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










Base de datos
Intervalo de año de publicación
1.
Oncogene ; 35(37): 4903-13, 2016 09 15.
Artículo en Inglés | MEDLINE | ID: mdl-26898758

RESUMEN

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.


Asunto(s)
Metilación de ADN/genética , Glioblastoma/genética , Proteína HMGA1a/genética , MicroARNs/genética , Factores de Transcripción SOXB1/genética , Animales , Línea Celular Tumoral , Proliferación Celular/genética , Ensamble y Desensamble de Cromatina/genética , Epigénesis Genética , Regulación Neoplásica de la Expresión Génica , Glioblastoma/tratamiento farmacológico , Glioblastoma/patología , Humanos , Ratones , Células Madre Neoplásicas/patología , Regiones Promotoras Genéticas , Ensayos Antitumor por Modelo de Xenoinjerto
2.
J Oncol Pharm Pract ; 22(3): 517-22, 2016 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-25616657

RESUMEN

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.


Asunto(s)
Adenoma/tratamiento farmacológico , Antineoplásicos Alquilantes/administración & dosificación , Dacarbazina/análogos & derivados , Recurrencia Local de Neoplasia/tratamiento farmacológico , Neoplasias Hipofisarias/tratamiento farmacológico , Prolactinoma/tratamiento farmacológico , Adenoma/sangre , Adenoma/diagnóstico por imagen , Dacarbazina/administración & dosificación , Femenino , Humanos , Imagen por Resonancia Magnética/tendencias , Persona de Mediana Edad , Recurrencia Local de Neoplasia/sangre , Recurrencia Local de Neoplasia/diagnóstico por imagen , Neoplasias Hipofisarias/sangre , Neoplasias Hipofisarias/diagnóstico por imagen , Prolactina/sangre , Prolactinoma/sangre , Prolactinoma/diagnóstico por imagen , Retratamiento/métodos , Temozolomida , Resultado del Tratamiento
3.
Magn Reson Med ; 74(3): 599-606, 2015 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-26151840

RESUMEN

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.


Asunto(s)
Procesamiento de Imagen Asistido por Computador/métodos , Imagen por Resonancia Magnética/métodos , Algoritmos , Encéfalo/patología , Neoplasias Encefálicas/patología , Humanos , Oligodendroglioma/patología , Fantasmas de Imagen
4.
Oncogene ; 34(30): 3994-4004, 2015 Jul 23.
Artículo en Inglés | MEDLINE | ID: mdl-25328136

RESUMEN

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.


Asunto(s)
Neoplasias Encefálicas/metabolismo , ADN (Citosina-5-)-Metiltransferasas/fisiología , Regulación Neoplásica de la Expresión Génica , Glioblastoma/metabolismo , MicroARNs/fisiología , Factor 3 de Transcripción de Unión a Octámeros/fisiología , Factores de Transcripción SOXB1/fisiología , Neoplasias Encefálicas/patología , Línea Celular Tumoral , Epigénesis Genética , Glioblastoma/patología , Humanos , Trasplante de Neoplasias , Células Madre Neoplásicas/metabolismo , Fenotipo
5.
Oncogene ; 31(50): 5132-43, 2012 Dec 13.
Artículo en Inglés | MEDLINE | ID: mdl-22310282

RESUMEN

Glioblastoma multiforme (GBM) is the most frequent and aggressive primary brain tumors in adults. Uncontrolled proliferation and abnormal cell migration are two prominent spatially and temporally disassociated characteristics of GBMs. In this study, we investigated the role of the receptor tyrosine kinase EphB2 in controlling the proliferation/migration dichotomy of GBM. We studied EphB2 gain of function and loss of function in glioblastoma-derived stem-like neurospheres, whose in vivo growth pattern closely replicates human GBM. EphB2 expression stimulated GBM neurosphere cell migration and invasion, and inhibited neurosphere cell proliferation in vitro. In parallel, EphB2 silencing increased tumor cell proliferation and decreased tumor cell migration. EphB2 was found to increase tumor cell invasion in vivo using an internally controlled dual-fluorescent xenograft model. Xenografts derived from EphB2-overexpressing GBM neurospheres also showed decreased cellular proliferation. The non-receptor tyrosine kinase focal adhesion kinase (FAK) was found to be co-associated with and highly activated by EphB2 expression, and FAK activation facilitated focal adhesion formation, cytoskeleton structure change and cell migration in EphB2-expressing GBM neurosphere cells. Taken together, our findings indicate that EphB2 has pro-invasive and anti-proliferative actions in GBM stem-like neurospheres mediated, in part, by interactions between EphB2 receptors and FAK. These novel findings suggest that tumor cell invasion can be therapeutically targeted by inhibiting EphB2 signaling, and that optimal antitumor responses to EphB2 targeting may require concurrent use of anti-proliferative agents.


Asunto(s)
Neoplasias Encefálicas/metabolismo , Neoplasias Encefálicas/patología , Movimiento Celular/fisiología , Proteína-Tirosina Quinasas de Adhesión Focal/metabolismo , Glioblastoma/metabolismo , Glioblastoma/patología , Receptor EphB2/metabolismo , Animales , Neoplasias Encefálicas/genética , Adhesión Celular/genética , Adhesión Celular/fisiología , Procesos de Crecimiento Celular/genética , Procesos de Crecimiento Celular/fisiología , Línea Celular Tumoral , Movimiento Celular/genética , Femenino , Quinasa 1 de Adhesión Focal/genética , Quinasa 1 de Adhesión Focal/metabolismo , Proteína-Tirosina Quinasas de Adhesión Focal/genética , Glioblastoma/enzimología , Glioblastoma/genética , Humanos , Ratones , Ratones SCID , Invasividad Neoplásica , Receptor EphB2/genética , Transducción de Señal , Ensayos Antitumor por Modelo de Xenoinjerto
6.
Oncogene ; 30(31): 3454-67, 2011 Aug 04.
Artículo en Inglés | MEDLINE | ID: mdl-21383690

RESUMEN

It is necessary to understand mechanisms by which differentiating agents influence tumor-initiating cancer stem cells. Toward this end, we investigated the cellular and molecular responses of glioblastoma stem-like cells (GBM-SCs) to all-trans retinoic acid (RA). GBM-SCs were grown as non-adherent neurospheres in growth factor supplemented serum-free medium. RA treatment rapidly induced morphology changes, induced growth arrest at G1/G0 to S transition, decreased cyclin D1 expression and increased p27 expression. Immunofluorescence and western blot analysis indicated that RA induced the expression of lineage-specific differentiation markers Tuj1 and GFAP and reduced the expression of neural stem cell markers such as CD133, Msi-1, nestin and Sox-2. RA treatment dramatically decreased neurosphere-forming capacity, inhibited the ability of neurospheres to form colonies in soft agar and inhibited their capacity to propagate subcutaneous and intracranial xenografts. Expression microarray analysis identified ∼350 genes that were altered within 48 h of RA treatment. Affected pathways included retinoid signaling and metabolism, cell-cycle regulation, lineage determination, cell adhesion, cell-matrix interaction and cytoskeleton remodeling. Notch signaling was the most prominent of these RA-responsive pathways. Notch pathway downregulation was confirmed based on the downregulation of HES and HEY family members. Constitutive activation of Notch signaling with the Notch intracellular domain rescued GBM neurospheres from the RA-induced differentiation and stem cell depletion. Our findings identify mechanisms by which RA targets GBM-derived stem-like tumor-initiating cells and novel targets applicable to differentiation therapies for glioblastoma.


Asunto(s)
Antineoplásicos/farmacología , Neoplasias Encefálicas/patología , Glioblastoma/patología , Células Madre Neoplásicas/efectos de los fármacos , Receptores Notch/antagonistas & inhibidores , Tretinoina/farmacología , Antígeno AC133 , Animales , Antígenos CD/biosíntesis , Antineoplásicos/uso terapéutico , Neoplasias Encefálicas/genética , Diferenciación Celular/efectos de los fármacos , Línea Celular Tumoral , Ciclina D1/biosíntesis , Femenino , Perfilación de la Expresión Génica , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Proteína Ácida Fibrilar de la Glía/biosíntesis , Glioblastoma/genética , Glicoproteínas/biosíntesis , Humanos , Proteínas de Filamentos Intermediarios/biosíntesis , Ratones , Ratones Desnudos , Células Madre Neoplásicas/metabolismo , Proteínas del Tejido Nervioso/biosíntesis , Nestina , Péptidos , Antígeno Nuclear de Célula en Proliferación/biosíntesis , Proteínas de Unión al ARN/biosíntesis , Factores de Transcripción SOXB1/biosíntesis , Tretinoina/uso terapéutico , Tubulina (Proteína)/biosíntesis
7.
Oncogene ; 26(33): 4774-96, 2007 Jul 19.
Artículo en Inglés | MEDLINE | ID: mdl-17297451

RESUMEN

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.


Asunto(s)
Daño del ADN , Factor de Crecimiento de Hepatocito/metabolismo , FN-kappa B/metabolismo , Transducción de Señal/fisiología , Proteínas ras/metabolismo , Animales , Antibióticos Antineoplásicos/farmacología , Línea Celular , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Doxorrubicina/farmacología , Vectores Genéticos/genética , Factor de Crecimiento de Hepatocito/genética , Humanos , Quinasa I-kappa B/genética , Quinasa I-kappa B/metabolismo , Proteínas Quinasas Activadas por Mitógenos/genética , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Mutación , FN-kappa B/genética , Fosforilación , Unión Proteica , Proteínas Proto-Oncogénicas c-raf/genética , Proteínas Proto-Oncogénicas c-raf/metabolismo , Interferencia de ARN , Transducción de Señal/genética , Transfección , Proteínas de Unión al GTP ral/genética , Proteínas de Unión al GTP ral/metabolismo , Proteínas ras/genética
8.
Oncogene ; 26(20): 2925-38, 2007 May 03.
Artículo en Inglés | MEDLINE | ID: mdl-17099727

RESUMEN

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.


Asunto(s)
Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Factor de Crecimiento de Hepatocito/fisiología , Proteína Oncogénica v-akt/antagonistas & inhibidores , Neoplasias de la Próstata/genética , ARN Interferente Pequeño/farmacología , Doxorrubicina/farmacología , Resistencia a Antineoplásicos/genética , Perfilación de la Expresión Génica , Humanos , Masculino , FN-kappa B/genética , FN-kappa B/fisiología , Análisis de Secuencia por Matrices de Oligonucleótidos , Proteína Oncogénica v-akt/genética , Proteína Oncogénica v-akt/fisiología , Neoplasias de la Próstata/patología , Proteínas Serina-Treonina Quinasas/genética , Proteínas Serina-Treonina Quinasas/fisiología , Células Tumorales Cultivadas , Quinasas p21 Activadas
9.
Nucl Instrum Methods Phys Res A ; 548(1-2): 30-37, 2005 Aug 11.
Artículo en Inglés | MEDLINE | ID: mdl-17369874

RESUMEN

Irradiation with parallel arrays of thin, planar slices of X-ray beams (microplanar beams, or microbeams) spares normal tissue, including the central nervous system (CNS), and preferentially damages tumors. The effects are mediated, at least in part, by the tissue's microvasculature that seems to effectively repair itself in normal tissue but fails to do so in tumors. Consequently, the therapeutic index of single-fraction unidirectional microbeam irradiations has been shown to be larger than that of single-fraction unidirectional unsegmented beams in treating the intracranial rat 9L gliosarcoma tumor model (9LGS) and the subcutaneous murine mammary carcinoma EMT-6. This paper presents results demonstrating that individual microbeams, or arrays of parallel ones, can also be used for targeted, selective cell ablation in the CNS, and also to induce demyelination. The results highlight the value of the method as a powerful tool for studying the CNS through selective cell ablation, besides its potential as a treatment modality in clinical oncology.

11.
Toxicol Appl Pharmacol ; 176(1): 34-53, 2001 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-11578147

RESUMEN

The toxic metal lead is a widespread environmental health hazard that can adversely affect human health. In an effort to better understand the cellular and molecular consequences of lead exposure, we have employed cDNA microarrays to analyze the effects of acute lead exposure on large-scale gene expression patterns in immortalized rat astrocytes. Our studies identified many genes previously reported to be differentially regulated by lead exposure. Additionally, we have identified novel putative targets of lead-mediated toxicity, including members of the family of calcium/phospholipid binding annexins, the angiogenesis-inducing thrombospondins, collagens, and tRNA synthetases. We demonstrate the ability to distinguish lead-exposed samples from control or sodium samples solely on the basis of large-scale gene expression patterns using two complementary clustering methods. We have confirmed the altered expression of candidate genes and their encoded proteins by RT-PCR and Western blotting, respectively. Finally, we show that the calcium-dependent phospholipid binding protein annexin A5, initially identified as a differentially regulated gene by our microarray analysis, is directly bound and activated by nanomolar concentrations of lead. We conclude that microarray technology is an effective tool for the identification of lead-induced patterns of gene expression and molecular targets of lead.


Asunto(s)
Astrocitos/efectos de los fármacos , Astrocitos/metabolismo , Regulación de la Expresión Génica/efectos de los fármacos , Plomo/farmacología , Análisis de Secuencia por Matrices de Oligonucleótidos , Aminoacil-ARNt Sintetasas/genética , Animales , Anexinas/genética , Anexinas/metabolismo , Western Blotting , Línea Celular Transformada , Colágeno/genética , ADN Complementario/análisis , Plomo/metabolismo , Plomo/toxicidad , Ratas , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Trombospondinas/genética
12.
J Biol Chem ; 276(50): 47257-65, 2001 Dec 14.
Artículo en Inglés | MEDLINE | ID: mdl-11571297

RESUMEN

The cytokine hepatocyte growth factor/scatter factor (HGF/SF) has been found to protect a variety of epithelial and cancer cell types against cytotoxicity and apoptosis induced by DNA damage, but the specific apoptotic signaling events and the levels at which they are blocked by HGF/SF have not been identified. We found that treatment of MDA-MB-453 human breast cancer cells with adriamycin (also known as doxorubicin, a DNA topoisomerase IIalpha inhibitor) induced a series of time-dependent events, including the mitochondrial release of cytochrome c and apoptosis-inducing factor, mitochondrial membrane depolarization, activation of a set of caspases (caspase-9, -3, -7, -2, and -8), cleavage of poly(ADP-ribose) polymerase (PARP), and up-regulation of expression of the Fas ligand. All of these events were blocked by preincubation of the cells with HGF/SF. In contrast, the pan-caspase inhibitor benzyloxycarbonyl-VAD-fluoromethylketone blocked some of these events (e.g. caspase-3 activation and PARP cleavage) but did not block cytochrome c release or mitochondrial depolarization. These findings suggest that HGF/SF functions, in part, upstream of the mitochondria to block mitochondrial apoptosis signaling, prevent activation of multiple caspases, and protect breast cancer cells against apoptosis.


Asunto(s)
Apoptosis , Neoplasias de la Mama/metabolismo , Factor de Crecimiento de Hepatocito/metabolismo , Mitocondrias/metabolismo , Clorometilcetonas de Aminoácidos/farmacología , Antineoplásicos/farmacología , Factor Inductor de la Apoptosis , Caspasa 3 , Caspasas/metabolismo , Colorantes/farmacología , Inhibidores de Cisteína Proteinasa/farmacología , Grupo Citocromo c/metabolismo , Doxorrubicina/farmacología , Activación Enzimática , Inhibidores Enzimáticos/farmacología , Proteína Ligando Fas , Flavoproteínas/metabolismo , Humanos , Glicoproteínas de Membrana/metabolismo , Potenciales de la Membrana , Proteínas de la Membrana/metabolismo , Proteínas Recombinantes/metabolismo , Transducción de Señal , Sales de Tetrazolio/farmacología , Tiazoles/farmacología , Factores de Tiempo , Células Tumorales Cultivadas , Regulación hacia Arriba
13.
Clin Cancer Res ; 7(8): 2555-62, 2001 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-11489839

RESUMEN

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.


Asunto(s)
Neoplasias de la Mama/patología , Proteínas Proto-Oncogénicas c-met/genética , ARN Catalítico/genética , Secuencia de Bases , Neoplasias de la Mama/genética , Neoplasias de la Mama/terapia , Movimiento Celular/genética , ADN sin Sentido/genética , Regulación Neoplásica de la Expresión Génica , Humanos , Datos de Secuencia Molecular , Invasividad Neoplásica/genética , Invasividad Neoplásica/prevención & control , Plásmidos/genética , Proteínas Proto-Oncogénicas c-met/metabolismo , ARN Catalítico/metabolismo , ARN Mensajero/genética , ARN Mensajero/metabolismo , ARN Nuclear Pequeño/genética , ARN Nuclear Pequeño/metabolismo , Factores de Tiempo , Transfección , Células Tumorales Cultivadas
14.
J Neurochem ; 76(5): 1497-508, 2001 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-11238734

RESUMEN

Scatter factor/hepatocyte growth factor (SF/HGF) and its tyrosine kinase receptor c-met are developmentally expressed, neuroprotective, and tumorigenic within the CNS. In the present study SF/HGF is shown to induce the expression of c-met in two human glioblastoma cell lines, U-373 MG and T98G, and the signaling pathways involved in this induction are dissected. SF/HGF activated mitogen-activated protein kinase (MAPK) and inhibition of either Ras or MAPK-kinase completely inhibited SF/HGF-mediated c-met induction. Inhibition of phospholipase-C (PLC) did not affect c-met induction in either cell line. Inhibition of phosphoinositide 3-kinase (PI3-kinase) substantially reduced c-met induction by SF/HGF in T98G cells but had no effect in U-373 MG cells. Protein kinase C (PKC) inhibition reduced c-met induction in T98G cells but not in U-373 MG cells. SF/HGF induced the expression of c-fos and c-jun mRNA and increased the levels of AP-1 transcription factor in both cells lines as determined by AP-1-luciferase reporter expression. Transfection of either cell line with TAM-67, a dominant negative for the jun transactivation domain, completely inhibited AP-1 and c-met induction by SF/HGF. These results support a model of c-met induction by SF/HGF in human glioma cells that uniformly involves Ras, MAPK, and AP-1 and additionally involves PI3-kinase and PKC in some cell lines.


Asunto(s)
Factor de Crecimiento de Hepatocito/farmacología , Proteínas Proto-Oncogénicas c-met/genética , Transducción de Señal/fisiología , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Regulación Neoplásica de la Expresión Génica/fisiología , Genes fos , Glioblastoma , Humanos , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo , Proteína Quinasa C/metabolismo , Proteínas Proto-Oncogénicas c-met/metabolismo , Proteínas Recombinantes/metabolismo , Transducción de Señal/efectos de los fármacos , Acetato de Tetradecanoilforbol/farmacología , Factor de Transcripción AP-1/metabolismo , Transcripción Genética , Transfección , Células Tumorales Cultivadas
15.
Cancer Res ; 60(15): 4277-83, 2000 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-10945642

RESUMEN

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.


Asunto(s)
Apoptosis/efectos de los fármacos , Glioblastoma/patología , Factor de Crecimiento de Hepatocito/farmacología , Fosfatidilinositol 3-Quinasas/fisiología , Proteínas Serina-Treonina Quinasas , Proteínas Proto-Oncogénicas/fisiología , Transducción de Señal/efectos de los fármacos , Animales , Apoptosis/fisiología , Expresión Génica , Técnicas de Transferencia de Gen , Glioblastoma/enzimología , Gliosarcoma/enzimología , Gliosarcoma/patología , Factor de Crecimiento de Hepatocito/fisiología , Humanos , Masculino , Proteínas Tirosina Quinasas/fisiología , Proteínas Proto-Oncogénicas c-akt , Proteínas Proto-Oncogénicas c-met/antagonistas & inhibidores , Proteínas Proto-Oncogénicas c-met/genética , Proteínas Proto-Oncogénicas c-met/fisiología , ARN Catalítico/genética , Ratas , Ratas Endogámicas F344 , Proteínas Recombinantes/farmacología , Ribonucleoproteína Nuclear Pequeña U1/genética , Transducción de Señal/fisiología , Células Tumorales Cultivadas
16.
J Biol Chem ; 275(36): 27874-82, 2000 Sep 08.
Artículo en Inglés | MEDLINE | ID: mdl-10882716

RESUMEN

The mechanism(s) underlying lead neurotoxicity are not fully elucidated. cDNA expression microarray analysis identified lead-sensitive genes in immortalized human fetal astrocytes (SV-FHA). Of the represented genes expressed, vascular endothelial growth factor (VEGF) was one of the most sensitive. Lead induced VEGF mRNA 3-fold and VEGF protein approximately 2-fold with maximum mRNA induction following incubation with 10 micrometer lead acetate for 24 h. Phorbol 12-myristate 13-acetate (PMA), a potent protein kinase C (PKC) activator, increased VEGF mRNA 2-fold and PKC inhibition by GF-109203 completely blocked VEGF induction by lead. Expression of dominant-negative PKC-epsilon, but not PKC-alpha, completely inhibited VEGF mRNA induction by lead. Lead activated the transcription factor AP-1 and increased AP-1-dependent luciferase expression >2-fold. Transfection of cells with a c-jun dominant-negative effectively inhibited both AP-1 activation and VEGF mRNA induction by lead. Hypoxia-inducible factor 1 (HIF-1) activity in SV-FHAs was moderately increased by lead (86%) and PMA (96%). Pretreatment with GF-109203 completely inhibited these effects of lead and PMA. However, lead did not alter HIF-1-dependent luciferase expression and a HIF-1alpha dominant-negative had no effects on the induction of VEGF mRNA by lead. These findings indicate that lead induces VEGF expression in SV-FHAs via a PKC/AP-1-dependent and HIF-1-independent signaling pathway.


Asunto(s)
Astrocitos/metabolismo , Proteínas de Unión al ADN/metabolismo , Factores de Crecimiento Endotelial/genética , Regulación de la Expresión Génica/efectos de los fármacos , Linfocinas/genética , Proteínas Nucleares/metabolismo , Compuestos Organometálicos/toxicidad , Proteína Quinasa C/metabolismo , Factor de Transcripción AP-1/metabolismo , Transcripción Genética/efectos de los fármacos , Astrocitos/efectos de los fármacos , Células Cultivadas , Feto , Humanos , Factor 1 Inducible por Hipoxia , Subunidad alfa del Factor 1 Inducible por Hipoxia , ARN Mensajero/genética , Transducción de Señal , Acetato de Tetradecanoilforbol/farmacología , Factores de Transcripción/metabolismo , Factor A de Crecimiento Endotelial Vascular , Factores de Crecimiento Endotelial Vascular
17.
Biochem Biophys Res Commun ; 273(1): 287-93, 2000 Jun 24.
Artículo en Inglés | MEDLINE | ID: mdl-10873600

RESUMEN

Strategies that antagonize growth factor signaling are attractive candidates for the biological therapy of brain tumors. HGF/NK2 is a secreted truncated splicing variant and potential antagonist of scatter factor/hepatocyte growth factor (SF/HGF), a multifunctional cytokine involved in the malignant progression of solid tumors including glioblastoma. U87 human malignant glioma cells that express an autocrine SF/HGF stimulatory loop were transfected with the human HGF/NK2 cDNA and clonal cell lines that secrete high levels of HGF/NK2 protein (U87-NK2) were isolated. The effects of HGF/NK2 gene transfer on the U87 malignant phenotype were examined. HGF/NK2 gene transfer had no effect on 2-dimensional anchorage-dependent cell growth. In contrast, U87-NK2 cell lines were approximately 20-fold less clonogenic in soft agar and approximately 4-fold less migratory than control-transfected cell lines. Intracranial tumor xenografts derived from U87-NK2 cells grew much slower than controls. U87-NK2 tumors were approximately 50-fold smaller than controls at 21 days post-implantation and HGF/NK2 gene transfer resulted in a trend toward diminished tumorigenicity. This report shows that the predominant effect of transgenic HGF/NK2 overexpression by glioma cells that are autocrine for SF/HGF stimulation is to inhibit their malignant phenotype.


Asunto(s)
Empalme Alternativo/genética , Neoplasias Encefálicas/patología , Glioma/patología , Factor de Crecimiento de Hepatocito/antagonistas & inhibidores , Factor de Crecimiento de Hepatocito/fisiología , Agar/metabolismo , Animales , Comunicación Autocrina , Neoplasias Encefálicas/tratamiento farmacológico , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/metabolismo , Adhesión Celular , División Celular , Movimiento Celular , Transformación Celular Neoplásica/patología , Técnicas de Transferencia de Gen , Glioma/tratamiento farmacológico , Glioma/genética , Glioma/metabolismo , Factor de Crecimiento de Hepatocito/química , Factor de Crecimiento de Hepatocito/genética , Humanos , Ratones , Ratones SCID , Peso Molecular , Trasplante de Neoplasias/patología , Fenotipo , Transfección , Trasplante Heterólogo/patología , Células Tumorales Cultivadas
18.
Oncogene ; 19(18): 2212-23, 2000 Apr 27.
Artículo en Inglés | MEDLINE | ID: mdl-10822371

RESUMEN

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.


Asunto(s)
Apoptosis/efectos de los fármacos , Reparación del ADN/efectos de los fármacos , Factor de Crecimiento de Hepatocito/farmacología , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas Serina-Treonina Quinasas , Antimutagênicos/farmacología , Antineoplásicos/farmacología , Proteína BRCA1 , Neoplasias de la Mama , Relación Dosis-Respuesta en la Radiación , Doxorrubicina/farmacología , Femenino , Rayos gamma , Humanos , Masculino , Mutágenos/farmacología , Neoplasias de la Próstata , Proteínas Proto-Oncogénicas/metabolismo , Proteínas Proto-Oncogénicas c-akt , Proteínas Proto-Oncogénicas c-bcl-2 , Transducción de Señal , Células Tumorales Cultivadas , Proteína bcl-X
19.
J Neurovirol ; 5(6): 695-702, 1999 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-10602410

RESUMEN

The neurological manifestations of HIV infection may be in part due to alterations in the blood-brain barrier. These may be caused by structural changes in the barrier or may consist of subtle metabolic or biochemical disturbances in barrier function. In the CNS, the family of glucose transporter proteins plays a key role in controlling movement of glucose across cell membranes. The 55 kDa isoform of glucose transporter 1 (GLUT1) regulates import of glucose from blood to brain across the endothelial cells of the blood-brain barrier (BBB), whereas the 45 kDa form of GLUT1 predominantly regulates nonvascular glial glucose uptake. In this study, expression of 55 and 45 kDa forms of GLUT1 in different regions of the brain from 18 SIV-infected macaques was measured by quantitative immunoblot and then compared with the severity of SIV encephalitis to determine whether neurologic disease is related to altered glucose metabolism at the BBB and in brain parenchyma. An inverse relationship was found between severity of SIV encephalitis and expression of the endothelial 55 kDa isoform of GLUT1 at the BBB in cortical grey matter, caudate nucleus, and cerebellum. A similar relationship also was found for the glial 45 kDa GLUT1 isoform in cortical grey matter. In addition, a significant increase in 55 kDa GLUT1 expression was found in caudate nucleus during the early stages of infection. In the brains of macaques with moderate to severe encephalitis, 55 kDa GLUT1 expression had declined to pre-infection levels. These GLUT1 alterations at the BBB and in glial cells may reflect severe disturbances in the CNS microenvironment that contribute to CNS dysfunction.


Asunto(s)
Barrera Hematoencefálica/fisiología , Encéfalo/metabolismo , Encefalitis Viral/metabolismo , Proteínas de Transporte de Monosacáridos/metabolismo , Síndrome de Inmunodeficiencia Adquirida del Simio/metabolismo , Animales , Encéfalo/irrigación sanguínea , Núcleo Caudado/metabolismo , Núcleo Caudado/patología , Cerebelo/metabolismo , Cerebelo/patología , Corteza Cerebral/metabolismo , Corteza Cerebral/patología , Encefalitis Viral/patología , Encefalitis Viral/virología , Endotelio Vascular/metabolismo , Transportador de Glucosa de Tipo 1 , Giro del Cíngulo/metabolismo , Giro del Cíngulo/patología , Immunoblotting , Macaca nemestrina , Síndrome de Inmunodeficiencia Adquirida del Simio/patología , Síndrome de Inmunodeficiencia Adquirida del Simio/virología , Factores de Tiempo
20.
Int J Dev Neurosci ; 17(5-6): 517-30, 1999.
Artículo en Inglés | MEDLINE | ID: mdl-10571413

RESUMEN

Scatter factor/hepatocyte growth factor (SF/HGF) is a pleiotrophic cytokine that stimulates motility and invasion of several cancer cell types and induces angiogenesis. Its receptor MET is a transmembrane tyrosine kinase encoded by the C-MET proto-oncogene. To assess the potential relevance of SF/HGF in gliomas we performed functional studies in vivo and in vitro, expression analyses and correlative studies. We showed that both SF/HGF and MET are expressed in gliomas in vivo and are upregulated during transition from low grade to malignant glioma. When SF/HGF cDNA was transfected into glioma cells that expressed the MET receptor the cells formed considerably larger and more vascularized intracranial tumors in vivo than SF/HGF negative control clones. In other glioma cells, which constitutively expressed both SF/HGF and MET, we abolished SF/HGF expression by antisense ribozyme-targeting, which led to a significant decrease in tumorigenicity and tumor growth. In vitro SF/HGF strongly stimulated glioma cell motility and to a lesser degree proliferation. SF/HGF also strongly increased endothelial cell motility in vitro and extracts of tumors derived from SF/HGF-transfected glioma cells were more mitogenic for endothelial cells and more angiogenic in the rat cornea angiogenesis assay than extracts from control tumors. In a three-dimensional in vitro angiogenesis assay basic fibroblast growth factor (bFGF) was found to synergize with either SF/HGF or vascular endothelial growth factor (VEGF) in inducing endothelial capillary-like tubes, whereas neither SF/HGF nor VEGF alone or in combination were effective. Interestingly, while both VEGF and SF/HGF levels appeared to be increased in malignant gliomas compared with low grade ones, this was not the case for bFGF of which biologically relevant levels were already present in low grade gliomas. It thus seems that bFGF alone is insufficient to induce angiogenesis in gliomas but may act synergistically with either VEGF and/or SF/HGF when these become upregulated during malignant progression. In conclusion, we showed that SF/HGF may contribute to glioma progression by stimulating tumor invasiveness, proliferation and neovascularization.


Asunto(s)
Neoplasias Encefálicas/química , Glioma/química , Factor de Crecimiento de Hepatocito/análisis , Animales , Modelos Animales de Enfermedad , Humanos , Ratones , Ratones Noqueados , Neovascularización Patológica , Proto-Oncogenes Mas , Proteínas Proto-Oncogénicas c-met/análisis , Ratas
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...