Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Adv Exp Med Biol ; 1444: 67-82, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38467973

RESUMEN

Regulatory T (Treg) cells, which specifically express the master transcription factor FoxP3, are indispensable for the maintenance of immunological self-tolerance and homeostasis. Their functional or numerical anomalies can be causative of autoimmune and other inflammatory diseases. Recent advances in the research of the cellular and molecular basis of how Treg cells develop, exert suppression, and maintain their function have enabled devising various ways for controlling physiological and pathological immune responses by targeting Treg cells. It is now envisaged that Treg cells as a "living drug" are able to achieve antigen-specific immune suppression of various immune responses and reestablish immunological self-tolerance in the clinic.


Asunto(s)
Autoinmunidad , Linfocitos T Reguladores , Autotolerancia , Tolerancia Inmunológica , Regulación de la Expresión Génica , Factores de Transcripción Forkhead/genética , Factores de Transcripción Forkhead/metabolismo
2.
Immunother Adv ; 3(1): ltad007, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37397971

RESUMEN

FoxP3-expressing regulatory T cells (Tregs), whether naturally generated in the immune system or unnaturally induced from conventional T cells (Tconvs) in the laboratory, have much therapeutic value in treating immunological diseases and establishing transplantation tolerance. Natural Tregs (nTregs) can be selectively expanded in vivo by administration of low-dose IL-2 or IL-2 muteins for immune suppression. For adoptive Treg cell therapy, nTregs can be expanded in vitro by strong antigenic stimulation in the presence of IL-2. Synthetic receptors such as CAR can be expressed in nTregs to equip them with a particular target specificity for suppression. In addition, antigen-specific Tconvs can be converted in vitro to functionally stable Treg-like cells by a combination of antigenic stimulation, FoxP3 induction, and establishment of the Treg-type epigenome. This review discusses current and prospective strategies for Treg-based immune suppression and the issues to be resolved for achieving stable antigen-specific immune suppression and tolerance induction in the clinic by targeting Tregs.

3.
Biol Pharm Bull ; 45(8): 1053-1060, 2022 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-35613869

RESUMEN

Combination treatment using fingolimod (FTY720), an immunomodulator, and a pathogenic antigen prevents the progression of glucose-6-phosphate isomerase (GPI)325-339-induced arthritis. In this study, we focused on myeloid-derived suppressor cells (MDSCs; CD11b+Gr-1+ cells) and investigated the effects of the combination treatment on these cells. DBA/1J mice with GPI325-339-induced arthritis were treated using FTY720 and/or GPI325-339 for five days. The expanded CD11b+Gr-1+ cell population and its inhibitory potential were examined. The percentage of CD369+CD11b+Gr-1+ cells effectively increased in the combination-treated mice. The inhibitory potential of CD369+CD11b+Gr-1+ cells was higher than that of cells not expressing CD369. Among bone marrow cells, the expression of CD369 in CD11b+Gr-1+ cells increased following stimulation with granulocyte-macrophage colony-stimulating factor, and the expression of CD11c increased accordingly. The increased CD11c expression indicated a decrease in the potential to suppress T cell proliferation based on the results of the suppression assay. The percentage of CD11c-CD369+ cells in CD11b+Gr-1+ cells that were induced by the combination treatment also increased, and these cells tended to have a higher capacity to inhibit T cell proliferation. In conclusion, the combination treatment using FTY720 and the pathogenic antigen effectively induces MDSC, which demonstrates a high potential for suppressing T cell proliferation in the lymph nodes, thereby establishing an immune-tolerant state.


Asunto(s)
Artritis Reumatoide , Células Supresoras de Origen Mieloide , Animales , Antígenos , Artritis Reumatoide/tratamiento farmacológico , Artritis Reumatoide/metabolismo , Antígeno CD11b/metabolismo , Antígeno CD11b/uso terapéutico , Clorhidrato de Fingolimod/farmacología , Clorhidrato de Fingolimod/uso terapéutico , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos DBA , Células Mieloides/metabolismo , Células Supresoras de Origen Mieloide/metabolismo
4.
Food Sci Nutr ; 9(6): 3200-3208, 2021 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-34136184

RESUMEN

FoxP3+ regulatory T cells (Tregs) are needed to suppress inflammatory diseases and maintain immune homeostasis. The suppressive function of Tregs can be used to control autoimmune or inflammatory diseases; therefore, it is well studied how Tregs can be artificially up- or downregulated in vitro and in vivo, by using antibodies, chemical compounds, foods, and natural resources. Propolis is a famous functional food that has an anti-inflammatory effect. However, the influences of propolis on Treg function have not been fully evaluated so far. Here, we demonstrated that Brazilian green propolis increases TNFR2 expression in Tregs via the IRF4/cMyc axis, and artepillin C was a major effective component of propolis on Tregs. These results indicate that propolis and artepillin C have the potential as Treg activators via TNFR2 expression and may be useful for the prevention and/or therapy of autoimmune or inflammatory diseases.

5.
Immunity ; 54(5): 947-961.e8, 2021 05 11.
Artículo en Inglés | MEDLINE | ID: mdl-33930308

RESUMEN

The transcription factor Foxp3 plays crucial roles for Treg cell development and function. Conserved non-coding sequences (CNSs) at the Foxp3 locus control Foxp3 transcription, but how they developmentally contribute to Treg cell lineage specification remains obscure. Here, we show that among Foxp3 CNSs, the promoter-upstream CNS0 and the intergenic CNS3, which bind distinct transcription factors, were activated at early stages of thymocyte differentiation prior to Foxp3 promoter activation, with sequential genomic looping bridging these regions and the promoter. While deletion of either CNS0 or CNS3 partially compromised thymic Treg cell generation, deletion of both completely abrogated the generation and impaired the stability of Foxp3 expression in residual Treg cells. As a result, CNS0 and CNS3 double-deleted mice succumbed to lethal systemic autoimmunity and inflammation. Thus, hierarchical and coordinated activation of Foxp3 CNS0 and CNS3 initiates and stabilizes Foxp3 gene expression, thereby crucially controlling Treg cell development, maintenance, and consequently immunological self-tolerance.


Asunto(s)
Elementos de Facilitación Genéticos/inmunología , Factores de Transcripción Forkhead/inmunología , Linfocitos T Reguladores/inmunología , Animales , Diferenciación Celular/inmunología , Linaje de la Célula/inmunología , Regulación de la Expresión Génica/inmunología , Humanos , Tolerancia Inmunológica/inmunología , Activación de Linfocitos/inmunología , Masculino , Ratones , Ratones Endogámicos C57BL , Regiones Promotoras Genéticas/inmunología , Autotolerancia/inmunología
6.
Curr Opin Immunol ; 67: 36-41, 2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-32827951

RESUMEN

Naturally occurring FoxP3+CD4+ regulatory T (Treg) cells indispensable for the maintenance of immunological self-tolerance and homeostasis are instrumental in treating autoimmune and other immunological disorders. Stable function of natural Treg cells requires not only the expression of Foxp3 and other Treg signature genes such as CD25 and CTLA-4 but also the generation of Treg-specific epigenetic changes, especially Treg-specific DNA hypomethylation, at these gene loci. Recent studies have shown that the Treg-specific transcriptional and epigenetic changes can be induced in antigen-specific conventional T cells in vivo and in vitro, converting them to functionally stable Treg cells. Such natural or induced Treg cells bear the potential to achieve stable antigen-specific immune suppression and reestablish immunological self-tolerance in treating and preventing autoimmune diseases.


Asunto(s)
Enfermedades Autoinmunes/terapia , Linfocitos T CD4-Positivos/inmunología , Linfocitos T Reguladores/inmunología , Animales , Enfermedades Autoinmunes/inmunología , Humanos
7.
Proc Natl Acad Sci U S A ; 117(22): 12258-12268, 2020 06 02.
Artículo en Inglés | MEDLINE | ID: mdl-32414925

RESUMEN

Foxp3-expressing regulatory T cells (Tregs) can be generated in vitro by antigenic stimulation of conventional T cells (Tconvs) in the presence of TGF-ß and IL-2. However, unlike Foxp3+ naturally occurring Tregs, such in vitro induced Tregs (iTregs) are functionally unstable mainly because of incomplete Treg-type epigenetic changes at Treg signature genes such as Foxp3 Here we show that deprivation of CD28 costimulatory signal at an early stage of iTreg generation is able to establish Treg-specific DNA hypomethylation at Treg signature genes. It was achieved, for example, by TCR/TGF-ß/IL-2 stimulation of CD28-deficient Tconvs or CD28-intact Tconvs without anti-CD28 agonistic mAb or with CD80/CD86-blocked or -deficient antigen-presenting cells. The signal abrogation could induce Treg-type hypomethylation in memory/effector as well as naive Tconvs, while hindering Tconv differentiation into effector T cells. Among various cytokines and signal activators/inhibitors, TNF-α and PKC agonists inhibited the hypomethylation. Furthermore, CD28 signal deprivation significantly reduced c-Rel expression in iTregs; and the specific genomic perturbation of a NF-κB binding motif at the Foxp3 CNS2 locus enhanced the locus-specific DNA hypomethylation even in CD28 signaling-intact iTregs. In addition, in vitro maintenance of such epigenome-installed iTregs with IL-2 alone, without additional TGF-ß or antigenic stimulation, enabled their expansion and stabilization of Treg-specific DNA hypomethylation. These iTregs indeed stably expressed Foxp3 after in vivo transfer and effectively suppressed antigen-specific immune responses. Taken together, inhibition of the CD28-PKC-NF-κB signaling pathway in iTreg generation enables de novo acquisition of Treg-specific DNA hypomethylation at Treg signature genes and abundant production of functionally stable antigen-specific iTregs for therapeutic purposes.


Asunto(s)
Antígenos CD28/metabolismo , Linfocitos T CD8-positivos/inmunología , Metilación de ADN , Epigénesis Genética , Factores de Transcripción Forkhead/fisiología , Regulación de la Expresión Génica , Linfocitos T Reguladores/inmunología , Animales , Linfocitos T CD8-positivos/metabolismo , Diferenciación Celular , Citocinas/metabolismo , Femenino , Interleucina-6/metabolismo , Masculino , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Transducción de Señal , Linfocitos T Reguladores/metabolismo , Factor de Crecimiento Transformador beta/metabolismo
8.
Sci Immunol ; 4(40)2019 10 25.
Artículo en Inglés | MEDLINE | ID: mdl-31653719

RESUMEN

A promising way to restrain hazardous immune responses, such as autoimmune disease and allergy, is to convert disease-mediating T cells into immunosuppressive regulatory T (Treg) cells. Here, we show that chemical inhibition of the cyclin-dependent kinase 8 (CDK8) and CDK19, or knockdown/knockout of the CDK8 or CDK19 gene, is able to induce Foxp3, a key transcription factor controlling Treg cell function, in antigen-stimulated effector/memory as well as naïve CD4+ and CD8+ T cells. The induction was associated with STAT5 activation, independent of TGF-ß action, and not affected by inflammatory cytokines. Furthermore, in vivo administration of a newly developed CDK8/19 inhibitor along with antigen immunization generated functionally stable antigen-specific Foxp3+ Treg cells, which effectively suppressed skin contact hypersensitivity and autoimmune disease in animal models. The results indicate that CDK8/19 is physiologically repressing Foxp3 expression in activated conventional T cells and that its pharmacological inhibition enables conversion of antigen-specific effector/memory T cells into Foxp3+ Treg cells for the treatment of various immunological diseases.


Asunto(s)
Linfocitos T CD4-Positivos/inmunología , Quinasa 8 Dependiente de Ciclina/antagonistas & inhibidores , Quinasas Ciclina-Dependientes/antagonistas & inhibidores , Factores de Transcripción Forkhead/genética , Inhibidores de Proteínas Quinasas/farmacología , Piridinas/farmacología , Pirimidinas/farmacología , Linfocitos T Reguladores/inmunología , Animales , Antígenos/inmunología , Células Cultivadas , Quinasa 8 Dependiente de Ciclina/deficiencia , Quinasa 8 Dependiente de Ciclina/inmunología , Quinasas Ciclina-Dependientes/deficiencia , Quinasas Ciclina-Dependientes/inmunología , Factores de Transcripción Forkhead/inmunología , Memoria Inmunológica/inmunología , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Endogámicos NOD , Ratones Noqueados , Ratones Transgénicos
9.
Nat Commun ; 10(1): 549, 2019 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-30710091

RESUMEN

The genome organizer, special AT-rich sequence-binding protein-1 (Satb1), plays a pivotal role in the regulation of global gene networks in a cell type-dependent manner and is indispensable for the development of multiple cell types, including mature CD4+ T, CD8+ T, and Foxp3+ regulatory T cells in the thymus. However, it remains unknown how the differentiation and effector program of the Th subsets in the periphery are regulated by Satb1. Here, we demonstrate that Satb1 differentially regulates gene expression profiles in non-pathogenic and pathogenic Th17 cells and promotes the pathogenic effector program of encephalitogenic Th17 cells by regulating GM-CSF via Bhlhe40 and inhibiting PD-1 expression. However, Satb1 is dispensable for the differentiation and non-pathogenic functions of Th17 cells. These results indicate that Satb1 regulates the specific gene expression and function of effector Th17 cells in tissue inflammation.


Asunto(s)
Inflamación/inmunología , Proteínas de Unión a la Región de Fijación a la Matriz/metabolismo , Células Th17/inmunología , Animales , Biomarcadores/metabolismo , Encefalitis , Encefalomielitis Autoinmune Experimental/patología , Regulación de la Expresión Génica , Sitios Genéticos , Inflamación/patología , Activación de Linfocitos/inmunología , Ratones Endogámicos C57BL , Ratones Noqueados , Receptor de Muerte Celular Programada 1/metabolismo , Regiones Promotoras Genéticas/genética , Unión Proteica , Médula Espinal/metabolismo
11.
Methods Mol Biol ; 1585: 141-153, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28477193

RESUMEN

IL-9-producing Th9 cell is a novel Th cell subset involved in type II allergic inflammations such as asthma. Th9 cells can be induced from naïve Th cells in the presence of IL-4 and TGF-ß. It is also well established that downstream signals of IL-4 and TGF-ß, including STAT6, IRF4, Smad, and PU.1, directly mediate IL-9 production in Th9 cells. In this chapter we describe the methods of flow cytometry, qPCR and western blot analysis to determine the expression or activation of these transcription factors downstream of IL-4 and TGF-ß.


Asunto(s)
Interleucina-4/metabolismo , Factor de Crecimiento Transformador beta/metabolismo , Western Blotting , Citometría de Flujo , Humanos , Interleucina-9/metabolismo , Reacción en Cadena en Tiempo Real de la Polimerasa , Linfocitos T Colaboradores-Inductores/metabolismo
13.
Nat Immunol ; 18(2): 173-183, 2017 02.
Artículo en Inglés | MEDLINE | ID: mdl-27992401

RESUMEN

Most Foxp3+ regulatory T (Treg) cells develop in the thymus as a functionally mature T cell subpopulation specialized for immune suppression. Their cell fate appears to be determined before Foxp3 expression; yet molecular events that prime Foxp3- Treg precursor cells are largely obscure. We found that Treg cell-specific super-enhancers (Treg-SEs), which were associated with Foxp3 and other Treg cell signature genes, began to be activated in Treg precursor cells. T cell-specific deficiency of the genome organizer Satb1 impaired Treg-SE activation and the subsequent expression of Treg signature genes, causing severe autoimmunity due to Treg cell deficiency. These results suggest that Satb1-dependent Treg-SE activation is crucial for Treg cell lineage specification in the thymus and that its perturbation is causative of autoimmune and other immunological diseases.


Asunto(s)
Diferenciación Celular/inmunología , Factores de Transcripción Forkhead/metabolismo , Proteínas de Unión a la Región de Fijación a la Matriz/metabolismo , Linfocitos T Reguladores/fisiología , Activación Transcripcional/inmunología , Animales , Autoinmunidad , Linaje de la Célula , Células Cultivadas , Elementos de Facilitación Genéticos/genética , Epigénesis Genética , Factores de Transcripción Forkhead/genética , Tolerancia Inmunológica , Masculino , Proteínas de Unión a la Región de Fijación a la Matriz/genética , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Especificidad de Órganos , Células Precursoras de Linfocitos T/fisiología
14.
Biochem Biophys Res Commun ; 477(3): 413-8, 2016 08 26.
Artículo en Inglés | MEDLINE | ID: mdl-27329810

RESUMEN

Human AlkB homolog 8 (ALKBH8) is highly expressed in high-grade, superficially and deeply invasive bladder cancer. Moreover, ALKBH8 knockdown induces apoptosis in bladder cancer cells. However, the underlying anti-apoptotic mechanism of ALKBH8 in bladder cancer cells has thus far remained unclear. Moreover, there is no direct evidence that highly expressed ALKBH8 is involved in tumor progression in vivo. We here show that ALKBH8 knockdown induced apoptosis via downregulating the protein expression of survivin, an anti-apoptotic factor also exhibiting increased levels in bladder cancer. We also clarify that ALKBH8 transgenic mice showed an accelerated rate of bladder tumor mass and invasiveness in an N-butyl-N-(4-hydroxybutyl)-nitrosamine-induced bladder cancer model. These findings suggest that the high expression of ALKBH8 is critical for the growth and progression of bladder cancer.


Asunto(s)
Homólogo 8 de AlkB ARNt Metiltransferasa/fisiología , Proteínas Inhibidoras de la Apoptosis/metabolismo , Neoplasias de la Vejiga Urinaria/patología , Homólogo 8 de AlkB ARNt Metiltransferasa/genética , Animales , Apoptosis/fisiología , Línea Celular Tumoral , Progresión de la Enfermedad , Humanos , Ratones , Ratones Transgénicos , Survivin , Neoplasias de la Vejiga Urinaria/metabolismo
15.
Mol Cancer Res ; 13(3): 565-74, 2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-25381221

RESUMEN

UNLABELLED: Renal cell carcinoma (RCC) is the most common neoplasm of the adult kidney, and clear cell RCC (ccRCC) represents its most common histological subtype. To identify a therapeutic target for ccRCC, miRNA expression signatures from ccRCC clinical specimens were analyzed. miRNA microarray and real-time PCR analyses revealed that miR-629 expression was significantly upregulated in human ccRCC compared with adjacent noncancerous renal tissue. Functional inhibition of miR-629 by a hairpin miRNA inhibitor suppressed ccRCC cell motility and invasion. Mechanistically, miR-629 directly targeted tripartite motif-containing 33 (TRIM33), which inhibits the TGFß/Smad signaling pathway. In clinical ccRCC specimens, downregulation of TRIM33 was observed with the association of both pathologic stages and grades. The miR-629 inhibitor significantly suppressed TGFß-induced Smad activation by upregulating TRIM33 expression and subsequently inhibited the association of Smad2/3 and Smad4. Moreover, a miR-629 mimic enhanced the effect of TGFß on the expression of epithelial-mesenchymal transition-related factors as well as on the motility and invasion in ccRCC cells. These findings identify miR-629 as a potent regulator of the TGFß/Smad signaling pathway via TRIM33 in ccRCC. IMPLICATIONS: This study suggests that miR-629 has biomarker potential through its ability to regulate TGFß/Smad signaling and accelerate ccRCC cell motility and invasion.


Asunto(s)
Carcinoma de Células Renales/genética , Carcinoma de Células Renales/patología , Neoplasias Renales/genética , Neoplasias Renales/patología , MicroARNs/genética , Factores de Transcripción/genética , Adulto , Anciano , Anciano de 80 o más Años , Proteínas Reguladoras de la Apoptosis , Línea Celular Tumoral , Femenino , Regulación Neoplásica de la Expresión Génica , Humanos , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Masculino , Persona de Mediana Edad , Proteínas Mitocondriales/metabolismo , Metástasis de la Neoplasia , Transducción de Señal , Factor de Crecimiento Transformador beta/metabolismo
16.
Mol Cancer Res ; 12(12): 1807-17, 2014 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-25092917

RESUMEN

UNLABELLED: Clear cell renal cell carcinoma (ccRCC) is the most common histologically defined subtype of renal cell carcinoma (RCC). To define the molecular mechanism in the progression of ccRCC, we focused on LOX-like protein 2 (LOXL2), which is critical for the first step in collagen and elastin cross-linking. Using exon array analysis and quantitative validation, LOXL2 was shown to be significantly upregulated in clinical specimens of human ccRCC tumor tissues, compared with adjacent noncancerous renal tissues, and this elevated expression correlated with the pathologic stages of ccRCC. RNAi-mediated knockdown of LOXL2 resulted in marked suppression of stress-fiber and focal adhesion formation in ccRCC cells. Moreover, LOXL2 siRNA knockdown significantly inhibited cell growth, migration, and invasion. Mechanistically, LOXL2 regulated the degradation of both integrins α5 (ITGAV5) and ß1 (ITGB1) via protease- and proteasome-dependent systems. In clinical ccRCC specimens, the expression levels of LOXL2 and integrin α5 correlated with the pathologic tumor grades. In conclusion, LOXL2 is a potent regulator of integrin α5 and integrin ß1 protein levels and functions in a tumor-promoting capacity in ccRCC. IMPLICATIONS: This is the first report demonstrating that LOXL2 is highly expressed and involved in ccRCC progression by regulating the levels of integrins α5 and ß1.


Asunto(s)
Aminoácido Oxidorreductasas/genética , Carcinoma de Células Renales/patología , Integrina alfa5/metabolismo , Integrina beta1/metabolismo , Neoplasias Renales/patología , Adulto , Anciano , Anciano de 80 o más Años , Aminoácido Oxidorreductasas/metabolismo , Carcinoma de Células Renales/genética , Carcinoma de Células Renales/metabolismo , Línea Celular Tumoral , Movimiento Celular , Proliferación Celular , Femenino , Regulación Neoplásica de la Expresión Génica , Células HEK293 , Humanos , Neoplasias Renales/genética , Neoplasias Renales/metabolismo , Masculino , Persona de Mediana Edad
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...