Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 26
Filter
Add more filters










Publication year range
1.
JCSM Rapid Commun ; 4(1): 40-56, 2021.
Article in English | MEDLINE | ID: mdl-34212132

ABSTRACT

BACKGROUND: During cancer cachexia, cytokines released from tumour cells can alter body's metabolism, which can lead to onset of this disease process. Biological basis of cachexia is multifactorial; hence, it is important to identify and modulate multiple targets to curtail the process of cachexia. Previously, we reported that the nuclear sirtuin, SIRT6, blocks expression of myostatin, a negative regulator of muscle growth, through modulation of the NF-κB signalling. This study was undertaken to test whether muscle-specific over-expression of SIRT6 can block the cancer-associated muscle wasting in vivo and to identify additional relevant targets of SIRT6, which can explain its ability to maintain muscle health. METHODS: We generated a skeletal muscle-specific SIRT6 over-expressing transgenic mouse line (Sk.T6Tg) expressing SIRT6 at a moderate (two-fold to four-fold) level, compared with its control littermates. To generate a cancer-cachexia model, B16F10 mouse melanoma cells were injected subcutaneously in the flanks of mice. Gastrocnemius muscle tissues from non-tumour and tumour controls and Sk.T6Tg mice (n = 5-20) were analysed by histology, immunoblotting, and RT-qPCR. Plasma samples of mice were evaluated using cytokine arrays and ELISA in both non-tumour and tumour conditions. RESULTS: Our results demonstrate dual benefits of muscle-specific moderate over-expression of SIRT6 in a mouse model of cancer-cachexia. In tumour-bearing mice, SIRT6 over-expression preserved muscle weight (P < 0.001) and fibre size (P < 0.005) as well as suppressed tumour growth (P < 0.05). SIRT6 over-expression significantly reduced myostatin expression and plasma free fatty acids levels but maintained plasma insulin levels in tumour-bearing mice. These positive effects of SIRT6 were associated with downregulation of the circulatory chemokine, CXCL10, and the myokine, WNT4. SIRT6 also upregulated expression of GLUT4, the major glucose transporter in the skeletal muscle. These results for the first time demonstrate that SIRT6 regulates multiple targets to limit tumour growth and cancer-associated muscle atrophy. CONCLUSION: Given the multifactorial nature of cachexia, SIRT6, which concurrently controls multiple pathways, can be a valuable therapeutic target to overcome this debilitating syndrome.

2.
Aging (Albany NY) ; 13(9): 12334-12358, 2021 05 02.
Article in English | MEDLINE | ID: mdl-33934090

ABSTRACT

Sirtuins have been shown to regulate the aging process. We have previously demonstrated that Sirt6 blocks the pressure overload-induced cardiac hypertrophy in mice. Here, we show that Sirt6 can also mitigate aging-induced cardiomyocyte senescence and cardiac hypertrophy. We found that aging is associated with altered Sirt6 activity along with development of cardiac hypertrophy and fibrosis. Compared to young mice (4-months), the hearts of aged mice (24-months) showed increased levels of mitochondrial DNA damage, shortened telomere length, and increased accumulation of 8-oxo-dG adducts, which are hallmarks of aging. The aged hearts also showed reduced levels of NAD+ and altered levels of mitochondrial fusion-fission proteins. Similar characteristics were observed in the hearts of Sirt6 deficient mice. Additionally, we found that doxorubicin (Dox) induced cardiomyocyte senescence, as measured by expression of p16INK4a, p53, and ß-galactosidase, was associated with loss of Sirt6. However, Sirt6 overexpression protected cardiomyocytes from developing Dox-induced senescence. Further, compared to wild-type mice, the hearts of Sirt6.Tg mice showed reduced expression of aging markers, and the development of aging-associated cardiac hypertrophy and fibrosis. Our data suggest that Sirt6 is a critical anti-aging molecule that regulates various cellular processes associated with aging and protects the heart from developing aging-induced cardiac hypertrophy and fibrosis.


Subject(s)
Aging/physiology , Cardiomegaly/metabolism , Myocytes, Cardiac/metabolism , Sirtuins/metabolism , Animals , Cardiomegaly/drug therapy , DNA Damage/drug effects , DNA Damage/physiology , Mice , Myocytes, Cardiac/drug effects , Protective Agents/pharmacology , Sirtuins/genetics , Telomere Shortening
3.
FASEB J ; 33(10): 10872-10888, 2019 10.
Article in English | MEDLINE | ID: mdl-31318577

ABSTRACT

Sirtuins (Sirts) are implicated in regulating a myriad of biologic functions ranging from cell growth and metabolism to longevity. Here, we show that nuclear Sirt, Sirt6, and mitochondrial Sirt, Sirt3, regulate each other's activity and protect the heart from developing diabetic cardiomyopathy. We found that expression of both Sirt6 and Sirt3 was reduced in cardiomyocytes treated with palmitate and in hearts of mice fed with a high-fat, high-sucrose (HF-HS) diet to develop obesity and diabetes. Conversely, whole-body overexpressing Sirt6 transgenic (Tg.Sirt6) mice were protected from developing obesity and insulin resistance when fed with the same HF-HS diet. The hearts of Tg.Sirt6 mice were also protected from mitochondrial fragmentation and decline of Sirt3, resulting otherwise from HF-HS diet feeding. Mechanistic studies showed that Sirt3 preserves Sirt6 levels by reducing oxidative stress, whereas Sirt6 maintains Sirt3 levels by up-regulating nuclear respiratory factor 2 (Nrf2)-dependent Sirt3 gene transcription. We found that Sirt6 regulates Nrf2-mediated cardiac gene expression in 2 ways; first, Sirt6 suppresses expression of Kelch-like ECH-associated protein 1 (Keap1), a negative regulator of Nrf2, and second, Sirt6 binds to Nrf2 and antagonizes its interaction with Keap1, thereby stabilizing Nrf2 levels in cardiomyocytes. Together, these studies demonstrate that Sirt6 and Sirt3 maintain each other's activity and protect the heart from developing diabetic cardiomyopathy.-Kanwal, A., Pillai, V. B., Samant, S., Gupta, M., Gupta, M. P. The nuclear and mitochondrial sirtuins, Sirt6 and Sirt3, regulate each other's activity and protect the heart from developing obesity-mediated diabetic cardiomyopathy.


Subject(s)
Diabetic Cardiomyopathies/metabolism , Obesity/metabolism , Sirtuin 3/metabolism , Sirtuins/metabolism , Animals , Cell Line , Cell Nucleus/metabolism , Diabetic Cardiomyopathies/complications , Diet, Carbohydrate Loading/adverse effects , Diet, High-Fat/adverse effects , Female , Kelch-Like ECH-Associated Protein 1/metabolism , Mice , Mice, Inbred C57BL , Mitochondria, Heart/metabolism , Myocytes, Cardiac/metabolism , NF-E2-Related Factor 2/metabolism , Obesity/complications , Obesity/etiology , Oxidative Stress , Protein Binding , Rats , Sirtuin 3/genetics , Sirtuins/genetics
4.
Can J Physiol Pharmacol ; 97(4): 235-245, 2019 Apr.
Article in English | MEDLINE | ID: mdl-30407871

ABSTRACT

Many chronic diseases are associated with unintentional loss of body weight, which is termed "cachexia". Cachexia is a complex multifactorial syndrome associated with the underlying primary disease, and characterized by loss of skeletal muscle with or without loss of fat tissue. Patients with cachexia face dire symptoms like dyspnea, fatigue, edema, exercise intolerance, and low responsiveness to medical therapy, which worsen quality of life. Because cachexia is not a stand-alone disorder, treating primary disease - such as cancer - takes precedence for the physician, and it remains mostly a neglected illness. Existing clinical trials have demonstrated limited success mostly because of their monotherapeutic approach and late detection of the syndrome. To conquer cachexia, it is essential to identify as many molecular targets as possible using the latest technologies we have at our disposal. In this review, we have discussed different aspects of cachexia, which include various disease settings, active molecular pathways, and recent novel advances made in this field to understand consequences of this illness. We also discuss roles of the sirtuins, the NAD+-dependent lysine deacetylases, microRNAs, certain dietary options, and epigenetic drugs as potential approaches, which can be used to tackle cachexia as early as possible in its course.


Subject(s)
Cachexia/enzymology , Cachexia/pathology , Sirtuins/metabolism , Animals , Cachexia/complications , Cachexia/therapy , Humans , Muscular Atrophy/complications , Signal Transduction
5.
Sci Rep ; 7(1): 11877, 2017 09 19.
Article in English | MEDLINE | ID: mdl-28928419

ABSTRACT

Muscle wasting, also known as cachexia, is associated with many chronic diseases, which worsens prognosis of primary illness leading to enhanced mortality. Molecular basis of this metabolic syndrome is not yet completely understood. SIRT6 is a chromatin-bound member of the sirtuin family, implicated in regulating many cellular processes, ranging from metabolism, DNA repair to aging. SIRT6 knockout (SIRT6-KO) mice display loss of muscle, fat and bone density, typical characteristics of cachexia. Here we report that SIRT6 depletion in cardiac as well as skeletal muscle cells promotes myostatin (Mstn) expression. We also observed upregulation of other factors implicated in muscle atrophy, such as angiotensin-II, activin and Acvr2b, in SIRT6 depleted cells. SIRT6-KO mice showed degenerated skeletal muscle phenotype with significant fibrosis, an effect consistent with increased levels of Mstn. Additionally, we observed that in an in vivo model of cancer cachexia, Mstn expression coupled with downregulation of SIRT6. Furthermore, SIRT6 overexpression downregulated the cytokine (TNFα-IFNγ)-induced Mstn expression in C2C12 cells, and promoted myogenesis. From the ChIP assay, we found that SIRT6 controls Mstn expression by attenuating NF-κB binding to the Mstn promoter. Together, these data suggest a novel role for SIRT6 in maintaining muscle mass by controlling expression of atrophic factors like Mstn and activin.


Subject(s)
Muscle, Skeletal/metabolism , Muscular Atrophy/metabolism , Myocardium/metabolism , Myostatin/biosynthesis , Sirtuins/metabolism , Up-Regulation , Activin Receptors, Type II/genetics , Activin Receptors, Type II/metabolism , Activins/genetics , Activins/metabolism , Angiotensin II/genetics , Angiotensin II/metabolism , Animals , Humans , Mice , Mice, Knockout , Muscle, Skeletal/pathology , Muscular Atrophy/genetics , Muscular Atrophy/pathology , Myostatin/genetics , NF-kappa B/genetics , Rats , Response Elements , Sirtuins/genetics
6.
Oncotarget ; 8(21): 34082-34098, 2017 May 23.
Article in English | MEDLINE | ID: mdl-28423723

ABSTRACT

Doxorubicin is the chemotherapeutic drug of choice for a wide variety of cancers, and cardiotoxicity is one of the major side effects of doxorubicin treatment. One of the main cellular targets of doxorubicin in the heart is mitochondria. Mitochondrial sirtuin, SIRT3 has been shown to protect against doxorubicin-induced cardiotoxicity. We have recently identified honokiol (HKL) as an activator of SIRT3, which protects the heart from developing pressure overload hypertrophy. Here, we show that HKL-mediated activation of SIRT3 also protects the heart from doxorubicin-induced cardiac damage without compromising the tumor killing potential of doxorubicin. Doxorubicin-induced cardiotoxicity is associated with increased ROS production and consequent fragmentation of mitochondria and cell death. HKL-mediated activation of SIRT3 prevented Doxorubicin induced ROS production, mitochondrial damage and cell death in rat neonatal cardiomyocytes. HKL also promoted mitochondrial fusion. We also show that treatment with HKL blocked doxorubicin-induced cardiac toxicity in mice. This was associated with reduced mitochondrial DNA damage and improved mitochondrial function. Furthermore, treatments of mice, bearing prostrate tumor-xenografts, with HKL and doxorubicin showed inhibition of tumor growth with significantly reduced cardiac toxicity. Our results suggest that HKL-mediated activation of SIRT3 protects the heart from doxorubicin-induced cardiotoxicity and represents a potentially novel adjunct for chemotherapy treatments.


Subject(s)
Biphenyl Compounds/administration & dosage , Cardiomyopathies/prevention & control , Doxorubicin/adverse effects , Lignans/administration & dosage , Mitochondria, Heart/drug effects , Animals , Biphenyl Compounds/pharmacology , Cardiomyopathies/chemically induced , Cell Line, Tumor , Cells, Cultured , Disease Models, Animal , Lignans/pharmacology , Mice , Mitochondria, Heart/metabolism , Myocytes, Cardiac/cytology , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/metabolism , Rats , Reactive Oxygen Species/metabolism , Sirtuin 3 , Up-Regulation
7.
Am J Physiol Lung Cell Mol Physiol ; 312(1): L68-L78, 2017 01 01.
Article in English | MEDLINE | ID: mdl-27815257

ABSTRACT

Myofibroblast differentiation is a key process in the pathogenesis of fibrotic diseases. Transforming growth factor-ß1 (TGF-ß1) is a powerful inducer of myofibroblast differentiation and is implicated in pathogenesis of tissue fibrosis. This study was undertaken to determine the role of mitochondrial deacetylase SIRT3 in TGF-ß1-induced myofibroblast differentiation in vitro and lung fibrosis in vivo. Treatment of human lung fibroblasts with TGF-ß1 resulted in increased expression of fibrosis markers, smooth muscle α-actin (α-SMA), collagen-1, and fibronectin. TGF-ß1 treatment also caused depletion of endogenous SIRT3, which paralleled with increased production of reactive oxygen species (ROS), DNA damage, and subsequent reduction in levels of 8-oxoguanine DNA glycosylase (OGG1), an enzyme that hydrolyzes oxidized guanine (8-oxo-dG) and thus protects DNA from oxidative damage. Overexpression of SIRT3 by adenovirus-mediated transduction reversed the effects of TGF-ß1 on ROS production and mitochondrial DNA damage and inhibited TGF-ß1-induced myofibroblast differentiation. To determine the antifibrotic role of SIRT3 in vivo, we used the bleomycin-induced mouse model of pulmonary fibrosis. Compared with wild-type controls, Sirt3-knockout mice showed exacerbated fibrosis after intratracheal instillation of bleomycin. Increased lung fibrosis was associated with decreased levels of OGG1 and concomitant accumulation of 8-oxo-dG and increased mitochondrial DNA damage. In contrast, the transgenic mice with whole body Sirt3 overexpression were protected from bleomycin-induced mtDNA damage and development of lung fibrosis. These data demonstrate a critical role of SIRT3 in the control of myofibroblast differentiation and lung fibrosis.


Subject(s)
Cell Differentiation , DNA Damage , DNA, Mitochondrial/metabolism , Myofibroblasts/pathology , Pulmonary Fibrosis/pathology , Sirtuin 3/metabolism , 8-Hydroxy-2'-Deoxyguanosine , Animals , Biomarkers/metabolism , Bleomycin , Cells, Cultured , Collagen Type I/metabolism , Cytoprotection/drug effects , DNA/metabolism , DNA Glycosylases/metabolism , Deoxyguanosine/analogs & derivatives , Deoxyguanosine/metabolism , Humans , Mice, Knockout , Models, Biological , Myofibroblasts/metabolism , Pulmonary Fibrosis/metabolism , Reactive Oxygen Species/metabolism , Transforming Growth Factor beta1/pharmacology
8.
Am J Physiol Heart Circ Physiol ; 310(8): H962-72, 2016 Apr 15.
Article in English | MEDLINE | ID: mdl-26873966

ABSTRACT

Doxorubicin (Doxo) is a chemotherapeutic drug widely used to treat variety of cancers. One of the most serious side effects of Doxo is its dose-dependent and delayed toxicity to the heart. Doxo is known to induce cardiac mitochondrial damage. Recently, the mitochondrial sirtuin SIRT3 has been shown to protect mitochondria from oxidative stress. Here we show that overexpression of SIRT3 protects the heart from toxicity of Doxo by preventing the drug-induced mitochondrial DNA (mtDNA) damage. Doxo treatment caused depletion of Sirt3 levels both in primary cultures of cardiomyocytes and in mouse hearts, which led to massive acetylation of mitochondrial proteins. Doxo-induced toxicity to cardiomyocytes was associated with increased reactive oxygen species (ROS) production, mitochondrial fragmentation, and cell death. Overexpression of SIRT3 helped to attenuate Doxo-induced ROS levels and cardiomyocyte death. Sirt3 knockout (Sirt3.KO) mice could not endure the full dose of Doxo treatment, developed exacerbated cardiac hypertrophy, and died during the course of treatment, whereas Sirt3 transgenic (Sirt3.tg) mice were protected against Doxo-induced cardiotoxicity. Along with Sirt3, we also observed a concomitant decrease in levels of oxoguanine-DNA glycosylase-1 (OGG1), a major DNA glycosylase that hydrolyzes oxidized-guanine (8-oxo-dG) to guanine. Depletion of OGG1 levels was associated with increased mtDNA damage. Sirt3.KO mice and Doxo-treated mice showed increased 8-oxo-dG adducts in DNA and corresponding increase in mtDNA damage, whereas, 8-oxo-dG adducts and mtDNA damage were markedly reduced in Sirt3 overexpressing transgenic mice hearts. These results thus demonstrated that Sirt3 activation protects the heart from Doxo-induced cardiotoxicity by maintaining OGG1 levels and protecting mitochondria from DNA damage.


Subject(s)
Cardiomyopathies/prevention & control , DNA Damage , DNA, Mitochondrial/metabolism , Doxorubicin , Mitochondria, Heart/enzymology , Myocytes, Cardiac/enzymology , Sirtuin 3/metabolism , 8-Hydroxy-2'-Deoxyguanosine , Animals , Cardiomegaly/chemically induced , Cardiomegaly/enzymology , Cardiomegaly/genetics , Cardiomegaly/pathology , Cardiomyopathies/chemically induced , Cardiomyopathies/enzymology , Cardiomyopathies/genetics , Cardiomyopathies/pathology , Cell Death , Cells, Cultured , DNA Adducts/metabolism , DNA Glycosylases/metabolism , DNA, Mitochondrial/genetics , Deoxyguanosine/analogs & derivatives , Deoxyguanosine/metabolism , Disease Models, Animal , Female , Fibroblasts/enzymology , Fibroblasts/pathology , Hydrolysis , Male , Mice, Knockout , Mitochondria, Heart/pathology , Myocytes, Cardiac/pathology , Oxidative Stress , Rats, Sprague-Dawley , Reactive Oxygen Species/metabolism , Sirtuin 3/deficiency , Sirtuin 3/genetics , Sirtuins/metabolism , Time Factors
9.
Mol Cell Biol ; 36(5): 678-92, 2015 Dec 14.
Article in English | MEDLINE | ID: mdl-26667039

ABSTRACT

Tissue fibrosis is a major cause of organ dysfunction during chronic diseases and aging. A critical step in this process is transforming growth factor ß1 (TGF-ß1)-mediated transformation of fibroblasts into myofibroblasts, cells capable of synthesizing extracellular matrix. Here, we show that SIRT3 controls transformation of fibroblasts into myofibroblasts via suppressing the profibrotic TGF-ß1 signaling. We found that Sirt3 knockout (KO) mice with age develop tissue fibrosis of multiple organs, including heart, liver, kidney, and lungs but not whole-body SIRT3-overexpressing mice. SIRT3 deficiency caused induction of TGF-ß1 expression and hyperacetylation of glycogen synthase kinase 3ß (GSK3ß) at residue K15, which negatively regulated GSK3ß activity to phosphorylate the substrates Smad3 and ß-catenin. Reduced phosphorylation led to stabilization and activation of these transcription factors regulating expression of the profibrotic genes. SIRT3 deacetylated and activated GSK3ß and thereby blocked TGF-ß1 signaling and tissue fibrosis. These data reveal a new role of SIRT3 to negatively regulate aging-associated tissue fibrosis and discloses a novel phosphorylation-independent mechanism controlling the catalytic activity of GSK3ß.


Subject(s)
Aging , Fibroblasts/pathology , Glycogen Synthase Kinase 3/metabolism , Myofibroblasts/pathology , Sirtuin 3/metabolism , Acetylation , Adult , Animals , Cells, Cultured , Enzyme Activation , Fibroblasts/cytology , Fibroblasts/metabolism , Fibrosis , Glycogen Synthase Kinase 3 beta , Humans , Kidney/cytology , Kidney/metabolism , Kidney/pathology , Liver/cytology , Liver/metabolism , Liver/pathology , Mice , Mice, Knockout , Myocardium/cytology , Myocardium/metabolism , Myocardium/pathology , Myofibroblasts/cytology , Myofibroblasts/metabolism , Phosphorylation , Signal Transduction , Sirtuin 3/genetics , Smad3 Protein/metabolism , Transforming Growth Factor beta1/metabolism , beta Catenin/metabolism
10.
J Biol Chem ; 290(25): 15559-15569, 2015 Jun 19.
Article in English | MEDLINE | ID: mdl-25911107

ABSTRACT

Reversible lysine acetylation is a widespread post-translational modification controlling the activity of proteins in different subcellular compartments. We previously demonstrated that a class II histone deacetylase (HDAC), HDAC4, and a histone acetyltransferase, p300/CREB-binding protein-associated factor, associate with cardiac sarcomeres and that a class I and II HDAC inhibitor, trichostatin A, enhances contractile activity of myofilaments. In this study we show that a class I HDAC, HDAC3, is also present at cardiac sarcomeres. By immunohistochemical and electron microscopic analyses, we found that HDAC3 was localized to A-band of sarcomeres and capable of deacetylating myosin heavy chain (MHC) isoforms. The motor domains of both cardiac α- and ß-MHC isoforms were found to be reversibly acetylated. Biomechanical studies revealed that lysine acetylation significantly decreased the Km for the actin-activated ATPase activity of MHC isoforms. By in vitro motility assay, we found that lysine acetylation increased the actin-sliding velocity of α-myosin by 20% and ß-myosin by 36% compared with their respective non-acetylated isoforms. Moreover, myosin acetylation was found to be sensitive to cardiac stress. During induction of hypertrophy, myosin isoform acetylation increased progressively with duration of stress stimuli independently of isoform shift, suggesting that lysine acetylation of myosin could be an early response of myofilaments to increase contractile performance of the heart. These studies provide the first evidence for localization of HDAC3 at myofilaments and uncover a novel mechanism modulating the motor activity of cardiac MHC isoforms.


Subject(s)
Histone Deacetylases/metabolism , Myocardial Contraction , Myocardium/enzymology , Myosin Heavy Chains/metabolism , Sarcomeres/enzymology , Acetylation , Animals , Cardiomegaly/enzymology , Cardiomegaly/pathology , Cardiomegaly/physiopathology , Isoenzymes/genetics , Isoenzymes/metabolism , Male , Mice , Myocardium/pathology , Sarcomeres/pathology
11.
Nat Commun ; 6: 6656, 2015 Apr 14.
Article in English | MEDLINE | ID: mdl-25871545

ABSTRACT

Honokiol (HKL) is a natural biphenolic compound derived from the bark of magnolia trees with anti-inflammatory, anti-oxidative, anti-tumour and neuroprotective properties. Here we show that HKL blocks agonist-induced and pressure overload-mediated, cardiac hypertrophic responses, and ameliorates pre-existing cardiac hypertrophy, in mice. Our data suggest that the anti-hypertrophic effects of HKL depend on activation of the deacetylase Sirt3. We demonstrate that HKL is present in mitochondria, enhances Sirt3 expression nearly twofold and suggest that HKL may bind to Sirt3 to further increase its activity. Increased Sirt3 activity is associated with reduced acetylation of mitochondrial Sirt3 substrates, MnSOD and oligomycin-sensitivity conferring protein (OSCP). HKL-treatment increases mitochondrial rate of oxygen consumption and reduces ROS synthesis in wild type, but not in Sirt3-KO cells. Moreover, HKL-treatment blocks cardiac fibroblast proliferation and differentiation to myofibroblasts in a Sirt3-dependent manner. These results suggest that HKL is a pharmacological activator of Sirt3 capable of blocking, and even reversing, the cardiac hypertrophic response.


Subject(s)
Biphenyl Compounds/pharmacology , Cardiomegaly/prevention & control , Cardiotonic Agents/pharmacology , Lignans/pharmacology , Mitochondria/drug effects , Myocytes, Cardiac/drug effects , Sirtuin 3/metabolism , Acetylation/drug effects , Adenosine Triphosphatases/antagonists & inhibitors , Adenosine Triphosphatases/genetics , Adenosine Triphosphatases/metabolism , Animals , Cardiomegaly/chemically induced , Cardiomegaly/genetics , Cardiomegaly/pathology , Carrier Proteins/antagonists & inhibitors , Carrier Proteins/genetics , Carrier Proteins/metabolism , Cell Differentiation/drug effects , Cell Proliferation/drug effects , Enzyme Activation , Fibroblasts/drug effects , Fibroblasts/enzymology , Fibroblasts/pathology , Gene Expression Regulation , Isoproterenol , Membrane Proteins/antagonists & inhibitors , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mice , Mitochondria/enzymology , Mitochondria/pathology , Mitochondrial Proton-Translocating ATPases , Myocardium/enzymology , Myocardium/pathology , Myocytes, Cardiac/enzymology , Myocytes, Cardiac/pathology , Myofibroblasts/drug effects , Myofibroblasts/enzymology , Myofibroblasts/pathology , Phenylephrine/pharmacology , Primary Cell Culture , Reactive Oxygen Species/antagonists & inhibitors , Reactive Oxygen Species/metabolism , Signal Transduction , Sirtuin 3/genetics , Superoxide Dismutase/antagonists & inhibitors , Superoxide Dismutase/genetics , Superoxide Dismutase/metabolism
12.
Mol Cell Biol ; 34(5): 807-19, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24344202

ABSTRACT

Mitochondrial morphology is regulated by the balance between two counteracting mitochondrial processes of fusion and fission. There is significant evidence suggesting a stringent association between morphology and bioenergetics of mitochondria. Morphological alterations in mitochondria are linked to several pathological disorders, including cardiovascular diseases. The consequences of stress-induced acetylation of mitochondrial proteins on the organelle morphology remain largely unexplored. Here we report that OPA1, a mitochondrial fusion protein, was hyperacetylated in hearts under pathological stress and this posttranslational modification reduced the GTPase activity of the protein. The mitochondrial deacetylase SIRT3 was capable of deacetylating OPA1 and elevating its GTPase activity. Mass spectrometry and mutagenesis analyses indicated that in SIRT3-deficient cells OPA1 was acetylated at lysine 926 and 931 residues. Overexpression of a deacetylation-mimetic version of OPA1 recovered the mitochondrial functions of OPA1-null cells, thus demonstrating the functional significance of K926/931 acetylation in regulating OPA1 activity. Moreover, SIRT3-dependent activation of OPA1 contributed to the preservation of mitochondrial networking and protection of cardiomyocytes from doxorubicin-mediated cell death. In summary, these data indicated that SIRT3 promotes mitochondrial function not only by regulating activity of metabolic enzymes, as previously reported, but also by regulating mitochondrial dynamics by targeting OPA1.


Subject(s)
GTP Phosphohydrolases/metabolism , Mitochondria/metabolism , Mitochondria/physiology , Mitochondrial Dynamics/physiology , Sirtuin 3/metabolism , Acetylation , Animals , Cell Death/genetics , Cell Death/physiology , Cell Line, Tumor , Cells, Cultured , Fibroblasts/metabolism , Fibroblasts/physiology , GTP Phosphohydrolases/genetics , HeLa Cells , Heart/physiology , Humans , Mice , Mice, Transgenic , Mitochondria/genetics , Mitochondrial Dynamics/genetics , Mitochondrial Proteins/genetics , Mitochondrial Proteins/metabolism , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/physiology , Protein Processing, Post-Translational/genetics , Protein Processing, Post-Translational/physiology , Sirtuin 3/genetics
13.
Am J Physiol Heart Circ Physiol ; 304(3): H415-26, 2013 Feb 01.
Article in English | MEDLINE | ID: mdl-23203961

ABSTRACT

Nicotinamide phosphoribosyltransferase (Nampt) is an important coenzyme involved in cellular redox reactions. Inside the cell, Nampt (iNampt) functions as a rate-limiting enzyme in the NAD salvage pathway, and outside the cell (eNampt), it acts as a proinflammatory cytokine. High-circulating levels of Nampt are reported in different pathological conditions. This study was designed to examine the role of Nampt in the development of cardiac hypertrophy and ventricular remodeling. We studied the hypertrophic response in Nampt heterozygous (+/-) knockout and cardiac-specific overexpressing Nampt transgenic mice. Whereas Nampt(+/-) mice were protected against agonist (isoproterenol and angiotensin II)-induced hypertrophy, Nampt transgenic mice spontaneously developed cardiac hypertrophy at 6 mo of age. Experiments conducted to gain insight into the mechanism revealed that treatment of cardiomyocytes with recombinant (eNampt) or overexpression with Nampt-synthesizing adenovirus vector (Ad.Nampt) induced cardiomyocyte hypertrophy. The prohypertrophic effects of eNampt and Ad.Nampt were blocked by the addition of a Nampt-blocking antibody into cultures, thus suggesting that Nampt was in fact invoking hypertrophic response of cardiomyocytes by acting on the cell surface receptors. We also found increased Nampt levels in the supernatant of cardiomyocyte cultures subjected to stress by either serum starvation or H(2)O(2) treatment. Exploration of signaling pathways in Nampt-induced cardiac hypertrophy and fibrosis revealed increased activation of mitogen-activated protein kinases, namely, JNK1, p38, and ERK. This was also associated with increased calcineurin levels and nuclear factor of activated T-cell localization into the nucleus. From these studies we conclude that cardiomyocytes are capable of secreting Nampt during stress, and exogenous Nampt is a positive regulator of cardiac hypertrophy and adverse ventricular remodeling.


Subject(s)
Cardiomegaly/enzymology , Myocytes, Cardiac/enzymology , Nicotinamide Phosphoribosyltransferase/physiology , Ventricular Remodeling/physiology , Animals , Animals, Newborn , Cardiomegaly/diagnostic imaging , Cardiomegaly/pathology , Coloring Agents , Echocardiography , Enzyme-Linked Immunosorbent Assay , Fibroblasts/pathology , Fibroblasts/physiology , Fibrosis , Immunohistochemistry , Leucine/metabolism , Mice , Mice, Knockout , Mice, Transgenic , Mitogen-Activated Protein Kinases/physiology , Myocytes, Cardiac/pathology , NFATC Transcription Factors/physiology , Nicotinamide Phosphoribosyltransferase/metabolism , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Rats , Real-Time Polymerase Chain Reaction , Signal Transduction/physiology , Tetrazolium Salts , Thiazoles , Ventricular Remodeling/genetics
14.
Nat Med ; 18(11): 1643-50, 2012 Nov.
Article in English | MEDLINE | ID: mdl-23086477

ABSTRACT

Abnormal activation of insulin-like growth factor (IGF)-Akt signaling is implicated in the development of various diseases, including heart failure. However, the molecular mechanisms that regulate activation of this signaling pathway are not completely understood. Here we show that sirtuin 6 (SIRT6), a nuclear histone deacetylase, functions at the level of chromatin to directly attenuate IGF-Akt signaling. SIRT6-deficient mice developed cardiac hypertrophy and heart failure, whereas SIRT6 transgenic mice were protected from hypertrophic stimuli, indicating that SIRT6 acts as a negative regulator of cardiac hypertrophy. SIRT6-deficient mouse hearts showed hyperactivation of IGF signaling-related genes and their downstream targets. Mechanistically, SIRT6 binds to and suppresses the promoter of IGF signaling-related genes by interacting with c-Jun and deacetylating histone 3 at Lys9 (H3K9). We also found reduced SIRT6 expression in human failing hearts. These findings disclose a new link between SIRT6 and IGF-Akt signaling and implicate SIRT6 in the development of cardiac hypertrophy and failure.


Subject(s)
Cardiomegaly , Heart Failure , JNK Mitogen-Activated Protein Kinases , Oncogene Protein v-akt , Sirtuins , Acetylation , Animals , Cardiomegaly/genetics , Cardiomegaly/metabolism , Chromatin/genetics , Chromatin/metabolism , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Heart Failure/genetics , Heart Failure/metabolism , Histone Demethylases/metabolism , Insulin-Like Growth Factor I/genetics , Insulin-Like Growth Factor I/metabolism , Mice , Mice, Transgenic , Oncogene Protein v-akt/genetics , Oncogene Protein v-akt/metabolism , Promoter Regions, Genetic , Signal Transduction , Sirtuins/deficiency , Sirtuins/genetics , Sirtuins/metabolism
15.
Sci Signal ; 4(182): ra46, 2011 Jul 19.
Article in English | MEDLINE | ID: mdl-21775285

ABSTRACT

Signaling through the kinase Akt regulates many biological functions. Akt is activated during growth factor stimulation through a process that requires binding of Akt to phosphatidylinositol 3,4,5-trisphosphate (PIP(3)), which promotes membrane localization and phosphorylation of Akt by the upstream kinase PDK1 (phosphoinositide-dependent protein kinase 1). We show that Akt and PDK1 are acetylated at lysine residues in their pleckstrin homology domains, which mediate PIP(3) binding. Acetylation blocked binding of Akt and PDK1 to PIP(3), thereby preventing membrane localization and phosphorylation of Akt. Deacetylation by SIRT1 enhanced binding of Akt and PDK1 to PIP(3) and promoted their activation. Mice injected with cells expressing a mutant that mimicked a constitutively acetylated form of Akt developed smaller tumors than those injected with cells expressing wild-type Akt. Furthermore, impaired Akt activation in the hearts of SIRT1-deficient mice was associated with reduced cardiac hypertrophy in response to physical exercise and angiotensin II. These findings uncover a key posttranslational modification of Akt that is important for its oncogenic and hypertrophic activities.


Subject(s)
Cardiomegaly/metabolism , Cell Membrane/metabolism , Cell Transformation, Neoplastic/metabolism , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Sirtuin 1/metabolism , 3-Phosphoinositide-Dependent Protein Kinases , Acetylation , Animals , Cardiomegaly/genetics , Cardiomegaly/pathology , Cell Membrane/genetics , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/pathology , Enzyme Activation/genetics , HEK293 Cells , HeLa Cells , Humans , Mice , Mice, Knockout , Mutation , Phosphatidylinositol Phosphates/genetics , Phosphatidylinositol Phosphates/metabolism , Phosphorylation/genetics , Protein Binding/genetics , Protein Serine-Threonine Kinases/genetics , Proto-Oncogene Proteins c-akt/genetics , Sirtuin 1/genetics
16.
Mol Cell Biol ; 31(11): 2349-63, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21444723

ABSTRACT

Like phosphorylation, acetylation of lysine residues within a protein is considered a biologically relevant modification that controls the activity of target proteins. During stress of cells, massive protein acetylation takes place. Here, we show that p38 mitogen-activated protein kinase (MAPK), which controls many biological functions during stress, is reversibly acetylated by PCAF/p300 and HDAC3. We identified two acetylated lysine residues, K152 and K53, located in the substrate binding domain and in the ATP-binding pocket of p38, respectively. Acetylation of lysine 53 enhanced the activity of p38 by increasing its affinity for ATP binding. The enhanced acetylation and activation of p38 were found to be in parallel with reduced intracellular ATP levels in cardiomyocytes under stress, as well as in vivo models of cardiac hypertrophy. Thus, our data show, for the first time, that p38 activity is critically regulated by, in addition to phosphorylation, reversible acetylation of a lysine residue, which is conserved in other kinases, implying the possibility of a similar mechanism regulating their activity.


Subject(s)
Myocytes, Cardiac/metabolism , Protein Processing, Post-Translational , p38 Mitogen-Activated Protein Kinases/chemistry , p38 Mitogen-Activated Protein Kinases/metabolism , Acetylation , Acetyltransferases , Adenosine Triphosphate/metabolism , Animals , Cells, Cultured , HEK293 Cells , HeLa Cells , Histone Deacetylases/metabolism , Humans , Hypertrophy , Mass Spectrometry , Mice , Myocytes, Cardiac/pathology , Myocytes, Cardiac/physiology , Rats , Stress, Physiological , p300-CBP Transcription Factors/metabolism
17.
J Biol Chem ; 286(7): 5567-77, 2011 Feb 18.
Article in English | MEDLINE | ID: mdl-21177250

ABSTRACT

Reversible lysine acetylation is a widespread post-translational modification controlling the activity of proteins in different subcellular compartments. We previously demonstrated that a class II histone deacetylase (HDAC), HDAC4, and a histone acetyltransferase, PCAF, associate with cardiac sarcomeres, and a class I and II HDAC inhibitor, trichostatin A, enhances contractile activity of myofilaments. In this study, we show that a class I HDAC, HDAC3, is also present at cardiac sarcomeres. By immunohistochemical and electron microscopic analyses, we found that HDAC3 was localized to the A band of sarcomeres and was capable of deacetylating myosin heavy chain (MHC) isoforms. The motor domains of both cardiac α- and ß-MHC isoforms were found to be reversibly acetylated. Biomechanical studies revealed that lysine acetylation significantly decreased the K(m) for the actin-activated ATPase activity of both α- and ß-MHC isoforms. By an in vitro motility assay, we found that lysine acetylation increased the actin sliding velocity of α-myosin by 20% and ß-myosin by 36%, compared to their respective non-acetylated isoforms. Moreover, myosin acetylation was found to be sensitive to cardiac stress. During induction of hypertrophy, myosin isoform acetylation increased progressively with duration of stress stimuli, independent of isoform shift, suggesting that lysine acetylation of myosin could be an early response of myofilaments to increase contractile performance of the heart. These studies provide the first evidence for localization of HDAC3 at myofilaments and uncover a novel mechanism modulating the motor activity of cardiac MHC isoforms.


Subject(s)
Actin Cytoskeleton/enzymology , Cardiac Myosins/metabolism , Histone Deacetylases/metabolism , Myocardium/enzymology , Myosin Heavy Chains/metabolism , Acetylation , Actin Cytoskeleton/genetics , Animals , Cardiac Myosins/genetics , Histone Deacetylases/genetics , Isoenzymes/genetics , Isoenzymes/metabolism , Mice , Myosin Heavy Chains/genetics , Stress, Physiological/genetics
18.
J Biol Chem ; 285(5): 3133-44, 2010 Jan 29.
Article in English | MEDLINE | ID: mdl-19940131

ABSTRACT

Since the discovery of NAD-dependent deacetylases, sirtuins, it has been recognized that maintaining intracellular levels of NAD is crucial for the management of stress response of cells. Here we show that agonist-induced cardiac hypertrophy is associated with loss of intracellular levels of NAD, but not exercise-induced physiologic hypertrophy. Exogenous addition of NAD was capable of maintaining intracellular levels of NAD and blocking the agonist-induced cardiac hypertrophic response in vitro as well as in vivo. NAD treatment blocked the activation of pro-hypertrophic Akt1 signaling, and augmented the activity of anti-hypertrophic LKB1-AMPK signaling in the heart, which prevented subsequent induction of mTOR-mediated protein synthesis. By using gene knock-out and transgenic mouse models of SIRT3 and SIRT1, we showed that the anti-hypertrophic effects of exogenous NAD are mediated through activation of SIRT3, but not SIRT1. SIRT3 deacetylates and activates LKB1, thus augmenting the activity of the LKB1-AMPK pathway. These results reveal a novel role of NAD as an inhibitor of cardiac hypertrophic signaling, and suggest that prevention of NAD depletion may be critical in the treatment of cardiac hypertrophy and heart failure.


Subject(s)
AMP-Activated Protein Kinases/metabolism , NAD/chemistry , Protein Serine-Threonine Kinases/metabolism , Sirtuin 3/metabolism , AMP-Activated Protein Kinase Kinases , Animals , Cardiomegaly/pathology , Heart Failure , Hypertrophy , Mice , Mice, Transgenic , Protein Binding , Rats , Reactive Oxygen Species
19.
PLoS One ; 4(7): e6161, 2009 Jul 07.
Article in English | MEDLINE | ID: mdl-19582148

ABSTRACT

MicroRNAs are increasingly being recognized as regulators of embryonic development; however, relatively few microRNAs have been identified to regulate cardiac development. FOG-2 (also known as zfpm2) is a transcriptional co-factor that we have previously shown is critical for cardiac development. In this report, we demonstrate that FOG-2 expression is controlled at the translational level by microRNA-130a. We identified a conserved region in the FOG-2 3' untranslated region predicted to be a target for miR-130a. To test the functional significance of this site, we generated an expression construct containing the luciferase coding region fused with the 3' untranslated region of FOG-2 or a mutant version lacking this microRNA binding site. When these constructs were transfected into NIH 3T3 fibroblasts (which are known to express miR-130a), we observed a 3.3-fold increase in translational efficiency when the microRNA target site was disrupted. Moreover, knockdown of miR-130a in fibroblasts resulted in a 3.6-fold increase in translational efficiency. We also demonstrate that cardiomyocytes express miR-130a and can attenuate translation of mRNAs with a FOG-2 3' untranslated region. Finally, we generated transgenic mice with cardiomyocyte over-expression of miR-130a. In the hearts of these mice, FOG-2 protein levels were reduced by as much as 80%. Histological analysis of transgenic embryos revealed ventricular wall hypoplasia and ventricular septal defects, similar to that seen in FOG-2 deficient hearts. These results demonstrate the importance of miR-130a for the regulation of FOG-2 protein expression and suggest that miR-130a may also play a role in the regulation of cardiac development.


Subject(s)
DNA-Binding Proteins/genetics , Gene Expression Regulation/physiology , MicroRNAs/physiology , Myocardium/metabolism , Protein Biosynthesis , Transcription Factors/genetics , 3' Untranslated Regions , Animals , Mice , Mice, Transgenic , Myocardium/cytology , NIH 3T3 Cells
20.
Mol Cell Biol ; 29(15): 4116-29, 2009 Aug.
Article in English | MEDLINE | ID: mdl-19470756

ABSTRACT

Poly(ADP-ribose) polymerase 1 (PARP1) and SIRT1 deacetylase are two NAD-dependent enzymes which play major roles in the decision of a cell to live or to die in a stress situation. Because of the dependence of both enzymes on NAD, cross talk between them has been suggested. Here, we show that PARP1 is acetylated after stress of cardiomyocytes, resulting in the activation of PARP1, which is independent of DNA damage. SIRT1 physically binds to and deacetylates PARP1. Increased acetylation of PARP1 was also detected in hearts of SIRT1(-/-) mice, compared to that detected in the hearts of SIRT1(+/+) mice, confirming a role of SIRT1 in regulating the PARP1 acetylation in vivo. SIRT1-dependent deacetylation blocks PARP1 activity, and it protects cells from PARP1-mediated cell death. We also show that SIRT1 negatively regulates the activity of the PARP1 gene promoter, thus suggesting that the deacetylase controls the PARP1 activity at the transcriptional level as well. These data demonstrate that the activity of PARP1 is under the control of SIRT1, which is necessary for survival of cells under stress conditions.


Subject(s)
Poly(ADP-ribose) Polymerases/metabolism , Sirtuins/metabolism , Acetylation/drug effects , Angiotensin II/pharmacology , Animals , Animals, Newborn , Blotting, Western , COS Cells , Cell Survival , Cells, Cultured , Chlorocebus aethiops , HeLa Cells , Humans , Mice , Mice, Knockout , Myocardium/cytology , Myocardium/metabolism , Myocytes, Cardiac/cytology , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/metabolism , NAD/metabolism , Phenylephrine/pharmacology , Poly (ADP-Ribose) Polymerase-1 , Poly(ADP-ribose) Polymerases/genetics , Protein Binding , RNA Interference , Rats , Sirtuin 1 , Sirtuins/genetics , Stress, Mechanical
SELECTION OF CITATIONS
SEARCH DETAIL
...