Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 95
Filter
Add more filters










Publication year range
1.
Biochem Biophys Res Commun ; 726: 150235, 2024 Sep 24.
Article in English | MEDLINE | ID: mdl-38908345

ABSTRACT

BACKGROUND: Diabetic ulcers (DUs) are characterized by chronic inflammation and delayed re-epithelialization, with a high incidence and weighty economic burden. The primary therapeutic strategies for refractory wounds include surgery, non-invasive wound therapy, and drugs, while the optimum regimen remains controversial. Sirtuin-6 (SIRT6) is a histone deacetylase and a key epigenetic factor that exerts anti-inflammatory and pro-proliferatory effects in wound healing. However, the exact function of SIRT6 in DUs remains unclear. METHODS: We generated tamoxifen-inducible SIRT6 knockout mice by crossing SIRT6flox/flox homozygous mice with UBC-creERT2+ transgenic mice. Systemic SIRT6 null mice, under either normal or diabetic conditions, were utilized to assess the effects of SIRT6 in DUs treatment. Gene and protein expressions of SIRT6 and inflammatory cytokines were measured by Western blotting and RT-qPCR. Histopathological examination confirmed the altered re-epithelialization (PCNA), inflammation (NF-κB p50 and F4/80), and angiogenesis (CD31) markers during DUs restoration. RESULTS: Knockout of SIRT6 inhibited the healing ability of DUs, presenting attenuated re-epithelialization (PCNA), exacerbated inflammation responses (NF-κB p50, F4/80, Il-1ß, Tnf-α, Il-6, Il-10, and Il-4), and hyperplasia vascular (CD31) compared with control mice. CONCLUSIONS: SIRT6 could boost impaired wound healing through improving epidermal proliferation, inflammation, and angiogenesis. Our study highlighted the therapeutic potential of the SIRT6 agonist for DUs treatment.


Subject(s)
Mice, Knockout , Sirtuins , Wound Healing , Animals , Wound Healing/genetics , Sirtuins/genetics , Sirtuins/metabolism , Sirtuins/deficiency , Mice , Diabetes Mellitus, Experimental/genetics , Diabetes Mellitus, Experimental/complications , Diabetes Mellitus, Experimental/metabolism , Diabetes Mellitus, Experimental/pathology , Cytokines/metabolism , Mice, Inbred C57BL , Inflammation/genetics , Inflammation/pathology , Inflammation/metabolism , Male
2.
Comput Math Methods Med ; 2022: 3200798, 2022.
Article in English | MEDLINE | ID: mdl-35035519

ABSTRACT

BACKGROUND: Data mining of current gene expression databases has not been previously performed to determine whether sirtuin 6 (SIRT6) expression participates in the pathological process of abdominal aortic aneurysm (AAA). The present study was aimed at investigating the role and mechanism of SIRT6 in regulating phenotype transformation of vascular smooth muscle cells (VSMC) in AAA. METHODS: Three gene expression microarray datasets of AAA patients in the Gene Expression Omnibus (GEO) database and one dataset of SIRT6-knockout (KO) mice were selected, and the differentially expressed genes (DEGs) were identified using GEO2R. Furthermore, Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) analyses of both the AAA-related DEGs and the SIRT6-related DEGs were conducted. RESULTS: GEO2R analysis showed that the expression of SIRT6 was downregulated for three groups and upregulated for one group in the three datasets, and none of them satisfied statistical significance. There were top 5 DEGs (KYNU, NPTX2, SCRG1, GRK5, and RGS5) in both of the human AAA group and SIRT6-KO mouse group. Top 25 ontology of the SIRT6-KO-related DEGs showed that several pathways including tryptophan catabolic process to kynurenine and negative regulation of cell growth were enriched in the tissues of thickness aortic wall biopsies of AAA patients. CONCLUSIONS: Although SIRT6 mRNA level itself did not change among AAA patients, SIRT6 may play an important role in regulating several signaling pathways with significant association with AAA, suggesting that SIRT6 mRNA upregulation is a protective factor for VSMC against AAA.


Subject(s)
Aortic Aneurysm, Abdominal/genetics , Aortic Aneurysm, Abdominal/metabolism , Myocytes, Smooth Muscle/metabolism , Sirtuins/genetics , Sirtuins/metabolism , Animals , Aortic Aneurysm, Abdominal/pathology , Computational Biology , Databases, Genetic/statistics & numerical data , Down-Regulation , Gene Ontology , Gene Regulatory Networks , Humans , Male , Mice , Mice, Knockout , Myocytes, Smooth Muscle/pathology , Phenotype , RNA, Messenger/genetics , RNA, Messenger/metabolism , Signal Transduction/genetics , Sirtuins/deficiency , Up-Regulation
3.
Exp Mol Med ; 53(9): 1298-1306, 2021 09.
Article in English | MEDLINE | ID: mdl-34493807

ABSTRACT

Intermittent fasting (IF) is gaining popularity for its effectiveness in improving overall health, including its effectiveness in achieving weight loss and euglycemia. The molecular mechanisms of IF, however, are not well understood. This study investigated the relationship between adipocyte sirtuin 6 (Sirt6) and the metabolic benefits of IF. Adipocyte-specific Sirt6-knockout (aS6KO) mice and wild-type littermates were fed a high-fat diet (HFD) ad libitum for four weeks and then subjected to 12 weeks on a 2:1 IF regimen consisting of two days of feeding followed by one day of fasting. Compared with wild-type mice, aS6KO mice subjected to HFD + IF exhibited a diminished response, as reflected by their glucose and insulin intolerance, reduced energy expenditure and adipose tissue browning, and increased inflammation of white adipose tissue. Sirt6 deficiency in hepatocytes or in myeloid cells did not impair adaptation to IF. Finally, the results indicated that the impaired adipose tissue browning and reduced expression of UCP1 in aS6KO mice were accompanied by downregulation of p38 MAPK/ATF2 signaling. Our findings indicate that Sirt6 in adipocytes is critical to obtaining the improved glucose metabolism and metabolic profiles conferred by IF and that maintaining high levels of Sirt6 in adipocytes may mimic the health benefits of IF.


Subject(s)
Adaptation, Biological , Adipocytes/metabolism , Adipose Tissue/metabolism , Fasting/metabolism , Sirtuins/deficiency , Adipose Tissue/cytology , Adipose Tissue, Brown/metabolism , Adipose Tissue, White/metabolism , Animals , Diet, High-Fat , Energy Metabolism , Gene Knockdown Techniques , Insulin Resistance , Mice , Mice, Knockout
4.
Gastroenterology ; 161(5): 1584-1600, 2021 11.
Article in English | MEDLINE | ID: mdl-34245764

ABSTRACT

BACKGROUND & AIMS: SIRT5 plays pleiotropic roles via post-translational modifications, serving as a tumor suppressor, or an oncogene, in different tumors. However, the role SIRT5 plays in the initiation and progression of pancreatic ductal adenocarcinoma (PDAC) remains unknown. METHODS: Published datasets and tissue arrays with SIRT5 staining were used to investigate the clinical relevance of SIRT5 in PDAC. Furthermore, to define the role of SIRT5 in the carcinogenesis of PDAC, we generated autochthonous mouse models with conditional Sirt5 knockout. Moreover, to examine the mechanistic role of SIRT5 in PDAC carcinogenesis, SIRT5 was knocked down in PDAC cell lines and organoids, followed by metabolomics and proteomics studies. A novel SIRT5 activator was used for therapeutic studies in organoids and patient-derived xenografts. RESULTS: SIRT5 expression negatively regulated tumor cell proliferation and correlated with a favorable prognosis in patients with PDAC. Genetic ablation of Sirt5 in PDAC mouse models promoted acinar-to-ductal metaplasia, precursor lesions, and pancreatic tumorigenesis, resulting in poor survival. Mechanistically, SIRT5 loss enhanced glutamine and glutathione metabolism via acetylation-mediated activation of GOT1. A selective SIRT5 activator, MC3138, phenocopied the effects of SIRT5 overexpression and exhibited antitumor effects on human PDAC cells. MC3138 also diminished nucleotide pools, sensitizing human PDAC cell lines, organoids, and patient-derived xenografts to gemcitabine. CONCLUSIONS: Collectively, we identify SIRT5 as a key tumor suppressor in PDAC, whose loss promotes tumorigenesis through increased noncanonic use of glutamine via GOT1, and that SIRT5 activation is a novel therapeutic strategy to target PDAC.


Subject(s)
Carcinoma, Pancreatic Ductal/enzymology , Energy Metabolism , Pancreatic Neoplasms/enzymology , Proto-Oncogene Proteins p21(ras)/metabolism , Sirtuins/deficiency , Animals , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Aspartate Aminotransferase, Cytoplasmic/genetics , Aspartate Aminotransferase, Cytoplasmic/metabolism , Carcinoma, Pancreatic Ductal/drug therapy , Carcinoma, Pancreatic Ductal/genetics , Carcinoma, Pancreatic Ductal/pathology , Cell Line, Tumor , Deoxycytidine/analogs & derivatives , Deoxycytidine/pharmacology , Disease Progression , Energy Metabolism/drug effects , Enzyme Activation , Enzyme Activators/pharmacology , Female , Gene Expression Regulation, Neoplastic , Humans , Male , Mice, Inbred C57BL , Mice, Inbred NOD , Mice, Knockout , Mice, SCID , Mutation , Pancreatic Neoplasms/drug therapy , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/pathology , Proto-Oncogene Proteins p21(ras)/genetics , Signal Transduction , Sirtuins/genetics , Tumor Burden , Tumor Cells, Cultured , Xenograft Model Antitumor Assays , Gemcitabine
5.
Cell Rep ; 35(9): 109190, 2021 06 01.
Article in English | MEDLINE | ID: mdl-34077730

ABSTRACT

Pathological lipid accumulation is often associated with enhanced uptake of free fatty acids via specific transporters in cardiomyocytes. Here, we identify SIRT6 as a critical transcriptional regulator of fatty acid transporters in cardiomyocytes. We find that SIRT6 deficiency enhances the expression of fatty acid transporters, leading to enhanced fatty acid uptake and lipid accumulation. Interestingly, the haploinsufficiency of SIRT6 is sufficient to induce the expression of fatty acid transporters and cause lipid accumulation in murine hearts. Mechanistically, SIRT6 depletion enhances the occupancy of the transcription factor PPARγ on the promoters of critical fatty acid transporters without modulating the acetylation of histone 3 at Lys 9 and Lys 56. Notably, the binding of SIRT6 to the DNA-binding domain of PPARγ is critical for regulating the expression of fatty acid transporters in cardiomyocytes. Our data suggest exploiting SIRT6 as a potential therapeutic target for protecting the heart from metabolic diseases.


Subject(s)
Fatty Acids/metabolism , PPAR gamma/metabolism , Sirtuins/metabolism , Transcription, Genetic , Adult , Animals , Biological Transport/genetics , Diabetic Cardiomyopathies/genetics , Diabetic Cardiomyopathies/pathology , Disease Models, Animal , Female , HEK293 Cells , Heart Failure/genetics , Humans , Male , Membrane Transport Proteins/metabolism , Mice , Mice, Inbred C57BL , Myocytes, Cardiac/metabolism , PPAR gamma/chemistry , Promoter Regions, Genetic/genetics , Protein Domains , Sirtuins/deficiency , Sirtuins/genetics
6.
Cartilage ; 13(2_suppl): 1185S-1199S, 2021 12.
Article in English | MEDLINE | ID: mdl-33567897

ABSTRACT

OBJECTIVE: Obesity accelerates the development of osteoarthritis (OA) during aging and is associated with altered chondrocyte cellular metabolism. Protein lysine malonylation (MaK) is a posttranslational modification (PTM) that has been shown to play an important role during aging and obesity. The objective of this study was to investigate the role of sirtuin 5 (Sirt5) in regulating MaK and cellular metabolism in chondrocytes under obesity-related conditions. METHODS: MaK and SIRT5 were immunostained in knee articular cartilage of obese db/db mice and different aged C57BL6 mice with or without destabilization of the medial meniscus surgery to induce OA. Primary chondrocytes were isolated from 7-day-old WT and Sirt5-/- mice and treated with varying concentrations of glucose and insulin to mimic obesity. Sirt5-dependent effects on MaK and metabolism were evaluated by western blot, Seahorse Respirometry, and gas/chromatography-mass/spectrometry (GC-MS) metabolic profiling. RESULTS: MaK was significantly increased in cartilage of db/db mice and in chondrocytes treated with high concentrations of glucose and insulin (GluhiInshi). Sirt5 was increased in an age-dependent manner following joint injury, and Sirt5 deficient primary chondrocytes had increased MaK, decreased glycolysis rate, and reduced basal mitochondrial respiration. GC-MS identified 41 metabolites. Sirt5 deficiency altered 13 distinct metabolites under basal conditions and 18 metabolites under GluhiInshi treatment. Pathway analysis identified a wide range of Sirt5-dependent altered metabolic pathways that include amino acid metabolism, TCA cycle, and glycolysis. CONCLUSION: This study provides the first evidence that Sirt5 broadly regulates chondrocyte metabolism. We observed changes in SIRT5 and MaK levels in cartilage with obesity and joint injury, suggesting that the Sirt5-MaK pathway may contribute to altered chondrocyte metabolism that occurs during OA development.


Subject(s)
Cartilage, Articular , Chondrocytes , Obesity , Sirtuins , Animals , Cartilage, Articular/metabolism , Cartilage, Articular/pathology , Chondrocytes/metabolism , Chondrocytes/pathology , Mice , Mice, Inbred C57BL , Obesity/metabolism , Obesity/pathology , Osteoarthritis/metabolism , Sirtuins/deficiency , Sirtuins/metabolism
7.
Neurochem Int ; 145: 104959, 2021 05.
Article in English | MEDLINE | ID: mdl-33444676

ABSTRACT

Sirtuin 6 (SIRT6), a member of the Sirtuin family, acts as nicotinamide adenine dinucleotide (NAD)-dependent protein deacetylase, mono-adenosine diphosphate (ADP)-ribosyltransferase, and fatty acid deacylase, and plays critical roles in inflammation, aging, glycolysis, and DNA repair. Accumulating evidence has suggested that SIRT6 is involved in brain functions such as neuronal differentiation, neurogenesis, and learning and memory. However, the precise molecular roles of SIRT6 during neuronal circuit formation are not yet well understood. In this study, we tried to elucidate molecular roles of SIRT6 on neurite development by using primary-cultured hippocampal neurons. We observed that SIRT6 was abundantly localized in the nucleus, and its expression was markedly increased during neurite outgrowth and synaptogenesis. By using shRNA-mediated SIRT6-knockdown, we show that both dendritic length and the number of dendrite branches were significantly reduced in the SIRT6-knockdown neurons. Microarray and subsequent gene ontology analysis revealed that reducing SIRT6 caused the downregulation of immediate early genes (IEGs) and alteration of several biological processes including MAPK (ERK1/2) signaling. We found that nuclear accumulation of phosphorylated ERK1/2 was significantly reduced in SIRT6-knockdown neurons. Overexpression of SIRT6 promoted dendritic length and branching, but the mutants lacking deacetylase activity had no significant effect on the dendritic morphology. Collectively, the presented findings reveal a role of SIRT6 in dendrite morphogenesis, and suggest that SIRT6 may act as an important regulator of ERK1/2 signaling pathway that mediates IEG expression, which leads to dendritic development.


Subject(s)
Dendrites/metabolism , Hippocampus/cytology , Hippocampus/metabolism , Neurogenesis/physiology , Neurons/metabolism , Sirtuins/biosynthesis , Animals , Animals, Newborn , Cells, Cultured , Gene Knockdown Techniques/methods , Rats , Sirtuins/deficiency , Sirtuins/genetics
8.
Nat Commun ; 11(1): 5927, 2020 11 23.
Article in English | MEDLINE | ID: mdl-33230181

ABSTRACT

Mitochondrial acyl-coenzyme A species are emerging as important sources of protein modification and damage. Succinyl-CoA ligase (SCL) deficiency causes a mitochondrial encephalomyopathy of unknown pathomechanism. Here, we show that succinyl-CoA accumulates in cells derived from patients with recessive mutations in the tricarboxylic acid cycle (TCA) gene succinyl-CoA ligase subunit-ß (SUCLA2), causing global protein hyper-succinylation. Using mass spectrometry, we quantify nearly 1,000 protein succinylation sites on 366 proteins from patient-derived fibroblasts and myotubes. Interestingly, hyper-succinylated proteins are distributed across cellular compartments, and many are known targets of the (NAD+)-dependent desuccinylase SIRT5. To test the contribution of hyper-succinylation to disease progression, we develop a zebrafish model of the SCL deficiency and find that SIRT5 gain-of-function reduces global protein succinylation and improves survival. Thus, increased succinyl-CoA levels contribute to the pathology of SCL deficiency through post-translational modifications.


Subject(s)
Acyl Coenzyme A/metabolism , Mitochondrial Diseases/pathology , Succinate-CoA Ligases/genetics , Animals , Cells, Cultured , Female , Humans , Infant , Lysine/metabolism , Male , Mice , Mice, Knockout , Mitochondria/metabolism , Mitochondrial Diseases/genetics , Mitochondrial Diseases/metabolism , Mutation , Proteomics , Sirtuins/deficiency , Sirtuins/genetics , Sirtuins/metabolism , Succinate-CoA Ligases/deficiency , Succinate-CoA Ligases/metabolism , Survival Analysis , Zebrafish
9.
Eur Rev Med Pharmacol Sci ; 24(18): 9246-9255, 2020 09.
Article in English | MEDLINE | ID: mdl-33015765

ABSTRACT

OBJECTIVE: Osteoarthritis (OA) is associated with decreased autophagy activity and imbalance of cell homeostasis in chondrocytes (CHs). Sirtuin 7 (Sirt7) is claimed to have the ability to activate the autophagy response. The aim of this study was to explore the function of Sirt7 in the development of OA involving autophagy by culturing human chondrocytes (CHs). PATIENTS AND METHODS: We collected knee joint cartilage from patients undergoing traumatic amputation and arthroscopic knee replacement. Protein and mRNA levels of collagen II, Sirt7, ULK1, Lc3, and Beclin1 were analyzed by Western blot and RT-PCR. CHs were isolated from the traumatic cartilage as a control group, and IL-1ß was used to induce CHs degeneration. The expression of Sirt7 gene was silenced by siRNA and upregulated by recombinant human Sirt7 protein (rh-Sirt7). An autophagy activator Tat-beclin 1 (Tat) was used to activate autophagy in cultural CHs. Expression of collagen II, Sirt7, ULK1, Lc3, and Beclin1 was determined by immunofluorescence, Western blot, and RT-PCR, respectively. RESULTS: The protein and mRNA levels of Collagen II, Sirt7, ULK1, Lc3-II, and Beclin1 expressions were decreased in OA cartilage compared with those in healthy cartilage. IL-1ß degenerated the CHs resulting in a reduction of collagen II, which also downregulated Sirt7, ULK1, Lc3-II, and Beclin1. Sirt7 deficiency accelerated the catabolism of collagen II and weakened the expression of ULK1, Lc3-II, and Beclin1. On the contrary, exogenous rh-Sirt7 played a positive role in these gene expressions. Finally, Tat alleviated the CHs degeneration by upregulating collagen II and activating ULK1, Lc3-II, and Beclin1, but incapable to mediate Sirt7 expression. CONCLUSIONS: Overall, these findings suggested that Sirt7 was suppressed in the degenerated cartilage. Sirt7 deficiency does harm to the autophagy level, affecting CHs metabolism, while the upregulation of Sirt7 activated autophagy and protected CHs degeneration. An appropriate increase in autophagy can protect CHs but has no effect on Sirt7 expression.


Subject(s)
Autophagy , Chondrocytes/metabolism , Osteoarthritis/metabolism , Sirtuins/metabolism , Cells, Cultured , Humans , Sirtuins/deficiency , Sirtuins/genetics
10.
Theranostics ; 10(16): 7465-7479, 2020.
Article in English | MEDLINE | ID: mdl-32642006

ABSTRACT

Disturbed renal lipid metabolism, especially cholesterol dysregulation plays a crucial role in the pathogenesis of chronic kidney disease (CKD). We recently reported that angiotensin (Ang) II could induce cholesterol accumulation and injury in podocytes. However, the underlying mechanisms for these alterations remain unknown. Methods: Bioinformatics analysis of renal biopsy specimens from patients with hypertensive nephropathy (HN) suggests the involvement of Sirtuin 6 (Sirt6) in Ang II-induced dysregulation of glomerular cholesterol. Using a podocyte-specific Sirt6 knockout mouse model, the effects of Sirt6 on Ang II-induced cholesterol accumulation in podocytes and the therapeutic efficacies of cholesterol-lowering agents were evaluated. Results: Cholesterol accumulation was detected in the podocytes of Ang II-infused mice, whereas selective deletion of Sirt6 in podocytes not only increased cholesterol accumulation in these cells but also exacerbated Ang II-induced kidney injury. Deletion of Sirt6 also attenuated the protective effect of cyclodextrin (CD) on Ang II-induced urinary albumin excretion, glomerulosclerosis and podocyte injury. In addition, we demonstrated that Sirt6 affected cholesterol efflux in podocytes by regulating the expression of ATP-binding cassette transporter G1 (ABCG1). Conclusions: These findings provide evidence that Sirt6 is a potential target for renin-angiotensin system (RAS)-associated podocyte injury and provide a rationale for the application of cholesterol-lowering agents in patients with CKD.


Subject(s)
Cholesterol/metabolism , Podocytes/pathology , Renal Insufficiency, Chronic/pathology , Sirtuins/deficiency , Sirtuins/metabolism , Adult , Angiotensin II/administration & dosage , Angiotensin II/toxicity , Animals , Anticholesteremic Agents/pharmacology , Anticholesteremic Agents/therapeutic use , Biopsy , Case-Control Studies , Cell Line , Cholesterol/blood , Datasets as Topic , Disease Models, Animal , Female , Gene Expression Profiling , Humans , Lipid Metabolism/drug effects , Male , Mice , Mice, Knockout , Middle Aged , Oligonucleotide Array Sequence Analysis , Podocytes/drug effects , Renal Insufficiency, Chronic/blood , Renal Insufficiency, Chronic/chemically induced , Renal Insufficiency, Chronic/drug therapy , Sirtuins/genetics
11.
Sci Rep ; 10(1): 12497, 2020 07 27.
Article in English | MEDLINE | ID: mdl-32719338

ABSTRACT

Acute lung injury (ALI), a common condition in critically ill patients, has limited treatments and high mortality. Aging is a risk factor for ALI. Sirtuins (SIRTs), central regulators of the aging process, decrease during normal aging and in aging-related diseases. We recently showed decreased SIRT7 expression in lung tissues and fibroblasts from patients with pulmonary fibrosis compared to controls. To gain insight into aging-related mechanisms in ALI, we investigated the effects of SIRT7 depletion on lipopolysaccharide (LPS)-induced inflammatory responses and endothelial barrier permeability in human primary pulmonary endothelial cells. Silencing SIRT7 in pulmonary artery or microvascular endothelial cells attenuated LPS-induced increases in ICAM1, VCAM1, IL8, and IL6 and induced endomesenchymal transition (EndoMT) with decreases in VE-Cadherin and PECAM1 and increases in collagen, alpha-smooth muscle actin, TGFß receptor 1, and the transcription factor Snail. Loss of endothelial adhesion molecules was accompanied by increased F-actin stress fibers and increased endothelial barrier permeability. Together, these results show that an aging phenotype induced by SIRT7 deficiency promotes EndoMT with impaired inflammatory responses and dysfunction of the lung vascular barrier.


Subject(s)
Capillary Permeability , Endothelial Cells/pathology , Epithelium/pathology , Inflammation/metabolism , Lung/pathology , Sirtuins/deficiency , Adult , Animals , Bleomycin , Cell Membrane Permeability , Cells, Cultured , Endothelial Cells/metabolism , Gene Expression Regulation , Gene Silencing , Humans , Inflammation Mediators/metabolism , Lipopolysaccharides , Mice, Inbred C57BL , NF-kappa B/metabolism , Pulmonary Fibrosis/genetics , Pulmonary Fibrosis/pathology , Pulmonary Fibrosis/physiopathology , RNA, Messenger/genetics , RNA, Messenger/metabolism , Signal Transduction , Sirtuins/genetics , Sirtuins/metabolism , Transforming Growth Factor beta/metabolism
12.
Protein Cell ; 11(7): 483-504, 2020 07.
Article in English | MEDLINE | ID: mdl-32504224

ABSTRACT

SIRT7, a sirtuin family member implicated in aging and disease, is a regulator of metabolism and stress responses. It remains elusive how human somatic stem cell populations might be impacted by SIRT7. Here, we found that SIRT7 expression declines during human mesenchymal stem cell (hMSC) aging and that SIRT7 deficiency accelerates senescence. Mechanistically, SIRT7 forms a complex with nuclear lamina proteins and heterochromatin proteins, thus maintaining the repressive state of heterochromatin at nuclear periphery. Accordingly, deficiency of SIRT7 results in loss of heterochromatin, de-repression of the LINE1 retrotransposon (LINE1), and activation of innate immune signaling via the cGAS-STING pathway. These aging-associated cellular defects were reversed by overexpression of heterochromatin proteins or treatment with a LINE1 targeted reverse-transcriptase inhibitor. Together, these findings highlight how SIRT7 safeguards chromatin architecture to control innate immune regulation and ensure geroprotection during stem cell aging.


Subject(s)
Cellular Senescence , Heterochromatin/metabolism , Mesenchymal Stem Cells/metabolism , Sirtuins/metabolism , Animals , Cells, Cultured , HEK293 Cells , Humans , Male , Mesenchymal Stem Cells/cytology , Mice , Mice, Nude , Sirtuins/deficiency
13.
Cell Mol Gastroenterol Hepatol ; 10(2): 341-364, 2020.
Article in English | MEDLINE | ID: mdl-32305562

ABSTRACT

BACKGROUND & AIMS: Nonalcoholic steatohepatitis (NASH) is a chronic liver disease that is manifested clinically by an increase in hepatic triglycerides, inflammation, and fibrosis. The pathogenesis of NASH remains incompletely understood. Sirtuin 6 (Sirt6), a nicotinamide adenine dinucleotide-dependent deacetylase, has been implicated in fatty liver disease; however, the underlying molecular mechanisms in the NASH pathogenesis are elusive. The aims of this study were to elucidate the role of hepatic Sirt6 in NASH. METHODS: Wild-type, liver-specific Sirt6 knockout (KO), hepatic stellate cell (HSC)-specific Sirt6 knockout (HSC-KO), and Sirt6 transgenic mice were subjected to a Western diet for 4 weeks. Hepatic phenotypes were characterized and underlying mechanisms were investigated. RESULTS: Remarkably, both the liver-KO and HSC-KO mice developed much worse NASH than the wild-type mice, whereas the transgenic mice were protected from the diet-induced NASH. Our cell signaling analysis showed that Sirt6 negatively regulates the transforming growth factor ß-Smad family member 3 (Smad3) pathway. Biochemical analysis showed a physical interaction between Sirt6 and Smad3 in hepatic stellate cells. Moreover, our molecular data further showed that Sirt6 deacetylated Smad3 at key lysine residues K333 and K378, and attenuated its transcriptional activity induced by transforming growth factor ß in hepatic stellate cells. CONCLUSIONS: Our data suggest that SIRT6 plays a critical role in the protection against NASH development and it may serve as a potential therapeutic target for NASH.


Subject(s)
Liver Cirrhosis/pathology , Non-alcoholic Fatty Liver Disease/pathology , Sirtuins/deficiency , Smad3 Protein/metabolism , Acetylation , Adult , Aged , Animals , Cell Line , Female , Gene Expression Regulation , Gene Knockdown Techniques , Hepatic Stellate Cells/pathology , Humans , Liver/cytology , Liver/pathology , Liver Cirrhosis/genetics , Male , Mice , Mice, Knockout , Middle Aged , Mutation , Non-alcoholic Fatty Liver Disease/genetics , Primary Cell Culture , Sirtuins/genetics , Smad3 Protein/genetics , Transcriptional Activation , Transforming Growth Factor beta1/metabolism
15.
Aging Cell ; 19(3): e13104, 2020 03.
Article in English | MEDLINE | ID: mdl-31967391

ABSTRACT

Mammalian sirtuin 6 (SIRT6) is involved in the regulation of many essential processes, especially metabolic homeostasis. SIRT6 knockout mice undergo premature aging and die at age ~4 weeks. Severe glycometabolic disorders have been found in SIRT6 knockout mice, and whether a dietary intervention can rescue SIRT6 knockout mice remains unknown. In our study, we found that at the same calorie intake, a high-fat diet dramatically increased the lifespan of SIRT6 knockout mice to 26 weeks (males) and 37 weeks (females), reversed multi-organ atrophy, and reduced body weight, hypoglycemia, and premature aging. Furthermore, the high-fat diet partially but significantly normalized the global gene expression profile in SIRT6 knockout mice. Regarding the mechanism, excessive glucose uptake and glycolysis induced by the SIRT6 deficiency were attenuated in skeletal muscle through inhibition of insulin and IGF1 signaling by the high-fat diet. Similarly, fatty acids but not ketone bodies inhibited glucose uptake, glycolysis, and senescence in SIRT6 knockout fibroblasts, whereas PI3K inhibition antagonized the effects of a high-fatty-acid medium in vitro. Overall, the high-fat diet dramatically reverses numerous consequences of SIRT6 deficiency through modulation of insulin and IGF1 signaling, providing a new basis for elucidation of SIRT6 and fatty-acid functions and supporting novel therapeutic approaches against metabolic disorders and aging-related diseases.


Subject(s)
Aging, Premature/metabolism , Diet, High-Fat , Insulin-Like Growth Factor I/metabolism , Insulin/metabolism , Metabolic Diseases/metabolism , Signal Transduction/genetics , Sirtuins/deficiency , Aging, Premature/genetics , Animals , Body Weight , Fatty Acids/metabolism , Female , Glucose/metabolism , Glycolysis , Longevity , Male , Metabolic Diseases/genetics , Mice , Mice, Knockout , Sirtuins/genetics
16.
J Am Soc Nephrol ; 30(12): 2384-2398, 2019 12.
Article in English | MEDLINE | ID: mdl-31575700

ABSTRACT

BACKGROUND: The primary site of damage during AKI, proximal tubular epithelial cells, are highly metabolically active, relying on fatty acids to meet their energy demands. These cells are rich in mitochondria and peroxisomes, the two organelles that mediate fatty acid oxidation. Emerging evidence shows that both fatty acid pathways are regulated by reversible posttranslational modifications, particularly by lysine acylation. Sirtuin 5 (Sirt5), which localizes to both mitochondria and peroxisomes, reverses post-translational lysine acylation on several enzymes involved in fatty acid oxidation. However, the role of the Sirt5 in regulating kidney energy metabolism has yet to be determined. METHODS: We subjected male Sirt5-deficient mice (either +/- or -/-) and wild-type controls, as well as isolated proximal tubule cells, to two different AKI models (ischemia-induced or cisplatin-induced AKI). We assessed kidney function and injury with standard techniques and measured fatty acid oxidation by the catabolism of 14C-labeled palmitate to 14CO2. RESULTS: Sirt5 was highly expressed in proximal tubular epithelial cells. At baseline, Sirt5 knockout (Sirt5-/- ) mice had modestly decreased mitochondrial function but significantly increased fatty acid oxidation, which was localized to the peroxisome. Although no overt kidney phenotype was observed in Sirt5-/- mice, Sirt5-/- mice had significantly improved kidney function and less tissue damage compared with controls after either ischemia-induced or cisplatin-induced AKI. This coincided with higher peroxisomal fatty acid oxidation compared with mitochondria fatty acid oxidation in the Sirt5-/- proximal tubular epithelial cells. CONCLUSIONS: Our findings indicate that Sirt5 regulates the balance of mitochondrial versus peroxisomal fatty acid oxidation in proximal tubular epithelial cells to protect against injury in AKI. This novel mechanism might be leveraged for developing AKI therapies.


Subject(s)
Acute Kidney Injury/metabolism , Fatty Acids/metabolism , Kidney Tubules, Proximal/metabolism , Mitochondria/metabolism , Peroxisomes/metabolism , Sirtuins/physiology , Acute Kidney Injury/etiology , Acute Kidney Injury/pathology , Animals , Cisplatin/toxicity , Kidney/blood supply , Male , Mice , Mice, Knockout , Oxidation-Reduction , Reperfusion Injury/metabolism , Reperfusion Injury/pathology , Sirtuins/deficiency , Sirtuins/genetics
17.
Nucleic Acids Res ; 47(15): 7870-7885, 2019 09 05.
Article in English | MEDLINE | ID: mdl-31226208

ABSTRACT

Long interspersed elements-1 (LINE-1, L1) are retrotransposons that hold the capacity of self-propagation in the genome with potential mutagenic outcomes. How somatic cells restrict L1 activity and how this process becomes dysfunctional during aging and in cancer cells is poorly understood. L1s are enriched at lamin-associated domains, heterochromatic regions of the nuclear periphery. Whether this association is necessary for their repression has been elusive. Here we show that the sirtuin family member SIRT7 participates in the epigenetic transcriptional repression of L1 genome-wide in both mouse and human cells. SIRT7 depletion leads to increased L1 expression and retrotransposition. Mechanistically, we identify a novel interplay between SIRT7 and Lamin A/C in L1 repression. Our results demonstrate that SIRT7-mediated H3K18 deacetylation regulates L1 expression and promotes L1 association with elements of the nuclear lamina. The failure of such activity might contribute to the observed genome instability and compromised viability in SIRT7 knockout mice. Overall, our results reveal a novel function of SIRT7 on chromatin organization by mediating the anchoring of L1 to the nuclear envelope, and a new functional link of the nuclear lamina with transcriptional repression.


Subject(s)
Genome , Lamin Type A/genetics , Long Interspersed Nucleotide Elements , Sirtuins/genetics , Transcription, Genetic , Animals , Cell Line , Cell Line, Tumor , Chromatin Immunoprecipitation , Epigenesis, Genetic , Fibroblasts/cytology , Fibroblasts/metabolism , Heterochromatin/chemistry , Heterochromatin/metabolism , Histones/genetics , Histones/metabolism , Humans , K562 Cells , Lamin Type A/metabolism , Liver/cytology , Liver/metabolism , Male , Mice , Mice, Knockout , Myocardium/cytology , Myocardium/metabolism , Nuclear Lamina/metabolism , Nuclear Lamina/ultrastructure , Sirtuins/deficiency , Sirtuins/metabolism , Testis/cytology , Testis/metabolism
18.
Nucleic Acids Res ; 47(15): 7914-7928, 2019 09 05.
Article in English | MEDLINE | ID: mdl-31216030

ABSTRACT

SIRT6 is critical for activating transcription of Nuclear factor (erythroid-derived 2)-like 2 (NRF2) responsive genes during oxidative stress. However, while the mechanism of SIRT6-mediated silencing is well understood, the mechanism of SIRT6-mediated transcriptional activation is unknown. Here, we employed SIRT6 separation of function mutants to reveal that SIRT6 mono-ADP-ribosylation activity is required for transcriptional activation. We demonstrate that SIRT6 mono-ADP-ribosylation of BAF170, a subunit of BAF chromatin remodeling complex, is critical for activation of a subset of NRF2 responsive genes upon oxidative stress. We show that SIRT6 recruits BAF170 to enhancer region of the Heme oxygenase-1 locus and promotes recruitment of RNA polymerase II. Furthermore, SIRT6 mediates the formation of the active chromatin 10-kb loop at the HO-1 locus, which is absent in SIRT6 deficient tissue. These results provide a novel mechanism for SIRT6-mediated transcriptional activation, where SIRT6 mono-ADP-ribosylates and recruits chromatin remodeling proteins to mediate the formation of active chromatin loop.


Subject(s)
Chromatin/metabolism , Chromosomal Proteins, Non-Histone/genetics , Heme Oxygenase-1/genetics , Membrane Proteins/genetics , NF-E2-Related Factor 2/genetics , Sirtuins/genetics , Transcription, Genetic , ADP-Ribosylation , Animals , Cell Line , Chromatin/chemistry , Chromatin/drug effects , Chromosomal Proteins, Non-Histone/metabolism , DNA-Binding Proteins , Embryo, Mammalian , Enhancer Elements, Genetic , Fibroblasts/cytology , Fibroblasts/drug effects , Fibroblasts/metabolism , Gene Expression Regulation , Heme Oxygenase-1/metabolism , Membrane Proteins/metabolism , Mice , Mice, Knockout , NF-E2-Related Factor 2/metabolism , Oxidative Stress , Paraquat/pharmacology , Protein Binding , RNA Polymerase II/genetics , RNA Polymerase II/metabolism , Signal Transduction , Sirtuins/deficiency , Transcription Factors
19.
Cell Metab ; 29(4): 871-885.e5, 2019 04 02.
Article in English | MEDLINE | ID: mdl-30853213

ABSTRACT

Mice deficient for SIRT6 exhibit a severely shortened lifespan, growth retardation, and highly elevated LINE1 (L1) activity. Here we report that SIRT6-deficient cells and tissues accumulate abundant cytoplasmic L1 cDNA, which triggers strong type I interferon response via activation of cGAS. Remarkably, nucleoside reverse-transcriptase inhibitors (NRTIs), which inhibit L1 retrotransposition, significantly improved health and lifespan of SIRT6 knockout mice and completely rescued type I interferon response. In tissue culture, inhibition of L1 with siRNA or NRTIs abrogated type I interferon response, in addition to a significant reduction of DNA damage markers. These results indicate that L1 activation contributes to the pathologies of SIRT6 knockout mice. Similarly, L1 transcription, cytoplasmic cDNA copy number, and type I interferons were elevated in the wild-type aged mice. As sterile inflammation is a hallmark of aging, we propose that modulating L1 activity may be an important strategy for attenuating age-related pathologies.


Subject(s)
Inflammation/metabolism , RNA-Binding Proteins/metabolism , Sirtuins/metabolism , Age Factors , Animals , Dideoxynucleotides/administration & dosage , Dideoxynucleotides/pharmacology , Female , Male , Mice , Mice, Inbred Strains , Mice, Knockout , RNA-Binding Proteins/antagonists & inhibitors , Sirtuins/deficiency , Stavudine/administration & dosage , Stavudine/pharmacology , Thymine Nucleotides/administration & dosage , Thymine Nucleotides/pharmacology , Zidovudine/administration & dosage , Zidovudine/analogs & derivatives , Zidovudine/pharmacology
20.
Circ Res ; 124(10): 1448-1461, 2019 05 10.
Article in English | MEDLINE | ID: mdl-30894089

ABSTRACT

RATIONALE: Endothelial dysfunction is an important determinant risk factor for the development of hypertension and its complications. Thus, identification of potential therapeutic targets for preventing endothelial dysfunction has major clinical importance. Emerging evidence indicates that epigenetic modifications are closely associated with the regulation of endothelial function. Among them, HDAC (histone deacetylase)-mediated epigenetic processes in vascular homeostasis and cardiovascular disease have attracted much attention. SIRT6 (sirtuin 6) is one member of SIRTs (class III HDAC) that are highly conserved NAD+-dependent deacetylases. OBJECTIVE: This study was designed to elucidate the role of SIRT6 in the pathogenesis of hypertension, discover the new targets of SIRT6, and explore related mechanisms on the regulation of endothelial function. METHODS AND RESULTS: The levels of endothelial SIRT6 were significantly reduced in 2 independent hypertension models: desoxycorticosterone acetate/salt-induced and Ang II (angiotensin II)-induced hypertensive mice. Utilizing genetically engineered endothelial-specific SIRT6 knockout (Cre+/SIRT6fl/fl) mice, we found that endothelial-specific deletion of SIRT6 significantly enhanced blood pressure, exacerbated endothelial dysfunction and cardiorenal injury in experimental hypertension. Functionally, SIRT6 has pleiotropic protective actions in endothelial cells, which include promoting endothelium-dependent vasodilatation and vascular NO bioavailability, reducing cellular permeability, ameliorating endothelial senescence and apoptosis, and facilitating autophagy. Mechanistically, SIRT6 induced the expression of GATA5 (GATA-binding protein 5), a novel regulator of blood pressure, through inhibiting Nkx3.2 (NK3 homeobox 2) transcription by deacetylating histone H3K9 (histone H3 lysine 9), thereby regulating GATA5-mediated signaling pathways to prevent endothelial injury. Finally, we provide direct evidence for the therapeutic potential of SIRT6 in desoxycorticosterone acetate/salt-induced hypertensive mice by overexpression of SIRT6 in vivo. CONCLUSIONS: This study for the first time demonstrates that SIRT6 prevents hypertension and its complications by maintaining endothelial function. Pharmacological targeting of SIRT6 may be an innovative therapeutic strategy for treating patients with hypertension.


Subject(s)
Endothelium, Vascular/physiology , Hypertension/prevention & control , Sirtuins/physiology , Acetylation , Angiotensin II , Animals , Desoxycorticosterone Acetate , Endothelium, Vascular/injuries , Epigenesis, Genetic , GATA5 Transcription Factor/metabolism , Histone Deacetylases , Histones/metabolism , Homeodomain Proteins/metabolism , Hypertension/chemically induced , Hypertension, Renal/metabolism , Kidney/injuries , Mice , Mice, Knockout , Nephritis/metabolism , Sirtuins/blood , Sirtuins/deficiency , Sirtuins/genetics , Sodium Chloride , Transcription Factors/metabolism , Vasoconstrictor Agents , Vasodilation
SELECTION OF CITATIONS
SEARCH DETAIL