Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
J Immunol ; 211(1): 34-42, 2023 07 01.
Artigo em Inglês | MEDLINE | ID: mdl-37212607

RESUMO

We recently discovered a (to our knowledge) new neuroimmune interaction named the gateway reflex, in which the activation of specific neural circuits establishes immune cell gateways at specific vessel sites in organs, leading to the development of tissue-specific autoimmune diseases, including a multiple sclerosis (MS) mouse model, experimental autoimmune encephalomyelitis (EAE). We have reported that peripheral-derived myeloid cells, which are CD11b+MHC class II+ and accumulate in the fifth lumbar (L5) cord during the onset of a transfer model of EAE (tEAE), play a role in the pain-mediated relapse via the pain-gateway reflex. In this study, we investigated how these cells survive during the remission phase to cause the relapse. We show that peripheral-derived myeloid cells accumulated in the L5 cord after tEAE induction and survive more than other immune cells. These myeloid cells, which highly expressed GM-CSFRα with common ß chain molecules, grew in number and expressed more Bcl-xL after GM-CSF treatment but decreased in number by blockade of the GM-CSF pathway, which suppressed pain-mediated relapse of neuroinflammation. Therefore, GM-CSF is a survival factor for these cells. Moreover, these cells were colocalized with blood endothelial cells (BECs) around the L5 cord, and BECs expressed a high level of GM-CSF. Thus, GM-CSF from BECs may have an important role in the pain-mediated tEAE relapse caused by peripheral-derived myeloid cells in the CNS. Finally, we found that blockade of the GM-CSF pathway after pain induction suppressed EAE development. Therefore, GM-CSF suppression is a possible therapeutic approach in inflammatory CNS diseases with relapse, such as MS.


Assuntos
Encefalomielite Autoimune Experimental , Esclerose Múltipla , Camundongos , Animais , Fator Estimulador de Colônias de Granulócitos e Macrófagos/metabolismo , Doenças Neuroinflamatórias , Células Endoteliais/metabolismo , Sistema Nervoso Central , Dor/metabolismo , Células Mieloides , Recidiva
2.
J Vis Exp ; (172)2021 06 14.
Artigo em Inglês | MEDLINE | ID: mdl-34180908

RESUMO

Patient-derived tumor organoids (PDOs) are expected to be a preclinical cancer model with better reproducibility of disease than traditional cell culture models. PDOs have been successfully generated from a variety of human tumors to recapitulate the architecture and function of tumor tissue accurately and efficiently. However, PDOs are unsuitable for an in vitro high-throughput assay system (HTS) or cell analysis using 96-well or 384-well plates when evaluating anticancer drugs because they are heterogeneous in size and form large clusters in culture. These cultures and assays use extracellular matrices, such as Matrigel, to create tumor tissue scaffolds. Therefore, PDOs have a low throughput and high cost, and it has been difficult to develop a suitable assay system. To address this issue, a simpler and more accurate HTS was established using PDOs to evaluate the potency of anticancer drugs and immunotherapy. An in vitro HTS was created that uses PDOs established from solid tumors cultured in 384-well plates. An HTS was also developed for assessment of antibody-dependent cellular cytotoxicity activity to represent the immune response using PDOs cultured in 96-well plates.


Assuntos
Antineoplásicos , Neoplasias , Humanos , Neoplasias/tratamento farmacológico , Organoides , Reprodutibilidade dos Testes
3.
Oncol Lett ; 21(5): 406, 2021 May.
Artigo em Inglês | MEDLINE | ID: mdl-33841567

RESUMO

An in vitro assay system using patient-derived tumor models represents a promising preclinical cancer model that replicates the disease better than traditional cell culture models. Patient-derived tumor organoid (PDO) and patient-derived tumor xenograft (PDX) models have been previously established from different types of human tumors to recapitulate accurately and efficiently their tissue architecture and function. However, these models have low throughput and are challenging to construct. Thus, the present study aimed to establish a simple in vitro high-throughput assay system using PDO and PDX models. Furthermore, the current study aimed to evaluate different classes of anticancer drugs, including chemotherapeutic, molecular targeted and antibody drugs, using PDO and PDX models. First, an in vitro high-throughput assay system was constructed using PDO and PDX established from solid and hematopoietic tumors cultured in 384-well plates to evaluate anticancer agents. In addition, an in vitro evaluation system of the immune response was developed using PDO and PDX. Novel cancer immunotherapeutic agents with marked efficacy have been used against various types of tumor. Thus, there is an urgent need for in vitro functional potency assays that can simulate the complex interaction of immune cells with tumor cells and can rapidly test the efficacy of different immunotherapies or antibody drugs. An evaluation system for the antibody-dependent cellular cytotoxic activity of anti-epidermal growth factor receptor antibody and the cytotoxic activity of activated lymphocytes, such as cytotoxic T lymphocytes and natural killer cells, was constructed. Moreover, immune response assay systems with bispecific T-cell engagers were developed using effector cells. The present results demonstrated that in vitro assay systems using PDO and PDX may be suitable for evaluating anticancer agents and immunotherapy potency with high reproducibility and simplicity.

4.
Cells ; 8(5)2019 05 20.
Artigo em Inglês | MEDLINE | ID: mdl-31137590

RESUMO

Patient-derived tumor organoids (PDOs) represent a promising preclinical cancer model that better replicates disease, compared with traditional cell culture models. We have established PDOs from various human tumors to accurately and efficiently recapitulate the tissue architecture and function. Molecular targeted therapies with remarkable efficacy are currently in use against various tumors. Thus, there is a need for in vitro functional-potency assays that can be used to test the efficacy of molecular targeted drugs and model complex interactions between immune cells and tumor cells to evaluate the potential for cancer immunotherapy. This study represents an in vitro evaluation of different classes of molecular targeted drugs, including small-molecule inhibitors, monoclonal antibodies, and an antibody-drug conjugate, using lung PDOs. We evaluated epidermal growth factor receptor and human epidermal growth factor receptor 2 (HER2) inhibitors using a suitable high-throughput assay system. Next, the antibody-dependent cellular cytotoxicity (ADCC) activity of an anti-HER2 monoclonal antibody was evaluated to visualize the interactions of immune cells with PDOs during ADCC responses. Moreover, an evaluation system was developed for the immune checkpoint inhibitors, nivolumab and pembrolizumab, using PDOs. Our results demonstrate that the in vitro assay systems using PDOs were suitable for evaluating molecular targeted drugs under conditions that better reflect pathological conditions.


Assuntos
Anticorpos Monoclonais Humanizados/uso terapêutico , Antineoplásicos/uso terapêutico , Carcinoma Adenoescamoso/tratamento farmacológico , Carcinoma de Células Escamosas/tratamento farmacológico , Avaliação de Medicamentos/métodos , Neoplasias Pulmonares/tratamento farmacológico , Terapia de Alvo Molecular , Organoides/efeitos dos fármacos , Anticorpos Monoclonais Humanizados/farmacologia , Antineoplásicos/farmacologia , Biópsia , Carcinoma Adenoescamoso/patologia , Carcinoma Adenoescamoso/cirurgia , Carcinoma de Células Escamosas/patologia , Carcinoma de Células Escamosas/cirurgia , Sobrevivência Celular/efeitos dos fármacos , Células Cultivadas , Receptores ErbB/antagonistas & inibidores , Humanos , L-Lactato Desidrogenase/análise , Neoplasias Pulmonares/patologia , Neoplasias Pulmonares/cirurgia , Receptor ErbB-2/antagonistas & inibidores
5.
Oncol Rep ; 40(2): 635-646, 2018 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-29917168

RESUMO

Patient-derived tumor xenograft models represent a promising preclinical cancer model that better replicates disease, compared with traditional cell culture; however, their use is low-throughput and costly. To overcome this limitation, patient-derived tumor organoids (PDOs) were established from human lung, ovarian and uterine tumor tissues, among others, to accurately and efficiently recapitulate the tissue architecture and function. PDOs were able to be cultured for >6 months, and formed cell clusters with similar morphologies to their source tumors. Comparative histological and comprehensive gene expression analyses proved that the characteristics of PDOs were similar to those of their source tumors, even following long-term expansion in culture. At present, 53 PDOs have been established by the Fukushima Translational Research Project, and were designated as Fukushima PDOs (F­PDOs). In addition, the in vivo tumorigenesis of certain F­PDOs was confirmed using a xenograft model. The present study represents a detailed analysis of three F­PDOs (termed REME9, 11 and 16) established from endometrial cancer tissues. These were used for cell growth inhibition experiments using anticancer agents. A suitable high-throughput assay system, with 96- or 384­well plates, was designed for each F­PDO, and the efficacy of the anticancer agents was subsequently evaluated. REME9 and 11 exhibited distinct responses and increased resistance to the drugs, as compared with conventional cancer cell lines (AN3 CA and RL95-2). REME9 and 11, which were established from tumors that originated in patients who did not respond to paclitaxel and carboplatin (the standard chemotherapy for endometrial cancer), exhibited high resistance (half-maximal inhibitory concentration >10 µM) to the two agents. Therefore, assay systems using F­PDOs may be utilized to evaluate anticancer agents using conditions that better reflect clinical conditions, compared with conventional methods using cancer cell lines, and to discover markers that identify the pharmacological effects of anticancer agents.


Assuntos
Antineoplásicos/farmacologia , Neoplasias do Endométrio/tratamento farmacológico , Organoides/efeitos dos fármacos , Animais , Carboplatina/farmacologia , Carcinogênese/efeitos dos fármacos , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Resistencia a Medicamentos Antineoplásicos/efeitos dos fármacos , Ensaios de Seleção de Medicamentos Antitumorais/métodos , Feminino , Expressão Gênica/efeitos dos fármacos , Humanos , Masculino , Camundongos , Paclitaxel/farmacologia , Ensaios Antitumorais Modelo de Xenoenxerto
6.
Oncol Rep ; 37(1): 66-76, 2017 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-27840973

RESUMO

Epidermal growth factor receptor (EGFR) overexpression and EGFR-mediated signaling pathway dysregulation have been observed in tumors from patients with various cancers, especially non-small cell lung cancer. Thus, several anti-EGFR drugs have been developed for cancer therapy. For patients with known EGFR activating mutations (EGFR exon 19 in-frame deletions and exon 21 L858R substitution), treatment with an EGFR tyrosine kinase inhibitor (EGFR TKI; gefitinib, erlotinib or afatinib) represents standard first-line therapy. However, the clinical efficacy of these TKIs is ultimately limited by the development of acquired drug resistance such as by mutation of the gatekeeper T790 residue (T790M). To overcome this acquired drug resistance and develop novel anti-EGFR drugs, a cell-based assay system for EGFR TKI resistance mutant-selective inhibitors is required. We constructed a novel cell-based assay for the evaluation of EGFR TKI efficacy against EGFR mutation. To this end, we established non-tumorigenic immortalized breast epithelial cells that proliferate dependent on EGF (MCF 10A cells), which stably overexpress mutant EGFR. We found that the cells expressing EGFR containing the T790M mutation showed higher resistance against gefitinib, erlotinib and afatinib compared with cells expressing wild-type EGFR. In contrast, L858R mutant-expressing cells exhibited higher TKI sensitivity. The effect of T790M-selective inhibitors (osimertinib and rociletinib) on T790M mutant-expressing cells was significantly higher than gefitinib and erlotinib. Finally, when compared with commercially available isogenic MCF 10A cell lines carrying introduced mutations in EGFR, our EGFR mutant-overexpressing cells exhibited obviously higher responsiveness to EGFR TKIs depending on the underlying mutations because of the higher levels of EGFR phosphorylation in the EGFR mutant-overexpressing cells than in the isogenic cell lines. In conclusion, we successfully developed a novel cell-based assay for evaluating the efficacy of anti-EGFR drugs against EGFR mutation.


Assuntos
Antineoplásicos/isolamento & purificação , Avaliação Pré-Clínica de Medicamentos/métodos , Receptores ErbB/antagonistas & inibidores , Receptores ErbB/genética , Inibidores de Proteínas Quinases/isolamento & purificação , Afatinib , Antineoplásicos/uso terapêutico , Técnicas de Cultura de Células , Células Cultivadas , Resistencia a Medicamentos Antineoplásicos/efeitos dos fármacos , Cloridrato de Erlotinib/farmacologia , Gefitinibe , Humanos , Mutação , Inibidores de Proteínas Quinases/uso terapêutico , Quinazolinas/farmacologia , Transfecção
7.
J Clin Biochem Nutr ; 51(3): 163-9, 2012 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-23170042

RESUMO

High-fat diet is one of the causes of nonalcoholic fatty liver disease. We have previously demonstrated that high-fat diet induces upregulation of adipose differentiation-related protein mRNA expression accompanied by lipid droplet formation in mouse liver. Vanin-1 is a ubiquitous epithelial ectoenzyme that has pantetheinase activity and produces cysteamine, a potent endogenous antioxidant. In the present study, we analyzed the expression of hepatic vanin-1 mRNA following the administration of a high-fat diet in mice as well as free fatty acids in hepatocyte cultures and speculated its possible mechanism. Vanin-1 mRNA levels in the livers of mice were upregulated within a day of the high-fat diet, even before the expression of adipose differentiation-related protein mRNA and lipid accumulation. An in vitro analysis using HuH-7 cells revealed a significant upregulation of vanin-1 mRNA by as low as 0.01 mM oleic acid; however, lipid accumulation in hepatocytes was not affected at this concentration. Furthermore, vanin-1 mRNA was differentially upregulated by various free fatty acids irrespective of the grade of lipid accumulation. These findings indicate that the upregulation of vanin-1 precedes lipid accumulation and is differentially mediated by various types of free fatty acids in the model, presenting vanin-1 as a novel player in the pathogenesis of nonalcoholic fatty liver disease.

8.
Biochem Biophys Res Commun ; 382(2): 348-52, 2009 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-19281795

RESUMO

Lipin-1 is a multifunctional metabolic regulator, involving in triacylglycerol and bioactive glycerolipids synthesis as an enzyme, transcriptional regulation as a coactivator, and adipogenesis. In obesity, adipose lipin-1 expression is decreased. Although lipin-1 is implicated in the pathogenesis of obesity, the mechanism is still not clear. Since TNF-alpha is deeply involved in the pathogenesis of obesity, insulin resistance, and diabetes, here we investigated the role of TNF-alpha on lipin-1 expression in adipocytes. Quantitative PCR studies showed that TNF-alpha suppressed both lipin-1A and -1B isoform expression in time- and dose-dependent manners in mature 3T3-L1 adpocytes. A Jak2 inhibitor, AG490, reversed the suppressive effect of TNF-alpha on both lipin-1A and -1B. In contrast, NF-kappaB, MAPKs, ceramide, and beta-catenin pathway tested were not involved in the mechanism. These results suggest that TNF-alpha could be involved in obesity-induced lipin-1 suppression in adipocytes and Jak2 may play an important role in the mechanism.


Assuntos
Janus Quinase 2/antagonistas & inibidores , Proteínas Nucleares/biossíntese , Obesidade/metabolismo , Fator de Necrose Tumoral alfa/fisiologia , Células 3T3-L1 , Animais , Camundongos , Fosfatidato Fosfatase , Inibidores de Proteínas Quinases/farmacologia , RNA Mensageiro/biossíntese , Fator de Necrose Tumoral alfa/antagonistas & inibidores , Fator de Necrose Tumoral alfa/farmacologia , Tirfostinas/farmacologia
9.
Biochem Biophys Res Commun ; 332(1): 89-94, 2005 Jun 24.
Artigo em Inglês | MEDLINE | ID: mdl-15896303

RESUMO

In the present study, we examined a role of mitogen-activated protein kinases (MAPKs), extracellular signal-related kinase (ERK), c-Jun N-terminal protein kinase, and p38 MAPK in troglitazone-induced inhibition of cell growth in human pancreatic cancer cells. Among the three kinases, troglitazone specifically inhibited the phosphorylation of ERK1/2 in a dose- and time-dependent manner. Troglitazone also down-regulated the protein expression of mitogen-activated protein kinase kinase (MEK)1/2, an upstream molecule that regulates ERK phosphorylation. Treatment of human pancreatic cancer cells with specific MEK inhibitor, PD98059 or U0126, inhibited ERK1/2 phosphorylation and cell growth. These results suggest for the first time that the inhibition of the MEK1/2-ERK1/2 signaling pathway may be implicated in the growth inhibitory effect by troglitazone in human pancreatic cancer cells.


Assuntos
Cromanos/administração & dosagem , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Quinases de Proteína Quinase Ativadas por Mitógeno/metabolismo , Neoplasias Pancreáticas/enzimologia , Neoplasias Pancreáticas/patologia , Transdução de Sinais/efeitos dos fármacos , Tiazolidinedionas/administração & dosagem , Antineoplásicos/administração & dosagem , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Relação Dose-Resposta a Droga , Humanos , Troglitazona
10.
J Gastroenterol ; 39(5): 461-8, 2004.
Artigo em Inglês | MEDLINE | ID: mdl-15175945

RESUMO

BACKGROUND: We have recently demonstrated that peroxisome proliferator activated receptor (PPAR) gamma activation by its selective ligand, troglitazone, potently inhibited cell proliferation in human pancreatic cancer cells. The present study was performed to clarify the role of PPARgamma in cell invasion/motility in human pancreatic cancer cells. METHODS: Cell invasive activity was assessed by an in vitro invasion assay, using a Transwell chamber, and by a wound-healing assay, in the human pancreatic cancer cell lines, PK-1 and PK-9. Cell morphology and actin structure were evaluated by phase-contrast and fluorescence microscopy. RESULTS: PPARgamma activation by troglitazone inhibited cell invasion and cell migration in PK-1 and PK-9 cells. We also examined the effect of troglitazone on cell morphology and actin structure because of its effect on cell motility. The size of PK-1 and PK-9 cells that had been incubated with troglitazone became smaller, and the in shape changed from flat to spindle, followed by round. The troglitazone-induced cell rounding was reversible by replacement with troglitazone-free medium. Rhodamine-phalloidin staining revealed a decreased number of actin filaments in PK-1 cells treated with troglitazone. In cells treated with mycalolide B, an actin depolymerizing agent, troglitazone failed to induce cell rounding. CONCLUSIONS: These results suggest that PPARgamma activation by troglitazone inhibited cell motility and changed cell morphology through modulating actin organization.


Assuntos
Antineoplásicos/farmacologia , Inibição de Migração Celular , Movimento Celular/efeitos dos fármacos , Cromanos/farmacologia , Neoplasias Pancreáticas/patologia , Receptores Citoplasmáticos e Nucleares/fisiologia , Tiazolidinedionas/farmacologia , Fatores de Transcrição/fisiologia , Actinas/metabolismo , Meios de Cultivo Condicionados , Citoesqueleto/efeitos dos fármacos , Humanos , Imuno-Histoquímica , Ligantes , Invasividade Neoplásica , Troglitazona , Células Tumorais Cultivadas
11.
Diabetes ; 53(7): 1671-9, 2004 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-15220189

RESUMO

Resistin was originally reported as an adipose tissue-specific hormone that provided a link between obesity and diabetes. Resistin protein level was elevated in obese mice and decreased by insulin-sensitizing thiazolidinediones. Immunoneutralization of resistin improved insulin sensitivity in diet-induced obese mice, while the administration of exogenous resistin induced insulin resistance. More recently, we have shown that ablation of the resistin gene in mice decreased fasting glucose through impairment of gluconeogenesis, while resistin treatment in these knockout mice increased hepatic glucose production. However, the link between resistin and glucose homeostasis has been questioned by studies demonstrating reduced, rather than increased, resistin mRNA expression in obese and diabetic mice. To better understand the regulation of resistin, we developed a sensitive and specific RIA resistin that could accurately measure serum resistin levels in several mouse models. We show that while resistin mRNA is indeed suppressed in obese mice, the circulating resistin level is significantly elevated and positively correlated with insulin, glucose, and lipids. Both resistin mRNA expression and protein levels in Lep(ob/ob) mice are suppressed by leptin treatment in parallel with reductions in glucose and insulin. In wild-type mice, serum resistin increases after nocturnal feeding, concordant with rising levels of insulin. Resistin mRNA and protein levels decline in parallel with glucose and insulin during fasting and are restored after refeeding. We performed clamp studies to determine whether resistin is causally related to insulin and glucose. Adipose resistin expression and serum resistin increased in response to hyperinsulinemia and further in response to hyperglycemia. Taken together, these findings suggest that the nutritional regulation of resistin and changes in resistin gene expression and circulating levels in obesity are mediated, at least in part, through insulin and glucose.


Assuntos
Diabetes Mellitus/metabolismo , Jejum/metabolismo , Hormônios Ectópicos/sangue , Obesidade/metabolismo , Tecido Adiposo/metabolismo , Animais , Glicemia/análise , Diabetes Mellitus/sangue , Diabetes Mellitus/etiologia , Dieta , Jejum/sangue , Feminino , Hormônios Ectópicos/genética , Hormônios Ectópicos/metabolismo , Hiperglicemia/metabolismo , Hiperinsulinismo/metabolismo , Insulina/sangue , Leptina/genética , Leptina/farmacologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Obesos , Obesidade/sangue , Obesidade/complicações , Obesidade/genética , RNA Mensageiro/sangue , RNA Mensageiro/metabolismo , Radioimunoensaio , Resistina
12.
Int J Cancer ; 108(1): 41-6, 2004 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-14618613

RESUMO

In our study, we examined whether human hepatocellular carcinoma (HCC) expresses peroxisome proliferator-activated receptor gamma (PPARgamma) and the effects of PPAR gamma activation by its selective ligands on cell growth and cell invasion in HCC cells. RT-PCR and Western blot analysis revealed that HCC-derived cell lines, HepG2 and HLF, express PPARgamma mRNA and protein. Luciferase assay in HLF cells showed that troglitazone, a selective ligand for PPAR gamma, transactivated the transcription of a peroxisome proliferator response element-driven promoter in a dose-dependent manner, suggesting that the expressed PPARgamma functions as a transcriptional factor. Not only troglitazone but pioglitazone dose-dependently inhibited cell growth in HepG2 and HLF cells. Invasion assay using a transwell chamber demonstrated that troglitazone also inhibited cell invasion in HCC cells. To examine the mechanism of the troglitazone-induced growth inhibition, we determined p27(Kip1), a cyclin dependent kinase inhibitor, expression by Western blot analysis in troglitazone-treated HLF cells. Troglitazone increased p27(Kip1) in time- and dose-dependent manners, suggesting that p27(Kip1) may be involved in the growth inhibition by troglitazone in HLF cells. To further examine the mechanism of the troglitazone-induced p27(Kip1) protein accumulation, 2 major systems for regulation of p27(Kip1) protein, proteasome activity and Skp2, an F-box protein that targets p27(Kip1) for degradation, were evaluated. Troglitazone potently inhibited proteasome activity and decreased Skp2 protein levels. All these results suggest that human HCC cells express functional PPAR gamma and PPARgamma activation resulted in growth inhibition. The growth inhibition was mediated by p27(Kip1) accumulation, which is induced by both inhibition of ubiquitylation of p27(Kip1) and reduction of degradation activity of p27Kip1 by proteasome.


Assuntos
Carcinoma Hepatocelular/patologia , Proteínas de Ciclo Celular/metabolismo , Cromanos/farmacologia , Cisteína Endopeptidases/metabolismo , Neoplasias Hepáticas/patologia , Complexos Multienzimáticos/metabolismo , Proteínas Quinases Associadas a Fase S/metabolismo , Tiazolidinedionas/farmacologia , Proteínas Supressoras de Tumor/metabolismo , Adenosina Trifosfatases/metabolismo , Carcinoma Hepatocelular/metabolismo , Divisão Celular , Inibidor de Quinase Dependente de Ciclina p27 , Relação Dose-Resposta a Droga , Humanos , Neoplasias Hepáticas/metabolismo , Complexo de Endopeptidases do Proteassoma , Receptores Citoplasmáticos e Nucleares/metabolismo , Fatores de Transcrição/metabolismo , Troglitazona , Células Tumorais Cultivadas
13.
Cancer Sci ; 94(4): 338-43, 2003 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-12824901

RESUMO

We have recently demonstrated that the PPAR gamma ligand troglitazone induced cell growth arrest and evoked apoptosis in a gastric cancer cell line, MKN-45. Since in general, p53 plays an important role in the induction of apoptosis and growth inhibition, we tried to clarify whether or not p53 mediates troglitazone-induced apoptosis and growth arrest in gastric cancer cells. Troglitazone increased the number of apoptoic cells in MKN-28, MKN-45 and MKN-74, but not in KATO-III cells. The troglitazone-induced apoptotic change was significantly reduced by coincubation with bisphenol A digycidyl ether (BADGE), a synthetic PPAR gamma antagonist, in MKN-74 cells, suggesting that PPAR gamma mediates the apoptotic effect of troglitazone. Since KATO-III lacks the p53 gene, we speculated that p53 might be implicated in the PPAR gamma ligand-induced apoptosis. Western blot analysis revealed that p53 expression was increased by troglitazone in a time-dependent manner in MKN-74 cells, further suggesting that p53 may mediate the apoptotic process induced by troglitazone. We next established a dominant-negative p53 mutant by stable transfection of p53 mutant into MKN-74 cells. In the dominant-negative p53 mutant cells, troglitazone failed to induce apoptosis, strongly supporting the hypothesis that p53 indeed mediates the process of the troglitazone-induced apoptosis. In the dominant-negative p53 mutant cells, troglitazone significantly induced cell growth arrest and increased expression of p27(Kip1) protein, which is thought to be the key molecule to evoke growth arrest, suggesting that p53 is not involved in the growth inhibition by troglitazone. All these results suggest that p53 mediates the PPAR gamma ligand-induced apoptosis, but not the cell growth inhibition.


Assuntos
Antineoplásicos/farmacologia , Apoptose/efeitos dos fármacos , Cromanos/farmacologia , Receptores Citoplasmáticos e Nucleares/metabolismo , Neoplasias Gástricas/patologia , Tiazolidinedionas/farmacologia , Fatores de Transcrição/metabolismo , Proteína Supressora de Tumor p53/metabolismo , Proteínas de Ciclo Celular/metabolismo , Divisão Celular/efeitos dos fármacos , Inibidor de Quinase Dependente de Ciclina p27 , Proteínas de Ligação a DNA/metabolismo , Genes Dominantes , Humanos , Ligantes , Neoplasias Gástricas/genética , Neoplasias Gástricas/metabolismo , Troglitazona , Células Tumorais Cultivadas/metabolismo , Células Tumorais Cultivadas/patologia , Proteína Supressora de Tumor p53/genética , Proteínas Supressoras de Tumor/metabolismo
14.
Jpn J Cancer Res ; 93(7): 774-82, 2002 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-12149143

RESUMO

We examined in the present study whether human gastric cancer cells express peroxisome proliferator-activated receptor gamma (PPARgamma), the effect of PPARgamma activation by troglitazone, a selective ligand, on cellular growth, and the mechanism of the growth arrest by troglitazone in gastric cancer cells. RT-PCR, northern blot and western blot analysis demonstrated that all four tested human gastric cancer cell lines, MKN-28, MKN-45, MKN-74 and KATO-III, expressed PPARgamma mRNA and protein. WST-1 assay and flow cytometric analysis revealed that troglitazone inhibited the growth and induced G1 arrest in all four gastric cancer cell lines. To examine the role of p27(Kip1), a cyclin-dependent kinase inhibitor, in the G1 arrest by troglitazone, we determined p27(Kip1) protein expression by western blot analysis in gastric cancer cells that had been treated with troglitazone. Troglitazone increased p27(Kip1) in all four gastric cancer cell lines. Since it has been reported that the ubiquitin-proteasome system plays a vital role in the degradation of p27(Kip1) protein, we evaluated the hypothesis that inhibition of proteasome mediates the troglitazone-induced p27(Kip1) accumulation. Lactacystin, a proteasome inhibitor, inhibited cell growth and increased p27(Kip1) expression in MKN-74 cells. It was further demonstrated that troglitazone inhibited proteasome activity in a dose-dependent manner in MKN-74 cells. All these results suggest that troglitazone inhibited proteasome activity, followed by induction of p27(Kip1), which arrests cells at the G1 phase of the cell cycle in gastric cancer cells. The troglitazone-mediated inhibition of the proteasome suggests a novel mechanism for the anti-proliferative effect of this agent in cancer cells.


Assuntos
Proteínas de Ciclo Celular/metabolismo , Cromanos/farmacologia , Fase G1 , Complexos Multienzimáticos/antagonistas & inibidores , Neoplasias Gástricas/metabolismo , Tiazóis/farmacologia , Tiazolidinedionas , Proteínas Supressoras de Tumor/metabolismo , Antineoplásicos/farmacologia , Northern Blotting , Western Blotting , Ciclo Celular , Divisão Celular , Inibidor de Quinase Dependente de Ciclina p27 , Cisteína Endopeptidases , Inibidores Enzimáticos/farmacologia , Citometria de Fluxo , Humanos , Ligantes , Complexo de Endopeptidases do Proteassoma , Proteínas Proto-Oncogênicas p21(ras)/metabolismo , RNA/metabolismo , RNA Mensageiro/metabolismo , Receptores Citoplasmáticos e Nucleares/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Fatores de Tempo , Fatores de Transcrição/metabolismo , Troglitazona , Células Tumorais Cultivadas
15.
Diabetes ; 51(7): 2099-104, 2002 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-12086938

RESUMO

The rise in obesity and its complications has generated enormous interest in the regulation of feeding and body weight. We show that a spermine metabolite of cholesterol (MSI-1436) decreases body weight, specifically fat, by suppressing feeding and preventing the reduction in energy expenditure, hormonal changes, and patterns of neuropeptide expression normally associated with weight loss. MSI-1436 enters the brain after peripheral injection and is more potent when injected into the cerebral ventricle (intracerebroventricular [ICV]). Systemic or ICV MSI-1436 administration induced similar patterns of Fos immunoreactivity in the brain, especially the paraventricular hypothalamic nucleus (PVN). This brain region integrates neural signals from hypothalamic and brain stem nuclei and regulates feeding behavior, autonomic function, and neuroendocrine function. Microinjection of MSI-1436 into the PVN potently suppressed feeding and reduced body weight for several days. Unlike caloric restriction, MSI-1436 decreased mRNA levels of agouti-related peptide and neuropeptide Y in the hypothalamus. These findings indicate that MSI-1436 acts in the brain to regulate food intake and energy expenditure, likely through suppression of orexigenic hypothalamic pathways.


Assuntos
Depressores do Apetite/farmacologia , Ventrículos Cerebrais/fisiologia , Colestanos/farmacologia , Ingestão de Energia/efeitos dos fármacos , Espermina/farmacologia , Redução de Peso/efeitos dos fármacos , Animais , Anticarcinógenos/administração & dosagem , Anticarcinógenos/farmacologia , Depressores do Apetite/administração & dosagem , Encéfalo/efeitos dos fármacos , Encéfalo/metabolismo , Ventrículos Cerebrais/efeitos dos fármacos , Colestanos/administração & dosagem , Colestanóis/administração & dosagem , Colestanóis/farmacologia , Metabolismo Energético/efeitos dos fármacos , Injeções Intraventriculares , Camundongos , Camundongos Endogâmicos C57BL , Núcleo Hipotalâmico Paraventricular/efeitos dos fármacos , Núcleo Hipotalâmico Paraventricular/fisiologia , Proteínas Proto-Oncogênicas c-fos/efeitos dos fármacos , Proteínas Proto-Oncogênicas c-fos/genética , Proteínas Proto-Oncogênicas c-fos/metabolismo , Ratos , Ratos Sprague-Dawley , Valores de Referência , Espermina/administração & dosagem , Espermina/análogos & derivados
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA