Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 22
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Aging (Albany NY) ; 7(6): 435-49, 2015 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-26142744

RESUMEN

Hippocampal neurogenesis undergoes dramatic age-related changes. Mice with targeted deletion of the clock geneBmal1 (Bmal1(-/-)) show disrupted regulation of reactive oxygen species homeostasis, accelerated aging, neurodegeneration and cognitive deficits. As proliferation of neuronal progenitor/precursor cells (NPCs) is enhanced in young Bmal1(-/-) mice, we tested the hypothesis that this results in premature aging of hippocampal neurogenic niche in adult Bmal1(-/-) mice as compared to wildtype littermates. We found significantly reduced pool of hippocampal NPCs, scattered distribution, enhanced survival of NPCs and an increased differentiation of NPCs into the astroglial lineage at the expense of the neuronal lineage. Immunoreaction of the redox sensitive histone deacetylase Sirtuine 1, peroxisomal membrane protein at 70 kDa and expression of the cell cycle inhibitor p21(Waf1/CIP1) were increased in adult Bmal1(-/-) mice. In conclusion, genetic disruption of the molecular clockwork leads to accelerated age-dependent decline in adult neurogenesis presumably as a consequence of oxidative stress.


Asunto(s)
Factores de Transcripción ARNTL/metabolismo , Envejecimiento Prematuro , Envejecimiento/fisiología , Hipocampo/fisiología , Factores de Transcripción ARNTL/genética , Transportadoras de Casetes de Unión a ATP/genética , Transportadoras de Casetes de Unión a ATP/metabolismo , Animales , Regulación de la Expresión Génica/fisiología , Ratones , Ratones Noqueados , Estrés Oxidativo , Sirtuina 1/genética , Sirtuina 1/metabolismo
2.
Acta Neuropathol Commun ; 3: 20, 2015 Apr 03.
Artículo en Inglés | MEDLINE | ID: mdl-25853624

RESUMEN

INTRODUCTION: Multiple sclerosis (MS) is a chronic demyelinating disorder of the central nervous system (CNS) leading to progressive neurological disability. Interferon ß (IFNß) represents a standard treatment for relapsing-remitting MS and exogenous administration of IFNß exhibits protective effects in experimentally induced CNS autoimmunity. Also, genetic deletion of IFNß in mice leads to an aggravation of disease symptoms in the MS model of experimental autoimmune encephalomyelitis (EAE). However, neither the underlying mechanisms mediating the beneficial effects nor the cellular source of IFNß have been fully elucidated. RESULTS: In this report, a subpopulation of activated microglia was identified as the major producers of IFNß in the CNS at the peak of EAE using an IFNß-fluorescence reporter mouse model. These IFNß expressing microglia specifically localized to active CNS lesions and were associated with myelin debris in demyelinated cerebellar organotypic slice cultures (OSCs). In response to IFNß microglia showed an enhanced capacity to phagocytose myelin in vitro and up-regulated the expression of phagocytosis-associated genes. IFNß treatment was further sufficient to stimulate association of microglia with myelin debris in OSCs. Moreover, IFNß-producing microglia mediated an enhanced removal of myelin debris when co-transplanted onto demyelinated OSCs as compared to IFNß non-producing microglia. CONCLUSIONS: These data identify activated microglia as the major producers of protective IFNß at the peak of EAE and as orchestrators of IFNß-induced clearance of myelin debris.


Asunto(s)
Cerebelo/citología , Encefalomielitis Autoinmune Experimental/inmunología , Interferón beta/inmunología , Microglía/metabolismo , Vaina de Mielina/metabolismo , Fagocitosis/genética , Animales , Células Cultivadas , Modelos Animales de Enfermedad , Encefalomielitis Autoinmune Experimental/patología , Interferón beta/metabolismo , Ratones , Ratones Endogámicos C57BL , Microglía/citología , Técnicas de Cultivo de Órganos , Regulación hacia Arriba
3.
PLoS One ; 9(7): e100871, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25013913

RESUMEN

The maturation status of dendritic cells determines whether interacting T cells are activated or if they become tolerant. Previously we could induce T cell tolerance by applying a 3-hydroxy-3-methylglutaryl-CoA (HMG-CoA) reductase inhibitor (HMGCRI) atorvastatin, which also modulates MHC class II expression and has therapeutic potential in autoimmune disease. Here, we aimed at elucidating the impact of this therapeutic strategy on T cell differentiation as a consequence of alterations in dendritic cell function. We investigated the effect of HMGCRI during differentiation of peripheral human monocytes and murine bone marrow precursors to immature DC in vitro and assessed their phenotype. To examine the stimulatory and tolerogenic capacity of these modulated immature dendritic cells, we measured proliferation and suppressive function of CD4+ T cells after stimulation with the modulated immature dendritic cells. We found that an HMGCRI, atorvastatin, prevents dendrite formation during the generation of immature dendritic cells. The modulated immature dendritic cells had a diminished capacity to take up and present antigen as well as to induce an immune response. Of note, the consequence was an increased capacity to differentiate naïve T cells towards a suppressor phenotype that is less sensitive to proinflammatory stimuli and can effectively inhibit the proliferation of T effector cells in vitro. Thus, manipulation of antigen-presenting cells by HMGCRI contributes to an attenuated immune response as shown by promotion of T cells with suppressive capacities.


Asunto(s)
Células Dendríticas/enzimología , Células Dendríticas/inmunología , Hidroximetilglutaril-CoA Reductasas/metabolismo , Animales , Atorvastatina , Movimiento Celular/efectos de los fármacos , Células Cultivadas , Células Dendríticas/efectos de los fármacos , Citometría de Flujo , Ácidos Heptanoicos/farmacología , Humanos , Inhibidores de Hidroximetilglutaril-CoA Reductasas/farmacología , Interleucina-10/metabolismo , Ratones , Ratones Endogámicos C57BL , Fagocitosis/efectos de los fármacos , Pirroles/farmacología , Linfocitos T Reguladores/efectos de los fármacos , Linfocitos T Reguladores/inmunología
4.
Proc Natl Acad Sci U S A ; 110(50): 20057-62, 2013 Dec 10.
Artículo en Inglés | MEDLINE | ID: mdl-24277839

RESUMEN

Embryonic development depends on complex and precisely orchestrated signaling pathways including specific reduction/oxidation cascades. Oxidoreductases of the thioredoxin family are key players conveying redox signals through reversible posttranslational modifications of protein thiols. The importance of this protein family during embryogenesis has recently been exemplified for glutaredoxin 2, a vertebrate-specific glutathione-disulfide oxidoreductase with a critical role for embryonic brain development. Here, we discovered an essential function of glutaredoxin 2 during vascular development. Confocal microscopy and time-lapse studies based on two-photon microscopy revealed that morpholino-based knockdown of glutaredoxin 2 in zebrafish, a model organism to study vertebrate embryogenesis, resulted in a delayed and disordered blood vessel network. We were able to show that formation of a functional vascular system requires glutaredoxin 2-dependent reversible S-glutathionylation of the NAD(+)-dependent protein deacetylase sirtuin 1. Using mass spectrometry, we identified a cysteine residue in the conserved catalytic region of sirtuin 1 as target for glutaredoxin 2-specific deglutathionylation. Thereby, glutaredoxin 2-mediated redox regulation controls enzymatic activity of sirtuin 1, a mechanism we found to be conserved between zebrafish and humans. These results link S-glutathionylation to vertebrate development and successful embryonic angiogenesis.


Asunto(s)
Sistema Cardiovascular/embriología , Glutarredoxinas/metabolismo , Glutatión/metabolismo , Neovascularización Fisiológica/fisiología , Transducción de Señal/fisiología , Sirtuina 1/metabolismo , Animales , Western Blotting , Cartilla de ADN/genética , Técnicas de Silenciamiento del Gen , Glutarredoxinas/genética , Células HeLa , Humanos , Espectrometría de Masas , Microscopía Confocal , Oxidación-Reducción , Reacción en Cadena en Tiempo Real de la Polimerasa , Transducción de Señal/genética , Imagen de Lapso de Tiempo , Pez Cebra
6.
Proc Natl Acad Sci U S A ; 109(17): 6650-5, 2012 Apr 24.
Artículo en Inglés | MEDLINE | ID: mdl-22492966

RESUMEN

The detection of pathological tissue alterations by manual palpation is a simple but essential diagnostic tool, which has been applied by physicians since the beginnings of medicine. Recently, the virtual "palpation" of the brain has become feasible using magnetic resonance elastography, which quantifies biomechanical properties of the brain parenchyma by analyzing the propagation of externally elicited shear waves. However, the precise molecular and cellular patterns underlying changes of viscoelasticity measured by magnetic resonance elastography have not been investigated up to date. We assessed changes of viscoelasticity in a murine model of multiple sclerosis, inducing reversible demyelination by feeding the copper chelator cuprizone, and correlated our results with detailed histological analyses, comprising myelination, extracellular matrix alterations, immune cell infiltration and axonal damage. We show firstly that the magnitude of the complex shear modulus decreases with progressive demyelination and global extracellular matrix degradation, secondly that the loss modulus decreases faster than the dynamic modulus during the destruction of the corpus callosum, and finally that those processes are reversible after remyelination.


Asunto(s)
Encéfalo/patología , Enfermedades Desmielinizantes/patología , Diagnóstico por Imagen de Elasticidad/métodos , Animales , Axones , Quelantes/administración & dosificación , Quelantes/química , Cobre/química , Cuprizona/administración & dosificación , Cuprizona/química , Femenino , Ratones , Ratones Endogámicos C57BL
7.
Front Neurol ; 2: 72, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-22203815

RESUMEN

We present a novel highly efficient protocol to magnetically label T cells applying electrostatically stabilized very small superparamagnetic iron oxide particles (VSOP). Our long-term aim is to use magnetic resonance imaging (MRI) to investigate T cell dynamics in vivo during the course of neuroinflammatory disorders such as experimental autoimmune encephalomyelitis (EAE), an animal model of multiple sclerosis. Encephalitogenic T cells were co-incubated with VSOP, or with protamine-complexed VSOP (VProt), respectively, at different conditions, optimizing concentrations and incubation times. Labeling efficacy was determined by atomic absorption spectrometry as well as histologically, and evaluated on a 7 T MR system. Furthermore, we investigated possible alterations of T cell physiology caused by the labeling procedure. T cell co-incubation with VSOP resulted in an efficient cellular iron uptake. T2 times of labeled cells dropped significantly, resulting in prominent hypointensity on T2*-weighted scans. Optimal labeling efficacy was achieved by VProt (1 mM Fe/ml, 8 h incubation; T2 time shortening of ∼80% compared to untreated cells). Although VSOP promoted T cell proliferation and altered the ratio of T cell subpopulations toward a CD4(+) phenotype, no effects on CD4 T cell proliferation or phenotypic stability were observed by labeling in vitro differentiated Th17 cells with VProt. Yet, high concentrations of intracellular iron oxide might induce alterations in T cell function, which should be considered in cell tagging studies. Moreover, we demonstrated that labeling of encephalitogenic T cells did not affect pathogenicity; labeled T cells were still capable of inducing EAE in susceptible recipient mice.

8.
Proc Natl Acad Sci U S A ; 108(51): 20532-7, 2011 Dec 20.
Artículo en Inglés | MEDLINE | ID: mdl-22139372

RESUMEN

Cellular functions and survival are dependent on a tightly controlled redox potential. Currently, an increasing amount of data supports the concept of local changes in the redox environment and specific redox signaling events controlling cell function. Specific protein thiol groups are the major targets of redox signaling and regulation. Thioredoxins and glutaredoxins catalyze reversible thiol-disulfide exchange reactions and are primary regulators of the protein thiol redox state. Here, we demonstrate that embryonic brain development depends on the enzymatic activity of glutaredoxin 2. Zebrafish with silenced expression of glutaredoxin 2 lost virtually all types of neurons by apoptotic cell death and the ability to develop an axonal scaffold. As demonstrated in zebrafish and in a human cellular model for neuronal differentiation, glutaredoxin 2 controls axonal outgrowth via thiol redox regulation of collapsin response mediator protein 2, a central component of the semaphorin pathway. This study provides an example of a specific thiol redox regulation essential for vertebrate embryonic development.


Asunto(s)
Encéfalo/embriología , Regulación del Desarrollo de la Expresión Génica , Glutarredoxinas/química , Pez Cebra/embriología , Animales , Apoptosis , Axones/fisiología , Línea Celular Tumoral , Biología Evolutiva , Glutarredoxinas/genética , Humanos , Neuritas/metabolismo , Oxidación-Reducción , Proteínas Recombinantes/química , Transducción de Señal , Vertebrados
9.
Glia ; 59(1): 132-42, 2011 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-20967885

RESUMEN

In chronic autoimmune diseases of the central nervous system (CNS) such as multiple sclerosis (MS) clinical signs of cognitive dysfunction have been associated with structural changes in the hippocampus. Moreover, experimental studies indicate that inflammatory responses within the CNS modulate the homeostasis of newborn cells in the adult dentate gyrus (DG). However, it remained open whether such changes happen regardless of the primary immunological target or whether a CNS antigen-directed T lymphocyte-mediated autoimmune response may exert a specific impact. We therefore induced experimental autoimmune encephalomyelitis (EAE), a common model of MS serving as a paradigm for a CNS-specific immune response, by immunizing C57BL/6 mice with encephalitogenic myelin oligodendrocyte glycoprotein (MOG) p35-55. In EAE animals, we found enhanced de novo generation and survival of doublecortin (DCX)-positive immature neurons when compared with controls immunized with CNS-irrelevant antigen (ovalbumine). However, despite activation of neurogenesis, we observed a reduced capacity of these cells to generate mature neurons. Moreover, the high number of newly born cells retained the expression of the glial marker GFAP. These effects were associated with downregulation of pro-neurogenic factors Neurogenin1 and Neurogenin2 and dysregulation of Notch, ß-catenin, Sonic Hedgehog (Shh) signaling as suggested by altered gene expression of effector molecules. Thus, a CNS antigen-specific immune response leads to an aberrant differentiation of neural precursors associated with dysbalance of signaling pathways relevant for adult hippocampal neurogenesis. These results may further extend our understanding of disturbed regeneration in the course of chronic inflammatory CNS diseases such as MS.


Asunto(s)
Encefalomielitis Autoinmune Experimental/inmunología , Hipocampo/inmunología , Inflamación/inmunología , Vaina de Mielina/inmunología , Neurogénesis/inmunología , Animales , Autoinmunidad/inmunología , Recuento de Células , Proteína Doblecortina , Encefalomielitis Autoinmune Experimental/fisiopatología , Femenino , Técnica del Anticuerpo Fluorescente , Hipocampo/fisiopatología , Ratones , Neuronas/inmunología , Distribución Aleatoria , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
10.
Cell ; 142(4): 613-24, 2010 Aug 20.
Artículo en Inglés | MEDLINE | ID: mdl-20723761

RESUMEN

Interferon (IFN)-induced immunoproteasomes (i-proteasomes) have been associated with improved processing of major histocompatibility complex (MHC) class I antigens. Here, we show that i-proteasomes function to protect cell viability under conditions of IFN-induced oxidative stress. IFNs trigger the production of reactive oxygen species, which induce protein oxidation and the formation of nascent, oxidant-damaged proteins. We find that the ubiquitylation machinery is concomitantly upregulated in response to IFNs, functioning to target defective ribosomal products (DRiPs) for degradation by i-proteasomes. i-proteasome-deficiency in cells and in murine inflammation models results in the formation of aggresome-like induced structures and increased sensitivity to apoptosis. Efficient clearance of these aggregates by the enhanced proteolytic activity of the i-proteasome is important for the preservation of cell viability upon IFN-induced oxidative stress. Our findings suggest that rather than having a specific role in the production of class I antigens, i-proteasomes increase the peptide supply for antigen presentation as part of a more general role in the maintenance of protein homeostasis.


Asunto(s)
Antígenos de Histocompatibilidad Clase I/metabolismo , Interferones/inmunología , Complejo de la Endopetidasa Proteasomal/inmunología , Complejo de la Endopetidasa Proteasomal/metabolismo , Proteínas/metabolismo , Animales , Presentación de Antígeno , Encefalomielitis Autoinmune Experimental/inmunología , Encefalomielitis Autoinmune Experimental/metabolismo , Antígenos de Histocompatibilidad Clase I/inmunología , Homeostasis , Humanos , Inflamación/metabolismo , Ratones , Ratones Endogámicos C57BL , Ubiquitinación
11.
Eur J Immunol ; 40(5): 1486-95, 2010 May.
Artículo en Inglés | MEDLINE | ID: mdl-20186879

RESUMEN

DC are professional APC that instruct T cells during the inflammatory course of EAE. We have previously shown that MAPK3 (Erk1) is important for the induction of T-cell anergy. Our goal was to determine the influence of MAPK3 on the capacity of DC to arm T-cell responses in autoimmunity. We report that DC from Mapk3(-/-) mice have a significantly higher membrane expression of CD86 and MHC-II and--when loaded with the myelin oligodendrocyte glycoprotein--show a superior capacity to prime naïve T cells towards an inflammatory phenotype than Mapk3(+/+) DC. Nonetheless and as previously described, Mapk3(-/-) mice were only slightly but not significantly more susceptible to myelin oligodendrocyte glycoprotein-induced EAE than WT littermate mice. However, Mapk3(+/+) mice engrafted with Mapk3(-/-) BM (KO-->WT) developed a severe form of EAE, in direct contrast to WT-->KO mice, which were even less sick than control WT-->WT mice. An infiltration of DC and accumulation of Th17 cells was also observed in the CNS of KO-->WT mice. Therefore, triggering of MAPK3 in the periphery might be a therapeutic option for the treatment of neuroinflammation since absence of this kinase in the immune system leads to severe EAE.


Asunto(s)
Autoinmunidad/fisiología , Células Dendríticas/enzimología , Encefalomielitis Autoinmune Experimental/enzimología , Proteína Quinasa 3 Activada por Mitógenos/fisiología , Subgrupos de Linfocitos T/inmunología , Animales , Antígeno B7-2/metabolismo , Citocinas/biosíntesis , Células Dendríticas/inmunología , Encefalomielitis Autoinmune Experimental/inmunología , Glicoproteínas/inmunología , Glicoproteínas/toxicidad , Antígenos de Histocompatibilidad Clase II/inmunología , Sistema de Señalización de MAP Quinasas , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Proteína Quinasa 3 Activada por Mitógenos/deficiencia , Proteína Quinasa 3 Activada por Mitógenos/genética , Glicoproteína Mielina-Oligodendrócito , Ovalbúmina/inmunología , Fragmentos de Péptidos/inmunología , Fragmentos de Péptidos/toxicidad , Quimera por Radiación , Organismos Libres de Patógenos Específicos , Especificidad del Receptor de Antígeno de Linfocitos T
12.
Nat Med ; 15(7): 788-93, 2009 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-19561616

RESUMEN

Previous proteomic and transcriptional analyses of multiple sclerosis lesions revealed modulation of the renin-angiotensin and the opposing kallikrein-kinin pathways. Here we identify kinin receptor B1 (Bdkrb1) as a specific modulator of immune cell entry into the central nervous system (CNS). We demonstrate that the Bdkrb1 agonist R838 (Sar-[D-Phe]des-Arg(9)-bradykinin) markedly decreases the clinical symptoms of experimental autoimmune encephalomyelitis (EAE) in SJL mice, whereas the Bdkrb1 antagonist R715 (Ac-Lys-[D-betaNal(7), Ile(8)]des-Arg(9)-bradykinin) resulted in earlier onset and greater severity of the disease. Bdkrb1-deficient (Bdkrb1(-/-)) C57BL/6 mice immunized with a myelin oligodendrocyte glycoprotein fragment, MOG(35-55), showed more severe disease with enhanced CNS-immune cell infiltration. The same held true for mixed bone marrow-chimeric mice reconstituted with Bdkrb1(-/-) T lymphocytes, which showed enhanced T helper type 17 (T(H)17) cell invasion into the CNS. Pharmacological modulation of Bdkrb1 revealed that in vitro migration of human T(H)17 lymphocytes across blood-brain barrier endothelium is regulated by this receptor. Taken together, these results suggest that the kallikrein-kinin system is involved in the regulation of CNS inflammation, limiting encephalitogenic T lymphocyte infiltration into the CNS, and provide evidence that Bdkrb1 could be a new target for the treatment of chronic inflammatory diseases such as multiple sclerosis.


Asunto(s)
Encéfalo/patología , Encefalomielitis Autoinmune Experimental/etiología , Receptor de Bradiquinina B1/fisiología , Linfocitos T/fisiología , Animales , Movimiento Celular , Encefalomielitis Autoinmune Experimental/patología , Encefalomielitis Autoinmune Experimental/terapia , Interleucina-17/fisiología , Ratones , Ratones Endogámicos C57BL , Receptor de Bradiquinina B1/agonistas , Células TH1/fisiología
13.
Nat Cell Biol ; 10(4): 385-94, 2008 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-18344989

RESUMEN

Repair processes that are activated in response to neuronal injury, be it inflammatory, ischaemic, metabolic, traumatic or other cause, are characterized by a failure to replenish neurons and by astrogliosis. The underlying molecular pathways, however, are poorly understood. Here, we show that subtle alterations of the redox state, found in different brain pathologies, regulate the fate of mouse neural progenitor cells (NPCs) through the histone deacetylase (HDAC) Sirt1. Mild oxidation or direct activation of Sirt1 suppressed proliferation of NPCs and directed their differentiation towards the astroglial lineage at the expense of the neuronal lineage, whereas reducing conditions had the opposite effect. Under oxidative conditions in vitro and in vivo, Sirt1 was upregulated in NPCs, bound to the transcription factor Hes1 and subsequently inhibited pro-neuronal Mash1. In utero shRNA-mediated knockdown of Sirt1 in NPCs prevented oxidation-mediated suppression of neurogenesis and caused upregulation of Mash1 in vivo. Our results provide evidence for an as yet unknown metabolic master switch that determines the fate of neural progenitors.


Asunto(s)
Diferenciación Celular/fisiología , Neuronas/fisiología , Sirtuinas/metabolismo , Células Madre/fisiología , Animales , Astrocitos/citología , Astrocitos/fisiología , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Encéfalo/citología , Encéfalo/metabolismo , Encéfalo/patología , Linaje de la Célula , Células Cultivadas , Proteínas Co-Represoras , Encefalomielitis Autoinmune Experimental/metabolismo , Encefalomielitis Autoinmune Experimental/patología , Femenino , Regulación del Desarrollo de la Expresión Génica , Histonas/metabolismo , Proteínas de Homeodominio/genética , Proteínas de Homeodominio/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Morfogénesis/fisiología , Neuronas/citología , Oxidación-Reducción , Embarazo , Regiones Promotoras Genéticas , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/metabolismo , Proteínas Represoras/genética , Proteínas Represoras/metabolismo , Sirtuina 1 , Sirtuinas/genética , Células Madre/citología , Factor de Transcripción HES-1 , Transcripción Genética
14.
J Immunol ; 179(9): 6024-32, 2007 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-17947676

RESUMEN

Rho GTPases orchestrate signaling pathways leading to cell migration. Their function depends on GTP loading and isoprenylation by geranylgeranyl pyrophosphate (GGpp). In this study, we show that in human T cells, geranylgeranylation-and not GTP loading-is necessary for RhoA-mediated downstream events. As a result of GGpp depletion with the 3-hydroxy-3-methylglutaryl-CoA reductase inhibitor atorvastatin, RhoA was sequestered from the membrane to the cytosol and, notwithstanding increased GTP loading, the constitutive activation of its substrate Rho-associated coiled-coil protein kinase-1 was blocked. In line with this, T cells expressing increased GTP-RhoA failed to form an intact cytoskeleton and to migrate toward a chemokine gradient. In vivo treatment with atorvastatin in the rodent model of multiple sclerosis markedly decreased the capacity of activated T cells to traffic within the brain, as demonstrated by multiphoton analysis. Thus, tethering of RhoA to the membrane by GGpp is determinant for T cell migration and provides a mechanism for preventing T cell infiltration into inflamed compartments by 3-hydroxy-3-methylglutaryl-CoA reductase inhibitors.


Asunto(s)
Movimiento Celular/efectos de los fármacos , Guanosina Trifosfato/farmacología , Prenilación , Linfocitos T/efectos de los fármacos , Linfocitos T/metabolismo , Proteína de Unión al GTP rhoA/metabolismo , Animales , Apoptosis/efectos de los fármacos , Encéfalo/citología , Encéfalo/efectos de los fármacos , Caspasa 3/metabolismo , Línea Celular , Citoesqueleto/metabolismo , Citosol/metabolismo , Activación Enzimática/efectos de los fármacos , Humanos , Ratones , Fenalenos/farmacología , Fosfatos de Poliisoprenilo/farmacología , Unión Proteica , Linfocitos T/citología , Quinasas Asociadas a rho/metabolismo
15.
Eur J Neurosci ; 26(1): 190-8, 2007 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-17596194

RESUMEN

The value of experimental autoimmune encephalomyelitis (EAE), an animal model of multiple sclerosis, in deriving novel diagnostic and therapeutic input has been subject to recent debate. This study is the first to report a disseminated distribution of plaques including cranial nerves, prior to or at early stages of disease in murine adoptive transfer EAE, irrespective of the development of clinical symptoms. We induced EAE by adoptive proteolipid protein-specific T-cell transfer in 26 female SJL/J mice, and applied high-field-strength magnetic resonance imaging (MRI) scans longitudinally, assessing blood-brain barrier (BBB) disruption by gadopentate dimeglumine enhancement. We visualized inflammatory nerve injury by gadofluorine M accumulation, and phagocytic cells in inflamed tissue by very small anionic iron oxide particles (VSOP-C184). MRI was correlated with immunohistological sections. In this study, we discovered very early BBB breakdown of white and grey brain matter in 25 mice; one mouse developed exclusively spinal cord inflammation. Widely disseminated contrast-enhancing lesions preceded the onset of disease in 10 animals. Such lesions were present despite the absence of any clinical disease formation in four mice, and coincided with the first detectable symptoms in others. Cranial nerves, predominantly the optic and trigeminal nerves, showed signal intensity changes in nuclei and fascicles of 14 mice. At all sites of MRI lesions we detected cellular infiltrates on corresponding histological sections. The discrepancy between the disease burden visualized by MRI and the extent of disability indeed mimics the human clinico-radiological paradox. MRI should therefore be implemented into evaluational in vivo routines of future therapeutic EAE studies.


Asunto(s)
Esclerosis Múltiple/diagnóstico por imagen , Esclerosis Múltiple/patología , Animales , Barrera Hematoencefálica/fisiología , Encéfalo/patología , Cerebelo/patología , Medios de Contraste , Nervios Craneales/patología , Modelos Animales de Enfermedad , Endocitosis/fisiología , Femenino , Fluorocarburos , Gadolinio DTPA , Procesamiento de Imagen Asistido por Computador , Inmunohistoquímica , Imagen por Resonancia Magnética , Ratones , Microglía/patología , Compuestos Organometálicos , Fagocitosis/efectos de los fármacos , Radiografía , Linfocitos T/fisiología
16.
J Clin Invest ; 117(7): 2004-13, 2007 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-17571163

RESUMEN

Apart from potential roles in anti-tumor surveillance, the TNF-related apoptosis-inducing ligand (TRAIL) has important regulatory functions in the host immune response. We studied antiinflammatory effects of endogenous and recombinant TRAIL (rTRAIL) in experimental meningitis. Following intrathecal application of pneumococcal cell wall, a TLR2 ligand, we found prolonged inflammation, augmented clinical impairment, and increased apoptosis in the hippocampus of TRAIL(-/-) mice. Administration of rTRAIL into the subarachnoid space of TRAIL(-/-) mice or reconstitution of hematopoiesis with wild-type bone marrow cells reversed these effects, suggesting an autoregulatory role of TRAIL within the infiltrating leukocyte population. Importantly, intrathecal application of rTRAIL in wild-type mice with meningitis also decreased inflammation and apoptosis. Moreover, patients suffering from bacterial meningitis showed increased intrathecal synthesis of TRAIL. Our findings provide what we believe is the first evidence that TRAIL may act as a negative regulator of acute CNS inflammation. The ability of TRAIL to modify inflammatory responses and to reduce neuronal cell death in meningitis suggests that it may be used as a novel antiinflammatory agent in invasive infections.


Asunto(s)
Meningitis Bacterianas/inmunología , Meningitis Bacterianas/metabolismo , Ligando Inductor de Apoptosis Relacionado con TNF/inmunología , Animales , Antígenos CD18/metabolismo , Supervivencia Celular , Citocinas/genética , Modelos Animales de Enfermedad , Regulación de la Expresión Génica , Genotipo , Cocos Grampositivos/fisiología , Granulocitos/metabolismo , Hipocampo/inmunología , Hipocampo/metabolismo , Hipocampo/patología , Humanos , Leucocitos/citología , Masculino , Meningitis Bacterianas/genética , Meningitis Bacterianas/patología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Neisseria/fisiología , Receptores del Ligando Inductor de Apoptosis Relacionado con TNF/metabolismo , Proteínas Recombinantes/farmacología , Solubilidad , Tasa de Supervivencia , Ligando Inductor de Apoptosis Relacionado con TNF/líquido cefalorraquídeo , Ligando Inductor de Apoptosis Relacionado con TNF/metabolismo , Ligando Inductor de Apoptosis Relacionado con TNF/farmacología
17.
Neuroscientist ; 12(4): 305-16, 2006 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-16840707

RESUMEN

Death ligands induce apoptosis, which is a cell suicide program leading mainly to selective elimination of an organism's useless cells. Importantly, the dying cell is an active participant in its own demise ("cellular suicide"). Under physiological conditions, apoptosis is most often found during normal cell turnover and tissue homeostasis, embryogenesis, induction and maintenance of immune tolerance, development of the nervous system, and endocrine-dependent tissue atrophy. However, apoptotic processes have also been suggested to contribute to the pathology of the autoimmune demyelinating disease multiple sclerosis (MS) and its animal model, experimental autoimmune encephalomyelitis. Here, apoptosis plays a double role. On one hand, impaired apoptosis may result in increased numbers or persistence of activated myelinspecific T cells. On the other hand, local tissue damage involves apoptosis of oligodendrocytes and neurons, leading to the clinical symptoms. In this article, an overview is given of the current knowledge of the roles of apoptosis-mediating and immune regulatory death ligands of the tumor necrosis factor (TNF) family (TNF, lymphotoxin-beta, OX40L [CD134L], CD154 [CD40L], CD95L, CD70 [CD27L], CD153 [CD30L], 4-1BBL [CD137L], TRAIL, TWEAK, BAFF, GITRL) in the pathogenesis of MS and of their implications for related therapeutic strategies.


Asunto(s)
Proteínas Reguladoras de la Apoptosis/metabolismo , Enfermedades Autoinmunes Desmielinizantes SNC/metabolismo , Enfermedades Desmielinizantes/metabolismo , Transducción de Señal/fisiología , Factor de Necrosis Tumoral alfa/metabolismo , Animales , Apoptosis/fisiología , Proteínas Reguladoras de la Apoptosis/inmunología , Enfermedades Autoinmunes Desmielinizantes SNC/inmunología , Enfermedades Desmielinizantes/inmunología , Humanos , Modelos Biológicos , Factor de Necrosis Tumoral alfa/inmunología
18.
FASEB J ; 19(13): 1902-4, 2005 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-16144955

RESUMEN

Multiple sclerosis (MS) is an autoimmune disease of the central nervous system (CNS) characterized by enormous variability in its clinical presentation and course, and for which clear diagnostic parameters are lacking. Here we performed an RNA screen in peripheral mononuclear cells from relapsing-remitting (RR) and primary progressive (PP) MS patients compared with healthy donors (HD) that indicated, among other findings, a role for the chemokine receptor CX3CR1 as a diagnostic marker. Gene expression and flow cytometric analyses demonstrated a significantly lower expression of CX3CR1 in MS patients compared with healthy individuals. The subpopulation of cells responsible for causing this reduced expression of CX3CR1 consisted exclusively of natural killer (NK) cells. Importantly, we found a correlation between disease activity and frequency of CX3CR1-positive NK cells in RRMS patients. These findings emphasize the role of NK cells in the development and course of MS and provide evidence for CX3CR1 expression as a marker for MS patients and disease activity.


Asunto(s)
Células Asesinas Naturales/citología , Proteínas de la Membrana/sangre , Esclerosis Múltiple/sangre , Receptores de Quimiocina/sangre , Adolescente , Adulto , Anciano , Anticuerpos Monoclonales/química , Biotinilación , Linfocitos T CD8-positivos/metabolismo , Receptor 1 de Quimiocinas CX3C , Supervivencia Celular , Regulación hacia Abajo , Femenino , Citometría de Flujo , Regulación de la Expresión Génica , Humanos , Inflamación , Masculino , Persona de Mediana Edad , Modelos Biológicos , Esclerosis Múltiple/diagnóstico , Esclerosis Múltiple Recurrente-Remitente/sangre , Análisis de Secuencia por Matrices de Oligonucleótidos , ARN/química , Recurrencia , Inducción de Remisión , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Factores de Tiempo , Regulación hacia Arriba
19.
Neuron ; 46(3): 421-32, 2005 May 05.
Artículo en Inglés | MEDLINE | ID: mdl-15882642

RESUMEN

Here, we provide evidence for a detrimental role of tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) in neural death in T cell-induced experimental autoimmune encephalomyelitis (EAE), an animal model of multiple sclerosis (MS). Clinical severity and neuronal apoptosis in brainstem motor areas were substantially reduced upon brain-specific blockade of TRAIL after induction of EAE through adoptive transfer of encephalitogenic T cells. Furthermore, TRAIL-deficient myelin-specific lymphocytes showed reduced encephalitogenicity when transferred to wild-type mice. Conversely, intracerebral delivery of TRAIL to animals with EAE increased clinical deficits, while naive mice were not susceptible to TRAIL. Using organotypic slice cultures as a model for living brain tissue, we found that neurons were susceptible to TRAIL-mediated injury induced by encephalitogenic T cells. Thus, in addition to its known immunoregulatory effects, the death ligand TRAIL contributes to neural damage in the inflamed brain.


Asunto(s)
Encéfalo/patología , Encefalomielitis Autoinmune Experimental/patología , Glicoproteínas de Membrana/metabolismo , Neuronas/patología , Linfocitos T/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo , Traslado Adoptivo , Animales , Apoptosis/inmunología , Proteínas Reguladoras de la Apoptosis , Western Blotting , Encéfalo/inmunología , Encéfalo/metabolismo , Modelos Animales de Enfermedad , Encefalomielitis Autoinmune Experimental/inmunología , Femenino , Citometría de Flujo , Inmunohistoquímica , Ratones , Esclerosis Múltiple , Neuronas/inmunología , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Linfocitos T/inmunología , Ligando Inductor de Apoptosis Relacionado con TNF
20.
J Immunol ; 174(9): 5630-5, 2005 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-15843562

RESUMEN

Modulation of T cell response is a novel property of 3-hydroxy-3-methylglutaryl (HMG)-CoA reductase inhibitors. Previously we reported the benefits of atorvastatin treatment in experimental autoimmune encephalomyelitis, the murine model of the T cell-mediated autoimmune disorder multiple sclerosis, in which a blockade of the T cell cycle by atorvastatin was attributed to an accumulation of the negative regulator p27(Kip1). We show in this report that, in line with the documented role of p27(Kip1) in T cell anergy, treatment with atorvastatin results in a deficient response to a second productive stimulus in human T cells. This effect of atorvastatin was dependent on HMG-CoA reduction and required IL-10 signaling. Importantly, atorvastatin induced an early and sustained phosphorylation of ERK1, but not ERK2, which was crucial for the induction of anergy. On the basis of the therapeutic impact of HMG-CoA reductase inhibitors, the present findings should pave the way for future therapeutic concepts related to tolerance induction in neuroinflammatory disorders such as multiple sclerosis.


Asunto(s)
Adyuvantes Inmunológicos/farmacología , Anergia Clonal/efectos de los fármacos , Anergia Clonal/inmunología , Ácidos Heptanoicos/farmacología , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Pirroles/farmacología , Linfocitos T/enzimología , Linfocitos T/inmunología , Atorvastatina , Butadienos/farmacología , Línea Celular , Humanos , Interleucina-10/fisiología , MAP Quinasa Quinasa 1/antagonistas & inhibidores , MAP Quinasa Quinasa 1/metabolismo , MAP Quinasa Quinasa 2/antagonistas & inhibidores , MAP Quinasa Quinasa 2/metabolismo , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Sistema de Señalización de MAP Quinasas/inmunología , Proteína Quinasa 1 Activada por Mitógenos/antagonistas & inhibidores , Nitrilos/farmacología , Fosforilación/efectos de los fármacos , Inhibidores de Proteínas Quinasas/farmacología , Linfocitos T/efectos de los fármacos , Factores de Tiempo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...