Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
1.
J Immunol ; 195(4): 1399-1407, 2015 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-26150529

RESUMEN

Multiple sclerosis (MS) is an immune-mediated demyelinating disease of the CNS that has been linked with defects in regulatory T cell function. Therefore, strategies to selectively target pathogenic cells via enhanced regulatory T cell activity may provide therapeutic benefit. Kv1.3 is a voltage-gated potassium channel expressed on myelin-reactive T cells from MS patients. Kv1.3-knockout (KO) mice are protected from experimental autoimmune encephalomyelitis, an animal model of MS, and Kv1.3-KO Th cells display suppressive capacity associated with increased IL-10. In this article, we demonstrate that myelin oligodendrocyte glycoprotein-specific Kv1.3-KO Th cells exhibit a unique regulatory phenotype characterized by high CD25, CTLA4, pSTAT5, FoxO1, and GATA1 expression without a corresponding increase in Foxp3. These phenotypic changes result from increased signaling through IL-2R. Moreover, myelin oligodendrocyte glycoprotein-specific Kv1.3-KO Th cells can ameliorate experimental autoimmune encephalomyelitis following transfer to wild-type recipients in a manner that is partially dependent on IL-2R and STAT5 signaling. The present study identifies a population of Foxp3(-) T cells with suppressive properties that arises in the absence of Kv1.3 and enhances the understanding of the molecular mechanism by which these cells are generated. This increased understanding could contribute to the development of novel therapies for MS patients that promote heightened immune regulation.


Asunto(s)
Antígenos/inmunología , Factores de Transcripción Forkhead/metabolismo , Canal de Potasio Kv1.3/deficiencia , Subgrupos de Linfocitos T/inmunología , Subgrupos de Linfocitos T/metabolismo , Linfocitos T Colaboradores-Inductores/inmunología , Linfocitos T Colaboradores-Inductores/metabolismo , Animales , Antígeno CTLA-4/genética , Antígeno CTLA-4/metabolismo , Calcio/metabolismo , Citocinas/biosíntesis , Modelos Animales de Enfermedad , Encefalomielitis Autoinmune Experimental/genética , Encefalomielitis Autoinmune Experimental/inmunología , Encefalomielitis Autoinmune Experimental/metabolismo , Proteína Forkhead Box O1 , Factores de Transcripción Forkhead/genética , Factor de Transcripción GATA1/genética , Factor de Transcripción GATA1/metabolismo , Expresión Génica , Inmunomodulación , Inmunofenotipificación , Subunidad alfa del Receptor de Interleucina-2/genética , Subunidad alfa del Receptor de Interleucina-2/metabolismo , Ratones , Ratones Noqueados , Esclerosis Múltiple/genética , Esclerosis Múltiple/inmunología , Esclerosis Múltiple/metabolismo , Glicoproteína Mielina-Oligodendrócito/inmunología , Factores de Transcripción NFATC/metabolismo , Fenotipo , Fosforilación , Factor de Transcripción STAT5/metabolismo , Transducción de Señal
2.
J Neurosci ; 35(22): 8626-39, 2015 Jun 03.
Artículo en Inglés | MEDLINE | ID: mdl-26041928

RESUMEN

Multiple sclerosis (MS) is a demyelinating disease of the CNS characterized by inflammation and neurodegeneration. Animal models that enable the study of remyelination in the context of ongoing inflammation are greatly needed for the development of novel therapies that target the pathological inhibitory cues inherent to the MS plaque microenvironment. We report the development of an innovative animal model combining cuprizone-mediated demyelination with transfer of myelin-reactive CD4(+) T cells. Characterization of this model reveals both Th1 and Th17 CD4(+) T cells infiltrate the CNS of cuprizone-fed mice, with infiltration of Th17 cells being more efficient. Infiltration correlates with impaired spontaneous remyelination as evidenced by myelin protein expression, immunostaining, and ultrastructural analysis. Electron microscopic analysis further reveals that demyelinated axons are preserved but reduced in caliber. Examination of the immune response contributing to impaired remyelination highlights a role for peripheral monocytes with an M1 phenotype. This study demonstrates the development of a novel animal model that recapitulates elements of the microenvironment of the MS plaque and reveals an important role for T cells and peripheral monocytes in impairing endogenous remyelination in vivo. This model could be useful for testing putative MS therapies designed to enhance remyelination in the setting of active inflammation, and may also facilitate modeling the pathophysiology of denuded axons, which has been a challenge in rodents because they typically remyelinate very quickly.


Asunto(s)
Sistema Nervioso Central/patología , Cuprizona/toxicidad , Enfermedades Desmielinizantes/terapia , Inhibidores de la Monoaminooxidasa/toxicidad , Vaina de Mielina/metabolismo , Células Th17/fisiología , Traslado Adoptivo , Animales , Polaridad Celular/efectos de los fármacos , Células Cultivadas , Sistema Nervioso Central/ultraestructura , Enfermedades Desmielinizantes/inducido químicamente , Modelos Animales de Enfermedad , Adyuvante de Freund/toxicidad , Interleucina-17/metabolismo , Antígenos Comunes de Leucocito/genética , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Monocitos/patología , Monocitos/ultraestructura , Proteínas de la Mielina/metabolismo , Glicoproteína Mielina-Oligodendrócito/toxicidad , Infiltración Neutrófila , Fragmentos de Péptidos/toxicidad , Regeneración/efectos de los fármacos , Células Th17/ultraestructura , Factores de Tiempo
3.
Proc Natl Acad Sci U S A ; 110(52): 21101-6, 2013 Dec 24.
Artículo en Inglés | MEDLINE | ID: mdl-24324134

RESUMEN

Pharmacologic targeting of T helper (TH) cell trafficking poses an attractive opportunity for amelioration of autoimmune diseases such as multiple sclerosis (MS). MS risk is associated with vitamin D deficiency, and its bioactive form, 1,25-dihydroxyvitamin D3 [1,25(OH)2D3], has been shown to prevent experimental autoimmune encephalomyelitis, a mouse model of MS, via an incompletely understood mechanism. Herein, we systematically examined 1,25(OH)2D3 effects on TH cells during their migration from the lymph nodes to the CNS. Our data demonstrate that myelin-reactive TH cells are successfully generated in the presence of 1,25(OH)2D3, secrete proinflammatory cytokines, and do not preferentially differentiate into suppressor T cells. These cells are able to leave the lymph node, enter the peripheral circulation, and migrate to the s.c. immunization sites. However, TH cells from 1,25(OH)2D3-treated mice are unable to enter the CNS parenchyma but are instead maintained in the periphery. Upon treatment cessation, mice rapidly develop experimental autoimmune encephalomyelitis, demonstrating that 1,25(OH)2D3 prevents the disease only temporarily likely by halting TH cell migration into the CNS.


Asunto(s)
Calcitriol/farmacología , Movimiento Celular/efectos de los fármacos , Sistema Nervioso Central/inmunología , Encefalomielitis Autoinmune Experimental/inmunología , Esclerosis Múltiple/inmunología , Linfocitos T Colaboradores-Inductores/efectos de los fármacos , Animales , Movimiento Celular/inmunología , Citocinas/metabolismo , Encefalomielitis Autoinmune Experimental/prevención & control , Ensayo de Inmunoadsorción Enzimática , Mediciones Luminiscentes , Ratones , Ratones Endogámicos C57BL , Estadísticas no Paramétricas , Linfocitos T Colaboradores-Inductores/inmunología , Linfocitos T Colaboradores-Inductores/metabolismo
4.
J Immunol ; 188(12): 5877-86, 2012 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-22581856

RESUMEN

Increasing evidence suggests ion channels have critical functions in the differentiation and plasticity of T cells. Kv1.3, a voltage-gated K(+) channel, is a functional marker and a pharmacological target for activated effector memory T cells. Selective Kv1.3 blockers have been shown to inhibit proliferation and cytokine production by human and rat effector memory T cells. We used Kv1.3 knockout (KO) mice to investigate the mechanism by which Kv1.3 blockade affects CD4(+) T cell differentiation during an inflammatory immune-mediated disease. Kv1.3 KO animals displayed significantly lower incidence and severity of myelin oligodendrocyte glycoprotein (MOG) peptide-induced experimental autoimmune encephalomyelitis. Kv1.3 was the only K(V) channel expressed in MOG 35-55-specific CD4(+) T cell blasts, and no K(V) current was present in MOG-specific CD4(+) T cell-blasts from Kv1.3 KO mice. Fewer CD4(+) T cells migrated to the CNS in Kv1.3 KO mice following disease induction, and Ag-specific proliferation of CD4(+) T cells from these mice was impaired with a corresponding cell-cycle delay. Kv1.3 was required for optimal expression of IFN-γ and IL-17, whereas its absence led to increased IL-10 production. Dendritic cells from Kv1.3 KO mice fully activated wild-type CD4(+) T cells, indicating a T cell-intrinsic defect in Kv1.3 KO mice. The loss of Kv1.3 led to a suppressive phenotype, which may contribute to the mechanism by which deletion of Kv1.3 produces an immunotherapeutic effect. Skewing of CD4(+) T cell differentiation toward Ag-specific regulatory T cells by pharmacological blockade or genetic suppression of Kv1.3 might be beneficial for therapy of immune-mediated diseases such as multiple sclerosis.


Asunto(s)
Encefalomielitis Autoinmune Experimental/inmunología , Encefalomielitis Autoinmune Experimental/metabolismo , Canal de Potasio Kv1.3/metabolismo , Activación de Linfocitos/inmunología , Linfocitos T Reguladores/inmunología , Animales , Western Blotting , Linfocitos T CD4-Positivos/citología , Linfocitos T CD4-Positivos/inmunología , Diferenciación Celular/inmunología , Electrofisiología , Encefalomielitis Autoinmune Experimental/patología , Ensayo de Inmunoadsorción Enzimática , Femenino , Citometría de Flujo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Fenotipo , Subgrupos de Linfocitos T/citología , Subgrupos de Linfocitos T/inmunología , Linfocitos T Reguladores/citología
5.
J Biol Chem ; 287(2): 1261-8, 2012 Jan 06.
Artículo en Inglés | MEDLINE | ID: mdl-22110135

RESUMEN

The maintenance of T cell memory is critical for the development of rapid recall responses to pathogens, but may also have the undesired side effect of clonal expansion of T effector memory (T(EM)) cells in chronic autoimmune diseases. The mechanisms by which lineage differentiation of T cells is controlled have been investigated, but are not completely understood. Our previous work demonstrated a role of the voltage-gated potassium channel Kv1.3 in effector T cell function in autoimmune disease. In the present study, we have identified a mechanism by which Kv1.3 regulates the conversion of T central memory cells (T(CM)) into T(EM). Using a lentiviral-dominant negative approach, we show that loss of function of Kv1.3 mediates reversion of T(EM) into T(CM), via a delay in cell cycle progression at the G2/M stage. The inhibition of Kv1.3 signaling caused an up-regulation of SMAD3 phosphorylation and induction of nuclear p21(cip1) with resulting suppression of Cdk1 and cyclin B1. These data highlight a novel role for Kv1.3 in T cell differentiation and memory responses, and provide further support for the therapeutic potential of Kv1.3 specific channel blockers in T(EM)-mediated autoimmune diseases.


Asunto(s)
Linfocitos T CD4-Positivos/inmunología , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/inmunología , Memoria Inmunológica , Canal de Potasio Kv1.3/inmunología , Transducción de Señal/inmunología , Proteína smad3/inmunología , Enfermedades Autoinmunes/genética , Enfermedades Autoinmunes/inmunología , Enfermedades Autoinmunes/metabolismo , Linfocitos T CD4-Positivos/metabolismo , Proteína Quinasa CDC2/genética , Proteína Quinasa CDC2/inmunología , Proteína Quinasa CDC2/metabolismo , Puntos de Control del Ciclo Celular/genética , Puntos de Control del Ciclo Celular/inmunología , División Celular/genética , División Celular/inmunología , Células Cultivadas , Ciclina B1/genética , Ciclina B1/inmunología , Ciclina B1/metabolismo , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/genética , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/metabolismo , Fase G2/genética , Fase G2/inmunología , Humanos , Canal de Potasio Kv1.3/genética , Canal de Potasio Kv1.3/metabolismo , Fosforilación/genética , Fosforilación/inmunología , Transducción de Señal/genética , Proteína smad3/genética , Proteína smad3/metabolismo
6.
Cell Immunol ; 284(1-2): 68-74, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23933188

RESUMEN

Double-negative T (DNT) cells are αßTCR(+)CD3(+)CD4(-)CD8(-)NK1.1(-) cells that constitute a small but significant proportion of the αßTCR(+) T cells. Their developmental pathway and pathological significance remain unclear. In the present study, we utilized chronic in vitro stimulation of CD4(+) T cells to mimic immune hyper-activation of autoimmune lymphoproliferative syndrome and systemic lupus erythematosus, conditions characterized by DNT cells accumulation. After approximately 4-5 rounds of stimulation, the CD3(+)CD4(-) population became apparent. These cells did not express CD8, NK1.1, γδTCR, or B220, exhibited a highly proliferative effector phenotype, and were dependent on T cell receptor (TCR) stimulation for survival. Moreover, CD3(+)CD4(-) cells expressed MHC class II-restricted αßTCR, indicative of their origin from a CD4(+) T cell population. The results presented herein illustrate a novel method of DNT cell generation in vitro and suggest that immune hyper-activation could also be implicated in the genesis of the disease-associated DNT cells in vivo.


Asunto(s)
Síndrome Linfoproliferativo Autoinmune/inmunología , Linfocitos T CD4-Positivos/inmunología , Lupus Eritematoso Sistémico/inmunología , Receptores de Antígenos de Linfocitos T alfa-beta/inmunología , Linfocitos T Colaboradores-Inductores/inmunología , Animales , Proliferación Celular , Regulación hacia Abajo , Femenino , Citometría de Flujo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Fenotipo
7.
Ann Neurol ; 67(4): 498-507, 2010 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-20437585

RESUMEN

OBJECTIVE: To determine if suppressing Nogo-A, an axonal inhibitory protein, will promote functional recovery in a murine model of multiple sclerosis (MS). METHODS: A small interfering RNA was developed to specifically suppress Nogo-A (siRNA-NogoA). The siRNA-NogoA silencing effect was evaluated in vitro and in vivo via immunohistochemistry. The siRNA was administered intravenously in 2 models of experimental autoimmune encephalomyelitis (EAE). Axonal repair was measured by upregulation of GAP43. Enzyme-linked immunosorbent assay, flow cytometry, and (3)H-thymidine incorporation were used to determine immunological changes in myelin-specific T cells in mice with EAE. RESULTS: The siRNA-NogoA suppressed Nogo-A expression in vitro and in vivo. Systemic administration of siRNA-NogoA ameliorated EAE and promoted axonal repair, as demonstrated by enhanced GAP43+ axons in the lesions. Myelin-specific T-cell proliferation and cytokine production were unchanged in the siRNA-NogoA-treated mice. INTERPRETATION: Silencing Nogo-A in EAE promotes functional recovery. The therapeutic benefit appears to be mediated by axonal growth and repair, and is not attributable to changes in the encephalitogenic capacity of the myelin-specific T cells. Silencing Nogo-A may be a therapeutic option for MS patients to prevent permanent functional deficits caused by immune-mediated axonal damage.


Asunto(s)
Encefalomielitis Autoinmune Experimental/tratamiento farmacológico , Proteínas de la Mielina/metabolismo , ARN Interferente Pequeño/uso terapéutico , Animales , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Modelos Animales de Enfermedad , Encefalomielitis Autoinmune Experimental/metabolismo , Encefalomielitis Autoinmune Experimental/patología , Ensayo de Inmunoadsorción Enzimática/métodos , Citometría de Flujo/métodos , Proteína GAP-43/genética , Proteína GAP-43/metabolismo , Regulación de la Expresión Génica/efectos de los fármacos , Regulación de la Expresión Génica/genética , Glicoproteínas/efectos adversos , Interferón gamma/metabolismo , Interleucina-10/metabolismo , Linfocitos/efectos de los fármacos , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Proteína Básica de Mielina/genética , Proteína Básica de Mielina/farmacología , Proteínas de la Mielina/genética , Glicoproteína Mielina-Oligodendrócito , Neuroblastoma , Proteínas Nogo , Fragmentos de Péptidos/efectos adversos , Fragmentos de Péptidos/genética , Fragmentos de Péptidos/farmacología , ARN Interferente Pequeño/genética , Médula Espinal/metabolismo , Acetato de Tetradecanoilforbol/análogos & derivados , Acetato de Tetradecanoilforbol/farmacología , Transfección/métodos
8.
J Immunol ; 182(7): 4479-87, 2009 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-19299749

RESUMEN

Peroxisome proliferator-activated receptor-alpha (PPARalpha) agonists have been shown to have a therapeutic benefit in experimental autoimmune encephalomyelitis (EAE), an animal model for multiple sclerosis (MS). In this study, we investigated the mechanism by which the PPARalpha agonist gemfibrozil induces immune deviation and protects mice from EAE. We demonstrated that treatment with gemfibrozil increases expression of the Th2 transcription factor GATA-3 and decreases expression of the Th1 transcription factor T-bet in vitro and directly ex vivo. These changes correlated with an increase in nuclear PPARalpha expression. Moreover, the protective effects of PPARalpha agonists in EAE were shown to be partially dependent on IL-4 and to occur in a receptor-dependent manner. PPARalpha was demonstrated, for the first time, to regulate the IL-4 and IL-5 genes and to bind the IL-4 promoter in the presence of steroid receptor coactivator-1, indicating that PPARalpha can directly transactivate the IL-4 gene. Finally, therapeutic administration of PPARalpha agonists ameliorated clinically established EAE, suggesting that PPARalpha agonists may provide a treatment option for immune-mediated inflammatory diseases.


Asunto(s)
Encefalomielitis Autoinmune Experimental/inmunología , Gemfibrozilo/farmacología , Factores Inmunológicos/farmacología , PPAR alfa/antagonistas & inhibidores , Transcripción Genética/efectos de los fármacos , Animales , Western Blotting , Citocinas/biosíntesis , Encefalomielitis Autoinmune Experimental/tratamiento farmacológico , Encefalomielitis Autoinmune Experimental/genética , Ensayo de Inmunoadsorción Enzimática , Fenofibrato/farmacología , Factor de Transcripción GATA3/biosíntesis , Factor de Transcripción GATA3/efectos de los fármacos , Regulación de la Expresión Génica/efectos de los fármacos , Regulación de la Expresión Génica/inmunología , Interleucina-4/genética , Interleucina-4/metabolismo , Interleucina-5/genética , Interleucina-5/metabolismo , Ratones , Ratones Transgénicos , Regiones Promotoras Genéticas/efectos de los fármacos , Proteínas de Dominio T Box/biosíntesis , Proteínas de Dominio T Box/efectos de los fármacos , Transfección
9.
Brain ; 133(Pt 2): 375-88, 2010 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-20145049

RESUMEN

The primary biological function of the endogenous cellular prion protein has remained unclear. We investigated its biological function in the generation of cellular immune responses using cellular prion protein gene-specific small interfering ribonucleic acid in vivo and in vitro. Our results were confirmed by blocking cellular prion protein with monovalent antibodies and by using cellular prion protein-deficient and -transgenic mice. In vivo prion protein gene-small interfering ribonucleic acid treatment effects were of limited duration, restricted to secondary lymphoid organs and resulted in a 70% reduction of cellular prion protein expression in leukocytes. Disruption of cellular prion protein signalling augmented antigen-specific activation and proliferation, and enhanced T cell receptor signalling, resulting in zeta-chain-associated protein-70 phosphorylation and nuclear factor of activated T cells/activator protein 1 transcriptional activity. In vivo prion protein gene-small interfering ribonucleic acid treatment promoted T cell differentiation towards pro-inflammatory phenotypes and increased survival of antigen-specific T cells. Cellular prion protein silencing with small interfering ribonucleic acid also resulted in the worsening of actively induced and adoptively transferred experimental autoimmune encephalomyelitis. Finally, treatment of myelin basic protein(1-11) T cell receptor transgenic mice with prion protein gene-small interfering ribonucleic acid resulted in spontaneous experimental autoimmune encephalomyelitis. Thus, central nervous system autoimmune disease was modulated at all stages of disease: the generation of the T cell effector response, the elicitation of T effector function and the perpetuation of cellular immune responses. Our findings indicate that cellular prion protein regulates T cell receptor-mediated T cell activation, differentiation and survival. Defects in autoimmunity are restricted to the immune system and not the central nervous system. Our data identify cellular prion protein as a regulator of cellular immunological homoeostasis and suggest cellular prion protein as a novel potential target for therapeutic immunomodulation.


Asunto(s)
Enfermedades Autoinmunes Desmielinizantes SNC/genética , Silenciador del Gen/inmunología , Priones/genética , Receptores de Antígenos de Linfocitos T/fisiología , Transducción de Señal/inmunología , Animales , Enfermedades Autoinmunes Desmielinizantes SNC/inmunología , Femenino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Priones/inmunología , ARN Interferente Pequeño/genética
10.
J Neuroimmunol ; 270(1-2): 13-21, 2014 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-24680062

RESUMEN

Fingolimod (FTY720) is a multiple sclerosis (MS) therapeutic that upon phosphorylation causes the internalization of sphingosine-1-phosphate receptors (S1PR) and traps CCR7+ T-cells in lymph nodes but relatively spares CCR7-effector T-cells. Nonetheless, FTY720-treated patients are more susceptible to viral infections, indicating a CD8 T-cell defect. Thus, the effects of FTY720 on CD8 T-cells were investigated. To this end, we utilized experimental autoimmune encephalomyelitis (EAE) and a murine influenza model. CD8 T-cell trafficking, IFNγ and Granzyme B (GrB) production were assessed by flow cytometry. CD8 T-cell cytotoxic function was assessed in vitro by an LDH release assay. FTY720 not only ameliorated EAE by sequestering T-cells, but also reduced IFNγ and Granzyme B (GrB) in splenic CD8 T-cells. Murine influenza infection was exacerbated and mortality was increased, as FTY720 inhibited CD8 T-cell GrB production and lung infiltration. Remarkably, only the unphosphorylated compound was able to reduce IFNγ and GrB levels in CD8 T-cells and inhibits their cytotoxic function in vitro. The phosphorylated moiety had no effect in vitro, indicating that CD8 T-cell suppression by FTY720 is independent of S1PR modulation. The addition of arachidonic acid rescued CD8 T-cell function, suggesting that this effect may be mediated via inhibition of cytosolic phospholipase A2. Herein, we demonstrate that FTY720 suppresses CD8 T-cells independently of its trafficking effects and S1PR modulation. This provides a novel explanation not only for the increased rate of viral infections in FTY720-treated patients, but also for its efficacy in MS, as CD8 T-cells have emerged as crucial mediators of MS pathogenesis.


Asunto(s)
Linfocitos T CD8-positivos/efectos de los fármacos , Encefalomielitis Autoinmune Experimental/inmunología , Inmunosupresores/farmacología , Infecciones por Orthomyxoviridae/inmunología , Glicoles de Propileno/farmacología , Esfingosina/análogos & derivados , Animales , Linfocitos T CD8-positivos/inmunología , Quimiotaxis de Leucocito/efectos de los fármacos , Femenino , Clorhidrato de Fingolimod , Citometría de Flujo , Granzimas/biosíntesis , Subtipo H1N1 del Virus de la Influenza A , Interferón gamma/biosíntesis , Lisofosfolípidos/metabolismo , Ratones , Ratones Endogámicos C57BL , Transducción de Señal/efectos de los fármacos , Esfingosina/metabolismo , Esfingosina/farmacología
11.
PLoS One ; 8(1): e54267, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23382885

RESUMEN

Increased expression of the voltage-gated potassium channel Kν1.3 on activated effector memory T cells (T(EM)) is associated with pathology in multiple sclerosis (MS). To date, most studies of Kν1.3 channels in MS have focused on CD4+ T(EM) cells. Much less is known about the functional relevance of Kv1.3 on CD8+ T(EM) cells. Herein, we examined the effects of Kν1.3 blockade on CD8+ T cell proliferation, differentiation into cytotoxic effector cells, and release of granzyme B (GrB), a key effector of CD8+ T cell-mediated cytotoxicity. We confirmed the expression of Kv1.3 channels on activated human CD8+ T lymphocytes by immunofluorescent staining. To test the functional relevance of the Kv1.3 channel in CD8+ T cells, we inhibited this channel via pharmacological blockers or a lentiviral-dominant negative (Kv1.xDN) approach and determined the effects of the blockade on critical pathogenic parameters of CD8+ T cells. We found that blockade of Kv1.3 with both lentivirus and pharmacologic agents effectively inhibited cytotoxic effector memory cells' proliferation, secretion of GrB, and their ability to kill neural progenitor cells. Intriguingly, the KvDN transduced T cells exhibited arrested differentiation from central memory (T(CM)) to effector memory (T(EM)) states. Transduction of cells that had already differentiated into T(EM) with KvDN led to their conversion into T(CM). CD8+ T(EM) have a critical role in MS and other autoimmune diseases. Our present results indicate a critical role for Kv1.3 in the conversion of CD8+ T cells into potential pathogenic effector cells with cytotoxic function.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Diferenciación Celular/efectos de los fármacos , Granzimas/metabolismo , Canal de Potasio Kv1.3/metabolismo , Linfocitos T CD8-positivos/citología , Linfocitos T CD8-positivos/efectos de los fármacos , Diferenciación Celular/genética , Proliferación Celular/efectos de los fármacos , Granzimas/genética , Granzimas/inmunología , Humanos , Memoria Inmunológica/efectos de los fármacos , Memoria Inmunológica/genética , Canal de Potasio Kv1.3/antagonistas & inhibidores , Canal de Potasio Kv1.3/genética , Activación de Linfocitos/inmunología , Bloqueadores de los Canales de Potasio/farmacología
12.
J Neuroimmunol ; 249(1-2): 86-92, 2012 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-22633193

RESUMEN

We investigated CD45RA and CCR7 expression in CD4+ and CD8+ subsets of cerebrospinal fluid (CSF) lymphocytes, both immediately ex vivo and after stimulation, from 134 patients with a variety of inflammatory and non-inflammatory neurological diseases. Most inflammatory diseases had a higher CD4+:CD8+ ratio and higher percentage of effector memory T cells (T(EM)) than non-inflammatory controls, excluding active infection. Moreover, we found that patients with highly elevated cell counts in the CSF tended to have a lower percentage of central memory T cells (T(CM)) than patients with low or absent pleocytosis, with a concomitant increase in T(EM). We also found that samples with elevated IgG index or presence of oligoclonal bands had a significantly higher CD4+:CD8+ ratio than normal samples, consistent with increased CD4+ help for intrathecal IgG synthesis by B cells.


Asunto(s)
Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD8-positivos/inmunología , Enfermedades del Sistema Nervioso Central/líquido cefalorraquídeo , Subgrupos de Linfocitos T/inmunología , Adolescente , Adulto , Anciano , Anciano de 80 o más Años , Linfocitos T CD4-Positivos/metabolismo , Linfocitos T CD8-positivos/metabolismo , Enfermedades del Sistema Nervioso Central/inmunología , Enfermedades del Sistema Nervioso Central/metabolismo , Niño , Preescolar , Femenino , Humanos , Inmunoglobulina G/líquido cefalorraquídeo , Lactante , Inflamación/líquido cefalorraquídeo , Inflamación/inmunología , Inflamación/metabolismo , Antígenos Comunes de Leucocito/biosíntesis , Antígenos Comunes de Leucocito/líquido cefalorraquídeo , Antígenos Comunes de Leucocito/inmunología , Masculino , Persona de Mediana Edad , Receptores CCR7/análisis , Receptores CCR7/biosíntesis , Receptores CCR7/inmunología , Subgrupos de Linfocitos T/metabolismo , Adulto Joven
13.
J Exp Med ; 208(11): 2291-303, 2011 Oct 24.
Artículo en Inglés | MEDLINE | ID: mdl-21987655

RESUMEN

Fumarates improve multiple sclerosis (MS) and psoriasis, two diseases in which both IL-12 and IL-23 promote pathogenic T helper (Th) cell differentiation. However, both diseases show opposing responses to most established therapies. First, we show in humans that fumarate treatment induces IL-4-producing Th2 cells in vivo and generates type II dendritic cells (DCs) that produce IL-10 instead of IL-12 and IL-23. In mice, fumarates also generate type II DCs that induce IL-4-producing Th2 cells in vitro and in vivo and protect mice from experimental autoimmune encephalomyelitis. Type II DCs result from fumarate-induced glutathione (GSH) depletion, followed by increased hemoxygenase-1 (HO-1) expression and impaired STAT1 phosphorylation. Induced HO-1 is cleaved, whereupon the N-terminal fragment of HO-1 translocates into the nucleus and interacts with AP-1 and NF-κB sites of the IL-23p19 promoter. This interaction prevents IL-23p19 transcription without affecting IL-12p35, whereas STAT1 inactivation prevents IL-12p35 transcription without affecting IL-23p19. As a consequence, GSH depletion by small molecules such as fumarates induces type II DCs in mice and in humans that ameliorate inflammatory autoimmune diseases. This therapeutic approach improves Th1- and Th17-mediated autoimmune diseases such as psoriasis and MS by interfering with IL-12 and IL-23 production.


Asunto(s)
Células Dendríticas/inmunología , Fumaratos/inmunología , Fumaratos/uso terapéutico , Esclerosis Múltiple/tratamiento farmacológico , Esclerosis Múltiple/inmunología , Psoriasis/tratamiento farmacológico , Psoriasis/inmunología , Animales , Diferenciación Celular/inmunología , Células Dendríticas/citología , Células Dendríticas/efectos de los fármacos , Encefalomielitis Autoinmune Experimental/inmunología , Femenino , Hemo-Oxigenasa 1/metabolismo , Humanos , Interleucina-12/inmunología , Interleucina-23/inmunología , Macrófagos/inmunología , Ratones , Células 3T3 NIH , Regiones Promotoras Genéticas , Especies Reactivas de Oxígeno/metabolismo , Transducción de Señal/inmunología , Linfocitos T/citología , Linfocitos T/inmunología , Factor de Transcripción ReIA/metabolismo
15.
Chem Biol ; 16(11): 1133-9, 2009 Nov 25.
Artículo en Inglés | MEDLINE | ID: mdl-19942136

RESUMEN

Antigen-specific T cells play a major role in mediating the pathogenesis of a variety of autoimmune conditions as well as other diseases. In the context of experimental autoimmune encephalomyelitis, a murine model of multiple sclerosis, we present here a general approach to the discovery of highly specific ligands for autoreactive cells. These ligands are obtained from a combinatorial library of hundreds of thousands of synthetic peptoids that is screened simultaneously against two populations of CD4+ T cells. Peptoids that recognize autoreactive T cells with extremely high specificity can be identified in the library. Since no specific knowledge is required regarding the nature of the native antigens recognized by the autoreactive T cells, this technology provides a powerful tool for the enrichment and inhibition of autoimmune cells in a variety of disease states.


Asunto(s)
Linfocitos T CD4-Positivos/inmunología , Encefalomielitis Autoinmune Experimental/inmunología , Peptoides/farmacología , Animales , Linfocitos T CD4-Positivos/efectos de los fármacos , Linfocitos T CD4-Positivos/metabolismo , Proliferación Celular , Técnicas Químicas Combinatorias , Encefalomielitis Autoinmune Experimental/metabolismo , Femenino , Citometría de Flujo , Ratones , Ratones Transgénicos , Proteína Básica de Mielina/inmunología , Fragmentos de Péptidos/inmunología , Biblioteca de Péptidos , Peptoides/química
16.
PPAR Res ; 2008: 546753, 2008.
Artículo en Inglés | MEDLINE | ID: mdl-18645614

RESUMEN

PPARs are members of the steroid hormone nuclear receptor superfamily and play an important role in the regulation of lipid metabolism, energy balance, artherosclerosis and glucose control. Recent studies suggest that they play an important role in regulating inflammation. This review will focus on PPAR-alpha regulation of the immune response. We describe how PPAR-alpha regulates differentiation of T cells by transactivation and/or interaction with other transcription factors. Moreover, PPAR-alpha agonists have been shown to ameliorate experimental autoimmune encephalomyelitis (EAE) in mice, suggesting that they could provide a therapy for human autoimmune diseases such as multiple sclerosis.

17.
J Immunol ; 178(3): 1341-8, 2007 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-17237380

RESUMEN

IL-17-producing T cells (Th17) have recently been implicated in the pathogenesis of experimental autoimmune encephalomyelitis (EAE), an animal model for the human disease multiple sclerosis. However, little is known about the transcription factors that regulate these cells. Although it is clear that the transcription factor T-bet plays an essential role in the differentiation of IFN-gamma-producing CD4(+) Th1 lymphocytes, the potential role of T-bet in the differentiation of Th17 cells is not completely understood. In this study, therapeutic administration of a small interfering RNA specific for T-bet significantly improved the clinical course of established EAE. The improved clinical course was associated with suppression of newly differentiated T cells that express IL-17 in the CNS as well as suppression of myelin basic protein-specific Th1 autoreactive T cells. Moreover, T-bet was found to directly regulate transcription of the IL-23R, and, in doing so, influenced the fate of Th17 cells, which depend on optimal IL-23 production for survival. We now show for the first time that suppression of T-bet ameliorates EAE by limiting the differentiation of autoreactive Th1 cells, as well as inhibiting pathogenic Th17 cells via regulation of IL-23R.


Asunto(s)
Autoinmunidad , Interleucina-17 , Interleucina-23/genética , Proteínas de Dominio T Box/fisiología , Linfocitos T Colaboradores-Inductores/inmunología , Células TH1/inmunología , Animales , Diferenciación Celular/inmunología , Encefalomielitis Autoinmune Experimental/tratamiento farmacológico , Encefalomielitis Autoinmune Experimental/etiología , Encefalomielitis Autoinmune Experimental/inmunología , Regulación de la Expresión Génica , Humanos , Ratones , Ratones Transgénicos , Proteína Básica de Mielina , Proteínas del Tejido Nervioso/inmunología , ARN Interferente Pequeño/farmacología , ARN Interferente Pequeño/uso terapéutico , Proteínas de Dominio T Box/inmunología , Subgrupos de Linfocitos T/inmunología , Factores de Transcripción/inmunología
18.
J Nutr ; 136(3): 700-3, 2006 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-16484546

RESUMEN

Experimental autoimmune encephalomyelitis (EAE) is a T-cell-mediated, autoimmune disorder characterized by central nervous system inflammation and demyelination, features reminiscent of the human disease, multiple sclerosis (MS). Prior work in the EAE model has suggested that encephalitogenic T cells are of the T helper (Th)-1 phenotype. Our group has performed several studies in the EAE model that suggest that a strategy for treating autoimmune disorders is to convert the pathogenic cells from the Th1 to Th2 phenotype. Peroxisome proliferator-activated receptors (PPARs) are members of a nuclear hormone receptor superfamily that include receptors for steroids, retinoids, and thyroid hormone, all of which are known to affect the immune response. Recently, we examined the role of PPARgamma in EAE and observed that administration of the PPARgamma agonist 15-deoxy-Delta(12,14) prostaglandin J2 exerted a significant therapeutic effect predominantly by inhibiting the activation and expansion of encephalitogenic T cells. One potential advantage in studying PPARalpha agonists is that they have been very well tolerated when used in humans to treat conditions such as elevated triglycerides. Building on prior work in immune deviation and with PPAR agonists, we have demonstrated that PPARalpha agonists can alter the cytokine phenotype of myelin-reactive T cells, alter their encephalitogenicity, and be useful in the treatment of EAE. The fact that PPARalpha agonists have been used as therapeutic agents in humans to treat metabolic conditions for over 25 years with little toxicity makes them attractive candidates for use as adjunctive therapies in MS.


Asunto(s)
Enfermedades Autoinmunes/inmunología , Esclerosis Múltiple/tratamiento farmacológico , Receptores Activados del Proliferador del Peroxisoma/agonistas , Receptores Citoplasmáticos y Nucleares/inmunología , Citocinas/genética , Regulación de la Expresión Génica/inmunología , Humanos , Modelos Inmunológicos , Esclerosis Múltiple/inmunología , PPAR alfa/inmunología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA