Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
1.
J Neurochem ; 136(1): 118-32, 2016 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-26466330

RESUMO

Thymosin beta 4 (Tß4), a secreted 43 amino acid peptide, promotes oligodendrogenesis, and improves neurological outcome in rat models of neurologic injury. We demonstrated that exogenous Tß4 treatment up-regulated the expression of the miR-200a in vitro in rat brain progenitor cells and in vivo in the peri-infarct area of rats subjected to middle cerebral artery occlusion (MCAO). The up-regulation of miR-200a down-regulated the expression of the following targets in vitro and in vivo models: (i) growth factor receptor-bound protein 2 (Grb2), an adaptor protein involved in epidermal growth factor receptor (EGFR)/Grb2/Ras/MEK/ERK1/c-Jun signaling pathway, which negatively regulates the expression of myelin basic protein (MBP), a marker of mature oligodendrocyte; (ii) ERRFI-1/Mig-6, an endogenous potent kinase inhibitor of EGFR, which resulted in activation/phosphorylation of EGFR; (iii) friend of GATA 2, and phosphatase and tensin homolog deleted in chromosome 10 (PTEN), which are potent inhibitors of the phosphatidylinositol-3-kinase (PI3K)/AKT signaling pathway, and resulted in marked activation of AKT; and (iv) transcription factor, p53, which induces pro-apoptotic genes, and possibly reduced apoptosis of the progenitor cells subjected to oxygen glucose deprivation (OGD). Anti-miR-200a transfection reversed all the effects of Tß4 treatment in vitro. Thus, Tß4 up-regulated MBP synthesis, and inhibited OGD-induced apoptosis in a novel miR-200a dependent EGFR signaling pathway. Our findings of miR-200a-mediated protection of progenitor cells may provide a new therapeutic importance for the treatment of neurologic injury. Tß4-induced micro-RNA-200a (miR-200a) regulates EGFR signaling pathways for MBP synthesis and apoptosis: up-regulation of miR-200a after Tß4 treatment, increases MBP synthesis after targeting Grb2 and thereby inactivating c-Jun from inhibition of MBP synthesis; and also inhibits OGD-mediated apoptosis after targeting EGFR inhibitor (Mig-6), PI3K inhibitors (FOG2 and Pten) and an inducer (p53) of pro-apoptotic genes, for AKT activation and down-regulation of p53. These findings may contribute the therapeutic benefits for stroke and other neuronal diseases associated with demyelination disorders.


Assuntos
Encéfalo/metabolismo , Diferenciação Celular/fisiologia , Sobrevivência Celular/fisiologia , MicroRNAs/biossíntese , Células-Tronco/metabolismo , Timosina/farmacologia , Animais , Encéfalo/efeitos dos fármacos , Diferenciação Celular/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Regulação da Expressão Gênica , Masculino , MicroRNAs/genética , Ratos , Ratos Wistar , Células-Tronco/efeitos dos fármacos , Regulação para Cima/efeitos dos fármacos , Regulação para Cima/fisiologia
2.
J Biol Chem ; 289(28): 19508-18, 2014 Jul 11.
Artigo em Inglês | MEDLINE | ID: mdl-24828499

RESUMO

Thymosin ß4 (Tß4), a G-actin-sequestering peptide, improves neurological outcome in rat models of neurological injury. Tissue inflammation results from neurological injury, and regulation of the inflammatory response is vital for neurological recovery. The innate immune response system, which includes the Toll-like receptor (TLR) proinflammatory signaling pathway, regulates tissue injury. We hypothesized that Tß4 regulates the TLR proinflammatory signaling pathway. Because oligodendrogenesis plays an important role in neurological recovery, we employed an in vitro primary rat embryonic cell model of oligodendrocyte progenitor cells (OPCs) and a mouse N20.1 OPC cell line to measure the effects of Tß4 on the TLR pathway. Cells were grown in the presence of Tß4, ranging from 25 to 100 ng/ml (RegeneRx Biopharmaceuticals Inc., Rockville, MD), for 4 days. Quantitative real-time PCR data demonstrated that Tß4 treatment increased expression of microRNA-146a (miR-146a), a negative regulator the TLR signaling pathway, in these two cell models. Western blot analysis showed that Tß4 treatment suppressed expression of IL-1 receptor-associated kinase 1 (IRAK1) and tumor necrosis factor receptor-associated factor 6 (TRAF6), two proinflammatory cytokines of the TLR signaling pathway. Transfection of miR-146a into both primary rat embryonic OPCs and mouse N20.1 OPCs treated with Tß4 demonstrated an amplification of myelin basic protein (MBP) expression and differentiation of OPC into mature MBP-expressing oligodendrocytes. Transfection of anti-miR-146a nucleotides reversed the inhibitory effect of Tß4 on IRAK1 and TRAF6 and decreased expression of MBP. These data suggest that Tß4 suppresses the TLR proinflammatory pathway by up-regulating miR-146a.


Assuntos
Diferenciação Celular , MicroRNAs/metabolismo , Oligodendroglia/metabolismo , Transdução de Sinais , Timosina/biossíntese , Receptores Toll-Like/metabolismo , Regulação para Cima , Animais , Citocinas/genética , Citocinas/metabolismo , Células Hep G2 , Humanos , Inflamação/genética , Inflamação/metabolismo , Inflamação/patologia , Quinases Associadas a Receptores de Interleucina-1/genética , Quinases Associadas a Receptores de Interleucina-1/metabolismo , Camundongos , MicroRNAs/genética , Proteína Básica da Mielina/genética , Proteína Básica da Mielina/metabolismo , Oligodendroglia/patologia , Ratos , Ratos Wistar , Fator 6 Associado a Receptor de TNF/genética , Fator 6 Associado a Receptor de TNF/metabolismo , Timosina/genética , Receptores Toll-Like/genética
3.
Sci Rep ; 14(1): 7676, 2024 04 01.
Artigo em Inglês | MEDLINE | ID: mdl-38561433

RESUMO

The conserved miR-183/96/182 cluster (miR-183C) is expressed in both corneal resident myeloid cells (CRMCs) and sensory nerves (CSN) and modulates corneal immune/inflammatory responses. To uncover cell type-specific roles of miR-183C in CRMC and CSN and their contributions to corneal physiology, myeloid-specific miR-183C conditional knockout (MS-CKO), and sensory nerve-specific CKO (SNS-CKO) mice were produced and characterized in comparison to the conventional miR-183C KO. Immunofluorescence and confocal microscopy of flatmount corneas, corneal sensitivity, and tear volume assays were performed in young adult naïve mice; 3' RNA sequencing (Seq) and proteomics in the trigeminal ganglion (TG), cornea and CRMCs. Our results showed that, similar to conventional KO mice, the numbers of CRMCs were increased in both MS-CKO and SNS-CKO vs age- and sex-matched WT control littermates, suggesting intrinsic and extrinsic regulations of miR-183C on CRMCs. The number of CRMCs was increased in male vs female MS-CKO mice, suggesting sex-dependent regulation of miR-183C on CRMCs. In the miR-183C KO and SNS-CKO, but not the MS-CKO mice, CSN density was decreased in the epithelial layer of the cornea, but not the stromal layer. Functionally, corneal sensitivity and basal tear volume were reduced in the KO and SNS-CKO, but not the MS-CKO mice. Tear volume in males is consistently higher than female WT mice. Bioinformatic analyses of the transcriptomes revealed a series of cell-type specific target genes of miR-183C in TG sensory neurons and CRMCs. Our data elucidate that miR-183C imposes intrinsic and extrinsic regulation on the establishment and function of CSN and CRMCs by cell-specific target genes. miR-183C modulates corneal sensitivity and tear production through its regulation of corneal sensory innervation.


Assuntos
MicroRNAs , Fenômenos Fisiológicos do Sistema Nervoso , Camundongos , Masculino , Feminino , Animais , Córnea/inervação , Gânglio Trigeminal/fisiologia , MicroRNAs/genética , Células Mieloides
4.
Glia ; 60(12): 1826-38, 2012 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-23073962

RESUMO

Thymosin beta 4 (Tß4), a G-actin sequestering peptide, increases oligodendrogenesis and improves functional outcome in models of neurological injury. The molecular mechanisms of Tß4 mediated oligodendrogenesis are unclear. The p38 mitogen-activated protein kinase (p38MAPK) regulates oligodendrocyte (OL) differentiation and myelin gene expression in other models. Therefore, we investigated p38MAPK signaling pathways. We used primary rat neural progenitor cells (NPCs) and a mouse oligodendrocyte progenitor cell (OPC) line (N20.1 cells) to investigate the molecular mechanisms of Tß4-enhanced oligodendrogenesis. NPCs were isolated from rat subventricular zone (SVZ) of the lateral ventricles (n = 12). Primary NPCs and N20.1 cells were grown in the presence of 0, 25, and 50 ng/mL of Tß4 (RegeneRx Biopharmaceuticals Inc, Rockville, MD) for 14 days. Quantitative real-time PCR and Western blot data showed significant induction of both expression and phosphorylation of p38MAPK with simultaneous inhibition of phosphorylation of extracellular signal regulated kinase (ERK1), c-Jun N-terminal kinase 1 (JNK1), leading to reduction of phosphorylation of c-Jun, a potent negative regulator of transcription of myelin genes. These effects were reversed with transfection of Tß4siRNA. Our data indicate that Tß4 treatment induces OL differentiation by inducing p38MAPK with parallel inactivation of ERK1 and JNK1, thus preventing the accumulation of phosphorylated c-Jun.


Assuntos
Diferenciação Celular/fisiologia , Sistema de Sinalização das MAP Quinases/fisiologia , Oligodendroglia/enzimologia , Timosina/fisiologia , Regulação para Cima/fisiologia , Proteínas Quinases p38 Ativadas por Mitógeno/biossíntese , Animais , Linhagem Celular , Células Cultivadas , Masculino , Camundongos , Fosforilação , Ratos , Células-Tronco/metabolismo
5.
Cancer Sci ; 102(7): 1350-7, 2011 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-21477071

RESUMO

Analysis of microarray probe data from glioma patient samples, in conjunction with patient Kaplan-Meier survival plots, indicates that expression of a glioma suppressor gene doublecortin (DCX) favors glioma patient survival. From neurosphere formation in culture, time-lapse microscopic video recording, and tumor xenograft, we show that DCX synthesis significantly reduces self-renewal of brain tumor stem cells (BTSC) in human primary glioma (YU-PG, HF66) cells from surgically removed human glioma specimens and U87 cells in vitro and in vivo. Time-lapse microscopic video recording revealed that double transfection of YU-PG, HF66, and U87 cells with DCX and neurabin II caused incomplete cell cycle with failure of cytokinesis, that is, endomitosis by dividing into three daughter cells from one mother BTSC. Activation of c-jun NH2-terminal kinase 1 (JNK1) after simvastatin (10 nM) treatment of DCX(+) neurabin II(+) BTSC from YU-PG, HF66, and U87 cells induced terminal differentiation into neuron-like cells. dUTP nick end labeling data indicated that JNK1 activation also induced apoptosis only in double transfected BTSC with DCX and neurabin II, but not in single transfected BTSC from YU-PG, HF66, and U87 cells. Western blot analysis showed that procaspase-3 was induced after DCX transfection and activated after simvastatin treatment in YU-PG, HF66, and U87 BTSC. Sequential immunoprecipitation and Western blot data revealed that DCX synthesis blocked protein phosphatase-1 (PP1)/caspase-3 protein-protein interaction and increased PP1-DCX interaction. These data show that DCX synthesis induces apoptosis in BTSC through a novel JNK1/neurabin II/DCX/PP1/caspase-3 pathway.


Assuntos
Neoplasias Encefálicas/patologia , Glioma/patologia , Proteínas Associadas aos Microtúbulos/fisiologia , Células-Tronco Neoplásicas/patologia , Neuropeptídeos/fisiologia , Animais , Apoptose/efeitos dos fármacos , Neoplasias Encefálicas/mortalidade , Caspase 3/fisiologia , Diferenciação Celular , Linhagem Celular Tumoral , Proteínas do Domínio Duplacortina , Proteína Duplacortina , Glioma/mortalidade , Humanos , Masculino , Proteínas dos Microfilamentos/fisiologia , Proteína Quinase 8 Ativada por Mitógeno/fisiologia , Proteínas do Tecido Nervoso/fisiologia , Proteína Fosfatase 1/fisiologia , Ratos , Sinvastatina/farmacologia
6.
J Neurosci Res ; 88(2): 304-14, 2010 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-19681167

RESUMO

We employed lentivirus-based doublecortin (DCX), as a glioma suppressor gene therapy in an intracranial glioma tumor xenograft model in nude rats. Single DCX-expressing lentivirus was directly administered into the tumor on day 8 after U87 tumor cell implantation. DCX treatment significantly reduced U87 glioma tumor volume (approximately 60%) on day 14 after DCX lentivirus injection and significantly improved median survival of tumor-bearing nude rats. DCX synthesis induced neuronal markers MAP2, TUJ1, and PSA-NCAM and the glial marker glial fibrillary acidic protein (GFAP) in the implanted U87 glioma tumors. DCX synthesis induced GFAP that colocalized with tubulin in the mitotic stage, inhibited cleavage furrow during cytokinesis, and blocked mitosis in glioma cells. DCX lentivirus infection did not induce apoptosis but significantly inhibited expression of the proliferation marker Ki-67 and the blood vessel marker von-Willebrand factor (vWF). U87 and other glioma cells except for brain tumor stem cells (BTSCs) do not express neuronal markers or both neuronal and glial markers. DCX-synthesizing glioma cells express a phenotype of antiangiogenic BTSC-like cells with terminal differentiation that causes remission of glioma cells by blocking mitosis via a novel DCX/GFAP pathway. Direct local delivery of lentivirus-based DCX gene therapy is a potential differentiation-based therapeutic approach for the treatment of glioma.


Assuntos
Neoplasias Encefálicas/terapia , Terapia Genética/métodos , Glioma/terapia , Proteínas Associadas aos Microtúbulos/genética , Proteínas Associadas aos Microtúbulos/uso terapêutico , Neuropeptídeos/genética , Neuropeptídeos/uso terapêutico , Animais , Astrócitos/patologia , Astrócitos/fisiologia , Encéfalo/patologia , Encéfalo/fisiopatologia , Neoplasias Encefálicas/patologia , Neoplasias Encefálicas/fisiopatologia , Linhagem Celular Tumoral , Citocinese/fisiologia , Proteínas do Domínio Duplacortina , Proteína Duplacortina , Vetores Genéticos , Proteína Glial Fibrilar Ácida/metabolismo , Glioma/patologia , Glioma/fisiopatologia , Lentivirus/genética , Transplante de Neoplasias , Neovascularização Patológica/patologia , Neovascularização Patológica/fisiopatologia , Neovascularização Patológica/terapia , Neurônios/patologia , Neurônios/fisiologia , Ratos , Ratos Nus , Resultado do Tratamento
7.
J Neurochem ; 108(1): 231-45, 2009 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-19094064

RESUMO

Doublecortin (DCX) is a microtubule (MT) binding protein that induces growth arrest at the G2-M phase of cell cycle in glioma and suppresses tumor xenograft in immunocompromised hosts. DCX expression was found in neuronal cells, but lacking in glioma cells. We tested the hypothesis that DCX inhibits glioma U87 cell mitosis and invasion. Our data showed that DCX synthesizing U87 cells underwent mitotic MT spindle catastrophe in a neurabin II dependent pathway. Synthesis of both DCX and neurabin II were required to induce apoptosis in U87 and human embryonic kidney 293T cells. In DCX expressing U87 cells, association of phosphorylated DCX with protein phosphatase-1 (PP1) in the cytosol disrupted the interaction between kinesin-13 and PP1 in the nucleus and yielded spontaneously active kinesin-13. The activated kinesin-13 caused mitotic MT catastrophe in spindle checkpoint. Phosphorylated-DCX induced depolymerization of actin filaments in U87 cells, down-regulated matrix metalloproteinases-2 and -9, and inhibited glioma U87 cell invasion in a neurabin II dependent pathway. Thus, localization of the DCX-neurabin II-PP1 complex in the cytosol of U87 tumor cells inhibited PP1 phosphatase activities leading to anti-glioma effects via (1) mitotic MT spindle catastrophe that blocks mitosis and (2) depolymerization of actin that inhibits glioma cell invasion.


Assuntos
Regulação Neoplásica da Expressão Gênica , Glioma/metabolismo , Proteínas Associadas aos Microtúbulos/metabolismo , Mitose , Neuropeptídeos/metabolismo , Fuso Acromático/fisiologia , Citoesqueleto de Actina , Adulto , Análise de Variância , Animais , Encéfalo/metabolismo , Linhagem Celular , Movimento Celular , Proliferação de Células , Células Cultivadas , Proteínas do Domínio Duplacortina , Proteína Duplacortina , Citometria de Fluxo , Humanos , Marcação In Situ das Extremidades Cortadas/métodos , Cinesinas/metabolismo , Masculino , Metaloproteinase 2 da Matriz/metabolismo , Metaloproteinase 9 da Matriz/metabolismo , Camundongos , Camundongos Nus , Proteínas dos Microfilamentos/metabolismo , Proteínas Associadas aos Microtúbulos/genética , Mitose/fisiologia , Proteínas do Tecido Nervoso/metabolismo , Neuroglia/metabolismo , Neuropeptídeos/genética , Proteína Fosfatase 1/metabolismo , RNA Mensageiro/metabolismo , RNA Interferente Pequeno/farmacologia , Ratos , Transfecção/métodos , Células Tumorais Cultivadas , Adulto Jovem
8.
J Neurochem ; 105(2): 324-37, 2008 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-18021292

RESUMO

We demonstrate that a proteoglycan decorin (DCN) up-regulates the vascular endothelial growth factor (VEGF) expression with activation of VEGF regulating transcription factors Sp1, hypoxia-inducible factor 1alpha (HIF1alpha), and signal transducer and activator of transcription 3 (Stat3) via epidermal growth factor receptor (EGFR), mitogen-activated protein kinase extracellular signal-regulated kinase 1/2 (ERK1/2), and protein kinase B (AKT) pathways in DCN transfected mouse cerebral endothelial (MCE) cells. Treatment with pharmacological inhibitors and small interfering RNAs reveal that induction and activation of Sp1, HIF1alpha, and Stat3 facilitate their nuclear localization and binding to their specific motifs of the VEGF promoter and induce VEGF expression via two independent pathways, DCN/EGFR/phosphoinositide-3 kinase/AKT and DCN/EGFR/ERK1/2, respectively, in DCN synthesizing MCE cells. The cell type specific glycosylation protects Sp1 and HIF1alpha from proteosome degradation and plays an important and novel role in the regulation of VEGF in DCN transfected MCE cells. Induction of gelatinases (matrix metalloproteinase 2 and 9), the serine protease tissue plasminogen activator and plasmin by DCN transfection in MCE cells leads to extracellular proteolysis and to release of matrix-bound VEGF and activation of angiogenesis. In this study, we demonstrate that two independent downstream signal pathways, DCN/EGFR/ERK1/2 and DCN/EGFR/phosphoinositide-3 kinase/AKT, mediate up-regulation and activation of transcription factors of VEGF such as HIF1alpha, Stat3, and Sp1 and increase VEGF transcription and angiogenesis in MCE cells.


Assuntos
Córtex Cerebral/citologia , Células Endoteliais/metabolismo , Proteínas da Matriz Extracelular/metabolismo , Proteoglicanas/metabolismo , Fatores de Transcrição/metabolismo , Regulação para Cima/efeitos dos fármacos , Fator A de Crescimento do Endotélio Vascular/metabolismo , Análise de Variância , Animais , Imunoprecipitação da Cromatina/métodos , Decorina , Ensaio de Desvio de Mobilidade Eletroforética/métodos , Ativação Enzimática/efeitos dos fármacos , Inibidores Enzimáticos/farmacologia , Proteínas da Matriz Extracelular/farmacologia , Subunidade alfa do Fator 1 Induzível por Hipóxia/genética , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Camundongos , Modelos Biológicos , Neovascularização Fisiológica/efeitos dos fármacos , Neovascularização Fisiológica/fisiologia , Proteoglicanas/farmacologia , RNA Interferente Pequeno/farmacologia , Fator de Transcrição STAT3/genética , Fator de Transcrição STAT3/metabolismo , Fator de Transcrição Sp1/genética , Fator de Transcrição Sp1/metabolismo , Fatores de Transcrição/genética , Transfecção/métodos , Regulação para Cima/fisiologia
9.
Brain Res ; 1226: 18-26, 2008 Aug 21.
Artigo em Inglês | MEDLINE | ID: mdl-18598677

RESUMO

The chemokine receptor CXCR4 and its ligand, stromal cell derived factor-1 alpha (SDF1 alpha) regulate neuroblast migration towards the ischemic boundary after stroke. Using loss- and gain-function, we investigated the biological effect of CXCR4/SDF1 alpha on neural progenitor cells. Neural progenitor cells, from the subventricular zone (SVZ) of the adult rat, were transfected with rat CXCR4-pLEGFP-C1 and pSIREN-RetroQ-CXCR4-siRNA retroviral vectors. Migration assay analysis showed that inhibition of CXCR4 by siRNA significantly reduced cell migration compared to the empty vector, indicating that CXCR4 mediated neural progenitor cell motility. When neural progenitor cells were cultured in growth medium containing bFGF (20 ng/ml), over-expression of CXCR4 significantly reduced the cell proliferation as measured by the number of bromodeoxyuridine+ (BrdU+) cells (26.4%) compared with the number in the control group (54.0%). Addition of a high concentration of SDF1 alpha (500 ng/ml) into the progenitor cells with over-expression of CXCR4 reversed the cell proliferation back to the control levels (57.6%). Immunostaining analysis showed that neither over-expression nor inhibition of CXCR4 altered the population of neurons and astrocytes, when neural progenitor cells were cultured in differentiation medium. These in vitro results suggest that CXCR4/SDF1 alpha primarily regulates adult neural progenitor cell motility but not differentiation, while over-expression of CXCR4 in the absence of SDF1 alpha decreases neural progenitor cell proliferation.


Assuntos
Células-Tronco Adultas/efeitos dos fármacos , Ventrículos Cerebrais/citologia , Quimiocina CXCL12/farmacologia , Neurônios/efeitos dos fármacos , Receptores CXCR4/metabolismo , Animais , Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo , Bromodesoxiuridina/metabolismo , Diferenciação Celular/efeitos dos fármacos , Diferenciação Celular/genética , Movimento Celular/efeitos dos fármacos , Movimento Celular/genética , Proliferação de Células/efeitos dos fármacos , Células Cultivadas , Fatores de Crescimento de Fibroblastos/farmacologia , Expressão Gênica/efeitos dos fármacos , Expressão Gênica/fisiologia , Perfilação da Expressão Gênica/métodos , Proteínas Luminescentes/genética , Proteínas Luminescentes/metabolismo , Análise de Sequência com Séries de Oligonucleotídeos/métodos , RNA Interferente Pequeno/metabolismo , Ratos , Ratos Wistar , Receptores CXCR4/genética , Transfecção/métodos
10.
Cancer Res ; 66(24): 11726-35, 2006 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-17178868

RESUMO

Doublecortin (DCX) is one of the three genes found from Affymetrix gene chip analysis related to glioma patient survival. Two other genes (e.g., osteonectin and semaphorin 3B) are well characterized as antioncogenic and tumor suppressor genes. However, there is no report about the involvement of DCX in cancer. Here, we show that gene transfer technology into DCX-deficient glioblastoma cell lines, such as A172, U87, U251N, RG2, and 9L, with DCX cDNA significantly suppressed growth of these glioma cells. U87 cells with ectopic expression of DCX exhibit a marked suppression of the transformed phenotype as growth arrested in the G(2) phase of the cell cycle progression, small colony formation in soft agar, and no tumor formation in nude rats. This transformed phenotype can be restored by knocking down DCX expression with DCX small interfering RNA. DCX was highly phosphorylated in glioma cells. Phosphorylation in the glioma cells was greater than in noncancer cells such as mouse NIH 3T3 and human embryonic kidney 293T cells. Coimmunoprecipitation of the phosphorylated DCX and spinophilin/neurabin II from DCX-synthesizing glioma cells indicated their interaction. This interaction would lead to a block of anchorage-independent growth as neurabin II is a synergistic inhibitor of anchorage-independent growth with p14ARF (ARF). Interaction between phosphorylated DCX and neurabin II may induce the association of the protein phosphatase 1 catalytic subunit (PP1) with neurabin II and inactivate PP1 and block mitosis during G(2) and M phases of the cell cycle progression. Thus, DCX seems to be a tumor suppressor of glioma.


Assuntos
Regulação Neoplásica da Expressão Gênica , Glioblastoma/genética , Proteínas Associadas aos Microtúbulos/genética , Neuropeptídeos/genética , Células 3T3 , Animais , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/patologia , Ciclo Celular/genética , Linhagem Celular Tumoral , Proteínas do Domínio Duplacortina , Proteína Duplacortina , Vetores Genéticos , Glioblastoma/patologia , Humanos , Lentivirus/genética , Camundongos , Proteínas Associadas aos Microtúbulos/metabolismo , Mitose/genética , Neuropeptídeos/metabolismo , Fosforilação , Biossíntese de Proteínas , RNA Mensageiro/genética , RNA Interferente Pequeno/genética , Ratos , Sobrevida , Transcrição Gênica
11.
J Cereb Blood Flow Metab ; 27(4): 669-78, 2007 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-16926839

RESUMO

Cerebral stroke induces proliferation of subventricular zone (SVZ) neural progenitor cells in adult rodent brain. Tumor necrosis factor-alpha-converting enzyme (TACE) proteolysis sheds the nonamyloidogenic soluble ectodomain of the amyloid precursor protein (APP) and is a convertase for tumor necrosis factor-alpha (TNFalpha). The resulting soluble peptides of APP and TNFalpha are mitogenic for neural progenitor cells of the SVZ. Therefore, we hypothesized a role for TACE proteolysis in stroke-induced neurogenesis. Using laser-capture microdissection, we found TACE transcription was increased in SVZ cells of ischemic brain. Immunohistochemistry revealed TACE protein was upregulated in SVZ neuroblasts. Intraventricular infusion of tumor necrosis factor-alpha protease inhibitor-2 (TAPI-2) decreased bromodeoxyuridine incorporation in SVZ cells of rats subjected to middle cerebral artery occlusion. Furthermore, primary culture SVZ neurospheres from ischemic brain overexpress TACE and its substrates APP and TNF-alpha. These cells proliferated more rapidly, possessed increased TACE protease-dependent alpha-secretase activity, and released more soluble APP and TNFalpha compared with nonischemic control. In addition, TAPI-2 reduced SVZ neuroblast migration out of SVZ explants in vitro. These findings indicate TACE proteolysis as a promoter of stroke-induced SVZ progenitor cell neurogenesis, and suggest this protease activity may represent an attractive therapeutic target for stroke recovery.


Assuntos
Proteínas ADAM/fisiologia , Ventrículos Cerebrais/patologia , Acidente Vascular Cerebral/patologia , Proteínas ADAM/metabolismo , Proteína ADAM17 , Precursor de Proteína beta-Amiloide/biossíntese , Animais , Western Blotting , Bromodesoxiuridina/metabolismo , Proliferação de Células , Células Cultivadas , Dissecação , Ensaio de Imunoadsorção Enzimática , Imuno-Histoquímica , Infarto da Artéria Cerebral Média/patologia , Lasers , Masculino , Ratos , Ratos Wistar , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Fator de Necrose Tumoral alfa/metabolismo
12.
J Cereb Blood Flow Metab ; 26(10): 1311-22, 2006 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-16467781

RESUMO

We employed an in vitro hypoxia cell culture model system and gene transfer technology to examine the effect of the decorin gene on cell survival against oxygen and glucose deprivation (OGD). Ectopic expression of decorin in subventricular zone (SVZ) cells from adult male mouse brain and human glioblastoma U-87 cells kept the cells viable against 24 h of OGD. Fewer than 1% of decorin-synthesizing cells were apoptotic after 12 h of OGD. In contrast, 100% of the control cells were apoptotic even after 4 h of OGD. De novo decorin synthesis in SVZ and U-87 cells induced expression of p21, p27 and Ras, AKT (acutely transforming retrovirus AKT8 in rodent T-cell lymphoma), and phosphorylated AKT. Blocking of phosphoinositide 3-kinase (PI-3K), Ras, and the epidermal growth factor receptor with specific inhibitors had no effect on induction of Ras, p21, and p27 at the messenger RNA level in decorin-synthesizing SVZ and U-87 cells. PI-3K inhibitors significantly increased apoptosis in decorin-expressing cells. Our data indicate that induction of p21, p27, Ras, AKT, and phosphorylated AKT by decorin inhibits apoptosis and protects U-87 and SVZ cells against OGD. Therefore, our data suggest that decorin is a potent trophic factor that protects neuronal progenitor cells and glioma cells from OGD.


Assuntos
Envelhecimento/fisiologia , Proteínas da Matriz Extracelular/metabolismo , Glioma/metabolismo , Glioma/patologia , Glucose/deficiência , Oxigênio/metabolismo , Proteoglicanas/metabolismo , Células-Tronco/metabolismo , Animais , Linhagem Celular , Sobrevivência Celular/efeitos dos fármacos , Inibidor de Quinase Dependente de Ciclina p21/metabolismo , Inibidor de Quinase Dependente de Ciclina p27/metabolismo , Decorina , Receptores ErbB/antagonistas & inibidores , Receptores ErbB/metabolismo , Proteínas da Matriz Extracelular/genética , Farnesiltranstransferase/antagonistas & inibidores , Farnesiltranstransferase/metabolismo , Glucose/farmacologia , Humanos , Camundongos , Modelos Biológicos , Oxigênio/farmacologia , Fosfatidilinositol 3-Quinases/metabolismo , Inibidores de Fosfoinositídeo-3 Quinase , Fosforilação , Proteoglicanas/genética , Proteínas Proto-Oncogênicas c-akt/metabolismo , Células-Tronco/citologia , Células-Tronco/efeitos dos fármacos , Regulação para Cima/efeitos dos fármacos , Proteínas ras/metabolismo
13.
Oncogene ; 21(31): 4765-77, 2002 Jul 18.
Artigo em Inglês | MEDLINE | ID: mdl-12101415

RESUMO

The progressive growth of most neoplasms is dependent upon the establishment of new blood vessels, a process regulated by tumor-secreted factors and matrix proteins. We examined the in vitro and in vivo angiogenic ability of conditioned media obtained from fibrosarcoma, carcinoma, and osteosarcoma cells and their decorin-transfected counterparts. Human endothelial cells were investigated in vitro by evaluating three essential steps of angiogenesis: migration, attachment, and differentiation. On the whole, wild-type tumor cell-secretions enhanced endothelial cell attachment, migration, and differentiation, whereas their decorin-expressing forms inhibited these processes. Similarly, decorin-containing media suppressed endothelial cell sprouting in an ex vivo aortic ring assay. Since angiogenesis is an important component of tumor expansion, the growth rate of these cells as tumor xenografts was examined by implantation in nude mice. In vivo, the decorin-expressing tumor xenografts grew at markedly lower rates and showed a significant suppression of neovascularization. Immunohistochemical, Northern and Western blot analyses indicated that the decorin-expressing cells produced vascular endothelial growth factor (VEGF) at markedly reduced rates vis-á-vis their wild-type counterparts. Specificity of this process was confirmed by experiments where addition of recombinant decorin to the wild-type tumor cells caused 80-95% suppression of VEGF mRNA and protein. These results provide a novel mechanism of action for decorin, and indicate that decorin could adversely affect in vivo tumor growth by suppressing the endogenous tumor cell production of a powerful angiogenic stimulus.


Assuntos
Neoplasias/irrigação sanguínea , Neovascularização Patológica , Proteoglicanas/fisiologia , Animais , Capilares/efeitos dos fármacos , Capilares/crescimento & desenvolvimento , Adesão Celular/efeitos dos fármacos , Divisão Celular , Células Cultivadas , Quimiotaxia , Meios de Cultivo Condicionados/química , Meios de Cultivo Condicionados/farmacologia , Técnicas de Cultura , Decorina , Relação Dose-Resposta a Droga , Fatores de Crescimento Endotelial/metabolismo , Endotélio Vascular/citologia , Endotélio Vascular/efeitos dos fármacos , Endotélio Vascular/fisiologia , Proteínas da Matriz Extracelular , Humanos , Laminina/metabolismo , Linfocinas/metabolismo , Camundongos , Camundongos Nus , Neoplasias/metabolismo , Neoplasias/patologia , Proteoglicanas/análise , Células Tumorais Cultivadas , Fator A de Crescimento do Endotélio Vascular , Fatores de Crescimento do Endotélio Vascular , Ensaios Antitumorais Modelo de Xenoenxerto
14.
PLoS One ; 5(6): e11016, 2010 Jun 11.
Artigo em Inglês | MEDLINE | ID: mdl-20552017

RESUMO

BACKGROUND: Erythropoietin (EPO), a hematopoietic cytokine, enhances neurogenesis and angiogenesis during stroke recovery. In the present study, we examined the effect of EPO on oligodendrogenesis in a rat model of embolic focal cerebral ischemia. METHODOLOGY AND PRINCIPAL FINDINGS: Recombinant human EPO (rhEPO) at a dose of 5,000 U/kg (n = 18) or saline (n = 18) was intraperitoneally administered daily for 7 days starting 24 h after stroke onset. Treatment with rhEPO augmented actively proliferating oligodendrocyte progenitor cells (OPCs) measured by NG2 immunoreactive cells within the peri-infarct white matter and the subventricular zone (SVZ), but did not protect against loss of myelinating oligodendrocytes measured by cyclic nucleotide phosphodiesterase (CNPase) positive cells 7 days after stroke. However, 28 and 42 days after stroke, treatment with rhEPO significantly increased myelinating oligodendrocytes and myelinated axons within the peri-infarct white matter. Using lentivirus to label subventricular zone (SVZ) neural progenitor cells, we found that in addition to the OPCs generated in the peri-infarct white matter, SVZ neural progenitor cells contributed to rhEPO-increased OPCs in the peri-infarct area. Using bromodeoxyuridine (BrdU) for birth-dating cells, we demonstrated that myelinating oligodendrocytes observed 28 days after stroke were derived from OPCs. Furthermore, rhEPO significantly improved neurological outcome 6 weeks after stroke. In vitro, rhEPO increased differentiation of adult SVZ neural progenitor cells into oligodendrocytes and enhanced immature oligodendrocyte cell proliferation. CONCLUSIONS: Our in vivo and in vitro data indicate that EPO amplifies stroke-induced oligodendrogenesis that could facilitate axonal re-myelination and lead to functional recovery after stroke.


Assuntos
Eritropoetina/farmacologia , Oligodendroglia/efeitos dos fármacos , Acidente Vascular Cerebral/patologia , Animais , Axônios , Divisão Celular , Oligodendroglia/patologia , Ratos , Proteínas Recombinantes
15.
J Biol Chem ; 277(38): 35671-81, 2002 Sep 20.
Artigo em Inglês | MEDLINE | ID: mdl-12105206

RESUMO

Decorin, a small leucine-rich proteoglycan, is a key regulator of tumor growth by acting as an antagonist of the epidermal growth factor receptor (EGFR) tyrosine kinase. To search for cell surface receptors interacting with decorin, we generated a decorin/alkaline phosphatase chimeric protein and used it to screen a cDNA library by expression cloning. We identified two strongly reactive clones that encoded either the full-length EGFR or its ectodomain. A physiologically relevant interaction between decorin and EGFR was confirmed in the yeast two-hybrid system and further validated by experiments using EGF/EGFR interaction and transient cell transfection assays. Using a panel of deletion mutants, decorin binding was mapped to a narrow region of the EGFR within its ligand-binding L2 domain. Moreover, the central leucine-rich repeat 6 of decorin was required for interaction with the EGFR. Site-directed mutagenesis of the EGFR L2 domain showed that a cluster of residues, His(394)-Ile(402), was essential for both decorin and EGF binding. In contrast, K465, previously shown to be cross-linked to epidermal growth factor (EGF), was required for EGF but not for decorin binding. Thus, decorin binds to a discrete region of the EGFR, partially overlapping with but distinct from the EGF-binding domain. These findings could lead to the generation of protein mimetics capable of suppressing EGFR function.


Assuntos
Fator de Crescimento Epidérmico/metabolismo , Epitopos/metabolismo , Receptores ErbB/metabolismo , Proteoglicanas/metabolismo , Sequência de Aminoácidos , Animais , Sítios de Ligação , Linhagem Celular , Decorina , Fator de Crescimento Epidérmico/imunologia , Receptores ErbB/química , Receptores ErbB/genética , Proteínas da Matriz Extracelular , Humanos , Modelos Moleculares , Dados de Sequência Molecular , Mutagênese Sítio-Dirigida , Ligação Proteica , Técnicas do Sistema de Duplo-Híbrido
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA