Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 26
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Int J Mol Sci ; 24(22)2023 Nov 17.
Artículo en Inglés | MEDLINE | ID: mdl-38003662

RESUMEN

Menin/MEN1 is a scaffold protein that participates in proliferation, regulation of gene transcription, DNA damage repair, and signal transduction. In hematological malignancies harboring the KMT2A/MLL1 (MLLr) chromosomal rearrangements, the interaction of the oncogenic fusion protein MLLr with MEN1 has been shown to be essential. MEN1 binders inhibiting the MEN1 and KMT2A interaction have been shown to be effective against MLLr AML and B-ALL in experimental models and clinical studies. We hypothesized that in addition to the MEN1-KMT2A interaction, alternative mechanisms might be instrumental in the MEN1 dependency of leukemia. We first mined and analyzed data from publicly available gene expression databases, finding that the dependency of B-ALL cell lines on MEN1 did not correlate with the presence of MLLr. Using shRNA-mediated knockdown, we found that all tested B-ALL cell lines were sensitive to MEN1 depletion, independent of the underlying driver mutations. Most multiple myeloma cell lines that did not harbor MLLr were also sensitive to the genetic depletion of MEN1. We conclude that the oncogenic role of MEN1 is not limited to the interaction with KMT2A. Our results suggest that targeted degradation of MEN1 or the development of binders that induce global changes in the MEN1 protein structure may be more efficient than the inhibition of individual MEN1 protein interactions.


Asunto(s)
Mieloma Múltiple , Leucemia-Linfoma Linfoblástico de Células Precursoras B , Humanos , Línea Celular Tumoral , Leucemia/metabolismo , Mieloma Múltiple/genética , Factores de Transcripción/genética
2.
Front Immunol ; 14: 1125503, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-36817488

RESUMEN

The germinal center (GC) reaction is a key process during an adaptive immune response to T cell specific antigens. GCs are specialized structures within secondary lymphoid organs, in which B cell proliferation, somatic hypermutation and antibody affinity maturation occur. As a result, high affinity antibody secreting plasma cells and memory B cells are generated. An effective GC response needs interaction between multiple cell types. Besides reticular cells and follicular dendritic cells, particularly B cells, T follicular helper (Tfh) cells as well as T follicular regulatory (Tfr) cells are a key player during the GC reaction. Whereas Tfh cells provide help to GC B cells in selection processes, Tfr cells, a specialized subset of regulatory T cells (Tregs), are able to suppress the GC reaction maintaining the balance between immune activation and tolerance. The formation and function of GCs is regulated by a complex network of signals and molecules at multiple levels. In this review, we highlight recent developments in GC biology by focusing on the transcriptional program regulating the GC reaction. This review focuses on the transcriptional co-activator BOB.1/OBF.1, whose important role for GC B, Tfh and Tfr cell differentiation became increasingly clear in recent years. Moreover, we outline how deregulation of the GC transcriptional program can drive lymphomagenesis.


Asunto(s)
Células T Auxiliares Foliculares , Linfocitos T Colaboradores-Inductores , Linfocitos B , Centro Germinal , Linfocitos T Reguladores , Antígenos/metabolismo
3.
Oncogenesis ; 11(1): 1, 2022 Jan 10.
Artículo en Inglés | MEDLINE | ID: mdl-35013097

RESUMEN

The D-type cyclins (CCND1, CCND2, and CCND3) in association with CDK4/6 are known drivers of cell cycle progression. We reported previously that inactivation of FOXO1 confers growth arrest and apoptosis in B-ALL, partially mediated by subsequent depletion of CCND3. Given that previously the canonical MYC target CCND2 has been considered to play the major role in B-ALL proliferation, further investigation of the role of FOXO1 in CCND3 transcription and the role of CCND3 in B-ALL is warranted. In this study, we demonstrated that CCND3 is essential for the proliferation and survival of B-ALL, independent of the mutational background. Respectively, its expression at mRNA level exceeds that of CCND1 and CCND2. Furthermore, we identified FOXO1 as a CCND3-activating transcription factor in B-ALL. By comparing the effects of CCND3 depletion and CDK4/6 inhibition by palbociclib on B-ALL cells harboring different driver mutations, we found that the anti-apoptotic effect of CCND3 is independent of the kinase activity of the CCND3-CDK4/6 complex. Moreover, we found that CCND3 contributes to CDK8 transcription, which in part might explain the anti-apoptotic effect of CCND3. Finally, we found that increased CCND3 expression is associated with the development of resistance to palbociclib. We conclude that CCND3 plays an essential role in the maintenance of B-ALL, regardless of the underlying driver mutation. Moreover, downregulation of CCND3 expression might be superior to inhibition of CDK4/6 kinase activity in terms of B-ALL treatment.

5.
Leukemia ; 34(3): 857-871, 2020 03.
Artículo en Inglés | MEDLINE | ID: mdl-31719683

RESUMEN

In addition to oncogenic MYC translocations, Burkitt lymphoma (BL) depends on the germinal centre (GC) dark zone (DZ) B cell survival and proliferation programme, which is characterized by relatively low PI3K-AKT activity. Paradoxically, PI3K-AKT activation facilitates MYC-driven lymphomagenesis in mice, and it has been proposed that PI3K-AKT activation is essential for BL. Here we show that the PI3K-AKT activity in primary BLs and BL cell lines does not exceed that of human non-neoplastic tonsillar GC DZ B cells. BLs were not sensitive to AKT1 knockdown, which induced massive cell death in pAKThigh DLBCL cell lines. Likewise, BL cell lines show low sensitivity to pan-AKT inhibitors. Moreover, hyper-activation of the PI3K-AKT pathway by overexpression of a constitutively active version of AKT (myrAKT) or knockdown of PTEN repressed the growth of BL cell lines. This was associated with increased AKT phosphorylation, NF-κB activation, and downregulation of DZ genes including the proto-oncogene MYB and the DZ marker CXCR4. In contrast to GCB-DLBCL, PTEN overexpression was tolerated by BL cell lines. We conclude that the molecular mechanisms instrumental to guarantee the survival of normal DZ B cells, including the tight regulation of the PTEN-PI3K-AKT axis, also operate in the survival/proliferation of BL.


Asunto(s)
Linfoma de Burkitt/metabolismo , Fosfohidrolasa PTEN/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Apoptosis , Linfocitos B/citología , Línea Celular Tumoral , Proliferación Celular , Supervivencia Celular , Humanos , Tonsila Palatina/citología , Fenotipo , Proto-Oncogenes Mas , Transducción de Señal
6.
Int J Cancer ; 147(1): 202-217, 2020 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-31846065

RESUMEN

Adenosine is a signaling molecule that exerts dual effects on tumor growth: while it inhibits immune cell function and thereby prevents surveillance by the immune system, it influences tumorigenesis directly via activation of adenosine receptors on tumor cells at the same time. However, the adenosine-mediated mechanisms affecting oncogenic processes particularly in head and neck squamous cell carcinomas (HNSCC) are not fully understood. Here, we investigated the role of adenosine receptor activity on HNSCC-derived cell lines. Targeting the adenosine receptor A2B (ADORA2B) on these cells with the inverse agonist/antagonist PSB-603 leads to inhibition of cell proliferation, transmigration as well as VEGFA secretion in vitro. At the molecular level, these effects were associated with cell cycle arrest as well as the induction of the apoptotic pathway. In addition, shRNA-mediated downmodulation of ADORA2B expression caused decreased proliferation. Moreover, in in vivo xenograft experiments, chemical and genetic abrogation of ADORA2B activity impaired tumor growth associated with decreased tumor vascularization. Together, our findings characterize ADORA2B as a crucial player in the maintenance of HNSCC and, therefore, as a potential therapeutic target for HNSCC treatment.


Asunto(s)
Neoplasias de Cabeza y Cuello/metabolismo , Receptor de Adenosina A2B/metabolismo , Carcinoma de Células Escamosas de Cabeza y Cuello/metabolismo , 5'-Nucleotidasa/biosíntesis , 5'-Nucleotidasa/metabolismo , Antagonistas del Receptor de Adenosina A2/farmacología , Animales , Apoptosis/efectos de los fármacos , Puntos de Control del Ciclo Celular/efectos de los fármacos , Procesos de Crecimiento Celular/efectos de los fármacos , Procesos de Crecimiento Celular/fisiología , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Movimiento Celular/fisiología , Embrión de Pollo , Membrana Corioalantoides/metabolismo , Proteínas Ligadas a GPI/biosíntesis , Proteínas Ligadas a GPI/metabolismo , Neoplasias de Cabeza y Cuello/irrigación sanguínea , Neoplasias de Cabeza y Cuello/patología , Humanos , Células Jurkat , Neovascularización Patológica/metabolismo , Neovascularización Patológica/patología , Receptor de Adenosina A2B/biosíntesis , Carcinoma de Células Escamosas de Cabeza y Cuello/irrigación sanguínea , Carcinoma de Células Escamosas de Cabeza y Cuello/patología , Sulfonamidas/farmacología , Xantinas/farmacología
7.
Cancers (Basel) ; 11(10)2019 Sep 25.
Artículo en Inglés | MEDLINE | ID: mdl-31557894

RESUMEN

The FOXO1 transcription factor plays a central role in the proliferation and survival of B cells at several stages of differentiation. B cell malignancies, with exception of classical Hodgkin lymphoma, maintain expression of FOXO1 at levels characteristic for their non-malignant counterparts. Extensive expression profiling had revealed that Burkitt lymphoma (BL) show many characteristics of the dark zone (DZ) germinal center (GC) B cell program. Here we show that FOXO1 knockdown inhibits proliferation of human BL cell lines. The anti-proliferative effect of the FOXO1 knockdown is associated with the repression of the DZ B cell program including expression of MYB, CCND3, RAG2, BACH2, and CXCR4. In addition, the induction of signaling pathways of the light zone (LZ) program like NF-κB and PI3K-AKT was observed. Using a rescue experiment we identified downregulation of the proto-oncogene MYB as a critical factor contributing to the antiproliferative effect of FOXO1 knockdown. In an attempt to estimate the feasibility of pharmacological FOXO1 repression, we found that the small molecular weight FOXO1 inhibitor AS1842856 induces cell death and growth arrest in BL cell lines at low concentrations. Interestingly, we found that overactivation of FOXO1 also induces growth inhibition in BL cell lines, indicating the importance of a tight regulation of FOXO1 activity in BL.

9.
Blood ; 131(26): 2929-2942, 2018 06 28.
Artículo en Inglés | MEDLINE | ID: mdl-29622548

RESUMEN

The FOXO1 transcription factor plays an essential role in the regulation of proliferation and survival programs at early stages of B-cell differentiation. Here, we show that tightly regulated FOXO1 activity is essential for maintenance of B-cell precursor acute lymphoblastic leukemia (BCP-ALL). Genetic and pharmacological inactivation of FOXO1 in BCP-ALL cell lines produced a strong antileukemic effect associated with CCND3 downregulation. Moreover, we demonstrated that CCND3 expression is critical for BCP-ALL survival and that overexpression of CCND3 protected BCP-ALL cell lines from growth arrest and apoptosis induced by FOXO1 inactivation. Most importantly, pharmacological inhibition of FOXO1 showed antileukemia activity on several primary, patient-derived, pediatric ALL xenografts with effective leukemia reduction in the hematopoietic, lymphoid, and central nervous system organ compartments, ultimately leading to prolonged survival without leukemia reoccurrence in a preclinical in vivo model of BCP-ALL. These results suggest that repression of FOXO1 might be a feasible approach for the treatment of BCP-ALL.


Asunto(s)
Proteína Forkhead Box O1/genética , Regulación Leucémica de la Expresión Génica , Leucemia-Linfoma Linfoblástico de Células Precursoras B/genética , Animales , Antineoplásicos/uso terapéutico , Linfocitos B/efectos de los fármacos , Linfocitos B/metabolismo , Linfocitos B/patología , Línea Celular Tumoral , Ciclina D3/genética , Proteína Forkhead Box O1/antagonistas & inhibidores , Proteína Forkhead Box O1/metabolismo , Regulación Leucémica de la Expresión Génica/efectos de los fármacos , Humanos , Ratones Endogámicos NOD , Ratones SCID , Leucemia-Linfoma Linfoblástico de Células Precursoras B/tratamiento farmacológico , Leucemia-Linfoma Linfoblástico de Células Precursoras B/metabolismo , Leucemia-Linfoma Linfoblástico de Células Precursoras B/patología , Proteínas Proto-Oncogénicas c-akt/metabolismo , Quinolonas/uso terapéutico , Transducción de Señal/efectos de los fármacos
10.
Blood ; 131(14): 1556-1567, 2018 04 05.
Artículo en Inglés | MEDLINE | ID: mdl-29439954

RESUMEN

We recently found that FOXO1 repression contributes to the oncogenic program of classical Hodgkin lymphoma (cHL). Interestingly, FOXO3A, another member of the FOXO family, was reported to be expressed in the malignant Hodgkin and Reed-Sternberg cells of cHL at higher levels than in non-Hodgkin lymphoma subtypes. We thus aimed to investigate mechanisms responsible for the maintenance of FOXO3A as well as the potential role of FOXO3A in cHL. Here, we show that high FOXO3A levels in cHL reflect a B-cell-differentiation-specific pattern. In B cells, FOXO3A expression increases during the process of centroblast to plasma cell (PC) differentiation. FOXO3A levels in cHL were found higher than in germinal center B cells, but lower than in terminally differentiated PCs. This intermediate FOXO3A expression in cHL might manifest the "abortive PC differentiation" phenotype. This assumption was further corroborated by the finding that overexpression of FOXO3A in cHL cell lines induced activation of the master PC transcription factor PRDM1α. As factors attenuating FOXO3A expression in cHL, we identified MIR155 and constitutive activation of extracellular signal-regulated kinase. Finally, we demonstrate the importance of FOXO3A expression in cHL using an RNA interference approach. We conclude that tightly regulated expression of FOXO3A contributes to the oncogenic program and to the specific phenotype of cHL.


Asunto(s)
Diferenciación Celular , Proteína Forkhead Box O3/biosíntesis , Regulación Neoplásica de la Expresión Génica , Enfermedad de Hodgkin/metabolismo , Proteínas de Neoplasias/biosíntesis , Células Plasmáticas/metabolismo , Línea Celular Tumoral , Supervivencia Celular , Proteína Forkhead Box O3/genética , Enfermedad de Hodgkin/genética , Enfermedad de Hodgkin/patología , Humanos , MicroARNs/genética , MicroARNs/metabolismo , Proteínas de Neoplasias/genética , Células Plasmáticas/patología , ARN Neoplásico/genética , ARN Neoplásico/metabolismo
11.
Semin Cancer Biol ; 50: 132-141, 2018 06.
Artículo en Inglés | MEDLINE | ID: mdl-28774833

RESUMEN

FOX O family transcription factors are important for differentiation and function of multiple cell types. In B lymphocytes they play a critical role. The activity of FOXOs is directly regulated both by signaling from B cell receptor (BCR) and cytokine receptors. FOXO1 action controls the transition between differentiation stages of B cell development. In comparison to other FOXO family members, FOXO1 plays a superior role in the regulation of early stages of B-cell differentiation. Although being known as a negative regulator of cell proliferation and therefore potential tumor suppressor, FOXO1 is downregulated only in Hodgkin lymphoma (HL) subtypes. In non-Hodgkin lymphoma (NHL) entities its expression is maintained at significant levels, raising the question on the role of FOXO-transcription factors in the proliferation and survival programs in the process of B cell differentiation as well as their contribution to the oncogenic programs of B-cell lymphomas. In particular, we discuss molecular mechanisms that might determine the switch between pro-apoptotic and pro-survival effects of FOXO1 and their interplay with specific differentiation programs.


Asunto(s)
Proteína Forkhead Box O1/genética , Enfermedad de Hodgkin/genética , Neoplasias/genética , Receptores de Antígenos de Linfocitos B/genética , Linfocitos B/patología , Proliferación Celular/genética , Factores de Transcripción Forkhead , Regulación Neoplásica de la Expresión Génica , Enfermedad de Hodgkin/patología , Humanos , Linfopoyesis/genética , Neoplasias/patología , Transducción de Señal/genética
12.
Int J Oncol ; 50(2): 555-566, 2017 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-28035374

RESUMEN

DNA methylation is an epigenetic control mechanism that contributes to the specific phenotype and to the oncogenic program of virtually all tumor entities. Although efficacy of demethylating agents in classical Hodgkin lymphoma (cHL) was not specifically tested, a case of regression of relapsed metastatic cHL was described as a fortunate side­effect of the demethylating agent 5­azacytidine in a patient with myelodysplastic syndrome. We investigated molecular mechanisms of decitabine (5­Aza­dC) antitumor activity in cHL using gene expression profiling followed by gene set enrichment analysis. We found that 5­Aza­dC inhibits growth of cHL cell lines at clinically relevant concentrations of 0.25­2 µM. The antitumor effect of 5­Aza­dC was associated with induction of genes, which negatively regulate cell cycle progression (e.g. CDKN1A and GADD45A). Surprisingly, we also observed significant enrichment of pro­survival pathways like MEK/ERK, JAK­STAT and NF­κB, as well as signatures comprising transcription­activating genes. Among the upregulated pro­survival genes were the anti­apoptotic genes BCL2 and BCL2L1, as well as genes involved in transduction of growth and survival signals like STAT1, TLR7, CD40 and IL-6. We therefore analyzed whether interference with these pro­survival pathways and genes would potentiate the antitumor effect of 5­Aza­dC. We could show that the BCL2/BCL2L1 inhibitor ABT263, the JAK­STAT inhibitors fedratinib and SH­4­54, the AKT inhibitor KP372­1, the NF­κB inhibitor QNZ, as well as the bromodomain and extraterminal (BET) family proteins inhibitor JQ1 acted synergistically with 5­Aza­dC. We conclude that targeting of oncogenic pathways of cHL may improve efficacy of DNA-demethylating therapy in cHL.


Asunto(s)
Azacitidina/análogos & derivados , Perfilación de la Expresión Génica/métodos , Enfermedad de Hodgkin/genética , Análisis de Secuencia por Matrices de Oligonucleótidos/métodos , Oncogenes/efectos de los fármacos , Azacitidina/farmacología , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Metilación de ADN/efectos de los fármacos , Decitabina , Epigénesis Genética/efectos de los fármacos , Regulación Neoplásica de la Expresión Génica/genética , Humanos , Transducción de Señal/efectos de los fármacos , Bibliotecas de Moléculas Pequeñas/farmacología
13.
Oncotarget ; 7(24): 36854-36864, 2016 Jun 14.
Artículo en Inglés | MEDLINE | ID: mdl-27166193

RESUMEN

Although Hodgkin and Reed-Sternberg (HRS) cells of classical Hodgkin lymphoma (cHL) derived from germinal or post germinal B cells, they have lost the B cell phenotype in the process of lymphomagenesis. The phenomenon can be at least partially explained by repression of B-cell-specific transcription factors including TCF3, early B-cell factor 1 (EBF1), SPI1/PU.1, and FOXO1, which are down-regulated by genetic and epigenetic mechanisms. The unique phenotype has been suspected to be advantageous for survival of HRS cells. Ectopic expression of some of these transcription factors (EBF1, PU.1, FOXO1) indeed impaired survival of cHL cells. Here we show that forced expression of TCF3 causes cell death and cell cycle arrest in cHL cell lines. Mechanistically, TCF3 overexpression modulated expression of multiple pro-apoptotic genes including BIK, APAF1, FASLG, BOK, and TNFRSF10A/DR4. We conclude that TCF3 inactivation contributes not only to extinguishing of B cell phenotype but also to cHL oncogenesis.


Asunto(s)
Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Carcinogénesis/metabolismo , Enfermedad de Hodgkin/patología , Linfocitos B/metabolismo , Línea Celular Tumoral , Enfermedad de Hodgkin/metabolismo , Humanos
15.
Blood ; 124(20): 3118-29, 2014 Nov 13.
Artículo en Inglés | MEDLINE | ID: mdl-25232062

RESUMEN

The survival of classical Hodgkin lymphoma (cHL) cells depends on activation of NF-κB, JAK/STAT, and IRF4. Whereas these factors typically induce the master regulator of plasma cell (PC) differentiation PRDM1/BLIMP-1, levels of PRDM1 remain low in cHL. FOXO1, playing a critical role in normal B-cell development, acts as a tumor suppressor in cHL, but has never been associated with induction of PC differentiation. Here we show that FOXO1 directly upregulates the full-length isoform PRDM1α in cHL cell lines. We also observed a positive correlation between FOXO1 and PRDM1 expression levels in primary Hodgkin-Reed-Sternberg cells. Further, we show that PRDM1α acts as a tumor suppressor in cHL at least partially by blocking MYC. Here we provide a link between FOXO1 repression and PRDM1α downregulation in cHL and identify PRDM1α as a tumor suppressor in cHL. The data support a potential role for FOXO transcription factors in normal PC differentiation.


Asunto(s)
Factores de Transcripción Forkhead/metabolismo , Regulación Neoplásica de la Expresión Génica , Enfermedad de Hodgkin/genética , Enfermedad de Hodgkin/patología , Células Plasmáticas/patología , Proteínas Represoras/metabolismo , Diferenciación Celular , Línea Celular Tumoral , Regulación hacia Abajo , Proteína Forkhead Box O1 , Factores de Transcripción Forkhead/genética , Enfermedad de Hodgkin/metabolismo , Humanos , Células Plasmáticas/citología , Células Plasmáticas/metabolismo , Factor 1 de Unión al Dominio 1 de Regulación Positiva , Proteínas Proto-Oncogénicas c-myc/metabolismo , Células de Reed-Sternberg/metabolismo , Células de Reed-Sternberg/patología , Proteínas Represoras/genética , Células Tumorales Cultivadas , Regulación hacia Arriba
16.
Oncotarget ; 5(14): 5392-402, 2014 Jul 30.
Artículo en Inglés | MEDLINE | ID: mdl-24977668

RESUMEN

Recently we have shown that the transcription factor FOXO1, highly expressed in B cells, is downregulated in classical Hodgkin lymphoma (cHL). As primary mediastinal B cell lymphoma (PMBL) has similarities with the cHL transcription program we investigated FOXO1 expression in this entity. By using immunohistochemistry we found that FOXO1 was absent or expressed at low levels in 19 of 20 primary PMBL cases. PMBL cell lines reproduce the low FOXO1 expression observed in primary cases. By analyzing gene expression profiling data we found that FOXO1 expression inversely correlated with JAK2 in PMBL cases. Targeting JAK2 activity by the small molecular weight inhibitor TG101348 resulted in upregulation of FOXO1 mRNA and protein expression in MedB-1 and U2940 cell lines, and the MYC inhibitor 10058-F4 increased FOXO1 mRNA in MedB-1 cells. Moreover, in MedB-1 cells FOXO1 expression was strongly upregulated by the inhibitor of DNA methylation 5-aza-2-deoxycytidine and by the histone deacetylase inhibitor trichostatin A. Since FOXO1 promoter was unmethylated, this effect is most likely indirect. FOXO1 activation in the FOXO1-negative Med-B1 cell line led to growth arrest and apoptosis, which was accompanied by repression of MYC and BCL2L1/BCLxL. Thus, FOXO1 repression might contribute to the oncogenic program and phenotype of PMBL.


Asunto(s)
Factores de Transcripción Forkhead/genética , Linfoma de Células B/genética , Neoplasias del Mediastino/genética , Apoptosis/genética , Azacitidina/análogos & derivados , Azacitidina/farmacología , Línea Celular Tumoral , Metilación de ADN/efectos de los fármacos , Decitabina , Regulación hacia Abajo , Epigenómica , Proteína Forkhead Box O1 , Factores de Transcripción Forkhead/metabolismo , Humanos , Linfoma de Células B/metabolismo , Linfoma de Células B/patología , Neoplasias del Mediastino/metabolismo , Neoplasias del Mediastino/patología , Transducción de Señal
17.
J Pathol ; 229(5): 775-83, 2013 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-23341364

RESUMEN

Burkitt lymphoma (BL) is caused by translocation of the MYC gene to an immunoglobulin locus resulting in its constitutive expression depending on the activity of the immunoglobulin (Ig) enhancer elements. Treatment of BL cell lines with epigenetic modifiers is known to repress B-cell-specific genes and to up-regulate B-cell-inappropriate genes including the transcription repressor ID2 expression. We found that the DNA methyltransferase inhibitor decitabine/5-aza-2-deoxycytidine (5-aza-dC) represses the MYC oncogene on RNA and protein levels by inducing ID2. Down-regulation of MYC was associated with repression of transcriptional activity of the Ig locus and with inhibition of proliferation. The induction of ID2 can be in part explained by activation of the transcription factor NF-κB. We conclude that up-regulation of ID2 contributes to anti-tumour activity of 5-aza-dC via repression of Ig locus activity and consequently MYC expression.


Asunto(s)
Antimetabolitos Antineoplásicos/farmacología , Azacitidina/análogos & derivados , Linfoma de Burkitt/genética , Metilasas de Modificación del ADN/antagonistas & inhibidores , Proteínas Proto-Oncogénicas c-myc/genética , Translocación Genética/efectos de los fármacos , Azacitidina/farmacología , Linfoma de Burkitt/enzimología , Linfoma de Burkitt/metabolismo , Linfoma de Burkitt/patología , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Decitabina , Relación Dosis-Respuesta a Droga , Represión Epigenética , Regulación Neoplásica de la Expresión Génica , Humanos , Inmunoglobulina M/genética , Inmunoglobulina M/metabolismo , Proteína 2 Inhibidora de la Diferenciación/genética , Proteína 2 Inhibidora de la Diferenciación/metabolismo , FN-kappa B/metabolismo , Proteínas Proto-Oncogénicas c-myc/metabolismo , Transcripción Genética , Transfección , Regulación hacia Arriba
18.
FASEB J ; 26(12): 4990-5001, 2012 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-22935140

RESUMEN

Inactivation of FoxO proteins by phosphorylation is the result of a number of stimuli, including the insulin/IGF pathway. We were interested in the consequence of blunting this pathway by employing transgenic mice with tetracycline-controllable conditional expression of a constitutively active allele of FOXO3 under the control of the forebrain-specific CaMKIIα promoter. Although transgene-expressing mice were viable, brain weight was reduced by 30% in adult animals. Brains showed an isocortex compression with normal cortical layering, and a size reduction in regions known to depend on adult neurogenesis, i.e., the olfactory bulbs and the dentate gyrus. On postnatal activation of the transgene, adult neurogenesis was also severely affected. Investigating the molecular basis of this phenotype, we observed enhanced apoptosis starting from embryonic day E10.5 and a subsequent loss of progenitors in the ventricular/subventricular zones, but not in the isocortex or the striatum of adult mice. The enhanced apoptosis was accompanied by increased expression of PIK3IP1, which we identified as a direct transcriptional target of FOXO3. Transfection of Pik3ip1 into differentiating neural progenitors resulted in a significant reduction of viable cells. We therefore conclude that neural progenitors are particularly vulnerable to FOXO3-induced apoptosis, which is mediated by PIK3IP1, a negative PI3 kinase regulator.


Asunto(s)
Factores de Transcripción Forkhead/genética , Células-Madre Neurales/metabolismo , Prosencéfalo/metabolismo , Animales , Apoptosis/genética , Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina/genética , Proteínas Portadoras/genética , Proteínas Portadoras/metabolismo , Células Cultivadas , Análisis por Conglomerados , Proteína Forkhead Box O3 , Factores de Transcripción Forkhead/metabolismo , Perfilación de la Expresión Génica , Regulación del Desarrollo de la Expresión Génica , Immunoblotting , Péptidos y Proteínas de Señalización Intracelular , Ventrículos Laterales/embriología , Ventrículos Laterales/crecimiento & desarrollo , Ventrículos Laterales/metabolismo , Proteínas de la Membrana , Ratones , Ratones Endogámicos , Ratones Transgénicos , Mutación , Análisis de Secuencia por Matrices de Oligonucleótidos , Regiones Promotoras Genéticas/genética , Prosencéfalo/embriología , Prosencéfalo/crecimiento & desarrollo , Interferencia de ARN , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Factores de Tiempo
19.
Blood ; 119(15): 3503-11, 2012 Apr 12.
Artículo en Inglés | MEDLINE | ID: mdl-22343918

RESUMEN

The FOXO transcription factors control proliferation and apoptosis in different cell types. Their activity is regulated by posttranslational modifications, mainly by the PI3K-PKB pathway, which controls nuclear export and degradation. We show that FOXO1 is highly expressed in normal germinal center B cells as well as in non-Hodgkin lymphomas, including follicular lymphoma, diffuse large B-cell lymphoma, mucosa-associated lymphoid tissue non-Hodgkin lymphoma, B-cell chronic lymphocytic leukemia, and mantle cell lymphoma. In contrast, in 31 of 32 classical Hodgkin lymphoma (cHL) cases, Hodgkin and Reed-Sternberg cells were FOXO1 negative. Neoplastic cells of nodular lymphocyte-predominant Hodgkin lymphoma were negative in 14 of 20 cases. FOXO1 was down-regulated in cHL cell lines, whereas it was expressed in non-Hodgkin lymphoma cell lines at levels comparable with normal B cells. Ectopic expression of a constitutively active FOXO1 induced apoptosis in cHL cell lines and blocked proliferation, accompanied with cell-cycle arrest in the G(0)/G(1) phase. We found that, in cHL cell lines, FOXO1 is inactivated by multiple mechanisms, including constitutive activation of AKT/PKB and MAPK/ERK kinases and up-regulation of microRNAs miR-96, miR-182, and miR-183. These results suggest that FOXO1 repression contributes to cHL lymphomagenesis.


Asunto(s)
Factores de Transcripción Forkhead/fisiología , Genes Supresores de Tumor , Enfermedad de Hodgkin/genética , Apoptosis/genética , Línea Celular Tumoral , Proliferación Celular , Quinasas MAP Reguladas por Señal Extracelular/genética , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Quinasas MAP Reguladas por Señal Extracelular/fisiología , Proteína Forkhead Box O1 , Factores de Transcripción Forkhead/genética , Factores de Transcripción Forkhead/metabolismo , Eliminación de Gen , Regulación Neoplásica de la Expresión Génica , Genes Supresores de Tumor/fisiología , Sitios Genéticos/genética , Enfermedad de Hodgkin/patología , Humanos , MicroARNs/genética , MicroARNs/fisiología , Proteína Oncogénica v-akt/genética , Proteína Oncogénica v-akt/metabolismo , Proteína Oncogénica v-akt/fisiología , Distribución Tisular
20.
Blood ; 116(9): 1469-78, 2010 Sep 02.
Artículo en Inglés | MEDLINE | ID: mdl-20519630

RESUMEN

The transcription factor KLF4 may act both as an oncogene and a tumor suppressor in a tissue-depending manner. In T- and pre-B-cell lymphoma, KLF4 was found to act as tumor suppressor. We found the KLF4 promoter methylated in B-cell lymphoma cell lines and in primary cases of B-cell lymphomas, namely, follicular lymphoma, diffuse large B-cell lymphoma, Burkitt lymphoma, and in classic Hodgkin lymphoma (cHL) cases. Promoter hypermethylation was associated with silencing of KLF4 expression. Conditional overexpression of KLF4 in Burkitt lymphoma cell lines moderately retarded proliferation, via cell-cycle arrest in G(0)/G(1). In the cHL cell lines, KLF4 induced massive cell death that could partially be inhibited with Z-VAD.fmk. A quantitative reverse-transcribed polymerase chain reaction array revealed KLF4 target genes, including the proapoptotic gene BAK1. Using an shRNA-mediated knock-down approach, we found that BAK1 is largely responsible for KLF4-induced apoptosis. In addition, we found that KLF4 negatively regulates CXCL10, CD86, and MSC/ABF-1 genes. These genes are specifically up-regulated in HRS cells of cHL and known to be involved in establishing the cHL phenotype. We conclude that epigenetic silencing of KLF4 in B-cell lymphomas and particularly in cHL may favor lymphoma survival by loosening cell-cycle control and protecting from apoptosis.


Asunto(s)
Linfoma de Burkitt/metabolismo , Genes Supresores de Tumor , Enfermedad de Hodgkin/metabolismo , Factores de Transcripción de Tipo Kruppel/fisiología , Linfoma Folicular/metabolismo , Linfoma de Células B Grandes Difuso/metabolismo , Apoptosis , Linfocitos B/metabolismo , Biomarcadores de Tumor/genética , Biomarcadores de Tumor/metabolismo , Western Blotting , Linfoma de Burkitt/genética , Linfoma de Burkitt/patología , Ciclo Celular , Línea Celular Tumoral , Proliferación Celular , Niño , Preescolar , Metilación de ADN , ADN de Neoplasias/genética , Perfilación de la Expresión Génica , Regulación Neoplásica de la Expresión Génica , Enfermedad de Hodgkin/genética , Enfermedad de Hodgkin/patología , Humanos , Factor 4 Similar a Kruppel , Linfoma Folicular/genética , Linfoma Folicular/patología , Linfoma de Células B Grandes Difuso/genética , Linfoma de Células B Grandes Difuso/patología , Análisis de Secuencia por Matrices de Oligonucleótidos , Regiones Promotoras Genéticas , ARN Mensajero/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Proteína Destructora del Antagonista Homólogo bcl-2/antagonistas & inhibidores , Proteína Destructora del Antagonista Homólogo bcl-2/genética , Proteína Destructora del Antagonista Homólogo bcl-2/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...