Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
1.
Blood ; 122(2): 219-26, 2013 Jul 11.
Artículo en Inglés | MEDLINE | ID: mdl-23699600

RESUMEN

Outcome in multiple myeloma is highly variable and a better understanding of the factors that influence disease biology is essential to understand and predict behavior in individual patients. In the present study, we analyzed combined genomewide DNA methylation and gene expression data of patients treated in the Medical Research Council Myeloma IX trial. We used these data to identify epigenetically repressed tumor suppressor genes with prognostic relevance in myeloma. We identified 195 genes with changes in methylation status that were significantly associated with prognosis. Combining DNA methylation and gene expression data led to the identification of the epigenetically regulated tumor modulating genes GPX3, RBP1, SPARC, and TGFBI. Hypermethylation of these genes was associated with significantly shorter overall survival, independent of age, International Staging System score, and adverse cytogenetics. The 4 differentially methylated and expressed genes are known to mediate important tumor suppressive functions including response to chemotherapy (TGFBI), interaction with the microenvironment (SPARC), retinoic acid signaling (RBP1), and the response to oxidative stress (GPX3), which could explain the prognostic impact of their differential methylation. Assessment of the DNA methylation status of the identified genes could contribute to the molecular characterization of myeloma, which is prerequisite for an individualized treatment approach.


Asunto(s)
Metilación de ADN , Epigénesis Genética , Perfilación de la Expresión Génica , Genes Supresores de Tumor , Mieloma Múltiple/genética , Adulto , Anciano , Anciano de 80 o más Años , Azacitidina/análogos & derivados , Azacitidina/farmacología , Aberraciones Cromosómicas , Metilación de ADN/efectos de los fármacos , Decitabina , Progresión de la Enfermedad , Femenino , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Masculino , Persona de Mediana Edad , Mieloma Múltiple/mortalidad , Osteonectina , Fenotipo , Pronóstico , Factor de Crecimiento Transformador beta1/genética , Proteínas Supresoras de Tumor/genética
2.
J Immunol ; 189(9): 4459-69, 2012 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-23024272

RESUMEN

The closely linked human IL-3 and GM-CSF genes are tightly regulated and are expressed in activated T cells and mast cells. In this study, we used transgenic mice to study the developmental regulation of this locus and to identify DNA elements required for its correct activity in vivo. Because these two genes are separated by a CTCF-dependent insulator, and the GM-CSF gene is regulated primarily by its own upstream enhancer, the main objective in this study was to identify regions of the locus required for correct IL-3 gene expression. We initially found that the previously identified proximal upstream IL-3 enhancers were insufficient to account for the in vivo activity of the IL-3 gene. However, an extended analysis of DNase I-hypersensitive sites (DHSs) spanning the entire upstream IL-3 intergenic region revealed the existence of a complex cluster of both constitutive and inducible DHSs spanning the -34- to -40-kb region. The tissue specificity of these DHSs mirrored the activity of the IL-3 gene, and included a highly inducible cyclosporin A-sensitive enhancer at -37 kb that increased IL-3 promoter activity 40-fold. Significantly, inclusion of this region enabled correct in vivo regulation of IL-3 gene expression in T cells, mast cells, and myeloid progenitor cells.


Asunto(s)
Elementos de Facilitación Genéticos/inmunología , Regulación del Desarrollo de la Expresión Génica/inmunología , Factor Estimulante de Colonias de Granulocitos y Macrófagos/biosíntesis , Factor Estimulante de Colonias de Granulocitos y Macrófagos/genética , Interleucina-3/biosíntesis , Interleucina-3/genética , Animales , Línea Celular , Línea Celular Tumoral , Células Cultivadas , Desoxirribonucleasa I/genética , Elementos de Facilitación Genéticos/genética , Sitios Genéticos/inmunología , Humanos , Células Jurkat , Ratones , Ratones Transgénicos , Distribución Tisular/genética , Distribución Tisular/inmunología
3.
Br J Haematol ; 162(3): 348-59, 2013 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-23718138

RESUMEN

Multiple myeloma (MM) is a heterogeneous disease. International Staging System/fluorescence hybridization (ISS/FISH)-based model and gene expression profiles (GEP) are effective approaches to define clinical outcome, although yet to be improved. The discovery of a class of small non-coding RNAs (micro RNAs, miRNAs) has revealed a new level of biological complexity underlying the regulation of gene expression. In this work, 163 presenting samples from MM patients were analysed by global miRNA profiling, and distinct miRNA expression characteristics in molecular subgroups with prognostic relevance (4p16, MAF and 11q13 translocations) were identified. Furthermore we developed an "outcome classifier", based on the expression of two miRNAs (MIR17 and MIR886-5p), which is able to stratify patients into three risk groups (median OS 19.4, 40.6 and 65.3 months, P = 0.001). The miRNA-based classifier significantly improved the predictive power of the ISS/FISH approach (P = 0.0004), and was independent of GEP-derived prognostic signatures (P < 0.002). Through integrative genomics analysis, we outlined the potential biological relevance of the miRNAs included in the classifier and their putative roles in regulating a large number of genes involved in MM biology. This is the first report showing that miRNAs can be built into molecular diagnostic strategies for risk stratification in MM.


Asunto(s)
Biomarcadores de Tumor/genética , MicroARNs/genética , Mieloma Múltiple/diagnóstico , Mieloma Múltiple/genética , ARN Neoplásico/genética , Anciano , Aberraciones Cromosómicas , Perfilación de la Expresión Génica/métodos , Regulación Neoplásica de la Expresión Génica , Humanos , Hibridación Fluorescente in Situ/métodos , Estimación de Kaplan-Meier , Persona de Mediana Edad , Pronóstico , Medición de Riesgo/métodos
4.
J Immunol ; 184(6): 3043-54, 2010 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-20147630

RESUMEN

The closely linked IL-3 and GM-CSF genes are located within a cluster of cytokine genes co-expressed in activated T cells. Their activation in response to TCR signaling pathways is controlled by specific, inducible upstream enhancers. To study the developmental regulation of this locus in T lineage cells, we created a transgenic mouse model encompassing the human IL-3 and GM-CSF genes plus the known enhancers. We demonstrated that the IL-3/GM-CSF locus undergoes progressive stages of activation, with stepwise increases in active modifications and the proportion of cytokine-expressing cells, throughout the course of T cell differentiation. Looking first at immature cells, we found that the IL-3/GM-CSF locus was epigenetically silent in CD4/CD8 double positive thymocytes, thereby minimizing the potential for inappropriate activation during the course of TCR selection. Furthermore, we demonstrated that the locus did not reach its maximal transcriptional potential until after T cells had undergone blast cell transformation to become fully activated proliferating T cells. Inducible locus activation in mature T cells was accompanied by noncoding transcription initiating within the enhancer elements. Significantly, we also found that memory CD4 positive T cells, but not naive T cells, maintain a remodeled chromatin structure resembling that seen in T blast cells.


Asunto(s)
Diferenciación Celular/inmunología , Epigénesis Genética/inmunología , Silenciador del Gen/inmunología , Sitios Genéticos/inmunología , Factor Estimulante de Colonias de Granulocitos y Macrófagos/genética , Interleucina-3/genética , Activación de Linfocitos/genética , Subgrupos de Linfocitos T/inmunología , Animales , Diferenciación Celular/genética , Células Cultivadas , Cromatina/química , Cromatina/metabolismo , Citocinas/biosíntesis , Desoxirribonucleasa I/genética , Factor Estimulante de Colonias de Granulocitos y Macrófagos/biosíntesis , Humanos , Memoria Inmunológica/genética , Interleucina-3/biosíntesis , Ratones , Ratones Transgénicos , Familia de Multigenes/inmunología , ARN/biosíntesis , Subgrupos de Linfocitos T/citología , Subgrupos de Linfocitos T/metabolismo , Timo/citología , Timo/inmunología , Timo/metabolismo , Activación Transcripcional/inmunología
5.
Clin Cancer Res ; 24(10): 2395-2407, 2018 05 15.
Artículo en Inglés | MEDLINE | ID: mdl-29391353

RESUMEN

Purpose: Myeloma is a plasma cell malignancy characterized by the overproduction of immunoglobulin, and is therefore susceptible to therapies targeting protein homeostasis. We hypothesized that heat shock factor 1 (HSF1) was an attractive therapeutic target for myeloma due to its direct regulation of transcriptional programs implicated in both protein homeostasis and the oncogenic phenotype. Here, we interrogate HSF1 as a therapeutic target in myeloma using bioinformatic, genetic, and pharmacologic means.Experimental Design: To assess the clinical relevance of HSF1, we analyzed publicly available patient myeloma gene expression datasets. Validation of this novel target was conducted in in vitro experiments using shRNA or inhibitors of the HSF1 pathway in human myeloma cell lines and primary cells as well as in in vivo human myeloma xenograft models.Results: Expression of HSF1 and its target genes were associated with poorer myeloma patient survival. ShRNA-mediated knockdown or pharmacologic inhibition of the HSF1 pathway with a novel chemical probe, CCT251236, or with KRIBB11, led to caspase-mediated cell death that was associated with an increase in EIF2α phosphorylation, CHOP expression and a decrease in overall protein synthesis. Importantly, both CCT251236 and KRIBB11 induced cytotoxicity in human myeloma cell lines and patient-derived primary myeloma cells with a therapeutic window over normal cells. Pharmacologic inhibition induced tumor growth inhibition and was well-tolerated in a human myeloma xenograft murine model with evidence of pharmacodynamic biomarker modulation.Conclusions: Taken together, our studies demonstrate the dependence of myeloma cells on HSF1 for survival and support the clinical evaluation of pharmacologic inhibitors of the HSF1 pathway in myeloma. Clin Cancer Res; 24(10); 2395-407. ©2018 AACRSee related commentary by Parekh, p. 2237.


Asunto(s)
Biomarcadores de Tumor , Supervivencia Celular/genética , Factores de Transcripción del Choque Térmico/genética , Mieloma Múltiple/genética , Animales , Antineoplásicos/farmacología , Apoptosis/genética , Caspasas/metabolismo , Línea Celular Tumoral , Biología Computacional/métodos , Modelos Animales de Enfermedad , Perfilación de la Expresión Génica , Técnicas de Silenciamiento del Gen , Factores de Transcripción del Choque Térmico/antagonistas & inhibidores , Factores de Transcripción del Choque Térmico/metabolismo , Humanos , Estimación de Kaplan-Meier , Ratones , Terapia Molecular Dirigida , Mieloma Múltiple/tratamiento farmacológico , Mieloma Múltiple/metabolismo , Mieloma Múltiple/mortalidad , Transducción de Señal , Ensayos Antitumor por Modelo de Xenoinjerto
6.
Blood Cancer J ; 9(1): 1, 2018 12 21.
Artículo en Inglés | MEDLINE | ID: mdl-30602759

RESUMEN

The clustering of different types of B-cell malignancies in families raises the possibility of shared aetiology. To examine this, we performed cross-trait linkage disequilibrium (LD)-score regression of multiple myeloma (MM) and chronic lymphocytic leukaemia (CLL) genome-wide association study (GWAS) data sets, totalling 11,734 cases and 29,468 controls. A significant genetic correlation between these two B-cell malignancies was shown (Rg = 0.4, P = 0.0046). Furthermore, four of the 45 known CLL risk loci were shown to associate with MM risk and five of the 23 known MM risk loci associate with CLL risk. By integrating eQTL, Hi-C and ChIP-seq data, we show that these pleiotropic risk loci are enriched for B-cell regulatory elements and implicate B-cell developmental genes. These data identify shared biological pathways influencing the development of CLL and, MM and further our understanding of the aetiological basis of these B-cell malignancies.


Asunto(s)
Estudios de Asociación Genética , Predisposición Genética a la Enfermedad , Leucemia Linfocítica Crónica de Células B/genética , Mieloma Múltiple/genética , Alelos , Estudios de Casos y Controles , Bases de Datos Genéticas , Ligamiento Genético , Estudio de Asociación del Genoma Completo , Humanos , Desequilibrio de Ligamiento , Especificidad de Órganos/genética , Polimorfismo de Nucleótido Simple , Sitios de Carácter Cuantitativo
7.
Nat Commun ; 7: 13656, 2016 11 24.
Artículo en Inglés | MEDLINE | ID: mdl-27882933

RESUMEN

Genome-wide association studies have identified several risk loci for multiple myeloma (MM); however, the mechanisms by which they influence MM are unknown. Here by using genetic association data and functional characterization, we demonstrate that rs4487645 G>T, the most highly associated variant (P = 5.30 × 10-25), resides in an enhancer element 47 kb upstream of the transcription start site of c-Myc-interacting CDCA7L. The G-risk allele, associated with increased CDCA7L expression (P=1.95 × 10-36), increases IRF4 binding and the enhancer interacts with the CDCA7L promoter. We show that suppression of CDCA7L limits MM proliferation through apoptosis, and increased CDCA7L expression is associated with adverse patient survival. These findings implicate IRF4-mediated CDCA7L expression in MM biology and indicate how germline variation might confer susceptibility to MM.


Asunto(s)
Apoptosis/genética , Elementos de Facilitación Genéticos/genética , Regulación Neoplásica de la Expresión Génica , Factores Reguladores del Interferón/metabolismo , Mieloma Múltiple/genética , Proteínas Represoras/genética , Alelos , Línea Celular Tumoral , Predisposición Genética a la Enfermedad , Humanos , Mieloma Múltiple/metabolismo , Polimorfismo de Nucleótido Simple , Regiones Promotoras Genéticas , Unión Proteica/genética , Proteínas Proto-Oncogénicas c-myc/metabolismo , Sitios de Carácter Cuantitativo , Proteínas Represoras/metabolismo
8.
J Clin Oncol ; 33(33): 3911-20, 2015 Nov 20.
Artículo en Inglés | MEDLINE | ID: mdl-26282654

RESUMEN

PURPOSE: At the molecular level, myeloma is characterized by copy number abnormalities and recurrent translocations into the immunoglobulin heavy chain locus. Novel methods, such as massively parallel sequencing, have begun to describe the pattern of tumor-acquired mutations, but their clinical relevance has yet to be established. METHODS: We performed whole-exome sequencing for 463 patients who presented with myeloma and were enrolled onto the National Cancer Research Institute Myeloma XI trial, for whom complete molecular cytogenetic and clinical outcome data were available. RESULTS: We identified 15 significantly mutated genes: IRF4, KRAS, NRAS, MAX, HIST1H1E, RB1, EGR1, TP53, TRAF3, FAM46C, DIS3, BRAF, LTB, CYLD, and FGFR3. The mutational spectrum is dominated by mutations in the RAS (43%) and nuclear factor-κB (17%) pathways, but although they are prognostically neutral, they could be targeted therapeutically. Mutations in CCND1 and DNA repair pathway alterations (TP53, ATM, ATR, and ZNFHX4 mutations) are associated with a negative impact on survival. In contrast, those in IRF4 and EGR1 are associated with a favorable overall survival. We combined these novel mutation risk factors with the recurrent molecular adverse features and international staging system to generate an international staging system mutation score that can identify a high-risk population of patients who experience relapse and die prematurely. CONCLUSION: We have refined our understanding of genetic events in myeloma and identified clinically relevant mutations that may be used to better stratify patients at presentation.


Asunto(s)
Variaciones en el Número de Copia de ADN/genética , Predisposición Genética a la Enfermedad/epidemiología , Mieloma Múltiple/genética , Mieloma Múltiple/mortalidad , Proteínas ras/genética , Adulto , Análisis Mutacional de ADN , Femenino , Secuenciación de Nucleótidos de Alto Rendimiento , Humanos , Masculino , Persona de Mediana Edad , Mieloma Múltiple/fisiopatología , Análisis Multivariante , Modelos de Riesgos Proporcionales , Estudios Prospectivos , Análisis de Supervivencia , Reino Unido , Adulto Joven
9.
PLoS One ; 9(6): e99493, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24923560

RESUMEN

The chromosomal translocation t(4;14) deregulates MMSET (WHSC1/NSD2) expression and is a poor prognostic factor in multiple myeloma (MM). MMSET encodes two major protein isoforms. We have characterized the role of the shorter isoform (REIIBP) in myeloma cells and identified a clear and novel interaction of REIIBP with members of the SMN (survival of motor neuron) complex that directly affects the assembly of the spliceosomal ribonucleic particles. Using RNA-seq we show that REIIBP influences the RNA splicing pattern of the cell. This new discovery provides novel insights into the understanding of MM pathology, and potential new leads for therapeutic targeting.


Asunto(s)
N-Metiltransferasa de Histona-Lisina/metabolismo , Procesamiento Postranscripcional del ARN , Proteínas Represoras/metabolismo , Proteínas del Complejo SMN/metabolismo , Proliferación Celular , Cromosomas Humanos Par 14/genética , Cromosomas Humanos Par 4/genética , Células HeLa , Histonas/metabolismo , Humanos , Intrones/genética , Espectrometría de Masas , Metilación , Mieloma Múltiple/genética , Mieloma Múltiple/patología , Fenotipo , Unión Proteica , Isoformas de Proteínas/metabolismo , Estructura Terciaria de Proteína , Empalme del ARN/genética , Ribonucleoproteínas Nucleares Pequeñas/metabolismo , Empalmosomas/metabolismo , Translocación Genética
10.
Leukemia ; 28(2): 384-390, 2014 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-23817176

RESUMEN

The mechanisms involved in the progression from monoclonal gammopathy of undetermined significance (MGUS) and smoldering myeloma (SMM) to malignant multiple myeloma (MM) and plasma cell leukemia (PCL) are poorly understood but believed to involve the sequential acquisition of genetic hits. We performed exome and whole-genome sequencing on a series of MGUS (n=4), high-risk (HR)SMM (n=4), MM (n=26) and PCL (n=2) samples, including four cases who transformed from HR-SMM to MM, to determine the genetic factors that drive progression of disease. The pattern and number of non-synonymous mutations show that the MGUS disease stage is less genetically complex than MM, and HR-SMM is similar to presenting MM. Intraclonal heterogeneity is present at all stages and using cases of HR-SMM, which transformed to MM, we show that intraclonal heterogeneity is a typical feature of the disease. At the HR-SMM stage of disease, the majority of the genetic changes necessary to give rise to MM are already present. These data suggest that clonal progression is the key feature of transformation of HR-SMM to MM and as such the invasive clinically predominant clone typical of MM is already present at the SMM stage and would be amenable to therapeutic intervention at that stage.


Asunto(s)
Transformación Celular Neoplásica/genética , Mieloma Múltiple/genética , Transformación Celular Neoplásica/metabolismo , Progresión de la Enfermedad , Humanos , Gammopatía Monoclonal de Relevancia Indeterminada/genética , Gammopatía Monoclonal de Relevancia Indeterminada/metabolismo , Mieloma Múltiple/diagnóstico , Mieloma Múltiple/metabolismo , Mutación , Translocación Genética
11.
Cancer Lett ; 339(1): 49-59, 2013 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-23887058

RESUMEN

Multiple myeloma (MM) cells rely on protein homeostatic mechanisms for survival. These mechanisms could be therapeutically targeted via modulation of the heat shock response. We studied the roles of Hsp72 and Hsc70, and show that the two major cytoplasmic Hsp70s play a key role in regulating protein homeostasis and controlling multiple oncogenic pathways in MM, and their inhibition can lead to myeloma cell death. Our study provides further evidence that targeting Hsp70 represents a novel therapeutic approach which may be effective in the treatment of MM.


Asunto(s)
Proteínas HSP70 de Choque Térmico/genética , Inmunoglobulinas/metabolismo , Mieloma Múltiple/genética , Mieloma Múltiple/metabolismo , Estrés Fisiológico , Muerte Celular/genética , Línea Celular Tumoral , Supervivencia Celular/genética , Silenciador del Gen , Proteínas del Choque Térmico HSC70/genética , Proteínas del Choque Térmico HSC70/metabolismo , Proteínas HSP70 de Choque Térmico/antagonistas & inhibidores , Proteínas HSP70 de Choque Térmico/metabolismo , Humanos , Espacio Intracelular/metabolismo , Interferencia de ARN , Estrés Fisiológico/genética , Ubiquitina/metabolismo
12.
Mol Cell Biol ; 29(7): 1682-93, 2009 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-19158269

RESUMEN

The human interleukin-3 (IL-3) and granulocyte-macrophage colony-stimulating-factor (GM-CSF, or CSF2) gene cluster arose by duplication of an ancestral gene. Although just 10 kb apart and responsive to the same signals, the IL-3 and GM-CSF genes are nevertheless regulated independently by separate, tissue-specific enhancers. To understand the differential regulation of the IL-3 and GM-CSF genes we have investigated a cluster of three ubiquitous DNase I-hypersensitive sites (DHSs) located between the two genes. We found that each site contains a conserved CTCF consensus sequence, binds CTCF, and recruits the cohesin subunit Rad21 in vivo. The positioning of these sites relative to the IL-3 and GM-CSF genes and their respective enhancers is conserved between human and mouse, suggesting a functional role in the organization of the locus. We found that these sites effectively block functional interactions between the GM-CSF enhancer and either the IL-3 or the GM-CSF promoter in reporter gene assays. These data argue that the regulation of the IL-3 and the GM-CSF promoters depends on the positions of their enhancers relative to the conserved CTCF/cohesin-binding sites. We suggest that one important role of these sites is to enable the independent regulation of the IL-3 and GM-CSF genes.


Asunto(s)
Secuencia Conservada , Proteínas de Unión al ADN/metabolismo , Regulación de la Expresión Génica , Factor Estimulante de Colonias de Granulocitos y Macrófagos/genética , Elementos Aisladores/genética , Interleucina-3/genética , Proteínas Nucleares/metabolismo , Fosfoproteínas/metabolismo , Proteínas Represoras/metabolismo , Animales , Emparejamiento Base , Secuencia de Bases , Sitios de Unión , Factor de Unión a CCCTC , Proteínas de Ciclo Celular/metabolismo , Proteínas Cromosómicas no Histona/metabolismo , Huella de ADN , Metilación de ADN , Desoxirribonucleasas/metabolismo , Ensayo de Cambio de Movilidad Electroforética , Elementos de Facilitación Genéticos , Humanos , Células Jurkat , Ratones , Datos de Secuencia Molecular , Unión Proteica , Cohesinas
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA