Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Bioorg Med Chem ; 26(17): 4823-4840, 2018 09 15.
Artigo em Inglês | MEDLINE | ID: mdl-30181028

RESUMO

The lack of any effective therapy along with the aging world population anticipates a growth of the worldwide incidence of Alzheimer's disease (AD) to more than 100 million cases by 2050. Accumulation of extracellular amyloid-ß (Aß) plaques, intracellular tangles in the brain, and formation of reactive oxygen species (ROS) are the major hallmarks of the disease. In the amyloidogenic process, a ß-secretase, known as BACE 1, plays a fundamental role in the production of Aß fragments, and therefore, inhibition of such enzymes represents a major strategy for the rational design of anti-AD drugs. In this work, a series of four multi-target compounds (1-4), inspired by previously described ionophoric polyphenols, have been synthesized and studied. These compounds have been designed to target important aspects of AD, including BACE 1 enzymatic activity, Aß aggregation, toxic concentrations of Cu2+ metal ions and/or ROS production. Two other compounds (5 and 6), previously reported by some of us as antimalarial agents, have also been studied because of their potential as multi-target species against AD. Interestingly, compounds 3 and 5 showed moderate to good ability to inhibit BACE 1 enzymatic activity in a FRET assay, with IC50's in the low micromolar range (4.4 ±â€¯0.3 and 1.7 ±â€¯0.3 µM, respectively), comparable to other multi-target species, and showing that the observed activity was in part due to a competitive binding of the compounds at the active site of the enzyme. Theoretical docking calculations overall agreed with FRET assay results, displaying the strongest binding affinities for 3 and 5 at the active site of the enzyme. In addition, all compounds selectively interacted with Cu2+ metal ions forming 2:1 complexes, inhibited the production of Aß-Cu2+ catalyzed hydroxyl radicals up to a ∼100% extent, and scavenged AAPH-induced peroxyl radical species comparably to resveratrol, a compound used as reference in this work. Our results also show good anti-amyloidogenic ability: compounds 1-6 inhibited both the Cu2+-induced and self-induced Aß(1-40) fibril aggregation to an extent that ranged from 31% to 77%, while they disaggregated pre-formed Aß(1-40) mature fibrils up to a 37% and a 69% extent in absence and presence of Cu2+, respectively. Cytotoxicity was additionally studied in Tetrahymena thermophila and HEK293 cells, and compared to that of resveratrol, showing that compounds 1-6 display lower toxicity than that of resveratrol, a well-known non-toxic polyphenol.


Assuntos
Doença de Alzheimer/tratamento farmacológico , Antioxidantes/uso terapêutico , Células HEK293 , Humanos , Modelos Teóricos , Simulação de Acoplamento Molecular
2.
Bioorg Med Chem ; 24(16): 3657-70, 2016 08 15.
Artigo em Inglês | MEDLINE | ID: mdl-27316544

RESUMO

Alzheimer's disease (AD) is the most common form of dementia affecting more than 28million people in the world. Only symptomatic treatments are currently available. Anticipated tri-fold increase of AD incidence in the next 50years has established the need to explore new possible treatments. Accumulation of extracellular amyloid-ß (Aß) plaques, intracellular tangles in the brain, and formation of reactive oxygen species (ROS) are the major hallmarks of the disease. The active role of some metal ions, especially Cu(2+), in promoting both Aß aggregation and reactive oxygen species formation has rendered ionophoric drugs as a promising treatment strategy. In this work, a series of 5 disease-modifying and multi-target ionophoric polyphenols (1-5), inspired on the structure of natural resveratrol, have been synthesized and characterized. All compounds bind Cu(2+) selectively over other biologically relevant metal ions. They form 2:1 (compound/Cu(2+)) complexes with association constants logKa 12-14 depending on the molecular design. Our results indicate that compounds 1-5 possess excellent antioxidant properties: they inhibit the Cu(2+)-catalyzed reactive oxygen species production between 47% and 100%, and they scavenge DPPH (1,1-diphenyl-2-picryl-hydrazyl) and AAPH (2,2'-azobis(2-amindino-propane)dihydrochloride) free radicals in general better than clioquinol, resveratrol and ascorbic acid. In addition, compounds 1-5 interact with Aß peptides and inhibit both the Cu(2+)-catalyzed aggregation and the self-assembly of Aß(1-40) up to a ∼92% extent. Interestingly, 1-5 are also able to disaggregate up to ∼91% of pre-formed Aß(1-40) aggregates. Furthermore, cytotoxic studies show remarkably low toxicity of 1-5 toward Tetrahymena thermophila with LD50 values higher than 150µM, comparable to non-toxic natural resveratrol.


Assuntos
Peptídeos beta-Amiloides/antagonistas & inibidores , Antioxidantes/farmacologia , Cobre/metabolismo , Polifenóis/metabolismo , Tetrahymena thermophila/efeitos dos fármacos , Doença de Alzheimer/tratamento farmacológico , Barreira Hematoencefálica , Espectroscopia de Ressonância Magnética Nuclear de Carbono-13 , Polifenóis/farmacologia , Polifenóis/uso terapêutico , Espectroscopia de Prótons por Ressonância Magnética , Espectrometria de Massas por Ionização por Electrospray
3.
Circ Res ; 100(10): 1512-21, 2007 May 25.
Artigo em Inglês | MEDLINE | ID: mdl-17446436

RESUMO

Silent information regulator (Sir)2, a class III histone deacetylase, mediates lifespan extension in model organisms and prevents apoptosis in mammalian cells. However, beneficial functions of Sir2 remain to be shown in mammals in vivo at the organ level, such as in the heart. We addressed this issue by using transgenic mice with heart-specific overexpression of Sirt1, a mammalian homolog of Sir2. Sirt1 was significantly upregulated (4- to 8-fold) in response to pressure overload and oxidative stress in nontransgenic adult mouse hearts. Low (2.5-fold) to moderate (7.5-fold) overexpression of Sirt1 in transgenic mouse hearts attenuated age-dependent increases in cardiac hypertrophy, apoptosis/fibrosis, cardiac dysfunction, and expression of senescence markers. In contrast, a high level (12.5-fold) of Sirt1 increased apoptosis and hypertrophy and decreased cardiac function, thereby stimulating the development of cardiomyopathy. Moderate overexpression of Sirt1 protected the heart from oxidative stress induced by paraquat, with increased expression of antioxidants, such as catalase, through forkhead box O (FoxO)-dependent mechanisms, whereas high levels of Sirt1 increased oxidative stress in the heart at baseline. Thus, mild to moderate expression of Sirt1 retards aging of the heart, whereas a high dose of Sirt1 induces cardiomyopathy. Furthermore, although high levels of Sirt1 increase oxidative stress, moderate expression of Sirt1 induces resistance to oxidative stress and apoptosis. These results suggest that Sirt1 could retard aging and confer stress resistance to the heart in vivo, but these beneficial effects can be observed only at low to moderate doses (up to 7.5-fold) of Sirt1.


Assuntos
Envelhecimento , Miocárdio/metabolismo , Estresse Oxidativo , Sirtuínas/fisiologia , Trifosfato de Adenosina/análise , Animais , Apoptose , Células Cultivadas , Ecocardiografia , Proteína Forkhead Box O1 , Fatores de Transcrição Forkhead/fisiologia , Camundongos , Camundongos Transgênicos , NAD/metabolismo , Ratos , Ratos Wistar , Sirtuína 1 , Sirtuínas/genética , Função Ventricular Esquerda
4.
Circ Res ; 95(10): 971-80, 2004 Nov 12.
Artigo em Inglês | MEDLINE | ID: mdl-15486319

RESUMO

Yeast silent information regulator 2 (Sir2), a nicotinamide adenine dinucleotide-dependent histone deacetylase (HDAC) and founding member of the HDAC class III family, functions in a wide array of cellular processes, including gene silencing, longevity, and DNA damage repair. We examined whether or not the mammalian ortholog Sir2 affects growth and death of cardiac myocytes. Cardiac myocytes express Sir2alpha predominantly in the nucleus. Neonatal rat cardiac myocytes were treated with 20 mmol/L nicotinamide (NAM), a Sir2 inhibitor, or 50 nmol/L Trichostatin A (TSA), a class I and II HDAC inhibitor. NAM induced a significant increase in nuclear fragmentation (2.2-fold) and cleaved caspase-3, as did sirtinol, a specific Sir2 inhibitor, and expression of dominant-negative Sir2alpha. TSA also modestly increased cell death (1.5-fold) but without accompanying caspase-3 activation. Although TSA induced a 1.5-fold increase in cardiac myocyte size and protein content, NAM reduced both. In addition, NAM caused acetylation and increases in the transcriptional activity of p53, whereas TSA did not. NAM-induced cardiac myocyte apoptosis was inhibited in the presence of dominant-negative p53, suggesting that Sir2alpha inhibition causes apoptosis through p53. Overexpression of Sir2alpha protected cardiac myocytes from apoptosis in response to serum starvation and significantly increased the size of cardiac myocytes. Furthermore, Sir2 expression was increased significantly in hearts from dogs with heart failure induced by rapid pacing superimposed on stable, severe hypertrophy. These results suggest that endogenous Sir2alpha plays an essential role in mediating cell survival, whereas Sir2alpha overexpression protects myocytes from apoptosis and causes modest hypertrophy. In contrast, inhibition of endogenous class I and II HDACs primarily causes cardiac myocyte hypertrophy and also induces modest cell death. An increase in Sir2 expression during heart failure suggests that Sir2 may play a cardioprotective role in pathologic hearts in vivo.


Assuntos
Inativação Gênica/fisiologia , Insuficiência Cardíaca/patologia , Hipertrofia Ventricular Esquerda/patologia , Longevidade/genética , Miócitos Cardíacos/enzimologia , Sirtuínas/fisiologia , Acetilação/efeitos dos fármacos , Alcaloides , Animais , Apoptose/efeitos dos fármacos , Apoptose/genética , Apoptose/fisiologia , Fator Natriurético Atrial/biossíntese , Fator Natriurético Atrial/genética , Benzamidas/farmacologia , Benzofenantridinas , Núcleo Celular/enzimologia , Tamanho Celular/efeitos dos fármacos , Sobrevivência Celular , Células Cultivadas/citologia , Células Cultivadas/efeitos dos fármacos , Células Cultivadas/metabolismo , Meios de Cultura Livres de Soro , Inibidores de Cisteína Proteinase/farmacologia , Cães , Genes Dominantes , Genes p53 , Insuficiência Cardíaca/enzimologia , Ventrículos do Coração/citologia , Ácidos Hidroxâmicos/farmacologia , Hipertrofia , Hipertrofia Ventricular Esquerda/enzimologia , Camundongos , Miócitos Cardíacos/citologia , Miócitos Cardíacos/efeitos dos fármacos , Naftóis/farmacologia , Niacinamida/farmacologia , Fenantridinas/farmacologia , Processamento de Proteína Pós-Traducional/efeitos dos fármacos , Processamento de Proteína Pós-Traducional/genética , Ratos , Ratos Wistar , Proteínas Recombinantes de Fusão/fisiologia , Sirtuína 1 , Sirtuínas/antagonistas & inibidores , Sirtuínas/genética , Transcrição Gênica/efeitos dos fármacos , Proteína Supressora de Tumor p53/fisiologia
5.
PLoS One ; 8(1): e51632, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23308102

RESUMO

Autophagy is a bulk degradation mechanism for cytosolic proteins and organelles. The heart undergoes hypertrophy in response to mechanical load but hypertrophy can regress upon unloading. We hypothesize that autophagy plays an important role in mediating regression of cardiac hypertrophy during unloading. Mice were subjected to transverse aortic constriction (TAC) for 1 week, after which the constriction was removed (DeTAC). Regression of cardiac hypertrophy was observed after DeTAC, as indicated by reduction of LVW/BW and cardiomyocyte cross-sectional area. Indicators of autophagy, including LC3-II expression, p62 degradation and GFP-LC3 dots/cell, were significantly increased after DeTAC, suggesting that autophagy is induced. Stimulation of autophagy during DeTAC was accompanied by upregulation of FoxO1. Upregulation of FoxO1 and autophagy was also observed in vitro when cultured cardiomyocytes were subjected to mechanical stretch followed by incubation without stretch (de-stretch). Transgenic mice with cardiac-specific overexpression of FoxO1 exhibited smaller hearts and upregulation of autophagy. Overexpression of FoxO1 in cultured cardiomyocytes significantly reduced cell size, an effect which was attenuated when autophagy was inhibited. To further examine the role of autophagy and FoxO1 in mediating the regression of cardiac hypertrophy, beclin1+/- mice and cultured cardiomyocytes transduced with adenoviruses harboring shRNA-beclin1 or shRNA-FoxO1 were subjected to TAC/stretch followed by DeTAC/de-stretch. Regression of cardiac hypertrophy achieved after DeTAC/de-stretch was significantly attenuated when autophagy was suppressed through downregulation of beclin1 or FoxO1. These results suggest that autophagy and FoxO1 play an essential role in mediating regression of cardiac hypertrophy during mechanical unloading.


Assuntos
Autofagia , Cardiomegalia/fisiopatologia , Coração/fisiopatologia , Animais , Autofagia/efeitos dos fármacos , Cardiomegalia/genética , Cardiomegalia/metabolismo , Cardiomegalia/patologia , Tamanho Celular , Células Cultivadas , Proteína Forkhead Box O1 , Fatores de Transcrição Forkhead/genética , Fatores de Transcrição Forkhead/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Miocárdio/metabolismo , Miocárdio/patologia , Miócitos Cardíacos/metabolismo , Miócitos Cardíacos/patologia , Ratos , Regulação para Cima , Suporte de Carga
6.
Cell Cycle ; 11(5): 856-64, 2012 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-22333581

RESUMO

Heart failure is a leading cause of death worldwide. Estrogen-related receptors (ERRs) are a nuclear receptor subfamily that facilitates the transcription of contractile and nucleus-encoded mitochondrial genes in the heart. Impaired expression of these ERR target genes is frequently observed in human heart failure patients. However, the responsible molecular mechanism is not well-understood. Recently, we have shown that PPARα forms a protein complex with Sirt1, which is involved in the downregulation of ERR targets through direct interaction with the ERR response element (ERRE) in the failing heart. Here, we provide additional lines of evidence supporting the pathological involvement of the PPARα/Sirt1 complex in heart failure. Pressure overload-induced left ventricular (LV) hypertrophy was attenuated in mice with heterozygous knockout of either PPARα (PPARα (+/-) ) or Sirt1 (Sirt1 (+/-) ), whereas cardiac-specific PPARα and Sirt1 bigenic mice showed LV hypertrophy accompanied by a high mortality rate even without pressure overload. Microarray analyses indicated that nuclear-encoded mitochondrial genes were largely downregulated and mitochondrial morphological abnormalities were observed in PPARα/Sirt1 bigenic mice. Those downregulated mitochondrial genes frequently harbor the ERRE in the promoter regions. Artificial and physiological PPARα ligands suppressed reporter genes driven by the ERREs. PPARα bound to and recruited Sirt1 to the genomic flanking region of the ERREs in the heart. Pressure overload downregulated many ERR targets, which were partly normalized by PPARα (+/-) and Sirt1 (+/-) mice. These results suggest that PPARα and Sirt1 downregulate ERR target gene expression through direct interaction with the ERRE in the failing heart.


Assuntos
Insuficiência Cardíaca/metabolismo , PPAR alfa/metabolismo , Receptores de Estrogênio/metabolismo , Sirtuína 1/metabolismo , Animais , Regulação para Baixo , Insuficiência Cardíaca/patologia , Humanos , Hipertrofia Ventricular Esquerda/metabolismo , Hipertrofia Ventricular Esquerda/mortalidade , Hipertrofia Ventricular Esquerda/patologia , Camundongos , Mitocôndrias/metabolismo , Mitocôndrias/patologia , PPAR alfa/genética , Regiões Promotoras Genéticas , Receptores de Estrogênio/genética , Elementos de Resposta , Sirtuína 1/genética , Transcrição Gênica
7.
Cell Metab ; 14(5): 598-611, 2011 Nov 02.
Artigo em Inglês | MEDLINE | ID: mdl-22055503

RESUMO

High energy production in mitochondria is essential for maintaining cardiac contraction in the heart. Genes regulating mitochondrial function are commonly downregulated during heart failure. Here we show that both PPARα and Sirt1 are upregulated by pressure overload in the heart. Haploinsufficiency of either PPARα or Sirt1 attenuated pressure overload-induced cardiac hypertrophy and failure, whereas simultaneous upregulation of PPARα and Sirt1 exacerbated the cardiac dysfunction. PPARα and Sirt1 coordinately suppressed genes involved in mitochondrial function that are regulated by estrogen-related receptors (ERRs). PPARα bound and recruited Sirt1 to the ERR response element (ERRE), thereby suppressing ERR target genes in an RXR-independent manner. Downregulation of ERR target genes was also observed during fasting, and this appeared to be an adaptive response of the heart. These results suggest that suppression of the ERR transcriptional pathway by PPARα/Sirt1, a physiological fasting response, is involved in the progression of heart failure by promoting mitochondrial dysfunction.


Assuntos
Cardiomegalia/metabolismo , Insuficiência Cardíaca/metabolismo , Miocárdio/metabolismo , PPAR alfa/metabolismo , Receptores de Estrogênio/metabolismo , Transdução de Sinais/genética , Sirtuína 1/metabolismo , Transcrição Gênica , Adaptação Fisiológica , Animais , Aorta/fisiopatologia , Aorta/cirurgia , Sítios de Ligação , Cardiomegalia/genética , Cardiomegalia/fisiopatologia , Jejum/metabolismo , Regulação da Expressão Gênica , Haploinsuficiência , Haplótipos , Insuficiência Cardíaca/genética , Insuficiência Cardíaca/fisiopatologia , Camundongos , Camundongos Transgênicos , Mitocôndrias Cardíacas , Miocárdio/patologia , Técnicas de Cultura de Órgãos , PPAR alfa/genética , Ligação Proteica , Receptores de Estrogênio/genética , Sirtuína 1/genética , Regulação para Cima
8.
Autophagy ; 5(8): 1229-31, 2009 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-19855187

RESUMO

Nicotinamide adenine dinucleotide (NAD(+)) acts as a transfer molecule for electrons, thereby acting as a key cofactor for energy production. NAD(+) also serves as a substrate for cellular enzymes, including poly (ADPribose) polymerase (PARP)-1 and Sirt1. Activation of PARP-1 by DNA damage depletes the cellular pool of NAD(+), leading to necrotic cell death. NAD(+) in the nucleus enhances the activity of Sirt1, thereby modulating transcription. NAD(+) is either synthesized de novo from amino acids, namely tryptophan and aspartic acid, or resynthesized from NAD(+) metabolites, such as nicotinamide (NAM), through the salvage pathway. NAM phosphoribosyltransferase (Nampt) is a rate-limiting enzyme in the NAD(+) salvage pathway. We have recently demonstrated that Nampt is an important regulator of NAD(+) and autophagy in cardiomyocytes. Here we discuss the role of Nampt in regulating autophagy and potential mechanisms by which NAD(+) regulates autophagy in the heart.


Assuntos
Autofagia , Miócitos Cardíacos/citologia , Miócitos Cardíacos/enzimologia , Nicotinamida Fosforribosiltransferase/metabolismo , Animais , Linhagem Celular Tumoral , Sobrevivência Celular , Transporte de Elétrons , Humanos , Camundongos , Modelos Biológicos , NAD/metabolismo
9.
Biol Chem ; 389(3): 221-31, 2008 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-18208353

RESUMO

The prevalence of heart diseases, such as coronary artery disease and congestive heart failure, increases with age. Optimal therapeutic interventions that antagonize aging may reduce the occurrence and mortality of adult heart diseases. We discuss here how molecular mechanisms mediating life span extension affect aging of the heart and its resistance to pathological insults. In particular, we review our recent findings obtained from transgenic mice with cardiac-specific overexpression of Sirt1, which demonstrated delayed aging and protection against oxidative stress in the heart. We propose that activation of known longevity mechanisms in the heart may represent a novel cardioprotection strategy against aging and certain types of cardiac stress, such as oxidative stress.


Assuntos
Envelhecimento/efeitos dos fármacos , Cardiopatias/prevenção & controle , Coração/efeitos dos fármacos , Coração/fisiologia , Sirtuínas/fisiologia , Animais , Apoptose/efeitos dos fármacos , Restrição Calórica , Insuficiência Cardíaca/tratamento farmacológico , Humanos , Longevidade/fisiologia , Camundongos , Nicotinamida Fosforribosiltransferase/metabolismo , Estresse Oxidativo/efeitos dos fármacos , Resveratrol , Saccharomyces cerevisiae/efeitos dos fármacos , Sirtuína 1 , Estilbenos/farmacologia , Regulação para Cima
10.
J Mol Cell Cardiol ; 40(5): 605-18, 2006 May.
Artigo em Inglês | MEDLINE | ID: mdl-16427076

RESUMO

Myocardial ischemia/reperfusion (IR) induces myocyte apoptosis, and the pro-apoptotic/tumor suppressor protein p53 may contribute to this process. However, the signaling mechanism by which IR induces p53 activation remains largely unknown. Here, we show that MEKK1 undergoes proteolytic cleavage in a caspase-3 dependent manner in both in vivo and in vitro models of ischemic injury. Overexpression studies both in vivo and in vitro indicated that the caspase-3 mediated cleavage of MEKK1 promotes phosphorylation and transcriptional activity of p53. In addition, caspase-3 inhibited the ability of the wild-type full-length form of MEKK1 to activate ATF2, suggesting that caspase-3, by way of proteolytic cleavage, abrogates the ability of MEKK1 to signal JNK. We propose that IR induces caspase-3 mediated proteolytic cleavage of MEKK1 and promotes p53 transcriptional activity via JNK-independent mechanisms, which in turn may contribute to pathological insults associated with IR injury, such as myocyte apoptosis.


Assuntos
Caspases/metabolismo , MAP Quinase Quinase Quinase 1/metabolismo , Transcrição Gênica , Proteína Supressora de Tumor p53/metabolismo , Fator 2 Ativador da Transcrição/metabolismo , Animais , Caspase 3 , Células Cultivadas , Isquemia/patologia , Luciferases/metabolismo , Fosforilação , Ratos , Ratos Wistar , Traumatismo por Reperfusão , Transdução de Sinais
11.
J Biol Chem ; 281(28): 19369-77, 2006 Jul 14.
Artigo em Inglês | MEDLINE | ID: mdl-16687393

RESUMO

In this study we have shown that the histone variant H2A.z is up-regulated during cardiac hypertrophy. Upon its knock-down with RNA interference, hypertrophy and the underlying increase in growth-related genes, protein synthesis, and cell size were down-regulated. During attempts to understand the mode of regulation of H2A.z, we found that overexpression of silent information regulator 2alpha (Sir2alpha) specifically induced down-regulation of H2A.z via NAD-dependent activity. This effect was reversed by the proteasome inhibitor epoxomicin, suggesting a Sir2alpha-mediated ubiquitin/proteasome-dependent mechanism for degradation of H2A.z. An increase in Sir2alpha also resulted in a dose-dependent reduction of the response to hypertrophic stimuli, whereas its inhibition resulted in enhanced hypertrophy and apoptosis. We have shown that Sir2alpha directly deacetylates H2A.z. Mutagenesis proved that lysines 4, 7, 11, and 13 do not play a role in the stability of H2A.z, whereas Lys-15 was indispensable. Meanwhile, Lys-115 and conserved, ubiquitinatable Lys-121 are critical for Sir2alpha-mediated degradation. Fusion of the C terminus of H2A.z (amino acids 115-127) to H2A.x or green fluorescence protein conferred Sir2alpha-inducible degradation to the former protein only. Because H2A.x and H2A.z have conserved N-tails, this implied that both the C and N termini are critical for mediating the effect of Sir2alpha. In short, the results suggest that H2A.z is required for cardiac hypertrophy, where its stability and the extent of cell growth and apoptosis are moderated by Sir2alpha. We also propose that Sir2alpha is involved in deacetylation of H2A.z, which results in ubiquitination of Lys-115 and Lys-121 and its degradation via a ubiquitin/proteasome-dependent pathway.


Assuntos
Regulação da Expressão Gênica , Histonas/fisiologia , Células Musculares/metabolismo , Sirtuínas/fisiologia , Animais , Apoptose , Histonas/metabolismo , Hipertrofia , Lisina/química , Camundongos , Mutagênese , Estrutura Terciária de Proteína , Ratos , Ratos Sprague-Dawley , Sirtuína 1 , Sirtuínas/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA