Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 22
Filtrar
1.
Cell ; 186(14): 3013-3032.e22, 2023 07 06.
Artigo em Inglês | MEDLINE | ID: mdl-37352855

RESUMO

Mitochondrial DNA (mtDNA) is a potent agonist of the innate immune system; however, the exact immunostimulatory features of mtDNA and the kinetics of detection by cytosolic nucleic acid sensors remain poorly defined. Here, we show that mitochondrial genome instability promotes Z-form DNA accumulation. Z-DNA binding protein 1 (ZBP1) stabilizes Z-form mtDNA and nucleates a cytosolic complex containing cGAS, RIPK1, and RIPK3 to sustain STAT1 phosphorylation and type I interferon (IFN-I) signaling. Elevated Z-form mtDNA, ZBP1 expression, and IFN-I signaling are observed in cardiomyocytes after exposure to Doxorubicin, a first-line chemotherapeutic agent that induces frequent cardiotoxicity in cancer patients. Strikingly, mice lacking ZBP1 or IFN-I signaling are protected from Doxorubicin-induced cardiotoxicity. Our findings reveal ZBP1 as a cooperative partner for cGAS that sustains IFN-I responses to mitochondrial genome instability and highlight ZBP1 as a potential target in heart failure and other disorders where mtDNA stress contributes to interferon-related pathology.


Assuntos
Cardiotoxicidade , DNA Mitocondrial , Animais , Camundongos , DNA Mitocondrial/metabolismo , Imunidade Inata , Interferons/metabolismo , Nucleotidiltransferases/genética , Nucleotidiltransferases/metabolismo , Fosforilação
2.
Pflugers Arch ; 466(3): 451-7, 2014 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-24442121

RESUMO

Diastolic dysfunction prominently contributes to heart failure with preserved ejection fraction (HFpEF). Owing partly to inadequate understanding, HFpEF does not have any effective treatments. Cardiac myosin-binding protein-C (cMyBP-C), a component of the thick filament of heart muscle that can modulate cross-bridge attachment/detachment cycling process by its phosphorylation status, appears to be involved in the diastolic dysfunction associated with HFpEF. In patients, cMyBP-C mutations are associated with diastolic dysfunction even in the absence of hypertrophy. cMyBP-C deletion mouse models recapitulate diastolic dysfunction despite in vitro evidence of uninhibited cross-bridge cycling. Reduced phosphorylation of cMyBP-C is also associated with diastolic dysfunction in patients. Mouse models of reduced cMyBP-C phosphorylation exhibit diastolic dysfunction while cMyBP-C phosphorylation mimetic mouse models show enhanced diastolic function. Thus, cMyBP-C phosphorylation mediates diastolic function. Experimental results of both cMyBP-C deletion and reduced cMyBP-C phosphorylation causing diastolic dysfunction suggest that cMyBP-C phosphorylation level modulates cross-bridge detachment rate in relation to ongoing attachment rate to mediate relaxation. Consequently, alteration in cMyBP-C regulation of cross-bridge detachment is a key mechanism that causes diastolic dysfunction. Regardless of the exact molecular mechanism, ample clinical and experimental data show that cMyBP-C is a critical mediator of diastolic function. Furthermore, targeting cMyBP-C phosphorylation holds potential as a future treatment for diastolic dysfunction.


Assuntos
Proteínas de Transporte/metabolismo , Diástole , Insuficiência Cardíaca/metabolismo , Processamento de Proteína Pós-Traducional , Animais , Proteínas de Transporte/genética , Insuficiência Cardíaca/fisiopatologia , Humanos , Mutação , Fosforilação
3.
Am J Physiol Heart Circ Physiol ; 306(9): H1353-63, 2014 May.
Artigo em Inglês | MEDLINE | ID: mdl-24585781

RESUMO

Contraction and relaxation are fundamental aspects of cardiomyocyte functional biology. They reflect the response of the contractile machinery to the systolic increase and diastolic decrease of the cytoplasmic Ca(2+) concentration. The analysis of contractile function and Ca(2+) transients is therefore important to discriminate between myofilament responsiveness and changes in Ca(2+) homeostasis. This article describes an automated technology to perform sequential analysis of contractile force and Ca(2+) transients in up to 11 strip-format, fibrin-based rat, mouse, and human fura-2-loaded engineered heart tissues (EHTs) under perfusion and electrical stimulation. Measurements in EHTs under increasing concentrations of extracellular Ca(2+) and responses to isoprenaline and carbachol demonstrate that EHTs recapitulate basic principles of heart tissue functional biology. Ca(2+) concentration-response curves in rat, mouse, and human EHTs indicated different maximal twitch forces (0.22, 0.05, and 0.08 mN in rat, mouse, and human, respectively; P < 0.001) and different sensitivity to external Ca(2+) (EC50: 0.15, 0.39, and 1.05 mM Ca(2+) in rat, mouse, and human, respectively; P < 0.001) in the three groups. In contrast, no difference in myofilament Ca(2+) sensitivity was detected between skinned rat and human EHTs, suggesting that the difference in sensitivity to external Ca(2+) concentration is due to changes in Ca(2+) handling proteins. Finally, this study confirms that fura-2 has Ca(2+) buffering effects and is thereby changing the force response to extracellular Ca(2+).


Assuntos
Sinalização do Cálcio , Microscopia de Fluorescência/métodos , Contração Miocárdica , Miocárdio/citologia , Miócitos Cardíacos/metabolismo , Engenharia Tecidual/métodos , Animais , Automação Laboratorial , Diferenciação Celular , Humanos , Células-Tronco Pluripotentes Induzidas/citologia , Camundongos , Camundongos Endogâmicos C57BL , Microscopia de Fluorescência/instrumentação , Miocárdio/metabolismo , Miócitos Cardíacos/citologia , Miócitos Cardíacos/fisiologia , Ratos , Ratos Endogâmicos Lew , Ratos Wistar
4.
J Mol Cell Cardiol ; 53(5): 609-16, 2012 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-22850286

RESUMO

Phosphorylation of cardiac myosin binding protein-C (cMyBP-C) is a regulator of pump function in healthy hearts. However, the mechanisms of regulation by cAMP-dependent protein kinase (PKA)-mediated cMyBP-C phosphorylation have not been completely dissociated from other myofilament substrates for PKA, especially cardiac troponin I (cTnI). We have used synchrotron X-ray diffraction in skinned trabeculae to elucidate the roles of cMyBP-C and cTnI phosphorylation in myocardial inotropy and lusitropy. Myocardium in this study was isolated from four transgenic mouse lines in which the phosphorylation state of either cMyBP-C or cTnI was constitutively altered by site-specific mutagenesis. Analysis of peak intensities in X-ray diffraction patterns from trabeculae showed that cross-bridges are displaced similarly from the thick filament and toward actin (1) when both cMyBP-C and cTnI are phosphorylated, (2) when only cMyBP-C is phosphorylated, and (3) when cMyBP-C phosphorylation is mimicked by replacement with negative charge in its PKA sites. These findings suggest that phosphorylation of cMyBP-C relieves a constraint on cross-bridges, thereby increasing the proximity of myosin to binding sites on actin. Measurements of Ca(2+)-activated force in myocardium defined distinct molecular effects due to phosphorylation of cMyBP-C or co-phosphorylation with cTnI. Echocardiography revealed that mimicking the charge of cMyBP-C phosphorylation protects hearts from hypertrophy and systolic dysfunction that develops with constitutive dephosphorylation or genetic ablation, underscoring the importance of cMyBP-C phosphorylation for proper pump function.


Assuntos
Proteínas de Transporte/metabolismo , Proteínas Quinases Dependentes de AMP Cíclico/fisiologia , Miocárdio/enzimologia , Processamento de Proteína Pós-Traducional , Troponina I/metabolismo , Citoesqueleto de Actina/metabolismo , Substituição de Aminoácidos , Animais , Proteínas de Transporte/genética , Ventrículos do Coração/diagnóstico por imagem , Camundongos , Camundongos Transgênicos , Miocárdio/metabolismo , Fosforilação , Volume Sistólico , Troponina I/genética , Ultrassonografia , Função Ventricular Esquerda , Difração de Raios X
5.
Cells ; 11(11)2022 06 02.
Artigo em Inglês | MEDLINE | ID: mdl-35681523

RESUMO

Organ-on-a-chip (OOAC) is an emerging technology based on microfluid platforms and in vitro cell culture that has a promising future in the healthcare industry. The numerous advantages of OOAC over conventional systems make it highly popular. The chip is an innovative combination of novel technologies, including lab-on-a-chip, microfluidics, biomaterials, and tissue engineering. This paper begins by analyzing the need for the development of OOAC followed by a brief introduction to the technology. Later sections discuss and review the various types of OOACs and the fabrication materials used. The implementation of artificial intelligence in the system makes it more advanced, thereby helping to provide a more accurate diagnosis as well as convenient data management. We introduce selected OOAC projects, including applications to organ/disease modelling, pharmacology, personalized medicine, and dentistry. Finally, we point out certain challenges that need to be surmounted in order to further develop and upgrade the current systems.


Assuntos
Inteligência Artificial , Dispositivos Lab-On-A-Chip , Materiais Biocompatíveis , Microfluídica , Engenharia Tecidual
6.
Methods Mol Biol ; 2319: 15-24, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34331238

RESUMO

Mouse models are extensively studied and well-used to study cardiomyopathies due to the genetic relevance of this model organism and increasing need to identify therapeutic targets. Cardiac myofibril preparation for whole hearts has proved to be an invaluable in vitro method for determining the fundamental molecular mechanisms in heart failure. The technique described below consistently yields intact cardiac myofibrils, which can be used in subsequent techniques such as western blotting, immunofluorescence, and mass spectrometry. Here, we describe a method to optimize the separation and yield of cardiac myofibrils from murine whole tissue samples and preparation for subsequent mass spectrometry. This method allows for quick visual identification of multiple cardiac myofibril proteins.


Assuntos
Espectrometria de Massas/métodos , Miocárdio/metabolismo , Miofibrilas/metabolismo , Animais , Coração , Camundongos
7.
Methods Mol Biol ; 2319: 143-152, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34331252

RESUMO

Heart disease is one of the leading causes of death in the United States. Isolation and culture adult cardiomyocytes are important for studying cardiomyocyte contractility, heart hypertrophy, and cardiac failure. In contrast to neonatal cardiomyocyte isolation, adult mice cardiomyocytes isolation is challenging due to firm connections among cardiomyocytes through intercalated discs. The availability of newly generated genetically modified mouse lines requires to establish protocols to isolation and culture adult mouse cardiomyocyte for in vitro studies. In this manuscript, we described a straightforward method of isolating adult mouse cardiomyocytes using Langendorff perfusion apparatus. Briefly, the hearts were harvested from adult mice and the heart was mounted to Lagendorff apparatus. After perfusion with calcium depletion and collagenase digestion, the left ventricles were minced and filtered. Lastly, the separated cardiomyocytes were treated with CaCl2. The isolated cardiac myocytes can be utilized in a broad range of experiments including screening for drugs.


Assuntos
Técnicas de Cultura de Células/métodos , Separação Celular/instrumentação , Separação Celular/métodos , Preparação de Coração Isolado/instrumentação , Preparação de Coração Isolado/métodos , Miócitos Cardíacos/citologia , Animais , Cálcio/farmacologia , Cloreto de Cálcio/farmacologia , Células Cultivadas , Colagenases/química , Colagenases/farmacologia , Camundongos , Miócitos Cardíacos/metabolismo , Perfusão/métodos
8.
Methods Mol Biol ; 2319: 31-44, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34331240

RESUMO

Isolated cardiac tissue allows investigators to study mechanisms underlying normal and pathological conditions, which would otherwise be difficult or impossible to perform in vivo. In contrast to ventricular muscle strip preparations, papillary muscles can be prepared without severely damaging the muscle tissue. In this preparation, the isolated papillary muscle is fixed in an environmentally controlled organ bath chamber and electrically stimulated. The evoked twitch force is recorded using a pressure transducer, and parameters such as twitch force amplitude and twitch kinetics are analyzed. A variety of experimental protocols can be performed to investigate the calcium- and frequency-dependent contractility as well as dose-response curves of contractile agents, as well as simulation of pathologic conditions such as acute cardiac ischemia. Mouse papillary muscle preparations have long been the mainstay for studying interactions between intracellular calcium regulation and contractile responses under a number of simulated pathophysiological conditions. These studies are often used to complement in vitro studies performed using isolated neonatal rat cardiac myocytes. In this procedure, we describe how neonatal rat papillary muscles can also be prepared for use in contractile studies.


Assuntos
Contração Miocárdica/fisiologia , Miócitos Cardíacos/fisiologia , Músculos Papilares/fisiologia , Animais , Animais Recém-Nascidos , Estimulação Elétrica , Ratos , Equipamentos Cirúrgicos
9.
Sci Adv ; 7(22)2021 05.
Artigo em Inglês | MEDLINE | ID: mdl-34039599

RESUMO

Mitochondrial dysfunction is a key driver of inflammatory responses in human disease. However, it remains unclear whether alterations in mitochondria-innate immune cross-talk contribute to the pathobiology of mitochondrial disorders and aging. Using the polymerase gamma (POLG) mutator model of mitochondrial DNA instability, we report that aberrant activation of the type I interferon (IFN-I) innate immune axis potentiates immunometabolic dysfunction, reduces health span, and accelerates aging in mutator mice. Mechanistically, elevated IFN-I signaling suppresses activation of nuclear factor erythroid 2-related factor 2 (NRF2), which increases oxidative stress, enhances proinflammatory cytokine responses, and accelerates metabolic dysfunction. Ablation of IFN-I signaling attenuates hyperinflammatory phenotypes by restoring NRF2 activity and reducing aerobic glycolysis, which combine to lessen cardiovascular and myeloid dysfunction in aged mutator mice. These findings further advance our knowledge of how mitochondrial dysfunction shapes innate immune responses and provide a framework for understanding mitochondria-driven immunopathology in POLG-related disorders and aging.


Assuntos
DNA Mitocondrial , Interferon Tipo I , Envelhecimento/genética , Envelhecimento/metabolismo , Animais , DNA Mitocondrial/genética , DNA Mitocondrial/metabolismo , Inflamação/genética , Inflamação/metabolismo , Interferon Tipo I/metabolismo , Camundongos , Mitocôndrias/genética , Mitocôndrias/metabolismo , Mutação , Fator 2 Relacionado a NF-E2/genética , Fator 2 Relacionado a NF-E2/metabolismo
10.
Circ Res ; 103(9): 974-82, 2008 Oct 24.
Artigo em Inglês | MEDLINE | ID: mdl-18802026

RESUMO

Normal cardiac function requires dynamic modulation of contraction. beta1-adrenergic-induced protein kinase (PK)A phosphorylation of cardiac myosin binding protein (cMyBP)-C may regulate crossbridge kinetics to modulate contraction. We tested this idea with mechanical measurements and echocardiography in a mouse model lacking 3 PKA sites on cMyBP-C, ie, cMyBP-C(t3SA). We developed the model by transgenic expression of mutant cMyBP-C with Ser-to-Ala mutations on the cMyBP-C knockout background. Western blots, immunofluorescence, and in vitro phosphorylation combined to show that non-PKA-phosphorylatable cMyBP-C expressed at 74% compared to normal wild-type (WT) and was correctly positioned in the sarcomeres. Similar expression of WT cMyBP-C at 72% served as control, ie, cMyBP-C(tWT). Skinned myocardium responded to stretch with an immediate increase in force, followed by a transient relaxation of force and finally a delayed development of force, ie, stretch activation. The rate constants of relaxation, k(rel) (s-1), and delayed force development, k(df) (s-1), in the stretch activation response are indicators of crossbridge cycling kinetics. cMyBP-C(t3SA) myocardium had baseline k(rel) and k(df) similar to WT myocardium, but, unlike WT, k(rel) and k(df) were not accelerated by PKA treatment. Reduced dobutamine augmentation of systolic function in cMyBP-C(t3SA) hearts during echocardiography corroborated the stretch activation findings. Furthermore, cMyBP-C(t3SA) hearts exhibited basal echocardiographic findings of systolic dysfunction, diastolic dysfunction, and hypertrophy. Conversely, cMyBP-C(tWT) hearts performed similar to WT. Thus, PKA phosphorylation of cMyBP-C accelerates crossbridge kinetics and loss of this regulation leads to cardiac dysfunction.


Assuntos
Cardiomegalia/enzimologia , Proteínas de Transporte/metabolismo , Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , Contração Miocárdica , Miocárdio/enzimologia , Agonistas Adrenérgicos beta/farmacologia , Animais , Cardiomegalia/diagnóstico por imagem , Cardiomegalia/fisiopatologia , Proteínas de Transporte/genética , Dobutamina/farmacologia , Ecocardiografia Doppler , Humanos , Cinética , Mecanotransdução Celular , Camundongos , Camundongos Transgênicos , Força Muscular , Mutação , Contração Miocárdica/efeitos dos fármacos , Miocárdio/patologia , Miofibrilas/enzimologia , Fosforilação , Sarcômeros/enzimologia , Troponina I/metabolismo
11.
JACC Basic Transl Sci ; 4(7): 817-830, 2019 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-31998850

RESUMO

Cardiac myosin binding protein-C (cMyBP-C) phosphorylation prevents aging-related cardiac dysfunction. We tested this hypothesis by aging genetic mouse models of hypophosphorylated cMyBP-C, wild-type equivalent, and phosphorylated-mimetic cMyBP-C for 18 to 20 months. Phosphorylated-mimetic cMyBP-C mice exhibited better survival, better preservation of systolic and diastolic functions, and unchanging wall thickness. Wild-type equivalent mice showed decreasing cMyBP-C phosphorylation along with worsening cardiac function and hypertrophy approaching those found in hypophosphorylated cMyBP-C mice. Intact papillary muscle experiments suggested that cMyBP-C phosphorylation increased cross-bridge detachment rates as the underlying mechanism. Thus, phosphorylating cMyBP-C is a novel mechanism with potential to treat aging-related cardiac dysfunction.

12.
Clin Transl Med ; 6(1): 40, 2017 Nov 03.
Artigo em Inglês | MEDLINE | ID: mdl-29101495

RESUMO

BACKGROUND: Cdc42 is a member of the Rho GTPase family and functions as a molecular switch in regulating cytoskeleton remodeling and cell polarity establishment. Inactivating Cdc42 in cardiomyocytes resulted in embryonic lethality with heart developmental defects, including ventricular septum defects and thin ventricle wall syndrome. FINDINGS: In this study, we have generated a Cdc42 cardiomyocyte knockout mouse line by crossing Cdc42/flox mice with myosin light chain 2a (MLC2a)-Cre mice. We found that the deletion of Cdc42 in embryonic cardiomyocytes resulted in an underdeveloped right ventricle. Microarray analysis and real-time PCR data analysis displayed that the deletion of Cdc42 decreased dHand expression level. In addition, we found evaginations in the ventricle walls of Cdc42 knockout hearts. CONCLUSION: We concluded that Cdc42 plays an essential role in right ventricle growth.

13.
Am J Cardiol ; 120(9): 1501-1507, 2017 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-28847594

RESUMO

Cardiac myosin binding protein-C (cMyBP-C) is a heart muscle-specific thick filament protein. Elevated level of serum cMyBP-C is an indicator of early myocardial infarction (MI), but its value as a predictor of future cardiovascular disease is unknown. Based on the presence of significant amount of cMyBP-C in the serum of previous study subjects independent of MI, we hypothesized that circulating cMyBP-C is a sensitive indicator of ongoing cardiovascular stress and disease. To test this hypothesis, 75 men and 83 women of similar ages were recruited for a prospective study. They underwent exercise stress echocardiography to provide pre- and poststress blood samples for subsequent determination of serum cMyBP-C levels. The subjects were followed for 1 to 1.5 years. Exercise stress increased serum cMyBP-C in all subjects. Twenty-seven primary events (such as death, MI, revascularization, invasive cardiovascular procedure, or cardiovascular-related hospitalization) and 7 critical events (CE; such as death, MI, stroke, or pulmonary embolism) occurred. After adjusting for sex and cardiovascular risk factors with multivariate Cox regression, a 96% sensitive prestress cMyBP-C threshold carried a hazard ratio of 8.1 with p = 0.041 for primary events. Most subjects (6 of 7) who had CE showed normal ejection fraction on echocardiography. Prestress cMyBP-C demonstrated area under receiver operating curve of 0.91 and multivariate Cox regression hazard ratio of 13.8 (p = 0.000472) for CE. Thus, basal cMyBP-C levels reflected susceptibility for a variety of cardiovascular diseases. Together with its high sensitivity, cMyBP-C holds potential as a screening biomarker for the existence of severe cardiovascular diseases.


Assuntos
Doenças Cardiovasculares/sangue , Doenças Cardiovasculares/etiologia , Proteínas de Transporte/sangue , Idoso , Doenças Cardiovasculares/diagnóstico , Teste de Esforço , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Valor Preditivo dos Testes , Estudos Prospectivos , Curva ROC , Volume Sistólico
14.
J Am Coll Cardiol ; 70(18): 2290-2303, 2017 Oct 31.
Artigo em Inglês | MEDLINE | ID: mdl-29073958

RESUMO

Early-career academic cardiologists, who many believe are an important component of the future of cardiovascular care, face myriad challenges. The Early Career Section Academic Working Group of the American College of Cardiology, with senior leadership support, assessed the progress of this cohort from 2013 to 2016 with a global perspective. Data consisted of accessing National Heart, Lung, and Blood Institute public information, data from the American Heart Association and international organizations, and a membership-wide survey. Although the National Heart, Lung, and Blood Institute increased funding of career development grants, only a small number of early-career American College of Cardiology members have benefited as funding of the entire cohort has decreased. Personal motivation, institutional support, and collaborators continued to be positive influential factors. Surprisingly, mentoring ceased to correlate positively with obtaining external grants. The totality of findings suggests that the status of early-career academic cardiologists remains challenging; therefore, the authors recommend a set of attainable solutions.


Assuntos
Cardiologistas/educação , Cardiologia/educação , Escolha da Profissão , Mentores/educação , Cardiologistas/economia , Cardiologistas/tendências , Cardiologia/economia , Cardiologia/tendências , Humanos , Apoio à Pesquisa como Assunto/economia , Apoio à Pesquisa como Assunto/tendências
15.
ASAIO J ; 61(3): 253-8, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25923576

RESUMO

Cardiogenic shock (CS) causes significant morbidity and mortality and such patients can deteriorate rapidly. Temporary left ventricular assist devices (LVADs) are a promising approach to manage these patients. The following is a case series in which patients stabilized with a temporary LVAD for CS improvement were analyzed retrospectively. Between June 2011 and January 2014, 15 patients received temporary devices through an axillary approach (mean age: 53 ± 15, 93% male). Mean survival time was 317.8 ± 359.5 days (range: 6-936 days). During support there were no major bleeding events, infectious complications at the axillary access site, upper extremity edema, or emboli. The most of the patients recovered from CS (93%) were mobilized (67%) and were extubated (73%) while on temporary device support. Median times to extubation, intensive care unit discharge, and discontinuation of inotropic medications were: 1.63, 18, and 15 days, respectively. Four patients recovered to no device support and five received a long-term LVAD, all of whom remain alive. Therefore, implantation of a temporary LVAD through an axillary approach is a promising therapy for improving outcomes in patients needing mechanical circulatory support as a bridge to recovery or a definitive LVAD.


Assuntos
Coração Auxiliar , Implantação de Prótese/métodos , Choque Cardiogênico/cirurgia , Adulto , Idoso , Axila , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Estudos Retrospectivos , Resultado do Tratamento , Adulto Jovem
16.
Circ Heart Fail ; 8(3): 595-604, 2015 May.
Artigo em Inglês | MEDLINE | ID: mdl-25740838

RESUMO

BACKGROUND: Mammalian hearts exhibit positive inotropic responses to ß-adrenergic stimulation as a consequence of protein kinase A-mediated phosphorylation or as a result of increased beat frequency (the Bowditch effect). Several membrane and myofibrillar proteins are phosphorylated under these conditions, but the relative contributions of these to increased contractility are not known. Phosphorylation of cardiac myosin-binding protein-C (cMyBP-C) by protein kinase A accelerates the kinetics of force development in permeabilized heart muscle, but its role in vivo is unknown. Such understanding is important because adrenergic responsiveness of the heart and the Bowditch effect are both depressed in heart failure. METHODS AND RESULTS: The roles of cMyBP-C phosphorylation were studied using mice in which either WT or nonphosphorylatable forms of cMyBP-C [ser273ala, ser282ala, ser302ala: cMyBP-C(t3SA)] were expressed at similar levels on a cMyBP-C null background. Force and [Ca(2+)]in measurements in isolated papillary muscles showed that the increased force and twitch kinetics because increased pacing or ß1-adrenergic stimulation were nearly absent in cMyBP-C(t3SA) myocardium, even though [Ca(2+)]in transients under each condition were similar to WT. Biochemical measurements confirmed that protein kinase A phosphorylated ser273, ser282, and ser302 in WT cMyBP-C. In contrast, CaMKIIδ, which is activated by increased pacing, phosphorylated ser302 principally, ser282 to a lesser degree, and ser273 not at all. CONCLUSIONS: Phosphorylation of cMyBP-C increases the force and kinetics of twitches in living cardiac muscle. Further, cMyBP-C is a principal mediator of increased contractility observed with ß-adrenergic stimulation or increased pacing because of protein kinase A and CaMKIIδ phosphorylations of cMyB-C.


Assuntos
Agonistas de Receptores Adrenérgicos beta 1/farmacologia , Estimulação Cardíaca Artificial , Cardiotônicos/farmacologia , Proteínas de Transporte/metabolismo , Contração Miocárdica/efeitos dos fármacos , Músculos Papilares/efeitos dos fármacos , Animais , Sinalização do Cálcio/efeitos dos fármacos , Proteína Quinase Tipo 2 Dependente de Cálcio-Calmodulina/metabolismo , Proteínas de Transporte/genética , Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , Ativação Enzimática , Genótipo , Cinética , Camundongos Transgênicos , Força Muscular/efeitos dos fármacos , Mutação , Músculos Papilares/metabolismo , Fenótipo , Fosforilação , Processamento de Proteína Pós-Traducional
17.
J Am Coll Cardiol ; 66(16): 1816-1827, 2015 Oct 20.
Artigo em Inglês | MEDLINE | ID: mdl-26483107

RESUMO

Nurturing the development of cardiovascular physician-scientist investigators is critical for sustained progress in cardiovascular science and improving human health. The transition from an inexperienced trainee to an independent physician-scientist is a multifaceted process requiring a sustained commitment from the trainee, mentors, and institution. A cornerstone of this training process is a career development (K) award from the National Institutes of Health (NIH). These awards generally require 75% of the awardee's professional effort devoted to research aims and diverse career development activities carried out in a mentored environment over a 5-year period. We report on recent success rates for obtaining NIH K awards, provide strategies for preparing a successful application and navigating the early career period for aspiring cardiovascular investigators, and offer cardiovascular division leadership perspectives regarding K awards in the current era. Our objective is to offer practical advice that will equip trainees considering an investigator path for success.


Assuntos
Distinções e Prêmios , Mobilidade Ocupacional , National Institutes of Health (U.S.) , Médicos , Pesquisa Biomédica , Cardiologia , Humanos , Mentores , Médicos/economia , Pesquisadores , Estados Unidos
18.
Circ Heart Fail ; 8(3): 582-94, 2015 May.
Artigo em Inglês | MEDLINE | ID: mdl-25740839

RESUMO

BACKGROUND: Heart failure (HF) with preserved ejection fraction (HFpEF) accounts for ≈50% of all cases of HF and currently has no effective treatment. Diastolic dysfunction underlies HFpEF; therefore, elucidation of the mechanisms that mediate relaxation can provide new potential targets for treatment. Cardiac myosin-binding protein-C (cMyBP-C) is a thick filament protein that modulates cross-bridge cycling rates via alterations in its phosphorylation status. Thus, we hypothesize that phosphorylated cMyBP-C accelerates the rate of cross-bridge detachment, thereby enhancing relaxation to mediate diastolic function. METHODS AND RESULTS: We compared mouse models expressing phosphorylation-deficient cMyBP-C(S273A/S282A/S302A)-cMyBP-C(t3SA), phosphomimetic cMyBP-C(S273D/S282D/S302D)-cMyBP-C(t3SD), and wild-type-control cMyBP-C(tWT) to elucidate the functional effects of cMyBP-C phosphorylation. Decreased voluntary running distances, increased lung/body weight ratios, and increased brain natriuretic peptide levels in cMyBP-C(t3SA) mice demonstrate that phosphorylation deficiency is associated with signs of HF. Echocardiography (ejection fraction and myocardial relaxation velocity) and pressure/volume measurements (-dP/dtmin, pressure decay time constant τ-Glantz, and passive filling stiffness) show that cMyBP-C phosphorylation enhances myocardial relaxation in cMyBP-C(t3SD) mice, whereas deficient cMyBP-C phosphorylation causes diastolic dysfunction with HFpEF in cMyBP-C(t3SA) mice. Simultaneous force and [Ca(2+)]i measurements on intact papillary muscles show that enhancement of relaxation in cMyBP-C(t3SD) mice and impairment of relaxation in cMyBP-C(t3SA) mice are not because of altered [Ca(2+)]i handling, implicating that altered cross-bridge detachment rates mediate these changes in relaxation rates. CONCLUSIONS: cMyBP-C phosphorylation enhances relaxation, whereas deficient phosphorylation causes diastolic dysfunction and phenotypes resembling HFpEF. Thus, cMyBP-C is a potential target for treatment of HFpEF.


Assuntos
Proteínas de Transporte/metabolismo , Insuficiência Cardíaca/metabolismo , Disfunção Ventricular Esquerda/metabolismo , Função Ventricular Esquerda , Animais , Pressão Sanguínea , Proteínas de Transporte/genética , Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , Diástole , Genótipo , Insuficiência Cardíaca/genética , Insuficiência Cardíaca/fisiopatologia , Cinética , Camundongos Transgênicos , Mutação , Fenótipo , Fosforilação , Processamento de Proteína Pós-Traducional , Disfunção Ventricular Esquerda/genética , Disfunção Ventricular Esquerda/fisiopatologia
19.
J Am Coll Cardiol ; 63(21): 2199-208, 2014 Jun 03.
Artigo em Inglês | MEDLINE | ID: mdl-24703919

RESUMO

Early career academic cardiologists currently face unprecedented challenges that threaten a highly valued career path. A team consisting of early career professionals and senior leadership members of American College of Cardiology completed this white paper to inform the cardiovascular medicine profession regarding the plight of early career cardiologists and to suggest possible solutions. This paper includes: 1) definition of categories of early career academic cardiologists; 2) general challenges to all categories and specific challenges to each category; 3) obstacles as identified by a survey of current early career members of the American College of Cardiology; 4) major reasons for the failure of physician-scientists to receive funding from National Institute of Health/National Heart Lung and Blood Institute career development grants; 5) potential solutions; and 6) a call to action with specific recommendations.


Assuntos
Centros Médicos Acadêmicos/tendências , Cardiologia/tendências , Escolha da Profissão , National Heart, Lung, and Blood Institute (U.S.)/tendências , Médicos/tendências , Centros Médicos Acadêmicos/economia , Cardiologia/economia , Cardiologia/educação , Humanos , Mentores/educação , National Heart, Lung, and Blood Institute (U.S.)/economia , Médicos/economia , Estados Unidos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA