Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 39
Filtrar
Mais filtros

Bases de dados
Tipo de documento
Intervalo de ano de publicação
1.
Cancer Sci ; 106(2): 160-70, 2015 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-25458015

RESUMO

Malignant gliomas can be counted to the most devastating tumors in humans. Novel therapies do not achieve significant prolonged survival rates. The cancer cells have an impact on the surrounding vital tissue and form tumor zones, which make up the tumor microenvironment. We investigated the effects of sunitinib, a small molecule multitargeted receptor tyrosine kinase inhibitor, on constituents of the tumor microenvironment such as gliomas, astrocytes, endothelial cells, and neurons. Sunitinib has a known anti-angiogenic effect. We found that sunitinib normalizes the aberrant tumor-derived vasculature and reduces tumor vessel pathologies (i.e. auto-loops). Sunitinib has only minor effects on the normal, physiological, non-proliferating vasculature. We found that neurons and astrocytes are protected by sunitinib against glutamate-induced cell death, whereas sunitinib acts as a toxin towards proliferating endothelial cells and tumor vessels. Moreover, sunitinib is effective in inducing glioma cell death. We determined the underlying pathways by which sunitinib operates as a toxin on gliomas and found vascular endothelial growth factor receptor 2 (VEGFR2, KDR/Flk1) as the main target to execute gliomatoxicity. The apoptosis-inducing effect of sunitinib can be mimicked by inhibition of VEGFR2. Knockdown of VEGFR2 can, in part, foster the resistance of glioma cells to receptor tyrosine kinase inhibitors. Furthermore, sunitinib alleviates tumor-induced neurodegeneration. Hence, we tested whether temozolomide treatment could be potentiated by sunitinib application. Here we show that sunitinib can amplify the effects of temozolomide in glioma cells. Thus, our data indicate that combined treatment with temozolomide does not abrogate the effects of sunitinib. In conclusion, we found that sunitinib acts as a gliomatoxic agent and at the same time carries out neuroprotective effects, reducing tumor-induced neurodegeneration. Thus, this report uncovered sunitinib's actions on the brain tumor microenvironment, revealing novel aspects for adjuvant approaches and new clinical assessment criteria when applied to brain tumor patients.


Assuntos
Antineoplásicos/farmacologia , Neoplasias Encefálicas/tratamento farmacológico , Glioma/tratamento farmacológico , Indóis/farmacologia , Fármacos Neuroprotetores/farmacologia , Pirróis/farmacologia , Microambiente Tumoral/efeitos dos fármacos , Inibidores da Angiogênese/farmacologia , Animais , Apoptose/efeitos dos fármacos , Astrócitos/efeitos dos fármacos , Astrócitos/metabolismo , Neoplasias Encefálicas/metabolismo , Morte Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Progressão da Doença , Células Endoteliais/efeitos dos fármacos , Células Endoteliais/metabolismo , Glioma/metabolismo , Humanos , Camundongos , Neovascularização Patológica/tratamento farmacológico , Neovascularização Patológica/metabolismo , Doenças Neurodegenerativas/tratamento farmacológico , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Ratos , Roedores , Sunitinibe , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/metabolismo
2.
J Cell Sci ; 123(Pt 20): 3535-46, 2010 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-20841379

RESUMO

In N1E-115 cells, neurite retraction induced by neurite remodelling factors such as lysophosphatidic acid, sphingosine 1-phosphate and semaphorin 3A require the activity of phosphatidylinositol 4-phosphate 5-kinases (PIP5Ks). PIP5Ks synthesise the phosphoinositide lipid second messenger phosphatidylinositol(4,5)bisphosphate [PtdIns(4,5)P2], and overexpression of active PIP5K is sufficient to induce neurite retraction in both N1E-115 cells and cerebellar granule neurones. However, how PIP5Ks are regulated or how they induce neurite retraction is not well defined. Here, we show that neurite retraction induced by PIP5Kß is dependent on its interaction with the low molecular weight G protein Rac. We identified the interaction site between PIP5Kß and Rac1 and generated a point mutant of PIP5Kß that no longer interacts with endogenous Rac. Using this mutant, we show that Rac controls the plasma membrane localisation of PIP5Kß and thereby the localised synthesis of PtdIns(4,5)P2 required to induce neurite retraction. Mutation of this residue in other PIP5K isoforms also attenuates their ability to induce neurite retraction and to localise at the membrane. To clarify how increased levels of PtdIns(4,5)P2 induce neurite retraction, we show that mutants of vinculin that are unable to interact with PtdIns(4,5)P2, attenuate PIP5K- and LPA-induced neurite retraction. Our findings support a role for PtdIns(4,5)P2 synthesis in the regulation of vinculin localisation at focal complexes and ultimately in the regulation of neurite dynamics.


Assuntos
Neuritos/metabolismo , Fosfatidilinositol 4,5-Difosfato/metabolismo , Fosfotransferases (Aceptor do Grupo Álcool)/metabolismo , Vinculina/metabolismo , Proteínas rac1 de Ligação ao GTP/metabolismo , Animais , Linhagem Celular Tumoral , Células Cultivadas , Immunoblotting , Imunoprecipitação , Camundongos , Microscopia Confocal , Proteínas rac1 de Ligação ao GTP/genética
3.
Nat Neurosci ; 6(6): 572-8, 2003 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-12730698

RESUMO

Outgrowth of axons in the central nervous system is governed by specific molecular cues. Molecules detected so far act as ligands that bind to specific receptors. Here, we report a new membrane-associated lipid phosphate phosphatase that we have named plasticity-related gene 1 (PRG-1), which facilitates axonal outgrowth during development and regenerative sprouting. PRG-1 is specifically expressed in neurons and is located in the membranes of outgrowing axons. There, it acts as an ecto-enzyme and attenuates phospholipid-induced axon collapse in neurons and facilitates outgrowth in the hippocampus. Thus, we propose a novel mechanism by which axons are able to control phospholipid-mediated signaling and overcome the growth-inhibiting, phospholipid-rich environment of the extracellular space.


Assuntos
Diferenciação Celular/fisiologia , Cones de Crescimento/enzimologia , Regeneração Nervosa/fisiologia , Plasticidade Neuronal/fisiologia , Fosfolipídeos/metabolismo , Monoéster Fosfórico Hidrolases/isolamento & purificação , Sequência de Aminoácidos/genética , Animais , Animais Recém-Nascidos , Sequência de Bases/genética , Proteínas de Ligação a Calmodulina , DNA Complementar/análise , DNA Complementar/genética , Espaço Extracelular/metabolismo , Feminino , Feto , Cones de Crescimento/ultraestrutura , Hipocampo/embriologia , Hipocampo/enzimologia , Hipocampo/crescimento & desenvolvimento , Lipídeos de Membrana/metabolismo , Camundongos , Dados de Sequência Molecular , Técnicas de Cultura de Órgãos , Monoéster Fosfórico Hidrolases/genética , Gravidez , RNA Mensageiro/metabolismo , Ratos , Ratos Sprague-Dawley , Células Tumorais Cultivadas , Regulação para Cima/genética
4.
Free Radic Biol Med ; 43(2): 191-201, 2007 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-17603929

RESUMO

Glutathione peroxidase-4 (GPx4) is a multifunctional selenoprotein expressed as mitochondrial, cytosolic, or nuclear isoforms. As a catalytically active enzyme it has been implicated in antioxidative defense, but during sperm development it functions as a structural protein. GPx4 null mice die in utero at midgestation and knockdown of GPx4 during embryogenesis disturbs brain development. To explore the cerebral function of GPx4 we profiled cell-specific enzyme expression at various stages of perinatal brain maturation and investigated its regulation following brain injury by immunohistochemistry, in situ hybridization, and quantitative RT-PCR. Large amounts of GPx4 mRNA were detected in all neuronal layers during perinatal brain development but expression became restricted during postnatal maturation. In adult brain mitochondrial and cytosolic GPx4 isoforms were detected in neurons of cerebral cortex, hippocampus, and cerebellum whereas glial cells were devoid of GPx4. Following selective brain injury expression of the enzyme was upregulated in reactive astrocytes of lesioned areas and deafferented regions but not in neurons. Selective knockdown of GPx4 by small interfering RNA induced depletion of phosphatidylinositol-(4,5)-bisphosphate in the neuronal plasma membrane and subsequently apoptosis as indicated by caspase-3 activation. We hypothesize that astrocytic upregulation of GPx4 in response to injury is part of a protective cascade counteracting further cell damage.


Assuntos
Apoptose/fisiologia , Astrócitos/enzimologia , Lesões Encefálicas/enzimologia , Encéfalo/enzimologia , Glutationa Peroxidase/metabolismo , Neurônios/fisiologia , Animais , Encéfalo/crescimento & desenvolvimento , Modelos Animais de Doenças , Indução Enzimática , Regulação Enzimológica da Expressão Gênica , Glutationa Peroxidase/genética , Imuno-Histoquímica , Masculino , Neurônios/citologia , Neurônios/enzimologia , Fosfolipídeo Hidroperóxido Glutationa Peroxidase , Ratos , Ratos Sprague-Dawley , Reação em Cadeia da Polimerase Via Transcriptase Reversa
5.
Ann N Y Acad Sci ; 1096: 179-83, 2007 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-17405929

RESUMO

Oxidative brain damage, such as excitotoxicity and stroke, leads to primary neuronal destruction. The primary damage is further potentiated by macrophages and microglial cells, which are attracted and invade into the zone of damage resulting in secondary neuronal death. Since the essential trace element selenium has anti-inflammatory properties, we analyzed the effects of selenium on these inflammatory cells. Here, we show that the essential trace element selenium abrogates the stress-induced migration of microglial cells. Thus, the antimigratory effects of selenium may attenuate the secondary cell death cascade by preventing microglial invasion.


Assuntos
Regulação da Expressão Gênica , Macrófagos/efeitos dos fármacos , Microglia/efeitos dos fármacos , Microglia/patologia , Selênio/farmacologia , Selenito de Sódio/farmacologia , Animais , Anti-Inflamatórios/farmacologia , Antígeno CD11a/biossíntese , Linhagem Celular , Movimento Celular , Separação Celular , Peróxido de Hidrogênio/farmacologia , Inflamação , Camundongos , Microglia/metabolismo , Neurônios/metabolismo
6.
Mol Cancer Ther ; 5(5): 1248-55, 2006 May.
Artigo em Inglês | MEDLINE | ID: mdl-16731757

RESUMO

Inhibitors of histone deacetylases are promising compounds for the treatment of cancer but have not been systematically explored in malignant brain tumors. Here, we characterize the benzamide MS-275, a class I histone deacetylase inhibitor, as potent drug for experimental therapy of glioblastomas. Treatment of four glioma cell lines (U87MG, C6, F98, and SMA-560) with MS-275 significantly reduced cell growth in a concentration-dependent manner (IC(90), 3.75 micromol/L). Its antiproliferative effect was corroborated using a bromodeoxyuridine proliferation assay and was mediated by G(0)-G(1) cell cycle arrest (i.e., up-regulation of p21/WAF) and apoptotic cell death. Implantation of enhanced green fluorescent protein-transfected F98 glioma cells into slice cultures of rat brain confirmed the cytostatic effect of MS-275 without neurotoxic damage to the organotypic neuronal environment in a dose escalation up to 20 micromol/L. A single intratumoral injection of MS-275 7 days after orthotopic implantation of glioma cells in syngeneic rats confirmed the chemotherapeutic efficacy of MS-275 in vivo. Furthermore, its propensity to pass the blood-brain barrier and to increase the protein level of acetylated histone H3 in brain tissue identifies MS-275 as a promising candidate drug in the treatment of malignant gliomas.


Assuntos
Antineoplásicos/uso terapêutico , Benzamidas/uso terapêutico , Glioma/tratamento farmacológico , Inibidores de Histona Desacetilases , Piridinas/uso terapêutico , Animais , Antineoplásicos/farmacocinética , Antineoplásicos/toxicidade , Benzamidas/farmacocinética , Benzamidas/toxicidade , Barreira Hematoencefálica/efeitos dos fármacos , Barreira Hematoencefálica/metabolismo , Encéfalo/efeitos dos fármacos , Encéfalo/metabolismo , Moléculas de Adesão Celular Neuronais/metabolismo , Dacarbazina/análogos & derivados , Dacarbazina/metabolismo , Dacarbazina/farmacologia , Modelos Animais de Doenças , Glioma/metabolismo , Glioma/patologia , Hipocampo/citologia , Hipocampo/metabolismo , Histona Desacetilases/metabolismo , Humanos , Piridinas/farmacocinética , Piridinas/toxicidade , Ratos , Ratos Endogâmicos F344 , Ratos Wistar , Temozolomida , Células Tumorais Cultivadas
7.
Curr Neuropharmacol ; 14(1): 48-54, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-26521944

RESUMO

Primary tumors of the brain account for 2 % of all cancers with malignant gliomas taking the lion's share with 70 %. Malignant gliomas (high grade gliomas WHO° III and °IV) belong to one of the most threatening tumor entities known with their disappointingly short median survival time of just 14 months despite maximum therapy according to current gold standards. Malignant gliomas secrete various factors, through which they adapt and manipulate the tumor microenvironment to their advantage. Epigenetic mechanisms operate on the tumor microenvironment by de- and methylation processes and imbalances between the histone deacetylases (HDAC) and histone acetylases (HAT). Many compounds have been discovered modulating epigenetically controlled signals. Recent studies indicate that xCT (system xc-, SLC7a11) and CD44 (H-CAM,ECM-III, HUTCH-1) functions as a bridge between these epigenetic regulatory mechanisms and the malignant glioma progression. The question that ensues is the extent to which therapeutic intervention on these signaling pathways would exert influence on the treatment of malignant gliomas as well as the extent to which manipulation of HDAC activity can sensitize tumor cells for chemotherapeutics through 'epigenetic priming'. Considering the current stagnation in the development of therapeutic options the need for new strategies in the treatment of gliomas has never been so urgent. Here, the possibility of pharmacological intervention on tumor-associated genes by epigenetic priming opens a novel path in combating primary brain tumors.


Assuntos
Neoplasias Encefálicas/genética , Neoplasias Encefálicas/metabolismo , Epigênese Genética/genética , Histona Desacetilases/genética , Histona Desacetilases/metabolismo , Animais , Antineoplásicos/farmacologia , Antineoplásicos/uso terapêutico , Neoplasias Encefálicas/tratamento farmacológico , Metilação de DNA/efeitos dos fármacos , Metilação de DNA/genética , Epigênese Genética/efeitos dos fármacos , Humanos , Microambiente Tumoral/efeitos dos fármacos , Microambiente Tumoral/genética
8.
Oncotarget ; 7(46): 74630-74647, 2016 11 15.
Artigo em Inglês | MEDLINE | ID: mdl-27612422

RESUMO

The glutamate exchanger xCT (SLC7a11) is causally linked with the malignancy grade of brain tumors and represents a key player in glutamate, cystine and glutathione metabolism. Although blocking xCT is not cytotoxic for brain tumors, xCT inhibition disrupts the neurodegenerative and microenvironment-toxifying activity of gliomas. Here, we report on the use of various xCT inhibitors as single modal drugs and in combination with the autophagy-inducing standard chemotherapeutic agent temozolomide (Temodal/Temcad®, TMZ). xCT overexpressing cells (xCTOE) are more resistant to the FDA and EMA approved drug sulfasalazine (Azulfidine/Salazopyrin/Sulazine®, SAS) and RNAi-mediated xCT knock down (xCTKD) in gliomas increases the susceptibility towards SAS in rodent gliomas. In human gliomas, challenged xCT expression had no impact on SAS-induced cytotoxicity. Noteworthy, other xCT inhibitors such as erastin and sorafenib showed enhanced efficacy on xCTKD gliomas. In contrast, cytotoxic action of TMZ operates independently from xCT expression levels on rodent gliomas. Human glioma cells with silenced xCT expression display higher vulnerability towards TMZ alone as well as towards combined TMZ and SAS. Hence, we tested the partial xCT blockers and ferroptosis inducing agents erastin and sorafenib (Nexavar®, FDA and EMA-approved drug for lung cancer). Noteworthy, xCTOE gliomas withstand erastin and sorafenib-induced cell death in a concentration-dependent manner, whereas siRNA-mediated xCT knock down increased susceptibility towards erastin and sorafenib. TMZ efficacy can be potentiated when combined with erastin, however not by sorafenib. Moreover, gliomas with high xCT expression are more vulnerable towards combinatorial treatment with erastin-temozolomide. These results disclose that ferroptosis inducers are valid compounds for potentiating the frontline therapeutic agent temozolomide in a multitoxic approach.


Assuntos
Sistema y+ de Transporte de Aminoácidos/metabolismo , Antineoplásicos Alquilantes/farmacologia , Dacarbazina/análogos & derivados , Sistema y+ de Transporte de Aminoácidos/genética , Animais , Apoptose/efeitos dos fármacos , Apoptose/genética , Astrócitos/metabolismo , Autofagia/efeitos dos fármacos , Autofagia/genética , Pontos de Checagem do Ciclo Celular/efeitos dos fármacos , Pontos de Checagem do Ciclo Celular/genética , Linhagem Celular Tumoral , Sobrevivência Celular/genética , Dacarbazina/farmacologia , Resistencia a Medicamentos Antineoplásicos/genética , Sinergismo Farmacológico , Expressão Gênica , Técnicas de Silenciamento de Genes , Glioma/metabolismo , Humanos , Camundongos , Niacinamida/análogos & derivados , Niacinamida/farmacologia , Compostos de Fenilureia/farmacologia , Piperazinas/farmacologia , Células Piramidais/efeitos dos fármacos , Células Piramidais/metabolismo , Ratos , Sorafenibe , Temozolomida
9.
Anticancer Res ; 36(3): 899-905, 2016 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-26976976

RESUMO

BACKGROUND/AIM: The combination of radiotherapy, temozolomide and valproic acid (VPA) has shown some promise in retrospective analyses of patients with glioblastoma, although their mechanisms of action remain unknown. MATERIALS AND METHODS: We investigated the in vitro and in vivo effects of pretreating glioma cells with temozolomide and VPA as an immunization strategy to boost an adaptive immune response in a syngeneic mouse model. RESULTS: Temozolomide and VPA induced autophagy in GL261 glioma cells, and caused tumor antigen-specific T-cells to become activated effector T-cells. Mice with a pre-existing glioma showed no improvement in clinical outcome when immunized with temozolomide- and VPA-treated glioma cells. CONCLUSION: Although temozolomide and VPA treatment of glioma cells can boost the adaptive immune response, in the context of a vaccine therapy, additional factors are necessary to eradicate the tumor and improve survival.


Assuntos
Imunidade Adaptativa/efeitos dos fármacos , Neoplasias Encefálicas/tratamento farmacológico , Dacarbazina/análogos & derivados , Glioblastoma/tratamento farmacológico , Ácido Valproico/administração & dosagem , Animais , Protocolos de Quimioterapia Combinada Antineoplásica/administração & dosagem , Protocolos de Quimioterapia Combinada Antineoplásica/farmacologia , Autofagia/efeitos dos fármacos , Neoplasias Encefálicas/imunologia , Neoplasias Encefálicas/mortalidade , Vacinas Anticâncer/administração & dosagem , Vacinas Anticâncer/farmacologia , Linhagem Celular Tumoral , Dacarbazina/administração & dosagem , Dacarbazina/farmacologia , Glioblastoma/imunologia , Glioblastoma/mortalidade , Camundongos , Estudos Retrospectivos , Análise de Sobrevida , Temozolomida , Resultado do Tratamento , Ácido Valproico/farmacologia , Ensaios Antitumorais Modelo de Xenoenxerto
10.
Sci Rep ; 6: 19799, 2016 Feb 02.
Artigo em Inglês | MEDLINE | ID: mdl-26831010

RESUMO

Brain tumors are fast proliferating and destructive within the brain microenvironment. Effective chemotherapeutic strategies are currently lacking which combat this deadly disease curatively. The glioma-specific chloride ion channel represents a specific target for therapy. Chlorotoxin (CTX), a peptide derived from scorpion venom, has been shown to be specific and efficacious in blocking glioma Cl(-) channel activity. Here, we report on two new derivatives (termed CA4 and CTX-23) designed and generated on the basis of the peptide sequence alignments of CTX and BmKCT. The novel peptides CA4 and CTX-23 are both effective in reducing glioma cell proliferation. In addition, CTX, CA4 and CTX-23 impact on cell migration and spheroid migration. These effects are accompanied by diminished cell extensions and increased nuclear sizes. Furthermore, we found that CA4 and CTX-23 are selective with low toxicity against primary neurons and astrocytes. In the ex vivo VOGiM, which maintain the entire brain tumor microenvironment, both CTX and CA4 display anti-tumor activity and reduce tumor volume. Hence, CTX and CA4 reveal anti-angiogenic properties with endothelial and angiogenic hotspots disrupting activities. These data report on the identification of two novel CTX derivatives with multiple anti-glioma properties including anti-angiogenesis.


Assuntos
Inibidores da Angiogênese/farmacologia , Antineoplásicos/farmacologia , Neoplasias Encefálicas , Proliferação de Células/efeitos dos fármacos , Glioma , Neovascularização Patológica , Venenos de Escorpião/farmacologia , Inibidores da Angiogênese/química , Animais , Antineoplásicos/química , Neoplasias Encefálicas/irrigação sanguínea , Neoplasias Encefálicas/tratamento farmacológico , Neoplasias Encefálicas/metabolismo , Neoplasias Encefálicas/patologia , Linhagem Celular Tumoral , Glioma/irrigação sanguínea , Glioma/tratamento farmacológico , Glioma/metabolismo , Glioma/patologia , Humanos , Neovascularização Patológica/tratamento farmacológico , Neovascularização Patológica/metabolismo , Neovascularização Patológica/patologia , Ratos , Venenos de Escorpião/química
11.
Oncotarget ; 7(2): 1838-53, 2016 Jan 12.
Artigo em Inglês | MEDLINE | ID: mdl-26673818

RESUMO

Primary brain tumors are hallmarked for their destructive activity on the microenvironment and vasculature. However, solely few experimental techniques exist to access the tumor microenvironment under anatomical intact conditions with remaining cellular and extracellular composition. Here, we detail an ex vivo vascular glioma impact method (VOGIM) to investigate the influence of gliomas and chemotherapeutics on the tumor microenvironment and angiogenesis under conditions that closely resemble the in vivo situation. We generated organotypic brain slice cultures from rats and transgenic mice and implanted glioma cells expressing fluorescent reporter proteins. In the VOGIM, tumor-induced vessels presented the whole range of vascular pathologies and tumor zones as found in human primary brain tumor specimens. In contrast, non-transformed cells such as primary astrocytes do not alter the vessel architecture. Vascular characteristics with vessel branching, junctions and vessel meshes are quantitatively assessable as well as the peritumoral zone. In particular, the VOGIM resembles the brain tumor microenvironment with alterations of neurons, microglia and cell survival. Hence, this method allows live cell monitoring of virtually any fluorescence-reporter expressing cell. We further analyzed the vasculature and microglia under the influence of tumor cells and chemotherapeutics such as Temozolamide (Temodal/Temcad®). Noteworthy, temozolomide normalized vasculare junctions and branches as well as microglial distribution in tumor-implanted brains. Moreover, VOGIM can be facilitated for implementing the 3Rs in experimentations. In summary, the VOGIM represents a versatile and robust technique which allows the assessment of the brain tumor microenvironment with parameters such as angiogenesis, neuronal cell death and microglial activity at the morphological and quantitative level.


Assuntos
Neoplasias Encefálicas/irrigação sanguínea , Glioma/irrigação sanguínea , Microglia/metabolismo , Neovascularização Patológica/diagnóstico por imagem , Técnicas de Cultura de Órgãos/métodos , Animais , Antineoplásicos Alquilantes/farmacologia , Neoplasias Encefálicas/patologia , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Proliferação de Células/genética , Dacarbazina/análogos & derivados , Dacarbazina/farmacologia , Glioma/patologia , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Humanos , Camundongos Transgênicos , Microscopia Confocal , Microscopia de Fluorescência , Neovascularização Patológica/genética , Neovascularização Patológica/metabolismo , Ratos Wistar , Reprodutibilidade dos Testes , Temozolomida , Microambiente Tumoral/efeitos dos fármacos , Microambiente Tumoral/genética
12.
Aging (Albany NY) ; 8(10): 2463-2487, 2016 10 15.
Artigo em Inglês | MEDLINE | ID: mdl-27744421

RESUMO

The Plasticity Related Gene family covers five, brain-specific, transmembrane proteins (PRG1-5, also termed LPPR1-5) that operate in neuronal plasticity during development, aging and brain trauma. Here we investigated the role of the PRG family on axonal and filopodia outgrowth. Comparative analysis revealed the strongest outgrowth induced by PRG3 (LPPR1). During development, PRG3 is ubiquitously located at the tip of neuronal processes and at the plasma membrane and declines with age. In utero electroporation of PRG3 induced dendritic protrusions and accelerated spine formations in cortical pyramidal neurons. The neurite growth promoting activity of PRG3 requires RasGRF1 (RasGEF1/Cdc25) mediated downstream signaling. Moreover, in axon collapse assays, PRG3-induced neurites resisted growth inhibitors such as myelin, Nogo-A (Reticulon/RTN-4), thrombin and LPA and impeded the RhoA-Rock-PIP5K induced neurite repulsion. Transgenic adult mice with constitutive PRG3 expression displayed strong axonal sprouting distal to a spinal cord lesion. Moreover, fostered PRG3 expression promoted complex motor-behavioral recovery compared to wild type controls as revealed in the Schnell swim test (SST). Thus, PRG3 emerges as a developmental RasGRF1-dependent conductor of filopodia formation and axonal growth enhancer. PRG3-induced neurites resist brain injury-associated outgrowth inhibitors and contribute to functional recovery after spinal cord lesions. Here, we provide evidence that PRG3 operates as an essential neuronal growth promoter in the nervous system. Maintaining PRG3 expression in aging brain may turn back the developmental clock for neuronal regeneration and plasticity.


Assuntos
Bainha de Mielina/metabolismo , Monoéster Fosfórico Hidrolases/metabolismo , Recuperação de Função Fisiológica/fisiologia , Traumatismos da Medula Espinal/metabolismo , Animais , Axônios/metabolismo , Camundongos , Camundongos Transgênicos , Neuritos/metabolismo , Neurônios/metabolismo , Monoéster Fosfórico Hidrolases/genética , Traumatismos da Medula Espinal/genética
13.
Oncotarget ; 7(18): 26692-708, 2016 May 03.
Artigo em Inglês | MEDLINE | ID: mdl-27058420

RESUMO

Malignant gliomas are one of the most devastating cancers in humans. One characteristic hallmark of malignant gliomas is their cellular heterogeneity with frequent genetic lesions and disturbed gene expression levels conferring selective growth advantage. Here, we report on the neuronal-associated growth promoting gene PRG3 executing oncogenic cooperation in gliomas. We have identified perturbed PRG3 levels in human malignant brain tumors displaying either elevated or down-regulated PRG3 levels compared to non-transformed specimens. Further, imbalanced PRG3 levels in gliomas foster Ras-driven oncogenic amplification with increased proliferation and cell migration although angiogenesis was unaffected. Hence, PRG3 interacts with RasGEF1 (RasGRF1/CDC25), undergoes Ras-induced challenges, whereas deletion of the C-terminal domain of PRG3 (PRG3ΔCT) inhibits Ras. Moreover PRG3 silencing makes gliomas resistant to Ras inhibition. In vivo disequilibrated PRG3 gliomas show aggravated proliferation, invasion, and deteriorate clinical outcome. Thus, our data show that the interference with PRG3 homeostasis amplifies oncogenic properties and foster the malignancy potential in gliomas.


Assuntos
Neoplasias Encefálicas/patologia , Glioma/patologia , Monoéster Fosfórico Hidrolases/biossíntese , Animais , Neoplasias Encefálicas/genética , Movimento Celular/genética , Glioma/genética , Humanos , Camundongos , Invasividade Neoplásica/genética , Neovascularização Patológica/genética , Proteínas Oncogênicas/biossíntese , Proteínas Oncogênicas/genética , Monoéster Fosfórico Hidrolases/genética , Ratos , ras-GRF1/metabolismo
14.
FASEB J ; 17(9): 1153-5, 2003 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-12692091

RESUMO

The Nogo gene encodes an integral membrane protein mainly responsible for the neurite inhibition properties of myelin. Here, we analyzed the expression pattern of Nogo-A, Nogo-B, and Nogo-C and Nogo-66 receptor (Ng66R) mRNA during hippocampal development and lesion-induced axonal sprouting. Nogo-A and Nogo-B and Ng66R transcripts preceded the progress of myelination and were highly expressed at postnatal day zero (P0) in all principal hippocampal cell layers, with the exception of dentate granule cells. Only a slight Nogo-C expression was found at P0 in the principal cell layers of the hippocampus. During adulthood, all Nogo splice variants and their receptor were expressed in the neuronal cell layers of the hippocampus, in contrast to the myelin basic protein mRNA expression pattern, which revealed a neuronal source of Nogo gene expression in addition to oligodendrocytes. After hippocampal denervation, the Nogo genes showed an isoform-specific temporal regulation. All Nogo genes were strongly regulated in the hippocampal cell layers, whereas the Ng66R transcripts showed a significant increase in the contralateral cortex. These data could be confirmed on protein levels. Furthermore, Nogo-A expression was up-regulated after kainate-induced seizures. Our data show that neurons express Nogo genes with a clearly distinguishable pattern during development. This expression is further dynamically and isoform-specifically altered after lesioning during the early phase of structural rearrengements. Thus, our results indicate a role for Nogo-A, -B, and -C during development and during the stabilization phase of hippocampal reorganization. Taken together with these data, the finding that neurons in a highly plastic brain region express Nogo genes supports the hypothesis that Nogo may function beyond its known neuronal growth inhibition activity in shaping neuronal circuits.


Assuntos
Hipocampo/crescimento & desenvolvimento , Hipocampo/metabolismo , Proteínas da Mielina/biossíntese , Convulsões/metabolismo , Animais , Denervação , Proteínas Ligadas por GPI , Regulação da Expressão Gênica , Hipocampo/citologia , Modelos Neurológicos , Proteínas da Mielina/genética , Fibras Nervosas Mielinizadas/química , Neurônios/química , Proteínas Nogo , Receptor Nogo 1 , Isoformas de Proteínas/biossíntese , Isoformas de Proteínas/genética , RNA Mensageiro/análise , RNA Mensageiro/metabolismo , Ratos , Receptores de Superfície Celular/biossíntese , Receptores de Superfície Celular/genética , Convulsões/genética , Regulação para Cima
15.
FASEB J ; 17(1): 112-4, 2003 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-12424220

RESUMO

Excitotoxic brain lesions, such as stroke and epilepsy, lead to increasing destruction of neurons hours after the insult. The deadly cascade of events involves detrimental actions by free radicals and the activation of proapoptotic transcription factors, which finally result in neuronal destruction. Here, we provide direct evidence that the nutritionally essential trace element selenium has a pivotal role in neuronal susceptibility to excitotoxic lesions. First, we observed in neuronal cell cultures that addition of selenium in the form of selenite within the physiological range protects against excitotoxic insults and even attenuates primary damage. The neuroprotective effect of selenium is not directly mediated via antioxidative effects of selenite but requires de novo protein synthesis. Gel shift analysis demonstrates that this effect is connected to the inhibition of glutamate-induced NF-kappaB and AP-1 activation. Furthermore, we provide evidence that selenium deficiency in vivo results in a massive increase in susceptibility to kainate-induced seizures and cell loss. These findings indicate the importance of selenium for prevention and therapy of excitotoxic brain damage.


Assuntos
Ácido Glutâmico/toxicidade , Síndromes Neurotóxicas/etiologia , Convulsões/etiologia , Selênio/deficiência , Animais , Morte Celular , Linhagem Celular , Suscetibilidade a Doenças , Antagonistas de Aminoácidos Excitatórios/farmacologia , Hipocampo/patologia , Modelos Neurológicos , NF-kappa B/metabolismo , Neurônios/citologia , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Fármacos Neuroprotetores/farmacologia , Síndromes Neurotóxicas/patologia , Estresse Oxidativo , Biossíntese de Proteínas , Ratos , Convulsões/induzido quimicamente , Convulsões/patologia , Selênio/farmacologia , Fator de Transcrição AP-1/metabolismo
16.
Curr Neuropharmacol ; 13(2): 258-65, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26411769

RESUMO

Malignant brain tumors are characterized by destructive growth and neuronal cell death making them one of the most devastating diseases. Neurodegenerative actions of malignant gliomas resemble mechanisms also found in many neurodegenerative diseases such as Alzheimer's and Parkinson's diseases and amyotrophic lateral sclerosis. Recent data demonstrate that gliomas seize neuronal glutamate signaling for their own growth advantage. Excessive glutamate release via the glutamate/cystine antiporter xCT (system xc-, SLC7a11) renders cancer cells resistant to chemotherapeutics and create the tumor microenvironment toxic for neurons. In particular the glutamate/cystine antiporter xCT takes center stage in neurodegenerative processes and sets this transporter a potential prime target for cancer therapy. Noteworthy is the finding, that reactive oxygen species (ROS) activate transient receptor potential (TRP) channels and thereby TRP channels can potentiate glutamate release. Yet another important biological feature of the xCT/glutamate system is its modulatory effect on the tumor microenvironment with impact on host cells and the cancer stem cell niche. The EMA and FDA-approved drug sulfasalazine (SAS) presents a lead compound for xCT inhibition, although so far clinical trials on glioblastomas with SAS were ambiguous. Here, we critically analyze the mechanisms of action of xCT antiporter on malignant gliomas and in the tumor microenvironment. Deciphering the impact of xCT and glutamate and its relation to TRP channels in brain tumors pave the way for developing important cancer microenvironmental modulators and drugable lead targets.


Assuntos
Sistemas de Transporte de Aminoácidos/metabolismo , Neoplasias Encefálicas/complicações , Neoplasias Encefálicas/patologia , Degeneração Neural/etiologia , Sistemas de Transporte de Aminoácidos/efeitos dos fármacos , Animais , Anti-Inflamatórios não Esteroides/uso terapêutico , Neoplasias Encefálicas/tratamento farmacológico , Ácido Glutâmico/metabolismo , Humanos , Modelos Moleculares , Degeneração Neural/tratamento farmacológico , Degeneração Neural/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Sulfassalazina/uso terapêutico , Canais de Potencial de Receptor Transitório/metabolismo
17.
Cancer Biol Ther ; 16(8): 1205-13, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26047211

RESUMO

Glioblastoma cells produce and release high amounts of glutamate into the extracellular milieu and subsequently can trigger seizure in patients. Tumor-associated microglia/macrophages (TAMs), consisting of both parenchymal microglia and monocytes-derived macrophages (MDMs) recruited from the blood, are known to populate up to 1/3 of the glioblastoma tumor environment and exhibit an alternative, tumor-promoting and supporting phenotype. However, it is unknown how TAMs respond to the excess extracellular glutamate in the glioblastoma microenvironment. We investigated the expressions of genes related to glutamate transport and metabolism in human TAMs freshly isolated from glioblastoma resections. Quantitative real-time PCR analysis showed (i) significant increases in the expressions of GRIA2 (GluA2 or AMPA receptor 2), SLC1A2 (EAAT2), SLC1A3 (EAAT1), (ii) a near-significant decrease in the expression of SLC7A11 (cystine-glutamate antiporter xCT) and (iii) a remarkable increase in GLUL expression (glutamine synthetase) in these cells compared to adult primary human microglia. TAMs co-cultured with glioblastoma cells also exhibited a similar glutamatergic profile as freshly isolated TAMs except for a slight increase in SLC7A11 expression. We next analyzed these genes expressions in cultured human MDMs derived from peripheral blood monocytes for comparison. In contrast, MDMs co-cultured with glioblastoma cells compared to MDMs co-cultured with normal astrocytes exhibited decreased expressions in the tested genes except for GLUL. This is the first study to demonstrate transcriptional changes in glutamatergic signaling of TAMs in a glioblastoma microenvironment, and the findings here suggest that TAMs and MDMs might potentially elicit different cellular responses in the presence of excess extracellular glutamate.


Assuntos
Neoplasias Encefálicas/patologia , Regulação da Expressão Gênica , Glioblastoma/patologia , Ácido Glutâmico/metabolismo , Macrófagos/fisiologia , Microglia/citologia , Antígenos CD/genética , Antígenos de Diferenciação Mielomonocítica/genética , Astrócitos/citologia , Astrócitos/fisiologia , Neoplasias Encefálicas/genética , Antígeno CD11b/genética , Proteínas de Ligação ao Cálcio , Técnicas de Cocultura , Proteínas de Ligação a DNA/genética , Glioblastoma/genética , Ácido Glutâmico/genética , Humanos , Antígenos Comuns de Leucócito/genética , Macrófagos/patologia , Proteínas dos Microfilamentos , Microglia/fisiologia , Receptores de AMPA/genética , Células Tumorais Cultivadas
18.
Sci Rep ; 5: 7958, 2015 Jan 22.
Artigo em Inglês | MEDLINE | ID: mdl-25609379

RESUMO

Malignant gliomas belong to the most threatening tumor entities and are hallmarked by rapid proliferation, hypervascularization and an invasive growth pattern. The primary obstacle in surgical treatment lies in differentiation between healthy and pathological tissue at the tumor margins, where current visualization methods reach their limits. Here, we report on a novel technique (vascular dual intraoperative visualization approach - vDIVA) enabling visualization of different tumor zones (TZ I-III) on the basis of angiogenic hotspots. We investigated glioblastoma patients who underwent 5-ALA fluorescence-guided surgery with simultaneous intraoperative ICG fluorescence angiography. This vDIVA technique revealed hypervascularized areas which were further histologically investigated. Neuropathological assessments revealed tissue areas at the resection margins corresponding to TZ II, and postoperative CD34- and Map2 immunostaining confirmed these angiogenic hotspots to be occupied by glioma cells. Hence, the vascular architecture in this transitional zone could be well differentiated from both primary tumor bulk and healthy brain parenchyma. These data demonstrate that ICG fluorescence angiography improves state-of-the-art glioma surgery techniques and facilitates the future characterization of polyclonal attributes of malignant gliomas.


Assuntos
Neoplasias Encefálicas/irrigação sanguínea , Neoplasias Encefálicas/cirurgia , Glioma/irrigação sanguínea , Glioma/cirurgia , Cuidados Intraoperatórios/métodos , Neovascularização Patológica/cirurgia , Procedimentos Cirúrgicos Vasculares/métodos , Antígenos CD34/metabolismo , Contagem de Células , Angiofluoresceinografia , Humanos , Verde de Indocianina/metabolismo , Proteínas Associadas aos Microtúbulos/metabolismo
19.
Sci Rep ; 5: 12373, 2015 Jul 21.
Artigo em Inglês | MEDLINE | ID: mdl-26197301

RESUMO

Despite advances in multimodal treatments, malignant gliomas remain characterized by a short survival time. Surgical treatment is accepted to be the first line of therapy, with recent studies revealing that maximal possible tumor reduction exerts significant impact on patient outcome. Consideration of tumor localization in relation to functionally eloquent brain areas has been gaining increasing importance. Despite existing assessment methods, the availability of a simple but reliable preoperative grading based on functional data would therefore prove to be indispensable for the prediction of postoperative outcome and hence for overall survival in glioma patients. We performed a clinical investigation comprising 322 patients with gliomas and developed a novel classification system of preoperative tumor status, which considers tumor operability based on two graduations (Friedlein Grading - FG): FGA with lesions at safe distance to eloquent regions which can be completely resected, and FGB referring to tumors which can only be partially resected or biopsied. Investigation of outcome revealed that FGA were characterized by a significantly longer overall survival time compared to FGB. We offer the opportunity to classify brain tumors in a dependable and reproducible manner. The FGA/B grading method provides high prognostic value with respect to overall survival time in relation to the extent of location-dependent tumor resection.


Assuntos
Neoplasias Encefálicas/patologia , Glioma/patologia , Adolescente , Adulto , Neoplasias Encefálicas/terapia , Criança , Terapia Combinada/métodos , Feminino , Glioma/terapia , Humanos , Masculino , Pessoa de Meia-Idade , Prognóstico , Adulto Jovem
20.
FEBS Lett ; 563(1-3): 41-8, 2004 Apr 09.
Artigo em Inglês | MEDLINE | ID: mdl-15063720

RESUMO

Damage to the central nervous system triggers rapid activation and specific migration of glial cells towards the lesion site. There, glial cells contribute heavily to secondary neuronal changes that take place after lesion. In an attempt to identify the molecular cues of glial activation following brain trauma we performed differential display reverse transcription-polymerase chain reaction screenings from lesioned and control hippocampus. Here we report on the identification of the macrophage/microglia activation factor (MAF), a new membrane protein with seven putative transmembrane domains. Expression analysis revealed that MAF is predominantly expressed in microglial cells in the brain, and is upregulated following brain lesion. Overexpression of MAF in non-glial cells shows an intracellular codistribution with the lysosomal marker endosome/lysosome-associated membrane protein-1 (lamp-1). Furthermore, MAF-transfected cells show that MAF is primarily associated with late endosomes/lysosomes, and that this association can be disrupted by activation of protein kinase C-dependent pathways. In conclusion, these results imply that MAF is involved in the dynamics of lysosomal membranes associated with microglial activation following brain lesion.


Assuntos
Endossomos/metabolismo , Lisossomos/metabolismo , Fatores Ativadores de Macrófagos/química , Fatores Ativadores de Macrófagos/metabolismo , Microglia/metabolismo , Sequência de Aminoácidos , Animais , Antígenos CD/metabolismo , Sequência de Bases , Células COS , Células Cultivadas , Chlorocebus aethiops , Códon de Iniciação , Códon de Terminação , Regulação da Expressão Gênica , Hipocampo/citologia , Hipocampo/lesões , Proteínas de Membrana Lisossomal , Fatores Ativadores de Macrófagos/genética , Masculino , Camundongos , Microglia/citologia , Dados de Sequência Molecular , Células NIH 3T3 , Neurônios/citologia , Neurônios/metabolismo , Células PC12 , Estrutura Terciária de Proteína , RNA Mensageiro/metabolismo , Ratos , Ratos Sprague-Dawley
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA