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












Base de datos
Intervalo de año de publicación
1.
Immunol Res ; 69(3): 275-284, 2021 06.
Artículo en Inglés | MEDLINE | ID: mdl-33959834

RESUMEN

Destabilizing and reprogramming regulatory T (Treg) cells have become a potential strategy to treat tumor. Mounting evidence indicates that the transcription factor Helios is required for the stable differentiation of Treg lineage. Hence, we investigated whether Helios suppression could be a potential treatment option for pancreatic cancer patients. We found that Helios+ cells were predominantly in Foxp3+ Treg cells. By contrast, Foxp3+ Treg cells can be Helios+ or Helios-, but the level of Foxp3 expression was significantly higher in Helios+Foxp3+ Treg cells than in Helios-Foxp3+ Treg cells. Resected pancreatic tumors were highly enriched with both Helios+Foxp3+ Treg cells and Helios-Foxp3+ Treg cells. Also, the proportion of Helios+ cells in total Foxp3+ Treg cells was significantly higher in peripheral blood mononuclear cells (PBMCs) of patients than in PBMCs of healthy controls and further increased in patient tumors. Using shRNA, we knocked down Helios expression without significant downregulation of Foxp3. After Helios knockdown, CD4+CD25+CD127- Treg cells presented significantly lower levels of TGF-ß secretion, lower levels of IL-10 secretion, and higher levels of IFN-γ secretion. In addition, Helios shRNA-transfected CD4+CD25+CD127- Treg cells presented lower capacity to inhibit CD4+CD25-CD127+ T conventional cell proliferation than control shRNA-transfected CD4+CD25+CD127- Treg cells. Of note, CD4+CD25+CD127- Treg cells from pancreatic cancer patients demonstrated higher TGF-ß expression and higher suppression capacity than the cells from healthy controls. Overall, these results suggest that in pancreatic cancer patients, Helios may serve as a candidate to suppress Treg function, which could be used as a target to treat pancreatic cancer.


Asunto(s)
Factor de Transcripción Ikaros/metabolismo , Neoplasias Pancreáticas/inmunología , Linfocitos T Reguladores/inmunología , Escape del Tumor/inmunología , Anciano , Estudios de Casos y Controles , Células Cultivadas , Femenino , Técnicas de Silenciamiento del Gen , Voluntarios Sanos , Humanos , Factor de Transcripción Ikaros/análisis , Factor de Transcripción Ikaros/antagonistas & inhibidores , Factor de Transcripción Ikaros/genética , Masculino , Persona de Mediana Edad , Neoplasias Pancreáticas/sangre , Neoplasias Pancreáticas/tratamiento farmacológico , Cultivo Primario de Células , Linfocitos T Reguladores/efectos de los fármacos , Linfocitos T Reguladores/metabolismo , Escape del Tumor/efectos de los fármacos
2.
Front Immunol ; 12: 783282, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-35003100

RESUMEN

Treg therapies are being tested in clinical trials in transplantation and autoimmune diseases, however, the impact of inflammation on Tregs remains controversial. We challenged human Tregs ex-vivo with pro-inflammatory cytokines IL-6 and TNFα and observed greatly enhanced proliferation stimulated by anti-CD3 and anti-CD28 (aCD3/28) beads or CD28 superagonist (CD28SA). The cytokine-exposed Tregs maintained high expression of FOXP3 and HELIOS, demethylated FOXP3 enhancer, and low IFNγ, IL-4, and IL-17 secretion. Blocking TNF receptor using etanercept or deletion of TNF receptor 2 using CRISPR/Cas9 blunted Treg proliferation and attenuated FOXP3 and HELIOS expression. These results prompted us to consider using CD28SA together with IL-6 and TNFα without aCD3/28 beads (beadless) as an alternative protocol for therapeutic Treg manufacturing. Metabolomics profiling revealed more active glycolysis and oxidative phosphorylation, increased energy production, and higher antioxidant potential during beadless Treg expansion. Finally, beadless expanded Tregs maintained suppressive functions in vitro and in vivo. These results demonstrate that human Tregs positively respond to proinflammatory cytokines with enhanced proliferation without compromising their lineage identity or function. This property can be harnessed for therapeutic Treg manufacturing.


Asunto(s)
Enfermedad Injerto contra Huésped/terapia , Inmunoterapia Adoptiva/métodos , Interleucina-6/metabolismo , Linfocitos T Reguladores/inmunología , Factor de Necrosis Tumoral alfa/metabolismo , Adulto , Anciano , Animales , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Modelos Animales de Enfermedad , Etanercept/farmacología , Femenino , Factores de Transcripción Forkhead/análisis , Factores de Transcripción Forkhead/metabolismo , Enfermedad Injerto contra Huésped/inmunología , Voluntarios Sanos , Humanos , Factor de Transcripción Ikaros/análisis , Factor de Transcripción Ikaros/metabolismo , Masculino , Ratones , Persona de Mediana Edad , Cultivo Primario de Células , Linfocitos T Reguladores/metabolismo , Linfocitos T Reguladores/trasplante , Trasplante Heterólogo/efectos adversos , Factor de Necrosis Tumoral alfa/antagonistas & inhibidores , Adulto Joven
4.
Leukemia ; 33(8): 2022-2033, 2019 08.
Artículo en Inglés | MEDLINE | ID: mdl-30992504

RESUMEN

Monitoring tumour burden and therapeutic response through analyses of circulating cell-free tumour DNA (ctDNA) and extracellular RNA (exRNA) in multiple myeloma (MM) patients were performed in a Phase Ib trial of 24 relapsed/refractory patients receiving oral azacitidine in combination with lenalidomide and dexamethasone. Mutational characterisation of paired BM and PL samples at study entry identified that patients with a higher number of mutations or a higher mutational fractional abundance in PL had significantly shorter overall survival (OS) (p = 0.005 and p = 0.018, respectively). A decrease in ctDNA levels at day 5 of cycle 1 of treatment (C1D5) correlated with superior progression-free survival (PFS) (p = 0.017). Evaluation of exRNA transcripts of candidate biomarkers indicated that high CRBN levels coupled with low levels of SPARC at baseline were associated with shorter OS (p = 0.000003). IKZF1 fold-change <0.05 at C1D5 was associated with shorter PFS (p = 0.0051) and OS (p = 0.0001). Furthermore, patients with high baseline CRBN coupled with low fold-change at C1D5 were at the highest risk of progression (p = 0.0001). In conclusion, this exploratory analysis has provided the first demonstration in MM of ctDNA for predicting disease outcome and of the utility of exRNA as a biomarker of therapeutic response.


Asunto(s)
ADN Tumoral Circulante/análisis , Mieloma Múltiple/tratamiento farmacológico , ARN/análisis , Proteínas Adaptadoras Transductoras de Señales , Costo de Enfermedad , Genes p53 , Humanos , Factor de Transcripción Ikaros/análisis , Mieloma Múltiple/sangre , Mieloma Múltiple/genética , Mieloma Múltiple/mortalidad , Mutación , Péptido Hidrolasas/análisis , Pronóstico , Ubiquitina-Proteína Ligasas
5.
Sci Rep ; 7(1): 11915, 2017 09 20.
Artículo en Inglés | MEDLINE | ID: mdl-28931834

RESUMEN

Regulatory T cells (Tregs) play crucial role in maintenance of peripheral tolerance. Recent clinical trials confirmed safety and efficacy of Treg treatment of deleterious immune responses. However, Tregs lose their characteristic phenotype and suppressive potential during expansion ex vivo. Therefore, multiple research teams have been studding Treg biology in aim to improve their stability in vitro. In the current paper, we demonstrate that mild hypothermia of 33 °C induces robust proliferation of Tregs, preserves expression of FoxP3, CD25 and Helios, and prevents TSDR methylation during culture in vitro. Tregs expanded at 33 °C have stronger immunosuppressive potential and remarkably anti-inflammatory phenotype demonstrated by the whole transcriptome sequencing. These observations shed new light on impact of temperature on regulation of immune response. We show that just a simple change in temperature can preserve Treg stability, function and accelerate their proliferation, responding to unanswered question- how to preserve Treg stability in vitro.


Asunto(s)
Hipotermia , Linfocitos T Reguladores/fisiología , Linfocitos T Reguladores/efectos de la radiación , Proliferación Celular/efectos de la radiación , Células Cultivadas , Frío , Factores de Transcripción Forkhead/análisis , Perfilación de la Expresión Génica , Humanos , Factor de Transcripción Ikaros/análisis , Tolerancia Inmunológica , Subunidad alfa del Receptor de Interleucina-2/análisis , Linfocitos T Reguladores/química
6.
Am J Hematol ; 92(3): 269-278, 2017 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-28052520

RESUMEN

Immunomodulatory drugs (IMiDs) are a cornerstone in the treatment of multiple myeloma (MM), but specific markers to predict outcome are still missing. Recent work pointed to a prognostic role for IMiD target genes (e.g. CRBN). Moreover, indirect activity of IMiDs on immune cells correlated with outcome, raising the possibility that cell populations in the bone marrow (BM) microenvironment could serve as biomarkers. We therefore analysed gene expression levels of six IMiD target genes in whole BM samples of 44 myeloma patients treated with lenalidomide-dexamethasone. Expression of CRBN (R = 0.30, P = .05), IKZF1 (R = 0.31, P = .04), IRF4 (R = 0.38, P = .01), MCT-1 (R = 0.30, P = .05), and CD147 (R = 0.38, P = .01), but not IKZF3 (R = -0.15, P = .34), was significantly associated with response. Interestingly, IKZF1 expression was elevated in BM environmental cells and thus selected for further investigation by multicolor flow cytometry. High IKAROS protein levels in total BM mononuclear cells (median OS 83.4 vs. 32.2 months, P = .02), CD19+ B cells (median OS 71.1 vs. 32.2 months, P = .05), CD3+ CD8+ T cells (median OS 83.4 vs 19.0 months, P = .008) as well as monocytes (median OS 53.9 vs 18.0 months, P = .009) were associated with superior overall survival (OS). In contrast, IKAROS protein expression in MM cells was not predictive for OS. Our data therefore corroborate the central role of immune cells for the clinical activity of IMiDs and built the groundwork for prospective analysis of IKAROS protein levels in distinct cell populations as a potential biomarker for IMiD based therapies.


Asunto(s)
Biomarcadores de Tumor/análisis , Células de la Médula Ósea/química , Factor de Transcripción Ikaros/análisis , Factores Inmunológicos/uso terapéutico , Mieloma Múltiple/tratamiento farmacológico , Dexametasona/uso terapéutico , Expresión Génica , Humanos , Factor de Transcripción Ikaros/metabolismo , Factores Inmunológicos/genética , Lenalidomida , Mieloma Múltiple/mortalidad , Mieloma Múltiple/patología , Tasa de Supervivencia , Talidomida/análogos & derivados , Talidomida/uso terapéutico , Resultado del Tratamiento
7.
Leukemia ; 31(8): 1760-1769, 2017 08.
Artículo en Inglés | MEDLINE | ID: mdl-27890933

RESUMEN

Inhibition of the bromodomain and extra-terminal (BET) proteins is a promising therapeutic strategy for various hematologic cancers. Previous studies suggest that BET inhibitors constrain tumor cell proliferation and survival mainly through the suppression of MYC transcription and activity. However, suppression of the transcription of additional genes also contributes to the antitumor activity of BET inhibitors but is less well understood. Here we examined the therapeutic potential of CPI-0610, a potent BET inhibitor currently undergoing phase I clinical testing, in multiple myeloma (MM). CPI-0610 displays potent cytotoxicity against MM cell lines and patient-derived MM cells through G1 cell cycle arrest and caspase-dependent apoptosis. CPI-0610-mediated BET inhibition overcomes the protective effects conferred by cytokines and bone marrow stromal cells. We also confirmed the in vivo efficacy of CPI-0610 in a MM xenograft mouse model. Our study found IKZF1 and IRF4 to be among the primary targets of CPI-0610, along with MYC. Given that immunomodulatory drugs (IMiDs) stabilize cereblon and facilitate Ikaros degradation in MM cells, we combined it with CPI-0610. Combination studies of CPI-0610 with IMiDs show in vitro synergism, in part due to concomitant suppression of IKZF1, IRF4 and MYC, providing a rationale for clinical testing of this drug combination in MM patients.


Asunto(s)
Benzazepinas/farmacología , Isoxazoles/farmacología , Mieloma Múltiple/tratamiento farmacológico , Proteínas Nucleares/antagonistas & inhibidores , Proteínas Serina-Treonina Quinasas/antagonistas & inhibidores , Factores de Transcripción/antagonistas & inhibidores , Animales , Proteínas de Ciclo Celular , Puntos de Control de la Fase G1 del Ciclo Celular , Humanos , Factor de Transcripción Ikaros/análisis , Factor de Transcripción Ikaros/genética , Factores Reguladores del Interferón/análisis , Factores Reguladores del Interferón/genética , Ratones , Mieloma Múltiple/patología , Proteínas Proto-Oncogénicas c-myc/análisis , Proteínas Proto-Oncogénicas c-myc/genética
8.
J Hematol Oncol ; 9(1): 123, 2016 11 21.
Artículo en Inglés | MEDLINE | ID: mdl-27881177

RESUMEN

The aim of this study is to assess nucleoprotein expression of IKZF1/3 in patients with relapsed/refractory multiple myeloma (MM) who received lenalidomide-based therapy and correlated them with their clinical outcomes. A total of 50 patients diagnosed with MM were entered in the study with the median follow-up of 86.4 months. By immunohistochemistry (IHC), IKZF1 and IKZF3 were expressed in 72 and 58% of the cases, respectively. IKZF1 and IKZF3 expressions were associated with longer median progression free survival (P = 0.0029 and P < 0.0001) and overall survival (P = 0.0014 and P < 0.0001). IKZF3 expression also appears predicted a favorable response to the lenalidomide-based therapy.


Asunto(s)
Factor de Transcripción Ikaros/análisis , Mieloma Múltiple/tratamiento farmacológico , Talidomida/análogos & derivados , Adulto , Anciano , Supervivencia sin Enfermedad , Femenino , Humanos , Lenalidomida , Masculino , Persona de Mediana Edad , Mieloma Múltiple/diagnóstico , Mieloma Múltiple/mortalidad , Pronóstico , Terapia Recuperativa , Tasa de Supervivencia , Talidomida/uso terapéutico , Resultado del Tratamiento
9.
PLoS One ; 9(8): e105027, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25119018

RESUMEN

The cancer microenvironment plays a pivotal role in oncogenesis, containing a number of regulatory cells that attenuate the anti-neoplastic immune response. While the negative prognostic impact of regulatory T cells (Tregs) in the context of most solid tissue tumors is well established, their role in lymphoid malignancies remains unclear. T cells expressing FOXP3 and Helios were documented in the fine needle aspirates of affected lymph nodes of dogs with spontaneous multicentric B cell lymphoma (BCL), proposed to be a model for human non-Hodgkin lymphoma. Multivariable analysis revealed that the frequency of lymph node FOXP3(+) T cells was an independent negative prognostic factor, impacting both progression-free survival (hazard ratio 1.10; p = 0.01) and overall survival (hazard ratio 1.61; p = 0.01) when comparing dogs showing higher than the median FOXP3 expression with those showing the median value of FOXP3 expression or less. Taken together, these data suggest the existence of a population of Tregs operational in canine multicentric BCL that resembles thymic Tregs, which we speculate are co-opted by the tumor from the periphery. We suggest that canine multicentric BCL represents a robust large animal model of human diffuse large BCL, showing clinical, cytological and immunophenotypic similarities with the disease in man, allowing comparative studies of immunoregulatory mechanisms.


Asunto(s)
Enfermedades de los Perros/diagnóstico , Factores de Transcripción Forkhead/análisis , Linfoma de Células B/diagnóstico , Linfoma de Células B/veterinaria , Linfocitos T Reguladores/patología , Animales , Antígenos CD8/análisis , Antígenos CD8/inmunología , Modelos Animales de Enfermedad , Enfermedades de los Perros/inmunología , Enfermedades de los Perros/patología , Perros , Femenino , Factores de Transcripción Forkhead/inmunología , Genes MHC Clase II , Factor de Transcripción Ikaros/análisis , Factor de Transcripción Ikaros/inmunología , Inmunofenotipificación , Linfoma de Células B/inmunología , Linfoma de Células B/patología , Masculino , Pronóstico , Linfocitos T Reguladores/inmunología , Microambiente Tumoral
11.
Haematologica ; 93(12): 1814-21, 2008 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-18838475

RESUMEN

BACKGROUND: Ikaros is the prototypic member of a Kruppel-like zinc finger transcription factor subfamily that is required for normal hematopoietic cell differentiation and proliferation, particularly in the lymphoid lineages. Alternative splicing can generate multiple Ikaros isoforms that lack different numbers of exons and have different functions. Shorter isoforms, which lack the amino-terminal domain that mediates sequence-specific DNA binding, exert a dominant negative effect and inhibit the ability of longer heterodimer partners to bind DNA. DESIGN AND METHODS: In this study, we developed a high-throughput capillary electrophoresis sizing method to detect and quantify different Ikaros cDNA transcripts. RESULTS: We demonstrated that Philadelphia chromosome-positive acute lymphoblastic leukemia cells expressed high levels of the non-DNA-binding isoform Ik6 that was generated following IKZF1 genomic deletions (19/46 patients, 41%). Furthermore, a recurring 60 bp insertion immediately upstream of exon 5, at the exon 3/exon 5 junction, was frequently detected in the Ik2 and Ik4 isoforms. This insertion occurred either alone or together with an in-frame ten amino acid deletion that was due to a 30 bp loss at the end of exon 7. Both the alterations are due to the selection of alternative cryptic splice sites and have been suggested to cause impaired DNA-binding activity. Non-DNA-binding isoforms were localized in the cytoplasm whereas the DNA-binding isoforms were localized in the nucleus. CONCLUSIONS: Our findings demonstrate that both aberrant splicing and genomic deletion leading to different non-DNA-binding Ikaros cDNA transcripts are common features of Philadelphia chromosome-positive acute lymphoblastic leukemia.


Asunto(s)
Electroforesis Capilar/métodos , Factor de Transcripción Ikaros/genética , Leucemia-Linfoma Linfoblástico de Células Precursoras/genética , Adolescente , Adulto , Anciano , ADN Complementario/análisis , Humanos , Factor de Transcripción Ikaros/análisis , Persona de Mediana Edad , ARN Mensajero/análisis , Adulto Joven
12.
Immunity ; 26(3): 335-44, 2007 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-17363301

RESUMEN

Ikaros DNA-binding proteins are critical for the development of lymphocytes and other hematopoietic lineages, but it remains unclear how they cooperate with other regulators of signaling and transcription to achieve ordered gene expression during development. Here, we show that Ikaros proteins regulate the pre-BCR component lambda5 in a stage-specific manner. In pre-BI cells, Ikaros modulated lambda5 expression in competition with the transcriptional activator EBF. This required Ikaros binding to the Igll1 (lambda5) promoter and was abolished either by mutation of the Ikaros DNA-binding domain or by deletion of a single Ikaros site from the Igll1 promoter. At the transition from the pre-BI to pre-BII stage, the expression of the Ikaros family member Aiolos was upregulated and required for the efficient silencing of Igll1. Aiolos expression was controlled by pre-BCR signals via the adaptor protein SLP-65. Thus, pre-BCR signaling regulates Aiolos and the silencing of Igll1 via a developmental-stage-specific feedback loop.


Asunto(s)
Linfocitos B/inmunología , Proteínas de Unión al ADN/fisiología , Regulación de la Expresión Génica , Factor de Transcripción Ikaros/fisiología , Cadenas Ligeras de Inmunoglobulina/genética , Glicoproteínas de Membrana/genética , Transactivadores/genética , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Animales , Linfocitos B/química , Células Cultivadas , Proteínas de Unión al ADN/genética , Silenciador del Gen , Factor de Transcripción Ikaros/análisis , Factor de Transcripción Ikaros/genética , Inmunoglobulina de Cadenas Ligeras Subrogadas , Activación de Linfocitos/genética , Ratones , Regiones Promotoras Genéticas/genética , Estructura Terciaria de Proteína/genética , Transactivadores/fisiología
13.
Proc Natl Acad Sci U S A ; 103(7): 2214-9, 2006 Feb 14.
Artículo en Inglés | MEDLINE | ID: mdl-16467156

RESUMEN

Ikaros transcription factors play critical functions in the control of lymphohematopoiesis and immune regulation. Family members contain multiple zinc fingers that mediate DNA binding and homooligomerization or heterooligomerization. Ikaros is abundantly expressed in pituitary mammosomatotrophs, where it deacetylates histone 3 sites on the proximal growth hormone (GH) promoter to silence gene expression. Ikaros-null mice display stunted growth with reduced circulating levels of the GH target factor insulin-like growth factor I (IGF-I). Ikaros-deficient mice have small anterior pituitary glands with a disproportionately reduced somatotroph population. Systemic administration of GH results in increased IGF-I levels and enhanced somatic growth. In contrast, reconstitution with WT lymphocytes was not sufficient to rescue the stunted growth phenotype of Ikaros-deficient mice. Ikaros was identified in mouse hypothalamic arcuate nuclei, where it colocalized with GH-releasing hormone (GHRH); in contrast, Ikaros-null mice lack GHRH immunoreactivity in the hypothalamus. Overexpression of Ikaros enhanced GHRH promoter activity and induced endogenous GHRH gene expression. These findings unmask a wider role for Ikaros in the neuroendocrine system, highlighting a critical contribution to the development of the hypothalamic-pituitary somatotrophic axis.


Asunto(s)
Regulación del Desarrollo de la Expresión Génica , Hormona Liberadora de Hormona del Crecimiento/genética , Sistema Hipotálamo-Hipofisario/crecimiento & desarrollo , Factor de Transcripción Ikaros/fisiología , Hipófisis/crecimiento & desarrollo , Animales , Núcleo Arqueado del Hipotálamo/citología , Núcleo Arqueado del Hipotálamo/metabolismo , Hormona del Crecimiento/deficiencia , Hormona del Crecimiento/farmacología , Hormona Liberadora de Hormona del Crecimiento/metabolismo , Hematopoyesis , Sistema Hipotálamo-Hipofisario/citología , Sistema Hipotálamo-Hipofisario/metabolismo , Hipotálamo/citología , Hipotálamo/metabolismo , Factor de Transcripción Ikaros/análisis , Factor de Transcripción Ikaros/genética , Factor I del Crecimiento Similar a la Insulina/análisis , Factor I del Crecimiento Similar a la Insulina/metabolismo , Ratones , Ratones Mutantes , Neuronas/química , Neuronas/metabolismo , Hipófisis/citología , Hipófisis/metabolismo
14.
Oncogene ; 25(7): 1118-24, 2006 Feb 16.
Artículo en Inglés | MEDLINE | ID: mdl-16205638

RESUMEN

Pre-B lymphoblastic leukemia cells carrying a BCR-ABL1 gene rearrangement exhibit an undifferentiated phenotype. Comparing the genome-wide gene expression profiles of normal B-cell subsets and BCR-ABL1+ pre-B lymphoblastic leukemia cells by SAGE, the leukemia cells show loss of B lymphoid identity and aberrant expression of myeloid lineage-specific molecules. Consistent with this, BCR-ABL1+ pre-B lymphoblastic leukemia cells exhibit defective expression of IKAROS, a transcription factor needed for early lymphoid lineage commitment. As shown by inducible expression of BCR-ABL1 in human and murine B-cell precursor cell lines, BCR-ABL1 induces the expression of a dominant-negative IKAROS splice variant, termed IK6. Comparing matched leukemia sample pairs from patients before and during therapy with the BCR-ABL1 kinase inhibitor STI571 (Imatinib), inhibition of BCR-ABL1 partially corrected aberrant expression of IK6 and lineage infidelity of the leukemia cells. To elucidate the contribution of IK6 to lineage infidelity in BCR-ABL1+ cell lines, IK6 expression was silenced by RNA interference. Upon inhibition of IK6, BCR-ABL1+ leukemia cells partially restored B lymphoid lineage commitment. Therefore, we propose that BCR-ABL1 induces aberrant splicing of IKAROS, which interferes with lineage identity and differentiation of pre-B lymphoblastic leukemia cells.


Asunto(s)
Empalme Alternativo , Factor de Transcripción Ikaros/genética , Leucemia-Linfoma Linfoblástico de Células Precursoras B/enzimología , Leucemia-Linfoma Linfoblástico de Células Precursoras B/genética , Proteínas Tirosina Quinasas/metabolismo , Animales , Antineoplásicos/farmacología , Antineoplásicos/uso terapéutico , Benzamidas , Línea Celular Tumoral , Linaje de la Célula/genética , Núcleo Celular/química , Proteínas de Fusión bcr-abl , Perfilación de la Expresión Génica , Silenciador del Gen , Humanos , Factor de Transcripción Ikaros/análisis , Factor de Transcripción Ikaros/metabolismo , Mesilato de Imatinib , Ratones , Piperazinas/farmacología , Leucemia-Linfoma Linfoblástico de Células Precursoras B/dietoterapia , Inhibidores de Proteínas Quinasas/farmacología , Inhibidores de Proteínas Quinasas/uso terapéutico , Proteínas Tirosina Quinasas/análisis , Pirimidinas/farmacología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...