Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
1.
N Engl J Med ; 363(11): 1025-37, 2010 Sep 09.
Artículo en Inglés | MEDLINE | ID: mdl-20825315

RESUMEN

BACKGROUND: The in vivo clinical significance of malignant stem cells remains unclear. METHODS: Patients who have the 5q deletion (del[5q]) myelodysplastic syndrome (interstitial deletions involving the long arm of chromosome 5) have complete clinical and cytogenetic remissions in response to lenalidomide treatment, but they often have relapse. To determine whether the persistence of rare but distinct malignant stem cells accounts for such relapses, we examined bone marrow specimens obtained from seven patients with the del(5q) myelodysplastic syndrome who became transfusion-independent while receiving lenalidomide treatment and entered cytogenetic remission. RESULTS: Virtually all CD34+, CD38+ progenitor cells and stem cells that were positive for CD34 and CD90, with undetectable or low CD38 (CD38−/low), had the 5q deletion before treatment. Although lenalidomide efficiently reduced these progenitors in patients in complete remission, a larger fraction of the minor, quiescent, CD34+,CD38-/low, CD90+ del(5q) stem cells as well as functionally defined del(5q) stem cells remained distinctly resistant to lenalidomide. Over time, lenalidomide resistance developed in most of the patients in partial and complete remission, with recurrence or expansion of the del(5q) clone and clinical and cytogenetic progression. CONCLUSIONS: In these patients with the del(5q) myelodysplastic syndrome, we identified rare and phenotypically distinct del(5q) myelodysplastic syndrome stem cells that were also selectively resistant to therapeutic targeting at the time of complete clinical and cytogenetic remission. (Funded by the EuroCancerStemCell Consortium and others.)


Asunto(s)
Antineoplásicos/farmacología , Deleción Cromosómica , Cromosomas Humanos Par 5/genética , Resistencia a Medicamentos/genética , Síndromes Mielodisplásicos/patología , Células Madre Neoplásicas/efectos de los fármacos , Talidomida/análogos & derivados , ADP-Ribosil Ciclasa 1/análisis , Anciano , Anciano de 80 o más Años , Antígenos CD34/análisis , Antineoplásicos/uso terapéutico , Femenino , Expresión Génica , Humanos , Lenalidomida , Masculino , Persona de Mediana Edad , Síndromes Mielodisplásicos/tratamiento farmacológico , Síndromes Mielodisplásicos/genética , Células Madre Neoplásicas/inmunología , Fenotipo , Inducción de Remisión , Talidomida/farmacología , Talidomida/uso terapéutico , Antígenos Thy-1/análisis
2.
Nat Med ; 11(6): 630-7, 2005 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-15908956

RESUMEN

The cellular targets of primary mutations and malignant transformation remain elusive in most cancers. Here, we show that clinically and genetically different subtypes of acute lymphoblastic leukemia (ALL) originate and transform at distinct stages of hematopoietic development. Primary ETV6-RUNX1 (also known as TEL-AML1) fusions and subsequent leukemic transformations were targeted to committed B-cell progenitors. Major breakpoint BCR-ABL1 fusions (encoding P210 BCR-ABL1) originated in hematopoietic stem cells (HSCs), whereas minor BCR-ABL1 fusions (encoding P190 BCR-ABL1) had a B-cell progenitor origin, suggesting that P190 and P210 BCR-ABL1 ALLs represent largely distinct tumor biological and clinical entities. The transformed leukemia-initiating stem cells in both P190 and P210 BCR-ABL1 ALLs had, as in ETV6-RUNX1 ALLs, a committed B progenitor phenotype. In all patients, normal and leukemic repopulating stem cells could successfully be separated prospectively, and notably, the size of the normal HSC compartment in ETV6-RUNX1 and P190 BCR-ABL1 ALLs was found to be unaffected by the expansive leukemic stem cell population.


Asunto(s)
Proteínas de Fusión bcr-abl/fisiología , Células Madre Hematopoyéticas/fisiología , Leucemia-Linfoma Linfoblástico de Células Precursoras/clasificación , ADP-Ribosil Ciclasa , ADP-Ribosil Ciclasa 1 , Adulto , Antígenos CD , Antígenos CD19 , Antígenos CD34 , Niño , Cromosomas Humanos Par 12 , Cromosomas Humanos Par 21 , Subunidad alfa 2 del Factor de Unión al Sitio Principal , Proteínas de Unión al ADN/fisiología , Citometría de Flujo , Proteínas de Fusión bcr-abl/genética , Humanos , Glicoproteínas de Membrana , Mutación , Proteínas Nucleares/fisiología , Proteínas de Fusión Oncogénica/fisiología , Fenotipo , Leucemia-Linfoma Linfoblástico de Células Precursoras/genética , Leucemia-Linfoma Linfoblástico de Células Precursoras/inmunología , Leucemia-Linfoma Linfoblástico de Células Precursoras/patología , Proteínas Proto-Oncogénicas c-ets , Proteínas Represoras/fisiología , Translocación Genética , Proteína ETS de Variante de Translocación 6
3.
Haematologica ; 91(9): 1287-8, 2006 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-16956839

RESUMEN

MLL/GAS7, resulting from t(11;17)(q23;p13), has been reported in one case of treatment-related acute myeloid leukemia (AML). We present a de novo case of t(11;17)-positive pediatric acute lymphoblastic leukemia. Fluorescent in situ hybridization and reverse transcriptase polymerase chain reaction analyses revealed an MLL/GAS7 chimera identical to the one previously described in AML. The molecular genetic features of MLL/GAS7 and the clinical impact of t(11;17) are discussed.


Asunto(s)
Proteínas de Fusión Oncogénica/genética , Leucemia-Linfoma Linfoblástico de Células Precursoras B/genética , Translocación Genética , Cromosomas Humanos Par 11 , Cromosomas Humanos Par 17 , Femenino , Humanos , Lactante , Proteína de la Leucemia Mieloide-Linfoide/genética , Proteínas del Tejido Nervioso/genética
4.
Int J Oncol ; 21(2): 321-6, 2002 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-12118328

RESUMEN

Rearrangements of chromosome bands 12q13-15 are frequent in various benign mesenchymal and epithelial tumors, and the gene HMGA2 seems to be the most common target within this chromosome region. In the majority of cases, the rearrangements result in a fusion of the first three exons of HMGA2 with different translocation partners. Despite the large number of HMGA2 mutations that have been reported, very little is known about the fusion partners. In this study, we have characterized a recurrent fusion of the first three exons of HMGA2 5' to the G protein-coupled receptor gene (RDC1) in lipomas with rearrangements involving chromosome bands 2q35-37 and 12q13-15, one of several recurrent chromosomal rearrangements in lipomas. The functional impact of the fusion is truncation of HMGA2, because the RDC1 part contributes with a stop codon one amino acid downstream of the breakpoint. The breakpoint within RDC1 was localized in a previously uncharacterized exon of the gene, and our data suggest that RDC1 is subject to alternative splicing.


Asunto(s)
Aberraciones Cromosómicas , Cromosomas Humanos Par 12 , Cromosomas Humanos Par 2 , Proteína HMGA2/genética , Lipoma/genética , Receptores de Superficie Celular/genética , Receptores de Quimiocina , Receptores Acoplados a Proteínas G , Empalme Alternativo , Secuencia de Aminoácidos , Fusión Artificial Génica , Secuencia de Bases , Cartilla de ADN/química , Proteína HMGA2/metabolismo , Humanos , Hibridación Fluorescente in Situ , Cariotipificación , Datos de Secuencia Molecular , Reacción en Cadena de la Polimerasa , Receptores CXCR , Receptores de Superficie Celular/metabolismo
5.
Cancer Genet Cytogenet ; 151(2): 178-81, 2004 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-15172757

RESUMEN

Chordoma is a rare, low- to intermediate-grade malignant tumor involving the axial spine. Cytogenetic data on these tumors have been limited to 25 cases. The findings of clonal chromosome aberrations in five new cases are presented. One of these and two previously reported cases have been studied with multicolor combined binary ratio labeling fluorescence in situ hybridization (COBRA-FISH). The karyotypes were near-diploid, mostly with several numerical and structural aberrations. There were multiple imbalances, with loss of segments from 1p, 3p, 3q, 9p, and chromosome 10 seen in two to four of the seven cases. No clustering of breakpoints was seen and no recurrent recombination between chromosomes was detected. The findings are consistent with previous data and indicate that chordoma tumor development is associated with multiple, nonrandom losses including chromosome segments that are frequently involved in many other solid tumors.


Asunto(s)
Cordoma/genética , Neoplasias de la Columna Vertebral/genética , Anciano , Femenino , Humanos , Hibridación Fluorescente in Situ , Cariotipificación , Masculino , Persona de Mediana Edad
6.
Cancer Genet Cytogenet ; 140(1): 66-9, 2003 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-12550762

RESUMEN

Trisomy 8 is the most common chromosomal aberration in myelocytic malignancies, occurring both as a sole change as well as in addition to other abnormalities. In spite of this, next to nothing is known about its pathogenetic importance or its molecular genetic consequences. Possible mechanisms involved in the transformation process include dosage effects of genes mapping to chromosome 8 and presence of specific mutations or cryptic fusion genes on the duplicated chromosome. In the latter case, +8 would be secondary to a cryptic primary rearrangement and not involved in leukemogenesis as such, but rather in tumor evolution. Although hidden genetic changes have been found in some trisomies, for example, mutations in KIT in acute myelocytic leukemia (AML) with +4 and in MET in hereditary papillary kidney carcinoma with trisomy 7, none associated with +8 have so far been discovered. To address this issue, we have investigated a total of 13 cases of AML, myelodysplastic syndromes, and chronic myeloproliferative disorders with trisomy 8 as the sole chromosomal anomaly. All cases were studied by combined binary ratio multicolor fluorescence in situ hybridization (FISH) and with FISH using locus-specific probes for both arms of chromosome 8, the subtelomeric regions of 8p and 8q, and the leukemia-associated genes FGFR1, MOZ, ETO, and MYC. No cryptic changes were detected, thus excluding the possibility of gross genetic rearrangements or aberrations involving these loci on chromosome 8.


Asunto(s)
Pintura Cromosómica/métodos , Cromosomas Humanos Par 8 , Leucemia Mieloide/genética , Síndromes Mielodisplásicos/genética , Trastornos Mieloproliferativos/genética , Trisomía , Enfermedad Aguda , Adulto , Anciano , Anciano de 80 o más Años , Anemia Refractaria/genética , Anemia Refractaria con Exceso de Blastos/genética , Sondas de ADN , Femenino , Humanos , Leucemia Mielomonocítica Crónica/genética , Masculino , Persona de Mediana Edad , Oncogenes
7.
Cancer Genet Cytogenet ; 150(2): 144-52, 2004 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-15066322

RESUMEN

Cytogenetic and molecular cytogenetic analyses were performed on four sublines derived from a newly established, SV40T-immortalized nasopharyngeal (NP) cell line, NP69, with two of the sublines expressing LMP1, an Epstein-Barr virus-encoded gene. A total of seven cytogenetically related subclones were identified, all having highly complex karyotypes with massive numerical and structural rearrangements. Centromeric rearrangements in the form of isochromosomes and whole-arm translocations were prevalent. A cytogenetic sign of gene amplification [i.e., homogeneously staining region (HSR)] was detected at 1q25 in all metaphase cells analyzed. Multicolor combined binary ratio labeling fluorescence in situ hybridization (COBRA-FISH) was used to confirm the karyotypic interpretations. Furthermore, multicolor COBRA-FISH also showed that part of the HSR contained chromosome 20 material. Extensive clonal evolution could be observed by the assessment of karyotypic variation among different subclones and individual metaphase cells. The evaluation of clonal evolution enabled the identification of the temporal order of chromosome aberrations during cell immortalization and malignant transformation. A striking karyotypic similarity was found between sublines expressing LMP1 and an NP carcinoma cell line, with loss of genetic material from chromosome arm 3p being an important recurrent observation. More interestingly, the karyotypic features of NP69 were also similar to those of many epithelial malignancies. Our observations suggest that serial transformation of NP cell lines might provide a useful in vitro model for the study of the multistep neoplastic transformation of NP cells.


Asunto(s)
Transformación Celular Viral/genética , Herpesvirus Humano 4/genética , Nasofaringe , Virus 40 de los Simios/genética , Proteínas de la Matriz Viral/genética , Técnicas de Cultivo de Célula/métodos , Mapeo Cromosómico , Humanos , Hibridación Fluorescente in Situ , Cariotipificación , Transfección
8.
Cancer Genet Cytogenet ; 148(1): 21-8, 2004 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-14697637

RESUMEN

Cytogenetic analyses of four squamous cell carcinomas (SCC) of the esophagus showed complex numerical and structural abnormalities. Chromosomal bands or regions preferentially involved were 11q13, 8q10, 21q10, 3p10 approximately p11, 1p11 approximately q11, 5p11 approximately q11, and 14p11 approximately q11. For the first time, to our knowledge, recurrent aberrations were identified in esophageal SCC, including homogeneous staining region (hsr), isochromosomes i(3q) and i(21q), and ring chromosome. Losses of chromosomal material dominated over gains. Recurrent imbalances included under-representation of 4p13 approximately pter, 5q14 approximately qter, 9p22 approximately pter, 10p, 11p13 approximately pter, 12p13 approximately pter, 17p10 approximately pter, 18p11 approximately pter, 21p, and 22p, as well as over-representation of 1q25 approximately qter, 3q, 7q, and 8q. Interestingly, hsr at different chromosomal regions occurred in three of four cases. With the application of fluorescence in situ hybridization (FISH) and multicolor combined binary ratio labeling-FISH with specific DNA probes, it could be shown that in two cases the hsr was derived from chromosome 11 material and that the amplicon included CCND1. Our results, together with previous molecular genetic findings, indicate that CCND1might be a prime target in 11q13 amplification, and that amplification of this gene might be crucial in the tumorigenesis of esophageal SCC. These observed chromosomal aberrations and imbalances thus provide important information for further molecular genetic investigation of esophageal SCC.


Asunto(s)
Carcinoma de Células Escamosas/genética , Aberraciones Cromosómicas , Ciclina D1/genética , Neoplasias Esofágicas/genética , Amplificación de Genes , Hibridación Fluorescente in Situ/métodos , Adulto , Anciano , Anciano de 80 o más Años , Femenino , Humanos , Cariotipificación , Masculino , Persona de Mediana Edad
9.
Genes Chromosomes Cancer ; 47(1): 64-70, 2008 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-17943971

RESUMEN

During 1995-2004, 209 children/adolescents were diagnosed with acute lymphoblastic or myeloid leukemia (ALL, AML) in Southern Sweden, of which 177 (85%), comprising 128 B-lineage ALL, 34 AML, and 15 T-cell ALL, could be analyzed for internal tandem duplications (ITD) and activating point mutations in the second tyrosine kinase domain (ATKD) of FLT3. Seventeen (10%) FLT3 mutations (6 ITD, 11 ATKD; mutually exclusive) were detected. None of the T-cell ALL harbored any mutations. ITD and ATKD were found in 2% and 6% of the B-lineage ALL and in 12% and 9% of the AML, being particularly common in high hyperdiploid ALL (14%), ALL (20%), and AML (23%) with 11q23/MLL rearrangements, and in AML with a normal karyotype (60%). All ATKD-positive AML with MLL rearrangements harbored the t(9;11)(p21;q23). Global gene expression data were available for 76 of the B-lineage ALL and 19 of the AML, of which 6 (8%) and 3 (16%) had FLT3 mutations, respectively. No distinct expression pattern associated with FLT3 mutations was identified.


Asunto(s)
Regulación Leucémica de la Expresión Génica/fisiología , Leucemia Mieloide Aguda/genética , Mutación , Leucemia-Linfoma Linfoblástico de Células Precursoras/genética , Tirosina Quinasa 3 Similar a fms/genética , Adolescente , Factores de Edad , Niño , Preescolar , Femenino , Frecuencia de los Genes , Humanos , Lactante , Recién Nacido , Leucemia Mieloide Aguda/enzimología , Masculino , Leucemia-Linfoma Linfoblástico de Células Precursoras/enzimología
10.
Genes Chromosomes Cancer ; 47(1): 26-33, 2008 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-17910045

RESUMEN

Although it has been suggested that mutations of the FLT3, NRAS, KRAS, and PTPN11 genes are particularly frequent in high hyperdiploid (>50 chromosomes) pediatric acute lymphoblastic leukemias (ALLs), this has as yet not been confirmed in a large patient cohort. Furthermore, it is unknown whether mutations of these genes coexist in hyperdiploid cases. We performed mutation analyses of FLT3, NRAS, KRAS, and PTPN11 in a consecutive series of 78 high hyperdiploid ALLs. Twenty-six (33%) of the cases harbored a mutation, comprising six activating point mutations and one internal tandem duplication of FLT3 (7/78 cases; 9.0%), eight codon 12, 13, or 61 NRAS mutations (8/78 cases; 10%), five codon 12 or 13 KRAS mutations (5/78 cases, 6.4%), and seven exon 3 or 13 PTPN11 mutations (7/78 cases; 9.0%). No association was seen between the presence of a mutation in FLT3, NRAS, KRAS, or PTPN11 and gender, age, white blood cell count, or relapse, suggesting that they do not confer a negative prognostic impact. Only one case harbored mutations in two different genes, suggesting that mutations of these four genes are generally mutually exclusive. In total, one third of the cases harbored a FLT3, NRAS, KRAS, or PTPN11 mutation, identifying the RTK-RAS signaling pathway as a potential target for novel therapies of high hyperdiploid pediatric ALLs.


Asunto(s)
Diploidia , Genes ras , Mutación , Leucemia-Linfoma Linfoblástico de Células Precursoras/enzimología , Leucemia-Linfoma Linfoblástico de Células Precursoras/genética , Proteína Tirosina Fosfatasa no Receptora Tipo 11/genética , Proteínas Proto-Oncogénicas/genética , Tirosina Quinasa 3 Similar a fms/genética , Proteínas ras/genética , Adolescente , Niño , Preescolar , Análisis Mutacional de ADN , Femenino , Humanos , Lactante , Masculino , Proteínas Proto-Oncogénicas p21(ras)
11.
Br J Haematol ; 136(2): 294-6, 2007 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-17233820

RESUMEN

Three NUP98 chimaeras have previously been reported in T cell acute lymphoblastic leukaemia (T-ALL): NUP98/ADD3, NUP98/CCDC28A, and NUP98/RAP1GDS1. We report a T-ALL with t(11;18)(p15;q12) resulting in a novel NUP98 fusion. Fluorescent in situ hybridisation showed NUP98 and SET binding protein 1(SETBP1) fusion signals; other analyses showed that exon 12 of NUP98 was fused in-frame with exon 5 of SETBP1. Nested polymerase chain reaction did not amplify the reciprocal SETBP1/NUP98, suggesting that NUP98/SETBP1 transcript is pathogenetically important. SETBP1 has previously not been implicated in leukaemias; however, it encodes a protein that specifically interacts with SET, fused to NUP214 in a case of acute undifferentiated leukaemia.


Asunto(s)
Proteínas Portadoras/genética , Cromosomas Humanos Par 11 , Cromosomas Humanos Par 18 , Leucemia-Linfoma de Células T del Adulto/genética , Proteínas de Complejo Poro Nuclear/genética , Proteínas Nucleares/genética , Proteínas de Fusión Oncogénica/genética , Translocación Genética , Niño , Humanos , Hibridación Fluorescente in Situ , Cariotipificación , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
12.
Blood ; 110(8): 3005-14, 2007 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-17616640

RESUMEN

Global gene expression profiling of highly purified 5q-deleted CD34+CD38(-)Thy1+ cells in 5q- myelodysplastic syndromes (MDSs) supported that they might originate from and outcompete normal CD34+CD38(-)Thy1+ hematopoietic stem cells. Few but distinct differences in gene expression distinguished MDS and normal stem cells. Expression of BMI1, encoding a critical regulator of self-renewal, was up-regulated in 5q- stem cells. Whereas multiple previous MDS genetic screens failed to identify altered expression of the gene encoding the myeloid transcription factor CEBPA, stage-specific and extensive down-regulation of CEBPA was specifically observed in MDS progenitors. These studies establish the importance of molecular characterization of distinct stages of cancer stem and progenitor cells to enhance the resolution of stage-specific dysregulated gene expression.


Asunto(s)
Proteínas Potenciadoras de Unión a CCAAT/biosíntesis , Cromosomas Humanos Par 5/genética , Expresión Génica , Células Madre Hematopoyéticas/fisiología , Síndromes Mielodisplásicos/genética , Proteínas Nucleares/biosíntesis , Proteínas Proto-Oncogénicas/biosíntesis , Proteínas Represoras/biosíntesis , ADP-Ribosil Ciclasa 1/metabolismo , Anciano , Anciano de 80 o más Años , Antígenos CD34/metabolismo , Linaje de la Célula/genética , Femenino , Perfilación de la Expresión Génica , Células Madre Hematopoyéticas/citología , Humanos , Procesamiento de Imagen Asistido por Computador , Hibridación Fluorescente in Situ , Masculino , Persona de Mediana Edad , Complejo Represivo Polycomb 1 , Reacción en Cadena de la Polimerasa , ARN/análisis , Antígenos Thy-1/metabolismo
13.
Br J Haematol ; 133(3): 270-5, 2006 May.
Artículo en Inglés | MEDLINE | ID: mdl-16643428

RESUMEN

ETV6 at 12p13 is rearranged in a variety of haematological malignancies and solid tumours, with more than 20 different partners having been reported. These fusions result in either chimeric proteins or activation of the partner gene. However, there are a few examples of abnormalities resulting in truncated and, most likely, unproductive ETV6 proteins, suggesting that haploinsufficiency of ETV6 and/or the partner is leukaemogenic. We present a novel ETV6 rearrangement, identified in a paediatric pre-B acute lymphoblastic leukaemia. Fluorescence in situ hybridisation (FISH) and molecular genetic analyses revealed a fusion of ETV6 and BAZ2A (at 12q13), generated through a cryptic rearrangement between 12p13 and 12q13, consisting of exons 1 and 2 of ETV6 and a sequence from intron 1 of BAZ2A. This transcript is not expected to produce any chimeric protein, but may encode a truncated form of ETV6, containing the first 54 amino acids (aa), followed by 16 aa from the 3' fusion sequence, reminiscent of ETV6 fusions with MDS2, LOC115548, PER1, and STL. The rearrangement might also modify the regulation of BAZ2A by either activating a cryptic promoter or by coming under the control of the ETV6 promoter. The present case emphasises that 'unproductive' ETV6 rearrangements may play an important pathogenetic role in leukaemia.


Asunto(s)
Linfoma de Burkitt/genética , Proteínas Cromosómicas no Histona/genética , Cromosomas Humanos Par 12/genética , Proteínas Proto-Oncogénicas c-ets/genética , Proteínas Represoras/genética , Secuencia de Aminoácidos , Secuencia de Bases , Preescolar , Femenino , Fusión Génica , Humanos , Hibridación Fluorescente in Situ , Datos de Secuencia Molecular , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa/métodos , Proteína ETS de Variante de Translocación 6
14.
Eur J Haematol ; 77(1): 27-34, 2006 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-16548914

RESUMEN

OBJECTIVES: The t(12;14)(p13;q11)--a recurrent translocation in childhood T-cell acute lymphoblastic leukemia (T-ALL)--has very recently been molecularly characterized in one case, which displayed overexpression of the cyclin D2 gene (CCND2). PATIENTS AND METHODS: We have characterized two pediatric t(12;14)-positive T-ALLs using fluorescence in situ hybridization (FISH), cDNA microarray, and real-time polymerase chain reaction (PCR). RESULTS: FISH revealed breakpoints (BPs) in the T-cell receptor alpha/delta locus (14q11) and in the vicinity of the CCND2 gene at 12p13. To investigate the expression of genes in 12p13, cDNA microarray analysis was performed. Expression data for eight genes, including CCND2, surrounding the 12p BP were compared with those in other T-ALLs. The t(12;14)-positive T-ALL displayed an increased expression of CCND2 compared to the controls, whereas the expression of the other genes was similar in all T-ALLs. Expression of CCND2 and two additional genes (PARP11 and FGF23), close to the 12p BP, was investigated with real-time PCR of the two t(12;14)-positive cases and four controls. Neither PARP11 nor FGF23 displayed expression differences among the T-ALLs, whereas CCND2 was clearly overexpressed in both t(12;14)-positive cases as compared to the mean expression level in the controls. CONCLUSION: We have confirmed, in two additional cases, that the recurrent T-ALL-associated t(12;14) results in overexpression of cyclin D2. The t(12;14) is the first neoplasia-associated translocation shown to result in overexpression of cyclin D2. Furthermore, it is the first example of a T-cell neoplasm with a targeted deregulation of a member of a cyclin-encoding gene family.


Asunto(s)
Ciclinas/genética , Regulación Neoplásica de la Expresión Génica , Leucemia-Linfoma Linfoblástico de Células Precursoras/genética , Receptores de Antígenos de Linfocitos T alfa-beta/genética , Receptores de Antígenos de Linfocitos T gamma-delta/genética , Translocación Genética , Adolescente , Cromosomas Humanos Par 12 , Cromosomas Humanos Par 14 , Ciclina D2 , Ciclinas/metabolismo , Femenino , Factor-23 de Crecimiento de Fibroblastos , Humanos , Hibridación Fluorescente in Situ , Lactante , Masculino , Reacción en Cadena de la Polimerasa , Receptores de Antígenos de Linfocitos T alfa-beta/metabolismo , Receptores de Antígenos de Linfocitos T gamma-delta/metabolismo
15.
Hum Mol Genet ; 15(6): 933-42, 2006 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-16452126

RESUMEN

Double minutes (dmin)-circular, extra-chromosomal amplifications of specific acentric DNA fragments-are relatively frequent in malignant disorders, particularly in solid tumors. In acute myeloid leukemia (AML) and myelodysplastic syndromes (MDS), dmin are observed in approximately 1% of the cases. Most of them consist of an amplified segment from chromosome band 8q24, always including the MYC gene. Besides this information, little is known about their internal structure. We have characterized in detail the genomic organization of 32 AML and two MDS cases with MYC-containing dmin. The minimally amplified region was shown to be 4.26 Mb in size, harboring five known genes, with the proximal and the distal amplicon breakpoints clustering in two regions of approximately 500 and 600 kb, respectively. Interestingly, in 23 (68%) of the studied cases, the amplified region was deleted in one of the chromosome 8 homologs at 8q24, suggesting excision of a DNA segment from the original chromosomal location according to the 'episome model'. In one case, sequencing of both the dmin and del(8q) junctions was achieved and provided definitive evidence in favor of the episome model for the formation of dmin. Expression status of the TRIB1 and MYC genes, encompassed by the minimally amplified region, was assessed by northern blot analysis. The TRIB1 gene was found over-expressed in only a subset of the AML/MDS cases, whereas MYC, contrary to expectations, was always silent. The present study, therefore, strongly suggests that MYC is not the target gene of the 8q24 amplifications.


Asunto(s)
Rotura Cromosómica , Marcación de Gen , Leucemia Mieloide Aguda/genética , Síndromes Mielodisplásicos/genética , Plásmidos/genética , Proteínas Proto-Oncogénicas c-myc/genética , Adulto , Anciano , Anciano de 80 o más Años , Secuencia de Bases , Cromosomas Humanos Par 8/genética , Biología Computacional/métodos , Femenino , Humanos , Hibridación Fluorescente in Situ , Péptidos y Proteínas de Señalización Intracelular/genética , Masculino , Persona de Mediana Edad , Modelos Genéticos , Datos de Secuencia Molecular , Proteínas Serina-Treonina Quinasas/biosíntesis , Proteínas Serina-Treonina Quinasas/genética , Proteínas Proto-Oncogénicas c-myc/metabolismo , Eliminación de Secuencia
16.
Genes Chromosomes Cancer ; 44(2): 113-22, 2005 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-15942938

RESUMEN

High hyperdiploidy (>50 chromosomes) in childhood acute lymphoblastic leukemia (ALL) is characterized by nonrandom multiple trisomies and tetrasomies involving in particular chromosomes X, 4, 6, 8, 10, 14, 17, 18, and 21. This characteristic karyotypic pattern, the most common in pediatric ALL, may arise via a tetraploid state with subsequent loss of chromosomes, by sequential gains of chromosomes in consecutive cell divisions, or by simultaneous gain of chromosomes in a single mitosis. These alternatives may be distinguished by investigation of the allelic ratios of loci on the tetrasomic and disomic chromosomes. Previous studies of tetrasomy 21 and of the occurrence of uniparental disomies (UPDs) have suggested that the most likely mechanism is simultaneous gain. However, the other pathways have not been definitely excluded because complete analyses of all disomies and tetrasomies have never been performed. In the present study, we investigated 27 hyperdiploid ALLs by using 58 polymorphic microsatellite markers mapped to 23 of the 24 human chromosomes. Twenty-six tetrasomies were analyzed (involving chromosomes X, 8, 10, 14, 18, and 21), and the frequency of UPDs was determined in 10 cases. In total, 200 chromosomes were studied. Equal allele dosage was observed in 24 of 26 tetrasomies, and only 7 UPDs were found. These data strongly suggest that hyperdiploidy in childhood ALL generally arises by a simultaneous gain of all additional chromosomes in a single abnormal mitosis.


Asunto(s)
Diploidia , Leucemia-Linfoma Linfoblástico de Células Precursoras/genética , División Celular , Niño , Marcadores Genéticos , Humanos , Hibridación Fluorescente in Situ , Repeticiones de Microsatélite/genética , Reacción en Cadena de la Polimerasa , Leucemia-Linfoma Linfoblástico de Células Precursoras/patología
17.
Genes Chromosomes Cancer ; 36(1): 107-12, 2003 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-12461755

RESUMEN

The NUP98 gene at 11p15 is known to be fused to DDX10, HOXA9, HOXA11, HOXA13, HOXD11, HOXD13, LEDGF, NSD1, NSD3, PMX1, RAP1GDS1, and TOP1 in various hematologic malignancies. The common theme in all NUP98 chimeras is a transcript consisting of the 5' part of NUP98 and the 3' portion of the partner gene; however, apart from the frequent fusion to different homeobox genes, there is no apparent similarity among the other partners. We here report a de novo acute myeloid leukemia with a t(11;12)(p15;q13), resulting in a novel NUP98/HOXC13 fusion. Fluorescence in situ hybridization analyses, by the use of probes covering NUP98 and the HOXC gene cluster at 12q13, revealed a fusion signal at the der(11)t(11;12), indicating a NUP98/HOXC chimera, whereas no fusion was found on the der(12)t(11;12), suggesting that the translocation was accompanied by a deletion of the reciprocal fusion gene. Reverse transcription-PCR and sequence analyses showed that exon 16 (nucleotide 2290) of NUP98 was fused in-frame with exon 2 (nucleotide 852) of HOXC13. Neither the HOXC13/NUP98 transcript nor the normal HOXC13 was expressed. The present results, together with previous studies of NUP98/homeobox gene fusions, strongly indicate that NUP98/HOXC13 is of pathogenetic importance in t(11;12)-positive acute myeloid leukemia.


Asunto(s)
Cromosomas Humanos Par 11/genética , Cromosomas Humanos Par 12/genética , Proteínas de Homeodominio/genética , Leucemia Mieloide/genética , Proteínas de Complejo Poro Nuclear/genética , Proteínas de Fusión Oncogénica/genética , Enfermedad Aguda , Secuencia de Aminoácidos , Secuencia de Bases , Femenino , Humanos , Leucemia Mieloide/diagnóstico , Leucemia Mieloide/patología , Persona de Mediana Edad , Datos de Secuencia Molecular , Translocación Genética/genética , Células Tumorales Cultivadas
18.
Hum Mol Genet ; 13(14): 1479-85, 2004 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-15163636

RESUMEN

Double minutes (dmin), the cytogenetic hallmark of genomic amplification, are found in approximately 1% of karyotypically abnormal acute myeloid leukemias (AML) and myelodysplastic syndromes (MDS). The MYC gene at 8q24 has been reported to be amplified in the majority of the cases, and generally it has been assumed that MYC is the target gene. However, only a few studies have focused on the extent of the amplicon or on the expression patterns of the amplified genes. We have studied six cases (five AML and one MDS) with MYC-containing dmin. Detailed fluorescence in situ hybridization analyses identified a common 4.3 Mb amplicon, with clustered proximal and distal breakpoints, harboring eight known genes (C8FW, NSE2, POU5FLC20, MYC, PVT1, AK093424, MGC27434 and MLZE). The corresponding region was deleted in one of the chromosome 8 homologues in five of the six cases, suggesting that the dmin originated through extra replication (or loop-formation)--excision--amplification. Northern blot analysis revealed that MYC was not overexpressed. Instead, the C8FW gene, encoding a phosphoprotein regulated by mitogenic pathways, displayed increased expression. These results exclude MYC as the target gene and indicate that overexpression of C8FW may be the functionally important consequence of 8q24 amplicons in AML and MDS.


Asunto(s)
Cromatina/genética , Amplificación de Genes , Genes myc , Síndromes Mielodisplásicos/genética , Fosfoproteínas/genética , Enfermedad Aguda , Anciano , Northern Blotting , Cromosomas Humanos Par 8 , Femenino , Humanos , Hibridación Fluorescente in Situ , Leucemia Mieloide/genética , Masculino , Fosfoproteínas/biosíntesis
19.
Gynecol Oncol ; 92(1): 183-91, 2004 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-14751156

RESUMEN

OBJECTIVES: This study aimed at identifying the genetic events involved in immortalization of ovarian epithelial cells, which might be important steps in ovarian carcinogenesis. METHODS: The genetic profiles of five human ovarian surface epithelial (HOSE) cell lines immortalized by retroviral transfection of the human papillomavirus (HPV) E6/E7 genes were thoroughly characterized by chromosome banding and fluorescence in situ hybridization (FISH), at various passages pre- and post-crisis. RESULTS: In pre-crisis, most cells had simple, non-clonal karyotypic changes. Telomere association was the commonest aberration, suggesting that tolermase dysfunction might be an important genetic event leading to cellular crisis. After immortalization post-crisis, however, the karyotypic patterns were non-random. Loss of genetic materials was a characteristic feature. The commonest numerical aberrations were -13, -14, -16, -17, -18, and +5. Among them, loss of chromosome 13 was common change observed in all lines. The only recurrent structural aberration was homogeneously staining regions (hsr) observed in three lines. FISH and combined binary ratio labeling (COBRA)-FISH showed in two cases that the hsrs were derived from chromosome 20. Clonal evolution was observed in four of the lines. In one line, hsr was the only change shared by all subclones, suggesting that it might be a primary event in cell immortalization. CONCLUSION: The results of the present study suggested that loss of chromosome 13 and the amplification of chromosome 20 might be early genetic events involved in ovarian cell immortalization, and might be useful targets for the study of genomic aberrations in ovarian carcinogenesis.


Asunto(s)
Línea Celular Transformada , Aberraciones Cromosómicas , Cromosomas Humanos Par 13/genética , Cromosomas Humanos Par 20/genética , Neoplasias Ováricas/genética , Ovario/ultraestructura , Transformación Celular Neoplásica/genética , Células Epiteliales/citología , Células Epiteliales/fisiología , Femenino , Humanos , Hibridación Fluorescente in Situ , Ovario/citología , Ovario/fisiología
20.
Genes Chromosomes Cancer ; 35(2): 127-37, 2002 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-12203776

RESUMEN

During the initial indolent chronic phase of chronic myeloid leukemia (CML), the t(9;22)(q34;q11), resulting in the Philadelphia chromosome (Ph), is usually the sole cytogenetic anomaly, but as the disease progresses into the accelerated phase (AP), and eventually into aggressive blast crisis (BC), secondary aberrations, mainly unbalanced changes such as +8, i(17q), and +Ph, are frequent. To date, molecular genetic studies of CML BC have mainly focused on alterations of well-known tumor-suppressor genes (e.g., TP53, CDKN2A, and RB1) and oncogenes (e.g., RAS and MYC), whereas limited knowledge is available about the molecular genetic correlates of the unbalanced chromosomal abnormalities. Balanced secondary changes are rare in CML AP/BC, but it is not known whether cryptic chromosomal translocations, generating fusion genes, may be responsible for disease progression in a subgroup of CML. To address this issue, we used multicolor combined binary ratio fluorescence in situ hybridization (FISH), which allows the simultaneous visualization of all 24 chromosomes in different colors, verified by locus-specific FISH in a series of 33 CML cases. Two cryptic balanced translocations, t(7;17)(q32-34;q23) and t(7;17)(p15;q23), were found in two of the five cases showing the t(9;22) as the only cytogenetic change. Using several BAC clones, the breakpoints at 17q23 in both cases were mapped within a 350-kb region. In the case with the 7p15 breakpoint, a BAC clone containing the HOXA gene cluster displayed a split signal, suggesting a possible creation of a fusion gene involving a member of the HOXA family. Furthermore, one case with a partially cryptic t(9;11)(p21-22;q23) and an MLL rearrangement as well as a previously unreported t(3;10)(p22;p12-13) were identified. Altogether, a refined karyotypic description was achieved in 12 (36%) of the 33 investigated cases, illustrating the value of using multicolor FISH for identifying pathogenetically important aberrations in CML AP/BC.


Asunto(s)
Hibridación Fluorescente in Situ/métodos , Leucemia Mielógena Crónica BCR-ABL Positiva/genética , Translocación Genética/genética , Adolescente , Adulto , Anciano , Anciano de 80 o más Años , Aberraciones Cromosómicas , Bandeo Cromosómico , Pintura Cromosómica , Progresión de la Enfermedad , Femenino , Humanos , Cariotipificación , Leucemia Mielógena Crónica BCR-ABL Positiva/patología , Masculino , Persona de Mediana Edad
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA