Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
FASEB J ; 30(3): 1065-75, 2016 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-26578687

RESUMEN

Obesity is associated with an increased risk of cardiovascular disease. C1q/TNF-related protein (CTRP)-1 is a poorly characterized adipokine that is up-regulated in association with ischemic heart disease. We investigated the role of CTRP1 in myocardial ischemia injury. CTRP1-knockout mice showed increased myocardial infarct size, cardiomyocyte apoptosis, and proinflammatory gene expression after I/R compared with wild-type (WT) mice. In contrast, systemic delivery of CTRP1 attenuated myocardial damage after I/R in WT mice. Treatment of cardiomyocytes with CTRP1 led to reduction of hypoxia-reoxygenation-induced apoptosis and lipopolysaccharide-stimulated expression of proinflammatory cytokines, which was reversed by inhibition of sphingosine-1-phosphate (S1P) signaling. Treatment of cardiomyocytes with CTRP1 also resulted in the increased production of cAMP, which was blocked by suppression of S1P signaling. The antiapoptotic and anti-inflammatory actions of CTRP1 were cancelled by inhibition of adenylyl cyclase or knockdown of adiponectin receptor 1. Furthermore, blockade of S1P signaling reversed CTRP1-mediated inhibition of myocardial infarct size, apoptosis, and inflammation after I/R in vivo. These data indicate that CTRP1 protects against myocardial ischemic injury by reducing apoptosis and inflammatory response through activation of the S1P/cAMP signaling pathways in cardiomyocytes, suggesting that CTRP1 plays a crucial role in the pathogenesis of ischemic heart disease.


Asunto(s)
Adipoquinas/metabolismo , Corazón/fisiopatología , Isquemia Miocárdica/metabolismo , Daño por Reperfusión Miocárdica/metabolismo , Miocardio/metabolismo , Miocitos Cardíacos/metabolismo , Sustancias Protectoras/metabolismo , Animales , Apoptosis/fisiología , AMP Cíclico/metabolismo , Citocinas/metabolismo , Modelos Animales de Enfermedad , Inflamación/metabolismo , Lisofosfolípidos/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Infarto del Miocardio/metabolismo , Transducción de Señal/fisiología , Esfingosina/análogos & derivados , Esfingosina/metabolismo
2.
Circ Heart Fail ; 8(2): 342-51, 2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-25654972

RESUMEN

BACKGROUND: Myocardial infarction (MI) is one of the major causes of death worldwide. Chronic heart failure is a serious complication of MI that leads to poor prognosis. We recently found that neuron-derived neurotrophic factor (NDNF) is a proangiogenic secretory protein that is upregulated in ischemic skeletal muscle. Here, we examined whether NDNF modulates cardiac remodeling in response to chronic ischemia. METHODS AND RESULTS: C57BL/6J wild-type mice were subjected to the permanent ligation of the left anterior descending coronary artery to create MI. Adenoviral vectors expressing NDNF or ß-galactosidase (control) were intramuscularly injected into mice 3 days before permanent left anterior descending coronary artery ligation. Intramuscular administration of adenoviral vectors expressing NDNF to mice resulted in increased levels of circulating NDNF. Adenoviral vectors expressing NDNF administration improved left ventricular systolic dysfunction and dilatation after MI surgery. Moreover, adenoviral vectors expressing NDNF enhanced capillary formation and reduced cardiomyocyte apoptosis and hypertrophy in the post-MI hearts. Treatment of cultured cardiomyocytes with recombinant NDNF protein led to reduced apoptosis under conditions of hypoxia. NDNF also promoted the phosphorylation of Akt and focal adhesion kinase in cardiomyocytes. Blockade of focal adhesion kinase activation blocked the stimulatory effects of NDNF on cardiomyocyte survival and Akt phosphorylation. Similarly, treatment of cultured endothelial cells with NDNF protein led to enhancement of network formation and Akt phosphorylation, which was diminished by focal adhesion kinase inhibition. CONCLUSIONS: These data suggest that NDNF ameliorates adverse myocardial remodeling after MI by its abilities to enhance myocyte survival and angiogenesis in the heart through focal adhesion kinase/Akt-dependent mechanisms.


Asunto(s)
Infarto del Miocardio/fisiopatología , Miocitos Cardíacos/fisiología , Factores de Crecimiento Nervioso/fisiología , Animales , Apoptosis/fisiología , Supervivencia Celular/fisiología , Modelos Animales de Enfermedad , Quinasa 1 de Adhesión Focal/fisiología , Inyecciones Intramusculares , Masculino , Ratones Endogámicos C57BL , Neovascularización Fisiológica/fisiología , Factores de Crecimiento Nervioso/administración & dosificación , Factores de Crecimiento Nervioso/metabolismo , Fosforilación/fisiología , Proteínas Proto-Oncogénicas c-akt/fisiología
3.
J Am Soc Nephrol ; 26(3): 636-46, 2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-25071081

RESUMEN

Heart disease contributes to the progression of CKD. Heart tissue produces a number of secreted proteins, also known as cardiokines, which participate in intercellular and intertissue communication. We recently reported that follistatin-like 1 (Fstl1) functions as a cardiokine with cardioprotective properties. Here, we investigated the role of cardiac Fstl1 in renal injury after subtotal nephrectomy. Cardiac-specific Fstl1-deficient (cFstl1-KO) mice and wild-type mice were subjected to subtotal (5/6) nephrectomy. cFstl1-KO mice showed exacerbation of urinary albumin excretion, glomerular hypertrophy, and tubulointerstitial fibrosis after subtotal renal ablation compared with wild-type mice. cFstl1-KO mice also exhibited increased mRNA levels of proinflammatory cytokines, including TNF-α and IL-6, NADPH oxidase components, and fibrotic mediators, in the remnant kidney. Conversely, systemic administration of adenoviral vectors expressing Fstl1 (Ad-Fstl1) to wild-type mice with subtotal nephrectomy led to amelioration of albuminuria, glomerular hypertrophy, and tubulointerstitial fibrosis, accompanied by reduced expression of proinflammatory mediators, NADPH oxidase components, and fibrotic markers in the remnant kidney. In cultured human mesangial cells, treatment with recombinant FSTL1 attenuated TNF-α-stimulated expression of proinflammatory cytokines. Treatment of mesangial cells with FSTL1 augmented the phosphorylation of AMP-activated protein kinase (AMPK), and inhibition of AMPK activation abrogated the anti-inflammatory effects of FSTL1. These data suggest that Fstl1 functions in cardiorenal communication and that the lack of Fstl1 production by myocytes promotes glomerular and tubulointerstitial damage in the kidney.


Asunto(s)
Proteínas Relacionadas con la Folistatina/fisiología , Insuficiencia Renal Crónica/etiología , Proteínas Quinasas Activadas por AMP/metabolismo , Animales , Modelos Animales de Enfermedad , Células Mesangiales/fisiología , Ratones Noqueados , Miocitos Cardíacos/metabolismo , Nefrectomía , Factor de Necrosis Tumoral alfa
4.
PLoS One ; 9(6): e99846, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24945145

RESUMEN

OBJECTIVE: Obesity is a major risk factor for cardiovascular disease. Recent evidence demonstrates that dysregulation of fat-derived hormones, also known as adipokines, is linked with the pathogenesis of obesity-related disorders including coronary artery disease (CAD). Here, we investigated whether circulating level of an adipokine C1q/TNF-related protein (CTRP) 1 is associated with the prevalence of CAD. METHODS AND RESULTS: Consecutive 76 male CAD patients were enrolled from inpatients that underwent coronary angiography. Sixty four healthy male subjects served as controls. Plasma CTRP1 concentration was determined by enzyme-linked immunosorbent assay. CTRP1 levels were correlated positively with systolic blood pressure (BP) and triglyceride levels, and negatively with HDL cholesterol levels in all subjects. Plasma levels of CTRP1 were significantly higher in CAD patients than in control subjects (CAD: 443.3±18.6 ng/ml, control: 307.8±21.5 ng/ml, p<0.001). Multiple logistic regression analysis with body mass index, systolic BP, glucose, total cholesterol, HDL cholesterol, triglyceride, adiponectin and CTRP1 revealed that CTRP1 levels, together with systolic BP and HDL cholesterol, correlated with CAD. CONCLUSIONS: Our data indicate the close association of high CTRP1 levels with CAD prevalence, suggesting that CTRP1 represents a novel biomarker for CAD.


Asunto(s)
Enfermedad de la Arteria Coronaria/sangre , Proteínas/metabolismo , Adiponectina/sangre , Anciano , Biomarcadores/sangre , Presión Sanguínea , Índice de Masa Corporal , Estudios de Casos y Controles , HDL-Colesterol/sangre , Angiografía Coronaria , Enfermedad de la Arteria Coronaria/fisiopatología , Humanos , Modelos Logísticos , Masculino , Persona de Mediana Edad , Triglicéridos/sangre
5.
Cardiovasc Res ; 103(1): 111-20, 2014 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-24743592

RESUMEN

AIMS: It is well-established that exercise diminishes cardiovascular risk, but whether humoral factors secreted by muscle confer these benefits has not been conclusively shown. We have shown that the secreted protein follistatin-like 1 (Fstl1) has beneficial actions on cardiac and endothelial function. However, the role of muscle-derived Fstl1 in proliferative vascular disease remains largely unknown. Here, we investigated whether muscle-derived Fstl1 modulates vascular remodelling in response to injury. METHODS AND RESULTS: The targeted ablation of Fstl1 in muscle led to an increase in neointimal formation following wire-induced arterial injury compared with control mice. Conversely, muscle-specific Fstl1 transgenic (TG) mice displayed a decrease in the neointimal thickening following arterial injury. Muscle-specific Fstl1 ablation and overexpression increased and decreased, respectively, the frequency of BrdU-positive proliferating cells in injured vessels. In cultured human aortic smooth muscle cells (HASMCs), treatment with human FSTL1 protein decreased proliferation and migration induced by stimulation with PDGF-BB. Treatment with FSTL1 enhanced AMPK phosphorylation, and inhibition of AMPK abrogated the inhibitory actions of FSTL1 on HASMC responses to PDGF-BB. The injured arteries of Fstl1-TG mice exhibited an increase in AMPK phosphorylation, and administration of AMPK inhibitor reversed the anti-proliferative actions of Fstl1 on the vessel wall. CONCLUSION: Our findings indicate that muscle-derived Fstl1 attenuates neointimal formation in response to arterial injury by suppressing SMC proliferation through an AMPK-dependent mechanism. Thus, the release of protein factors from muscle, such as Fstl1, may partly explain why the maintenance of muscle function can have a therapeutic effect on the cardiovascular system.


Asunto(s)
Arteria Femoral/lesiones , Proteínas Relacionadas con la Folistatina/fisiología , Neointima/patología , Neointima/fisiopatología , Proteínas Quinasas Activadas por AMP/antagonistas & inhibidores , Proteínas Quinasas Activadas por AMP/metabolismo , Animales , Becaplermina , Movimiento Celular , Proliferación Celular , Células Cultivadas , Modelos Animales de Enfermedad , Arteria Femoral/patología , Arteria Femoral/fisiopatología , Proteínas Relacionadas con la Folistatina/antagonistas & inhibidores , Proteínas Relacionadas con la Folistatina/genética , Técnicas de Silenciamiento del Gen , Humanos , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Músculo Esquelético/fisiología , Miocitos del Músculo Liso/patología , Miocitos del Músculo Liso/fisiología , Neointima/prevención & control , Proteínas Proto-Oncogénicas c-sis/metabolismo
6.
J Am Coll Cardiol ; 63(24): 2722-33, 2014 Jun 24.
Artículo en Inglés | MEDLINE | ID: mdl-24768874

RESUMEN

OBJECTIVES: This study examined the impact of omentin on myocardial injury in a mouse model of ischemia/reperfusion (I/R) and explored its underlying mechanisms. BACKGROUND: Obesity is a major risk factor for ischemic heart disease. Omentin is a circulating adipokine that is down-regulated by obesity. METHODS: In patients who underwent successful reperfusion treatment after acute myocardial infarction, cardiac function and perfusion defect were assessed by using scintigraphic images. Mice were subjected to myocardial ischemia followed by reperfusion. RESULTS: This study found that high levels of plasma omentin were associated with improvement of heart damage and function after reperfusion therapy in patients with acute myocardial infarction. Systemic administration of human omentin to mice led to a reduction in myocardial infarct size and apoptosis after I/R, which was accompanied by enhanced phosphorylation of AMP-activated protein kinase (AMPK) and Akt in the ischemic heart. Fat-specific overexpression of human omentin also resulted in reduction of infarct size after I/R. Blockade of AMPK or Akt activity reversed omentin-induced inhibition of myocardial ischemic damage and apoptosis in mice. In cultured cardiomyocytes, omentin suppressed hypoxia/reoxygenation-induced apoptosis, which was blocked by inactivation of AMPK or Akt. CONCLUSIONS: Our data indicate that omentin functions as an adipokine that ameliorates acute ischemic injury in the heart by suppressing myocyte apoptosis through both AMPK- and Akt-dependent mechanisms.


Asunto(s)
Proteínas Quinasas Activadas por AMP/fisiología , Citocinas/uso terapéutico , Lectinas/uso terapéutico , Daño por Reperfusión Miocárdica/enzimología , Daño por Reperfusión Miocárdica/prevención & control , Proteína Oncogénica v-akt/fisiología , Animales , Apoptosis/genética , Biomarcadores/sangre , Citocinas/sangre , Citocinas/genética , Modelos Animales de Enfermedad , Femenino , Proteínas Ligadas a GPI/sangre , Proteínas Ligadas a GPI/genética , Proteínas Ligadas a GPI/uso terapéutico , Humanos , Lectinas/sangre , Lectinas/genética , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Persona de Mediana Edad , Reperfusión Miocárdica/métodos , Daño por Reperfusión Miocárdica/patología , Intervención Coronaria Percutánea/métodos , Fosforilación/genética
7.
J Biol Chem ; 289(20): 14132-44, 2014 May 16.
Artículo en Inglés | MEDLINE | ID: mdl-24706764

RESUMEN

Strategies to stimulate revascularization are valuable for cardiovascular diseases. Here we identify neuron-derived neurotrophic factor (NDNF)/epidermacan as a secreted molecule that is up-regulated in endothelial cells in ischemic limbs of mice. NDNF was secreted from cultured human endothelial cells, and its secretion was stimulated by hypoxia. NDNF promoted endothelial cell network formation and survival in vitro through activation of Akt/endothelial NOS (eNOS) signaling involving integrin αvß3. Conversely, siRNA-mediated knockdown of NDNF in endothelial cells led to reduction of cellular responses and basal Akt signaling. Intramuscular overexpression of NDNF led to enhanced blood flow recovery and capillary density in ischemic limbs of mice, which was accompanied by enhanced phosphorylation of Akt and eNOS. The stimulatory actions of NDNF on perfusion recovery in ischemic muscles of mice were abolished by eNOS deficiency or NOS inhibition. Furthermore, siRNA-mediated reduction of NDNF in muscles of mice resulted in reduction of perfusion recovery and phosphorylation of Akt and eNOS in response to ischemia. Our data indicate that NDNF acts as an endogenous modulator that promotes endothelial cell function and ischemia-induced revascularization through eNOS-dependent mechanisms. Thus, NDNF can represent a therapeutic target for the manipulation of ischemic vascular disorders.


Asunto(s)
Células Endoteliales/citología , Neovascularización Fisiológica , Factores de Crecimiento Nervioso/metabolismo , Animales , Circulación Sanguínea , Vasos Sanguíneos/citología , Vasos Sanguíneos/patología , Vasos Sanguíneos/fisiología , Vasos Sanguíneos/fisiopatología , Células COS , Supervivencia Celular , Chlorocebus aethiops , Células Endoteliales/patología , Humanos , Isquemia/metabolismo , Isquemia/patología , Isquemia/fisiopatología , Masculino , Ratones , Ratones Endogámicos C57BL , Factores de Crecimiento Nervioso/genética , Óxido Nítrico Sintasa de Tipo III/metabolismo , Fenotipo , Fosforilación , Proteínas Proto-Oncogénicas c-akt/metabolismo
8.
PLoS One ; 8(12): e83183, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-24358263

RESUMEN

Obese states characterized by chronic inflammation are closely linked to the development of metabolic dysfunction. We identified adipolin/CTRP12 as an insulin-sensitizing and anti-inflammatory adipokine. Although obese conditions down-regulate adipolin expression, its molecular mechanism is largely unknown. Here we show that the transcriptional regulator Krüppel-like factor (KLF) 15 is involved in the regulation of adipolin expression in adipocytes. White adipose tissue from diet-induced obese (DIO) mice showed decreased expression of KLF9 and KLF15 among several KLFs, which was accompanied by reduced expression of adipolin. In cultured 3T3L1 adipocytes, treatment with TNFα significantly reduced the mRNA levels of KLF9, KLF15 and adipolin. Adenovirus-mediated overexpression of KLF15 but not KLF9 reversed TNFα-induced reduction of adipolin expression in adipocytes. Conversely, gene targeting ablation of KLF15 attenuated adipolin expression in adipocytes. Expression of KLF15 but not KLF9 enhanced the promoter activity of adipolin in HEK293 cells. Pretreatment of 3T3L1 adipocytes with the JNK inhibitor SP600125, but not p38 MAPK inhibitor SB203580 blocked the inhibitory effects of TNFα on adipolin and KLF15 expression. These data suggest that adipose inflammation under conditions of obesity suppresses adipolin expression via JNK-dependent down-regulation of KLF15 in adipocytes.


Asunto(s)
Adipocitos/metabolismo , Adipoquinas/genética , Proteínas de Unión al ADN/fisiología , Regulación de la Expresión Génica , Factores de Transcripción/fisiología , Células 3T3-L1 , Adipocitos/efectos de los fármacos , Adipoquinas/metabolismo , Animales , Regulación de la Expresión Génica/efectos de los fármacos , Células HEK293 , Humanos , Factores de Transcripción de Tipo Kruppel , Ratones , Ratones Endogámicos C57BL , Obesidad/genética , Obesidad/metabolismo , Transcripción Genética/efectos de los fármacos , Factor de Necrosis Tumoral alfa/farmacología
9.
J Am Heart Assoc ; 2(5): e000438, 2013 Sep 19.
Artículo en Inglés | MEDLINE | ID: mdl-24052499

RESUMEN

BACKGROUND: Obesity is linked with an increased risk of lymphedema, which is a serious clinical problem. Adiponectin is a circulating adipokine that is down-regulated in obese states. We investigated the effects of adiponectin on lymphatic vessel formation in a model of lymphedema and dissected its mechanisms. METHODS AND RESULTS: A mouse model of lymphedema was created via ablation of tail surface lymphatic network. Adiponectin-knockout mice showed the greater diameter of the injured tail compared with wild-type mice, which was associated with lower numbers of lymphatic endothelial cells (LECs). Systemic delivery of adiponectin reduced the thickness of the injured tail and enhanced LEC formation in wild-type and adiponectin-knockout mice. Adiponectin administration also improved the edema of injured tails in obese KKAy mice. Treatment with adiponectin protein stimulated the differentiation of human LECs into tubelike structures and increased LEC viability. Adiponectin treatment promoted the phosphorylation of AMP-activated protein kinase (AMPK), Akt, and endothelial nitric oxide synthase n LECs. Blockade of AMPK or Akt activity abolished adiponectin-stimulated increase in LEC differentiation and viability and endothelial nitric oxide synthase phosphorylation. Inhibition of AMPK activation also suppressed adiponectin-induced Akt phosphorylation in LECs. In contrast, inactivation of Akt signaling had no effects on adiponectin-mediated AMPK phosphorylation in LECs. Furthermore, adiponectin administration did not affect the thickening of the damaged tail in endothelial nitric oxide synthase-knockout mice. CONCLUSIONS: Adiponectin can promote lymphatic vessel formation via activation of AMPK/Akt/endothelial nitric oxide synthase signaling within LECs, thereby leading to amelioration of lymphedema.


Asunto(s)
Adiponectina/fisiología , Linfangiogénesis , Linfedema/etiología , Animales , Células Endoteliales/fisiología , Masculino , Ratones , Ratones Noqueados , Óxido Nítrico Sintasa de Tipo III/fisiología , Proteínas Proto-Oncogénicas c-akt/fisiología
10.
Sci Rep ; 3: 1418, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23475393

RESUMEN

Angiogenic cell therapy represents a novel strategy for ischemic diseases, but some patients show poor responses. We investigated the therapeutic potential of an induced pluripotent stem (iPS) cell sheet created by a novel magnetite tissue engineering technology (Mag-TE) for reparative angiogenesis. Mouse iPS cell-derived Flk-1(+) cells were incubated with magnetic nanoparticle-containing liposomes (MCLs). MCL-labeled Flk-1(+) cells were mixed with diluted extracellular matrix (ECM) precursor and a magnet was placed on the reverse side. Magnetized Flk-1(+) cells formed multi-layered cell sheets according to magnetic force. Implantation of the Flk-1(+) cell sheet accelerated revascularization of ischemic hindlimbs relative to the contralateral limbs in nude mice as measured by laser Doppler blood flow and capillary density analyses. The Flk-1(+) cell sheet also increased the expressions of VEGF and bFGF in ischemic tissue. iPS cell-derived Flk-1(+) cell sheets created by this novel Mag-TE method represent a promising new modality for therapeutic angiogenesis.


Asunto(s)
Células Madre Pluripotentes Inducidas/citología , Nanopartículas de Magnetita/química , Ingeniería de Tejidos , Animales , Técnicas de Cultivo de Célula , Células Cultivadas , Matriz Extracelular/metabolismo , Factor 2 de Crecimiento de Fibroblastos/metabolismo , Miembro Posterior/metabolismo , Células Madre Pluripotentes Inducidas/metabolismo , Células Madre Pluripotentes Inducidas/trasplante , Isquemia/metabolismo , Isquemia/patología , Isquemia/terapia , Flujometría por Láser-Doppler , Liposomas/química , Ratones , Ratones Desnudos , Neovascularización Fisiológica , Factor A de Crecimiento Endotelial Vascular/metabolismo
11.
FASEB J ; 27(1): 25-33, 2013 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-22972916

RESUMEN

Obesity is closely associated with the progression of vascular disorders, including atherosclerosis and postangioplasty restenosis. C1q/TNF-related protein (CTRP) 9 is an adipocytokine that is down-regulated in obese mice. Here we investigated whether CTRP9 modulates neointimal hyperplasia and vascular smooth muscle cell (VSMC) proliferation in vivo and in vitro. Left femoral arteries of wild-type (WT) mice were injured by a steel wire. An adenoviral vector expressing CTRP9 (Ad-CTRP9) or ß-galactosidase as a control was intravenously injected into WT mice 3 d before vascular injury. Delivery of Ad-CTRP9 significantly attenuated the neointimal thickening and the number of bromodeoxyuridine-positive proliferating cells in the injured arteries compared with that of control. Treatment of VSMCs with CTRP9 protein attenuated the proliferative and chemotactic activities induced by growth factors including platelet-derived growth factor (PDGF)-BB, and suppressed PDGF-BB-stimulated phosphorylation of ERK. CTRP9 treatment dose-dependently increased cAMP levels in VSMCs. Blockade of cAMP-PKA pathway reversed the inhibitory effect of CTRP9 on DNA synthesis and ERK phosphorylation in response to PDGF-BB. The present data indicate that CTRP9 functions to attenuate neointimal formation following vascular injury through its ability to inhibit VSMC growth via cAMP-dependent mechanism, suggesting that the therapeutic approaches to enhance CTRP9 production could be valuable for prevention of vascular restenosis after angioplasty.


Asunto(s)
Adiponectina/fisiología , Tejido Adiposo/metabolismo , Proliferación Celular , Glicoproteínas/fisiología , Músculo Liso Vascular/citología , Túnica Íntima/crecimiento & desarrollo , Animales , Western Blotting , Células Cultivadas , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Fosforilación , Proteínas Recombinantes/metabolismo , Péptidos y Proteínas Asociados a Receptores de Factores de Necrosis Tumoral
12.
Biochem Biophys Res Commun ; 428(1): 155-9, 2012 Nov 09.
Artículo en Inglés | MEDLINE | ID: mdl-23068097

RESUMEN

Obesity is highly associated with the development of insulin resistance and type 2 diabetes. Recently we found that adipolin/CRTP12 is an adipocytokine that exerts beneficial actions on glucose metabolism. Here we investigated the regulation of circulating adipolin under conditions of obesity and assessed its potential mechanisms. Both full and cleaved forms of adipolin were observed in mouse plasma. Diet-induced obese (DIO) mice showed a significant reduction of plasma levels of full and total (full and cleaved) adipolin compared with control mice, resulting in an increase in the ratio of cleaved to full isoform. In vitro gene transfection studies using HEK293 cells revealed that a deletion mutant of adipolin gene (Δaa90-93) caused a reduction of cleaved production of adipolin in media. A bioinformatics analysis of adipolin amino acid sequence indicated the potential involvement of the family of proprotein convertases (PCs) in cleavage of adipolin. Treatment of 3T3-L1 adipocytes with an inhibitor for PCs abolished the expression of cleaved adipolin form in the media. The expression of furin, the member of PCs, was increased in adipose tissue of DIO mice. Furin expression was also increased in cultured adipocytes by treatment with an inducer of inflammation. These data suggest that obesity states facilitate the cleavage of adipolin presumably through upregulation of furin in adipose tissue.


Asunto(s)
Adipoquinas/metabolismo , Furina/biosíntesis , Obesidad/metabolismo , Células 3T3-L1 , Adipoquinas/sangre , Adipoquinas/genética , Tejido Adiposo/enzimología , Secuencias de Aminoácidos , Animales , Dieta/efectos adversos , Células HEK293 , Humanos , Ratones , Ratones Endogámicos C57BL , Obesidad/sangre , Proproteína Convertasas/antagonistas & inhibidores , Proproteína Convertasas/metabolismo , Eliminación de Secuencia , Delgadez/sangre , Delgadez/metabolismo , Regulación hacia Arriba
13.
Circulation ; 126(14): 1728-38, 2012 Oct 02.
Artículo en Inglés | MEDLINE | ID: mdl-22929303

RESUMEN

BACKGROUND: Acute coronary syndrome is a leading cause of death in developed countries. Follistatin-like 1 (FSTL1) is a myocyte-derived secreted protein that is upregulated in the heart in response to ischemic insult. Here, we investigated the therapeutic impact of FSTL1 on acute cardiac injury in small and large preclinical animal models of ischemia/reperfusion and dissected its molecular mechanism. METHODS AND RESULTS: Administration of human FSTL1 protein significantly attenuated myocardial infarct size in a mouse or pig model of ischemia/reperfusion, which was associated with a reduction of apoptosis and inflammatory responses in the ischemic heart. Administration of FSTL1 enhanced the phosphorylation of AMP-activated protein kinase in the ischemia/reperfusion-injured heart. In cultured cardiac myocytes, FSTL1 suppressed apoptosis in response to hypoxia/reoxygenation and lipopolysaccharide-stimulated expression of proinflammatory genes through its ability to activate AMP-activated protein kinase. Ischemia/reperfusion led to enhancement of bone morphogenetic protein-4 expression and Smad1/5/8 phosphorylation in the heart, and FSTL1 suppressed the increased phosphorylation of Smad1/5/8 in ischemic myocardium. Treating cardiac myocytes with FSTL1 abolished the bone morphogenetic protein-4-stimulated increase in apoptosis, Smad1/5/8 phosphorylation, and proinflammatory gene expression. In cultured macrophages, FSTL1 diminished lipopolysaccharide-stimulated expression of proinflammatory genes via activation of AMP-activated protein kinase and abolished bone morphogenetic protein-4-dependent induction of proinflammatory mediators. CONCLUSIONS: Our data indicate that FSTL1 can prevent myocardial ischemia/reperfusion injury by inhibiting apoptosis and inflammatory response through modulation of AMP-activated protein kinase- and bone morphogenetic protein-4-dependent mechanisms, suggesting that FSTL1 could represent a novel therapeutic target for post-myocardial infarction, acute coronary syndrome.


Asunto(s)
Modelos Animales de Enfermedad , Proteínas Relacionadas con la Folistatina/administración & dosificación , Isquemia Miocárdica/tratamiento farmacológico , Animales , Apoptosis/fisiología , Células Cultivadas , Proteínas Relacionadas con la Folistatina/biosíntesis , Proteínas Relacionadas con la Folistatina/fisiología , Humanos , Infusiones Intravenosas , Ratones , Ratones Endogámicos C57BL , Isquemia Miocárdica/metabolismo , Isquemia Miocárdica/patología , Porcinos , Resultado del Tratamiento
14.
J Biol Chem ; 287(23): 18965-73, 2012 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-22514273

RESUMEN

Ischemic heart disease is the major cause of death in Western countries. CTRP9 (C1q/TNF-related protein 9) is a fat-derived plasma protein that has salutary effects on glucose metabolism and vascular function. However, the functional role of CTRP9 in ischemic heart disease has not been clarified. Here, we examined the regulation of CTRP9 in response to acute cardiac injury and investigated whether CTRP9 modulates cardiac damage after ischemia and reperfusion. Myocardial ischemia-reperfusion injury resulted in reduced plasma CTRP9 levels and increased plasma free fatty acid levels, which were accompanied by a decrease in CTRP9 expression and an increase in NADPH oxidase component expression in fat tissue. Treatment of cultured adipocytes with palmitic acid or hydrogen peroxide reduced CTRP9 expression. Systemic administration of CTRP9 to wild-type mice, before the induction of ischemia or at the time of reperfusion, led to a reduction in myocardial infarct size following ischemia-reperfusion. Administration of CTRP9 also attenuated myocyte apoptosis in ischemic heart, which was accompanied by increased phosphorylation of AMP-activated protein kinase (AMPK). Treatment of cardiac myocytes with CTRP9 protein reduced apoptosis in response to hypoxia/reoxygenation and stimulated AMPK phosphorylation. Blockade of AMPK activity reversed the suppressive actions of CTRP9 on cardiomyocyte apoptosis. Knockdown of adiponectin receptor 1 diminished CTRP9-induced increases in AMPK phosphorylation and survival of cardiac myocytes. Our data suggest that CTRP9 protects against acute cardiac injury following ischemia-reperfusion via an AMPK-dependent mechanism.


Asunto(s)
Proteínas Quinasas Activadas por AMP/metabolismo , Adiponectina/sangre , Apoptosis , Glicoproteínas/sangre , Daño por Reperfusión Miocárdica/metabolismo , Miocardio/metabolismo , Miocitos Cardíacos/metabolismo , Células 3T3-L1 , Adipocitos/metabolismo , Adipocitos/patología , Adiponectina/farmacología , Animales , Ácidos Grasos no Esterificados/sangre , Regulación de la Expresión Génica , Glicoproteínas/farmacología , Masculino , Ratones , Ratones Obesos , Daño por Reperfusión Miocárdica/patología , Daño por Reperfusión Miocárdica/prevención & control , Miocardio/patología , Miocitos Cardíacos/patología , Ácido Palmítico/metabolismo , Fosforilación/efectos de los fármacos , Ratas
15.
J Biol Chem ; 287(1): 408-417, 2012 Jan 02.
Artículo en Inglés | MEDLINE | ID: mdl-22081609

RESUMEN

Obesity-related diseases are associated with vascular dysfunction and impaired revascularization. Omentin is a fat-derived secreted protein, which is down-regulated in association with obese complications. Here, we investigated whether omentin modulates endothelial cell function and revascularization processes in vitro and in vivo. Systemic delivery of an adenoviral vector expressing omentin (Ad-omentin) enhanced blood flow recovery and capillary density in ischemic limbs of wild-type mice in vivo, which were accompanied by increased phosphorylation of Akt and endothelial nitric oxide synthase (eNOS). In cultured human umbilical vein endothelial cells (HUVECs), a physiological concentration of recombinant omentin protein increased differentiation into vascular-like structures and decreased apoptotic activity under conditions of serum starvation. Treatment with omentin protein stimulated the phosphorylation of Akt and eNOS in HUVECs. Inhibition of Akt signaling by treatment with dominant-negative Akt or LY294002 blocked the stimulatory effects of omentin on differentiation and survival of HUVECs and reversed omentin-stimulated eNOS phosphorylation. Pretreatment with the NOS inhibitor also reduced the omentin-induced increase in HUVEC differentiation and survival. Omentin protein also stimulated the phosphorylation of AMP-activated protein kinase in HUVECs. Transduction with dominant-negative AMP-activated protein kinase diminished omentin-induced phosphorylation of Akt and omentin-stimulated increase in HUVEC differentiation and survival. Of importance, in contrast to wild-type mice, systemic administration of Ad-omentin did not affect blood flow in ischemic muscle in eNOS-deficient mice in vivo. These data indicate that omentin promotes endothelial cell function and revascularization in response to ischemia through its ability to stimulate an Akt-eNOS signaling pathway.


Asunto(s)
Tejido Adiposo/metabolismo , Citocinas/metabolismo , Células Endoteliales de la Vena Umbilical Humana/patología , Isquemia/patología , Isquemia/fisiopatología , Lectinas/metabolismo , Neovascularización Fisiológica , Óxido Nítrico Sintasa de Tipo III/metabolismo , Proteínas Quinasas Activadas por AMP/metabolismo , Adenoviridae/genética , Animales , Vasos Sanguíneos/fisiopatología , Diferenciación Celular , Supervivencia Celular , Citocinas/genética , Activación Enzimática , Proteínas Ligadas a GPI/genética , Proteínas Ligadas a GPI/metabolismo , Vectores Genéticos/genética , Células Endoteliales de la Vena Umbilical Humana/enzimología , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Humanos , Espacio Intracelular/metabolismo , Isquemia/enzimología , Isquemia/metabolismo , Lectinas/genética , Ratones , Fosforilación , Proteínas Proto-Oncogénicas c-akt/metabolismo , Transducción de Señal
16.
Atherosclerosis ; 219(2): 811-4, 2011 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-21925659

RESUMEN

OBJECTIVE: Obesity is closely associated with an increased risk for cardiovascular morbidity and mortality. Omentin is a fat-derived secreted factor that is downregulated in obesity. We investigated whether circulating omentin associates with the prevalence of coronary artery disease (CAD). METHODS: The consecutive 78 male subjects were enrolled from patients who underwent coronary angiography. Sixty one age-matched male subjects served as controls. Plasma omentin concentration was measured by enzyme-linked immunosorbent assay. RESULTS: Plasma levels of omentin correlated negatively with body mass index (BMI), systolic blood pressure, hemoglobin A1c and total cholesterol levels, and positively with HDL cholesterol and adiponectin levels. Circulating omentin was independently associated with hemoglobin A1c and HDL cholesterol in a multiple regression analysis. Plasma levels of omentin were markedly lower in CAD patients than in control subjects (CAD: 102.8 ± 69.0 ng/ml, control: 454.7 ± 128.6 ng/ml, P < 0.001). Multiple logistic regression analysis with BMI, systolic blood pressure, glucose, hemoglobin A1c, HDL cholesterol, adiponectin and omentin revealed that plasma omentin levels were independently correlated with CAD. CONCLUSION: These data indicate that low levels of omentin are closely linked with the presence of CAD and that omentin serves as a novel biomarker for CAD.


Asunto(s)
Estenosis Coronaria/sangre , Citocinas/sangre , Lectinas/sangre , Anciano , Anciano de 80 o más Años , Biomarcadores/sangre , Estudios de Casos y Controles , Angiografía Coronaria , Estenosis Coronaria/diagnóstico por imagen , Estenosis Coronaria/epidemiología , Regulación hacia Abajo , Ensayo de Inmunoadsorción Enzimática , Proteínas Ligadas a GPI/sangre , Humanos , Japón/epidemiología , Modelos Logísticos , Masculino , Oportunidad Relativa , Valor Predictivo de las Pruebas , Prevalencia , Medición de Riesgo , Factores de Riesgo , Índice de Severidad de la Enfermedad
17.
Hypertens Res ; 34(12): 1309-12, 2011 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-21814208

RESUMEN

Obesity is causally linked with the development of atherosclerosis. Omentin is an adipocytokine whose concentrations are reduced in obese individuals. Here we examined the relationship between plasma omentin levels and carotid intima-media thickness (IMT), a marker of early atherosclerosis, in apparently healthy Japanese men. Participants were 100 Japanese men who underwent a medical checkup. Maximal IMT (max-IMT) and mean-IMT in common carotid artery were measured by high-resolution carotid ultrasound system. Plasma omentin concentrations were determined by enzyme-linked immunosorbent assay. Circulating omentin levels correlated negatively with body mass index, waist circumference, fasting glucose, creatinine, max-IMT and mean-IMT, and positively with estimated glomerular filtration rates (eGFR). Single regression analysis demonstrated that max-IMT associated with age, eGFR and omentin levels, and that mean-IMT associated with age, fasting glucose, eGFR and omentin levels. Multiple regression analysis revealed that omentin levels, together with age, correlated with max-IMT and mean-IMT. Our data document that circulating omentin levels independently and negatively associate with carotid IMT in this population, suggesting that measurement of omentin may be useful for assessment of carotid IMT.


Asunto(s)
Arterias Carótidas/patología , Grosor Intima-Media Carotídeo , Citocinas/sangre , Citocinas/genética , Lectinas/sangre , Lectinas/genética , Envejecimiento/fisiología , Glucemia/metabolismo , Índice de Masa Corporal , Arterias Carótidas/diagnóstico por imagen , Creatinina/sangre , Ensayo de Inmunoadsorción Enzimática , Femenino , Proteínas Ligadas a GPI/sangre , Proteínas Ligadas a GPI/genética , Tasa de Filtración Glomerular , Humanos , Japón , Masculino , Persona de Mediana Edad , Circunferencia de la Cintura
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA