Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 37
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Acta Neuropathol ; 142(5): 899-915, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-34487221

RESUMEN

Multiple sclerosis (MS) is an inflammatory demyelinating disease of the central nervous system (CNS) characterized by varying degrees of secondary neurodegeneration. Retinal ganglion cells (RGC) are lost in MS in association with optic neuritis but the mechanisms of neuronal injury remain unclear. Complement component C3 has been implicated in retinal and cerebral synaptic pathology that may precede neurodegeneration. Herein, we examined post-mortem MS retinas, and then used a mouse model, experimental autoimmune encephalomyelitis (EAE), to examine the role of C3 in the pathogenesis of RGC loss associated with optic neuritis. First, we show extensive C3 expression in astrocytes (C3+/GFAP+ cells) and significant RGC loss (RBPMS+ cells) in post-mortem retinas from people with MS compared to retinas from non-MS individuals. A patient with progressive MS with a remote history of optic neuritis showed marked reactive astrogliosis with C3 expression in the inner retina extending into deeper layers in the affected eye more than the unaffected eye. To study whether C3 mediates retinal degeneration, we utilized global C3-/- EAE mice and found that they had less RGC loss and partially preserved neurites in the retina compared with C3+/+ EAE mice. C3-/- EAE mice had fewer axonal swellings in the optic nerve, reflecting reduced axonal injury, but had no changes in demyelination or T cell infiltration into the CNS. Using a C3-tdTomato reporter mouse line, we show definitive evidence of C3 expression in astrocytes in the retina and optic nerves of EAE mice. Conditional deletion of C3 in astrocytes showed RGC protection replicating the effects seen in the global knockouts. These data implicate astrocyte C3 expression as a critical mediator of retinal neuronal pathology in EAE and MS, and are consistent with recent studies showing C3 gene variants are associated with faster rates of retinal neurodegeneration in human disease.


Asunto(s)
Complemento C3/metabolismo , Esclerosis Múltiple/patología , Enfermedades Neuroinflamatorias/patología , Células Ganglionares de la Retina/patología , Animales , Astrocitos/inmunología , Astrocitos/metabolismo , Encefalomielitis Autoinmune Experimental/inmunología , Encefalomielitis Autoinmune Experimental/patología , Humanos , Ratones , Esclerosis Múltiple/inmunología , Degeneración Nerviosa/inmunología , Degeneración Nerviosa/patología , Enfermedades Neuroinflamatorias/inmunología , Nervio Óptico/patología , Neuritis Óptica/inmunología , Neuritis Óptica/patología
2.
Brain ; 142(9): 2722-2736, 2019 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-31289819

RESUMEN

Multiple sclerosis is a heterogeneous disease with an unpredictable course and a wide range of severity; some individuals rapidly progress to a disabled state whereas others experience only mild symptoms. Though genetic studies have identified variants that are associated with an increased risk of developing multiple sclerosis, no variants have been consistently associated with multiple sclerosis severity. In part, the lack of findings is related to inherent limitations of clinical rating scales; these scales are insensitive to early degenerative changes that underlie disease progression. Optical coherence tomography imaging of the retina and low-contrast letter acuity correlate with and predict clinical and imaging-based outcomes in multiple sclerosis. Therefore, they may serve as sensitive phenotypes to discover genetic predictors of disease course. We conducted a set of genome-wide association studies of longitudinal structural and functional visual pathway phenotypes in multiple sclerosis. First, we assessed genetic predictors of ganglion cell/inner plexiform layer atrophy in a discovery cohort of 374 patients with multiple sclerosis using mixed-effects models adjusting for age, sex, disease duration, optic neuritis and genetic ancestry and using a combination of single-variant and network-based analyses. For candidate variants identified in discovery, we conducted a similar set of analyses of ganglion cell/inner plexiform layer thinning in a replication cohort (n = 376). Second, we assessed genetic predictors of sustained loss of 5-letters in low-contrast letter acuity in discovery (n = 582) using multivariable-adjusted Cox proportional hazards models. We then evaluated candidate variants/pathways in a replication cohort. (n = 253). Results of both studies revealed novel subnetworks highly enriched for connected genes in early complement activation linked to measures of disease severity. Within these networks, C3 was the gene most strongly associated with ganglion cell/inner plexiform layer atrophy (P = 0.004) and C1QA and CR1 were top results in analysis of sustained low-contrast letter acuity loss. Namely, variant rs158772, linked to C1QA, and rs61822967, linked to CR1, were associated with 71% and 40% increases in risk of sustained LCLA loss, respectively, in meta-analysis pooling discovery and replication cohorts (rs158772: hazard ratio: 1.71; 95% confidence interval 1.30-2.25; P = 1.3 × 10-4; rs61822967: hazard ratio: 1.40; 95% confidence interval: 1.16-1.68; P = 4.1 × 10-4). In conclusion, early complement pathway gene variants were consistently associated with structural and functional measures of multiple sclerosis severity. These results from unbiased analyses are strongly supported by several prior reports that mechanistically implicated early complement factors in neurodegeneration.


Asunto(s)
Proteínas del Sistema Complemento/genética , Redes Reguladoras de Genes/genética , Esclerosis Múltiple/genética , Degeneración Nerviosa/genética , Vías Visuales/fisiopatología , Adulto , Ensayos Clínicos Fase III como Asunto , Método Doble Ciego , Femenino , Estudios de Seguimiento , Heterogeneidad Genética , Estudio de Asociación del Genoma Completo , Genotipo , Humanos , Masculino , Persona de Mediana Edad , Esclerosis Múltiple/inmunología , Esclerosis Múltiple/patología , Polimorfismo de Nucleótido Simple , Modelos de Riesgos Proporcionales , Estudios Prospectivos , Ensayos Clínicos Controlados Aleatorios como Asunto , Retina/patología , Tomografía de Coherencia Óptica
3.
Stem Cells ; 32(10): 2744-55, 2014 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-24911892

RESUMEN

Mesenchymal stem cells (MSC) have emerged as a promising candidate for inflammatory suppression and disease amelioration, especially of neuro-inflammatory diseases such as multiple sclerosis (MS). Auto-reactive CD4+ and CD8+ T cells acquire pathogenic IFNγ-producing- (Type I) and IL-17A-producing- (Type 17) effector phenotypes in MS and its animal model experimental autoimmune encephalomyelitis (EAE). Although MSC have been extensively demonstrated to suppress pathogenic effector CD4+ T cells and CD4+ T cell-mediated EAE, surprisingly few studies have addressed their modulation of effector CD8+ T cells represented in MS or their impact on CD8+ T cell-mediated EAE. We find that MSC differentially modulate CD8+ T cell development depending on effector T cell subtype. MSC drive activated low-IFNγ producers toward an enhanced high-IFNγ Tc1-like phenotype but strongly inhibit the production of IL-17A and Tc17 polarization in vitro. These observations are underscored by differential MSC modulation of T cell activation, proliferation, and signature transcription factor up-regulation. In addition, effector CD8+ T cells co-cultured with MSC exhibited increased production of IL-2, a molecule known to enhance IFNγ, yet suppress IL-17A, production. Based on these in vitro effects on CD8+ T cells, we next evaluated their impact on the severity of EAE. To better evaluate CD8+ T cells, we immunized mice with MOG37-50 , which is a CD8-targeted epitope. Our results revealed a worsening of disease, consistent with their in vitro stimulation of Tc1 cells. These findings highlight the emerging duality of MSC in immune modulation and provide implications for their future use in immune-related diseases.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Progresión de la Enfermedad , Encefalomielitis Autoinmune Experimental/inmunología , Encefalomielitis Autoinmune Experimental/patología , Células Madre Mesenquimatosas/citología , Subgrupos de Linfocitos T/inmunología , Animales , Apoptosis , Linfocitos T CD8-positivos/citología , Proliferación Celular , Técnicas de Cocultivo , Femenino , Interferón gamma/biosíntesis , Interleucina-2/biosíntesis , Activación de Linfocitos/inmunología , Ratones Endogámicos C57BL , Glicoproteína Mielina-Oligodendrócito/inmunología , Proteínas de Dominio T Box/metabolismo , Subgrupos de Linfocitos T/citología
4.
J Biol Chem ; 288(33): 23868-74, 2013 Aug 16.
Artículo en Inglés | MEDLINE | ID: mdl-23846700

RESUMEN

The initiation and maintenance of the immune response require a coordinated regulation of signal transduction pathways. Identifying the mechanisms by which these pathways are controlled and modulated is a significant goal of immunology. In the present report, we show a novel role for the zinc finger transcription factor Kruppel-like factor 4 (KLF4) in the modulation of the inflammatory immune response via its regulation of IL-6. We analyzed the role of KLF4 in the production of IL-6 by dendritic cells. Our data indicate that KLF4 can act in a dual function manner. It acts as a transcription factor in that it can bind to and activate the IL-6 promoter at specific binding sites. KLF4 also has a role in the chromatin remodeling of the IL-6 promoter in that cells deficient in KLF4 exhibited a relative hypoacetylation. These results indicate a molecular role for KLF4 in modulating the intensity of the inflammatory response and help to explain its pleiotropic role in different settings.


Asunto(s)
Epigénesis Genética , Interleucina-6/genética , Factores de Transcripción de Tipo Kruppel/metabolismo , Regiones Promotoras Genéticas , Acetilación , Animales , Secuencia de Bases , Sitios de Unión , Secuencia de Consenso/genética , Células Dendríticas/metabolismo , Células HEK293 , Histonas/metabolismo , Humanos , Interleucina-6/metabolismo , Factor 4 Similar a Kruppel , Factores de Transcripción de Tipo Kruppel/deficiencia , Ratones , Ratones Noqueados , Datos de Secuencia Molecular , FN-kappa B/metabolismo , Unión Proteica/genética , Factor de Transcripción STAT3/metabolismo , Transducción de Señal/genética , Transcripción Genética
5.
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
6.
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
7.
Nature ; 445(7126): 433-6, 2007 Jan 25.
Artículo en Inglés | MEDLINE | ID: mdl-17230191

RESUMEN

Feedback regulation of adaptive immunity is a fundamental mechanism for controlling the overall output of different signal transduction pathways, including that mediated by the T-cell antigen receptor (TCR). Calcineurin and Ras are known to have essential functions during T-cell activation. However, how the calcineurin signalling pathway is terminated in the process is still largely unknown. Although several endogenous inhibitors of calcineurin have been reported, none fulfils the criteria of a feedback inhibitor, as their expression is not responsive to TCR signalling. Here we identify an endogenous inhibitor of calcineurin, named Carabin, which also inhibits the Ras signalling pathway through its intrinsic Ras GTPase-activating protein (GAP) activity. Expression of Carabin is upregulated on TCR signalling in a manner that is sensitive to inhibitors of calcineurin, indicating that Carabin constitutes part of a negative regulatory loop for the intracellular TCR signalling pathway. Knockdown of Carabin by short interfering RNA led to a significant enhancement of interleukin-2 production by antigen-specific T cells in vitro and in vivo. Thus, Carabin is a negative feedback inhibitor of the calcineurin signalling pathway that also mediates crosstalk between calcineurin and Ras.


Asunto(s)
Inhibidores de la Calcineurina , Proteínas Portadoras/metabolismo , Retroalimentación Fisiológica , Proteínas ras/antagonistas & inhibidores , Proteínas Adaptadoras Transductoras de Señales , Animales , Linfocitos T CD4-Positivos/citología , Linfocitos T CD4-Positivos/metabolismo , Calcineurina/metabolismo , Proteínas Portadoras/genética , Células Cultivadas , Proteínas Activadoras de GTPasa , Regulación de la Expresión Génica , Humanos , Interleucina-2/biosíntesis , Células Jurkat , Ratones , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Fosforilación , Unión Proteica , Transducción de Señal , Proteínas Activadoras de ras GTPasa/metabolismo , Proteínas ras/metabolismo
8.
J Immunol ; 185(12): 7161-4, 2010 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-21076063

RESUMEN

Th17 cells play a significant role in inflammatory and autoimmune responses. Although a number of molecular pathways that contribute to the lineage differentiation of T cells have been discovered, the mechanisms by which lineage commitment occurs are not fully understood. Transcription factors play a key role in driving T cells toward specific lineages. We have identified a role for the transcription factor Kruppel-like factor (KLF) 4 in the development of IL-17-producing CD4(+) T cells. KLF4 was required for the production of IL-17, and further, chromatin immunoprecipitation analysis demonstrated binding of KLF4 to the IL-17 promoter, indicating a direct effect on the regulation of IL-17. Further, KLF4-deficient T cells upregulated expression of retinoic acid-related orphan receptor γt similar to wild-type during the polarization process toward Th17, suggesting that these two transcription factors are regulated independently.


Asunto(s)
Diferenciación Celular/inmunología , Factores de Transcripción de Tipo Kruppel/inmunología , Miembro 3 del Grupo F de la Subfamilia 1 de Receptores Nucleares/inmunología , Regiones Promotoras Genéticas/inmunología , Células Th17/inmunología , Regulación hacia Arriba/inmunología , Animales , Diferenciación Celular/genética , Interleucina-17/genética , Interleucina-17/inmunología , Factor 4 Similar a Kruppel , Factores de Transcripción de Tipo Kruppel/genética , Ratones , Ratones Noqueados , Miembro 3 del Grupo F de la Subfamilia 1 de Receptores Nucleares/genética , Regiones Promotoras Genéticas/genética , Células Th17/citología , Regulación hacia Arriba/genética
9.
Blood ; 113(13): 2906-13, 2009 Mar 26.
Artículo en Inglés | MEDLINE | ID: mdl-19164597

RESUMEN

Activation of dendritic cells (DCs) leads to cell maturation, which is accompanied by a regulated pattern of gene expression changes. Two significant and contradictory consequences of DC activation are that, although activation is necessary for maximal T-cell stimulation, it also leads to the initiation of gene expression that results ultimately in cell death. We have identified a gene, MINOR (mitogen-inducible nuclear orphan receptor), that becomes highly up-regulated on activation and whose expression leads to apoptosis in mature DCs. MINOR is a member of the Nur77 family of nuclear orphan receptors, which includes Nur77 and Nurr1. Although Nur77 and Nurr1 are expressed in macrophages and DCs, their expression levels do not change on DC activation. We thus tested the hypothesis that induction of MINOR would lead to an activation-induced cell death in DCs and that its inhibition would increase the lifespan of DCs and improve their vaccine efficacy. To block natural expression of MINOR by DCs, we generated a lentiviral vector that expresses a small interfering RNA. Our results indicate that blockade of MINOR expression dramatically decreases apoptosis in DCs and suggest that this approach may be a novel means to improve the potency of ex vivo-generated DC vaccines.


Asunto(s)
Proteínas de Unión al ADN/antagonistas & inhibidores , Células Dendríticas/efectos de los fármacos , Células Dendríticas/inmunología , Células Dendríticas/fisiología , Inmunoterapia Adoptiva , Proteínas del Tejido Nervioso/antagonistas & inhibidores , ARN Interferente Pequeño/farmacología , Receptores de Esteroides/antagonistas & inhibidores , Receptores de Hormona Tiroidea/antagonistas & inhibidores , Animales , Presentación de Antígeno/efectos de los fármacos , Presentación de Antígeno/genética , Vacunas contra el Cáncer/genética , Vacunas contra el Cáncer/inmunología , Vacunas contra el Cáncer/metabolismo , Muerte Celular/efectos de los fármacos , Muerte Celular/genética , Muerte Celular/inmunología , Células Cultivadas , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Células Dendríticas/metabolismo , Regulación hacia Abajo/efectos de los fármacos , Regulación de la Expresión Génica/efectos de los fármacos , Genes Codificadores de los Receptores de Linfocitos T , Linfoma de Células B/genética , Linfoma de Células B/inmunología , Linfoma de Células B/metabolismo , Linfoma de Células B/terapia , Ratones , Ratones Endogámicos BALB C , Ratones Transgénicos , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/metabolismo , Receptores de Esteroides/genética , Receptores de Esteroides/metabolismo , Receptores de Hormona Tiroidea/genética , Receptores de Hormona Tiroidea/metabolismo , Resultado del Tratamiento
10.
J Immunol ; 182(7): 4192-9, 2009 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-19299717

RESUMEN

IL-17- and IFN-gamma-secreting T cells play an important role in autoimmune responses in multiple sclerosis and the model system experimental autoimmune encephalomyelitis (EAE). Dendritic cells (DCs) in the periphery and microglia in the CNS are responsible for cytokine polarization and expansion of this T cell subset. Our results indicate that in vivo administration of a signal transduction inhibitor that targets DCs to mice with EAE led to a decrease in CNS infiltration of pathogenic Ag-specific T cells. Since this approach does not target T cells directly, we assessed the effects on the APCs that are involved in generating the T cell responses. Since in EAE and multiple sclerosis, both microglia and peripheral DCs are likely to contribute to disease, we utilized a bone marrow chimera system to distinguish between these two populations. These studies show that peripheral DCs are the primary target but that microglia are also modestly affected by CEP-701, as numbers and activation states of the cells in the CNS are decreased after therapy. Our results also showed a decrease in secretion of TNF-alpha, IL-6, and IL-23 by DCs as well as a decrease in expression of costimulatory molecules. We further determined that levels of phospho-Stat1, Stat3, Stat5, and NF-kappaB, which are signaling molecules that have been implicated in these pathways, were decreased. Thus, use of this class of signal transduction inhibitors may represent a novel method to treat autoimmunity by dampening the autoreactive polarizing condition driven by DCs.


Asunto(s)
Células Presentadoras de Antígenos/efectos de los fármacos , Carbazoles/farmacología , Encefalomielitis Autoinmune Experimental/tratamiento farmacológico , Inhibidores Enzimáticos/farmacología , Transducción de Señal/efectos de los fármacos , Células TH1/efectos de los fármacos , Animales , Células Presentadoras de Antígenos/inmunología , Western Blotting , Encéfalo/inmunología , Encéfalo/metabolismo , Encéfalo/patología , Quimiotaxis de Leucocito/efectos de los fármacos , Citocinas/biosíntesis , Citocinas/inmunología , Encefalomielitis Autoinmune Experimental/inmunología , Encefalomielitis Autoinmune Experimental/patología , Femenino , Furanos , Interleucina-17/inmunología , Interleucina-17/metabolismo , Ratones , Microglía/efectos de los fármacos , Microglía/inmunología , Microglía/metabolismo , Transducción de Señal/inmunología , Subgrupos de Linfocitos T/efectos de los fármacos , Subgrupos de Linfocitos T/inmunología , Células TH1/inmunología , Tirosina Quinasa 3 Similar a fms/deficiencia , Tirosina Quinasa 3 Similar a fms/inmunología
11.
Nat Med ; 9(7): 952-8, 2003 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-12778137

RESUMEN

A major focus of cancer immunotherapy is to develop strategies to induce T-cell responses through presentation of tumor antigens by dendritic cells (DCs). Current vaccines are limited in their ability to efficiently transfer antigens to DCs in vivo. Ex vivo-generated DCs can be efficiently loaded with antigen but after reinjection, few DCs traffic to secondary lymphoid organs, the critical sites for antigen presentation. To enhance efficiency and durability of antigen presentation by DCs, we transduced hematopoietic stem-progenitor cells (HSCs) with a model tumor antigen and then transplanted the gene-modified cells into irradiated recipient mice, which resulted in efficient expression of the transgene in a large proportion of donor derived DCs in lymphoid organs. The combination of bone marrow transplantation (BMT) using transduced HSCs, systemic agents that generate and activate DCs, and mature T-cell infusion resulted in substantial expansion and activation of antigen-specific T cells. This tripartite strategy provided potent antigen-specific immunotherapy for an aggressive established tumor.


Asunto(s)
Trasplante de Médula Ósea/métodos , Trasplante de Células Madre Hematopoyéticas/métodos , Inmunoterapia Adoptiva/métodos , Linfoma de Células B/inmunología , Linfoma de Células B/terapia , Neoplasias Experimentales/inmunología , Neoplasias Experimentales/terapia , Animales , Antígenos/metabolismo , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD4-Positivos/metabolismo , Antígenos CD40/genética , Antígenos CD40/inmunología , Antígenos CD40/metabolismo , Linfocitos T CD8-positivos/fisiología , Células Cultivadas , Células Dendríticas/fisiología , Hemaglutinación/genética , Células Madre Hematopoyéticas/fisiología , Interferón gamma/metabolismo , Lentivirus/genética , Linfoma de Células B/genética , Ratones , Ratones Endogámicos BALB C , Neoplasias Experimentales/genética , Bazo/citología , Tasa de Supervivencia , Linfocitos T/inmunología , Linfocitos T/fisiología , Transducción Genética , Trasplante Autólogo
12.
Nat Commun ; 12(1): 652, 2021 01 28.
Artículo en Inglés | MEDLINE | ID: mdl-33510160

RESUMEN

Injury and loss of oligodendrocytes can cause demyelinating diseases such as multiple sclerosis. To improve our understanding of human oligodendrocyte development, which could facilitate development of remyelination-based treatment strategies, here we describe time-course single-cell-transcriptomic analysis of developing human stem cell-derived oligodendrocyte-lineage-cells (hOLLCs). The study includes hOLLCs derived from both genome engineered embryonic stem cell (ESC) reporter cells containing an Identification-and-Purification tag driven by the endogenous PDGFRα promoter and from unmodified induced pluripotent (iPS) cells. Our analysis uncovers substantial transcriptional heterogeneity of PDGFRα-lineage hOLLCs. We discover sub-populations of human oligodendrocyte progenitor cells (hOPCs) including a potential cytokine-responsive hOPC subset, and identify candidate regulatory genes/networks that define the identity of these sub-populations. Pseudotime trajectory analysis defines developmental pathways of oligodendrocytes vs astrocytes from PDGFRα-expressing hOPCs and predicts differentially expressed genes between the two lineages. In addition, pathway enrichment analysis followed by pharmacological intervention of these pathways confirm that mTOR and cholesterol biosynthesis signaling pathways are involved in maturation of oligodendrocytes from hOPCs.


Asunto(s)
Heterogeneidad Genética , Variación Genética , Células Madre Pluripotentes Inducidas/metabolismo , Células Precursoras de Oligodendrocitos/metabolismo , Análisis de la Célula Individual/métodos , Transcriptoma/genética , Astrocitos/citología , Astrocitos/metabolismo , Diferenciación Celular/genética , Línea Celular , Linaje de la Célula/genética , Colesterol/biosíntesis , Células Madre Embrionarias/citología , Células Madre Embrionarias/metabolismo , Redes Reguladoras de Genes/genética , Humanos , Células Madre Pluripotentes Inducidas/citología , Células Precursoras de Oligodendrocitos/citología , Receptor alfa de Factor de Crecimiento Derivado de Plaquetas/genética , Receptor alfa de Factor de Crecimiento Derivado de Plaquetas/metabolismo , Transducción de Señal/genética , Serina-Treonina Quinasas TOR/genética , Serina-Treonina Quinasas TOR/metabolismo
13.
Exp Mol Pathol ; 89(2): 109-16, 2010 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-20566414

RESUMEN

Inflammatory cell infiltration and resident microglial activation within the central nervous system (CNS) are pathological events in multiple sclerosis (MS) and experimental autoimmune encephalomyelitis (EAE). While MS therapies target the peripheral immune system, no treatment is currently known to also modulate microglia. FMS-like tyrosine-3 (FLT-3) is expressed on hematopoietic and dendritic cells. We reported that FLT-3 inhibition ameliorates early actively induced EAE by predominantly modulating dendritic cell function as compared to microglia. We demonstrate in this report that FLT-3 is expressed in perivascular cuffs, brain parenchyma and in non-lesioned gray and white matter within MS brain but not in these regions within control brain. Furthermore, we demonstrate that FLT-3 is expressed on two populations of cells within MS brain; one which expresses the dendritic cell marker CD209, and the other which does not, suggesting that FLT-3 within MS brain is expressed on infiltrating dendritic cells and a non-dendritic cell such as microglia. Additionally, we report that FLT-3 inhibition in murine microglia blocks, in a dose-dependent manner, IFN-γ-induced expression of MHC class II and CD86, and LPS-induced secretion of IL-6. These data suggest that FLT-3 is involved in microglial cells' capacity to respond to environmental cues to function as antigen presenting cells and mediate CNS inflammation. Furthermore these data suggest that FLT-3 may be a therapeutic target on microglia to mitigate CNS inflammation.


Asunto(s)
Microglía/metabolismo , Esclerosis Múltiple/metabolismo , Tirosina Quinasa 3 Similar a fms/metabolismo , Animales , Encéfalo/inmunología , Encéfalo/patología , Encéfalo/fisiología , Células Dendríticas/metabolismo , Células Dendríticas/patología , Células Dendríticas/fisiología , Encefalomielitis Autoinmune Experimental/metabolismo , Encefalomielitis Autoinmune Experimental/patología , Femenino , Masculino , Ratones , Ratones Endogámicos C57BL , Microglía/citología , Microglía/fisiología , Esclerosis Múltiple/inmunología , Esclerosis Múltiple/patología
14.
J Neuroimmunol ; 339: 577115, 2020 02 15.
Artículo en Inglés | MEDLINE | ID: mdl-31778849

RESUMEN

Multiple sclerosis (MS) is an autoimmune disease that remains in need of effective therapies. Plant-derived medicines have appealing properties for the treatment of autoimmune diseases. MYMD-1 is a synthetic plant alkaloid that has been shown to ameliorate the course of autoimmune thyroiditis. The goal of the present study was to determine whether MYMD-1 would produce similar beneficial effects in a mouse model of MS, experimental autoimmune encephalomyelitis (EAE) induced by immunization with myelin oligodendrocyte glycoprotein. MYMD-1 improved the course of EAE and suppressed activation of effector T cells without causing global immunosuppression or toxicity. These results suggest that MYMD-1 may be of interest for evaluating for the treatment of autoimmune diseases.


Asunto(s)
Alcaloides/uso terapéutico , Encefalomielitis Autoinmune Experimental/tratamiento farmacológico , Encefalomielitis Autoinmune Experimental/inmunología , Alcaloides/síntesis química , Alcaloides/farmacología , Animales , Células Cultivadas , Relación Dosis-Respuesta a Droga , Encefalomielitis Autoinmune Experimental/metabolismo , Femenino , Ratones , Ratones Endogámicos C57BL , Linfocitos T/efectos de los fármacos , Linfocitos T/inmunología , Linfocitos T/metabolismo
15.
PLoS One ; 15(6): e0233980, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32511247

RESUMEN

Multiple sclerosis (MS) is an inflammatory and demyelinating disease of the central nervous system (CNS) that results in variable severities of neurodegeneration. The understanding of MS has been limited by the inaccessibility of the affected cells and the lengthy timeframe of disease development. However, recent advances in stem cell technology have facilitated the bypassing of some of these challenges. Towards gaining a greater understanding of the innate potential of stem cells from people with varying degrees of disability, we generated induced pluripotent stem cells (iPSCs) from peripheral blood mononuclear cells derived from stable and progressive MS patients, and then further differentiated them into oligodendrocyte (OL) lineage cells. We analyzed differentiation under both homeostatic and inflammatory conditions via sustained exposure to low-dose interferon gamma (IFNγ), a prominent cytokine in MS. We found that all iPSC lines differentiated into mature myelinating OLs, but chronic exposure to IFNγ dramatically inhibited differentiation in both MS groups, particularly if exposure was initiated during the pre-progenitor stage. Low-dose IFNγ was not toxic but led to an early upregulation of interferon response genes in OPCs followed by an apparent redirection in lineage commitment from OL to a neuron-like phenotype in a significant portion of the treated cells. Our results reveal that a chronic low-grade inflammatory environment may have profound effects on the efficacy of regenerative therapies.


Asunto(s)
Células Madre Pluripotentes Inducidas/citología , Esclerosis Múltiple Crónica Progresiva/patología , Oligodendroglía/citología , Diferenciación Celular/efectos de los fármacos , Células Cultivadas , Homeostasis , Humanos , Células Madre Pluripotentes Inducidas/efectos de los fármacos , Células Madre Pluripotentes Inducidas/fisiología , Inflamación/patología , Interferón gamma/farmacología , Leucocitos Mononucleares/citología , Regeneración
16.
J Leukoc Biol ; 105(5): 829-841, 2019 05.
Artículo en Inglés | MEDLINE | ID: mdl-30762897

RESUMEN

Myeloid-derived suppressor cells (MDSCs) are a diverse group of cells that are recognized for their remarkable suppressive effects on pro-inflammatory T cells. The pleiotropic nature of these cells, however, has been demonstrated by their differential effects on immune responses in different settings. Our and others' work has demonstrated suppressive effects of these cells. We previously demonstrated that these cells were mobilized to the lungs during experimental autoimmune encephalomyelitis (EAE), which is a murine model of multiple sclerosis, and potently inhibited CD8+ T cell responses against influenza infection. Interestingly, they appeared to have a lesser effect on CD4+ T cells, and in fact, others have demonstrated that spleen-derived MDSCs could actually promote Th17 differentiation. We sought to determine the role of lung-derived MDSCs on EAE pathogenesis, as excursion through the lungs by pathologic CNS-Ag targeted T cells was shown to be critical for EAE induction. Our results indicate a robust accumulation of granulocytic MDSCs in the lungs of mice during EAE, which could promote Th17 polarization, and which coincided with the trafficking of autoimmune-targeted T cells through the lungs. These studies underscore the pleiotropic effect of MDSCs on T cells and their potential pro-inflammatory phenotypes in neuro-inflammatory disease. Understanding both the intrinsic multifunctional nature of these cells and the ability to influence organ-specific targets such as the CNS from remote organs such as lungs will help to elucidate both mechanisms of disease and possible new therapeutic approaches.


Asunto(s)
Sistema Nervioso Central/inmunología , Encefalomielitis Autoinmune Experimental/inmunología , Pulmón/inmunología , Células Supresoras de Origen Mieloide/inmunología , Células Th17/inmunología , Animales , Autoinmunidad , Comunicación Celular/inmunología , Linaje de la Célula/genética , Linaje de la Célula/inmunología , Movimiento Celular , Sistema Nervioso Central/patología , Técnicas de Cocultivo , Modelos Animales de Enfermedad , Encefalomielitis Autoinmune Experimental/inducido químicamente , Encefalomielitis Autoinmune Experimental/genética , Encefalomielitis Autoinmune Experimental/patología , Femenino , Expresión Génica , Humanos , Interleucina-17/genética , Interleucina-17/inmunología , Pulmón/patología , Ratones Endogámicos C57BL , Esclerosis Múltiple/genética , Esclerosis Múltiple/inmunología , Esclerosis Múltiple/patología , Glicoproteína Mielina-Oligodendrócito/administración & dosificación , Células Supresoras de Origen Mieloide/efectos de los fármacos , Células Supresoras de Origen Mieloide/patología , Fragmentos de Péptidos/administración & dosificación , Toxina del Pertussis/administración & dosificación , Transducción de Señal/inmunología , Células Th17/efectos de los fármacos , Células Th17/patología , Factor de Crecimiento Transformador beta/farmacología
17.
PLoS One ; 14(9): e0221747, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31490950

RESUMEN

Multiple sclerosis (MS) is characterized by demyelinated lesions in the central nervous system. Destruction of myelin and secondary damage to axons and neurons leads to significant disability, particularly in people with progressive MS. Accumulating evidence suggests that the potential for myelin repair exists in MS, although for unclear reasons this process fails. The cells responsible for producing myelin, the oligodendrocytes, and their progenitors, oligodendrocyte precursor cells (OPCs), have been identified at the site of lesions, even in adults. Their presence suggests the possibility that endogenous remyelination without transplantation of donor stem cells may be a mechanism for myelin repair in MS. Strategies to develop novel therapies have focused on induction of signaling pathways that stimulate OPCs to mature into myelin-producing oligodendrocytes that could then possibly remyelinate lesions. We have been investigating pharmacological approaches to enhance OPC differentiation, and have identified that the combination of two agents, triiodothyronine (T3) and quetiapine, leads to an additive effect on OPC differentiation and consequent myelin production via both overlapping and distinct signaling pathways. While the ultimate production of myelin requires cholesterol biosynthesis, we identified that quetiapine enhances gene expression in this pathway more potently than T3. Two blockers of cholesterol production, betulin and simvastatin, reduced OPC differentiation into myelin producing oligodendrocytes. Elucidating the nature of agents that lead to complementary and additive effects on oligodendrocyte differentiation and myelin production may pave the way for more efficient induction of remyelination in people with MS.


Asunto(s)
Diferenciación Celular/efectos de los fármacos , Colesterol/biosíntesis , Células Precursoras de Oligodendrocitos/citología , Células Precursoras de Oligodendrocitos/efectos de los fármacos , Fumarato de Quetiapina/farmacología , Triyodotironina/farmacología , Animales , Sinergismo Farmacológico , Regulación de la Expresión Génica/efectos de los fármacos , Proteína Básica de Mielina/metabolismo , Células Precursoras de Oligodendrocitos/metabolismo , Ratas , Ratas Sprague-Dawley , Transducción de Señal/efectos de los fármacos , Simvastatina/farmacología , Triterpenos/farmacología
18.
Biomaterials ; 190-191: 24-37, 2019 01.
Artículo en Inglés | MEDLINE | ID: mdl-30391800

RESUMEN

Microvessels of the blood-brain barrier (BBB) regulate transport into the brain. The highly specialized brain microvascular endothelial cells, a major component of the BBB, express tight junctions and efflux transporters which regulate paracellular and transcellular permeability. However, most existing models of BBB microvessels fail to exhibit physiological barrier function. Here, using (iPSC)-derived human brain microvascular endothelial cells (dhBMECs) within templated type I collagen channels we mimic the cylindrical geometry, cell-extracellular matrix interactions, and shear flow typical of human brain post-capillary venules. We characterize the structure and barrier function in comparison to non-brain-specific microvessels, and show that dhBMEC microvessels recapitulate physiologically low solute permeability and quiescent endothelial cell behavior. Transcellular permeability is increased two-fold using a clinically relevant dose of a p-glycoprotein inhibitor tariquidar, while paracellular permeability is increased using a bolus dose of hyperosmolar agent mannitol. Lastly, we show that our human BBB microvessels are responsive to inflammatory cytokines via upregulation of surface adhesion molecules and increased leukocyte adhesion, but no changes in permeability. Human iPSC-derived blood-brain barrier microvessels support quantitative analysis of barrier function and endothelial cell dynamics in quiescence and in response to biologically- and clinically-relevant perturbations.


Asunto(s)
Barrera Hematoencefálica/citología , Células Endoteliales/citología , Células Madre Pluripotentes Inducidas/citología , Microvasos/citología , Barrera Hematoencefálica/metabolismo , Permeabilidad Capilar , Diferenciación Celular , Línea Celular , Células Endoteliales/metabolismo , Diseño de Equipo , Humanos , Células Madre Pluripotentes Inducidas/metabolismo , Microvasos/metabolismo , Ingeniería de Tejidos/instrumentación , Ingeniería de Tejidos/métodos
19.
J Neuroimmunol ; 316: 7-16, 2018 03 15.
Artículo en Inglés | MEDLINE | ID: mdl-29274729

RESUMEN

Pro-inflammatory T cells are critical to the pathogenesis of multiple sclerosis (MS). We investigated the potential for the anti-proliferative, pro-apoptotic drug gemcitabine to affect development of MS-relevant effector TH1, TH17, and Treg cells. Gemcitabine directly suppressed proliferation, activation, and induced apoptosis of all effector subsets in subtype and dose-dependent fashion. This drug also prevented development of disease in the MS model experimental autoimmune encephalomyelitis (EAE) and significantly reduced the abundance of TH1 and TH17 cells. Our results indicate that pathogenic CD4+ T cells may be viable targets by gemcitabine for therapeutic benefit in MS.


Asunto(s)
Linfocitos T CD4-Positivos/efectos de los fármacos , Desoxicitidina/análogos & derivados , Encefalomielitis Autoinmune Experimental/inmunología , Inmunosupresores/farmacología , Activación de Linfocitos/efectos de los fármacos , Animales , Linfocitos T CD4-Positivos/inmunología , Desoxicitidina/farmacología , Femenino , Activación de Linfocitos/inmunología , Ratones , Ratones Endogámicos C57BL , Gemcitabina
20.
J Exp Med ; 214(2): 297-307, 2017 02.
Artículo en Inglés | MEDLINE | ID: mdl-28057805

RESUMEN

The discovery that central nervous system (CNS)-targeted autoreactive T cells required a process of licensing in the lung revealed an unexpected relationship between these organs. The clinical and immunological significance of this finding is bidirectional in that it showed not only a mechanism by which T cells become pathogenic before entering the CNS, but also the potential for this process to influence lung immunity as well. Epidemiological studies have shown that people with multiple sclerosis (MS) suffer from increased morbidity and mortality from infectious diseases, independent of immunosuppressive therapies. Respiratory infections account for a large percentage of deaths of people with MS. In this study, to investigate the mechanisms responsible for this enhanced susceptibility, we established a comorbid model system in which mice with experimental autoimmune encephalomyelitis (EAE) were administered a sublethal dose of influenza. Whereas mice with either EAE alone or influenza alone survived, 70% of comorbid mice died as a result of uncontrolled viral replication. Immunological analyses revealed that the induction of EAE led to a surprising alteration of the lung milieu, converting an effective stimulatory influenza-reactive environment into a suppressive one. These results provide mechanistic information that may help to explain the unexpected immunological interactions.


Asunto(s)
Autoinmunidad , Encéfalo/inmunología , Infecciones por Orthomyxoviridae/mortalidad , Animales , Movimiento Celular , Comorbilidad , Encefalomielitis Autoinmune Experimental/mortalidad , Femenino , Pulmón/virología , Activación de Linfocitos , Ratones , Ratones Endogámicos C57BL , Células Supresoras de Origen Mieloide/fisiología , Óxido Nítrico Sintasa de Tipo II/fisiología , Replicación Viral
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA