Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Proc Natl Acad Sci U S A ; 118(9)2021 03 02.
Artigo em Inglês | MEDLINE | ID: mdl-33619082

RESUMO

Encephalitis associated with antibodies against the neuronal gamma-aminobutyric acid A receptor (GABAA-R) is a rare form of autoimmune encephalitis. The pathogenesis is still unknown but autoimmune mechanisms were surmised. Here we identified a strongly expanded B cell clone in the cerebrospinal fluid of a patient with GABAA-R encephalitis. We expressed the antibody produced by it and showed by enzyme-linked immunosorbent assay (ELISA) and immunohistochemistry that it recognizes the GABAA-R. Patch-clamp recordings revealed that it tones down inhibitory synaptic transmission and causes increased excitability of hippocampal CA1 pyramidal neurons. Thus, the antibody likely contributed to clinical disease symptoms. Hybridization to a protein array revealed the cross-reactive protein LIM-domain-only protein 5 (LMO5), which is related to cell-cycle regulation and tumor growth. We confirmed LMO5 recognition by immunoprecipitation and ELISA and showed that cerebrospinal fluid samples from two other patients with GABAA-R encephalitis also recognized LMO5. This suggests that cross-reactivity between GABAA-R and LMO5 is frequent in GABAA-R encephalitis and supports the hypothesis of a paraneoplastic etiology.


Assuntos
Antígenos de Neoplasias/imunologia , Autoanticorpos/imunologia , Reações Cruzadas/imunologia , Suscetibilidade a Doenças , Encefalite/etiologia , Receptores de GABA-A/imunologia , Autoantígenos/imunologia , Doenças Autoimunes do Sistema Nervoso/etiologia , Doenças Autoimunes do Sistema Nervoso/metabolismo , Autoimunidade , Linfócitos B/imunologia , Linfócitos B/metabolismo , Biomarcadores , Suscetibilidade a Doenças/imunologia , Encefalite/metabolismo , Encefalite/patologia , Humanos , Células Piramidais/imunologia , Células Piramidais/metabolismo
2.
Int J Mol Sci ; 18(9)2017 Sep 08.
Artigo em Inglês | MEDLINE | ID: mdl-28885567

RESUMO

Acquired epilepsies can arise as a consequence of brain injury and result in unprovoked seizures that emerge after a latent period of epileptogenesis. These epilepsies pose a major challenge to clinicians as they are present in the majority of patients seen in a common outpatient epilepsy clinic and are prone to pharmacoresistance, highlighting an unmet need for new treatment strategies. Metabolic and homeostatic changes are closely linked to seizures and epilepsy, although, surprisingly, no potential treatment targets to date have been translated into clinical practice. We summarize here the current knowledge about metabolic and homeostatic changes in seizures and acquired epilepsy, maintaining a particular focus on mitochondria, calcium dynamics, reactive oxygen species and key regulators of cellular metabolism such as the Nrf2 pathway. Finally, we highlight research gaps that will need to be addressed in the future which may help to translate these findings into clinical practice.


Assuntos
Cálcio/metabolismo , Metabolismo Energético , Epilepsia/metabolismo , Homeostase , Mitocôndrias/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Convulsões/metabolismo , Trifosfato de Adenosina/metabolismo , Animais , Canais de Cálcio/metabolismo , Morte Celular , Membrana Celular/metabolismo , Retículo Endoplasmático/metabolismo , Epilepsia/etiologia , Humanos , Fator 2 Relacionado a NF-E2/metabolismo , Estresse Oxidativo , Pesquisa , Convulsões/etiologia
3.
Sci Transl Med ; 8(362): 362ra146, 2016 10 26.
Artigo em Inglês | MEDLINE | ID: mdl-27797962

RESUMO

In inflammation-associated progressive neuroinflammatory disorders, such as multiple sclerosis (MS), inflammatory infiltrates containing T helper 1 (TH1) and TH17 cells cause demyelination and neuronal degeneration. Regulatory T cells (Treg) control the activation and infiltration of autoreactive T cells into the central nervous system (CNS). In MS and experimental autoimmune encephalomyelitis (EAE) in mice, Treg function is impaired. We show that a recently approved drug, Nle4-d-Phe7-α-melanocyte-stimulating hormone (NDP-MSH), induced functional Treg, resulting in amelioration of EAE progression in mice. NDP-MSH also prevented immune cell infiltration into the CNS by restoring the integrity of the blood-brain barrier. NDP-MSH exerted long-lasting neuroprotective effects in mice with EAE and prevented excitotoxic death and reestablished action potential firing in mouse and human neurons in vitro. Neuroprotection by NDP-MSH was mediated via signaling through the melanocortin-1 and orphan nuclear 4 receptors in mouse and human neurons. NDP-MSH may be of benefit in treating neuroinflammatory diseases such as relapsing-remitting MS and related disorders.


Assuntos
Encefalomielite Autoimune Experimental/tratamento farmacológico , Fármacos Neuroprotetores/farmacologia , Receptor Tipo 1 de Melanocortina/metabolismo , alfa-MSH/análogos & derivados , Potenciais de Ação , Animais , Barreira Hematoencefálica , Células da Medula Óssea/metabolismo , Proliferação de Células , Sistema Nervoso Central/imunologia , Progressão da Doença , Citometria de Fluxo , Perfilação da Expressão Gênica , Ácido Glutâmico/química , Hipocampo/metabolismo , Imageamento por Ressonância Magnética , Camundongos , Camundongos Endogâmicos C57BL , Neurônios/metabolismo , Ligação Proteica , Receptor Tipo 1 de Melanocortina/genética , Linfócitos T Reguladores/citologia , alfa-MSH/farmacologia
4.
Eur J Immunol ; 45(9): 2602-14, 2015 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-26140335

RESUMO

Natural killer (NK) cells are a subset of cytotoxic lymphocytes that recognize and kill tumor- and virus-infected cells without prior stimulation. Killing of target cells is a multistep process including adhesion to target cells, formation of an immunological synapse, and polarization and release of cytolytic granules. The role of distinct potassium channels in this orchestrated process is still poorly understood. The current study reveals that in addition to the voltage-gated KV 1.3 and the calcium-activated KCa 3.1 channels, human NK cells also express the two-pore domain K2 P channel TASK2 (TWIK-related acid-sensitive potassium channel). Expression of Task2 varies among NK-cell subsets and depends on their differentiation and activation state. Despite its different expression in TASK2(high) CD56(bright) CD16(-) and TASK2(low) CD56(dim) CD16(+) NK cells, TASK2 is involved in cytokine-induced proliferation and cytolytic function of both subsets. TASK2 is crucial for leukocyte functional antigen (LFA-1) mediated adhesion of both resting and cytokine-activated NK cells to target cells, an early step in killing of target cells. With regard to the following mechanism, TASK2 plays a role in release of cytotoxic granules by resting, but not IL-15-induced NK cells. Taken together, our data exhibit two-pore potassium channels as important players in NK-cell activation and effector function.


Assuntos
Citotoxicidade Imunológica , Sinapses Imunológicas/metabolismo , Células Matadoras Naturais/imunologia , Antígeno-1 Associado à Função Linfocitária/imunologia , Canais de Potássio de Domínios Poros em Tandem/imunologia , Antígeno CD56/genética , Antígeno CD56/imunologia , Adesão Celular/efeitos dos fármacos , Diferenciação Celular , Linhagem Celular Tumoral , Proliferação de Células , Técnicas de Cocultura , Proteínas Ligadas por GPI/genética , Proteínas Ligadas por GPI/imunologia , Expressão Gênica , Células HEK293 , Humanos , Interleucina-15/farmacologia , Células K562 , Células Matadoras Naturais/citologia , Células Matadoras Naturais/efeitos dos fármacos , Células Matadoras Naturais/metabolismo , Ativação Linfocitária/efeitos dos fármacos , Antígeno-1 Associado à Função Linfocitária/genética , Antígeno-1 Associado à Função Linfocitária/metabolismo , Canais de Potássio de Domínios Poros em Tandem/genética , Canais de Potássio de Domínios Poros em Tandem/metabolismo , Cultura Primária de Células , Receptores de IgG/genética , Receptores de IgG/imunologia , Transdução de Sinais , Análise de Célula Única
5.
Stroke ; 45(11): 3395-402, 2014 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-25236871

RESUMO

BACKGROUND AND PURPOSE: Brain injury during stroke results in oxidative stress and the release of factors that include extracellular Ca(2+), hydrogen peroxide, adenosine diphosphate ribose, and nicotinic acid adenine dinucleotide phosphate. These alterations of the extracellular milieu change the activity of transient receptor potential melastatin subfamily member 2 (TRPM2), a nonselective cation channel expressed in the central nervous system and the immune system. Our goal was to evaluate the contribution of TRPM2 to the tissue damage after stroke. METHODS: In accordance with current quality guidelines, we independently characterized Trpm2 in a murine ischemic stroke model in 2 different laboratories. RESULTS: Gene deficiency of Trpm2 resulted in significantly improved neurological outcome and decreased infarct size. Besides an already known moderate neuroprotective effect of Trpm2 deficiency in vitro, ischemic brain invasion by neutrophils and macrophages was particularly reduced in Trpm2-deficient mice. Bone marrow chimeric mice revealed that Trpm2 deficiency in the peripheral immune system is responsible for the protective phenotype. Furthermore, experiments with mixed bone marrow chimeras demonstrated that Trpm2 is essential for the migration of neutrophils and, to a lesser extent, also of macrophages into ischemic hemispheres. Notably, the pharmacological TRPM2 inhibitor, N-(p-amylcinnamoyl)anthranilic acid, was equally protective in the stroke model. CONCLUSIONS: Although a neuroprotective effect of TRPM2 in vitro is well known, we can show for the first time that the detrimental role of TRPM2 in stroke primarily depends on its role in activating peripheral immune cells. Targeting TRPM2 systemically represents a promising therapeutic approach for ischemic stroke.


Assuntos
Isquemia Encefálica/imunologia , Movimento Celular/imunologia , Imunidade Celular/imunologia , Acidente Vascular Cerebral/imunologia , Canais de Cátion TRPM/fisiologia , Animais , Isquemia Encefálica/patologia , Células Cultivadas , Hipocampo/imunologia , Hipocampo/patologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Técnicas de Cultura de Órgãos , Distribuição Aleatória , Acidente Vascular Cerebral/patologia
6.
FASEB J ; 28(8): 3435-45, 2014 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-24744146

RESUMO

CD4(+) T cells expressing the immunotolerizing molecule HLA-G have been described as a unique human thymus-derived regulatory T (tTreg) cell subset involved in immunoregulation and parenchymal homeostasis during infectious and autoimmune inflammation. We compared properties and molecular characteristics of human CD4(+)HLA-G(+) with those of CD4(+)CD25(+)FoxP3-expressing tTreg cells using in vitro studies of T-cell receptor (TCR) signaling, single-cell electrophysiology, and functional in vivo studies. Both tTreg populations are characterized by alterations in proximal-signaling pathways on TCR stimulation and a hyperpolarization of the plasma membrane when compared to conventional CD4(+) T cells. However, both clearly differ in phenotype and pattern of secreted cytokines, which results in distinct mechanisms of suppression: While CD4(+)HLA-G(+) cells secrete high levels of inhibitory molecules (IL-10, soluble HLA-G, IL-35), CD4(+)CD25(+)FoxP3(+) cells express these molecules at significantly lower levels and seem to exert their function mainly in a contact-dependent manner via cyclic adenosine-monophosphate. Finally we demonstrate that human CD4(+)HLA-G(+) tTreg cells significantly ameliorated graft-versus-host disease in a humanized mouse model as a first proof of their in vivo relevance. Our data further characterize and establish CD4(+)HLA-G(+) cells as a potent human tTreg population that can modulate polyclonal adaptive immune responses in vivo and thus being a promising candidate for potential clinical applications in the future.


Assuntos
Transferência Adotiva , Linfócitos T CD4-Positivos/imunologia , Doença Enxerto-Hospedeiro/prevenção & controle , Terapia de Imunossupressão/métodos , Leucócitos Mononucleares/transplante , Subpopulações de Linfócitos T/imunologia , Linfócitos T Reguladores/imunologia , Animais , Linfócitos T CD4-Positivos/transplante , Sinalização do Cálcio , Citocinas/biossíntese , Citocinas/metabolismo , Fatores de Transcrição Forkhead/análise , Doença Enxerto-Hospedeiro/imunologia , Antígenos HLA-G/imunologia , Xenoenxertos , Humanos , Subunidade alfa de Receptor de Interleucina-2/análise , Ativação Linfocitária , Potenciais da Membrana , Camundongos , Camundongos Endogâmicos NOD , Organismos Livres de Patógenos Específicos , Subpopulações de Linfócitos T/transplante , Linfócitos T Reguladores/transplante
7.
Nat Med ; 19(9): 1161-5, 2013 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-23933981

RESUMO

The blood-brain barrier (BBB) is an integral part of the neurovascular unit (NVU). The NVU is comprised of endothelial cells that are interconnected by tight junctions resting on a parenchymal basement membrane ensheathed by pericytes, smooth muscle cells and a layer of astrocyte end feet. Circulating blood cells, such as leukocytes, complete the NVU. BBB disruption is common in several neurological diseases, but the molecular mechanisms involved remain largely unknown. We analyzed the role of TWIK-related potassium channel-1 (TREK1, encoded by KCNK2) in human and mouse endothelial cells and the BBB. TREK1 was downregulated in endothelial cells by treatment with interferon-γ (IFN-γ) and tumor necrosis factor-α (TNF-α). Blocking TREK1 increased leukocyte transmigration, whereas TREK1 activation had the opposite effect. We identified altered mitogen-activated protein (MAP) kinase signaling, actin remodeling and upregulation of cellular adhesion molecules as potential mechanisms of increased migration in TREK1-deficient (Kcnk2(-/-)) cells. In Kcnk2(-/-) mice, brain endothelial cells showed an upregulation of the cellular adhesion molecules ICAM1, VCAM1 and PECAM1 and facilitated leukocyte trafficking into the CNS. Following the induction of experimental autoimmune encephalomyelitis (EAE) by immunization with a myelin oligodendrocyte protein (MOG)35-55 peptide, Kcnk2(-/-) mice showed higher EAE severity scores that were accompanied by increased cellular infiltrates in the central nervous system (CNS). The severity of EAE was attenuated in mice given the amyotrophic lateral sclerosis drug riluzole or fed a diet enriched with linseed oil (which contains the TREK-1 activating omega-3 fatty acid α-linolenic acid). These beneficial effects were reduced in Kcnk2(-/-) mice, suggesting TREK-1 activating compounds may be used therapeutically to treat diseases related to BBB dysfunction.


Assuntos
Barreira Hematoencefálica/metabolismo , Encéfalo/metabolismo , Células Endoteliais/metabolismo , Canais de Potássio de Domínios Poros em Tandem/metabolismo , Actinas/metabolismo , Animais , Anticonvulsivantes/farmacologia , Barreira Hematoencefálica/imunologia , Encéfalo/imunologia , Moléculas de Adesão Celular/biossíntese , Moléculas de Adesão Celular/imunologia , Movimento Celular , Células Cultivadas , Técnicas de Cocultura , Células Dendríticas , Regulação para Baixo , Encefalomielite Autoimune Experimental/tratamento farmacológico , Encefalomielite Autoimune Experimental/metabolismo , Feminino , Células HEK293 , Humanos , Molécula 1 de Adesão Intercelular/imunologia , Molécula 1 de Adesão Intercelular/metabolismo , Interferon-alfa/farmacologia , Leucócitos/metabolismo , Óleo de Semente do Linho/administração & dosagem , Sistema de Sinalização das MAP Quinases , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Glicoproteína Mielina-Oligodendrócito , Fragmentos de Peptídeos , Canais de Potássio de Domínios Poros em Tandem/genética , Riluzol/farmacologia , Migração Transendotelial e Transepitelial
8.
Am J Pathol ; 177(6): 3051-60, 2010 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-21037084

RESUMO

Glatiramer acetate (GA) is a synthetic, random, basic copolymer capable of modulating adaptive T cell responses. In animal models of various inflammatory and degenerative central nervous system disorders, GA-induced T cells cross the blood-brain barrier, secrete high levels of anti-inflammatory cytokines and neurotrophins, and thus both reduce neuronal damage and promote neurogenesis. Recently, it has been suggested that GA itself may permeate the (impaired) blood-brain-barrier and directly protect neurons under conditions of inflammation-mediated neurodegeneration. To test this hypothesis, we examined the direct effects of GA on neuronal functionality and T cell-mediated neuronal apoptosis in culture, acute brain slices, and focal experimental autoimmune encephalomyelitis. GA caused a depolarization of the resting membrane potential and led to an immediate impairment of action potential generation in neurons. Moreover, GA-incubated neurons underwent dose-dependent apoptosis. Apoptosis of ovalbumin peptide-loaded major histocompatibility complex class I-expressing neurons induced by ovalbumin-specific effector T cells could be reduced by pre-incubation of T cells, but not neurons with GA. Similar results could be found using acute brain slices. In focal experimental autoimmune encephalomyelitis, lesion size and neuronal apoptosis could be limited by pretreating rats with GA, whereas intracerebral GA application into the inflammatory lesion had no effect on neuronal survival. Our data suggest that GA attenuates adaptive pro-inflammatory T cell responses, but does not exert direct neuroprotective effects.


Assuntos
Inflamação/patologia , Neurônios/efeitos dos fármacos , Peptídeos/farmacologia , Linfócitos T/efeitos dos fármacos , Animais , Morte Celular/efeitos dos fármacos , Morte Celular/imunologia , Células Cultivadas , Citoproteção/efeitos dos fármacos , Citoproteção/imunologia , Regulação para Baixo/efeitos dos fármacos , Regulação para Baixo/imunologia , Avaliação Pré-Clínica de Medicamentos , Embrião de Mamíferos , Encefalomielite Autoimune Experimental/imunologia , Encefalomielite Autoimune Experimental/metabolismo , Encefalomielite Autoimune Experimental/patologia , Acetato de Glatiramer , Inflamação/imunologia , Inflamação/metabolismo , Mediadores da Inflamação/metabolismo , Potenciais da Membrana/efeitos dos fármacos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Neurônios/imunologia , Neurônios/metabolismo , Neurônios/patologia , Ratos , Ratos Transgênicos , Linfócitos T/imunologia , Linfócitos T/metabolismo
9.
Glia ; 58(4): 469-80, 2010 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-19780193

RESUMO

Demyelination and death of oligodendrocytes accompanied by transection of neurites and neuronal apoptosis are pathological hallmarks of cortical and subcortical gray matter lesions in demyelinating viral and autoimmune inflammatory CNS disorders. In these disorders, leukocortical lesions, containing the perikarya of most efferent neurons, display pronounced infiltration by CD8(+) T cells of putative specificity for oligodendrocyte- and myelin-related antigens. Hence, neuronal apoptosis in gray matter lesions may be a collateral effect of an oligodendrocyte-directed attack by CD8(+) T cells. To challenge this hypothesis, we transferred activated antigen-specific CD8(+) T cells (OT-I T cells) into acute coronal brain slices from mice selectively expressing ovalbumin as a cytosolic neo-self-antigen in oligodendrocytes (ODC-OVA mice). We studied mechanisms and kinetics of oligodendroglial and neuronal apoptosis in the neocortex and hippocampus, using multicolor staining for different cell types and activated caspase-3. Within the gray matter, a single OT-I T cell caused simultaneous caspase-3 activation in about 30 ODCs and 10 neurons within 6 h in a strictly antigen-dependent manner. Experiments with OT-I T cells genetically deficient for perforin or the granzyme B-cluster and with blocking anti-FasL antibodies as well as proinflammatory cytokines revealed, that collateral apoptosis of neurons was likely due to a spillover of perforin and granzyme(s) from the OT-I T cell itself or the immunological synapse that it selectively formed with antigen-presenting oligodendrocytes. Collateral neuronal apoptosis could contribute to substantial neuronal loss in gray matter lesions and cause persistent neurological impairment in both acute and chronic gray matter lesions in various inflammatory CNS disorders.


Assuntos
Apoptose/fisiologia , Encéfalo/fisiologia , Antígenos CD8/metabolismo , Neurônios/fisiologia , Oligodendroglia/fisiologia , Linfócitos T/fisiologia , Animais , Caspase 3/metabolismo , Movimento Celular , Citocinas/metabolismo , Proteína Ligante Fas/metabolismo , Granzimas/genética , Granzimas/metabolismo , Hipocampo/fisiologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Neocórtex/fisiologia , Fibras Nervosas Amielínicas/fisiologia , Proteínas Citotóxicas Formadoras de Poros/genética , Proteínas Citotóxicas Formadoras de Poros/metabolismo , Serpinas/metabolismo
10.
J Neuroimmunol ; 194(1-2): 62-9, 2008 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-18164423

RESUMO

Multiple sclerosis is a chronic disabling CNS disorder, characterized by autoimmune inflammatory demyelination and neurodegeneration. CD200, broadly expressed on neurons and endothelial cells, mediates inhibitory signals through its receptor, CD200R, on cells of myeloid origin. Antibody-mediated blockade of CD200R leads to an aggravated clinical course of rodent experimental autoimmune encephalomyelitis in vivo, accompanied by profoundly augmented cellular infiltrates consisting of T cells and activated iNOS(+) macrophages in inflammatory spinal cord lesions. In vitro blockade of CD200R on macrophages leads to enhanced IFN-gamma-induced release of IL6 and neuronal cell death in co-cultures with hippocampal neurons expressing CD200. CD200 and its receptor could also be detected on neurons and macrophages in human MS plaques. Therefore the CD200-CD200R pathway seems of critical relevance for macrophage-mediated damage in autoimmune inflammation of the CNS.


Assuntos
Antígenos CD/fisiologia , Encefalomielite Autoimune Experimental/patologia , Macrófagos Peritoneais/fisiologia , Glicoproteínas de Membrana/fisiologia , Esclerose Múltipla/metabolismo , Receptores Imunológicos/fisiologia , Animais , Anticorpos/farmacologia , Anticorpos/uso terapêutico , Antígenos CD/análise , Antígenos de Superfície/análise , Antígenos de Superfície/fisiologia , Apoptose , Células Cultivadas/metabolismo , Células Cultivadas/patologia , Técnicas de Cocultura , Citocinas/biossíntese , Encefalomielite Autoimune Experimental/metabolismo , Hipocampo/citologia , Humanos , Interferon gama/fisiologia , Interleucina-6/metabolismo , Macrófagos Peritoneais/química , Glicoproteínas de Membrana/biossíntese , Camundongos , Camundongos Endogâmicos C57BL , Pessoa de Meia-Idade , Esclerose Múltipla/patologia , Neurônios/química , Neurônios/patologia , Receptores de Orexina , Ratos , Ratos Long-Evans , Receptores de Superfície Celular/análise , Receptores de Superfície Celular/fisiologia , Receptores Imunológicos/antagonistas & inibidores , Receptores Imunológicos/biossíntese , Receptores Imunológicos/imunologia , Subpopulações de Linfócitos T/imunologia
11.
J Neurooncol ; 87(3): 263-70, 2008 May.
Artigo em Inglês | MEDLINE | ID: mdl-18217213

RESUMO

Two-pore domain K(+) channels, a recently discovered family of ion channels with a unique membrane topology, have been shown to be critically involved in cell death. We here address the functional role of TASK3 (TWIK-related acid-sensitive K(+) channel, KCNK9) in human glioblastoma in vitro and in vivo. Human glioma cell lines (n = 5) as well as glioma specimens (n = 5) constitutively express TASK3 mRNA and protein. The functional impact of the potassium channel on cell survival was investigated using a medium with high (25 mM) extracellular potassium over 7 days. Using flow cytometric assessment, we show that under these culture conditions 97 +/- 0.76% of all glioma cells survived. Application of the TASK channel opener isoflurane (1 vol%) resulted in a 30 +/- 4% reduction of cell survival in different glioma cell lines. Simultaneous application of isoflurane and the TASK channel blockers bupivacaine (20 microM) and spermine (500 microM) completely reversed this effect. Our results demonstrate the expression of TASK3 in glioma cells in vitro and in vivo and provide a direct link between the TASK3 channel function and glioma cell survival. This implies that TASK3 channels may possibly represent a novel molecular target for the treatment of this type of cancer.


Assuntos
Neoplasias Encefálicas/metabolismo , Glioma/metabolismo , Canais de Potássio de Domínios Poros em Tandem/biossíntese , Neoplasias Encefálicas/patologia , Morte Celular/fisiologia , Citometria de Fluxo , Glioma/patologia , Humanos , Imuno-Histoquímica , Proteínas do Tecido Nervoso/biossíntese , RNA Mensageiro/análise , Reação em Cadeia da Polimerase Via Transcriptase Reversa
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA