Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Cell Mol Neurobiol ; 37(2): 183-194, 2017 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-26993510

RESUMO

Oligodendrocyte progenitor cell (OPC) migration is critical for effective myelination of the central nervous system. Not only during normal myelination but also during remyelination, the growth factors (GFs) and extracellular matrix (ECM) protein affect the OPC migration. Studies showed the altered levels of GFs and ECM in the demyelinating lesions. In our earlier studies, we have shown that the effect of platelet-derived growth factor alpha (PDGF-A) on OPC migration is dose- and time-dependent. In that we have shown that the physiological concentration (1 ng/ml) of PDGF-A was unable to induce OPC migration at transient exposure (30 min). However, the involvement of ECM in the regulation of PDGF-A mediated OPC migration was not clear. In the present study, we have used fibronectin (FN) as ECM. PDGF-A and FN have similar and overlapping intracellular signaling pathways including the extracellular regulated kinases 1 and 2 (ERK1/2). Here we demonstrate how physiological concentration of PDGF-A combines with FN to augment OPC migration in vitro. The present study is first of its kind to show the importance of the synergistic effects of PDGF-A and FN on peripheral recruitment of phosphorylated/activated ERK1/2 (pERK1/2), actin-pERK1/2 co-localization, and filopodia formation, which are essential for the enhanced OPC migration. These findings were further confirmed by ERK1/2 inhibition studies, using the pharmacological inhibitor U0126. An understanding of these complex interactions may lead to additional strategies for transplanting genetically modified OPCs to repair widespread demyelinated lesions.


Assuntos
Fibronectinas/metabolismo , Sistema de Sinalização das MAP Quinases/fisiologia , Oligodendroglia/metabolismo , Fator de Crescimento Derivado de Plaquetas/metabolismo , Pseudópodes/metabolismo , Células-Tronco/metabolismo , Animais , Butadienos/farmacologia , Movimento Celular/efeitos dos fármacos , Movimento Celular/fisiologia , Células Cultivadas , Fibronectinas/farmacologia , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Nitrilas/farmacologia , Oligodendroglia/efeitos dos fármacos , Fator de Crescimento Derivado de Plaquetas/farmacologia , Ligação Proteica/fisiologia , Pseudópodes/efeitos dos fármacos , Ratos , Células-Tronco/efeitos dos fármacos
2.
J Cell Mol Med ; 16(8): 1856-65, 2012 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-22050733

RESUMO

Multiple sclerosis (MS) is characterized by focal destruction of the white matter of the brain and spinal cord. The exact mechanisms underlying the pathophysiology of the disease are unknown. Many studies have shown that MS is predominantly an autoimmune disease with an inflammatory phase followed by a demyelinating phase. Recent studies alongside current treatment strategies, including glatiramer acetate, have revealed a potential role for brain-derived neurotrophic factor (BDNF) in MS. However, the exact role of BDNF is not fully understood. We used the experimental autoimmune encephalomyelitis (EAE) model of MS in adolescent female Lewis rats to identify the role of BDNF in disease progression. Dorsal root ganglia (DRG) and spinal cords were harvested for protein and gene expression analysis every 3 days post-disease induction (pdi) up to 15 days. We show significant increases in BDNF protein and gene expression in the DRG of EAE animals at 12 dpi, which correlates with peak neurological disability. BDNF protein expression in the spinal cord was significantly increased at 12 dpi, and maintained at 15 dpi. However, there was no significant change in mRNA levels. We show evidence for the anterograde transport of BDNF protein from the DRG to the dorsal horn of the spinal cord via the dorsal roots. Increased levels of BDNF within the DRG and spinal cord in EAE may facilitate myelin repair and neuroprotection in the CNS. The anterograde transport of DRG-derived BDNF to the spinal cord may have potential implications in facilitating central myelin repair and neuroprotection.


Assuntos
Fator Neurotrófico Derivado do Encéfalo/metabolismo , Gânglios Espinais/metabolismo , Esclerose Múltipla/metabolismo , Animais , Fator Neurotrófico Derivado do Encéfalo/genética , Avaliação da Deficiência , Encefalomielite Autoimune Experimental/metabolismo , Encefalomielite Autoimune Experimental/patologia , Ensaio de Imunoadsorção Enzimática , Feminino , Gânglios Espinais/patologia , Regulação da Expressão Gênica , Imuno-Histoquímica , Vértebras Lombares/metabolismo , Vértebras Lombares/patologia , Esclerose Múltipla/patologia , Ratos , Ratos Endogâmicos Lew
3.
Mult Scler ; 17(4): 397-403, 2011 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-21212089

RESUMO

BACKGROUND: Staphylococcus aureus may produce superantigens that can non-specifically activate CD4(+) cells to potentially target the myelin basic protein. OBJECTIVE: This study examined the association between individuals with multiple sclerosis (MS) and colonization with S. aureus harbouring superantigens. METHODS: Nasal swabs were collected from non-MS subjects and patients with MS who had not experienced a relapse in the past six months (MS stable group) and who had suffered a relapse within 30 days of study recruitment (MS exacerbation group). S. aureus was isolated from the anterior nares of participants following standard procedures and staphylococcal superantigen genes (sea, seb, and tsst-1) were detected using standard laboratory PCR techniques. RESULTS: The study enrolled 204 patients, 80 in the non-MS and MS stable groups and 44 patients in the MS exacerbation group. Overall, 27.0% of patients were colonized with S. aureus with no significant differences identified between study groups. Amongst individuals colonized with S. aureus, the prevalence of sea was significantly greater in the MS exacerbation versus non-MS study group (p < 0.05; odds ratio 7.9; 95% confidence interval 1.2-49.5). CONCLUSIONS: The ability to rapidly screen patients for the presence of S. aureus producing sea may serve as a useful marker of a potential MS exacerbation.


Assuntos
Enterotoxinas/imunologia , Esclerose Múltipla/microbiologia , Infecções Estafilocócicas/imunologia , Staphylococcus aureus/imunologia , Superantígenos/imunologia , Adulto , Feminino , Humanos , Modelos Logísticos , Masculino , Pessoa de Meia-Idade , Esclerose Múltipla/imunologia , Cavidade Nasal/imunologia , Fatores de Risco
4.
Int J Neurosci ; 120(10): 631-40, 2010 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-20942577

RESUMO

INTRODUCTION: Fatigue and cognitive deficits are common symptoms affecting patients with multiple sclerosis. METHODS: The effects of interferon beta on fatigue and cognitive deficits were assessed in 50 patients with relapsing multiple sclerosis (recruited at a single center). The pre-treatment assessments were performed on visits 1 and 2 (Months 0 and 3). Patients started treatment with subcutaneous interferon beta-1a or beta-1b, or intramuscular interferon beta-1a at Month 3, with reassessment at visits 3 and 4 (6 and 12 months, respectively). Co-primary endpoints were change in fatigue (Modified Fatigue Impact Scale) and change in cognition (Brief Repeatable Battery of Neuropsychological Tests) from pre-treatment to visits 3 and 4. Follow-up data were obtained for 40 patients. RESULTS: The pre-treatment demographic and disease characteristics did not differ between groups. Improvements in fatigue levels were reported for patients receiving subcutaneous interferon beta-1a versus patients in the intramuscular interferon beta-1a group (p = .04) and in the interferon beta-1b group (p = .09). Improvements were also reported in five out of 17 cognitive indices for all the treatment groups. CONCLUSION: The data suggest that interferon beta may reduce fatigue and cognitive deficits in patients with relapsing multiple sclerosis. Larger, randomized, and controlled studies are required to confirm our findings.


Assuntos
Transtornos Cognitivos/tratamento farmacológico , Fadiga/tratamento farmacológico , Interferon beta/administração & dosagem , Esclerose Múltipla Recidivante-Remitente/tratamento farmacológico , Adolescente , Adulto , Idoso , Transtornos Cognitivos/etiologia , Progressão da Doença , Fadiga/etiologia , Feminino , Humanos , Injeções Intramusculares , Injeções Subcutâneas , Interferon beta-1a , Interferon beta-1b , Masculino , Pessoa de Meia-Idade , Esclerose Múltipla Recidivante-Remitente/complicações , Projetos Piloto , Estudos Prospectivos , Adulto Jovem
5.
J Neurochem ; 109(5): 1285-99, 2009 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-19476544

RESUMO

The germinal matrix of human brain gives rise to oligodendrocytes and astrocytes after mid-gestation. Hemorrhage in the germinal matrix of premature infants is associated with suppressed cell proliferation. We hypothesize that soluble blood constituents have an adverse effect on the proliferation of cultured rat subventricular zone (SVZ) cells and the proliferation, migration, and differentiation of oligodendrocyte progenitor cells (OPC). Using caspase 3 activation and lactate dehydrogenase release assays, rat plasma, serum, thrombin, and kallikrein killed SVZ cells when grown in the presence (but not absence) of platelet derived growth factor. Plasma and serum killed OPC at 1:1 to 1:100 dilutions. Using a bromodeoxyuridine incorporation assay OPC proliferation was reduced by plasma, serum, thrombin and plasmin. Blood proteins also suppressed OPC migration in a concentration dependent manner. However, differentiation of OPC into myelin basic protein expressing cells was suppressed only by thrombin. We conclude that soluble blood components, particularly thrombin, have an adverse effect on maturing SVZ cells and OPC derived from newborn rat brain.


Assuntos
Diferenciação Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Ventrículos Cerebrais/citologia , Neurônios/efeitos dos fármacos , Oligodendroglia/efeitos dos fármacos , Plasma/química , Células-Tronco/efeitos dos fármacos , Animais , Animais Recém-Nascidos , Sangue , Encéfalo/citologia , Bromodesoxiuridina/metabolismo , Caspase 3/metabolismo , Morte Celular/efeitos dos fármacos , Relação Dose-Resposta a Droga , Gangliosídeos/metabolismo , Proteína Glial Fibrilar Ácida/metabolismo , L-Lactato Desidrogenase/metabolismo , Antígenos Comuns de Leucócito/metabolismo , Fosfopiruvato Hidratase/metabolismo , Ratos , Ratos Sprague-Dawley , Soro/química
6.
Neurochem Res ; 34(1): 169-81, 2009 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-18512152

RESUMO

During CNS development, oligodendrocyte progenitor (OP) cells migrate from germinal zones to presumptive white matter tracts to generate myelinating oligodendrocytes. In vitro and in vivo studies indicate that platelet-derived growth factor-A (PDGF-A) is a potent chemoattractant for OP cells and important for normal distribution throughout the developing CNS. However, PDGF-A does not localize in concentration gradients corresponding to OP migratory pathways, as would be expected for a chemoattractant to direct migration. Therefore, the mechanism by which PDGF-A regulates OP distribution remains to be clarified. Here we show that PDGF-A induces OP migration and continuous exposure to PDGF-A is not required to maintain migration. Using pharmacological inhibitors, we show that a self-sustaining extracellular-regulated-kinase signaling pathway drives OP migration for up to 72 hours after the initial PDGF stimulus. These findings indicate PDGF-A may act to mobilize OP cells that then respond to distinct directional signals to distribute appropriately within the CNS.


Assuntos
Movimento Celular/fisiologia , MAP Quinases Reguladas por Sinal Extracelular/fisiologia , Oligodendroglia/fisiologia , Fator de Crescimento Derivado de Plaquetas/fisiologia , Células-Tronco/fisiologia , Animais , Sobrevivência Celular/efeitos dos fármacos , Retroalimentação Fisiológica , Oligodendroglia/efeitos dos fármacos , Fosfatidilinositol 3-Quinases/fisiologia , Fosfolipases A2/fisiologia , Ratos , Transdução de Sinais/fisiologia , Células-Tronco/efeitos dos fármacos , Ácido alfa-Amino-3-hidroxi-5-metil-4-isoxazol Propiônico/farmacologia , Proteínas Quinases p38 Ativadas por Mitógeno/fisiologia
7.
J Mol Neurosci ; 67(4): 564-573, 2019 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-30649683

RESUMO

Oligodendrocyte progenitor cells (OPCs) originate from the sub-ventricular zone of the developing brain. They migrate and proliferate to occupy the white matter tracts of the central nervous system and transform into myelinating oligodendrocytes. Along their route of migration, OPCs are guided and controlled by several growth factors and chemokines. PDGF-A (platelet-derived growth factor), a growth factor, serves as a monogenic and mitogenic cue during the process and activates intracellular signaling pathways inside the cell. Activation of extracellular signal regulated kinase (ERK) signaling is one of the mechanisms by which PDGF-A induces the migration of OPCs. However, the mechanisms governing the PDGF-A-induced ERK-driven OPCs migration are still unclear. In the current study, we investigated further the role of PDGF-A-induced ERK signaling in OPC migration. First, we confirmed the role of PDGF-A-activated ERK signaling in OPC migration using the pharmacological inhibitor U0126, or siRNA-mediated suppression of ERK expression. Then, we demonstrated that PDGF-A-induced actin reorganization and interaction of focal adhesion kinase (FAK), Paxillin, and pERK signals are impaired in OPCs treated with the MEK inhibitor U0126. Thus, our findings demonstrated that PDGF-A induces OPC migration in an ERK-dependent mechanism via regulation of actin reorganization and FAK-Paxillin interaction.


Assuntos
Movimento Celular , Sistema de Sinalização das MAP Quinases , Células-Tronco Neurais/metabolismo , Oligodendroglia/metabolismo , Animais , Células Cultivadas , Proteína-Tirosina Quinases de Adesão Focal/metabolismo , Proteína Quinase 1 Ativada por Mitógeno/metabolismo , Proteína Quinase 3 Ativada por Mitógeno/metabolismo , Células-Tronco Neurais/fisiologia , Oligodendroglia/fisiologia , Paxilina/metabolismo , Fator de Crescimento Derivado de Plaquetas/metabolismo , Ratos
8.
Neurosci Lett ; 674: 42-48, 2018 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-29540297

RESUMO

MethylCpG binding protein-2 (MeCP2) is an epigenetic regulator and essential for brain development. MeCP2 mutations are associated with a spectrum of neuro-developmental disorders that vary depending on the patient gender, most notably Rett Syndrome. MeCP2 is essential for normal neuronal maturation, and glial cell function in the brain. Besides, its role in neurodevelopmental disorders, MeCP2 is involved in many cancers such as breast, colorectal, lung, liver, and prostate cancer. Glioma is the most lethal form of brain cancer. Studies have shown that dysfunctional epigenetic regulation plays a crucial role in glioma progression. Further, previous studies have suggested a role for MeCP2 in glioma pathogenesis. In this study, we show that MeCP2 may play a critical role in the suppression of glioma progression. Stable overexpression of MeCP2in C6 glioma cells inhibits proliferation, migration, invasion, and adhesion. Moreover, MeCP2 overexpression inhibits pERKand BDNF expression while inducing GFAP expression in C6 glioma. These findings suggest that MeCP2 may play a crucial role in suppression of glioma progression.


Assuntos
Neoplasias Encefálicas/metabolismo , Expressão Gênica , Glioma/metabolismo , Sistema de Sinalização das MAP Quinases , Proteína 2 de Ligação a Metil-CpG/metabolismo , Animais , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/patologia , Fator Neurotrófico Derivado do Encéfalo/metabolismo , Linhagem Celular Tumoral , Movimento Celular , Epigênese Genética , Proteína Glial Fibrilar Ácida/metabolismo , Glioma/genética , Glioma/patologia , Invasividade Neoplásica , Ratos
9.
Acta Neurobiol Exp (Wars) ; 78(1): 30-40, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29694339

RESUMO

avMethyl­CpG binding protein 2 (MeCP2) is an epigenetic regulator, which preferentially binds to methylated CpG dinucleotides in DNA. MeCP2 mutations have been linked to Rett syndrome, a neurodevelopmental disorder characterized by severe intellectual disability in females. Earlier studies indicated that loss of MeCP2 function in neuronal cells was the sole cause of Rett syndrome. Subsequent studies have linked MeCP2 expression in CNS glial cells to Rett syndrome pathogenesis. In this review, we have discussed the role of MeCP2 in glial subtypes, astrocytes, oligodendrocytes and microglia, and how loss of MeCP2 function in these cells has a profound influence on both glial and neuronal function.


Assuntos
Sistema Nervoso Central/citologia , Proteína 2 de Ligação a Metil-CpG/metabolismo , Neuroglia/metabolismo , Animais , Sistema Nervoso Central/patologia , Humanos , Proteína 2 de Ligação a Metil-CpG/genética , Síndrome de Rett/genética , Síndrome de Rett/patologia
10.
J Neurosci ; 22(19): 8574-85, 2002 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-12351731

RESUMO

This study takes advantage of fibroblast growth factor 2 (FGF2) knock-out mice to determine the contribution of FGF2 to the regeneration of oligodendrocytes in the adult CNS. The role of FGF2 during spontaneous remyelination was examined using two complementary mouse models of experimental demyelination. The murine hepatitis virus strain A59 (MHV-A59) model produces focal areas of spinal cord demyelination with inflammation. The cuprizone neurotoxicant model causes extensive corpus callosum demyelination without a lymphocytic cell response. In both models, FGF2 expression is upregulated in areas of demyelination in wild-type mice. Surprisingly, in both models, oligodendrocyte repopulation of demyelinated white matter was significantly increased in FGF2 -/- mice compared with wild-type mice and even surpassed the oligodendrocyte density of nonlesioned mice. This dramatic result indicated that the absence of FGF2 promoted oligodendrocyte regeneration, possibly by enhancing oligodendrocyte progenitor proliferation and/or differentiation. FGF2 -/- and +/+ mice had similar oligodendrocyte progenitor densities in normal adult CNS, as well as similar progenitor proliferation and accumulation during demyelination. To directly analyze progenitor differentiation, glial cultures from spinal cords of wild-type mice undergoing remyelination after MHV-A59 demyelination were treated for 3 d with either exogenous FGF2 or an FGF2 neutralizing antibody. Elevating FGF2 favored progenitor proliferation, whereas attenuating endogenous FGF2 activity promoted the differentiation of progenitors into oligodendrocytes. These in vitro results are consistent with enhanced progenitor differentiation in FGF2 -/- mice. These studies demonstrate that the FGF2 genotype regulates oligodendrocyte regeneration and that FGF2 appears to inhibit oligodendrocyte lineage differentiation during remyelination.


Assuntos
Doenças Desmielinizantes/fisiopatologia , Fator 2 de Crescimento de Fibroblastos/deficiência , Oligodendroglia/citologia , Animais , Anticorpos/farmacologia , Contagem de Células , Diferenciação Celular/efeitos dos fármacos , Divisão Celular/efeitos dos fármacos , Linhagem da Célula/efeitos dos fármacos , Células Cultivadas , Corpo Caloso/metabolismo , Corpo Caloso/patologia , Cuprizona , Doenças Desmielinizantes/induzido quimicamente , Doenças Desmielinizantes/genética , Modelos Animais de Doenças , Feminino , Fator 2 de Crescimento de Fibroblastos/antagonistas & inibidores , Fator 2 de Crescimento de Fibroblastos/metabolismo , Fator 2 de Crescimento de Fibroblastos/farmacologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Vírus da Hepatite Murina/patogenicidade , Bainha de Mielina/metabolismo , Regeneração Nervosa/efeitos dos fármacos , Oligodendroglia/efeitos dos fármacos , Células-Tronco/citologia , Células-Tronco/efeitos dos fármacos
11.
J Mol Neurosci ; 57(2): 176-84, 2015 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-26140854

RESUMO

Methyl CpG binding protein 2 (MeCP2) is a multifunctional protein which binds to methylated CpG, mutation of which cause a neurodevelopmental disorder, Rett syndrome. MeCP2 can function as both transcriptional activator and repressor of target gene. MeCP2 regulate gene expression in both neuron and glial cells in central nervous system (CNS). Oligodendrocytes, the myelinating cells of CNS, are required for normal functioning of neurons and are regulated by several transcription factors during their differentiation. In current study, we focused on the role of MeCP2 as transcription regulator of myelin genes in cultured rat oligodendrocytes. We have observed expression of MeCP2 at all stages of oligodendrocyte development. MeCP2 knockdown in cultured oligodendrocytes by small interference RNA (siRNA) has shown increase in myelin genes (myelin basic protein (MBP), proteolipid protein (PLP), myelin oligodendrocyte glycoprotein (MOG), and myelin-associated oligodendrocyte basic protein (MOBP)), neurotrophin (brain-derived neurotrophic factor (BDNF)), and transcriptional regulator (YY1) transcripts level, which are involved in regulation of oligodendrocyte differentiation and myelination. Further, we also found that protein levels of MBP, PLP, DM-20, and BDNF also significantly upregulated in MeCP2 knockdown oligodendrocytes. Our study suggests that the MeCP2 acts as a negative regulator of myelin protein expression.


Assuntos
Proteína 2 de Ligação a Metil-CpG/metabolismo , Proteína Básica da Mielina/metabolismo , Proteína Proteolipídica de Mielina/metabolismo , Glicoproteína Mielina-Oligodendrócito/metabolismo , Oligodendroglia/metabolismo , Animais , Fator Neurotrófico Derivado do Encéfalo/genética , Fator Neurotrófico Derivado do Encéfalo/metabolismo , Células Cultivadas , Córtex Cerebral/citologia , Córtex Cerebral/crescimento & desenvolvimento , Córtex Cerebral/metabolismo , Proteína 2 de Ligação a Metil-CpG/genética , Proteína Básica da Mielina/genética , Proteína Proteolipídica de Mielina/genética , Glicoproteína Mielina-Oligodendrócito/genética , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Ratos , Fator de Transcrição YY1/genética , Fator de Transcrição YY1/metabolismo
12.
Exp Neurol ; 236(2): 259-67, 2012 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-22554866

RESUMO

Cell migration is an indispensable aspect of tissue patterning during embryonic development. Oligodendrocytes, the myelinating cells of the central nervous system, migrate significantly during development of the brain. Several growth factors have been identified as being critical regulators of oligodendrocyte progenitor migration, including platelet derived growth factor-A (PDGFA), and fibroblast growth factor-2 (FGF2). Further, the chemokine CXCL1 has been shown to play a critical role in regulating the dispersal of oligodendrocyte progenitors during development, although the mechanisms underlying this regulation are unknown. Previous studies have also shown that calcium flux is required for oligodendrocyte progenitor migration. CXCL1 induces calcium flux in cells; therefore, we hypothesized that CXCL1 inhibition of oligodendrocyte progenitor migration is regulated via changes in intracellular calcium flux. The current study shows that CXCL1 inhibition of oligodendrocyte progenitor migration is independent of calcium signaling. Further, we show that CXCL1 inhibition of oligodendrocyte progenitor migration is specific to PDGFA induced migration. Finally, we show that CXCL1 inhibition of oligodendrocyte progenitor migration is independent of activation of the cell cycle. Our results provide intriguing results relevant to specific aspects of patterning of white matter tracts in the central nervous system, and may further the understanding of tissue remodeling seen during disease-related processes.


Assuntos
Sinalização do Cálcio/fisiologia , Inibição de Migração Celular/fisiologia , Quimiocina CXCL1/fisiologia , Oligodendroglia/fisiologia , Células-Tronco/fisiologia , Animais , Animais Recém-Nascidos , Cálcio/química , Cálcio/metabolismo , Cálcio/fisiologia , Células Cultivadas , Líquido Intracelular/metabolismo , Líquido Intracelular/fisiologia , Oligodendroglia/citologia , Oligodendroglia/metabolismo , Fator de Crescimento Derivado de Plaquetas/fisiologia , Ratos , Células-Tronco/citologia , Células-Tronco/metabolismo
13.
Eur J Cell Biol ; 90(8): 649-56, 2011 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-21616555

RESUMO

Oligodendrocytes are myelinating cells of the CNS that originate as progenitor cells (OP) in discrete areas of the developing brain. During brain development, OP migrate significant distances prior to proliferating and myelinating the axons of the putative white matter tracts. Growth factors play a major regulatory role in the behavior of OP. Specifically, platelet-derived growth factor A (PDGF-A) and fibroblast growth factor 2 (FGF2) are two of the most well characterized regulators of OP development. Both growth factors interact with tyrosine kinase receptors, activating various intracellular signaling pathways. The current study advances our earlier research by comparing the effects of both PDGF-A and FGF2 on OP migration. Our results show that activation of ERK is required for OP migration. These findings correlate well with our previous demonstration of the ERK pathway mediating PDGF-A induced OP migration. We also demonstrate the significance of threshold levels of growth factors and temporal regulation for OP migration. In addition, ERK activation alone is not sufficient to induce OP migration. The current research supports the involvement of the non-ERK mediated signaling pathway in OP migration.


Assuntos
Movimento Celular/efeitos dos fármacos , Fator 2 de Crescimento de Fibroblastos/farmacologia , Oligodendroglia/metabolismo , Fator de Crescimento Derivado de Plaquetas/farmacologia , Células-Tronco/metabolismo , Animais , Western Blotting , Encéfalo/embriologia , Butadienos/farmacologia , Proliferação de Células/efeitos dos fármacos , Células Cultivadas , Inibidores Enzimáticos/farmacologia , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Fator 2 de Crescimento de Fibroblastos/metabolismo , MAP Quinase Quinase 1/antagonistas & inibidores , MAP Quinase Quinase 2/antagonistas & inibidores , Nitrilas/farmacologia , Fator de Crescimento Derivado de Plaquetas/metabolismo , Ratos , Receptores Proteína Tirosina Quinases/metabolismo , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/fisiologia , Células-Tronco/efeitos dos fármacos
14.
Neuroreport ; 21(14): 917-21, 2010 Oct 06.
Artigo em Inglês | MEDLINE | ID: mdl-20697302

RESUMO

Myelination is critical for normal functioning of mammalian central nervous system. Central nervous system myelin is created and maintained by oligodendrocytes. Protein expression patterns change as the oligodendrocyte progenitors differentiate into myelinating oligodendrocytes. Several proteins, including the cell surface proteoglycan NG2, proteolipid protein, myelin basic protein, and myelin-associated glycoprotein are critical for normal myelination. The molecular regulation of myelination is for the most part unknown, although several transcription factors have been identified as regulating myelin protein expression. We have identified a known transcriptional regulator, methyl-CpG-binding protein 2, as regulating myelin specific gene expression in a transgenic mouse. Our findings show a potential role for myelin in the pathophysiology of methyl-CpG-binding protein 2 mutation-associated disorders.


Assuntos
Deficiências do Desenvolvimento/genética , Regulação da Expressão Gênica no Desenvolvimento/genética , Proteína 2 de Ligação a Metil-CpG/genética , Bainha de Mielina/genética , Animais , Animais Recém-Nascidos , Química Encefálica/genética , Células Cultivadas , Deficiências do Desenvolvimento/metabolismo , Feminino , Proteína 2 de Ligação a Metil-CpG/biossíntese , Proteína 2 de Ligação a Metil-CpG/deficiência , Proteína 2 de Ligação a Metil-CpG/isolamento & purificação , Camundongos , Camundongos Knockout , Camundongos Transgênicos , Bainha de Mielina/metabolismo , Fibras Nervosas Mielinizadas/metabolismo , Fibras Nervosas Mielinizadas/patologia , Ratos , Ratos Sprague-Dawley
15.
J Neurosci Methods ; 187(1): 59-66, 2010 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-20043948

RESUMO

Histological analysis of bone encased tissue is severely hampered by technical difficulties associated with sectioning calcified tissue. Cryosectioning of bone is possible but requires significant technical adaptation and expensive materials and is often time-consuming. Some decalcifying reagents in common use result in successful cryosectioning in less time but the integrity of the soft tissue of the spinal column is often compromised during processing. This can result in significant loss of cellular detail. In order to find a method that would allow cryosectioning of the bone without loss of structural integrity of the underlying soft tissue we assessed the efficacy of four different decalcifying reagents with respect to their effects on the cellular structure of the myelin of the grey and white matter of the spinal cord. The antigenic integrity of the spinal cord white matter was evaluated using tissue structural integrity and quality of myelin basic protein immunostaining. The result of this research shows that 6% TCA not only decalcifies intact spinal column suitably for cryosectioning but does so without compromising the antigenic integrity of the tissue. The ease of application, speed of processing and a favorable cost-effective profile were secondary benefits noted with the use of the 6% TCA decalcifying solution. The ability to utilize a decalcifying solution that allows for both histomorphometry and immunohistochemistry in the same spinal column segment represents a novel technique that will provide new insights into pathophysiological aspects and therapeutic approaches ispinal cord damage or disease.


Assuntos
Crioultramicrotomia/métodos , Técnica de Descalcificação/métodos , Medula Espinal/citologia , Coluna Vertebral , Ácido Tricloroacético , Animais , Crioultramicrotomia/economia , Técnica de Descalcificação/economia , Feminino , Imuno-Histoquímica , Hibridização In Situ , Indicadores e Reagentes/economia , Proteína Básica da Mielina/metabolismo , Bainha de Mielina , Fibras Nervosas Mielinizadas , Fibras Nervosas Amielínicas , Vias Neurais/citologia , Sistema Nervoso Periférico/citologia , Ratos , Ratos Sprague-Dawley , Fatores de Tempo , Ácido Tricloroacético/economia
16.
Magn Reson Med ; 57(1): 164-71, 2007 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-17191231

RESUMO

Neural stem cell (NSC) transplantation has been shown to attenuate the severity of experimental autoimmune encephalomyelitis (EAE), an animal model of multiple sclerosis (MS). Central to the future success of NSC transplantation in MS is the ability of transplanted cells to migrate from the site of transplantation to relevant foci of disease. Using magnetically labeled mouse neurospheres and human embryonic stem cell (hESC)-derived neurospheres, we applied serial magnetic resonance imaging (MRI) to assess the biodynamics of transplanted cell migration in a chronic mouse EAE model. Magnetic labeling did not affect the in vitro and in vivo characteristics of cells as multipotential precursors. Cell migration occurred along white matter (WM) tracts (especially the corpus callosum (CC), fimbria, and internal capsule), predominantly early in the acute phase of disease, and in an asymmetric manner. The distance of cell migration correlated well with clinical severity of disease and the number of microglia in the WM tracts, supporting the notion that inflammatory signals promote transplanted cell migration. This study shows for the first time that hESC-derived neural precursors also respond to tissue signals in an MS model, similarly to rodent cells. The results are directly relevant for designing and optimizing cell therapies for MS, and achieving a better understanding of in vivo cell dynamics and cell-tissue interactions.


Assuntos
Encefalomielite Autoimune Experimental/terapia , Imageamento por Ressonância Magnética , Neurônios/transplante , Esferoides Celulares/transplante , Animais , Linhagem da Célula , Movimento Celular , Células Cultivadas , Modelos Animais de Doenças , Encefalomielite Autoimune Experimental/induzido quimicamente , Encefalomielite Autoimune Experimental/fisiopatologia , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Neurônios/citologia , Neurônios/fisiologia , Índice de Gravidade de Doença , Esferoides Celulares/citologia , Transplante de Células-Tronco , Transplante Heterólogo
17.
Neoplasia ; 8(3): 207-13, 2006 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-16611414

RESUMO

We have developed a noninvasive magnetic resonance imaging (MRI) assay to characterize human umbilical vein endothelial cell (HUVEC) motility, invasion, and network formation in response to the presence of cancer cells. HUVECs were labeled with a superparamagnetic iron oxide T(2) contrast agent and cocultured with MDA-MB-231 breast cancer cells in the presence of an extracellular matrix (ECM) gel. Invasion into the ECM gel by HUVECs in response to paracrine factors secreted by MDA-MB-231 cancer cells, as well as network formation by HUVECs, was easily tracked with MRI. The invasive behavior of HUVECs was not observed in the absence of cancer cells. This noninvasive assay used to characterize the response of endothelial cells (ECs) can be used to understand the role of proangiogenic or antiangiogenic stimuli, and to study the interactions between ECs and other disease-specific cells in pathologies with aberrant angiogenesis, such as retinopathy and arthritis.


Assuntos
Adenocarcinoma/patologia , Neoplasias da Mama/patologia , Células Endoteliais/citologia , Imageamento por Ressonância Magnética/métodos , Neovascularização Patológica/patologia , Comunicação Parácrina/fisiologia , Adenocarcinoma/metabolismo , Proteínas Angiogênicas/metabolismo , Neoplasias da Mama/metabolismo , Linhagem Celular Tumoral/citologia , Linhagem Celular Tumoral/metabolismo , Movimento Celular , Técnicas de Cocultura , Meios de Contraste , Dextranos , Endotélio Vascular/citologia , Matriz Extracelular/fisiologia , Feminino , Óxido Ferroso-Férrico , Géis , Substâncias de Crescimento/metabolismo , Humanos , Ferro , Nanopartículas de Magnetita , Microscopia de Fluorescência , Microscopia de Contraste de Fase , Invasividade Neoplásica , Proteínas de Neoplasias/metabolismo , Óxidos , Polilisina , Veias Umbilicais/citologia
18.
Radiology ; 238(1): 200-10, 2006 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-16373768

RESUMO

PURPOSE: To assess renal glomerular homing of intravenously injected superparamagnetic iron oxide (SPIO)-labeled mesenchymal stem cells (MSCs) at in vivo and ex vivo magnetic resonance (MR) imaging in an experimental rat model of mesangiolysis. MATERIALS AND METHODS: Animal procedures were performed in accordance with protocols approved by Institutional Animal Care and Use Committee. Fourteen rats were divided into two groups: one pathologic (n = 10), with persistent mesangiolysis following simultaneous injection of OX-7 monoclonal antibody and puromycin aminonucleoside in which 10(7) SPIO- and DiI-labeled MSCs were injected, and one control (n = 4). In vivo and ex vivo MR imaging examinations were performed with 4.7- and 9.4-T spectrometers, respectively, and T2*-weighted sequences. In vivo signal intensity variations were measured in the liver and kidney before and 6 days after MSC injection. Intrarenal signal intensity variations were correlated with histopathologic data by means of colocalization of DiI fluorescence, alpha-actin, and Prussian blue stain-positive cells. Histologic differences between the glomerular homing of MSCs in different kidney portions were correlated to the areas of MR signal intensity decrease with nonparametric statistical tests. RESULTS: On in vivo images, signal intensity measurements of pathologic kidneys following MSC injection did not show any signal intensity decrease (P = .7), whereas a 34% +/- 14 (mean +/- standard deviation) signal intensity decrease was observed in the liver (P < .01), where a substantial number of labeled cells were trapped. On ex vivo images, pathologic kidneys showed focal cortical (glomerular) areas of signal intensity loss, which was absent in controls. The areas of low signal intensity correlated well with alpha-actin and Prussian blue stain- and DiI-positive areas (P < .01), which indicates that MSCs specifically home to injured tissue. No MSCs were detected in the kidneys of control animals. CONCLUSION: Intravenously injected MSCs specifically home to focal areas of glomerular damage and can be detected at ex vivo MR imaging.


Assuntos
Nefropatias/terapia , Imageamento por Ressonância Magnética/métodos , Transplante de Células-Tronco Mesenquimais/métodos , Animais , Técnicas de Cultura de Células , Meios de Contraste , Dextranos , Óxido Ferroso-Férrico , Processamento de Imagem Assistida por Computador , Imuno-Histoquímica , Ferro , Nefropatias/patologia , Glomérulos Renais/citologia , Nanopartículas de Magnetita , Masculino , Óxidos , Ratos , Ratos Endogâmicos Lew , Coloração e Rotulagem
19.
J Neurobiol ; 54(3): 457-72, 2003 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-12532397

RESUMO

Acute demyelination of adult CNS, resulting from trauma or disease, is initially followed by remyelination. However, chronic lesions with subsequent functional impairment result from eventual failure of the remyelination process, as seen in multiple sclerosis. Studies using animal models of successful remyelination delineate a progression of events facilitating remyelination. A universal feature of this repair process is extensive proliferation of oligodendrocyte progenitor cells (OPs) in response to demyelination. To investigate signals that regulate OP proliferation in response to demyelination we used murine hepatitis virus-A59 (MHV-A59) infection of adult mice to induce focal demyelination throughout the spinal cord followed by spontaneous remyelination. We cultured glial cells directly from demyelinating and remyelinating spinal cords using conditions that maintain the dramatically enhanced OP proliferative response prior to CNS remyelination. We identify PDGF and FGF2 as significant mitogens regulating this proliferative response. Furthermore, we demonstrate endogenous PDGF and FGF2 activity in these glial cultures isolated from demyelinated CNS tissue. These findings correlate well with our previous demonstration of increased in vivo expression of PDGF and FGF2 ligand and corresponding receptors in MHV-A59 lesions. Together these studies support the potential of these pathways to function in vivo as critical factors in regulating remyelination.


Assuntos
Doenças Desmielinizantes/metabolismo , Fator 2 de Crescimento de Fibroblastos/metabolismo , Oligodendroglia/metabolismo , Fator de Crescimento Derivado de Plaquetas/metabolismo , Células-Tronco/metabolismo , Animais , Animais Recém-Nascidos , Anticorpos/farmacologia , Antígenos/metabolismo , Diferenciação Celular/efeitos dos fármacos , Divisão Celular/efeitos dos fármacos , Linhagem da Célula , Doenças Desmielinizantes/patologia , Doenças Desmielinizantes/virologia , Interações Medicamentosas , Feminino , Imunoquímica , Hibridização In Situ , Técnicas In Vitro , Camundongos , Camundongos Endogâmicos C57BL , Vírus da Hepatite Murina/patogenicidade , Regeneração Nervosa/efeitos dos fármacos , Oligorribonucleotídeos Antissenso , Proteoglicanas/metabolismo , Medula Espinal/metabolismo , Fatores de Tempo , Tirfostinas/farmacologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA