Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
1.
BMC Neurosci ; 23(1): 30, 2022 05 25.
Artigo em Inglês | MEDLINE | ID: mdl-35614392

RESUMO

BACKGROUND: Therapeutic agents stimulating the process of myelination could be beneficial for the treatment of demyelinating diseases, such as multiple sclerosis. The efficient translation of compounds promoting myelination in vitro to efficacy in vivo is inherently time-consuming and expensive. Thyroid hormones accelerate the differentiation and maturation of oligodendrocytes, thereby promoting myelination. Systemic administration of the thyroid hormone thyroxine (T4) accelerates brain maturation, including myelination, during early postnatal development. The objective of this study was to validate an animal model for rapid testing of promyelinating therapeutic candidates for their effects on early postnatal development by using T4 as a reference compound. METHODS: Daily subcutaneous injections of T4 were given to Sprague Dawley rat pups from postnatal day (PND) 2 to PND10. Changes in white matter were determined at PND10 using diffusion tensor magnetic resonance imaging (DTI). Temporal changes in myelination from PND3 to PND11 were also assessed by quantifying myelin basic protein (MBP) expression levels in the brain using the resonance Raman spectroscopy/enzyme-linked immunosorbent assay (RRS-ELISA) and quantitative immunohistochemistry. RESULTS: DTI of white matter tracts showed significantly higher fractional anisotropy in the internal capsule of T4-treated rat pups. The distribution of total FA values in the forebrain was significantly shifted towards higher values in the T4-treated group, suggesting increased myelination. In vivo imaging data were supported by in vitro observations, as T4 administration significantly potentiated the developmental increase in MBP levels in brain lysates starting from PND8. MBP levels in the brain of animals that received treatment for 9 days correlated with the FA metric determined in the same pups in vivo a day earlier. Furthermore, accelerated developmental myelination following T4 administration was confirmed by immunohistochemical staining for MBP in coronal brain sections of treated rat pups. CONCLUSIONS: T4-treated rat pups had increased MBP expression levels and higher MRI fractional anisotropy values, both indications of accelerated myelination. This simple developmental myelination model affords a rapid test of promyelinating activity in vivo within several days, which could facilitate in vivo prescreening of candidate therapeutic compounds for developmental hypomyelinating diseases. Further research will be necessary to assess the utility of this platform for screening promyelination compounds in more complex demyelination disease models, such us multiple sclerosis.


Assuntos
Esclerose Múltipla , Substância Branca , Animais , Encéfalo/metabolismo , Modelos Animais de Doenças , Esclerose Múltipla/metabolismo , Oligodendroglia/metabolismo , Ratos , Ratos Sprague-Dawley , Substância Branca/patologia
2.
BMC Neurosci ; 17: 16, 2016 Apr 22.
Artigo em Inglês | MEDLINE | ID: mdl-27103572

RESUMO

BACKGROUND: Regeneration of new myelin is impaired in persistent multiple sclerosis (MS) lesions, leaving neurons unable to function properly and subject to further degeneration. Current MS therapies attempt to ameliorate autoimmune-mediated demyelination, but none directly promote the regeneration of lost and damaged myelin of the central nervous system (CNS). Development of new drugs that stimulate remyelination has been hampered by the inability to evaluate axonal myelination in a rapid CNS culture system. RESULTS: We established a high throughput cell-based assay to identify compounds that promote myelination. Culture methods were developed for initiating myelination in vitro using primary embryonic rat cortical cells. We developed an immunofluorescent phenotypic image analysis method to quantify the morphological alignment of myelin characteristic of the initiation of myelination. Using γ-secretase inhibitors as promoters of myelination, the optimal growth, time course and compound treatment conditions were established in a 96 well plate format. We have characterized the cortical myelination assay by evaluating the cellular composition of the cultures and expression of markers of differentiation over the time course of the assay. We have validated the assay scalability and consistency by screening the NIH clinical collection library of 727 compounds and identified ten compounds that promote myelination. Half maximal effective concentration (EC50) values for these compounds were determined to rank them according to potency. CONCLUSIONS: We have designed the first high capacity in vitro assay that assesses myelination of live axons. This assay will be ideal for screening large compound libraries to identify new drugs that stimulate myelination. Identification of agents capable of promoting the myelination of axons will likely lead to the development of new therapeutics for MS patients.


Assuntos
Axônios/efeitos dos fármacos , Córtex Cerebral/efeitos dos fármacos , Avaliação Pré-Clínica de Medicamentos/métodos , Ensaios de Triagem em Larga Escala/métodos , Esclerose Múltipla/tratamento farmacológico , Bainha de Mielina/efeitos dos fármacos , Regeneração Nervosa/efeitos dos fármacos , Secretases da Proteína Precursora do Amiloide/farmacologia , Animais , Axônios/fisiologia , Técnicas de Cultura de Células , Diferenciação Celular/efeitos dos fármacos , Córtex Cerebral/fisiologia , Meios de Cultivo Condicionados/farmacologia , Imunofluorescência/métodos , Esclerose Múltipla/fisiopatologia , Bainha de Mielina/fisiologia , Oligodendroglia/efeitos dos fármacos , Oligodendroglia/fisiologia , Ratos
3.
Mol Pharmacol ; 88(2): 357-67, 2015 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-26018904

RESUMO

Erythropoietin (EPO) and its receptor are expressed in a wide variety of tissues, including the central nervous system. Local expression of both EPO and its receptor is upregulated upon injury or stress and plays a role in tissue homeostasis and cytoprotection. High-dose systemic administration or local injection of recombinant human EPO has demonstrated encouraging results in several models of tissue protection and organ injury, while poor tissue availability of the protein limits its efficacy. Here, we describe the discovery and characterization of the nonpeptidyl compound STS-E412 (2-[2-(4-chlorophenoxy)ethoxy]-5,7-dimethyl-[1,2,4]triazolo[1,5-a]pyrimidine), which selectively activates the tissue-protective EPO receptor, comprising an EPO receptor subunit (EPOR) and the common ß-chain (CD131). STS-E412 triggered EPO receptor phosphorylation in human neuronal cells. STS-E412 also increased phosphorylation of EPOR, CD131, and the EPO-associated signaling molecules JAK2 and AKT in HEK293 transfectants expressing EPOR and CD131. At low nanomolar concentrations, STS-E412 provided EPO-like cytoprotective effects in primary neuronal cells and renal proximal tubular epithelial cells. The receptor selectivity of STS-E412 was confirmed by a lack of phosphorylation of the EPOR/EPOR homodimer, lack of activity in off-target selectivity screening, and lack of functional effects in erythroleukemia cell line TF-1 and CD34(+) progenitor cells. Permeability through artificial membranes and Caco-2 cell monolayers in vitro and penetrance across the blood-brain barrier in vivo suggest potential for central nervous system availability of the compound. To our knowledge, STS-E412 is the first nonpeptidyl, selective activator of the tissue-protective EPOR/CD131 receptor. Further evaluation of the potential of STS-E412 in central nervous system diseases and organ protection is warranted.


Assuntos
Encéfalo/embriologia , Eritropoetina/metabolismo , Neurônios/metabolismo , Pirimidinas/farmacologia , Pirimidinas/farmacocinética , Receptores da Eritropoetina/agonistas , Triazóis/farmacologia , Triazóis/farmacocinética , Animais , Disponibilidade Biológica , Barreira Hematoencefálica/efeitos dos fármacos , Encéfalo/citologia , Células CACO-2 , Células Cultivadas , Subunidade beta Comum dos Receptores de Citocinas/metabolismo , Células HEK293 , Humanos , Ratos , Transdução de Sinais/efeitos dos fármacos
4.
Glia ; 63(5): 768-79, 2015 May.
Artigo em Inglês | MEDLINE | ID: mdl-25557204

RESUMO

Inflammatory signals present in demyelinated multiple sclerosis lesions affect the reparative remyelination process conducted by oligodendrocyte progenitor cells (OPCs). Interferon-γ (IFN-γ), tumor necrosis factor-α (TNF-α), and interleukin (IL)-6 have differing effects on the viability and growth of OPCs, however the effects of IL-17A are largely unknown. Primary murine OPCs were stimulated with IL-17A and their viability, proliferation, and maturation were assessed in culture. IL-17A-stimulated OPCs exited the cell cycle and differentiated with no loss in viability. Expression of the myelin-specific protein, proteolipid protein, increased in a cerebellar slice culture assay in the presence of IL-17A. Downstream, IL-17A activated ERK1/2 within 15 min and induced chemokine expression in 2 days. These results demonstrate that IL-17A exposure stimulates OPCs to mature and participate in the inflammatory response.


Assuntos
Diferenciação Celular/efeitos dos fármacos , Encefalomielite Autoimune Experimental/patologia , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Oligodendroglia/efeitos dos fármacos , Oligodendroglia/enzimologia , Células-Tronco/efeitos dos fármacos , Animais , Células Cultivadas , Cerebelo/citologia , Cerebelo/metabolismo , Citocinas/genética , Citocinas/metabolismo , Modelos Animais de Doenças , Encefalomielite Autoimune Experimental/induzido quimicamente , Citometria de Fluxo , Adjuvante de Freund/toxicidade , Regulação da Expressão Gênica/efeitos dos fármacos , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Técnicas In Vitro , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Proteína Proteolipídica de Mielina/genética , Glicoproteína Mielina-Oligodendrócito/toxicidade , Técnicas de Cultura de Órgãos , Fragmentos de Peptídeos/toxicidade , Receptores de Interleucina-17/deficiência , Receptores de Interleucina-17/genética , Células-Tronco/fisiologia
5.
BMC Res Notes ; 16(1): 348, 2023 Nov 25.
Artigo em Inglês | MEDLINE | ID: mdl-38007440

RESUMO

OBJECTIVES: Animal models of skin disease are used to evaluate therapeutics to alleviate disease. One common clinical dermatological complaint is pruritus (itch), but there is a lack of standardization in the characterization of pre-clinical models and scratching behavior, a key itch endpoint, is often neglected. One such model is the widely used imiquimod (IMQ) mouse model of psoriasis. However, it lacks characterized behavioral attributes like scratching, nor has widely expanded to other species like rats. Given these important attributes, this study was designed to broaden the characterization beyond the expected IMQ-induced psoriasis-like skin inflammatory skin changes and to validate the role of a potential therapeutic agent for pruritus in our genetic rat model. The study included female Wistar rats and genetically modified knockin (humanized proteinase-activated receptor 2 (F2RL1) female rats, with the widely used C57BL/6 J mice as a methodology control for typical IMQ dosing. RESULTS: We demonstrate that the IMQ model can be reproduced in rats, including their genetically modified derivatives, and how scratching can be used as a key behavioral endpoint. We systemically delivered an anti-PAR2 antibody (P24E1102) which reversed scratching bouts-validating this behavioral methodology and have shown its feasibility and value in identifying effective antipruritic drugs.


Assuntos
Antipruriginosos , Psoríase , Camundongos , Ratos , Feminino , Animais , Antipruriginosos/farmacologia , Antipruriginosos/uso terapêutico , Imiquimode/efeitos adversos , Ratos Wistar , Camundongos Endogâmicos C57BL , Prurido/induzido quimicamente , Prurido/tratamento farmacológico , Prurido/genética , Pele , Psoríase/induzido quimicamente , Psoríase/tratamento farmacológico , Modelos Animais de Doenças
6.
Neurotherapeutics ; 16(3): 808-827, 2019 07.
Artigo em Inglês | MEDLINE | ID: mdl-30815844

RESUMO

The development of neuroprotective therapies is a sought-after goal. By screening combinatorial chemical libraries using in vitro assays, we identified the small molecule BN201 that promotes the survival of cultured neural cells when subjected to oxidative stress or when deprived of trophic factors. Moreover, BN201 promotes neuronal differentiation, the differentiation of precursor cells to mature oligodendrocytes in vitro, and the myelination of new axons. BN201 modulates several kinases participating in the insulin growth factor 1 pathway including serum-glucocorticoid kinase and midkine, inducing the phosphorylation of NDRG1 and the translocation of the transcription factor Foxo3 to the cytoplasm. In vivo, BN201 prevents axonal and neuronal loss, and it promotes remyelination in models of multiple sclerosis, chemically induced demyelination, and glaucoma. In summary, we provide a new promising strategy to promote neuroaxonal survival and remyelination, potentially preventing disability in brain diseases.


Assuntos
Amidas/uso terapêutico , Axônios/efeitos dos fármacos , Encefalite/tratamento farmacológico , Bainha de Mielina/efeitos dos fármacos , Fármacos Neuroprotetores/uso terapêutico , Peptoides/uso terapêutico , Pirrolidinonas/uso terapêutico , Animais , Modelos Animais de Doenças , Encefalomielite Autoimune Experimental/tratamento farmacológico , Feminino , Imunofluorescência , Glaucoma/tratamento farmacológico , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Nervo Óptico/efeitos dos fármacos , Proguanil , Ratos , Ratos Sprague-Dawley , Reação em Cadeia da Polimerase em Tempo Real , Triazinas
7.
Methods Mol Biol ; 1791: 179-192, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30006710

RESUMO

The formation of new myelin in persistent multiple sclerosis (MS) lesions is compromised, leading to a reduction in neuron function and subject to degeneration and death. Current MS therapies can control autoimmune-mediated demyelination, but none directly promote the regeneration of myelin in the central nervous system (CNS). To identify new drugs that stimulate remyelination, we established a high-throughput cell-based assay to identify compounds that promote myelination. Methods were developed for initiating myelination in vitro using a preparation of primary embryonic rat cortical cells. We developed an immunofluorescent phenotypic image analysis method to quantify the morphological alignment of myelin characteristic of the initiation of myelination. The assay scalability and consistency was validated by screening the NIH clinical collection library of 727 compounds and identified ten compounds that promote myelination (Lariosa-Willingham et al., BMC Neurosci 17:16, 2016). Here, we present the detailed methods for a high capacity in vitro assay that assesses myelination of live axons.


Assuntos
Axônios , Sistema Nervoso Central/fisiologia , Ensaios de Triagem em Larga Escala , Bainha de Mielina/fisiologia , Neurogênese , Animais , Encéfalo/metabolismo , Técnicas de Cultura de Células , Descoberta de Drogas , Imuno-Histoquímica/métodos , Imagem Molecular/métodos , Bainha de Mielina/efeitos dos fármacos , Neurogênese/efeitos dos fármacos , Oligodendroglia/fisiologia , Ratos
8.
Curr Protoc Cell Biol ; 79(1): e49, 2018 06.
Artigo em Inglês | MEDLINE | ID: mdl-29924487

RESUMO

Multiple sclerosis (MS) is an autoimmune disease that involves an immune-mediated inflammatory response in the central nervous system and optic nerve resulting in demyelination and neural degeneration, the cause of which is unknown. The adult central nervous system has the capacity to remyelinate axons by generating new oligodendrocytes (OLs). To identify clinical candidate compounds that may promote remyelination, we have developed a high-throughput screening (HTS) assay to identify compounds that promote the differentiation of oligodendrocyte precursor cells (OPCs) into OLs. Using acutely dissociated and purified rat OPCs coupled with immunofluorescent image quantification, we have developed an OL differentiation assay. Building on OPC culturing techniques developed over the past 30 years, we have scaled up the isolation and purification process to generate sufficient quantities for HTS. We then describe the use of these acutely derived OPCs in an assay designed to identify compounds that promote differentiation into OLs. We have validated this assay with a known promoter of differentiation, thyroid hormone, and subsequently used the assay to screen the NIH clinical collection library (Lariosa-Willingham, et al., 2016). © 2018 by John Wiley & Sons, Inc.


Assuntos
Diferenciação Celular , Separação Celular/métodos , Avaliação Pré-Clínica de Medicamentos/métodos , Ensaios de Triagem em Larga Escala/métodos , Células Precursoras de Oligodendrócitos/citologia , Oligodendroglia/citologia , Animais , Dissecação , Proteína Básica da Mielina/metabolismo , Oxigênio/farmacologia , Perfusão , Ratos Sprague-Dawley , Tripsina/metabolismo
9.
BMC Res Notes ; 9(1): 419, 2016 Sep 05.
Artigo em Inglês | MEDLINE | ID: mdl-27592856

RESUMO

BACKGROUND: Multiple sclerosis is caused by an autoimmune response resulting in demyelination and neural degeneration. The adult central nervous system has the capacity to remyelinate axons in part through the generation of new oligodendrocytes (OLs). To identify clinical candidate compounds that may promote remyelination, we have developed a high throughput screening (HTS) assay to identify compounds that promote the differentiation of oligodendrocyte precursor cells (OPCs) into OLs. RESULTS: Using acutely dissociated and purified rat OPCs coupled with immunofluorescent image quantification, we have developed an OL differentiation assay. We have validated this assay with a known promoter of differentiation, thyroid hormone, and subsequently used the assay to screen the NIH clinical collection library. We have identified twenty-seven hit compounds which were validated by dose response analysis and the generation of half maximal effective concentration (EC50) values allowed for the ranking of efficacy. The assay identified novel promoters of OL differentiation which we attribute to (1) the incorporation of an OL toxicity pre-screen to allow lowering the concentrations of toxic compounds and (2) the utilization of freshly purified, non-passaged OPCs. These features set our assay apart from other OL differentiation assays used for drug discovery efforts. CONCLUSIONS: This acute primary OL-based differentiation assay should be of use to those interested in screening large compound libraries for the identification of drugs for the treatment of MS and other demyelinating diseases.


Assuntos
Diferenciação Celular/efeitos dos fármacos , Avaliação Pré-Clínica de Medicamentos/métodos , Oligodendroglia/citologia , Oligodendroglia/efeitos dos fármacos , Células-Tronco/efeitos dos fármacos , Animais , Técnicas In Vitro , Esclerose Múltipla/tratamento farmacológico , Ratos , Células-Tronco/citologia
10.
BMC Res Notes ; 9(1): 444, 2016 Sep 05.
Artigo em Inglês | MEDLINE | ID: mdl-27629829

RESUMO

BACKGROUND: Newly proliferated oligodendrocyte precursor cells (OPCs) migrate and surround lesions of patients with multiple sclerosis (MS) and other demyelinating diseases, but fail to differentiate into oligodendrocytes (OLs) and remyelinate remaining viable axons. The abundance of secreted inflammatory factors within and surrounding these lesions likely plays a major inhibitory role, promoting cell death and preventing OL differentiation and axon remyelination. To identify clinical candidate compounds that may protect existing and differentiating OLs in patients, we have developed a high throughput screening (HTS) assay that utilizes purified rat OPCs. RESULTS: Using a fluorescent indicator of cell viability coupled with image quantification, we developed an assay to allow the identification of compounds that promote OL viability and differentiation in the presence of the synergistic inflammatory cytokines, tumor necrosis factor α and interferon-γ. We have utilized this assay to screen the NIH clinical collection library and identify compounds that protect OLs and promote OL differentiation in the presence of these inflammatory cytokines. CONCLUSION: This primary OL-based cytokine protection assay is adaptable for HTS and may be easily modified for profiling of compounds in the presence of other potentially inhibitory molecules found in MS lesions. This assay should be of use to those interested in identifying drugs for the treatment of MS and other demyelinating diseases.


Assuntos
Avaliação Pré-Clínica de Medicamentos/métodos , Oligodendroglia/efeitos dos fármacos , Animais , Diferenciação Celular/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Feminino , Inflamação , Interferon gama/metabolismo , Masculino , Esclerose Múltipla/patologia , Oligodendroglia/citologia , Oligodendroglia/metabolismo , Ratos , Fator de Necrose Tumoral alfa/metabolismo
11.
Dev Cell ; 34(2): 152-67, 2015 Jul 27.
Artigo em Inglês | MEDLINE | ID: mdl-26166300

RESUMO

Myelin is essential in vertebrates for the rapid propagation of action potentials, but the molecular mechanisms driving its formation remain largely unknown. Here we show that the initial stage of process extension and axon ensheathment by oligodendrocytes requires dynamic actin filament assembly by the Arp2/3 complex. Unexpectedly, subsequent myelin wrapping coincides with the upregulation of actin disassembly proteins and rapid disassembly of the oligodendrocyte actin cytoskeleton and does not require Arp2/3. Inducing loss of actin filaments drives oligodendrocyte membrane spreading and myelin wrapping in vivo, and the actin disassembly factor gelsolin is required for normal wrapping. We show that myelin basic protein, a protein essential for CNS myelin wrapping whose role has been unclear, is required for actin disassembly, and its loss phenocopies loss of actin disassembly proteins. Together, these findings provide insight into the molecular mechanism of myelin wrapping and identify it as an actin-independent form of mammalian cell motility.


Assuntos
Citoesqueleto de Actina/metabolismo , Complexo 2-3 de Proteínas Relacionadas à Actina/metabolismo , Sistema Nervoso Central/crescimento & desenvolvimento , Bainha de Mielina/fisiologia , Oligodendroglia/fisiologia , Complexo 2-3 de Proteínas Relacionadas à Actina/genética , Actinas/metabolismo , Animais , Axônios/fisiologia , Membrana Celular/fisiologia , Movimento Celular/fisiologia , Células Cultivadas , Sistema Nervoso Central/embriologia , Cofilina 1/genética , Gelsolina/genética , Gelsolina/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Proteína Básica da Mielina/genética , Proteína Básica da Mielina/metabolismo , Nervo Óptico/metabolismo , Nervo Óptico/fisiologia , Interferência de RNA , RNA Interferente Pequeno , Ratos , Ratos Sprague-Dawley
12.
J Neuroimmunol ; 153(1-2): 108-21, 2004 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-15265669

RESUMO

FTY720 is a prodrug for FTY-phosphate, an agonist at four of the five known receptors for sphingosine-1-phosphate (S1P). We show that administration of either FTY720 or FTY-P to SJL mice with established relapsing-remitting experimental autoimmune encephalitis (EAE) results in a rapid and sustained improvement in their clinical status, and a reversal of changes in expression of mRNAs encoding some myelin proteins and inflammatory mediators. EAE produced by adoptively transferring lymph node cells from immunized mice to naïve hosts is similarly ameliorated by FTY-P. Treatment with FTY-P is accompanied by a dose-responsive peripheral lymphopoenia.


Assuntos
Encefalomielite Autoimune Experimental/tratamento farmacológico , Imunossupressores/uso terapêutico , Propilenoglicóis/uso terapêutico , Receptores Acoplados a Proteínas G/agonistas , Animais , Antineoplásicos/uso terapêutico , Modelos Animais de Doenças , Relação Dose-Resposta a Droga , Interações Medicamentosas , Encefalomielite Autoimune Experimental/sangue , Encefalomielite Autoimune Experimental/induzido quimicamente , Encefalomielite Autoimune Experimental/metabolismo , Feminino , Cloridrato de Fingolimode , Regulação da Expressão Gênica/imunologia , Fator Estimulador de Colônias de Granulócitos e Macrófagos/genética , Fator Estimulador de Colônias de Granulócitos e Macrófagos/metabolismo , Imunossupressores/farmacologia , Interferon gama/genética , Interferon gama/metabolismo , Linfócitos/efeitos dos fármacos , Linfócitos/fisiologia , Linfopenia/tratamento farmacológico , Linfopenia/etiologia , Camundongos , Camundongos Endogâmicos , Mitoxantrona/uso terapêutico , Dados de Sequência Molecular , Proteínas da Mielina/genética , Proteínas da Mielina/metabolismo , Proteína Proteolipídica de Mielina , Óxido Nítrico Sintase/genética , Óxido Nítrico Sintase/metabolismo , Óxido Nítrico Sintase Tipo II , Fragmentos de Peptídeos , Propilenoglicóis/farmacologia , RNA Mensageiro/biossíntese , Receptores de Lisofosfolipídeos , Reação em Cadeia da Polimerase Via Transcriptase Reversa/métodos , Esfingosina/análogos & derivados , Fatores de Tempo
13.
Brain Res ; 978(1-2): 213-22, 2003 Jul 18.
Artigo em Inglês | MEDLINE | ID: mdl-12834916

RESUMO

Glia play a pivotal role in glutaminergic excitatory neurotransmission in the central nervous system by regulating synaptic levels of glutamate and by providing glutamine as the sole precursor for the neurotransmitter pool glutamate to neurons through the glutamate-glutamine cycle. In the present investigation, we examined the influence of glutamate application on glutamine, serine and aspartate release from rat cortical glial cultures. The glial glutamate transporters rapidly cleared exogenously applied glutamate and this was accompanied by rapid increases in aspartate and glutamine, and a delayed increase in serine levels in the glial-conditioned medium. While glutamate-induced increases in glutamine and serine were sustained for up to 24 h, increases in aspartate lasted only for up to 6 h. The glutamate-induced increases in aspartate and glutamine were dependent both on the concentration and the duration of glutamate stimulus, but were largely insensitive to the inhibition of non-N-methyl-D-aspartate receptors or the metabotropic glutamate receptor 5. Inhibition of the glutamate transporter function by L-trans-pyrrolidine 2,4-dicarboxylate decreased the rate of glutamate uptake but not completely abrogated the uptake process, and this resulted in the attenuation of rate of glutamate induced glutamine synthesis. Dexamethasone treatment increased serine and glutamine levels in conditioned medium and increased glutamate induced glutamine release suggesting an upregulation of glutamine synthase activity. These results further substantiate coupling between glutamate and glutamine, and shed light on glutamate-dependent release of serine and aspartate, which may further contribute to excitatory neurotransmission.


Assuntos
Ácido Aspártico/metabolismo , Ácido Glutâmico/farmacologia , Glutamina/metabolismo , Neuroglia/efeitos dos fármacos , Serina/metabolismo , Análise de Variância , Animais , Animais Recém-Nascidos , Anti-Inflamatórios/farmacologia , Células Cultivadas , Córtex Cerebral , Dexametasona/farmacologia , Ácidos Dicarboxílicos/farmacologia , Relação Dose-Resposta a Droga , Interações Medicamentosas , Ácido Glutâmico/análise , Neuroglia/metabolismo , Inibidores da Captação de Neurotransmissores/farmacologia , Pirrolidinas/farmacologia , Ratos , Ratos Sprague-Dawley , Fatores de Tempo
14.
Brain Res ; 990(1-2): 182-94, 2003 Nov 14.
Artigo em Inglês | MEDLINE | ID: mdl-14568343

RESUMO

Lysophosphatidic acid (1-acyl-2-lyso-sn-glycero-3-phosphate; LPA) and sphingosine-1-phosphate (S1P) are bioactive phospholipids which respectively act as agonists for the G-protein-coupled lpA receptors (LPA1, LPA2, and LPA3) and s1p receptors (S1P1, S1P2, S1P3, S1P4, and S1P5), collectively referred to as lysophospholipid receptors (lpR). Since astrocytes are responsive to LPA and S1P, we examined mechanisms of lpR signaling in rat cortical secondary astrocytes. Rat cortical astrocyte mRNA expression by quantitative TaqMan polymerase chain reaction (PCR) analysis revealed the following order of relative expression of lpR mRNAs: s1p3>s1p1>lpa1>s1p2=lpa3>>s1p5. Activation of lpRs by LPA or S1P led to multiple pharmacological effects, including the influx of calcium, phosphoinositide (PI) hydrolysis, phosphorylation of extracellular receptor regulated kinase (ERK) and release of [3H]-arachidonic acid (AA). These signalling events downstream of lpR activation were inhibited to varying degrees by pertussis toxin (PTX) pretreatment or by the inhibition of sphingosine kinase (SK), a rate-limiting enzyme in the biosynthesis of S1P from sphingosine. These results suggest that astrocyte lpR signalling mechanisms likely involve both Gi- and Gq-coupled GPCRs and that receptor-mediated activation of SK leads to intracellular generation of S1P, which in turn amplifies the lpR signalling in a paracrine/autocrine manner.


Assuntos
Astrócitos/fisiologia , Córtex Cerebral/fisiologia , Lisofosfolipídeos/metabolismo , Receptores de Lipoproteínas/efeitos dos fármacos , Transdução de Sinais/efeitos dos fármacos , Esfingosina/análogos & derivados , Actinas/biossíntese , Actinas/genética , Animais , Ácido Araquidônico/metabolismo , Astrócitos/efeitos dos fármacos , Western Blotting , Sinalização do Cálcio/efeitos dos fármacos , Células Cultivadas , Córtex Cerebral/citologia , Córtex Cerebral/efeitos dos fármacos , Clonagem Molecular , Ativação Enzimática/efeitos dos fármacos , Ativação Enzimática/fisiologia , Fosfatos de Inositol/metabolismo , Lisofosfolipídeos/farmacologia , Proteína Quinase 1 Ativada por Mitógeno/metabolismo , Fosforilação , Fosfotransferases (Aceptor do Grupo Álcool)/metabolismo , RNA Mensageiro/biossíntese , Ratos , Ratos Sprague-Dawley , Receptor Cross-Talk/efeitos dos fármacos , Receptores de Lipoproteínas/biossíntese , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Esfingosina/farmacologia
15.
Int J Dev Neurosci ; 22(3): 131-5, 2004 May.
Artigo em Inglês | MEDLINE | ID: mdl-15140466

RESUMO

Reactive gliosis is an aspect of neural plasticity and growth factor (GF) stimulation of astrocytes in vitro is widely regarded as a model system to study astrocyte plasticity. Astrocytes express receptors for several ligands including lysophosphatidic acid (LPA) and sphingosine-1-phosphate (S1P), agonists for the G-protein-coupled lysophospholipid receptors (lpRs). Activation of lpRs by LPA or S1P leads to multiple pharmacological effects including the influx of calcium, phosphoinositide (PI) hydrolysis, phosphorylation of extracellular receptor regulated kinase (ERK), release of arachidonic acid, and induces mitogenesis. Treatment of astrocytes in vitro with a growth factor cocktail (containing epidermal growth factor [EGF], basic fibroblast growth factor [bFGF] and insulin) led to a marked attenuation of lpR-induced PI hydrolysis. In contrast, under identical conditions, GF treatment led to marked potentiation of PI hydrolysis downstream of activation of another abundantly expressed G-protein coupled receptor, mGluR5. Quantitative gene expression analysis of GF-treated or control astrocytes by TaqMan RT-PCR indicated that GF treatment did not change gene expression of lpa1 and s1p1, but increased gene expression of s1p5 which is expressed at very low levels in basal conditions. These results suggest that GF differentially affected PLC activation downstream of mGluR5 versus lpR activation and that the changes in mRNA levels of lpRs do not account for marked attenuation of agonist-induced phosphoinositide turnover.


Assuntos
Astrócitos/efeitos dos fármacos , Astrócitos/metabolismo , Córtex Cerebral/efeitos dos fármacos , Córtex Cerebral/metabolismo , Substâncias de Crescimento/farmacologia , Fosfatidilinositóis/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Receptores de Glutamato Metabotrópico/metabolismo , Animais , Células Cultivadas , Relação Dose-Resposta a Droga , Fator de Crescimento Epidérmico/farmacologia , Fator 2 de Crescimento de Fibroblastos/farmacologia , Homeostase/efeitos dos fármacos , Homeostase/fisiologia , Fosfatos de Inositol/metabolismo , Insulina/farmacologia , Fosforilação , Ratos , Ratos Sprague-Dawley , Receptor de Glutamato Metabotrópico 5 , Receptores de Lisofosfolipídeos , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/fisiologia
16.
J Neurochem ; 94(3): 819-27, 2005 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-16033424

RESUMO

Minocycline is neuroprotective in animal models of a number of acute CNS injuries and neurodegenerative diseases. While anti-inflammatory and anti-apoptotic effects of minocycline have been characterized, the molecular basis for the neuroprotective effects of minocycline remains unclear. We report here that minocycline and a number of antioxidant compounds protect mixed neuronal cultures in an oxidative stress assay. To evaluate the role of minocycline's direct antioxidant properties in neuroprotection, we determined potencies for minocycline, other tetracycline antibiotics, and reference antioxidant compounds using a panel of in vitro radical scavenging assays. Data from in vitro rat brain homogenate lipid peroxidation and 2,2-diphenyl-1-picrylhydrazyl (DPPH) radical scavenging assays show that minocycline, in contrast to tetracycline, is an effective antioxidant with radical scavenging potency similar to vitamin E. Our findings suggest that the direct antioxidant activity of minocycline may contribute to its neuroprotective effects in some cell-based assays and animal models of neuronal injury.


Assuntos
Antioxidantes/farmacologia , Ácido Glutâmico/farmacologia , Minociclina/farmacologia , Neurônios/efeitos dos fármacos , Estresse Oxidativo/efeitos dos fármacos , Animais , Antipirina/análogos & derivados , Antipirina/farmacologia , Benzotiazóis , Morte Celular/efeitos dos fármacos , Células Cultivadas , Córtex Cerebral/citologia , Cromanos/farmacologia , Desoxirribose/metabolismo , Maleato de Dizocilpina/farmacologia , Relação Dose-Resposta a Droga , Interações Medicamentosas , Edaravone , Embrião de Mamíferos , Sequestradores de Radicais Livres/farmacologia , Radicais Livres/farmacologia , Concentração Inibidora 50 , L-Lactato Desidrogenase/metabolismo , Peroxidação de Lipídeos/efeitos dos fármacos , Minociclina/química , Fármacos Neuroprotetores/farmacologia , Estresse Oxidativo/fisiologia , Fenetilaminas/metabolismo , Ratos , Ratos Sprague-Dawley , Ácidos Sulfônicos/metabolismo , Vitamina E/farmacologia
17.
Glia ; 45(1): 17-27, 2004 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-14648542

RESUMO

Lysophosphatidic acid (LPA) and sphingosine-1-phosphate (S1P) have been proposed to play a key role in oligodendrocyte maturation and myelinogenesis. In this study, we examined lysophospholipid receptor gene expression in differentiated rat oligodendrocyte cultures and signaling downstream of lysophospholipid receptor activation by LPA and S1P. Differentiated oligodendrocytes express mRNAs encoding lysophospholipid receptors with the relative abundance of lpa1>s1p5>s1p1=s1p2=lpa3>s1p3. LPA and S1P transiently increased phosphorylation of extracellular signal-regulated kinase (ERK) with EC50 values of 956 and 168 nM, respectively. LPA- and S1P-induced ERK phosphorylation was dependent on the activation of mitogen-activated protein kinase (MAPK), phospholipase C (PLC), and protein kinase C (PKC), but was insensitive to pertussis toxin (PTX). LPA increased intracellular calcium levels in oligodendrocytes and these increases were partially blocked by a PLC inhibitor but not by PTX. In contrast, S1P was not found to induce measurable changes of intracellular calcium. These results taken together suggest that lysophospholipid receptor activation involves receptor coupling to heterotrimeric Gq subunits with consequent activation of PLC, PKC, and MAPK pathways leading to ERK phosphorylation.


Assuntos
Córtex Cerebral/efeitos dos fármacos , Lisofosfolipídeos/farmacologia , Oligodendroglia/efeitos dos fármacos , Transdução de Sinais/efeitos dos fármacos , Esfingosina/análogos & derivados , Esfingosina/farmacologia , Animais , Córtex Cerebral/metabolismo , Relação Dose-Resposta a Droga , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Sistema de Sinalização das MAP Quinases/fisiologia , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Oligodendroglia/metabolismo , Ratos , Ratos Sprague-Dawley , Transdução de Sinais/fisiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA