Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
1.
Genes Chromosomes Cancer ; 61(10): 629-634, 2022 10.
Artigo em Inglês | MEDLINE | ID: mdl-35639830

RESUMO

The diagnosis of acute promyelocytic leukemia (APL) relies on the identification of PML::RARA fusion. While the majority of APL cases harbor a typical t(15;17)(q24;q21), atypical genetic mechanisms leading to the oncogenic PML::RARA fusion have been reported yet their frequency and scope remain poorly characterized. We assessed the genetic findings of 831 cases with APL investigated with concurrent chromosome banding analysis and dual-color dual-fusion fluorescence in situ hybridization (D-FISH) analysis at our institution over an 18.5-year timeframe. Seven hundred twenty-three (87%) cases had a typical balanced t(15;17) with both testing modalities. Atypical karyotypic results including complex translocations, unbalanced rearrangements and insertional events occurred in 50 (6%) cases, while 6 (0.7%) cases were cryptic by conventional chromosome studies despite PML::RARA fusion by D-FISH evaluation. Atypical FISH patterns were observed in 48 (6%) cases despite apparently balanced t(15;17) on chromosome banding analysis. Two hundred fifty (30%) cases displayed additional chromosome abnormalities of which trisomy/tetrasomy 8 (37%), del(7q)/add(7q) (12%), and del(9q) (7%) were most frequent. Complex and very complex karyotypes were observed in 81 (10%) and 34 (4%) cases, respectively. In addition, 4 (0.5%) cases presented as an apparently doubled, near-tetraploid stemline clone. This report provides the largest appraisal of cytogenetic findings in APL with conventional chromosome and PML::RARA D-FISH analysis. By characterizing the frequency and breadth of typical and atypical results through the lens of these cytogenetic testing modalities, this study serves as a pragmatic source of information for those involved in the investigation of APL in both the clinical and research laboratory settings.


Assuntos
Leucemia Promielocítica Aguda , Cromossomos Humanos Par 15/genética , Cromossomos Humanos Par 17/genética , Cromossomos Humanos Par 8 , Humanos , Hibridização in Situ Fluorescente , Leucemia Promielocítica Aguda/genética , Proteínas de Fusão Oncogênica/genética , Estudos Retrospectivos , Translocação Genética , Trissomia
2.
Eur J Haematol ; 102(1): 87-96, 2019 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-30270457

RESUMO

OBJECTIVE: Acute myeloid leukemia (AML) can be subtyped based on recurrent cytogenetic and molecular genetic abnormalities with diagnostic and prognostic significance. Although cytogenetic characterization classically involves conventional chromosome and/or fluorescence in situ hybridization (FISH) assays, limitations of these techniques include poor resolution and the inability to precisely identify breakpoints. METHOD: We evaluated whether an NGS-based methodology that detects structural abnormalities and copy number changes using mate pair sequencing (MPseq) can enhance the diagnostic yield for patients with AML. RESULTS: Using 68 known abnormal and 20 karyotypically normal AML samples, each recurrent primary AML-specific abnormality previously identified in the abnormal samples was confirmed using MPseq. Importantly, in eight cases with abnormalities that could not be resolved by conventional cytogenetic studies, MPseq was utilized to molecularly define eight recurrent AML-fusion events. In addition, MPseq uncovered two cryptic abnormalities that were missed by conventional cytogenetic studies. Thus, MPseq improved the diagnostic yield in the detection of AML-specific structural rearrangements in 10/88 (11%) of cases analyzed. CONCLUSION: Utilization of MPseq represents a precise, molecular-based technique that can be used as an alternative to conventional cytogenetic studies for newly diagnosed AML patients with the potential to revolutionize the diagnosis of hematologic malignancies.


Assuntos
Aberrações Cromossômicas , Genômica , Leucemia Mieloide Aguda/diagnóstico , Leucemia Mieloide Aguda/genética , Análise de Sequência de DNA , Idoso , Biologia Computacional/métodos , Feminino , Genômica/métodos , Sequenciamento de Nucleotídeos em Larga Escala , Humanos , Hibridização in Situ Fluorescente , Cariotipagem , Masculino , Proteínas de Fusão Oncogênica/genética
3.
Am J Surg Pathol ; 42(12): 1708-1714, 2018 12.
Artigo em Inglês | MEDLINE | ID: mdl-30303818

RESUMO

Perineuriomas are rare nerve sheath tumors, divided into intraneural and extraneural (soft tissue) types. Intraneural perineuriomas frequently contain TRAF7 mutations, and rarely, chr22q12 deletions. While chr22q losses can occur in soft tissue perineuriomas, comprehensive high-resolution molecular profiling has not been reported in these tumors and TRAF7 status is unknown. We used whole-exome sequencing and OncoScan single nucleotide polymorphism (SNP) array to evaluate 14 soft tissue perineuriomas. Thirteen cases showed 2 or more chromosomal abnormalities, composed primarily of large deletions. Recurrent chr22q deletions, containing the NF2 locus (n=6) and the previously unreported finding of chr17q deletions, with the NF1 locus (n=4) were frequent events and were mutually exclusive in all but1 case. In addition, 5 cases had varying chr2 deletions; and 4 cases had chr6 deletions. A chr10 deletion (previously reported in the sclerosing variant of soft tissue perineurioma) was observed in one case and another case had chr7 chromothripsis as the sole chromosomal abnormality. No TRAF7 mutations or alterations were identified in any case and no other evaluated gene (MAF<0.0001) had recurrent, deleterious mutations in >2 cases. The molecular genetic profiles showed no association with patient sex, age, tumoral histology or anatomic site. OncoScan SNP array analysis was performed on 10 cases and showed high concordance with the whole exome data, validating the large-scale deletions, duplications, and chr7 chromothripsis findings. In soft tissue perineuriomas, recurrent 22q12 deletions (with NF2) and 17q11 deletions (with NF1) appear to be mutually exclusive events, and alterations in NF1 or NF2 likely contribute to perineurioma pathogenesis, similar to other nerve sheath tumors. Moreover, the lack of TRAF7 mutations in soft tissue perineuriomas indicates divergent pathogenetic mechanisms from those of intraneural perineuriomas.


Assuntos
Biomarcadores Tumorais/genética , Deleção Cromossômica , Cromotripsia , Neoplasias de Bainha Neural/genética , Mutação Puntual , Polimorfismo de Nucleotídeo Único , Neoplasias de Tecidos Moles/genética , Adulto , Idoso de 80 Anos ou mais , Criança , Análise Mutacional de DNA/métodos , Feminino , Predisposição Genética para Doença , Humanos , Masculino , Pessoa de Meia-Idade , Neoplasias de Bainha Neural/patologia , Neurofibromina 1/genética , Neurofibromina 2 , Análise de Sequência com Séries de Oligonucleotídeos , Fenótipo , Neoplasias de Tecidos Moles/patologia , Transcriptoma , Peptídeos e Proteínas Associados a Receptores de Fatores de Necrose Tumoral/genética , Sequenciamento do Exoma , Adulto Jovem
4.
Proc Natl Acad Sci U S A ; 110(37): 14990-4, 2013 Sep 10.
Artigo em Inglês | MEDLINE | ID: mdl-23980137

RESUMO

Obesity is a highly heritable condition and a risk factor for other diseases, including type 2 diabetes, cardiovascular disease, hypertension, and cancer. Recently, genomic copy number variation (CNV) has been implicated in cases of early onset obesity that may be comorbid with intellectual disability. Here, we describe a recurrent CNV that causes a syndrome associated with intellectual disability, seizures, macrocephaly, and obesity. This unbalanced chromosome translocation leads to duplication of over 100 genes on chromosome 12, including the obesity candidate gene G protein ß3 (GNB3). We generated a transgenic mouse model that carries an extra copy of GNB3, weighs significantly more than its wild-type littermates, and has excess intraabdominal fat accumulation. GNB3 is highly expressed in the brain, consistent with G-protein signaling involved in satiety and/or metabolism. These functional data connect GNB3 duplication and overexpression to elevated body mass index and provide evidence for a genetic syndrome caused by a recurrent CNV.


Assuntos
Duplicação Gênica , Proteínas Heterotriméricas de Ligação ao GTP/genética , Obesidade Infantil/genética , Adolescente , Adulto , Animais , Encéfalo/metabolismo , Criança , Pré-Escolar , Deleção Cromossômica , Cromossomos Humanos Par 12/genética , Cromossomos Humanos Par 8/genética , Modelos Animais de Doenças , Feminino , Proteínas de Ligação ao GTP/metabolismo , Proteínas Heterotriméricas de Ligação ao GTP/metabolismo , Humanos , Masculino , Camundongos , Camundongos Transgênicos , Obesidade Infantil/metabolismo , Obesidade Infantil/patologia , Linhagem , Síndrome , Translocação Genética
5.
J Clin Invest ; 123(6): 2502-8, 2013 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-23676460

RESUMO

Small intestine neuroendocrine tumors (SI-NETs) are the most common malignancy of the small bowel. Several clinical trials target PI3K/Akt/mTOR signaling; however, it is unknown whether these or other genes are genetically altered in these tumors. To address the underlying genetics, we analyzed 48 SI-NETs by massively parallel exome sequencing. We detected an average of 0.1 somatic single nucleotide variants (SNVs) per 106 nucleotides (range, 0-0.59), mostly transitions (C>T and A>G), which suggests that SI-NETs are stable cancers. 197 protein-altering somatic SNVs affected a preponderance of cancer genes, including FGFR2, MEN1, HOOK3, EZH2, MLF1, CARD11, VHL, NONO, and SMAD1. Integrative analysis of SNVs and somatic copy number variations identified recurrently altered mechanisms of carcinogenesis: chromatin remodeling, DNA damage, apoptosis, RAS signaling, and axon guidance. Candidate therapeutically relevant alterations were found in 35 patients, including SRC, SMAD family genes, AURKA, EGFR, HSP90, and PDGFR. Mutually exclusive amplification of AKT1 or AKT2 was the most common event in the 16 patients with alterations of PI3K/Akt/mTOR signaling. We conclude that sequencing-based analysis may provide provisional grouping of SI-NETs by therapeutic targets or deregulated pathways.


Assuntos
Neoplasias Intestinais/genética , Tumores Neuroendócrinos/genética , Sequência de Bases , Exoma , Genes Neoplásicos , Genômica , Humanos , Neoplasias Intestinais/mortalidade , Mutação , Tumores Neuroendócrinos/mortalidade , Polimorfismo de Nucleotídeo Único , Proteínas Proto-Oncogênicas c-akt/genética , Sítios de Splice de RNA , Análise de Sequência de DNA , Transdução de Sinais/genética
6.
Am J Hum Genet ; 87(5): 618-30, 2010 Nov 12.
Artigo em Inglês | MEDLINE | ID: mdl-21055719

RESUMO

Autism spectrum disorders (ASD) and schizophrenia are neurodevelopmental disorders for which recent evidence indicates an important etiologic role for rare copy number variants (CNVs) and suggests common genetic mechanisms. We performed cytogenomic array analysis in a discovery sample of patients with neurodevelopmental disorders referred for clinical testing. We detected a recurrent 1.4 Mb deletion at 17q12, which harbors HNF1B, the gene responsible for renal cysts and diabetes syndrome (RCAD), in 18/15,749 patients, including several with ASD, but 0/4,519 controls. We identified additional shared phenotypic features among nine patients available for clinical assessment, including macrocephaly, characteristic facial features, renal anomalies, and neurocognitive impairments. In a large follow-up sample, the same deletion was identified in 2/1,182 ASD/neurocognitive impairment and in 4/6,340 schizophrenia patients, but in 0/47,929 controls (corrected p = 7.37 × 10⁻5). These data demonstrate that deletion 17q12 is a recurrent, pathogenic CNV that confers a very high risk for ASD and schizophrenia and show that one or more of the 15 genes in the deleted interval is dosage sensitive and essential for normal brain development and function. In addition, the phenotypic features of patients with this CNV are consistent with a contiguous gene syndrome that extends beyond RCAD, which is caused by HNF1B mutations only.


Assuntos
Cromossomos Humanos Par 17 , Variações do Número de Cópias de DNA , Esquizofrenia/genética , Deleção de Sequência , Criança , Transtornos Globais do Desenvolvimento Infantil/genética , Pré-Escolar , Fácies , Feminino , Humanos , Masculino , Fenótipo
7.
Am J Med Genet A ; 152A(10): 2623-7, 2010 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-20815035

RESUMO

Juvenile polyposis syndrome (JPS) is a hereditary condition characterized by development of gastrointestinal polyps, and caused by mutations in SMAD4 or BMPR1A genes. Juvenile polyps can also be found in a related group of syndromes with multisystemic involvement including Cowden disease, Lhermitte-Duclos disease, Bannayan-Riley-Ruvalcaba syndrome, and Proteus-like syndrome, all grouped as PTEN hamartoma tumor syndromes (PHTS). In all these conditions including JPS, polyps manifest in older childhood or early adulthood. Infantile juvenile polyposis (JPI) is a rare entity, presenting in the first year of life with severe gastrointestinal symptoms. Many of these patients have associated macrocephaly, hypotonia, and congenital anomalies. It was recently recognized that patients with infantile polyposis have a 10q23 microdeletion, involving both BMPR1A and PTEN genes. There is a major risk for gastrointestinal malignancies in these patients, but the risk for development of other tumors is not known. We describe a patient with a history of infantile polyposis, macrocephaly, developmental delay, hypotonia, and a 10q23 microdeletion. At age 14 she presented with bilateral mucinous cystadenoma of the ovary. This type of tumor was not previously reported in association with JPS, 10q23 microdeletion syndrome, or infantile polyposis. We believe that ovarian cystadenomas may be another neoplastic complication of infantile polyposis, and that our report widens the spectrum of the 10q23 microdeletion phenotype.


Assuntos
Polipose Adenomatosa do Colo/genética , Cromossomos Humanos Par 10 , Cistadenoma Mucinoso/genética , Neoplasias Ovarianas/genética , Deleção de Sequência , Adulto , Idade de Início , Pré-Escolar , Hibridização Genômica Comparativa , Face/anormalidades , Feminino , Humanos , Lactente , Recém-Nascido , Masculino , Fenótipo
8.
Mayo Clin Proc ; 84(9): 801-10, 2009 09.
Artigo em Inglês | MEDLINE | ID: mdl-19720778

RESUMO

OBJECTIVE: To use fluorescence in situ hybridization (FISH) to visualize genetic abnormalities in interphase cell nuclei (interphase FISH) of acinar cell carcinoma, ductal adenocarcinoma, and islet cell carcinoma of the pancreas. PATIENTS AND METHODS: Between April 4, 2007, and December 4, 2008, interphase FISH was used to study paraffin-embedded preparations of tissue obtained from 18 patients listed in the Mayo Clinic Biospecimen Resource for Pancreas Research with a confirmed diagnosis of acinar cell carcinoma, ductal adenocarcinoma, islet cell carcinoma, or pancreas without evidence of neoplasia. FISH probes were used for chromosome loci of APC (see glossary at end of article for expansion of all gene symbols), BRCA2, CTNNB1, EGFR, ERBB2, CDKN2A, TP53, TYMP, and TYMS. These FISH probes were used with control probes to distinguish among various kinds of chromosome abnormalities of number and structure. RESULTS: FISH abnormalities were observed in 12 (80%) of 15 patients with pancreatic cancer: 5 of 5 patients with acinar cell carcinoma, 5 of 5 patients with ductal adenocarcinoma, and 2 (40%) of 5 patients with islet cell carcinoma. All 3 specimens of pancreatic tissue without neoplasia had normal FISH results. Gains of CTNNB1 due to trisomy 3 occurred in each tumor with acinar cell carcinoma but in none of the other tumors in this study. FISH abnormalities of all other cancer genes studied were observed in all forms of pancreatic tumors in this investigation. CONCLUSION: FISH abnormalities of CTNNB1 due to trisomy 3 were observed only in acinar cell carcinoma. FISH abnormalities of genes implicated in familial cancer, tumor progression, and the 5-fluorouracil pathway were common but were not associated with specific types of pancreatic cancer.


Assuntos
Carcinoma de Células Acinares/genética , Carcinoma de Células das Ilhotas Pancreáticas/genética , Carcinoma Ductal Pancreático/genética , Aberrações Cromossômicas , Neoplasias Pancreáticas/genética , Carcinoma de Células Acinares/patologia , Carcinoma de Células das Ilhotas Pancreáticas/patologia , Carcinoma Ductal Pancreático/patologia , Feminino , Humanos , Hibridização in Situ Fluorescente , Interfase/genética , Masculino , Neoplasias Pancreáticas/patologia
9.
Leuk Res ; 33(5): 662-4, 2009 May.
Artigo em Inglês | MEDLINE | ID: mdl-18937974

RESUMO

Comparative genomic hybridization (CGH), using oligo arrays with either 44,000 or 105,000 oligonucleotides, was performed on granulocyte-derived DNA from 71 patients with BCR-ABL-negative classic myeloproliferative neoplasms (MPNs): 32 primary myelofibrosis (PMF), 26 polycythemia vera (PV) and 13 essential thrombocythemia (ET). Copy number changes (CNCs) were detected in 44%, 35%, and 15% of the cases with PMF, PV and ET, respectively. In ET and PMF, CNCs were more frequently detected in the presence of JAK2V617F (50% vs. 19%; p=0.05). Conventional chromosome analysis was obtained in 57 patients either at diagnosis or within 1 year of the array CGH study; all 21 patients with PV and 11 with ET displayed normal cytogenetic findings despite the presence of CNCs in 29% and 18%, respectively. In PMF, the respective rates of CNCs and abnormal karyotype were 48% and 36%; karyotypic abnormalities, including unbalanced translocations, were often detected by array CGH as chromosomal gains or losses. This preliminary report suggests a potential value for array CGH in terms of both clinical diagnostics and genomic research in MPNs.


Assuntos
Aberrações Cromossômicas , Janus Quinase 2/genética , Mutação , Transtornos Mieloproliferativos/genética , Hibridização de Ácido Nucleico , Análise de Sequência com Séries de Oligonucleotídeos , Adolescente , Adulto , Idoso , Idoso de 80 Anos ou mais , Feminino , Humanos , Cariotipagem , Masculino , Pessoa de Meia-Idade
10.
Am J Clin Pathol ; 130(2): 178-85, 2008 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-18628085

RESUMO

Translocations involving the T-cell receptor (TCR) and TCL1 genes occur in T-cell precursor lymphoblastic leukemia/lymphoma and prolymphocytic leukemia; isochromosome 7q has been associated with hepatosplenic T-cell lymphoma. However, the incidence of these abnormalities in peripheral T-cell lymphomas (PTCLs) as a whole has not been well defined. We studied genetic abnormalities in 124 PTCLs seen at the Mayo Clinic, Rochester, MN, between 1987 and 2007. Tissue microarrays were screened using 2-color break-apart fluorescence in situ hybridization probes flanking the TCRalpha (TCRA, 14q11), TCRbeta (TCRB, 7q35), and TCRgamma (TCRG, 7p15) genes and the TCL1 gene (14q32). Isochromosome 7q was analyzed by using a 2-color probe to 7p and 7q32.1. Translocations involved TCRA in 3 (2.9%) of 102 cases and TCRB in 1 (1%) of 88. Isochromosome 7q was detected in 2 cases of extranodal NK/T-cell lymphoma, nasal type, and 2 cases of anaplastic lymphoma kinase-negative anaplastic large cell lymphoma. One of the latter cases also had a translocation of TCRA, and further studies confirmed a novel t(5;14) translocation.


Assuntos
Proteínas Proto-Oncogênicas/genética , Adulto , Idoso , Cromossomos Humanos Par 7 , Feminino , Humanos , Hibridização in Situ Fluorescente , Isocromossomos , Linfoma de Células T Periférico , Masculino , Pessoa de Meia-Idade , Receptores de Antígenos de Linfócitos T , Análise Serial de Tecidos , Translocação Genética
11.
Mayo Clin Proc ; 83(7): 825-46, 2008 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-18613999

RESUMO

Malignant melanoma presents a substantial clinical challenge. Current diagnostic methods are limited in their ability to diagnose early disease and accurately predict individual risk of disease progression and outcome. The lack of adequate approaches to properly define disease subgroups precludes rational treatment design and selection. Better tools are urgently needed to provide more accurate and personalized melanoma patient management. Recent progress in the understanding of the molecular aberrations that underlie melanoma oncogenesis will likely advance the diagnosis, prognosis, and treatment of melanoma. The emerging pattern of molecular complexity in melanoma tumors mirrors the clinical diversity of the disease and highlights the notion that melanoma, like other cancers, is not a single disease but a heterogeneous group of disorders that arise from complex molecular changes. Understanding of molecular aberrations involving important cellular processes, such as cellular signaling networks, cell cycle regulation, and cell death, will be essential for better diagnosis, accurate assessment of prognosis, and rational design of effective therapeutics. Defining an individual patient's unique tumor characteristics may lead to personalized prediction of outcomes and selection of therapy. We review the emerging molecular landscape of melanoma and its implications for better management of patients with melanoma.


Assuntos
Biomarcadores Tumorais/genética , Melanoma , Biologia Molecular/métodos , Apoptose , Ciclo Celular/genética , Proliferação de Células , Diagnóstico Diferencial , Progressão da Doença , Técnicas Genéticas , Humanos , Melanoma/diagnóstico , Melanoma/genética , Melanoma/terapia , Prognóstico
12.
Cancer Genet Cytogenet ; 167(2): 177-81, 2006 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-16737921

RESUMO

Loss of tumor protein 53 (TP53) has been associated with aggressive disease and poor response to therapy in B-cell chronic lymphocytic leukemia (B-CLL). TP53 is located at chromosome band 17p13 and its absence can be detected by fluorescence in situ hybridization (FISH) in the interphase nuclei of 8-10% patients with B-CLL. To study the cytogenetic mechanism for loss of TP53, metaphase and interphase FISH studies were conducted on 16 B-CLL patients to investigate 17p10 to 17p12, a chromosome region known to be rich in low-copy DNA repeats. Loss of TP53 was caused by an isochromosome with breakpoints between 17p10 and 17p11.2 in four patients, an unbalanced translocation involving 17p10 to 17p11.2 in nine patients, and an unbalanced translocation involving 17p11.2 to 17p12 in three patients. These findings indicate that loss of TP53 results from the absence of nearly the entire chromosome 17 p-arm rather than to monosomy 17 or deletions of TP53. Translocations or isochromosome formations at sites of low-copy DNA repeats in 17p10 to 17p12 appear to be the mechanism for the loss of TP53 in B-CLL.


Assuntos
Deleção Cromossômica , Cromossomos Humanos Par 17 , Genes p53 , Isocromossomos , Leucemia Linfocítica Crônica de Células B/genética , Translocação Genética , Idoso , Cromossomos Humanos Par 17/ultraestrutura , Feminino , Deleção de Genes , Humanos , Hibridização in Situ Fluorescente , Masculino , Pessoa de Meia-Idade
13.
Genet Med ; 8(1): 16-23, 2006 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-16418595

RESUMO

PURPOSE: Validation of fluorescence in situ hybridization assays is required before using them in clinical practice. Yet, there are few published examples that describe the validation process, leading to inconsistent and sometimes inadequate validation practices. The purpose of this article is to describe a broadly applicable preclinical validation process. METHODS: Validation is performed using four consecutive experiments. The Familiarization experiment tests probe performance on metaphase cells to measure analytic sensitivity and specificity for normal blood specimens. The Pilot Study tests a variety of normal and abnormal specimens, using the intended tissue type, to set a preliminary normal cutoff and establish the analytic sensitivity. The Clinical Evaluation experiment tests these parameters in a series of normal and abnormal specimens to simulate clinical practice, establish the normal cutoff and abnormal reference ranges, and finalize the standard operating procedure. The Precision experiment measures the reproducibility of the new assay over 10 consecutive working days. To illustrate documentation and analysis of data with this process, the results for a new assay to detect fusion of IGH and BCL3 associated with t(14;19)(q32;q13.3) in lymphoproliferative disorders are provided in this report. RESULTS: These four experiments determine the analytic sensitivity and specificity, normal values, precision, and reportable reference ranges for validation of the new test. CONCLUSION: This report describes a method for preclinical validation of fluorescence in situ hybridization studies of metaphase cells and interphase nuclei using commercial or home brew probes.


Assuntos
Técnicas de Laboratório Clínico , Sondas de DNA , Corantes Fluorescentes , Hibridização in Situ Fluorescente , Transtornos Linfoproliferativos/diagnóstico , Proteína 3 do Linfoma de Células B , Cromossomos Humanos Par 14/genética , Cromossomos Humanos Par 19/genética , Humanos , Hibridização in Situ Fluorescente/métodos , Transtornos Linfoproliferativos/genética , Proteínas de Fusão Oncogênica/genética , Projetos Piloto , Valor Preditivo dos Testes , Proteínas Proto-Oncogênicas/genética , Reprodutibilidade dos Testes , Fatores de Transcrição , Translocação Genética
14.
Oncogene ; 22(46): 7233-42, 2003 Oct 16.
Artigo em Inglês | MEDLINE | ID: mdl-14562053

RESUMO

The development of cervical cancer is highly associated with human papillomavirus (HPV) infection. Greater than 99% of all cervical tumors contain HPV DNA. Integration of high-risk HPV has been temporally associated with the acquisition of a malignant phenotype. Recent work from our lab has shown that HPV16, the most common high-risk HPV associated with cervical carcinoma, preferentially integrates at loci containing human common fragile sites (CFSs). CFSs are regions of genomic instability that have also been associated with deletions, translocations, and gene amplification during cancer development. The current work shows that HPV18, the second most prevalent high-risk HPV type found in cervical tumors, preferentially targets the CFSs. We identified 27 unique HPV18 integrations in cervical tumors, of which 63% (P<0.001) occur in CFSs. However, the distribution of HPV18 integrations found were profoundly different from those found for HPV16. Specifically, 30% of all HPV18 integrations occurred within the chromosomal band 8q24 near the c-myc proto-oncogene. None of the HPV16 integrations occurred in this region. Previous low-resolution mapping suggested that c-myc may be a target of HPV integration. Our data at nucleotide resolution confirm that in HPV18-positive cervical tumors, the region surrounding c-myc is indeed a hot spot of viral integration. These results demonstrate that CFSs are preferred sites of integration for HPV18 in cervical tumors. In addition, we have identified multiple cellular genes that have been disrupted by HPV18 integration in cervical tumors. Our results suggest that the sites of HPV18 integration are nonrandom and may play an important role in the development of cervical tumors.


Assuntos
Cromossomos Humanos Par 8 , Genes myc , Papillomaviridae/genética , Neoplasias do Colo do Útero/genética , Neoplasias do Colo do Útero/virologia , Mapeamento Cromossômico , Troca Genética , Feminino , Humanos , Papillomaviridae/classificação , Papillomaviridae/isolamento & purificação , Proto-Oncogene Mas , Integração Viral
15.
Oncogene ; 22(8): 1225-37, 2003 Feb 27.
Artigo em Inglês | MEDLINE | ID: mdl-12606949

RESUMO

The development of cervical cancer is highly associated with human papillomavirus (HPV) infection. HPV integration into the genome of infected cervical cells is temporally associated with the acquisition of the malignant phenotype. A relationship between the sites of HPV integration in cervical cancer and the position of the common fragile sites (CFSs) has been observed at both the cytogenetic and molecular levels. To further explore this relationship at the molecular level, we used RS-PCR to rapidly isolate cellular sequences flanking the sites of HPV16 integration in 26 primary cervical tumors. Human bacterial artificial chromosome clones were isolated based on these flanking sequences and used as probes for fluorescence in situ hybridization on aphidicolin-stimulated metaphases. Our data demonstrate that 11/23 HPV16 integrations in cervical tumors occurred within CFSs (P&<0.001). In addition, we show that deletions and complex rearrangements frequently occur in the cellular sequences targeted by the integrations and that integrations cluster in FRA13C (13q22), FRA3B (3p14.2), and FRA17B (17q23). Finally, our data suggest that cellular genes, such as Notch 1, are disrupted by the HPV16 integrations, which may contribute to the malignant phenotype.


Assuntos
Carcinoma de Células Escamosas/virologia , Transformação Celular Viral/genética , Fragilidade Cromossômica , Cromossomos Humanos/virologia , Papillomaviridae/fisiologia , Infecções por Papillomavirus/virologia , Infecções Tumorais por Vírus/virologia , Neoplasias do Colo do Útero/virologia , Integração Viral/genética , Carcinoma de Células Escamosas/genética , Sítios Frágeis do Cromossomo , Cromossomos Artificiais Bacterianos , DNA Viral/análise , Feminino , Humanos , Mutagênese Insercional , Proteínas de Neoplasias/genética , Papillomaviridae/classificação , Papillomaviridae/isolamento & purificação , Infecções por Papillomavirus/genética , Reação em Cadeia da Polimerase/métodos , Deleção de Sequência , Infecções Tumorais por Vírus/genética , Neoplasias do Colo do Útero/genética
16.
Oncogene ; 21(57): 8713-22, 2002 Dec 12.
Artigo em Inglês | MEDLINE | ID: mdl-12483524

RESUMO

FRA3B is the most frequently expressed common fragile site localized within human chromosomal band 3p14.2, which is frequently deleted in many different cancers, including cervical cancer. Previous reports indicate aphidicolin-induced FRA3B instability occurs over approximately 500 kb which is spanned by the 1.5 Mb fragile histidine triad (FHIT) gene. Recently an HPV16 cervical tumor integration, 2 Mb centromeric to the published FRA3B region, has been identified. FISH-based analysis with a BAC spanning the integration has demonstrated this integration occurs within the FRA3B region of instability. These data suggest that the unstable FRA3B region is much larger than previously reported. FISH-based analysis of aphidicolin-induced metaphase chromosomes allowed for a complete characterization of instability associated with FRA3B. This analysis indicates that fragility extends for 4 Mb. Within this region are a total of five genes, including FHIT. FRA3B gene expression analysis on a panel of cervical tumor-derived cell lines revealed that three of the five genes within FRA3B were aberrantly regulated. A similar analysis of genes outside of FRA3B indicated that the surrounding genes were not aberrantly expressed. These data provide additional support that regions of instability associated with CFSs and the genes contained within them, may play an important role in cancer development.


Assuntos
Hidrolases Anidrido Ácido , Fragilidade Cromossômica , Proteínas de Neoplasias/genética , Sequência de Bases , Carcinoma de Células Escamosas/virologia , Sítios Frágeis do Cromossomo , Cromossomos Humanos Par 3 , DNA de Neoplasias , DNA Viral , Feminino , Humanos , Hibridização in Situ Fluorescente , Dados de Sequência Molecular , Papillomaviridae/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Neoplasias do Colo do Útero/virologia , Integração Viral
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA