Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
1.
Sci Rep ; 12(1): 8852, 2022 05 25.
Artigo em Inglês | MEDLINE | ID: mdl-35614155

RESUMO

Renewal of the myocardium by preexisting cardiomyocytes is a powerful strategy for restoring the architecture and function of hearts injured by myocardial infarction. To advance this strategy, we show that combining two clinically approved drugs, but neither alone, muscularizes the heart through cardiomyocyte proliferation. Specifically, in adult murine cardiomyocytes, metoprolol, a cardioselective ß1-adrenergic receptor blocker, when given with triiodothyronine (T3, a thyroid hormone) accentuates the ability of T3 to stimulate ERK1/2 phosphorylation and proliferative signaling by inhibiting expression of the nuclear phospho-ERK1/2-specific phosphatase, dual-specificity phosphatase-5. While short-duration metoprolol plus T3 therapy generates new heart muscle in healthy mice, in mice with myocardial infarction-induced left ventricular dysfunction and pathological remodeling, it remuscularizes the heart, restores contractile function and reverses chamber dilatation; outcomes that are enduring. If the beneficial effects of metoprolol plus T3 are replicated in humans, this therapeutic strategy has the potential to definitively address ischemic heart failure.


Assuntos
Infarto do Miocárdio , Disfunção Ventricular Esquerda , Antagonistas de Receptores Adrenérgicos beta 1/farmacologia , Antagonistas de Receptores Adrenérgicos beta 1/uso terapêutico , Animais , Metoprolol/farmacologia , Metoprolol/uso terapêutico , Camundongos , Infarto do Miocárdio/patologia , Miocárdio/metabolismo , Miócitos Cardíacos/metabolismo , Tri-Iodotironina/metabolismo , Tri-Iodotironina/farmacologia , Disfunção Ventricular Esquerda/patologia , Remodelação Ventricular
2.
Theranostics ; 11(10): 4790-4808, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33754028

RESUMO

Rationale: Doxorubicin is a widely used anticancer drug. However, its major side effect, cardiotoxicity, results from cardiomyocyte loss that causes left ventricle (LV) wall thinning, chronic LV dysfunction and heart failure. Cardiomyocyte number expansion by thyroid hormone (T3) during preadolescence is suppressed by the developmental induction of an ERK1/2-specific dual specificity phosphatase 5 (DUSP5). Here, we sought to determine if a brief course of combined DUSP5 suppression plus T3 therapy replaces cardiomyocytes lost due to preexisting doxorubicin injury and reverses heart failure. Methods: We used in vivo-jetPEI to deliver DUSP5 or scrambled siRNA to ~5-week-old C57BL6 mice followed by 5 daily injections of T3 (2 ng/µg body weight). Genetic lineage tracing using Myh6-MerCreMer::Rosa26fs-Confetti mice and direct cardiomyocyte number counting, along with cell cycle inhibition (danusertib), was used to test if this treatment leads to de novo cardiomyocyte generation and improves LV contractile function. Three doses of doxorubicin (20 µg/g) given at 2-weekly intervals, starting at 5-weeks of age in C57BL6 mice, caused severe heart failure, as evident by a decrease in LV ejection fraction. Mice with an ~40 percentage point decrease in LVEF post-doxorubicin injury were randomized to receive either DUSP5 siRNA plus T3, or scrambled siRNA plus vehicle for T3. Age-matched mice without doxorubicin injury served as controls. Results: In uninjured adult mice, transient therapy with DUSP5 siRNA and T3 increases cardiomyocyte numbers, which is required for the associated increase in LV contractile function, since both are blocked by danusertib. In mice with chronic doxorubicin injury, DUSP5 siRNA plus T3 therapy rebuilds LV muscle by increasing cardiomyocyte numbers, which reverses LV dysfunction and prevents progressive chamber dilatation. Conclusion: RNA therapies are showing great potential. Importantly, a GMP compliant in vivo-jetPEI system for delivery of siRNA is already in use in humans, as is T3. Given these considerations, our findings provide a potentially highly translatable strategy for addressing doxorubicin cardiomyopathy, a currently untreatable condition.


Assuntos
Fosfatases de Especificidade Dupla/genética , Contração Miocárdica/efeitos dos fármacos , Miócitos Cardíacos/efeitos dos fármacos , Tri-Iodotironina/farmacologia , Função Ventricular Esquerda/efeitos dos fármacos , Animais , Antibióticos Antineoplásicos/toxicidade , Benzamidas/farmacologia , Cardiotoxicidade/etiologia , Contagem de Células , Proliferação de Células/efeitos dos fármacos , Proliferação de Células/genética , Doxorrubicina/toxicidade , Fosfatases de Especificidade Dupla/antagonistas & inibidores , Fator de Crescimento Insulin-Like I/metabolismo , Camundongos , Contração Miocárdica/genética , Miócitos Cardíacos/citologia , Inibidores de Proteínas Quinases/farmacologia , Pirazóis/farmacologia , RNA Interferente Pequeno , Disfunção Ventricular Esquerda/induzido quimicamente , Função Ventricular Esquerda/genética , Remodelação Ventricular/efeitos dos fármacos , Remodelação Ventricular/genética
3.
Sci Rep ; 10(1): 21918, 2020 12 14.
Artigo em Inglês | MEDLINE | ID: mdl-33318551

RESUMO

Cardiomyocytes of newborn mice proliferate after injury or exposure to growth factors. However, these responses are diminished after postnatal day-6 (P6), representing a barrier to building new cardiac muscle in adults. We have previously shown that exogenous thyroid hormone (T3) stimulates cardiomyocyte proliferation in P2 cardiomyocytes, by activating insulin-like growth factor-1 receptor (IGF-1R)-mediated ERK1/2 signaling. But whether exogenous T3 functions as a mitogen in post-P6 murine hearts is not known. Here, we show that exogenous T3 increases the cardiomyocyte endowment of P8 hearts, but the proliferative response is confined to cardiomyocytes of the left ventricular (LV) apex. Exogenous T3 stimulates proliferative ERK1/2 signaling in apical cardiomyocytes, but not in those of the LV base, which is inhibited by expression of the nuclear phospho-ERK1/2-specific dual-specificity phosphatase, DUSP5. Developmentally, between P7 and P14, DUSP5 expression increases in the myocardium from the LV base to its apex; after this period, it is uniformly expressed throughout the LV. In young adult hearts, exogenous T3 increases cardiomyocyte numbers after DUSP5 depletion, which might be useful for eliciting cardiac regeneration.


Assuntos
Fosfatases de Especificidade Dupla/biossíntese , Regulação Enzimológica da Expressão Gênica/efeitos dos fármacos , Ventrículos do Coração/enzimologia , Miocárdio/enzimologia , Miócitos Cardíacos/enzimologia , Tri-Iodotironina/farmacologia , Animais , Sistema de Sinalização das MAP Quinases , Camundongos , Camundongos Transgênicos , Proteína Quinase 1 Ativada por Mitógeno/metabolismo , Proteína Quinase 3 Ativada por Mitógeno/metabolismo
4.
Sci Rep ; 9(1): 17731, 2019 11 27.
Artigo em Inglês | MEDLINE | ID: mdl-31776360

RESUMO

Mitochondria-generated reactive oxygen species (mROS) are frequently associated with DNA damage and cell cycle arrest, but physiological increases in mROS serve to regulate specific cell functions. T3 is a major regulator of mROS, including hydrogen peroxide (H2O2). Here we show that exogenous thyroid hormone (T3) administration increases cardiomyocyte numbers in neonatal murine hearts. The mechanism involves signaling by mitochondria-generated H2O2 (mH2O2) acting via the redox sensor, peroxiredoxin-1, a thiol peroxidase with high reactivity towards H2O2 that activates c-Jun N-terminal kinase-2α2 (JNK2α2). JNK2α2, a relatively rare member of the JNK family of mitogen-activated protein kinases (MAPK), phosphorylates c-Jun, a component of the activator protein 1 (AP-1) early response transcription factor, resulting in enhanced insulin-like growth factor 1 (IGF-1) expression and activation of proliferative ERK1/2 signaling. This non-canonical mechanism of MAPK activation couples T3 actions on mitochondria to cell cycle activation. Although T3 is regarded as a maturation factor for cardiomyocytes, these studies identify a novel redox pathway that is permissive for T3-mediated cardiomyocyte proliferation-this because of the expression of a pro-proliferative JNK isoform that results in growth factor elaboration and ERK1/2 cell cycle activation.


Assuntos
Proliferação de Células , Sistema de Sinalização das MAP Quinases , Proteína Quinase 9 Ativada por Mitógeno/metabolismo , Miócitos Cardíacos/metabolismo , Hormônios Tireóideos/farmacologia , Animais , Células Cultivadas , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Fator de Crescimento Insulin-Like I/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Mitocôndrias Cardíacas/metabolismo , Miócitos Cardíacos/efeitos dos fármacos , Miócitos Cardíacos/fisiologia , Oxirredução , Peroxirredoxinas/metabolismo
6.
NPJ Regen Med ; 3: 6, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29507774

RESUMO

Stimulating regeneration of complex tissues and organs after injury to effect complete structural and functional repair, is an attractive therapeutic option that would revolutionize clinical medicine. Compared to many metazoan phyla that show extraordinary regenerative capacity, which in some instances persists throughout life, regeneration in mammalians, particularly humans, is limited or absent. Here we consider recent insights in the elucidation of molecular mechanisms of regeneration that have come from studies of tissue homeostasis and injury repair in mammalian tissues that span the spectrum from little or no self-renewal, to those showing active cell turnover throughout life. These studies highlight the diversity of factors that constrain regeneration, including immune responses, extracellular matrix composition, age, injury type, physiological adaptation, and angiogenic and neurogenic capacity. Despite these constraints, much progress has been made in elucidating key molecular mechanisms that may provide therapeutic targets for the development of future regenerative therapies, as well as previously unidentified developmental paradigms and windows-of-opportunity for improved regenerative repair.

7.
Int J Cardiol ; 250: 133-138, 2018 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-29169751

RESUMO

The advent of the subcutaneous implantable cardioverter-defibrillator (SQ-ICD) provides an alternative to transvenous and epicardial ICD therapy. Particularly germane to the young patient with congenital heart disease or inheritable arrhythmia syndromes, the SQ-ICD may be ideal for those who do not require permanent cardiac pacing. The serious complications associated with transvenous ICD systems are largely driven by the intravascular components of these devices and are avoided by this extravascular technique. Multiple clinical trials have shown that SQ-ICDs are effective in detecting and terminating ventricular arrhythmias, yet nuanced issues must be considered, especially in the context of congenital cardiovascular anomalies. This review aims to contextualize the role of this technology in contrast with traditional ICDs, and provide a logical approach to appropriate device selection.


Assuntos
Desfibriladores Implantáveis/normas , Cardiopatias Congênitas/diagnóstico por imagem , Cardiopatias Congênitas/terapia , Pericárdio/diagnóstico por imagem , Veia Subclávia/diagnóstico por imagem , Tela Subcutânea/diagnóstico por imagem , Fatores Etários , Criança , Desfibriladores Implantáveis/efeitos adversos , Desfibriladores Implantáveis/tendências , Humanos
8.
J Clin Lipidol ; 12(1): 110-115, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29198934

RESUMO

BACKGROUND: Hyperalphalipoproteinemia (HALP) is inversely correlated with coronary heart disease (CHD) although genetic variants associated with high serum levels of high-density lipoprotein cholesterol (HDL-C) have not been shown to be cardioprotective. OBJECTIVE: The objective of the study was to uncover novel genetic variants associated with HALP and possibly with reduced risk of CHD. METHODS: Exome sequencing data, HDL-C, and triglyceride levels were analyzed in 1645 subjects. They included the University of Maryland outpatients with high HDL-C (n = 12), Cardiovascular Health Study (n = 210), Jackson Heart Study (n = 402), Multi-Ethnic Study of Atherosclerosis (n = 404), Framingham Heart Study (n = 463), and Old Order Amish (n = 154). RESULTS: Novel nonsynonymous single-nucleotide polymorphisms (nsSNPs) were identified in men and women with primary HALP (mean HDL-C, 145 ± 30 mg/dL). Using PolyPhen-2 and Combined Annotation Dependent Depletion to estimate the predictive effect of each nsSNP on the gene product, rare, deleterious polymorphisms in UGT1A3, PLLP, PLEKHH1, ANK2, DIS3L, ACACB, and LRP4 were identified in 16 subjects with HALP but not in any tested subject with low HDL-C (<40 mg/dL). In addition, a single novel polymorphism, rs376849274, was found in OSBPL1A. The majority of these candidate genes have been implicated in fat and lipid metabolism, and none of these subjects has a history of CHD despite 75% of subjects having risk factors for CHD. Overall, the probability of finding these nsSNPs in a non-high HDL-C population ranges from 1 × 10-17 to 1 × 10-25. CONCLUSION: Novel functional polymorphisms in 8 candidate genes are associated with HALP in the absence of CHD. Future study is required to examine the extent to which these genes may affect HDL function and serve as potential therapeutic targets for CHD risk reduction.


Assuntos
Hiperlipoproteinemias/patologia , Polimorfismo de Nucleotídeo Único , Idoso , Idoso de 80 Anos ou mais , HDL-Colesterol/sangue , Feminino , Frequência do Gene , Glucuronosiltransferase/genética , Humanos , Hiperlipoproteinemias/epidemiologia , Hiperlipoproteinemias/genética , Masculino , Pessoa de Meia-Idade , Proteínas Proteolipídicas Associadas a Linfócitos e Mielina/genética , Linhagem , Fenótipo , Receptores de Esteroides/genética , Triglicerídeos/sangue , Sequenciamento do Exoma
11.
Echocardiography ; 31(6): 752-8, 2014 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-24372693

RESUMO

BACKGROUND: Patent foramen ovale (PFO) is a remnant of the fetal circulation present in 20% of the population. Right-to-left shunting (RLS) through a PFO has been linked to the pathophysiology of stroke, migraine with aura, and hypoxemia. While different imaging modalities including transcranial Doppler, intra-cardiac echo, and transthoracic echo (TTE) have often been used to detect RLS, transesophageal echo (TEE) bubble study remains the gold standard for diagnosing PFO. The aim of this study was to determine the relative accuracy of TEE in the detection of PFO. METHODS AND RESULTS: A systematic review of Medline, using a standard approach for meta-analysis, was performed for all prospective studies assessing accuracy of TEE in the detection of PFO using confirmation by autopsy, cardiac surgery, and/or catheterization as the reference. Search results revealed 3105 studies; 4 met inclusion criteria. A total of 164 patients were included. TEE had a weighted sensitivity of 89.2% (95% CI: 81.1-94.7%) and specificity of 91.4% (95% CI: 82.3-96.8%) to detect PFO. The overall positive likelihood ratio (LR+) was 5.93 (95% CI: 1.30-27.09) and the overall negative likelihood ratio (LR-) was 0.22 (95% CI: 0.08-0.56). CONCLUSION: While TEE bubble study is considered to be the gold standard modality for diagnosing PFO, some PFOs may still be missed or misdiagnosed. It is important to understand the limitations of TEE and perhaps use other highly sensitive screening tests, such as transcranial doppler (TCD), in conjunction with TEE before scheduling a patient for transcatheter PFO closure.


Assuntos
Ecocardiografia Transesofagiana/estatística & dados numéricos , Forame Oval Patente/diagnóstico por imagem , Forame Oval Patente/epidemiologia , Feminino , Humanos , Masculino , Prevalência , Reprodutibilidade dos Testes , Fatores de Risco , Sensibilidade e Especificidade
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA