Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 48
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Medicine (Baltimore) ; 100(27): e26580, 2021 Jul 09.
Artigo em Inglês | MEDLINE | ID: mdl-34232204

RESUMO

ABSTRACT: Lung cancer (LC) is a prevalent malignancy worldwide with increased morbidity and mortality. Mitochondrial nicotinamide adenine dinucleotide-dependent deacetylase sirtuin-3 (SIRT3) has been reported to be involved in tumorigenesis. In this retrospective study, we measured the expression and diagnostic value of SIRT3 in LC patients.Quantitative real-time polymerase chain reaction (qRT-PCR) was performed to measure serum SIRT3 mRNA level in 150 LC patients and 52 healthy volunteers. SIRT3 protein level was detected using western blot for 84 pairs of LC and adjacent normal tissues. The association of SIRT3 mRNA level with clinical parameters of LC patients was estimated via chi-square test. Receiver operating characteristic curve (ROC) was plotted to evaluate the diagnostic performance of serum SIRT3 in LC patients.SIRT3 mRNA and protein levels were significantly decreased in LC tissues and serum samples, compared with corresponding controls (P < .05). Moreover, the expression of SIRT3 mRNA was negatively associated with tumor size (P = .002), tumor node metastasis stage (P < .001), and metastasis (P < .001). ROC curve demonstrated that serum SIRT3 could distinguish LC patients from healthy individuals, with an area under the curve of 0.918. The optimal cutoff value was 3.12, reaching a sensitivity of 86.4%, and a specificity of 94%.SIRT3 expression is significantly down-regulated in LC serum and tissues. SIRT3 may be employed as a promising biomarker in the early diagnosis of LC.


Assuntos
Regulação Neoplásica da Expressão Gênica , Neoplasias Pulmonares/diagnóstico , Sirtuína 3/genética , Biomarcadores Tumorais/biossíntese , Biomarcadores Tumorais/genética , Western Blotting , Feminino , Seguimentos , Humanos , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/metabolismo , Masculino , Pessoa de Meia-Idade , RNA Neoplásico/genética , Estudos Retrospectivos , Sirtuína 3/biossíntese
2.
Anticancer Res ; 41(3): 1377-1386, 2021 03.
Artigo em Inglês | MEDLINE | ID: mdl-33788729

RESUMO

BACKGROUND/AIM: The purpose of this study was to examine the expression of estrogen receptor α (ERα) and ß (ERß), androgen receptor (AR), SIRT1, SIRT2 and SIRT3 in prostate cancer (PCa). MATERIALS AND METHODS: From October 2010 to January 2015, 70 patients who had undergone radical prostatectomy following a PCa diagnosis were enrolled in our study. Normal prostate tissue (NPT) and prostate cancer tissues (PCAT) were separated, and the expression of each receptor in each tissue was analyzed with immunochemical staining. Univariate and multivariate analyses were performed to identify factors affecting the development of PCa. RESULTS: ERß and AR were highly expressed in PCAT compared with NPT (p<0.05). SIRT2 was highly expressed in NPT and PCAT (p<0.05). Univariate and multivariate analyses showed that AR and SIRT2 affect PCa development. CONCLUSION: AR is a risk factor for PC, and SIRT2 is associated with a lower incidence of PCa.


Assuntos
Receptor alfa de Estrogênio/biossíntese , Receptor beta de Estrogênio/biossíntese , Neoplasias da Próstata/metabolismo , Receptores Androgênicos/biossíntese , Sirtuína 1/biossíntese , Sirtuína 2/biossíntese , Sirtuína 3/biossíntese , Idoso , Humanos , Imuno-Histoquímica/métodos , Masculino , Pessoa de Meia-Idade , Análise Multivariada , Prostatectomia/métodos , Neoplasias da Próstata/cirurgia
3.
J Am Heart Assoc ; 10(5): e018913, 2021 02.
Artigo em Inglês | MEDLINE | ID: mdl-33586458

RESUMO

Background Impairment of glycolytic metabolism is suggested to contribute to diabetic cardiomyopathy. In this study, we explored the roles of SIRT3 (Sirtuin 3) on cardiomyocyte glucose metabolism and cardiac function. Methods and Results Exposure of H9c2 cardiomyocyte cell lines to high glucose (HG) (30 mmol/L) resulted in a gradual decrease in SIRT3 and 6-phosphofructo-2-kinase/fructose-2,6-bisphosphatase isoform 3 (PFKFB3) expression together with increases in p53 acetylation and TP53-induced glycolysis and apoptosis regulator (TIGAR) expression. Glycolysis was significantly reduced in the cardiomyocyte exposed to HG. Transfection with adenovirus-SIRT3 significantly increased PFKFB3 expression and reduced HG-induced p53 acetylation and TIGAR expression. Overexpression of SIRT3 rescued impaired glycolysis and attenuated HG-induced reactive oxygen species formation and apoptosis. Knockdown of TIGAR in cardiomyocytes by using siRNA significantly increased PFKFB3 expression and glycolysis under hyperglycemic conditions. This was accompanied by a significant suppression of HG-induced reactive oxygen species formation and apoptosis. In vivo, overexpression of SIRT3 by an intravenous jugular vein injection of adenovirus-SIRT3 resulted in a significant reduction of p53 acetylation and TIGAR expression together with upregulation of PFKFB3 expression in the heart of diabetic db/db mice at day 14. Overexpression of SIRT3 further reduced reactive oxygen species formation and blunted microvascular rarefaction in the diabetic db/db mouse hearts. Overexpression of SIRT3 significantly blunted cardiac fibrosis and hypertrophy and improved cardiac function at day 14. Conclusions Our study demonstrated that SIRT3 attenuated diabetic cardiomyopathy via regulating p53 acetylation and TIGAR expression. Therefore, SIRT3 may be a novel target for abnormal energy metabolism in diabetes mellitus.


Assuntos
Proteínas Reguladoras de Apoptose/genética , DNA/genética , Diabetes Mellitus Experimental , Cardiomiopatias Diabéticas/genética , Regulação da Expressão Gênica , Miócitos Cardíacos/metabolismo , Monoéster Fosfórico Hidrolases/genética , Sirtuína 3/genética , Animais , Proteínas Reguladoras de Apoptose/biossíntese , Western Blotting , Células Cultivadas , Cardiomiopatias Diabéticas/metabolismo , Cardiomiopatias Diabéticas/patologia , Masculino , Camundongos , Miócitos Cardíacos/patologia , Fosfofrutoquinase-2/biossíntese , Fosfofrutoquinase-2/genética , Monoéster Fosfórico Hidrolases/biossíntese , Sirtuína 3/biossíntese
4.
J Neuroinflammation ; 18(1): 41, 2021 Feb 04.
Artigo em Inglês | MEDLINE | ID: mdl-33541361

RESUMO

BACKGROUND: Postoperative cognitive dysfunction (POCD) is a very common complication that might increase the morbidity and mortality of elderly patients after surgery. However, the mechanism of POCD remains largely unknown. The NAD-dependent deacetylase protein Sirtuin 3 (SIRT3) is located in the mitochondria and regulates mitochondrial function. SIRT3 is the only sirtuin that specifically plays a role in extending lifespan in humans and is associated with neurodegenerative diseases. Therefore, the aim of this study was to evaluate the effect of SIRT3 on anesthesia/surgery-induced cognitive impairment in aged mice. METHODS: SIRT3 expression levels were decreased after surgery. For the interventional study, an adeno-associated virus (AAV)-SIRT3 vector or an empty vector was microinjected into hippocampal CA1 region before anesthesia/surgery. Western blotting, immunofluorescence staining, and enzyme-linked immune-sorbent assay (ELISA) were used to measure the oxidative stress response and downstream microglial activation and proinflammatory cytokines, and Golgi staining and long-term potentiation (LTP) recording were applied to evaluate synaptic plasticity. RESULTS: Overexpression of SIRT3 in the CA1 region attenuated anesthesia/surgery-induced learning and memory dysfunction as well as synaptic plasticity dysfunction and the oxidative stress response (superoxide dismutase [SOD] and malondialdehyde [MDA]) in aged mice with POCD. In addition, microglia activation (ionized calcium binding adapter molecule 1 [Iba1]) and neuroinflammatory cytokine levels (tumor necrosis factor-alpha [TNF-α], interleukin [IL]-1ß and IL-6) were regulated after anesthesia/surgery in a SIRT3-dependent manner. CONCLUSION: The results of the current study demonstrate that SIRT3 has a critical effect in the mechanism of POCD in aged mice by suppressing hippocampal neuroinflammation and reveal that SIRT3 may be a promising therapeutic and diagnostic target for POCD.


Assuntos
Envelhecimento/metabolismo , Anestésicos Inalatórios/toxicidade , Hipocampo/metabolismo , Mediadores da Inflamação/metabolismo , Complicações Cognitivas Pós-Operatórias/metabolismo , Sirtuína 3/biossíntese , Envelhecimento/efeitos dos fármacos , Envelhecimento/patologia , Animais , Hipocampo/efeitos dos fármacos , Hipocampo/patologia , Mediadores da Inflamação/antagonistas & inibidores , Isoflurano/toxicidade , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Neuroproteção/efeitos dos fármacos , Neuroproteção/fisiologia , Complicações Cognitivas Pós-Operatórias/induzido quimicamente , Complicações Cognitivas Pós-Operatórias/etiologia , Complicações Cognitivas Pós-Operatórias/prevenção & controle , Fraturas da Tíbia/cirurgia
5.
Cell Mol Neurobiol ; 41(6): 1203-1215, 2021 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-32500353

RESUMO

Sirtuin 3 (Sirt3) is a member of the Sirtuin family proteins and known to regulate multiple physiological processes such as metabolism and aging. As stroke is an aging-related disease, in this work, we attempt to examine the role and potential mechanism of Sirt3 in regulating ischemic stroke by using a permanent middle cerebral artery occlusion (pMCAO) model in wild type (WT) and Sirt3 knockout (KO) mice, coupled with oxygen glucose deprivation (OGD) experiments in cultured primary astrocytes. Sirt3 deficiency aggravated neuronal cell apoptosis and neurological deficits after brain ischemia. In addition, Sirt3 KO mice showed more severe blood-brain barrier (BBB) disruption and inflammatory responses compared with WT group in the acute phase. Furthermore, specific overexpression of Sirt3 in astrocytes by injecting glial fibrillary acidic protein (GFAP)::Sirt3 virus in ischemic region showed protective effect against stroke-induced damage. Mechanistically, Sirt3 could regulate vascular endothelial growth factor (VEGF) expression by inhibiting hypoxia inducible factor-1α (HIF-1α) signaling after ischemia (OGD). Our results have shown that Sirt3 plays a protective role in ischemic stroke via regulating HIF-1α/VEGF signaling in astrocytes, and reversal of the Sirt3 expression at the acute phase could be a worthy direction for stroke therapy.


Assuntos
Barreira Hematoencefálica/metabolismo , Isquemia Encefálica/metabolismo , Subunidade alfa do Fator 1 Induzível por Hipóxia/biossíntese , Neuroproteção/fisiologia , Sirtuína 3/biossíntese , Fator A de Crescimento do Endotélio Vascular/biossíntese , Animais , Barreira Hematoencefálica/patologia , Isquemia Encefálica/patologia , Isquemia Encefálica/prevenção & controle , Células Cultivadas , Subunidade alfa do Fator 1 Induzível por Hipóxia/antagonistas & inibidores , Masculino , Camundongos , Camundongos da Linhagem 129 , Camundongos Endogâmicos C57BL , Camundongos Knockout , Transdução de Sinais/fisiologia , Sirtuína 3/deficiência
6.
Cancer Biol Ther ; 22(1): 30-39, 2021 01 02.
Artigo em Inglês | MEDLINE | ID: mdl-33241954

RESUMO

Chemokine (C-X-C motif) ligand 6 (CXCL6), a member of the CXC chemokine family, reportedly mediates several processes such as inflammation, immunoreaction, cell growth, and metastasis through interaction with the chemokine receptors CXCR1 and CXCR2 in humans; further, CXCR1 and CXCR2 can promote repair and regeneration of organs or tissues after ischemia-reperfusion injury (IRI). In this study, we found that HIF-1α, CXCL6, and CXCR2 expression levels were elevated in human brain microvascular endothelial cells (HBMECs) after IRI, whereas silent information regulator of transcription (Sirt) 3 expression level had reduced. HIF-1α inhibition in an IRI model potently promoted HBMEC proliferation, accompanied by increased Sirt3 and decreased CXCL6/CXCR2 expression levels. CXCL6 knockdown in the IRI model significantly decreased HBMEC permeability and promoted HBMEC proliferation, concurrent with a decrease in apoptosis; it also increased Sirt3 expression levels and decreased CXCL6/CXCR2 protein and phosphorylated AKT (p-AKT) and class O of forkhead box (FOXO) 3a (p-FOXO3a) levels. In addition, CXCL6-induced HBMEC permeability and inhibition of HBMEC proliferation were counteracted by Sirt3 overexpression, and the AKT inhibitor LY294002 counteracted the effect of CXCL6 recombinant proteins on Sirt3, p-AKT, and p-FOXO3a expressions. These results suggest that CXCL6 and Sirt3 are downstream of HIF-1α and that CXCL6 regulatesHBMEC permeability, proliferation, and apoptosis after IRI by modulating Sirt3 expression via AKT/FOXO3a activation.


Assuntos
Quimiocina CXCL6/metabolismo , Proteína Forkhead Box O3/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Traumatismo por Reperfusão/metabolismo , Sirtuína 3/biossíntese , Apoptose/fisiologia , Permeabilidade da Membrana Celular , Proliferação de Células/fisiologia , Humanos , Traumatismo por Reperfusão/patologia , Transdução de Sinais
7.
J Biochem Mol Toxicol ; 35(3): e22662, 2021 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-33147367

RESUMO

Cancer and diabetes, the two mitochondria-related diseases, have recently been linked to silent mating-type information regulation 2 homolog 3 (SIRT3) activity irregularities. In this study, the effect of metformin, an antidiabetic with anticancer properties, has been evaluated on mitochondrial functionality markers, cell death pathways, and SIRT3 enzyme activity in the colon cancer cell line, HT-29, and human embryonic kidney cells (HEK 293). HT-29 cells were treated with metformin (5, 10, 20, 40, and 80 µM) for 24, 48, and 72 h for measuring the IC50 concentration. Reactive oxygen species (ROS) production, apoptosis, mitochondrial membrane potential, SIRT3 activity, and expression were evaluated against the colon cancer cell line, HT-29. Results indicated a higher ROS production at 6 than 12 h with metformin treatment. Metformin modified the mitochondrial membrane potential, resulting in cell death induction. Results from SIRT3 activity and expression showed that metformin increased its activity and expression in cancer cells. In conclusion, metformin in HT-29 cells disturbed the mitochondrial activity via increased ROS levels and SIRT3 activity, and these rapid modifications may play a key role in its cytotoxic property.


Assuntos
Neoplasias do Colo/enzimologia , Regulação Enzimológica da Expressão Gênica/efeitos dos fármacos , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Metformina/farmacologia , Mitocôndrias/metabolismo , Proteínas de Neoplasias/biossíntese , Sirtuína 3/biossíntese , Regulação para Cima/efeitos dos fármacos , Neoplasias do Colo/tratamento farmacológico , Neoplasias do Colo/patologia , Células HT29 , Humanos , Mitocôndrias/patologia
8.
Neuroreport ; 31(3): 220-225, 2020 02 05.
Artigo em Inglês | MEDLINE | ID: mdl-31876685

RESUMO

Recent studies revealed that increased oxidative stress is one of the major mechanisms underlying the cognitive dysfunction induced by long-term intermittent hypoxia (LTIH). Locus ceruleus (LC) neurons, which fire at high rate across wakefulness, are essential for optimal cognitive function. The aim of this study was to investigate whether sirtuin type 3 (SirT3), a redox responses coordinator, plays a role in LTIH-induced neurocognitive impairment. Mice were subjected to LTIH or room air [normal control (NC)] for 10 weeks (10 h/day). Morris water maze test was used to detect spatial learning and memory ability. The oxidative stress was evaluated through the level of superoxide dismutase 2 (SOD2) and dihydroethidium and ethidium (DHE). Then the correlation between the number of platform crossing and SirT3 content measured by western blot was analyzed. Results showed that performance on the Morris water maze test was significantly worse for LTIH mice than for NC mice. LTIH exposure downregulated SirT3 and SOD2 in LC neurons, increasing DHE immunodensity. In addition, the SirT3 protein levels in LC neurons were positively related to the number of platform crossing. These observations suggest that SirT3-SOD2-intracellular superoxide is a key component associated with the cognitive dysfunction induced by LTIH. Moreover, they lend support to a rational basis for targeting upregulation of SirT3 in LC as a disease modifying strategy.Video abstract: http://links.lww.com/WNR/A577.


Assuntos
Disfunção Cognitiva/metabolismo , Hipóxia/complicações , Locus Cerúleo/metabolismo , Estresse Oxidativo/fisiologia , Sirtuína 3/biossíntese , Animais , Disfunção Cognitiva/etiologia , Hipóxia/metabolismo , Masculino , Aprendizagem em Labirinto , Camundongos , Camundongos Endogâmicos C57BL
9.
Eur Rev Med Pharmacol Sci ; 23(21): 9180-9188, 2019 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-31773668

RESUMO

OBJECTIVE: The objective of this paper is to determine whether SIRT3 could retard intervertebral disc degeneration and study the mechanism. MATERIALS AND METHODS: We chose the 3-month mice to establish intervertebral disc degeneration model and study the effect of SIRT3 on the intervertebral disc by Western blotting, quantitative Real Time-Polymerase Chain Reaction (qRT-PCR), immunohistochemistry. Mouse nucleus pulposus cells were cultured to study the exact mechanism. RESULTS: The expression of SIRT3 was decreased in degenerated human nucleus pulposus. Intervertebral discs of mice treated with theacrine expressed more collagen II and less collagen X. In addition, nucleus pulposus cells stimulated with interleukin-1ß (IL-1ß) expressed less SIRT3 than that in the control group and nucleus pulposus cells with SIRT3 overexpress vectors expressed more collagen II FOXO3a and superoxide dismutase 2 (SOD2), indicating that SIRT3 could improve the intervertebral disc degeneration by anti-oxidative stress. CONCLUSIONS: SIRT3 is a protective factor for intervertebral discs and can reduce oxidative stress in the intervertebral disc.


Assuntos
Proteína Forkhead Box O3/biossíntese , Degeneração do Disco Intervertebral/fisiopatologia , Sirtuína 3/fisiologia , Superóxido Dismutase/biossíntese , Animais , Colágeno/biossíntese , Colágeno Tipo II/biossíntese , Humanos , Interleucina-1beta/farmacologia , Disco Intervertebral , Degeneração do Disco Intervertebral/metabolismo , Camundongos , Núcleo Pulposo , Estresse Oxidativo/fisiologia , Fatores de Proteção , Transdução de Sinais/fisiologia , Sirtuína 3/biossíntese
10.
Biosci Rep ; 39(10)2019 10 30.
Artigo em Inglês | MEDLINE | ID: mdl-31652451

RESUMO

Isoflurane/surgery (I/S) may induce neurocognitive disorders, but detailed mechanisms and appropriate treatment remain largely unknown. This experiment was designed to determine whether ginsenoside Rg1 could attenuate I/S-induced neurocognitive disorders and Sirtuin3 (Sirt3) dysfunction. C57BL/6J male mice received 1.4% isoflurane plus abdominal surgery for 2 h. Ginsenoside Rg1 10 mg/kg was intraperitoneally given for 8 days before surgery. Neurocognitive function was assessed by the Barnes Maze test. Levels of reactive oxygen species (ROS), oxygen consumption rate (OCR), mitochondrial membrane potential (MMP), expression and deacetylation activity of Sirt3 in the hippocampus tissues were measured. Results showed that I/S induced hippocampus-dependent learning and memory impairments, with increased ROS levels, and reduced OCR, MMP, and expression and deacetylation activity of Sirt3 in hippocampus tissues. Ginsenoside Rg1 treatment before I/S intervention significantly ameliorated learning and memory performance, reduced ROS levels and improved the OCR, MMP, expression and deacetylation activity of Sirt3. In conclusion, this experiment demonstrates that ginsenoside Rg1 treatment can attenuate I/S-induced neurocognitive disorders and Sirt3 dysfunction.


Assuntos
Regulação Enzimológica da Expressão Gênica/efeitos dos fármacos , Ginsenosídeos/farmacologia , Hipocampo/enzimologia , Isoflurano/efeitos adversos , Complicações Cognitivas Pós-Operatórias , Sirtuína 3/biossíntese , Animais , Hipocampo/patologia , Isoflurano/farmacologia , Masculino , Aprendizagem em Labirinto/efeitos dos fármacos , Potencial da Membrana Mitocondrial/efeitos dos fármacos , Camundongos , Complicações Cognitivas Pós-Operatórias/induzido quimicamente , Complicações Cognitivas Pós-Operatórias/tratamento farmacológico , Complicações Cognitivas Pós-Operatórias/enzimologia , Complicações Cognitivas Pós-Operatórias/patologia , Espécies Reativas de Oxigênio/metabolismo
11.
Pulm Pharmacol Ther ; 57: 101803, 2019 08.
Artigo em Inglês | MEDLINE | ID: mdl-31085231

RESUMO

Pulmonary fibrosis is a progressive fibrotic lung disease with a paucity of therapeutic options. Here we investigated the potential roles of probucol, a cholesterol-lowering drug with potent anti-oxidation properties, on pulmonary epithelial-mesenchymal transition (EMT) and fibrosis. We found that bleomycin-induced lung fibrosis was associated with increased transforming growth factor (TGF)-ß1, α-smooth muscle actin (α-SMA) and decreased E-cadherin expression in lung tissues, indicating EMT formation. Bleomycin treatment resulted in an induction of oxidative stress in lung tissues. Probucol treatment attenuated bleomycin-induced TGF-ß1 production, EMT and pulmonary fibrosis, meanwhile it suppressed bleomycin-induced oxidative stress. Bleomycin treatment resulted in decreases in protein expressions of Sirtuin 3 (SIRT3) in the lung, which were restored by ROS scavenger NAC and probucol treatment, suggesting that probucol might restore SIRT3 expression by suppressing bleomycin-induced oxidative stress. In the mouse alveolar type II epithelial cell line MLE-12, probucol treatment leads to an increase in SIRT3 expression in bleomycin-treated AT-II cells, which might contribute to the inhibitory effect of probucol on EMT through suppressing hypoxia inducible factor (HIF)-1α/TGF-ß1 pathway. In addition, probucol inhibited bleomycin-induced macrophage infiltration in the lung. Bleomycin decreased SIRT3 protein expression, whereas increased HIF-1α activation and TGF-ß1 release in the mouse macrophage cell line RAW264.7, which were attenuated by probucol treatment. Taken together, the present study suggests that probucol may ameliorate EMT and lung fibrosis through restoration of SIRT3 expression. The data obtained in this study provides proof for the idea that probucol may be a potential therapeutic option for the treatment of pulmonary fibrosis.


Assuntos
Transição Epitelial-Mesenquimal/efeitos dos fármacos , Probucol/farmacologia , Fibrose Pulmonar/tratamento farmacológico , Sirtuína 3/metabolismo , Actinas/metabolismo , Células Epiteliais Alveolares , Animais , Bleomicina/farmacologia , Caderinas/metabolismo , Técnicas de Cultura de Células , Colágeno Tipo I/metabolismo , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Camundongos , Modelos Animais , Estresse Oxidativo/efeitos dos fármacos , Fibrose Pulmonar/induzido quimicamente , Fibrose Pulmonar/metabolismo , Fibrose Pulmonar/patologia , Células RAW 264.7 , Transdução de Sinais/efeitos dos fármacos , Sirtuína 3/biossíntese , Fator de Crescimento Transformador beta1/metabolismo
12.
J Alzheimers Dis ; 69(2): 355-362, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-30958373

RESUMO

BACKGROUND: Emerging evidence shows tau acetylation has been observed in Alzheimer's disease (AD) brain at early Braak stages and is involved in regulating tau early accumulation. However, the effects of deacetylase Sirtuin 3 (Sirt3) on tau acetylation and its aggregations are unclear. OBJECTIVE: We studied the effects of Sirt3 on tau acetylation and its aggregations. METHODS: We investigated the protein levels of Sirt3 and tangle tau in human postmortem brains slices from AD, mild cognitive impairment, and age- and education-matched cognitively normal subjects, and AD model mice. We also measured tau acetylation levels in hippocampal HT22 cells after Sirt3 knockdown or overexpression. RESULTS: The level of Sirt3 was inversely related with tau protein in brain slices from both human being and AD model mice. Mechanistically, tau acetylation decreased dramatically with Sirt3 overexpression, while tau acetylation increased after Sirt3 knockdown in hippocampal HT22 cells. CONCLUSIONS: Sirt3 may play a role in tau acetylation and could be a potential target for novel therapy to alleviate tau accumulation.


Assuntos
Encéfalo/metabolismo , Sirtuína 3/biossíntese , Proteínas tau/biossíntese , Acetilação , Animais , Encéfalo/patologia , Linhagem Celular , Humanos , Camundongos , Camundongos Transgênicos , Sirtuína 3/genética , Proteínas tau/genética
13.
Exp Eye Res ; 182: 1-9, 2019 05.
Artigo em Inglês | MEDLINE | ID: mdl-30849386

RESUMO

Acetylation of lysine residues occurs in lens proteins. Previous studies have shown an improvement in the chaperone activity of αA-crystallin upon acetylation. Sirtuins are NAD+-dependent enzymes that can deacylate proteins. The roles of sirtuins in regulating the acetylation of lens proteins and their impacts on the function of α-crystallin are not known. Here, we detected sirtuin activity in mouse lenses, and SIRT3 and SIRT5 were present primarily in the mitochondria of cultured primary mouse lens epithelial cells. Western blotting showed higher levels of protein acetylation in the lenses of SIRT3 KO and SIRT5 KO mice than in lenses of WT mice. Mass spectrometry analyses revealed a greater number of acetylated lysine residues in α-crystallin isolated from the SIRT3 and SIRT5 KO lenses than from WT lenses. α-Crystallin isolated from SIRT3 and SIRT5 KO lenses displayed a higher surface hydrophobicity and higher chaperone activity than the protein isolated from WT lenses. Thus, SIRTs regulate the acetylation levels of crystallins in mouse lenses, and acetylation in lenses enhances the chaperone activity of α-crystallin.


Assuntos
Catarata/genética , Regulação da Expressão Gênica , Cristalino/metabolismo , Chaperonas Moleculares/metabolismo , Sirtuína 3/genética , Sirtuínas/genética , alfa-Cristalinas/genética , Acetilação , Animais , Western Blotting , Catarata/metabolismo , Modelos Animais de Doenças , Espectrometria de Massas , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , RNA/genética , Sirtuína 3/biossíntese , Sirtuínas/biossíntese , alfa-Cristalinas/metabolismo
14.
Life Sci ; 222: 78-87, 2019 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-30797821

RESUMO

AIMS: Acute kidney injury (AKI) can limit the clinical use of cisplatin in cancer treatment. The drivers of cisplatin-induced AKI include oxidative stress, mitochondrial dysfunction and apoptosis. Previous studies showed renalase protected cultured human renal proximal tubular cell (HK-2) against cisplatin induced necrosis, and renalase-knockout mice subjected to cisplatin showed exacerbated kidney injury. Therefore, it is necessary to determine the exact mechanisms of renalase in cisplatin-induced nephrotoxicity. MAIN METHODS: To study the protective effect of renalase on cell viability, renal function, apoptosis, reactive oxygen species (ROS) production and mitochondrial dynamics, cultured HK-2 cells and male mice were subjected to cisplatin. Signaling proteins related to apoptosis, survival, and mitochondrial fission were analyzed by Western blot. KEY FINDINGS: In this study, we showed that the protective effect of recombinant renalase in cisplatin-induced AKI was associated with the regulation of ROS production, mitochondrial dynamics and sirtuin-3 (Sirt3) levels in vivo and in vitro. After cisplatin treatment, recombinant renalase restored Sirt3 expression, reduced mitochondrial fission and ROS generation. In HK-2 cells, downregulation of endogenous Sirt3 expression by siRNA transfection abrogated the renalase cytoprotection. SIGNIFICANCE: Our study suggests that renalase protects against cisplatin-induced AKI by improving mitochondrial function and inhibiting oxidative stress, and in vitro, it functions in a Sirt3-dependent manner.


Assuntos
Injúria Renal Aguda/metabolismo , Injúria Renal Aguda/prevenção & controle , Cisplatino/toxicidade , Dinâmica Mitocondrial/efeitos dos fármacos , Monoaminoxidase/uso terapêutico , Sirtuína 3/biossíntese , Injúria Renal Aguda/induzido quimicamente , Animais , Antineoplásicos/toxicidade , Linhagem Celular , Humanos , Túbulos Renais Proximais/efeitos dos fármacos , Túbulos Renais Proximais/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Dinâmica Mitocondrial/fisiologia , Monoaminoxidase/farmacologia , Distribuição Aleatória , Espécies Reativas de Oxigênio/antagonistas & inibidores , Espécies Reativas de Oxigênio/metabolismo , Sirtuína 3/antagonistas & inibidores
15.
Exp Eye Res ; 181: 223-231, 2019 04.
Artigo em Inglês | MEDLINE | ID: mdl-30794763

RESUMO

We aim to investigate how Sirt3 (silent mating type information regulation 2 homolog 3) promoting diabetic corneal epithelial wound healing by regulating mitophagy. The effect of HG(High Glucose, 25 mM D-glucose) on Sirt3 and LC3B(light chain 3 beta)which representing of mitophagy were investigated in TKE2 cells (a murine limbal/corneal epithelium-derived progenitor cell line) and corneal epithelium from C57BL/6J-Ins2Akita (Ins2Akita/+) mice using RT-PCR and Western blotting. How overexpression of Sirt3 promoting diabetic corneal epithelial wound healing was investigated with cell migration assay、immunofluorescence、 immunofluorescence colocalization and corneal injury model. We found that HG reduced the expression of Sirt3 as well the mitophagy both in TKE2 cells and corneas from Ins2Akita/+ mice. And overexpression of Sirt3 prominently promoted wound healing speed under HG condition via upregulating the level of mitophagy. Mitophagy level was increased dramatically when the Foxo3a (Forkhead box O3)/PINK1(PTEN Induced putative kinase protein 1)-Parkin pathway was activated by Sirt3 overexpression which suggested that the mitophagy was involved in cell injury under HG condition. This study demonstrated the mechanism of Sirt3 regulating mitophagy to promote diabetic corneal epithelial wound healing in vivo and in vitro, which suggested that Sirt3 may positively impact diabetic keratopathy(DK).


Assuntos
Lesões da Córnea/genética , Diabetes Mellitus Experimental , Epitélio Corneano/metabolismo , Mitofagia/genética , Sirtuína 3/genética , Regulação para Cima/genética , Cicatrização , Animais , Western Blotting , Células Cultivadas , Lesões da Córnea/metabolismo , Lesões da Córnea/patologia , Epitélio Corneano/patologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Sirtuína 3/biossíntese
16.
Diabetes Metab Syndr ; 13(1): 582-589, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-30641770

RESUMO

INTRODUCTION: Sirtuins regulate energy metabolism and insulin sensitivity through their ability to act as energy sensors and regulators in several metabolic tissues. AIM: To evaluate the expression levels of sirtuin genes SIRT1, SIRT2, SIRT3 and SIRT6 and their target genes (PPAR-α, PGC1-α, NRF1, DGAT1, PPAR-γ and FOXO3a) in subcutaneous adipose tissue collected from individuals with normoweight, overweight and obesity. METHODS: Adipose tissue samples, obtained by lipoaspiration during liposuction surgery, were processed to obtain RNA, which was reverse-transcribed to cDNA. Then, we measured the expression levels of each gene by qPCR. RESULTS: We found differences in the mRNA expression of SIRT1, SIRT2, SIRT3 and SIRT6 and their target genes (PPAR-α, PGC1-α, NRF1, DGAT1, PPAR-γ and FOXO3a) in adipose tissue from overweight or obese subjects when compared to normoweight subjects. All genes analyzed, except SIRT2, showed correlation with BMI. CONCLUSIONS: Our findings in human subcutaneous adipose tissue show that increased body mass index modifies the expression of genes encoding sirtuins and their target genes, which are metabolic regulators of adipose tissue. Therefore, these could be used as biomarkers to predict the ability of adipose tissue to gain mass of adipose tissue.


Assuntos
Tecido Adiposo/fisiologia , Obesidade/genética , Sirtuína 1/genética , Sirtuína 2/genética , Sirtuína 3/genética , Sirtuínas/genética , Adulto , Índice de Massa Corporal , Feminino , Humanos , Pessoa de Meia-Idade , Obesidade/diagnóstico , Obesidade/metabolismo , Sirtuína 1/biossíntese , Sirtuína 2/biossíntese , Sirtuína 3/biossíntese , Sirtuínas/biossíntese , Adulto Jovem
17.
Hypertension ; 72(2): 350-360, 2018 08.
Artigo em Inglês | MEDLINE | ID: mdl-29915018

RESUMO

Endothelial-to-mesenchymal transition (EndoMT) has recently emerged as a potentially important contributor in promoting fibrosis in chronic kidney disease. However, little is known about the role and molecular basis of its involvement in hypertensive renal injury. Here, we aim to determine the role of SIRT (sirtuin) 3 on EndoMT in hypertensive renal injury and to explore its underlying mechanisms. We found that SIRT3 expression was significantly reduced in Ang II (angiotensin II)-induced hypertensive model, accompanied with induction of EndoMT and increased reactive oxygen species and renal fibrosis. In SIRT3-/- (SIRT3 knockout) mice subjected to Ang II infusion, renal dysfunction was aggravated with an increased EndoMT and reactive oxygen species level, whereas in SIRT3-TgEC (SIRT3 endothelial cell-specific transgenic) mice, the Ang II-induced renal fibrosis and EndoMT and oxidative stress were ameliorated. With primary mouse glomerular endothelial cells, we confirmed that Ang II treatment initiated EndoMT and decreased catalase expression, which were suppressed by SIRT3 overexpression. Using immunoprecipitation, luciferase, and chromatin immunoprecipitation assay, we demonstrated that SIRT3-mediated deacetylation and nuclear localization of Foxo3a (forkhead box O3a) resulted in activated Foxo3a-dependent catalase expression. Moreover, Foxo3a knockdown abolished SIRT3-mediated suppression of EndoMT. In conclusion, these results established the SIRT3-Foxo3a-catalase pathway as a critical factor in the maintenance of endothelial homeostasis and point to an important role of EndoMT in the vascular pathology of renal fibrosis, which may provide a new therapeutic target to impede the progression of hypertensive renal injury.


Assuntos
Células Endoteliais/metabolismo , Regulação da Expressão Gênica , Hipertensão/complicações , Nefropatias/etiologia , Estresse Oxidativo , RNA/genética , Sirtuína 3/genética , Animais , Modelos Animais de Doenças , Células Endoteliais/patologia , Hipertensão/genética , Hipertensão/metabolismo , Nefropatias/genética , Nefropatias/metabolismo , Masculino , Camundongos , Camundongos Transgênicos , Espécies Reativas de Oxigênio/metabolismo , Transdução de Sinais , Sirtuína 3/biossíntese
18.
Proc Natl Acad Sci U S A ; 115(27): 7057-7062, 2018 07 03.
Artigo em Inglês | MEDLINE | ID: mdl-29915029

RESUMO

Metastasis remains the leading cause of cancer mortality, and reactive oxygen species (ROS) signaling promotes the metastatic cascade. However, the molecular pathways that control ROS signaling relevant to metastasis are little studied. Here, we identify SIRT3, a mitochondrial deacetylase, as a regulator of cell migration via its control of ROS signaling. We find that, although mitochondria are present at the leading edge of migrating cells, SIRT3 expression is down-regulated during migration, resulting in elevated ROS levels. This SIRT3-mediated control of ROS represses Src oxidation and attenuates focal adhesion kinase (FAK) activation. SIRT3 overexpression inhibits migration and metastasis in breast cancer cells. Finally, in human breast cancers, SIRT3 expression is inversely correlated with metastatic outcome and Src/FAK signaling. Our results reveal a role for SIRT3 in cell migration, with important implications for breast cancer progression.


Assuntos
Neoplasias da Mama/metabolismo , Movimento Celular , Células Epiteliais/metabolismo , Quinase 1 de Adesão Focal/metabolismo , Proteínas de Neoplasias/metabolismo , Sirtuína 3/biossíntese , Quinases da Família src/metabolismo , Neoplasias da Mama/patologia , Linhagem Celular Tumoral , Ativação Enzimática , Células Epiteliais/patologia , Feminino , Humanos , Metástase Neoplásica , Espécies Reativas de Oxigênio , Sirtuína 3/metabolismo
19.
Exp Cell Res ; 367(2): 137-149, 2018 06 15.
Artigo em Inglês | MEDLINE | ID: mdl-29580688

RESUMO

Increasing evidence suggests that mitochondrial respiratory chain complex I participates in carcinogenesis and cancer progression by providing energy and maintaining mitochondrial function. However, the role of complex I in ovarian cancer is largely unknown. In this study we showed that metformin, considered to be an inhibitor of complex I, simultaneously inhibited cell growth and induced mitochondrial-related apoptosis in human ovarian cancer cells. Metformin interrupted cellular energy metabolism mainly by causing damage to complex I that impacted mitochondrial function. Additionally, treatment with metformin increased the activation of sirtuin 3 (SIRT3), a mitochondrial deacetylase. We demonstrated that SIRT3 overexpression aggravated metformin-induced apoptosis, energy stress and mitochondrial dysfunction. Moreover, treatment with metformin or SIRT3 overexpression increased activation of AMP-activated protein kinase (AMPK), a major sensor of cellular energy status. AMPK compensated for energy loss by increasing glycolysis. The impact of this was assessed by reducing glucose levels in the media or by using inhibitors (2-deoxyglucose, Compound C) of glycolysis and AMPK. The combination of these factors with metformin intensified cytotoxicity through further downregulation of ATP. Our study outlines an important role for SIRT3 in the antitumor effect of mitochondrial complex I inhibitors in human ovarian cancer cells. This effect appears to be mediated by induction of energy stress and apoptosis. Strategies that target the mitochondria could be enhanced by modulating glycolysis to further aggravate energy stress that may increase the antitumor effect.


Assuntos
Antineoplásicos/farmacologia , Apoptose , Metformina/farmacologia , Mitocôndrias/efeitos dos fármacos , Neoplasias Ovarianas/metabolismo , Sirtuína 3/metabolismo , Proteínas Quinases Ativadas por AMP/metabolismo , Linhagem Celular Tumoral , Complexo I de Transporte de Elétrons/metabolismo , Feminino , Glucose/metabolismo , Humanos , Mitocôndrias/metabolismo , Neoplasias Ovarianas/patologia , Sirtuína 3/biossíntese , Estresse Fisiológico
20.
Biochim Biophys Acta Gen Subj ; 1862(5): 1157-1167, 2018 May.
Artigo em Inglês | MEDLINE | ID: mdl-29452236

RESUMO

BACKGROUND: Intrauterine growth restriction (IUGR) is associated with cardiovascular remodeling persisting into adulthood. Mitochondrial bioenergetics, essential for embryonic development and cardiovascular function, are regulated by nuclear effectors as sirtuins. A rabbit model of IUGR and cardiovascular remodeling was generated, in which heart mitochondrial alterations were observed by microscopic and transcriptomic analysis. We aimed to evaluate if such alterations are translated at a functional mitochondrial level to establish the etiopathology and potential therapeutic targets for this obstetric complication. METHODS: Hearts and placentas from 16 IUGR-offspring and 14 controls were included to characterize mitochondrial function. RESULTS: Enzymatic activities of complexes II, IV and II + III in IUGR-hearts (-11.96 ±â€¯3.16%; -15.58 ±â€¯5.32%; -14.73 ±â€¯4.37%; p < 0.05) and II and II + III in IUGR-placentas (-17.22 ±â€¯3.46%; p < 0.005 and -29.64 ±â€¯4.43%; p < 0.001) significantly decreased. This was accompanied by a not significant reduction in CI-stimulated oxygen consumption and significantly decreased complex II SDHB subunit expression in placenta (-44.12 ±â€¯5.88%; p < 0.001). Levels of mitochondrial content, Coenzyme Q and cellular ATP were conserved. Lipid peroxidation significantly decreased in IUGR-hearts (-39.02 ±â€¯4.35%; p < 0.001), but not significantly increased in IUGR-placentas. Sirtuin3 protein expression significantly increased in IUGR-hearts (84.21 ±â€¯31.58%; p < 0.05) despite conserved anti-oxidant SOD2 protein expression and activity in both tissues. CONCLUSIONS: IUGR is associated with cardiac and placental mitochondrial CII dysfunction. Up-regulated expression of Sirtuin3 may explain attenuation of cardiac oxidative damage and preserved ATP levels under CII deficiency. GENERAL SIGNIFICANCE: These findings may allow the design of dietary interventions to modulate Sirtuin3 expression and consequent regulation of mitochondrial imbalance associated with IUGR and derived cardiovascular remodeling.


Assuntos
Retardo do Crescimento Fetal/metabolismo , Mitocôndrias Cardíacas/metabolismo , Proteínas Mitocondriais/biossíntese , Placenta/metabolismo , Sirtuína 3/biossíntese , Animais , Modelos Animais de Doenças , Feminino , Retardo do Crescimento Fetal/patologia , Mitocôndrias Cardíacas/patologia , Placenta/patologia , Gravidez , Coelhos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...