hearts, which was associated with increased capillary density, but no effects on cardiac mass or fibrosis. Taken together, these data demonstrate that the aged C57BL/6 male mouse is a valuable model for studying the role of aging biology in HFpEF pathophysiology, and provide a molecular framework for how ExT potentially reverses cardiac aging phenotypes in HFpEF.


Assuntos
Envelhecimento/genética , Ecocardiografia Doppler/métodos , Exercício Físico/fisiologia , Insuficiência Cardíaca/fisiopatologia , Volume Sistólico/fisiologia , Animais , Humanos , Masculino , Camundongos , Fenótipo
17.
Arterioscler Thromb Vasc Biol ; 28(1): 68-76, 2008 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-17962628

RESUMO

OBJECTIVE: We evaluated whether phosphatidylinositol 3-kinase gamma (PI3Kgamma) plays a role in reparative neovascularization and endothelial progenitor cell (EPC) function. METHODS AND RESULTS: Unilateral limb ischemia was induced in mice lacking the PI3Kgamma gene (PI3Kgamma-/-) or expressing a catalytically inactive mutant (PI3Kgamma(KD/KD)) and wild-type controls (WT). Capillarization and arteriogenesis were reduced in PI3Kgamma-/- ischemic muscles resulting in delayed reperfusion compared with WT, whereas reparative neovascularization was preserved in PI3Kgamma(KD/KD). In PI3Kgamma-/- muscles, endothelial cell proliferation was reduced, apoptosis was increased, and interstitial space was infiltrated with leukocytes but lacked cKit+ progenitor cells that in WT muscles typically surrounded arterioles. PI3Kgamma is constitutively expressed by WT EPCs, with expression levels being upregulated by hypoxia. PI3Kgamma-/- EPCs showed a defect in proliferation, survival, integration into endothelial networks, and migration toward SDF-1. The dysfunctional phenotype was associated with nuclear constraining of FOXO1, reduced Akt and eNOS phosphorylation, and decreased nitric oxide (NO) production. Pretreatment with an NO donor corrected the migratory defect of PI3Kgamma-/- EPCs. PI3Kgamma(KD/KD) EPCs showed reduced Akt phosphorylation, but constitutive activation of eNOS and preserved proliferation, survival, and migration. CONCLUSIONS: We newly demonstrated that PI3Kgamma modulates angiogenesis, arteriogenesis, and vasculogenesis by mechanisms independent from its kinase activity.


Assuntos
Células Endoteliais/fisiologia , Isquemia/fisiopatologia , Neovascularização Fisiológica/fisiologia , Fosfatidilinositol 3-Quinases/fisiologia , Células-Tronco/fisiologia , Animais , Classe Ib de Fosfatidilinositol 3-Quinase , Modelos Animais de Doenças , Extremidades/irrigação sanguínea , Isoenzimas/genética , Isoenzimas/fisiologia , Masculino , Camundongos , Camundongos Knockout , Músculo Liso/fisiologia , Fosfatidilinositol 3-Quinases/genética , Transplantes
18.
PLoS One ; 14(8): e0220717, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31404087

RESUMO

Silencing Mediator of Retinoid and Thyroid Hormone Receptors (SMRT) and the nuclear receptor co-repressor1 (NCoR1) are paralogs and regulate nuclear receptor (NR) function through the recruitment of a multiprotein complex that includes histone deacetylase activity. Previous genetic strategies which deleted SMRT in a specific tissue or which altered the interaction between SMRT and NRs have suggested that it may regulate adiposity and insulin sensitivity. However, the full role of SMRT in adult mice has been difficult to establish because its complete deletion during embryogenesis is lethal. To elucidate the specific roles of SMRT in mouse target tissues especially in the context of thyroid hormone (TH) signaling, we used a tamoxifen-inducible post-natal disruption strategy. We found that global SMRT deletion causes dramatic obesity even though mice were fed a standard chow diet and exhibited normal food intake. This weight gain was associated with a decrease in energy expenditure. Interestingly, the deletion of SMRT had no effect on TH action in any tissue but did regulate retinoic acid receptor (RAR) function in the liver. We also demonstrate that the deletion of SMRT leads to profound hepatic steatosis in the setting of obesity. This is unlike NCoR1 deletion, which results in hepatic steatosis due to the upregulation of lipogenic gene expression. Taken together, our data demonstrate that SMRT plays a unique and CoR specific role in the regulation of body weight and has no role in TH action. This raises the possibility that additional role of CoRs besides NCoR1 and SMRT may exist to regulate TH action.


Assuntos
Peso Corporal/fisiologia , Correpressor 2 de Receptor Nuclear/fisiologia , Hormônios Tireóideos/fisiologia , Animais , Western Blotting , Colesterol/análise , Ecocardiografia , Metabolismo Energético , Teste de Tolerância a Glucose , Lipídeos/sangue , Fígado/química , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Reação em Cadeia da Polimerase em Tempo Real , Hormônios Tireóideos/sangue , Tireotropina/sangue , Tireotropina/fisiologia , Tiroxina/sangue , Tiroxina/fisiologia , Triglicerídeos/análise , Aumento de Peso/fisiologia
19.
Sci Transl Med ; 11(482)2019 03 06.
Artigo em Inglês | MEDLINE | ID: mdl-30842316

RESUMO

Activin type II receptor (ActRII) ligands have been implicated in muscle wasting in aging and disease. However, the role of these ligands and ActRII signaling in the heart remains unclear. Here, we investigated this catabolic pathway in human aging and heart failure (HF) using circulating follistatin-like 3 (FSTL3) as a potential indicator of systemic ActRII activity. FSTL3 is a downstream regulator of ActRII signaling, whose expression is up-regulated by the major ActRII ligands, activin A, circulating growth differentiation factor-8 (GDF8), and GDF11. In humans, we found that circulating FSTL3 increased with aging, frailty, and HF severity, correlating with an increase in circulating activins. In mice, increasing circulating activin A increased cardiac ActRII signaling and FSTL3 expression, as well as impaired cardiac function. Conversely, ActRII blockade with either clinical-stage inhibitors or genetic ablation reduced cardiac ActRII signaling while restoring or preserving cardiac function in multiple models of HF induced by aging, sarcomere mutation, or pressure overload. Using unbiased RNA sequencing, we show that activin A, GDF8, and GDF11 all induce a similar pathologic profile associated with up-regulation of the proteasome pathway in mammalian cardiomyocytes. The E3 ubiquitin ligase, Smurf1, was identified as a key downstream effector of activin-mediated ActRII signaling, which increased proteasome-dependent degradation of sarcoplasmic reticulum Ca2+ ATPase (SERCA2a), a critical determinant of cardiomyocyte function. Together, our findings suggest that increased activin/ActRII signaling links aging and HF pathobiology and that targeted inhibition of this catabolic pathway holds promise as a therapeutic strategy for multiple forms of HF.


Assuntos
Receptores de Activinas Tipo II/metabolismo , Envelhecimento/metabolismo , Insuficiência Cardíaca/metabolismo , Miocárdio/metabolismo , Miocárdio/patologia , Transdução de Sinais , Ativinas/sangue , Adulto , Idoso , Idoso de 80 Anos ou mais , Envelhecimento/sangue , Animais , Constrição Patológica , Modelos Animais de Doenças , Proteínas Relacionadas à Folistatina/metabolismo , Fragilidade , Insuficiência Cardíaca/sangue , Insuficiência Cardíaca/patologia , Insuficiência Cardíaca/fisiopatologia , Ventrículos do Coração/patologia , Ventrículos do Coração/fisiopatologia , Humanos , Ligantes , Masculino , Camundongos Endogâmicos C57BL , Pessoa de Meia-Idade , Miócitos Cardíacos/metabolismo , Pressão , Complexo de Endopeptidases do Proteassoma/metabolismo , Proteólise , Ratos , ATPases Transportadoras de Cálcio do Retículo Sarcoplasmático , Índice de Gravidade de Doença , Sístole
20.
J Clin Invest ; 126(8): 2989-3005, 2016 08 01.
Artigo em Inglês | MEDLINE | ID: mdl-27348588

RESUMO

Hypertrophic cardiomyopathy is a common cause of mortality in congenital heart disease (CHD). Many gene abnormalities are associated with cardiac hypertrophy, but their function in cardiac development is not well understood. Loss-of-function mutations in PTPN11, which encodes the protein tyrosine phosphatase (PTP) SHP2, are implicated in CHD and cause Noonan syndrome with multiple lentigines (NSML), a condition that often presents with cardiac hypertrophic defects. Here, we found that NSML-associated hypertrophy stems from aberrant signaling mechanisms originating in developing endocardium. Trabeculation and valvular hyperplasia were diminished in hearts of embryonic mice expressing a human NSML-associated variant of SHP2, and these defects were recapitulated in mice expressing NSML-associated SHP2 specifically in endothelial, but not myocardial or neural crest, cells. In contrast, mice with myocardial- but not endothelial-specific NSML SHP2 expression developed ventricular septal defects, suggesting that NSML-associated mutations have both cell-autonomous and nonautonomous functions in cardiac development. However, only endothelial-specific expression of NSML-associated SHP2 induced adult-onset cardiac hypertrophy. Further, embryos expressing the NSML-associated SHP2 mutation exhibited aberrant AKT activity and decreased downstream forkhead box P1 (FOXP1)/FGF and NOTCH1/EPHB2 signaling, indicating that SHP2 is required for regulating reciprocal crosstalk between developing endocardium and myocardium. Together, our data provide functional and disease-based evidence that aberrant SHP2 signaling during cardiac development leads to CHD and adult-onset heart hypertrophy.


Assuntos
Cardiomegalia/metabolismo , Regulação da Expressão Gênica , Lentigo/metabolismo , Síndrome de Noonan/metabolismo , Proteína Tirosina Fosfatase não Receptora Tipo 11/metabolismo , Animais , Apoptose , Linhagem da Célula , Modelos Animais de Doenças , Endocárdio/metabolismo , Feminino , Heterozigoto , Homozigoto , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Mutação , Miocárdio/metabolismo , Miócitos Cardíacos/metabolismo , Transdução de Sinais
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA