Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 5 de 5
Filter
Add more filters










Database
Language
Publication year range
1.
Circulation ; 136(21): 2051-2067, 2017 Nov 21.
Article in English | MEDLINE | ID: mdl-28947430

ABSTRACT

BACKGROUND: Pathological cardiac hypertrophy induced by stresses such as aging and neurohumoral activation is an independent risk factor for heart failure and is considered a target for the treatment of heart failure. However, the mechanisms underlying pathological cardiac hypertrophy remain largely unknown. We aimed to investigate the roles of SIRT2 in aging-related and angiotensin II (Ang II)-induced pathological cardiac hypertrophy. METHODS: Male C57BL/6J wild-type and Sirt2 knockout mice were subjected to the investigation of aging-related cardiac hypertrophy. Cardiac hypertrophy was also induced by Ang II (1.3 mg/kg/d for 4 weeks) in male C57BL/6J Sirt2 knockout mice, cardiac-specific SIRT2 transgenic (SIRT2-Tg) mice, and their respective littermates (8 to ≈12 weeks old). Metformin (200 mg/kg/d) was used to treat wild-type and Sirt2 knockout mice infused with Ang II. Cardiac hypertrophy, fibrosis, and cardiac function were examined in these mice. RESULTS: SIRT2 protein expression levels were downregulated in hypertrophic hearts from mice. Sirt2 knockout markedly exaggerated cardiac hypertrophy and fibrosis and decreased cardiac ejection fraction and fractional shortening in aged (24-month-old) mice and Ang II-infused mice. Conversely, cardiac-specific SIRT2 overexpression protected the hearts against Ang II-induced cardiac hypertrophy and fibrosis and rescued cardiac function. Mechanistically, SIRT2 maintained the activity of AMP-activated protein kinase (AMPK) in aged and Ang II-induced hypertrophic hearts in vivo as well as in cardiomyocytes in vitro. We identified the liver kinase B1 (LKB1), the major upstream kinase of AMPK, as the direct target of SIRT2. SIRT2 bound to LKB1 and deacetylated it at lysine 48, which promoted the phosphorylation of LKB1 and the subsequent activation of LKB1-AMPK signaling. Remarkably, the loss of SIRT2 blunted the response of AMPK to metformin treatment in mice infused with Ang II and repressed the metformin-mediated reduction of cardiac hypertrophy and protection of cardiac function. CONCLUSIONS: SIRT2 promotes AMPK activation by deacetylating the kinase LKB1. Loss of SIRT2 reduces AMPK activation, promotes aging-related and Ang II-induced cardiac hypertrophy, and blunts metformin-mediated cardioprotective effects. These findings indicate that SIRT2 will be a potential target for therapeutic interventions in aging- and stress-induced cardiac hypertrophy.


Subject(s)
Cardiomegaly/prevention & control , Metformin/pharmacology , Myocardium/enzymology , Sirtuin 2/metabolism , AMP-Activated Protein Kinase Kinases , AMP-Activated Protein Kinases/metabolism , Acetylation , Age Factors , Aging/metabolism , Angiotensin II , Animals , Cardiomegaly/chemically induced , Cardiomegaly/enzymology , Cardiomegaly/physiopathology , Cells, Cultured , Disease Models, Animal , Fibrosis , Genetic Predisposition to Disease , Lysine , Male , Mice, Inbred C57BL , Mice, Knockout , Myocardial Contraction/drug effects , Myocardium/pathology , Phenotype , Protein Binding , Protein Serine-Threonine Kinases/metabolism , Protein Serine-Threonine Kinases/pharmacology , Rats , Signal Transduction/drug effects , Sirtuin 2/deficiency , Sirtuin 2/genetics , Stroke Volume/drug effects , Ventricular Remodeling/drug effects
2.
Sci Rep ; 6: 36576, 2016 11 07.
Article in English | MEDLINE | ID: mdl-27819261

ABSTRACT

The homeodomain transcription factor Nkx2.5/Csx is critically essential for heart specification, morphogenesis, and homeostasis. Acetylation/deacetylation is important for the localization, stability and activation of transcription factors. It remains unknown how Nkx2.5 is deacetylated and how Nkx2.5 acetylation determines its activity. In this study, we provide evidence that the NAD+-dependent class III protein deacetylase SIRT1 deacetylates Nkx2.5 in cardiomyocytes and represses the transcriptional activity of Nkx2.5. We show that SIRT1 interacts with the C-terminus of Nkx2.5 and deacetylates Nkx2.5 at lysine 182 in the homeodomain. The mutation of Nkx2.5 at lysine 182 reduces its transcriptional activity. Furthermore, SIRT1 inhibits the transcriptional activity of Nkx2.5 and represses the expression of its target genes partly by reducing Nkx2.5 binding to its co-factors, including SRF and TBX5. Taken together, these findings demonstrate that SIRT1 deacetylates Nkx2.5 and inhibits the transcriptional activity of Nkx2.5.


Subject(s)
Homeobox Protein Nkx-2.5/genetics , Homeodomain Proteins/genetics , Sirtuin 1/genetics , Transcription, Genetic/genetics , Acetylation , Animals , HEK293 Cells , Humans , Myocardium/metabolism , Myocytes, Cardiac/metabolism , Rats , Rats, Sprague-Dawley , Transcription Factors/genetics
3.
Basic Res Cardiol ; 108(4): 364, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23744058

ABSTRACT

Nkx2.5 plays protective roles in cardiac homeostasis and survival in the postnatal hearts. However, the underlying molecular mechanisms that mediate the protective functions of Nkx2.5 remain unknown. Here, we showed that Nkx2.5 was downregulated in response to various stresses and was required for protection against the stress-induced apoptosis of cardiomyocytes. SIRT1, a member of the sirtuin family of proteins, was found to be a direct transcriptional target of Nkx2.5 and was required for the Nkx2.5-mediated protection of cardiomyocytes from doxorubicin (DOX)-induced apoptosis. Moreover, using chromatin immunoprecipitation assays, we found that Nkx2.5 was able to bind to the SIRT1 promoter and that this binding was significantly decreased in DOX-treated mouse hearts. Furthermore, the cardiac-specific overexpression of SIRT1 decreased the DOX-induced apoptosis of cardiomyocytes in SIRT1 transgenic mouse hearts compared with the hearts of their wild-type littermates. These findings demonstrate that SIRT1 acts as a direct transcriptional target of Nkx2.5 that maintains cardiomyocyte homeostasis and survival.


Subject(s)
Homeodomain Proteins/physiology , Myocytes, Cardiac/physiology , Sirtuin 1/physiology , Stress, Physiological/physiology , Transcription Factors/physiology , Animals , Apoptosis/drug effects , Apoptosis/physiology , Cell Survival/physiology , Cells, Cultured , Doxorubicin/pharmacology , Homeobox Protein Nkx-2.5 , Homeostasis/physiology , Mice , Mice, Transgenic , Models, Animal , Myocytes, Cardiac/cytology , Myocytes, Cardiac/drug effects , Rats , Rats, Sprague-Dawley , Signal Transduction/physiology , Sirtuin 1/genetics , Up-Regulation/physiology
4.
Circ Res ; 109(6): 639-48, 2011 Sep 02.
Article in English | MEDLINE | ID: mdl-21778425

ABSTRACT

RATIONALE: Inactivation of the p66Shc adaptor protein confers resistance to oxidative stress and protects mice from aging-associated vascular diseases. However, there is limited information about the negative regulating mechanisms of p66Shc expression in the vascular system. OBJECTIVE: In this study, we investigated the role of SIRT1, a class III histone deacetylase, in the regulation of p66Shc expression and hyperglycemia-induced endothelial dysfunction. METHODS AND RESULTS: Expressions of p66Shc gene transcript and protein were significantly increased by different kinds of class III histone deacetylase (sirtuin) inhibitors in human umbilical vein endothelial cells and 293A cells. Adenoviral overexpression of SIRT1 inhibited high-glucose-induced p66Shc upregulation in human umbilical vein endothelial cells. Knockdown of SIRT1 increased p66Shc expression and also increased the expression levels of plasminogen activator inhibitor-1 expression, but decreased manganese superoxide dismutase expression in high-glucose conditions. However, knockdown of p66Shc significantly reversed the effects of SIRT1 knockdown. In addition, p66Shc overexpression significantly decreased manganese superoxide dismutase expression and increased plasminogen activator inhibitor-1 expression in high-glucose conditions, which were recovered by SIRT1 overexpression. Moreover, compared to streptozotocin-induced wild-type diabetic mice, endothelium-specific SIRT1 transgenic diabetic mice had decreased p66Shc expression at both the mRNA and the protein levels, improved endothelial function, and reduced accumulation of nitrotyrosine and 8-OHdG (markers of oxidative stress). We further found that SIRT1 was able to bind to the p66Shc promoter (-508 bp to -250 bp), resulting in a decrease in the acetylation of histone H3 bound to the p66Shc promoter region. CONCLUSION: Our findings indicate that repression of p66Shc expression by SIRT1 contributes to the protection of hyperglycemia-induced endothelial dysfunction.


Subject(s)
Down-Regulation/genetics , Endothelium, Vascular/metabolism , Hyperglycemia/genetics , Shc Signaling Adaptor Proteins/antagonists & inhibitors , Sirtuin 1/physiology , Aging/genetics , Animals , Diabetes Mellitus, Experimental/genetics , Diabetes Mellitus, Experimental/metabolism , Diabetes Mellitus, Experimental/pathology , Endothelium, Vascular/pathology , HEK293 Cells , Humans , Hyperglycemia/pathology , Hyperglycemia/prevention & control , Immunity, Innate/genetics , Male , Mice , Mice, Transgenic , Oxidative Stress/genetics , Protein Stability , Shc Signaling Adaptor Proteins/biosynthesis , Src Homology 2 Domain-Containing, Transforming Protein 1
5.
J Biol Chem ; 280(24): 22606-15, 2005 Jun 17.
Article in English | MEDLINE | ID: mdl-15817480

ABSTRACT

Insulin-like growth factor binding protein-3 (IGFBP-3) is a well documented growth inhibitor and pro-apoptotic factor. IGFBP-3 mRNA and its protein are overexpressed by senescent human diploid fibroblasts. However, the mechanism responsible for the up-regulation of its expression is still unclear. This report describes a novel transcriptional regulatory element, IGFBP-3 enhancer element (IEE), identified in the 5' untranslated region of the IGFBP-3 gene. This element differentially activates IGFBP-3 expression in senescent versus young fibroblasts. Electrophoretic mobility shift assays revealed abundant complexes in senescent cell nuclear extracts compared with young cell nuclear extracts. Similar to young proliferative cells, young quiescent cells showed reduced binding activity; enhancement of this activity was specific to senescent cells and not an effect of cell cycle arrest. The DNase I footprint revealed the protein-binding core sequence within the IEE through which the protein binds the IEE. Site-directed mutagenesis within IEE abolished binding activity and selectively decreased IGFBP-3 promoter activity in senescent (but not young) cells. Furthermore, introduction of an IEE decoy suppressed the endogenous IGFBP-3 gene expression specifically in senescent cells. These results point to the IEE as being a positive transcription regulatory element that contributes to the up-regulation of IGFBP-3 during cellular senescence.


Subject(s)
Gene Expression Regulation , Genes, Regulator , Insulin-Like Growth Factor Binding Protein 3/biosynthesis , Insulin-Like Growth Factor Binding Protein 3/genetics , 5' Untranslated Regions , Base Sequence , Blotting, Northern , Cell Nucleus/metabolism , Cell Proliferation , Cells, Cultured , Cellular Senescence , Cloning, Molecular , Cross-Linking Reagents/pharmacology , Deoxyribonuclease I/metabolism , Down-Regulation , Enhancer Elements, Genetic , Fibroblasts/metabolism , Gene Deletion , Genes, Reporter , Humans , Luciferases/metabolism , Models, Genetic , Molecular Sequence Data , Mutagenesis, Site-Directed , Mutation , Oligonucleotides/chemistry , Promoter Regions, Genetic , Protein Binding , RNA, Messenger/metabolism , Transfection , Ultraviolet Rays , Up-Regulation
SELECTION OF CITATIONS
SEARCH DETAIL
...