Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 43
Filtrar
1.
Int J Mol Sci ; 24(10)2023 May 20.
Artigo em Inglês | MEDLINE | ID: mdl-37240382

RESUMO

All-trans retinoic acid (ATRA) promotes myoblast differentiation into myotubes. Leucine-rich repeat-containing G-protein-coupled receptor 6 (LGR6) is a candidate ATRA-responsive gene; however, its role in skeletal muscles remains unclear. Here, we demonstrated that during the differentiation of murine C2C12 myoblasts into myotubes, Lgr6 mRNA expression transiently increased before the increase in the expression of the mRNAs encoding myogenic regulatory factors, such as myogenin, myomaker, and myomerger. The loss of LGR6 decreased the differentiation and fusion indices. The exogenous expression of LGR6 up to 3 and 24 h after the induction of differentiation increased and decreased the mRNA levels of myogenin, myomaker, and myomerger, respectively. Lgr6 mRNA was transiently expressed after myogenic differentiation in the presence of a retinoic acid receptor α (RARα) agonist and an RARγ agonist in addition to ATRA, but not in the absence of ATRA. Furthermore, a proteasome inhibitor or Znfr3 knockdown increased exogenous LGR6 expression. The loss of LGR6 attenuated the Wnt/ß-catenin signaling activity induced by Wnt3a alone or in combination with Wnt3a and R-spondin 2. These results indicate that LGR6 promotes myogenic differentiation and that ATRA is required for the transient expression of LGR6 during differentiation. Furthermore, LGR6 expression appeared to be downregulated by the ubiquitin-proteasome system involving ZNRF3.


Assuntos
Tretinoína , Via de Sinalização Wnt , Camundongos , Animais , Miogenina/genética , Miogenina/metabolismo , Tretinoína/farmacologia , Tretinoína/metabolismo , Mioblastos/metabolismo , RNA Mensageiro/genética , Diferenciação Celular/genética , Receptores Acoplados a Proteínas G/genética , Receptores Acoplados a Proteínas G/metabolismo
2.
Br J Nutr ; 126(4): 481-491, 2021 08 28.
Artigo em Inglês | MEDLINE | ID: mdl-33143796

RESUMO

Skeletal muscle atrophy causes decreased physical activity and increased risk of metabolic diseases. We investigated the effects of oleamide (cis-9,10-octadecanamide) treatment on skeletal muscle health. The plasma concentration of endogenous oleamide was approximately 30 nm in male ddY mice under normal physiological conditions. When the stable isotope-labelled oleamide was orally administered to male ddY mice (50 mg/kg), the plasma concentration of exogenous oleamide reached approximately 170 nm after 1 h. Male ddY mice were housed in small cages (one-sixth of normal size) to enforce sedentary behaviour and orally administered oleamide (50 mg/kg per d) for 4 weeks. Housing in small cages decreased tibialis anterior (TA) muscle mass and the cross-sectional area of the myofibres in TA muscle. Dietary oleamide alleviated the decreases in TA muscle and resulted in plasma oleamide concentration of approximately 120 nm in mice housed in small cages. Housing in small cages had no influence on the phosphorylation levels of Akt serine/threonine kinase (Akt), mechanistic target of rapamycin (mTOR) and ribosomal protein S6 kinase (p70S6K) in TA muscle; nevertheless, oleamide increased the phosphorylation levels of the proteins. Housing in small cages increased the expression of microtubule-associated protein 1 light chain 3 (LC3)-II and sequestosome 1 (p62), but not LC3-I, in TA muscle, and oleamide reduced LC3-I, LC3-II and p62 expression levels. In C2C12 myotubes, oleamide increased myotube diameter at ≥100 nm. Furthermore, the mTOR inhibitor, Torin 1, suppressed oleamide-induced increases in myotube diameter and protein synthesis. These results indicate that dietary oleamide rescued TA muscle atrophy in mice housed in small cages, possibly by activating the phosphoinositide 3-kinase/Akt/mTOR signalling pathway and restoring autophagy flux.


Assuntos
Músculo Esquelético/efeitos dos fármacos , Atrofia Muscular , Ácidos Oleicos/farmacologia , Fosfatidilinositol 3-Quinases , Animais , Autofagia , Abrigo para Animais , Masculino , Camundongos , Fibras Musculares Esqueléticas/metabolismo , Músculo Esquelético/metabolismo , Atrofia Muscular/tratamento farmacológico , Fosfatidilinositol 3-Quinases/metabolismo , Fosforilação , Proteínas Proto-Oncogênicas c-akt/metabolismo , Serina-Treonina Quinases TOR/metabolismo
3.
Nutrients ; 12(12)2020 Dec 18.
Artigo em Inglês | MEDLINE | ID: mdl-33353230

RESUMO

Several recent experimental studies have investigated the effects of caffeine and chlorogenic acid (CGA), representative ingredients of coffee, on nonalcoholic fatty liver disease (NAFLD)/nonalcoholic steatohepatitis (NASH). However, the results are conflicting, and their effects are yet to be clarified. In the present study, we examined the effects of caffeine and CGA on choline-deficient, L-amino acid-defined, high-fat diet (CDAHFD)-fed mice, relatively new model mice of NASH. Seven-week-old male C57BL/6J mice were divided into the following groups: Control diet (control), CDAHFD (CDAHFD), CDAHFD supplemented with 0.05% (w/w) caffeine (caffeine), and CDAHFD supplemented with 0.1% (w/w) CGA (CGA). After seven weeks, the mice were killed and serum biochemical, histopathological, and molecular analyses were performed. Serum alanine aminotransferase (ALT) levels were significantly higher in the caffeine and CGA groups than in the CDAHFD group. On image analysis, the prevalence of Oil red O-positive areas (reflecting steatosis) was significantly higher in the caffeine group than in the CDAHFD group, and that of CD45R-positive areas (reflecting lymphocytic infiltration) in the hepatic lobule was significantly higher in the caffeine and CGA groups than in the CDAHFD group. Hepatic expression of interleukin (IL)-6 mRNA was higher in the caffeine and CGA groups than in the CDAHFD group, and the difference was statistically significant for the caffeine group. In conclusion, in the present study, caffeine and CGA significantly worsened the markers of liver cell injury, inflammation, and/or steatosis in NASH lesions in mice.


Assuntos
Cafeína/farmacologia , Ácido Clorogênico/farmacologia , Dieta Hiperlipídica , Hepatopatia Gordurosa não Alcoólica/tratamento farmacológico , Alanina Transaminase/sangue , Aminoácidos , Animais , Deficiência de Colina , Ingestão de Alimentos , Ingestão de Energia , Interleucina-6/genética , Interleucina-6/metabolismo , Antígenos Comuns de Leucócito/análise , Fígado/diagnóstico por imagem , Fígado/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Hepatopatia Gordurosa não Alcoólica/diagnóstico por imagem , Hepatopatia Gordurosa não Alcoólica/metabolismo , Hepatopatia Gordurosa não Alcoólica/patologia , RNA Mensageiro/metabolismo
4.
Molecules ; 25(14)2020 Jul 17.
Artigo em Inglês | MEDLINE | ID: mdl-32709013

RESUMO

Adenyl cobamide (commonly known as pseudovitamin B12) is synthesized by intestinal bacteria or ingested from edible cyanobacteria. The effect of pseudovitamin B12 on the activities of cobalamin-dependent enzymes in mammalian cells has not been studied well. This study was conducted to investigate the effects of pseudovitamin B12 on the activities of the mammalian vitamin B12-dependent enzymes methionine synthase and methylmalonyl-CoA mutase in cultured mammalian COS-7 cells to determine whether pseudovitamin B12 functions as an inhibitor or a cofactor of these enzymes. Although the hydoroxo form of pseudovitamin B12 functions as a coenzyme for methionine synthase in cultured cells, pseudovitamin B12 does not activate the translation of methionine synthase, unlike the hydroxo form of vitamin B12 does. In the second enzymatic reaction, the adenosyl form of pseudovitamin B12 did not function as a coenzyme or an inhibitor of methylmalonyl-CoA mutase. Experiments on the cellular uptake were conducted with human transcobalamin II and suggested that treatment with a substantial amount of pseudovitamin B12 might inhibit transcobalamin II-mediated absorption of a physiological trace concentration of vitamin B12 present in the medium.


Assuntos
5-Metiltetra-Hidrofolato-Homocisteína S-Metiltransferase/genética , Metilmalonil-CoA Mutase/genética , Vitamina B 12/análogos & derivados , Vitamina B 12/metabolismo , Animais , Transporte Biológico/efeitos dos fármacos , Células COS , Chlorocebus aethiops , Coenzimas/genética , Regulação Enzimológica da Expressão Gênica/efeitos dos fármacos , Humanos , Vitamina B 12/genética , Vitamina B 12/farmacologia
5.
Biochim Biophys Acta Mol Cell Res ; 1867(2): 118563, 2020 02.
Artigo em Inglês | MEDLINE | ID: mdl-31666191

RESUMO

Skeletal muscle secretes biologically active proteins that contribute to muscle hypertrophy in response to either exercise or dietary intake. The identification of skeletal muscle-secreted proteins that induces hypertrophy can provide critical information regarding skeletal muscle health. Dietary provitamin A, ß-carotene, induces hypertrophy of the soleus muscle in mice. Here, we hypothesized that skeletal muscle produces hypertrophy-inducible secretory proteins via dietary ß-carotene. Knockdown of retinoic acid receptor (RAR) γ inhibited the ß-carotene-induced increase soleus muscle mass in mice. Using RNA sequencing, bioinformatic analyses, and literature searching, we predicted transglutaminase 2 (TG2) to be an all-trans retinoic acid (ATRA)-induced secretory protein in cultured C2C12 myotubes. Tg2 mRNA expression increased in ATRA- or ß-carotene-stimulated myotubes and in the soleus muscle of ß-carotene-treated mice. Knockdown of RARγ inhibited ß-carotene-increased mRNA expression of Tg2 in the soleus muscle. ATRA increased endogenous TG2 levels in conditioned medium from myotubes. Extracellular TG2 promoted the phosphorylation of Akt, mechanistic target of rapamycin (mTOR), and ribosomal p70 S6 kinase (p70S6K), and inhibitors of mTOR, phosphatidylinositol 3-kinase, and Src (rapamycin, LY294002, and Src I1, respectively) inhibited TG2-increased phosphorylation of mTOR and p70S6K. Furthermore, extracellular TG2 promoted protein synthesis and hypertrophy in myotubes. TG2 mutant lacking transglutaminase activity exerted the same effects as wild-type TG2. Knockdown of G protein-coupled receptor 56 (GPR56) inhibited the effects of TG2 on mTOR signaling, protein synthesis, and hypertrophy. These results indicated that TG2 expression was upregulated through ATRA-mediated RARγ and that extracellular TG2 induced myotube hypertrophy by activating mTOR signaling-mediated protein synthesis through GPR56, independent of transglutaminase activity.


Assuntos
Proteínas de Ligação ao GTP/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Transglutaminases/metabolismo , Animais , Crescimento Celular/efeitos dos fármacos , Linhagem Celular , Proteínas de Ligação ao GTP/genética , Fator de Crescimento Insulin-Like I/genética , Fator de Crescimento Insulin-Like I/metabolismo , Camundongos , Músculo Esquelético/efeitos dos fármacos , Músculo Esquelético/metabolismo , Mioblastos/citologia , Mioblastos/metabolismo , Fosforilação/efeitos dos fármacos , Proteína 2 Glutamina gama-Glutamiltransferase , Proteínas Proto-Oncogênicas c-akt/metabolismo , Interferência de RNA , RNA Interferente Pequeno/metabolismo , Receptores Acoplados a Proteínas G/antagonistas & inibidores , Receptores Acoplados a Proteínas G/genética , Receptores do Ácido Retinoico/antagonistas & inibidores , Receptores do Ácido Retinoico/genética , Receptores do Ácido Retinoico/metabolismo , Receptor alfa de Ácido Retinoico/antagonistas & inibidores , Receptor alfa de Ácido Retinoico/genética , Receptor alfa de Ácido Retinoico/metabolismo , Proteínas Quinases S6 Ribossômicas 70-kDa/metabolismo , Transdução de Sinais/efeitos dos fármacos , Serina-Treonina Quinases TOR/metabolismo , Transglutaminases/genética , Tretinoína/farmacologia , beta Caroteno/administração & dosagem , beta Caroteno/farmacologia , Receptor gama de Ácido Retinoico
6.
Arch Biochem Biophys ; 646: 98-106, 2018 05 15.
Artigo em Inglês | MEDLINE | ID: mdl-29625124

RESUMO

Adipose tissues in obese individuals are characterized by a state of chronic low-grade inflammation. Pre-adipocytes and adipocytes in this state secrete pro-inflammatory adipokines, such as interleukin 6 (IL-6), which induce insulin resistance and hyperglycemia. Theophylline (1,3-dimethylxanthine) exerts anti-inflammatory effects, but its effects on pro-inflammatory adipokine secretion by pre-adipocytes and adipocytes have not been examined. In this study, we found that theophylline decreased IL-6 secretion by 3T3-L1 pre-adipocytes and mouse-derived primary pre-adipocytes. The synthetic glucocorticoid dexamethasone (DEX) induced IL-6 expression in 3T3-L1 pre-adipocytes, and this effect was suppressed by theophylline at the mRNA level. Knockdown of CCAAT/enhancer binding protein (C/EBP) δ inhibited DEX-induced IL-6 expression, and theophylline suppressed C/EBPδ expression. Furthermore, theophylline suppressed transcriptional activity of the glucocorticoid receptor (GR) through suppression of nuclear localization of GR. In vivo, glucocorticoid corticosterone treatment (100 µg/mL) increased fasting blood glucose and plasma IL-6 levels in C57BL/6 N mice. Theophylline administration (0.1% diet) reduced corticosterone-increased fasting blood glucose, plasma IL-6 levels, and Il6 gene expression in adipose tissues. These results show that theophylline administration attenuated glucocorticoid-induced hyperglycemia and IL-6 production by inhibiting GR activity. The present findings indicate the potential of theophylline as a candidate therapeutic agent to treat insulin resistance and hyperglycemia.


Assuntos
Adipócitos/efeitos dos fármacos , Interleucina-6/metabolismo , Receptores de Glucocorticoides/metabolismo , Transdução de Sinais/efeitos dos fármacos , Teofilina/farmacologia , Células 3T3-L1 , Animais , Proteína delta de Ligação ao Facilitador CCAAT/metabolismo , Núcleo Celular/metabolismo , Dexametasona/farmacologia , Interleucina-6/sangue , Interleucina-6/genética , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Regiões Promotoras Genéticas/efeitos dos fármacos
7.
Mol Med Rep ; 17(4): 5912-5920, 2018 04.
Artigo em Inglês | MEDLINE | ID: mdl-29436684

RESUMO

Lactoferrin (Lf) is a multifunctional glycoprotein, which promotes the proliferation of murine C2C12 myoblasts. In the present study, it was investigated how Lf promotes myoblast proliferation and whether Lf promotes myoblast differentiation or induces myotube hypertrophy. Lf promoted the proliferation of myoblasts in a dose­dependent manner. Myoblast proliferation increased on day 3 when myoblasts were cultured in the presence of Lf for three days and also when myoblasts were cultured in the presence of Lf for the first day and in the absence of Lf for the subsequent two days. In addition, Lf induced the phosphorylation of extracellular signal­regulated kinase (ERK)1/2 in myoblasts. The mitogen­activated protein kinase kinase 1/2 inhibitor U0126 inhibited Lf­induced ERK1/2 phosphorylation and repressed Lf­promoted myoblast proliferation. C2C12 myoblasts, myotubes and skeletal muscle expressed low­density lipoprotein receptor­related protein (LRP)1 mRNA and Lf­promoted myoblast proliferation was attenuated by an LRP1 antagonist or LRP1 gene silencing. The knockdown of LRP1 repressed Lf­induced phosphorylation of ERK1/2. Furthermore, when myoblasts were induced to differentiate, Lf increased the expression of the myotube­specific structural protein, myosin heavy chain (MyHC) and promoted myotube formation. Knockdown of LRP1 repressed Lf­induced MyHC expression. Lf also increased myotube size following differentiation. These results indicate that Lf promotes myoblast proliferation and differentiation, at least partially through LRP1 and also stimulates myotube hypertrophy.


Assuntos
Diferenciação Celular/efeitos dos fármacos , Lactoferrina/farmacologia , Fibras Musculares Esqueléticas/efeitos dos fármacos , Fibras Musculares Esqueléticas/metabolismo , Mioblastos/efeitos dos fármacos , Mioblastos/metabolismo , Animais , Linhagem Celular , Proliferação de Células/efeitos dos fármacos , Hipertrofia , Proteína-1 Relacionada a Receptor de Lipoproteína de Baixa Densidade , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Masculino , Camundongos , Proteína Quinase 3 Ativada por Mitógeno/metabolismo , Fibras Musculares Esqueléticas/citologia , Mioblastos/citologia , Receptores de LDL/metabolismo , Proteínas Supressoras de Tumor/metabolismo
8.
J Nutr Biochem ; 49: 63-70, 2017 11.
Artigo em Inglês | MEDLINE | ID: mdl-28886438

RESUMO

Ubiquitin-specific protease 19 (USP19) is a key player in the negative regulation of muscle mass during muscle atrophy. Loss-of-function approaches demonstrate that 17ß-estradiol (E2) increases USP19 expression through estrogen receptor (ER) α and consequently decreases soleus muscle mass in young female mice under physiological conditions. Daidzein is one of the main isoflavones in soy, and activates ERß-dependent transcription. Here, we investigated the effects of daidzein on E2-increased USP19 expression and E2-decreased soleus muscle mass in young female mice. Daidzein stimulated the transcriptional activity of ERß in murine C2C12 cells and down-regulated USP19 expression. Consistently, daidzein inhibited E2-induced USP19 expression in a reporter activity using a functional half-estrogen response element (hERE) from Usp19. Daidzein inhibited E2-induced recruitment of ERα and promoted recruitment of ERß to the Usp19 hERE. Dietary daidzein down-regulated the expression of USP19 at the mRNA and protein levels and increased soleus muscle mass in female mice, but not in males. In soleus muscle from ovariectomized (OVX) female mice, dietary daidzein inhibited E2-increased USP19 mRNA expression and E2-decreased muscle mass. Furthermore, E2 induced the recruitment of ERα and ERß to the hERE, whereas daidzein inhibited E2-induced recruitment of ERα, and enhanced E2-increased recruitment of ERß, to the Usp19 hERE. These results demonstrate that dietary daidzein decreases USP19 mRNA expression through ERß and increases soleus muscle mass in young female mice, but not in male mice, under physiological conditions.


Assuntos
Suplementos Nutricionais , Receptor beta de Estrogênio/agonistas , Isoflavonas/uso terapêutico , Músculo Esquelético/metabolismo , Fitoestrógenos/uso terapêutico , Sarcopenia/prevenção & controle , Proteases Específicas de Ubiquitina/antagonistas & inibidores , Transporte Ativo do Núcleo Celular , Animais , Animais não Endogâmicos , Linhagem Celular , Endopeptidases , Repressão Enzimática , Receptor beta de Estrogênio/genética , Receptor beta de Estrogênio/metabolismo , Feminino , Genes Reporter , Masculino , Camundongos , Músculo Esquelético/enzimologia , Músculo Esquelético/patologia , Mioblastos Esqueléticos/citologia , Mioblastos Esqueléticos/enzimologia , Mioblastos Esqueléticos/metabolismo , Mioblastos Esqueléticos/patologia , Ovariectomia/efeitos adversos , Distribuição Aleatória , Elementos de Resposta , Sarcopenia/etiologia , Sarcopenia/metabolismo , Sarcopenia/patologia , Caracteres Sexuais , Transdução de Sinais , Proteases Específicas de Ubiquitina/genética , Proteases Específicas de Ubiquitina/metabolismo
9.
Cell Biol Int ; 41(12): 1325-1334, 2017 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-28833753

RESUMO

Dermal fibroblasts generate the extracellular matrix component elastin, which is synthesized as tropoelastin (TE) and play a critical role in maintaining skin elasticity. Lactoferrin (Lf), an 80-kDa iron-binding glycoprotein, has biological functions such as anti-bacterial, -inflammatory, and -cancer activities. We previously reported that bovine Lf increases TE mRNA expression in human dermal fibroblasts. However, it remains unclear how Lf up-regulates TE expression. Here, we investigated molecular mechanisms underlying this effect. Lf promoted the phosphorylation of Akt1 and extracellular signal-regulated protein kinase (ERK)1/2. As expected, the phosphatidylinositol 3-kinase (PI3K) inhibitor LY294002 and the MAPK inhibitor U0126 inhibited Lf-induced phosphorylation of Akt1 and ERK1/2, respectively. In contrast, LY294002, but not U0126, inhibited Lf-induced TE expression. Human dermal fibroblasts expressed lipoprotein receptor-related protein 1 (LRP-1) mRNA, and the LRP1 inhibitor receptor-associated protein attenuated Lf-induced increases in TE expression. Furthermore, siRNA-mediated knockdown of LRP-1 significantly suppressed Lf-increased TE expression and Lf-induced Akt1 phosphorylation. Iron-saturated Lf (holo-Lf) increased TE expression and promoted Akt1 phosphorylation, when compared to those parameters in cells treated with iron-free Lf (apo-Lf). Transforming growth factor (TGF)-ß1 also increased TE expression. LY294002 inhibited TGF-ß1-mediated TE upregulation, whereas TGF-ß1 activated Akt2, but not Akt1, phosphorylation. These results indicate that holo-Lf, but not apo-Lf, increases TE expression through LRP-1 in human dermal fibroblasts and suggest that holo-Lf and TGF-ß1 enhance TE expression by activating the PI3K/Akt1 and PI3K/Akt2 pathways, respectively.


Assuntos
Lactoferrina/metabolismo , Proteína-1 Relacionada a Receptor de Lipoproteína de Baixa Densidade/metabolismo , Fosfatidilinositol 3-Quinase/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Tropoelastina/biossíntese , Células Cultivadas , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Fibroblastos/citologia , Fibroblastos/metabolismo , Humanos , Lactoferrina/farmacologia , Proteína-1 Relacionada a Receptor de Lipoproteína de Baixa Densidade/genética , Sistema de Sinalização das MAP Quinases , Fosforilação/efeitos dos fármacos , Ligação Proteica , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Pele/citologia , Pele/metabolismo , Fator de Crescimento Transformador beta1/metabolismo , Fator de Crescimento Transformador beta1/farmacologia , Tropoelastina/metabolismo
10.
J Nutr ; 147(9): 1631-1639, 2017 09.
Artigo em Inglês | MEDLINE | ID: mdl-28768836

RESUMO

Background:S-equol, which is enantioselectively produced from daidzein by gut microbiota, has been suggested as a chemopreventive agent against type 2 diabetes mellitus (T2DM), but the underlying mechanisms remain unclear.Objective: We investigated the effects of S-equol on pancreatic ß-cell function.Methods: ß-Cell growth and insulin secretion were evaluated with male Institute of Cancer Research mice and isolated pancreatic islets from the mice, respectively. The mechanisms by which S-equol stimulated ß-cell response were examined in INS-1 ß-cells. The effect of S-equol treatment on ß-cell function was assessed in low-dose streptozotocin-treated mice. S-equol was used at 10 µmol/L for in vitro and ex vivo studies and was administered by oral gavage (20 mg/kg, 2 times/d throughout the experimental period) for in vivo studies.Results:S-equol administration for 7 d increased Ki67-positive ß-cells by 27% (P < 0.01) in mice. S-equol enantioselectively enhanced glucose-stimulated insulin secretion in mouse pancreatic islets by 41% (P < 0.001). In INS-1 cells, S-equol exerted stronger effects than daidzein on cell growth, insulin secretion, and cAMP-response element (CRE)-mediated transcription. These S-equol effects were diminished by inhibiting protein kinase A. The effective concentration of S-equol for stimulating cAMP production at the plasma membrane was lower than that for phosphodiesterase inhibition. S-equol-stimulated CRE activation was negatively controlled by the knockdown of G-protein α subunit group S (stimulatory) and positively controlled by that of G-protein-coupled receptor kinase-3 and -6. Compared with vehicle-treated controls, S-equol gavage treatment resulted in an increase in ß-cell mass of 104% (P < 0.05), a trend toward high plasma insulin concentrations (by 118%; P = 0.06), and resistance to hyperglycemia after streptozotocin treatment (78% of AUC after glucose challenge; P < 0.01). S-equol administration significantly increased the number of Ki67-positive proliferating ß-cells by 62% (P < 0.01) and decreased that of terminal deoxynucleotidyl transferase-mediated dUTP nick-end labeling-positive apoptotic ß-cells by 75% (P < 0.05).Conclusions: Our results show that S-equol boosts ß-cell function and prevents hypoglycemia in mice, suggesting its potential for T2DM prevention.


Assuntos
Glicemia/metabolismo , Membrana Celular/efeitos dos fármacos , AMP Cíclico/metabolismo , Diabetes Mellitus Experimental/tratamento farmacológico , Equol/farmacologia , Células Secretoras de Insulina/efeitos dos fármacos , Insulina/metabolismo , Animais , Área Sob a Curva , Crescimento Celular/efeitos dos fármacos , Membrana Celular/metabolismo , Diabetes Mellitus Experimental/sangue , Diabetes Mellitus Experimental/patologia , Diabetes Mellitus Tipo 2/sangue , Diabetes Mellitus Tipo 2/prevenção & controle , Hiperglicemia/sangue , Hiperglicemia/induzido quimicamente , Hiperglicemia/etiologia , Hiperglicemia/prevenção & controle , Secreção de Insulina , Células Secretoras de Insulina/citologia , Células Secretoras de Insulina/metabolismo , Isoflavonas/metabolismo , Isoflavonas/farmacologia , Masculino , Camundongos Endogâmicos ICR , Ratos , Transdução de Sinais
11.
Biochem Biophys Res Commun ; 478(3): 1292-7, 2016 09 23.
Artigo em Inglês | MEDLINE | ID: mdl-27553280

RESUMO

The majority of studies on possible roles for collagen hydrolysates in human health have focused on their effects on bone and skin. Hydroxyprolyl-glycine (Hyp-Gly) was recently identified as a novel collagen hydrolysate-derived dipeptide in human blood. However, any possible health benefits of Hyp-Gly remain unclear. Here, we report the effects of Hyp-Gly on differentiation and hypertrophy of murine skeletal muscle C2C12 cells. Hyp-Gly increased the fusion index, the myotube size, and the expression of the myotube-specific myosin heavy chain (MyHC) and tropomyosin structural proteins. Hyp-Gly increased the phosphorylation of Akt, mTOR, and p70S6K in myoblasts, whereas the phosphatidylinositol 3-kinase (PI3K) inhibitor LY294002 inhibited their phosphorylation by Hyp-Gly. LY294002 and the mammalian target of rapamycin (mTOR) inhibitor rapamycin repressed the enhancing effects of Hyp-Gly on MyHC and tropomyosin expression. The peptide/histidine transporter 1 (PHT1) was highly expressed in both myoblasts and myotubes, and co-administration of histidine inhibited Hyp-Gly-induced phosphorylation of p70S6K in myoblasts and myotubes. These results indicate that Hyp-Gly can induce myogenic differentiation and myotube hypertrophy and suggest that Hyp-Gly promotes myogenic differentiation by activating the PI3K/Akt/mTOR signaling pathway, perhaps depending on PHT1 for entry into cells.


Assuntos
Diferenciação Celular/efeitos dos fármacos , Colágeno/farmacologia , Dipeptídeos/farmacologia , Fibras Musculares Esqueléticas/patologia , Mioblastos/citologia , Animais , Tamanho Celular/efeitos dos fármacos , Histidina/farmacologia , Hipertrofia , Masculino , Proteínas de Membrana Transportadoras/metabolismo , Camundongos , Fibras Musculares Esqueléticas/efeitos dos fármacos , Fibras Musculares Esqueléticas/metabolismo , Mioblastos/efeitos dos fármacos , Mioblastos/metabolismo , Cadeias Pesadas de Miosina/metabolismo , Fosfatidilinositol 3-Quinases/metabolismo , Fosforilação , Proteínas Proto-Oncogênicas c-akt/metabolismo , Proteínas Quinases S6 Ribossômicas 70-kDa/metabolismo , Transdução de Sinais/efeitos dos fármacos , Serina-Treonina Quinases TOR/metabolismo , Tropomiosina/metabolismo
12.
FEBS Lett ; 590(16): 2690-9, 2016 08.
Artigo em Inglês | MEDLINE | ID: mdl-27323688

RESUMO

Estrogen receptor α (ERα) functions as a transcription factor for genes involved in estrogen-dependent development of breast cancer cells. We demonstrate here that knockdown of hypoxia-inducible factor (HIF)-2α, but not of HIF-1α, increases endogenous ERα protein expression in normoxia and hypoxia. The von Hippel-Lindau protein (pVHL)-dependent degradation of HIF-2α participates in the regulation of ERα expression. Additionally, HIF-2α forms a protein complex with ERα, and amino acids 396-823 of HIF-2α physically interact with the ligand-binding domain of ERα. These results indicate that HIF-2α functions as a negative regulator of ERα expression in breast cancer, especially in normoxia.


Assuntos
Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Neoplasias da Mama/genética , Receptor alfa de Estrogênio/metabolismo , Proteína Supressora de Tumor Von Hippel-Lindau/metabolismo , Aminoácidos/genética , Aminoácidos/metabolismo , Fatores de Transcrição Hélice-Alça-Hélice Básicos/genética , Neoplasias da Mama/patologia , Hipóxia Celular/genética , Receptor alfa de Estrogênio/genética , Feminino , Regulação Neoplásica da Expressão Gênica , Humanos , Ligantes , Células MCF-7 , Complexos Multiproteicos/genética , Complexos Multiproteicos/metabolismo , Proteólise , Proteína Supressora de Tumor Von Hippel-Lindau/genética
13.
J Biol Chem ; 290(43): 26072-87, 2015 Oct 23.
Artigo em Inglês | MEDLINE | ID: mdl-26359500

RESUMO

Alzheimer disease (AD) is a progressive neurodegenerative disorder characterized by loss of neurons and formation of pathological extracellular deposits induced by amyloid-ß peptide (Aß). Numerous studies have established Aß amyloidogenesis as a hallmark of AD pathogenesis, particularly with respect to mitochondrial dysfunction. We have previously shown that glycolytic glyceraldehyde-3-phosphate dehydrogenase (GAPDH) forms amyloid-like aggregates upon exposure to oxidative stress and that these aggregates contribute to neuronal cell death. Here, we report that GAPDH aggregates accelerate Aß amyloidogenesis and subsequent neuronal cell death both in vitro and in vivo. Co-incubation of Aß40 with small amounts of GAPDH aggregates significantly enhanced Aß40 amyloidogenesis, as assessed by in vitro thioflavin-T assays. Similarly, structural analyses using Congo red staining, circular dichroism, and atomic force microscopy revealed that GAPDH aggregates induced Aß40 amyloidogenesis. In PC12 cells, GAPDH aggregates augmented Aß40-induced cell death, concomitant with disruption of mitochondrial membrane potential. Furthermore, mice injected intracerebroventricularly with Aß40 co-incubated with GAPDH aggregates exhibited Aß40-induced pyramidal cell death and gliosis in the hippocampal CA3 region. These observations were accompanied by nuclear translocation of apoptosis-inducing factor and cytosolic release of cytochrome c from mitochondria. Finally, in the 3×Tg-AD mouse model of AD, GAPDH/Aß co-aggregation and mitochondrial dysfunction were consistently detected in an age-dependent manner, and Aß aggregate formation was attenuated by GAPDH siRNA treatment. Thus, this study suggests that GAPDH aggregates accelerate Aß amyloidogenesis, subsequently leading to mitochondrial dysfunction and neuronal cell death in the pathogenesis of AD.


Assuntos
Doença de Alzheimer/metabolismo , Peptídeos beta-Amiloides/metabolismo , Gliceraldeído-3-Fosfato Desidrogenases/metabolismo , Peptídeos beta-Amiloides/biossíntese , Animais , Humanos , Camundongos , Camundongos Transgênicos , Microscopia de Força Atômica , Mitocôndrias/fisiologia , Células PC12 , Ratos
14.
Cell Signal ; 27(10): 1994-2001, 2015 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-26186973

RESUMO

Prostate cancer grows under hypoxic conditions. Hypoxia decreases androgen receptor (AR) protein levels. However, the molecular mechanism remains unclear. Here, we report that p62-mediated autophagy degrades AR protein and suppresses apoptosis in prostate cancer LNCaP cells in hypoxia. In LNCaP cells, hypoxia decreased AR at the protein level, but not at the mRNA level. Hypoxia-induced AR degradation was inhibited not only by knockdown of LC3, a key component of the autophagy machinery, but also by knockdown of p62. Depletion of p62 enhanced hypoxia-induced poly(ADP-ribose) polymerase cleavage and caspase-3 cleavage, markers of apoptosis, whereas simultaneous knockdown of p62 and AR suppressed hypoxia-induced apoptosis. Hypoxia increased the formation of a cytosolic p62-AR complex and enhanced sequestration of AR from the nucleus. Formation of this complex was promoted by the increased phosphorylation of serine 403 in the ubiquitin-associated domain of p62 during hypoxia. An antioxidant and an AMP-activated protein kinase (AMPK) inhibitor reduced hypoxia-induced p62 phosphorylation at serine 403 and suppressed hypoxia-induced complex formation between AR and p62. These results demonstrate that hypoxia enhances the complex formation between p62 and AR by promoting phosphorylation of p62 at serine 403, probably through activating AMPK, and that p62-mediated autophagy degrades AR protein for cell survival in hypoxia.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Apoptose , Receptores Androgênicos/metabolismo , Autofagia , Hipóxia Celular , Linhagem Celular Tumoral , Humanos , Fosforilação , Ligação Proteica , Processamento de Proteína Pós-Traducional , Proteólise , Espécies Reativas de Oxigênio/metabolismo , Proteína Sequestossoma-1
15.
J Endocrinol ; 225(3): 135-45, 2015 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-25901042

RESUMO

17ß-Estradiol (E2) is thought to be responsible for sex-specific differences in skeletal muscle mass. The biological function of E2 is exerted through its binding to estrogen receptor α (ERα). The expression of ubiquitin-specific peptidase 19 (USP19) is upregulated during muscle atrophy and by E2-activated ERα. Here, we investigated the involvement of USP19 in sex difference in muscle mass in young mice. Knockdown of USP19 in hindlimb muscles increased the mass and fiber size in soleus muscle in females but not males. Using Usp19 promoter reporter constructs, a functional half-estrogen response element (hERE) was identified in intron 1 of Usp19. ERα bound to hERE in an E2-dependent manner in C2C12 myoblasts and in soleus muscle in ovariectomized (OVX) female mice. Furthermore, under normal physiological conditions, ERα bound to hERE in soleus muscle only in females. In contrast, administration of E2 resulted in increased Usp19 mRNA expression, decreased muscle mass, and recruitment of ERα to hERE in soleus muscle in males. Knockdown of ERα in hindlimb muscles decreased Usp19 mRNA expression and increased the mass of soleus muscle only in females. Knockdown of USP19 resulted in increased levels of ubiquitin conjugates in soleus muscle in females. OVX increased the levels of ubiquitin conjugates and administration of E2 decreased OVX-induced levels of ubiquitin conjugates. These results demonstrate that in soleus muscle in young female mice under physiological conditions, E2 upregulates USP19 expression through ERα and consequently leads to decreases in ubiquitin conjugates and muscle mass.


Assuntos
Endopeptidases/metabolismo , Indução Enzimática , Estradiol/metabolismo , Receptor alfa de Estrogênio/agonistas , Desenvolvimento Muscular , Músculo Esquelético/crescimento & desenvolvimento , Proteases Específicas de Ubiquitina/metabolismo , Ubiquitinação , Animais , Linhagem Celular , Endopeptidases/genética , Receptor alfa de Estrogênio/antagonistas & inibidores , Receptor alfa de Estrogênio/genética , Receptor alfa de Estrogênio/metabolismo , Feminino , Membro Posterior , Íntrons , Masculino , Camundongos Endogâmicos , Proteínas Musculares/metabolismo , Músculo Esquelético/metabolismo , Ovariectomia , Regiões Promotoras Genéticas , Interferência de RNA , Distribuição Aleatória , Proteínas Recombinantes/química , Proteínas Recombinantes/metabolismo , Elementos de Resposta , Caracteres Sexuais , Proteases Específicas de Ubiquitina/antagonistas & inibidores , Proteases Específicas de Ubiquitina/genética
16.
Biosci Biotechnol Biochem ; 79(9): 1430-7, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25799006

RESUMO

Androgen receptor (AR) signaling is the master regulator of prostate cell growth. Here, to better understand AR signaling, we searched for AR-interacting proteins by yeast two-hybrid screening and identified protein arginine methyltransferase 10 (PRMT10) as one of the interacting proteins. PRMT10 was highly expressed in reproductive tissues, such as prostate. Immunostaining showed that PRMT10 was expressed in the nucleus of both epithelia and stroma of rat prostate. In human prostate cancer LNCaP cells, PRMT10 co-immunoprecipitated with AR in both the presence and absence of dihydrotestosterone (DHT). Knockdown of PRMT10 by siRNA decreased DHT-dependent LNCaP cell growth and induction of prostate-specific antigen, an AR-target gene, without apparent loss of AR. DHT decreased PRMT10 at both the mRNA and protein levels. The decrease in PRMT10 was canceled by knockdown of AR or an AR antagonist. These results indicate that PRMT10 plays an important role in androgen-dependent proliferation of prostate cancer cells.


Assuntos
Proliferação de Células/genética , Neoplasias da Próstata/genética , Proteína-Arginina N-Metiltransferases/biossíntese , Receptores Androgênicos/biossíntese , Animais , Linhagem Celular Tumoral , Regulação Neoplásica da Expressão Gênica , Humanos , Masculino , Próstata/metabolismo , Próstata/patologia , Neoplasias da Próstata/patologia , Proteína-Arginina N-Metiltransferases/genética , RNA Mensageiro/biossíntese , Ratos , Receptores Androgênicos/genética
17.
J Nutr Sci Vitaminol (Tokyo) ; 60(4): 276-82, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25297617

RESUMO

Androgen-dependent prostate cancer inevitably progresses to incurable castration-resistant prostate cancer (CRPC) after androgen deprivation therapy. Because castration-induced hypoxia-inducible factor (HIF)-1α enhances the transcriptional activity of androgen receptor (AR) at low androgen levels mimicking the castration-resistant stage, HIF-1α is expected to be a promising target for suppression of growth of CRPC. We investigated the effect of resveratrol (3,4',5-trihydroxy-trans-stilbene) on the growth of human prostate cancer LNCaP xenografts in castrated male BALB/cSlc-nu/nu mice (5 wk old). The mice were administered a control diet or a resveratrol diet (4 g/kg diet) for 40 d. The resveratrol diet significantly suppressed tumor growth compared to the control diet. In LNCaP xenografts, dietary resveratrol decreased the protein level of HIF-1α, but not the AR coactivator ß-catenin, and reduced the mRNA levels of androgen-responsive genes. In the control group, ß-catenin was predominantly localized in the nucleus with HIF-1α in LNCaP xenografts, whereas dietary resveratrol inhibited the nuclear accumulation of ß-catenin. In hypoxic LNCaP cells at a low androgen level mimicking the castration-resistant stage, hypoxia-induced nuclear accumulation of ß-catenin was inhibited by resveratrol. Furthermore, resveratrol repressed the expression level of HIF-1α even in the presence of a proteasome inhibitor and suppressed hypoxia-enhanced AR transactivation. These results indicate that dietary resveratrol represses nuclear localization of ß-catenin by decreasing the HIF-1α expression, perhaps in a proteasome-independent manner, and inhibits ß-catenin-mediated AR signaling; this contributes to suppression of tumor growth of CRPC.


Assuntos
Antineoplásicos Fitogênicos/uso terapêutico , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Fitoterapia , Extratos Vegetais/uso terapêutico , Neoplasias de Próstata Resistentes à Castração/tratamento farmacológico , Receptores Androgênicos/metabolismo , Estilbenos/uso terapêutico , Animais , Antineoplásicos Fitogênicos/farmacologia , Linhagem Celular Tumoral , Núcleo Celular , Dieta , Humanos , Masculino , Camundongos Endogâmicos BALB C , Camundongos Nus , Extratos Vegetais/farmacologia , Neoplasias de Próstata Resistentes à Castração/genética , Neoplasias de Próstata Resistentes à Castração/metabolismo , RNA Mensageiro/metabolismo , Resveratrol , Transdução de Sinais , Estilbenos/farmacologia , Ativação Transcricional , Vitis/química , beta Catenina/metabolismo
18.
J Nutr Sci Vitaminol (Tokyo) ; 60(4): 291-6, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25297619

RESUMO

S-Equol is enantioselectively produced from the isoflavone daidzein by gut microflora and is absorbed by the body. An increase of pancreatic ß-cell death is directly associated with defects in insulin secretion and an increased risk of type 2 diabetes mellitus. In the present study, we demonstrate that only the S-enantiomer has suppressive effects against alloxan-induced oxidative stress in INS-1 pancreatic ß-cells. S-Equol reduced alloxan-induced cell death in a dose-dependent manner, whereas R-equol had no effects. In contrast, no significant differences were observed between the enantiomers in estrogenic activity. The cytoprotective effects of S-equol were stronger than those of its precursor daidzein and were blocked by the protein synthesis inhibitor cycloheximide. The cytoprotection was diminished when cells were incubated with a protein kinase A (PKA) inhibitor (H89), but not an estrogen receptor inhibitor. S-Equol increased intracellular cAMP levels in an enantioselective manner. S-Equol, but not R-equol, induced phosphorylation of cAMP-response element-binding protein at Ser 133, and induced cAMP-response element-mediated transcription, both of which were diminished in the presence of H89. Taken together, these results show that S-equol enantioselectively increases the survival of INS-1 cells presumably through activating PKA signaling. Thus, S-equol might have applications as an anti-type 2 diabetic agent.


Assuntos
Morte Celular/efeitos dos fármacos , Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , Equol/farmacologia , Células Secretoras de Insulina/efeitos dos fármacos , Isoflavonas/farmacologia , Estresse Oxidativo/efeitos dos fármacos , Extratos Vegetais/farmacologia , Aloxano , Animais , Bactérias/metabolismo , Linhagem Celular , AMP Cíclico/metabolismo , Diabetes Mellitus Tipo 2/metabolismo , Equol/química , Insulina/metabolismo , Isoflavonas/metabolismo , Isomerismo , Fosforilação , Fitoestrógenos/farmacologia , Extratos Vegetais/metabolismo , Ratos , Transdução de Sinais
19.
J Nutr Sci Vitaminol (Tokyo) ; 60(2): 122-8, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24975222

RESUMO

Resveratrol (3,4',5-trihydroxy-trans-stilbene) is known to enhance the cytotoxicity of the anticancer drug doxorubicin. On the other hand, breast cancer MCF-7 cells acquire resistance to doxorubicin under hypoxic conditions. In this study, we investigated the effect of resveratrol on hypoxia-induced resistance to doxorubicin in MCF-7 cells. Resveratrol and its derivative 3,5-dihydroxy-4'-methoxy-trans-stilbene, but not 3,5-dimethoxy-4'-hydroxy-trans-stilbene, cancelled hypoxia-induced resistance to doxorubicin at a concentration of 10 µM. Carbonyl reductase 1 (CBR1) catalyzes the conversion of doxorubicin to its metabolite doxorubicinol, which is much less effective than doxorubicin. Hypoxia increased the expression of CBR1 at both mRNA and protein levels, and knockdown of CBR1 inhibited hypoxia-induced resistance to doxorubicin in MCF-7 cells. Knockdown of hypoxia-inducible factor (HIF)-1α repressed the hypoxia-induced expression of CBR1. Resveratrol repressed the expression of HIF-1α protein, but not HIF-1α mRNA, and decreased hypoxia-activated HIF-1 activity. Resveratrol repressed the hypoxia-induced expression of CBR1 at both mRNA and protein levels. Likewise, 3,5-dihydroxy-4'-methoxy-trans-stilbene decreased the hypoxia-induced expression of CBR1 protein, but not 3,5-dimethoxy-4'-hydroxy-trans-stilbene. Furthermore, resveratrol decreased the expression of HIF-1α protein even in the presence of the proteasome inhibitor MG132 in hypoxia. Theses results indicate that in MCF-7 cells, HIF-1α-increased CBR1 expression plays an important role in hypoxia-induced resistance to doxorubicin and that resveratrol and 3,5-dihydroxy-4'-methoxy-trans-stilbene decrease CBR1 expression by decreasing HIF-1α protein expression, perhaps through a proteasome-independent pathway, and consequently repress hypoxia-induced resistance to doxorubicin.


Assuntos
Oxirredutases do Álcool/metabolismo , Antineoplásicos/uso terapêutico , Neoplasias da Mama/tratamento farmacológico , Doxorrubicina/uso terapêutico , Resistencia a Medicamentos Antineoplásicos/efeitos dos fármacos , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Estilbenos/uso terapêutico , Oxirredutases do Álcool/genética , Antineoplásicos/farmacologia , Neoplasias da Mama/metabolismo , Doxorrubicina/análogos & derivados , Doxorrubicina/farmacologia , Feminino , Humanos , Hipóxia , Subunidade alfa do Fator 1 Induzível por Hipóxia/genética , Leupeptinas/farmacologia , Células MCF-7 , Fitoterapia , Extratos Vegetais/farmacologia , Extratos Vegetais/uso terapêutico , RNA Mensageiro/metabolismo , Resveratrol , Estilbenos/farmacologia
20.
J Nutr Sci Vitaminol (Tokyo) ; 59(4): 358-64, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-24064738

RESUMO

The mechanisms by which resveratrol (3,4',5-trihydroxy-trans-stilbene) elicits diverse health benefits remain unclear because the intracellular target molecules of resveratrol are poorly defined. We screened resveratrol-binding proteins from lysates of MCF-7 breast cancer cells using resveratrol-affinity resin, which was constructed by immobilizing 4'-amino-3,5-dihydroxy-trans-stilbene on activated CH-Sepharose. On SDS-PAGE, two bands were detected as proteins that specifically bound to the resveratrol-affinity resin. One of these, a 30-kDa protein, was identified as human carbonyl reductase 1 (CBR1) by hybrid linear ion trap/time-of-flight mass spectrometry. Similarly, recombinant CBR1 bound to the resveratrol-affinity resin in the absence of resveratrol, but not in the presence of resveratrol. Among its activities, CBR1 catalyzes a NADPH-dependent reduction of the anticancer drug doxorubicin to the cardiotoxin doxorubicinol. The effects of doxorubicin on viability of MCF-7 cells were enhanced by resveratrol, 3,5-dihydroxy-4'-methoxy-trans-stilbene, 3,4'-dihydroxy-5-methoxy-trans-stilbene, and 4'-amino-3,5-dihydroxy-trans-stilbene at concentrations of 1 and 10 µM. Resveratrol and these derivatives inhibited CBR1 activities to a similar degree at concentrations of 100 and 200 µM. However, 3,5-dimethoxy-4'-hydroxy-trans-stilbene and m-hydroquinone had no influence on doxorubicin cytotoxicity or CBR1 activity. Resveratrol inhibited CBR1 activity through an apparent mix of competitive (Ki=55.8 µM) and noncompetitive (αKi=164 µM; α=2.98) inhibition kinetics. These results indicate that (i) resveratrol enhances the cytotoxic effects of doxorubicin on MCF-7 cells; (ii) the moiety that contains the 3,5-dihydroxyl groups of resveratrol, but not the m-hydroquinone structure alone, is required to bind CBR1; and (iii) resveratrol acts as a mixed-type inhibitor of CBR1 activity on doxorubicin.


Assuntos
Oxirredutases do Álcool/antagonistas & inibidores , Antibióticos Antineoplásicos/metabolismo , Antineoplásicos Fitogênicos/farmacologia , Neoplasias da Mama/metabolismo , Doxorrubicina/metabolismo , Extratos Vegetais/farmacologia , Estilbenos/farmacologia , Antibióticos Antineoplásicos/uso terapêutico , Antineoplásicos Fitogênicos/química , Antineoplásicos Fitogênicos/uso terapêutico , Ligação Competitiva , Neoplasias da Mama/tratamento farmacológico , Cromatografia de Afinidade/métodos , Doxorrubicina/análogos & derivados , Doxorrubicina/uso terapêutico , Feminino , Humanos , Hidroquinonas/farmacologia , Células MCF-7 , NADP/metabolismo , Fitoterapia , Extratos Vegetais/química , Extratos Vegetais/uso terapêutico , Ligação Proteica/efeitos dos fármacos , Resveratrol , Estilbenos/química , Estilbenos/uso terapêutico
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA