Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 56
Filtrar
Mais filtros

Base de dados
País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
Physiology (Bethesda) ; 39(5): 0, 2024 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-38713091

RESUMO

Insulin-like growth factor-1 (IGF-1) signaling has multiple physiological roles in cellular growth, metabolism, and aging. Myocardial hypertrophy, cell death, senescence, fibrosis, and electrical remodeling are hallmarks of various heart diseases and contribute to the progression of heart failure. This review highlights the critical role of IGF-1 and its cognate receptor in cardiac hypertrophy, aging, and remodeling.


Assuntos
Fator de Crescimento Insulin-Like I , Transdução de Sinais , Humanos , Fator de Crescimento Insulin-Like I/metabolismo , Animais , Transdução de Sinais/fisiologia , Receptor IGF Tipo 1/metabolismo , Miocárdio/metabolismo , Envelhecimento/metabolismo , Envelhecimento/fisiologia , Coração/fisiologia , Cardiomegalia/metabolismo , Cardiomegalia/fisiopatologia
2.
Biochem Biophys Res Commun ; 516(2): 350-356, 2019 08 20.
Artigo em Inglês | MEDLINE | ID: mdl-31208720

RESUMO

Receptor activator of NF-κB ligand (RANKL) is a member of the TNF superfamily. RANKL increases endothelial permeability and induces angiogenesis, suggesting its critical roles in the vasculature. Despite the evidence implicating RANKL in vascular pathology, its role in ischemic retinopathy has not been previously reported. In this study, neonatal mice were exposed to 75% oxygen from postnatal day (P)7 to P12 to induce vaso-obliteration, and then returned to room air from P12 to P17, causing the retina to become hypoxic and inducing vascular endothelial growth factor (VEGF) signaling, which produces pathological neovascularization. On P12, the mice received a single intravitreal injection of control IgG1 or RANK-Fc, and retinas were obtained at P17. On P17, RANKL was expressed strongly and selectively in the neovascular tufts (NVT) area. RANKL colocalized with αSMA or PDGFRß in NVT. However, co-immunostaining revealed that CD31-positive areas were not the same as RANKL, which indicates that RANKL might be produced by retinal pericytes, not endothelial cells. Consistent with this finding, chemical hypoxia upregulated RANKL expression in cultured human retinal pericytes but not in endothelial cells. Treatment with RANK-Fc markedly reduced the NVT area compared to that in mice administered the IgG1 injection. In contrast, the central avascular region of RANKL-Fc retina was comparable to the controls. In addition, we assessed retinal vascular permeability using FITC-labeled dextran. RANK-Fc treated mice displayed decreased vascular leakages compared to those injected with IgG1. Our work supports the use of an RANKL blockade as a potential therapeutic approach against ischemic retinopathies.


Assuntos
Isquemia/patologia , Neovascularização Patológica/patologia , Ligante RANK/antagonistas & inibidores , Doenças Retinianas/patologia , Animais , Animais Recém-Nascidos , Permeabilidade Capilar , Hipóxia Celular , Modelos Animais de Doenças , Células Endoteliais/metabolismo , Isquemia/complicações , Camundongos Endogâmicos C57BL , Neovascularização Patológica/complicações , Pericitos/metabolismo , Ligante RANK/metabolismo , Doenças Retinianas/complicações , Células Ganglionares da Retina/metabolismo
3.
Int J Cancer ; 143(10): 2458-2469, 2018 11 15.
Artigo em Inglês | MEDLINE | ID: mdl-30070361

RESUMO

Insulin and insulin-like growth factor (IGF)-1 signaling in the thyroid are thought to be permissive for the coordinated regulation by thyroid-stimulating hormone (TSH) of thyrocyte proliferation and hormone production. However, the integrated role of insulin receptor (IR) and IGF-1 receptor (IGF-1R) in thyroid development and function has not been explored. Here, we generated thyrocyte-specific IR and IGF-1R double knockout (DTIRKO) mice to precisely evaluate the coordinated functions of these receptors in the thyroid of neonates and adults. Neonatal DTIRKO mice displayed smaller thyroids, paralleling defective folliculogenesis associated with repression of the thyroid-specific transcription factor Foxe1. By contrast, at postnatal day 14, absence of IR and IGF-1R paradoxically induced thyrocyte proliferation, which was mediated by mTOR-dependent signaling pathways. Furthermore, we found elevated production of TSH during the development of follicular hyperplasia at 8 weeks of age. By 50 weeks, all DTIRKO mice developed papillary thyroid carcinoma (PTC)-like lesions that correlated with induction of the ErbB pathway. Taken together, these data define a critical role for IR and IGF-1R in neonatal thyroid folliculogenesis. They also reveal an important reciprocal relationship between IR/IGF-1R and TSH/ErbB signaling in the pathogenesis of thyroid follicular hyperplasia and, possibly, of papillary carcinoma.


Assuntos
Receptores ErbB/metabolismo , Receptor IGF Tipo 1/deficiência , Receptor de Insulina/deficiência , Câncer Papilífero da Tireoide/metabolismo , Células Epiteliais da Tireoide/metabolismo , Neoplasias da Glândula Tireoide/metabolismo , Animais , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Receptor IGF Tipo 1/genética , Receptor IGF Tipo 1/metabolismo , Receptor de Insulina/genética , Receptor de Insulina/metabolismo , Transdução de Sinais , Câncer Papilífero da Tireoide/patologia , Células Epiteliais da Tireoide/patologia , Neoplasias da Glândula Tireoide/patologia , Tireotropina/biossíntese , Tireotropina/metabolismo
4.
Biochim Biophys Acta Mol Basis Dis ; 1864(5 Pt B): 1931-1938, 2018 May.
Artigo em Inglês | MEDLINE | ID: mdl-28847512

RESUMO

Cardiovascular disease (CVD) is the leading cause of death in most developed countries. Aging is associated with enhanced risk of CVD. Insulin-like growth factor-1 (IGF-1) binds to its cognate receptor, IGF-1 receptor (IGF-1R), and exerts pleiotropic effects on cell growth, differentiation, development, and tissue repair. Importantly, IGF-1/IGF-1R signaling is implicated in cardiac aging and longevity. Cardiac aging is an intrinsic process that results in cardiac dysfunction, accompanied by molecular and cellular changes. In this review, we summarize the current state of knowledge regarding the link between the IGF-1/IGF-1R system and cardiac aging. The biological effects of IGF-1R and insulin receptor will be discussed and compared. Furthermore, we describe data regarding how deletion of IGF-1R in cardiomyocytes of aged knockout mice may delay the development of senescence-associated myocardial pathologies. This article is part of a Special issue entitled Cardiac adaptations to obesity, diabetes and insulin resistance, edited by Professors Jan F.C. Glatz, Jason R.B. Dyck and Christine Des Rosiers.


Assuntos
Envelhecimento/metabolismo , Doenças Cardiovasculares/metabolismo , Fator de Crescimento Insulin-Like I/metabolismo , Miocárdio/metabolismo , Fatores Etários , Envelhecimento/patologia , Animais , Antígenos CD/metabolismo , Doenças Cardiovasculares/genética , Doenças Cardiovasculares/patologia , Doenças Cardiovasculares/fisiopatologia , Senescência Celular , Humanos , Camundongos Knockout , Receptor IGF Tipo 1 , Receptor de Insulina/metabolismo , Receptores de Somatomedina/genética , Receptores de Somatomedina/metabolismo , Transdução de Sinais
5.
Biochim Biophys Acta Mol Basis Dis ; 1864(4 Pt A): 1183-1191, 2018 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-29378301

RESUMO

While deletion of Akt1 results in a smaller heart size and Akt2-/- mice are mildly insulin resistant, Akt1-/-/Akt2-/- mice exhibit perinatal lethality, indicating a large degree of functional overlap between the isoforms of the serine/threonine kinase Akt. The present study aimed to determine the cooperative contribution of Akt1 and Akt2 on the structure and contractile function of adult hearts. To generate an inducible, cardiomyocyte-restricted Akt2 knockout (KO) model, Akt2flox/flox mice were crossed with tamoxifen-inducible MerCreMer transgenic (MCM) mice and germline Akt1-/- mice to generate the following genotypes:Akt1+/+; Akt2flox/flox (WT), Akt2flox/flox; α-MHC-MCM (iAkt2 KO), Akt1-/-, and Akt1-/-; Akt2flox/flox; α-MHC-MCM mice (Akt1-/-/iAkt2 KO). At 28 days after the first tamoxifen injection, Akt1-/-/iAkt2 KO mice developed contractile dysfunction paralleling increased atrial and brain natriuretic peptide (ANP and BNP) levels, and repressed mitochondrial gene expression. Neither cardiac fibrosis nor apoptosis were detected in Akt1-/-/iAkt2 KO hearts. To explore potential molecular mechanisms for contractile dysfunction, we investigated myocardial microstructure before the onset of heart failure. At 3 days after the first tamoxifen injection, Akt1-/-/iAkt2 KO hearts showed decreased expression of connexin43 (Cx43) and connexin-interacting protein zonula occludens-1 (ZO-1). Furthermore, Akt1/2 silencing significantly decreased both Cx43 and ZO-1 expression in cultured neonatal rat cardiomyocytes in concert with reduced beating frequency. Akt1 and Akt2 are required to maintain cardiac contraction. Loss of Akt signaling disrupts gap junction protein, which might precipitate early contractile dysfunction prior to heart failure in the absence of myocardial remodeling, such as hypertrophy, fibrosis, or cell death.


Assuntos
Cardiomiopatias/metabolismo , Conexina 43/metabolismo , Contração Miocárdica , Miocárdio/metabolismo , Miócitos Cardíacos/metabolismo , Proteínas Proto-Oncogênicas c-akt/deficiência , Proteína da Zônula de Oclusão-1/metabolismo , Animais , Fator Natriurético Atrial/genética , Fator Natriurético Atrial/metabolismo , Cardiomiopatias/genética , Cardiomiopatias/patologia , Conexina 43/genética , Fibrose , Camundongos , Camundongos Knockout , Miocárdio/patologia , Miócitos Cardíacos/patologia , Peptídeo Natriurético Encefálico/genética , Peptídeo Natriurético Encefálico/metabolismo , Ratos , Proteína da Zônula de Oclusão-1/genética
6.
Am J Physiol Heart Circ Physiol ; 313(6): H1098-H1108, 2017 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-28822962

RESUMO

Pathological cardiac hypertrophy may be associated with reduced expression of glucose transporter 4 (GLUT4) in contrast to exercise-induced cardiac hypertrophy, where GLUT4 levels are increased. However, mice with cardiac-specific deletion of GLUT4 (G4H-/-) have normal cardiac function in the unstressed state. This study tested the hypothesis that cardiac GLUT4 is required for myocardial adaptations to hemodynamic demands. G4H-/- and control littermates were subjected to either a pathological model of left ventricular pressure overload [transverse aortic constriction (TAC)] or a physiological model of endurance exercise (swim training). As predicted after TAC, G4H-/- mice developed significantly greater hypertrophy and more severe contractile dysfunction. Somewhat surprisingly, after exercise training, G4H-/- mice developed increased fibrosis and apoptosis that was associated with dephosphorylation of the prosurvival kinase Akt in concert with an increase in protein levels of the upstream phosphatase protein phosphatase 2A (PP2A). Exercise has been shown to decrease levels of ceramide; G4H-/- hearts failed to decrease myocardial ceramide in response to exercise. Furthermore, G4H-/- hearts have reduced levels of the transcriptional coactivator peroxisome proliferator-activated receptor-γ coactivator-1, lower carnitine palmitoyl-transferase activity, and reduced hydroxyacyl-CoA dehydrogenase activity. These basal changes may also contribute to the impaired ability of G4H-/- hearts to adapt to hemodynamic stresses. In conclusion, GLUT4 is required for the maintenance of cardiac structure and function in response to physiological or pathological processes that increase energy demands, in part through secondary changes in mitochondrial metabolism and cellular stress survival pathways such as Akt.NEW & NOTEWORTHY Glucose transporter 4 (GLUT4) is required for myocardial adaptations to exercise, and its absence accelerates heart dysfunction after pressure overload. The requirement for GLUT4 may extend beyond glucose uptake to include defects in mitochondrial metabolism and survival signaling pathways that develop in its absence. Therefore, GLUT4 is critical for responses to hemodynamic stresses.


Assuntos
Cardiomegalia Induzida por Exercícios , Cardiomegalia/metabolismo , Transportador de Glucose Tipo 4/deficiência , Hemodinâmica , Miocárdio/metabolismo , Função Ventricular Esquerda , Remodelação Ventricular , 3-Hidroxiacil-CoA Desidrogenases/metabolismo , Adaptação Fisiológica , Animais , Aorta/fisiopatologia , Aorta/cirurgia , Cardiomegalia/etiologia , Cardiomegalia/patologia , Cardiomegalia/fisiopatologia , Carnitina O-Palmitoiltransferase/metabolismo , Constrição , Modelos Animais de Doenças , Predisposição Genética para Doença , Transportador de Glucose Tipo 4/genética , Camundongos Knockout , Contração Miocárdica , Miocárdio/patologia , Coativador 1-alfa do Receptor gama Ativado por Proliferador de Peroxissomo/genética , Coativador 1-alfa do Receptor gama Ativado por Proliferador de Peroxissomo/metabolismo , Fenótipo , Esforço Físico , Proteína Fosfatase 2/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo
7.
Diabetes Obes Metab ; 19(5): 635-643, 2017 05.
Artigo em Inglês | MEDLINE | ID: mdl-28026912

RESUMO

AIMS: To assess the efficacy and safety of gemigliptin, a dipeptidyl peptidase-4 inhibitor, added to metformin and sulphonylurea in patients with type 2 diabetes (T2DM). MATERIALS AND METHODS: We conducted a randomized, double-blind, placebo-controlled trial in 219 Korean patients inadequately controlled with metformin and glimepiride. Participants were randomized to gemigliptin 50 mg once daily or placebo added to metformin and glimepiride. The primary endpoint was change in glycated haemoglobin (HbA1c) level from baseline to week 24. RESULTS: The baseline HbA1c was 8.2% in both groups. The addition of gemigliptin to metformin and glimepiride significantly reduced HbA1c levels at week 24 compared with placebo (between-group difference in adjusted mean change -0.87%, 95% confidence interval [CI] -1.09% to -0.64%). Fasting plasma glucose level was also significantly reduced with gemigliptin (-0.93 mmol/L, 95% CI -1.50 to -0.35 mmol/L), and a higher proportion of participants achieved an HbA1c level of <7% (39.3% vs 5.5%; P <.001) in the gemigliptin group than in the placebo group. Total cholesterol and LDL cholesterol were modestly but significantly reduced in the gemigliptin group compared with the placebo group (-0.21 mmol/L, 95% CI -0.38 to -0.03 mmol/L for total cholesterol, -0.18 mmol/L, 95% CI -0.34 to -0.01 mmol/L for LDL cholesterol). The incidence of hypoglycaemia was 9.4% in the gemigliptin group and 2.7% in the placebo group. CONCLUSIONS: Gemigliptin significantly improved glycaemic control in patients with T2DM inadequately controlled with metformin and sulphonylurea. The incidence of hypoglycaemia was higher with gemigliptin than with placebo, which highlights the importance of optimal dose adjustment for sulphonylurea.


Assuntos
Diabetes Mellitus Tipo 2/tratamento farmacológico , Inibidores da Dipeptidil Peptidase IV/uso terapêutico , Resistência a Medicamentos , Hiperglicemia/prevenção & controle , Hipoglicemia/prevenção & controle , Piperidonas/uso terapêutico , Pirimidinas/uso terapêutico , Idoso , Diabetes Mellitus Tipo 2/sangue , Inibidores da Dipeptidil Peptidase IV/efeitos adversos , Método Duplo-Cego , Monitoramento de Medicamentos , Quimioterapia Combinada/efeitos adversos , Feminino , Hemoglobinas Glicadas/análise , Humanos , Hipoglicemia/induzido quimicamente , Hipoglicemia/epidemiologia , Hipoglicemiantes/efeitos adversos , Hipoglicemiantes/uso terapêutico , Incidência , Masculino , Metformina/efeitos adversos , Metformina/uso terapêutico , Pessoa de Meia-Idade , Piperidonas/efeitos adversos , Pirimidinas/efeitos adversos , República da Coreia/epidemiologia , Risco , Compostos de Sulfonilureia/efeitos adversos , Compostos de Sulfonilureia/uso terapêutico
8.
J Biol Chem ; 289(8): 4839-52, 2014 Feb 21.
Artigo em Inglês | MEDLINE | ID: mdl-24398673

RESUMO

Doxorubicin is one of the most widely used anti-cancer drugs, but its clinical application is compromised by severe adverse effects in different organs including cardiotoxicity. In the present study we explored mechanisms of doxorubicin-induced cytotoxicity by revealing a novel role for the AMP-activated protein kinase α2 (AMPKα2) in mouse embryonic fibroblasts (MEFs). Doxorubicin robustly induced the expression of AMPKα2 in MEFs but slightly reduced AMPKα1 expression. Our data support the previous notion that AMPKα1 harbors survival properties under doxorubicin treatment. In contrast, analyses of Ampkα2(-/-) MEFs, gene knockdown of AMPKα2 by shRNA, and inhibition of AMPKα2 activity with an AMPK inhibitor indicated that AMPKα2 functions as a pro-apoptotic molecule under doxorubicin treatment. Doxorubicin induced AMPKα2 at the transcription level via E2F1, a transcription factor that regulates apoptosis in response to DNA damage. E2F1 directly transactivated the Ampkα2 gene promoter. In turn, AMPKα2 significantly contributed to stabilization and activation of E2F1 by doxorubicin, forming a positive signal amplification loop. AMPKα2 directly interacted with and phosphorylated E2F1. This signal loop was also detected in H9c2, C2C12, and ECV (human epithelial cells) cells as well as mouse liver under doxorubicin treatment. Resveratrol, which has been suggested to attenuate doxorubicin-induced cytotoxicity, significantly blocked induction of AMPKα2 and E2F1 by doxorubicin, leading to protection of these cells. This signal loop appears to be non-carcinoma-specific because AMPKα2 was not induced by doxorubicin in five different tested cancer cell lines. These results suggest that AMPKα2 may serve as a novel target for alleviating the cytotoxicity of doxorubicin.


Assuntos
Proteínas Quinases Ativadas por AMP/metabolismo , Doxorrubicina/farmacologia , Fator de Transcrição E2F1/metabolismo , Embrião de Mamíferos/citologia , Fibroblastos/metabolismo , Transdução de Sinais/efeitos dos fármacos , Proteínas Quinases Ativadas por AMP/genética , Animais , Morte Celular/efeitos dos fármacos , Linhagem Celular , Citoproteção/efeitos dos fármacos , Dano ao DNA , Fibroblastos/citologia , Fibroblastos/efeitos dos fármacos , Regulação Enzimológica da Expressão Gênica/efeitos dos fármacos , Humanos , Camundongos , Modelos Biológicos , Fosforilação/efeitos dos fármacos , Ligação Proteica/efeitos dos fármacos , Estabilidade Proteica/efeitos dos fármacos , Resveratrol , Estilbenos/farmacologia
9.
Endocr J ; 62(5): 449-62, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25819061

RESUMO

The aim of this study was to evaluate the efficacy and safety of anagliptin in drug-naïve patients with type 2 diabetes in a double-blind randomized placebo-controlled study. A total of 109 patients were randomized to 100 mg (n=37) or 200 mg (n=33) anagliptin twice daily or placebo (n=39). The primary objective was to alter HbA1c levels from baseline at a 24-week endpoint. The overall baseline mean age and body mass index were 56.20 ± 9.77 years and 25.01 ± 2.97 kg/m(2), respectively, and the HbA1c level was of 7.14 ± 0.69 %. Anagliptin at 100 mg and 200 mg produced significant reductions in HbA1c (-0.50 ± 0.45 % and -0.51 ± 0.55%, respectively), and the placebo treatment resulted in an increase in HbA1c by 0.23 ± 0.62 %. Both doses of anagliptin produced significant decreases in fasting plasma glucose (-0.53 ± 1.25 mmol/L and -0.72 ± 1.25 mmol/L, respectively) and the proinsulin/insulin ratio (-0.04 ± 0.15 and -0.07 ± 0.18, respectively) compared with placebo. No meaningful body weight changes from baseline were observed in three groups. Plasma dipeptidyl peptidase (DPP)-4 activity was significantly inhibited after 24 weeks of anagliptin treatment, and >75% and >90% inhibitions were observed during the meal tolerance tests with 100 mg and 200 mg anagliptin, respectively. The incidences of adverse or serious adverse events were similar among the three study groups. Twice-daily anagliptin therapy effectively inhibited DPP-4 activity and improved glycemic control and was well-tolerated in patients with type 2 diabetes.


Assuntos
Diabetes Mellitus Tipo 2/tratamento farmacológico , Inibidores da Dipeptidil Peptidase IV , Pirimidinas/uso terapêutico , Idoso , Glicemia/análise , Índice de Massa Corporal , Diabetes Mellitus Tipo 2/sangue , Dipeptidil Peptidase 4/sangue , Método Duplo-Cego , Jejum , Feminino , Hemoglobinas Glicadas/análise , Humanos , Insulina/sangue , Masculino , Pessoa de Meia-Idade , Placebos , Proinsulina/sangue , Pirimidinas/efeitos adversos
10.
FASEB J ; 27(12): 4899-908, 2013 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-23982142

RESUMO

Although thyroid-stimulating hormone (TSH) is known to be a major regulator of thyroid hormone biosynthesis and thyroid growth, insulin-like growth factor 1 (IGF-1) is required for mediating thyrocyte growth in concert with TSH in vitro. We generated mice with thyrocyte-selective ablation of IGF-1 receptor (TIGF1RKO) to explore the role of IGF-1 receptor signaling on thyroid function and growth. In 5-wk-old TIGF1RKO mice, serum thyroxine (T4) concentrations were decreased by 30% in concert with a 43% down-regulation of the monocarboxylate transporter 8 (MCT8), which is involved in T4 secretion. Despite a 3.5-fold increase in circulating concentrations of TSH, thyroid architecture and size were normal. Furthermore, thyrocyte area was increased by 40% in WT thyroids after 10 d TSH injection, but this effect was absent in TSH-injected TIGF1RKO mice. WT mice treated with methimazole and sodium perchlorate for 2 or 6 wk exhibited pronounced goiter development (2.0 and 5.4-fold, respectively), but in TIGF1RKO mice, goiter development was completely abrogated. These data reveal an essential role for IGF-1 receptor signaling in the regulation of thyroid function and TSH-stimulated goitrogenesis.


Assuntos
Bócio/metabolismo , Receptor IGF Tipo 1/genética , Tireotropina/metabolismo , Tiroxina/metabolismo , Animais , Antitireóideos/farmacologia , Regulação para Baixo , Bócio/induzido quimicamente , Proteínas de Membrana Transportadoras/genética , Proteínas de Membrana Transportadoras/metabolismo , Metimazol/farmacologia , Camundongos , Camundongos Knockout , Transportadores de Ácidos Monocarboxílicos , Percloratos/toxicidade , Receptor IGF Tipo 1/deficiência , Compostos de Sódio/toxicidade , Simportadores , Glândula Tireoide/efeitos dos fármacos , Glândula Tireoide/metabolismo , Glândula Tireoide/patologia
11.
Mol Ther ; 20(8): 1540-9, 2012 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-22617107

RESUMO

Practical methods to deliver proteins systemically in animals have been hampered by poor tissue penetration and inefficient cytoplasmic localization of internalized proteins. We therefore pursued the development of improved macromolecule transduction domains (MTDs) and tested their ability to deliver therapeutically active p18(INK4c). MTD103 was identified from a screen of 1,500 signal peptides; tested for the ability to promote protein uptake by cells and tissues; and analyzed with regard to the mechanism of protein uptake and the delivery of biologically active p18(INK4c) into cancer cells. The therapeutic potential of cell-permeable MTD103p18(INK4c) (CP-p18(INK4c)) was tested in the HCT116 tumor xenograft model. MTD103p18(INK4c) appeared to traverse plasma membranes directly, was transferred from cell-to-cell and was therapeutically effective against cancer xenografts, inhibiting tumor growth by 86-98% after 5 weeks (P < 0.05). The therapeutic responses to CP-p18(INK4c) were accompanied by high levels of apoptosis in tumor cells. In addition to enhancing systemic delivery of CP-p18(INK4c) to normal tissues and cancer xenografts, the MTD103 sequence delayed protein clearance from the blood, liver and spleen. These results demonstrate that macromolecule intracellular transduction technology (MITT), enabled by MTDs, may provide novel protein therapies against cancer and other diseases.


Assuntos
Antineoplásicos/administração & dosagem , Antineoplásicos/uso terapêutico , Inibidor de Quinase Dependente de Ciclina p18/administração & dosagem , Inibidor de Quinase Dependente de Ciclina p18/uso terapêutico , Peptídeos/química , Animais , Antineoplásicos/química , Neoplasias Colorretais/tratamento farmacológico , Inibidor de Quinase Dependente de Ciclina p18/química , Feminino , Humanos , Camundongos , Camundongos Endogâmicos BALB C , Peptídeos/administração & dosagem , Ensaios Antitumorais Modelo de Xenoenxerto
12.
Exp Mol Med ; 55(5): 965-973, 2023 05.
Artigo em Inglês | MEDLINE | ID: mdl-37121973

RESUMO

Insulin and insulin-like growth factor 1 (IGF-1) signaling regulate cellular growth and glucose metabolism in the myocardium. However, their physiological role in the cells of the cardiac conduction system has never been explored. Therefore, we sought to determine the spatiotemporal function of insulin/IGF-1 receptors in the sinoatrial node (SAN). We generated cardiac conduction cell-specific inducible IGF-1 receptor (IGF-1R) knockout (KO) (CSIGF1RKO), insulin receptor (IR) KO (CSIRKO), and IR/IGF-1R double-KO (CSDIRKO) mice and evaluated their phenotypes. Telemetric electrocardiography revealed regular sinus rhythm in CSIGF1RKO mice, indicating that IGF-1R is dispensable for normal pacemaking. In contrast, CSIRKO and CSDIRKO mice exhibited profound sinus bradycardia. CSDIRKO mice showed typical sinus node dysfunction characterized by junctional rhythm and sinus pauses on electrocardiography. Interestingly, the lack of an insulin receptor in the SAN cells of CSIRKO and CSDIRKO mice caused sinus nodal fibrosis. Mechanistically, hyperpolarization-activated cyclic nucleotide-gated channel 4 (HCN4) protein expression significantly decreased in the CSIRKO and CSDIRKO mice relative to the controls. A patch-clamp study of the SAN cells of CSIRKO mice revealed a significant decrease in the funny current, which is responsible for spontaneous diastolic depolarization in the SAN. This result suggested that insulin receptor loss reduces the heart rate via downregulation of the HCN4 channel. Additionally, HCN1 expression was decreased in CSDIRKO mice, explaining their sinus node dysfunction. Our results reveal a previously unrecognized role of insulin/IGF-1 signaling in sinus node structural maintenance and pacemaker function.


Assuntos
Síndrome do Nó Sinusal , Nó Sinoatrial , Camundongos , Animais , Nó Sinoatrial/metabolismo , Síndrome do Nó Sinusal/metabolismo , Receptor de Insulina/metabolismo , Fator de Crescimento Insulin-Like I/metabolismo , Insulina/metabolismo
13.
J Mol Cell Cardiol ; 52(3): 718-26, 2012 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-22197309

RESUMO

Despite its significant clinical implications, physiological hypertrophy remains poorly understood. In this study, the transcription coactivator Eya2 was shown to be up-regulated during physiological hypertrophy. Transgene- or adenovirus-mediated overexpression of Eya2 led to up-regulation of mTOR, a critical mediator of physiological hypertrophy. Luciferase reporter and chromatin immunoprecipitation assays revealed that Eya2 directly binds to and activates mTOR expression. The phosphorylation of mTOR downstream molecules was significantly enhanced in Eya2 transgenic (TG) hearts, implying that the Eya2-mediated induction of mTOR expression leads to an elevated mTOR activity. The transcription factor Six1 was also up-regulated during physiological hypertrophy and formed a complex with Eya2. Luciferase reporter and electrophoretic mobility shift assays revealed that the Eya2-Six1 complex binds to and enhances the expression of mTOR in a synergistic manner. Under pressure overload, Eya2 transgenic hearts developed hypertrophy which exhibited important molecular signatures of physiological hypertrophy, as assessed by gene expression profiling and measurements of expression levels of physiological hypertrophy-related genes by quantitative (q) RT-PCR. Examination of heart sections under electron microscopy revealed that the mitochondrial integrity remained largely intact in Eya2 transgenic mice, but not in wild-type littermates, under pressure overload. This finding was confirmed by measurements of mitochondrial DNA contents and the expression levels of mitochondrial function-related genes by qRT-PCR. These data suggest that Eya2 in a physical complex with Six1 plays a critical role in physiological hypertrophy. The cardioprotective effect of Eya2 appears to be due, at least in part, to its preservation of mitochondrial integrity upon pressure overload.


Assuntos
Crescimento Celular , Peptídeos e Proteínas de Sinalização Intracelular/genética , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Miócitos Cardíacos/metabolismo , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Proteínas Tirosina Fosfatases/genética , Proteínas Tirosina Fosfatases/metabolismo , Animais , Linhagem Celular , Células Cultivadas , Regulação da Expressão Gênica , Proteínas de Homeodomínio/metabolismo , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Mitocôndrias Cardíacas/ultraestrutura , Ligação Proteica , Ratos , Ratos Sprague-Dawley , Serina-Treonina Quinases TOR/genética , Serina-Treonina Quinases TOR/metabolismo
14.
J Mol Cell Cardiol ; 53(1): 53-63, 2012 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-22449794

RESUMO

Protein kinase C (PKC)-interacting cousin of thioredoxin (PICOT) has distinct anti-hypertrophic and inotropic functions. We have previously shown that PICOT exerts its anti-hypertrophic effect by inhibiting calcineurin-NFAT signaling through its C-terminal glutaredoxin domain. However, the mechanism underlying the inotropic effect of PICOT is unknown. The results of protein pull-down experiments showed that PICOT directly binds to the catalytic domain of PKCζ through its N-terminal thioredoxin-like domain. Purified PICOT protein inhibited the kinase activity of PKCζ in vitro, which indicated that PICOT is an endogenous inhibitor of PKCζ. The inhibition of PKCζ activity with a PKCζ-specific pseudosubstrate peptide inhibitor was sufficient to increase the cardiac contractility in vitro and ex vivo. Overexpression of PICOT or inhibition of PKCζ activity down-regulated PKCα activity, which led to the elevation of sarcoplasmic reticulum Ca(2+)-ATPase (SERCA) 2a activity, concomitant with the increased phosphorylation of phospholamban (PLB). Overexpression of PICOT or inhibition of PKCζ activity also down-regulated protein phosphatase (PP) 2A activity, which subsequently resulted in the increased phosphorylation of troponin (Tn) I and T, key myofilament proteins associated with the regulation of contractility. PICOT appeared to inhibit PP2A activity through the disruption of the functional PKCζ/PP2A complex. In contrast to the overexpression of PICOT or inhibition of PKCζ, reduced PICOT expression resulted in up-regulation of PKCα and PP2A activities, followed by decreased phosphorylation of PLB, and TnI and T, respectively, supporting the physiological relevance of these events. Transgene- or adeno-associated virus (AAV)-mediated overexpression of PICOT restored the impaired contractility and prevented further morphological and functional deterioration of the failing hearts. Taken together, the results of the present study suggest that PICOT exerts its inotropic effect by negatively regulating PKCα and PP2A activities through the inhibition of PKCζ activity. This finding provides a novel insight into the regulation of cardiac contractility.


Assuntos
Proteínas de Transporte/metabolismo , Contração Miocárdica/fisiologia , Proteína Quinase C/metabolismo , Animais , Proteínas de Ligação ao Cálcio/metabolismo , Proteínas de Transporte/genética , Linhagem Celular , Ativação Enzimática , Humanos , Masculino , Camundongos , Modelos Biológicos , Contração Miocárdica/efeitos dos fármacos , Contração Miocárdica/genética , Fragmentos de Peptídeos/farmacologia , Ligação Proteica , Proteína Quinase C/antagonistas & inibidores , Proteína Quinase C-alfa/metabolismo , Ratos , Ratos Sprague-Dawley , Transdução de Sinais , Troponina I/metabolismo , Troponina T/metabolismo , Produtos do Gene tat do Vírus da Imunodeficiência Humana/farmacologia
15.
Cell Metab ; 6(4): 294-306, 2007 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-17908558

RESUMO

Physiological cardiac hypertrophy is associated with mitochondrial adaptations that are characterized by activation of PGC-1alpha and increased fatty acid oxidative (FAO) capacity. It is widely accepted that phosphatidylinositol 3-kinase (PI3K) signaling to Akt1 is required for physiological cardiac growth. However, the signaling pathways that coordinate physiological hypertrophy and metabolic remodeling are incompletely understood. We show here that activation of PI3K is sufficient to increase myocardial FAO capacity and that inhibition of PI3K signaling prevents mitochondrial adaptations in response to physiological hypertrophic stimuli despite increased expression of PGC-1alpha. We also show that activation of the downstream kinase Akt is not required for the mitochondrial adaptations that are secondary to PI3K activation. Thus, in physiological cardiac growth, PI3K is an integrator of cellular growth and metabolic remodeling. Although PI3K signaling to Akt1 is required for cellular growth, Akt-independent pathways mediate the accompanying mitochondrial adaptations.


Assuntos
Cardiomegalia/enzimologia , Mitocôndrias/fisiologia , Fosfatidilinositol 3-Quinases/fisiologia , Condicionamento Físico Animal , Proteínas Proto-Oncogênicas c-akt/fisiologia , Adaptação Fisiológica , Animais , Ácidos Graxos/metabolismo , Regulação da Expressão Gênica , Isoenzimas/antagonistas & inibidores , Camundongos , Camundongos Mutantes , Miócitos Cardíacos/enzimologia , Oxirredução , Fosfatidilinositol 3-Quinases/genética , Isoformas de Proteínas/antagonistas & inibidores , Isoformas de Proteínas/genética , Isoformas de Proteínas/fisiologia , Proteína Quinase C/antagonistas & inibidores , Proteínas Proto-Oncogênicas c-akt/antagonistas & inibidores , Proteínas Proto-Oncogênicas c-akt/genética , Transdução de Sinais
16.
Curr Hypertens Rep ; 14(6): 510-6, 2012 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-22986910

RESUMO

To further understand chronic heart disease, such as heart failure and cardiomyopathy, we must fully define signaling pathways within the myocardium. Recent studies suggest that some forms of heart disease are associated with a chronic low-grade inflammation that promotes adverse ventricular remodeling and correlates with disease progression. Several inflammatory mediators, including TNF-α, IL-1ß, and IL-6, are involved in cardiac injury subsequent to myocardial ischemia and reperfusion, sepsis, viral myocarditis, and transplant rejection. Once activated, components of the inflammatory response can have both beneficial and deleterious effects on the heart. In this review, we discuss the complex inflammatory signaling pathways in the myocardium and potential therapeutic implications.


Assuntos
Cardiomiopatias/metabolismo , Insuficiência Cardíaca/metabolismo , Mediadores da Inflamação/metabolismo , Inflamação/metabolismo , Cardiomiopatias/imunologia , Insuficiência Cardíaca/imunologia , Humanos , Inflamação/imunologia , Mediadores da Inflamação/imunologia , Traumatismo por Reperfusão Miocárdica/imunologia , Traumatismo por Reperfusão Miocárdica/metabolismo , Miocárdio/imunologia , Miocárdio/metabolismo , Transdução de Sinais , Remodelação Ventricular/imunologia
17.
Nanomaterials (Basel) ; 12(15)2022 Aug 02.
Artigo em Inglês | MEDLINE | ID: mdl-35957086

RESUMO

Air pollution exerts several deleterious effects on the cardiovascular system, with cardiovascular disease (CVD) accounting for 80% of all premature deaths caused by air pollution. Short-term exposure to particulate matter 2.5 (PM2.5) leads to acute CVD-associated deaths and nonfatal events, whereas long-term exposure increases CVD-associated risk of death and reduces longevity. Here, we summarize published data illustrating how PM2.5 may impact the cardiovascular system to provide information on the mechanisms by which it may contribute to CVDs. We provide an overview of PM2.5, its associated health risks, global statistics, mechanistic underpinnings related to mitochondria, and hazardous biological effects. We elaborate on the association between PM2.5 exposure and CVD development and examine preventive PM2.5 exposure measures and future strategies for combating PM2.5-related adverse health effects. The insights gained can provide critical guidelines for preventing pollution-related CVDs through governmental, societal, and personal measures, thereby benefitting humanity and slowing climate change.

18.
Endocr J ; 58(11): 1013-9, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-21908931

RESUMO

Thyroid-stimulating hormone (TSH) is the primary regulator of thyroid growth and function acting via cyclic AMP signaling cascades. In cultured thyrocytes, insulin and/or insulin-like growth factor-1 (IGF-1) are required for mediating thyrocyte proliferation in concert with TSH. To determine the role of insulin signaling in thyroid, growth in vivo, mice with thyrocyte-selective ablation of the insulin receptor (IR) were generated by crossing mice homozygous for a floxed IR allele with transgenic mice in which thyrocyte-specific expression of Cre recombinase was driven by the human thyroid peroxidase (TPO) gene promoter. Immunohistochemistry and Western blot analysis confirmed near complete loss of IR expression in the thyroid of thyrocyte IR knockout mice. These mice are viable and have no obvious thyroid dysfunction and macro- and microscopic thyroid morphology was normal. Thus, insulin signaling in thyrocytes does not play an essential role in the architecture and function of the thyroid in vivo.


Assuntos
Receptor de Insulina/deficiência , Glândula Tireoide/fisiologia , Animais , Imuno-Histoquímica , Fator de Crescimento Insulin-Like I/genética , Fator de Crescimento Insulin-Like I/fisiologia , Integrases/genética , Integrases/fisiologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , RNA/química , RNA/genética , Receptor de Insulina/genética , Receptor de Insulina/fisiologia , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transdução de Sinais , Organismos Livres de Patógenos Específicos , Glândula Tireoide/citologia , Tireotropina/sangue , Tiroxina/sangue
19.
Diabetes Metab J ; 45(2): 129-145, 2021 03.
Artigo em Inglês | MEDLINE | ID: mdl-33813812

RESUMO

Diabetic heart disease is a growing and important public health risk. Apart from the risk of coronary artery disease or hypertension, diabetes mellitus (DM) is a well-known risk factor for heart failure in the form of diabetic cardiomyopathy (DiaCM). Currently, DiaCM is defined as myocardial dysfunction in patients with DM in the absence of coronary artery disease and hypertension. The underlying pathomechanism of DiaCM is partially understood, but accumulating evidence suggests that metabolic derangements, oxidative stress, increased myocardial fibrosis and hypertrophy, inflammation, enhanced apoptosis, impaired intracellular calcium handling, activation of the renin-angiotensin-aldosterone system, mitochondrial dysfunction, and dysregulation of microRNAs, among other factors, are involved. Numerous animal models have been used to investigate the pathomechanisms of DiaCM. Despite some limitations, animal models for DiaCM have greatly advanced our understanding of pathomechanisms and have helped in the development of successful disease management strategies. In this review, we summarize the current pathomechanisms of DiaCM and provide animal models for DiaCM according to its pathomechanisms, which may contribute to broadening our understanding of the underlying mechanisms and facilitating the identification of possible new therapeutic targets.


Assuntos
Diabetes Mellitus , Cardiomiopatias Diabéticas , Insuficiência Cardíaca , Animais , Cardiomiopatias Diabéticas/terapia , Humanos , Modelos Animais , Estresse Oxidativo , Sistema Renina-Angiotensina
20.
Life (Basel) ; 11(12)2021 Nov 26.
Artigo em Inglês | MEDLINE | ID: mdl-34947831

RESUMO

NAD(P)H:quinone oxidoreductase (NQO) is an antioxidant flavoprotein that catalyzes the reduction of highly reactive quinone metabolites by employing NAD(P)H as an electron donor. There are two NQO enzymes-NQO1 and NQO2-in mammalian systems. In particular, NQO1 exerts many biological activities, including antioxidant activities, anti-inflammatory effects, and interactions with tumor suppressors. Moreover, several recent studies have revealed the promising roles of NQO1 in protecting against cardiovascular damage and related diseases, such as dyslipidemia, atherosclerosis, insulin resistance, and metabolic syndrome. In this review, we discuss recent developments in the molecular regulation and biochemical properties of NQO1, and describe the potential beneficial roles of NQO1 in diseases associated with oxidative stress.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA