Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Cancer Med ; 13(16): e70124, 2024 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-39158384

RESUMO

BACKGROUND: Despite advances in the treatment of cancer, pancreatic ductal adenocarcinoma (PDAC) remains highly lethal due to the lack of effective therapies. Our previous study showed that Luteolin (Lut), a flavonoid, suppressed pancreatocarcinogenesis and reduced the expression of dihydropyrimidine dehydrogenase (DPYD), an enzyme that degrades pyrimidines such as 5-fluorouracil (5-FU), in PDACs. In this study, we investigated the role of DPYD and evaluated the therapeutic potential of combining 5-FU with Lut in PDACs. METHODS AND RESULTS: PDAC cells overexpressing DPYD showed increased proliferation, and invasiveness, adding to the resistance to 5-FU. The xenograft tumors of DPYD-overexpressing PDAC cells also exhibit enhanced growth and invasion compared to the control xenograft tumors. RNA-seq analysis of the DPYD-overexpressing PDAC xenograft tumors revealed an upregulation of genes associated with metallopeptidase activity-MMP9 and MEP1A. Furthermore, the overexpression of MEP1A in PDAC was associated with invasion. Next, we investigated the combined effects of Lut, a DPYD suppressor, and 5-FU on DPYD-overexpressing xenograft tumors and PDAC of Pdx1-Cre; LSL-KrasG12D/+; Trp53flox/flox(KPPC) mice. Neither single administration of 5-FU nor Lut showed significant inhibitory effects; however, the combined administration of 5-FU and Lut exhibited a significant tumor-suppressive effect in both the xenograft tumors and KPPC models. CONCLUSION: We have elucidated that DPYD expression contributes to proliferation, invasiveness, and 5-FU resistance, in PDACs. The combination therapy of Lut and 5-FU holds the potential for enhanced efficacy against PDACs.


Assuntos
Carcinoma Ductal Pancreático , Proliferação de Células , Di-Hidrouracila Desidrogenase (NADP) , Fluoruracila , Luteolina , Neoplasias Pancreáticas , Ensaios Antitumorais Modelo de Xenoenxerto , Fluoruracila/farmacologia , Fluoruracila/uso terapêutico , Animais , Humanos , Di-Hidrouracila Desidrogenase (NADP)/genética , Di-Hidrouracila Desidrogenase (NADP)/metabolismo , Neoplasias Pancreáticas/tratamento farmacológico , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/patologia , Neoplasias Pancreáticas/metabolismo , Camundongos , Carcinoma Ductal Pancreático/tratamento farmacológico , Carcinoma Ductal Pancreático/patologia , Carcinoma Ductal Pancreático/genética , Carcinoma Ductal Pancreático/metabolismo , Luteolina/farmacologia , Luteolina/uso terapêutico , Proliferação de Células/efeitos dos fármacos , Linhagem Celular Tumoral , Resistencia a Medicamentos Antineoplásicos , Protocolos de Quimioterapia Combinada Antineoplásica/farmacologia , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Camundongos Nus , Invasividade Neoplásica
2.
Food Funct ; 15(16): 8562-8571, 2024 Aug 12.
Artigo em Inglês | MEDLINE | ID: mdl-39072556

RESUMO

Nonalcoholic fatty liver disease (NAFLD) is fatty liver mainly related to metabolic syndrome. NAFLD with inflammation and hepatocellular damage is defined as nonalcoholic steatohepatitis (NASH), which can progress to cirrhosis and hepatocellular carcinoma. We have previously reported that a hexane insoluble fraction from an anthocyanin-rich purple rice ethanolic extract (PRE-HIF) can suppress prostate carcinogenesis. However, the extract's effect on NASH has not yet been established. In the present study, we investigated the chemopreventive effect of a PRE-HIF on NASH and liver fibrosis using a connexin 32 (Cx32) dominant negative transgenic (Cx32ΔTg) rat NASH model. Seven-week-old male Cx32ΔTg rats were fed a control diet, a high-fat diet (HFD), or an HFD with 1% PRE-HIF and intraperitoneal administration of dimethylnitrosamine for 17 weeks. Histological findings of NASH such as fat deposition, lobular inflammation, hepatocyte ballooning injury, and bridging fibrosis were observed in the HFD group but not in the control group, and all histological parameters were significantly improved by PRE-HIF treatment. Corresponding to the histological changes, increased expression of inflammatory cytokine mRNAs (TNF-α, IL-6, IL-18, IFN-γ, IL-1ß, TGF-ß1, TIMP1, TIMP2, COL1A1), along with and activation of nuclear factor-κB (NF-κB) and c-Jun N-terminal kinase (JNK) signaling were observed in the HFD group, which was significantly decreased by PRE-HIF. The number and area of hepatic precancerous glutathione S-transferase placental form-positive foci tended to be decreased by PRE-HIF. These results indicate that intake of purple rice as a dietary supplement may reduce steatohepatitis, liver injury, and fibrosis in NASH by inactivation of NF-κB or JNK.


Assuntos
Cirrose Hepática , NF-kappa B , Hepatopatia Gordurosa não Alcoólica , Oryza , Extratos Vegetais , Animais , Masculino , NF-kappa B/metabolismo , NF-kappa B/genética , Extratos Vegetais/farmacologia , Ratos , Hepatopatia Gordurosa não Alcoólica/tratamento farmacológico , Hepatopatia Gordurosa não Alcoólica/metabolismo , Cirrose Hepática/tratamento farmacológico , Cirrose Hepática/metabolismo , Oryza/química , Humanos , Fígado/efeitos dos fármacos , Fígado/metabolismo , Fígado/patologia , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Dieta Hiperlipídica/efeitos adversos , Modelos Animais de Doenças , Transdução de Sinais/efeitos dos fármacos , Citocinas/metabolismo , Citocinas/genética
3.
Virchows Arch ; 479(2): 277-284, 2021 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-33638656

RESUMO

Colorectal cancer (CRC) is one of the most common gastrointestinal cancers worldwide with high morbidity and mortality rates. The discovery of small molecule anticancer reagents has significantly affected cancer therapy. However, the anticancer effects of these therapies are not sufficient to completely cure CRC. PDZ-binding kinase (PBK) was initially identified as a mitotic kinase for mitogen-activated protein kinase and is involved in cytokinesis and spermatogenesis. Aberrant expression of PBK has been reported to be closely associated with malignant phenotypes of many cancers and/or patient survival. However, the expression of PBK and its association to patient survival in CRC have not been fully elucidated. In the present study, 269 primary CRCs were evaluated immunohistochemically for PBK expression to assess its ability as a prognostic factor. CRC tumor cells variably expressed PBK (range, 0-100%; median, 32%) in the nucleus and cytoplasm. Univariate analyses identified a significant inverse correlation between PBK expression and pT stage (P<0.0001). Furthermore, patients carrying CRC with higher PBK expression showed significantly favorable survival (P=0.0094). Multivariate Cox proportional hazards regression analysis revealed high PBK expression (HR, 0.52; P=0.015) as one of the potential favorable factors for CRC patients. PBK expression showed significant correlation to Ki-67 labeling indices (ρ=0.488, P<0.0001). In vitro, the PBK inhibitor OTS514 suppressed cellular proliferation of CRC cells with PBK expression through downregulation of P-ERK and induction of apoptosis. These results suggest that PBK-targeting therapeutics may be useful for the treatment of PBK-expressing CRC patients.


Assuntos
Biomarcadores Tumorais/metabolismo , Neoplasias Colorretais/enzimologia , Quinases de Proteína Quinase Ativadas por Mitógeno/metabolismo , Idoso , Idoso de 80 Anos ou mais , Antineoplásicos/farmacologia , Apoptose/efeitos dos fármacos , Biomarcadores Tumorais/antagonistas & inibidores , Células CACO-2 , Proliferação de Células/efeitos dos fármacos , Neoplasias Colorretais/mortalidade , Neoplasias Colorretais/patologia , Neoplasias Colorretais/terapia , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Feminino , Células HCT116 , Humanos , Imuno-Histoquímica , Masculino , Pessoa de Meia-Idade , Quinases de Proteína Quinase Ativadas por Mitógeno/antagonistas & inibidores , Estadiamento de Neoplasias , Fosforilação , Inibidores de Proteínas Quinases/farmacologia , Quinolonas/farmacologia , Medição de Risco , Fatores de Risco , Tiofenos/farmacologia , Fatores de Tempo , Resultado do Tratamento
4.
Nutrients ; 14(1)2021 Dec 23.
Artigo em Inglês | MEDLINE | ID: mdl-35010924

RESUMO

Non-alcoholic steatohepatitis (NASH) can cause liver cirrhosis and hepatocellular carcinoma (HCC), with cases increasing worldwide. To reduce the incidence of liver cirrhosis and HCC, NASH is targeted for the development of treatments, along with viral hepatitis and alcoholic hepatitis. Lactoferrin (LF) has antioxidant, anti-cancer, and anti-inflammatory activities. However, whether LF affects NASH and fibrosis remains unelucidated. We aimed to clarify the chemopreventive effect of LF on NASH progression. We used a NASH model with metabolic syndrome established using connexin 32 (Cx32) dominant negative transgenic (Cx32ΔTg) rats. Cx32ΔTg rats (7 weeks old) were fed a high-fat diet and intraperitoneally injected with dimethylnitrosamine (DMN). Rats were divided into three groups for LF treatment at 0, 100, or 500 mg/kg/day for 17 weeks. Lactoferrin significantly protected steatosis and lobular inflammation in Cx32ΔTg rat livers and attenuated bridging fibrosis or liver cirrhosis induced by DMN. By quantitative RT-PCR, LF significantly down-regulated inflammatory (Tnf-α, Il-6, Il-18, and Il-1ß) and fibrosis-related (Tgf-ß1, Timp2, and Col1a1) cytokine mRNAs. Phosphorylated nuclear factor (NF)-κB protein decreased in response to LF, while phosphorylated JNK protein was unaffected. These results indicate that LF might act as a chemopreventive agent to prevent hepatic injury, inflammation, and fibrosis in NASH via NF-κB inactivation.


Assuntos
Lactoferrina/farmacologia , Cirrose Hepática/prevenção & controle , NF-kappa B/metabolismo , Hepatopatia Gordurosa não Alcoólica/tratamento farmacológico , Animais , Anticarcinógenos/farmacologia , Carcinogênese/efeitos dos fármacos , Carcinoma Hepatocelular/tratamento farmacológico , Conexinas/metabolismo , Citocinas/metabolismo , Dimetilnitrosamina/efeitos adversos , Fibrose/prevenção & controle , Lactoferrina/administração & dosagem , Fígado/lesões , Neoplasias Hepáticas/tratamento farmacológico , Masculino , Hepatopatia Gordurosa não Alcoólica/metabolismo , Ratos , Fator de Crescimento Transformador beta1/metabolismo , Fator de Necrose Tumoral alfa/metabolismo , Proteína beta-1 de Junções Comunicantes
5.
Arch Toxicol ; 94(12): 4085-4097, 2020 12.
Artigo em Inglês | MEDLINE | ID: mdl-32833043

RESUMO

Non-alcoholic steatohepatitis (NASH) is a recognized risk factor for liver fibrosis and malignancies, and is associated with features of metabolic syndrome, such as obesity and insulin resistance (IR). We previously demonstrated that the disturbance of connexin 32 (Cx32), a gap junctional protein of hepatocytes, exacerbated NASH in Cx32 dominant-negative transgenic (Cx32ΔTg) rats fed methionine choline-deficient diet (MCDD). MCDD is well-established means of inducing NASH in rodents; however, the Cx32ΔTg-MCDD NASH model does not reproduce obesity and IR. In this study, we aimed to establish an improved NASH model. Eight-week-old male Cx32ΔTg and wild-type (Wt) rats received a high-fat diet (HFD) with dimethylnitrosamine (DMN) for 12 weeks. The HFD with DMN led to gains in body, liver, and visceral fat weights in both genotypes. IR was significantly greater in Cx32ΔTg than in Wt rats. Elevation of serum hepatic enzymes (AST, ALT), inflammatory cytokine expressions (Tnfα, Il-6, Tgf-ß1, Il-1ß, Timp2, and Col1a1), steatohepatitis, and fibrosis were significantly greater in Cx32ΔTg as compared with Wt rats. Regarding carcinogenesis, the number and area of glutathione S-transferase placental form (GST-P)-positive preneoplastic hepatic foci were significantly increased in Cx32ΔTg versus Wt rats. Moreover, activation of NF-κB and JNK contributed to the progression of NASH in Cx32ΔTg rats. These results suggest that Cx32 dysfunction promoted the progression of NASH, metabolic syndrome, and carcinogenesis. Therefore, the novel Cx32ΔTg-HFD-DMN NASH model may be a rapid and useful tool for evaluating the progression of NASH.


Assuntos
Transformação Celular Neoplásica/metabolismo , Conexinas/metabolismo , Cirrose Hepática Experimental/etiologia , Neoplasias Hepáticas Experimentais/etiologia , Fígado/metabolismo , Hepatopatia Gordurosa não Alcoólica/etiologia , Animais , Transformação Celular Neoplásica/genética , Transformação Celular Neoplásica/patologia , Conexinas/genética , Citocinas/metabolismo , Dieta Hiperlipídica , Dimetilnitrosamina , Progressão da Doença , Mediadores da Inflamação/metabolismo , Resistência à Insulina , Proteínas Quinases JNK Ativadas por Mitógeno/metabolismo , Fígado/patologia , Cirrose Hepática Experimental/genética , Cirrose Hepática Experimental/metabolismo , Cirrose Hepática Experimental/patologia , Neoplasias Hepáticas Experimentais/genética , Neoplasias Hepáticas Experimentais/metabolismo , Neoplasias Hepáticas Experimentais/patologia , Masculino , NF-kappa B/metabolismo , Hepatopatia Gordurosa não Alcoólica/genética , Hepatopatia Gordurosa não Alcoólica/metabolismo , Hepatopatia Gordurosa não Alcoólica/patologia , Ratos Transgênicos , Transdução de Sinais , Proteína beta-1 de Junções Comunicantes
6.
Cancer Sci ; 111(4): 1165-1179, 2020 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-31994822

RESUMO

Luteolin is a natural flavonoid with strong anti-oxidative properties that is reported to have an anti-cancer effect in several malignancies other than bladder cancer. In this study, we describe the effect of luteolin on a human bladder cancer cell line, T24, in the context of the regulation of p21, thioredoxin-1 (TRX1) and the mechanistic target of rapamycin (mTOR) pathway. Luteolin inhibited cell survival and induced G2/M cell-cycle arrest, p21 upregulation and downregulation of phospho(p)-S6, which is downstream of mTOR signaling. Luteolin also upregulated TRX1 and reduced intracellular reactive oxygen species production. In a subcutaneous xenograft mouse model using the rat bladder cancer cell line, BC31, tumor volumes were significantly decreased in mice orally administered luteolin compared to control. Immunohistochemical analysis revealed that increased p21 and decreased p-S6 expression were induced in the luteolin treatment group. Moreover, in another in vivo N-butyl-N-(4-hydroxybutyl) nitrosamine (BBN)-induced rat bladder cancer model, the oral administration of luteolin led to a trend of decreased bladder tumor dimension and significantly decreased the Ki67-labeling index and p-S6 expression. Furthermore, the major findings on the metabolism of luteolin suggest that both plasma and urine luteolin-3'-O-glucuronide concentrations are strongly associated with the inhibition of cell proliferation and mTOR signaling. Moreover, a significant decrease in the squamous differentiation of bladder cancer is attributed to plasma luteolin-3'-glucuronide concentration. In conclusion, luteolin, and in particular its metabolized product, may represent another natural product-derived therapeutic agent that acts against bladder cancer by upregulating p21 and inhibiting mTOR signaling.


Assuntos
Luteolina/farmacologia , Serina-Treonina Quinases TOR/genética , Neoplasias da Bexiga Urinária/tratamento farmacológico , Proteínas rho de Ligação ao GTP/genética , Animais , Apoptose/efeitos dos fármacos , Butilidroxibutilnitrosamina/toxicidade , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Modelos Animais de Doenças , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Humanos , Antígeno Ki-67/genética , Luteolina/metabolismo , Masculino , Camundongos , Proteínas Proto-Oncogênicas c-akt/genética , Ratos , Transdução de Sinais/efeitos dos fármacos , Tiorredoxinas/genética , Neoplasias da Bexiga Urinária/induzido quimicamente , Neoplasias da Bexiga Urinária/genética , Neoplasias da Bexiga Urinária/patologia , Ensaios Antitumorais Modelo de Xenoenxerto
7.
Carcinogenesis ; 41(8): 1145-1157, 2020 08 12.
Artigo em Inglês | MEDLINE | ID: mdl-31805186

RESUMO

A need exists for seeking effective treatments for castration-resistant prostate cancer (CRPC) in response to its emergence following androgen deprivation therapy as a major clinical problem. In the present study, we investigated the chemopreventive and chemotherapeutic potential of luteolin, a flavonoid with antioxidative properties, on prostate cancer, including CRPC. Luteolin inhibited the progression of rat prostate carcinogenesis by induction of apoptosis in a transgenic rat for adenocarcinoma of prostate (TRAP) model. Luteolin decreased cell proliferation in a dose-dependent manner and induced apoptosis with the activation of caspases 3 and 7 in both rat (PCai1, established from a TRAP prostate tumor) and human (22Rv1) CRPC cells. Dietary luteolin also suppressed tumor growth via an increase in apoptosis and inhibition of angiogenesis in PCai1 and 22Rv1 xenografts implanted in castrated nude mice. We also focused on androgen receptor splice variant 7 (AR-V7), which contributes to cell proliferation and therapeutic resistance in CRPC. Luteolin dramatically suppressed AR-V7 protein expression in 22Rv1 cells in vitro and ex vivo. Microarray analysis identified MiR-8080, which contains a possible target sequence for AR-V7 3'-UTR, as a gene upregulated by luteolin. MiR-8080 transfection decreased the AR-V7 expression level and the induction of apoptosis in 22Rv1 cells. Furthermore, miR-8080 knockdown canceled luteolin decreasing AR-V7 and the cell growth of 22Rv1. MiR-8080 induced by luteolin intake enhanced the therapeutic effect of enzalutamide on 22Rv1 xenografts under castration conditions. These results indicate luteolin inhibits CRPC by AR-V7 suppression through miR-8080, highlighting luteolin and miR-8080 as promising therapeutic agents for this disease.


Assuntos
Antagonistas de Receptores de Andrógenos/farmacologia , Antioxidantes/farmacologia , Luteolina/farmacologia , MicroRNAs/metabolismo , Neoplasias de Próstata Resistentes à Castração/tratamento farmacológico , Receptores Androgênicos/metabolismo , Antagonistas de Receptores de Andrógenos/uso terapêutico , Animais , Antioxidantes/uso terapêutico , Apoptose/efeitos dos fármacos , Carcinogênese/efeitos dos fármacos , Caspase 3/metabolismo , Caspase 7/metabolismo , Linhagem Celular Tumoral , Quimioprevenção , Humanos , Luteolina/uso terapêutico , Masculino , Camundongos , Camundongos Nus , MicroRNAs/genética , Neovascularização Patológica/tratamento farmacológico , Neoplasias de Próstata Resistentes à Castração/prevenção & controle , Isoformas de Proteínas/antagonistas & inibidores , Ratos , Ratos Sprague-Dawley , Ratos Transgênicos , Receptores Androgênicos/genética , Ensaios Antitumorais Modelo de Xenoenxerto
8.
J Clin Med ; 8(4)2019 Mar 31.
Artigo em Inglês | MEDLINE | ID: mdl-30935141

RESUMO

Recent studies have shown that epigenetic alterations lead to oncogenic activation, thus indicating that these are therapeutic targets. Herein, we analyzed the efficacy and therapeutic potential of our developed histone lysine demethylase 1 (LSD1) inhibitor, NCL1, in castration-resistant prostate cancer (CRPC). The CRPC cell lines 22Rv1, PC3, and PCai1CS were treated with NCL1, and LSD1 expression and cell viability were assessed. The epigenetic effects and mechanisms of NCL1 were also evaluated. CRPC cells showed strong LSD1 expression, and cell viability was decreased by NCL1 in a dose-dependent manner. Chromatin immunoprecipitation analysis indicated that NCL1 induced histone H3 lysine 9 dimethylation accumulation at promoters of P21. As shown by Western blot and flow cytometry analyses, NCL1 also dose-dependently induced caspase-dependent apoptosis. The stimulation of autophagy was observed in NCL1-treated 22Rv1 cells by transmission electron microscopy and LysoTracker analysis. Furthermore, WST-8 assay revealed that the anti-tumor effect of NCL1 was reinforced when autophagy was inhibited by chloroquine in 22Rv1 cells. Combination index analysis revealed that a concurrent use of these drugs had a synergistic effect. In ex vivo analysis, castrated nude mice were injected subcutaneously with PCai1 cells and intraperitoneally with NCL1. Tumor volume was found to be reduced with no adverse effects in NCL1-treated mice compared with controls. Finally, immunohistochemical analysis using consecutive human specimens in pre- and post-androgen deprivation therapy demonstrated that LSD1 expression levels in CRPC, including neuroendocrine differentiation cases, were very high, and identical to levels observed in previously examined prostate biopsy specimens. NCL1 effectively suppressed prostate cancer growth in vitro and ex vivo without adverse events via the regulation of apoptosis and autophagy, suggesting that NCL1 is a potential therapeutic agent for CRPC.

9.
Toxicology ; 398-399: 31-40, 2018 04 01.
Artigo em Inglês | MEDLINE | ID: mdl-29501575

RESUMO

Tobacco smoking is a major risk factor for human cancers including urinary bladder carcinoma. Cigarette smoke inhalation in mice and orally administered nicotine in rats and mice increased urothelial cell proliferation. Nicotine, a major component of smoke, induced cell proliferation in multiple cell types in vitro. In the present study, the enhancing effects of nicotine on F344 rat bladder carcinogenesis induced by N-butyl-N-(4-hydroxybutyl)nitrosamine (BBN) were examined. Nicotine administered in drinking water for 32 weeks following 4 weeks of BBN treatment significantly increased the incidence and number of urothelial carcinomas dose-dependently. Ki67 and pSTAT3 labeling indices and expression of nicotinic acetylcholine receptor alpha 7 (nAChRα7) in non-tumor bladder urothelial lesions were significantly increased by nicotine, but the TUNEL assay for apoptosis showed no increase. In a 4 week study, inhibitors of nicotinic acetylcholine receptor decreased nicotine-induced urothelial simple hyperplasia and Ki67 labeling index in the bladder and kidney pelvis at a single cytotoxic dose of nicotine (40 ppm). Urothelial cytotoxicity with regenerative proliferation was observed by light and scanning electron microscopy. In vitro, nicotine was not cytotoxic to rat or human immortalized urothelial cells (do not express nicotine receptors) below millimolar concentrations, nor in human RT4, T24 or UMUC3 urothelial carcinoma cells (express nicotine receptors). However, nicotine slightly, but statistically significantly, increased cell proliferation at micromolar concentrations in human urothelial carcinoma cells. These data suggest that nicotine enhances urinary bladder carcinogenesis by inducing cytotoxicity with regenerative proliferation. The possible role of direct mitogenesis, involving nAChR and STAT3 signaling and of nicotine receptors requires further investigation at non-cytotoxic doses of nicotine.


Assuntos
Nicotina/toxicidade , Neoplasias da Bexiga Urinária/induzido quimicamente , Bexiga Urinária/efeitos dos fármacos , Administração Oral , Animais , Peso Corporal/efeitos dos fármacos , Carcinogênese/induzido quimicamente , Linhagem Celular , Proliferação de Células/efeitos dos fármacos , Humanos , Masculino , Nicotina/administração & dosagem , Distribuição Aleatória , Ratos , Ratos Endogâmicos F344 , Bexiga Urinária/patologia , Neoplasias da Bexiga Urinária/patologia
10.
Int J Mol Sci ; 17(12)2016 Dec 10.
Artigo em Inglês | MEDLINE | ID: mdl-27973395

RESUMO

Pioglitazone (PGZ), a peroxisome proliferator-activated receptor γ agonist, which is known as a type 2 diabetes drug, inhibits cell proliferation in various cancer cell lines, including prostate carcinomas. This study focused on the effect of PGZ on prostate carcinogenesis using a transgenic rat for an adenocarcinoma of prostate (TRAP) model. Adenocarcinoma lesions as a percentage of overall lesions in the ventral prostate were significantly reduced by PGZ treatment in a dose-dependent manner. The number of adenocarcinomas per given area in the ventral prostate was also significantly reduced by PGZ treatment. The Ki67 labeling index in the ventral prostate was also significantly reduced by PGZ. Decreased cyclin D1 expression in addition to the inactivation of both p38 mitogen-activated protein kinase (MAPK) and nuclear factor (NF)κB were detected in PGZ-treated TRAP rat groups. In LNCaP, a human androgen-dependent prostate cancer cell line, PGZ also inhibited cyclin D1 expression and the activation of both p38 MAPK and NFκB. The suppression of cultured cell growth was mainly regulated by the NFκB pathway as detected using specific inhibitors in both LNCaP and PC3, a human androgen-independent prostate cancer cell line. These data suggest that PGZ possesses a chemopreventive potential for prostate cancer.


Assuntos
Carcinogênese/patologia , PPAR gama/agonistas , Neoplasias da Próstata/patologia , Tiazolidinedionas/farmacologia , Adenocarcinoma/tratamento farmacológico , Adenocarcinoma/patologia , Animais , Peso Corporal/efeitos dos fármacos , Carcinogênese/efeitos dos fármacos , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Progressão da Doença , Humanos , Masculino , NF-kappa B/metabolismo , Tamanho do Órgão/efeitos dos fármacos , PPAR gama/metabolismo , Pioglitazona , Neoplasias da Próstata/tratamento farmacológico , Ratos Sprague-Dawley , Ratos Transgênicos , Transdução de Sinais/efeitos dos fármacos , Tiazolidinedionas/uso terapêutico , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
11.
Nutrients ; 8(7)2016 Jul 11.
Artigo em Inglês | MEDLINE | ID: mdl-27409632

RESUMO

Fermented brown rice and rice bran with Aspergillus oryzae (FBRA) is considered to have the potential to prevent chemically-induced carcinogenesis in multiple organs of rodents. In the present study, we evaluated the possible chemopreventive effects of FBRA against prostate tumorigenesis. Six-week-old male rats of the transgenic rat for adenocarcinoma of prostate (TRAP) strain were fed diets containing 5% or 10% FBRA for 15 weeks. Animals were sacrificed at 21 weeks of age, and the ventral and lateral prostate were removed for histopathological evaluation and immunoblot analyses. FBRA decreased the incidence of adenocarcinoma in the lateral prostate and suppressed the progression of prostate carcinogenesis. Treatment with FBRA induced apoptosis and inhibited cell proliferation in histologically high-grade prostatic intraepithelial neoplasias. Phospho-AMP-activated kinase α (Thr172) was up-regulated in the prostate of rats fed the diet supplemented with FBRA. These results indicate that FBRA controls tumor growth by activating pathways responsive to energy deprivation and suggest that FBRA has translational potential for the prevention of human prostate cancer.


Assuntos
Adenocarcinoma/prevenção & controle , Ração Animal , Aspergillus oryzae/fisiologia , Fibras na Dieta/administração & dosagem , Fermentação , Oryza/microbiologia , Neoplasia Prostática Intraepitelial/prevenção & controle , Neoplasias da Próstata/prevenção & controle , Proteínas Quinases Ativadas por AMP/metabolismo , Adenocarcinoma/enzimologia , Adenocarcinoma/genética , Adenocarcinoma/patologia , Animais , Antígenos Transformantes de Poliomavirus/genética , Apoptose , Proliferação de Células , Modelos Animais de Doenças , Metabolismo Energético , Ativação Enzimática , Heterozigoto , Masculino , Fosforilação , Próstata/metabolismo , Próstata/patologia , Neoplasia Prostática Intraepitelial/enzimologia , Neoplasia Prostática Intraepitelial/genética , Neoplasia Prostática Intraepitelial/patologia , Neoplasias da Próstata/enzimologia , Neoplasias da Próstata/genética , Neoplasias da Próstata/patologia , Ratos Sprague-Dawley , Ratos Transgênicos , Transdução de Sinais , Fatores de Tempo
12.
Brain Res ; 1615: 1-11, 2015 Jul 30.
Artigo em Inglês | MEDLINE | ID: mdl-25881893

RESUMO

Oxidative stress induced by the treatment with 100 µM hydrogen peroxide (H2O2) for 10 min enhances release of cytosolic proteins along with fibroblast growth factor 1 (FGF-1) from rat astrocytes without inducing apoptosis. FGF-1 promotes the generation of apolipoprotein E-containing high-density lipoprotein-like particles (apoE/HDL) in astrocytes, which contributes to cholesterol homeostasis in the brain. In this work, we studied various effects of oxidative stress on rat astrocyte׳s membrane to understand the mechanism underlying release of cytosolic proteins and FGF-1. The oxidative stress using 100 µM H2O2 enhanced lipid release from rat astrocytes in addition to suppression of lipid synthesis. The lipid synthesis, however, was hardly suppressed by H2O2 in the cell lines such as bovine endothelial cells and HepG2 cells from which the release of cytosolic proteins is not increased by H2O2 unlike rat astrocytes. The treatment of rat astrocytes with H2O2 changed the distribution of lipids and proteins in the caveolin-1-rich domain of membrane to the non-raft domain, which was canceled by the pretreatment of cells with low-density lipoproteins (LDL). These findings suggest that oxidative stress induced by H2O2 changes lipid level of the plasma membrane to make the membrane structure fragile in rat astrocytes. The direct treatment with H2O2 of membrane fraction prepared from rat astrocytes did not enhance lipid release from the membrane. The lipid release, however, was enhanced from the isolated membrane fraction, after the cells were treated with H2O2 and incubated in H2O2-free DPBS. Hydrogen peroxide enhanced phosphorylation of protein kinases such as Akt, MEK, and ERK in intact astrocytes without injury and stress. A MEK inhibitor, U0126, suppressed not only the H2O2-induced ERK phosphorylation but also cytosolic protein release from rat astrocytes. These findings suggest that the H2O2-induced release of cytosolic proteins depends on imbalance of lipid level in the membrane through suppressing lipid synthesis and increasing lipid release induced by the intracellular biochemical reaction such as signal transduction generated in intact rat astrocytes treated with H2O2.


Assuntos
Astrócitos/metabolismo , Lipídeos de Membrana/metabolismo , Estresse Oxidativo , Animais , Fenômenos Bioquímicos , Células Cultivadas , Células Hep G2 , Humanos , Peróxido de Hidrogênio , Lipídeos de Membrana/biossíntese , Ratos , Ratos Wistar , Transdução de Sinais
13.
Brain Res ; 1522: 12-21, 2013 Jul 19.
Artigo em Inglês | MEDLINE | ID: mdl-23727614

RESUMO

We previously observed that the production and release of fibroblast growth factor (FGF-1) are increased in rat astrocytes during in vitro long-term culture, that FGF-1 enhances the generation of apoE-containing high density lipoproteins (apoE/HDL), and that the wound healing of brain cryoinjury delays in apoE-deficient mouse. The detail mechanism underlying these phenomena remains unknown. In this study, we examined effects of oxidative stress on release of FGF-1 from cultured rat astrocytes. The treatment of rat astrocytes with 100µM hydrogen peroxide (H2O2) for 10min enhanced FGF-1 release without inducing apoptosis. The conditioned medium prepared from the cells cultured in a fresh medium after the treatment with H2O2 had the FGF-1-like activities, which enhanced cholesterol synthesis, signalings to phosphorylate Akt and ERK, and apoE secretion. The oxidative stress induced by H2O2 enhanced the release of cytosolic proteins such as HSP70 and HSP90 in addition to FGF-1. Antioxidants such as ascorbic acid and ebselen suppressed the release of cytosolic proteins induced by H2O2 treatment. The addition of lipoproteins such as low density lipoproteins (LDL), furthermore, canceled H2O2-induced release of FGF-1 and cytosolic proteins. Proteolysis of cytosolic proteins in the H2O2-treated rat astrocytes was enhanced in the presence of exogenous trypsin, which was attenuated by the pretreatment with LDL, suggesting that H2O2 increases the permeability of the membrane of cells, which was prevented by the addition of lipoproteins. These findings suggest that oxidative stress is one of the candidates which triggers FGF-1 release from astrocytes in the brain, and that the lipid homeostasis in the cell membrane may regulate H2O2-induced release of FGF-1.


Assuntos
Transplante de Medula Óssea/métodos , Isquemia Encefálica/patologia , Região CA1 Hipocampal/patologia , Leucócitos Mononucleares/transplante , Degeneração Neural/patologia , Animais , Masculino
14.
Biochim Biophys Acta ; 1811(12): 1062-9, 2011 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-21907307

RESUMO

In the previous paper, we reported that apolipoprotein (apo) A-I enhances generation of HDL-like lipoproteins in rat astrocytes to be accompanied with both increase in tyrosine phosphorylation of phospholipase Cγ (PL-Cγ) and PL-Cγ translocation to cytosolic lipid-protein particles (CLPP) fraction. In this paper, we studied the interaction between apoA-I and ATP-binding cassette transporter A1 (ABCA1) to relate with PL-Cγ function for generation of HDL-like lipoproteins in the apoA-I-stimulated astrocytes. ABCA1 co-migrated with exogenous apoA-I with apparent molecular weight over 260kDa on SDS-PAGE when rat astrocytes were treated with apoA-I and then with a cross-linker, BS3. The solubilized ABCA1 of rat astrocytes was associated with the apoA-I-immobilized Affi-Gel 15. An LXR agonist, To901317, increased the cellular level of ABCA1, association of apoA-I with ABCA1 and apoA-I-mediated lipid release in rat astrocytoma GA-1/Mock cells where ABCA1 expression at baseline is very low. PL-Cγ was co-isolated by apoA-I-immobilized Affi-Gel 15 and co-immunoprecipitated by anti-ABCA1 antibody along with ABCA1 from the solubilized membrane fraction of rat astrocytes. The SiRNA of ABCA1 suppressed not only the PL-Cγ binding to ABCA1 but also the tyrosine phosphorylation of PL-Cγ. A PL-C inhibitor, U73122, prevented generation of apoA-I-mediated HDL-like lipoproteins in rat astrocytes. To901317 increased the association of PL-Cγ with ABCA1 in GA-1/Mock cells dependently on the increase of cellular level of ABCA1 without changing that of PL-Cγ. These findings suggest that the exogenous apoA-I augments the interaction between PL-Cγ and ABCA1 to stimulate tyrosine phosphorylation and activation of PL-Cγ for generation of HDL-like lipoproteins in astrocytes.


Assuntos
Transportadores de Cassetes de Ligação de ATP/metabolismo , Astrócitos/metabolismo , Lipoproteínas HDL/biossíntese , Fosfolipase C gama/metabolismo , Transdução de Sinais/fisiologia , Transportador 1 de Cassete de Ligação de ATP , Transportadores de Cassetes de Ligação de ATP/antagonistas & inibidores , Transportadores de Cassetes de Ligação de ATP/genética , Animais , Apolipoproteína A-I/metabolismo , Apolipoproteína A-I/farmacologia , Astrócitos/citologia , Astrócitos/efeitos dos fármacos , Linhagem Celular , Reagentes de Ligações Cruzadas/química , Citosol/metabolismo , Eletroforese em Gel de Poliacrilamida , Estrenos/farmacologia , Feto , Inativação Gênica , Proteínas Imobilizadas/química , Proteínas Imobilizadas/metabolismo , Imunoprecipitação , Camundongos , Inibidores de Fosfodiesterase/farmacologia , Fosfolipase C gama/antagonistas & inibidores , Fosforilação , Transporte Proteico/efeitos dos fármacos , Pirrolidinonas/farmacologia , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/metabolismo , Ratos
15.
J Biochem ; 146(6): 881-6, 2009 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-19713443

RESUMO

Fibroblast growth factor-1 (FGF-1) is released from astrocytes in stress and stimulates MEK/ERK and PI3K/Akt pathways in autocrine fashion to increase synthesis of cholesterol and 25-OH-cholesterol, and to induce transport and secretion of apoE, respectively. FGF-1-induced phosphorylation of Src, and phosphorylation of MEK, ERK and Ark was inhibited by Src inhibitors in rat astrocytes. Src inhibitors also suppressed FGF-1-induced increase of biosynthesis and release of cholesterol and increase of apolipoprotein E (apoE) secretion. The results were reproduced in rat astrocytoma cells transfected by rat apoE and in 3T3-L1 cells. Down-regulation of Src expression reduced FGF-1-induced phosphorylation of the signalling protein and subsequent reactions. Increase by FGF-1 of messages of apoE and HMG-CoA reductase was not influenced by Src inhibitors or by its down-regulation. We conclude that FGF-1 activates Src for activation of MEK/ERK and PI3K/Akt pathways, while Src may not be involved in enhancement of transcription of the cholesterol-related genes.


Assuntos
Apolipoproteínas E/metabolismo , Astrócitos/metabolismo , Fator 1 de Crescimento de Fibroblastos/metabolismo , Lipoproteínas HDL/metabolismo , Quinases da Família src/metabolismo , Animais , Encéfalo/citologia , Células Cultivadas , Embrião de Mamíferos , Feminino , Modelos Biológicos , RNA Interferente Pequeno , Ratos , Ratos Wistar , Transdução de Sinais
16.
J Neurochem ; 97(4): 1034-43, 2006 May.
Artigo em Inglês | MEDLINE | ID: mdl-16606364

RESUMO

Apolipoprotein (apo) A-I induces rapid translocation of protein kinase Calpha and phospholipase Cgamma, and slow translocation of caveolin-1 and newly synthesized cholesterol to the cytosolic lipid-protein particle (CLPP) fraction in rat astrocytes. In order to understand the function of CLPP, we investigated the interaction with cytoskeletons of CLPP-related proteins such as caveolin-1 and protein kinase Calpha and of CLPP-related lipids in rat astrocytes. Under the conditions that microtubules were depolymerized, association of cytosolic caveolin-1 with protein kinase Calpha and alpha-tubulin was enhanced when the cells were treated with apoA-I for 5 min. This association was suppressed by a scaffolding domain-peptide of caveolin-1. Association with the microtubule-like filaments of cytosolic lipids, caveolin-1 and protein kinase Calpha was also increased by the apoA-I treatment and inhibited by the scaffolding domain peptide. Paclitaxel (taxol), a compound to stabilize microtubules, suppressed the apoA-I-mediated intracellular translocation and release from the cells of the de novo synthesized cholesterol and phospholipid. The findings suggested that the association of CLPP with microtubules is mediated by a scaffolding domain of caveolin-1, induced by apoA-I and involved in regulation of intracellular cholesterol trafficking for assembly of cellular lipids to apoA-I-high-density lipoprotein (HDL).


Assuntos
Apolipoproteína A-I/metabolismo , Astrócitos/metabolismo , Caveolina 1/metabolismo , Colesterol/metabolismo , Citosol/metabolismo , Microtúbulos/metabolismo , Animais , Apolipoproteína A-I/farmacologia , Astrócitos/efeitos dos fármacos , Astrócitos/ultraestrutura , Cavéolas/efeitos dos fármacos , Cavéolas/metabolismo , Cavéolas/ultraestrutura , Caveolina 1/química , Células Cultivadas , Citoesqueleto/efeitos dos fármacos , Citoesqueleto/metabolismo , Citoesqueleto/ultraestrutura , Citosol/efeitos dos fármacos , Citosol/ultraestrutura , Lipoproteínas HDL/metabolismo , Microscopia Eletrônica de Transmissão , Microtúbulos/efeitos dos fármacos , Microtúbulos/ultraestrutura , Paclitaxel/farmacologia , Peptídeos/metabolismo , Peptídeos/farmacologia , Proteína Quinase C-alfa/metabolismo , Estrutura Terciária de Proteína/fisiologia , Transporte Proteico/efeitos dos fármacos , Transporte Proteico/fisiologia , Ratos , Ratos Wistar , Frações Subcelulares/metabolismo , Tubulina (Proteína)/metabolismo , Regulação para Cima/efeitos dos fármacos , Regulação para Cima/fisiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA