Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 420
Filter
1.
Elife ; 122023 10 18.
Article in English | MEDLINE | ID: mdl-37850626

ABSTRACT

Enterovirus D68 (EV-D68) is a re-emerging enterovirus that causes acute respiratory illness in infants and has recently been linked to Acute Flaccid Myelitis. Here, we show that the histone deacetylase, SIRT-1, is essential for autophagy and EV-D68 infection. Knockdown of SIRT-1 inhibits autophagy and reduces EV-D68 extracellular titers. The proviral activity of SIRT-1 does not require its deacetylase activity or functional autophagy. SIRT-1's proviral activity is, we demonstrate, mediated through the repression of endoplasmic reticulum stress (ER stress). Inducing ER stress through thapsigargin treatment or SERCA2A knockdown in SIRT-1 knockdown cells had no additional effect on EV-D68 extracellular titers. Knockdown of SIRT-1 also decreases poliovirus and SARS-CoV-2 titers but not coxsackievirus B3. In non-lytic conditions, EV-D68 is primarily released in an enveloped form, and SIRT-1 is required for this process. Our data show that SIRT-1, through its translocation to the cytosol, is critical to promote the release of enveloped EV-D68 viral particles.


Subject(s)
Enterovirus D, Human , Enterovirus Infections , Sirtuin 1 , Virus Activation , Humans , COVID-19 , Enterovirus/genetics , Enterovirus/physiology , Enterovirus D, Human/genetics , Enterovirus D, Human/physiology , Enterovirus Infections/genetics , Enterovirus Infections/physiopathology , Neuromuscular Diseases , Proviruses , SARS-CoV-2 , Viral Envelope/metabolism , Viral Envelope/physiology , Virus Activation/genetics , Virus Activation/physiology , Sirtuin 1/genetics , Sirtuin 1/physiology
2.
Int J Cancer ; 150(7): 1198-1211, 2022 04 01.
Article in English | MEDLINE | ID: mdl-34751438

ABSTRACT

Angiogenesis plays an important role during tumor growth and metastasis. We could previously show that Type I interferon (IFN)-deficient tumor-associated neutrophils (TANs) show strong pro-angiogenic activity, and stimulate tumor angiogenesis and growth. However, the exact mechanism responsible for their pro-angiogenic shift is not clear. Here, we set out to delineate the molecular mechanism and factors regulating pro-angiogenic properties of neutrophils in the context of Type I IFN availability. We demonstrate that neutrophils from IFN-deficient (Ifnar1-/- ) mice efficiently release pro-angiogenic factors, such as VEGF, MMP9 or BV8, and thus significantly support the vascular normalization of tumors by increasing the maturation of perivascular cells. Mechanistically, we could show here that the expression of pro-angiogenic factors in neutrophils is controlled by the transcription factor forkhead box protein O3a (FOXO3a), which activity depends on its post-translational modifications, such as deacetylation or phosphorylation. In TANs isolated from Ifnar1-/- mice, we observe significantly elevated SIRT1, resulting in SIRT1-mediated deacetylation of FOXO3a, its nuclear retention and activation. Activated FOXO3a supports in turn the transcription of pro-angiogenic genes in TANs. In the absence of SIRT1, or after its inhibition in neutrophils, elevated kinase MEK/ERK and PI3K/AKT activity is observed, leading to FOXO3a phosphorylation, cytoplasmic transfer and inactivation. In summary, we have found that FOXO3a is a key transcription factor controlling the angiogenic switch of neutrophils. Post-translational FOXO3a modifications regulate its transcriptional activity and, as a result, the expression of pro-angiogenic factors supporting development of vascular network in growing tumors. Therefore, targeting FOXO3a activity could provide a novel strategy of antiangiogenic targeted therapy for cancer.


Subject(s)
Forkhead Box Protein O3/metabolism , Interferon Type I/physiology , Neoplasms/blood supply , Neovascularization, Pathologic/etiology , Neutrophils/physiology , Sirtuin 1/physiology , Acetylation , Animals , Cell Line, Tumor , Humans , Mice , Mice, Inbred C57BL , Protein Processing, Post-Translational
3.
Front Endocrinol (Lausanne) ; 12: 756909, 2021.
Article in English | MEDLINE | ID: mdl-34867800

ABSTRACT

Introduction: Using transgenic collagen type II-specific Sirt1 knockout (CKO) mice we studied the role of Sirt1 in nutritional induced catch up growth (CUG) and we found that these mice have a less organized growth plate and reduced efficiency of CUG. In addition, we noted that they weigh more than control (CTL) mice. Studying the reason for the increased weigh, we found differences in activity and brain function. Methods: Several tests for behavior and activity were used: open field; elevated plus maze, Morris water maze, and home cage running wheels. The level of Glu- osteocalcin, known to connect bone and brain function, was measured by Elisa; brain Sirt1 was analyzed by western blot. Results: We found that CKO mice had increased anxiety, with less spatial memory, learning capabilities and reduced activity in their home cages. No significant differences were found between CKO and CTL mice in Glu- osteocalcin levels; nor in the level of brain SIRT1. Discussion/Conclusion: Using transgenic collagen type II-specific Sirt1 knockout (CKO) mice we found a close connection between linear growth and brain function. Using a collagen type II derived system we affected a central regulatory mechanism leading to hypo activity, increased anxiety, and slower learning, without affecting circadian period. As children with idiopathic short stature are more likely to have lower IQ, with substantial deficits in working memory than healthy controls, the results of the current study suggest that SIRT1 may be the underlying factor connecting growth and brain function.


Subject(s)
Anxiety , Locomotion , Maze Learning , Musculoskeletal Development , Sirtuin 1/physiology , Animals , Cartilage, Articular/physiology , Cognition , Male , Mice, Knockout
4.
Cell Rep ; 37(8): 110038, 2021 11 23.
Article in English | MEDLINE | ID: mdl-34818543

ABSTRACT

Cellular senescence is associated with pleiotropic physiopathological processes, including aging and age-related diseases. The persistent DNA damage is a major stress leading to senescence, but the underlying molecular link remains elusive. Here, we identify La Ribonucleoprotein 7 (LARP7), a 7SK RNA binding protein, as an aging antagonist. DNA damage-mediated Ataxia Telangiectasia Mutated (ATM) activation triggers the extracellular shuttling and downregulation of LARP7, which dampens SIRT1 deacetylase activity, enhances p53 and NF-κB (p65) transcriptional activity by augmenting their acetylation, and thereby accelerates cellular senescence. Deletion of LARP7 leads to senescent cell accumulation and premature aging in rodent model. Furthermore, we show this ATM-LARP7-SIRT1-p53/p65 senescence axis is active in vascular senescence and atherogenesis, and preventing its activation substantially alleviates senescence and atherogenesis. Together, this study identifies LARP7 as a gatekeeper of senescence, and the altered ATM-LARP7-SIRT1-p53/p65 pathway plays an important role in DNA damage response (DDR)-mediated cellular senescence and atherosclerosis.


Subject(s)
Cellular Senescence/physiology , Ribonucleoproteins/metabolism , Sirtuin 1/metabolism , Acetylation , Aging/metabolism , Aging/physiology , Animals , Ataxia Telangiectasia Mutated Proteins/metabolism , Cell Line, Tumor , DNA Damage , Female , Humans , Male , Mice , Mice, Inbred C57BL , Oxidative Stress/genetics , RNA-Binding Proteins/metabolism , RNA-Binding Proteins/physiology , Ribonucleoproteins/physiology , Signal Transduction , Sirtuin 1/physiology , Transcription Factor RelA/metabolism , Tumor Suppressor Protein p53/metabolism
5.
J Pharmacol Sci ; 147(4): 376-385, 2021 Dec.
Article in English | MEDLINE | ID: mdl-34663520

ABSTRACT

Systemic sirtuin 1 (SIRT1) activation alleviates muscle wasting and improves muscle function by downregulation of myotropic and proteolytic markers. In this study, we evaluated the effects of the intestinal Sirt1 deletion on the dysregulated gutmuscle axis in cirrhotic mice. Cirrhosis-related muscle wasting was induced by common bile duct ligated (BDL) in either wild-type (WT) or intestine-specific Sirt1-deleted (Sirt1IEC-KO) mice, including WT-BDL, WT-sham, Sirt1IEC-KO-BDL and Sirt1IEC-KO-sham mice. Compared with WT-BDL mice, Sirt1IEC-KO-BDL mice showed worsened low lean mass, exacerbated muscle wasting, increased expression of myotropic markers, increased muscular protein degradation, and decreased expression of myogenic markers through aggravation of intestinal inflammation (as evidenced by increased fecal calprotectin/lipocalin-2 levels, increased intestinal macrophage infiltration, and increased intestinal TNFα/IL-6 levels), decrease in abundance of short-chain fatty acid (SCFA)-producing bacteria, decrease in levels of intestinal SCFAs (with anti-inflammatory effects), and downregulation of SCFA receptor GPR43. In biliary cirrhotic mice, a decrease in the abundance of SCFA-producing bacteria and an increase in the levels of intestinal/muscular inflammatory markers are involved in the pathogenesis of dysregulated gut-muscle axis-related muscle wasting, and intestinal deletion of Sirt1 exacerbated these changes.


Subject(s)
Fatty Acids, Volatile/metabolism , Gene Deletion , Intestines/metabolism , Liver Cirrhosis/complications , Sarcopenia/genetics , Sirtuin 1/metabolism , Sirtuin 1/physiology , Animals , Disease Models, Animal , Disease Progression , Gastrointestinal Microbiome/physiology , Inflammation , Male , Mice, Inbred C57BL , Muscle Proteins/metabolism , Muscles/metabolism , Sarcopenia/etiology , Sarcopenia/metabolism
6.
Mol Neurobiol ; 58(11): 5890-5906, 2021 Nov.
Article in English | MEDLINE | ID: mdl-34415486

ABSTRACT

Alzheimer's disease (AD) is associated with a very large burden on global healthcare systems. Thus, it is imperative to find effective treatments of the disease. One feature of AD is the accumulation of neurotoxic ß-amyloid peptide (Aß). Aß induces multiple pathological processes that are deleterious to nerve cells. Despite the development of medications that target the reduction of Aß to treat AD, none has proven to be effective to date. Non-pharmacological interventions, such as physical exercise, are also being studied. The benefits of exercise on AD are widely recognized. Experimental and clinical studies have been performed to verify the role that exercise plays in reducing Aß deposition to alleviate AD. This paper reviewed the various mechanisms involved in the exercise-induced reduction of Aß, including the regulation of amyloid precursor protein cleaved proteases, the glymphatic system, brain-blood transport proteins, degrading enzymes and autophagy, which is beneficial to promote exercise therapy as a means of prevention and treatment of AD and indicates that exercise may provide new therapeutic targets for the treatment of AD.


Subject(s)
Amyloid beta-Peptides/metabolism , Amyloid beta-Protein Precursor/metabolism , Exercise , Alzheimer Disease/metabolism , Alzheimer Disease/therapy , Animals , Autophagy , Blood-Brain Barrier , Brain-Derived Neurotrophic Factor/physiology , Carrier Proteins/metabolism , Disease Models, Animal , Exercise/physiology , Fibronectins/physiology , Glymphatic System , Humans , Membrane Microdomains/physiology , Mice , Nerve Tissue Proteins/physiology , Neurodegenerative Diseases/physiopathology , Neurodegenerative Diseases/prevention & control , Neuroinflammatory Diseases/physiopathology , Peptide Hydrolases/metabolism , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/physiology , Physical Conditioning, Animal , Proteolysis , Signal Transduction/physiology , Sirtuin 1/physiology , Unfolded Protein Response/physiology
7.
Radiat Res ; 196(6): 647-657, 2021 12 01.
Article in English | MEDLINE | ID: mdl-34459925

ABSTRACT

Radiotherapy is an important method for the treatment of malignant tumors. It can directly or indirectly lead to the formation of free radicals and DNA damage, resulting in a series of biological effects, including tumor cell death and normal tissue damage. These radiation effects are typically accompanied by the abnormal expression of sirtuin 1 (Sirt1), which deacetylates histones and non-histones. These Sirt1 substrates, including transcription factors and some catalytic enzymes, play a crucial role in anti-oxidative stress, DNA damage repair, autophagy regulation, anti-senescence, and apoptosis, which are closely related to triggering cell defense and survival in radiation-induced damage. In this article, we review the mechanisms underlying cellular responses to ionizing radiation and the role of Sirt1 in the process, with the aim of providing a theoretical basis for protection against radiation by Sirt1 as well as novel targets for developing radioprotective agents.


Subject(s)
Neoplasms/radiotherapy , Radiation Injuries/prevention & control , Sirtuin 1/physiology , Autophagy , DNA Damage , DNA Repair , Humans , Mitochondria/radiation effects , Oxidative Stress/physiology , Oxidative Stress/radiation effects , Radiotherapy/adverse effects
8.
Mol Brain ; 14(1): 128, 2021 08 20.
Article in English | MEDLINE | ID: mdl-34416891

ABSTRACT

Machado-Joseph disease (MJD, also known as spinocerebellar ataxia type 3) is a fatal neurodegenerative disease that impairs control and coordination of movement. Here we tested whether treatment with the histone deacetylase inhibitor sodium valproate (valproate) prevented a movement phenotype that develops in larvae of a transgenic zebrafish model of the disease. We found that treatment with valproate improved the swimming of the MJD zebrafish, affected levels of acetylated histones 3 and 4, but also increased expression of polyglutamine expanded human ataxin-3. Proteomic analysis of protein lysates generated from the treated and untreated MJD zebrafish also predicted that valproate treatment had activated the sirtuin longevity signaling pathway and this was confirmed by findings of increased SIRT1 protein levels and sirtuin activity in valproate treated MJD zebrafish and HEK293 cells expressing ataxin-3 84Q, respectively. Treatment with resveratrol (another compound known to activate the sirtuin pathway), also improved swimming in the MJD zebrafish. Co-treatment with valproate alongside EX527, a SIRT1 activity inhibitor, prevented induction of autophagy by valproate and the beneficial effects of valproate on the movement in the MJD zebrafish, supporting that they were both dependent on sirtuin activity. These findings provide the first evidence of sodium valproate inducing activation of the sirtuin pathway. Further, they indicate that drugs that target the sirtuin pathway, including sodium valproate and resveratrol, warrant further investigation for the treatment of MJD and related neurodegenerative diseases.


Subject(s)
Histone Deacetylase Inhibitors/therapeutic use , Machado-Joseph Disease/drug therapy , Sirtuins/drug effects , Valproic Acid/therapeutic use , Acetylation , Animals , Animals, Genetically Modified , Ataxin-3/antagonists & inhibitors , Ataxin-3/genetics , Ataxin-3/metabolism , Autophagy/drug effects , Carbazoles/pharmacology , Carbazoles/therapeutic use , Disease Models, Animal , Drug Evaluation, Preclinical , Drug Synergism , Genes, Reporter , HEK293 Cells , Histone Deacetylase Inhibitors/pharmacology , Histones/metabolism , Humans , Peptides/genetics , Protein Processing, Post-Translational , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Repressor Proteins/genetics , Repressor Proteins/metabolism , Resveratrol/pharmacology , Resveratrol/therapeutic use , Signal Transduction , Sirtuin 1/physiology , Sirtuins/physiology , Swimming , Trinucleotide Repeat Expansion , Valproic Acid/pharmacology , Zebrafish , Zebrafish Proteins/antagonists & inhibitors , Zebrafish Proteins/genetics , Zebrafish Proteins/metabolism
9.
Mediators Inflamm ; 2021: 6639252, 2021.
Article in English | MEDLINE | ID: mdl-33927570

ABSTRACT

Oxidized low-density lipoprotein (oxLDL) induced a foam-cell-like phenotype of the vascular smooth muscle cells (VSMCs), leading to the inflammatory responses incorporating Toll-like receptor- (Tlr-) mediated cellular alterations. However, the role of Tlr4 in foam cell formation and underlying molecular pathways has not been comprehensively elucidated. To further investigate the mechanism, VSMCs were incubated with different doses of oxLDL, and then, the lipid, reactive oxygen species (ROS) accumulation, Tlr family genes, and the foam cell phenotype were explored. We observed that oxLDL induced foam cell-like phenotype in VSMCs and led to lipid and ROS accumulation in a dose-dependent manner. Furthermore, in the Tlr family, Tlr4 demonstrated the strongest upregulation under oxLDL stimulation. Simultaneously, oxLDL induced activation of Src, higher expression of Nox2, and lower expression of Mnsod, Sirt1, and Sirt3. By interfering the TLR4 expression, the phenotype alteration, lipid accumulation in VSMCs, and Src kinase activation induced by oxLDL were abolished. After interfering Src activation, the oxLDL-induced lipid accumulation and foam cell phenotype in VSMCs were also alleviated. Furthermore, the ROS accumulation, upregulated Nox2 expression, downregulated Sirt1, Sirt3, and Mnsod expression in VSMCs under oxLDL stimulation were also relieved after the knockdown of Tlr4. Additionally, overexpression of Sirt1 and Sirt3 ameliorated the ROS accumulation and foam cell-like marker expression in VSMCs. These results demonstrated that beyond its familiar role in regulating inflammation response, Tlr4 is a critical regulator in oxLDL-induced foam cell formation in VSMCs via regulating Src kinase activation as well as Sirt1 and Sirt3 expression.


Subject(s)
Foam Cells/drug effects , Lipoproteins, LDL/toxicity , Muscle, Smooth, Vascular/drug effects , Sirtuin 1/physiology , Sirtuin 3/physiology , Toll-Like Receptor 4/physiology , src-Family Kinases/physiology , Animals , Lipid Metabolism/drug effects , Mice , Mice, Inbred C57BL , Muscle, Smooth, Vascular/pathology , Myocytes, Smooth Muscle/drug effects , Myocytes, Smooth Muscle/pathology , Oxidative Stress/drug effects
10.
Mol Nutr Food Res ; 65(9): e2000672, 2021 05.
Article in English | MEDLINE | ID: mdl-33686759

ABSTRACT

INTRODUCTION: Calorie restriction (CR) exerts multiple effects on health, including the amelioration of systemic insulin resistance. Although the precise mechanisms by which CR improves glucose homeostasis remain poorly defined, SIRT1 has been suggested to act as a central mediator of the cellular responses to CR. Here, we aim at identifying the mechanisms by which CR and SIRT1 modulate white adipose tissue (WAT) function, a key tissue in the control of glucose homeostasis. MATERIAL AND METHODS: A gene expression profiling study using DNA microarrays is conducted in WAT of control and SIRT1 transgenic mice fed ad libitum (AL) and mice subjected to 40% CR. RESULTS: Gene expression profiling reveals a relatively low degree of overlap between the transcriptional programs regulated by SIRT1 and CR. Gene networks related to extracellular matrix appear commonly downregulated by SIRT1/CR, whereas mitochondrial biogenesis is enhanced exclusively by CR. Moreover, WAT inflammation is reduced by CR and SIRT1, although their anti-inflammatory effects appeared to be achieved by regulating different gene networks related to the immune system. CONCLUDING REMARKS: In WAT, SIRT1 does not mediate most of the effects of CR on gene expression. Still, gene networks differentially regulated by SIRT1 and CR converge to reduce WAT inflammation.


Subject(s)
Adipose Tissue, White/metabolism , Caloric Restriction , Sirtuin 1/physiology , Transcriptome , Animals , Extracellular Matrix Proteins/metabolism , Glucose/metabolism , Inflammation/prevention & control , Male , Mice
11.
Cancer Lett ; 505: 24-36, 2021 05 01.
Article in English | MEDLINE | ID: mdl-33617947

ABSTRACT

The NAD+-dependent deacetylase, Sirtuin 1 (SIRT1) is involved in prostate cancer pathogenesis. However, the actual contribution is unclear as some reports propose a protective role while others suggest it is harmful. We provide evidence for a contextual role for SIRT1 in prostate cancer. Our data show that (i) mice orthotopically implanted with SIRT1-silenced LNCaP cells produced smaller tumors; (ii) SIRT1 suppression mimicked AR inhibitory effects in hormone responsive LNCaP cells; and (iii) caused significant reduction in gene signatures associated with E2F and MYC targets in AR-null PC-3 and E2F and mTORC1 signaling in castrate-resistant ARv7 positive 22Rv1 cells. Our findings further show increased nuclear SIRT1 (nSIRT1) protein under androgen-depleted relative to androgen-replete conditions in prostate cancer cell lines. Silencing SIRT1 resulted in decreased recruitment of AR to PSA enhancer selectively under androgen-deprivation conditions. Prostate cancer outcome data show that patients with higher levels of nSIRT1 progress to advanced disease relative to patients with low nSIRT1 levels. Collectively, we demonstrate that lowering SIRT1 levels potentially provides new avenues to effectively prevent prostate cancer recurrence.


Subject(s)
Prostatic Neoplasms/pathology , Receptors, Androgen/physiology , Sirtuin 1/physiology , Aged , Animals , Cell Line, Tumor , Cell Survival , Disease Progression , Humans , Male , Mice , Middle Aged , Orchiectomy , Signal Transduction/physiology
12.
Can J Physiol Pharmacol ; 99(9): 875-884, 2021 Sep.
Article in English | MEDLINE | ID: mdl-33517853

ABSTRACT

Metformin has been demonstrated to be beneficial for the treatment of an impaired myocardium as a result of ischemia/reperfusion (I/R) injury, and miR-34a may be involved in this process. The aim of the present study was to determine the mechanisms by which metformin attenuated myocardial I/R injury-induced apoptosis. In the in vivo I/R model using Sprague-Dawley rats, metformin reduced the area of damaged myocardium and serum creatine MB isoform (CKMB) activity resulting in protection of the myocardium. Metformin also reduced apoptosis and the expression of apoptosis associated proteins, including caspase 3 and cleaved caspase, and decreased the expression of miR-34a, which is upregulated during I/R injury, which in turn resulted in corresponding changes in expression of Bcl-2, a direct target of miR-34a both in vitro and in vivo. To further examine the role of miR-34a in this process, H9C2 cells were transfected by a miR-34a mimic and inhibitor. Overexpression of miR-34a increased apoptosis in H9C2 cells induced by oxygen-glucose deprivation/recovery and knockdown of miR-34a expression-reduced apoptosis under the same conditions. Therefore, the effect of metformin on miR-34a in vitro were assessed. Metformin decreased the deacetylation activity of silent information regulator 1 resulting in reduced Ac-p53 levels, which reduced the levels of pri-miR-34a, and thus in turn reduced miR-34a levels. To confirm these results clinically, 90 patients with ST-segment elevation myocardial infarction following percutaneous coronary intervention were recruited. Patients who took metformin regularly before infarction had lower miR-34a levels and lower serum CKMB activity. Metformin also improved the sum ST-segment recovery following I/R injury. In conclusion, metformin may be helpful in the treatment of myocardial I/R.


Subject(s)
Apoptosis/drug effects , Metformin/pharmacology , MicroRNAs/physiology , Myocardial Reperfusion Injury/drug therapy , Myocytes, Cardiac/drug effects , Sirtuin 1/physiology , Tumor Suppressor Protein p53/physiology , AMP-Activated Protein Kinases/physiology , Adult , Aged , Animals , Creatine Kinase, MB Form/blood , Down-Regulation , Female , Humans , Male , MicroRNAs/analysis , Middle Aged , Myocardial Reperfusion Injury/pathology , Myocytes, Cardiac/pathology , Percutaneous Coronary Intervention , Rats , Rats, Sprague-Dawley , ST Elevation Myocardial Infarction/drug therapy
13.
Can J Physiol Pharmacol ; 99(9): 900-909, 2021 Sep.
Article in English | MEDLINE | ID: mdl-33529089

ABSTRACT

Vascular endothelial cell senescence is a leading cause of age-associated diseases and cardiovascular diseases. Interventions and therapies targeting endothelial cell senescence and dysfunction would have important clinical implications. This study evaluated the effect of 10 resveratrol analogues, including pterostilbene (Pts) and its derivatives, against endothelial senescence and dysfunction. All the tested compounds at the concentrations from 10-9 M to 10-6 M did not show cytotoxicity in endothelial cells by MTT assay. Among the 10 resveratrol analogues, Pts and Pts nicotinate attenuated the expression of senescence-associated ß-galactosidase, downregulated p21 and p53, and increased the production of nitric oxide (NO) in both angiotensin II - and hydrogen peroxide - induced endothelial senescence models. In addition, Pts and Pts nicotinate elicited endothelium-dependent relaxations, which were attenuated in the presence of endothelial NO synthase (eNOS) inhibitor L-NAME or sirtuin 1 (SIRT1) inhibitor sirtinol. Pts and Pts nicotinate did not alter SIRT1 expression but enhanced its activity. Both Pts and Pts nicotinate have high binding activities with SIRT1, according to surface plasmon resonance results and the molecular docking analysis. Inhibition of SIRT1 by sirtinol reversed the anti-senescent effects of Pts and Pts nicotinate. Moreover, Pts and Pts nicotinate shared similar ADME (absorption, distribution, metabolism, excretion) profiles and physiochemical properties. This study suggests that the Pts and Pts nicotinate ameliorate vascular endothelial senescence and elicit endothelium-dependent relaxations via activation of SIRT1. These two compounds may be potential drugs for the treatment of cardiovascular diseases related to endothelial senescence and dysfunction.


Subject(s)
Cellular Senescence/drug effects , Endothelial Cells/drug effects , Resveratrol/analogs & derivatives , Sirtuin 1/physiology , Stilbenes/pharmacology , Vasodilation/drug effects , Animals , Cells, Cultured , Endothelial Cells/physiology , Humans , Male , Niacin/analogs & derivatives , Rats , Rats, Sprague-Dawley
14.
Neuropeptides ; 87: 102134, 2021 Jun.
Article in English | MEDLINE | ID: mdl-33639357

ABSTRACT

Parkinson's disease (PD) is the second most common neurodegenerative disorder, characterized by the loss of dopaminergic neurons in the substantia nigra and the deposition of Lewy bodies. Mitochondrial dysfunction, oxidative stress, and autophagy dysfunction are involved in the pathogenesis of PD. Ghrelin is a brain-gut peptide that has been reported that protected against 1-methyl-4-phenyl-1,2,3,6- tetrahydropyran (MPTP)/MPP+-induced toxic effects. In the present work, human neuroblastoma SH-SY5Y cells were exposed to rotenone as a PD model to explore the underlying mechanism of ghrelin. We found that ghrelin inhibited rotenone-induced cytotoxicity, mitochondrial dysfunction, and apoptosis by improving cell viability, increasing the ratio of red/green of JC-1, inhibiting the production of reactive oxidative species (ROS), and regulating Bcl-2, Bax, Cytochrome c, caspase-9, and caspase-3 expression. Besides, ghrelin promoted mitophagy accompanied by up-regulating microtubule-associated protein 1 Light Chain 3B-II/I(LC3B-II/I) and Beclin1 but decreasing the expression of p62. Moreover, ghrelin promoted PINK1/Parkin mitochondrial translocation. Additionally, we investigated that ghrelin activated the AMPK/SIRT1/PGC1α pathway and pharmacological inhibition of AMPK and SIRT1 abolished the cytoprotection of ghrelin, decreased the level of mitophagy, and PINK1/Parkin mitochondrial translocation. Taken together, our findings suggested that mitophagy and AMPK/SIRT1/PGC1α pathways were related to the cytoprotection of ghrelin. These findings provided novel insights into the underlying mechanisms of ghrelin, further mechanistic studies on preclinical and clinical levels are required to be conducted with ghrelin to avail and foresee it as a potential agent in the treatment and management of PD.


Subject(s)
Ghrelin/physiology , Mitochondria/drug effects , Mitophagy/physiology , Nerve Tissue Proteins/physiology , Rotenone/toxicity , Signal Transduction/drug effects , AMP-Activated Protein Kinases/antagonists & inhibitors , AMP-Activated Protein Kinases/physiology , Apoptosis/drug effects , Cell Line, Tumor , Drug Evaluation, Preclinical , Gene Expression Regulation/drug effects , Ghrelin/pharmacology , Humans , Membrane Potential, Mitochondrial/drug effects , Mitochondria/metabolism , Nerve Tissue Proteins/antagonists & inhibitors , Neuroblastoma , Oxidative Stress/drug effects , Parkinson Disease , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/physiology , Protein Kinases/metabolism , Protein Transport/drug effects , Reactive Oxygen Species , Rotenone/antagonists & inhibitors , Sirtuin 1/antagonists & inhibitors , Sirtuin 1/physiology , Ubiquitin-Protein Ligases/metabolism , alpha-Synuclein/biosynthesis , alpha-Synuclein/genetics
15.
Int J Infect Dis ; 105: 49-53, 2021 Apr.
Article in English | MEDLINE | ID: mdl-33578018

ABSTRACT

BACKGROUND/OBJECTIVES: A dysregulated inflammatory profile plays an important role in coronavirus disease-2019 (COVID-19) pathogenesis. Moreover, the depletion of lymphocytes is typically associated with an unfavourable disease course. We studied the role and impact of p53 and deacetylase Sirtuin 1 (SIRT1) on lymph-monocyte homeostasis and their possible effect on T and B cell signalling. METHODS: Gene expression analysis and flow cytometry were performed on peripheral blood mononuclear cells (PBMC) of 35 COVID-19 patients and 10 healthy donors (HD). Inflammatory cytokines, the frequency of Annexin+ cells among CD3+ T cells and CD19+ B cell subsets were quantified. RESULTS: PBMC from COVID-19 patients had a higher p53 expression, and higher concentrations of plasma proinflammatory cytokines (IL1ß, TNF-α, IL8, and IL6) than HD. Deacetylase Sirtuin 1 (SIRT1) expression was significantly decreased in COVID-19 patients and was negatively correlated with p53 (p = 0.003 and r = -0.48). A lower expression of IL-7R and B Cell linker (BLNK), key genes for lymphocyte homeostasis and function, was observed in COVID-19 than in HD. The reduction of IgK and IgL chains was seen in lymphopenic COVID-19 patients. A significant increase in both apoptotic B and T cells were observed. Inflammatory cytokines correlated positively with p53 (IL-1ß: r = 0.5 and p = 0.05; IL-8: r = 0.5 and p = 0.05) and negatively with SIRT1 (IL1-ß: r = -0.5 and p = 0.04; TNF-α: r = -0.4 and p = 0.04). CONCLUSIONS: Collectively, our data indicate that the inflammatory environment, the dysregulated p53/SIRT1 axis and low expression of IL7R and BLNK may impact cell survival, B cell signalling and antibody production in COVID-19 patients. Further studies are required to define the functional impact of low BLNK/IL7R expression during severe acute respiratory syndrome coronavirus-2 infection.


Subject(s)
COVID-19/immunology , Homeostasis , Lymphocytes/immunology , SARS-CoV-2 , Sirtuin 1/physiology , Tumor Suppressor Protein p53/physiology , Aged , Cytokines/blood , Female , Humans , Male , Middle Aged
16.
Int Urol Nephrol ; 53(11): 2299-2310, 2021 Nov.
Article in English | MEDLINE | ID: mdl-33458788

ABSTRACT

Acute kidney injury (AKI) is a common syndrome associated with high morbidity and mortality, despite progress in medical care. Many studies have shown that there are sex differences and different role of sex hormones particularly estrogens in kidney injury. In this regard, the incidence and rate of progression of kidney diseases are higher in men compared with women. These observations suggest that female sex hormone may be renoprotective. Silent information regulator 2 homolog 1 (SIRT1) is a histone deacetylase, which is implicated in multiple biologic processes in several organisms. In the kidneys, SIRT1 inhibits renal cell apoptosis, inflammation, and fibrosis. Studies have reported a link between SIRT1 and estrogen. In addition, SIRT1 regulates ERα expression and inhibition of SIRT1 activity suppresses ERα expression. This effect leads to inhibition of estrogen-responsive gene expression. In this text, we review the role of SIRT1 in mediating the protective effects of estrogen in the onset and progression of AKI.


Subject(s)
Acute Kidney Injury/etiology , Estrogens/physiology , Sirtuin 1/physiology , Acute Kidney Injury/prevention & control , Female , Humans , Male
17.
Exp Cell Res ; 399(2): 112421, 2021 02 15.
Article in English | MEDLINE | ID: mdl-33412164

ABSTRACT

Postovulatory oocyte aging has a major influence on the development potential of embryos. Many antioxidants can delay oocyte aging by regulating the expression of SIRT1. However, there is a lack of knowledge on SIRT1 function in postovulatory oocyte aging. In vitro transcribed RNA of Sirt1 was injected into fresh oocytes to investigate the function of SIRT1 during postovulatory oocyte aging. In the present study, SIRT1 was found to be down-regulated in aged oocytes compared with fresh oocytes. Meanwhile the intensity of acetylation of H3K9 (H3K9ac) and H3K4 methylation increased in postovulatory aged oocytes. After the oocytes were injected with SIRT1 and aged for 12 h, the intensity of H3K9ac and H3K4 methylation markedly decreased compared with controls. Furthermore, SIRT1 overexpression also reduced the aging-induced oocyte morphological changes and reactive oxygen species accumulation, maintained the spindle normal morphology and attenuated the aging-associated abnormalities of mitochondrial function. The role of SIRT1 in protecting oocyte aging was diminished when oocytes with overexpressed SIRT1 were cultured with SIRT1 inhibitor EX-527. Briefly, these present results show that SIRT1 not only reduced the non-epigenetic changes such as abnormal oocyte morphology, ROS accumulation, spindle defects and mitochondrial dysfunctions but also regulated the epigenetic changes in order to maintain the quality of postovulatory aged oocytes.


Subject(s)
Cellular Senescence/genetics , Epigenesis, Genetic/genetics , Oocytes/physiology , Sirtuin 1/physiology , Acetylation , Animals , Antioxidants/metabolism , Cells, Cultured , DNA Methylation/genetics , Female , Histone Acetyltransferases/metabolism , Histones/metabolism , Mice , Mice, Inbred ICR , Oocytes/cytology , Ovulation/physiology , Reactive Oxygen Species/metabolism
18.
Cell Biol Int ; 45(5): 1050-1059, 2021 May.
Article in English | MEDLINE | ID: mdl-33438275

ABSTRACT

Endometrial cancer (EC) constitutes a common female genital tract tumor with a rising incidence rate. Sirtuin 1 (SIRT1) is a member of histone deacetylase, which extensively participates in the progression of aging, cell death, and tumorigenesis. This study explored the effect of SIRT1-mediated LC3 acetylation on autophagy and proliferation of EC cells. SIRT1 expression in EC tissues and adjacent tissues, EC cell lines and normal human epithelial cells was detected. SIRT1 expression was elevated in EC cell lines and tissues. Knockdown of SIRT1 inhibited proliferation, migration, and invasion of EC cells. Then, EC cells were starved in serum-free medium, and levels of autophagy-related proteins were detected. Starvation induced autophagy of EC cells. The starvation-treated EC cells showed an increased SIRT1 expression, a decreased LC3 acetylation level and an increased autophagy level. The proliferation and autophagy of EC cells under different treatments were evaluated. In EC cells transfected with overexpressing SIRT1, LC3 acetylation was inhibited and cell proliferation was promoted. Moreover, overexpressing SIRT1 facilitated growth and autophagy of transplanted tumors in nude mice. In conclusion, SIRT1 promoted autophagy and proliferation of EC cells by reducing acetylation level of LC3.


Subject(s)
Endometrial Neoplasms/metabolism , Microtubule-Associated Proteins/metabolism , Sirtuin 1/metabolism , Acetylation , Animals , Autophagy/physiology , Autophagy-Related Proteins/metabolism , Cell Line, Tumor , Cell Proliferation/physiology , Endometrium/metabolism , Endometrium/physiology , Female , Gene Expression/genetics , Humans , Mice , Mice, Nude , Microtubule-Associated Proteins/physiology , Sirtuin 1/physiology , Transcriptome/genetics , Xenograft Model Antitumor Assays
19.
Curr Neuropharmacol ; 19(7): 1019-1037, 2021.
Article in English | MEDLINE | ID: mdl-32727328

ABSTRACT

Neurodegeneration is a complex neurological phenomenon characterized by disturbed coherence in neuronal efflux. Progressive neuronal loss and brain damage due to various age-related pathological hallmarks perturb the behavioral balance and quality of life. Sirtuins have been widely investigated for their neuroprotective role, with SIRT1 being the most contemplated member of the family. SIRT1 exhibits significant capabilities to enhance neurogenesis and cellular lifespan by regulating various pathways, which makes it an exciting therapeutic target to inhibit neurodegenerative disease progression. SIRT1 mediated neuronal fortification involves modulation of molecular co-factors and biochemical pathways responsible for the induction and sustenance of pro-inflammatory and pro-oxidative environment in the cellular milieu. In this review, we present the major role played by SIRT1 in maintaining cellular strength through the regulation of genomic stability, neuronal growth, energy metabolism, oxidative stress, inhibiting mechanisms and anti-inflammatory responses. The therapeutic significance of SIRT1 has been put into perspective through a comprehensive discussion about its ameliorating potential against neurodegenerative stimuli in a variety of diseases that characteristically impair cognition, memory and motor coordination. This review enhances the acquaintance concerned with the neuroprotective potential of SIRT1 and thus promotes the development of novel SIRT1 regulating therapeutic agents and strategies.


Subject(s)
Neurodegenerative Diseases , Neurons/physiology , Sirtuin 1/physiology , Humans , Longevity , Neurodegenerative Diseases/drug therapy , Quality of Life
20.
Cartilage ; 13(2_suppl): 1237S-1248S, 2021 12.
Article in English | MEDLINE | ID: mdl-31072129

ABSTRACT

OBJECTIVE: The aim of this study is to investigate the role of Sirtuin1 (Sirt1) in the regulation of autophagy for human osteoarthritis (OA) chondrocytes. DESIGN: All cartilage samples were collected from human donors, including young group, aged group, and OA group. Primary chondrocytes were isolated and cultured with Sirt1 activator or inhibitor. Sirt1 expression in cartilage tissue and chondrocytes was evaluated, and the deacetylation activity of Sirt1 was determined. The alteration of autophagy activity after upregulating or downregulating Sirt1 was detected. Chondrocytes were treated with autophagy activator and inhibitor, and then the protein level of Sirt1 was examined. The interactions between Sirt1 and autophagy-related proteins Atg7, microtubule associated protein 1 light chain 3 (LC3), and Beclin-1 were determined by using immunoprecipitation. RESULTS: The assay of articular cartilage revealed that the expression of Sirt1 might be age-related: highly expressed in of younger people, and respectively decreased in the elderly people and OA patients. In vitro study was also validated this result. Further study confirmed that higher levels of Sirt1 significantly increased autophagy in aged chondrocytes, while the lower expression of Sirt1 reduced autophagy in young chondrocytes. Of note, the high levels of Sirt1 reduced autophagy in OA chondrocytes. When the chondrocytes were treated with autophagy activator or inhibitor, we found the expression of Sirt1 was not affected. In addition, we found that Sirt1 could interact with Atg7. CONCLUSION: These results suggest that Sirt1 in human chondrocytes regulates autophagy by interacting with autophagy related Atg7, and Sirt1 may become a more important target in OA treatment.


Subject(s)
Cartilage, Articular , Osteoarthritis , Sirtuin 1 , Aged , Autophagy/physiology , Cartilage, Articular/metabolism , Chondrocytes/metabolism , Humans , Osteoarthritis/metabolism , Sirtuin 1/metabolism , Sirtuin 1/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...