Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 45
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
2.
Cell Metab ; 36(2): 354-376, 2024 02 06.
Artigo em Inglês | MEDLINE | ID: mdl-38181790

RESUMO

Here, we summarize the current knowledge on eight promising drugs and natural compounds that have been tested in the clinic: metformin, NAD+ precursors, glucagon-like peptide-1 receptor agonists, TORC1 inhibitors, spermidine, senolytics, probiotics, and anti-inflammatories. Multiple clinical trials have commenced to evaluate the efficacy of such agents against age-associated diseases including diabetes, cardiovascular disease, cancer, and neurodegenerative diseases. There are reasonable expectations that drugs able to decelerate or reverse aging processes will also exert broad disease-preventing or -attenuating effects. Hence, the outcome of past, ongoing, and future disease-specific trials may pave the way to the development of new anti-aging medicines. Drugs approved for specific disease indications may subsequently be repurposed for the treatment of organism-wide aging consequences.


Assuntos
Doenças Cardiovasculares , Metformina , Neoplasias , Humanos , NAD , Envelhecimento , Metformina/farmacologia , Metformina/uso terapêutico
3.
Cell ; 173(1): 74-89.e20, 2018 03 22.
Artigo em Inglês | MEDLINE | ID: mdl-29570999

RESUMO

A decline in capillary density and blood flow with age is a major cause of mortality and morbidity. Understanding why this occurs is key to future gains in human health. NAD precursors reverse aspects of aging, in part, by activating sirtuin deacylases (SIRT1-SIRT7) that mediate the benefits of exercise and dietary restriction (DR). We show that SIRT1 in endothelial cells is a key mediator of pro-angiogenic signals secreted from myocytes. Treatment of mice with the NAD+ booster nicotinamide mononucleotide (NMN) improves blood flow and increases endurance in elderly mice by promoting SIRT1-dependent increases in capillary density, an effect augmented by exercise or increasing the levels of hydrogen sulfide (H2S), a DR mimetic and regulator of endothelial NAD+ levels. These findings have implications for improving blood flow to organs and tissues, increasing human performance, and reestablishing a virtuous cycle of mobility in the elderly.


Assuntos
Envelhecimento , Sulfeto de Hidrogênio/metabolismo , NAD/metabolismo , Animais , Células Endoteliais/citologia , Células Endoteliais/metabolismo , Humanos , Camundongos , Camundongos Knockout , Microvasos/metabolismo , Mitocôndrias/metabolismo , Músculo Esquelético/metabolismo , Neovascularização Fisiológica , Coativador 1-alfa do Receptor gama Ativado por Proliferador de Peroxissomo/metabolismo , Condicionamento Físico Animal , Interferência de RNA , RNA Interferente Pequeno/metabolismo , Receptores Notch/metabolismo , Transdução de Sinais , Sirtuína 1/antagonistas & inibidores , Sirtuína 1/genética , Sirtuína 1/metabolismo , Fator A de Crescimento do Endotélio Vascular/metabolismo
4.
Hepatology ; 68(2): 496-514, 2018 08.
Artigo em Inglês | MEDLINE | ID: mdl-29457836

RESUMO

Alcoholic liver disease (ALD) is characterized by lipid accumulation and liver injury. However, how chronic alcohol consumption causes hepatic lipid accumulation remains elusive. The present study demonstrates that activation of the mechanistic target of rapamycin complex 1 (mTORC1) plays a causal role in alcoholic steatosis, inflammation, and liver injury. Chronic-plus-binge ethanol feeding led to hyperactivation of mTORC1, as evidenced by increased phosphorylation of mTOR and its downstream kinase S6 kinase 1 (S6K1) in hepatocytes. Aberrant activation of mTORC1 was likely attributed to the defects of the DEP domain-containing mTOR-interacting protein (DEPTOR) and the nicotinamide adenine dinucleotide-dependent deacetylase sirtuin 1 (SIRT1) in the liver of chronic-plus-binge ethanol-fed mice and in the liver of patients with ALD. Conversely, adenoviral overexpression of hepatic DEPTOR suppressed mTORC1 signaling and ameliorated alcoholic hepatosteatosis, inflammation, and acute-on-chronic liver injury. Mechanistically, the lipid-lowering effect of hepatic DEPTOR was attributable to decreased proteolytic processing, nuclear translocation, and transcriptional activity of the lipogenic transcription factor sterol regulatory element-binding protein-1 (SREBP-1). DEPTOR-dependent inhibition of mTORC1 also attenuated alcohol-induced cytoplasmic accumulation of the lipogenic regulator lipin 1 and prevented alcohol-mediated inhibition of fatty acid oxidation. Pharmacological intervention with rapamycin alleviated the ability of alcohol to up-regulate lipogenesis, to down-regulate fatty acid oxidation, and to induce steatogenic phenotypes. Chronic-plus-binge ethanol feeding led to activation of SREBP-1 and lipin 1 through S6K1-dependent and independent mechanisms. Furthermore, hepatocyte-specific deletion of SIRT1 disrupted DEPTOR function, enhanced mTORC1 activity, and exacerbated alcoholic fatty liver, inflammation, and liver injury in mice. CONCLUSION: The dysregulation of SIRT1-DEPTOR-mTORC1 signaling is a critical determinant of ALD pathology; targeting SIRT1 and DEPTOR and selectively inhibiting mTORC1-S6K1 signaling may have therapeutic potential for treating ALD in humans. (Hepatology 2018).


Assuntos
Fígado Gorduroso Alcoólico/metabolismo , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Lipogênese/genética , Alvo Mecanístico do Complexo 1 de Rapamicina/metabolismo , Animais , Etanol/farmacologia , Fígado Gorduroso Alcoólico/patologia , Hepatócitos/metabolismo , Humanos , Fígado/metabolismo , Fígado/patologia , Camundongos , Proteínas Nucleares/metabolismo , Fosfatidato Fosfatase/metabolismo , Transdução de Sinais , Sirtuína 1/metabolismo , Proteína de Ligação a Elemento Regulador de Esterol 1/metabolismo , Serina-Treonina Quinases TOR/metabolismo
5.
NPJ Aging Mech Dis ; 3: 17, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-29184669

RESUMO

NRPT is a combination of nicotinamide riboside (NR), a nicotinamide adenine dinucleotide (NAD+) precursor vitamin found in milk, and pterostilbene (PT), a polyphenol found in blueberries. Here, we report this first-in-humans clinical trial designed to assess the safety and efficacy of a repeat dose of NRPT (commercially known as Basis). NRPT was evaluated in a randomized, double-blind, and placebo-controlled study in a population of 120 healthy adults between the ages of 60 and 80 years. The study consisted of three treatment arms: placebo, recommended dose of NRPT (NRPT 1X), and double dose of NRPT (NRPT 2X). All subjects took their blinded supplement daily for eight weeks. Analysis of NAD+ in whole blood demonstrated that NRPT significantly increases the concentration of NAD+ in a dose-dependent manner. NAD+ levels increased by approximately 40% in the NRPT 1X group and approximately 90% in the NRPT 2X group after 4 weeks as compared to placebo and baseline. Furthermore, this significant increase in NAD+ levels was sustained throughout the entire 8-week trial. NAD+ levels did not increase for the placebo group during the trial. No serious adverse events were reported in this study. This study shows that a repeat dose of NRPT is a safe and effective way to increase NAD+ levels sustainably.

6.
PLoS One ; 12(5): e0178520, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28542607

RESUMO

Activation of SIRT1 has previously been shown to protect mice against osteoporosis through yet ill-defined mechanisms. In this study, we outline a role for SIRT1 as a positive regulator of the master osteoblast transcription factor, RUNX2. We find that ex vivo deletion of sirt1 leads to decreased expression of runx2 downstream targets, but not runx2 itself, along with reduced osteoblast differentiation. Reciprocally, treatment with a SIRT1 agonist promotes osteoblast differentiation, as well as the expression of runx2 downstream targets, in a SIRT1-dependent manner. Biochemical and luciferase reporter assays demonstrate that SIRT1 interacts with and promotes the transactivation potential of RUNX2. Intriguingly, mice treated with the SIRT1 agonist, resveratrol, show similar increases in the expression of RUNX2 targets in their calvaria (bone tissue), validating SIRT1 as a physiologically relevant regulator of RUNX2.


Assuntos
Subunidade alfa 1 de Fator de Ligação ao Core/metabolismo , Osteoblastos/metabolismo , Fatores de Transcrição/metabolismo , Animais , Diferenciação Celular/efeitos dos fármacos , Diferenciação Celular/fisiologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Osteoblastos/efeitos dos fármacos , Osteogênese/efeitos dos fármacos , Osteogênese/fisiologia , Resveratrol , Sirtuína 1/antagonistas & inibidores , Sirtuína 1/metabolismo , Estilbenos/farmacologia
7.
Nat Cell Biol ; 19(5): 445-456, 2017 May.
Artigo em Inglês | MEDLINE | ID: mdl-28436968

RESUMO

A hallmark of cancer cells is the metabolic switch from oxidative phosphorylation (OXPHOS) to glycolysis, a phenomenon referred to as the 'Warburg effect', which is also observed in primed human pluripotent stem cells (hPSCs). Here, we report that downregulation of SIRT2 and upregulation of SIRT1 is a molecular signature of primed hPSCs and that SIRT2 critically regulates metabolic reprogramming during induced pluripotency by targeting glycolytic enzymes including aldolase, glyceraldehyde-3-phosphate dehydrogenase, phosphoglycerate kinase, and enolase. Remarkably, knockdown of SIRT2 in human fibroblasts resulted in significantly decreased OXPHOS and increased glycolysis. In addition, we found that miR-200c-5p specifically targets SIRT2, downregulating its expression. Furthermore, SIRT2 overexpression in hPSCs significantly affected energy metabolism, altering stem cell functions such as pluripotent differentiation properties. Taken together, our results identify the miR-200c-SIRT2 axis as a key regulator of metabolic reprogramming (Warburg-like effect), via regulation of glycolytic enzymes, during human induced pluripotency and pluripotent stem cell function.


Assuntos
Diferenciação Celular , Metabolismo Energético , MicroRNAs/metabolismo , Células-Tronco Pluripotentes/enzimologia , Sirtuína 2/metabolismo , Acetilação , Linhagem da Célula , Sobrevivência Celular , Reprogramação Celular , Biologia Computacional , Bases de Dados Genéticas , Regulação Enzimológica da Expressão Gênica , Glicólise , Células HEK293 , Humanos , MicroRNAs/genética , Fosforilação Oxidativa , Fenótipo , Processamento de Proteína Pós-Traducional , Transdução de Sinais , Sirtuína 1/genética , Sirtuína 1/metabolismo , Sirtuína 2/genética , Fatores de Tempo , Transfecção
8.
Cell ; 166(2): 436-450, 2016 Jul 14.
Artigo em Inglês | MEDLINE | ID: mdl-27345368

RESUMO

Longevity-promoting caloric restriction is thought to trigger downregulation of mammalian target of rapamycin complex 1 (mTORC1) signaling and upregulation of SIRT1 activity with associated health benefits. Here, we show that mTORC1 signaling in intestinal stem cells (ISCs) is instead upregulated during calorie restriction (CR). SIRT1 deacetylates S6K1, thereby enhancing its phosphorylation by mTORC1, which leads to an increase in protein synthesis and an increase in ISC number. Paneth cells in the ISC niche secrete cyclic ADP ribose that triggers SIRT1 activity and mTORC1 signaling in neighboring ISCs. Notably, the mTOR inhibitor rapamycin, previously reported to mimic effects of CR, abolishes this expansion of ISCs. We suggest that Paneth cell signaling overrides any direct nutrient sensing in ISCs to sculpt the observed response to CR. Moreover, drugs that modulate pathways important in CR may exert opposing effects on different cell types.


Assuntos
Células-Tronco Adultas/metabolismo , Restrição Calórica , Complexos Multiproteicos/metabolismo , Transdução de Sinais , Sirtuína 1/metabolismo , Serina-Treonina Quinases TOR/metabolismo , Animais , Proliferação de Células , ADP-Ribose Cíclica/metabolismo , Dieta , Mucosa Intestinal/metabolismo , Intestinos/citologia , Alvo Mecanístico do Complexo 1 de Rapamicina , Camundongos , Camundongos Endogâmicos C57BL , NAD/metabolismo , Organoides/metabolismo , Fosforilação , Proteínas Quinases S6 Ribossômicas 90-kDa/metabolismo , Sirtuína 2/metabolismo
9.
Nat Commun ; 7: 11445, 2016 05 11.
Artigo em Inglês | MEDLINE | ID: mdl-27165717

RESUMO

Machado-Joseph disease (MJD) is a neurodegenerative disorder characterized by an abnormal expansion of the CAG triplet in the ATXN3 gene, translating into a polyglutamine tract within the ataxin-3 protein. The available treatments only ameliorate symptomatology and do not block disease progression. In this study we find that caloric restriction dramatically rescues the motor incoordination, imbalance and the associated neuropathology in transgenic MJD mice. We further show that caloric restriction rescues SIRT1 levels in transgenic MJD mice, whereas silencing SIRT1 is sufficient to prevent the beneficial effects on MJD pathology. In addition, the re-establishment of SIRT1 levels in MJD mouse model, through the gene delivery approach, significantly ameliorates neuropathology, reducing neuroinflammation and activating autophagy. Furthermore, the pharmacological activation of SIRT1 with resveratrol significantly reduces motor incoordination of MJD mice. The pharmacological SIRT1 activation could provide important benefits to treat MJD patients.


Assuntos
Restrição Calórica , Doença de Machado-Joseph/metabolismo , Doença de Machado-Joseph/patologia , Atividade Motora , Sistema Nervoso/patologia , Sirtuína 1/metabolismo , Animais , Ataxina-3/metabolismo , Autofagia/efeitos dos fármacos , Linhagem Celular Tumoral , Cerebelo/metabolismo , Cerebelo/patologia , Modelos Animais de Doenças , Marcha , Inflamação/patologia , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Atividade Motora/efeitos dos fármacos , Proteínas Mutantes/metabolismo , Sistema Nervoso/efeitos dos fármacos , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Neurônios/patologia , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Resveratrol , Sirtuína 1/genética , Estilbenos/administração & dosagem , Estilbenos/farmacologia , Estilbenos/uso terapêutico
10.
Nat Rev Cancer ; 15(10): 608-24, 2015 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-26383140

RESUMO

The sirtuins (SIRTs; of which there are seven in mammals) are NAD(+)-dependent enzymes that regulate a large number of cellular pathways and forestall the progression of ageing and age-associated diseases. In recent years, the role of sirtuins in cancer biology has become increasingly apparent, and growing evidence demonstrates that sirtuins regulate many processes that go awry in cancer cells, such as cellular metabolism, the regulation of chromatin structure and the maintenance of genomic stability. In this article, we review recent advances in our understanding of how sirtuins affect cancer metabolism, DNA repair and the tumour microenvironment and how activating or inhibiting sirtuins may be important in preventing or treating cancer.


Assuntos
Neoplasias/enzimologia , Sirtuínas/fisiologia , Animais , Reparo do DNA/fisiologia , DNA de Neoplasias/fisiologia , Humanos , Microambiente Tumoral/fisiologia
11.
Am J Physiol Lung Cell Mol Physiol ; 306(9): L816-28, 2014 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-24633890

RESUMO

Oxidative and carbonyl stress is increased in lungs of smokers and patients with chronic obstructive pulmonary disease (COPD), as well as in cigarette smoke (CS)-exposed rodent lungs. We previously showed that sirtuin1 (SIRT1), an antiaging protein, is reduced in lungs of CS-exposed mice and patients with COPD and that SIRT1 attenuates CS-induced lung inflammation and injury. It is not clear whether SIRT1 protects against CS-induced lung oxidative stress. Therefore, we determined the effect of SIRT1 on lung oxidative stress and antioxidants in response to CS exposure using loss- and gain-of-function approaches, as well as a pharmacological SIRT1 activation by SRT1720. We found that CS exposure increased protein oxidation and lipid peroxidation in lungs of wild-type (WT) mice, which was further augmented in SIRT1-deficient mice. Furthermore, both SIRT1 genetic overexpression and SRT1720 treatment significantly decreased oxidative stress induced by CS exposure. FOXO3 deletion augmented lipid peroxidation products but reduced antioxidants in response to CS exposure, which was not affected by SRT1720. Interestingly, SRT1720 treatment exhibited a similar effect on lipid peroxidation and antioxidants (i.e., manganese superoxide dismutase, heme oxygenase-1, and NADPH quinone oxidoreductase-1) in WT and nuclear factor (erythroid-derived 2)-like 2 (Nrf2)-deficient mice in response to CS exposure. This indicates that SIRT1 protects against CS-induced oxidative stress, which is mediated by FOXO3, but is independent of Nrf2. Overall, these findings reveal a novel function of SIRT1, which is to reduce CS-induced oxidative stress, and this may contribute to its protective effects against lung inflammation and subsequent development of COPD.


Assuntos
Fatores de Transcrição Forkhead/metabolismo , Pneumopatias/prevenção & controle , Estresse Oxidativo , Sirtuína 1/fisiologia , Fumaça/efeitos adversos , Animais , Antioxidantes/metabolismo , Western Blotting , Feminino , Proteína Forkhead Box O3 , Fatores de Transcrição Forkhead/genética , Humanos , Peroxidação de Lipídeos/efeitos dos fármacos , Pneumopatias/induzido quimicamente , Pneumopatias/patologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Fator 2 Relacionado a NF-E2/fisiologia , Carbonilação Proteica/efeitos dos fármacos , RNA Mensageiro/genética , Reação em Cadeia da Polimerase em Tempo Real , Reação em Cadeia da Polimerase Via Transcriptase Reversa
13.
Gastroenterology ; 146(2): 539-49.e7, 2014 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-24184811

RESUMO

BACKGROUND & AIMS: The hepatocyte-derived hormone fibroblast growth factor 21 (FGF21) is a hormone-like regulator of metabolism. The nicotinamide adenine dinucleotide-dependent deacetylase SIRT1 regulates fatty acid metabolism through multiple nutrient sensors. Hepatic overexpression of SIRT1 reduces steatosis and glucose intolerance in obese mice. We investigated mechanisms by which SIRT1 controls hepatic steatosis in mice. METHODS: Liver-specific SIRT1 knockout (SIRT1 LKO) mice and their wild-type littermates (controls) were divided into groups that were placed on a normal chow diet, fasted for 24 hours, or fasted for 24 hours and then fed for 6 hours. Liver tissues were collected and analyzed by histologic examination, gene expression profiling, and real-time polymerase chain reaction assays. Human HepG2 cells were incubated with pharmacologic activators of SIRT1 (resveratrol or SRT1720) and mitochondrion oxidation consumption rate and immunoblot analyses were performed. FGF21 was overexpressed in SIRT1 LKO mice using an adenoviral vector. Energy expenditure was assessed by indirect calorimetry. RESULTS: Prolonged fasting induced lipid deposition in livers of control mice, but severe hepatic steatosis in SIRT1 LKO mice. Gene expression analysis showed that fasting up-regulated FGF21 in livers of control mice but not in SIRT1 LKO mice. Decreased hepatic and circulating levels of FGF21 in fasted SIRT1 LKO mice were associated with reduced hepatic expression of genes involved in fatty acid oxidation and ketogenesis, and increased expression of genes that control lipogenesis, compared with fasted control mice. Resveratrol or SRT1720 each increased the transcriptional activity of the FGF21 promoter (-2070/+117) and levels of FGF21 messenger RNA and protein in HepG2 cells. Surprisingly, SIRT1 LKO mice developed late-onset obesity with impaired whole-body energy expenditure. Hepatic overexpression of FGF21 in SIRT1 LKO mice increased the expression of genes that regulate fatty acid oxidation, decreased fasting-induced steatosis, reduced obesity, increased energy expenditure, and promoted browning of white adipose tissue. CONCLUSIONS: SIRT1-mediated activation of FGF21 prevents liver steatosis caused by fasting. This hepatocyte-derived endocrine signaling appears to regulate expression of genes that control a brown fat-like program in white adipose tissue, energy expenditure, and adiposity. Strategies to activate SIRT1 or FGF21 could be used to treat fatty liver disease and obesity.


Assuntos
Metabolismo Energético/fisiologia , Fígado Gorduroso/metabolismo , Fatores de Crescimento de Fibroblastos/metabolismo , Sirtuína 1/metabolismo , Animais , Biomarcadores/metabolismo , Calorimetria Indireta , Jejum , Fígado Gorduroso/etiologia , Fígado Gorduroso/prevenção & controle , Perfilação da Expressão Gênica , Células Hep G2 , Humanos , Immunoblotting , Fígado/metabolismo , Fígado/patologia , Camundongos , Camundongos Knockout , Obesidade/etiologia , Obesidade/metabolismo , Obesidade/prevenção & controle , Reação em Cadeia da Polimerase em Tempo Real , Regulação para Cima
14.
Annu Rev Pharmacol Toxicol ; 54: 363-80, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24160699

RESUMO

The mammalian sirtuins (SIRT1-7) are NAD(+)-dependent lysine deacylases that play central roles in cell survival, inflammation, energy metabolism, and aging. Members of this family of enzymes are considered promising pharmaceutical targets for the treatment of age-related diseases including cancer, type 2 diabetes, inflammatory disorders, and Alzheimer's disease. SIRT1-activating compounds (STACs), which have been identified from a variety of chemical classes, provide health benefits in animal disease models. Recent data point to a common mechanism of allosteric activation by natural and synthetic STACs that involves the binding of STACs to a conserved N-terminal domain in SIRT1. Compared with polyphenols such as resveratrol, the synthetic STACs show greater potency, solubility, and target selectivity. Although considerable progress has been made regarding SIRT1 allosteric activation, key questions remain, including how the molecular contacts facilitate SIRT1 activation, whether other sirtuin family members will be amenable to activation, and whether STACs will ultimately prove safe and efficacious in humans.


Assuntos
Terapia de Alvo Molecular , Sirtuínas/metabolismo , Regulação Alostérica , Animais , Benzimidazóis/farmacologia , Modelos Animais de Doenças , Humanos , Inflamação/terapia , Resveratrol , Estilbenos/farmacologia
15.
Am J Physiol Lung Cell Mol Physiol ; 305(9): L615-24, 2013 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-24039251

RESUMO

Sirtuin1 (SIRT1), a protein/histone deacetylase, protects against the development of pulmonary emphysema. However, the molecular mechanisms underlying this observation remain elusive. The imbalance of tissue inhibitor of matrix metalloproteinases (TIMPs)/matrix metalloproteinases (MMPs) plays an important role in the pathogenesis of chronic obstructive pulmonary disease (COPD)/emphysema. We hypothesized that SIRT1 protects against emphysema by redressing the imbalance between MMPs and TIMPs. To test this hypothesis, SIRT1-deficient and overexpressing/transgenic mice were exposed to cigarette smoke (CS). The protein level and activity of MMP-9 were increased in lungs of SIRT1-deficient mice exposed to CS compared with wild-type (WT) littermates, and these effects were attenuated by SIRT1 overexpression. SIRT1 deficiency decreased the level of TIMP-1, which was augmented in SIRT1 transgenic mice compared with WT littermates by CS. However, the level of MMP-2, MMP-12, TIMP-2, TIMP-3, or TIMP-4 was not altered by SIRT1 in response to CS exposure. SIRT1 reduction was associated with imbalance of TIMP-1 and MMP-9 in lungs of smokers and COPD patients. Mass spectrometry and immunoprecipitation analyses revealed that TIMP-1 acetylation on specific lysine residues was increased, whereas its interaction with SIRT1 and MMP-9 was reduced in mouse lungs with emphysema, as well as in lungs of smokers and COPD patients. SIRT1 deficiency increased CS-induced TIMP-1 acetylation, and these effects were attenuated by SIRT1 overexpression. These results suggest that SIRT1 protects against COPD/emphysema, in part, via redressing the TIMP-1/MMP-9 imbalance involving TIMP-1 deacetylation. Thus redressing the TIMP-1/MMP-9 imbalance by pharmacological activation of SIRT1 is an attractive approach in the intervention of COPD.


Assuntos
Enfisema/metabolismo , Metaloproteinase 9 da Matriz/metabolismo , Doença Pulmonar Obstrutiva Crônica/metabolismo , Sirtuína 1/genética , Sirtuína 1/metabolismo , Inibidor Tecidual de Metaloproteinase-1/metabolismo , Sequência de Aminoácidos , Animais , Enfisema/patologia , Enfisema/fisiopatologia , Humanos , Pulmão/metabolismo , Pulmão/patologia , Pulmão/fisiopatologia , Camundongos , Camundongos da Linhagem 129 , Camundongos Endogâmicos C57BL , Camundongos Knockout , Dados de Sequência Molecular , Doença Pulmonar Obstrutiva Crônica/patologia , Doença Pulmonar Obstrutiva Crônica/fisiopatologia , Inibidor Tecidual de Metaloproteinase-1/genética , Poluição por Fumaça de Tabaco/efeitos adversos
16.
Nat Commun ; 4: 2236, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23900562

RESUMO

Reductively metabolized glutamine is a major cellular carbon source for fatty acid synthesis during hypoxia or when mitochondrial respiration is impaired. Yet, a mechanistic understanding of what determines reductive metabolism is missing. Here we identify several cellular conditions where the α-ketoglutarate/citrate ratio is changed due to an altered acetyl-CoA to citrate conversion, and demonstrate that reductive glutamine metabolism is initiated in response to perturbations that result in an increase in the α-ketoglutarate/citrate ratio. Thus, targeting reductive glutamine conversion for a therapeutic benefit might require distinct modulations of metabolite concentrations rather than targeting the upstream signalling, which only indirectly affects the process.


Assuntos
Células/metabolismo , Ácido Cítrico/metabolismo , Glutamina/metabolismo , Ácidos Cetoglutáricos/metabolismo , Acetatos/metabolismo , Hipóxia Celular , Linhagem Celular Tumoral , Ciclo do Ácido Cítrico , Ácidos Graxos/metabolismo , Humanos , Ácido Láctico/metabolismo , Modelos Biológicos , NAD/metabolismo , Mononucleotídeo de Nicotinamida/metabolismo , Oxirredução , Proteínas Serina-Treonina Quinases/metabolismo , Piruvato Desidrogenase Quinase de Transferência de Acetil
17.
Cancer Res ; 73(14): 4429-38, 2013 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-23687346

RESUMO

Metformin inhibits cancer cell proliferation, and epidemiology studies suggest an association with increased survival in patients with cancer taking metformin; however, the mechanism by which metformin improves cancer outcomes remains controversial. To explore how metformin might directly affect cancer cells, we analyzed how metformin altered the metabolism of prostate cancer cells and tumors. We found that metformin decreased glucose oxidation and increased dependency on reductive glutamine metabolism in both cancer cell lines and in a mouse model of prostate cancer. Inhibition of glutamine anaplerosis in the presence of metformin further attenuated proliferation, whereas increasing glutamine metabolism rescued the proliferative defect induced by metformin. These data suggest that interfering with glutamine may synergize with metformin to improve outcomes in patients with prostate cancer.


Assuntos
Glucose/metabolismo , Glutamina/metabolismo , Metformina/farmacologia , Neoplasias da Próstata/metabolismo , Animais , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Ciclo do Ácido Cítrico/efeitos dos fármacos , Modelos Animais de Doenças , Masculino , Camundongos , Camundongos Transgênicos , Oxirredução , Neoplasias da Próstata/tratamento farmacológico , Neoplasias da Próstata/patologia
18.
Cell Res ; 23(6): 746-58, 2013 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-23689277

RESUMO

Sirtuin enzymes are a family of highly conserved protein deacetylases that depend on nicotinamide adenine dinucleotide (NAD+) for their activity. There are seven sirtuins in mammals and these proteins have been linked with caloric restriction and aging by modulating energy metabolism, genomic stability and stress resistance. Sirtuin enzymes are potential therapeutic targets in a variety of human diseases including cancer, diabetes, inflammatory disorders and neurodegenerative disease. Modulation of sirtuin activity has been shown to impact the course of several aggregate-forming neurodegenerative disorders including Alzheimer's disease, Parkinson's disease, Huntington's disease, amyotrophic lateral sclerosis and spinal and bulbar muscular atrophy. Sirtuins can influence the progression of neurodegenerative disorders by modulating transcription factor activity and directly deacetylating proteotoxic species. Here, we describe sirtuin protein targets in several aggregate-forming neurodegenerative diseases and discuss the therapeutic potential of compounds that modulate sirtuin activity in these disorders.


Assuntos
Metabolismo Energético , Doenças Neurodegenerativas/enzimologia , Doenças Neurodegenerativas/metabolismo , Sirtuínas/metabolismo , Envelhecimento , Restrição Calórica , Histona Desacetilases , Humanos , NAD/metabolismo
19.
Cell Rep ; 3(4): 1175-86, 2013 Apr 25.
Artigo em Inglês | MEDLINE | ID: mdl-23583181

RESUMO

The epithelial-to-mesenchymal transition (EMT) is important for the development of cancer metastases and organ fibrosis, conditions prevalent in aging. Because sirtuins affect the pathology of aging, we tested the effect of SirT1 on EMT. Reduced SIRT1 levels in HMLER breast cancer cells led to increased metastases in nude mice, and the loss of SIRT1 in kidney tubular epithelial cells exacerbated injury-induced kidney fibrosis. SIRT1 reduces EMT in cancer and fibrosis by deacetylating Smad4 and repressing the effect of TGF-ß signaling on MMP7, a Smad4 target gene. Consequently, less E-cadherin is cleaved from the cell surface and ß-catenin remains bound to E-cadherin at the cell-cell junctions. Our findings suggest that the SIRT1/Smad4/ß-catenin axis may be a target for diseases driven by EMT.


Assuntos
Sirtuína 1/metabolismo , Animais , Neoplasias da Mama/metabolismo , Neoplasias da Mama/patologia , Caderinas/metabolismo , Linhagem Celular , Movimento Celular , Transição Epitelial-Mesenquimal , Feminino , Fibrose , Humanos , Rim/patologia , Metaloproteinase 7 da Matriz/química , Metaloproteinase 7 da Matriz/genética , Metaloproteinase 7 da Matriz/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Nus , Camundongos SCID , Camundongos Transgênicos , Metástase Neoplásica , Interferência de RNA , RNA Interferente Pequeno/metabolismo , Transdução de Sinais , Sirtuína 1/antagonistas & inibidores , Sirtuína 1/genética , Proteína Smad4/antagonistas & inibidores , Proteína Smad4/genética , Proteína Smad4/metabolismo , Fator de Crescimento Transformador beta/metabolismo , Células Tumorais Cultivadas , beta Catenina/metabolismo
20.
EMBO Mol Med ; 5(3): 430-40, 2013 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-23364955

RESUMO

Mesenchymal stem cells (MSCs) are multi-potent cells that can differentiate into osteoblasts, adipocytes, chondrocytes and myocytes. This potential declines with aging. We investigated whether the sirtuin SIRT1 had a function in MSCs by creating MSC specific SIRT1 knock-out (MSCKO) mice. Aged MSCKO mice (2.2 years old) showed defects in tissues derived from MSCs; i.e. a reduction in subcutaneous fat, cortical bone thickness and trabecular volume. Young mice showed related but less pronounced effects. MSCs isolated from MSCKO mice showed reduced differentiation towards osteoblasts and chondrocytes in vitro, but no difference in proliferation or apoptosis. Expression of ß-catenin targets important for differentiation was reduced in MSCKO cells. Moreover, while ß-catenin itself (T41A mutant resistant to cytosolic turnover) accumulated in the nuclei of wild-type MSCs, it was unable to do so in MSCKO cells. However, mutating K49R or K345R in ß-catenin to mimic deacetylation restored nuclear localization and differentiation potential in MSCKO cells. We conclude that SIRT1 deacetylates ß-catenin to promote its accumulation in the nucleus leading to transcription of genes for MSC differentiation.


Assuntos
Diferenciação Celular , Células-Tronco Mesenquimais/enzimologia , Sirtuína 1/metabolismo , beta Catenina/metabolismo , Acetilação , Adipócitos/metabolismo , Adipogenia , Fatores Etários , Animais , Apoptose , Blastômeros/metabolismo , Diferenciação Celular/genética , Núcleo Celular/metabolismo , Células Cultivadas , Condrócitos/metabolismo , Condrogênese , Transferência Embrionária , Feminino , Genótipo , Proteínas de Fluorescência Verde/biossíntese , Proteínas de Fluorescência Verde/genética , Masculino , Transplante de Células-Tronco Mesenquimais , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos SCID , Camundongos Transgênicos , Mutação , Osteoblastos/metabolismo , Osteogênese , Fenótipo , Transdução de Sinais , Sirtuína 1/deficiência , Sirtuína 1/genética , Fatores de Tempo , Transcrição Gênica , beta Catenina/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA