Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 63
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
J Oncol Pharm Pract ; 22(3): 517-22, 2016 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-25616657

RESUMO

BACKGROUND: Temozolomide is an oral alkylating agent with schedule-dependent antitumor activity against high-grade malignancies including high-grade glioma. Increasingly, reports have suggested that temozolomide may have activity as a salvage therapy for aggressive, recurrent pituitary adenomas or carcinomas that fail surgery, radiation and other pharmacotherapy. To our knowledge, temozolomide retreatment following initial responsiveness has not previously been demonstrated. CASE REPORT: A woman was diagnosed with a prolactin-secreting pituitary adenoma in 1995 (age 44). Despite bromocriptine therapy, transphenoidal resection, radiotherapy, and cabergoline treatment she experienced continued clinico-radiographic progression, and temozolomide was initiated in 2011. She received three treatment cycles with rapid, dramatic clinico-radiographic response, and 99.3% reduction in serum prolactin. After three years of close observation, she developed recurrent radiographic progression and prolactin elevation. She was re-initiated on temozolomide, and after four cycles, clinical, radiographic and hormonal response was observed with a 92.2% reduction in serum prolactin. CONCLUSIONS/SUMMARY: Temozolomide is an increasingly described treatment option for refractory pituitary adenomas and carcinomas. In the current report, we document rapid biochemical response following retreatment with temozolomide in aggressive pituitary adenoma. When "off label" salvage therapy with temozolomide is offered for patients with recurrent prolactinomas, retreatment at the time of recurrence can be considered.


Assuntos
Adenoma/tratamento farmacológico , Antineoplásicos Alquilantes/administração & dosagem , Dacarbazina/análogos & derivados , Recidiva Local de Neoplasia/tratamento farmacológico , Neoplasias Hipofisárias/tratamento farmacológico , Prolactinoma/tratamento farmacológico , Adenoma/sangue , Adenoma/diagnóstico por imagem , Dacarbazina/administração & dosagem , Feminino , Humanos , Imageamento por Ressonância Magnética/tendências , Pessoa de Meia-Idade , Recidiva Local de Neoplasia/sangue , Recidiva Local de Neoplasia/diagnóstico por imagem , Neoplasias Hipofisárias/sangue , Neoplasias Hipofisárias/diagnóstico por imagem , Prolactina/sangue , Prolactinoma/sangue , Prolactinoma/diagnóstico por imagem , Retratamento/métodos , Temozolomida , Resultado do Tratamento
2.
Magn Reson Med ; 74(3): 599-606, 2015 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-26151840

RESUMO

PURPOSE: To develop the simultaneous acquisition of multiple voxels in localized MR spectroscopy (MRS) using sensitivity encoding, allowing reduced total scan time compared to conventional sequential single voxel (SV) acquisition methods. METHODS: Dual volume localization was used to simultaneously excite voxels in both hemispheres. Receiver coil sensitivity profiles were used to unfold the data. To demonstrate the method, MRS voxels in the left and right hippocampus were measured at 3 tesla (T) and the left and right motor cortices at 7T. Spectra were compared to conventional SV acquisitions. Spectra were also recorded from the lesion and contralateral hemisphere of a patient with a low-grade oligodendroglioma at 7T. RESULTS: It was possible to generate signal in two voxels simultaneously and separate the signal originating from the different locations, with spectral results almost identical to those observed using conventional single voxel methods. The method results in an increased chemical shift displacement artifact, which might be improved by advanced pulse designs, and a noise increase due to the unfolding g-factor, which was larger at 3T than 7T. CONCLUSION: The simultaneous acquisition of voxels for MRS is possible by using modulated slice-selective pulses and receive coil sensitivity profiles to unfold the resulting signals.


Assuntos
Processamento de Imagem Assistida por Computador/métodos , Imageamento por Ressonância Magnética/métodos , Algoritmos , Encéfalo/patologia , Neoplasias Encefálicas/patologia , Humanos , Oligodendroglioma/patologia , Imagens de Fantasmas
3.
J Cell Biol ; 96(1): 112-23, 1983 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-6219115

RESUMO

Proteins with affinities for specific glycosaminoglycans (GAC's) were used as probes for testing the potential of cell surface GAG's to mediate cell adhesive responses to extracellular matrices (ECM). Plasma fibronectin (FN) and proteins that bind hyaluronate (cartilage proteo-glycan core and link proteins) or heparan sulfate (platelet factor 4 [PF4]) were adsorbed to inert substrata to evaluate attachment and spreading of several 3T3 cell lines. Cells failed to attach to hyaluronate-binding substrata. The rates of attachment on PF4 were identical to those on FN; however, PF4 stimulated formation of broad convex lamellae but not tapered cell processes fibers during the spreading response. PF4-mediated responses were blocked by treating the PF4-adsorbed substratum with heparin (but not chondroitin sulfate), or alternatively the cells with Flavobacter heparinum heparinase (but not chondroitinase ABC). Heparinase treatment did not inhibit cell attachment to FN but did inhibit spreading. Cells spread on PF4 or FN contained similar Ca2+-independent cell-substratum adhesions, as revealed by EGTA-mediated retraction of their substratum-bound processes. Microtubular networks reorganized in cells on PF4 but failed to extend into the broadly spread lamellae, where fine microfilament bundles had developed. Stress fibers, common on FN, failed to develop on PF4. These experiments indicate that (a) heparan sulfate proteoglycans are critical mediators of cell adhesion and heparan sulfate-dependent adhesion via PF4 is comparable in some, but not all, ways to FN-mediated adhesion, (b) the uncharacterized and heparan sulfate-independent "cell surface" receptor for FN permits some but not all aspects of adhesion, and (c) physiologically compatible and complete adhesion of fibroblasts requires binding of extracellular matrix FN to both the unidentified "cell surface" receptor and heparan sulfate proteoglycans.


Assuntos
Fatores de Coagulação Sanguínea/farmacologia , Adesão Celular , Corrente Citoplasmática , Fibronectinas/farmacologia , Glicosaminoglicanos/fisiologia , Heparitina Sulfato/fisiologia , Fator Plaquetário 4/farmacologia , Animais , Adesão Celular/efeitos dos fármacos , Linhagem Celular , Sulfatos de Condroitina/farmacologia , Condroitinases e Condroitina Liases/farmacologia , Citoplasma/ultraestrutura , Corrente Citoplasmática/efeitos dos fármacos , Citoesqueleto/ultraestrutura , Fibroblastos , Heparina/farmacologia , Heparina Liase , Ácido Hialurônico/fisiologia , Camundongos , Polissacarídeo-Liases/farmacologia
4.
Oncogene ; 26(33): 4774-96, 2007 Jul 19.
Artigo em Inglês | MEDLINE | ID: mdl-17297451

RESUMO

Scatter factor (SF) (hepatocyte growth factor) is a pleiotrophic cytokine that accumulates within tumors in vivo and protects tumor cells against cytotoxicity and apoptosis due to DNA damaging agents in vitro. Previous studies have established that SF-mediated cell protection involves antiapoptotic signaling from its receptor (c-Met) to PI3 kinase --> c-Akt --> Pak1 (p21-activated kinase -1) --> NF-kappaB (nuclear factor-kappa B). Here, we found that Ras proteins (H-Ras and R-Ras) enhance SF-mediated activation of NF-kappaB and protection of DU-145 and MDCK (Madin-Darby canine kidney) cells against the topoisomerase IIalpha inhibitor adriamycin. Studies of Ras effector loop mutants and their downstream effectors suggest that Ras/PI3 kinase and Ras/Raf1 pathways contribute to SF stimulation of NF-kappaB signaling and cell protection. Further studies revealed that Raf1 positively regulates the ability of SF to stimulate NF-kappaB activity and cell protection. The ability of Raf1 to stimulate NF-kappaB activity was not due to the classical Raf1 --> MEK1/2 --> ERK1/2 pathway. However, we found that a MEK3/6 --> p38 pathway contributes to SF-mediated activation of NF-kappaB. In contrast, RalA, a target of the Ras/RalGDS pathway negatively regulated the ability of SF to stimulate NF-kappaB activity and cell protection. Ras, Raf1 and RalA modulate SF stimulation of NF-kappaB activity, in part, by regulating IkappaB kinase (IKK)-beta kinase activity. These findings suggest that Ras/Raf1/RalA pathways may converge to modulate NF-kappaB activation and SF-mediated survival signaling at the IKK complex and/or a kinase upstream of this complex.


Assuntos
Dano ao DNA , Fator de Crescimento de Hepatócito/metabolismo , NF-kappa B/metabolismo , Transdução de Sinais/fisiologia , Proteínas ras/metabolismo , Animais , Antibióticos Antineoplásicos/farmacologia , Linhagem Celular , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Relação Dose-Resposta a Droga , Doxorrubicina/farmacologia , Vetores Genéticos/genética , Fator de Crescimento de Hepatócito/genética , Humanos , Quinase I-kappa B/genética , Quinase I-kappa B/metabolismo , Proteínas Quinases Ativadas por Mitógeno/genética , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Mutação , NF-kappa B/genética , Fosforilação , Ligação Proteica , Proteínas Proto-Oncogênicas c-raf/genética , Proteínas Proto-Oncogênicas c-raf/metabolismo , Interferência de RNA , Transdução de Sinais/genética , Transfecção , Proteínas ral de Ligação ao GTP/genética , Proteínas ral de Ligação ao GTP/metabolismo , Proteínas ras/genética
5.
Oncogene ; 26(20): 2925-38, 2007 May 03.
Artigo em Inglês | MEDLINE | ID: mdl-17099727

RESUMO

The cytokine scatter factor (SF) (hepatocyte growth factor) transduces various biologic actions, including cell motility, invasion, angiogenesis and apoptosis inhibition. The latter is relevant to understanding the role of SF in promoting tumor cell survival in different contexts, for example, detachment from basement membrane, growth in metastatic sites and responses to chemo- and radiotherapy. Previously, we showed that SF protects cells against apoptosis owing to DNA damage, by a mechanism involving phosphoinositol-3-kinase/c-Akt signaling. Here, we used DNA microarray assays to identify c-Akt-regulated genes that might contribute to cell protection. DU-145 human prostate cancer cells were transfected+/-a dominant-negative mutant Akt, treated+/-SF and analysed for gene expression using Affymetrix arrays. These studies identified SF-regulated genes for which induction was c-Akt-dependent vs -independent. Selected microarray findings were confirmed by semiquantitative and quantitative reverse transcription-polymerase chain reaction. We tested the contribution of four SF-inducible/c-Akt-dependent genes (AMPD3, EPHB2, MX1 and WNT4) to protection against adriamycin (a DNA topoisomerase IIalpha inhibitor) using RNA interference. Knockdown of each gene except EPHB2 caused a small but significant reduction in the SF cell protection. The lack of effect of EPHB2 knockdown may be due to the fact that DU-145 cells contain a single-mutant EPHB2 allele. A combination of three small interfering RNAs blocked most of the protection by SF in both DU-145 and T47D cells. These findings identify novel c-Akt-regulated genes, some of which contribute to SF-mediated cytoprotection.


