Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Cell Immunol ; 384: 104674, 2023 02.
Artículo en Inglés | MEDLINE | ID: mdl-36706656

RESUMEN

Second mitochondria-derived activator of caspases (SMAC) mimetics are small molecule drugs that mimic the activity of the endogenous SMAC protein. SMAC and SMAC mimetics antagonize inhibitors of apoptosis proteins (IAPs), thereby sensitizing cells to apoptosis. As such, SMAC mimetics are being tested in numerous clinical trials for cancer. In addition to their direct anti-cancer effect, it has been suggested that SMAC mimetics may activate T cells, thereby promoting anti-tumor immunity. Here, we tested the effect of three clinically relevant SMAC mimetics on activation of primary human T cells. As previously reported, SMAC mimetics killed tumor cells and activated non-canonical NF-κB in T cells at clinically relevant doses. Surprisingly, none of the SMAC mimetics augmented T cell responses. Rather, SMAC mimetics impaired T cell proliferation and decreased the proportion of IFNγ/TNFα double-producing T cells. These results question the assumption that SMAC mimetics are likely to boost anti-tumor immunity in cancer patients.


Asunto(s)
Caspasas , Neoplasias , Humanos , Caspasas/farmacología , Caspasas/uso terapéutico , Proteínas Inhibidoras de la Apoptosis/metabolismo , Proteínas Inhibidoras de la Apoptosis/farmacología , Proteínas Inhibidoras de la Apoptosis/uso terapéutico , Citocinas , Neoplasias/tratamiento farmacológico , Apoptosis , Mitocondrias/metabolismo , Proliferación Celular , Proteínas Mitocondriales/metabolismo , Línea Celular Tumoral
2.
PLoS Pathog ; 19(1): e1010351, 2023 01.
Artículo en Inglés | MEDLINE | ID: mdl-36689486

RESUMEN

CMV, a ubiquitous herpesvirus, elicits an extraordinarily large T cell response that is sustained or increases over time, a phenomenon termed 'memory inflation.' Remarkably, even latent, non-productive infection can drive memory inflation. Despite intense research on this phenomenon, the infected cell type(s) involved are unknown. To identify the responsible cell type(s), we designed a Cre-lox murine CMV (MCMV) system, where a spread-deficient (ΔgL) virus expresses recombinant SIINFEKL only in Cre+ host cells. We found that latent infection of endothelial cells (ECs), but not dendritic cells (DCs) or hepatocytes, was sufficient to drive CD8 T cell memory inflation. Infection of Lyve-1-Cre and Prox1-CreERT2 mice revealed that amongst EC subsets, infection of lymphatic ECs was sufficient. Genetic ablation of ß2m on lymphatic ECs did not prevent inflation, suggesting another unidentified cell type can also present antigen to CD8 T cells during latency. This novel system definitively shows that antigen presentation by lymphatic ECs drives robust CD8 T cell memory inflation.


Asunto(s)
Infecciones por Citomegalovirus , Infección Latente , Muromegalovirus , Animales , Ratones , Células Endoteliales , Linfocitos T CD8-positivos , Antígenos , Memoria Inmunológica
3.
Nature ; 606(7915): 791-796, 2022 06.
Artículo en Inglés | MEDLINE | ID: mdl-35322234

RESUMEN

Immune checkpoint blockade has revolutionized the field of oncology, inducing durable anti-tumour immunity in solid tumours. In patients with advanced prostate cancer, immunotherapy treatments have largely failed1-5. Androgen deprivation therapy is classically administered in these patients to inhibit tumour cell growth, and we postulated that this therapy also affects tumour-associated T cells. Here we demonstrate that androgen receptor (AR) blockade sensitizes tumour-bearing hosts to effective checkpoint blockade by directly enhancing CD8 T cell function. Inhibition of AR activity in CD8 T cells prevented T cell exhaustion and improved responsiveness to PD-1 targeted therapy via increased IFNγ expression. AR bound directly to Ifng and eviction of AR with a small molecule significantly increased cytokine production in CD8 T cells. Together, our findings establish that T cell intrinsic AR activity represses IFNγ expression and represents a novel mechanism of immunotherapy resistance.


Asunto(s)
Linfocitos T CD8-positivos , Inmunoterapia , Neoplasias de la Próstata , Receptores Androgénicos , Antagonistas de Andrógenos/farmacología , Antagonistas de Andrógenos/uso terapéutico , Linfocitos T CD8-positivos/inmunología , Humanos , Inhibidores de Puntos de Control Inmunológico/farmacología , Inhibidores de Puntos de Control Inmunológico/uso terapéutico , Interferón gamma , Masculino , Neoplasias de la Próstata/tratamiento farmacológico , Neoplasias de la Próstata/inmunología , Neoplasias de la Próstata/metabolismo , Receptores Androgénicos/metabolismo , Insuficiencia del Tratamiento
4.
J Immunol ; 203(7): 2011-2019, 2019 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-31434709

RESUMEN

OX40 is a costimulatory molecule from the TNFR family. In mice, it is expressed on Foxp3+ regulatory T cells (Tregs) constitutively and on conventional CD4 (Tconv) and CD8 T cells after Ag encounter. OX40 agonists are in clinical development to enhance antitumor immune responses, and one proposed mechanism of action is loss of Treg suppressive function. Studies have postulated that agonist OX40 therapy can impair Treg suppressive function. Using tools developed since the initial studies were published, we evaluated a direct effect of OX40 agonism on Treg function. We conclude that OX40 agonist Abs do not intrinsically impair Treg function but rather enhance Tconv cell IL-2 production, increasing Treg and Tconv cell proliferation. OX40-stimulated Tregs retain suppressive function, but also gain IFN-γ, TNF-α, and granzyme B expression. These data help resolve mechanistic questions regarding OX40 agonist immunotherapy and thus are relevant to developing combination therapies that target distinct T cell functions.


Asunto(s)
Anticuerpos Antineoplásicos/farmacología , Linfocitos T CD8-positivos/inmunología , Inmunoterapia , Proteínas de Neoplasias , Neoplasias , Receptores OX40 , Linfocitos T Reguladores/inmunología , Animales , Anticuerpos Antineoplásicos/inmunología , Linfocitos T CD8-positivos/patología , Línea Celular Tumoral , Citocinas/genética , Citocinas/inmunología , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Regulación Neoplásica de la Expresión Génica/inmunología , Ratones , Ratones Transgénicos , Proteínas de Neoplasias/antagonistas & inhibidores , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/inmunología , Neoplasias/genética , Neoplasias/inmunología , Neoplasias/patología , Neoplasias/terapia , Receptores OX40/antagonistas & inhibidores , Receptores OX40/genética , Receptores OX40/inmunología , Linfocitos T Reguladores/patología
5.
Sci Rep ; 7(1): 14779, 2017 11 07.
Artículo en Inglés | MEDLINE | ID: mdl-29116141

RESUMEN

CD4+Foxp3+ regulatory T cells (Tregs) are indispensable negative regulators of immune responses. To understand Treg biology in health and disease, it is critical to elucidate factors that affect Treg homeostasis and suppressive function. Tregs express several costimulatory TNF receptor family members that activate non-canonical NF-κB via accumulation of NF-κB inducing kinase (NIK). We previously showed that constitutive NIK expression in all T cells causes fatal multi-organ autoimmunity associated with hyperactive conventional T cell responses and poor Treg-mediated suppression. Here, we show that constitutive NIK expression that is restricted to Tregs via a Cre-inducible transgene causes an autoimmune syndrome. We found that constitutive NIK expression decreased expression of numerous Treg signature genes and microRNAs involved in Treg homeostasis and suppressive phenotype. NIK transgenic Tregs competed poorly with WT Tregs in vivo and produced pro-inflammatory cytokines upon stimulation. Lineage tracing experiments revealed accumulation of ex-Foxp3+ T cells in mice expressing NIK constitutively in Tregs, and these former Tregs produced copious IFNγ and IL-2. Our data indicate that under inflammatory conditions in which NIK is activated, Tregs may lose suppressive function and may actively contribute to inflammation.


Asunto(s)
Citocinas/metabolismo , Mediadores de Inflamación/metabolismo , FN-kappa B/metabolismo , Linfocitos T Reguladores/metabolismo , Animales , Ratones , Ratones Transgénicos , Linfocitos T Reguladores/inmunología
6.
J Exp Med ; 214(7): 1889-1899, 2017 Jul 03.
Artículo en Inglés | MEDLINE | ID: mdl-28566275

RESUMEN

Cytomegalovirus (CMV)-based vaccines have shown remarkable efficacy in the rhesus macaque model of acquired immune deficiency syndrome, enabling 50% of vaccinated monkeys to clear a subsequent virulent simian immunodeficiency virus challenge. The protective vaccine elicited unconventional CD8 T cell responses that were entirely restricted by MHC II or the nonclassical MHC I molecule, MHC-E. These unconventional responses were only elicited by a fibroblast-adapted rhesus CMV vector with limited tissue tropism; a repaired vector with normal tropism elicited conventional responses. Testing whether these unusual protective CD8 T responses could be elicited in humans requires vaccinating human subjects with a fibroblast-adapted mutant of human CMV (HCMV). In this study, we describe the CD8 T cell responses of human subjects vaccinated with two fibroblast-adapted HCMV vaccines. Most responses were identified as conventional classically MHC I restricted, and we found no evidence for MHC II or HLA-E restriction. These results indicate that fibroblast adaptation alone is unlikely to explain the unconventional responses observed in macaques.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Infecciones por Citomegalovirus/inmunología , Vacunas contra Citomegalovirus/inmunología , Citomegalovirus/inmunología , Fibroblastos/inmunología , Secuencia de Aminoácidos , Línea Celular , Línea Celular Tumoral , Células Cultivadas , Citomegalovirus/fisiología , Infecciones por Citomegalovirus/prevención & control , Infecciones por Citomegalovirus/virología , Vacunas contra Citomegalovirus/administración & dosificación , Vacunas contra Citomegalovirus/genética , Epítopos/inmunología , Fibroblastos/virología , Citometría de Flujo , Antígenos de Histocompatibilidad Clase I/inmunología , Interacciones Huésped-Patógeno/efectos de los fármacos , Interacciones Huésped-Patógeno/inmunología , Humanos , Células K562 , Leucocitos Mononucleares/inmunología , Leucocitos Mononucleares/virología , Masculino , Microscopía Fluorescente , Mutación , Vacunación
7.
J Infect Dis ; 214(9): 1341-1348, 2016 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-27521362

RESUMEN

BACKGROUND: Human cytomegalovirus (HCMV) infection causes disease in newborns and transplant recipients. A HCMV vaccine (Towne) protects transplant recipients. METHODS: The genomes of Towne and the nonattenuated Toledo strain were recombined, yielding 4 Towne/Toledo chimera vaccines. Each of 36 HCMV-seronegative men received 1 subcutaneous dose of 10, 100, or 1000 plaque-forming units (PFU) in cohorts of 3. Safety and immunogenicity were evaluated over 12 weeks after immunization and for 52 weeks for those who seroconverted. RESULTS: There were no serious local or systemic reactions. No subject had HCMV in urine or saliva. For chimera 3, none of 9 subjects seroconverted. For chimera 1, 1 of 9 seroconverted (the seroconverter received 100 PFU). For chimera 2, 3 subjects seroconverted (1 received 100 PFU, and 2 received 1000 PFU). For chimera 4, 7 subjects seroconverted (1 received 10 PFU, 3 received 100 PFU, and 3 received 1000 PFU). All 11 seroconverters developed low but detectable levels of neutralizing activity. CD4+ T-cell responses were detectable in 1 subject (who received 100 PFU of chimera 4). Seven subjects receiving chimera 2 or 4 had detectable CD8+ T-cell responses to IE1; 3 responded to 1-2 additional antigens. CONCLUSIONS: The Towne/Toledo chimera vaccine candidates were well tolerated and were not excreted. Additional human trials of chimeras 2 and 4 are appropriate. CLINICAL TRIALS REGISTRATION: NCT01195571.


Asunto(s)
Quimera/inmunología , Infecciones por Citomegalovirus/inmunología , Vacunas contra Citomegalovirus/inmunología , Citomegalovirus/inmunología , Vacunas Sintéticas/inmunología , Adulto , Anticuerpos Antivirales/inmunología , Linfocitos T CD4-Positivos/inmunología , Humanos , Activación de Linfocitos/inmunología , Masculino , Persona de Mediana Edad , Vacunación/métodos , Vacunas Atenuadas/inmunología , Vacunas Virales/inmunología , Adulto Joven
8.
PLoS One ; 8(9): e76216, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-24073289

RESUMEN

NF-κB inducing kinase (NIK, MAP3K14) is a key signaling molecule in non-canonical NF-κB activation, and NIK deficient mice have been instrumental in deciphering the immunologic role of this pathway. Global ablation of NIK prevents lymph node development, impairs thymic stromal development, and drastically reduces B cells. Despite altered thymic selection, T cell numbers are near normal in NIK deficient mice. The exception is CD4(+) regulatory T cells (Tregs), which are reduced in the thymus and periphery. Defects in thymic stroma are known to contribute to impaired Treg generation, but whether NIK also plays a cell intrinsic role in Tregs is unknown. Here, we compared intact mice with single and mixed BM chimeric mice to assess the intrinsic role of NIK in Treg generation and maintenance. We found that while NIK expression in stromal cells suffices for normal thymic Treg development, NIK is required cell-intrinsically to maintain peripheral Tregs. In addition, we unexpectedly discovered a cell-intrinsic role for NIK in memory phenotype conventional T cells that is masked in intact mice, but revealed in BM chimeras. These results demonstrate a novel role for NIK in peripheral regulatory and memory phenotype T cell homeostasis.


Asunto(s)
Linfocitos T CD4-Positivos/inmunología , Factores de Transcripción Forkhead/metabolismo , Proteínas Serina-Treonina Quinasas/fisiología , Linfocitos T Reguladores/citología , Linfocitos T Reguladores/inmunología , Timo/citología , Animales , Médula Ósea/inmunología , Médula Ósea/metabolismo , Linfocitos T CD4-Positivos/citología , Linfocitos T CD4-Positivos/metabolismo , Diferenciación Celular , Proteínas de Unión al ADN/metabolismo , Memoria Inmunológica/inmunología , Ratones , Ratones Noqueados , Datos de Secuencia Molecular , FN-kappa B/metabolismo , Proteínas Nucleares/metabolismo , Fenotipo , Transducción de Señal , Linfocitos T Reguladores/metabolismo , Timo/inmunología , Timo/metabolismo , Ubiquitina-Proteína Ligasas , Quinasa de Factor Nuclear kappa B
9.
J Immunol ; 191(7): 3663-72, 2013 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-24006459

RESUMEN

NF-κB-inducing kinase [(NIK), MAP3K14] is an essential kinase linking a subset of TNFR family members to the noncanonical NF-κB pathway. To assess the cell-intrinsic role of NIK in murine T cell function, we generated mixed bone marrow chimeras using bone marrow from NIK knockout (KO) and wild-type (WT) donor mice and infected the chimeras with lymphocytic choriomeningitis virus (LCMV). The chimeras possess an apparently normal immune system, including a mixture of NIK KO and WT T cells, and the virus was cleared normally. Comparison of the NIK KO and WT CD4 and CD8 T cell responses at 8 d post infection revealed modest but significant differences in the acute response. In both CD4 and CD8 compartments, relatively fewer activated (CD44(hi)) NIK KO T cells were present, but within the CD44(hi) population, a comparable percentage of the activated cells produced IFN-γ in response to ex vivo stimulation with antigenic LCMV peptides, although IL-7R expression was reduced in the NIK KO CD8 T cells. Assessment of the LCMV-specific memory at 65 d post infection revealed many more LCMV-specific WT memory T cells than NIK KO memory T cells in both the CD4 and the CD8 compartments, although the small number of surviving NIK KO memory T cells responded to secondary challenge with virus. These results demonstrate a cell-intrinsic requirement for NIK in the generation and/or maintenance of memory T cells in response to acute viral infection.


Asunto(s)
Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD4-Positivos/metabolismo , Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/metabolismo , Memoria Inmunológica , Proteínas Serina-Treonina Quinasas/metabolismo , Animales , Infecciones por Arenaviridae/inmunología , Linfocitos T CD4-Positivos/virología , Linfocitos T CD8-positivos/virología , Quimera , Femenino , Memoria Inmunológica/genética , Virus de la Coriomeningitis Linfocítica/inmunología , Masculino , Ratones , Ratones Noqueados , Proteínas Serina-Treonina Quinasas/genética , Receptores de Interleucina-7/metabolismo , Quinasa de Factor Nuclear kappa B
10.
Eur J Immunol ; 43(7): 1818-27, 2013 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-23532986

RESUMEN

B cells are efficient APCs when they internalize antigen via BCR-mediated uptake. Adoptively transferred antigen-presenting B cells can induce T-cell tolerance to foreign and self antigens; however, it is unknown whether endogenous B cells presenting self-peptides interact with naïve T cells and contribute to peripheral T-cell self-tolerance. Moreover, the relative abilities of mature B-cell subsets to induce T-cell tolerance have not been examined. To address these questions, we created a new mouse model wherein a very small fraction of B cells expresses an antigen transgene that cannot be transferred to other APCs. We limited antigen expression to follicular, marginal zone, or B-1 B-cell subsets and found that small numbers of each subset interacted with naïve antigen-specific T cells. Although antigen expressed by B-1 B cells induced the most T-cell division, divided T cells subsequently disappeared from secondary lymphoid tissues. Independent of which B-cell subset presented antigen, the remaining T cells were rendered hypo-responsive, and this effect was not associated with Foxp3 expression. Our data show that physiologically relevant proportions of B cells can mediate peripheral T-cell tolerance, and suggest that the mechanisms of tolerance induction might differ among follicular, marginal zone, and B-1 B-cell subsets.


Asunto(s)
Subgrupos de Linfocitos B/inmunología , Linfocitos B/inmunología , Linfocitos T CD4-Positivos/inmunología , Tolerancia Inmunológica/inmunología , Traslado Adoptivo , Animales , Presentación de Antígeno/inmunología , Separación Celular , Citometría de Flujo , Activación de Linfocitos/inmunología , Ratones , Ratones Transgénicos , Bazo/citología , Bazo/inmunología
11.
Int Immunol ; 25(4): 259-69, 2013 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-23334952

RESUMEN

The duration of signaling through the MAP kinase (or ERK pathway) cascade has been implicated in thymic development, particularly positive and negative selection. In T cells, two isoforms of the MAP kinase kinase kinase Raf function to transmit signals from the T-cell receptor to ERK: C-Raf and B-Raf. In this study, we conditionally ablated B-Raf expression within thymocytes to assess the effects on ERK activation and thymocyte development. The complete loss of B-Raf is accompanied by a dramatic loss of ERK activation in both the double positive (DP) and single positive (SP) thymocytes, as well as peripheral splenocytes. There was a significant decrease in the cellularity of KO thymi, largely due to a loss of pre-selected DP cells, a decrease in DP cells undergoing positive selection, and a defect in SP maturation. B-Raf plays significant roles in survival of DP thymocytes and function of SP cells in the periphery. Surprisingly, we saw no effect of B-Raf deficiency on negative selection of autoreactive SP thymocytes, despite the greatly reduced ERK activation in these cells.


Asunto(s)
Proteínas Proto-Oncogénicas B-raf/metabolismo , Receptores de Antígenos de Linfocitos T/metabolismo , Subgrupos de Linfocitos T/inmunología , Linfocitos T/inmunología , Timo/inmunología , Animales , Antígenos CD4/metabolismo , Antígenos CD8/metabolismo , Diferenciación Celular/genética , Supervivencia Celular/genética , Células Cultivadas , Supresión Clonal/genética , Activación Enzimática/genética , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Sistema de Señalización de MAP Quinasas/genética , Sistema de Señalización de MAP Quinasas/inmunología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proteínas Proto-Oncogénicas B-raf/genética , Proteínas Proto-Oncogénicas B-raf/inmunología
12.
J Clin Invest ; 121(12): 4775-86, 2011 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-22045568

RESUMEN

NF-κB­inducing kinase (NIK) is an essential upstream kinase in noncanonical NF-κB signaling. NIK-dependent NF-κB activation downstream of several TNF receptor family members mediates lymphoid organ development and B cell homeostasis. Peripheral T cell populations are normal in the absence of NIK, but the role of NIK during in vivo T cell responses to antigen has been obscured by other developmental defects in NIK-deficient mice. Here, we have identified a T cell­intrinsic requirement for NIK in graft-versus-host disease (GVHD), wherein NIK-deficient mouse T cells transferred into MHC class II mismatched recipients failed to cause GVHD. Although NIK was not necessary for antigen receptor signaling, it was absolutely required for costimulation through the TNF receptor family member OX40 (also known as CD134). When we conditionally overexpressed NIK in T cells, mice suffered rapid and fatal autoimmunity characterized by hyperactive effector T cells and poorly suppressive Foxp3(+) Tregs. Together, these data illuminate a critical T cell­intrinsic role for NIK during immune responses and suggest that its tight regulation is critical for avoiding autoimmunity.


Asunto(s)
Enfermedades Autoinmunes/enzimología , Enfermedad Injerto contra Huésped/enzimología , Proteínas Serina-Treonina Quinasas/fisiología , Linfocitos T/enzimología , Traslado Adoptivo , Animales , Enfermedades Autoinmunes/genética , Enfermedades Autoinmunes/inmunología , Autoinmunidad/inmunología , Linfocitos T CD4-Positivos/inmunología , Femenino , Enfermedad Injerto contra Huésped/inmunología , Isoantígenos/inmunología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , FN-kappa B/fisiología , Fosforilación , Procesamiento Proteico-Postraduccional , Proteínas Serina-Treonina Quinasas/deficiencia , Proteínas Serina-Treonina Quinasas/genética , Quimera por Radiación , Receptores de Antígenos de Linfocitos T/inmunología , Receptores OX40/fisiología , Transducción de Señal/inmunología , Linfocitos T/inmunología , Linfocitos T Reguladores/inmunología , Linfocitos T Reguladores/patología , Quinasa de Factor Nuclear kappa B
13.
J Immunol ; 178(12): 7694-702, 2007 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-17548606

RESUMEN

Ag-specific CD4 T cells transferred into unirradiated Ag-bearing recipients proliferate, but survival and accumulation of proliferating cells is not extensive and the donor cells do not acquire effector functions. We previously showed that a single costimulatory signal delivered by an agonist Ab to OX40 (CD134) promotes accumulation of proliferating cells and promotes differentiation to effector CD4 T cells capable of secreting IFN-gamma. In this study, we determined whether OX40 costimulation requires supporting costimulatory or differentiation signals to drive acquisition of effector T cell function. We report that OX40 engagement drives effector T cell differentiation in the absence of CD28 and CD40 signals. Two important regulators of Th1 differentiation, IL-12R and T-bet, also are not required for acquisition of effector function in CD4 T cells responsive to OX40 stimulation. Finally, we show that CD25-deficient CD4 T cells produce little IFN-gamma in the presence of OX40 costimulation compared with wild type, suggesting that IL-2R signaling is required for efficient OX40-mediated differentiation to IFN-gamma secretion.


Asunto(s)
Linfocitos T CD4-Positivos/inmunología , Activación de Linfocitos , Receptores de Interleucina-2/fisiología , Receptores OX40/fisiología , Animales , Células Presentadoras de Antígenos/inmunología , Antígenos CD28/genética , Antígenos CD28/fisiología , Antígenos CD40/genética , Antígenos CD40/fisiología , Diferenciación Celular , Interferón gamma/metabolismo , Subunidad beta 2 del Receptor de Interleucina-12/genética , Subunidad beta 2 del Receptor de Interleucina-12/fisiología , Ratones , Ratones Mutantes , Receptores de Interleucina-2/genética , Receptores OX40/agonistas , Transducción de Señal , Proteínas de Dominio T Box/genética , Proteínas de Dominio T Box/fisiología
14.
J Neuroimmunol ; 156(1-2): 31-41, 2004 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-15465594

RESUMEN

Corticotropin-releasing hormone (CRH) is a central mediator in the response to stress, coordinating behavioral, autonomic and neuroendocrine activation. CRH overproduction is implicated in several affective disorders, including major depression, panic-anxiety disorder and anorexia--diseases also associated with altered immune function. We investigated the link between CRH overdrive and immune function using CRH transgenic mice. Following immunization, CRH transgenic mice fail to form germinal centers; chronic glucocorticoid administration recapitulates this effect in wild-type mice. Regulation of germinal centers by glucocorticoids appears to be mediated, in part, through effects on follicular dendritic cells (FDC), providing a novel mechanism by which CRH dysregulation may significantly impair humoral immune responses.


Asunto(s)
Corticosterona/fisiología , Hormona Liberadora de Corticotropina/biosíntesis , Hormona Liberadora de Corticotropina/fisiología , Células Dendríticas Foliculares/inmunología , Centro Germinal/patología , Animales , Diferenciación Celular/inmunología , Corticosterona/farmacología , Células Dendríticas Foliculares/efectos de los fármacos , Células Dendríticas Foliculares/metabolismo , Relación Dosis-Respuesta a Droga , Centro Germinal/efectos de los fármacos , Centro Germinal/inmunología , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA