Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 60
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.
Am J Physiol Endocrinol Metab ; 320(3): E399-E414, 2021 03 01.
Article in English | MEDLINE | ID: mdl-33308014

ABSTRACT

The ability to ward off pathogens with minimal damage to the host determines the immune system's robustness. Multiple factors, including pathogen processing, identification, secretion of mediator and effector molecules, and immune cell proliferation and differentiation into various subsets, constitute the success of mounting an effective immune response. Cellular metabolism controls all of these intricate processes. Cells utilize diverse fuel sources and switch back and forth between different metabolic pathways depending on their energy needs. The three most critical metabolic pathways on which immune cells depend to meet their energy needs are oxidative metabolism, glycolysis, and glutaminolysis. Dynamic switching between these metabolic pathways is needed for optimal function of the immune cells. Moreover, switching between these metabolic pathways needs to be tightly regulated to achieve the best results. Immune cells depend on the Warburg effect for their growth, proliferation, secretory, and effector functions. Here, we hypothesize that the sirtuin, SIRT6, could be a negative regulator of the Warburg effect. We also postulate that SIRT6 could act as a master regulator of immune cell metabolism and function by regulating critical signaling pathways.


Subject(s)
Immune System/physiology , Sirtuins/physiology , Animals , Cell Nucleus/metabolism , Energy Metabolism/physiology , Humans , Metabolic Networks and Pathways/physiology , Sirtuins/metabolism
4.
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
5.
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
6.
Elife ; 72018 03 05.
Article in English | MEDLINE | ID: mdl-29504933

ABSTRACT

Glycogen synthase kinase 3 (GSK3) is a critical regulator of diverse cellular functions involved in the maintenance of structure and function. Enzymatic activity of GSK3 is inhibited by N-terminal serine phosphorylation. However, alternate post-translational mechanism(s) responsible for GSK3 inactivation are not characterized. Here, we report that GSK3α and GSK3ß are acetylated at Lys246 and Lys183, respectively. Molecular modeling and/or molecular dynamics simulations indicate that acetylation of GSK3 isoforms would hinder both the adenosine binding and prevent stable interactions of the negatively charged phosphates. We found that SIRT2 deacetylates GSK3ß, and thus enhances its binding to ATP. Interestingly, the reduced activity of GSK3ß is associated with lysine acetylation, but not with phosphorylation at Ser9 in hearts of SIRT2-deficient mice. Moreover, GSK3 is required for the anti-hypertrophic function of SIRT2 in cardiomyocytes. Overall, our study identified lysine acetylation as a novel post-translational modification regulating GSK3 activity.


Subject(s)
Glycogen Synthase Kinase 3/metabolism , Protein Isoforms/metabolism , Protein Processing, Post-Translational , Sirtuin 2/metabolism , Animals , Cell Line , Glycogen Synthase Kinase 3/chemistry , Humans , Mice , Mice, Knockout , Models, Molecular , Molecular Dynamics Simulation , Phosphorylation
7.
Cell Death Differ ; 25(9): 1638-1656, 2018 09.
Article in English | MEDLINE | ID: mdl-29449643

ABSTRACT

c-Jun NH2-terminal kinases (JNKs) are responsive to stress stimuli and their activation regulate key cellular functions, including cell survival, growth, differentiation and aging. Previous studies demonstrate that activation of JNK requires dual phosphorylation by the mitogen-activated protein kinase kinases. However, other post-translational mechanisms involved in regulating the activity of JNK have been poorly understood. In this work, we studied the functional significance of reversible lysine acetylation in regulating the kinase activity of JNK. We found that the acetyl transferase p300 binds to, acetylates and inhibits kinase activity of JNK. Using tandem mass spectrometry, molecular modelling and molecular dynamics simulations, we found that acetylation of JNK at Lys153 would hinder the stable interactions of the negatively charged phosphates and prevent the adenosine binding to JNK. Our screening for the deacetylases found SIRT2 as a deacetylase for JNK. Mechanistically, SIRT2-dependent deacetylation enhances ATP binding and enzymatic activity of JNK towards c-Jun. Furthermore, SIRT2-mediated deacetylation favours the phosphorylation of JNK by MKK4, an upstream kinase. Our results indicate that deacetylation of JNK by SIRT2 promotes oxidative stress-induced cell death. Conversely, SIRT2 inhibition attenuates H2O2-mediated cell death in HeLa cells. SIRT2-deficient (SIRT2-KO) mice exhibit increased acetylation of JNK, which is associated with markedly reduced catalytic activity of JNK in the liver. Interestingly, SIRT2-KO mice were resistant to acetaminophen-induced liver toxicity. SIRT2-KO mice show lower cell death, minimal degenerative changes, improved liver function and survival following acetaminophen treatment. Overall, our work identifies SIRT2-mediated deacetylation of JNK as a critical regulator of cell survival during oxidative stress.


Subject(s)
Apoptosis , Mitogen-Activated Protein Kinase 8/metabolism , Oxidative Stress , Sirtuin 2/metabolism , Acetaminophen/toxicity , Acetylation/drug effects , Adenosine Triphosphate/metabolism , Animals , Apoptosis/drug effects , Chemical and Drug Induced Liver Injury/etiology , Chemical and Drug Induced Liver Injury/mortality , Crystallography, X-Ray , E1A-Associated p300 Protein/metabolism , Hydrogen Peroxide/toxicity , MAP Kinase Kinase 4/metabolism , Mice , Mice, Knockout , Mitogen-Activated Protein Kinase 8/genetics , Molecular Dynamics Simulation , Mutagenesis, Site-Directed , Oxidative Stress/drug effects , Phosphorylation , Protein Binding , Protein Structure, Tertiary , Sirtuin 2/deficiency , Sirtuin 2/genetics
8.
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
9.
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
12.
Biochim Biophys Acta Mol Cell Res ; 1864(7): 1260-1273, 2017 Jul.
Article in English | MEDLINE | ID: mdl-28342806

ABSTRACT

Mitochondrial homeostasis is critical for keeping functional heart in response to metabolic or environmental stresses. Mitochondrial fission and fusion (mitochondrial dynamics) play essential roles in maintaining mitochondrial homeostasis, defects in mitochondrial dynamics lead to cardiac diseases such as ischemia-reperfusion injury (IRI), heart failure and diabetic cardiomyopathy. Mitochondrial dynamics is determined by mitochondrial fission and fusion proteins, including OPA1, mitofusins and Drp1. These proteins are tightly regulated by a series of signaling pathways through different aspects such as transcription, post translation modifications (PTMs) and proteasome-dependent protein degradation. By modulating these mitochondrial fission and fusion proteins, mitochondria fine-tune their metabolic status to meet the energy demands of the heart. Moreover, these mitochondrial fission and fusion proteins are essential for mediating mitochondrial autophagy (mitophagy), leading to clearance of damaged mitochondria to maintain a healthy population of mitochondria in heart under stressed conditions. Mitochondrial dynamics dependent improvement in mitochondrial metabolism and quality could partially reverse the pathological conditions of heart. This review describes an overview of mechanisms on mitochondrial dynamics regulation and provides potential therapeutic targets for treating cardiovascular diseases.


Subject(s)
Heart Diseases/metabolism , Mitochondrial Dynamics , Animals , Dynamins/genetics , Dynamins/metabolism , GTP Phosphohydrolases/genetics , GTP Phosphohydrolases/metabolism , Heart Diseases/genetics , Humans , Mitochondrial Proteins/genetics , Mitochondrial Proteins/metabolism
13.
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
14.
Oncotarget ; 7(43): 69321-69336, 2016 Oct 25.
Article in English | MEDLINE | ID: mdl-27732568

ABSTRACT

Constitutive fibroblast activation is responsible for organ fibrosis in fibrotic disorders including systemic sclerosis (SSc), but the underlying mechanisms are not fully understood, and effective therapies are lacking. We investigated the expression of the mitochondrial deacetylase sirtuin 3 (SIRT3) and its modulation by hexafluoro, a novel fluorinated synthetic honokiol analogue, in the context of fibrosis. We find that augmenting cellular SIRT3 by forced expression in normal lung and skin fibroblasts, or by hexafluoro treatment, blocked intracellular TGF-ß signaling and fibrotic responses, and mitigated the activated phenotype of SSc fibroblasts. Moreover, hexafluoro attenuated mitochondrial and cytosolic reactive oxygen species (ROS) accumulation in TGF-ß-treated fibroblasts. Remarkably, we found that the expression of SIRT3 was significantly reduced in SSc skin biopsies and explanted fibroblasts, and was suppressed by TGF-ß treatment in normal fibroblasts. Moreover, tissue levels of acetylated MnSOD, a sensitive marker of reduced SIRT3 activity, were dramatically enhanced in lesional skin and lung biopsies from SSc patients. Mice treated with hexafluoro showed substantial attenuation of bleomycin-induced fibrosis in the lung and skin. Our findings reveal a cell-autonomous function for SIRT3 in modulating fibrotic responses, and demonstrate the ability of a novel pharmacological SIRT3 agonist to attenuate fibrosis in vitro and in vivo. In light of the impaired expression and activity of SIRT3 associated with organ fibrosis in SSc, pharmacological approaches for augmenting SIRT3 might have therapeutic potential.


Subject(s)
Lung/enzymology , Scleroderma, Systemic/enzymology , Sirtuin 3/metabolism , Skin/enzymology , Adult , Aged , Animals , Bleomycin , Cells, Cultured , Enzyme Activation/drug effects , Female , Fibroblasts/drug effects , Fibroblasts/enzymology , Fibrosis/chemically induced , Fibrosis/prevention & control , Humans , Hydrocarbons, Fluorinated/pharmacology , Lung/pathology , Male , Mice, Inbred C57BL , Middle Aged , RNA Interference , Scleroderma, Systemic/genetics , Scleroderma, Systemic/pathology , Sirtuin 3/genetics , Skin/pathology
15.
Trends Endocrinol Metab ; 27(8): 563-573, 2016 08.
Article in English | MEDLINE | ID: mdl-27210897

ABSTRACT

Cardiovascular diseases (CVDs) are expanding at an alarming rate and people's propensity to develop them increases with age. Growing evidence indicates that sirtuins play a pivotal role in regulating a multitude of age-related diseases. Sirtuins are versatile molecules conserved from archaea to mammals. They are regulated by various metabolic and environmental stimuli. Seven sirtuin homologs (SIRT1-7) are present in mammals, with diverse cellular locations. Recent studies have delineated roles of sirtuins in regulating cardiac pathophysiological conditions under various stressors. SIRT1 is the most extensively studied sirtuin, while the role of other sirtuins in maintaining cardiac growth and function is still emerging. In this review we discuss the present understanding of the role of sirtuins in regulating pathophysiological conditions of the heart.


Subject(s)
Heart/physiopathology , Sirtuins/metabolism , Animals , Cardiovascular Diseases/metabolism , Heart/growth & development , Humans , Sirtuin 1/metabolism
16.
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
17.
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
18.
J Thorac Cardiovasc Surg ; 150(5): 1280-90.e2, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26344683

ABSTRACT

OBJECTIVE: In an effort to expand treatment for advanced heart failure, we sought to develop a tissue-engineered cardiac patch for constructive and functional in situ myocardial regeneration. METHODS: An extracellular matrix patch derived from porcine small intestine submucosa was incorporated with a controlled release of basic fibroblast growth factor. The patch was surgically implanted into the porcine right ventricle (group B, n = 5). Untreated extracellular matrix (group U) and Dacron (group D) patches served as control (n = 5/group). Cardiovascular magnetic resonance was performed in all 3 groups 60 days postsurgery to evaluate regional contractility with peak longitudinal strain, perfusion with relative maximum upslope, and extent of fibrosis/edema with extracellular volume fraction. Electrophysiologic-anatomic mapping was performed in group B. Histology and quantitative reverse transcription-polymerase chain reaction were performed for further tissue characterization. RESULTS: Cardiovascular magnetic resonance-derived parameters were significantly better in group B compared with groups U and D (strain: group B = -16.6% ± 1.8%, group U = -14.7% ± 1.2%, group D = -9.0% ± 1.5%, P < .001; upslope: group B = 13.7% ± 1.1%, group U = 10.8% ± 1.3%, group D = 6.4% ± 1.8%, P < .001; extracellular volume: group B = 45% ± 7%, group U = 54% ± 10%, group D = 70% ± 10%, P = .003). Histology in group B showed a homogenous distribution of host cells, including tropomyosin and α-sarcomeric actinin-positive maturing cardiomyocytes. Group B demonstrated the greatest degree of vasculogenesis as determined by capillary density analysis (group B = 19.5 ± 6.2/mm(3), group U = 12.7 ± 2.5/mm(3), group D = 6.9 ± 3.7/mm(3), P < .001). Quantitative reverse transcription-polymerase chain reaction supported the histologic findings. Electrophysiologic-anatomic mapping in group B indicated positive electrical conductivity in the patch area. CONCLUSIONS: The extracellular matrix patch enhanced with controlled release of fibroblast growth factor facilitated in situ constructive repopulation of the host cells, including cardiomyocyte and functional regeneration, increased regional contractility and tissue perfusion, and positive electrical activity in a porcine preparation.


Subject(s)
Cardiac Surgical Procedures , Drug Carriers , Extracellular Matrix/transplantation , Fibroblast Growth Factor 2/administration & dosage , Heart Ventricles/drug effects , Heart Ventricles/surgery , Intestine, Small/transplantation , Regeneration/drug effects , Tissue Engineering/methods , Ventricular Remodeling/drug effects , Action Potentials , Animals , Delayed-Action Preparations , Electrophysiologic Techniques, Cardiac , Female , Gene Expression Regulation , Heart Ventricles/metabolism , Heart Ventricles/pathology , Heart Ventricles/physiopathology , Magnetic Resonance Imaging , Models, Animal , Myocardial Contraction/drug effects , Sus scrofa , Time Factors , Ventricular Function, Right/drug effects
19.
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
20.
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
SELECTION OF CITATIONS
SEARCH DETAIL
...