Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
1.
Growth Factors ; 32(6): 176-89, 2014 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-25391995

RESUMEN

Eph receptor tyrosine kinases control cell-cell interactions during normal and oncogenic development, and are implicated in a range of processes including angiogenesis, stem cell maintenance and metastasis. They are thus of great interest as targets for cancer therapy. EphA3, originally isolated from leukemic and melanoma cells, is presently one of the most promising therapeutic targets, with multiple tumor-promoting roles in a variety of cancer types. This review focuses on EphA3, its functions in controlling cellular behavior, both in normal and pathological development, and most particularly in cancer.


Asunto(s)
Neoplasias/metabolismo , Receptores de la Familia Eph/metabolismo , Animales , Antineoplásicos/farmacología , Antineoplásicos/uso terapéutico , Humanos , Neoplasias/tratamiento farmacológico , Inhibidores de Proteínas Quinasas/farmacología , Inhibidores de Proteínas Quinasas/uso terapéutico , Receptores de la Familia Eph/antagonistas & inhibidores , Receptores de la Familia Eph/química , Receptores de la Familia Eph/genética
2.
Blood ; 119(12): 2883-94, 2012 Mar 22.
Artículo en Inglés | MEDLINE | ID: mdl-22323452

RESUMEN

Constitutive activation of FLT3 by internal tandem duplication (ITD) is one of the most common molecular alterations in acute myeloid leukemia (AML). FLT3/ITD mutations have also been observed in myelodysplastic syndrome patients both before and during progression to AML. Previous work has shown that insertion of an FLT3/ITD mutation into the murine Flt3 gene induces a myeloproliferative neoplasm, but not progression to acute leukemia, suggesting that additional cooperating events are required. We therefore combined the FLT3/ITD mutation with a model of myelodysplastic syndrome involving transgenic expression of the Nup98-HoxD13 (NHD13) fusion gene. Mice expressing both the FLT3/ITD and NHD13 transgene developed AML with 100% penetrance and short latency. These leukemias were driven by mutant FLT3 expression and were susceptible to treatment with FLT3 tyrosine kinase inhibitors. We also observed a spontaneous loss of the wild-type Flt3 allele in these AMLs, further modeling the loss of the heterozygosity phenomenon that is seen in human AML with FLT3-activating mutations. Because resistance to FLT3 inhibitors remains an important clinical issue, this model may help identify new molecular targets in collaborative signaling pathways.


Asunto(s)
Modelos Animales de Enfermedad , Proteínas de Homeodominio/genética , Leucemia Mieloide Aguda/genética , Proteínas de Complejo Poro Nuclear/genética , Proteínas de Fusión Oncogénica/genética , Tirosina Quinasa 3 Similar a fms/genética , Animales , Western Blotting , Citometría de Flujo , Técnicas de Sustitución del Gen , Humanos , Ratones , Ratones Transgénicos , Mutación , Reacción en Cadena en Tiempo Real de la Polimerasa , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
3.
Blood ; 120(12): 2475-83, 2012 Sep 20.
Artículo en Inglés | MEDLINE | ID: mdl-22855610

RESUMEN

Programmed cell death or apoptosis is a prominent feature of low-risk myelodysplastic syndromes (MDS), although the underlying mechanism remains controversial. High-risk MDS have less apoptosis associated with increased expression of the prosurvival BCL2-related proteins. To address the mechanism and pathogenic role of apoptosis and BCL2 expression in MDS, we used a mouse model resembling human MDS, in which the fusion protein NUP98-HOXD13 (NHD13) of the chromosomal translocation t(2;11)(q31;p15) is expressed in hematopoietic cells. Hematopoietic stem and progenitor cells from 3-month-old mice had increased rates of apoptosis associated with increased cell cycling and DNA damage. Gene expression profiling of these MDS progenitors revealed a specific reduction in Bcl2. Restoration of Bcl2 expression by a BCL2 transgene blocked apoptosis of the MDS progenitors, which corrected the macrocytic anemia. Blocking apoptosis also restored cell-cycle quiescence and reduced DNA damage in the MDS progenitors. We expected that preventing apoptosis would accelerate malignant transformation to acute myeloid leukemia (AML). However, contrary to expectations, preventing apoptosis of premalignant cells abrogated transformation to AML. In contrast to the current dogma that overcoming apoptosis is an important step toward cancer, this work demonstrates that gaining a survival advantage of premalignant cells may delay or prevent leukemic progression.


Asunto(s)
Apoptosis , Transformación Celular Neoplásica/patología , Células Madre Hematopoyéticas/patología , Síndromes Mielodisplásicos/patología , Proteínas de Fusión Oncogénica/fisiología , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Animales , Biomarcadores de Tumor/genética , Biomarcadores de Tumor/metabolismo , Western Blotting , Proliferación Celular , Perfilación de la Expresión Génica , Células Madre Hematopoyéticas/metabolismo , Humanos , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Síndromes Mielodisplásicos/genética , Síndromes Mielodisplásicos/metabolismo , Análisis de Secuencia por Matrices de Oligonucleótidos , Proteínas Proto-Oncogénicas c-bcl-2/genética , ARN Mensajero/genética , Reacción en Cadena en Tiempo Real de la Polimerasa , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
4.
Blood ; 118(24): 6247-57, 2011 Dec 08.
Artículo en Inglés | MEDLINE | ID: mdl-21948299

RESUMEN

Structural chromosomal rearrangements of the Nucleoporin 98 gene (NUP98), primarily balanced translocations and inversions, are associated with a wide array of hematopoietic malignancies. NUP98 is known to be fused to at least 28 different partner genes in patients with hematopoietic malignancies, including acute myeloid leukemia, chronic myeloid leukemia in blast crisis, myelodysplastic syndrome, acute lymphoblastic leukemia, and bilineage/biphenotypic leukemia. NUP98 gene fusions typically encode a fusion protein that retains the amino terminus of NUP98; in this context, it is important to note that several recent studies have demonstrated that the amino-terminal portion of NUP98 exhibits transcription activation potential. Approximately half of the NUP98 fusion partners encode homeodomain proteins, and at least 5 NUP98 fusions involve known histone-modifying genes. Several of the NUP98 fusions, including NUP98-homeobox (HOX)A9, NUP98-HOXD13, and NUP98-JARID1A, have been used to generate animal models of both lymphoid and myeloid malignancy; these models typically up-regulate HOXA cluster genes, including HOXA5, HOXA7, HOXA9, and HOXA10. In addition, several of the NUP98 fusion proteins have been shown to inhibit differentiation of hematopoietic precursors and to increase self-renewal of hematopoietic stem or progenitor cells, providing a potential mechanism for malignant transformation.


Asunto(s)
Neoplasias Hematológicas/genética , Proteínas de Complejo Poro Nuclear/genética , Proteínas de Fusión Oncogénica/genética , Animales , Transformación Celular Neoplásica/metabolismo , Neoplasias Hematológicas/metabolismo , Proteínas de Homeodominio/genética , Humanos , Poro Nuclear/fisiología , Proteínas de Complejo Poro Nuclear/fisiología , Inversión de Secuencia , Transcripción Genética , Translocación Genética
5.
Blood ; 115(6): 1194-203, 2010 Feb 11.
Artículo en Inglés | MEDLINE | ID: mdl-20007546

RESUMEN

The t(10;11) translocation results in a CALM-AF10 fusion gene in a subset of leukemia patients. Expression of a CALM-AF10 transgene results in leukemia, with prolonged latency and incomplete penetrance, suggesting that additional events are necessary for leukemic transformation. CALM-AF10 mice infected with the MOL4070LTR retrovirus developed acute leukemia, and ligation-mediated polymerase chain reaction was used to identify retroviral insertions at 19 common insertion sites, including Zeb2, Nf1, Mn1, Evi1, Ift57, Mpl, Plag1, Kras, Erg, Vav1, and Gata1. A total of 26% (11 of 42) of the mice had retroviral integrations near Zeb2, a transcriptional corepressor leading to overexpression of the Zeb2-transcript. A total of 91% (10 of 11) of mice with Zeb2 insertions developed B-lineage acute lymphoblastic leukemia, suggesting that Zeb2 activation promotes the transformation of CALM-AF10 hematopoietic precursors toward B-lineage leukemias. More than half of the mice with Zeb2 integrations also had Nf1 integrations, suggesting cooperativity among CALM-AF10, Zeb2, and Ras pathway mutations. We searched for Nras, Kras, and Ptpn11 point mutations in the CALM-AF10 leukemic mice. Three mutations were identified, all of which occurred in mice with Zeb2 integrations, consistent with the hypothesis that Zeb2 and Ras pathway activation promotes B-lineage leukemic transformation in concert with CALM-AF10.


Asunto(s)
Proteínas de Homeodominio/metabolismo , Leucemia Experimental/genética , Mutagénesis Insercional , Proteínas de Fusión Oncogénica/genética , Leucemia-Linfoma Linfoblástico de Células Precursoras/genética , Proteínas Represoras/metabolismo , Retroviridae/genética , Animales , Southern Blotting , Western Blotting , Proliferación Celular , Proteínas de Homeodominio/antagonistas & inhibidores , Proteínas de Homeodominio/genética , Humanos , Inmunofenotipificación , Leucemia de Células B/genética , Leucemia de Células B/metabolismo , Leucemia de Células B/patología , Leucemia Experimental/metabolismo , Leucemia Experimental/patología , Ratones , Ratones Transgénicos , Mutación/genética , Leucemia-Linfoma Linfoblástico de Células Precursoras/metabolismo , Leucemia-Linfoma Linfoblástico de Células Precursoras/patología , ARN Mensajero/genética , ARN Mensajero/metabolismo , ARN Interferente Pequeño/farmacología , Proteínas Represoras/antagonistas & inhibidores , Proteínas Represoras/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transducción de Señal , Caja Homeótica 2 de Unión a E-Box con Dedos de Zinc , Proteínas ras/genética
6.
J Immunol ; 183(10): 6227-35, 2009 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-19841179

RESUMEN

Expression of a NUP98-HOXD13 (NHD13) fusion gene leads to myelodysplastic syndrome in mice. In addition to ineffective hematopoiesis, we observed that NHD13 mice were lymphopenic; the lymphopenia was due to a decrease in both T and B lymphocytes. Although the pro-B cell (B220(+)/CD43(+)) populations from the NHD13 and wild-type mice were similar, the NHD13 mice showed decreased pre-B cells (B220(+)/CD43(-)), indicating impaired differentiation at the pro-B to pre-B stage. Thymi from NHD13 mice were smaller and overexpressed Hoxa cluster genes, including Hoxa7, Hoxa9, and Hoxa10. In addition, the NHD13 thymi contained fewer thymocytes, with an increased percentage of CD4(-)/CD8(-) (double-negative (DN)) cells and a decreased percentage of CD4(+)/CD8(+) (double-positive) cells; the DN1/DN2 population was increased and the DN3/DN4 population was decreased, suggesting a partial block at the DN2 to DN3 transition. To determine clonality of the thymocytes, we used degenerate RT-PCR to identify clonal Tcrb gene rearrangements. Five of six NHD13 thymi showed an unusual Tcrb gene rearrangement pattern with common, clonal DJ rearrangements, but distinct V-D junctions, suggesting a marked clonal expansion of thymocytes that had undergone a DJ rearrangement, but not completed a VDJ rearrangement. Taken together, these findings demonstrate that expression of the NHD13 transgene inhibits lymphoid as well as myeloid and erythroid differentiation, results in overexpression of Hoxa cluster genes, and leads to a precursor T cell lymphoblastic leukemia/lymphoma.


Asunto(s)
Linfocitos B/inmunología , Reordenamiento Génico de la Cadena beta de los Receptores de Antígenos de los Linfocitos T , Proteínas de Homeodominio/metabolismo , Síndromes Mielodisplásicos/inmunología , Proteínas de Complejo Poro Nuclear/metabolismo , Proteínas de Fusión Oncogénica/metabolismo , Receptores de Antígenos de Linfocitos T alfa-beta/genética , Linfocitos T/inmunología , Factores de Transcripción/metabolismo , Animales , Apoptosis/genética , Apoptosis/inmunología , Linfocitos B/metabolismo , Diferenciación Celular/genética , Diferenciación Celular/inmunología , Expresión Génica/genética , Expresión Génica/inmunología , Proteínas de Homeodominio/genética , Proteínas de Homeodominio/inmunología , Ratones , Síndromes Mielodisplásicos/genética , Síndromes Mielodisplásicos/metabolismo , Proteínas de Complejo Poro Nuclear/genética , Proteínas de Complejo Poro Nuclear/inmunología , Proteínas de Fusión Oncogénica/genética , Receptores de Antígenos de Linfocitos T alfa-beta/inmunología , Linfocitos T/metabolismo , Timo/inmunología , Timo/metabolismo , Timo/patología , Factores de Transcripción/genética , Factores de Transcripción/inmunología
7.
Proc Natl Acad Sci U S A ; 105(37): 14088-93, 2008 Sep 16.
Artículo en Inglés | MEDLINE | ID: mdl-18768819

RESUMEN

The myelodysplastic syndromes (MDS) comprise a group of premalignant hematologic disorders characterized by ineffective hematopoiesis, dysplasia, and transformation to acute myeloid leukemia (AML). Although it is well established that many malignancies can be transplanted, there is little evidence to demonstrate that a premalignant disease entity, such as MDS or colonic polyps, can be transplanted and subsequently undergo malignant transformation in vivo. Using mice that express a NUP98-HOXD13 (NHD13) transgene in hematopoietic tissues, we show that a MDS can be transplanted to WT recipients. Recipients of the MDS bone marrow displayed all of the critical features of MDS, including peripheral blood cytopenias, dysplasia, and transformation to AML. Even when transplanted with a 10-fold excess of WT cells, the NHD13 cells outcompeted the WT cells over a 38-week period. Limiting-dilution experiments demonstrated that the frequency of the cell that could transmit the disease was approximately 1/6,000-1/16,000 and that the MDS was also transferable to secondary recipients as a premalignant condition. Transformation to AML in primary transplant recipients was generally delayed (46-49 weeks after transplant); however, 6 of 10 secondary transplant recipients developed AML. These findings demonstrate that MDS originates in a transplantable, premalignant, long-term repopulating, MDS-initiating cell.


Asunto(s)
Linaje de la Célula , Trasplante de Células Madre Hematopoyéticas , Sistema Hematopoyético/citología , Síndromes Mielodisplásicos/patología , Animales , Médula Ósea/inmunología , Separación Celular , Leucemia/patología , Ratones , Ratones Endogámicos C57BL , Síndromes Mielodisplásicos/cirugía , Factores de Tiempo
8.
Cancers (Basel) ; 13(6)2021 Mar 18.
Artículo en Inglés | MEDLINE | ID: mdl-33803727

RESUMEN

Pre-leukemia is a catch-all term for any haematological condition which predisposes the individual towards developing leukemia [...].

9.
Cancers (Basel) ; 13(15)2021 Jul 31.
Artículo en Inglés | MEDLINE | ID: mdl-34359759

RESUMEN

We recently characterised the NUP98-HOXD13 (NHD13) mouse as a model of T-cell pre-leukaemia, featuring thymocytes that can engraft in recipient animals and progress to T-cell acute lymphoblastic leukaemia (T-ALL). However, loss of this engraftment ability by deletion of Lyl1 did not result in any loss of leukemogenesis activity. In the present study, we observe that NHD13 thymocytes overexpress EPHA3, and we characterise thymocyte behaviour in NHD13 mice with deletion of EphA3, which show a markedly reduced incidence of T-ALL. Deletion of EphA3 from the NHD13 mice does not prevent the abnormal accumulation or transplantation ability of these thymocytes. However, upon transplantation, these cells are unable to block the normal progression of recipient wild type (WT) progenitor cells through the normal developmental pathway. This is in contrast to the EphA3+/+ NHD13 thymocytes, which block the progression of incoming WT progenitors past the DN1 stage. Therefore, EphA3 is not critical for classical self-renewal, but is essential for mediating an interaction between the abnormally self-renewing cells and healthy progenitors-an interaction that results in a failure of the healthy cells to differentiate normally. We speculate that this may orchestrate a loss of healthy cell competition, which in itself has been demonstrated to be oncogenic, and that this may explain the decrease in T-ALL incidence in the absence of EphA3. We suggest that pre-leukaemic self-renewal in this model is a complex interplay of cell-intrinsic and -extrinsic factors, and that multiple redundant pathways to leukaemogenesis are active.

10.
Blood ; 112(5): 2017-9, 2008 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-18566322

RESUMEN

The NUP98-HOXD13 (NHD13) fusion gene occurs in patients with myelodysplastic syndrome (MDS) and acute nonlymphocytic leukemia (ANLL). We reported that transgenic mice expressing NHD13 develop MDS, and that more than half of these mice eventually progress to acute leukemia. The latency period suggests a requirement for at least 1 complementary event before leukemic transformation. We conducted a candidate gene search for complementary events focused on genes that are frequently mutated in human myeloid leukemia. We investigated 22 ANLL samples and found a high frequency of Nras and Kras mutations, an absence of Npm1, p53, Runx1, Kit and Flt3 mutations, and a single Cbl mutation. Our findings support a working hypothesis that predicts that ANLL cases have one mutation which inhibits differentiation, and a complementary mutation which enhances proliferation or inhibit apoptosis. In addition, we provide the first evidence for spontaneous collaborating mutations in a genetically engineered mouse model of ANLL.


Asunto(s)
Genes ras , Leucemia Experimental/genética , Mutación , Proteínas de Fusión Oncogénica/genética , Proteínas Proto-Oncogénicas c-cbl/genética , Animales , Secuencia de Bases , Transformación Celular Neoplásica/genética , Cocarcinogénesis , ADN de Neoplasias/genética , Modelos Animales de Enfermedad , Humanos , Leucemia Experimental/etiología , Leucemia Mieloide Aguda/etiología , Leucemia Mieloide Aguda/genética , Ratones , Ratones Transgénicos , Modelos Biológicos , Síndromes Mielodisplásicos/complicaciones , Síndromes Mielodisplásicos/genética , Nucleofosmina
11.
Cancer Res ; 67(11): 5148-55, 2007 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-17545593

RESUMEN

The t(2;11)(q31;p15) chromosomal translocation results in a fusion between the NUP98 and HOXD13 genes and has been observed in patients with myelodysplastic syndrome (MDS) or acute myelogenous leukemia. We previously showed that expression of the NUP98-HOXD13 (NHD13) fusion gene in transgenic mice results in an invariably fatal MDS; approximately one third of mice die due to complications of severe pancytopenia, and about two thirds progress to a fatal acute leukemia. In the present study, we used retroviral insertional mutagenesis to identify genes that might collaborate with NHD13 as the MDS transformed to an acute leukemia. Newborn NHD13 transgenic mice and littermate controls were infected with the MOL4070LTR retrovirus. The onset of leukemia was accelerated, suggesting a synergistic effect between the NHD13 transgene and the genes neighboring retroviral insertion events. We identified numerous common insertion sites located near protein-coding genes and confirmed dysregulation of a subset of these by expression analyses. Among these genes were Meis1, a known collaborator of HOX and NUP98-HOX fusion genes, and Mn1, a transcriptional coactivator involved in human leukemia through fusion with the TEL gene. Other putative collaborators included Gata2, Erg, and Epor. Of note, we identified a common insertion site that was >100 kb from the nearest coding gene, but within 20 kb of the miR29a/miR29b1 microRNA locus. Both of these miRNA were up-regulated, demonstrating that retroviral insertional mutagenesis can target miRNA loci as well as protein-coding loci. Our data provide new insights into NHD13-mediated leukemogenesis as well as retroviral insertional mutagenesis mechanisms.


Asunto(s)
Proteínas de Homeodominio/genética , Leucemia Mieloide/genética , Proteínas de Complejo Poro Nuclear/genética , Proteínas de Fusión Oncogénica/genética , Leucemia-Linfoma Linfoblástico de Células Precursoras/genética , Factores de Transcripción/genética , Animales , Transformación Celular Neoplásica/genética , Clonación Molecular , Canal de Potasio ERG1 , Canales de Potasio Éter-A-Go-Go/genética , Factor de Transcripción GATA2/genética , Ratones , Ratones Transgénicos , MicroARNs/genética , Virus de la Leucemia Murina de Moloney/genética , Mutagénesis Insercional , Proteína 1 del Sitio de Integración Viral Ecotrópica Mieloide , Células 3T3 NIH , Proteínas de Neoplasias/genética , Proteínas Oncogénicas/genética , Transactivadores , Proteínas Supresoras de Tumor
12.
Leukemia ; 33(8): 1868-1880, 2019 08.
Artículo en Inglés | MEDLINE | ID: mdl-30700838

RESUMEN

T cell acute lymphoblastic leukaemia (T-ALL) cases include subfamilies that overexpress the TAL1/LMO, TLX1/3 and HOXA transcription factor oncogenes. While it has been shown that TAL1/LMO transcription factors induce self-renewal of thymocytes, whether this is true for other transcription factor oncogenes is unknown. To address this, we have studied NUP98-HOXD13-transgenic (NHD13-Tg) mice, which overexpress HOXA transcription factors throughout haematopoiesis and develop both myelodysplastic syndrome (MDS) progressing to acute myeloid leukaemia (AML) as well as T-ALL. We find that thymocytes from preleukaemic NHD13-Tg mice can serially transplant, demonstrating that they have self-renewal capacity. Transcriptome analysis shows that NHD13-Tg thymocytes exhibit a stem cell-like transcriptional programme closely resembling that induced by Lmo2, including Lmo2 itself and its critical cofactor Lyl1. To determine whether Lmo2/Lyl1 are required for NHD13-induced thymocyte self-renewal, NHD13-Tg mice were crossed with Lyl1 knockout mice. This showed that Lyl1 is essential for expression of the stem cell-like gene expression programme in thymocytes and self-renewal. Surprisingly however, NHD13 transgenic mice lacking Lyl1 showed accelerated T-ALL and absence of transformation to AML, associated with a loss of multipotent progenitors in the bone marrow. Thus multiple T cell oncogenes induce thymocyte self-renewal via Lmo2/Lyl1; however, NHD13 can also promote T-ALL via an alternative pathway.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/fisiología , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/fisiología , Proteínas de Homeodominio/genética , Proteínas con Dominio LIM/fisiología , Proteínas de Neoplasias/fisiología , Proteínas de Complejo Poro Nuclear/genética , Proteínas de Fusión Oncogénica/genética , Leucemia-Linfoma Linfoblástico de Células T Precursoras/genética , Timocitos/fisiología , Factores de Transcripción/genética , Animales , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos
13.
J Natl Cancer Inst Monogr ; (39): 64-8, 2008.
Artículo en Inglés | MEDLINE | ID: mdl-18648006

RESUMEN

The myelodysplastic syndromes (MDS) are a group of clonal hematopoietic stem cell disorders characterized by ineffective hematopoiesis, peripheral blood cytopenias, dysplasia, and a propensity for transformation to acute myeloid leukemia (AML). A wide spectrum of genetic aberrations has been associated with MDS, including chromosomal translocations involving the NUP98 gene, most commonly leading to fusions of NUP98 with abd-b group HOX genes, including HOXD13. We used vav regulatory elements to direct expression of a NUP98-HOXD13 (NHD13) fusion gene in hematopoietic tissues. NHD13 transgenic mice faithfully recapitulate all the key features of MDS, including peripheral blood cytopenias, bone marrow dysplasia and apoptosis, and transformation to acute leukemia. The MDS that develops in NHD13 transgenic mice is highly lethal; within 14 months, 90% of the mice died of either leukemic transformation or severe anemia and leukopenia due to progressive MDS. These mice provide a preclinical model that can be used for the evaluation of MDS therapy and biology.


Asunto(s)
Proteínas de Homeodominio/genética , Síndromes Mielodisplásicos/genética , Proteínas de Complejo Poro Nuclear/genética , Proteínas de Fusión Oncogénica/genética , Factores de Transcripción/genética , Translocación Genética , Animales , Aberraciones Cromosómicas , Modelos Animales de Enfermedad , Humanos , Ratones , Ratones Transgénicos
14.
Haematologica ; 93(9): 1394-7, 2008 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-18603548

RESUMEN

Expression of a NUP98-HOXD13 (NHD13) fusion gene, initially identified in a patient with myelodysplastic syndrome, leads to a highly penetrant myelodysplastic syndrome in mice that recapitulates all of the key features of the human disease. Expansion of undifferentiated lineage negative (lin(neg)) hematopoietic precursors that express NHD13 was markedly inhibited (30-fold) in vitro. Decreased expansion was accompanied by decreased production of terminally differentiated cells, indicating impaired differentiation of NHD13 precursors. Rather than differentiate, the majority (80%) of NHD13 lin(neg) precursors underwent apoptotic cell death when induced to differentiate. These findings demonstrate that NHD13 lin(neg) cells provide a tractable in vitro system for studies of myelodysplastic syndrome.


Asunto(s)
Apoptosis , Diferenciación Celular , Sistema Hematopoyético/citología , Síndromes Mielodisplásicos/patología , Animales , Línea Celular , Modelos Animales de Enfermedad , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos
15.
Cancer Res ; 65(16): 7151-8, 2005 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-16103065

RESUMEN

OLIG2 (originally designated BHLHB1) encodes a transcription factor that contains the basic helix-loop-helix motif. Although expression of OLIG2 is normally restricted to neural tissues, overexpression of OLIG2 has been shown in patients with precursor T-cell lymphoblastic lymphoma/leukemia (pre-T LBL). In the current study, we found that overexpression of OLIG2 was not only found in oligodendroglioma samples and normal neural tissue but also in a wide spectrum of malignant cell lines including leukemia, non-small cell lung carcinoma, melanoma, and breast cancer cell lines. To investigate whether enforced expression of OLIG2 is oncogenic, we generated transgenic mice that overexpressed OLIG2 in the thymus. Ectopic OLIG2 expression in the thymus was only weakly oncogenic as only 2 of 85 mice developed pre-T LBL. However, almost 60% of transgenic mice that overexpressed both OLIG2 and LMO1 developed pre-T LBL with large thymic tumor masses. Gene expression profiling of thymic tumors that developed in OLIG2/LMO1 mice revealed up-regulation of Notch1 as well as Deltex1 (Dtx1) and pre T-cell antigen receptor alpha (Ptcra), two genes that are considered to be downstream of Notch1. Of note, we found mutations in the Notch1 heterodimerization or proline-, glutamic acid-, serine-, and threonine-rich domain in three of six primary thymic tumors. In addition, growth of leukemic cell lines established from OLIG2/LMO1 transgenic mice was suppressed by a gamma-secretase inhibitor, suggesting that Notch1 up-regulation is important for the proliferation of OLIG2-LMO1 leukemic cells.


Asunto(s)
Transformación Celular Neoplásica/metabolismo , Proteínas de Unión al ADN/genética , Leucemia de Células T/genética , Proteínas del Tejido Nervioso/biosíntesis , Proteínas Oncogénicas/genética , Leucemia-Linfoma Linfoblástico de Células Precursoras/genética , Secretasas de la Proteína Precursora del Amiloide , Animales , Ácido Aspártico Endopeptidasas/antagonistas & inhibidores , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico , Procesos de Crecimiento Celular/efectos de los fármacos , Procesos de Crecimiento Celular/fisiología , Línea Celular Tumoral , Transformación Celular Neoplásica/genética , Endopeptidasas , Perfilación de la Expresión Génica , Humanos , Proteínas con Dominio LIM , Leucemia de Células T/metabolismo , Ratones , Ratones Transgénicos , Mutación , Proteínas del Tejido Nervioso/genética , Proteínas Nucleares , Factor de Transcripción 2 de los Oligodendrocitos , Leucemia-Linfoma Linfoblástico de Células Precursoras/metabolismo , Receptor Notch1 , Receptores de Superficie Celular/genética , Neoplasias del Timo/genética , Neoplasias del Timo/metabolismo , Factores de Transcripción/genética
17.
Leuk Lymphoma ; 45(7): 1341-50, 2004 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-15359631

RESUMEN

Chromosomal aberrations occur with great frequency and some specificity in leukemia and other hematologic malignancies. The most common outcome of these rearrangements is the formation of a fusion gene, comprising portions of 2 genes normally present in the cell. These fusion proteins are presumed to be oncogenic; in many cases, animal models have proven them to be oncogenic. One of the most promiscuous fusion partner genes is the newly identified NUP98 gene, located on chromosome 11p15.5, which to date has been observed fused to 15 different fusion partners. NUP98 encodes a 98 kD protein that is an important component of the nuclear pore complex, which mediates nucleo-cytoplasmic transport of protein and RNA. The fusion partners of NUP98 form 2 distinct groups: homeobox genes and non-homeobox genes. All NUP98 fusions join the N-terminal GLFG repeats of NUP98 to the C-terminal portion of the partner gene, which, in the case of the homeobox gene partners, includes the homeodomain. Clinical findings are reviewed here, along with the findings of several in vivo and in vitro models have been employed to investigate the mechanisms by which NUP98 fusion genes contribute to the pathogenesis of leukemia.


Asunto(s)
Neoplasias Hematológicas/genética , Proteínas de Complejo Poro Nuclear/genética , Proteínas de Fusión Oncogénica/genética , Enfermedad Aguda , Antineoplásicos/farmacología , Transformación Celular Neoplásica/genética , Rotura Cromosómica , Cromosomas Humanos Par 11/genética , ADN-Topoisomerasas de Tipo II/fisiología , Inhibidores Enzimáticos/farmacología , Genes Homeobox , Neoplasias Hematológicas/metabolismo , Proteínas de Homeodominio/genética , Proteínas de Homeodominio/fisiología , Humanos , Leucemia/genética , Leucemia/metabolismo , Modelos Genéticos , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/fisiología , Poro Nuclear/fisiología , Proteínas de Complejo Poro Nuclear/fisiología , Proteínas de Fusión Oncogénica/fisiología , Estructura Terciaria de Proteína , Inhibidores de Topoisomerasa II , Translocación Genética
18.
Leuk Res ; 37(12): 1668-73, 2013 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-24090997

RESUMEN

NUP98 is among the most promiscuously translocated genes in hematological diseases. Among the 28 known fusion partners, there are two categories: homeobox genes and non-homeobox genes. The homeobox fusion partners are well-studied in animal models, resulting in HoxA cluster overexpression and hematological disease. The non-homeobox fusion partners are less well studied. We created transgenic animal models for three NUP98 fusion genes (one homeobox, two non-homeobox), and show that in this system, the NUP98-homeobox fusion promotes self-renewal and aberrant gene expression to a significantly greater extent. We conclude that homeobox partners create more potent NUP98 fusion oncogenes than do non-homeobox partners.


Asunto(s)
Transformación Celular Neoplásica/genética , Proteínas de Homeodominio/genética , Proteínas de Complejo Poro Nuclear/genética , Proteínas de Fusión Oncogénica/fisiología , Translocación Genética/fisiología , Proteínas Adaptadoras Transductoras de Señales/genética , Animales , Células Cultivadas , Proteínas del Citoesqueleto/genética , ADN-Topoisomerasas de Tipo I/genética , Factores de Intercambio de Guanina Nucleótido/genética , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos
19.
Exp Hematol ; 40(12): 1016-27, 2012 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-22885519

RESUMEN

NUP98-HOXD13 (NHD13) and CALM-AF10 (CA10) are oncogenic fusion proteins produced by recurrent chromosomal translocations in patients with acute myeloid leukemia (AML). Transgenic mice that express these fusions develop AML with a long latency and incomplete penetrance, suggesting that collaborating genetic events are required for leukemic transformation. We employed genetic techniques to identify both preleukemic abnormalities in healthy transgenic mice as well as collaborating events leading to leukemic transformation. Candidate gene resequencing revealed that 6 of 27 (22%) CA10 AMLs spontaneously acquired a Ras pathway mutation and 8 of 27 (30%) acquired an Flt3 mutation. Two CA10 AMLs acquired an Flt3 internal-tandem duplication, demonstrating that these mutations can be acquired in murine as well as human AML. Gene expression profiles revealed a marked upregulation of Hox genes, particularly Hoxa5, Hoxa9, and Hoxa10 in both NHD13 and CA10 mice. Furthermore, mir196b, which is embedded within the Hoxa locus, was overexpressed in both CA10 and NHD13 samples. In contrast, the Hox cofactors Meis1 and Pbx3 were differentially expressed; Meis1 was increased in CA10 AMLs but not NHD13 AMLs, whereas Pbx3 was consistently increased in NHD13 but not CA10 AMLs. Silencing of Pbx3 in NHD13 cells led to decreased proliferation, increased apoptosis, and decreased colony formation in vitro, suggesting a previously unexpected role for Pbx3 in leukemic transformation.


Asunto(s)
Transformación Celular Neoplásica/genética , Perfilación de la Expresión Génica , Leucemia/genética , Mutación , Proteínas de Fusión Oncogénica/genética , Animales , Apoptosis/genética , Proliferación Celular , Modelos Animales de Enfermedad , Regulación Leucémica de la Expresión Génica , Técnicas de Silenciamiento del Gen , Sitios Genéticos , Proteínas de Homeodominio/genética , Humanos , Leucemia Mieloide Aguda/genética , Ratones , Proteína 1 del Sitio de Integración Viral Ecotrópica Mieloide , Proteínas de Neoplasias/genética , Proteínas Proto-Oncogénicas/genética , Proteínas Represoras/genética , Tirosina Quinasa 3 Similar a fms/genética
20.
Neoplasia ; 9(4): 315-21, 2007 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-17460775

RESUMEN

In the most common form of stem cell leukemia (SCL) gene rearrangement, an interstitial deletion of 82 kb brings SCL under the control of regulatory elements that normally govern expression of the ubiquitously expressed SCL interrupting locus (SIL) gene, which is located directly upstream of SCL. To investigate the effect of this fusion in a mouse model, a bacterial artificial chromosome (BAC) clone containing both human SIL and SCL genes was isolated, and loxP sites were inserted into intron 1 of both the SIL and SCL genes, corresponding to the sites at which recombination occurs in human T-cell acute lymphocytic leukemia patients. This BAC clone was used to generate transgenic SILloxloxSCL mice. These transgenic mice were subsequently bred to Lck-Cre mice that express the Cre recombinase specifically in the thymus. The BAC transgene was recombined between the two loxP sites in over 50% of the thymocytes from SILloxloxSCL/Cre double-transgenic mice, bringing the SCL gene under the direct control of SIL regulatory elements. Aberrant SCL gene expression in the thymus was verified by reverse transcription-polymerase chain reaction. Using FACS analysis, we found that mice carrying both SILloxloxSCL and Cre transgenes have increased CD4-/CD8- thymocytes compared with transgene-negative mice. In the spleen, these transgenic mice show a marked reduction in the number of mature CD4+ or CD8+ cells. These results demonstrate that conditional activation of SCL under control of SIL regulatory elements can impair normal T-cell development.


Asunto(s)
Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Linfocitos T CD4-Positivos/enzimología , Linfocitos T CD8-positivos/enzimología , Diferenciación Celular/genética , Proteínas de la Matriz Extracelular/fisiología , Reordenamiento Génico de Linfocito T/genética , Integrasas/genética , Péptidos y Proteínas de Señalización Intracelular/genética , Proteína-Lisina 6-Oxidasa/fisiología , Proteínas Proto-Oncogénicas/genética , Animales , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/biosíntesis , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Antígenos CD4/biosíntesis , Antígenos CD4/genética , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD4-Positivos/patología , Antígenos CD8/biosíntesis , Antígenos CD8/genética , Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/patología , Humanos , Integrasas/biosíntesis , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Ratones , Ratones SCID , Ratones Transgénicos , Proteínas Proto-Oncogénicas/biosíntesis , Proteínas Proto-Oncogénicas/metabolismo , Recombinación Genética/inmunología , Proteína 1 de la Leucemia Linfocítica T Aguda
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA