Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 69
Filtrar
1.
Immunity ; 54(2): 308-323.e6, 2021 02 09.
Artículo en Inglés | MEDLINE | ID: mdl-33421362

RESUMEN

Th17 cells are known to exert pathogenic and non-pathogenic functions. Although the cytokine transforming growth factor ß1 (TGF-ß1) is instrumental for Th17 cell differentiation, it is dispensable for generation of pathogenic Th17 cells. Here, we examined the T cell-intrinsic role of Activin-A, a TGF-ß superfamily member closely related to TGF-ß1, in pathogenic Th17 cell differentiation. Activin-A expression was increased in individuals with relapsing-remitting multiple sclerosis and in mice with experimental autoimmune encephalomyelitis. Stimulation with interleukin-6 and Activin-A induced a molecular program that mirrored that of pathogenic Th17 cells and was inhibited by blocking Activin-A signaling. Genetic disruption of Activin-A and its receptor ALK4 in T cells impaired pathogenic Th17 cell differentiation in vitro and in vivo. Mechanistically, extracellular-signal-regulated kinase (ERK) phosphorylation, which was essential for pathogenic Th17 cell differentiation, was suppressed by TGF-ß1-ALK5 but not Activin-A-ALK4 signaling. Thus, Activin-A drives pathogenic Th17 cell differentiation, implicating the Activin-A-ALK4-ERK axis as a therapeutic target for Th17 cell-related diseases.


Asunto(s)
Activinas/metabolismo , Encefalomielitis Autoinmune Experimental/inmunología , Esclerosis Múltiple/inmunología , Inflamación Neurogénica/inmunología , Células Th17/inmunología , Factor de Crecimiento Transformador beta/metabolismo , Receptores de Activinas Tipo I/genética , Receptores de Activinas Tipo I/metabolismo , Activinas/genética , Animales , Diferenciación Celular , Células Cultivadas , Humanos , Ratones , Ratones Noqueados , Terapia Molecular Dirigida , Transducción de Señal
2.
Immunity ; 49(5): 886-898.e5, 2018 11 20.
Artículo en Inglés | MEDLINE | ID: mdl-30446383

RESUMEN

Pathogenic Th17 (pTh17) cells drive inflammation and immune-pathology, but whether pTh17 cells are a Th17 cell subset whose generation is under specific molecular control remains unaddressed. We found that Ras p21 protein activator 3 (RASA3) was highly expressed by pTh17 cells relative to non-pTh17 cells and was required specifically for pTh17 generation in vitro and in vivo. Mice conditionally deficient for Rasa3 in T cells showed less pathology during experimental autoimmune encephalomyelitis. Rasa3-deficient T cells acquired a Th2 cell-biased program that dominantly trans-suppressed pTh17 cell generation via interleukin 4 production. The Th2 cell bias of Rasa3-deficient T cells was due to aberrantly elevated transcription factor IRF4 expression. RASA3 promoted proteasome-mediated IRF4 protein degradation by facilitating interaction of IRF4 with E3-ubiquitin ligase Cbl-b. Therefore, a RASA3-IRF4-Cbl-b pathway specifically directs pTh17 cell generation by balancing reciprocal Th17-Th2 cell programs. These findings indicate that a distinct molecular program directs pTh17 cell generation and reveals targets for treating pTh17 cell-related pathology and diseases.


Asunto(s)
Diferenciación Celular/genética , Proteínas Activadoras de GTPasa/genética , Células Th17/citología , Células Th17/metabolismo , Células Th2/citología , Células Th2/metabolismo , Animales , Autoinmunidad , Biomarcadores , Encefalomielitis Autoinmune Experimental/etiología , Encefalomielitis Autoinmune Experimental/metabolismo , Encefalomielitis Autoinmune Experimental/patología , Proteínas Activadoras de GTPasa/metabolismo , Expresión Génica , Inmunofenotipificación , Factores Reguladores del Interferón/genética , Factores Reguladores del Interferón/metabolismo , Ratones , Proteolisis , ARN Mensajero , Células Th17/inmunología , Células Th2/inmunología
3.
Nature ; 591(7849): 300-305, 2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-33505023

RESUMEN

The inflammasome initiates innate defence and inflammatory responses by activating caspase-1 and pyroptotic cell death in myeloid cells1,2. It consists of an innate immune receptor/sensor, pro-caspase-1, and a common adaptor molecule, ASC. Consistent with their pro-inflammatory function, caspase-1, ASC and the inflammasome component NLRP3 exacerbate autoimmunity during experimental autoimmune encephalomyelitis by enhancing the secretion of IL-1ß and IL-18 in myeloid cells3-6. Here we show that the DNA-binding inflammasome receptor AIM27-10 has a T cell-intrinsic and inflammasome-independent role in the function of T regulatory (Treg) cells. AIM2 is highly expressed by both human and mouse Treg cells, is induced by TGFß, and its promoter is occupied by transcription factors that are associated with Treg cells such as RUNX1, ETS1, BCL11B and CREB. RNA sequencing, biochemical and metabolic analyses demonstrated that AIM2 attenuates AKT phosphorylation, mTOR and MYC signalling, and glycolysis, but promotes oxidative phosphorylation of lipids in Treg cells. Mechanistically, AIM2 interacts with the RACK1-PP2A phosphatase complex to restrain AKT phosphorylation. Lineage-tracing analysis demonstrates that AIM2 promotes the stability of Treg cells during inflammation. Although AIM2 is generally accepted as an inflammasome effector in myeloid cells, our results demonstrate a T cell-intrinsic role of AIM2 in restraining autoimmunity by reducing AKT-mTOR signalling and altering immune metabolism to enhance the stability of Treg cells.


Asunto(s)
Autoinmunidad/inmunología , Proteínas de Unión al ADN/inmunología , Encefalomielitis Autoinmune Experimental/inmunología , Encefalomielitis Autoinmune Experimental/prevención & control , Linfocitos T Reguladores/inmunología , Linfocitos T Reguladores/metabolismo , Animales , Proteínas Adaptadoras de Señalización CARD/deficiencia , Proteínas de Unión al ADN/deficiencia , Proteínas de Unión al ADN/genética , Encefalomielitis Autoinmune Experimental/metabolismo , Femenino , Glucólisis , Humanos , Inflamasomas , Inflamación/inmunología , Ratones , Fosforilación Oxidativa , Fosforilación , Proteína Fosfatasa 2/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Proteínas Proto-Oncogénicas c-myc/metabolismo , Receptores de Cinasa C Activada/metabolismo , Serina-Treonina Quinasas TOR/metabolismo , Factores de Transcripción/metabolismo , Factor de Crecimiento Transformador beta
4.
Nat Immunol ; 14(7): 714-22, 2013 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-23708251

RESUMEN

GATA-3 controls T helper type 2 (TH2) differentiation. However, whether GATA-3 regulates the function of mature T cells beyond TH2 determination remains poorly understood. We found that signaling via the T cell antigen receptor (TCR) and cytokine stimulation promoted GATA-3 expression in CD8(+) T cells, which controlled cell proliferation. Although GATA-3-deficient CD8(+) T cells were generated, their peripheral maintenance was impaired, with lower expression of the receptor for interleukin 7 (IL-7R). GATA-3-deficient T cells had defective responses to viral infection and alloantigen. The proto-oncoprotein c-Myc was a critical target of GATA-3 in promoting T cell proliferation. Our study thus demonstrates an essential role for GATA-3 in controlling the maintenance and proliferation of T cells and provides insight into immunoregulation.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Factor de Transcripción GATA3/inmunología , Activación de Linfocitos/inmunología , Proteínas Proto-Oncogénicas c-myc/inmunología , Receptores de Antígenos de Linfocitos T/inmunología , Receptores de Interleucina-7/inmunología , Animales , Proliferación Celular , Inmunoprecipitación de Cromatina , Citometría de Flujo , Enfermedad Injerto contra Huésped/inmunología , Virus de la Coriomeningitis Linfocítica/inmunología , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Noqueados , ARN/química , ARN/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
5.
J Biol Chem ; 299(1): 102739, 2023 01.
Artículo en Inglés | MEDLINE | ID: mdl-36435197

RESUMEN

Recent discovery of the ribosomal protein (RP) RPL11 interacting with and inhibiting the E3 ubiquitin ligase function of MDM2 established the RP-MDM2-p53 signaling pathway, which is linked to biological events, including ribosomal biogenesis, nutrient availability, and metabolic homeostasis. Mutations in RPs lead to a diverse array of phenotypes known as ribosomopathies in which the role of p53 is implicated. Here, we generated conditional RPL11-deletion mice to investigate in vivo effects of impaired RP expression and its functional connection with p53. While deletion of one Rpl11 allele in germ cells results in embryonic lethality, deletion of one Rpl11 allele in adult mice does not affect viability but leads to acute anemia. Mechanistically, we found RPL11 haploinsufficiency activates p53 in hematopoietic tissues and impedes erythroid precursor differentiation, resulting in insufficient red blood cell development. We demonstrated that reducing p53 dosage by deleting one p53 allele rescues RPL11 haploinsufficiency-induced inhibition of erythropoietic precursor differentiation and restores normal red blood cell levels in mice. Furthermore, blocking the RP-MDM2-p53 pathway by introducing an RP-binding mutation in MDM2 prevents RPL11 haploinsufficiency-caused p53 activation and rescues the anemia in mice. Together, these findings demonstrate that the RP-MDM2-p53 pathway is a critical checkpoint for RP homeostasis and that p53-dependent cell cycle arrest of erythroid precursors is the molecular basis for the anemia phenotype commonly associated with RP deficiency.


Asunto(s)
Anemia , Proteína p53 Supresora de Tumor , Animales , Ratones , Anemia/genética , Haploinsuficiencia , Mutación , Proteínas Proto-Oncogénicas c-mdm2/genética , Proteínas Proto-Oncogénicas c-mdm2/metabolismo , Proteínas Ribosómicas/genética , Proteínas Ribosómicas/metabolismo , Proteína p53 Supresora de Tumor/genética , Proteína p53 Supresora de Tumor/metabolismo
6.
Immunity ; 42(1): 68-79, 2015 Jan 20.
Artículo en Inglés | MEDLINE | ID: mdl-25577439

RESUMEN

Transforming growth factor-beta (TGF-ß) suppresses T cell function to maintain self-tolerance and to promote tumor immune evasion. Yet how Smad4, a transcription factor component of TGF-ß signaling, regulates T cell function remains unclear. Here we have demonstrated an essential role for Smad4 in promoting T cell function during autoimmunity and anti-tumor immunity. Smad4 deletion rescued the lethal autoimmunity resulting from transforming growth factor-beta receptor (TGF-ßR) deletion and compromised T-cell-mediated tumor rejection. Although Smad4 was dispensable for T cell generation, homeostasis, and effector function, it was essential for T cell proliferation after activation in vitro and in vivo. The transcription factor Myc was identified to mediate Smad4-controlled T cell proliferation. This study thus reveals a requirement of Smad4 for T-cell-mediated autoimmunity and tumor rejection, which is beyond the current paradigm. It highlights a TGF-ßR-independent role for Smad4 in promoting T cell function, autoimmunity, and anti-tumor immunity.


Asunto(s)
Enfermedad Injerto contra Huésped/inmunología , Receptores de Factores de Crecimiento Transformadores beta/metabolismo , Proteína Smad4/metabolismo , Subgrupos de Linfocitos T/fisiología , Linfocitos T Reguladores/fisiología , Animales , Autoinmunidad/genética , Proliferación Celular/genética , Células Cultivadas , Tolerancia Inmunológica/genética , Activación de Linfocitos/genética , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Noqueados , Proteínas Proto-Oncogénicas c-myc/metabolismo , Receptores de Factores de Crecimiento Transformadores beta/genética , Transducción de Señal/genética , Transducción de Señal/inmunología , Proteína Smad4/genética , Subgrupos de Linfocitos T/trasplante , Linfocitos T Reguladores/trasplante , Quimera por Trasplante , Escape del Tumor
7.
J Immunol ; 209(1): 49-56, 2022 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-35750335

RESUMEN

The objective of this study is to determine the mechanism of action of anti-CD52 mAb treatment in patients with relapsing-remitting multiple sclerosis (RRMS). Experimental autoimmune encephalomyelitis (EAE), an animal model of the disease, was used to address the role of T regulatory cells (Tregs) in the anti-CD52 mAb-induced suppression of the disease. In vitro studies on PBMCs from RRMS patients and matched healthy controls determined the effect of IL-7 on the expansion of CD4+CD25+CD127- Tregs and induction of their suppressive phenotype. This study using EAE animal models of MS has shown that mouse anti-CD52 mAb suppression of clinical disease was augmented by coadministration of IL-7 and partially reversed by anti-IL-7 mAb. In vitro human studies showed that IL-7 induced expansion of CD4+CD25+CD127- Tregs and increased their FOXP3, GITIR, CD46, CTLA-4, granzyme B, and perforin expression. Anti-CD52 mAb treatment of mice with relapsing-remitting EAE induced expansion of Foxp3+CD4+ Tregs and the suppression of IL-17A+CD4+ and IFN-γ+CD4+ cells in peripheral immune organs and CNS infiltrates. The effect was detected immediately after the treatment and maintained over long-term follow-up. Foxp3+CD4+ Treg-mediated suppression of IL-17A+CD4+ and IFN-γ+CD4+ cells in the spinal cord infiltrates was reversed after inducible Foxp3 depletion. Our results demonstrated that the therapeutic effect of U.S. Food and Drug Administration-approved anti-CD52 mAb is dependent on the presence of Tregs.


Asunto(s)
Encefalomielitis Autoinmune Experimental , Esclerosis Múltiple Recurrente-Remitente , Esclerosis Múltiple , Alemtuzumab/uso terapéutico , Animales , Factores de Transcripción Forkhead/metabolismo , Interleucina-17/metabolismo , Ratones , Esclerosis Múltiple Recurrente-Remitente/tratamiento farmacológico , Linfocitos T Reguladores
8.
Nature ; 551(7678): 105-109, 2017 11 02.
Artículo en Inglés | MEDLINE | ID: mdl-29072299

RESUMEN

T helper 17 (TH17) cells are critically involved in host defence, inflammation, and autoimmunity. Transforming growth factor ß (TGFß) is instrumental in TH17 cell differentiation by cooperating with interleukin-6 (refs 6, 7). Yet, the mechanism by which TGFß enables TH17 cell differentiation remains elusive. Here we reveal that TGFß enables TH17 cell differentiation by reversing SKI-SMAD4-mediated suppression of the expression of the retinoic acid receptor (RAR)-related orphan receptor γt (RORγt). We found that, unlike wild-type T cells, SMAD4-deficient T cells differentiate into TH17 cells in the absence of TGFß signalling in a RORγt-dependent manner. Ectopic SMAD4 expression suppresses RORγt expression and TH17 cell differentiation of SMAD4-deficient T cells. However, TGFß neutralizes SMAD4-mediated suppression without affecting SMAD4 binding to the Rorc locus. Proteomic analysis revealed that SMAD4 interacts with SKI, a transcriptional repressor that is degraded upon TGFß stimulation. SKI controls histone acetylation and deacetylation of the Rorc locus and TH17 cell differentiation via SMAD4: ectopic SKI expression inhibits H3K9 acetylation of the Rorc locus, Rorc expression, and TH17 cell differentiation in a SMAD4-dependent manner. Therefore, TGFß-induced disruption of SKI reverses SKI-SMAD4-mediated suppression of RORγt to enable TH17 cell differentiation. This study reveals a critical mechanism by which TGFß controls TH17 cell differentiation and uncovers the SKI-SMAD4 axis as a potential therapeutic target for treating TH17-related diseases.


Asunto(s)
Diferenciación Celular , Proteínas de Unión al ADN/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Proteínas Represoras/metabolismo , Proteína Smad4/metabolismo , Células Th17/citología , Células Th17/metabolismo , Factor de Crecimiento Transformador beta/metabolismo , Animales , Diferenciación Celular/genética , Femenino , Eliminación de Gen , Humanos , Interleucina-6/metabolismo , Masculino , Ratones , Complejos Multiproteicos/química , Complejos Multiproteicos/metabolismo , Miembro 3 del Grupo F de la Subfamilia 1 de Receptores Nucleares/deficiencia , Miembro 3 del Grupo F de la Subfamilia 1 de Receptores Nucleares/genética , Miembro 3 del Grupo F de la Subfamilia 1 de Receptores Nucleares/metabolismo , Receptores de Factores de Crecimiento Transformadores beta/metabolismo , Transducción de Señal , Proteína Smad4/deficiencia , Proteína Smad4/genética
9.
Nat Immunol ; 11(10): 962-8, 2010 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-20802482

RESUMEN

Induced regulatory T cells (iT(reg) cells) can be generated by peripheral dendritic cells (DCs) that mediate T cell unresponsiveness to rechallenge with antigen. The molecular factors required for the function of such iT(reg) cells remain unknown. We report a critical role for the transcription cofactor homeodomain-only protein (Hop; also known as Hopx) in iT(reg) cells to mediate T cell unresponsiveness in vivo. Hopx-sufficient iT(reg) cells downregulated expression of the transcription factor AP-1 complex and suppressed other T cells. In the absence of Hopx, iT(reg) cells had high expression of the AP-1 complex, proliferated and failed to mediate T cell unresponsiveness to rechallenge with antigen. Thus, Hopx is required for the function of T(reg) cells induced by DCs and the promotion of DC-mediated T cell unresponsiveness in vivo.


Asunto(s)
Células Dendríticas/inmunología , Proteínas de Homeodominio/inmunología , Linfocitos T Reguladores/inmunología , Animales , Proteínas de Homeodominio/genética , Tolerancia Inmunológica , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Factor de Transcripción AP-1/metabolismo
11.
Immunity ; 35(3): 337-48, 2011 Sep 23.
Artículo en Inglés | MEDLINE | ID: mdl-21924928

RESUMEN

Forkhead Box P3 (Foxp3)-expressing regulatory T (Treg) cells are central to maintaining self-tolerance and immune homeostasis. How Treg cell function and Foxp3 expression are regulated is an important question under intensive investigation. Here, we have demonstrated an essential role for the transcription factor GATA-3, a previously recognized Th2 cell master regulator, in controlling Treg cell function. Treg cell-specific GATA-3 deletion led to a spontaneous inflammatory disorder in mice. GATA-3-null Treg cells were defective in peripheral homeostasis and suppressive function, gained Th17 cell phenotypes, and expressed reduced amounts of Foxp3. In addition, GATA-3 controlled Foxp3 expression by binding to and promoting the activity of cis-acting elements of Foxp3. Furthermore, the combined function of GATA-3 and Foxp3 was essential for Foxp3 expression. These findings provide insights into immune regulatory mechanisms and uncover a critical function of GATA-3 in Treg cells and immune tolerance.


Asunto(s)
Factor de Transcripción GATA3/genética , Regulación de la Expresión Génica , Tolerancia Inmunológica , Linfocitos T Reguladores/inmunología , Animales , Proliferación Celular , Células Cultivadas , Citocinas/metabolismo , Regulación hacia Abajo , Citometría de Flujo , Factores de Transcripción Forkhead/genética , Factores de Transcripción Forkhead/metabolismo , Homeostasis/genética , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Unión Proteica , Linfocitos T Reguladores/citología
12.
J Immunol ; 198(3): 1130-1141, 2017 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-28003377

RESUMEN

IL-10 is essential to maintain intestinal homeostasis. CD4+ T regulatory type 1 (TR1) cells produce large amounts of this cytokine and are therefore currently being examined in clinical trials as T cell therapy in patients with inflammatory bowel disease. However, factors and molecular signals sustaining TR1 cell regulatory activity still need to be identified to optimize the efficiency and ensure the safety of these trials. We investigated the role of IL-10 signaling in mature TR1 cells in vivo. Double IL-10eGFP Foxp3mRFP reporter mice and transgenic mice with impairment in IL-10 receptor signaling were used to test the activity of TR1 cells in a murine inflammatory bowel disease model, a model that resembles the trials performed in humans. The molecular signaling was elucidated in vitro. Finally, we used human TR1 cells, currently employed for cell therapy, to confirm our results. We found that murine TR1 cells expressed functional IL-10Rα. TR1 cells with impaired IL-10 receptor signaling lost their regulatory activity in vivo. TR1 cells required IL-10 receptor signaling to activate p38 MAPK, thereby sustaining IL-10 production, which ultimately mediated their suppressive activity. Finally, we confirmed these data using human TR1 cells. In conclusion, TR1 cell regulatory activity is dependent on IL-10 receptor signaling. These data suggest that to optimize TR1 cell-based therapy, IL-10 receptor expression has to be taken into consideration.


Asunto(s)
Receptores de Interleucina-10/fisiología , Transducción de Señal/fisiología , Linfocitos T Reguladores/inmunología , Animales , Interleucina-10/fisiología , Ratones , Ratones Endogámicos C57BL , Fosforilación , Factor de Transcripción STAT3/metabolismo , Células Th17/inmunología , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
13.
J Immunol ; 197(11): 4325-4333, 2016 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-27799308

RESUMEN

Bromodomain PHD finger transcription factor (BPTF), a ubiquitously expressed ATP-dependent chromatin-remodeling factor, is critical for epigenetically regulating DNA accessibility and gene expression. Although BPTF is important for the development of thymocytes, its function in mature T cells remains largely unknown. By specifically deleting BPTF from late double-negative 3/double-negative 4 stage of developing T cells, we found that BPTF was critical for the homeostasis of T cells via a cell-intrinsic manner. In addition, BPTF was essential for the maintenance and function of regulatory T (Treg) cells. Treg cell-specific BPTF deletion led to reduced Foxp3 expression, increased lymphocyte infiltration in the nonlymphoid organs, and a systemic autoimmune syndrome. These findings therefore reveal a vital role for BPTF in T and Treg cell function and immune homeostasis.


Asunto(s)
Antígenos Nucleares/inmunología , Regulación de la Expresión Génica/inmunología , Homeostasis/inmunología , Proteínas del Tejido Nervioso/inmunología , Linfocitos T Reguladores/inmunología , Factores de Transcripción/inmunología , Animales , Antígenos Nucleares/genética , Enfermedades Autoinmunes/genética , Enfermedades Autoinmunes/inmunología , Enfermedades Autoinmunes/patología , Factores de Transcripción Forkhead/inmunología , Homeostasis/genética , Ratones , Ratones Noqueados , Proteínas del Tejido Nervioso/genética , Síndrome , Factores de Transcripción/genética
14.
EMBO J ; 32(3): 395-408, 2013 Feb 06.
Artículo en Inglés | MEDLINE | ID: mdl-23321680

RESUMEN

Treg activation in response to environmental cues is necessary for regulatory T cells (Tregs) to suppress inflammation, but little is known about the transcription mechanisms controlling Treg activation. We report that despite the known proinflammatory role of the chromatin-remodelling factor BRG1 in CD4 cells, deleting Brg1 in all αß T cell lineages led to fatal inflammation, which reflected essential roles of BRG1 in Tregs. Brg1 deletion impaired Treg activation, concomitant with the onset of the inflammation. Remarkably, as the inflammation progressed, Tregs became increasingly activated, but the activation levels could not catch up with the severity of inflammation. In vitro assays indicate that BRG1 regulates a subset of TCR target genes including multiple chemokine receptor genes. Finally, using a method that can create littermates bearing either a tissue-specific point mutation or deletion, we found the BRG1 ATPase activity partially dispensable for BRG1 function. Collectively, these data suggest that BRG1 acts in part via remodelling-independent functions to sensitize Tregs to inflammatory cues, thus allowing Tregs to promptly and effectively suppress autoimmunity.


Asunto(s)
Ensamble y Desensamble de Cromatina/inmunología , ADN Helicasas/inmunología , Tolerancia Inmunológica/inmunología , Proteínas Nucleares/inmunología , Linfocitos T Reguladores/inmunología , Factores de Transcripción/inmunología , Animales , Inmunoprecipitación de Cromatina , Concanavalina A , Citocinas/inmunología , ADN Helicasas/genética , Cartilla de ADN/genética , Femenino , Eliminación de Gen , Técnicas Histológicas , Activación de Linfocitos/inmunología , Masculino , Ratones , Ratones Noqueados , Proteínas Nucleares/genética , Factores de Transcripción/genética
15.
Trends Immunol ; 35(6): 233-42, 2014 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-24786134

RESUMEN

GATA3 has conventionally been regarded as a transcription factor that drives the differentiation of T helper (Th) 2 cells. Increasing evidence points to a function for GATA3 beyond controlling Th2 differentiation. GATA3 regulates T cell development, proliferation, and maintenance. Furthermore, recent studies have demonstrated important roles for GATA3 in innate lymphoid cells. Thus, GATA3 emerges as a factor with diverse functions in immune regulation, which are in some cases cell-type specific and in others shared by multiple cell types. Here, I discuss recent discoveries and the current understanding of the functions of GATA3 in immune regulation.


Asunto(s)
Factor de Transcripción GATA3/metabolismo , Inmunidad/fisiología , Inmunidad Adaptativa , Animales , Factor de Transcripción GATA3/genética , Regulación de la Expresión Génica , Humanos , Inmunidad Innata , Activación de Linfocitos , Subgrupos Linfocitarios/inmunología , Subgrupos Linfocitarios/metabolismo , Transducción de Señal
16.
Nature ; 475(7357): 514-8, 2011 Jul 17.
Artículo en Inglés | MEDLINE | ID: mdl-21765430

RESUMEN

Interleukin (IL)-17-producing T helper cells (T(H)17) are a recently identified CD4(+) T cell subset distinct from T helper type 1 (T(H)1) and T helper type 2 (T(H)2) cells. T(H)17 cells can drive antigen-specific autoimmune diseases and are considered the main population of pathogenic T cells driving experimental autoimmune encephalomyelitis (EAE), the mouse model for multiple sclerosis. The factors that are needed for the generation of T(H)17 cells have been well characterized. However, where and how the immune system controls T(H)17 cells in vivo remains unclear. Here, by using a model of tolerance induced by CD3-specific antibody, a model of sepsis and influenza A viral infection (H1N1), we show that pro-inflammatory T(H)17 cells can be redirected to and controlled in the small intestine. T(H)17-specific IL-17A secretion induced expression of the chemokine CCL20 in the small intestine, facilitating the migration of these cells specifically to the small intestine via the CCR6/CCL20 axis. Moreover, we found that T(H)17 cells are controlled by two different mechanisms in the small intestine: first, they are eliminated via the intestinal lumen; second, pro-inflammatory T(H)17 cells simultaneously acquire a regulatory phenotype with in vitro and in vivo immune-suppressive properties (rT(H)17). These results identify mechanisms limiting T(H)17 cell pathogenicity and implicate the gastrointestinal tract as a site for control of T(H)17 cells.


Asunto(s)
Intestino Delgado/inmunología , Células Th17/inmunología , Animales , Anticuerpos/inmunología , Anticuerpos/farmacología , Complejo CD3/inmunología , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD4-Positivos/trasplante , Movimiento Celular/efectos de los fármacos , Quimiocina CCL20/inmunología , Modelos Animales de Enfermedad , Encefalomielitis Autoinmune Experimental/inmunología , Femenino , Perfilación de la Expresión Génica , Regulación de la Expresión Génica/inmunología , Virus de la Influenza A/inmunología , Interleucina-17/inmunología , Intestino Delgado/citología , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Transgénicos , Infecciones por Orthomyxoviridae/inmunología , Receptores CCR6/inmunología , Sepsis/inmunología , Infecciones Estafilocócicas/inmunología
17.
Adv Exp Med Biol ; 1011: 163-196, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28875490

RESUMEN

Tumor microenvironment (TME) is composed of tumor cells, immune cells, cytokines, extracellular matrix, etc. The immune system and the metabolisms of glucose, lipids, amino acids, and nucleotides are integrated in the tumorigenesis and development. Cancer cells and immune cells show metabolic reprogramming in the TME, which intimately links immune cell functions and edits tumor immunology. Recent findings in immune cell metabolism hold the promising possibilities toward clinical therapeutics for treating cancer. This chapter introduces the updated understandings of metabolic reprogramming of immune cells in the TME and suggests new directions in manipulation of immune responses for cancer diagnosis and therapy.


Asunto(s)
Sistema Inmunológico/metabolismo , Neoplasias/inmunología , Microambiente Tumoral , Carcinogénesis , Transformación Celular Neoplásica , Humanos
18.
Proc Natl Acad Sci U S A ; 109(3): 905-10, 2012 Jan 17.
Artículo en Inglés | MEDLINE | ID: mdl-22219364

RESUMEN

TGF-ß modulates immune response by suppressing non-regulatory T (Treg) function and promoting Treg function. The question of whether TGF-ß achieves distinct effects on non-Treg and Treg cells through discrete signaling pathways remains outstanding. In this study, we investigated the requirements of Smad-dependent and -independent TGF-ß signaling for T-cell function. Smad2 and Smad3 double deficiency in T cells led to lethal inflammatory disorder in mice. Non-Treg cells were spontaneously activated and produced effector cytokines in vivo on deletion of both Smad2 and Smad3. In addition, TGF-ß failed to suppress T helper differentiation efficiently and to promote induced Treg generation of non-Treg cells lacking both Smad2 and Smad3, suggesting that Smad-dependent signaling is obligatory to mediate TGF-ß function in non-Treg cells. Unexpectedly, however, the development, homeostasis, and function of Treg cells remained intact in the absence of Smad2 and Smad3, suggesting that the Smad-independent pathway is important for Treg function. Indeed, Treg-specific deletion of TGF-ß-activated kinase 1 led to failed Treg homeostasis and lethal immune disorder in mice. Therefore, Smad-dependent and -independent TGF-ß signaling discretely controls non-Treg and Treg function to modulate immune tolerance and immune homeostasis.


Asunto(s)
Transducción de Señal/inmunología , Proteína Smad2/metabolismo , Proteína smad3/metabolismo , Linfocitos T/inmunología , Factor de Crecimiento Transformador beta/metabolismo , Animales , Eliminación de Gen , Homeostasis/inmunología , Inflamación/patología , Integrasas/metabolismo , Quinasas Quinasa Quinasa PAM/metabolismo , Ratones , Ratones Noqueados , Fenotipo , Proteína Smad2/deficiencia , Proteína smad3/deficiencia , Linfocitos T/citología , Linfocitos T Reguladores/citología , Linfocitos T Reguladores/inmunología
19.
Int J Mol Sci ; 16(5): 10267-80, 2015 May 06.
Artículo en Inglés | MEDLINE | ID: mdl-25955647

RESUMEN

Interleukin-7 (IL-7) is a non-hematopoietic cell-derived cytokine with a central role in the adaptive immune system. It promotes lymphocyte development in the thymus and maintains survival of naive and memory T cell homeostasis in the periphery. Moreover, it is important for the organogenesis of lymph nodes (LN) and for the maintenance of activated T cells recruited into the secondary lymphoid organs (SLOs). The immune capacity of cancer patients is suppressed that is characterized by lower T cell counts, less effector immune cells infiltration, higher levels of exhausted effector cells and higher levels of immunosuppressive cytokines, such as transforming growth factor ß (TGF-ß). Recombinant human IL-7 (rhIL-7) is an ideal solution for the immune reconstitution of lymphopenia patients by promoting peripheral T cell expansion. Furthermore, it can antagonize the immunosuppressive network. In animal models, IL-7 has been proven to prolong the survival of tumor-bearing hosts. In this review, we will focus on the mechanism of action and applications of IL-7 in cancer immunotherapy and the potential restrictions for its usage.


Asunto(s)
Inmunoterapia/métodos , Interleucina-7/metabolismo , Neoplasias/terapia , Animales , Humanos , Interleucina-7/uso terapéutico , Neoplasias/metabolismo , Transducción de Señal , Linfocitos T/inmunología
20.
J Immunol ; 189(9): 4417-25, 2012 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-22993204

RESUMEN

Dihydroartemisinin (DHA) is an important derivative of the herb medicine Artemisia annua L., used in ancient China. DHA is currently used worldwide to treat malaria by killing malaria-causing parasites. In addition to this prominent effect, DHA is thought to regulate cellular functions, such as angiogenesis, tumor cell growth, and immunity. Nonetheless, how DHA affects T cell function remains poorly understood. We found that DHA potently suppressed Th cell differentiation in vitro. Unexpectedly, however, DHA greatly promoted regulatory T cell (Treg) generation in a manner dependent on the TGF-ßR:Smad signal. In addition, DHA treatment effectively reduced onset of experimental autoimmune encephalomyelitis (EAE) and ameliorated ongoing EAE in mice. Administration of DHA significantly decreased Th but increased Tregs in EAE-inflicted mice, without apparent global immune suppression. Moreover, DHA modulated the mammalian target of rapamycin (mTOR) pathway, because mTOR signal was attenuated in T cells upon DHA treatment. Importantly, enhanced Akt activity neutralized DHA-mediated effects on T cells in an mTOR-dependent fashion. This study therefore reveals a novel immune regulatory function of DHA in reciprocally regulating Th and Treg cell generation through the modulating mTOR pathway. It addresses how DHA regulates immune function and suggests a new type of drug for treating diseases in which mTOR activity is to be tempered.


Asunto(s)
Artemisininas/farmacología , Inflamación/prevención & control , Transducción de Señal/inmunología , Linfocitos T Colaboradores-Inductores/efectos de los fármacos , Linfocitos T Colaboradores-Inductores/inmunología , Linfocitos T Reguladores/efectos de los fármacos , Linfocitos T Reguladores/inmunología , Serina-Treonina Quinasas TOR/metabolismo , Secuencia de Aminoácidos , Animales , Encefalomielitis Autoinmune Experimental/inmunología , Encefalomielitis Autoinmune Experimental/patología , Encefalomielitis Autoinmune Experimental/prevención & control , Inflamación/inmunología , Inflamación/patología , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Datos de Secuencia Molecular , Transducción de Señal/efectos de los fármacos , Linfocitos T Reguladores/patología , Serina-Treonina Quinasas TOR/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA