Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 22
Filtrar
1.
Intern Med ; 62(19): 2813-2820, 2023 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-36823091

RESUMO

Objective In recent decades, living conditions have changed drastically. However, there are few data regarding the interaction between living conditions and the risk of ischemic stroke (IS) in young adults. The present study explored the association between living conditions or marital status and the risk factors, etiology, and outcome of IS in young adults. Methods We prospectively enrolled patients with incident IS who were 20-49 years old from 37 clinical stroke centers. We collected the demographic data, living conditions, marital status, vascular risk factors, disease etiology, treatment, and outcomes at discharge. A comparison group was established using the official statistics of Japan. We categorized patients into the two groups based on living conditions: solitary group and cohabiting group. Clinical characteristics were then compared between living conditions. Results In total, 303 patients were enrolled (224 men; median age at the onset: 44 years old). Significant factors associated with the incidence of IS were as follows: solitary status, body mass index >30 kg/m2, current smoking, heavy alcohol consumption, hypertension, diabetes mellitus, and dyslipidemia. Furthermore, in the solitary group, the proportions of men, unmarried individuals, and current smokers were significantly higher than in the cohabiting group. In addition, poor outcomes (modified Rankin Scale ≥4) of IS were more common in the solitary group than in the cohabiting group. Conclusion Our study showed that not only conventional vascular risk factors but also living conditions, especially living alone while unmarried, were independent risk factors for IS in young adults.


Assuntos
AVC Isquêmico , Acidente Vascular Cerebral , Masculino , Humanos , Adulto Jovem , Adulto , Pessoa de Meia-Idade , Condições Sociais , Fatores de Risco , Acidente Vascular Cerebral/epidemiologia , Acidente Vascular Cerebral/etiologia , Fumar/efeitos adversos , Fumar/epidemiologia
2.
Intern Med ; 61(6): 801-810, 2022 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-34483213

RESUMO

Objective Limited data exist regarding the comparative detailed clinical characteristics of patients with ischemic stroke (IS)/transient ischemic attack (TIA) and intracerebral hemorrhage (ICH) receiving oral anticoagulants (OACs). Methods The prospective analysis of stroke patients taking oral anticoagulants (PASTA) registry, a multicenter registry of 1,043 stroke patients receiving OACs [vitamin K antagonists (VKAs) or non-vitamin K antagonist oral anticoagulant (NOACs)] across 25 medical institutions throughout Japan, was used. Univariate and multivariable analyses were used to analyze differences in clinical characteristics between IS/TIA and ICH patients with atrial fibrillation (AF) who were registered in the PASTA registry. Results There was no significant differences in cardiovascular risk factors, such as hypertension, diabetes mellitus, dyslipidemia, smoking, or alcohol consumption (all p>0.05), between IS/TIA and ICH among both NOAC and VKA users. Cerebral microbleeds (CMBs) [odds ratio (OR), 4.77; p<0.0001] were independently associated with ICH, and high brain natriuretic peptide/N-terminal pro B-type natriuretic peptide levels (OR, 1.89; p=0.0390) were independently associated with IS/TIA among NOAC users. A history of ICH (OR, 13.59; p=0.0279) and the high prothrombin time-international normalized ratio (PT-INR) (OR, 1.17; p<0.0001) were independently associated with ICH, and a history of IS/TIA (OR, 3.37; 95% CI, 1.34-8.49; p=0.0101) and high D-dimer levels (OR, 2.47; 95% CI, 1.05-5.82; p=0.0377) were independently associated with IS/TIA among VKA users. Conclusion The presence of CMBs, a history of stroke, natriuretic peptide and D-dimer levels, and PT-INR may be useful for risk stratification of either IS/TIA or ICH development in patients with AF receiving OACs.


Assuntos
Fibrilação Atrial , Acidente Vascular Cerebral Hemorrágico , Acidente Vascular Cerebral , Administração Oral , Anticoagulantes/efeitos adversos , Fibrilação Atrial/complicações , Fibrilação Atrial/tratamento farmacológico , Humanos , Acidente Vascular Cerebral/epidemiologia , Acidente Vascular Cerebral/etiologia , Vitamina K/uso terapêutico
3.
J Neurol Sci ; 417: 117068, 2020 10 15.
Artigo em Inglês | MEDLINE | ID: mdl-32745720

RESUMO

PURPOSE: This study aimed to evaluate the risk factors, etiology, and outcomes of ischemic stroke (IS) in Japanese young adults. METHODS: This was a prospective multicenter study. We enrolled patients aged 16 to 55 years with IS within seven days of the onset of symptoms. We assessed the demographic data, risk factors, stroke etiology, and outcome at discharge. The clinical characteristics were compared between sexes and among age groups. RESULTS: We prospectively enrolled 519 patients (median age, 48 years: 139 females). The mean National Institute of Health Stroke Scale score was 3.6 ± 0.2. The most common risk factors were hypertension (HT) (55%), dyslipidemia (DL) (47%), and current smoking (42%). Body mass index, incidence of current smoking, and heavy alcohol consumption were higher in males. The prevalence of current smoking, HT, DL, and diabetes mellitus increased with aging. The most common etiologic subgroup of IS was small vessel disease (145/510, 28%). Intracranial arterial dissection (IAD) was the most common among the other determined causes (56/115, 49%). The outcome at discharge was relatively good (mRS 0-1, 71.7%); however, poor outcome (mRS ≥ 4) was observed at an incidence of 9.5%. CONCLUSIONS: Most young adults with IS had modifiable risk factors, of which prevalence increased with age. This emphasizes lifestyle improvement to prevent IS in the young population. Furthermore, we indicated that the incidence rate of IAD was high among the other determined causes.


Assuntos
Isquemia Encefálica , AVC Isquêmico , Acidente Vascular Cerebral , Adolescente , Adulto , Isquemia Encefálica/epidemiologia , Feminino , Humanos , Japão/epidemiologia , Masculino , Pessoa de Meia-Idade , Estudos Prospectivos , Fatores de Risco , Acidente Vascular Cerebral/epidemiologia , Acidente Vascular Cerebral/etiologia , Adulto Jovem
4.
Int J Oncol ; 33(1): 5-15, 2008 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-18575745

RESUMO

Focal necrosis is a key pathologic feature that distinguishes glioblastoma from lower grade glioma. The presence of necrosis in a glioblastoma could promote its rapid growth and clinical progression. Focal necrosis of glioblastoma seems to be associated with thrombosis that result from hyper-coagulability. In the present study, we found that glioblastoma cells had a high level of constitutive nuclear factor (NF)-kappaB activity, which was directly correlated with necrosis in glioblastomas. We also found a direct correlation between NF-kappaB activity and the expression of tissue factor (TF), a potent procoagulant factor in gliomas. Inhibition of TF by an inhibitory antibody prevented the procoagulant activity of glioblastoma cells, indicating a TF-dependent mechanism. Blockade of NF-kappaB activation significantly inhibited TF expression and the procoagulant activity of glioblastoma cells in vitro. Blockade of NF-kappaB activation also significantly inhibited in vivo expression of TF, which was directly correlated with decreased necrosis formation and tumor growth of glioblastoma cells in nude mice. Collectively, these results suggest that elevated NF-kappaB activity in glioblastomas cells plays a critical role in necrosis formation of glioblastoma and that inhibition of NF-kappaB activity in glioblastoma can suppress necrosis formation and progressive growth.


Assuntos
Glioblastoma/patologia , NF-kappa B/fisiologia , Tromboplastina/análise , Linhagem Celular Tumoral , Glioblastoma/química , Glioblastoma/metabolismo , Humanos , Proteínas I-kappa B/metabolismo , Imuno-Histoquímica , Interleucina-8/análise , Inibidor de NF-kappaB alfa , NF-kappa B/antagonistas & inibidores , Necrose , Transfecção , Fator A de Crescimento do Endotélio Vascular/análise
5.
J Natl Cancer Inst ; 97(21): 1589-600, 2005 Nov 02.
Artigo em Inglês | MEDLINE | ID: mdl-16264179

RESUMO

BACKGROUND: Several tumor suppressor pathways have been identified as modulators of telomerase function. We examined the functional role of the retinoblastoma-E2F1 pathway in regulating telomerase activity in malignant gliomas. METHODS: Adenovirus vectors were used to transfer cDNAs into human glioblastoma and sarcoma cells. Telomerase activity was assessed with a telomere repeat amplification protocol. Promoter activity in cancer cells was assessed with promoter-luciferase reporter constructs. Promoter binding was assessed with the chromatin immunoprecipitation (ChIP) assay. We isolated astrocytes from E2F1 transgenic mice and normal mice for in vivo studies. We evaluated the expression of E2F1 and hTERT (the catalytic subunit of human telomerase) mRNAs by reverse transcriptase-polymerase chain reaction and proteins in human glioblastoma samples by immunoblot analysis. Associations between survival among 61 glioblastoma multiforme patients and expression of E2F1 and hTERT mRNA and protein were examined with Kaplan-Meier analysis, the log-rank test, and Cox proportional hazards regression models. All statistical tests were two-sided. RESULTS: Ectopic E2F1 expression increased hTERT promoter activity in cancer cells. We detected an interaction between E2F1 protein and the hTERT promoter. Transgenic E2F1 astrocytes contained functional telomerase protein. E2F1 mRNA expression and hTERT mRNA expression were statistically significantly correlated in human glioblastoma specimens (R = .8; P < .001). Longer median survival was statistically significantly associated with lower E2F1 mRNA expression in tumors (103.6 weeks) rather than with higher expression (46.1 weeks) (difference = 57.5 weeks; 95% confidence interval [CI] = 14.7 to 159.7; log-rank P = .002). E2F1 mRNA was the only factor that was statistically significantly associated with overall survival in a multivariable model (P = .04). Among 27 patients with glioblastoma multiforme samples, the expression of E2F1 protein was statistically significantly associated with survival (log-rank P < .001). CONCLUSIONS: E2F1 may participate in telomerase activity regulation in malignant glioma cells. Its expression appears to be strongly associated with the survival of patients with malignant brain tumors.


Assuntos
Neoplasias Encefálicas/metabolismo , Proteínas de Ligação a DNA/metabolismo , Fator de Transcrição E2F1/metabolismo , Ligação Genética , Glioblastoma/metabolismo , Sarcoma/metabolismo , Telomerase/metabolismo , Animais , Neoplasias Encefálicas/enzimologia , Linhagem Celular Tumoral , Imunoprecipitação da Cromatina , Fator de Transcrição E2F1/genética , Regulação Enzimológica da Expressão Gênica , Regulação Neoplásica da Expressão Gênica , Glioblastoma/enzimologia , Humanos , Immunoblotting , Camundongos , Camundongos Transgênicos , Valor Preditivo dos Testes , Prognóstico , Regiões Promotoras Genéticas , RNA Mensageiro/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Sarcoma/enzimologia
6.
Int J Hyperthermia ; 21(7): 615-29, 2005 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-16278167

RESUMO

Adenoviral replacement of the p53 gene has already been proved effective for the treatment of various tumours, including malignant gliomas. However, it is difficult to treat malignant glioma with p53 gene therapy alone because of problems with resistance or a less-than-satisfactory response to the treatment. This study investigated whether heat shock at 43 degrees C (mild hyperthermia) augments the cytotoxic effect of p53 gene transfer on malignant glioma cells expressing wild-type p53 (D54) or mutant p53 (U373-MG and U251-MG). The combination of mild hyperthermia and adenoviral p53 over-expression had an additive inhibitory effect on cellular proliferation in all three cell lines studied. Further, both cell cycle analysis and a DNA fragmentation assay showed that apoptosis was induced by p53 over-expression alone but not by heat shock at 43 degrees C alone. However, p53 over-expression followed by mild hyperthermia additively increased the proportion of cells in which apoptosis was induced, regardless of the endogenous p53 status of the tumour cells. Interestingly, a caspase-independent mechanism was observed to be involved in the p53-induced apoptosis in U251-MG and D54 cells. Taken together, the findings showed that combining adenoviral p53 transfer with mild hyperthermia inhibits the proliferation of malignant glioma cells in an additive manner, irrespective of their endogenous p53 status, suggesting a novel treatment strategy for this malignancy.


Assuntos
Sobrevivência Celular/fisiologia , Terapia Genética , Glioma/terapia , Hipertermia Induzida , Proteína Supressora de Tumor p53/biossíntese , Adenoviridae/genética , Clorometilcetonas de Aminoácidos/farmacologia , Apoptose/fisiologia , Inibidores de Caspase , Terapia Combinada , Inibidor de Quinase Dependente de Ciclina p21/biossíntese , Genes p53 , Humanos , Células Tumorais Cultivadas
7.
Nat Rev Cancer ; 5(9): 726-34, 2005 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-16148885

RESUMO

Autophagy is a process in which subcellular membranes undergo dynamic morphological changes that lead to the degradation of cellular proteins and cytoplasmic organelles. This process is an important cellular response to stress or starvation. Many studies have shed light on the importance of autophagy in cancer, but it is still unclear whether autophagy suppresses tumorigenesis or provides cancer cells with a rescue mechanism under unfavourable conditions. What is the present state of our knowledge about the role of autophagy in cancer development, and in response to therapy? And how can the autophagic process be manipulated to improve anticancer therapeutics?


Assuntos
Autofagia/fisiologia , Morte Celular/fisiologia , Neoplasias/fisiopatologia , Transdução de Sinais/fisiologia , Animais , Antineoplásicos/farmacologia , Humanos , Neoplasias/tratamento farmacológico
8.
Hum Gene Ther ; 16(6): 685-98, 2005 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-15960600

RESUMO

Replacement of the p53 tumor suppressor gene is a rational approach to the management of malignant gliomas because p53 is frequently mutated or inactivated in these cancers. Major weaknesses of this approach are that malignant gliomas are mixtures of cells with wild-type and mutant p53, and that tumor cells exhibiting wildtype p53 are resistant to p53 gene transfer. An effective alternative is needed to overcome these difficulties. p53-upregulated modulator of apoptosis (PUMA) was identified as a p53-inducible proapoptotic molecule. Our purpose was to elucidate a role for PUMA in p53 gene therapy and to investigate whether PUMA is an efficient substitute for p53 in cancer therapy. We demonstrated that PUMA was upregulated in mutant p53 malignant glioma cells (U373-MG and T98G) undergoing apoptosis but was not upregulated in apoptosis-resistant wild-type p53 malignant glioma cells (U87-MG and D54) after adenoviral transfer of p53. Overexpression of PUMA resulted in massive apoptosis associated with mitochondrial damage and caspase-3 activation in all tumor cells tested. Use of the human telomerase reverse transcriptase (hTERT) promoter system induced apoptosis only in malignant glioma cells with telomerase activity, while sparing normal cells lacking telomerase. The ability of PUMA to induce apoptosis was greater than that of caspase-6 or caspase-8 transfer, using the same system. Moreover, exogenous expression of PUMA under the hTERT promoter system significantly suppressed the growth of subcutaneous U87-MG tumors in nude mice and did not induce apoptosis in surrounding nontumor tissues. These results indicate that PUMA, which is regulated under a tumor-specific expression system such as the hTERT promoter, may be better than p53 as a therapeutic tool for malignant gliomas.


Assuntos
Neoplasias do Sistema Nervoso Central/terapia , Terapia Genética/métodos , Glioma/terapia , Proteínas Proto-Oncogênicas/genética , Proteína Supressora de Tumor p53/genética , Adenoviridae/genética , Animais , Apoptose/genética , Proteínas Reguladoras de Apoptose , Caspase 3 , Caspases/genética , Neoplasias do Sistema Nervoso Central/genética , Proteínas de Ligação a DNA/genética , Feminino , Regulação da Expressão Gênica , Vetores Genéticos/genética , Vetores Genéticos/farmacologia , Glioma/genética , Humanos , Camundongos , Camundongos Nus , Mutação , Regiões Promotoras Genéticas , Proteínas Proto-Oncogênicas/farmacologia , Telomerase/genética , Células Tumorais Cultivadas , Proteína Supressora de Tumor p53/metabolismo , Proteína Supressora de Tumor p53/farmacologia
9.
Int J Oncol ; 26(5): 1401-10, 2005 May.
Artigo em Inglês | MEDLINE | ID: mdl-15809734

RESUMO

Autophagy is a novel response of cancer cells to ionizing radiation (IR) or chemotherapy, but its significance or mechanism remains largely elusive. Autophagy is characterized with the prominent formation of autophagic vacuoles in the cytoplasm. It is a protein degradation system that involves autophagic/lysosomal compartment. The process begins with sequestering a portion of the cytoplasm, forming the autophagosome. The autophagosome then fuses with the lysosome and lyses its contents. To study radiation-induced autophagy with specific molecules, we assessed changes in the expression of microtubule-associated protein light chain 3 (LC3) and its intracellular distribution after IR in comparison with starvation-induced autophagy. First, we showed that IR induced cell cycle arrest and autophagy, but not apoptosis, in human malignant glioma U373-MG cells. Type II LC3, that is specifically associated with the membrane of the autophagosome, increased after IR and amino acid starvation. Exogenous LC3 distributed on punctate structures, indicative of the formation of autophagosomes. Autophagy inhibitors, 3-methyladenine and bafilomycin A1, radiosensitized U373-MG cells. Furthermore, gammaH2AX foci, that show the extent of DNA double-strand breaks, were more pronounced and prolonged in the cells treated with IR and autophagy inhibitors than in those cells treated with IR only. Our results suggest that autophagy inhibitors may represent a new application of radiosensitization for malignant glioma cells.


Assuntos
Adenina/análogos & derivados , Autofagia/efeitos da radiação , Neoplasias Encefálicas/patologia , Neoplasias Encefálicas/radioterapia , Glioma/patologia , Glioma/radioterapia , Proteínas Associadas aos Microtúbulos/biossíntese , Adenina/farmacologia , Apoptose , Ciclo Celular , Dano ao DNA , Inibidores Enzimáticos/farmacologia , Regulação Neoplásica da Expressão Gênica/efeitos da radiação , Humanos , Macrolídeos/farmacologia , Proteínas Associadas aos Microtúbulos/antagonistas & inibidores , Tolerância a Radiação
10.
Cancer Res ; 65(8): 3336-46, 2005 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-15833867

RESUMO

The mammalian target of rapamycin (mTOR) is a downstream effector of the phosphatidylinositol 3-kinase (PI3K)/protein kinase B (Akt) signaling pathway and a central modulator of cell proliferation in malignant gliomas. Therefore, the targeting of mTOR signaling is considered a promising therapy for malignant gliomas. However, the mechanisms underlying the cytotoxic effects of a selective mTOR inhibitor, rapamycin, on malignant glioma cells are poorly understood. The purpose of this study was thus to elucidate how rapamycin exerts its cytotoxic effects on malignant glioma cells. We showed that rapamycin induced autophagy but not apoptosis in rapamycin-sensitive malignant glioma U87-MG and T98G cells by inhibiting the function of mTOR. In contrast, in rapamycin-resistant U373-MG cells, the inhibitory effect of rapamycin was minor, although the phosphorylation of p70S6 kinase, a molecule downstream of mTOR, was remarkably inhibited. Interestingly, a PI3K inhibitor, LY294002, and an Akt inhibitor, UCN-01 (7-hydroxystaurosporine), both synergistically sensitized U87-MG and T98G cells as well as U373-MG cells to rapamycin by stimulating the induction of autophagy. Enforced expression of active Akt in tumor cells suppressed the combined effects of LY294002 or UCN-01, whereas dominant-negative Akt expression was sufficient to increase the sensitivity of tumor cells to rapamycin. These results indicate that rapamycin exerts its antitumor effect on malignant glioma cells by inducing autophagy and suggest that in malignant glioma cells a disruption of the PI3K/Akt signaling pathway could greatly enhance the effectiveness of mTOR inhibitors.


Assuntos
Antibióticos Antineoplásicos/farmacologia , Protocolos de Quimioterapia Combinada Antineoplásica/farmacologia , Inibidores Enzimáticos/farmacologia , Glioma/tratamento farmacológico , Inibidores de Fosfoinositídeo-3 Quinase , Proteínas Serina-Treonina Quinases/antagonistas & inibidores , Proteínas Proto-Oncogênicas/antagonistas & inibidores , Sirolimo/farmacologia , Estaurosporina/análogos & derivados , Antibióticos Antineoplásicos/administração & dosagem , Apoptose/efeitos dos fármacos , Autofagia/efeitos dos fármacos , Linhagem Celular Tumoral , Cromonas/administração & dosagem , Sinergismo Farmacológico , Inibidores Enzimáticos/administração & dosagem , Glioma/enzimologia , Glioma/patologia , Humanos , Morfolinas/administração & dosagem , Inibidores de Proteínas Quinases/farmacologia , Proteínas Quinases/metabolismo , Proteínas Proto-Oncogênicas c-akt , Sirolimo/administração & dosagem , Estaurosporina/administração & dosagem , Serina-Treonina Quinases TOR
11.
Int J Oncol ; 26(3): 589-96, 2005 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-15703812

RESUMO

D-24851 is a recently developed microtubule inhibitor that induces G2/M cell-cycle arrest and has an antitumor effect in many cancer cell types. It is expected to be a promising chemotherapeutic agent against a broad range of tumors. However, the precise mechanisms underlying its antitumor effect remain to be determined. Here, we investigated the in vitro effect of D-24851 on tumor growth and the apoptosis mechanism in human malignant glioma cells. Because both p53-dependent and -independent pathways of apoptosis have been reported, we used cell lines with wild-type p53 (U87-MG and D54) and cell lines with mutant p53 (U373-MG and T98G) and compared their responses to D-24851. D-24851 substantially inhibited the proliferation of the four glioma cell lines tested in a dose- and time-dependent manner. The inhibitory effect of D-24851 on tumor growth was associated with cell-cycle arrest in G2/M, subsequently inducing apoptosis. D-24851 treatment induced phosphorylated Bcl-2 and translocated Bax from the cytoplasm to the mitochondria, resulting in apoptotic cell death. These events took place regardless of the p53 status of tumor cells. Our results indicated that D-24851 effectively induces apoptosis through Bcl-2 phosphorylation and Bax translocation in human malignant glioma cells in a p53-independent manner. The results of this study make D-24851 even more promising as a therapeutic agent, especially because many malignant gliomas have a heterogeneous p53 status.


Assuntos
Acetamidas/farmacologia , Apoptose/efeitos dos fármacos , Neoplasias Encefálicas/patologia , Ciclo Celular/efeitos dos fármacos , Glioma/patologia , Indóis/farmacologia , Proteína Supressora de Tumor p53/farmacologia , Proliferação de Células , Relação Dose-Resposta a Droga , Humanos , Fosforilação , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , Células Tumorais Cultivadas , Proteína X Associada a bcl-2
12.
Oncogene ; 24(6): 980-91, 2005 Feb 03.
Artigo em Inglês | MEDLINE | ID: mdl-15592527

RESUMO

Arsenic trioxide (As(2)O(3)) has shown considerable efficacy in treating hematological malignancies with induction of programmed cell death (PCD) type I, apoptosis. However, the mechanisms underlying the antitumor effect of As(2)O(3) on solid tumors are poorly defined. Previously, we reported that As(2)O(3) induced autophagic cell death (PCD type II) but not apoptosis in human malignant glioma cell lines. The purpose of this study was to elucidate the molecular pathway leading to autophagic cell death. In this study, we demonstrated that the cell death was accompanied by involvement of autophagy-specific marker, microtubule-associated protein light chain 3 (LC3), and damage of mitochondrial membrane integrity, but not by caspase activation. Analysis by cDNA microarray, RT-PCR, and Western blot showed that cell death members of Bcl-2 family, Bcl-2/adenovirus E1B 19-kDa-interacting protein 3 (BNIP3) and its homologue BNIP3-like (BNIP3L), were upregulated in As(2)O(3)-induced autophagic cell death. Exogenous expression of BNIP3, but not BNIP3L, induced autophagic cell death in malignant glioma cells without As(2)O(3) treatment. When upregulation of BNIP3 induced by As(2)O(3) was suppressed by a dominant-negative effect of the transmembrane-deleted BNIP3 (BNIP3 Delta TM), autophagic cell death was inhibited. In contrast, BNIP3 transfection augmented As(2)O(3)-induced autophagic cell death. These results suggest that BNIP3 plays a central role in As(2)O(3)-induced autophagic cell death in malignant glioma cells. This study adds a new concept to characterize the pathways by which As(2)O(3) acts to induce autophagic cell death in malignant glioma cells.


Assuntos
Antineoplásicos/farmacologia , Apoptose/efeitos dos fármacos , Apoptose/genética , Arsenicais/farmacologia , Autofagia/efeitos dos fármacos , Neoplasias Encefálicas/patologia , Perfilação da Expressão Gênica , Glioma/patologia , Proteínas de Membrana/biossíntese , Óxidos/farmacologia , Proteínas Proto-Oncogênicas/biossíntese , Trióxido de Arsênio , Western Blotting , Humanos , Proteínas de Membrana/genética , Proteínas de Membrana/farmacologia , Análise de Sequência com Séries de Oligonucleotídeos , Proteínas Proto-Oncogênicas/genética , Proteínas Proto-Oncogênicas/farmacologia , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Células Tumorais Cultivadas , Regulação para Cima
13.
Int J Oncol ; 25(1): 57-63, 2004 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-15201989

RESUMO

Recently, we have reported the therapeutic efficacy of delivering initiator caspase (caspase-8) or executioner active caspase (rev-caspase-6) to telomerase-positive malignant glioma cells using the human telomerase reverse transcriptase (hTERT) gene promoter system (hTERT/caspase-8 or hTERT/rev-caspase-6). In the present study, we investigated if conventional treatments for malignant gliomas augment the efficacy of the hTERT/caspase therapy. First, we demonstrated that hTERT/rev-caspase-6 exhibited a greater ability to induce apoptosis in malignant glioma U87-MG and U373-MG cells than hTERT/caspase-8. Next, as conventional treatments to combine with hTERT/rev-caspase-6, apoptosis-inducing agents [cisplatin (CDDP), paclitaxel (PTX), and BCNU] and non-apoptosis-inducing therapies [temozolomide (TMZ) and gamma-irradiation (IR)] were used. Combination of hTERT/rev-caspase-6 gene therapy with PTX yielded a dose-dependent additive effect, while CDDP and BCNU had additive effect only when tumor cells were treated at IC75 of each agent. A decline in the combination effect of CDDP and BCNU at IC50 was due to decreased activity of telomerase in treated tumor cells prior to the hTERT/rev-caspase-6 transfer. On the other hand, TMZ or IR had no significant additive effect on induction of apoptosis. These results suggest that agents, which induce apoptosis without inhibiting telomerase activity are a promising counterpart to combine with hTERT/rev-caspase-6 therapy for the management of malignant gliomas.


Assuntos
Caspases/metabolismo , Regiões Promotoras Genéticas/genética , Telomerase/genética , Antineoplásicos/farmacologia , Carmustina/farmacologia , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Sobrevivência Celular/efeitos da radiação , Cisplatino/farmacologia , Proteínas de Ligação a DNA , Raios gama , Glioma , Humanos , Toxina Pertussis/farmacologia , Proteínas Recombinantes de Fusão/metabolismo , Telomerase/metabolismo , Transfecção
14.
J Neurooncol ; 68(2): 101-11, 2004 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-15218946

RESUMO

Although hyperthermia has been used as a treatment of malignant brain tumors, it is not yet clear what is the mechanism of the cell growth inhibition by heat shock, especially by the temperature which has clinically been applied to tumor-brain border-zone, 42-43 degrees C. Therefore, we evaluated the change of U251-MG and U87-MG human malignant glioma cells after 43 degrees C-heat shock comparing with that of 45 degrees C. First, we observed that cell growth was transiently inhibited after 43 degrees C-heat shock for 3 or 5 days, in U251-MG or U87-MG cells, respectively, which was followed by regrowth. During the period of transient growth inhibition, mild G2/M arrest was observed. However, apoptosis was observed in only 2.7% or 1.5%, of 43 degrees C-heated cells, in U251-MG or U87-MG cells, respectively. Instead, transmission electron micrography showed the formation of vacuoles, degeneration of mitochondria, and autophagosomes. Moreover, in the both cell lines, flow-cytometric analysis with acridine orange revealed the induction of acidic vesicle organelles, which was blocked by 3-methyladenine (3-MA), suggesting the involvement of autophagy. Furthermore, while 3-MA did not increase the anti-tumor effect of 43 degrees C-heat shock, bafilomycin A1, another autophagy inhibitor, did significantly enhance the effect in U251-MG cells. Taken together, mild heat shock (43 degrees C for 2 h) causes autophagy and mild G2/M arrest, but does not induce apparent apoptosis in U251-MG and U87-MG glioma cells. Inhibition of autophagy with bafilomycin A1 may increase the anti-tumor efficacy of mild heat shock against some malignant glioma cells.


Assuntos
Adenina/análogos & derivados , Apoptose/fisiologia , Autofagia/fisiologia , Neoplasias Encefálicas/patologia , Divisão Celular/fisiologia , Glioma/patologia , Adenina/farmacologia , Apoptose/efeitos dos fármacos , Autofagia/efeitos dos fármacos , Neoplasias Encefálicas/ultraestrutura , Ciclo Celular/efeitos dos fármacos , Divisão Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Inibidores Enzimáticos/farmacologia , Temperatura Alta , Humanos , Macrolídeos/farmacologia , Organelas/patologia , Organelas/ultraestrutura , Termodinâmica
15.
Cancer Res ; 64(12): 4286-93, 2004 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-15205343

RESUMO

The sphingolipid ceramide has been recognized as an important second messenger implicated in regulating diverse signaling pathways especially for apoptosis. Very little is known, however, about the molecular mechanisms underlying nonapoptotic cell death induced by ceramide. In the present study, we first demonstrate that ceramide induces nonapoptotic cell death in malignant glioma cells. The cell death was accompanied by several specific features characteristic of autophagy: presence of numerous autophagic vacuoles in the cytoplasm, development of the acidic vesicular organelles, autophagosome membrane association of microtubule-associated protein light chain 3 (LC3), and a marked increase in expression levels of two forms of LC3 protein (LC3-I and LC3-II). We additionally demonstrate that ceramide decreases mitochondrial membrane potential and activates the transcription of death-inducing mitochondrial protein, BNIP3, resulting in increased expression levels of its mRNA and protein in malignant glioma cells. Moreover, tumor cells transfected with BNIP3 gene undergo autophagy in the absence of ceramide. These results suggest that ceramide induces autophagic cell death in malignant glioma cells via activation of BNIP3. This study adds a new concept to characterize the pathways by which ceramide acts to induce nonapoptotic autophagic cell death in malignant gliomas.


Assuntos
Glioma/patologia , Proteínas de Membrana/fisiologia , Proteínas Proto-Oncogênicas/fisiologia , Esfingosina/análogos & derivados , Esfingosina/farmacologia , Autofagia/efeitos dos fármacos , Autofagia/fisiologia , Morte Celular/efeitos dos fármacos , Morte Celular/fisiologia , Linhagem Celular Tumoral , Fase G2/efeitos dos fármacos , Fase G2/fisiologia , Glioma/tratamento farmacológico , Glioma/genética , Glioma/metabolismo , Humanos , Membranas Intracelulares/efeitos dos fármacos , Membranas Intracelulares/fisiologia , Potenciais da Membrana/efeitos dos fármacos , Proteínas de Membrana/biossíntese , Proteínas de Membrana/genética , Mitocôndrias/efeitos dos fármacos , Mitocôndrias/fisiologia , Mitose/efeitos dos fármacos , Mitose/fisiologia , Proteínas Proto-Oncogênicas/biossíntese , Proteínas Proto-Oncogênicas/genética , Transfecção
16.
Int J Oncol ; 23(6): 1703-8, 2003 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-14612944

RESUMO

The cyclin-dependent kinase inhibitor p27KIP1 is considered not only a prognostic factor in cancer, but also a promising anti-cancer agent. However, the relationship between p27KIP1 and telomerase, that has potential as tumor-marker, remains to be elucidated. In this study, using the recombinant adenoviral vector expressing p27KIP1 (Adp27KIP1), we investigated whether p27KIP1 affects telomerase activity in malignant glioma U373-MG cells. Overexpression of p27KIP1 suppressed telomerase activity in tumor cells. The down-regulation of telomerase was due to inhibition of the human telomerase reverse transcriptase (hTERT) gene expression at the transcriptional level. This inhibitory effect was partially induced by interfering with binding sites of the hTERT core promoter for transcription factors Myc and Sp1. These findings identify a novel role for p27KIP1 in down-regulation of telomerase activity.


Assuntos
Neoplasias Encefálicas/enzimologia , Proteínas de Ciclo Celular/metabolismo , Regulação para Baixo , Glioma/enzimologia , Telomerase/metabolismo , Proteínas Supressoras de Tumor/metabolismo , Sítios de Ligação , Linhagem Celular Tumoral , Inibidor de Quinase Dependente de Ciclina p27 , Proteínas de Ligação a DNA , Fibroblastos/metabolismo , Regulação da Expressão Gênica , Humanos , Luciferases/metabolismo , Prognóstico , Proteínas Proto-Oncogênicas c-myc/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Fator de Transcrição Sp1
17.
Cancer Res ; 63(9): 2103-8, 2003 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-12727826

RESUMO

Recent clinical data shows that arsenic trioxide (As(2)O(3)) causes remission in patients with acute promyelocytic leukemia and multiple myeloma without severe side effects. Laboratory data suggest that As(2)O(3) induces apoptosis or cell differentiation of hematopoietic or solid tumor cells. To date, there has been no study on the effects of As(2)O(3) on glioma cells. In this study, we investigated the in vitro effect of As(2)O(3) on cell growth inhibition and cell death mechanisms in human glioma cells. As(2)O(3) significantly inhibited the proliferation of all six of the glioma cell lines (U373, U87, U251, GB1, A-172, and T98G) tested in this study in a dose-dependent manner. The IC(50) of As(2)O(3) for all of the tumor cell lines was <2 micro M. Previous studies have shown that this is a clinically safe concentration. Treatment with 2 micro M As(2)O(3) induced G(2)/M arrest in all of the glioma cell lines. Autophagy (programmed cell death type II), but not apoptosis (programmed cell death type I), was detected by electron microscopy in U-373-MG cells treated with 2 micro M As(2)O(3). Caspase inhibitors did not halt As(2)O(3)-induced cell death. Furthermore, combination of As(2)O(3) with bafilomycin A1 autophagy inhibitor enhanced the antitumor effect of As(2)O(3) through induction of apoptosis. These findings suggest that As(2)O(3) at a clinically safe concentration may be an effective chemotherapeutic agent for malignant gliomas.


Assuntos
Antineoplásicos/farmacologia , Arsenicais/farmacologia , Glioma/tratamento farmacológico , Macrolídeos , Óxidos/farmacologia , Clorometilcetonas de Aminoácidos/farmacologia , Antibacterianos/farmacologia , Apoptose/efeitos dos fármacos , Trióxido de Arsênio , Autofagia/efeitos dos fármacos , Inibidores de Caspase , Divisão Celular/efeitos dos fármacos , Citoplasma/efeitos dos fármacos , Citoplasma/metabolismo , Interações Medicamentosas , Inibidores Enzimáticos/farmacologia , Fase G2/efeitos dos fármacos , Glioma/patologia , Humanos , Mitose/efeitos dos fármacos , Células Tumorais Cultivadas
18.
J Biomed Biotechnol ; 2003(1): 25-34, 2003.
Artigo em Inglês | MEDLINE | ID: mdl-12686720

RESUMO

Malignant gliomas are the most common neoplasm in the central nervous system. When treated with conventional treatments including surgery, irradiation, and chemotherapy, the average life expectancy of the most malignant type, glioblastoma multiforme is usually less than 1 year. Therefore, gene therapy is expected to be an effective and possibly curative treatment. Many gene therapeutic approaches have demonstrated efficacy in experimental animal models. However, the current clinical trials are disappointing. This review focuses on current therapeutic genes/vectors/delivery systems/targeting strategies in order to introduce updated trends and hopefully indicate prospective gene therapy for malignant gliomas.

19.
J Neurosurg ; 98(2): 378-84, 2003 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-12593626

RESUMO

OBJECT: Ionizing radiation is the gold-standard adjuvant treatment for glioblastoma multiforme (GBM), the most aggressive primary brain tumor. The mechanisms underlying neoplastic glial cell growth inhibition after administration of ionizing radiation, however, remain largely unknown. In this report, the authors characterize the response of GBM cells to ionizing radiation and elucidate factors that correlate with the radiosensitivity of these tumors. METHODS: Six human GBM cell lines were subjected to increasing doses of radiation. Each demonstrated a dose-dependent suppression of cell proliferation. In the most radiosensitive cell line, the authors demonstrated a transient increase in the expression of the cyclin-dependent kinase inhibitors (CDKIs) p21 and p27, which corresponded with a G1 cell-cycle arrest. In contrast, the most radioresistant cell line demonstrated a decrease in p21 and p27 expression levels, which correlated with a failure to arrest. Apoptosis did not occur in any cell line following irradiation. Instead, autophagic cell changes were observed following administration of radiation, regardless of the relative radiosensitivity of the cell line. CONCLUSIONS: These findings elucidate some of the molecular responses of GBMs to irradiation and suggest novel targets for future therapy.


Assuntos
Autofagia/genética , Autofagia/efeitos da radiação , Neoplasias Encefálicas/patologia , Neoplasias Encefálicas/radioterapia , Proteínas de Ciclo Celular/efeitos da radiação , Ciclinas/efeitos da radiação , Inibidores Enzimáticos/efeitos da radiação , Fase G1/genética , Fase G1/efeitos da radiação , Glioblastoma/patologia , Glioblastoma/radioterapia , Radiação Ionizante , Proteínas Supressoras de Tumor/efeitos da radiação , Neoplasias Encefálicas/genética , Proteínas de Ciclo Celular/análise , Inibidor de Quinase Dependente de Ciclina p21 , Inibidor de Quinase Dependente de Ciclina p27 , Ciclinas/análise , Inibidores Enzimáticos/análise , Glioblastoma/genética , Humanos , Marcação In Situ das Extremidades Cortadas , Técnicas In Vitro , Organelas/genética , Organelas/efeitos da radiação , Tolerância a Radiação/genética , Tolerância a Radiação/efeitos da radiação , Fatores de Tempo , Células Tumorais Cultivadas/patologia , Células Tumorais Cultivadas/efeitos da radiação , Proteínas Supressoras de Tumor/análise
20.
J Neurosurg ; 99(6): 1047-52, 2003 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-14705733

RESUMO

OBJECT: Temozolomide (TMZ) is a DNA alkylating agent currently used as adjuvant treatment for anaplastic astrocytomas. Its use in managing glioblastoma multiforme has been halted because of the lack of therapeutic effects due to cell resistance. Note that O6-alkylguanine-DNA alkyltranferase (AGT) is a DNA repair enzyme that limits the efficacy of TMZ. In this study the authors investigated the ability of O6-benzylguanine (BG), an AGT inhibitor, to sensitize a glioblastoma cell line resistant to TMZ. METHODS: The effects of TMZ alone (100 microg) and after exposure to BG (50 microg) were assessed in two glioblastoma cell lines, U373-MG and T98G, respectively, sensitive and resistant to TMZ. Cell viability was assessed using trypan blue; cell cycle analysis by fluorescence-activated cell sorter; and apoptosis and autophagy by terminal deoxynucleotidyl transferase-mediated deoxyuridine triphosphate nick-end labeling (TUNEL) and acridine orange staining, respectively. Furthermore, the involvement of an autophagy marker, microtubule-associated light chain 3 (LC3), was assessed. Temozolomide suppressed the growth of and caused cell cycle arrest in the G2-M phase of U373-MG cells but not T98G cells. Exposure to BG prior to TMZ resulted in a significant decrease in cell viability as well as cell cycle arrest in the G2-M phase in T98G cells (p < 0.05). Although apoptosis was not detected on TUNEL staining, programmed cell death Type II (autophagy) was detected after exposure to BG and TMZ in T98G cells. CONCLUSIONS: These results indicate that inhibition of AGT by BG can render previously resistant glioma cells sensitive to TMZ treatment. The mechanism of cell demise following BG-TMZ treatment seems to be autophagy and not apoptosis. Combination therapy involving TMZ and an AGT inhibitor may be an effective strategy to treat resistant gliomas.


Assuntos
Antineoplásicos Alquilantes/uso terapêutico , Reparo do DNA/efeitos dos fármacos , Dacarbazina/análogos & derivados , Dacarbazina/uso terapêutico , Inibidores Enzimáticos/farmacologia , Glioma/genética , Guanina/análogos & derivados , Guanina/farmacologia , Apoptose/genética , Linhagem Celular Tumoral , Sobrevivência Celular/genética , Resistencia a Medicamentos Antineoplásicos/efeitos dos fármacos , Glioma/tratamento farmacológico , Humanos , O(6)-Metilguanina-DNA Metiltransferase/antagonistas & inibidores , Temozolomida
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA