Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 32
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Cell Biochem Biophys ; 74(1): 59-65, 2016 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-26972302

RESUMO

Patients with hyperhomocysteinemia (HHcy), or elevated plasma homocysteine (Hcy), are at higher risk of developing arrhythmias and sudden cardiac death; however, the mechanisms are unknown. In this study, the effects of HHcy on sinus node function, atrioventricular conduction, and ventricular vulnerability were investigated by electrophysiological (EP) analysis, and the role of magnesium (Mg(2+)), an endogenous N-methyl-D-aspartate (NMDA) receptor antagonist, in attenuating EP changes due to HHcy was explored. Wild-type mice (WT) and mice receiving Hcy in the drinking water for 12 weeks (DW) were subjected to electrocardiographic and EP studies. DW compared to WT had significantly shorter RR, PR, QT, and HV intervals, corrected sinus node recovery times (CSNRT), Wenckebach periodicity (WP), atrioventricular nodal effective refractory periods (AVNERP), and right ventricular effective refractory periods (RVERP). To examine the role of Mg(2+) in mitigating conduction changes in HHcy, WT, DW, and heterozygous cystathionine-ß-synthase knockout mice (CBS (+/-) ) were subjected to repeat EP studies before and after administration of low-dose magnesium sulfate (20 mg/kg). Mg(2+) had no effect on EP variables in WT, but significantly slowed CSNRT, WP, and AVNERP in DW, as well as WP and AVNERP in CBS (+/-) . These findings suggest that ionic channels modulated by Mg(2+) may contribute to HHcy-induced conduction abnormalities.


Assuntos
Hiper-Homocisteinemia/fisiopatologia , Magnésio/metabolismo , Nó Sinoatrial/fisiopatologia , Potenciais de Ação , Animais , Cistationina beta-Sintase/genética , Cistationina beta-Sintase/metabolismo , Hiper-Homocisteinemia/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Período Refratário Eletrofisiológico , Nó Sinoatrial/metabolismo
2.
J Mol Cell Cardiol ; 92: 163-173, 2016 03.
Artigo em Inglês | MEDLINE | ID: mdl-26827898

RESUMO

AIMS: Although the cardiovascular benefits of exercise are well known, exercise induced effects and mechanisms in prevention of cardiomyopathy are less clear during obesity associated type-2 diabetes. The current study assessed the impact of moderate intensity exercise on diabetic cardiomyopathy by examining cardiac function and structure and mitochondrial function. METHODS: Obese-diabetic (db/db), and lean control (db/+) mice, were subjected to a 5 week, 300 m run on a tread-mill for 5 days/week at the speeds of 10-11 m/min. Various physiological parameters were recorded and the heart function was evaluated with M-mode echocardiography. Contraction parameters and calcium transits were examined on isolated cardiomyocytes. At the molecular level: connexin 43 and 37 (Cx43 and 37) levels, mitochondrial biogenesis regulators: Mfn2 and Drp-1 levels, mitochondrial trans-membrane potential and cytochrome c leakage were assessed through western blotting immunohistochemistry and flow cytometry. Ability of exercise to reverse oxygen consumption rate (OCR), tissue ATP levels, and cardiac fibrosis were also determined. RESULTS: The exercise regimen was able to prevent diabetic cardiac functional deficiencies: ejection fraction (EF) and fractional shortening (FS). Improvements in contraction velocity and contraction maximum were noted with the isolated cardiomyocytes. Restoration of interstitial and micro-vessels associated Cx43 levels and improved gap junction intercellular communication (GJIC) were observed. The decline in the Mfn2/Drp-1 ratio in the db/db mice hearts was prevented after exercise. The exercise regimen further attenuated transmembrane potential decline and cytochrome c leakage. These corrections further led to improvements in OCR and tissue ATP levels and reduction in cardiac fibrosis. CONCLUSIONS: Moderate intensity exercise produced significant cardiovascular benefits by improving mitochondrial function through restoration of Cx43 networks and mitochondrial trans-membrane potential and prevention of excessive mitochondrial fission.


Assuntos
Conexina 43/metabolismo , Diabetes Mellitus Tipo 2/terapia , Cardiomiopatias Diabéticas/terapia , Mitocôndrias/metabolismo , Condicionamento Físico Animal , Trifosfato de Adenosina/metabolismo , Animais , Conexina 43/genética , Conexinas/genética , Conexinas/metabolismo , Diabetes Mellitus Tipo 2/complicações , Diabetes Mellitus Tipo 2/fisiopatologia , Cardiomiopatias Diabéticas/fisiopatologia , Humanos , Potencial da Membrana Mitocondrial/genética , Camundongos , Camundongos Obesos , Miócitos Cardíacos/metabolismo , Obesidade , Estresse Oxidativo/genética , Consumo de Oxigênio/genética , Proteína alfa-4 de Junções Comunicantes
3.
Can J Physiol Pharmacol ; 93(9): 755-63, 2015 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-26176406

RESUMO

Paraoxanase-1 (PON1) is an HDL-associated enzyme that contributes to the antioxidant and antiatherosclerotic properties of HDL. Lack of PON1 results in dysfunctional HDL. HHcy is a risk factor for cardiovascular disorders, and instigates vascular dysfunction and ECM remodeling. Although studies have reported HHcy during atherosclerosis, the exact mechanism is unclear. Here, we hypothesize that dysfunctional HDL due to lack of PON1 contributes to endothelial impairment and atherogenesis through HHcy-induced ECM re-modeling. To verify this hypothesis, we used C57BL6/J and PON1 knockout mice (KO) and fed them an atherogenic diet. The expression of Akt, ADMA, and DDAH, as well as endothelial gap junction proteins such as Cx-37 and Cx-40 and eNOS was measured for vascular dysfunction and inflammation. We observed that cardiac function was decreased and plasma Hcy levels were increased in PON1 KO mice fed the atherogenic diet compared with the controls. Expression of Akt, eNOS, DDAH, Cx-37, and Cx-40 was decreased, and the expression of MMP-9 and ADMA was increased in PON1 KO mice fed an atherogenic diet compared with the controls. Our results suggest that HHcy plays an intricate role in dysfunctional HDL, owing to the lack of PON1. This contributes to vascular endothelial impairment and atherosclerosis through MMP-9-induced vascular remodeling.


Assuntos
Arildialquilfosfatase/metabolismo , Aterosclerose/fisiopatologia , Hiper-Homocisteinemia/sangue , Lipoproteínas HDL/sangue , Amidoidrolases/biossíntese , Animais , Arginina/análogos & derivados , Arginina/biossíntese , Arildialquilfosfatase/deficiência , Arildialquilfosfatase/genética , Aterosclerose/sangue , Aterosclerose/genética , Aterosclerose/patologia , Pressão Sanguínea/genética , Pressão Sanguínea/fisiologia , Conexinas/biossíntese , Dieta Aterogênica , Endotélio Vascular/metabolismo , Fibrose/induzido quimicamente , Fibrose/patologia , Masculino , Metaloproteinase 9 da Matriz/biossíntese , Camundongos , Camundongos Knockout , Óxido Nítrico Sintase Tipo III/biossíntese , Proteínas Proto-Oncogênicas c-akt/biossíntese , Disfunção Ventricular Esquerda/genética , Disfunção Ventricular Esquerda/fisiopatologia , Proteína alfa-5 de Junções Comunicantes , Proteína alfa-4 de Junções Comunicantes
4.
Can J Physiol Pharmacol ; 93(7): 577-84, 2015 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-26118387

RESUMO

INTRODUCTION: Hyperhomocysteinemia (HHcy) is associated with inflammatory diseases and is known to increase the production of reactive oxygen species (ROS), matrix metalloproteinase (MMP)-9, and inducible nitric oxide synthase, and to decrease endothelial nitric oxide production. However, the impact of HHcy on macrophage phenotype differentiation is not well-established. It has been documented that macrophages have 2 distinct phenotypes: the "classically activated/destructive" (M1), and the "alternatively activated/constructive" (M2) subtypes. We hypothesize that HHcy increases M1 macrophage differentiation through extracellular matrix metalloproteinase inducer (EMMPRIN), a known inducer of matrix metalloproteinases. METHODS: murine J774A.1 and Raw 264.7 macrophages were treated with 100 and 500 µmol/L Hcy, respectively, for 24 h. Samples were analyzed using Western blotting and immunocytochemistry. RESULTS: Homocysteine treatment increased cluster of differentiation 40 (CD40; M1 marker) in J774A.1 and Raw 264.7 macrophages. MMP-9 was induced in both cell lines. EMMPRIN protein expression was also increased in both cell lines. Blocking EMMPRIN function by pre-treating cells with anti-EMMPRIN antibody, with or without Hcy, resulted in significantly lower expression of CD40 in both cell lines by comparison with the controls. A DCFDA assay demonstrated increased ROS production in both cell lines with Hcy treatment when compared with the controls. CONCLUSION: Our results suggest that HHcy results in an increase of the M1 macrophage phenotype. This effect seems to be at least partially mediated by EMMPRIN induction.


Assuntos
Basigina/biossíntese , Diferenciação Celular/efeitos dos fármacos , Homocisteína/farmacologia , Macrófagos/efeitos dos fármacos , Espécies Reativas de Oxigênio/metabolismo , Animais , Anticorpos Monoclonais/farmacologia , Western Blotting , Antígenos CD40/biossíntese , Técnicas de Cultura de Células , Linhagem Celular , Relação Dose-Resposta a Droga , Imuno-Histoquímica , Macrófagos/metabolismo , Metaloproteinase 9 da Matriz/metabolismo , Camundongos , Transdução de Sinais
5.
Can J Physiol Pharmacol ; 92(7): 575-82, 2014 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-24923386

RESUMO

Regular exercise is a great medicine with its benefits encompassing everything from prevention of cardiovascular risk to alleviation of different muscular myopathies. Interestingly, elevated levels of homocysteine (Hcy), also known as hyperhomocysteinemia (HHcy), antagonizes beta-2 adrenergic receptors (ß2AR), gamma amino butyric acid (GABA), and peroxisome proliferator-activated receptor-gamma (PPARγ) receptors. HHcy also stimulates an elevation of the M1/M2 macrophage ratio, resulting in a more inflammatory profile. In this review we discuss several potential targets altered by HHcy that result in myopathy and excessive fat accumulation. Several of these HHcy mediated changes can be countered by exercise and culminate into mitigation of HHcy induced myopathy and metabolic syndrome. We suggest that exercise directly impacts levels of Hcy, matrix metalloproteinase 9 (MMP-9), macrophages, and G-protein coupled receptors (GPCRs, especially Gs). While HHcy promotes the M1 macrophage phenotype, it appears that exercise may diminish the M1/M2 ratio, resulting in a less inflammatory phenotype. HHcy through its influence on GPCRs, specifically ß2AR, PPARγ and GABA receptors, promotes accumulation of white fat, whereas exercise enhances the browning of white fat and counters HHcy-mediated effects on GPCRs. Alleviation of HHcy-associated pathologies with exercise also includes reversal of excessive MMP-9 activation. Moreover, exercise, by reducing plasma Hcy levels, may prevent skeletal muscle myopathy, improve exercise capacity and rescue the obese phenotype. The purpose of this review is to summarize the pathological conditions surrounding HHcy and to clarify the importance of regular exercise as a method of disease prevention.


Assuntos
Adipócitos Brancos/metabolismo , Exercício Físico , Hiper-Homocisteinemia/metabolismo , Hiper-Homocisteinemia/patologia , Macrófagos/metabolismo , Metaloproteinase 9 da Matriz/metabolismo , Músculo Esquelético/metabolismo , Animais , Humanos , Hiper-Homocisteinemia/complicações , Inflamação/metabolismo , Síndrome Metabólica/etiologia , Síndrome Metabólica/prevenção & controle , Doenças Musculares/etiologia , Doenças Musculares/prevenção & controle , Receptores Acoplados a Proteínas G/metabolismo
6.
Can J Physiol Pharmacol ; 92(7): 583-91, 2014 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-24905188

RESUMO

Therapeutic approaches for cardiac regenerative mechanisms have been explored over the past decade to target various cardiovascular diseases (CVD). Structural and functional aberrations of mitochondria have been observed in CVD. The significance of mitochondrial maturation and function in cardiomyocytes is distinguished by their attribution to embryonic stem cell differentiation into adult cardiomyocytes. An abnormal fission process has been implicated in heart failure, and treatment with mitochondrial division inhibitor 1 (Mdivi-1), a specific inhibitor of dynamin related protein-1 (Drp-1), has been shown to improve cardiac function. We recently observed that the ratio of mitofusin 2 (Mfn2; a fusion protein) and Drp-1 (a fission protein) was decreased during heart failure, suggesting increased mitophagy. Treatment with Mdivi-1 improved cardiac function by normalizing this ratio. Aberrant mitophagy and enhanced oxidative stress in the mitochondria contribute to abnormal activation of MMP-9, leading to degradation of the important gap junction protein connexin-43 (Cx-43) in the ventricular myocardium. Reduced Cx-43 levels were associated with increased fibrosis and ventricular dysfunction in heart failure. Treatment with Mdivi-1 restored MMP-9 and Cx-43 expression towards normal. In this review, we discuss mitochondrial dynamics, its relation to MMP-9 and Cx-43, and the therapeutic role of fission inhibition in heart failure.


Assuntos
GTP Fosfo-Hidrolases/genética , Insuficiência Cardíaca/metabolismo , Proteínas Associadas aos Microtúbulos/genética , Proteínas Mitocondriais/genética , Animais , Conexina 43/metabolismo , Dinaminas , GTP Fosfo-Hidrolases/metabolismo , Insuficiência Cardíaca/tratamento farmacológico , Insuficiência Cardíaca/fisiopatologia , Humanos , Metaloproteinase 9 da Matriz/metabolismo , Proteínas Associadas aos Microtúbulos/metabolismo , Dinâmica Mitocondrial , Proteínas Mitocondriais/metabolismo , Quinazolinonas/farmacologia , Quinazolinonas/uso terapêutico
7.
Mol Biol Rep ; 41(8): 4889-98, 2014 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-24874304

RESUMO

Hyperhomocysteinemia (HHcy) is associated with cognitive decline and hearing loss due to vascular dysfunction. Although we have shown that HHcy-induced increased expression of matrix metalloproteinase-9 (MMP-9) is associated with cochlear pathology in cystathionine-ß-synthase heterozygous (CBS(+/-)) mice, it is still unclear whether MMP-9 contributes to functional deficit in cognition and hearing. Therefore, we hypothesize that HHcy-induced MMP-9 activation causes vascular, cerebral and cochlear remodeling resulting in diminished cognition and hearing. Wildtype (WT), CBS(+/-), MMP-9(-/-) and CBS(+/-)/MMP-9(-/-) double knock-out (DKO) mice were genotyped and used. Doppler flowmetry of internal carotid artery (ICA) was performed for peak systolic velocity [PSV], pulsatility index [PI] and resistive index [RI]. Cognitive functions were assessed by Novel Object Recognition Test (NORT) and for cochlear function Auditory brainstem response (ABR) was elicited. Peak systolic velocity, pulsatility and resistive indices of ICA were decreased in CBS(+/-) mice, indicating reduced perfusion. ABR threshold was increased and maximum ABR amplitude and NORT indices (recognition, discrimination) were decreased in CBS(+/-) mice compared to WT and MMP-9(-/-). All these parameters were attenuated in DKO mice suggesting a significant role of MMP-9 in HHcy-induced vascular, neural and cochlear pathophysiology. Regression analysis of PSV with ABR and cognitive parameters revealed significant correlation (0.44-0.58). For the first time, MMP-9 has been correlated directly to functional deficits of brain and cochlea, and found to have a significant role. Our data suggests a dual pathology of HHcy occurring due to a decrease in blood supply (vasculo-neural and vasculo-cochlear) and direct tissue remodeling.


Assuntos
Transtornos Cognitivos/etiologia , Deleção de Genes , Transtornos da Audição/etiologia , Hiper-Homocisteinemia/complicações , Metaloproteinase 9 da Matriz/genética , Animais , Velocidade do Fluxo Sanguíneo , Artéria Carótida Interna/fisiologia , Genótipo , Fluxometria por Laser-Doppler , Camundongos , Camundongos Knockout , Fluxo Pulsátil , Análise de Regressão
8.
Int J Biomed Sci ; 10(1): 1-7, 2014 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-24711742

RESUMO

UNLABELLED: Parstatin, a novel protease activated receptor-1 (PAR-1) derived peptide is a potent inhibitor of angiogenesis. We and others have reported that imbalance between angiogenic growth factors and anti-angiogenic factors results in transition from compensatory cardiac hypertrophy to heart failure in a pressure overload condition. Though cardio protective role of parstatin was shown previously in ischemic cardiac injury, its role in pressure overload cardiac injury is yet to unveil. We hypothesize that supplementing anti-parstatin antibody during pressure overload condition augments angiogenesis and ameliorate left ventricular dysfunction and heart failure. To verify this, we created ascending aortic banding in mice to mimic pressure overload condition and then treated mice with anti-parstatin antibody. Left ventricular function was assessed by echocardiography and pressure-volume loop study. Angiogenic growth factors and anti-angiogenic factors along with MMP-2,-9 were evaluated by western blot and immunohistochemistry. RESULTS: our results showed an improved left ventricular function in anti-parstatin treated aortic banding hearts compared to their corresponding wild type controls. Expression of angiogenic growth factor, VEGF, MMP-2 and CD31 expression was increased in treated aortic banding hearts compared to their corresponding wild type controls. Our results suggest that treating pressure overload mice with anti-parstatin antibody augments angiogenesis and ameliorates left ventricular dysfunction.

9.
FASEB J ; 28(8): 3411-22, 2014 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-24739303

RESUMO

Hyperhomocysteinemia (HHcy) is prevalent in patients with hypertension and is an independent risk factor for aortic pathologies. HHcy is known to cause an imbalance between matrix metalloproteinases (MMPs) and tissue inhibitors of metalloproteinases (TIMPs), leading to the accumulation of collagen in the aorta and resulting in stiffness and development of hypertension. Although the exact mechanism of extracellular matrix (ECM) remodeling is unclear, emerging evidence implicates epigenetic regulation involving DNA methylation. Our purpose was to investigate whether 5-aza-2'-deoxycytidine (Aza), a DNA methyltransferase (DNMT1) inhibitor, reduces high blood pressure (BP) by regulating aortic ECM remodeling in HHcy. Wild-type and cystathionine ß-synthase (CBS)(+/-) HHcy mice were treated with Aza (0.5 mg/kg body weight). In HHcy mice, Aza treatment normalized the plasma homocysteine (Hcy) level and BP. Thoracic and abdominal aorta ultrasound revealed a reduction in the resistive index and wall-to-lumen ratio. Vascular response to phenylephrine, acetylcholine, and sodium nitroprusside improved after Aza in HHcy mice. Histology showed a marked reduction in collagen deposition in the aorta. Aza treatment decreased the expression of DNMT1, MMP9, TIMP1, and S-adenosyl homocysteine hydrolase (SAHH) and upregulated methylene tetrahydrofolate reductase (MTHFR). We conclude that reduction of DNA methylation by Aza in HHcy reduces adverse aortic remodeling to mitigate hypertension.


Assuntos
Aorta/fisiopatologia , Azacitidina/análogos & derivados , Metilação de DNA , Epigênese Genética/fisiologia , Hiper-Homocisteinemia/genética , Hipertensão/prevenção & controle , Resistência Vascular/efeitos dos fármacos , Acetilcolina/farmacologia , Adenosil-Homocisteinase/biossíntese , Adenosil-Homocisteinase/genética , Animais , Aorta/química , Aorta/diagnóstico por imagem , Aorta/efeitos dos fármacos , Azacitidina/farmacologia , Colágeno/metabolismo , DNA (Citosina-5-)-Metiltransferase 1 , DNA (Citosina-5-)-Metiltransferases/antagonistas & inibidores , DNA (Citosina-5-)-Metiltransferases/biossíntese , DNA (Citosina-5-)-Metiltransferases/genética , Metilação de DNA/efeitos dos fármacos , Decitabina , Endotélio Vascular/fisiopatologia , Epigênese Genética/genética , Proteínas da Matriz Extracelular/metabolismo , Regulação Enzimológica da Expressão Gênica/efeitos dos fármacos , Homocistinúria/complicações , Homocistinúria/tratamento farmacológico , Homocistinúria/genética , Hiper-Homocisteinemia/complicações , Hiper-Homocisteinemia/fisiopatologia , Hipertensão/etiologia , Hipertensão/genética , Masculino , Metaloproteinase 9 da Matriz/biossíntese , Metaloproteinase 9 da Matriz/genética , Metilenotetra-Hidrofolato Redutase (NADPH2)/biossíntese , Metilenotetra-Hidrofolato Redutase (NADPH2)/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Nitroprussiato/farmacologia , Fenilefrina/farmacologia , Inibidor Tecidual de Metaloproteinase-1/biossíntese , Inibidor Tecidual de Metaloproteinase-1/genética , Ultrassonografia , Resistência Vascular/genética
10.
Am J Physiol Heart Circ Physiol ; 306(8): H1116-27, 2014 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-24585779

RESUMO

Hyperhomocysteinemia (HHcy) is associated with elderly frailty, skeletal muscle injury and malfunction, reduced vascular integrity and function, and mortality. Although HHcy has been implicated in the impairment of angiogenesis after hindlimb ischemia in murine models, the underlying mechanisms are still unclear. We hypothesized that HHcy compromises skeletal muscle perfusion, collateral formation, and arteriogenesis by diminishing postischemic vasculogenic responses in muscle fibers. To test this hypothesis, we created femoral artery ligation in wild-type and heterozygous cystathionine ß-synthase (CBS(+/-)) mice (a model for HHcy) and assessed tissue perfusion, collateral vessel formation, and skeletal muscle function using laser-Doppler perfusion imaging, barium angiography, and fatigue tests. In addition, we assessed postischemic levels of VEGF and levels of its muscle-specific regulators: hypoxia-inducible factor (HIF)-1α and peroxisome proliferator-activated receptor-γ coactivator (PGC)-1α. The observations indicated dysregulation of VEGF, HIF-1α, and PGC-1α levels in ischemic skeletal muscles of CBS(+/-) mice. Concomitant with the reduced ischemic angiogenic responses, we also observed diminished leptin expression and attenuated Akt signaling in ischemic muscle fibers of CBS(+/-) mice. Moreover, there was enhanced atrogene, ubiquitin ligases that conjugate proteins for degradation during muscle atrophy, transcription, and reduced muscle function after ischemia in CBS(+/-) mice. These results suggest that HHcy adversely affects muscle-specific ischemic responses and contributes to muscle frailty.


Assuntos
Membro Posterior/irrigação sanguínea , Hiper-Homocisteinemia/fisiopatologia , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Isquemia/fisiopatologia , Neovascularização Fisiológica/fisiologia , Fatores de Transcrição/metabolismo , Animais , Comportamento Animal , Cistationina beta-Sintase/deficiência , Cistationina beta-Sintase/genética , Artéria Femoral/cirurgia , Expressão Gênica , Ligadura , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Proteínas Musculares/genética , Músculo Esquelético/irrigação sanguínea , Músculo Esquelético/química , Coativador 1-alfa do Receptor gama Ativado por Proliferador de Peroxissomo , Proteínas Proto-Oncogênicas c-akt/metabolismo , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Proteínas Ligases SKP Culina F-Box/genética , Natação , Proteínas com Motivo Tripartido , Ubiquitina-Proteína Ligases/genética , Fator A de Crescimento do Endotélio Vascular
11.
Physiol Genomics ; 46(7): 245-55, 2014 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-24495916

RESUMO

The mechanisms of homocysteine-mediated cardiac threats are poorly understood. Homocysteine, being the precursor to S-adenosyl methionine (a methyl donor) through methionine, is indirectly involved in methylation phenomena for DNA, RNA, and protein. We reported previously that cardiac-specific deletion of N-methyl-d-aspartate receptor-1 (NMDAR1) ameliorates homocysteine-posed cardiac threats, and in this study, we aim to explore the role of NMDAR1 in epigenetic mechanisms of heart failure, using cardiomyocytes during hyperhomocysteinemia (HHcy). High homocysteine levels activate NMDAR1, which consequently leads to abnormal DNA methylation vs. histone acetylation through modulation of DNA methyltransferase 1 (DNMT1), HDAC1, miRNAs, and MMP9 in cardiomyocytes. HL-1 cardiomyocytes cultured in Claycomb media were treated with 100 µM homocysteine in a dose-dependent manner. NMDAR1 antagonist (MK801) was added in the absence and presence of homocysteine at 10 µM in a dose-dependent manner. The expression of DNMT1, histone deacetylase 1 (HDAC1), NMDAR1, microRNA (miR)-133a, and miR-499 was assessed by real-time PCR as well as Western blotting. Methylation and acetylation levels were determined by checking 5'-methylcytosine DNA methylation and chromatin immunoprecipitation. Hyperhomocysteinemic mouse models (CBS+/-) were used to confirm the results in vivo. In HHcy, the expression of NMDAR1, DNMT1, and matrix metalloproteinase 9 increased with increase in H3K9 acetylation, while HDAC1, miR-133a, and miR-499 decreased in cardiomyocytes. Similar results were obtained in heart tissue of CBS+/- mouse. High homocysteine levels instigate cardiovascular remodeling through NMDAR1, miR-133a, miR-499, and DNMT1. A decrease in HDAC1 and an increase in H3K9 acetylation and DNA methylation are suggestive of chromatin remodeling in HHcy.


Assuntos
Metilação de DNA , Epigênese Genética , Histonas , Miócitos Cardíacos/metabolismo , Acetilação , Animais , Técnicas In Vitro , Camundongos , Camundongos Endogâmicos C57BL , MicroRNAs/metabolismo , Estresse Oxidativo
12.
J Mol Neurosci ; 52(2): 202-15, 2014 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-24122186

RESUMO

Epigenetic mechanisms underlying nutrition (nutrition epigenetics) are important in understanding human health. Nutritional supplements, for example folic acid, a cofactor in one-carbon metabolism, regulate epigenetic alterations and may play an important role in the maintenance of neuronal integrity. Folic acid also ameliorates hyperhomocysteinemia, which is a consequence of elevated levels of homocysteine. Hyperhomocysteinemia induces oxidative stress that may epigenetically mediate cerebrovascular remodeling and leads to neurodegeneration; however, the mechanisms behind such alterations remain unclear. Therefore, the present study was designed to observe the protective effects of folic acid against hyperhomocysteinemia-induced epigenetic and molecular alterations leading to neurotoxic cascades. To test this hypothesis, we employed 8-weeks-old male wild-type (WT) cystathionine-beta-synthase heterozygote knockout methionine-fed (CBS+/− + Met), WT, and CBS+/− + Met mice supplemented with folic acid (FA) [WT + FA and CBS+/− + Met + FA, respectively, 0.0057-µg g−1 day−1 dose in drinking water/4 weeks]. Hyperhomocysteinemia in CBS+/− + Met mouse brain was accompanied by a decrease in methylenetetrahydrofolate reductase and an increase in S-adenosylhomocysteine hydrolase expression, symptoms of oxidative stress, upregulation of DNA methyltransferases, rise in matrix metalloproteinases, a drop in the tissue inhibitors of metalloproteinases, decreased expression of tight junction proteins, increased permeability of the blood-brain barrier, neurodegeneration, and synaptotoxicity. Supplementation of folic acid to CBS+/− + Met mouse brain led to a decrease in the homocysteine level and rescued pathogenic and epigenetic alterations, showing its protective efficacy against homocysteine-induced neurotoxicity.


Assuntos
Barreira Hematoencefálica/efeitos dos fármacos , Epigênese Genética , Ácido Fólico/uso terapêutico , Hiper-Homocisteinemia/dietoterapia , Fármacos Neuroprotetores/uso terapêutico , Adenosil-Homocisteinase/genética , Adenosil-Homocisteinase/metabolismo , Animais , Barreira Hematoencefálica/patologia , Cistationina beta-Sintase/genética , Dieta , Ácido Fólico/administração & dosagem , Ácido Fólico/farmacologia , Heterozigoto , Hiper-Homocisteinemia/genética , Hiper-Homocisteinemia/metabolismo , Metaloproteinases da Matriz/genética , Metaloproteinases da Matriz/metabolismo , Metionina/administração & dosagem , Metionina/farmacologia , Metilenotetra-Hidrofolato Redutase (NADPH2)/genética , Metilenotetra-Hidrofolato Redutase (NADPH2)/metabolismo , Metiltransferases/genética , Metiltransferases/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Fármacos Neuroprotetores/administração & dosagem , Fármacos Neuroprotetores/farmacologia , Estresse Oxidativo
13.
J Hypertens ; 31(11): 2270-81; discussion 2281, 2013 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-24077247

RESUMO

BACKGROUND: Sustained hypertension induces renovascular remodelling by altering extracellular matrix (ECM) components. Matrix metalloproteinases (MMPs) are Zn-dependent enzymes that regulate ECM turnover in concert with their inhibitors, tissue inhibitors of metalloproteinases (TIMPs). Increased MMP-2 and MMP-9 have been implicated in hypertensive complications; however, the contribution of individual MMPs/TIMPs in renal remodelling has not been fully elucidated. The purpose of this study was to determine the effect of TIMP2 deficiency and thus MMP-2 on angiotensin-II (Ang-II) induced renal remodelling. METHOD: C57BL/6J (wild-type) and TIMP2 knockout mice were infused with Ang-II at 250 ng/kg per min for 4 weeks. Blood pressure was measured weekly and end-point laser Doppler flowmetry was done to assess cortical blood flow. Immunohistochemical staining was performed for collagen and elastin analyses. The activity of MMP-9 and MMP-2 was determined by Gelatin zymography. RESULTS: Ang-II induced similar elevation in mean blood pressure in TIMP2 and wild-type mice. In TIMP2 mice, Ang-II treatment was associated with a greater reduction in renal cortical blood flow and barium angiography demonstrated decreased vascular density compared with Ang-II treated wild-type mice. Peri-glomerular and vascular collagen deposition was increased and elastin content was decreased causing increased wall-to-lumen ratio in TIMP2 mice compared with wild-type mice receiving Ang-II. Ang-II increased the expression and activity of MMP-9 predominantly in TIMP2 mice than in wild-type mice. CONCLUSION: These results suggest that TIMP2 deficiency exacerbates renovascular remodelling in agonist-induced hypertension by a mechanism that may, in part, be attributed to increased activity of MMP-9.


Assuntos
Angiotensina II/farmacologia , Hipertensão/induzido quimicamente , Rim/efeitos dos fármacos , Metaloproteinase 2 da Matriz , Inibidor Tecidual de Metaloproteinase-2/deficiência , Animais , Pressão Arterial , Pressão Sanguínea/efeitos dos fármacos , Colágeno/análise , Elastina/análise , Hipertensão/metabolismo , Hipertensão/fisiopatologia , Rim/irrigação sanguínea , Rim/química , Masculino , Metaloproteinase 9 da Matriz/metabolismo , Metaloproteinases da Matriz/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Inibidor Tecidual de Metaloproteinase-2/metabolismo , Inibidores Teciduais de Metaloproteinases/metabolismo
14.
Biochim Biophys Acta ; 1832(12): 2271-6, 2013 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-24055000

RESUMO

Cardiac muscle is unique because it contracts ceaselessly throughout the life and is highly resistant to fatigue. The marvelous nature of the cardiac muscle is attributed to its matrix that maintains structural and functional integrity and provides ambient micro-environment required for mechanical, cellular and molecular activities in the heart. Cardiac matrix dictates the endothelium myocyte (EM) coupling and contractility of cardiomyocytes. The matrix metalloproteinases (MMPs) and their tissue inhibitor of metalloproteinases (TIMPs) regulate matrix degradation that determines cardiac fibrosis and myocardial performance. We have shown that MMP-9 regulates differential expression of micro RNAs (miRNAs), calcium cycling and contractility of cardiomyocytes. The differential expression of miRNAs is associated with angiogenesis, hypertrophy and fibrosis in the heart. MMP-9, which is involved in the degradation of cardiac matrix and induction of fibrosis, is also implicated in inhibition of survival and differentiation of cardiac stem cells (CSC). Cardiac matrix is distinct because it renders mechanical properties and provides a framework essential for differentiation of cardiac progenitor cells (CPC) into specific lineage. Cardiac matrix regulates myocyte contractility by EM coupling and calcium transients and also directs miRNAs required for precise regulation of continuous and synchronized beating of cardiomyocytes that is indispensible for survival. Alteration in the matrix homeostasis due to induction of MMPs, altered expression of specific miRNAs or impaired signaling for contractility of cardiomyocytes leads to catastrophic effects. This review describes the mechanisms by which cardiac matrix regulates myocardial performance and suggests future directions for the development of treatment strategies in cardiovascular diseases.


Assuntos
Doenças Cardiovasculares/terapia , Metaloproteinases da Matriz/química , Miocárdio/patologia , Miócitos Cardíacos/patologia , Inibidores Teciduais de Metaloproteinases/antagonistas & inibidores , Animais , Doenças Cardiovasculares/metabolismo , Matriz Extracelular/metabolismo , Humanos , Metaloproteinases da Matriz/metabolismo , Inibidores Teciduais de Metaloproteinases/metabolismo
15.
PLoS One ; 7(3): e32388, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22479323

RESUMO

BACKGROUND: We have previously reported the role of anti-angiogenic factors in inducing the transition from compensatory cardiac hypertrophy to heart failure and the significance of MMP-9 and TIMP-3 in promoting this process during pressure overload hemodynamic stress. Several studies reported the evidence of cardiac autophagy, involving removal of cellular organelles like mitochondria (mitophagy), peroxisomes etc., in the pathogenesis of heart failure. However, little is known regarding the therapeutic role of mitochondrial division inhibitor (Mdivi) in the pressure overload induced heart failure. We hypothesize that treatment with mitochondrial division inhibitor (Mdivi) inhibits abnormal mitophagy in a pressure overload heart and thus ameliorates heart failure condition. MATERIALS AND METHODS: To verify this, ascending aortic banding was done in wild type mice to create pressure overload induced heart failure and then treated with Mdivi and compared with vehicle treated controls. RESULTS: Expression of MMP-2, vascular endothelial growth factor, CD31, was increased, while expression of anti angiogenic factors like endostatin and angiostatin along with MMP-9, TIMP-3 was reduced in Mdivi treated AB 8 weeks mice compared to vehicle treated controls. Expression of mitophagy markers like LC3 and p62 was decreased in Mdivi treated mice compared to controls. Cardiac functional status assessed by echocardiography showed improvement and there is also a decrease in the deposition of fibrosis in Mdivi treated mice compared to controls. CONCLUSION: Above results suggest that Mdivi inhibits the abnormal cardiac mitophagy response during sustained pressure overload stress and propose the novel therapeutic role of Mdivi in ameliorating heart failure.


Assuntos
Insuficiência Cardíaca/prevenção & controle , Coração/efeitos dos fármacos , Miocárdio/metabolismo , Quinazolinonas/farmacologia , Angiostatinas/genética , Angiostatinas/metabolismo , Animais , Aorta/patologia , Apoptose/efeitos dos fármacos , Constrição Patológica/complicações , Ecocardiografia , Complexo IV da Cadeia de Transporte de Elétrons/metabolismo , Endostatinas/genética , Endostatinas/metabolismo , Expressão Gênica/efeitos dos fármacos , Coração/fisiopatologia , Insuficiência Cardíaca/diagnóstico por imagem , Insuficiência Cardíaca/etiologia , Hipertrofia Ventricular Esquerda/diagnóstico por imagem , Hipertrofia Ventricular Esquerda/fisiopatologia , Hipertrofia Ventricular Esquerda/prevenção & controle , Imuno-Histoquímica , Metaloproteinase 2 da Matriz/genética , Metaloproteinase 2 da Matriz/metabolismo , Metaloproteinase 9 da Matriz/genética , Metaloproteinase 9 da Matriz/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Mitocôndrias Cardíacas/efeitos dos fármacos , Mitocôndrias Cardíacas/metabolismo , Miocárdio/patologia , Pressão , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Inibidor Tecidual de Metaloproteinase-3/genética , Inibidor Tecidual de Metaloproteinase-3/metabolismo , Fator A de Crescimento do Endotélio Vascular/genética , Fator A de Crescimento do Endotélio Vascular/metabolismo
16.
Am J Physiol Cell Physiol ; 303(1): C41-51, 2012 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-22517358

RESUMO

Hydrogen sulfide (H(2)S) has recently been identified as a regulator of various physiological events, including vasodilation, angiogenesis, antiapoptotic, and cellular signaling. Endogenously, H(2)S is produced as a metabolite of homocysteine (Hcy) by cystathionine ß-synthase (CBS), cystathionine γ-lyase (CSE), and 3-mercaptopyruvate sulfurtransferase (3MST). Although Hcy is recognized as vascular risk factor at an elevated level [hyperhomocysteinemia (HHcy)] and contributes to vascular injury leading to renovascular dysfunction, the exact mechanism is unclear. The goal of the current study was to investigate whether conversion of Hcy to H(2)S improves renovascular function. Ex vivo renal artery culture with CBS, CSE, and 3MST triple gene therapy generated more H(2)S in the presence of Hcy, and these arteries were more responsive to endothelial-dependent vasodilation compared with nontransfected arteries treated with high Hcy. Cross section of triple gene-delivered renal arteries immunostaining suggested increased expression of CD31 and VEGF and diminished expression of the antiangiogenic factor endostatin. In vitro endothelial cell culture demonstrated increased mitophagy during high levels of Hcy and was mitigated by triple gene delivery. Also, dephosphorylated Akt and phosphorylated FoxO3 in HHcy were reversed by H(2)S or triple gene delivery. Upregulated matrix metalloproteinases-13 and downregulated tissue inhibitor of metalloproteinase-1 in HHcy were normalized by overexpression of triple genes. Together, these results suggest that H(2)S plays a key role in renovasculopathy during HHcy and is mediated through Akt/FoxO3 pathways. We conclude that conversion of Hcy to H(2)S by CBS, CSE, or 3MST triple gene therapy improves renovascular function in HHcy.


Assuntos
Cistationina beta-Sintase/genética , Cistationina gama-Liase/genética , Terapia Genética , Sulfeto de Hidrogênio/metabolismo , Hiper-Homocisteinemia/terapia , Sulfurtransferases/genética , Animais , Células Cultivadas , Cistationina beta-Sintase/metabolismo , Cistationina gama-Liase/metabolismo , Endostatinas/biossíntese , Proteína Forkhead Box O3 , Fatores de Transcrição Forkhead/metabolismo , Homocisteína/metabolismo , Hiper-Homocisteinemia/genética , Hiper-Homocisteinemia/metabolismo , Hipertensão Renovascular/genética , Hipertensão Renovascular/terapia , Metaloproteinase 13 da Matriz/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Técnicas de Cultura de Órgãos , Molécula-1 de Adesão Celular Endotelial a Plaquetas/biossíntese , Proteínas Proto-Oncogênicas c-akt/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Artéria Renal/metabolismo , Sulfurtransferases/metabolismo , Inibidor Tecidual de Metaloproteinase-1/metabolismo , Fator A de Crescimento do Endotélio Vascular/biossíntese , Lesões do Sistema Vascular
17.
J Mol Neurosci ; 47(1): 128-38, 2012 May.
Artigo em Inglês | MEDLINE | ID: mdl-22212488

RESUMO

High levels of homocysteine (Hcy) known as hyperhomocysteinemia (HHcy), contribute to autophagy and ischemia/reperfusion injury (I/R). Previous studies have shown that I/R injury and HHcy cause increased cerebrovascular permeability; however, the associated mechanism remains obscure. Interestingly, during HHcy, cytochome-c becomes homocysteinylated (Hcy-cyto-c). Cytochrome-c (cyto-c) transports electrons and facilitates bioenergetics in the system. However, its role in autophagy during ischemia/reperfusion injury is unclear. Tetrahydrocurcumin (THC) is a major herbal antioxidant and anti-inflammatory agent. Therefore, the objective of this study was to determine whether THC ameliorates autophagy during ischemia/reperfusion injury by reducing homocysteinylation of cyto-c in hyperhomocysteinemia pathological condition. To test this hypothesis, we employed 8-10-week-old male cystathionine-beta-synthase heterozygote knockout (CBS⁺/⁻) mice (genetically hyperhomocystemic mice). Experimental group was: CBS⁺/⁻, CBS⁺/⁻ + THC (25 mg/kg in 0.1% DMSO dose); CBS ⁺/⁻/I/R, and CBS⁺/⁻/I/R + THC (25 mg/kg in 0.1% DMSO dose). Ischemia was performed for 30 min and reperfusion for 72 h. THC was injected intra-peritoneally (I.P.) once daily for a period of 3 days after 30 min of ischemia. The infarct area was measured using 2,3,5-triphenyltetrazolium chloride staining. Permeability was determined by brain edema and Evans Blue extravasation. The brain tissues were analyzed for oxidative stress, matrix metalloproteinase-9 (MMP-9), damage-regulated autophagy modulator (DRAM), and microtubule-associated protein 1 light chain 3 (LC3) by Western blot. The mRNA levels of S-adenosyl-L-homocysteine hydrolases (SAHH) and methylenetetrahydrofolate reductase (MTHFR) genes were measured by quantitative real-time polymerase chain reaction. Co-immunoprecipitation was used to determine the homocysteinylation of cyto-c. We found that brain edema and Evans Blue leakage were reduced in I/R + THC-treated groups as compared to sham-operated groups along with reduced brain infarct size. THC also decreased oxidative damage and ameliorated the homocysteinylation of cyto-c in-part by MMP-9 activation which leads to autophagy in I/R groups as compared to sham-operated groups. This study suggests a potential therapeutic role of dietary THC in cerebral ischemia.


Assuntos
Antioxidantes/farmacologia , Autofagia/efeitos dos fármacos , Isquemia Encefálica/tratamento farmacológico , Curcumina/análogos & derivados , Citocromos c/fisiologia , Hiper-Homocisteinemia/tratamento farmacológico , Animais , Autofagia/fisiologia , Isquemia Encefálica/enzimologia , Isquemia Encefálica/patologia , Curcumina/farmacologia , Citocromos c/metabolismo , Hiper-Homocisteinemia/enzimologia , Hiper-Homocisteinemia/patologia , Masculino , Camundongos , Camundongos Knockout , Traumatismo por Reperfusão/tratamento farmacológico , Traumatismo por Reperfusão/enzimologia , Traumatismo por Reperfusão/patologia
18.
J Cereb Blood Flow Metab ; 32(1): 150-63, 2012 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-21989482

RESUMO

Elevated blood level of Fibrinogen (Fg) is commonly associated with vascular dysfunction. We tested the hypothesis that at pathologically high levels, Fg increases cerebrovascular permeability by activating matrix metalloproteinases (MMPs). Fibrinogen (4 mg/mL blood concentration) or equal volume of phosphate-buffered saline (PBS) was infused into male wild-type (WT; C57BL/6J) or MMP-9 gene knockout (MMP9-/-) mice. Pial venular leakage of fluorescein isothiocyanate-bovine serum albumin to Fg or PBS alone and to topically applied histamine (10(-5) mol/L) were assessed. Intravital fluorescence microscopy and image analysis were used to assess cerebrovascular protein leakage. Pial venular macromolecular leakage increased more after Fg infusion than after infusion of PBS in both (WT and MMP9-/-) mice but was more pronounced in WT compared with MMP9-/- mice. Expression of vascular endothelial cadherin (VE-cadherin) was less and plasmalemmal vesicle-associated protein-1 (PV-1) was greater in Fg-infused than in PBS-infused both mice groups. However, in MMP9-/- mice, VE-cadherin expression was greater and PV-1 expression was less than in WT mice. These data indicate that at higher levels, Fg compromises microvascular integrity through activation of MMP-9 and downregulation of VE-cadherin and upregulation of PV-1. Our results suggest that elevated blood level of Fg could have a significant role in cerebrovascular dysfunction and remodeling.


Assuntos
Permeabilidade Capilar/fisiologia , Veias Cerebrais/metabolismo , Fibrinogênio/farmacologia , Metaloproteinase 9 da Matriz/fisiologia , Animais , Antígenos CD/biossíntese , Caderinas/biossíntese , Permeabilidade Capilar/efeitos dos fármacos , Proteínas de Transporte/biossíntese , Veias Cerebrais/efeitos dos fármacos , Regulação para Baixo , Endotélio Vascular/efeitos dos fármacos , Endotélio Vascular/metabolismo , Fibrinogênio/fisiologia , Fluoresceína-5-Isotiocianato/análogos & derivados , Fluoresceína-5-Isotiocianato/farmacocinética , Infusões Intra-Arteriais , Masculino , Metaloproteinase 9 da Matriz/genética , Proteínas de Membrana/biossíntese , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Microscopia de Fluorescência , Soroalbumina Bovina/farmacocinética , Regulação para Cima , Vênulas/efeitos dos fármacos , Vênulas/metabolismo
19.
Arch Physiol Biochem ; 117(5): 270-82, 2011 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-21838575

RESUMO

In the present study we tested the hypothesis whether hyperhomocysteinemia, an elevated homocysteine level, induces venous phenotype in artery. To test our hypothesis, we employed wild type (WT) and cystathionine ß-synthase heterozygous (+/-) (CBS+/-) mice treatment with or without folic acid (FA). Aortic blood flow and velocity were significantly lower in CBS+/-mice compared to WT. Aortic lumen diameter was significantly decreased in CBS+/-mice, whereas FA treatment normalized it. Medial thickness and collagen were significantly increased in CBS+/-aorta, whereas elastin/collagen ratio was significantly decreased. Superoxide and gelatinase activity was significantly high in CBS+/-aorta vs WT. Western blot showed significant increase in MMP-2, -9,-12, TIMP-2 and decrease in TIMP-4 in aorta. RT-PCR revealed significant increase of vena cava marker EphB4, MMP-13 and TIMP-3 in aorta. We summarize that chronic HHcy causes vascular remodelling that transduces changes in vascular wall in a way that artery expresses vein phenotype.


Assuntos
Aorta/metabolismo , Cistationina beta-Sintase/deficiência , Ácido Fólico/administração & dosagem , Expressão Gênica/efeitos dos fármacos , Hiper-Homocisteinemia/metabolismo , Veias/metabolismo , Animais , Aorta/patologia , Western Blotting , Doença Crônica , Colágeno/genética , Colágeno/metabolismo , Cistationina beta-Sintase/genética , Elastina/genética , Elastina/metabolismo , Efrina-B2/genética , Efrina-B2/metabolismo , Ácido Fólico/uso terapêutico , Genótipo , Hemorreologia/efeitos dos fármacos , Hiper-Homocisteinemia/genética , Hiper-Homocisteinemia/patologia , Hiper-Homocisteinemia/fisiopatologia , Masculino , Metaloproteinases da Matriz/genética , Metaloproteinases da Matriz/metabolismo , Camundongos , Camundongos Transgênicos , Fenótipo , Receptor EphB4/genética , Receptor EphB4/metabolismo , Inibidor Tecidual de Metaloproteinase-2/genética , Inibidor Tecidual de Metaloproteinase-2/metabolismo , Inibidores Teciduais de Metaloproteinases/genética , Inibidores Teciduais de Metaloproteinases/metabolismo , Túnica Íntima/anatomia & histologia , Veias/patologia , Inibidor Tecidual 4 de Metaloproteinase
20.
J Immunol ; 187(5): 2723-31, 2011 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-21810612

RESUMO

Duchenne muscular dystrophy (DMD), caused by mutations in the dystrophin gene, is a common and lethal form of muscular dystrophy. With progressive disease, most patients succumb to death from respiratory or heart failure, or both. However, the mechanisms, especially those governing cardiac inflammation and fibrosis in DMD, remain less understood. Matrix metalloproteinase (MMPs) are a group of extracellular matrix proteases involved in tissue remodeling in both physiologic and pathophysiologic conditions. Previous studies have shown that MMP-9 exacerbates myopathy in dystrophin-deficient mdx mice. However, the role and the mechanisms of action of MMP-9 in cardiac tissue and the biochemical mechanisms leading to increased levels of MMP-9 in mdx mice remain unknown. Our results demonstrate that the levels of MMP-9 are increased in the heart of mdx mice. Genetic ablation of MMP-9 attenuated cardiac injury, left ventricle dilation, and fibrosis in 1-y-old mdx mice. Echocardiography measurements showed improved heart function in Mmp9-deficient mdx mice. Deletion of the Mmp9 gene diminished the activation of ERK1/2 and Akt kinase in the heart of mdx mice. Ablation of MMP-9 also suppressed the expression of MMP-3 and MMP-12 in the heart of mdx mice. Finally, our experiments have revealed that osteopontin, an important immunomodulator, contributes to the increased amounts of MMP-9 in cardiac and skeletal muscle of mdx mice. This study provides a novel mechanism for development of cardiac dysfunction and suggests that MMP-9 and OPN are important therapeutic targets to mitigating cardiac abnormalities in patients with DMD.


Assuntos
Cardiomiopatias/metabolismo , Metaloproteinase 9 da Matriz/metabolismo , Distrofia Muscular de Duchenne/metabolismo , Osteopontina/metabolismo , Animais , Western Blotting , Cardiomiopatias/genética , Cardiomiopatias/patologia , Imuno-Histoquímica , Camundongos , Camundongos Endogâmicos mdx , Camundongos Knockout , Distrofia Muscular de Duchenne/complicações , Distrofia Muscular de Duchenne/patologia , Reação em Cadeia da Polimerase Via Transcriptase Reversa
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...