Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 32
Filtrar
1.
Nature ; 459(7249): 1000-4, 2009 Jun 18.
Artigo em Inglês | MEDLINE | ID: mdl-19536265

RESUMO

T-cell acute lymphoblastic leukaemia (T-ALL) is a blood malignancy afflicting mainly children and adolescents. T-ALL patients present at diagnosis with increased white cell counts and hepatosplenomegaly, and are at an increased risk of central nervous system (CNS) relapse. For that reason, T-ALL patients usually receive cranial irradiation in addition to intensified intrathecal chemotherapy. The marked increase in survival is thought to be worth the considerable side-effects associated with this therapy. Such complications include secondary tumours, neurocognitive deficits, endocrine disorders and growth impairment. Little is known about the mechanism of leukaemic cell infiltration of the CNS, despite its clinical importance. Here we show, using T-ALL animal modelling and gene-expression profiling, that the chemokine receptor CCR7 (ref. 5) is the essential adhesion signal required for the targeting of leukaemic T-cells into the CNS. Ccr7 gene expression is controlled by the activity of the T-ALL oncogene Notch1 and is expressed in human tumours carrying Notch1-activating mutations. Silencing of either CCR7 or its chemokine ligand CCL19 (ref. 6) in an animal model of T-ALL specifically inhibits CNS infiltration. Furthermore, murine CNS-targeting by human T-ALL cells depends on their ability to express CCR7. These studies identify a single chemokine-receptor interaction as a CNS 'entry' signal, and open the way for future pharmacological targeting. Targeted inhibition of CNS involvement in T-ALL could potentially decrease the intensity of CNS-targeted therapy, thus reducing its associated short- and long-term complications.


Assuntos
Sistema Nervoso Central/metabolismo , Sistema Nervoso Central/patologia , Leucemia de Células T/metabolismo , Leucemia de Células T/patologia , Receptores CCR7/metabolismo , Transdução de Sinais , Animais , Adesão Celular , Linhagem Celular Tumoral , Quimiocina CCL19/deficiência , Quimiocina CCL19/metabolismo , Quimiocina CCL21/metabolismo , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Leucemia-Linfoma Linfoblástico de Células T Precursoras/metabolismo , Leucemia-Linfoma Linfoblástico de Células T Precursoras/patologia , Receptor Notch1/genética , Receptor Notch1/metabolismo , Receptores CCR7/deficiência
2.
Mol Cancer ; 9: 133, 2010 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-20515450

RESUMO

BACKGROUND: Glioblastoma (GBM) is the most common and malignant primary intracranial human neoplasm. GBMs are characterized by the presence of extensive areas of necrosis and hypoxia. Hypoxia and its master regulator, hypoxia inducible factor 1 (HIF-1) play a key role in glioma invasion. RESULTS: To further elucidate the functional role of HIF-1alpha in glioma cell migration in vitro and in invasion in vivo, we used a shRNA approach to knock down HIF-1alpha expression complemented with genome-wide expression profiling, performed in both normoxic and hypoxic conditions. Our data show that knock down of HIF-1alpha in glioma cells significantly impairs their migration in vitro as well as their ability to invade into the brain parenchyma in vivo. Next, we assessed the role that HIF-1alpha plays in maintaining the characteristics of cancer stem cells (CSCs). By using the tumor sphere forming assay, we demonstrate that HIF-1alpha plays a role in the survival and self-renewal potential of CSCs. Finally, expression profiling experiments in glioma cells provided detailed insight into a broad range of specific biological pathways and processes downstream of HIF-1alpha. We discuss the role of these processes in the migratory and invasive properties, as well as the stem cell biology of glioblastomas CONCLUSIONS: Our data show that knock down of HIF-1alpha in human and murine glioma cells impairs their migration in vitro and their invasion in vivo. In addition, our data suggest that HIF-1alpha plays a role in the survival and self-renewal potential of CSCs and identify genes that might further elucidate the role of HIF-1alpha in tumor migration, invasion and stem cell biology.


Assuntos
Neoplasias Encefálicas/metabolismo , Glioma/metabolismo , Subunidade alfa do Fator 1 Induzível por Hipóxia/biossíntese , Células-Tronco Neoplásicas/metabolismo , Animais , Western Blotting , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/patologia , Hipóxia Celular , Linhagem Celular Tumoral , Movimento Celular/genética , Feminino , Perfilação da Expressão Gênica , Regulação Neoplásica da Expressão Gênica , Técnicas de Silenciamento de Genes , Glioma/genética , Glioma/patologia , Humanos , Subunidade alfa do Fator 1 Induzível por Hipóxia/genética , Imuno-Histoquímica , Camundongos , Camundongos Endogâmicos C57BL , Invasividade Neoplásica/genética , Invasividade Neoplásica/patologia , Células-Tronco Neoplásicas/patologia , Análise de Sequência com Séries de Oligonucleotídeos , RNA Interferente Pequeno
3.
J Neurooncol ; 99(1): 33-40, 2010 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-20157762

RESUMO

Invasion is a hallmark of malignant gliomas and is the main reason for therapeutic failure and recurrence of the tumor. CXCR4 is a key chemokine receptor implicated in glioma cell migration whose expression is regulated by hypoxia. Here, we report that hepatocyte growth factor (HGF) upregulated CXCR4 protein expression in glioma cells. HGF pre-treatment increased migration of U87MG and LN229 glioma cells towards the CXCR4 ligand, stromal cell-derived factor-1alpha (SDF-1alpha). AMD3100, a CXCR4 inhibitor, inhibited the increased migration of HGF pre-treated LN229 glioma cells towards SDF-1alpha. Following exposure to HGF and hypoxia, both cell lines showed nuclear translocation of NF-kappaB (p65). The HGF- and hypoxia-induced nuclear translocation of NF-kappaB (p65) involved phosphorylation and degradation of IkappaB-alpha. Knock-down of NF-kappaB expression inhibited the induction of CXCR4 expression in response to HGF, but not to hypoxia. However, knock-down of NF-kappaB expression inhibited the induction of CXCR4 expression in response to hypoxia in the presence of HGF. NF-kappaB mediated migration towards SDF-1alpha in response to HGF. Knock-down of NF-kappaB expression resulted in decreased migration of HGF pre-treated glioma cells towards SDF-1alpha. Therefore, HGF upregulates CXCR4 expression via NF-kappaB and leads to enhanced migration. To our knowledge, this is the first report to show that a crosstalk mediated by NF-kappaB exists between the SDF-1alpha/CXCR4 and HGF/c-Met axes relevant to glioma cell migration. These findings imply that effective inhibition of glioma invasion should be directed against several ligand/receptor signaling pathways.


Assuntos
Movimento Celular/efeitos dos fármacos , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Glioma/metabolismo , NF-kappa B/metabolismo , Fator de Crescimento Neural/farmacologia , Receptores CXCR4/metabolismo , Benzilaminas , Hipóxia Celular/fisiologia , Linhagem Celular Tumoral , Quimiocina CXCL12/metabolismo , Ciclamos , Interações Medicamentosas , Compostos Heterocíclicos/farmacologia , Humanos , RNA Interferente Pequeno/fisiologia , Receptores CXCR4/antagonistas & inibidores , Receptores CXCR4/genética , Fatores de Tempo , Transfecção/métodos
4.
J Neurooncol ; 99(2): 195-200, 2010 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-20730556

RESUMO

Hemangioblastomas frequently develop in patients with von Hippel-Lindau (VHL) disease, an autosomal dominant genetic disorder. The tumors are characterized by a dense network of blood capillaries, often in association with cysts. Although activation of receptor tyrosine kinase (RTK) signaling, including epidermal growth factor receptor (EGFR) has been implicated in the development of malignant brain tumors such as high-grade gliomas, little is known about the role of RTK signaling in hemangioblastomas. To address this issue, we examined hemangioblastoma tumor specimens using receptor tyrosine kinase (RTK) activation profiling and immunohistochemistry. Six human hemangioblastomas were analyzed with a phospho-RTK antibody array, revealing EGFR phosphorylation in all tumors. EGFR expression was confirmed by immunohistochemistry in all tumors analyzed and downstream effector pathway activation was demonstrated by positive staining for phospho-AKT. Our findings suggest that, in primary hemangioblastomas, RTK upregulation and signaling predominantly involves EGFR, providing an attractive molecular target for therapeutic intervention.


Assuntos
Neoplasias Cerebelares/metabolismo , Receptores ErbB/metabolismo , Hemangioblastoma/metabolismo , Neoplasias da Medula Espinal/metabolismo , Adulto , Neoplasias Cerebelares/patologia , Feminino , Hemangioblastoma/patologia , Humanos , Técnicas Imunoenzimáticas , Masculino , Fosforilação , Prognóstico , Análise Serial de Proteínas , Neoplasias da Medula Espinal/patologia , Adulto Jovem
5.
J Neurooncol ; 97(2): 159-69, 2010 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-19802719

RESUMO

Adults diagnosed with Glioblastoma multiforme (GBM) are frequently faced with a 7% chance of surviving 2 years compared with pediatric patients with GBM who have a 26% survival rate. Our recent screen of possible glioma-associated antigen precursor protein (TAPP) profiles displayed from different types of pediatric brain tumors showed that pediatric patients contained a subset of the tumor antigens displayed by adult GBM patients. Adult GBM possess at least 27 tumor antigens that can potentially stimulate T cell immune responses, suggesting that these tumors are quite antigenic. In contrast, pediatric brain tumors only expressed nine tumor antigens with mRNA levels that were equivalent to those displayed by adult GBM. These tumor-associated antigens could be used as possible targets of therapeutic immunization for pediatric brain cancer patients. Children have developing immune systems that peak at puberty. An immune response mounted by these pediatric patients might account for their extended life spans, even though the pediatric brain tumors express far fewer total tumor-associated antigens. Here we present a hypothesis that pediatric brain tumor patients might be the best patients to show that immunotherapy can be used to successfully treat established cancers. We speculate that immunotherapy should include a panel of tumor antigens that might prevent the out-growth of more malignant tumor cells and thereby prevent the brain tumor relapse. Thus, pediatric brain tumor patients might provide an opportunity to prove the concept of immunoprevention.