Assuntos
Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Fator de Crescimento de Hepatócito/fisiologia , Proteína Oncogênica v-akt/antagonistas & inibidores , Neoplasias da Próstata/genética , RNA Interferente Pequeno/farmacologia , Doxorrubicina/farmacologia , Resistencia a Medicamentos Antineoplásicos/genética , Perfilação da Expressão Gênica , Humanos , Masculino , NF-kappa B/genética , NF-kappa B/fisiologia , Análise de Sequência com Séries de Oligonucleotídeos , Proteína Oncogênica v-akt/genética , Proteína Oncogênica v-akt/fisiologia , Neoplasias da Próstata/patologia , Proteínas Serina-Treonina Quinases/genética , Proteínas Serina-Treonina Quinases/fisiologia , Células Tumorais Cultivadas , Quinases Ativadas por p21
6.
J Natl Cancer Inst ; 91(18): 1548-56, 1999 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-10491431

RESUMO

BACKGROUND: Expression of scatter factor (SF), also known as hepatocyte growth factor (HGF), and its receptor, c-met, is often associated with malignant progression of human tumors, including gliomas. Overexpression of SF/HGF in experimental gliomas enhances tumorigenicity and tumor-associated angiogenesis (i.e., growth of new blood vessels). However, the role of endogenous SF/HGF or c-met expression in the malignant progression of gliomas has not been examined directly. In this study, we tested the hypothesis that human glioblastomas can be SF/HGF-c-met dependent and that a reduction in endogenous SF/HGF or c-met expression can lead to inhibition of tumor growth and tumorigenicity. METHODS: Expression of the SF/HGF and c-met genes was inhibited by transfecting glioblastoma cells with chimeric transgenes consisting of U1 small nuclear RNA, a hammerhead ribozyme, and antisense sequences. The effects of reduced SF/HGF and c-met expression on 1) SF/HGF-dependent induction of immediate early genes (c-fos and c-jun), indicative of signal transduction; 2) anchorage-independent colony formation (clonogenicity), an in vitro correlate of solid tumor malignancy; and 3) intracranial tumor formation in immunodeficient mice were quantified. Statistical tests were two-sided. RESULTS: Introduction of the transgenes into glioblastoma cells reduced expression of the SF/HGF and c-met genes to as little as 2% of control cell levels. Reduction in c-met expression specifically inhibited SF/HGF-dependent signal transduction (P<.01). Inhibition of SF/HGF or c-met expression in glioblastoma cells possessing an SF/HGF-c-met autocrine loop reduced tumor cell clonogenicity (P =.005 for SF/HGF and P=.009 for c-met) and substantially inhibited tumorigenicity (P<.0001) and tumor growth in vivo (P<.0001). CONCLUSIONS: To our knowledge, this is the first successful inhibition of SF/HGF and c-met expression in a tumor model directly demonstrating a role for endogenous SF/HGF and c-met in human glioblastoma. Our results suggest that targeting the SF/HGF-c-met signaling pathway may be an important approach in controlling tumor progression.


Assuntos
Regulação Neoplásica da Expressão Gênica , Glioblastoma/genética , Fator de Crescimento de Hepatócito/antagonistas & inibidores , Proteínas Proto-Oncogênicas c-met/antagonistas & inibidores , RNA Catalítico/genética , RNA Neoplásico/metabolismo , RNA Nuclear Pequeno/genética , Animais , Northern Blotting , Adesão Celular , Divisão Celular , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Terapia Genética , Glioblastoma/metabolismo , Glioblastoma/patologia , Glioblastoma/terapia , Fator de Crescimento de Hepatócito/genética , Fator de Crescimento de Hepatócito/metabolismo , Humanos , Hibridização In Situ , Camundongos , Camundongos Nus , Transplante de Neoplasias , Fenótipo , Proteínas Proto-Oncogênicas c-met/genética , Proteínas Proto-Oncogênicas c-met/metabolismo , RNA Antissenso/genética , RNA Antissenso/uso terapêutico , RNA Catalítico/metabolismo , RNA Neoplásico/genética , RNA Nuclear Pequeno/uso terapêutico , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/uso terapêutico , Transdução de Sinais , Células Tumorais Cultivadas , Regulação para Cima/efeitos dos fármacos
7.
Cancer Res ; 60(15): 4277-83, 2000 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-10945642

RESUMO

We have shown recently that the multifunctional growth factor, scatter factor/hepatocyte growth factor (SF/HGF), and its receptor c-met enhance the malignancy of human glioblastoma through an autocrine stimulatory loop (R. Abounader et al., J. Natl. Cancer Inst., 91: 1548-1556, 1999). This report examines the effects of SF/HGF:c-met signaling on human glioma cell responses to DNA-damaging agents. Pretreating U373 human glioblastoma cells with recombinant SF/HGF partially abrogated their cytotoxic responses to gamma irradiation, cisplatin, camptothecin, Adriamycin, and Taxol in vitro. This cytoprotective effect of SF/HGF occurred at least in part through an inhibition of apoptosis, as evidenced by diminished terminal deoxynucleotidyl transferase-mediated dUTP nick-end labeling index and reduced DNA laddering. Anti-c-met U1/ribozyme gene transfer inhibited the ability of SF/HGF to protect against single-strand DNA breakage, DNA fragmentation, and glioblastoma cell death caused by DNA-damaging agents, demonstrating a requirement for c-met receptor function. Phosphorylation of the cell survival-promoting kinase Akt (protein kinase B) resulted from SF/HGF treatment of U373 cells, and both Akt phosphorylation and cell survival induced by SF/HGF were inhibited by phosphatidylinositol 3-kinase inhibitors but not by inhibitors of mitogen-activated protein kinase kinase or protein kinase C. Cytoprotection by SF/HGF in vitro was also inhibited by transient expression of dominant-negative Akt. Transgenic SF/HGF expression by intracranial 9L gliosarcomas reduced tumor cell sensitivity to gamma irradiation, confirming the cytoprotective effect of SF/HGF in vivo. These findings demonstrate that c-met receptor activation by SF/HGF protects certain glioblastoma cells from DNA-damaging agents by activating phosphoinositol 3-kinase-dependent and Akt-dependent antiapoptotic pathways.


Assuntos
Apoptose/efeitos dos fármacos , Glioblastoma/patologia , Fator de Crescimento de Hepatócito/farmacologia , Fosfatidilinositol 3-Quinases/fisiologia , Proteínas Serina-Treonina Quinases , Proteínas Proto-Oncogênicas/fisiologia , Transdução de Sinais/efeitos dos fármacos , Animais , Apoptose/fisiologia , Expressão Gênica , Técnicas de Transferência de Genes , Glioblastoma/enzimologia , Gliossarcoma/enzimologia , Gliossarcoma/patologia , Fator de Crescimento de Hepatócito/fisiologia , Humanos , Masculino , Proteínas Tirosina Quinases/fisiologia , Proteínas Proto-Oncogênicas c-akt , Proteínas Proto-Oncogênicas c-met/antagonistas & inibidores , Proteínas Proto-Oncogênicas c-met/genética , Proteínas Proto-Oncogênicas c-met/fisiologia , RNA Catalítico/genética , Ratos , Ratos Endogâmicos F344 , Proteínas Recombinantes/farmacologia , Ribonucleoproteína Nuclear Pequena U1/genética , Transdução de Sinais/fisiologia , Células Tumorais Cultivadas
8.
Cancer Res ; 58(13): 2784-92, 1998 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-9661892

RESUMO

The novel aminosterol, squalamine, inhibits angiogenesis and tumor growth in multiple animal models. This effect is mediated, at least in part, by blocking mitogen-induced proliferation and migration of endothelial cells, thus preventing neovascularization of the tumor. Squalamine has no observable effect on unstimulated endothelial cells, is not directly cytotoxic to tumor cells, does not alter mitogen production by tumor cells, and has no obvious effects on the growth of newborn vertebrates. Squalamine was also found to have remarkable effects on the primitive vascular bed of the chick chorioallantoic membrane, which has striking similarities to tumor capillaries. Squalamine may thus be well suited for treatment of tumors and other diseases characterized by neovascularization in humans.


Assuntos
Anticarcinógenos/farmacologia , Neovascularização Fisiológica/efeitos dos fármacos , Animais , Divisão Celular/efeitos dos fármacos , Movimento Celular/efeitos dos fármacos , Embrião de Galinha/efeitos dos fármacos , Colestanóis/farmacologia , Colágeno , Córnea , Neovascularização da Córnea/prevenção & controle , Combinação de Medicamentos , Fatores de Crescimento Endotelial/metabolismo , Fatores de Crescimento Endotelial/farmacologia , Endotélio Vascular/efeitos dos fármacos , Neoplasias Oculares/prevenção & controle , Fator 2 de Crescimento de Fibroblastos/farmacologia , Glioma/tratamento farmacológico , Glioma/patologia , Laminina , Linfocinas/efeitos dos fármacos , Linfocinas/metabolismo , Linfocinas/farmacologia , Camundongos , Camundongos Endogâmicos BALB C , Fator de Crescimento Derivado de Plaquetas/farmacologia , Proteoglicanas , Coelhos , Ratos , Ratos Endogâmicos F344 , Transplante Heterólogo , Fator A de Crescimento do Endotélio Vascular , Fatores de Crescimento do Endotélio Vascular
9.
Oncogene ; 35(37): 4903-13, 2016 09 15.
Artigo em Inglês | MEDLINE | ID: mdl-26898758

RESUMO

Solid malignancies contain subsets of multipotent cells that grow as spheres and efficiently propagate tumors in xenograft models, reflecting a stem-like, self-renewing and tumor-propagating phenotype. These cancer 'stem cells (SCs)' have been shown to maintain tumor growth, contribute to resistance and drive tumor recurrence. Cancer cell stemness is dynamically influenced by epigenetic mechanisms and differentially regulated coding and noncoding RNAs. How these mechanisms specifically contribute to the generation and/or maintenance of cancer SCs remains unclear. This study identifies a novel epigenetically regulated circuit that integrates microRNA, chromatin remodeling and the reprogramming transcription factor Sox2 to regulate glioblastoma (GBM)-propagating SCs. We show that miR-296-5p expression is repressed in a DNA methylation-dependent manner under conditions that promote GBM cell stemness and that miR-296-5p inhibits GBM cell stemness and their capacity to self-renew as spheres and propagate glioma xenografts in vivo. We show that the chromatin remodeling protein HMGA1 functions as a downstream effector of these biological responses to miR-296-5p and regulates Sox2 expression, a master driver of cell stemness, by modifying chromatin architecture at the Sox2 promoter. These results show for the first time that miR-296-5p inhibits transcriptional mechanisms that support GBM SCs and identify a miR-296-5p:HMGA1:Sox2 axis as a novel regulator of GBM SCs and candidate pathway for targeting therapies directed at depleting tumors of their tumor-propagating stem cell subsets.


Assuntos
Metilação de DNA/genética , Glioblastoma/genética , Proteína HMGA1a/genética , MicroRNAs/genética , Fatores de Transcrição SOXB1/genética , Animais , Linhagem Celular Tumoral , Proliferação de Células/genética , Montagem e Desmontagem da Cromatina/genética , Epigênese Genética , Regulação Neoplásica da Expressão Gênica , Glioblastoma/tratamento farmacológico , Glioblastoma/patologia , Humanos , Camundongos , Células-Tronco Neoplásicas/patologia , Regiões Promotoras Genéticas , Ensaios Antitumorais Modelo de Xenoenxerto
10.
Oncogene ; 19(18): 2212-23, 2000 Apr 27.
Artigo em Inglês | MEDLINE | ID: mdl-10822371

RESUMO

Scatter factor (SF) [aka. hepatocyte growth factor (HGF)] (designated HGF/SF) is a multifunctional cytokine that stimulates tumor cell invasion and angiogenesis. We recently reported that HGF/SF protects epithelial and carcinoma cells against cytotoxicity from DNA-damaging agents and that HGF/SF-mediated cytoprotection was associated with up-regulation of the anti-apoptotic protein Bcl-XL in cells exposed to adriamycin. We now report that in addition to blocking apoptosis, HGF/SF markedly enhances the repair of DNA strand breaks caused by adriamycin or gamma radiation. Constitutive expression of Bcl-XL in MDA-MB-453 breast cancer cells not only simulated the HGF/SF-mediated chemoradioresistance, but also enhanced the repair of DNA strand breaks. The ability of HGF/SF to induce both chemoresistance and DNA repair was inhibited by wortmannin, suggesting that these activities of HGF/SF are due, in part, to a phosphatidylinositol-3'-kinase (PI3K) dependent signaling pathway. Consistent with this finding, HGF/SF induced the phosphorylation of c-Akt (protein kinase-B), a PI3K substrate implicated in apoptosis inhibition; and an expression vector encoding a dominant negative kinase inactive Akt partially but significantly inhibited HGF/SF-mediated cell protection and DNA repair. These findings suggest that HGF/SF activates a cell survival and DNA repair pathway that involves signaling through PI3K and c-Akt and stabilization of the expression of Bcl-XL; and they implicate Bcl-XL in the DNA repair process.


Assuntos
Apoptose/efeitos dos fármacos , Reparo do DNA/efeitos dos fármacos , Fator de Crescimento de Hepatócito/farmacologia , Fosfatidilinositol 3-Quinases/metabolismo , Proteínas Serina-Treonina Quinases , Antimutagênicos/farmacologia , Antineoplásicos/farmacologia , Proteína BRCA1 , Neoplasias da Mama , Relação Dose-Resposta à Radiação , Doxorrubicina/farmacologia , Feminino , Raios gama , Humanos , Masculino , Mutagênicos/farmacologia , Neoplasias da Próstata , Proteínas Proto-Oncogênicas/metabolismo , Proteínas Proto-Oncogênicas c-akt , Proteínas Proto-Oncogênicas c-bcl-2 , Transdução de Sinais , Células Tumorais Cultivadas , Proteína bcl-X
11.
Clin Cancer Res ; 7(8): 2555-62, 2001 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-11489839

RESUMO

PURPOSE: Hepatocyte growth factor/scatter factor (HGF/SF), via its receptor c-MET, has been implicated to play a pivotal role in breast cancer development and progression. This study examined a transgene-consisting of a combination of U1snRNA, hammerhead ribozyme, and antisense, designed to inhibit c-met expression-and its impact on the migration and in vitro invasion of breast cancer cells. EXPERIMENTAL DESIGN: A hammerhead ribozyme targeting human c-MET was cloned into a modified pZeoU1EcoSpe vector and transfected into breast cancer cells MDA MB 231 and MCF-7 by electroporation. Expression of MET mRNA and protein was determined. Migration and in vitro invasiveness of transfected cells were also analyzed. RESULTS: Breast cancer cells were transfected with the ribozyme-containing plasmids. Stable transfectants manifested an almost complete loss of MET mRNA and protein, as shown by reverse transcription-PCR, Northern blotting, and Western blotting, respectively, whereas the wild-type plasmid had no effects. Met-ribozyme transfected cells exhibited reduced migration and in vitro invasiveness through extracellular matrix (Matrigel), compared with the wild-type cells and cells transfected with empty plasmid. CONCLUSIONS: These data show that targeting c-MET by way of a hammerhead ribozyme encoding antisense to c-MET is an effective approach in reducing the invasiveness of breast cancer cells.


Assuntos
Neoplasias da Mama/patologia , Proteínas Proto-Oncogênicas c-met/genética , RNA Catalítico/genética , Sequência de Bases , Neoplasias da Mama/genética , Neoplasias da Mama/terapia , Movimento Celular/genética , DNA Antissenso/genética , Regulação Neoplásica da Expressão Gênica , Humanos , Dados de Sequência Molecular , Invasividade Neoplásica/genética , Invasividade Neoplásica/prevenção & controle , Plasmídeos/genética , Proteínas Proto-Oncogênicas c-met/metabolismo , RNA Catalítico/metabolismo , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , RNA Nuclear Pequeno/genética , RNA Nuclear Pequeno/metabolismo , Fatores de Tempo , Transfecção , Células Tumorais Cultivadas
12.
Oncogene ; 34(30): 3994-4004, 2015 Jul 23.
Artigo em Inglês | MEDLINE | ID: mdl-25328136

RESUMO

Cancer stem-like cells represent poorly differentiated multipotent tumor-propagating cells that contribute disproportionately to therapeutic resistance and tumor recurrence. Transcriptional mechanisms that control the phenotypic conversion of tumor cells lacking tumor-propagating potential to tumor-propagating stem-like cells remain obscure. Here we show that the reprogramming transcription factors Oct4 and Sox2 induce glioblastoma cells to become stem-like and tumor-propagating via a mechanism involving direct DNA methyl transferase (DNMT) promoter transactivation, resulting in global DNA methylation- and DNMT-dependent downregulation of multiple microRNAs (miRNAs). We show that one such downregulated miRNA, miRNA-148a, inhibits glioblastoma cell stem-like properties and tumor-propagating potential. This study identifies a novel and targetable molecular circuit by which glioma cell stemness and tumor-propagating capacity are regulated.


Assuntos
Neoplasias Encefálicas/metabolismo , DNA (Citosina-5-)-Metiltransferases/fisiologia , Regulação Neoplásica da Expressão Gênica , Glioblastoma/metabolismo , MicroRNAs/fisiologia , Fator 3 de Transcrição de Octâmero/fisiologia , Fatores de Transcrição SOXB1/fisiologia , Neoplasias Encefálicas/patologia , Linhagem Celular Tumoral , Epigênese Genética , Glioblastoma/patologia , Humanos , Transplante de Neoplasias , Células-Tronco Neoplásicas/metabolismo , Fenótipo
13.
J Neuroimmunol ; 92(1-2): 50-9, 1998 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-9916879

RESUMO

This study examines the effects of interleukin-10 (IL-10) and combination IL-10 + IL-2 gene transfer on experimental brain tumor growth in vivo. 9L gliosarcoma cells were engineered to stably express murine IL-10 (9L-IL-10 cells) and implanted subcutaneously or to the caudate/putamen of syngeneic rats. The growth of tumors expressing IL-10 was substantially reduced compared to that of control tumors (p < 0.05). Intracranial tumors expressing IL-10 and IL-2 were established by co-implanting 9L-IL-10 cells with endothelial cells engineered to express IL-2. At 14 days post-implantation, tumors expressing IL-10 + IL-2 were 99% smaller than control-transfected tumors (p < 0.0001). This extent of anti-tumor effect could not be achieved by expression of IL-10 or IL-2 alone within tumors. Neither IL-10 nor a combination of IL-10 + IL-2 gene delivery inhibited tumor growth in severe combined immunodeficient (SCID-Beige) mice (p > 0.05). Immunohistochemical analysis revealed that IL-10 + IL-2 gene delivery markedly increased T-cell infiltration within the striatum ipsilateral to tumor cell implantation. These findings establish that IL-10 expression, particularly in combination with IL-2 expression, can have significant immune-dependent anti-tumor actions within intracranial gliomas.


Assuntos
Neoplasias Encefálicas/genética , Técnicas de Transferência de Genes , Glioma/genética , Interleucina-10/genética , Interleucina-2/genética , Animais , Formação de Anticorpos/efeitos dos fármacos , Formação de Anticorpos/fisiologia , Neoplasias Encefálicas/imunologia , Sinergismo Farmacológico , Glioma/imunologia , Glioma/patologia , Interleucina-10/farmacologia , Interleucina-2/farmacologia , Camundongos , Ratos , Células Tumorais Cultivadas
14.
J Histochem Cytochem ; 42(7): 953-6, 1994 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-8014479

RESUMO

Cell transplantation is commonly used in studies of CNS development, tumor biology, and gene therapy. Fluorescent dyes and the E. coli lacZ reporter gene are used to identify transplanted cells in host tissues. The usefulness of these methods depends on host autofluorescence and beta-galactosidase (beta-Gal) activity. Our interest in the CNS vasculature led us to examine vascular autofluorescence and beta-Gal activity in postnatal and adult rat brains. Brains were perfusion-fixed (3.7% paraformaldehyde), cryoprotected, and cryostat-sectioned (12 microns). Autofluorescent vessel profiles were quantitated in sections using rhodamine filter sets and beta-Gal-positive vessels were quantitated under bright-field after incubation of sections with X-Gal chromogenic substrate for 1-18 hr at 37 degrees C. Multifocal vessel autofluorescence appeared in postnatal Day (PND) 18 Lewis rats (0.6 +/- 0.4 vessels/field) and increased tenfold in adults (6.8 +/- 0.3/field). The numbers of beta-Gal-positive vessels in PND 18 and adult sections incubated with X-Gal for 18 hr were 21.1 +/- 1.7 and 119 +/- 17, respectively. Host beta-Gal staining was similar to that produced by implanted endothelial cells expressing the bacterial lacZ reporter gene. Reducing incubation times in X-Gal to less than 4 hr eliminated endogenous staining and retained lacZ-specific staining. The presence of vascular autofluorescence and endogenous beta-Gal activity must be considered when either fluorescence- or lacZ-dependent cell markers are used in rat brain.


Assuntos
Encéfalo/irrigação sanguínea , beta-Galactosidase/metabolismo , Animais , Transplante de Células , Endotélio Vascular/citologia , Fluorescência , Genes Reporter , Microcirculação/enzimologia , Microcirculação/crescimento & desenvolvimento , Microscopia de Fluorescência , Ratos , Ratos Endogâmicos Lew
15.
EXS ; 79: 47-64, 1997.
Artigo em Inglês | MEDLINE | ID: mdl-9002220

RESUMO

Gliomas are highly resistant to conventional therapeutic measures, requiring the development of novel treatments. Since gliomas are particularly vascular tumors, one approach involves treatments directed at inhibiting angiogenic mechanisms. Although multiple factors contribute to the ultimate vascularization of any tumor, some are especially relevant to gliomas. Early experimental work directed at inhibiting angiogenic pathways has shown promise toward achieving control of tumor growth. This article focuses on the evidence that angiogenesis and related vascular cell responses play important roles in glioma biology, and reviews those biochemical pathways known through experimentation to be involved in the vascular response to gliomas. Finally, contemporary vessel-targeted approaches that have been used to inhibit glioma growth are discussed.


Assuntos
Neoplasias Encefálicas/irrigação sanguínea , Glioma/irrigação sanguínea , Substâncias de Crescimento/fisiologia , Neovascularização Patológica/prevenção & controle , Neovascularização Patológica/fisiopatologia , Animais , Neoplasias Encefálicas/patologia , Neoplasias Encefálicas/terapia , Endopeptidases/metabolismo , Fatores de Crescimento Endotelial/fisiologia , Matriz Extracelular/fisiologia , Fator 2 de Crescimento de Fibroblastos/fisiologia , Glioma/patologia , Glioma/terapia , Fator de Crescimento de Hepatócito/fisiologia , Humanos , Linfocinas/fisiologia , Neovascularização Patológica/patologia , Ativadores de Plasminogênio/fisiologia , Inativadores de Plasminogênio/fisiologia , Fator de Crescimento Derivado de Plaquetas/fisiologia , Fator A de Crescimento do Endotélio Vascular , Fatores de Crescimento do Endotélio Vascular
16.
Science ; 254(5035): 1091, 1991 Nov 22.
Artigo em Inglês | MEDLINE | ID: mdl-17776395
17.
Brain Res ; 640(1-2): 98-104, 1994 Mar 21.
Artigo em Inglês | MEDLINE | ID: mdl-8004469

RESUMO

Blood-brain barrier (BBB) properties of endothelial cells have on impact on brain tumor behavior, diagnosis, and response to therapy. Biochemical BBB properties are expressed by endothelial cells within intracerebral (IC) gliomas but little is known regarding the expression of BBB-associated proteins within gliomas established subcutaneously (SC), a site that is frequently used in experimental glioma models. We compared the expression of two BBB proteins, glucose transporter type-1 (Glut1) and endothelial barrier antigen (EBA), in IC and SC rat 9L and F98 gliomas. The percentage of microvessels with immunohistochemically-detectable Glut1 and EBA in IC 9L tumors (31-98%) contrasted with that found in SC 9L tumors (0-3.9%) (P < 0.0001). Likewise, the percentage of immunohistochemically-positive vessels in IC F98 tumors (35-66%) differed markedly from that in SC F98 tumors (0%) (P < 0.0001). These differences were not explained by effects of tumor location on vessel density or tumor histology. These findings demonstrate that the peritumoral environment influences endothelial differentiation within glial tumors and suggest that glioma cells maintain but do not induce the expression of barrier properties in vessels that infiltrate tumor from surrounding tissue.


Assuntos
Neoplasias Encefálicas/fisiopatologia , Endotélio Vascular/fisiologia , Glioma/fisiopatologia , Neoplasias Cutâneas/fisiopatologia , Animais , Antígenos de Neoplasias/biossíntese , Antígenos de Neoplasias/imunologia , Barreira Hematoencefálica/fisiologia , Neoplasias Encefálicas/irrigação sanguínea , Neoplasias Encefálicas/patologia , Diferenciação Celular/fisiologia , Endotélio Vascular/imunologia , Endotélio Vascular/patologia , Glioma/irrigação sanguínea , Glioma/patologia , Transportador de Glucose Tipo 1 , Imuno-Histoquímica , Masculino , Proteínas de Transporte de Monossacarídeos/biossíntese , Proteínas do Tecido Nervoso/biossíntese , Proteínas do Tecido Nervoso/imunologia , Ratos , Ratos Endogâmicos F344 , Neoplasias Cutâneas/irrigação sanguínea , Neoplasias Cutâneas/patologia
18.
Brain Res ; 833(2): 173-80, 1999 Jul 03.
Artigo em Inglês | MEDLINE | ID: mdl-10375692

RESUMO

Malignant gliomas are associated with a dysfunctional blood-tumor barrier (BTB) that causes substantial morbidity. Scatter factor/hepatocyte growth factor (SF/HGF) is a multifunctional growth factor that correlates with glioma malignancy and has several biological properties that suggest a role in enhancing blood-glioma barrier permeability. In this study, we examined the effects of glioma cell SF/HGF expression on BTB permeability to horseradish peroxidase (HRP). Fischer 344 rats bearing intrastriatal 9L tumors engineered to secrete SF/HGF (9L-SF) and SF/HGF-negative control tumors (9L-neo) received intracardiac injections of HRP and were rapidly decapitated. Densitometric analysis of brain sections reacted with diaminobenzidine showed significantly greater extravascular HRP surrounding SF/HGF-secreting tumors than 9L-neo tumors of comparable size (p<0.05). HRP enzymatic activity associated with striata containing SF/HGF-expressing tumors was 1. 6-fold greater than that of striata containing control tumors (p<0. 05). Northern analysis showed that expression of vascular endothelial growth factor/vascular permeability factor (VEGF/VPF) did not differ between 9L-neo and 9L-SF tumors. These data demonstrate that SF/HGF expression by intracerebral glial tumors can enhance BTB permeability independent of changes in VEGF/VPF expression.


Assuntos
Neoplasias Encefálicas/irrigação sanguínea , Permeabilidade Capilar/fisiologia , Técnicas de Transferência de Genes , Glioma/irrigação sanguínea , Fator de Crescimento de Hepatócito/genética , Peroxidase do Rábano Silvestre/farmacocinética , Animais , Fatores de Crescimento Endotelial/genética , Regulação Neoplásica da Expressão Gênica , Fator de Crescimento de Hepatócito/metabolismo , Linfocinas/genética , Proteínas Proto-Oncogênicas c-met/genética , Ratos , Ratos Endogâmicos F344 , Células Tumorais Cultivadas , Fator A de Crescimento do Endotélio Vascular , Fatores de Crescimento do Endotélio Vascular
19.
Brain Res ; 731(1-2): 161-70, 1996 Aug 26.
Artigo em Inglês | MEDLINE | ID: mdl-8883866

RESUMO

Malignant brain neoplasms present great therapeutic challenges due to their extremely aggressive behavior and relative isolation by the blood-brain and blood-tumor barriers. Endothelial cells may be versatile platforms for delivering genes to solid tumors by virtue of their location at blood-tissue interfaces and their proliferation in response to endothelial mitogens produced by tumors. Immortalized rat brain endothelial cells that express the E. coli lacZ reporter gene and the gene for murine interleukin-2 (RBEZ-IL2) were co-inoculated with 9L glioma cells to Fisher rats to examine the effects of endothelial cell-based cytokine delivery on glioma growth in vivo. 9L glioma growth was not affected by the implantation of control RBEZ cells. The growth of subcutaneous and intracranial 9L gliomas was significantly inhibited by RBEZ-IL2 cells (P < 0.005 and P < 0.01, respectively) when compared to control transfected RBEZ cells. Rats receiving intracranial 9L glioma cells with RBEZ-IL2 cells showed increased survival (P < 0.001). Histologic and immunohistologic analysis showed enhanced activation of microglia/macrophages and CD8-positive T lymphocytes and/or natural killer cells within brain at sites of 9L inoculation with RBEZ-IL2 cells. This report establishes that immortalized endothelial cells can be used for cytokine gene delivery and to activate anti-tumor host responses to experimental gliomas within the central nervous system.


Assuntos
Técnicas de Transferência de Genes , Terapia Genética , Glioma/terapia , Interleucina-2/genética , Animais , Divisão Celular/imunologia , Transplante de Células , Endotélio Vascular/citologia , Endotélio Vascular/metabolismo , Expressão Gênica , Glioma/imunologia , Inibidores do Crescimento/imunologia , Interleucina-2/imunologia , Ratos , Ratos Endogâmicos Lew , Transfecção , Transgenes , Células Tumorais Cultivadas
20.
Brain Res ; 604(1-2): 79-85, 1993 Feb 26.
Artigo em Inglês | MEDLINE | ID: mdl-7681348

RESUMO

Angiogenesis, a process dependent upon perivascular proteolysis, is required for solid tumor growth and is inhibited by certain steroids including glucocorticoids. We examined the relationship between tumor growth and vessel density in experimental rat brain 9L glial tumors following chronic treatment with the glucocorticoid dexamethasone. Tumor growth was inhibited by intraperitoneal administration of 3 mg/kg/day dexamethasone. Maximal cross-sectional areas of post-implantation day 9 tumors were 4.6 +/- 1.0 mm2 in dexamethasone-treated animals and 17.0 +/- 3.4 mm2 in controls (P < 0.01). Microvessel density assessed by laminin immunohistochemistry was 59% lower in dexamethasone-treated tumors (P < 0.01). Plasminogen activator (PA) activity, a proteolytic enzyme related to endothelial migration and vessel growth, was 4.2 +/- 0.9 IU/micrograms protein in dexamethasone-treated tumors and 9.0 +/- 1.0 IU/micrograms protein in control tumors (P < 0.01). Exposure of cultured 9L and central nervous system microvessel endothelial cells to dexamethasone concentrations comparable to those achieved in vivo had no effect on cell growth, but reduced the PA activity of culture supernatant fractions by 78% and 99%, respectively. These findings suggest that inhibition of proteolytic steps involved in vessel growth may underlie, in part, the mechanism by which glucocorticoids decrease brain tumor growth.


Assuntos
Neoplasias Encefálicas/irrigação sanguínea , Neoplasias Encefálicas/enzimologia , Dexametasona/farmacologia , Ativadores de Plasminogênio/metabolismo , Animais , Neoplasias Encefálicas/patologia , Divisão Celular/efeitos dos fármacos , Linhagem Celular , Células Cultivadas , Endotélio Vascular/enzimologia , Masculino , Neovascularização Patológica , Ratos , Ratos Endogâmicos F344
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA