Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 109
Filter
1.
Brain Res Bull ; 178: 37-48, 2022 01.
Article in English | MEDLINE | ID: mdl-34774993

ABSTRACT

OBJECTIVE: Astrocytes mediate brain defense against oxidative stress-induced injury. Silent information regulator 1 (SIRT1) has anti-oxidative stress effects in many diseases and is highly expressed in astrocytes. However, the neuroprotective effects of SIRT1 on astrocytes after cerebral ischemia/reperfusion injury are unclear. Therein, we aim to investigate the protective effect of SIRT1 on oxidative stress injury after ischemic stroke and possible mechanisms. METHODS: We evaluated the effects of SIRT1 in astrocytes after cerebral ischemia/reperfusion injury using oxygen-glucose deprivation/recovery (OGD/R) in astrocytes in vitro and middle cerebral artery occlusion in rats in vivo. siRNA was injected intracerebroventricularly 24 h before Middle cerebral artery (MCA) occlusion (MCAO)/reperfusion(R) to silence SIRT1. RESULTS: SIRT1 knockdown reduced cell viability, increased oxidative stress, and decreased PGC-1α, PPARγ, Nrf2, heme oxygenase (HO)-1, and NAD(P)H: oxidoreductase-1 (NQO1) expression. Moreover, SIRT1 knockdown also suppressed PGC-1α activity, the PGC-1α/PPARγ interaction, and the PPARγ/PPRE interaction. Similarly, in our in vivo experiments, SIRT1 overexpression and PGC-1α or PPARγ knockdown reduced PGC-1α, PPARγ, Nrf2, HO-1, and NQO1 protein expression and blocked the PGC-1α/PPARγ interaction. SIRT1 overexpression plus PPARγ knockdown inhibited the interaction of PPARγ with PPRE. Nrf2 knockdown blocked Nrf2 expression and downstream proteins induced by SIRT1 overexpression. CONCLUSION: Overall, our data indicated that SIRT1 directly mediated the PGC-1α/PPARγ pathway in response to focal cerebral ischemia/reperfusion-induced neurological deficit, providing insights into the treatment of focal cerebral ischemia/reperfusion injury.


Subject(s)
Ischemic Stroke/metabolism , NF-E2-Related Factor 2/metabolism , Oxidative Stress/physiology , PPAR gamma/metabolism , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/metabolism , Reperfusion Injury/metabolism , Sirtuin 1/metabolism , Animals , Disease Models, Animal , Rats , Rats, Transgenic , Signal Transduction/physiology , Sirtuin 1/deficiency
2.
Eur J Pharmacol ; 910: 174498, 2021 Nov 05.
Article in English | MEDLINE | ID: mdl-34506778

ABSTRACT

BACKGROUND: Phosphodiesterase inhibitors can be used to enhance second messenger signaling to regulate intracellular Ca2+ cycling. This study investigated whether ITI-214, a selective phosphodiesterase-1 inhibitor, modulates intracellular Ca2+ regulation, resulting in a positive inotropic effect in sirtuin 1 (Sirt1)-deficient cardiomyocytes. METHODS: Mice with cardiac-specific Sirt1 knockout (Sirt1-/-) were used, with Sirt1flox/flox mice serving as controls. Electromechanical analyses of ventricular tissues were conducted, and we monitored intracellular Ca2+ using Fluo-3 as well as reactive oxygen species production in isolated cardiomyocytes. RESULTS: Sirt1-/- ventricles showed prolonged action potential duration at 90% repolarization and increased contractile force after treatment with ITI-214. The rates and sustained durations of burst firing in ventricles were higher and longer, respectively, in Sirt1-/- ventricles than in controls. ITI-214 treatment decreased the rates and shortened the durations of burst firing in Sirt1-/- mice. Sirt1-/- cardiomyocytes showed reduced Ca2+ transient amplitudes and sarcoplasmic reticulum (SR) Ca2+ stores compared to those in control cardiac myocytes, which was reversed after ITI-214 treatment. SR Ca2+ leakage was larger in Sirt1-/- cardiac myocytes than in control myocytes. ITI-214 reduced SR Ca2+ leakage in Sirt1-/- cardiac myocytes. Increased levels of reactive oxygen species in Sirt1-/- cardiomyocytes compared to those in controls were reduced after ITI-214 treatment. Levels of Ca2+ regulatory proteins, including ryanodine receptor 2, phospholamban, and sarcoplasmic/endoplasmic reticulum Ca2+ ATPase 2a were not affected by ITI-214 administration. CONCLUSIONS: Our results suggest that ITI-214 improves intracellular Ca2+ regulation, which in turn exerts inotropic effects and suppresses arrhythmic events in Sirt1-deficient ventricular myocytes.


Subject(s)
Arrhythmias, Cardiac/drug therapy , Heterocyclic Compounds, 4 or More Rings/pharmacology , Myocytes, Cardiac/drug effects , Phosphodiesterase Inhibitors/pharmacology , Sirtuin 1/deficiency , Action Potentials/drug effects , Animals , Arrhythmias, Cardiac/genetics , Arrhythmias, Cardiac/pathology , Calcium/metabolism , Calcium Signaling/drug effects , Disease Models, Animal , Heterocyclic Compounds, 4 or More Rings/therapeutic use , Humans , Male , Mice , Mice, Knockout , Myocytes, Cardiac/pathology , Phosphodiesterase Inhibitors/therapeutic use , Reactive Oxygen Species/metabolism , Sirtuin 1/genetics
3.
Oncogene ; 40(41): 6023-6033, 2021 10.
Article in English | MEDLINE | ID: mdl-34433910

ABSTRACT

Sirtuin-1 (SIRT1) is involved in various metabolic pathways, including fatty acid synthesis and gluconeogenesis in the liver. However, its role in initiation and progression of liver cancer remains unclear. Studying Sirt1 liver-specific knockout (LKO) mice in combination with diethylnitrosamine (DEN) treatment, we demonstrated that loss of Sirt1 rendered mice resistant to DEN-induced hepatocellular carcinoma (HCC) development. RNA-seq revealed that livers from LKO mice exhibited an enrichment in glutathione metabolism eight months after DEN challenge. Sirt1 deficiency elevated the expression of glutathione-s-transferase family genes by increasing the level of Nrf2, a key regulator of glutathione metabolism. Hence, LKO livers displayed a reductive environment with an increased ratio of GSH to GSSG and an elevated GSH level. Furthermore, using CRISPR knockout techniques, we confirmed that the impairment of HCC formation in LKO mice is mainly dependent on NRF2 signaling. Meanwhile, HCC induced by DEN could be blocked by the administration of N-acetyl cysteine (NAC) when administered one month after DEN challenge. However, NAC treatment starting five months after DEN injection was not able to prevent tumor development. In conclusion, our findings indicate that a reductive environment orchestrated by glutathione metabolism at an early stage can prevent the initiation of HCC.


Subject(s)
Glutathione/metabolism , Liver Neoplasms, Experimental/metabolism , Sirtuin 1/deficiency , Animals , Liver Neoplasms, Experimental/pathology , Male , Mice , Mice, Knockout , Sirtuin 1/metabolism , Up-Regulation
4.
Aging Cell ; 20(7): e13419, 2021 07.
Article in English | MEDLINE | ID: mdl-34216536

ABSTRACT

Sirtuin1 (SIRT1) and Sirtuin3 (SIRT3) protects cardiac function against ischemia/reperfusion (I/R) injury. Mitochondria are critical in response to myocardial I/R injury as disturbance of mitochondrial dynamics contributes to cardiac dysfunction. It is hypothesized that SIRT1 and SIRT3 are critical components to maintaining mitochondria homeostasis especially mitochondrial dynamics to exert cardioprotective actions under I/R stress. The results demonstrated that deficiency of SIRT1 and SIRT3 in aged (24-26 months) mice hearts led to the exacerbated cardiac dysfunction in terms of cardiac systolic dysfunction, cardiomyocytes contractile defection, and abnormal cardiomyocyte calcium flux during I/R stress. Moreover, the deletion of SIRT1 or SIRT3 in young (4-6 months) mice hearts impair cardiomyocyte contractility and shows aging-like cardiac dysfunction upon I/R stress, indicating the crucial role of SIRT1 and SIRT3 in protecting myocardial contractility from I/R injury. The biochemical and seahorse analysis showed that the deficiency of SIRT1/SIRT3 leads to the inactivation of AMPK and alterations in mitochondrial oxidative phosphorylation (OXPHOS) that causes impaired mitochondrial respiration in response to I/R stress. Furthermore, the remodeling of the mitochondria network goes together with hypoxic stress, and mitochondria undergo the processes of fusion with the increasing elongated branches during hypoxia. The transmission electron microscope data showed that cardiac SIRT1/SIRT3 deficiency in aging alters mitochondrial morphology characterized by the impairment of mitochondria fusion under I/R stress. Thus, the age-related deficiency of SIRT1/SIRT3 in the heart affects mitochondrial dynamics and respiration function that resulting in the impaired contractile function of cardiomyocytes in response to I/R.


Subject(s)
Mitochondrial Dynamics/genetics , Myocytes, Cardiac/metabolism , Sirtuin 1/deficiency , Sirtuin 3/deficiency , Aging , Animals , Humans , Mice
5.
Biochem Biophys Res Commun ; 546: 7-14, 2021 03 26.
Article in English | MEDLINE | ID: mdl-33556638

ABSTRACT

SIRT1 is involved in the regulation of a variety of biological processes such as metabolism, stress response, autophagy and differentiation. Although progenitor cells of oligodendrocytes (OPCs) express high level of SIRT1, its function on differentiation is unknown. Because we have shown that SIRT1 plays a pivotal role in differentiation of neural precursor cells, we hypothesized that SIRT1 may also participate in the differentiation of oligodendrocytes (OLGs). We examined whether SIRT1 was expressed in two human oligodendrocyte cell lines: KG-1-C and MO 3.13 OLG. Transfection of cell lines with SIRT1-siRNA and SIRT2-siRNA promoted the extension of cellular processes. SIRT1-siRNA and SIRT2-siRNA increased acetyl-α-tubulin level, conversely, over expression of SIRTs resulted in decreased the ratio of acetyl-α-tubulin to α-tubulin. We also found knockdown of SIRT1 and SIRT2 induced overexpression of ßIV-tubulin and tubulin polymerization promoting protein (TPPP) (OLG-specific cytoskeleton-related molecules) that distributed widely in cell bodies. Taken together, SIRT1 may play a role in oligodenroglial differentiation and myelinogenesis.


Subject(s)
Cell Shape , Cytoskeleton/metabolism , Gene Expression Regulation , Oligodendroglia/cytology , Oligodendroglia/metabolism , Sirtuin 1/metabolism , Acetylation , Cell Differentiation/genetics , Cell Line , Humans , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , RNA, Small Interfering/genetics , Sirtuin 1/deficiency , Sirtuin 1/genetics , Sirtuin 2/genetics , Sirtuin 2/metabolism , Tubulin/chemistry , Tubulin/genetics , Tubulin/metabolism
6.
Int J Mol Sci ; 22(3)2021 Jan 27.
Article in English | MEDLINE | ID: mdl-33513830

ABSTRACT

In advanced cirrhosis, the TNFα-mediated intestinal inflammation and bacteria dysbiosis are involved in the development of inflammation and vasoconstriction-related renal dysfunction. In colitis and acute kidney injury models, activation of SIRT1 attenuates the TNFα-mediated intestinal and renal abnormalities. This study explores the impacts of intestinal SIRT1 deficiency and TNFα-mediated intestinal abnormalities on the development of cirrhosis-related renal dysfunction. Systemic and renal hemodynamics, intestinal dysbiosis [cirrhosis dysbiosis ratio (CDR) as marker of dysbiosis], and direct renal vasoconstrictive response (renal vascular resistance (RVR) and glomerular filtration rate (GFR)) to cumulative doses of TNFα were measured in bile duct ligated (BDL)-cirrhotic ascitic mice. In SIRT1IEC-KO-BDL-ascitic mice, the worsening of intestinal dysbiosis exacerbates intestinal inflammation/barrier dysfunction, the upregulation of the expressions of intestinal/renal TNFα-related pathogenic signals, higher TNFα-induced increase in RVR, and decrease in GFR in perfused kidney. In intestinal SIRT1 knockout groups, the positive correlations were identified between intestinal SIRT1 activity and CDR. Particularly, the negative correlations were identified between CDR and RVR, with the positive correlation between CDR and GFR. In mice with advanced cirrhosis, the expression of intestinal SIRT1 is involved in the linkage between intestinal dysbiosis and vasoconstriction/hypoperfusion-related renal dysfunction through the crosstalk between intestinal/renal TNFα-related pathogenic inflammatory signals.


Subject(s)
Inflammation/metabolism , Intestinal Mucosa/metabolism , Kidney/abnormalities , Liver Cirrhosis/metabolism , Sirtuin 1/deficiency , Tumor Necrosis Factor-alpha/metabolism , Urogenital Abnormalities/metabolism , Animals , Gastrointestinal Microbiome/genetics , Glomerular Filtration Rate/genetics , Inflammation/genetics , Inflammation/pathology , Intestinal Mucosa/microbiology , Intestinal Mucosa/physiopathology , Intestines/microbiology , Intestines/physiopathology , Kidney/metabolism , Kidney/physiopathology , Liver Cirrhosis/genetics , Liver Cirrhosis/physiopathology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Sirtuin 1/genetics , Sirtuin 1/metabolism , Tumor Necrosis Factor-alpha/pharmacology , Urogenital Abnormalities/genetics , Urogenital Abnormalities/physiopathology , Vascular Resistance/genetics
7.
Cardiovasc Res ; 117(3): 836-849, 2021 02 22.
Article in English | MEDLINE | ID: mdl-32402066

ABSTRACT

AIMS: Vascular calcification is a recognized predictor of cardiovascular risk in the diabetic patient, with DNA damage and accelerated senescence linked to oxidative stress-associated pathological calcification. Having previously shown that systemic SIRT1 is reduced in diabetes, the aim was to establish whether SIRT1 is protective against a DNA damage-induced senescent and calcified phenotype in diabetic vascular smooth muscle cells (vSMCs). METHODS AND RESULTS: Immunohistochemistry revealed decreased SIRT1 and increased DNA damage marker expression in diabetic calcified arteries compared to non-diabetic and non-calcified controls, strengthened by findings that vSMCs isolated from diabetic patients show elevated DNA damage and senescence, assessed by the Comet assay and telomere length. Hyperglycaemic conditions were used and induced DNA damage and enhanced senescence in vSMCs in vitro. Using H2O2 as a model of oxidative stress-induced DNA damage, pharmacological activation of SIRT1 reduced H2O2 DNA damage-induced calcification, prevented not only DNA damage, as shown by reduced comet tail length, but also decreased yH2AX foci formation, and attenuated calcification. While Ataxia Telanglectasia Mutated (ATM) expression was reduced following DNA damage, in contrast, SIRT1 activation significantly increased ATM expression, phosphorylating both MRE11 and NBS1, thus allowing formation of the MRN complex and increasing activation of the DNA repair pathway. CONCLUSION: DNA damage-induced calcification is accelerated within a diabetic environment and can be attenuated in vitro by SIRT1 activation. This occurs through enhancement of the MRN repair complex within vSMCs and has therapeutic potential within the diabetic patient.


Subject(s)
DNA Damage , Diabetes Mellitus/enzymology , Muscle, Smooth, Vascular/enzymology , Myocytes, Smooth Muscle/enzymology , Sirtuin 1/deficiency , Vascular Calcification/enzymology , Acid Anhydride Hydrolases/metabolism , Ataxia Telangiectasia Mutated Proteins/metabolism , Calcium Chloride/toxicity , Case-Control Studies , Cell Cycle Proteins/metabolism , Cells, Cultured , Cellular Senescence , DNA Repair , DNA-Binding Proteins/metabolism , Diabetes Mellitus/genetics , Diabetes Mellitus/pathology , Disease Progression , Glucose/toxicity , Histones/metabolism , Humans , Hydrogen Peroxide/toxicity , MRE11 Homologue Protein/metabolism , Muscle, Smooth, Vascular/drug effects , Muscle, Smooth, Vascular/pathology , Myocytes, Smooth Muscle/drug effects , Myocytes, Smooth Muscle/pathology , Nuclear Proteins/metabolism , Osteogenesis , Phenotype , Phosphorylation , Popliteal Artery/drug effects , Popliteal Artery/enzymology , Popliteal Artery/pathology , Signal Transduction , Sirtuin 1/genetics , Time Factors , Vascular Calcification/genetics , Vascular Calcification/pathology
8.
Life Sci ; 265: 118787, 2021 Jan 15.
Article in English | MEDLINE | ID: mdl-33249095

ABSTRACT

AIM: The regulation of secreted osteopontin (OPN) expression by genistein and its functional sequel in the metastatic cancer cells (MDA-MB-435 and MDA-MB-231) was ascertained. MAIN METHODS: Western blot and Real-Time PCR were used to analyse the proteins and mRNA transcripts, respectively. Possible transcriptional regulation of secreted OPN was analyzed by chromatin immunoprecipitation assay, bioinformatics analysis, transfection and luciferase reporter assay. The specific siRNAs and constitutive p-ERKs were used to evaluate the role of the MAPK pathway. The functional sequel of genistein in these cells was analyzed by colony formation-, migration- and invasion- assay. KEY FINDINGS: Secreted OPN expression was inhibited (up to ~0.7-fold) by genistein in these cells. Genistein (50 µM) displayed a reduction in the aggressiveness of these cells concerning colony formation rate, migration, and invasion. The p-ERK½ was increased by ~2.5-fold and ~1.5-fold upon 50 µM genistein and 15 µM resveratrol treatments at 24 h, respectively. Knockdown of ERK½ and PD98059, the inhibitor of MEK, promoted secreted OPN expression in vitro in these cells; while, the transfection of the constitutive active ERK2 (L73P and S151D) decreased the secreted OPN expression. Further, silent mating type information regulation 2 homolog 1 (SIRT1) expression in the cells was increased (~1.6-fold) upon genistein treatment (50 µM) likewise with resveratrol (~1.5-fold), an activator for SIRT1. Knockdown of SIRT1 increased OPN mRNA transcripts expression level and secreted OPN protein level in these cells. SIGNIFICANCE: MAPK pathway and SIRT1 activation are involved in the regulation of secreted OPN by genistein in these cells.


Subject(s)
Anticarcinogenic Agents/pharmacology , Breast Neoplasms/metabolism , Genistein/pharmacology , MAP Kinase Signaling System/physiology , Osteopontin/biosynthesis , Sirtuin 1/biosynthesis , Breast Neoplasms/genetics , Cell Line, Tumor , Cell Movement/drug effects , Cell Movement/physiology , Dose-Response Relationship, Drug , Down-Regulation/drug effects , Down-Regulation/physiology , Female , Gene Expression Regulation, Neoplastic , Gene Knockdown Techniques/methods , Humans , MAP Kinase Signaling System/drug effects , Osteopontin/genetics , Sirtuin 1/deficiency , Sirtuin 1/genetics
10.
J Biol Chem ; 295(39): 13640-13650, 2020 09 25.
Article in English | MEDLINE | ID: mdl-32747445

ABSTRACT

Progranulin (PGRN) is an autocrine growth factor that exerts crucial roles within cartilage tissue; however, the molecular mechanisms underlying PGRN-mediated cartilage homeostasis remain elusive. In the present study, we investigated the role of PGRN in regulating chondrocyte homeostasis and its therapeutic potential for managing osteoarthritis (OA). We found that PGRN levels are significantly increased in human cartilage in mild OA and that its expression is decreased in the cartilage in severe OA. In vitro, treatment of primary rat chondrocytes with recombinant PGRN significantly enhanced the levels of collagen type II α 1 chain (COL2A1) and aggrecan, and attenuated TNFα-induced up-regulation of matrix metallopeptidase 13 (MMP13) and ADAM metallopeptidase with thrombospondin type 1 motif 5 (ADAMTS5) in chondrocytes. These effects were abrogated in SIRT1-/- cells, indicating a causative role of SIRT1 in the effects of PGRN on protein expression in chondrocytes. Mechanistically, PGRN increased SIRT1 expression and activity, which reduced the acetylation levels of SRY-box transcription factor (SOX9) and transcription factor P65 (P65) and thereby promoted nuclear translocation of SOX9 and inhibited TNFα-induced P65 nuclear accumulation to maintain chondrocyte homeostasis. In conclusion, our findings reveal a mechanism of action for PGRN that maintains cartilage homeostasis and supports the notion that PGRN up-regulation may be a promising strategy for managing OA.


Subject(s)
Cartilage, Articular/metabolism , Neoplasm Proteins/metabolism , Nucleocytoplasmic Transport Proteins/metabolism , Progranulins/metabolism , SOX9 Transcription Factor/metabolism , Sirtuin 1/metabolism , Acetylation , Aged , Animals , Cells, Cultured , Chondrocytes/metabolism , Humans , Rats , Rats, Sprague-Dawley , Recombinant Proteins/metabolism , Sirtuin 1/deficiency , Sirtuin 1/genetics
11.
J Am Soc Nephrol ; 31(5): 907-919, 2020 05.
Article in English | MEDLINE | ID: mdl-32276962

ABSTRACT

Growing evidence indicates that oxidative and endoplasmic reticular stress, which trigger changes in ion channels and inflammatory pathways that may undermine cellular homeostasis and survival, are critical determinants of injury in the diabetic kidney. Cells are normally able to mitigate these cellular stresses by maintaining high levels of autophagy, an intracellular lysosome-dependent degradative pathway that clears the cytoplasm of dysfunctional organelles. However, the capacity for autophagy in both podocytes and renal tubular cells is markedly impaired in type 2 diabetes, and this deficiency contributes importantly to the intensity of renal injury. The primary drivers of autophagy in states of nutrient and oxygen deprivation-sirtuin-1 (SIRT1), AMP-activated protein kinase (AMPK), and hypoxia-inducible factors (HIF-1α and HIF-2α)-can exert renoprotective effects by promoting autophagic flux and by exerting direct effects on sodium transport and inflammasome activation. Type 2 diabetes is characterized by marked suppression of SIRT1 and AMPK, leading to a diminution in autophagic flux in glomerular podocytes and renal tubules and markedly increasing their susceptibility to renal injury. Importantly, because insulin acts to depress autophagic flux, these derangements in nutrient deprivation signaling are not ameliorated by antihyperglycemic drugs that enhance insulin secretion or signaling. Metformin is an established AMPK agonist that can promote autophagy, but its effects on the course of CKD have been demonstrated only in the experimental setting. In contrast, the effects of sodium-glucose cotransporter-2 (SGLT2) inhibitors may be related primarily to enhanced SIRT1 and HIF-2α signaling; this can explain the effects of SGLT2 inhibitors to promote ketonemia and erythrocytosis and potentially underlies their actions to increase autophagy and mute inflammation in the diabetic kidney. These distinctions may contribute importantly to the consistent benefit of SGLT2 inhibitors to slow the deterioration in glomerular function and reduce the risk of ESKD in large-scale randomized clinical trials of patients with type 2 diabetes.


Subject(s)
Autophagy/physiology , Diabetes Mellitus, Type 2/complications , Diabetic Nephropathies/etiology , Nutrients/metabolism , Oxygen/metabolism , Podocytes/metabolism , Sodium-Glucose Transporter 2 Inhibitors/pharmacology , Adenylate Kinase/deficiency , Adenylate Kinase/physiology , Autophagy/drug effects , Diabetes Mellitus, Type 2/drug therapy , Diabetes Mellitus, Type 2/metabolism , Diabetic Nephropathies/metabolism , Diabetic Nephropathies/prevention & control , Disease Progression , Endoplasmic Reticulum Stress , Humans , Hypoglycemic Agents/pharmacology , Hypoglycemic Agents/therapeutic use , Ion Transport/drug effects , Kidney Tubules/cytology , Kidney Tubules/metabolism , Metformin/pharmacology , Metformin/therapeutic use , Mitochondria/metabolism , Oxidative Stress , Oxygen Consumption , Podocytes/pathology , Renal Circulation/drug effects , Signal Transduction , Sirtuin 1/deficiency , Sirtuin 1/physiology , Sodium/metabolism , Sodium-Glucose Transporter 2 Inhibitors/therapeutic use
12.
J Cell Mol Med ; 24(12): 6762-6772, 2020 06.
Article in English | MEDLINE | ID: mdl-32342656

ABSTRACT

This study addressed the hypothesis that cardiac Sirtuin 1 (Sirt1) deficiency alters cardiomyocyte Ca2+ and Na+ regulation, leading to cardiac dysfunction and arrhythmogenesis. We used mice with cardiac-specific Sirt1 knockout (Sirt1-/- ). Sirt1flox/flox mice were served as control. Sirt1-/- mice showed impaired cardiac ejection fraction with increased ventricular spontaneous activity and burst firing compared with those in control mice. The arrhythmic events were suppressed by KN93 and ranolazine. Reduction in Ca2+ transient amplitudes and sarcoplasmic reticulum (SR) Ca2+ stores, and increased SR Ca2+ leak were shown in the Sirt1-/- mice. Electrophysiological measurements were performed using patch-clamp method. While L-type Ca2+ current (ICa, L ) was smaller in Sirt1-/- myocytes, reverse-mode Na+ /Ca2+ exchanger (NCX) current was larger compared with those in control myocytes. Late Na+ current (INa, L ) was enhanced in the Sirt1-/- mice, alongside with elevated cytosolic Na+ level. Increased cytosolic and mitochondrial reactive oxygen species (ROS) were shown in Sirt1-/- mice. Sirt1-/- cardiomyocytes showed down-regulation of L-type Ca2+ channel α1c subunit (Cav1.2) and sarcoplasmic/endoplasmic reticulum Ca2+  ATPase 2a (SERCA2a), but up-regulation of Ca2+ /calmodulin-dependent protein kinase II and NCX. In conclusions, these findings suggest that deficiency of Sirt1 impairs the regulation of intracellular Ca2+ and Na+ in cardiomyocytes, thereby provoking cardiac dysfunction and arrhythmogenesis.


Subject(s)
Calcium/metabolism , Heart Ventricles/cytology , Myocytes, Cardiac/metabolism , Sirtuin 1/deficiency , Sodium/metabolism , Action Potentials , Animals , Calcium Channels, L-Type/metabolism , Cytosol/metabolism , Electrocardiography , Intracellular Space/metabolism , Ion Channel Gating , Mice, Inbred C57BL , Mice, Knockout , Oxidative Stress , Sarcoplasmic Reticulum/metabolism , Sirtuin 1/metabolism , Sodium-Calcium Exchanger/metabolism
13.
Mol Psychiatry ; 25(5): 1094-1111, 2020 05.
Article in English | MEDLINE | ID: mdl-30705425

ABSTRACT

Sirtuin 1 (SIRT1), an NAD+-dependent deacetylase, is a key regulator of cellular metabolism. Recent genome-wide association studies identified genetic variants of SIRT1 linked to major depressive disorders. SIRT1 is widely expressed in the brain; however, neuronal substrates that mediate SIRT1 action on depressive behaviors remain largely unknown. Here we show that selective deletion of SIRT1 in forebrain excitatory neurons causes depression-like phenotypes in male but not female mice. AAV-Cre-mediated SIRT1 knockdown in the medial prefrontal cortex (mPFC) of adult male mice induces depressive-like behaviors. Whole-cell patch-clamp recordings demonstrate that loss of SIRT1 decreases intrinsic excitability and spontaneous excitatory synaptic transmission in layer V pyramidal neurons in the prelimbic mPFC. Consistent with neuronal hypoexcitability, SIRT1 knockout reduces mitochondrial density and expression levels of genes involved in mitochondrial biogenesis and dynamics in the prelimbic mPFC. When a SIRT1 activator (SRT2104) is injected into the mPFC or lateral ventricle of wild-type mice, it reverses chronic unpredictable stress-induced anhedonia and behavioral despair, indicating an antidepressant-like effect. These results suggest that SIRT1 in mPFC excitatory neurons is required for normal neuronal excitability and synaptic transmission and regulates depression-related behaviors in a sex-specific manner.


Subject(s)
Depression/metabolism , Neurons/metabolism , Prefrontal Cortex/metabolism , Prefrontal Cortex/pathology , Sex Characteristics , Sirtuin 1/metabolism , Synaptic Transmission , Animals , Depression/genetics , Depression/pathology , Depressive Disorder, Major/genetics , Depressive Disorder, Major/metabolism , Depressive Disorder, Major/pathology , Excitatory Postsynaptic Potentials , Female , Male , Mice , Mice, Inbred C57BL , Pyramidal Cells/metabolism , Sirtuin 1/deficiency , Sirtuin 1/genetics
14.
Cell Death Differ ; 27(2): 482-496, 2020 02.
Article in English | MEDLINE | ID: mdl-31209362

ABSTRACT

Both the stress-response protein, SIRT1, and the cell cycle checkpoint kinase, CHK2, play critical roles in aging and cancer via the modulation of cellular homeostasis and the maintenance of genomic integrity. However, the underlying mechanism linking the two pathways remains elusive. Here, we show that SIRT1 functions as a modifier of CHK2 in cell cycle control. Specifically, SIRT1 interacts with CHK2 and deacetylates it at lysine 520 residue, which suppresses CHK2 phosphorylation, dimerization, and thus activation. SIRT1 depletion induces CHK2 hyperactivation-mediated cell cycle arrest and subsequent cell death. In vivo, genetic deletion of Chk2 rescues the neonatal lethality of Sirt1-/- mice, consistent with the role of SIRT1 in preventing CHK2 hyperactivation. Together, these results suggest that CHK2 mediates the function of SIRT1 in cell cycle progression, and may provide new insights into modulating cellular homeostasis and maintaining genomic integrity in the prevention of aging and cancer.


Subject(s)
Checkpoint Kinase 2/metabolism , Sirtuin 1/metabolism , Acetylation , Animals , Cell Cycle , Cells, Cultured , Checkpoint Kinase 2/deficiency , Humans , Mice , Mice, Knockout , Phosphorylation , Sirtuin 1/deficiency
15.
Cell Metab ; 30(2): 385-401.e9, 2019 08 06.
Article in English | MEDLINE | ID: mdl-31390551

ABSTRACT

The mechanisms by which steatosis of the liver progresses to non-alcoholic steatohepatitis and end-stage liver disease remain elusive. Metabolic derangements in hepatocytes controlled by SIRT1 play a role in the development of fatty liver in inbred animals. The ability to perform similar studies using human tissue has been limited by the genetic variability in man. We generated human induced pluripotent stem cells (iPSCs) with controllable expression of SIRT1. By differentiating edited iPSCs into hepatocytes and knocking down SIRT1, we found increased fatty acid biosynthesis that exacerbates fat accumulation. To model human fatty livers, we repopulated decellularized rat livers with human mesenchymal cells, fibroblasts, macrophages, and human SIRT1 knockdown iPSC-derived hepatocytes and found that the human iPSC-derived liver tissue developed macrosteatosis, acquired proinflammatory phenotype, and shared a similar lipid and metabolic profiling to human fatty livers. Biofabrication of genetically edited human liver tissue may become an important tool for investigating human liver biology and disease.


Subject(s)
Cell Engineering , Fatty Liver/metabolism , Pluripotent Stem Cells/metabolism , Sirtuin 1/metabolism , Adult , Animals , Cell Differentiation , Cells, Cultured , Fatty Acids/biosynthesis , Humans , Male , Pluripotent Stem Cells/cytology , Rats , Rats, Sprague-Dawley , Sirtuin 1/deficiency , Sirtuin 1/genetics
16.
PLoS One ; 14(8): e0220581, 2019.
Article in English | MEDLINE | ID: mdl-31386694

ABSTRACT

Sirt1, a member of the sirtuin gene family, encodes the most conserved mammalian NAD+-dependent deacetylase enzyme responsible for removing acetyl groups from many proteins. The Sirt1 gene is known as a longevity gene whose knockout in mice leads to decreased lifespan relative to the wild type. This study aimed to explore phenotypic changes in zebrafish Sirt1-knockouts and to investigate the function of the Sirt1 gene. Targeted knockout of Sirt1 in zebrafish (Danio rerio) was achieved using the CRISPR-Cas9 genome editing system. We created a 4-bp insertion-homozygote Sirt1-knockout zebrafish. Although there was no evident difference in appearance in the early stages of development, a significant increase in reactive oxygen species and in the extent of apoptosis in Sirt1-knockout zebrafish was observed. Sirt1 knockout caused inflammatory genes, including IL-1b, IL-6 and TNF-α to be highly expressed. Additionally, the lack of Sirt1 caused chronic inflammation and intestinal atrophy, thereby increasing pro-apoptotic events, which ultimately reduced the lifespan of transgenic zebrafish. Overall, our data demonstrate that lack of Sirt1 caused a significantly increased generation of reactive oxygen species that resulted in chronic inflammation and regeneration. Continuous repetition of these events played an important role in accelerating aging, thereby decreasing lifespan. Our findings using the knockout zebrafish model confirmed the association of the Sirt1 gene to aging processes and lifespan. Furthermore, the Sirt1-knockout mutant zebrafish developed in our study will surely be a valuable model to explore the mechanism of chronic inflammation.


Subject(s)
Inflammation/genetics , Longevity/drug effects , Oxidative Stress/drug effects , Sirtuin 1/genetics , Aging/genetics , Animals , Gene Knockout Techniques , Reactive Oxygen Species/metabolism , Sirtuin 1/deficiency , Sirtuin 1/pharmacology , Zebrafish
17.
J Pharmacol Sci ; 140(1): 79-85, 2019 May.
Article in English | MEDLINE | ID: mdl-31178326

ABSTRACT

Bone marrow failure is a disease syndrome with the disability to produce mature blood cells. Pancytopenia is the most common manifestation of bone marrow failure. Sirt1 is important for the function of hematopoietic stem cells, we hypothesized that Sirt1 activation may improve hematopoiesis. The Sirt1 heterozygous and wild type mice were exposed to lethal 6.5 Gy 60Co-γ rays. The survival time and hematopoietic indexes were evaluated. The survival time of Sirt1 deficiency mice was significantly decreased. The numbers of platelets (PLT), reticulocytes (RET) and white blood cells (WBC) were significantly decreased. C57BL/6 mice were exposed to 6.5 Gy 60Co-γ rays then administrated with resveratrol (20 mg/kg/d) or vehicle. Resveratrol increased the survival time and protective against irradiation induced hematopoietic damage. Resveratrol also significantly increased the numbers of PLT, RET and WBC of mice. It also increased the hematopoietic area and karyocytes number. In HEK293T cells, the expression of LKB1 was significantly increased in cytoplasm but not in nuclei when treated with resveratrol (50 µM). These results suggest that Sirt1 deficiency might aggravate bone marrow failure. Resveratrol corrected this hematopoietic defect and LKB1 might involve in the protective effect on bone marrow failure.


Subject(s)
Gamma Rays/adverse effects , Hematopoiesis/drug effects , Hematopoiesis/genetics , Pancytopenia/blood , Pancytopenia/etiology , Radiation Exposure/adverse effects , Radiation-Protective Agents/pharmacology , Resveratrol/pharmacology , Sirtuin 1/metabolism , AMP-Activated Protein Kinase Kinases , Animals , Gene Expression/drug effects , HEK293 Cells , Humans , Leukocyte Count , Mice, Knockout , Platelet Count , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Reticulocyte Count , Sirtuin 1/deficiency , Sirtuin 1/physiology , Stimulation, Chemical
18.
Mol Biol Rep ; 46(4): 4225-4234, 2019 Aug.
Article in English | MEDLINE | ID: mdl-31102152

ABSTRACT

Sirt1, also known as the longevity gene, is an NAD+-dependent class III histone deacetylase that has been extensively studied in multiple areas of research including cellular metabolism, longevity, cancer, autoimmunity, and immunity. However, little is known about the function of Sirt1 in B cells. This study aimed to investigate the role of Sirt1 in the expression pattern of mRNAs in the resting B cells of mice. CD19+ B cell-specific inducible Sirt1 knockout (KO) mice were divided into tamoxifen-treated Sirt1 KO group (S19T) or control group (S19). mRNAs extracted from resting B cells of both groups were analyzed for differentially expressed genes (DEG) using microarray. DEG analysis showed significant differential expression of 20 genes, of which Hspa1a and Hspa1b showed the highest fold change (FC) in S19T compared with S19 (p value < 0.01 and FC > 3). Further, Kyoto Encyclopedia of Genes and Genomes analysis identified pathways associated with diseases, organismal systems, and antigen processing and presentation. Additionally, the pathways known to involve Hspa1a and Hspa1b were also activated in the S19T group. On the other hand, after in vitro stimulation with lipopolysaccharide, cell viability and IgM production were significantly decreased in Sirt1 KO B cells, while expressions of TNF-α, IL-6, and IL-10 were increased. In summary, our study reveals that Sirt1 may maintain the quiescent state in resting B cells by suppressing the increase of Hspa1a and Hspa1b. This work provides a foundation for further studies on the functional roles of Sirt1 in B cells.


Subject(s)
B-Lymphocytes/metabolism , HSP70 Heat-Shock Proteins/genetics , Sirtuin 1/deficiency , Animals , B-Lymphocytes/physiology , Cell Survival , Female , HSP70 Heat-Shock Proteins/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , RNA, Messenger/genetics , RNA, Messenger/metabolism , Sirtuin 1/genetics , Sirtuin 1/metabolism
19.
Physiol Rep ; 7(9): e14044, 2019 05.
Article in English | MEDLINE | ID: mdl-31087539

ABSTRACT

Glomerular filtration rate (GFR) declines with age such that the prevalence of chronic kidney disease is much higher in the elderly. SIRT1 is the leading member of the sirtuin family of NAD+ -dependent lysine deacetylases that mediate the health span extending properties of caloric restriction. Since reduction in energy intake has also been shown to decrease age-related kidney disease in rodents, we hypothesized that a diminution in SIRT1 activity would accelerate the GFR decline and structural injury with age. To test this hypothesis, we compared changes in the kidney structure and function in control mice and mice that carry a point mutation at a conserved histidine (H355Y) of SIRT1 that renders the enzyme catalytically inactive. Taking advantage of this mouse model along with the disector/fractionator technique for glomerular counting and direct measurements of GFR by inulin clearance, we assessed the impact of SIRT1 inactivity on kidney aging. At 14 months of age, SIRT1 catalytically inactive (Sirt1Y/Y ) mice had lower GFRs and fewer glomeruli than their wild-type (Sirt1+/+ ) counterparts. This was not, however, due to either accelerated GFR decline or increased glomerulosclerosis and loss, but rather to reduced glomerular endowment in Sirt1Y/Y mice. Moreover, the compensatory glomerular hypertrophy and elevated single nephron GFR that customarily accompany reduction in nephron number were absent in Sirt1Y/Y mice. These findings suggest a role for SIRT1 not only in determining nephron endowment but also in orchestrating the response to it.


Subject(s)
Aging/physiology , Glomerular Filtration Rate/physiology , Kidney Glomerulus/pathology , Sirtuin 1/physiology , Aging/pathology , Animals , Body Weight/physiology , Disease Models, Animal , Histone Deacetylases/metabolism , Kidney/pathology , Male , Mice, Mutant Strains , Nephrons/pathology , Organ Size/physiology , Point Mutation , Sirtuin 1/deficiency , Sirtuin 1/genetics
20.
Rheumatology (Oxford) ; 58(9): 1674-1683, 2019 09 01.
Article in English | MEDLINE | ID: mdl-31106362

ABSTRACT

OBJECTIVES: Acute gout is an inflammatory response to MSU crystals. In our previous research, Sirt1 was shown to have an effect in preventing acute gouty inflammation. In the current study, we aimed to investigate the underlying mechanism involving Sirt1 in acute gout. METHODS: The cytological changes and Sirt1 expression in the synovium were observed in patients with acute or intermittent gout. The effect of Sirt1 and its mechanism in gout were studied in macrophages, C57BL/6 mice and Sirt1+/- mice. RESULTS: Sirt1 expression was increased in the peripheral blood mononuclear cells (PBMCs) of patients with acute gout but not in the chronic tophus tissue. The arthritis score and numbers of inflammatory cells in injured paw tissue from murine gout models were upregulated in Sirt1+/- mice compared with wild-type mice. A PCR array of the paw tissue from murine gout models indicated that Sirt1 activation might attenuate MSU-induced inflammation by altering the polarization state of macrophages. Furthermore, in patients with acute gout, the phagocytosis of MSU crystals by a macrophage was found in a smear of the joint fluid and large amounts of macrophages were also found in the synovium. The activation of Sirt1 in gouty mice actually decreased the tendency toward M1 polarization. The inhibition of PI3K/Akt partially blocked the anti-inflammatory effect of Sirt1 and the translocation of STAT6, and phosphorylated STAT6 expression was decreased in RAW 264.7 cells treated with MSU crystals. CONCLUSION: Our studies revealed that Sirt1 ameliorates MSU-induced inflammation by altering macrophage polarization via the PI3K/Akt/STAT6 pathway.


Subject(s)
Arthritis, Experimental/pathology , Arthritis, Gouty/pathology , Macrophages/pathology , Sirtuin 1/physiology , Acute Disease , Adult , Animals , Arthritis, Experimental/chemically induced , Arthritis, Experimental/metabolism , Arthritis, Gouty/chemically induced , Arthritis, Gouty/metabolism , Cell Polarity/physiology , Gout/metabolism , Gout/pathology , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Middle Aged , Phagocytosis/physiology , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , RAW 264.7 Cells , STAT6 Transcription Factor/metabolism , Sirtuin 1/blood , Sirtuin 1/deficiency , Synovial Membrane/metabolism , Synovial Membrane/pathology , Uric Acid
SELECTION OF CITATIONS
SEARCH DETAIL
...