Assuntos
Neoplasias Encefálicas/imunologia , Neoplasias Encefálicas/terapia , Glioblastoma/imunologia , Glioblastoma/terapia , Imunoterapia/métodos , Adulto , Antígenos de Neoplasias/imunologia , Criança , Humanos
6.
Am J Pathol ; 173(2): 545-60, 2008 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-18599607

RESUMO

The morphological patterns of glioma cell invasion are known as the secondary structures of Scherer. In this report, we propose a biologically based mechanism for the nonrandom formation of Scherer's secondary structures based on the differential expression of stromal cell-derived factor (SDF)-1alpha and CXCR4 at the invading edge of glioblastomas. The chemokine SDF-1alpha was highly expressed in neurons, blood vessels, subpial regions, and white matter tracts that form the basis of Scherer's secondary structures. In contrast, the SDF-1alpha receptor, CXCR4, was highly expressed in invading glioma cells organized around neurons and blood vessels, in subpial regions, and along white matter tracts. Neuronal and endothelial cells exposed to vascular endothelial growth factor up-regulated the expression of SDF-1alpha. CXCR4-positive tumor cells migrated toward a SDF-1alpha gradient in vitro, whereas inhibition of CXCR4 expression decreased their migration. Similarly, inhibition of CXCR4 decreased levels of SDF-1alpha-induced phosphorylation of FAK, AKT, and ERK1/2, suggesting CXCR4 involvement in glioma invasion signaling. These studies offer one plausible molecular basis and explanation of the formation of Scherer's structures in glioma patients.


Assuntos
Neoplasias Encefálicas/metabolismo , Encéfalo/metabolismo , Quimiocina CXCL12/metabolismo , Glioblastoma/metabolismo , Receptores CXCR4/fisiologia , Fator A de Crescimento do Endotélio Vascular/fisiologia , Adulto , Idoso , Idoso de 80 Anos ou mais , Animais , Encéfalo/patologia , Neoplasias Encefálicas/patologia , Hipóxia Celular , Linhagem Celular Tumoral , Quimiotaxia , Células Endoteliais/metabolismo , Endotélio Vascular/citologia , Feminino , Glioblastoma/patologia , Humanos , Masculino , Camundongos , Pessoa de Meia-Idade , Neurônios/metabolismo , Transdução de Sinais , Fator A de Crescimento do Endotélio Vascular/farmacologia
7.
J Cell Physiol ; 217(3): 584-9, 2008 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-18651562

RESUMO

Over the past decades considerable advances have been made in neurosurgery, radiotherapy and chemotherapy resulting in improved survival and cure rates for children with brain tumors. Here we review four of the most common subtypes of pediatric brain tumors, low-grade and high-grade astrocytomas, medulloblastomas and ependymomas, highlighting their molecular features regarding their tumor biology, and promising potential therapeutic targets that may hold promise for finding new "molecular targeted" drugs. Importantly, appropriate clinical trial design will play a critical role in the evaluation of new and novel treatment approaches for pediatric brain tumors.


Assuntos
Neoplasias Encefálicas/terapia , Astrocitoma/terapia , Criança , Ensaios Clínicos como Assunto , Ependimoma/terapia , Glioma/terapia , Humanos , Meduloblastoma/terapia
8.
Int J Radiat Oncol Biol Phys ; 71(5): 1477-84, 2008 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-18640497

RESUMO

PURPOSE: To assess the effects of noscapine, a tubulin-binding drug, in combination with radiation in a murine glioma model. METHODS AND MATERIALS: The human T98G and murine GL261 glioma cell lines treated with noscapine, radiation, or both were assayed for clonogenic survival. Mice with established GL261 hind limb tumors were treated with noscapine, radiation, or both to evaluate the effect of noscapine on radioresponse. In a separate experiment with the same treatment groups, 7 days after radiation, tumors were resected and immunostained to measure proliferation rate, apoptosis, and angiogenic activity. RESULTS: Noscapine reduced clonogenic survival without enhancement of radiosensitivity in vitro. Noscapine combined with radiation significantly increased tumor growth delay: 5, 8, 13, and 18 days for control, noscapine alone, radiation alone, and the combination treatment, respectively (p < 0.001). To assess the effect of the combination of noscapine plus radiation on the tumor vasculature, tubule formation by the murine endothelial 2H11 cells was tested. Noscapine with radiation significantly inhibited tubule formation compared with radiation alone. By immunohistochemistry, tumors treated with the combination of noscapine plus radiation showed a decrease in BrdU incorporation, an increase in apoptosis by terminal deoxynucleotidyl transferase-mediated deoxyuridine triphosphate nick end labeling, and a decrease in tumor vessel density compared with tumors treated with radiation alone. CONCLUSION: Noscapine enhanced the sensitivity of GL261 glioma tumors to radiation, resulting in a significant tumor growth delay. An antiangiogenic mechanism contributed to the effect. These findings are clinically relevant, particularly in view of the mild toxicity profile of this drug.


Assuntos
Inibidores da Angiogênese/uso terapêutico , Glioma/radioterapia , Noscapina/uso terapêutico , Tolerância a Radiação/efeitos dos fármacos , Animais , Apoptose , Bromodesoxiuridina/farmacocinética , Linhagem Celular Tumoral , Proliferação de Células , Terapia Combinada/métodos , Feminino , Glioma/irrigação sanguínea , Glioma/tratamento farmacológico , Glioma/patologia , Humanos , Marcação In Situ das Extremidades Cortadas , Camundongos , Camundongos Endogâmicos C57BL , Neovascularização Patológica/tratamento farmacológico , Ensaios Antitumorais Modelo de Xenoenxerto/métodos
9.
Biochem Pharmacol ; 73(10): 1528-36, 2007 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-17324379

RESUMO

Cell cycle progression requires precise expression and activation of several cyclins and cyclin-dependent kinases. Geldanamycin (GA) affects cell cycle progression in various kinds of cells. We analyzed GA-induced cell cycle regulation in glioblastoma cells. GA-induced G2 or M arrest in glioblastoma cells in a cell line-dependent manner. GA decreased the expression of Cdc2 and cyclin B1 in U87MG cells. And phosphorylated Cdc2 decreased along with Cdc2 in the GA-treated cells. This cell line showed G2 arrest after GA treatment. In contrast, GA failed to down-regulate these cell cycle regulators in U251MG cells. In U251MG cells, the cell cycle was arrested at M phase in addition to G2 by GA. Next, we analyzed the mechanism of the GA-induced regulation of Cdc2 and cyclin B1 in U87MG cells. Cdc2 and cyclin B1 were ubiquitinated by GA. MG132 abrogated the GA-induced decrease of Cdc2 and cyclin B1 indicating that these proteins were degraded by proteasomes. In conclusion, GA controls the stability of Cdc2 and cyclin B1 in glioblastomas cell species-dependently. Cdc2 and cyclin B1 might be responsible for the different responses of glioblastoma cell lines to GA.


Assuntos
Antibióticos Antineoplásicos/farmacologia , Benzoquinonas/farmacologia , Proteína Quinase CDC2/metabolismo , Ciclina B/metabolismo , Fase G2/efeitos dos fármacos , Glioblastoma/patologia , Lactamas Macrocíclicas/farmacologia , Proteína Quinase CDC2/genética , Ciclo Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Ciclina B/genética , Ciclina B1 , Regulação para Baixo/efeitos dos fármacos , Fase G2/fisiologia , Meia-Vida , Humanos , Complexo de Endopeptidases do Proteassoma/metabolismo , Complexo de Endopeptidases do Proteassoma/farmacologia , Células Tumorais Cultivadas , Ubiquitina/metabolismo
10.
Clin Cancer Res ; 12(15): 4730-7, 2006 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-16899624

RESUMO

PURPOSE: High-grade glioma treatment includes ionizing radiation therapy. The high invasiveness of glioma cells precludes their eradication and is responsible for the dismal prognosis. Recently, we reported the down-regulation of MHC class I (MHC-I) products in invading tumor cells in human and mouse GL261 gliomas. Here, we tested the hypothesis that whole-brain radiotherapy (WBRT) up-regulates MHC-I expression on GL261 tumors and enhances the effectiveness of immunotherapy. EXPERIMENTAL DESIGN: MHC-I molecule expression on GL261 cells was analyzed in vitro and in vivo by flow cytometry and immunohistochemistry, respectively. To test the response of established GL261 gliomas to treatment, mice with measurable (at CT imaging) brain tumors were randomly assigned to four groups receiving (a) no treatment, (b) WBRT in two fractions of 4 Gy, (c) vaccination with irradiated GL261 cells secreting granulocyte-macrophage colony-stimulating factor, or (d) WBRT and vaccination. Endpoints were tumor response and survival. RESULTS: An ionizing radiation dose of 4 Gy maximally up-regulated MHC-I molecules on GL261 cells in vitro. In vivo, WBRT induced the expression of the beta2-microglobulin light chain subunit of the MHC class I complex on glioma cells invading normal brain and increased CD4+ and CD8+ T cell infiltration. However, the survival advantage obtained with WBRT or vaccination alone was minimal. In contrast, WBRT in combination with vaccination increased long-term survival to 40% to 80%, compared with 0% to 10% in the other groups (P < 0.002). Surviving animals showed antitumor immunity by rejecting challenge tumors. CONCLUSION: Ionizing radiation can be successfully combined with peripheral vaccination for the treatment of established high-grade gliomas.


Assuntos
Neoplasias Encefálicas/terapia , Vacinas Anticâncer/uso terapêutico , Glioma/terapia , Radiação Ionizante , Animais , Neoplasias Encefálicas/diagnóstico , Neoplasias Encefálicas/imunologia , Linhagem Celular Tumoral , Terapia Combinada , Modelos Animais de Doenças , Intervalo Livre de Doença , Relação Dose-Resposta à Radiação , Feminino , Citometria de Fluxo , Glioma/diagnóstico , Glioma/imunologia , Antígenos de Histocompatibilidade Classe I/biossíntese , Antígenos de Histocompatibilidade Classe I/efeitos da radiação , Humanos , Cadeias Leves de Imunoglobulina/biossíntese , Cadeias Leves de Imunoglobulina/efeitos da radiação , Imuno-Histoquímica , Imunoterapia , Camundongos , Tomografia Computadorizada por Raios X , Transplante Heterólogo , Regulação para Cima/efeitos da radiação , Ensaios Antitumorais Modelo de Xenoenxerto , Microglobulina beta-2/biossíntese , Microglobulina beta-2/efeitos da radiação
11.
Int J Oncol ; 28(5): 1121-30, 2006 May.
Artigo em Inglês | MEDLINE | ID: mdl-16596228

RESUMO

Overexpression of hypoxia-inducible factor-1 (HIF-1) is a common feature in solid malignancies related to oxygen deficiency. Since increased HIF-1 expression correlates with advanced disease stage, increased angiogenesis and poor prognosis, HIF-1 and its signaling pathway have become targets for cancer chemotherapy. In this study, we identified noscapine to be a novel small molecule inhibitor of the HIF-1 pathway based on its structure-function relation-ships with HIF-1 pathway inhibitors belonging to the benzylisoquinoline class of plant metabolites and/or to microtubule binding agents. We demonstrate that noscapine treatment of human glioma U87MG and T98G cell lines exposed to the hypoxic mimetic agent, CoCl2, inhibits hypoxia-mediated HIF-1alpha expression and transcriptional activity as measured by decreased secretion of VEGF, a HIF-1 target gene. Inhibition of hypoxia-mediated HIF-1alpha expression was due, in part, to its ability to inhibit accumulation of HIF-1alpha in the nucleus and target it for degradation via the proteasome. One mechanism of action of microtubule binding agents is their antiangiogenic activity associated with disruption of endothelial tubule formation. We show that noscapine has similar properties in vitro. Thus, noscapine may possess novel antiangiogenic activity associated with two broad mechanisms of action: first, by decreasing HIF-1alpha expression in hypoxic tumor cells, upregulation of target genes, such as VEGF, would be decreased concomitant with its associated angiogenic activity; second, by inhibiting endothelial cells from forming blood vessels in response to VEGF stimulation, it may limit the process of neo-vascularization, correlating with antitumor activity in vivo. For more than 75 years, noscapine has traditionally been used as an oral cough suppressant with no known toxic side effects in man. Thus, the studies reported here have found a novel function for an old drug. Given its low toxicity profile, its demonstrated antitumor activity in several animal models of cancer and its potential to inhibit the HIF-1 pathway, noscapine should be considered as an antiangiogenic chemotherapy for glioma.


Assuntos
Inibidores da Angiogênese/farmacologia , Subunidade alfa do Fator 1 Induzível por Hipóxia/antagonistas & inibidores , Subunidade alfa do Fator 1 Induzível por Hipóxia/genética , Neovascularização Patológica/prevenção & controle , Noscapina/farmacologia , Neoplasias Encefálicas , Linhagem Celular Tumoral , Cobalto/farmacologia , Primers do DNA , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Glioma , Humanos , RNA Neoplásico/genética , RNA Neoplásico/isolamento & purificação
12.
Brain Pathol ; 15(4): 297-310, 2005 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-16389942

RESUMO

Glioblastoma multiforme (GBM) is characterized by exuberant angiogenesis, a key event in tumor growth and progression. The pathologic mechanisms driving this change and the biological behavior of gliomas remain unclear. One mechanism may involve cooption of native blood vessels by glioma cells inducing expression of angiopoietin-2 by endothelial cells. Subsequently, vascular apoptosis and involution leads to necrosis and hypoxia. This in turn induces angiogenesis that is associated with expression of hypoxia-inducible factor (HIF)-1alpha and vascular endothelial growth factor (VEGF) in perinecrotic pseudopalisading glioma cells. Here we review the molecular and cellular mechanisms implicated in HIF-1-dependent and HIF-1-independent glioma-associated angiogenesis. In GBMs, both tumor hypoxia and genetic alterations commonly occur and act together to induce the expression of HIF-1. The angiogenic response of the tumor to HIF-1 is mediated by HIF-1-regulated target genes leading to the upregulation of several proangiogenic factors such as VEGF and other adaptive response molecules. Understanding the roles of these regulatory processes in tumor neovascularization, tumor growth and progression, and resistance to therapy will ultimately lead to the development of improved antiangiogenic therapies for GBMs.


Assuntos
Neoplasias Encefálicas/irrigação sanguínea , Glioma/irrigação sanguínea , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Neovascularização Patológica/fisiopatologia , Fator A de Crescimento do Endotélio Vascular/metabolismo , Animais , Neoplasias Encefálicas/patologia , Glioma/patologia , Humanos
13.
Brain Pathol ; 15(4): 342-63, 2005 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-16389946

RESUMO

Much of the interest in angiogenesis and hypoxia has led to investigating diagnostic imaging methodologies and developing efficacious agents against angiogenesis in gliomas. In many ways, because of the cytostatic effects of these agents on tumor growth and tumor-associated endothelial cells, the effects of therapy are not immediately evident. Hence finding clinically applicable imaging tools and pathologic surrogate markers is an important step in translating glioma biology to therapeutics. There are a variety of strategies in the approach to experimental therapeutics that target the hypoxia-inducible factor pathway, the endogenous antiangiogenic and proangiogenic factors and their receptors, adhesion molecules, matrix proteases and cytokines, and the existing vasculature. We discuss the rationale for antiangiogenesis as a treatment strategy, the preclinical and clinical assessment of antiangiogenic interventions and finally focus on the various treatment strategies, including combining antiangiogenic drugs with radiation and chemotherapy.


Assuntos
Neoplasias Encefálicas/irrigação sanguínea , Neoplasias Encefálicas/diagnóstico por imagem , Glioma/irrigação sanguínea , Glioma/diagnóstico por imagem , Neovascularização Patológica/fisiopatologia , Algoritmos , Inibidores da Angiogênese/uso terapêutico , Animais , Neoplasias Encefálicas/tratamento farmacológico , Espectroscopia de Ressonância de Spin Eletrônica , Feminino , Glioma/tratamento farmacológico , Humanos , Imageamento por Ressonância Magnética , Espectroscopia de Ressonância Magnética , Pessoa de Meia-Idade , Radiografia
14.
Neuro Oncol ; 7(3): 225-35, 2005 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-16053697

RESUMO

Angiogenesis is a critical step required for sustained tumor growth and tumor progression. The stimulation of endothelial cells by cytokines secreted by tumor cells such as vascular endothelial growth factor (VEGF) induces their proliferation and migration. This is a prominent feature of high-grade gliomas. The secretion of VEGF is greatly upregulated under conditions of hypoxia because of the transcription factor hypoxiainducible factor (HIF)-1alpha, which controls the expression of many genes, allowing rapid adaptation of cells to their hypoxic microenvironment. Flavopiridol, a novel cyclin-dependent kinase inhibitor, has been attributed with antiangiogenic properties in some cancer cell lines by its ability to inhibit VEGF production. Here, we show that flavopiridol treatment of human U87MG and T98G glioma cell lines decreases hypoxia-mediated HIF-1alpha expression, VEGF secretion, and tumor cell migration. These in vitro results correlate with reduced vascularity of intracranial syngeneic GL261 gliomas from animals treated with flavopiridol. In addition, we show that flavopiridol downregulates HIF-1alpha expression in the presence of a proteasome inhibitor, an agent that normally results in the accumulation and overexpression of HIF-1alpha. The potential to downregulate HIF-1alpha expression with flavopiridol treatment in combination with a proteasome inhibitor makes this an extremely attractive anticancer treatment strategy for tumors with high angiogenic activity, such as gliomas.


Assuntos
Neoplasias Encefálicas/tratamento farmacológico , Flavonoides/farmacologia , Glioma/tratamento farmacológico , Piperidinas/farmacologia , Inibidores de Proteases/farmacologia , Inibidores de Proteínas Quinases/farmacologia , Fatores de Transcrição/efeitos dos fármacos , Animais , Western Blotting , Ácidos Borônicos/farmacologia , Neoplasias Encefálicas/irrigação sanguínea , Linhagem Celular Tumoral , Movimento Celular/efeitos dos fármacos , Regulação para Baixo , Feminino , Glioma/irrigação sanguínea , Humanos , Hipóxia/complicações , Subunidade alfa do Fator 1 Induzível por Hipóxia , Metaloproteinase 2 da Matriz/efeitos dos fármacos , Camundongos , Neovascularização Patológica/tratamento farmacológico , Reação em Cadeia da Polimerase , Complexo de Endopeptidases do Proteassoma/efeitos dos fármacos , Complexo de Endopeptidases do Proteassoma/metabolismo , RNA Mensageiro/análise , Transcrição Gênica/efeitos dos fármacos , Fator A de Crescimento do Endotélio Vascular/efeitos dos fármacos
15.
Brain Res Mol Brain Res ; 137(1-2): 98-103, 2005 Jun 13.
Artigo em Inglês | MEDLINE | ID: mdl-15950766

RESUMO

Polymorphisms in codon 72 of the p53 tumor suppressor gene have been associated with susceptibility to human cancer. We wished to evaluate whether variant allelic forms of the p53 protein were associated with brain tumors. In this study, we scored 135 brain tumor samples (92 adult and 43 pediatric cases consisting of 64 high-grade astrocytomas and 71 non-astrocytomas) for the P53 Arg72Pro polymorphisms. Our data show that the genotype frequencies of P53 Arg72Pro vary not only between patients with brain tumors and controls, but also between different histological subtypes of brain tumors. Specifically, we found (i) that the genotype distributions of the P53 Arg72Pro between all brain tumors and controls were statistically significant (P < 0.001) as well as their variant allele frequencies between cases and controls (P < 0.001); (ii) that there was a significant increase in the Arg/Pro heterozygous genotype among high-grade astrocytomas compared with non-astrocytomas (P = 0.002); and (iii) that there was a significant increase in the Arg/Pro heterozygous genotype among high-grade astrocytomas containing transdominant as well as recessive p53 mutations compared with controls (P = 0.002). Our results suggest a possible association between P53 Arg72Pro polymorphisms and susceptibility to brain tumors, particularly high-grade astrocytomas.


Assuntos
Astrocitoma/genética , Neoplasias Encefálicas/genética , Códon/genética , Predisposição Genética para Doença/genética , Polimorfismo Genético/genética , Proteína Supressora de Tumor p53/genética , Adolescente , Adulto , Idoso , Substituição de Aminoácidos/genética , Astrocitoma/metabolismo , Astrocitoma/fisiopatologia , Neoplasias Encefálicas/metabolismo , Neoplasias Encefálicas/fisiopatologia , Criança , Pré-Escolar , Análise Mutacional de DNA , Feminino , Frequência do Gene/genética , Testes Genéticos , Genótipo , Heterozigoto , Humanos , Lactente , Recém-Nascido , Masculino , Pessoa de Meia-Idade , Mutação/genética , Proteína Supressora de Tumor p53/metabolismo
16.
Cell Cycle ; 3(2): 230-4, 2004 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-14712094

RESUMO

The mechanism of action of many chemotherapeutic agents targets the cell cycle. Recently, we demonstrated cytotoxic and other anti-tumor effects of flavopiridol, the first synthetic cyclin dependent kinase (CDK) inhibitor to enter clinical trials, on the murine GL261 glioma cell line in vitro (Newcomb et al., Cell Cycle 2003; 2:243). Given that flavopiridol has demonstrated anti-tumor activity in several human xenograft models, we wanted to evaluate it for anti-glioma activity in vivo in our established subcutaneous and intracranial GL261 experimental tumor models. In particular, the intracranial animal model recapitulates many of the histopathological and biological features of human high-grade glioma including both necrosis with pseudopalisading and invasion of the brain adjacent to tumor. Here we tested the activity of flavopiridol against tumors formed by GL261 cells, first as subcutaneous implants, and then in the intracranial model. We demonstrate efficacy of flavopiridol as a single modality treatment in delaying tumor growth in both animal models. We hypothesize that flavopiridol treatment induced tumor growth delay by two possible mechanisms involving growth arrest combined with recruitment of tumor cells to S-phase. Based on our findings, flavopiridol should be considered as a treatment approach for patients with high-grade glioma.


Assuntos
Neoplasias Encefálicas/tratamento farmacológico , Divisão Celular/fisiologia , Flavonoides/farmacologia , Glioma/tratamento farmacológico , Piperidinas/farmacologia , Animais , Antineoplásicos/farmacologia , Antineoplásicos/uso terapêutico , Apoptose/fisiologia , Neoplasias Encefálicas/metabolismo , Morte Celular , Feminino , Flavonoides/uso terapêutico , Glioma/metabolismo , Camundongos , Modelos Animais , Piperidinas/uso terapêutico , Células Tumorais Cultivadas
17.
Cell Cycle ; 2(3): 243-50, 2003.
Artigo em Inglês | MEDLINE | ID: mdl-12734434

RESUMO

Glioblastoma (GBM) remains one of the most challenging solid cancers to treat due to its highly proliferative, angiogenic and invasive nature. The small molecule CDK inhibitor, flavopiridol, has demonstrated antitumor activity in human xenograft models and is currently in clinical trials showing efficacy in patients with advanced disease. We have developed an experimental animal model using the murine glioma GL261 cells as a novel in vivo system to screen potential therapeutic agents for GBM. Results of in vitro testing demonstrate that flavopiridol has several relevant clinical characteristics such as its ability to: 1. inhibit cell growth; 2. inhibit cell migration; 3. decrease expression of cyclin D1, CDK4 and p21; 4. induce apoptosis in cells with high levels of p27 expression; and 5. decrease the expression of the anti-apoptotic protein Bcl-2. The mechanism by which flavopiridol induces apoptosis is mitochondrial-mediated. We demonstrate by electron microscopy and immunohistochemistry that drug treatment induces mitochondrial damage that was accompanied by the release of cytochrome c into the cytosol together with the translocation of apoptosis inducing factor (AIF) into the nucleus. This finding in murine glioma cells differs from the mechanism of flavopiridolinduced cell death reported by us for human glioma cells (Alonso et al., Mol Cancer Ther 2003; 2:139) where drug treatment induced a caspase- and cytochrome c-independent pathway in the absence of detectable damage to mitochondria. In apoptotic human glioma cells only translocation of AIF into the nucleus occurred. Thus, the same drug kills different types of glioma cells by different mitochondrial-dependent pathways.


Assuntos
Antineoplásicos/farmacologia , Apoptose/efeitos dos fármacos , Células Eucarióticas/efeitos dos fármacos , Flavonoides/farmacologia , Glioblastoma/tratamento farmacológico , Mitocôndrias/efeitos dos fármacos , Piperidinas/farmacologia , Animais , Apoptose/fisiologia , Fator de Indução de Apoptose , Proteínas de Ciclo Celular/efeitos dos fármacos , Proteínas de Ciclo Celular/metabolismo , Divisão Celular/efeitos dos fármacos , Divisão Celular/fisiologia , Movimento Celular/efeitos dos fármacos , Movimento Celular/fisiologia , Grupo dos Citocromos c/metabolismo , Citotoxinas/toxicidade , Modelos Animais de Doenças , Relação Dose-Resposta a Droga , Células Eucarióticas/metabolismo , Flavoproteínas/metabolismo , Fase G2/efeitos dos fármacos , Glioblastoma/genética , Glioblastoma/metabolismo , Proteínas de Membrana/metabolismo , Camundongos , Mitocôndrias/metabolismo , Células Tumorais Cultivadas
18.
Mol Cancer Ther ; 2(2): 139-50, 2003 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-12589031

RESUMO

Flavopiridol is a synthetic flavone, which inhibits growth in vitro and in vivo of several solid malignancies such as renal, prostate, and colon cancers. It is a potent cyclin-dependent kinase inhibitor presently in clinical trials. In this study, we examined the effect of flavopiridol on a panel of glioma cell lines having different genetic profiles: five of six have codeletion of p16(INK4a) and p14(ARF); three of six have p53 mutations; and one of six shows overexpression of mouse double minute-2 (MDM2) protein. Independent of retinoblastoma and p53 tumor suppressor pathway alterations, flavopiridol induced apoptosis in all cell lines but through a caspase-independent mechanism. No cleavage products for caspase 3 or its substrate poly(ADP-ribose) polymerase or caspase 8 were detected. The pan-caspase inhibitor Z-VAD-fmk did not inhibit flavopiridol-induced apoptosis. Mitochondrial damage measured by cytochrome c release and transmission electron microscopy was not observed in drug-treated glioma cells. In contrast, flavopiridol treatment induced translocation of apoptosis-inducing factor from the mitochondria to the nucleus. The proteins cyclin D(1) and MDM2 involved in the regulation of retinoblastoma and p53 activity, respectively, were down-regulated early after flavopiridol treatment. Given that MDM2 protein can confer oncogenic properties under certain circumstances, loss of MDM2 expression in tumor cells could promote increased chemosensitivity. After drug treatment, a low Bcl-2/Bax ratio was observed, a condition that may favor apoptosis. Taken together, the data indicate that flavopiridol has activity against glioma cell lines in vitro and should be considered for clinical development in the treatment of glioblastoma multiforme.


Assuntos
Antineoplásicos/farmacologia , Apoptose/efeitos dos fármacos , Neoplasias Encefálicas/patologia , Quinases Ciclina-Dependentes/antagonistas & inibidores , Inibidores Enzimáticos/farmacologia , Flavonoides/farmacologia , Glioma/patologia , Proteínas Nucleares , Piperidinas/farmacologia , Proteína do Retinoblastoma/fisiologia , Proteína Supressora de Tumor p53/fisiologia , Fator de Indução de Apoptose , Northern Blotting , Neoplasias Encefálicas/tratamento farmacológico , Neoplasias Encefálicas/metabolismo , Caspase 8 , Caspase 9 , Caspases/metabolismo , Proteínas de Ciclo Celular/metabolismo , Divisão Celular/efeitos dos fármacos , Grupo dos Citocromos c/metabolismo , Flavoproteínas/metabolismo , Citometria de Fluxo , Glioma/tratamento farmacológico , Glioma/metabolismo , Humanos , Técnicas Imunoenzimáticas , Proteínas de Membrana/metabolismo , Microscopia Eletrônica , Proteínas Proto-Oncogênicas/genética , Proteínas Proto-Oncogênicas/metabolismo , Proteínas Proto-Oncogênicas c-mdm2 , Células Tumorais Cultivadas
19.
Brain Pathol ; 13(1): 34-7, 2003 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-12580543

RESUMO

In vivo animal models of primary brain tumors are necessary to advance knowledge related to the complex interactions between glioma cells and the adjacent brain. A cardinal feature of glioma growth, and a major reason why neurosurgical and adjunctive therapies ultimately fail in most patients is their invasive properties. We have adapted a previously described animal model developed by one of us to give better histological detail while preserving the identification of single infiltrating glioma cells. GL261 glioma cells were first transfected with the plasmid encoding green fluorescent protein (GFP) and then implanted into the brains of syngeneic C57BL/6 mice. Identification of GFP-positive tumor cells in paraffin sections of the brains of tumor-bearing animals utilized an antibody for conventional immunoperoxidase immunohistochemistry. This method is a more powerful technique compared with the prior use of frozen sections and fluorescence microscopy to identify GFP-tagged tumor cells. We find that this new method provides improved morphology and proves to be a sensitive and reliable system for detection of invading glioma cells. Using this methodology with other advanced technologies (eg, laser capture microdissection) holds out the promise of helping to elucidate the molecular mechanisms of glioma cell infiltration and invasion into the surrounding brain.


Assuntos
Glioma/patologia , Imuno-Histoquímica , Proteínas Luminescentes/metabolismo , Animais , Neoplasias Encefálicas/metabolismo , Neoplasias Encefálicas/patologia , Glioma/metabolismo , Proteínas de Fluorescência Verde , Imuno-Histoquímica/métodos , Camundongos , Camundongos Endogâmicos C57BL , Microscopia de Fluorescência , Invasividade Neoplásica , Transplante de Neoplasias , Plasmídeos , Transfecção , Células Tumorais Cultivadas
20.
Hum Pathol ; 34(1): 48-53, 2003 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-12605366

RESUMO

Numerous studies examining the prognostic significance of p27KIP1 expression in human cancer have shown that decreased expression often is an independent prognostic factor associated with worse survival. However, the prognostic value of p27KIP1 expression in gliomas is less well established. To further address this issue, we evaluated the relationship between p27KIP1 protein expression in a series of 50 astrocytomas with clinicopathologic parameters including age, tumor grade, MIB-1 proliferation index, and patient survival using both Western blot analysis and immunohistochemistry. The level of p27KIP1 protein expression in 9 nonneoplastic brain tissue specimens served as a control. Sixteen high-grade astrocytomas were analyzed by Western blot, and 26 high-grade astrocytomas were analyzed by immunohistochemistry for levels of p27KIP1 protein expression. Regardless of the technique used to measure p27KIP1, approximately 50% of the high-grade tumors were low expressors and the other 50% were high expressors. Thus, expression of p27KIP1 was independent of tumor grade. Loss of p27KIP1 expression is often associated with an increase in proliferative activity. We measured the rate of tumor cell proliferation using MIB-1 immunostaining in 16 high-grade astrocytomas to determine whether there was an inverse correlation between p27KIP1 expression and proliferation. No correlation between p27KIP1 expression and MIB-1 labeling index or patient survival was found. Using immunohistochemistry, we noted that the staining pattern of p27KIP1 in glioblastomas was mainly in the pseudopalisading cells that outline areas of necrosis. Because p27KIP1 can be up-regulated by hypoxia, this staining pattern would be consistent with our observation that hypoxia-inducible factor 1alpha is expressed primarily in pseudopalisading tumor cells around necrotic zones. It has been shown that a high level of p27KIP1 prevents apoptosis in hypoxic cells. Thus, maintenance of high levels of p27KIP1 in gliomas could result from the hypoxic microenvironment present within the tumor. No correlation was found between p27KIP1 expression and any of the clinicopathologic parameters tested, including patient age and tumor grade, the 2 strongest predictors of survival among glioma patients.


Assuntos
Neoplasias Encefálicas/metabolismo , Proteínas de Ciclo Celular/metabolismo , Glioma/metabolismo , Proteínas Supressoras de Tumor/metabolismo , Adolescente , Adulto , Idoso , Idoso de 80 Anos ou mais , Western Blotting , Neoplasias Encefálicas/mortalidade , Neoplasias Encefálicas/patologia , Divisão Celular , Criança , Pré-Escolar , Inibidor de Quinase Dependente de Ciclina p27 , Feminino , Glioma/mortalidade , Glioma/patologia , Humanos , Técnicas Imunoenzimáticas , Lactente , Antígeno Ki-67/metabolismo , Masculino , Pessoa de Meia-Idade , Estadiamento de Neoplasias , Prognóstico , Taxa de Sobrevida
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA