Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 36
Filtrar
Más filtros













Base de datos
Intervalo de año de publicación
1.
Br J Haematol ; 204(3): 945-958, 2024 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-38296260

RESUMEN

EVI1 expression is associated with poor prognosis in myeloid leukaemia, which can result from Chr.3q alterations that juxtapose enhancers to induce EVI1 expression via long-range chromatin interactions. More often, however, EVI1 expression occurs unrelated to 3q alterations, and it remained unclear if, in these cases, EVI1 expression is similarly caused by aberrant enhancer activation. Here, we report that, in EVI1+3q- myeloid leukaemia cells, the EVI1 promoter interacts via long-range chromatin interactions with promoters of distally located, active genes, rather than with enhancer elements. Unlike in 3q+ cells, EVI1 expression and long-range interactions appear to not depend on CTCF/cohesin, though EVI1+3q- cells utilise an EVI1 promoter-proximal site to enhance its expression that is also involved in CTCF-mediated looping in 3q+ cells. Long-range interactions in 3q- cells connect EVI1 to promoters of multiple genes, whose transcription correlates with EVI1 in EVI1+3q- cell lines, suggesting a shared mechanism of transcriptional regulation. In line with this, CRISPR interference-induced silencing of two of these sites minimally, but consistently reduced EVI1 expression. Together, we provide novel evidence of features associated with EVI1 expression in 3q- leukaemia and consolidate the view that EVI1 in 3q- leukaemia is largely promoter-driven, potentially involving long-distance promoter clustering.


Asunto(s)
Leucemia Mieloide , Factores de Transcripción , Humanos , Factores de Transcripción/genética , Proteínas de Unión al ADN/genética , Cromatina , Proteína del Locus del Complejo MDS1 y EV11/genética , Leucemia Mieloide/genética , Proto-Oncogenes
2.
Blood Adv ; 7(21): 6395-6410, 2023 11 14.
Artículo en Inglés | MEDLINE | ID: mdl-37224458

RESUMEN

Deregulated expression of lineage-affiliated transcription factors (TFs) is a major mechanism of oncogenesis. However, how the deregulation of nonlineage affiliated TF affects chromatin to initiate oncogenic transcriptional programs is not well-known. To address this, we studied the chromatin effects imposed by oncogenic MAF as the cancer-initiating driver in the plasma cell cancer multiple myeloma. We found that the ectopically expressed MAF endows myeloma plasma cells with migratory and proliferative transcriptional potential. This potential is regulated by the activation of enhancers and superenhancers, previously inactive in healthy B cells and plasma cells, and the cooperation of MAF with the plasma cell-defining TF IRF4. Forced ectopic MAF expression confirms the de novo ability of oncogenic MAF to convert transcriptionally inert chromatin to active chromatin with the features of superenhancers, leading to the activation of the MAF-specific oncogenic transcriptome and the acquisition of cancer-related cellular phenotypes such as CCR1-dependent cell migration. These findings establish oncogenic MAF as a pioneer transcription factor that can initiate as well as sustain oncogenic transcriptomes and cancer phenotypes. However, despite its pioneer function, myeloma cells remain MAF-dependent, thus validating oncogenic MAF as a therapeutic target that would be able to circumvent the challenges of subsequent genetic diversification driving disease relapse and drug resistance.


Asunto(s)
Mieloma Múltiple , Humanos , Mieloma Múltiple/tratamiento farmacológico , Regulación de la Expresión Génica , Células Plasmáticas/metabolismo , Linfocitos B/metabolismo , Cromatina
3.
bioRxiv ; 2023 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-36993619

RESUMEN

In most cell types, nuclear ß-catenin functions as prominent oncogenic driver and pairs with TCF7-family factors for transcriptional activation of MYC. Surprisingly, B-lymphoid malignancies not only lacked expression and activating lesions of ß-catenin but critically depended on GSK3ß for effective ß-catenin degradation. Our interactome studies in B-lymphoid tumors revealed that ß-catenin formed repressive complexes with lymphoid-specific Ikaros factors at the expense of TCF7. Instead of MYC-activation, ß-catenin was essential to enable Ikaros-mediated recruitment of nucleosome remodeling and deacetylation (NuRD) complexes for transcriptional repression of MYC. To leverage this previously unrecognized vulnerability of B-cell-specific repressive ß-catenin-Ikaros-complexes in refractory B-cell malignancies, we examined GSK3ß small molecule inhibitors to subvert ß-catenin degradation. Clinically approved GSK3ß-inhibitors that achieved favorable safety prof les at micromolar concentrations in clinical trials for neurological disorders and solid tumors were effective at low nanomolar concentrations in B-cell malignancies, induced massive accumulation of ß-catenin, repression of MYC and acute cell death. Preclinical in vivo treatment experiments in patient-derived xenografts validated small molecule GSK3ß-inhibitors for targeted engagement of lymphoid-specific ß-catenin-Ikaros complexes as a novel strategy to overcome conventional mechanisms of drug-resistance in refractory malignancies. HIGHLIGHTS: Unlike other cell lineages, B-cells express nuclear ß-catenin protein at low baseline levels and depend on GSK3ß for its degradation.In B-cells, ß-catenin forms unique complexes with lymphoid-specific Ikaros factors and is required for Ikaros-mediated tumor suppression and assembly of repressive NuRD complexes. CRISPR-based knockin mutation of a single Ikaros-binding motif in a lymphoid MYC superenhancer region reversed ß-catenin-dependent Myc repression and induction of cell death. The discovery of GSK3ß-dependent degradation of ß-catenin as unique B-lymphoid vulnerability provides a rationale to repurpose clinically approved GSK3ß-inhibitors for the treatment of refractory B-cell malignancies. GRAPHICAL ABSTRACT: Abundant nuclear ß-cateninß-catenin pairs with TCF7 factors for transcriptional activation of MYCB-cells rely on efficient degradation of ß-catenin by GSK3ßB-cell-specific expression of Ikaros factors Unique vulnerability in B-cell tumors: GSK3ß-inhibitors induce nuclear accumulation of ß-catenin.ß-catenin pairs with B-cell-specific Ikaros factors for transcriptional repression of MYC.

5.
Nat Commun ; 12(1): 512, 2021 01 21.
Artículo en Inglés | MEDLINE | ID: mdl-33479235

RESUMEN

To achieve replicative immortality, cancer cells must activate telomere maintenance mechanisms to prevent telomere shortening. ~85% of cancers circumvent telomeric attrition by re-expressing telomerase, while the remaining ~15% of cancers induce alternative lengthening of telomeres (ALT), which relies on break-induced replication (BIR) and telomere recombination. Although ALT tumours were first reported over 20 years ago, the mechanism of ALT induction remains unclear and no study to date has described a cell-based model that permits the induction of ALT. Here, we demonstrate that infection with Kaposi's sarcoma herpesvirus (KSHV) induces sustained acquisition of ALT-like features in previously non-ALT cell lines. KSHV-infected cells acquire hallmarks of ALT activity that are also observed in KSHV-associated tumour biopsies. Down-regulating BIR impairs KSHV latency, suggesting that KSHV co-opts ALT for viral functionality. This study uncovers KSHV infection as a means to study telomere maintenance by ALT and reveals features of ALT in KSHV-associated tumours.


Asunto(s)
Neoplasias/genética , Homeostasis del Telómero/genética , Acortamiento del Telómero/genética , Telómero/genética , Carcinogénesis , Línea Celular , Línea Celular Tumoral , Daño del ADN , Replicación del ADN/genética , Células HeLa , Herpesvirus Humano 8/fisiología , Interacciones Huésped-Patógeno , Humanos , Hibridación Fluorescente in Situ , Neoplasias/patología , Neoplasias/virología , Proteoma/genética , Proteoma/metabolismo , Telomerasa/genética , Telomerasa/metabolismo
6.
J Immunol ; 202(12): 3423-3433, 2019 06 15.
Artículo en Inglés | MEDLINE | ID: mdl-31085591

RESUMEN

Induction of programmed DNA damage and its recognition and repair are fundamental for B cell development. The ssDNA-binding protein SSB1 has been described in human cells as essential for the recognition and repair of DNA damage. To study its relevance for B cells, we recently developed Ssb1 -/- and conditional Ssb1 -/- mice. Although SSB1 loss did not affect B cell development, Ssb1 -/- cells exhibited compensatory expression of its homolog SSB2. We have now generated Ssb2 -/- mice and show in this study that SSB2 is also dispensable for B cell development and DNA damage response activation. In contrast to the single loss of Ssb1 or Ssb2, however, combined SSB1/2 deficiency caused a defect in early B cell development. We relate this to the sensitivity of B cell precursors as mature B cells largely tolerated their loss. Toxicity of combined genetic SSB1/2 loss can be rescued by ectopic expression of either SSB1 or SSB2, mimicked by expression of SSB1 ssDNA-binding mutants, and attenuated by BCL2-mediated suppression of apoptosis. SSB1/2 loss in B cell precursors further caused increased exposure of ssDNA associated with disruption of genome fragile sites, inefficient cell cycle progression, and increased DNA damage if apoptosis is suppressed. As such, our results establish SSB1/2 as safeguards of B cell development and unveil their differential requirement in immature and mature B lymphocytes.


Asunto(s)
Linfocitos B/fisiología , Proteínas de Unión al ADN/metabolismo , Células Precursoras de Linfocitos B/fisiología , Proteínas Supresoras de la Señalización de Citocinas/metabolismo , Animales , Apoptosis , Diferenciación Celular , Células Cultivadas , Daño del ADN , Reparación del ADN , Proteínas de Unión al ADN/genética , Genoma/genética , Activación de Linfocitos , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Mutación/genética , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Proteínas Supresoras de la Señalización de Citocinas/genética
7.
Oncogene ; 38(17): 3216-3231, 2019 04.
Artículo en Inglés | MEDLINE | ID: mdl-30626938

RESUMEN

VCP/p97 regulates numerous cellular functions by mediating protein degradation through its segregase activity. Its key role in governing protein homoeostasis has made VCP/p97 an appealing anticancer drug target. Here, we provide evidence that VCP/p97 acts as a regulator of cellular metabolism. We found that VCP/p97 was tied to multiple metabolic processes on the gene expression level in a diverse range of cancer cell lines and in patient-derived multiple myeloma cells. Cellular VCP/p97 dependency to maintain proteostasis was increased under conditions of glucose and glutamine limitation in a range of cancer cell lines from different tissues. Moreover, glutamine depletion led to increased VCP/p97 expression, whereas VCP/p97 inhibition perturbed metabolic processes and intracellular amino acid turnover. GCN2, an amino acid-sensing kinase, attenuated stress signalling and cell death triggered by VCP/p97 inhibition and nutrient shortages and modulated ERK activation, autophagy, and glycolytic metabolite turnover. Together, our data point to an interconnected role of VCP/p97 and GCN2 in maintaining cancer cell metabolic and protein homoeostasis.


Asunto(s)
Adenosina Trifosfatasas/metabolismo , Proteínas Nucleares/metabolismo , Nutrientes/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Proteostasis/fisiología , Proteína que Contiene Valosina/metabolismo , Células A549 , Autofagia/fisiología , Línea Celular Tumoral , Expresión Génica/fisiología , Glucosa/metabolismo , Glutamina/metabolismo , Humanos , Sistema de Señalización de MAP Quinasas/fisiología , Células MCF-7 , Mieloma Múltiple/metabolismo , Células PC-3 , Proteolisis , Transducción de Señal/fisiología
8.
Cell Metab ; 27(3): 588-601.e4, 2018 03 06.
Artículo en Inglés | MEDLINE | ID: mdl-29514067

RESUMEN

Visceral adipose tissue (VAT) has multiple roles in orchestrating whole-body energy homeostasis. In addition, VAT is now considered an immune site harboring an array of innate and adaptive immune cells with a direct role in immune surveillance and host defense. We report that conventional dendritic cells (cDCs) in VAT acquire a tolerogenic phenotype through upregulation of pathways involved in adipocyte differentiation. While activation of the Wnt/ß-catenin pathway in cDC1 DCs induces IL-10 production, upregulation of the PPARγ pathway in cDC2 DCs directly suppresses their activation. Combined, they promote an anti-inflammatory milieu in vivo delaying the onset of obesity-induced chronic inflammation and insulin resistance. Under long-term over-nutrition, changes in adipocyte biology curtail ß-catenin and PPARγ activation, contributing to VAT inflammation.


Asunto(s)
Adipocitos/metabolismo , Células Dendríticas/metabolismo , Homeostasis/inmunología , Grasa Intraabdominal/inmunología , Obesidad/metabolismo , Animales , Diferenciación Celular , Inflamación/inmunología , Resistencia a la Insulina/inmunología , Interleucina-10/inmunología , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , PPAR gamma/inmunología , Vía de Señalización Wnt
9.
Cell Rep ; 18(7): 1687-1698, 2017 02 14.
Artículo en Inglés | MEDLINE | ID: mdl-28199841

RESUMEN

In human leukemia, lineage-specific genes represent predominant targets of deletion, with lymphoid-specific genes frequently affected in lymphoid leukemia and myeloid-specific genes in myeloid leukemia. To investigate the basis of lineage-specific alterations, we analyzed global DNA damage in primary B cell precursors expressing leukemia-inducing oncogenes by ChIP-seq. We identified more than 1,000 sensitive regions, of which B lineage-specific genes constitute the most prominent targets. Identified hotspots at B lineage genes relate to DNA-DSBs, affect genes that harbor genomic lesions in human leukemia, and associate with ectopic deletion in successfully transformed cells. Furthermore, we show that most identified regions overlap with gene bodies of highly expressed genes and that induction of a myeloid lineage phenotype in transformed B cell precursors promotes de novo DNA damage at myeloid loci. Hence, we demonstrate that lineage-specific transcription predisposes lineage-specific genes in transformed B cell precursors to DNA damage, which is likely to promote the frequent alteration of lineage-specific genes in human leukemia.


Asunto(s)
Linaje de la Célula/genética , Daño del ADN/genética , Leucemia/genética , Células Precursoras de Linfocitos B/patología , Transformación Genética/genética , Animales , Células Cultivadas , Expresión Génica/genética , Leucemia/patología , Linfocitos/patología , Ratones , Oncogenes/genética , Fenotipo , Transcripción Genética/genética
10.
Cell ; 162(4): 727-37, 2015 Aug 13.
Artículo en Inglés | MEDLINE | ID: mdl-26276629

RESUMEN

Chronic infection with Plasmodium falciparum was epidemiologically associated with endemic Burkitt's lymphoma, a mature B cell cancer characterized by chromosome translocation between the c-myc oncogene and Igh, over 50 years ago. Whether infection promotes B cell lymphoma, and if so by which mechanism, remains unknown. To investigate the relationship between parasitic disease and lymphomagenesis, we used Plasmodium chabaudi (Pc) to produce chronic malaria infection in mice. Pc induces prolonged expansion of germinal centers (GCs), unique compartments in which B cells undergo rapid clonal expansion and express activation-induced cytidine deaminase (AID), a DNA mutator. GC B cells elicited during Pc infection suffer widespread DNA damage, leading to chromosome translocations. Although infection does not change the overall rate, it modifies lymphomagenesis to favor mature B cell lymphomas that are AID dependent and show chromosome translocations. Thus, malaria infection favors mature B cell cancers by eliciting protracted AID expression in GC B cells. PAPERCLIP.


Asunto(s)
Inestabilidad Genómica , Linfoma de Células B/genética , Malaria/complicaciones , Malaria/genética , Plasmodium chabaudi/fisiología , Animales , Linfocitos B/patología , Enfermedad Crónica , Citidina Desaminasa/metabolismo , Replicación del ADN , Genes p53 , Centro Germinal/parasitología , Malaria/parasitología , Malaria/patología , Ratones , Translocación Genética
11.
Hum Mutat ; 36(2): 187-90, 2015 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-25504470

RESUMEN

Cerebro-costo-mandibular syndrome (CCMS) is a developmental disorder characterized by the association of Pierre Robin sequence and posterior rib defects. Exome sequencing and Sanger sequencing in five unrelated CCMS patients revealed five heterozygous variants in the small nuclear ribonucleoprotein polypeptides B and B1 (SNRPB) gene. This gene includes three transcripts, namely transcripts 1 and 2, encoding components of the core spliceosomal machinery (SmB' and SmB) and transcript 3 undergoing nonsense-mediated mRNA decay. All variants were located in the premature termination codon (PTC)-introducing alternative exon of transcript 3. Quantitative RT-PCR analysis revealed a significant increase in transcript 3 levels in leukocytes of CCMS individuals compared to controls. We conclude that CCMS is due to heterozygous mutations in SNRPB, enhancing inclusion of a SNRPB PTC-introducing alternative exon, and show that this developmental disease is caused by defects in the splicing machinery. Our finding confirms the report of SNRPB mutations in CCMS patients by Lynch et al. (2014) and further extends the clinical and molecular observations.


Asunto(s)
Discapacidad Intelectual/genética , Micrognatismo/genética , Costillas/anomalías , Proteínas Nucleares snRNP/genética , Adolescente , Adulto , Secuencia de Bases , Preescolar , Estudios de Asociación Genética , Heterocigoto , Humanos , Masculino , Mutación Missense , Adulto Joven
12.
Mol Cell ; 49(4): 623-31, 2013 Feb 21.
Artículo en Inglés | MEDLINE | ID: mdl-23290917

RESUMEN

Deficiencies in factors that regulate the DNA damage response enhance the incidence of malignancy by destabilizing the genome. However, the precise influence of the DNA damage response on regulation of cancer-associated rearrangements is not well defined. Here we examine the genome-wide impact of tumor protein P53-binding protein 1 (53BP1) deficiency in lymphoma and translocation. While both activation-induced cytidine deaminase (AID) and 53BP1 have been associated with cancer in humans, neither AID overexpression nor loss of 53BP1 is sufficient to produce malignancy. However, the combination of 53BP1 deficiency and AID deregulation results in B cell lymphoma. Deep sequencing of the genome of 53BP1(-/-) cancer cells and translocation capture sequencing (TC-Seq) of primary 53BP1(-/-) B cells revealed that their chromosomal rearrangements differ from those found in wild-type cells in that they show increased DNA end resection. Moreover, loss of 53BP1 alters the translocatome by increasing rearrangements to intergenic regions.


Asunto(s)
Transformación Celular Neoplásica/genética , Proteínas Cromosómicas no Histona/fisiología , Citidina Desaminasa/fisiología , Proteínas de Unión al ADN/fisiología , Reordenamiento Génico , Linfoma de Células B/metabolismo , Animales , Células Cultivadas , Proteínas Cromosómicas no Histona/deficiencia , Proteínas Cromosómicas no Histona/genética , Cromosomas de los Mamíferos/genética , Citidina Desaminasa/genética , Citidina Desaminasa/metabolismo , Proteínas de Unión al ADN/deficiencia , Proteínas de Unión al ADN/genética , Epigénesis Genética , Genes Supresores de Tumor , Estudio de Asociación del Genoma Completo , Linfoma de Células B/genética , Ratones , Ratones Noqueados , Mutación , Análisis de Secuencia de ADN , Transcripción Genética , Translocación Genética , Proteína 1 de Unión al Supresor Tumoral P53
13.
Science ; 339(6120): 711-5, 2013 Feb 08.
Artículo en Inglés | MEDLINE | ID: mdl-23306439

RESUMEN

DNA double-strand breaks (DSBs) represent a threat to the genome because they can lead to the loss of genetic information and chromosome rearrangements. The DNA repair protein p53 binding protein 1 (53BP1) protects the genome by limiting nucleolytic processing of DSBs by a mechanism that requires its phosphorylation, but whether 53BP1 does so directly is not known. Here, we identify Rap1-interacting factor 1 (Rif1) as an ATM (ataxia-telangiectasia mutated) phosphorylation-dependent interactor of 53BP1 and show that absence of Rif1 results in 5'-3' DNA-end resection in mice. Consistent with enhanced DNA resection, Rif1 deficiency impairs DNA repair in the G(1) and S phases of the cell cycle, interferes with class switch recombination in B lymphocytes, and leads to accumulation of chromosome DSBs.


Asunto(s)
Proteínas Cromosómicas no Histona/metabolismo , Roturas del ADN de Doble Cadena , Proteínas de Unión al ADN/metabolismo , ADN/metabolismo , Cambio de Clase de Inmunoglobulina , Proteínas de Unión a Telómeros/metabolismo , Animales , Proteínas de la Ataxia Telangiectasia Mutada , Linfocitos B/inmunología , Linfocitos B/metabolismo , Proteínas de Ciclo Celular/antagonistas & inhibidores , Proteínas de Ciclo Celular/metabolismo , Células Cultivadas , Reparación del ADN , Proteínas de Unión al ADN/antagonistas & inhibidores , Fase G1 , Fase G2 , Inestabilidad Genómica , Ratones , Fosforilación , Proteínas Serina-Treonina Quinasas/antagonistas & inhibidores , Proteínas Serina-Treonina Quinasas/metabolismo , Fase S , Proteínas Supresoras de Tumor/antagonistas & inhibidores , Proteínas Supresoras de Tumor/metabolismo , Proteína 1 de Unión al Supresor Tumoral P53
14.
EMBO J ; 31(20): 4045-56, 2012 Oct 17.
Artículo en Inglés | MEDLINE | ID: mdl-22940690

RESUMEN

Human single-stranded DNA-binding protein 1 (hSSB1), encoded by OBFC2B, was recently characterized as an essential factor for the initiation of DNA damage checkpoints and the maintenance of genomic stability. Here, we report that loss of Obfc2b in mice results in perinatal lethality characterized by growth delay and skeletal abnormalities. These abnormalities are associated with accumulation of γH2ax, apoptosis and defective pre-cartilage condensation, which is essential for normal bone formation. However, deficiency of Obfc2b does not affect the initiation of DNA damage checkpoints, Atm activation, or the maintenance of genomic stability in B lymphocytes and primary fibroblasts. Loss of Obfc2b results in increased expression of its homologue Obfc2a (hSSB2). In contrast to Obfc2b deficiency, depletion of Obfc2a in fibroblasts results in impaired proliferation, accumulation of γH2ax and increased genomic instability. Thus, the hSSB1 orthologue Obfc2b has a unique function during embryogenesis limited to cell types that contribute to bone formation. While being dispensable in most other cell lineages, its absence leads to a compensatory increase in Obfc2a protein, a homologue required for the maintenance of genomic integrity.


Asunto(s)
Daño del ADN , Reparación del ADN/genética , Proteínas de Unión al ADN/deficiencia , Proteínas Mitocondriales/deficiencia , Osteogénesis/genética , Animales , Apoptosis/genética , Linfocitos B/metabolismo , Huesos/anomalías , Huesos/embriología , División Celular , Condrocitos/metabolismo , Replicación del ADN , Proteínas de Unión al ADN/biosíntesis , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/fisiología , Fibroblastos/metabolismo , Regulación del Desarrollo de la Expresión Génica , Genes Letales , Genes p53 , Histonas/metabolismo , Humanos , Mesodermo/anomalías , Ratones , Ratones Noqueados , Proteínas Mitocondriales/genética , Proteínas Mitocondriales/fisiología , Especificidad de Órganos , Osteoblastos/metabolismo , Osteoclastos/metabolismo
15.
Oncoimmunology ; 1(5): 618-629, 2012 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-22934254

RESUMEN

Although cure rates for acute lymphoblastic leukemia (ALL) have increased, development of resistance to drugs and patient relapse are common. The environment in which the leukemia cells are present during the drug treatment is known to provide significant survival benefit. Here, we have modeled this process by culturing murine Bcr/Abl-positive acute lymphoblastic leukemia cells in the presence of stroma while treating them with a moderate dose of two unrelated drugs, the farnesyltransferase inhibitor lonafarnib and the tyrosine kinase inhibitor nilotinib. This results in an initial large reduction in cell viability of the culture and inhibition of cell proliferation. However, after a number of days, cell death ceases and the culture becomes drug-tolerant, enabling cell division to resume. Using gene expression profiling, we found that the development of drug resistance was accompanied by massive transcriptional upregulation of genes that are associated with general inflammatory responses such as the metalloproteinase MMP9. MMP9 protein levels and enzymatic activity were also increased in ALL cells that had become nilotinib-tolerant. Activation of p38, Akt and Erk correlated with the development of environment-mediated drug resistance (EMDR), and inhibitors of Akt and Erk in combination with nilotinib reduced the ability of the cells to develop resistance. However, inhibition of p38 promoted increased resistance to nilotinib. We conclude that development of EMDR by ALL cells involves changes in numerous intracellular pathways. Development of tolerance to drugs such as nilotinib may therefore be circumvented by simultaneous treatment with other drugs having divergent targets.

16.
BMC Genomics ; 13: 418, 2012 Aug 23.
Artículo en Inglés | MEDLINE | ID: mdl-22916712

RESUMEN

BACKGROUND: Acute lymphoblastic leukemia (ALL) cells treated with drugs can become drug-tolerant if co-cultured with protective stromal mouse embryonic fibroblasts (MEFs). RESULTS: We performed transcriptional profiling on these stromal fibroblasts to investigate if they were affected by the presence of drug-treated ALL cells. These mitotically inactivated MEFs showed few changes in gene expression, but a family of sequences of which transcription is significantly increased was identified. A sequence related to this family, which we named cassini, was selected for further characterization. We found that cassini was highly upregulated in drug-treated ALL cells. Analysis of RNAs from different normal mouse tissues showed that cassini expression is highest in spleen and thymus, and can be further enhanced in these organs by exposure of mice to bacterial endotoxin. Heat shock, but not other types of stress, significantly induced the transcription of this locus in ALL cells. Transient overexpression of cassini in human 293 embryonic kidney cells did not increase the cytotoxic or cytostatic effects of chemotherapeutic drugs but provided some protection. Database searches revealed that sequences highly homologous to cassini are present in rodents, apicomplexans, flatworms and primates, indicating that they are conserved in evolution. Moreover, CASSINI RNA was induced in human ALL cells treated with vincristine. Surprisingly, cassini belongs to the previously reported murine family of γ-satellite/major satellite DNA sequences, which were not known to be present in other species. CONCLUSIONS: Our results show that the transcription of at least one member of these sequences is regulated, suggesting that this has a function in normal and transformed immune cells. Expression of these sequences may protect cells when they are exposed to specific stress stimuli.


Asunto(s)
Proteínas/metabolismo , ARN/metabolismo , Animales , Antineoplásicos Fitogénicos/toxicidad , Apicomplexa , Evolución Biológica , Línea Celular Tumoral , Técnicas de Cocultivo , Bases de Datos Genéticas , Endotoxinas/toxicidad , Fibroblastos/metabolismo , Perfilación de la Expresión Génica , Sitios Genéticos , Células HEK293 , Humanos , Ratones , Ratones Transgénicos , Platelmintos , Leucemia-Linfoma Linfoblástico de Células Precursoras/genética , Leucemia-Linfoma Linfoblástico de Células Precursoras/metabolismo , Células Precursoras de Linfocitos B/efectos de los fármacos , Células Precursoras de Linfocitos B/metabolismo , Primates , Proteínas/genética , Pirimidinas/toxicidad , ARN/análisis , ARN/genética , Temperatura , Transfección , Regulación hacia Arriba/efectos de los fármacos , Vincristina/toxicidad
17.
Mol Cell ; 42(3): 319-29, 2011 May 06.
Artículo en Inglés | MEDLINE | ID: mdl-21549309

RESUMEN

53BP1 is a DNA damage protein that forms phosphorylated H2AX (γ-H2AX) dependent foci in a 1 Mb region surrounding DNA double-strand breaks (DSBs). In addition, 53BP1 promotes genomic stability by regulating the metabolism of DNA ends. We have compared the joining rates of paired DSBs separated by 1.2 kb to 27 Mb on chromosome 12 in the presence or absence of 53BP1. 53BP1 facilitates joining of intrachromosomal DSBs but only at distances corresponding to γ-H2AX spreading. In contrast, DNA end protection by 53BP1 is distance independent. Furthermore, analysis of 53BP1 mutants shows that chromatin association, oligomerization, and N-terminal ATM phosphorylation are all required for DNA end protection and joining as measured by immunoglobulin class switch recombination. These data elucidate the molecular events that are required for 53BP1 to maintain genomic stability and point to a model wherein 53BP1 and H2AX cooperate to repress resection of DSBs.


Asunto(s)
Proteínas Cromosómicas no Histona/genética , Roturas del ADN de Doble Cadena , Proteínas de Unión al ADN/genética , Cambio de Clase de Inmunoglobulina/genética , Animales , Linfocitos B/metabolismo , Sitios de Unión , Western Blotting , Células Cultivadas , Cromatina/metabolismo , Proteínas Cromosómicas no Histona/química , Proteínas Cromosómicas no Histona/metabolismo , Reparación del ADN , Proteínas de Unión al ADN/química , Proteínas de Unión al ADN/metabolismo , Citometría de Flujo , Histonas/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Modelos Genéticos , Fosforilación , Multimerización de Proteína , Recombinación Genética , Proteína 1 de Unión al Supresor Tumoral P53
18.
Cell ; 141(2): 243-54, 2010 Apr 16.
Artículo en Inglés | MEDLINE | ID: mdl-20362325

RESUMEN

Defective DNA repair by homologous recombination (HR) is thought to be a major contributor to tumorigenesis in individuals carrying Brca1 mutations. Here, we show that DNA breaks in Brca1-deficient cells are aberrantly joined into complex chromosome rearrangements by a process dependent on the nonhomologous end-joining (NHEJ) factors 53BP1 and DNA ligase 4. Loss of 53BP1 alleviates hypersensitivity of Brca1 mutant cells to PARP inhibition and restores error-free repair by HR. Mechanistically, 53BP1 deletion promotes ATM-dependent processing of broken DNA ends to produce recombinogenic single-stranded DNA competent for HR. In contrast, Lig4 deficiency does not rescue the HR defect in Brca1 mutant cells but prevents the joining of chromatid breaks into chromosome rearrangements. Our results illustrate that HR and NHEJ compete to process DNA breaks that arise during DNA replication and that shifting the balance between these pathways can be exploited to selectively protect or kill cells harboring Brca1 mutations.


Asunto(s)
Proteína BRCA1/genética , Reparación del ADN , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Animales , Linfocitos B/metabolismo , Proteínas Cromosómicas no Histona , Roturas del ADN , Proteínas de Unión al ADN , Femenino , Inestabilidad Genómica , Humanos , Ratones , Proteína 1 de Unión al Supresor Tumoral P53
19.
J Exp Med ; 207(4): 855-65, 2010 Apr 12.
Artículo en Inglés | MEDLINE | ID: mdl-20368578

RESUMEN

Class switch recombination (CSR) diversifies antibodies by joining highly repetitive DNA elements, which are separated by 60-200 kbp. CSR is initiated by activation-induced cytidine deaminase, an enzyme that produces multiple DNA double-strand breaks (DSBs) in switch regions. Switch regions are joined by a mechanism that requires an intact DNA damage response and classical or alternative nonhomologous end joining (A-NHEJ). Among the DNA damage response factors, 53BP1 has the most profound effect on CSR. We explore the role of 53BP1 in intrachromosomal DNA repair using I-SceI to introduce paired DSBs in the IgH locus. We find that the absence of 53BP1 results in an ataxia telangiectasia mutated-dependent increase in DNA end resection and that resected DNA is preferentially repaired by microhomology-mediated A-NHEJ. We propose that 53BP1 favors long-range CSR in part by protecting DNA ends against resection, which prevents A-NHEJ-dependent short-range rejoining of intra-switch region DSBs.


Asunto(s)
Reparación del ADN/fisiología , Cambio de Clase de Inmunoglobulina/fisiología , Péptidos y Proteínas de Señalización Intracelular/fisiología , Recombinación Genética/fisiología , Animales , Proteínas de la Ataxia Telangiectasia Mutada , Linfocitos B/efectos de los fármacos , Linfocitos B/metabolismo , Proteínas de Ciclo Celular/antagonistas & inhibidores , Proteínas de Ciclo Celular/metabolismo , Proteínas Cromosómicas no Histona , Emparejamiento Cromosómico/genética , Citidina Desaminasa/genética , Roturas del ADN de Doble Cadena , Proteínas de Unión al ADN/antagonistas & inhibidores , Proteínas de Unión al ADN/metabolismo , Desoxirribonucleasas de Localización Especificada Tipo II/genética , Cadenas Pesadas de Inmunoglobulina/genética , Integrasas/genética , Interleucina-4/farmacología , Lipopolisacáridos/farmacología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Proteínas Serina-Treonina Quinasas/antagonistas & inhibidores , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Supresoras de Tumor/antagonistas & inhibidores , Proteínas Supresoras de Tumor/metabolismo , Proteína 1 de Unión al Supresor Tumoral P53
20.
Mol Cell ; 36(4): 631-41, 2009 Nov 25.
Artículo en Inglés | MEDLINE | ID: mdl-19941823

RESUMEN

Cancer-initiating translocations such as those associated with lymphomas require the formation of paired DNA double-strand breaks (DSBs). Activation-induced cytidine deaminase (AID) produces widespread somatic mutation in mature B cells; however, the extent of "off-target" DSB formation and its role in translocation-associated malignancy is unknown. Here, we show that deregulated expression of AID causes widespread genome instability, which alone is insufficient to induce B cell lymphoma; transformation requires concomitant loss of the tumor suppressor p53. Mature B cell lymphomas arising as a result of deregulated AID expression are phenotypically diverse and harbor clonal reciprocal translocations involving a group of Immunoglobulin (Ig) and non-Ig genes that are direct targets of AID. This group includes miR-142, a previously unknown micro-RNA target that is translocated in human B cell malignancy. We conclude that AID produces DSBs throughout the genome, which can lead to lymphoma-associated chromosome translocations in mature B cells.


Asunto(s)
Cromosomas de los Mamíferos/genética , Citidina Desaminasa/metabolismo , Roturas del ADN de Doble Cadena , Genes de Inmunoglobulinas/genética , Linfoma de Células B/enzimología , Linfoma de Células B/genética , Translocación Genética , Animales , Linfocitos B/citología , Linfocitos B/enzimología , Diferenciación Celular/genética , Células Cultivadas , Inestabilidad Cromosómica/genética , Daño del ADN , Humanos , Cambio de Clase de Inmunoglobulina/genética , Cariotipificación , Linfoma de Células B/patología , Ratones , Ratones Transgénicos , MicroARNs/metabolismo , Fenotipo , Proteínas Proto-Oncogénicas c-myc/genética , Hipermutación Somática de Inmunoglobulina/genética , Proteína p53 Supresora de Tumor/deficiencia
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA