Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 112
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Brain Behav Immun ; 77: 110-126, 2019 03.
Artigo em Inglês | MEDLINE | ID: mdl-30582962

RESUMO

Remyelination is an endogenous process by which functional recovery of damaged neurons is achieved by reinstating the myelin sheath around axons. Remyelination has been documented in multiple sclerosis (MS) lesions and experimental models, although it is often incomplete or fails to affect the integrity of the axon, thereby leading to progressive disability. Microglia play a crucial role in the clearance of the myelin debris produced by demyelination and in inflammation-dependent OPC activation, two processes necessary for remyelination to occur. We show here that following corpus callosum demyelination in the TMEV-IDD viral murine model of MS, there is spontaneous and partial remyelination that involves a temporal discordance between OPC mobilization and microglia activation. Pharmacological treatment with the endocannabinoid 2-AG enhances the clearance of myelin debris by microglia and OPC differentiation, resulting in complete remyelination and a thickening of the myelin sheath. These results highlight the importance of targeting microglia during the repair processes in order to enhance remyelination.


Assuntos
Ácidos Araquidônicos/farmacologia , Endocanabinoides/farmacologia , Glicerídeos/farmacologia , Microglia/efeitos dos fármacos , Remielinização/efeitos dos fármacos , Animais , Ácidos Araquidônicos/metabolismo , Axônios/metabolismo , Diferenciação Celular/fisiologia , Corpo Caloso/patologia , Corpo Caloso/fisiologia , Doenças Desmielinizantes/fisiopatologia , Modelos Animais de Doenças , Endocanabinoides/metabolismo , Feminino , Glicerídeos/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos , Microglia/metabolismo , Esclerose Múltipla/metabolismo , Esclerose Múltipla/fisiopatologia , Bainha de Mielina/metabolismo , Células Precursoras de Oligodendrócitos/fisiologia , Oligodendroglia/metabolismo , Theilovirus/patogenicidade
2.
Eur J Neurol ; 25(4): 651-658, 2018 04.
Artigo em Inglês | MEDLINE | ID: mdl-29316044

RESUMO

BACKGROUND AND PURPOSE: Genetic research in multiple sclerosis (MS) mostly compares patients with MS with healthy controls, but does not differentiate between MS disease courses. We compared peripheral blood gene expression patterns between extremes of MS phenotypes, i.e. patients with mild relapsing-remitting MS (mRRMS) and primary progressive MS (PPMS). METHODS: We analyzed global gene expression profiles of peripheral blood samples of age- and gender-matched patients with mRRMS and PPMS. Detailed bioinformatic and gene set enrichment analysis, pathway and principle component analyses were used to identify differentially expressed genes and pathways. RESULTS: A total of 84 genes were significantly deregulated between the groups. Of those, 19 had been previously reported to be deregulated in patients with MS as compared with healthy controls, including major histocompatibility complex, interferon receptor 2 and interleukin 6 receptor. Detailed molecular pathway analysis revealed significant up-regulation of antigen processing and presentation, leukocyte transendothelial migration, nucleotide-binding oligomerization domain-like receptor signaling, chemokine signaling and down-regulation of RNA transport, spliceosome and aminoacyl-tRNA biosynthesis pathways in PPMS compared with mRRMS. CONCLUSION: Our analyses show significant differences between mRRMS and PPMS gene expression. Surprisingly, the differentially expressed genes were mostly involved in immunological and inflammatory pathways, suggesting that the difference in MS phenotypes is caused primarily by a difference in immune responses. It should be kept in mind that our analyses were in peripheral blood only, and that the observed differences in inflammatory pathways may be a substrate of the analysed tissue. Further research into gene expression differences between disease courses including analyses in central nervous system tissue is warranted.


Assuntos
Perfilação da Expressão Gênica , Esclerose Múltipla Crônica Progressiva/genética , Esclerose Múltipla Recidivante-Remitente/genética , Estudos de Coortes , Biologia Computacional , Bases de Dados Genéticas , Feminino , Expressão Gênica , Humanos , Masculino , Análise em Microsséries , Pessoa de Meia-Idade , Transdução de Sinais/genética
3.
J Neurol Sci ; 297(1-2): 76-81, 2010 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-20708201

RESUMO

Abnormally decreased deep gray matter (GM) signal intensity on T2-weighted MRI (T2 hypointensity) is associated with brain atrophy and disability progression in patients with multiple sclerosis (MS) and is believed to represent excessive iron deposition. We investigated the time course of deep GM T2 hypointensity and its relationship with disability at 3T in 8 stable relapsing-remitting (RR) MS patients treated with minocycline over 3years. MRI and disability measurements were compared at baseline, 6, 12, 24, and 36months. Grand mean deep GM T2 hypointensity was negatively correlated with EDSS over time (r=-0.94, P=0.02). This correlation was strongest in the head of caudate (r=-0.95, P=0.01) and putamen (r=-0.89, P=0.04). Additionally, baseline grand mean deep GM T2 hypointensity appears to predict third year EDSS (r=-0.72, P=0.04). These results suggest that iron associated deep GM injury correlates with patient disability in stable RRMS. Measurements of deep GM T2 hypointensity at high field MRI may prove to be useful in monitoring individuals with MS. Further studies are required to confirm these results in a large sample and to determine if T2 hypointensity changes in clinically active MS patients.


Assuntos
Encéfalo/patologia , Pessoas com Deficiência , Esclerose Múltipla Recidivante-Remitente/complicações , Esclerose Múltipla Recidivante-Remitente/patologia , Fibras Nervosas Amielínicas/patologia , Adulto , Análise de Variância , Antibacterianos/uso terapêutico , Atrofia/patologia , Avaliação da Deficiência , Feminino , Humanos , Estudos Longitudinais , Imageamento por Ressonância Magnética/métodos , Masculino , Pessoa de Meia-Idade , Minociclina/uso terapêutico , Esclerose Múltipla Recidivante-Remitente/tratamento farmacológico , Projetos Piloto , Índice de Gravidade de Doença , Estatística como Assunto , Fatores de Tempo
4.
Mult Scler ; 15(10): 1183-94, 2009 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-19776092

RESUMO

Minocycline is proposed as an add-on therapy to improve the efficacy of glatiramer acetate in relapsing-remitting multiple sclerosis. The effect of minocycline plus glatiramer acetate was evaluated in this double-blind, placebo-controlled study by determining the total number of T1 gadolinium-enhanced lesions at months 8 and 9 in patients who were starting glatiramer acetate and had at least one T1 gadolinium-enhanced lesion on screening magnetic resonance imaging. Forty-four participants were randomized to either minocycline 100 mg twice daily or matching placebo for 9 months as add-on therapy. They were assessed at screening and months 1, 3, 6, 8 and 9. Forty participants completed the study. Compared with glatiramer acetate/placebo, glatiramer acetate/minocycline reduced the total number of T1 gadolinium-enhanced lesions by 63% (mean 1.47 versus 2.95; p = 0.08), the total number of new and enlarging T2 lesions by 65% (mean 1.84 versus 5.14; p = 0.06), and the total T2 disease burden (p = 0.10). A higher number of gadolinium-enhanced lesions were present in the glatiramer acetate/minocycline group at baseline; this was incorporated into the analysis of the primary endpoint but makes interpretation of the data more challenging. The risk of relapse tended to be lower in the combination group (0.19 versus 0.41; p = NS). Treatment was safe and well tolerated. We conclude that efficacy endpoints showed a consistent trend favoring combination treatment. As minocycline is a relatively safe oral therapy, further study of this combination is warranted in relapsing-remitting multiple sclerosis.


Assuntos
Adjuvantes Imunológicos/uso terapêutico , Antibacterianos/uso terapêutico , Minociclina/uso terapêutico , Esclerose Múltipla Recidivante-Remitente/tratamento farmacológico , Peptídeos/uso terapêutico , Adjuvantes Imunológicos/efeitos adversos , Adulto , Antibacterianos/efeitos adversos , Encéfalo/patologia , Método Duplo-Cego , Quimioterapia Combinada , Feminino , Gadolínio , Acetato de Glatiramer , Humanos , Imageamento por Ressonância Magnética , Masculino , Pessoa de Meia-Idade , Minociclina/efeitos adversos , Esclerose Múltipla Recidivante-Remitente/diagnóstico , Peptídeos/efeitos adversos , Resultado do Tratamento , Adulto Jovem
5.
Neurology ; 68(22 Suppl 3): S32-7; discussion S43-54, 2007 May 29.
Artigo em Inglês | MEDLINE | ID: mdl-17548566

RESUMO

Activated T cells, particularly those of the T-helper (Th) 1 subset, have the capacity to kill neurons. Strategies for preventing such damage may include deviation of activated T cells into the Th2 subset (e.g., via use of glatiramer acetate), alteration of functional properties of Th1 cells (e.g., through use of interferon [INF]-beta or IV immunoglobulin), and inhibition of activated cell migration into the CNS (e.g., by employing INF-beta or natalizumab). Matrix metalloproteinase-9 (MMP-9) also causes neuron death in neurotoxicity models, and examination of medications with MMP inhibitory activity indicates that minocycline is capable of preventing such damage. Minocycline also has other properties relevant to conferring neuroprotection, such as inhibition of microglial activity and apoptosis pathways. In a small pilot study in patients with relapsing-remitting multiple sclerosis, minocycline treatment produced favorable outcomes in terms of gadolinium-enhancing lesions and clinical course. Further studies are needed to establish whether experimental neuroprotection strategies involving these mechanisms may be translated into preventing neurodegeneration in multiple sclerosis.


Assuntos
Modelos Animais de Doenças , Esclerose Múltipla/imunologia , Esclerose Múltipla/prevenção & controle , Fármacos Neuroprotetores/uso terapêutico , Animais , Morte Celular/efeitos dos fármacos , Morte Celular/imunologia , Humanos , Esclerose Múltipla/tratamento farmacológico , Esclerose Múltipla/fisiopatologia , Fármacos Neuroprotetores/farmacologia
6.
Mult Scler ; 13(4): 517-26, 2007 May.
Artigo em Inglês | MEDLINE | ID: mdl-17463074

RESUMO

Minocycline has immunomodulatory and neuroprotective activities in vitro and in an animal model of multiple sclerosis (MS). We have previously reported that minocycline decreased gadolinium-enhancing activity over six months in a small trial of patients with active relapsing-remitting MS (RRMS). Here we report the impact of oral minocycline on clinical and magnetic resonance imaging (MRI) outcomes and serum immune molecules in this cohort over 24 months of open-label minocycline treatment. Despite a moderately high pretreatment annualized relapse rate (1.3/year pre-enrolment; 1.2/year during a three-month baseline period) prior to treatment, no relapses occurred between months 6 and 24. Also, despite very active MRI activity pretreatment (19/40 scans had gadolinium-enhancing activity during a three-month run-in), the only patient with gadolinium-enhancing lesions on MRI at 12 and 24 months was on half-dose minocycline. Levels of the p40 subunit of interleukin (IL)-12, which at high levels might antagonize the proinflammatory IL-12 receptor, were elevated over 18 months of treatment, as were levels of soluble vascular cell adhesion molecule-1. The activity of matrix metalloproteinase-9 was decreased by treatment. Thus, clinical and MRI outcomes are supported by systemic immunological changes and call for further investigation of minocycline in MS.


Assuntos
Minociclina/uso terapêutico , Esclerose Múltipla/tratamento farmacológico , Esclerose Múltipla/imunologia , Adulto , Citocinas/sangue , Feminino , Seguimentos , Humanos , Fatores Imunológicos/uso terapêutico , Imageamento por Ressonância Magnética , Masculino , Esclerose Múltipla/patologia , Fármacos Neuroprotetores/uso terapêutico , Projetos Piloto
7.
Cell Death Differ ; 13(7): 1203-12, 2006 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-16273078

RESUMO

Cyclin-dependent kinase-5 (Cdk5) is required for neuronal survival, but its targets in the apoptotic pathways remain unknown. Here, we show that Cdk5 kinase activity prevents neuronal apoptosis through the upregulation of Bcl-2. Treatment of SH-SY5Y cells with retinoid acid (RA) and brain-derived neurotrophic factor (BDNF) generates differentiated neuron-like cells. DNA damage triggers apoptosis in the undifferentiated cells through mitochondrial pathway; however, RA/BDNF treatment results in Bcl-2 upregulation and inhibition of the mitochondrial pathway in the differentiated cells. RA/BDNF treatment activates Cdk5-mediated PI3K/Akt and ERK pathways. Inhibition of Cdk5 inhibits PI3K/Akt and ERK phosphorylation and Bcl-2 expression, and thus sensitizes the differentiated cells to DNA-damage. Inhibition of ERK, but not PI3K/Akt, abrogates Cdk5-medidated Bcl-2 upregulation and the protection of the differentiated cells. This study suggests that ERK-mediated Bcl-2 upregulation contributes to BDNF-induced Cdk5-mediated neuronal survival.


Assuntos
Apoptose/efeitos dos fármacos , Quinase 5 Dependente de Ciclina/metabolismo , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Neurônios/efeitos dos fármacos , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , Western Blotting , Fator Neurotrófico Derivado do Encéfalo/farmacologia , Diferenciação Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Células Cultivadas , Citometria de Fluxo , Humanos , Modelos Biológicos , Neurônios/citologia , Neurônios/metabolismo , Fosfatidilinositol 3-Quinases/metabolismo , Fosforilação/efeitos dos fármacos , Plasmídeos/genética , Proteínas Proto-Oncogênicas c-akt/metabolismo , Proteínas Proto-Oncogênicas c-bcl-2/genética , RNA Interferente Pequeno/genética , Transdução de Sinais/efeitos dos fármacos , Transfecção , Tretinoína/farmacologia , Regulação para Cima/efeitos dos fármacos
8.
J Neurochem ; 89(5): 1092-100, 2004 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-15147501

RESUMO

The prevailing view of the astrocytic response to injury is that reactive astrocytes impede the regenerative process by forming scar tissue. As the levels of many cytokines dramatically increase following CNS insult and as this increase in cytokine expression precedes the production of the glial scar, a long-standing view has been that cytokines diminish neuronal survival and regeneration by stimulating the formation of astrogliotic scar tissue. However, there is a wealth of data indicating that cytokines "activate" astrocytes, and that cytokine-stimulated astrocytes can promote the recovery of CNS function. Supporting evidence demonstrates that cytokine-activated astrocytes produce energy substrates and trophic factors for neurons and oligodendrocytes, act as free radical and excess glutamate scavengers, actively restore the blood-brain barrier, promote neovascularization, restore CNS ionic homeostasis, promote remyelination and also stimulate neurogenesis from neural stem cells. Accordingly, a re-assessment of cytokine-activated astrocytes is necessary. Here, we review studies that promote the thesis that cytokines elicit potent neuroprotective and regenerative responses from astrocytes.


Assuntos
Astrócitos/fisiologia , Citocinas/fisiologia , Regeneração Nervosa/fisiologia , Animais , Astrócitos/efeitos dos fármacos , Sistema Nervoso Central/lesões , Sistema Nervoso Central/metabolismo , Citocinas/farmacologia , Gliose/metabolismo , Humanos
9.
Int MS J ; 10(4): 122-30, 2003 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-14977489

RESUMO

It has become evident that multiple sclerosis (MS) has significant neurodegenerative components. An increasing number of reports show neuronal and axonal damage in MS patients and experimental allergic encephalomyelitis (EAE) in an animal model of MS. The mechanisms behind this neurodegeneration are unknown, but evidence suggests immune-mediated damage. In this review we analyse the findings of immune-mediated injury in MS and focus on axonal and neuronal injury. The potential neuroprotective role of some currently available MS drug treatments is also discussed.


Assuntos
Esclerose Múltipla/complicações , Esclerose Múltipla/tratamento farmacológico , Degeneração Neural/etiologia , Degeneração Neural/prevenção & controle , Fármacos Neuroprotetores/uso terapêutico , Animais , Autoimunidade , Humanos , Esclerose Múltipla/fisiopatologia
10.
Mult Scler ; 8(4): 299-306, 2002 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-12166500

RESUMO

The efficacy of glatiramer acetate in multiple sclerosis (MS) is thought to involve the production of Th2 regulatory lymphocytes that secrete anti-inflammatory cytokines; however, other mechanisms cannot be excluded Given that activated T lymphocytes infiltrate into the CNS and become in dose proximity to microglia, we evaluated whether glatiramer acetate affects the potential interaction between T cells and microglia. We report that the co-culture of activated T lymphocytes with microglia led to the induction of several cytokines, and that these were reduced by glatiramer acetate treatment Morphological transformation of bipolar/ramified microglia into an activated ameboid form was attenuated by glatiramer acetate. These results reveal a novel mechanism for glatiramer acetate: the impairment of activated T cells to effectively interact with microglia to produce cytokines. The net result of a non-inflammatory milieu within the CNS, in spite of T cell infiltration, may help account for the amelioration of disease activity in MS patients on glatiramer acetate therapy.


Assuntos
Comunicação Celular/efeitos dos fármacos , Imunossupressores/farmacologia , Microglia/citologia , Esclerose Múltipla/tratamento farmacológico , Peptídeos/farmacologia , Linfócitos T/citologia , Adulto , Adesão Celular/efeitos dos fármacos , Adesão Celular/imunologia , Comunicação Celular/imunologia , Células Cultivadas , Técnicas de Cocultura , Citocinas/metabolismo , Acetato de Glatiramer , Humanos , Macrófagos/citologia , Esclerose Múltipla/imunologia , Linfócitos T/metabolismo
11.
Glia ; 36(3): 281-94, 2001 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-11746766

RESUMO

Oligodendrocyte (OL) death occurs in many disorders of the CNS, including multiple sclerosis and brain trauma. Factors reported to induce OL death include deprivation of growth factors, elevation of cytokines, oxidative stress, and glutamate excitotoxicity. Because astrocytes produce a large amount of growth factors and antioxidants and are a major source of glutamate uptake, we tested the hypothesis that astrocytes may have a protective role for OL survival. We report that when OLs from the adult mouse brain were initiated into tissue culture, DNA fragmentation and chromatin condensation resulted, indicative of apoptosis. OL death was significantly reduced in coculture with astrocytes, but not with fibroblasts, which provided a similar monolayer of cells as astrocytes. The protection of OL demise by astrocytes was not reproduced by its conditioned medium and was not accounted for by several neurotrophic factors. In contrast, interference with the alpha(6) integrin subunit, but not the alpha(1), alpha(2), alpha(3), alpha(4), alpha(5), or alpha(v) integrin chains, negated astrocyte protection of OLs. Furthermore, a function-blocking antibody to alpha(6)beta(1) integrin reduced the ability of astrocytes to promote OL survival. The extracellular matrix ligand for alpha(6)beta(1) is laminin, which is expressed by astrocytes. Significantly, neutralizing antibodies to laminin-2 and laminin-5 inhibited the astrocyte mediation of OL survival. These results implicate astrocytes in promoting OL survival through a mechanism involving the interaction of alpha(6)beta(1) integrin on OLs with laminin on astrocytes. Enhancing this interaction may provide for OL survival in neurological injury.


Assuntos
Antígenos CD/metabolismo , Astrócitos/metabolismo , Comunicação Celular/fisiologia , Morte Celular/fisiologia , Doenças do Sistema Nervoso Central/metabolismo , Laminina/metabolismo , Oligodendroglia/metabolismo , Animais , Antígenos CD/imunologia , Sobrevivência Celular/fisiologia , Células Cultivadas/citologia , Células Cultivadas/metabolismo , Doenças do Sistema Nervoso Central/patologia , Doenças do Sistema Nervoso Central/fisiopatologia , Técnicas de Cocultura , Fragmentação do DNA/fisiologia , Inibidores Enzimáticos/farmacologia , Marcação In Situ das Extremidades Cortadas , Integrina alfa6 , Camundongos , Camundongos Endogâmicos , Oligodendroglia/patologia , Fosfotransferases/antagonistas & inibidores , Fosfotransferases/metabolismo , Transdução de Sinais/fisiologia
12.
Oncogene ; 20(50): 7398-407, 2001 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-11704869

RESUMO

Protein kinase C (PKC) is a family of serine/threonine kinases involved in the transduction of a variety of signals. There is increasing evidence to indicate that specific PKC isoforms are involved in the regulation of distinct cellular processes. In glioma cells, PKC alpha was found to be a critical regulator of proliferation and cell cycle progression, while PKC epsilon was found to regulate adhesion and migration. Herein, we report that specific PKC isoforms are able to differentially activate extracellular-signal regulated kinase (ERK) in distinct cellular locations: while PKC alpha induces the activation of nuclear ERK, PKC epsilon induces the activation of ERK at focal adhesions. Inhibition of the ERK pathway completely abolished the PKC-induced integrin-mediated adhesion and migration. Thus, we present the first evidence that PKC epsilon is able to activate ERK at focal adhesions to mediate glioma cell adhesion and motility, providing a molecular mechanism to explain the different biological functions of PKC alpha and epsilon in glioma cells.


Assuntos
Neoplasias Encefálicas/patologia , Adesão Celular/efeitos dos fármacos , Adesões Focais/fisiologia , Glioma/patologia , Isoenzimas/metabolismo , Proteína Quinase 1 Ativada por Mitógeno/metabolismo , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Proteínas de Neoplasias/metabolismo , Proteína Quinase C/metabolismo , Acetato de Tetradecanoilforbol/farmacologia , Androstadienos/farmacologia , Neoplasias Encefálicas/enzimologia , Butadienos/farmacologia , Movimento Celular/efeitos dos fármacos , Ativação Enzimática , Inibidores Enzimáticos/farmacologia , Glioma/enzimologia , Humanos , MAP Quinase Quinase 1 , MAP Quinase Quinase 2 , Proteína Quinase 3 Ativada por Mitógeno , Quinases de Proteína Quinase Ativadas por Mitógeno/antagonistas & inibidores , Quinases de Proteína Quinase Ativadas por Mitógeno/metabolismo , Nitrilas/farmacologia , Fosforilação , Proteína Quinase C-épsilon , Processamento de Proteína Pós-Traducional , Proteínas Serina-Treonina Quinases/antagonistas & inibidores , Proteínas Serina-Treonina Quinases/metabolismo , Proteínas Tirosina Quinases/antagonistas & inibidores , Proteínas Tirosina Quinases/metabolismo , Pirimidinas/farmacologia , Transdução de Sinais/efeitos dos fármacos , Células Tumorais Cultivadas/citologia , Células Tumorais Cultivadas/efeitos dos fármacos , Células Tumorais Cultivadas/enzimologia , Wortmanina
13.
J Neuropathol Exp Neurol ; 60(10): 961-71, 2001 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-11589427

RESUMO

The CNS response to injury is characterized by the rapid activation of astrocytes in a process known as astrogliosis. The function of reactive astrocytes is controversial, in that both beneficial and detrimental properties are postulated. Identification of the molecules involved in regulating astrogliosis is an important step towards understanding astrocyte functions and establishing suitable conditions for CNS regeneration. We previously reported that inflammatory cytokines are regulators of astrogliosis but the key cytokine involved in initiating astrogliosis was unclear. We describe here that the elevation of glial fibrillary acid protein (GFAP) transcripts follows the very early rise of interleukin (IL)-1beta mRNA in a murine corticectomy model of CNS lesion. Furthermore, the injury-induced upregulation of GFAP mRNA and protein did not occur in mice genetically deficient for IL-1beta compared to wild-type animals. This was correlated with an absence of an increase in GFAP-immunoreactivity (GFAP-ir) in IL-1beta-null mice at 2 and 3 days of injury. However, by 5 to 7 days after the lesion, GFAP-ir was not different between cytokine-deficient and wild-type controls. Functionally, mice lacking IL-1beta exhibited a significant impairment in reformation of the blood-brain barrier (BBB) following corticectomy compared to wild-type controls. These findings suggest that the rapid production of IL-1beta following trauma plays a beneficial role in initiating astrogliosis in an attempt to restore the integrity of the BBB and seal off the wound site.


Assuntos
Sistema Nervoso Central/metabolismo , Sistema Nervoso Central/patologia , Proteína Glial Fibrilar Ácida/biossíntese , Gliose/metabolismo , Interleucina-1/deficiência , Animais , Barreira Hematoencefálica/genética , Córtex Cerebral/metabolismo , Córtex Cerebral/patologia , Feminino , Gliose/genética , Gliose/patologia , Interleucina-1/genética , Camundongos , Camundongos Mutantes , RNA Mensageiro/biossíntese
14.
J Neurosci ; 21(18): 7135-42, 2001 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-11549724

RESUMO

Leukocyte infiltration in the CNS after trauma or inflammation is triggered in part by upregulation of the chemokine, monocyte chemoattractant protein-1 (MCP-1), in astrocytes. However the signals that induce the upregulation of MCP-1 in astrocytes are unknown. We have investigated the roles for ATP P2X7 receptor activation because ATP is an intercellular signaling transmitter that is released in both trauma and inflammation and P2X7 receptors are involved in immune system signaling. Astrocytes in primary cell culture and acutely isolated from the hippocampus were immunopositive for P2X7 receptors. In astrocyte cultures, application of the selective P2X7 agonist, benzoyl-benzoyl ATP (Bz-ATP), activated MAP kinases extracellular signal receptor-activated kinase 1 (ERK1), ERK2, and p38. Purinergic antagonists depressed this activation with a profile suggesting P2X7 receptors. Bz-ATP also increased MCP-1 expression in cultured astrocytes, and again P2X7 antagonists prevented this increase. Blocking either the ERK1/ERK2 or the p38 pathway (with PD98059 or SB203580, respectively) significantly inhibited Bz-ATP-induced MCP-1 expression. Coapplication of both antagonists caused a greater depression. We also tested the roles for ATP receptor activation in inducing MCP-1 upregulation in corticectomy, an in vivo model of trauma. This model of cortical trauma was previously shown to increase MCP-1 expression in vivo principally in astrocytes. Suramin, a wide-spectrum purinergic receptor antagonist, significantly depressed the rapid (3 hr) trauma-induced increase in MCP-1 mRNA. These data indicate that purinergic transmitter receptors in astrocytes are important in regulating chemokine synthesis. The regulation of MCP-1 in astrocytes by ATP may be important in mediating communication with hematopoietic inflammatory cells.


Assuntos
Astrócitos/metabolismo , Quimiocina CCL2/biossíntese , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Receptores Purinérgicos P2/metabolismo , Trifosfato de Adenosina/análogos & derivados , Trifosfato de Adenosina/metabolismo , Trifosfato de Adenosina/farmacologia , Animais , Astrócitos/citologia , Astrócitos/efeitos dos fármacos , Células Cultivadas , Descorticação Cerebral , Quimiocina CCL2/genética , Relação Dose-Resposta a Droga , Proteína Glial Fibrilar Ácida/metabolismo , Hipocampo/citologia , Hipocampo/efeitos dos fármacos , Hipocampo/metabolismo , Imuno-Histoquímica , Masculino , Proteína Quinase 1 Ativada por Mitógeno/metabolismo , Proteína Quinase 3 Ativada por Mitógeno , Agonistas do Receptor Purinérgico P2 , Antagonistas do Receptor Purinérgico P2 , RNA Mensageiro/biossíntese , Ratos , Ratos Sprague-Dawley , Receptores Purinérgicos P2X7 , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/fisiologia , Proteínas Quinases p38 Ativadas por Mitógeno
17.
J Neurooncol ; 51(3): 245-64, 2001 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-11407596

RESUMO

The activity and regulation of a number of mitogenic signaling pathways is aberrant in astrocytomas, and this is thought to play a crucial role in the development of these tumors. The cascade of events leading to the formation and the progression from low-grade to high-grade astrocytomas is well characterized. These events include activating mutations, amplification, and overexpression of various growth factor receptors (e.g. epidermal growth factor receptor (EGFR), platelet derived growth factor receptor (PDGFR), c-Met), signaling intermediates (e.g. Ras and Protein kinase C (PKC)), and cell cycle regulatory molecules (e.g. mouse double minute-2 (Mdm2), cyclin-dependent kinase-4 (CDK4), and CDK6), that positively regulate proliferation and cell cycle progression. Inactivating mutations and deletions of signaling and cell cycle regulatory molecules that negatively regulate proliferation and cell cycle progression (e.g. p53, p16/INK4a, p14/ARF, p15/INK4b, retinoblastoma protein (Rb), and Phosphatase and tensin homologue deleted from chromosome 10 (PTEN)) also participate actively in the development of the transformed phenotype. Several mitogenic pathways are also stimulated via an autocrine loop, with astrocytoma cells expressing both the receptors and the respective cognate ligand. Due to the multitude of factors involved in astrocytoma pathogenesis, attempts to target a single pathway have not given satisfactory results. The simultaneous targeting of several pathways or the targeting of signaling intermediates, such as Ras or PKC, situated downstream of many growth factor receptor signaling pathways may show more efficacy in astrocytoma therapy. We will give an overview of how the combination of these aberrations drive astrocytoma cells into a relentless proliferation and how these signaling molecules may constitute relevant therapeutic targets.


Assuntos
Astrocitoma/patologia , Astrocitoma/fisiopatologia , Mitógenos/fisiologia , Transdução de Sinais/fisiologia , Animais , Ciclo Celular/fisiologia , Citoplasma/metabolismo , Humanos , Integrinas/fisiologia , Proteínas/fisiologia , Receptores de Fatores de Crescimento/fisiologia
18.
J Neurosci Methods ; 105(2): 111-20, 2001 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-11275268

RESUMO

Cognate interactions between human adult microglia and activated T lymphocytes induce the production of inflammatory cytokines. Since this interaction can occur in a non-antigen-dependent manner, it is relevant to a variety of CNS diseases where activated T cells, regardless of specificities, come into contact with microglia; these disorders include multiple sclerosis, trauma, stroke and Alzheimer's disease. A model cell line would facilitate studies of the engagement between T cells and human adult microglia, since the latter are difficult to obtain in substantial quantity or frequency. This study shows that the PMA/IFN gamma-treated U937 cell line shows similarities to microglia in its interaction with activated T lymphocytes, in that the production of tumor necrosis factor (TNF)-alpha, interleukin (IL)-4, IL-10 and IL-12 is induced. Morphological features and mechanisms of cytokine production resemble those observed in microglia--T cell co-cultures since CTLA-4 and CD40--CD40L blockades reduce TNF-alpha and IL-10 levels, while anti-CD23 inhibits IL-10 only in U937--T cell interactions. We propose that PMA/IFN gamma-treated U937 cells can serve as a model of human adult microglia to study cytokine generation in response to interactions with activated T cells.


Assuntos
Carcinógenos/farmacologia , Técnicas de Cultura de Células/métodos , Diferenciação Celular/efeitos dos fármacos , Citocinas/efeitos dos fármacos , Microglia/efeitos dos fármacos , Linfócitos T/imunologia , Acetato de Tetradecanoilforbol/farmacologia , Anticorpos/farmacologia , Antígenos CD/efeitos dos fármacos , Antígenos CD/metabolismo , Antígeno B7-1/efeitos dos fármacos , Antígeno B7-1/metabolismo , Antígeno B7-2 , Antígenos CD40/efeitos dos fármacos , Antígenos CD40/metabolismo , Comunicação Celular/efeitos dos fármacos , Comunicação Celular/imunologia , Diferenciação Celular/fisiologia , Tamanho Celular/efeitos dos fármacos , Tamanho Celular/imunologia , Doenças do Sistema Nervoso Central/imunologia , Doenças do Sistema Nervoso Central/metabolismo , Doenças do Sistema Nervoso Central/fisiopatologia , Citocinas/biossíntese , Encefalite/imunologia , Encefalite/metabolismo , Encefalite/fisiopatologia , Humanos , Interferon gama/farmacologia , Glicoproteínas de Membrana/efeitos dos fármacos , Glicoproteínas de Membrana/metabolismo , Microglia/citologia , Microglia/imunologia , Receptores de IgE/efeitos dos fármacos , Receptores de IgE/metabolismo , Linfócitos T/citologia , Células U937
19.
Ann Neurol ; 49(2): 230-41, 2001 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-11220743

RESUMO

The release of potentially neurotoxic molecules by HIV-infected brain macrophages is accompanied by neuronal injury and death that results in the development of HIV-associated dementia (HAD). Among the potential neurotoxins implicated in the development of HAD is the HIV-1 transactivating protein, Tat. To investigate the mechanism by which Tat causes neurotoxicity, brain-derived Tat sequences from nondemented (Tat-ND) and demented (Tat-HAD) AIDS patients, which differed primarily in the augmenting region of Tat, were expressed in U937 monoblastoid cells and primary human macrophages. Cells expressing Tat-HAD protein exhibited elevated matrix metalloproteinase (MMP)-2 and -7 release and activation, but cells expressing Tat-ND did not exhibit enhanced MMP expression. Conditioned media from Tat-HAD-transfected cells caused significantly greater neuronal death (15.4 +/- 4.3%) than did Tat-ND (4.4 +/- 2.1%) or nontransfected (2.1 +/- 0.8%) cell-derived conditioned media. The neurotoxicity induced by Tat-HAD was inhibited by anti-MMP-2 or -7 antibodies (p < 0.005) but not by antibodies against MMP-9 or Tat. Similarly, scid/nod mice receiving striatal implants of Tat-HAD-transfected cells exhibited greater neurobehavioral abnormalities and neuronal loss (p < 0.005) than did animals receiving Tat-ND or nontransfected cells, which were reduced by treatment with the MMP inhibitor prinomastat (p < 0.005). These findings indicate that Tat causes neuronal death through an indirect mechanism that is Tat sequence dependent and involves the induction of MMPs.


Assuntos
Complexo AIDS Demência/metabolismo , Produtos do Gene tat/toxicidade , HIV-1/metabolismo , Metaloproteinase 2 da Matriz/metabolismo , Metaloproteinase 7 da Matriz/metabolismo , Inibidores de Metaloproteinases de Matriz , Fragmentos de Peptídeos/toxicidade , Complexo AIDS Demência/genética , Sequência de Aminoácidos , Animais , Morte Celular/efeitos dos fármacos , Humanos , Imuno-Histoquímica , Camundongos , Dados de Sequência Molecular , Neurônios/metabolismo , Reação em Cadeia da Polimerase , Produtos do Gene tat do Vírus da Imunodeficiência Humana
20.
Cancer Res ; 61(3): 1162-70, 2001 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-11221847

RESUMO

Tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) preferentially triggers apoptosis in tumor cells versus normal cells, thus providing a therapeutic potential. In this study, we examined a large panel of human malignant glioma cell lines and primary cultures of normal human astrocytes for their sensitivity to TRAIL. Of 13 glioma cell lines, 3 were sensitive (80-100% death), 4 were partially resistant (30-79% death), and 6 were resistant (< 30% death). Normal astrocytes were also resistant. TRAIL-induced cell death was characterized by activation of caspase-8 and -3, poly(ADP-ribose) polymerase cleavage, and DNA fragmentation. Decoy receptor (DcR1 and DcR2) expression was limited in the glioma cell lines and did not correlate with TRAIL sensitivity. Both sensitive and resistant cell lines expressed TRAIL death receptor (DR5), adapter protein Fas-associated death domain (FADD), and caspase-8; but resistant cell lines expressed 2-fold higher levels of the apoptosis inhibitor phosphoprotein enriched in diabetes/phosphoprotein enriched in astrocytes-15 kDa (PED/PEA-15). In contrast, cellular FADD-like IL-1beta-converting enzyme-like inhibitory protein (cFLIP) expression was similar in sensitive and resistant cells. Transfection of sense PED/PEA-15 cDNA in sensitive cells resulted in cell resistance, whereas transfection of antisense in resistant cells rendered them sensitive. Inhibition of protein kinase C (PKC) activity restored TRAIL sensitivity in resistant cells, suggesting that PED/ PEA-15 function might be dependent on PKC-mediated phosphorylation. In summary, TRAIL induces apoptosis in > 50% of glioma cell lines, and this killing occurs through activation of the DR pathway. This caspase-8-induced apoptotic cascade is regulated by intracellular PED/PEA-15, but not by cFLIP or decoy receptors. This pathway may be exploitable for glioma and possibly for other cancer therapies.


Assuntos
Apoptose/efeitos dos fármacos , Glioma/patologia , Peptídeos e Proteínas de Sinalização Intracelular , Glicoproteínas de Membrana/farmacologia , Fator de Necrose Tumoral alfa/farmacologia , Apoptose/fisiologia , Proteínas Reguladoras de Apoptose , Astrócitos/citologia , Astrócitos/efeitos dos fármacos , Proteína Reguladora de Apoptosis Semelhante a CASP8 e FADD , Proteínas de Transporte/biossíntese , Caspase 8 , Caspase 9 , Caspases/metabolismo , Caspases/fisiologia , Células Cultivadas , Ativação Enzimática , Proteínas Ligadas por GPI , Glioma/genética , Glioma/metabolismo , Humanos , Glicoproteínas de Membrana/fisiologia , Oligodesoxirribonucleotídeos Antissenso/genética , Oligodesoxirribonucleotídeos Antissenso/farmacologia , Fosfoproteínas/genética , Inibidores da Síntese de Proteínas/farmacologia , Receptores do Fator de Necrose Tumoral/biossíntese , Membro 10c de Receptores do Fator de Necrose Tumoral , Proteínas Recombinantes/farmacologia , Ligante Indutor de Apoptose Relacionado a TNF , Transfecção , Células Tumorais Cultivadas , Receptores Chamariz do Fator de Necrose Tumoral , Fator de Necrose Tumoral alfa/fisiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...