Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
1.
JCI Insight ; 9(3)2024 Jan 04.
Artículo en Inglés | MEDLINE | ID: mdl-38175731

RESUMEN

Dissemination within the peritoneal cavity is a main determinant of poor patient outcomes from high-grade serous carcinomas (HGSCs). The dissemination process is poorly understood from a cancer evolutionary perspective. We reconstructed the evolutionary trajectories across a median of 5 tumor sites and regions from each of 23 patients based on deep whole-exome sequencing. Polyclonal cancer origin was detected in 1 patient. Ovarian tumors had more complex subclonal architectures than other intraperitoneal tumors in each patient, which indicated that tumors developed earlier in the ovaries. Three common modes of dissemination were identified, including monoclonal or polyclonal dissemination of monophyletic (linear) or polyphyletic (branched) subclones. Mutation profiles of initial or disseminated clones varied greatly among cancers, but recurrent mutations were found in 7 cancer-critical genes, including TP53, BRCA1, BRCA2, and DNMT3A, and in the PI3K/AKT1 pathway. Disseminated clones developed late in the evolutionary trajectory models of most cancers, in particular in cancers with DNA damage repair deficiency. Polyclonal dissemination was predicted to occur predominantly as a single and rapid wave, but chemotherapy exposure was associated with higher genomic diversity of disseminated clones. In conclusion, we described three common evolutionary dissemination modes across HGSCs and proposed factors associated with dissemination diversity.


Asunto(s)
Carcinoma , Neoplasias Ováricas , Femenino , Humanos , Mutación , Neoplasias Ováricas/genética , Neoplasias Ováricas/patología
2.
Endocr Relat Cancer ; 27(9): 457-468, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-32580154

RESUMEN

Testicular germ cell tumours (TGCTs) appear as different histological subtypes or mixtures of these. They show similar, multiple DNA copy number changes, where gain of 12p is pathognomonic. However, few high-resolution analyses have been performed and focal DNA copy number changes with corresponding candidate target genes remain poorly described for individual subtypes. We present the first high-resolution DNA copy number aberration (CNA) analysis on the subtype embryonal carcinomas (ECs), including 13 primary ECs and 5 EC cell lines. We identified recurrent gains and losses and allele-specific CNAs. Within these regions, we nominate 30 genes that may be of interest to the EC subtype. By in silico analysis of data from 150 TGCTs from The Cancer Genome Atlas (TCGA), we further investigated CNAs, RNA expression, somatic mutations and fusion transcripts of these genes. Among primary ECs, ploidy ranged between 2.3 and 5.0, and the most common aberrations were DNA copy number gains at chromosome (arm) 7, 8, 12p, and 17, losses at 4, 10, 11, and 18, replicating known TGCT genome characteristics. Gain of whole or parts of 12p was found in all samples, including a highly amplified 100 kbp segment at 12p13.31, containing SLC2A3. Gain at 7p21, encompassing ETV1, was the second most frequent aberration. In conclusion, we present novel CNAs and the genes located within these regions, where the copy number gain of SLC2A3 and ETV1 are of interest, and which copy number levels also correlate with expression in TGCTs.


Asunto(s)
Variaciones en el Número de Copia de ADN/genética , Proteínas de Unión al ADN/genética , Transportador de Glucosa de Tipo 3/genética , Neoplasias de Células Germinales y Embrionarias/genética , Neoplasias Testiculares/genética , Factores de Transcripción/genética , Humanos
3.
PLoS One ; 12(7): e0182030, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28759630

RESUMEN

The pan lymphocyte marker CD45 exists in various isoforms arising from alternative splicing of the exons 4, 5 and 6. While naïve T cells express CD45RA translated from an mRNA containing exon 4, exons 4-6 are spliced out to encode the shorter CD45R0 in antigen-experienced effector/memory T cells. The SNP C77G (rs17612648) is located in exon 4 and blocks the exon's differential splicing from the pre-mRNA, enforcing expression of CD45RA. Several studies have linked C77G to autoimmune diseases but lack of validation in other cohorts has left its role elusive. An incidental finding in an ovarian cancer patient cohort from West Norway (Bergen region, n = 312), suggested that the frequency of C77G was higher among ovarian cancer patients than in healthy Norwegians (n = 1,357) (3.0% vs. 1.8% allele frequency). However, this finding could not be validated in a larger patient cohort from South-East Norway (Oslo region, n = 1,198) with 1.2% allele frequency. Hence, C77G is not associated with ovarian cancer in the Norwegian population. However, its frequency was increased in patients with FIGO stage II, endometrioid histology or an age at diagnosis of 60 years or older indicating a possible association with a less aggressive cancer type.


Asunto(s)
Antígenos Comunes de Leucocito/genética , Mutación Missense , Neoplasias Ováricas/genética , Adulto , Anciano , Estudios de Casos y Controles , Femenino , Frecuencia de los Genes , Humanos , Persona de Mediana Edad , Noruega , Neoplasias Ováricas/patología , Polimorfismo de Nucleótido Simple
4.
Neoplasia ; 17(2): 167-74, 2015 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-25748235

RESUMEN

Intratubular germ cell neoplasia, the precursor of testicular germ cell tumors (TGCTs), is hypothesized to arise during embryogenesis from developmentally arrested primordial germ cells (PGCs) or gonocytes. In early embryonal life, the PGCs migrate from the yolk sac to the dorsal body wall where the cell population separates before colonizing the genital ridges. However, whether the malignant transformation takes place before or after this separation is controversial. We have explored the somatic exome-wide mutational spectra of bilateral TGCT to provide novel insight into the in utero critical time frame of malignant transformation and TGCT pathogenesis. Exome sequencing was performed in five patients with bilateral TGCT (eight tumors), of these three patients in whom both tumors were available (six tumors) and two patients each with only one available tumor (two tumors). Selected loci were explored by Sanger sequencing in 71 patients with bilateral TGCT. From the exome-wide mutational spectra, no identical mutations in any of the three bilateral tumor pairs were identified. Exome sequencing of all eight tumors revealed 87 somatic non-synonymous mutations (median 10 per tumor; range 5-21), some in already known cancer genes such as CIITA, NEB, platelet-derived growth factor receptor α (PDGFRA), and WHSC1. SUPT6H was found recurrently mutated in two tumors. We suggest independent development lineages of bilateral TGCT. Thus, malignant transformation into intratubular germ cell neoplasia is likely to occur after the migration of PGCs. We reveal possible drivers of TGCT pathogenesis, such as mutated PDGFRA, potentially with therapeutic implications for TGCT patients.


Asunto(s)
Linaje de la Célula , Exoma/genética , Neoplasias de Células Germinales y Embrionarias/genética , Neoplasias Testiculares/genética , Adulto , Análisis Mutacional de ADN , Mutación de Línea Germinal , Humanos , Masculino , Proteínas de Neoplasias/genética , Neoplasias de Células Germinales y Embrionarias/patología , Reacción en Cadena de la Polimerasa/métodos , Proteínas Proto-Oncogénicas c-kit/genética , Receptores del Factor de Crecimiento Derivado de Plaquetas/genética , Análisis de Secuencia de ADN , Neoplasias Testiculares/patología
5.
Nat Genet ; 45(6): 680-5, 2013 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-23666239

RESUMEN

We conducted a meta-analysis to identify new susceptibility loci for testicular germ cell tumor (TGCT). In the discovery phase, we analyzed 931 affected individuals and 1,975 controls from 3 genome-wide association studies (GWAS). We conducted replication in 6 independent sample sets comprising 3,211 affected individuals and 7,591 controls. In the combined analysis, risk of TGCT was significantly associated with markers at four previously unreported loci: 4q22.2 in HPGDS (per-allele odds ratio (OR) = 1.19, 95% confidence interval (CI) = 1.12-1.26; P = 1.11 × 10(-8)), 7p22.3 in MAD1L1 (OR = 1.21, 95% CI = 1.14-1.29; P = 5.59 × 10(-9)), 16q22.3 in RFWD3 (OR = 1.26, 95% CI = 1.18-1.34; P = 5.15 × 10(-12)) and 17q22 (rs9905704: OR = 1.27, 95% CI = 1.18-1.33; P = 4.32 × 10(-13) and rs7221274: OR = 1.20, 95% CI = 1.12-1.28; P = 4.04 × 10(-9)), a locus that includes TEX14, RAD51C and PPM1E. These new TGCT susceptibility loci contain biologically plausible genes encoding proteins important for male germ cell development, chromosomal segregation and the DNA damage response.


Asunto(s)
Neoplasias de Células Germinales y Embrionarias/genética , Neoplasias Testiculares/genética , Algoritmos , Estudios de Casos y Controles , Cromosomas Humanos , Sitios Genéticos , Predisposición Genética a la Enfermedad , Estudio de Asociación del Genoma Completo , Humanos , Desequilibrio de Ligamiento , Masculino , Modelos Genéticos , Polimorfismo de Nucleótido Simple , Factores de Riesgo
6.
Pharmacogenet Genomics ; 19(10): 751-9, 2009 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-19741569

RESUMEN

PURPOSE: The pathogenesis of testicular germ cell tumor (TGCT) remains unknown. The aim of this study was to evaluate the pathogenic role of functional polymorphisms in detoxification enzymes among TGCT patients, through association studies of constitutive genotypes and medical parameters before and after chemotherapy. EXPERIMENTAL DESIGN: Germline deletion polymorphisms in the glutathione S-transferase mu 1 (GSTM1) and the GST theta 1 (GSTT1), and a functional single nucleotide polymorphism in GST pi 1 (GSTP1, Ile105Val), were analyzed in TGCT survivors (TCSs) (n = 675) and controls (n = 189). Statistical analyses were performed for the genotype distributions between the TCSs and control populations, and between the genotypes and clinicopathological parameters of the TCSs. RESULTS: The GST genotypes showed comparable distributions among the TCSs and the control population. However, the genotype combination GSTT1positive/GSTP1-GG or GSTP1-AG/GSTM1positive was more frequent among the TCSs [P = 0.050, odds ratio (OR): 1.47, 95% confidence interval (CI): 0.998-2.165]. The combined genotype GSTT1positive/GSTP1AA/GSTM1positive was associated with decreased risk of development of pure embryonal carcinoma (P = 0.009, OR: 0.309, 95% CI: 0.122-0.784) and the GSTP1-A-allele (i.e. genotypes GSTP-AA or GSTP-AG) was also associated with decreased risk for development of pure teratoma (P = 0.032, OR: 0.326, 95% CI: 0.122-0.873). Furthermore, the GSTP1-A-allele was overrepresented within the 'good prognosis group' (P = 0.032, OR: 2.407, 95% CI: 1.060-5.469), whereas the GSTM1nulltype was associated with the extent of TC qualifying as 'poor prognosis group' (P = 0.025, OR: 2.839, 95% CI: 1.104-7.301). The GSTP1-AG genotype was associated with necrosis in the tumor's post-chemotherapy histology (P = 0.001, OR: 16.087, 95% CI: 1.930-134.087). Failure, after platinum-based chemotherapy, was associated with the GSTT1positive/GSTP-AA or GSTP-GG/GSTM1-positive genotype (P = 0.019, OR: 2.168, 95% CI: 1.130-4.160). CONCLUSION: This study confirms an association between the GSTP1-G-allele and TGCT. Combinations of GST genotypes were associated with primary and post-chemotherapy tumor histology, and prognostic group presentation.


Asunto(s)
Germinoma/genética , Glutatión Transferasa/genética , Neoplasias Testiculares/tratamiento farmacológico , Neoplasias Testiculares/genética , Adolescente , Adulto , Anciano , Predisposición Genética a la Enfermedad , Genotipo , Germinoma/patología , Gutatión-S-Transferasa pi/genética , Gutatión-S-Transferasa pi/metabolismo , Glutatión Transferasa/metabolismo , Humanos , Masculino , Persona de Mediana Edad , Estadificación de Neoplasias , Polimorfismo Genético , Neoplasias Testiculares/patología
7.
J Transl Med ; 5: 70, 2007 Dec 27.
Artículo en Inglés | MEDLINE | ID: mdl-18162130

RESUMEN

BACKGROUND: To assess the impact of polymorphisms in Glutathione S-transferase (GST) -P1, -M1, and -T1 on self-reported chemotherapy-induced long-term toxicities in testicular cancer survivors (TCSs). METHODS: A total of 238 TCSs, who had received cisplatin-based chemotherapy at median twelve years earlier, had participated in a long-term follow-up survey which assessed the prevalence of self-reported paresthesias in fingers/toes, Raynaud-like phenomena in fingers/toes, tinnitus, and hearing impairment. From all TCSs lymphocyte-derived DNA was analyzed for the functional A-->G polymorphism at bp 304 in GSTP1, and deletions in GST-M1 and GST-T1. Evaluation of associations between GST polymorphisms and self-reported toxicities included adjustment for prior treatment. RESULTS: All six evaluated toxicities were significantly associated with the cumulative dose of cisplatin and/or bleomycin. Compared to TCSs with either GSTP1-AG or GSTP1-AA, the 37 TCSs with the genotype GSTP1-GG, were significantly less bothered by paresthesias in fingers and toes (p = 0.039, OR 0.46 [0.22-0.96] and p = 0.023, OR 0.42 [0.20-0.88], respectively), and tinnitus (p = 0.008, OR 0.33 [0.14-0.74]). Furthermore, absence of functional GSTM1 protected against hearing impairment (p = 0.025, OR 1.81 [1.08-3.03]). CONCLUSION: In TCSs long-term self-reported chemotherapy-induced toxicities are associated with functional polymorphisms in GSTP1 and GSTM1. Hypothetically, absence of GST-M1 leaves more glutathione as substrate for the co-expressed GST-P1. Also intracellular inactivation of pro-apoptotic mediators represents a possible explanation of our findings. Genotyping of these GSTs might be a welcomed step towards a more individualized treatment of patients with metastatic testicular cancer.


Asunto(s)
Cisplatino/toxicidad , Gutatión-S-Transferasa pi/genética , Glutatión Transferasa/genética , Neoplasias Testiculares/genética , Adolescente , Adulto , Anciano , Antineoplásicos/uso terapéutico , Antineoplásicos/toxicidad , Cisplatino/uso terapéutico , Estudios Transversales , Predisposición Genética a la Enfermedad , Genotipo , Humanos , Masculino , Persona de Mediana Edad , Polimorfismo de Longitud del Fragmento de Restricción , Estudios Retrospectivos , Sobrevivientes , Neoplasias Testiculares/tratamiento farmacológico , Neoplasias Testiculares/enzimología
8.
Mol Cancer ; 6: 12, 2007 Feb 02.
Artículo en Inglés | MEDLINE | ID: mdl-17274819

RESUMEN

BACKGROUND: Ovarian germ cell tumours (OGCTs) typically arise in young females and their pathogenesis remains poorly understood. We investigated the origin of malignant OGCTs and underlying molecular events in the development of the various histological subtypes of this neoplasia. RESULTS: We examined in situ expression of stem cell-related (NANOG, OCT-3/4, KIT, AP-2gamma) and germ cell-specific proteins (MAGE-A4, NY-ESO-1, TSPY) using a tissue microarray consisting of 60 OGCT tissue samples and eight ovarian small cell carcinoma samples. Developmental pattern of expression of NANOG, TSPY, NY-ESO-1 and MAGE-A4 was determined in foetal ovaries (gestational weeks 13-40). The molecular genetic part of our study included search for the presence of Y-chromosome material by fluorescence in situ hybridisation (FISH), and mutational analysis of the KIT oncogene (exon 17, codon 816), which is often mutated in testicular GCTs, in a subset of tumour DNA samples. We detected a high expression of transcription factors related to the embryonic stem cell-like pluripotency and undifferentiated state in OGCTs, but not in small cell carcinomas, supporting the view that the latter do not arise from a germ cell progenitor. Bilateral OGCTs expressed more stem cell markers than unilateral cases. However, KIT was mutated in 5/13 unilateral dysgerminomas, whereas all bilateral dysgerminomas (n = 4) and all other histological types (n = 22) showed a wild type sequence. Furthermore, tissue from five phenotypic female patients harbouring combined dysgerminoma/gonadoblastoma expressed TSPY and contained Y-chromosome material as confirmed by FISH. CONCLUSION: This study provides new data supporting two distinct but overlapping pathways in OGCT development; one involving spontaneous KIT mutation(s) leading to increased survival and proliferation of undifferentiated oogonia, the other related to presence of Y chromosome material and ensuing gonadal dysgenesis in phenotypic females.


Asunto(s)
Biomarcadores de Tumor/análisis , Disgerminoma/patología , Regulación Neoplásica de la Expresión Génica , Mutación , Neoplasias Ováricas/patología , Células Madre Pluripotentes/patología , Proteínas Proto-Oncogénicas c-kit/genética , Antígenos de Neoplasias/análisis , Carcinoma Embrionario/química , Carcinoma Embrionario/genética , Carcinoma Embrionario/patología , Carcinoma de Células Pequeñas/química , Carcinoma de Células Pequeñas/genética , Carcinoma de Células Pequeñas/patología , Proteínas de Ciclo Celular/análisis , Diferenciación Celular , Linaje de la Célula , Transformación Celular Neoplásica/química , Transformación Celular Neoplásica/genética , Transformación Celular Neoplásica/patología , Proteínas de Unión al ADN/análisis , Disgerminoma/química , Disgerminoma/genética , Células Madre de Carcinoma Embrionario , Femenino , Regulación del Desarrollo de la Expresión Génica , Edad Gestacional , Gonadoblastoma/química , Gonadoblastoma/genética , Gonadoblastoma/patología , Proteínas de Homeodominio/análisis , Humanos , Proteínas de la Membrana/análisis , Proteína Homeótica Nanog , Proteínas de Neoplasias/análisis , Células Madre Neoplásicas/química , Células Madre Neoplásicas/patología , Factor 3 de Transcripción de Unión a Octámeros/análisis , Oogonios/química , Oogonios/patología , Neoplasias Ováricas/química , Neoplasias Ováricas/genética , Ovario/química , Ovario/embriología , Células Madre Pluripotentes/química , Proteínas Proto-Oncogénicas c-kit/análisis , Factor de Transcripción AP-2/análisis
9.
J Clin Oncol ; 25(6): 708-14, 2007 Feb 20.
Artículo en Inglés | MEDLINE | ID: mdl-17228018

RESUMEN

PURPOSE: Cisplatin, a cornerstone of combination chemotherapy in the treatment of testicular cancer, induces hearing impairment with considerable interindividual variations. These differences might be a result of functional polymorphisms in cisplatin-detoxifying enzymes like glutathione S-transferases (GSTs). PATIENTS AND METHODS: We identified 173 cisplatin-treated testicular cancer survivors (TCSs) who had participated in a long-term survey that included audiometric testing and lymphocyte sampling. The hearing decibel thresholds at 4,000 Hz were categorized into leveled scales by normative decibel percentiles. Known functional polymorphisms (positive or negative) in GSTT1 and GSTM1 and codon 105 A/G (Ile/Val) in GSTP1 were analyzed by multiplex polymerase chain reaction, followed by restriction enzyme cutting, and separated by gel electrophoresis. RESULTS: The risk of having an inferior audiometric result was more than four times higher in TCSs with 105Ile/105Ile-GSTP1 or 105Val/105Ile-GSTP1 compared with 105Val/105Val-GSTP1 (odds ratio [OR] = 4.21; 95% CI, 1.99 to 8.88; P < .001 when modeled by ordinal logistic regression [OLR]). GSTM1 positivity was detrimental for hearing ability. Two combined genotypes were associated with hearing ability. The presence of pattern 1 (GSTT1 positive, GSTM1 positive, and 105Ile/105Ile-GSTP1) was associated with hearing impairment (OR = 2.76; 95% CI, 1.35 to 5.64; P = .005, OLR). TCSs with pattern 2 (GSTT1 positive, GSTM1 positive, and 105Val/105Val-GSTP1) had better hearing ability than TCSs without this pattern (OR = 5.35; 95% CI, 2.25 to 12.76; P < .001, OLR). CONCLUSION: The presence of both alleles of 105Val-GSTP1 offered protection against cisplatin-induced hearing impairment. Two genotype patterns with good and poor protection against cisplatin-induced ototoxicity were identified.


Asunto(s)
Cisplatino/efectos adversos , Gutatión-S-Transferasa pi/genética , Glutatión Transferasa/genética , Pérdida Auditiva/inducido químicamente , Neoplasias Testiculares/tratamiento farmacológico , Neoplasias Testiculares/genética , Adolescente , Adulto , Anciano , Audiometría , Estudios de Casos y Controles , Cisplatino/uso terapéutico , Intervalos de Confianza , Estudios de Seguimiento , Predisposición Genética a la Enfermedad , Genotipo , Pérdida Auditiva/diagnóstico , Pérdida Auditiva/genética , Humanos , Masculino , Persona de Mediana Edad , Estadificación de Neoplasias , Oportunidad Relativa , Probabilidad , Estudios Retrospectivos , Índice de Severidad de la Enfermedad , Sobrevivientes , Neoplasias Testiculares/mortalidad , Neoplasias Testiculares/patología , Factores de Tiempo
10.
Cell Oncol ; 28(5-6): 315-26, 2006.
Artículo en Inglés | MEDLINE | ID: mdl-17167184

RESUMEN

INTRODUCTION: Testicular germ cell tumors of adolescent and young adult men (TGCTs) generally have near triploid and complex karyotypes. The actual genes driving the tumorigenesis remain essentially to be identified. MATERIALS AND METHODS: To determine the detailed DNA copy number changes, and investigate their impact on gene expression levels, we performed an integrated microarray profiling of TGCT genomes and transcriptomes. We analyzed 17 TGCTs, three precursor lesions, and the embryonal carcinoma cell lines, NTERA2 and 2102Ep, by comparative genomic hybridization microarrays (array-CGH), and integrated the data with transcriptome profiles of the same samples. RESULTS: The gain of chromosome arm 12p was, as expected, the most common aberration, and we found CCND2, CD9, GAPD, GDF3, NANOG, and TEAD4 to be the therein most highly over-expressed genes. Additional frequent genomic aberrations revealed some shorter chromosomal segments, which are novel to TGCT, as well as known aberrations for which we here refined boundaries. These include gains from 7p15.2 and 21q22.2, and losses of 4p16.3 and 22q13.3. Integration of DNA copy number information to gene expression profiles identified that BRCC3, FOS, MLLT11, NES, and RAC1 may act as novel oncogenes in TGCT. Similarly, DDX26, ERCC5, FZD4, NME4, OPTN, and RB1 were both lost and under-expressed genes, and are thus putative TGCT suppressor genes. CONCLUSION: This first genome-wide integrated array-CGH and gene expression profiling of TGCT provides novel insights into the genome biology underlying testicular tumorigenesis.


Asunto(s)
Aberraciones Cromosómicas , Cromosomas Humanos Par 12/genética , Regulación Neoplásica de la Expresión Génica , Genoma Humano/genética , Neoplasias de Células Germinales y Embrionarias/genética , Análisis de Secuencia por Matrices de Oligonucleótidos/métodos , Neoplasias Testiculares/genética , Adolescente , Adulto , ADN de Neoplasias/genética , Dosificación de Gen , Perfilación de la Expresión Génica , Genes Relacionados con las Neoplasias , Humanos , Masculino , Hibridación de Ácido Nucleico , Reproducibilidad de los Resultados , Regulación hacia Arriba/genética
11.
Cancer Genet Cytogenet ; 147(1): 1-8, 2003 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-14580764

RESUMEN

Allelic imbalance (AI) at loci on chromosome 11 has been shown in several types of human solid tumors, including testicular germ cell tumors (TGCTs). In this study we have focused on the 11p15 region, which is known for its high density of imprinted genes. Highly polymorphic microsatellite markers were analyzed in a series of 71 TGCTs, and AI was observed in 28 of the tumors (39%) at one or more of the loci analyzed. The AI data were evaluated against the chromosome 11 copy number, determined by fluorescence in situ hybridization with a centromere-specific probe. To evaluate preferential parental allele alterations, the patients' normal and tumor genotypes were compared with the parental genotypes. Both losses and gains of both paternal and maternal alleles were found, and this lack of parental origin specificity of the altered allele suggests that the remaining allele is not inactivated by imprinting. A smallest region of overlapping changes was identified between the markers D11S2351 and D11S2347. In summary, our results support the theory that a nonimprinted 11p15 tumor suppressor gene is involved in the development of a subgroup of TGCTs.


Asunto(s)
Cromosomas Humanos Par 11 , Impresión Genómica/genética , Germinoma/genética , Neoplasias Testiculares/genética , Alelos , Aberraciones Cromosómicas , Mapeo Cromosómico , Cartilla de ADN , Marcadores Genéticos , Germinoma/patología , Humanos , Hibridación Fluorescente in Situ , Masculino , Repeticiones de Microsatélite/genética , Reacción en Cadena de la Polimerasa , Neoplasias Testiculares/patología
12.
Oncogene ; 21(24): 3909-16, 2002 May 30.
Artículo en Inglés | MEDLINE | ID: mdl-12032829

RESUMEN

The genetic nature of testicular germ cell tumors and the molecular mechanisms underlying the morphological and clinical differences between the two subtypes, seminomas and nonseminomas, remains unclear. Genetic studies show that both subtypes exhibit many of the same regional genomic disruptions, although the frequencies vary and few clear differences are found. We demonstrate significant epigenetic differences between seminomas and nonseminomas by restriction landmark genomic scanning. Seminomas show almost no CpG island methylation, in contrast to nonseminomas that show CpG island methylation at a level similar to other solid tumors. We find an average of 1.11% of CpG islands methylation in nonseminomas, but only 0.08% methylated in seminomas. Furthermore, we demonstrate that seminomas are more highly hypomethylated than nonseminomas throughout their genome. Since both subtypes are thought to arise from primordial germ cells, the epigenetic differences seen between these subtypes may reflect the normal developmental switch in primordial germ cells from an undermethylated genome to a normally methylated genome. We discuss these findings in relation to different developmental models for seminomatous and nonseminomatous testicular germ cell tumors.


Asunto(s)
Islas de CpG , Seminoma/genética , Seminoma/metabolismo , Neoplasias Testiculares/genética , Neoplasias Testiculares/metabolismo , Southern Blotting , Clonación Molecular , Citosina/metabolismo , Metilación de ADN , Genoma Humano , Humanos , Masculino , Modelos Biológicos , Fenotipo
13.
Genes Chromosomes Cancer ; 33(3): 304-9, 2002 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-11807988

RESUMEN

About 20% of breast carcinomas show no clonal chromosome abnormalities when analyzed after short-term culturing. An interesting question is whether this subset of breast carcinomas really is karyotypically normal or if selection for normal cells occurred in vitro. To address this issue, 26 breast carcinomas that had shown no cytogenetic changes by chromosome banding analysis were examined by comparative genomic hybridization (CGH), a technique that does not require culturing or tumor metaphase cells. All but one case showed copy number changes by CGH (median, four). A comparison of these findings with those of a karyotypically abnormal series analyzed using the same CGH protocol found that the cytogenetically "normal" cases were typically genetically less complex (median, four and eight, respectively; P = 0.0058). Although largely the same alterations were found in both series, some differences with respect to the frequencies of specific imbalances were seen. Gains of 3p and 6q and losses of 10q, 14q, and 17p more often were found in the cytogenetically abnormal series than in the normal tumors. We conclude that in most instances cells found to be normal by chromosome banding analysis after short-term culture do not belong to the tumor parenchyma. Furthermore, when we compared the distribution of the number of imbalances detected by CGH in the total data set according to the mitotic index in vivo (scored from 1 to 3), the median values were three, seven, and 18, respectively (P < 0.001). These data indicate not only that karyotypically normal breast carcinomas may represent a genetically simpler subgroup that grows poorly in vitro but also that this subset of tumors already has a slow growth rate in vivo.


Asunto(s)
Neoplasias de la Mama/genética , Dosificación de Gen , Genoma Humano , Neoplasias de la Mama/patología , Aberraciones Cromosómicas/estadística & datos numéricos , Femenino , Humanos , Hibridación in Situ/estadística & datos numéricos , Cariotipificación , Hibridación de Ácido Nucleico/métodos , Células Tumorales Cultivadas
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA