Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 41
Filtrar
1.
Front Immunol ; 11: 1291, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32655569

RESUMEN

The zinc finger protein 36-like 2, ZFP36L2, is a member of a small family of RNA-binding proteins composed by ZFP36 (also known as tristetraprolin, TTP), ZFP36L1 and ZFP36L2 in humans, with corresponding murine orthologs. These proteins bind to adenine uridine-rich element (ARE) in the 3'untranslated region of target messenger RNA and stimulate target degradation. ZFP36 functions as an anti-inflammatory modulator in murine models of inflammatory diseases by down-regulating the production of inflammatory cytokines such as tumor necrosis factor-α. However, how ZFP36L1 and ZFP36L2 alter the function of CD4+ T cells is not completely understood. We addressed this issue by searching for the target genes of ZFP36L2 by comprehensive transcriptome analysis. We observed that ZFP36L2 is highly expressed in naïve CD4+ T cells; however, when CD4+ T cells are stimulated through their T cell receptors, ZFP36L2 expression is rapidly reduced in both humans and mice. Among CD4+ T cell populations, the expression levels of ZFP36L2 in regulatory T cells (Tregs) were significantly lower than those in naïve or effector CD4+ T cells. RNA-sequence analysis revealed that the forced expression of ZFP36L2 decreased Ikzf2 (encoding Helios) expression in Foxp3+ Tregs and inhibited the ability of induced Tregs (iTregs). ZFP36L2 directly bound to and destabilized the 3'untranslated region of Ikzf2 mRNA, which contains AU-rich elements. These results indicate that ZFP36L2 reduces the expression of Ikzf2 and suppresses iTreg function, raising the interesting possibility that the inhibition of ZFP36L2 in iTregs could be a therapeutic strategy for autoimmune diseases.


Asunto(s)
Proteínas de Unión al ADN/biosíntesis , Regulación de la Expresión Génica/inmunología , Factor de Transcripción Ikaros/biosíntesis , Linfocitos T Reguladores/inmunología , Factores de Transcripción/biosíntesis , Factores de Transcripción/inmunología , Tristetraprolina/inmunología , Animales , Proteínas de Unión al ADN/inmunología , Regulación hacia Abajo , Humanos , Factor de Transcripción Ikaros/inmunología , Ratones , Factores de Transcripción/metabolismo , Tristetraprolina/metabolismo
2.
PLoS One ; 15(5): e0233191, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32453736

RESUMEN

The Ikzf1 locus encodes the lymphoid specific transcription factor Ikaros, which plays an essential role in both T and B cell differentiation, while deregulation or mutation of IKZF1/Ikzf1 is involved in leukemia. Tissue-specific and cell identity genes are usually associated with clusters of enhancers, also called super-enhancers, which are believed to ensure proper regulation of gene expression throughout cell development and differentiation. Several potential regulatory regions have been identified in close proximity of Ikzf1, however, the full extent of the regulatory landscape of the Ikzf1 locus is not yet established. In this study, we combined epigenomics and transcription factor binding along with high-throughput enhancer assay and 4C-seq to prioritize an enhancer element located 120 kb upstream of the Ikzf1 gene. We found that deletion of the E120 enhancer resulted in a significant reduction of Ikzf1 mRNA. However, the epigenetic landscape and 3D topology of the locus were only slightly affected, highlighting the complexity of the regulatory landscape regulating the Ikzf1 locus.


Asunto(s)
Elementos de Facilitación Genéticos/fisiología , Regulación de la Expresión Génica/fisiología , Sitios Genéticos/fisiología , Factor de Transcripción Ikaros/biosíntesis , Animales , Línea Celular , Epigenómica , Genes Reporteros , Factor de Transcripción Ikaros/genética , Ratones , ARN Mensajero/biosíntesis , ARN Mensajero/genética
3.
Exp Hematol ; 86: 21-27.e2, 2020 06.
Artículo en Inglés | MEDLINE | ID: mdl-32437909

RESUMEN

Our previous study revealed that expression of G protein-coupled receptor 68 (GPR68) was upregulated in MDSL cells, a cell line representing myelodysplastic syndromes (MDS), in response to lenalidomide (LEN), and mediated a calcium/calpain proapoptotic pathway. Isx, a GPR68 agonist, enhanced the sensitivity to LEN in MDSL cells. The fact that Isx is not a U.S. Food and Drug Administration-approved drug prompts us to look for alternative candidates that could enhance the sensitivity of LEN in MDS as well as other hematologic malignancies, such as acute myeloid leukemia (AML). In the study described here, we found that regulator of calcineurin 1 (RCAN1), an endogenous inhibitor of calcineurin (CaN), was upregulated in MDSL cells in response to LEN, possibly through degradation of IKZF1. Consistently, cyclosporin (Cys), a pharmacological inhibitor of CaN, inhibited the activity of CaN and induced apoptosis in MDSL cells, indicating that CaN provided a prosurvival signal in MDSL cells. In addition, Cys enhanced the cytotoxic effect of LEN in MDS/AML cell lines as well as primary bone marrow cells from MDS patients and AML patient-derived xenograft models. Intriguingly, pretreatment with LEN reversed the suppressive effect of Cys on T-cell activation. Our study suggests a novel mechanism of action of LEN in mediating cytotoxicity in MDS/AML via upregulation of RCAN1, thus inhibiting the CaN prosurvival pathway. Our study also suggests that Cys enhances the sensitivity to LEN in MDS/AML cells without compromising T-cell activation.


Asunto(s)
Ciclosporina/farmacología , Resistencia a Antineoplásicos/efectos de los fármacos , Lenalidomida/farmacología , Leucemia Mieloide Aguda , Síndromes Mielodisplásicos , Animales , Apoptosis/efectos de los fármacos , Línea Celular Tumoral , Ciclosporina/agonistas , Proteínas de Unión al ADN/biosíntesis , Regulación Leucémica de la Expresión Génica/efectos de los fármacos , Humanos , Factor de Transcripción Ikaros/biosíntesis , Lenalidomida/agonistas , Leucemia Mieloide Aguda/tratamiento farmacológico , Leucemia Mieloide Aguda/metabolismo , Leucemia Mieloide Aguda/patología , Ratones , Ratones Endogámicos NOD , Proteínas Musculares/biosíntesis , Síndromes Mielodisplásicos/tratamiento farmacológico , Síndromes Mielodisplásicos/metabolismo , Síndromes Mielodisplásicos/patología , Proteínas de Neoplasias/biosíntesis , Regulación hacia Arriba/efectos de los fármacos , Ensayos Antitumor por Modelo de Xenoinjerto
5.
Immunol Invest ; 47(6): 632-642, 2018 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-29851536

RESUMEN

BACKGROUND: The control of auto-reactive cells is defective in rheumatoid arthritis (RA). Regulatory T (Treg) cells which play a key role in the modulation of immune responses have an impaired function in RA. Foxp3 is a master regulator of Treg cells which its expression is under the tight control of epigenetic mechanisms. In the current study, we analyzed the epigenetic modulation of the Foxp3 Treg-specific demethylated region (TSDR) and Helios gene expression to determine Treg cells alteration in RA patients. METHODS: We have recruited 20 newly diagnosed patients with RA and 41 healthy controls in our study. The measurement of Foxp3 and Helios gene expression was performed by the real-time PCR technique and the methylation level of TSDR was analyzed by bisulfite treatment and quantitative methylation-specific PCR (Q-MSP). RESULTS: We found that RA patients had significantly lower level of Foxp3 gene expression and TSDR demethylation compared to healthy subjects (P < 0.001 and P = 0.006, respectively). Inversely, the Helios gene expression was elevated significantly in RA patients group (P = 0.048). We also observed a significant correlation between Foxp3 and Helios gene expression (P = 0.016) as well as a significant correlation between FoxP3 expression and demethylation rate of TSDR (P = 0.010). CONCLUSION: Our results suggested that both epigenetic modifications and Helios gene expression may have important roles in the pathogenesis of RA through their effects on Foxp3 gene expression.


Asunto(s)
Artritis Reumatoide/genética , Metilación de ADN/genética , Factores de Transcripción Forkhead/genética , Regulación de la Expresión Génica/genética , Factor de Transcripción Ikaros/genética , Linfocitos T Reguladores/inmunología , Actinas/genética , Actinas/metabolismo , Adulto , Estudios de Casos y Controles , Epigénesis Genética/genética , Femenino , Factores de Transcripción Forkhead/metabolismo , Humanos , Factor de Transcripción Ikaros/biosíntesis , Masculino
6.
Haematologica ; 103(10): 1688-1697, 2018 10.
Artículo en Inglés | MEDLINE | ID: mdl-29954930

RESUMEN

The immunomodulatory drugs, lenalidomide and pomalidomide yield high response rates in multiple myeloma patients, but are associated with a high rate of thrombocytopenia and increased risk of secondary hematologic malignancies. Here, we demonstrate that the immunomodulatory drugs induce self-renewal of hematopoietic progenitors and upregulate megakaryocytic colonies by inhibiting apoptosis and increasing proliferation of early megakaryocytic progenitors via down-regulation of IKZF1. In this process, the immunomodulatory drugs degrade IKZF1 and subsequently down-regulate its binding partner, GATA1. This results in the decrease of GATA1 targets such as ZFPM1 and NFE2, leading to expansion of megakaryocytic progenitors with concomitant inhibition of maturation of megakaryocytes. The down-regulation of GATA1 further decreases CCND1 and increases CDKN2A expression. Overexpression of GATA1 abrogated the effects of the immunomodulatory drugs and restored maturation of megakaryocytic progenitors. Our data not only provide the mechanism for the immunomodulatory drugs induced thrombocytopenia but also help to explain the higher risk of secondary malignancies and long-term cytopenia induced by enhanced cell cycling and subsequent exhaustion of the stem cell pool.


Asunto(s)
Apoptosis/efectos de los fármacos , Regulación hacia Abajo/efectos de los fármacos , Células Madre Hematopoyéticas/metabolismo , Factor de Transcripción Ikaros/biosíntesis , Factores Inmunológicos/farmacología , Megacariocitos/metabolismo , Femenino , Células Madre Hematopoyéticas/citología , Humanos , Masculino , Megacariocitos/citología
7.
Front Immunol ; 9: 19, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29403500

RESUMEN

Human immunodeficiency virus type-1 (HIV-1) elite controllers (ELCs) represent a unique population that control viral replication in the absence of antiretroviral therapy (cART). It is well established that expression of multiple inhibitory receptors on CD8+ T cells is associated with HIV-1 disease progression. However, whether reduced co-expression of inhibitory receptors on CD4+ T cells is linked to natural viral control and slow HIV-1 disease progression remains undefined. Here, we report on the expression pattern of numerous measurable inhibitory receptors, associated with T cell exhaustion (programmed cell death-1, CTLA-4, and TIGIT), on different CD4+ T cell memory populations in ELCs and HIV-infected subjects with or without long-term cART. We found that the co-expression pattern of inhibitory receptors was significantly reduced in ELCs compared with HIV-1 cART-treated and viremic subjects, and similar to healthy controls. Markers associated with T cell exhaustion varied among different memory CD4+ T cell subsets and highest levels were found mainly on transitional memory T cells. CD4+ T cells co-expressing all inhibitory markers were positively correlated to T cell activation (CD38+ HLA-DR+) as well as the transcription factors Helios and FoxP3. Finally, clinical parameters such as CD4 count, HIV-1 viral load, and the CD4/CD8 ratio all showed significant associations with CD4+ T cell exhaustion. We demonstrate that ELCs are able to maintain lower levels of CD4+ T cell exhaustion despite years of ongoing viral replication compared with successfully cART-treated subjects. Our findings suggest that ELCs harbor a "healthy" state of inhibitory receptor expression on CD4+ T cells that might play part in maintenance of their control status.


Asunto(s)
Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD8-positivos/inmunología , VIH-1/inmunología , Memoria Inmunológica/inmunología , Activación de Linfocitos/inmunología , ADP-Ribosil Ciclasa 1/biosíntesis , Recuento de Linfocito CD4 , Relación CD4-CD8 , Antígeno CTLA-4/biosíntesis , Factores de Transcripción Forkhead/biosíntesis , Antígenos HLA-DR/biosíntesis , Humanos , Factor de Transcripción Ikaros/biosíntesis , Glicoproteínas de Membrana/biosíntesis , Receptor de Muerte Celular Programada 1/biosíntesis , Receptores Inmunológicos/biosíntesis , Carga Viral , Replicación Viral
8.
Pediatr Blood Cancer ; 64(10)2017 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-28371317

RESUMEN

BACKGROUND: A high-level expression of the CRLF2 gene is frequent in precursor B-cell acute lymphoblastic leukemia (pB-ALL) and can be caused by different genetic aberrations. The presence of the most frequent alteration, the P2RY8/CRLF2 fusion, was shown to be associated with a high relapse incidence in children treated according to ALL-Berlin-Frankfurt-Münster (BFM) protocols, which is poorly understood. Moreover, the frequency of other alterations has not been systematically analyzed yet. PROCEDURE: CRLF2 mRNA expression and potential genetic aberrations causing a CRLF2 high expression were prospectively assessed in 1,105 patients treated according to the Associazione Italiana Ematologia Oncologia Pediatrica (AIEOP)-BFM ALL 2009 protocol. Additionally, we determined copy number alterations in selected B-cell differentiation genes for all CRLF2 high-expressing pB-ALL cases, as well as JAK2 and CRLF2 mutations. RESULTS: A CRLF2 high expression was detected in 26/178 (15%) T-cell acute lymphoblastic leukemia (T-ALL) cases, 21 of them (81%) had been stratified as high-risk patients by treatment response. In pB-ALL, a CRLF2 high expression was determined in 91/927 (10%) cases; the P2RY8/CRLF2 rearrangement in 44/91 (48%) of them, supernumerary copies of CRLF2 in 18/91 (20%), and, notably, the IGH/CRLF2 translocation was detected in 16/91 (18%). Remarkably, 7 of 16 (44%) patients with IGH/CRLF2 translocation had already relapsed. P2RY8/CRLF2- and IGH/CRLF2-positive samples (70 and 94%, respectively) were characterized by a high frequency of additional deletions in B-cell differentiation genes such as IKZF1 or PAX5. CONCLUSION: Our data suggest that this high frequency of genetic aberrations in the context of a high CRLF2 expression could contribute to the high risk of relapse in P2RY8/CRLF2- and IGH/CRLF2-positive ALL.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/administración & dosificación , Regulación Leucémica de la Expresión Génica/efectos de los fármacos , Leucemia-Linfoma Linfoblástico de Células Precursoras/tratamiento farmacológico , Leucemia-Linfoma Linfoblástico de Células Precursoras/metabolismo , Receptores de Citocinas/biosíntesis , Adolescente , Asparaginasa/administración & dosificación , Niño , Preescolar , Daunorrubicina/administración & dosificación , Femenino , Reordenamiento Génico , Humanos , Factor de Transcripción Ikaros/biosíntesis , Factor de Transcripción Ikaros/genética , Lactante , Masculino , Proteínas de Fusión Oncogénica/biosíntesis , Proteínas de Fusión Oncogénica/genética , Factor de Transcripción PAX5/biosíntesis , Factor de Transcripción PAX5/genética , Leucemia-Linfoma Linfoblástico de Células Precursoras/genética , Prednisona/administración & dosificación , Receptores de Citocinas/genética , Receptores Purinérgicos P2Y/biosíntesis , Receptores Purinérgicos P2Y/genética , Vincristina/administración & dosificación
9.
J Cancer Res Clin Oncol ; 142(11): 2375-82, 2016 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-27576507

RESUMEN

PURPOSE: Helios belongs to Ikaros family, which plays an important role in the cell-fate decision and control cell proliferation; abnormal expressions in leukemia are associated with poor prognosis. In this study, we investigated the Helios expression between Helicobacter pylori infection and prognosis in gastric cancer patients. METHODS: A total of 67 gastric cancer patients who received partial or full gastrectomies were enrolled. Helios expression by immunohistochemistry and mRNA was investigated with the clinical stage, Helicobacter pylori infection, CD4 expression, FoxP3 expression and prognosis. RESULTS: From the immunohistochemistry stain, we found that the Helios was expressed in both cancer cell and tumor-infiltrated lymphocytes. The high expression of Helios in gastric tumor cells had a better median overall survival rate in gastric cancer patients (50.7 ± 3.2 vs. 34.1 ± 4.9 months; P = 0.015), Helicobacter pylori-infected patients (51.1 ± 3.5 vs. 30.4 ± 5.1 months; P = 0.007) and advanced gastric cancer patients (42.1 ± 5.5 vs. 23.2 ± 4.8 months; P = 0.043). From multivariate analysis, the Helios expression in gastric tumor cells was an independent factor to predict better survival in all gastric cancers (HR = 2.78; 95 % confidence interval [CI], 1.09-7.09; P = 0.032) and advanced gastric cancer patients (HR = 2.85; 95 % confidence interval [CI], 1.00-8.13; P = 0.03). CONCLUSIONS: Higher expression of Helios in gastric tumor cells predicts better survival in gastric cancer patients, especially for Helicobacter pylori-infected and advanced-stage gastric cancer patients.


Asunto(s)
Factor de Transcripción Ikaros/biosíntesis , Neoplasias Gástricas/metabolismo , Anciano , Anciano de 80 o más Años , Biomarcadores de Tumor/biosíntesis , Femenino , Infecciones por Helicobacter/metabolismo , Infecciones por Helicobacter/patología , Helicobacter pylori/aislamiento & purificación , Humanos , Inmunohistoquímica , Estimación de Kaplan-Meier , Linfocitos Infiltrantes de Tumor/metabolismo , Masculino , Persona de Mediana Edad , Estadificación de Neoplasias , Pronóstico , Neoplasias Gástricas/microbiología , Neoplasias Gástricas/patología
10.
PLoS One ; 11(6): e0157767, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27315244

RESUMEN

The Ikaros transcription factor is a tumor suppressor that is also important for lymphocyte development. How post-translational modifications influence Ikaros function remains partially understood. We show that Ikaros undergoes sumoylation in developing T cells that correspond to mono-, bi- or poly-sumoylation by SUMO1 and/or SUMO2/3 on three lysine residues (K58, K240 and K425). Sumoylation occurs in the nucleus and requires DNA binding by Ikaros. Sumoylated Ikaros is less effective than unsumoylated forms at inhibiting the expansion of murine leukemic cells, and Ikaros sumoylation is abundant in human B-cell acute lymphoblastic leukemic cells, but not in healthy peripheral blood leukocytes. Our results suggest that sumoylation may be important in modulating the tumor suppressor function of Ikaros.


Asunto(s)
Proteínas de Unión al ADN/genética , Factor de Transcripción Ikaros/genética , Leucemia-Linfoma Linfoblástico de Células Precursoras/genética , Transcripción Genética , Animales , Linfocitos B/patología , Línea Celular Tumoral , Proteínas de Unión al ADN/biosíntesis , Regulación Neoplásica de la Expresión Génica , Humanos , Factor de Transcripción Ikaros/biosíntesis , Linfocitos/patología , Ratones , Leucemia-Linfoma Linfoblástico de Células Precursoras/patología , Procesamiento Proteico-Postraduccional/genética , Proteína SUMO-1/genética , Sumoilación/genética , Proteínas Supresoras de Tumor
11.
PLoS One ; 11(2): e0148826, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-26866695

RESUMEN

CD4+ T regulatory cells (Tregs) comprise a heterogeneous population of cells the regulate immune responses and prevent autoimmunity. Most reports on human Tregs are derived from studies of peripheral blood, although Tregs mainly exert their functions in the periphery. Here we performed a detailed analysis of Tregs in the human upper airway mucosa under non-inflammatory conditions, and found that 10% of all CD4+ T cells expressed the transcription factor FOXP3 and the memory marker CD45RO, as well as high levels of CTLA-4. The majority of FOXP3+CD4+ T cells co-expressed the transcription factor Helios and produced very little cytokines, compatible with being thymus-derived Tregs. FOXP3+Helios-CD4+ T cells were more heterogeneous. A mean of 24% produced the immunomodulatory cytokine IL-10, whereas a large fraction also produced IL-2, IFN-µ or IL-17. A significant population (6%) of FOXP3-negative T cells also produced IL-10, usually in combination with IFN-µ. Together, we found that CD4+ T cells in the upper airways differed functionally from their counterparts in peripheral blood, including higher expression of IL-10. Moreover, our findings suggest that several subsets of CD4+ T cells with functionally distinct regulatory properties reside in the upper airway mucosa which should be taken into account when targeting Tregs for therapy.


Asunto(s)
Linfocitos T CD4-Positivos/citología , Mucosa Nasal/metabolismo , Linfocitos T Reguladores/citología , Adolescente , Adulto , Antígenos CD/biosíntesis , Antígenos CD/metabolismo , Femenino , Citometría de Flujo , Factores de Transcripción Forkhead/biosíntesis , Factores de Transcripción Forkhead/metabolismo , Humanos , Factor de Transcripción Ikaros/biosíntesis , Factor de Transcripción Ikaros/metabolismo , Integrina alfa2/biosíntesis , Integrina alfa2/metabolismo , Interferones/biosíntesis , Interferones/metabolismo , Interleucina-10/biosíntesis , Interleucina-10/metabolismo , Interleucina-17/biosíntesis , Interleucina-17/metabolismo , Interleucina-2/biosíntesis , Interleucina-2/metabolismo , Antígenos Comunes de Leucocito/biosíntesis , Antígenos Comunes de Leucocito/metabolismo , Leucocitos Mononucleares/citología , Masculino , Persona de Mediana Edad , Fenotipo , Sistema Respiratorio/patología , Adulto Joven , Proteína del Gen 3 de Activación de Linfocitos
12.
Oncogene ; 35(14): 1797-810, 2016 Apr 07.
Artículo en Inglés | MEDLINE | ID: mdl-26119939

RESUMEN

Primary effusion lymphoma (PEL) is an aggressive type of non-Hodgkin lymphoma localized predominantly in body cavities. Kaposi's sarcoma-associated herpes virus (KSHV) is the causative agent of PEL. PEL is an incurable malignancy and has extremely poor prognosis when treated with conventional chemotherapy. Immunomodulatory drugs (IMiDs) lenalidomide and pomalidomide are Food and Drug Administration-approved drugs for the treatment of various ailments. IMiDs display pronounced antiproliferative effect against majority of PEL cell lines within their clinically achievable concentrations, by arresting cells at G0/G1 phase of cell cycle and without any induction of KSHV lytic cycle reactivation. Although microarray examination of PEL cells treated with lenalidomide revealed activation of interferon (IFN) signaling, blocking the IFN pathway did not block the anti-PEL activity of IMiDs. The anti-PEL effects of IMiDs involved cereblon-dependent suppression of IRF4 and rapid degradation of IKZF1, but not IKZF3. Small hairpin RNA-mediated knockdown of MYC enhanced the cytotoxicity of IMiDs. Bromodomain (BRD) and extra-terminal domain (BET) proteins are epigenetic readers, which perform a vital role in chromatin remodeling and transcriptional regulation. BRD4, a widely expressed transcriptional coactivator, belongs to the BET family of proteins, which has been shown to co-occupy the super enhancers associated with MYC. Specific BRD4 inhibitors were developed, which suppress MYC transcriptionally. Lenalidomide displayed synergistic cytotoxicity with several structurally distinct BRD4 inhibitors (JQ-1, IBET151 and PFI-1). Furthermore, combined administration of lenalidomide and BRD4 inhibitor JQ-1 significantly increased the survival of PEL bearing NOD-SCID mice in an orthotopic xenograft model as compared with either agent alone. These results provide compelling evidence for clinical testing of IMiDs alone and in combination with BRD4 inhibitors for PEL.


Asunto(s)
Factor de Transcripción Ikaros/genética , Factores Reguladores del Interferón/genética , Linfoma de Efusión Primaria/tratamiento farmacológico , Proteínas Nucleares/genética , Péptido Hidrolasas/genética , Proteínas Proto-Oncogénicas c-myc/genética , Factores de Transcripción/genética , Proteínas Adaptadoras Transductoras de Señales , Animales , Apoptosis/efectos de los fármacos , Azepinas/administración & dosificación , Proteínas de Ciclo Celular , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Sinergismo Farmacológico , Técnicas de Silenciamiento del Gen , Humanos , Factor de Transcripción Ikaros/biosíntesis , Factores Inmunológicos/administración & dosificación , Factores Reguladores del Interferón/biosíntesis , Lenalidomida , Linfoma de Efusión Primaria/genética , Linfoma de Efusión Primaria/patología , Ratones , Proteínas Proto-Oncogénicas c-myc/antagonistas & inhibidores , Talidomida/administración & dosificación , Talidomida/análogos & derivados , Triazoles/administración & dosificación , Ubiquitina-Proteína Ligasas , Ensayos Antitumor por Modelo de Xenoinjerto
13.
Int J Oncol ; 47(6): 2082-90, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-26460798

RESUMEN

The functions of different regulatory T cell (Treg) types in cancer progression are unclear. Recently, expression of the transcription factor Helios was proposed as a marker for natural (non-induced) Tregs. The present study investigated the clinical significance of Helios expression in patients with non-small cell lung cancer (NSCLC). We enrolled 64 patients with NSCLC, of whom 45 were treated surgically and 19 received chemotherapy because of advanced/recurrent disease. Their peripheral blood mononuclear cells were examined by flow cytometry. From the 45 surgery patients, we matched 9 patients with recurrent disease with 9 stage-matched patients without recurrence (n=18), compared their specimens immunohistochemically for tumor infiltrating lymphocytes (TILs) and analyzed these data against clinicopathological factors. Helios expression in Foxp3+ Tregs was 47.5±13.3% in peripheral blood and 18.1±13.4% in tumor specimens. Percentage of Helios- Tregs among CD4+ T cells were significantly higher in the cancer patients (2.4%), especially those with stage IA disease (2.6%) than in healthy donors (1.5%; P<0.001). Patients with low levels of Helios expression in Tregs among their TILs had significantly poorer survival (P=0.038). Helios- Tregs may affect immune suppression, even in early stage NSCLC; they could also be a useful prognostic biomarker in patients with NSCLC, and possibly a novel cancer immunotherapy target.


Asunto(s)
Carcinoma de Pulmón de Células no Pequeñas/inmunología , Neoplasias Pulmonares/inmunología , Linfocitos Infiltrantes de Tumor/inmunología , Subgrupos de Linfocitos T/inmunología , Linfocitos T Reguladores/inmunología , Anciano , Carcinoma de Pulmón de Células no Pequeñas/mortalidad , Carcinoma de Pulmón de Células no Pequeñas/patología , Femenino , Citometría de Flujo , Factores de Transcripción Forkhead/biosíntesis , Factores de Transcripción Forkhead/inmunología , Humanos , Factor de Transcripción Ikaros/biosíntesis , Factor de Transcripción Ikaros/inmunología , Inmunohistoquímica , Inmunofenotipificación , Estimación de Kaplan-Meier , Neoplasias Pulmonares/mortalidad , Neoplasias Pulmonares/patología , Masculino , Persona de Mediana Edad
14.
J Immunol ; 194(11): 5282-93, 2015 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-25917092

RESUMEN

Expansion of Ag-specific naturally occurring regulatory T cells (nTregs) is required to obtain sufficient numbers of cells for cellular immunotherapy. In this study, different allogeneic stimuli were studied for their capacity to generate functional alloantigen-specific nTregs. A highly enriched nTreg fraction (CD4(+)CD25(bright)CD127(-) T cells) was alloantigen-specific expanded using HLA-mismatched immature, mature monocyte-derived dendritic cells (moDCs), or PBMCs. The allogeneic mature moDC-expanded nTregs were fully characterized by analysis of the demethylation status within the Treg-specific demethylation region of the FOXP3 gene and the expression of both protein and mRNA of FOXP3, HELIOS, CTLA4, and cytokines. In addition, the Ag-specific suppressive capacity of these expanded nTregs was tested. Allogeneic mature moDCs and skin-derived DCs were superior in inducing nTreg expansion compared with immature moDCs or PBMCs in an HLA-DR- and CD80/CD86-dependent way. Remarkably, the presence of exogenous IL-15 without IL-2 could facilitate optimal mature moDC-induced nTreg expansion. Allogeneic mature moDC-expanded nTregs were at low ratios (<1:320), potent suppressors of alloantigen-induced proliferation without significant suppression of completely HLA-mismatched, Ag-induced proliferation. Mature moDC-expanded nTregs were highly demethylated at the Treg-specific demethylation region within the FOXP3 gene and highly expressed of FOXP3, HELIOS, and CTLA4. A minority of the expanded nTregs produced IL-10, IL-2, IFN-γ, and TNF-α, but few IL-17-producing nTregs were found. Next-generation sequencing of mRNA of moDC-expanded nTregs revealed a strong induction of Treg-associated mRNAs. Human allogeneic mature moDCs are highly efficient stimulator cells, in the presence of exogenous IL-15, for expansion of stable alloantigen-specific nTregs with superior suppressive function.


Asunto(s)
Células Dendríticas/efectos de los fármacos , Interleucina-15/farmacología , Linfocitos T Reguladores/inmunología , Traslado Adoptivo , Antígeno B7-1/metabolismo , Antígeno B7-2/metabolismo , Secuencia de Bases , Antígenos CD4/metabolismo , Antígeno CTLA-4/biosíntesis , Antígeno CTLA-4/genética , Diferenciación Celular/inmunología , Proliferación Celular , Células Cultivadas , Metilación de ADN , Células Dendríticas/inmunología , Factores de Transcripción Forkhead/biosíntesis , Factores de Transcripción Forkhead/genética , Antígenos HLA-DR/inmunología , Humanos , Factor de Transcripción Ikaros/biosíntesis , Factor de Transcripción Ikaros/genética , Interferón gamma/biosíntesis , Interleucina-10/biosíntesis , Interleucina-15/inmunología , Interleucina-17/biosíntesis , Interleucina-2/biosíntesis , Interleucina-2/metabolismo , Subunidad alfa del Receptor de Interleucina-2/metabolismo , Subunidad alfa del Receptor de Interleucina-7/metabolismo , Análisis de Secuencia de ARN , Piel/citología , Piel/inmunología , Factor de Necrosis Tumoral alfa/biosíntesis
15.
Oncol Rep ; 33(3): 1319-25, 2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-25524659

RESUMEN

T-cell acute lymphoblastic leukemia (T-ALL) is an aggressive neoplastic disorder of immature hematopoietic precursors committed to T-cell lineage. T-ALL accounts for ~15% of pediatric ALL cases and is prone to early relapse. With new and improved treatment protocols, the prognosis of T-ALL has improved particularly in children; however, the outcome of relapsed T-ALL cases remains poor. The AIOLOS gene is necessary to control lymphocyte differentiation and may be a potential target of T-ALL therapy. In the present study, Jurkat cells were divided into three groups: untransfected (UT) control, lentiviral vector control (Lenti-Mock) and AIOLOS-overexpressing (Lenti-AIOLOS) groups. Lenti-AIOLOS Jurkat cells were constructed by lentiviral transduction; cell cycle analysis, apoptosis and cytotoxicity assays were then performed to evaluate the effects of AIOLOS on cell cycle distribution, apoptosis and cell chemosensitivity to etoposide of Jurkat cells in vitro. Moreover, the expression levels of genes associated with apoptosis and cell cycle were investigated by quantitative reverse transcription-polymerase chain reaction. Results showed that the percentage of Jurkat cells in the G0/G1 phase increased from 71.5 (UT) to 85.4% (Lenti-AIOLOS; P<0.05), yet the percentage of cells in the S-phase decreased from 15.1 (UT) to 11.6% (Lenti­AIOLOS; P<0.05). The percentage of total apoptotic cells was significantly increased in the AIOLOS-transfected Jurkat cells (21.93%) compared with this percentage in the Lenti-Mock (13.35%) or the UT group (13.30%; P<0.05). Consistent with these results, AIOLOS overexpression induced P21 and P27 upregulation and CCND3 and SKP2 downregulation. Furthermore, AIOLOS overexpression synergistically increased the cytotoxic effects of etoposide and downregulated NF-κB expression. Our findings revealed that lentivirus-mediated AIOLOS overexpression in Jurkat cells induced cell apoptosis, arrested the cell cycle at the G0/G1 phase, and synergistically increased the sensitivity of Jurkat cells to etoposide by inhibiting NF-κB activity.


Asunto(s)
Apoptosis/genética , Etopósido/farmacología , Puntos de Control de la Fase G1 del Ciclo Celular/genética , Factor de Transcripción Ikaros/biosíntesis , Leucemia-Linfoma Linfoblástico de Células T Precursoras/tratamiento farmacológico , Leucemia-Linfoma Linfoblástico de Células T Precursoras/genética , Antineoplásicos Fitogénicos/farmacología , Diferenciación Celular/inmunología , Línea Celular Tumoral , Ciclina D3/biosíntesis , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/biosíntesis , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/genética , Inhibidor p27 de las Quinasas Dependientes de la Ciclina/biosíntesis , Inhibidor p27 de las Quinasas Dependientes de la Ciclina/genética , Resistencia a Antineoplásicos , Humanos , Factor de Transcripción Ikaros/genética , Células Jurkat , Lentivirus/genética , FN-kappa B/biosíntesis , FN-kappa B/metabolismo , Leucemia-Linfoma Linfoblástico de Células T Precursoras/patología , Proteínas Quinasas Asociadas a Fase-S/biosíntesis , Linfocitos T/citología , Linfocitos T/metabolismo , Regulación hacia Arriba
16.
Clin Exp Immunol ; 180(1): 108-17, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25425428

RESUMEN

Forkhead box protein 3 (FoxP3)(+) regulatory T cells (Tregs ) are important not only in regulating the development of autoimmune conditions, but also in chronic infectious diseases. Given their cardinal function in suppressing immune activation, research has focused upon whether they play a detrimental role in chronic infections, particularly HIV. While the role of Tregs in HIV has been investigated intensively, it remains an unresolved topic. However, it is generally accepted that Tregs are susceptible to HIV infection and are preferentially preserved over conventional CD4(+) T cells. It is unknown whether the peripheral-induced or the thymic-derived Tregs are more susceptible to HIV cytotoxicity. It has been recognized that Tregs can be segregated into two subsets based on Helios expression, with the vast majority being Helios(+) . This study examines the impact of HIV infection on total Tregs and their Helios subsets in a perinatal-acquired HIV-infected paediatric population. The finding indicates a selective expansion or survival of Tregs in association with CD4 depletion and increased viraemia. The Helios(+) and Helios(-) subsets within Tregs appear to be equally affected. However, the Helios(+) Tregs seem to be more preserved in patients with low CD4(+) ≤ 25% and detectable plasma HIV RNA >20 copies/ml. In this group, the frequencies of Tregs are increased, but their numbers appear insufficient to restrain immune activation. In conclusion, our findings suggest that both Helios subsets of Tregs are susceptible to HIV infection and are preferentially preserved compared to conventional CD4(+) T cells.


Asunto(s)
Factores de Transcripción Forkhead/inmunología , Regulación de la Expresión Génica/inmunología , Infecciones por VIH/inmunología , VIH-1/inmunología , Factor de Transcripción Ikaros/inmunología , Linfocitos T Reguladores/inmunología , Adolescente , Adulto , Niño , Preescolar , Enfermedad Crónica , Femenino , Factores de Transcripción Forkhead/biosíntesis , Infecciones por VIH/sangre , Infecciones por VIH/congénito , Infecciones por VIH/patología , VIH-1/metabolismo , Humanos , Factor de Transcripción Ikaros/biosíntesis , Lactante , Masculino , ARN Viral/sangre , ARN Viral/inmunología , Linfocitos T Reguladores/metabolismo , Linfocitos T Reguladores/patología , Timo/inmunología , Timo/metabolismo , Timo/patología
17.
J Immunol ; 194(1): 68-75, 2015 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-25480564

RESUMEN

Upregulation of IL-17 immunity and detrimental effects of IL-17 on human islets have been implicated in human type 1 diabetes. In animal models, the plasticity of Th1/Th17 cells contributes to the development of autoimmune diabetes. In this study, we demonstrate that the upregulation of the IL-17 pathway and Th1/Th17 plasticity in peripheral blood are markers of advanced ß cell autoimmunity and impaired ß cell function in human type 1 diabetes. Activated Th17 immunity was observed in the late stage of preclinical diabetes in children with ß cell autoimmunity and impaired glucose tolerance, but not in children with early ß cell autoimmunity. We found an increased ratio of IFN-γ/IL-17 expression in Th17 cells in children with advanced ß cell autoimmunity, which correlated with HbA1c and plasma glucose concentrations in an oral glucose tolerance test, and thus impaired ß cell function. Low expression of Helios was seen in Th17 cells, suggesting that Th1/Th17 cells are not converted thymus-derived regulatory T cells. Our results suggest that the development of Th1/Th17 plasticity may serve as a biomarker of disease progression from ß cell autoantibody positivity to type 1 diabetes. These data in human type 1 diabetes emphasize the role of Th1/Th17 plasticity as a potential contributor to tissue destruction in autoimmune conditions.


Asunto(s)
Diabetes Mellitus Tipo 1/inmunología , Intolerancia a la Glucosa/inmunología , Células Secretoras de Insulina/inmunología , Interleucina-17/biosíntesis , Células TH1/inmunología , Células Th17/inmunología , Autoanticuerpos/inmunología , Autoinmunidad/inmunología , Biomarcadores/sangre , Células Cultivadas , Niño , Preescolar , Progresión de la Enfermedad , Femenino , Humanos , Factor de Transcripción Ikaros/biosíntesis , Interferón gamma/biosíntesis , Interleucina-17/inmunología , Interleucina-9/biosíntesis , Interleucina-9/inmunología , Masculino , Linfocitos T Reguladores/inmunología , Regulación hacia Arriba/inmunología
18.
EMBO Rep ; 15(9): 991-9, 2014 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-25074018

RESUMEN

Regulatory T cells (Tregs) control autoreactive T cells by inhibiting activation-induced proliferation and cytokine expression. The molecular mechanisms responsible for the inactivation of effector T cells by Tregs remain yet to be fully characterized. We report that T-helper cells stimulated in the presence of Tregs quickly activate NFAT1 and have increased NFAT1-dependent expression of the transcription repressor Ikaros. NFAT1 deficiency or dominant-negative Ikaros compromises Treg-mediated inhibition of T-helper cells in vitro and in vivo. Thus, our results place NFAT-dependent mechanisms as general regulators of T-cell tolerance and show that Treg-mediated suppression of T-helper cells results from the activation of NFAT-regulated gene expression.


Asunto(s)
Linfocitos T CD4-Positivos/inmunología , Factor de Transcripción Ikaros/biosíntesis , Factores de Transcripción NFATC/inmunología , Linfocitos T Reguladores/inmunología , Animales , Linfocitos T CD4-Positivos/metabolismo , Proliferación Celular , Citocinas/biosíntesis , Regulación de la Expresión Génica , Factor de Transcripción Ikaros/genética , Activación de Linfocitos/inmunología , Ratones , Factores de Transcripción NFATC/biosíntesis , Linfocitos T Colaboradores-Inductores/inmunología , Linfocitos T Reguladores/metabolismo
19.
PLoS Genet ; 10(2): e1004167, 2014 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-24586197

RESUMEN

Retroviral insertional mutagenesis (RIM) is a powerful tool for cancer genomics that was combined in this study with deep sequencing (RIM/DS) to facilitate a comprehensive analysis of lymphoma progression. Transgenic mice expressing two potent collaborating oncogenes in the germ line (CD2-MYC, -Runx2) develop rapid onset tumours that can be accelerated and rendered polyclonal by neonatal Moloney murine leukaemia virus (MoMLV) infection. RIM/DS analysis of 28 polyclonal lymphomas identified 771 common insertion sites (CISs) defining a 'progression network' that encompassed a remarkably large fraction of known MoMLV target genes, with further strong indications of oncogenic selection above the background of MoMLV integration preference. Progression driven by RIM was characterised as a Darwinian process of clonal competition engaging proliferation control networks downstream of cytokine and T-cell receptor signalling. Enhancer mode activation accounted for the most efficiently selected CIS target genes, including Ccr7 as the most prominent of a set of chemokine receptors driving paracrine growth stimulation and lymphoma dissemination. Another large target gene subset including candidate tumour suppressors was disrupted by intragenic insertions. A second RIM/DS screen comparing lymphomas of wild-type and parental transgenics showed that CD2-MYC tumours are virtually dependent on activation of Runx family genes in strong preference to other potent Myc collaborating genes (Gfi1, Notch1). Ikzf1 was identified as a novel collaborating gene for Runx2 and illustrated the interface between integration preference and oncogenic selection. Lymphoma target genes for MoMLV can be classified into (a) a small set of master regulators that confer self-renewal; overcoming p53 and other failsafe pathways and (b) a large group of progression genes that control autonomous proliferation in transformed cells. These findings provide insights into retroviral biology, human cancer genetics and the safety of vector-mediated gene therapy.


Asunto(s)
Genes myb/genética , Linfoma/genética , Virus de la Leucemia Murina de Moloney/genética , Mutagénesis Insercional/genética , Proteína p53 Supresora de Tumor/genética , Animales , Carcinogénesis , Perfilación de la Expresión Génica , Regulación Neoplásica de la Expresión Génica , Redes Reguladoras de Genes , Células Germinativas , Secuenciación de Nucleótidos de Alto Rendimiento , Humanos , Factor de Transcripción Ikaros/biosíntesis , Factor de Transcripción Ikaros/genética , Linfoma/patología , Linfoma/virología , Ratones
20.
Cancer Sci ; 104(8): 1097-106, 2013 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-23600753

RESUMEN

Molecular abnormalities involved in the multistep leukemogenesis of adult T-cell leukemia (ATL) remain to be clarified. Based on our integrated database, we focused on the expression patterns and levels of Ikaros family genes, Ikaros, Helios, and Aiolos, in ATL patients and HTLV-1 carriers. The results revealed profound deregulation of Helios expression, a pivotal regulator in the control of T-cell differentiation and activation. The majority of ATL samples (32/37 cases) showed abnormal splicing of Helios expression, and four cases did not express Helios. In addition, novel genomic loss in Helios locus was observed in 17/168 cases. We identified four ATL-specific short Helios isoforms and revealed their dominant-negative function. Ectopic expression of ATL-type Helios isoform as well as knockdown of normal Helios or Ikaros promoted T-cell growth. Global mRNA profiling and pathway analysis showed activation of several signaling pathways important for lymphocyte proliferation and survival. These data provide new insights into the molecular involvement of Helios function in the leukemogenesis and phenotype of ATL cells, indicating that Helios deregulation is one of the novel molecular hallmarks of ATL.


Asunto(s)
Factor de Transcripción Ikaros/biosíntesis , Leucemia-Linfoma de Células T del Adulto/metabolismo , Leucemia-Linfoma de Células T del Adulto/patología , Linfocitos T/metabolismo , Linfocitos T/patología , Procesos de Crecimiento Celular/fisiología , Línea Celular , Línea Celular Tumoral , Citoplasma/genética , Citoplasma/metabolismo , Exones , Células HEK293 , Células HeLa , Virus Linfotrópico T Tipo 1 Humano/genética , Virus Linfotrópico T Tipo 1 Humano/metabolismo , Humanos , Factor de Transcripción Ikaros/genética , Factor de Transcripción Ikaros/metabolismo , Leucemia-Linfoma de Células T del Adulto/genética , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Transducción de Señal/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA