Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Biomolecules ; 13(12)2023 11 23.
Artigo em Inglês | MEDLINE | ID: mdl-38136568

RESUMO

Elevated levels of saturated very long-chain fatty acids (VLCFAs) in cell membranes and secreted lipoparticles have been associated with neurotoxicity and, therefore, require tight regulation. Excessive VLCFAs are imported into peroxisomes for degradation by ß-oxidation. Impaired VLCFA catabolism due to primary or secondary peroxisomal alterations is featured in neurodegenerative and neuroinflammatory disorders such as X-linked adrenoleukodystrophy and multiple sclerosis (MS). Here, we identified that healthy human macrophages upregulate the peroxisomal genes involved in ß-oxidation during myelin phagocytosis and pro-inflammatory activation, and that this response is impaired in peripheral macrophages and phagocytes in brain white matter lesions in MS patients. The pharmacological targeting of VLCFA metabolism and peroxisomes in innate immune cells could be favorable in the context of neuroinflammation and neurodegeneration. We previously identified the epigenetic histone deacetylase (HDAC) inhibitors entinostat and vorinostat to enhance VLCFA degradation and pro-regenerative macrophage polarization. However, adverse side effects currently limit their use in chronic neuroinflammation. Here, we focused on tefinostat, a monocyte/macrophage-selective HDAC inhibitor that has shown reduced toxicity in clinical trials. By using a gene expression analysis, peroxisomal ß-oxidation assay, and live imaging of primary human macrophages, we assessed the efficacy of tefinostat in modulating VLCFA metabolism, phagocytosis, chemotaxis, and immune function. Our results revealed the significant stimulation of VLCFA degradation with the upregulation of genes involved in peroxisomal ß-oxidation and interference with immune cell recruitment; however, tefinostat was less potent than the class I HDAC-selective inhibitor entinostat in promoting a regenerative macrophage phenotype. Further research is needed to fully explore the potential of class I HDAC inhibition and downstream targets in the context of neuroinflammation.


Assuntos
Transportadores de Cassetes de Ligação de ATP , Inibidores de Histona Desacetilases , Humanos , Inibidores de Histona Desacetilases/farmacologia , Transportadores de Cassetes de Ligação de ATP/metabolismo , Doenças Neuroinflamatórias , Ácidos Graxos/metabolismo , Membro 1 da Subfamília D de Transportadores de Cassetes de Ligação de ATP , Ácidos Graxos não Esterificados , Macrófagos/metabolismo , Imunidade
2.
Netw Neurosci ; 5(2): 337-357, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34189368

RESUMO

Identifying the nodes able to drive the state of a network is crucial to understand, and eventually control, biological systems. Despite recent advances, such identification remains difficult because of the huge number of equivalent controllable configurations, even in relatively simple networks. Based on the evidence that in many applications it is essential to test the ability of individual nodes to control a specific target subset, we develop a fast and principled method to identify controllable driver-target configurations in sparse and directed networks. We demonstrate our approach on simulated networks and experimental gene networks to characterize macrophage dysregulation in human subjects with multiple sclerosis.

3.
Mol Neurobiol ; 58(2): 470-482, 2021 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-32974731

RESUMO

Multiple sclerosis (MS) is a neuroinflammatory disease whose pathogenesis remains unclear. Lysophosphatidic acid (LPA) is an endogenous phospholipid involved in multiple immune cell functions and dysregulated in MS. Its receptor LPA1 is expressed in macrophages and regulates their activation, which is of interest due to the role of macrophage activation in MS in both destruction and repair. In this study, we studied the genetic deletion and pharmaceutical inhibition of LPA1 in the mouse MS model, experimental autoimmune encephalomyelitis (EAE). LPA1 expression was analyzed in EAE mice and MS patient immune cells. The effect of LPA and LPA1 on macrophage activation was studied in human monocyte-derived macrophages. We show that lack of LPA1 activity induces milder clinical EAE course and that Lpar1 expression in peripheral blood mononuclear cells (PBMC) correlates with onset of relapses and severity in EAE. We see the same over-expression in PBMC from MS patients during relapse compared with progressive forms of the disease and in stimulated monocyte-derived macrophages. LPA induced a proinflammatory-like response in macrophages through LPA1, providing a plausible way in which LPA and LPA1 dysregulation can lead to the inflammation in MS. These data show a new mechanism of LPA signaling in the MS pathogenesis, prompting further research into its use as a therapeutic target biomarker.


Assuntos
Lisofosfolipídeos/metabolismo , Ativação de Macrófagos , Macrófagos/metabolismo , Esclerose Múltipla/metabolismo , Receptores de Ácidos Lisofosfatídicos/metabolismo , Adolescente , Adulto , Idoso , Animais , Polaridade Celular , Sistema Nervoso Central/metabolismo , Sistema Nervoso Central/patologia , Encefalomielite Autoimune Experimental/metabolismo , Encefalomielite Autoimune Experimental/patologia , Feminino , Humanos , Inflamação/patologia , Camundongos Endogâmicos C57BL , Pessoa de Meia-Idade , Monócitos/patologia , PPAR gama/metabolismo , Fenótipo , Receptores de Ácidos Lisofosfatídicos/antagonistas & inibidores , Recidiva , Adulto Jovem
5.
Acta Neuropathol ; 138(3): 457-476, 2019 09.
Artigo em Inglês | MEDLINE | ID: mdl-31011859

RESUMO

Schwann cells (SC) enter the central nervous system (CNS) in pathophysiological conditions. However, how SC invade the CNS to remyelinate central axons remains undetermined. We studied SC migratory behavior ex vivo and in vivo after exogenous transplantation in the demyelinated spinal cord. The data highlight for the first time that SC migrate preferentially along blood vessels in perivascular extracellular matrix (ECM), avoiding CNS myelin. We demonstrate in vitro and in vivo that this migration route occurs by virtue of a dual mode of action of Eph/ephrin signaling. Indeed, EphrinB3, enriched in myelin, interacts with SC Eph receptors, to drive SC away from CNS myelin, and triggers their preferential adhesion to ECM components, such as fibronectin via integrinß1 interactions. This complex interplay enhances SC migration along the blood vessel network and together with lesion-induced vascular remodeling facilitates their timely invasion of the lesion site. These novel findings elucidate the mechanism by which SC invade and contribute to spinal cord repair.


Assuntos
Vasos Sanguíneos , Movimento Celular/fisiologia , Efrina-B3/metabolismo , Remielinização/fisiologia , Células de Schwann/fisiologia , Medula Espinal/metabolismo , Animais , Doenças Desmielinizantes/patologia , Feminino , Fibronectinas/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Transdução de Sinais/fisiologia , Medula Espinal/patologia
6.
J Neurosci ; 37(24): 5885-5899, 2017 06 14.
Artigo em Inglês | MEDLINE | ID: mdl-28522736

RESUMO

Remyelination of CNS axons by Schwann cells (SCs) is not efficient, in part due to the poor migration of SCs into the adult CNS. Although it is known that migrating SCs avoid white matter tracts, the molecular mechanisms underlying this exclusion have never been elucidated. We now demonstrate that myelin-associated glycoprotein (MAG), a well known inhibitor of neurite outgrowth, inhibits rat SC migration and induces their death via γ-secretase-dependent regulated intramembrane proteolysis of the p75 neurotrophin receptor (also known as p75 cleavage). Blocking p75 cleavage using inhibitor X (Inh X), a compound that inhibits γ-secretase activity before exposing to MAG or CNS myelin improves SC migration and survival in vitro Furthermore, mouse SCs pretreated with Inh X migrate extensively in the demyelinated mouse spinal cord and remyelinate axons. These results suggest a novel role for MAG/myelin in poor SC-myelin interaction and identify p75 cleavage as a mechanism that can be therapeutically targeted to enhance SC-mediated axon remyelination in the adult CNS.SIGNIFICANCE STATEMENT Numerous studies have used Schwann cells, the myelin-making cells of the peripheral nervous system to remyelinate adult CNS axons. Indeed, these transplanted cells successfully remyelinate axons, but unfortunately they do not migrate far and so remyelinate only a few axons in the vicinity of the transplant site. It is believed that if Schwann cells could be induced to migrate further and survive better, they may represent a valid therapy for remyelination. We show that myelin-associated glycoprotein or CNS myelin, in general, inhibit rodent Schwann cell migration and induce their death via cleavage of the neurotrophin receptor p75. Blockade of p75 cleavage using a specific inhibitor significantly improves migration and survival of the transplanted Schwann cells in vivo.


Assuntos
Apoptose/fisiologia , Movimento Celular/fisiologia , Glicoproteína Associada a Mielina/metabolismo , Crescimento Neuronal/fisiologia , Células de Schwann/citologia , Células de Schwann/fisiologia , Animais , Células Cultivadas , Feminino , Camundongos , Camundongos Nus , Bainha de Mielina/metabolismo
7.
Brain ; 140(4): 967-980, 2017 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-28334918

RESUMO

One major challenge in multiple sclerosis is to understand the cellular and molecular mechanisms leading to disease severity progression. The recently demonstrated correlation between disease severity and remyelination emphasizes the importance of identifying factors leading to a favourable outcome. Why remyelination fails or succeeds in multiple sclerosis patients remains largely unknown, mainly because remyelination has never been studied within a humanized pathological context that would recapitulate major events in plaque formation such as infiltration of inflammatory cells. Therefore, we developed a new paradigm by grafting healthy donor or multiple sclerosis patient lymphocytes in the demyelinated lesion of nude mice spinal cord. We show that lymphocytes play a major role in remyelination whose efficacy is significantly decreased in mice grafted with multiple sclerosis lymphocytes compared to those grafted with healthy donors lymphocytes. Mechanistically, we demonstrated in vitro that lymphocyte-derived mediators influenced differentiation of oligodendrocyte precursor cells through a crosstalk with microglial cells. Among mice grafted with lymphocytes from different patients, we observed diverse remyelination patterns reproducing for the first time the heterogeneity observed in multiple sclerosis patients. Comparing lymphocyte secretory profile from patients exhibiting high and low remyelination ability, we identified novel molecules involved in oligodendrocyte precursor cell differentiation and validated CCL19 as a target to improve remyelination. Specifically, exogenous CCL19 abolished oligodendrocyte precursor cell differentiation observed in patients with high remyelination pattern. Multiple sclerosis lymphocytes exhibit intrinsic capacities to coordinate myelin repair and further investigation on patients with high remyelination capacities will provide new pro-regenerative strategies.


Assuntos
Imunidade Adaptativa/fisiologia , Doenças Desmielinizantes/imunologia , Bainha de Mielina/imunologia , Adolescente , Adulto , Idoso , Animais , Transplante de Células , Quimiocina CCL19/imunologia , Feminino , Humanos , Linfócitos/imunologia , Lisofosfatidilcolinas/farmacologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Nus , Pessoa de Meia-Idade , Esclerose Múltipla/imunologia , Esclerose Múltipla/patologia , Células-Tronco Neurais/imunologia , Oligodendroglia/imunologia , Oligodendroglia/patologia , Adulto Jovem
8.
Stem Cells ; 34(4): 984-96, 2016 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-26676415

RESUMO

Pelizaeus-Merzbacher disease (PMD) results from an X-linked misexpression of proteolipid protein 1 (PLP1). This leukodystrophy causes severe hypomyelination with progressive inflammation, leading to neurological dysfunctions and shortened life expectancy. While no cure exists for PMD, experimental cell-based therapy in the dysmyelinated shiverer model suggested that human oligodendrocyte progenitor cells (hOPCs) or human neural precursor cells (hNPCs) are promising candidates to treat myelinopathies. However, the fate and restorative advantages of human NPCs/OPCs in a relevant model of PMD has not yet been addressed. Using a model of Plp1 overexpression, resulting in demyelination with progressive inflammation, we compared side-by-side the therapeutic benefits of intracerebrally grafted hNPCs and hOPCs. Our findings reveal equal integration of the donor cells within presumptive white matter tracks. While the onset of exogenous remyelination was earlier in hOPCs-grafted mice than in hNPC-grafted mice, extended lifespan occurred only in hNPCs-grafted animals. This improved survival was correlated with reduced neuroinflammation (microglial and astrocytosis loads) and microglia polarization toward M2-like phenotype followed by remyelination. Thus modulation of neuroinflammation combined with myelin restoration is crucial to prevent PMD pathology progression and ensure successful rescue of PMD mice. These findings should help to design novel therapeutic strategies combining immunomodulation and stem/progenitor cell-based therapy for disorders associating hypomyelination with inflammation as observed in PMD.


Assuntos
Imunidade Inata , Inflamação/terapia , Células-Tronco Neurais/transplante , Oligodendroglia/transplante , Doença de Pelizaeus-Merzbacher/terapia , Animais , Doenças Desmielinizantes/imunologia , Doenças Desmielinizantes/patologia , Regulação da Expressão Gênica no Desenvolvimento , Humanos , Imunomodulação , Inflamação/imunologia , Inflamação/patologia , Camundongos , Microglia/imunologia , Microglia/patologia , Proteína Proteolipídica de Mielina/biossíntese , Bainha de Mielina/metabolismo , Células-Tronco Neurais/imunologia , Oligodendroglia/imunologia , Doença de Pelizaeus-Merzbacher/imunologia , Doença de Pelizaeus-Merzbacher/patologia , Regeneração
9.
Stem Cells ; 33(6): 2011-24, 2015 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-25786382

RESUMO

It has been proposed that the adult dorsal root ganglia (DRG) harbor neural stem/progenitor cells (NPCs) derived from the neural crest. However, the thorough characterization of their stemness and differentiation plasticity was not addressed. In this study, we investigated adult DRG-NPC stem cell properties overtime, and their fate when ectopically grafted in the central nervous system. We compared them in vitro and in vivo to the well-characterized adult spinal cord-NPCs derived from the same donors. Using micro-dissection and neurosphere cultures, we demonstrate that adult DRG-NPCs have quasi unlimited self-expansion capacities without compromising their tissue specific molecular signature. Moreover, they differentiate into multiple peripheral lineages in vitro. After transplantation, adult DRG-NPCs generate pericytes in the developing forebrain but remyelinating Schwann cells in response to spinal cord demyelination. In addition, we show that axonal and endothelial/astrocytic factors as well astrocytes regulate the fate of adult DRG-NPCs in culture. Although the adult DRG-NPC multipotency is restricted to the neural crest lineage, their dual responsiveness to developmental and lesion cues highlights their impressive adaptive and repair potentials making them valuable targets for regenerative medicine.


Assuntos
Diferenciação Celular/fisiologia , Doenças Desmielinizantes/patologia , Gânglios Espinais/citologia , Bainha de Mielina/metabolismo , Pericitos/citologia , Células de Schwann/citologia , Células-Tronco Adultas/citologia , Animais , Células Cultivadas , Doenças Desmielinizantes/terapia , Gânglios Espinais/metabolismo , Camundongos Endogâmicos C57BL , Camundongos Nus , Regeneração Nervosa/fisiologia , Crista Neural/citologia , Neurônios/citologia
10.
PLoS One ; 7(9): e42667, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22984406

RESUMO

Schwann cell (SC) transplantation is currently being discussed as a strategy that may promote functional recovery in patients with multiple sclerosis (MS) and other inflammatory demyelinating diseases of the central nervous system (CNS). However this assumes they will not only survive but also remyelinate demyelinated axons in the chronically inflamed CNS. To address this question we investigated the fate of transplanted SCs in myelin oligodendrocyte glycoprotein (MOG)-induced experimental autoimmune encephalomyelitis (EAE) in the Dark Agouti rat; an animal model that reproduces the complex inflammatory demyelinating immunopathology of MS. We now report that SCs expressing green fluorescent protein (GFP-SCs) allografted after disease onset not only survive but also migrate to remyelinate lesions in the inflamed CNS. GFP-SCs were detected more frequently in the parenchyma after direct injection into the spinal cord, than via intra-thecal delivery into the cerebrospinal fluid. In both cases the transplanted cells intermingled with astrocytes in demyelinated lesions, aligned with axons and by twenty one days post transplantation had formed Pzero protein immunoreactive internodes. Strikingly, GFP-SCs transplantation was associated with marked decrease in clinical disease severity in terms of mortality; all GFP-SCs transplanted animals survived whilst 80% of controls died within 40 days of disease.


Assuntos
Movimento Celular , Encefalomielite Autoimune Experimental/fisiopatologia , Encefalomielite Autoimune Experimental/terapia , Bainha de Mielina/metabolismo , Recuperação de Função Fisiológica , Células de Schwann/citologia , Células de Schwann/transplante , Animais , Astrócitos/metabolismo , Astrócitos/patologia , Sobrevivência Celular , Feminino , Proteínas de Fluorescência Verde/metabolismo , Injeções Espinhais , Bainha de Mielina/patologia , Glicoproteína Mielina-Oligodendrócito , Ratos , Medula Espinal/irrigação sanguínea , Medula Espinal/patologia , Medula Espinal/fisiopatologia , Transdução Genética
11.
Proc Natl Acad Sci U S A ; 108(26): 10714-9, 2011 Jun 28.
Artigo em Inglês | MEDLINE | ID: mdl-21670295

RESUMO

Boundary cap cells (BC), which express the transcription factor Krox20, participate in the formation of the boundary between the central nervous system and the peripheral nervous system. To study BC stemness, we developed a method to purify and amplify BC in vitro from Krox20(Cre/+), R26R(YFP/+) mouse embryos. We show that BC progeny are EGF/FGF2-responsive, form spheres, and express neural crest markers. Upon growth factor withdrawal, BC progeny gave rise to multiple neural crest and CNS lineages. Transplanted into the developing murine forebrain, they successfully survived, migrated, and integrated within the host environment. Surprisingly, BC progeny generated exclusively CNS cells, including neurons, astrocytes, and myelin-forming oligodendrocytes. In vitro experiments indicated that a sequential combination of ventralizing morphogens and glial growth factors was necessary to reprogram BC into oligodendrocytes. Thus, BC progeny are endowed with differentiation plasticity beyond the peripheral nervous system. The demonstration that CNS developmental cues can reprogram neural crest-derived stem cells into CNS derivatives suggests that BC could serve as a source of cell type-specific lineages, including oligodendrocytes, for cell-based therapies to treat CNS disorders.


Assuntos
Diferenciação Celular , Sistema Nervoso Periférico/citologia , Células-Tronco/citologia , Animais , Linhagem da Célula , Movimento Celular , Células Cultivadas , Citometria de Fluxo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos DBA , Oligodendroglia/metabolismo
12.
Neuroscientist ; 13(4): 383-91, 2007 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-17644768

RESUMO

The loss of myelin, a major element involved in the saltatory conduction of the electrical impulse of the nervous system, is a major target of current research. Serious long-term disabilities are observed in patients with demyelinating disease of the central nervous system, such as multiple sclerosis. New therapeutic strategies aimed at overcoming myelin damage and axonal loss focus on the repair potential of myelin-forming cells. This review examines the use of peripheral myelin-forming cells, the Schwann cells, to promote myelin repair.


Assuntos
Sistema Nervoso Central/fisiologia , Bainha de Mielina/metabolismo , Nervos Periféricos/fisiologia , Regeneração/fisiologia , Animais , Terapia Baseada em Transplante de Células e Tecidos/história , Terapia Baseada em Transplante de Células e Tecidos/métodos , Doenças Desmielinizantes/metabolismo , Doenças Desmielinizantes/fisiopatologia , Doenças Desmielinizantes/terapia , Modelos Animais de Doenças , História do Século XIX , História do Século XX , História do Século XXI , Humanos
13.
J Neurosci Res ; 82(5): 581-91, 2005 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-16267826

RESUMO

Facial nerve axotomy (FNA) is a well-established experimental model of motoneuron regeneration. After peripheral nerve axotomy, a sequence of events including glial activation and axonal regrowth leads to functional recovery of the afflicted pool of motoneurons. Using microarray analysis we identified an increase in the expression of 60 genes (at a false discovery rate of 0.1, genes were significant P < 0.004) within the facial nucleus as a consequence of nerve injury. In situ hybridization analysis validated the increased expression of many of these axotomy-induced genes. One specific gene, encoding a unique primary amino acid sequence, termed hemopoietic- and neurologic-expressed sequence-1 (Hn1), was evaluated more extensively using several additional nerve injury paradigms. Hn1 mRNA was upregulated in injured facial motoneurons in both rats and mice. Sustained upregulation of Hn1 mRNA was evident after nerve resection whereas levels of Hn1 mRNA returned to baseline in animals subjected to nerve crush or nerve transection. Hn1 was also increased in the dorsal motor nucleus and the nucleus ambiguous after vagus nerve axotomy, another regeneration model. No upregulation of Hn1 expression was observed, however, in two nonregeneration models: FNA in newborn rats and rubrospinal tractotomy. Hn1 mRNA was ubiquitous in the developing central nervous system whereas its expression in adult brain was confined to neurons of the hippocampus, cortex and cerebellum. These findings identify Hn1 as a gene associated with nervous system development and nerve regeneration.


Assuntos
Traumatismos do Nervo Facial/genética , Nervo Facial/metabolismo , Regeneração Nervosa/genética , Proteínas do Tecido Nervoso/genética , Proteínas/genética , Ativação Transcricional/genética , Animais , Axotomia , Proteínas de Ciclo Celular , Nervo Facial/citologia , Traumatismos do Nervo Facial/metabolismo , Regulação da Expressão Gênica/genética , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Proteínas Associadas aos Microtúbulos , Neurônios Motores/metabolismo , Análise de Sequência com Séries de Oligonucleotídeos , RNA Mensageiro/análise , RNA Mensageiro/metabolismo , Ratos , Ratos Sprague-Dawley , Rombencéfalo/citologia , Rombencéfalo/metabolismo , Regulação para Cima/fisiologia , Nervo Vago/citologia , Nervo Vago/metabolismo , Doenças do Nervo Vago/genética , Doenças do Nervo Vago/metabolismo
14.
Mol Pharmacol ; 66(6): 1431-9, 2004 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-15361546

RESUMO

Fractalkine (FKN/CX3CL1) is a cell surface-expressed chemokine involved in many aspects of leukocyte trafficking and activation. The various structural domains of FKN play distinct roles in its ability to bind and activate its receptor, CX3CR1. A human herpesvirus 8-encoded chemokine, termed viral macrophage inflammatory protein (vMIP)-II, is structurally similar to FKN; vMIP-II is a nonselective chemokine receptor antagonist (binding multiple chemokine receptors, including CX3CR1). The goal of this study was to identify FKN determinants of selectivity for its receptor and to further refine domains important in affinity and efficacy at CX3CR1. Chimeric and insertional mutagenesis was used to generate mutants of both vMIP-II and FKN, and the expressed proteins were evaluated for chemokine receptor binding affinities and efficacy at CX3CR1. Modification of the intervening amino acids between the first two conserved cysteine residues of FKN or vMIP-II indicated a role of the X3 bulge of FKN in affinity and selectivity for CX3CR1. Substitution of the vMIP-II N terminus with that of FKN created an agonist that was just as potent and efficacious as FKN for binding and stimulating CX3CR1, whereas replacement of the FKN N terminus with the cognate domain of vMIP-II disrupted the ability of FKN to bind CX3CR1. Furthermore, the entire N terminus of FKN was necessary for the high-affinity and full agonist properties of FKN at CX3CR1. These results refine the pharmacophore for chemokine binding to and activation of CX3CR1 and demonstrate the usefulness of modified virally encoded chemokines as templates for the development of selective chemokine receptor antagonists.


Assuntos
Quimiocinas CX3C/metabolismo , Quimiocinas/metabolismo , Proteínas de Membrana/metabolismo , Receptores de Quimiocinas/metabolismo , Sequência de Aminoácidos , Antivirais/metabolismo , Sítios de Ligação , Ligação Competitiva , Receptor 1 de Quimiocina CX3C , Linhagem Celular , Quimiocina CX3CL1 , Humanos , Cinética , Dados de Sequência Molecular , Fosforilação , Proteínas Recombinantes de Fusão/metabolismo , Sarcoma de Kaposi/patologia , Transfecção
15.
Methods ; 29(4): 345-50, 2003 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-12725801

RESUMO

In order to understand processes involved in central nervous system inflammatory diseases, a critical appreciation of mechanisms involved in the control of immune function in the brain is needed. Microglial cells are watchful eyes for unusual events and detecting the presence of pathogens but are also alert to signals emanating from damaged neurons. Fractalkine (CX3CL1) is a chemokine which is expressed predominantly in the central nervous system, being localized on neurons, while its receptor, CX3CR1, is found on microglial cells. We have developed a strategy to investigate the role of this chemokine in neuronal-microglia interactions. Because fractalkine is expressed both as a soluble and as a membrane-attached protein, we have established various protocols involving different levels of cell-to-cell communication. Three experimental systems were instituted, including (1) a conditioned medium transfer system in which no cell-cell communication or contact is possible, (2) a transwell system that permits cell-contact-independent communication through diffusible soluble factors only, and (3) a coculture system allowing cell-to-cell communication via direct microglial-neuronal contacts. Using these in vitro cocultured systems, we have investigated the role of a soluble and/or cell-associated chemokine, such as fractalkine, in order to obtain insights into the role of glia-neuron interactions in cerebral inflammation.


Assuntos
Quimiocinas/biossíntese , Microglia/metabolismo , Neurônios/metabolismo , Animais , Comunicação Celular , Quimiocina CX3CL1 , Quimiocinas CX3C/metabolismo , Técnicas de Cocultura , Meios de Cultivo Condicionados/farmacologia , Feminino , Lipopolissacarídeos/metabolismo , Proteínas de Membrana/metabolismo , Modelos Biológicos , Ratos , Ratos Sprague-Dawley
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...