Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 35
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Nat Genet ; 39(5): 593-5, 2007 May.
Artículo en Inglés | MEDLINE | ID: mdl-17435759

RESUMEN

We identified a duplication of the MYB oncogene in 8.4% of individuals with T cell acute lymphoblastic leukemia (T-ALL) and in five T-ALL cell lines. The duplication is associated with a threefold increase in MYB expression, and knockdown of MYB expression initiates T cell differentiation. Our results identify duplication of MYB as an oncogenic event and suggest that MYB could be a therapeutic target in human T-ALL.


Asunto(s)
Diferenciación Celular/genética , Duplicación de Gen , Genes myb/genética , Leucemia-Linfoma de Células T del Adulto/genética , Linfocitos T/patología , Diferenciación Celular/inmunología , Línea Celular Tumoral , Cromosomas Artificiales/genética , Citometría de Flujo , Dosificación de Gen , Regulación Neoplásica de la Expresión Génica/genética , Pruebas Genéticas , Humanos , Hibridación Fluorescente in Situ , Cariotipificación , Mutación/genética , Hibridación de Ácido Nucleico/genética , ARN Interferente Pequeño/genética , Estadísticas no Paramétricas
2.
Haematologica ; 100(10): 1301-10, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26206799

RESUMEN

T-cell acute lymphoblastic leukemia is caused by the accumulation of multiple oncogenic lesions, including chromosomal rearrangements and mutations. To determine the frequency and co-occurrence of mutations in T-cell acute lymphoblastic leukemia, we performed targeted re-sequencing of 115 genes across 155 diagnostic samples (44 adult and 111 childhood cases). NOTCH1 and CDKN2A/B were mutated/deleted in more than half of the cases, while an additional 37 genes were mutated/deleted in 4% to 20% of cases. We found that IL7R-JAK pathway genes were mutated in 27.7% of cases, with JAK3 mutations being the most frequent event in this group. Copy number variations were also detected, including deletions of CREBBP or CTCF and duplication of MYB. FLT3 mutations were rare, but a novel extracellular mutation in FLT3 was detected and confirmed to be transforming. Furthermore, we identified complex patterns of pairwise associations, including a significant association between mutations in IL7R-JAK genes and epigenetic regulators (WT1, PRC2, PHF6). Our analyses showed that IL7R-JAK genetic lesions did not confer adverse prognosis in T-cell acute lymphoblastic leukemia cases enrolled in the UK ALL2003 trial. Overall, these results identify interconnections between the T-cell acute lymphoblastic leukemia genome and disease biology, and suggest a potential clinical application for JAK inhibitors in a significant proportion of patients with T-cell acute lymphoblastic leukemia.


Asunto(s)
Epigénesis Genética , Quinasas Janus/genética , Mutación , Leucemia-Linfoma Linfoblástico de Células T Precursoras/genética , Receptores de Interleucina-7/genética , Adulto , Niño , Evolución Clonal/genética , Variaciones en el Número de Copia de ADN , Femenino , Perfilación de la Expresión Génica , Regulación Leucémica de la Expresión Génica , Estudios de Asociación Genética , Variación Genética , Secuenciación de Nucleótidos de Alto Rendimiento , Humanos , Quinasas Janus/metabolismo , Masculino , Polimorfismo de Nucleótido Simple , Leucemia-Linfoma Linfoblástico de Células T Precursoras/diagnóstico , Leucemia-Linfoma Linfoblástico de Células T Precursoras/metabolismo , Pronóstico , Receptores de Interleucina-7/metabolismo , Reproducibilidad de los Resultados , Transducción de Señal
3.
Blood ; 117(15): 4056-64, 2011 Apr 14.
Artículo en Inglés | MEDLINE | ID: mdl-21325169

RESUMEN

The genetics of classical Hodgkin lymphoma (cHL) is poorly understood. The finding of a JAK2-involving t(4;9)(q21;p24) in 1 case of cHL prompted us to characterize this translocation on a molecular level and to determine the prevalence of JAK2 rearrangements in cHL. We showed that the t(4;9)(q21;p24) leads to a novel SEC31A-JAK2 fusion. Screening of 131 cHL cases identified 1 additional case with SEC31A-JAK2 and 2 additional cases with rearrangements involving JAK2. We demonstrated that SEC31A-JAK2 is oncogenic in vitro and acts as a constitutively activated tyrosine kinase that is sensitive to JAK inhibitors. In vivo, SEC31A-JAK2 was found to induce a T-lymphoblastic lymphoma or myeloid phenotype in a murine bone marrow transplantation model. Altogether, we identified SEC31A-JAK2 as a chromosomal aberration characteristic for cHL and provide evidence that JAK2 rearrangements occur in a minority of cHL cases. Given the proven oncogenic potential of this novel fusion, our studies provide new insights into the pathogenesis of cHL and indicate that in at least some cases, constitutive activation of the JAK/STAT pathway is caused by JAK2 rearrangements. The finding that SEC31A-JAK2 responds to JAK inhibitors indicates that patients with cHL and JAK2 rearrangements may benefit from targeted therapies.


Asunto(s)
Reordenamiento Génico/genética , Enfermedad de Hodgkin/genética , Janus Quinasa 2/genética , Proteínas de Fusión Oncogénica/genética , Proteínas de Transporte Vesicular/genética , Adulto , Anciano de 80 o más Años , Animales , Trasplante de Médula Ósea , Cromosomas Humanos Par 4 , Cromosomas Humanos Par 9 , Modelos Animales de Enfermedad , Femenino , Predisposición Genética a la Enfermedad/epidemiología , Células HEK293 , Enfermedad de Hodgkin/epidemiología , Humanos , Masculino , Ratones , Persona de Mediana Edad , Leucemia-Linfoma Linfoblástico de Células Precursoras/genética , Prevalencia , Proteínas Tirosina Quinasas/metabolismo , Translocación Genética , Adulto Joven
4.
Haematologica ; 96(10): 1448-56, 2011 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-21750091

RESUMEN

BACKGROUND: The EVI1 gene (3q26) codes for a zinc finger transcription factor with important roles in both mammalian development and leukemogenesis. Over-expression of EVI1 through either 3q26 rearrangements, MLL fusions, or other unknown mechanisms confers a poor prognosis in acute myeloid leukemia. DESIGN AND METHODS: We analyzed the prevalence and prognostic impact of EVI1 over-expression in a series of 476 patients with acute myeloid leukemia, and investigated the epigenetic modifications of the EVI1 locus which could be involved in the transcriptional regulation of this gene. RESULTS: Our data provide further evidence that EVI1 over-expression is a poor prognostic marker in acute myeloid leukemia patients less than 65 years old. Moreover, we found that patients with no basal expression of EVI1 had a better prognosis than patients with expression/over-expression (P=0.036). We also showed that cell lines with over-expression of EVI1 had no DNA methylation in the promoter region of the EVI1 locus, and had marks of active histone modifications: H3 and H4 acetylation, and trimethylation of histone H3 lysine 4. Conversely, cell lines with no expression of EVI1 have DNA hypermethylation and are marked by repressive trimethylation of histone H3 lysine 27 at the EVI1 promoter. CONCLUSIONS: Our results identify EVI1 over-expression as a poor prognostic marker in a large, independent cohort of acute myeloid leukemia patients less than 65 years old, and show that the total absence of EVI1 expression has a prognostic impact on the outcome of such patients. Furthermore, we demonstrated for the first time that an aberrant epigenetic pattern involving DNA methylation, H3 and H4 acetylation, and trimethylation of histone H3 lysine 4 and histone H3 lysine 27 might play a role in the transcriptional regulation of EVI1 in acute myeloid leukemia. This study opens new avenues for a better understanding of the regulation of EVI1 expression at a transcriptional level.


Asunto(s)
Proteínas de Unión al ADN/genética , Regulación hacia Abajo/genética , Epigénesis Genética , Leucemia Mieloide Aguda/diagnóstico , Leucemia Mieloide Aguda/genética , Proto-Oncogenes/genética , Factores de Transcripción/genética , Anciano , Anciano de 80 o más Años , Empalme Alternativo , Línea Celular Tumoral , Cromosomas Humanos Par 3 , Femenino , Perfilación de la Expresión Génica , Regulación Leucémica de la Expresión Génica , Reordenamiento Génico , Humanos , Estimación de Kaplan-Meier , Leucemia Mieloide Aguda/mortalidad , Proteína del Locus del Complejo MDS1 y EV11 , Masculino , Persona de Mediana Edad , Pronóstico
5.
J Clin Invest ; 117(12): 3696-707, 2007 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-18037989

RESUMEN

Transgenic expression of the abnormal products of acute myeloid leukemia-associated (AML-associated) primary chromosomal translocations in hematopoietic stem/progenitor cells initiates leukemogenesis in mice, yet additional mutations are needed for leukemia development. We report here aberrant expression of PR domain containing 16 (PRDM16) in AML cells with either translocations of 1p36 or normal karyotype. These carried, respectively, relatively high prevalence of mutations in the TP53 tumor suppressor gene and in the nucleophosmin (NPM) gene, which regulates p53. Two protein isoforms are expressed from PRDM16, which differ in the presence or absence of the PR domain. Overexpression of the short isoform, sPRDM16, in mouse bone marrow induced AML with full penetrance, but only in the absence of p53. The mouse leukemias were characterized by multilineage cellular abnormalities and megakaryocyte dysplasia, a common feature of human AMLs with 1p36 translocations or NPM mutations. Overexpression of sPRDM16 increased the pool of HSCs in vivo, and in vitro blocked myeloid differentiation and prolonged progenitor life span. Loss of p53 augmented the effects of sPRDM16 on stem cell number and induced immortalization of progenitors. Thus, overexpression of sPRDM16 induces abnormal growth of stem cells and progenitors and cooperates with disruption of the p53 pathway in the induction of myeloid leukemia.


Asunto(s)
Transformación Celular Neoplásica/metabolismo , Proteínas de Unión al ADN/biosíntesis , Células Madre Hematopoyéticas/metabolismo , Leucemia Mieloide Aguda/metabolismo , Células Madre Neoplásicas/metabolismo , Factores de Transcripción/biosíntesis , Proteína p53 Supresora de Tumor , Animales , Médula Ósea/metabolismo , Médula Ósea/patología , Diferenciación Celular/genética , Transformación Celular Neoplásica/genética , Transformación Celular Neoplásica/patología , Senescencia Celular/genética , Proteínas de Unión al ADN/genética , Regulación Leucémica de la Expresión Génica , Células Madre Hematopoyéticas/patología , Humanos , Leucemia Mieloide Aguda/genética , Leucemia Mieloide Aguda/patología , Ratones , Ratones Noqueados , Células Madre Neoplásicas/patología , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Nucleofosmina , Isoformas de Proteínas/biosíntesis , Isoformas de Proteínas/genética , Estructura Terciaria de Proteína/genética , Factores de Transcripción/genética , Translocación Genética , Proteína p53 Supresora de Tumor/genética
6.
Haematologica ; 100(3): e102-5, 2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-25527561
7.
Haematologica ; 93(4): 533-42, 2008 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-18322257

RESUMEN

BACKGROUND: Activating NOTCH1 mutations are common in T-cell acute lymphoblastic leukemia. Inhibition of NOTCH1 signaling with gamma-secretase inhibitors causes cell cycle block, but only after treatment for several days. We further documented the effects of gamma-secretase inhibitor treatment on T-cell acute lymphoblastic leukemia cell lines and tested whether combining gamma-secretase inhibitors with other anti-cancer drugs offers a therapeutic advantage. DESIGN AND METHODS: The effect of gamma-secretase inhibitor treatment and combinations of gamma-secretase inhibitors with chemotherapy or glucocorticoids was assessed on T-cell acute lymphoblastic leukemia cell lines. We sequenced NOTCH1 in T-cell acute lymphoblastic leukemia cases with ABL1 fusions and tested combinations of gamma-secretase inhibitors and the ABL1 inhibitor imatinib in a T-cell acute lymphoblastic leukemia cell line. RESULTS: gamma-secretase inhibitor treatment for 5-7 days reversibly inhibited cell proliferation, caused cell cycle block in sensitive T-cell acute lymphoblastic leukemia cell lines, and caused differentiation of some T-cell acute lymphoblastic leukemia cell lines. Treatment for 14 days or longer was required to induce significant apoptosis. The cytotoxic effects of the chemotherapeutic agent vincristine were not significantly enhanced by addition of gamma-secretase inhibitors to T-cell acute lymphoblastic leukemia cell lines, but gamma-secretase inhibitor treatment sensitized cells to the effect of dexamethasone. NOTCH1 mutations were identified in all T-cell acute lymphoblastic leukemia patients with ABL1 fusions and in a T-cell acute lymphoblastic leukemia cell line expressing NUP214-ABL1. In this cell line, the anti-proliferative effect of imatinib was increased by pre-treatment with gamma-secretase inhibitors. CONCLUSIONS: Short-term treatment of T-cell acute lymphoblastic leukemia cell lines with gamma-secretase inhibitors had limited effects on cell proliferation and survival. Combinations of gamma-secretase inhibitors with other drugs may be required to obtain efficient therapeutic effects in T-cell acute lymphoblastic leukemia, and not all combinations may be useful.


Asunto(s)
Secretasas de la Proteína Precursora del Amiloide/antagonistas & inhibidores , Antineoplásicos/farmacología , Benzodiazepinonas/farmacología , Carbamatos/farmacología , Dipéptidos/farmacología , Inhibidores Enzimáticos/farmacología , Leucemia-Linfoma de Células T del Adulto/tratamiento farmacológico , Proteínas de Neoplasias/antagonistas & inhibidores , Receptor Notch1/antagonistas & inhibidores , Antineoplásicos/administración & dosificación , Apoptosis/efectos de los fármacos , Benzamidas , Carbamatos/administración & dosificación , Ciclo Celular/efectos de los fármacos , División Celular/efectos de los fármacos , Línea Celular Tumoral/efectos de los fármacos , Línea Celular Tumoral/enzimología , ADN de Neoplasias/genética , Daunorrubicina/administración & dosificación , Daunorrubicina/farmacología , Dexametasona/administración & dosificación , Dexametasona/farmacología , Dipéptidos/administración & dosificación , Ensayos de Selección de Medicamentos Antitumorales , Sinergismo Farmacológico , Inhibidores Enzimáticos/administración & dosificación , Humanos , Mesilato de Imatinib , Técnicas In Vitro , Leucemia-Linfoma de Células T del Adulto/genética , Leucemia-Linfoma de Células T del Adulto/patología , Mutación , Proteínas de Neoplasias/genética , Proteínas de Fusión Oncogénica/antagonistas & inhibidores , Proteínas de Fusión Oncogénica/genética , Piperazinas/administración & dosificación , Piperazinas/farmacología , Pirimidinas/administración & dosificación , Pirimidinas/farmacología , Receptor Notch1/genética , Análisis de Secuencia de ADN , Vincristina/administración & dosificación , Vincristina/farmacología
8.
Haematologica ; 93(1): 49-56, 2008 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-18166785

RESUMEN

BACKGROUND: Translocations involving region 5q31-32 (PDGFRB) have been reported in a variety of myeloproliferative diseases and are often associated with significant peripheral eosinophilia. We report an unusual case of a patient presenting with peripheral basophilia and systemic mastocytosis in whom cytogenetic analysis revealed a t(4;5)(q21.1;q31.3). DESIGN AND METHODS: We used molecular analyses to determine the role of PDGFRB in this case. The patient was treated with imatinib. RESULTS: Fluorescence in situ hybridization (FISH) documented a breakpoint in PDGFRB. In agreement with this, the patient responded very well to imatinib with resolution of clinical symptoms, basophilia, and mast cell disease. Molecular analyses revealed that PDGFRB, encoding an imatinib-sensitive tyrosine kinase, was fused to PRKG2. The fusion gene incorporates the first two exons of PRKG2 fused to the truncated exon 12 of PDGFRB, resulting in the disruption of its juxtamembrane domain. Functional studies confirmed that the activity and transforming properties of PRKG2-PDGFRbeta were dependent on the disruption of the auto-inhibitory juxtamembrane domain. CONCLUSIONS: Our results identify a second case of the PRKG2-PDGFRB fusion and confirm the unusual PDGFRB breakpoint associated with this fusion. This work also illustrates the use of imatinib for the treatment of specific cases of systemic mastocytosis.


Asunto(s)
Regulación Leucémica de la Expresión Génica , Leucemia Mielógena Crónica BCR-ABL Positiva/tratamiento farmacológico , Leucemia Mielógena Crónica BCR-ABL Positiva/genética , Mastocitosis Sistémica/tratamiento farmacológico , Mastocitosis Sistémica/genética , Proteínas de Fusión Oncogénica/genética , Piperazinas/farmacología , Pirimidinas/farmacología , Receptor beta de Factor de Crecimiento Derivado de Plaquetas/química , Animales , Benzamidas , Médula Ósea/patología , Citogenética , Humanos , Mesilato de Imatinib , Inmunofenotipificación , Hibridación Fluorescente in Situ , Leucemia Mielógena Crónica BCR-ABL Positiva/complicaciones , Masculino , Mastocitosis Sistémica/complicaciones , Ratones , Persona de Mediana Edad
10.
Haematologica ; 92(1): 27-34, 2007 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-17229632

RESUMEN

BACKGROUND AND OBJECTIVES: Activated tyrosine kinases are implicated in the pathogenesis of chronic and acute leukemia, and represent attractive targets for therapy. Sorafenib (BAY43-9006, Nexavar) is a small molecule B-RAF inhibitor that is used for the treatment of renal cell carcinoma, and has been shown to have activity against receptor tyrosine kinases from the platelet-derived growth factor receptor (PDGFR) and vascular endothelial growth factor receptor (VEGFR) families. We investigated the efficacy of sorafenib at inhibiting mutants of the receptor tyrosine kinases PDGFRbeta, KIT, and FLT3, which are implicated in the pathogenesis of myeloid malignancies. DESIGN AND METHODS: We tested the effect of sorafenib on the proliferation of hematopoietic cells transformed by ETV6-PDGFRbeta, FLT3 with an internal tandem duplication or D835Y point mutation, and the KIT(D816V) mutant. The direct effect of sorafenib on the activity of these kinases and their downstream signaling was tested using phospho-specific antibodies. RESULTS: We show that sorafenib is a potent inhibitor of ETV6-PDGFRbeta and FLT3 mutants, including some of the mutants that confer resistance to PKC412 and other FLT3 inhibitors. Sorafenib induced a cell cycle block and apoptosis in the acute myeloid leukemia cell lines MV4-11 and MOLM-13, both expressing FLT3 with an internal tandem duplication, whereas no effect was observed on four other acute myeloid leukemia cell lines. The imatinib-resistant KIT(D816V) mutant, associated with systemic mastocytosis, was found to be resistant to sorafenib. INTERPRETATION AND CONCLUSIONS: These results warrant further clinical studies of sorafenib for the treatment of myeloid malignancies expressing activated forms of PDGFRbeta and FLT3.


Asunto(s)
Antineoplásicos/farmacología , Bencenosulfonatos/farmacología , Resistencia a Antineoplásicos , Piridinas/farmacología , Receptor beta de Factor de Crecimiento Derivado de Plaquetas/biosíntesis , Receptor beta de Factor de Crecimiento Derivado de Plaquetas/genética , Tirosina Quinasa 3 Similar a fms/biosíntesis , Tirosina Quinasa 3 Similar a fms/genética , Apoptosis , Ciclo Celular , Línea Celular , Línea Celular Transformada , Línea Celular Tumoral , Regulación Neoplásica de la Expresión Génica , Células Madre Hematopoyéticas/metabolismo , Humanos , Mutación , Niacinamida/análogos & derivados , Compuestos de Fenilurea , Sorafenib
11.
Cancer Genet Cytogenet ; 172(1): 70-3, 2007 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-17175383

RESUMEN

Rearrangement of the BCL6 gene is found in follicular lymphomas and in diffuse large B cell lymphomas of follicular center cell origin. The breakpoints cluster mainly in a region spanning the first noncoding exon of the gene (the major breakpoint region). A second breakpoint cluster has also been identified upstream of the first BCL6 noncoding exon (the alternative breakpoint region [ABR]). To date, eight different rearrangements involving the ABR have been reported. Here, we describe a novel rearrangement involving a t(2;3)(p11;q27) translocation that affects the ABR in an unusual combination with the IGK locus.


Asunto(s)
Cromosomas Humanos Par 2/genética , Cromosomas Humanos Par 3/genética , Linfoma Folicular/genética , Translocación Genética/genética , Anciano , Reordenamiento Génico , Humanos , Inmunoglobulinas/genética , Linfoma Folicular/clasificación , Masculino , Reacción en Cadena de la Polimerasa , Proteínas Proto-Oncogénicas c-bcl-6/genética
12.
Cancer Genet Cytogenet ; 175(1): 65-8, 2007 May.
Artículo en Inglés | MEDLINE | ID: mdl-17498561

RESUMEN

The simultaneous occurrence of two different translocations affecting both alleles of the IGH gene has rarely been reported in multiple myeloma. In such a case, two different oncogenes might become transcriptionally deregulated. To investigate this hypothesis, we have characterized the plasma cell leukemia cell line SK-MM2 and a primary myeloma both carrying simultaneous IGH-FGFR3/MMSET and IGH-CCND1 fusions as shown by multicolor fluorescence in situ hybridization. Remarkably, quantitative real-time polymerase chain reaction demonstrated that only one of the oncogene loci was transcriptionally upregulated in both instances. Moreover, the upregulated oncogenes differed between both samples. Thus, biallelic IGH translocations might exert different pathogenetic effects in plasma cell disorders.


Asunto(s)
Ciclinas/genética , N-Metiltransferasa de Histona-Lisina/genética , Cadenas Pesadas de Inmunoglobulina/genética , Mieloma Múltiple/genética , Receptor Tipo 3 de Factor de Crecimiento de Fibroblastos/genética , Proteínas Represoras/genética , Translocación Genética , Alelos , Línea Celular , Bandeo Cromosómico , Pintura Cromosómica , Ciclina D , Regulación Neoplásica de la Expresión Génica , Humanos , Cariotipificación , ARN Mensajero/genética , ARN Mensajero/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
13.
Cancer Res ; 64(8): 2673-6, 2004 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-15087377

RESUMEN

We describe a new PDGFRB fusion associated with a t(5;14)(q33;q24) in a patient with a longstanding chronic myeloproliferative disorder with eosinophilia. After confirmation of PDGFRB involvement and definition of the chromosome 14 breakpoint by fluorescence in situ hybridization, candidate partner genes were selected on the basis of the presence of predicted oligomerization domains believed to be an essential feature of tyrosine kinase fusion proteins. We demonstrate that the t(5;14) fuses PDGFRB to NIN, a gene encoding a centrosomal protein with CEP110-like function. After treatment with imatinib, the patient achieved hematological and cytogenetical remission, but NIN-PDGFRB mRNA remained detectable by reverse transcription-PCR.


Asunto(s)
Antineoplásicos/uso terapéutico , Trastornos Mieloproliferativos/tratamiento farmacológico , Trastornos Mieloproliferativos/genética , Proteínas de Fusión Oncogénica/genética , Piperazinas/uso terapéutico , Pirimidinas/uso terapéutico , Proteínas Tirosina Quinasas Receptoras/genética , Receptor beta de Factor de Crecimiento Derivado de Plaquetas/genética , Adulto , Secuencia de Bases , Benzamidas , Cromosomas Humanos Par 14/genética , Cromosomas Humanos Par 5/genética , Reordenamiento Génico , Humanos , Mesilato de Imatinib , Hibridación Fluorescente in Situ , Masculino , Datos de Secuencia Molecular , Receptor Tipo 1 de Factor de Crecimiento de Fibroblastos , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Translocación Genética
14.
Cancer Res ; 63(12): 3079-83, 2003 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-12810632

RESUMEN

The nucleoporin 98 gene (NUP98) has been reported to be fused to 13 partner genes in hematological malignancies with 11p15 translocations. Twelve of them have been identified in patients with myeloid neoplasias and only 1, RAP1GDS1 (4q21), is fused with NUP98 in five patients with T-cell acute lymphoblastic leukemia (T-ALL). Three of these patients coexpressed T and myeloid markers, suggesting the specific association of t(4;11)(q21;p15) with a subset of T-ALL originating from an early progenitor, which has the potential to express mature T-cell antigens as well as myeloid markers. We describe here a new NUP98 partner involved in a t(10;11)(q25;p15) in a patient with acute biphenotypic leukemia, showing coexpression of mature T and myeloid markers. The gene involved, located in 10q25, was identified as ADD3 using 3'-RACE. ADD3 codes for the ubiquitous expressed subunit gamma of the adducin protein, and it seems to play an important role in the skeletal organization of the cell membrane. Both NUP98-ADD3 and ADD3-NUP98 fusion transcripts are expressed in the patient. This is the second partner of NUP98 described in T-ALL. Adducin shares with the product of RAP1GDS1, and with all of the nonhomeobox NUP98 partners, the presence of a region with significant probability of adopting a coiled-coil conformation. This region is always retained in the fusion transcript with the NH(2) terminus FG repeats of NUP98, suggesting an important role in the mechanism of leukemogenesis.


Asunto(s)
Cromosomas Humanos Par 10/ultraestructura , Cromosomas Humanos Par 11/ultraestructura , Leucemia-Linfoma de Células T del Adulto/genética , Proteínas de Fusión Oncogénica/genética , Translocación Genética , Adulto , Secuencias de Aminoácidos , Secuencia de Aminoácidos , Antígenos CD/análisis , Antígenos de Neoplasias/análisis , Transformación Celular Neoplásica/genética , Cromosomas Humanos Par 10/genética , Cromosomas Humanos Par 11/genética , Humanos , Inmunofenotipificación , Hibridación Fluorescente in Situ , Leucemia-Linfoma de Células T del Adulto/patología , Masculino , Datos de Secuencia Molecular , Células Madre Neoplásicas/inmunología , Proteínas de Fusión Oncogénica/química , Proteínas de Fusión Oncogénica/fisiología , Conformación Proteica , Estructura Terciaria de Proteína
15.
PLoS One ; 11(1): e0146100, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-26745281

RESUMEN

The four members of the epidermal growth factor receptor (EGFR/ERBB) family form homo- and heterodimers which mediate ligand-specific regulation of many key cellular processes in normal and cancer tissues. While signaling through the EGFR has been extensively studied on the molecular level, signal transduction through ERBB3/ERBB4 heterodimers is less well understood. Here, we generated isogenic mouse Ba/F3 cells that express full-length and functional membrane-integrated ERBB3 and ERBB4 or ERBB4 alone, to serve as a defined cellular model for biological and phosphoproteomics analysis of ERBB3/ERBB4 signaling. ERBB3 co-expression significantly enhanced Ba/F3 cell proliferation upon neuregulin-1 (NRG1) treatment. For comprehensive signaling studies we performed quantitative mass spectrometry (MS) experiments to compare the basal ERBB3/ERBB4 cell phosphoproteome to NRG1 treatment of ERBB3/ERBB4 and ERBB4 cells. We employed a workflow comprising differential isotope labeling with mTRAQ reagents followed by chromatographic peptide separation and final phosphopeptide enrichment prior to MS analysis. Overall, we identified 9686 phosphorylation sites which could be confidently localized to specific residues. Statistical analysis of three replicate experiments revealed 492 phosphorylation sites which were significantly changed in NRG1-treated ERBB3/ERBB4 cells. Bioinformatics data analysis recapitulated regulation of mitogen-activated protein kinase and Akt pathways, but also indicated signaling links to cytoskeletal functions and nuclear biology. Comparative assessment of NRG1-stimulated ERBB4 Ba/F3 cells revealed that ERBB3 did not trigger defined signaling pathways but more broadly enhanced phosphoproteome regulation in cells expressing both receptors. In conclusion, our data provide the first global picture of ERBB3/ERBB4 signaling and provide numerous potential starting points for further mechanistic studies.


Asunto(s)
Linfocitos B/metabolismo , Fosfoproteínas/genética , Proteoma/genética , Receptor ErbB-3/genética , Receptor ErbB-4/genética , Transducción de Señal , Secuencia de Aminoácidos , Animales , Linfocitos B/citología , Linfocitos B/efectos de los fármacos , Línea Celular , Regulación de la Expresión Génica , Redes Reguladoras de Genes , Ingeniería Genética , Humanos , Ratones , Datos de Secuencia Molecular , Neurregulina-1/metabolismo , Neurregulina-1/farmacología , Fosfoproteínas/metabolismo , Fosforilación , Unión Proteica , Mapeo de Interacción de Proteínas , Proteoma/metabolismo , Receptor ErbB-3/metabolismo , Receptor ErbB-4/metabolismo
16.
Oncogene ; 23(1): 311-6, 2004 Jan 08.
Artículo en Inglés | MEDLINE | ID: mdl-14712237

RESUMEN

Patients with myeloid malignancies and either the 3q21q26 syndrome or t(1;3)(p36;q21) have been reported to share similar clinicopathological features and a common molecular mechanism for leukemogenesis. Overexpression of MDS1/EVI1 (3q26) or MEL1/PRDM16 (1p36), both members of the PR-domain family, has been directly implicated in the malignant transformation of this subset of neoplasias. The breakpoints in both entities are outside the genes, and the 3q21 region, where RPN1 is located, seems to act as an enhancer. MEL1 has been reported to be expressed in leukemia cells with t(1;3) and in the normal uterus and fetal kidney, but neither in bone marrow (BM) nor in other tissues, suggesting that this gene is specific to t(1;3)-positive MDS/AML. We report the molecular characterization of a t(1;3)(p36;q21) in a patient with MDS (RAEB-2). In contrast to previous studies, we demonstrate that MEL1, the PR-containing form, and MEL1S, the PR-lacking form, are widely expressed in normal tissues, including BM. The clinicopathological features and the breakpoint on 1p36 are different from cases previously described, and MEL1 is not overexpressed, suggesting a heterogeneity in myeloid neoplasias with t(1;3).


Asunto(s)
Cromosomas Humanos Par 1 , Cromosomas Humanos Par 3 , Proteínas de Unión al ADN/genética , Síndromes Mielodisplásicos/genética , Proteínas de Fusión Oncogénica , Factores de Transcripción/genética , Translocación Genética , Anciano , Médula Ósea/metabolismo , Femenino , Regulación de la Expresión Génica , Humanos , Proteínas Recombinantes de Fusión/genética
17.
Cancer Genet Cytogenet ; 157(2): 151-6, 2005 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-15721637

RESUMEN

The t(7;11)(p15;p15.4) has been reported to fuse the NUP98 gene (11p15), a component of the nuclear pore complex, with the class-1 homeobox gene HOXA9 at 7p15. This translocation has been associated with myeloid leukemias, predominantly acute myeloid leukemia (AML) M2 subtype with trilineage myelodysplastic features, and with a poor prognosis. The derived fusion protein retains the FG repeat motif of NUP98 N-terminus and the homeodomain shared by the HOX genes, acting as an oncogenic transcription factor critical for leukemogenesis. We report here a new complex t(7;11)-variant, i.e., t(7;11;13;17)(p15;p15;p?;p1?2) in a patient with AML-M2 and poor prognosis. The NUP98-HOXA9 fusion transcript was detected by RT-PCR, suggesting its role in the malignant transformation as it has been postulated for other t(7;11)-associated leukemias. No other fusion transcripts involving the NUP98 or HOXA9 genes were present, although other mechanisms involving several genes on chromosomes 13 and 17 may also be involved. To our knowledge, this is the first t(7;11) variant involving NUP98 described in hematological malignancies.


Asunto(s)
Cromosomas Humanos Par 11 , Cromosomas Humanos Par 7 , Proteínas de Homeodominio/genética , Proteínas de Homeodominio/metabolismo , Leucemia Mieloide Aguda/genética , Proteínas de Complejo Poro Nuclear/metabolismo , Proteínas de Fusión Oncogénica/metabolismo , Translocación Genética , Anciano , Secuencia de Bases , Cromosomas Humanos Par 13 , Cromosomas Humanos Par 17 , Femenino , Variación Genética , Humanos , Datos de Secuencia Molecular
18.
Cancer Genet Cytogenet ; 160(1): 22-6, 2005 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-15949566

RESUMEN

In this study, we report the case of a Philadelphia (Ph) positive chronic myelogenous leukemia (CML) patient with the presence of p190 and p210 BCR-ABL1 mRNA fusion transcripts derived from e1a2 and b3a2 BCR-ABL1 genomic rearrangements, respectively. The presence of e1a2 BCR-ABL1 genomic rearrangement was seen in 2 different clones, one with the rearrangement and another one with the rearrangement and deletion of the BCR gene of the non-rearranged chromosome 22. After treatment with imatinib, the p210 transcript could not be detected, whereas p190 was still present 6 months after initiation of imatinib therapy and progression to blast phase. The absence of p210 transcript post treatment indicates that the clone with b3a2 responded to imatinib and that the observed resistance was associated to cells harboring the e1a2 genomic rearrangement. Despite resistance of this patient to imatinib, no evidence of mutations in the kinase domain of ABL1 was found. Loss of normal BCR in one cell clone may contribute to the resistance to imatinib due to the lack of BCR mediated inhibition of BCR-ABL1.


Asunto(s)
Proteínas de Fusión bcr-abl/genética , Leucemia Mielógena Crónica BCR-ABL Positiva/genética , Piperazinas/uso terapéutico , Pirimidinas/uso terapéutico , ARN Mensajero/análisis , Anciano , Benzamidas , Femenino , Reordenamiento Génico , Humanos , Mesilato de Imatinib , Hibridación Fluorescente in Situ , Leucemia Mielógena Crónica BCR-ABL Positiva/tratamiento farmacológico
19.
Cancer Genet Cytogenet ; 142(2): 115-9, 2003 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-12699886

RESUMEN

Structural rearrangements of the short arm of chromosome 12 are frequent cytogenetic findings in various hematologic malignancies. The ETV6 gene is the most common target for rearrangements in 12p13. Fluorescence in situ hybridization (FISH) investigations have shown that translocations of 12p other than t(12;21) are frequently accompanied by small interstitial deletions that include ETV6. Unbalanced translocations involving ETV6 have rarely been described, and breakpoints outside ETV6 appear to be strongly associated with complex karyotypes. We studied bone marrow samples from 19 patients known to have 12p unbalanced translocations and complex karyotypes, using FISH and spectral karyotyping. FISH analysis confirmed the hemizygous deletion of the ETV6 and CDKN1B genes in 74% of cases. We found four cases with interstitial deletions. In these four cases and in two others (6/19, 31.5%), the fusion with the partner chromosome was in the subtelomeric region of 12p13.3, confirming that there is a recurrent breakpoint in this region.


Asunto(s)
Rotura Cromosómica/genética , Cromosomas Humanos Par 12/genética , Neoplasias Hematológicas/genética , Translocación Genética/genética , Adolescente , Adulto , Médula Ósea/metabolismo , Niño , Femenino , Humanos , Hibridación Fluorescente in Situ , Ganglios Linfáticos/metabolismo , Masculino , Persona de Mediana Edad , Cariotipificación Espectral
20.
Cancer Discov ; 4(4): 415-22, 2014 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-24469108

RESUMEN

UNLABELLED: We discovered a novel somatic gene fusion, CD74-NRG1, by transcriptome sequencing of 25 lung adenocarcinomas of never smokers. By screening 102 lung adenocarcinomas negative for known oncogenic alterations, we found four additional fusion-positive tumors, all of which were of the invasive mucinous subtype. Mechanistically, CD74-NRG1 leads to extracellular expression of the EGF-like domain of NRG1 III-ß3, thereby providing the ligand for ERBB2-ERBB3 receptor complexes. Accordingly, ERBB2 and ERBB3 expression was high in the index case, and expression of phospho-ERBB3 was specifically found in tumors bearing the fusion (P < 0.0001). Ectopic expression of CD74-NRG1 in lung cancer cell lines expressing ERBB2 and ERBB3 activated ERBB3 and the PI3K-AKT pathway, and led to increased colony formation in soft agar. Thus, CD74-NRG1 gene fusions are activating genomic alterations in invasive mucinous adenocarcinomas and may offer a therapeutic opportunity for a lung tumor subtype with, so far, no effective treatment. SIGNIFICANCE: CD74­NRG1 fusions may represent a therapeutic opportunity for invasive mucinous lung adenocarcinomas, a tumor with no effective treatment that frequently presents with multifocal unresectable disease.


Asunto(s)
Adenocarcinoma Mucinoso/genética , Adenocarcinoma/genética , Antígenos de Diferenciación de Linfocitos B/genética , Antígenos de Histocompatibilidad Clase II/genética , Neoplasias Pulmonares/genética , Neurregulina-1/genética , Proteínas de Fusión Oncogénica/genética , Adenocarcinoma/patología , Adenocarcinoma del Pulmón , Adenocarcinoma Mucinoso/patología , Adulto , Anciano , Anciano de 80 o más Años , Animales , Secuencia de Bases , Línea Celular Tumoral , Femenino , Perfilación de la Expresión Génica , Humanos , Neoplasias Pulmonares/patología , Masculino , Ratones , Persona de Mediana Edad , Datos de Secuencia Molecular , Células 3T3 NIH , Proteínas de Fusión Oncogénica/metabolismo , Análisis de Secuencia de ADN , Transducción de Señal/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA