Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
1.
J Mol Diagn ; 23(10): 1269-1278, 2021 10.
Artigo em Inglês | MEDLINE | ID: mdl-34325058

RESUMO

Alterations in the BCOR gene, including internal tandem duplications (ITDs) of exon 15 have emerged as important oncogenic changes that define several diagnostic entities. In pediatric cancers, BCOR ITDs have recurrently been described in clear cell sarcoma of kidney (CCSK), primitive myxoid mesenchymal tumor of infancy (PMMTI), and central nervous system high-grade neuroepithelial tumor with BCOR ITD in exon 15 (HGNET-BCOR ITDex15). In adults, BCOR ITDs are also reported in endometrial and other sarcomas. The utility of multiplex targeted RNA sequencing for the identification of BCOR ITD in pediatric cancers was investigated. All available archival cases of CCSK, PMMTI, and HGNET-BCOR ITDex15 were collected. Each case underwent anchored multiplex PCR library preparation with a custom-designed panel, with BCOR targeted for both fusions and ITDs. BCOR ITD was detected in all cases across three histologic subtypes using the RNA panel, with no other fusions identified in any of the cases. All BCOR ITDs occurred in the final exon, within 16 codons from the stop sequence. Multiplex targeted RNA sequencing from formalin-fixed, paraffin-embedded tissue is successful at identifying BCOR internal tandem duplications. This analysis supports the use of anchored multiplex PCR targeted RNA next-generation sequencing panels for identification of BCOR ITDs in pediatric tumors. The use of post-analytic algorithms to improve the detection of BCOR ITD using DNA panels was also explored.


Assuntos
Neoplasias Encefálicas/genética , Sequenciamento de Nucleotídeos em Larga Escala/métodos , Neoplasias Renais/genética , Neoplasias Neuroepiteliomatosas/genética , Proteínas Proto-Oncogênicas/genética , Proteínas Repressoras/genética , Sarcoma de Células Claras/genética , Análise de Sequência de RNA/métodos , Neoplasias de Tecidos Moles/genética , Sequências de Repetição em Tandem/genética , Criança , Pré-Escolar , Códon/genética , Éxons , Feminino , Humanos , Lactente , Masculino , Reação em Cadeia da Polimerase Multiplex/métodos , Oncogenes , Reprodutibilidade dos Testes
2.
Mod Pathol ; 33(5): 775-780, 2020 05.
Artigo em Inglês | MEDLINE | ID: mdl-31827231

RESUMO

Cranial fasciitis is a benign myofibroproliferative lesion of the scalp and underlying bones typically occurring in the pediatric population. Histologically, it is characterized by loose fascicles of stellate cells in a fibromyxoid background, findings similar to those described in the closely related variant nodular fasciitis. Previously characterized as a reactive process, the identification of USP6 translocations in over 90% of nodular fasciitis cases prompted their reclassification as a clonal neoplastic process. Unlike nodular fasciitis, the molecular underpinnings of cranial fasciitis are less clear. While a subset of cranial fasciitis has been associated with Wnt/ß-catenin pathway dysregulation, recent case reports suggest that this entity may also harbor USP6 fusions, a finding we sought to further investigate. We identified fifteen archival cases of cranial fasciitis, five females and ten males ranging in age from 3 months to 9 years (median 11 months), composed of formalin-fixed paraffin-embedded and fresh frozen tissues (11 and 4 cases respectively). Samples were evaluated on an RNA-based targeted sequencing panel targeting genes recurrently rearranged in neoplasia, including USP6. Five of fifteen cases (33%) were positive for USP6 rearrangements predicted to result in the fusion of the entire USP6 coding region to the promoter of the 5' partner, (three of which were novel):  two SERPINH1-USP6 (novel) and one each of COL3A1-USP6 (novel), SPARC-USP6, and MYH9-USP6. These results demonstrate the recurrent nature of USP6 rearrangements in cranial fasciitis, and highlight the success of targeted RNA sequencing in identifying known and novel fusion partners. The identification of USP6 promoter-swapping rearrangements is helpful in understanding the underlying biology of cranial fasciitis, and reinforces its biologic relationship to nodular fasciitis. Targeted RNA sequencing is a helpful tool in diagnosing this pseudosarcomatous lesion.


Assuntos
Fasciite/genética , Couro Cabeludo/patologia , Crânio/patologia , Ubiquitina Tiolesterase/genética , Criança , Pré-Escolar , Fasciite/patologia , Feminino , Humanos , Lactente , Masculino , Proteínas Recombinantes de Fusão/genética
3.
Artigo em Inglês | MEDLINE | ID: mdl-30364844

RESUMO

Purpose: Next-generation sequencing technologies are actively applied in clinical oncology. Bioinformatics pipeline analysis is an integral part of this process; however, humans cannot yet realize the full potential of the highly complex pipeline output. As a result, the decision to include a variant in the final report during routine clinical sign-out remains challenging. Methods: We used an artificial intelligence approach to capture the collective clinical sign-out experience of six board-certified molecular pathologists to build and validate a decision support tool for variant reporting. We extracted all reviewed and reported variants from our clinical database and tested several machine learning models. We used 10-fold cross-validation for our variant call prediction model, which derives a contiguous prediction score from 0 to 1 (no to yes) for clinical reporting. Results: For each of the 19,594 initial training variants, our pipeline generates approximately 500 features, which results in a matrix of > 9 million data points. From a comparison of naive Bayes, decision trees, random forests, and logistic regression models, we selected models that allow human interpretability of the prediction score. The logistic regression model demonstrated 1% false negativity and 2% false positivity. The final models' Youden indices were 0.87 and 0.77 for screening and confirmatory cutoffs, respectively. Retraining on a new assay and performance assessment in 16,123 independent variants validated our approach (Youden index, 0.93). We also derived individual pathologist-centric models (virtual consensus conference function), and a visual drill-down functionality allows assessment of how underlying features contributed to a particular score or decision branch for clinical implementation. Conclusion: Our decision support tool for variant reporting is a practically relevant artificial intelligence approach to harness the next-generation sequencing bioinformatics pipeline output when the complexity of data interpretation exceeds human capabilities.

5.
Mol Cell ; 60(4): 571-83, 2015 Nov 19.
Artigo em Inglês | MEDLINE | ID: mdl-26481663

RESUMO

Phosphoenolpyruvate carboxykinase (PEPCK) is well known for its role in gluconeogenesis. However, PEPCK is also a key regulator of TCA cycle flux. The TCA cycle integrates glucose, amino acid, and lipid metabolism depending on cellular needs. In addition, biosynthetic pathways crucial to tumor growth require the TCA cycle for the processing of glucose and glutamine derived carbons. We show here an unexpected role for PEPCK in promoting cancer cell proliferation in vitro and in vivo by increasing glucose and glutamine utilization toward anabolic metabolism. Unexpectedly, PEPCK also increased the synthesis of ribose from non-carbohydrate sources, such as glutamine, a phenomenon not previously described. Finally, we show that the effects of PEPCK on glucose metabolism and cell proliferation are in part mediated via activation of mTORC1. Taken together, these data demonstrate a role for PEPCK that links metabolic flux and anabolic pathways to cancer cell proliferation.


Assuntos
Neoplasias Colorretais/patologia , Glucose/metabolismo , Glutamina/metabolismo , Complexos Multiproteicos/metabolismo , Fosfoenolpiruvato Carboxiquinase (ATP)/metabolismo , Serina-Treonina Quinases TOR/metabolismo , Animais , Linhagem Celular Tumoral , Proliferação de Células , Neoplasias Colorretais/metabolismo , Glicólise , Células HT29 , Humanos , Alvo Mecanístico do Complexo 1 de Rapamicina , Camundongos , Transplante de Neoplasias
6.
J Clin Invest ; 123(1): 517-25, 2013 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-23257362

RESUMO

Because of the high risk of recurrence in high-grade serous ovarian carcinoma (HGS-OvCa), the development of outcome predictors could be valuable for patient stratification. Using the catalog of The Cancer Genome Atlas (TCGA), we developed subtype and survival gene expression signatures, which, when combined, provide a prognostic model of HGS-OvCa classification, named "Classification of Ovarian Cancer" (CLOVAR). We validated CLOVAR on an independent dataset consisting of 879 HGS-OvCa expression profiles. The worst outcome group, accounting for 23% of all cases, was associated with a median survival of 23 months and a platinum resistance rate of 63%, versus a median survival of 46 months and platinum resistance rate of 23% in other cases. Associating the outcome prediction model with BRCA1/BRCA2 mutation status, residual disease after surgery, and disease stage further optimized outcome classification. Ovarian cancer is a disease in urgent need of more effective therapies. The spectrum of outcomes observed here and their association with CLOVAR signatures suggests variations in underlying tumor biology. Prospective validation of the CLOVAR model in the context of additional prognostic variables may provide a rationale for optimal combination of patient and treatment regimens.


Assuntos
Perfilação da Expressão Gênica , Regulação Neoplásica da Expressão Gênica , Modelos Biológicos , Neoplasias Ovarianas/genética , Neoplasias Ovarianas/metabolismo , Neoplasias Ovarianas/mortalidade , Adulto , Idoso , Proteína BRCA1/biossíntese , Proteína BRCA1/genética , Proteína BRCA2/biossíntese , Proteína BRCA2/genética , Intervalo Livre de Doença , Feminino , Humanos , Pessoa de Meia-Idade , Mutação , Gradação de Tumores , Neoplasias Ovarianas/classificação , Neoplasias Ovarianas/terapia , Valor Preditivo dos Testes , Estudos Prospectivos , Taxa de Sobrevida
7.
PLoS One ; 7(9): e46486, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-23029531

RESUMO

p150, product of the SALL2 gene, is a binding partner of the polyoma virus large T antigen and a putative tumor suppressor. p150 binds to the nuclease hypersensitive element of the c-MYC promoter and represses c-MYC transcription. Overexpression of p150 in human ovarian surface epithelial cells leads to decreased expression, and downregulation to increased expression, of c-MYC. c-MYC is repressed upon restoration of p150 to ovarian carcinoma cells. Induction of apoptosis by etoposide results in recruitment of p150 to the c-MYC promoter and to repression of c-MYC. Analysis of data in The Cancer Genome Atlas shows negative correlations between SALL2 and c-MYC expression in four common solid tumor types.


Assuntos
Genes Supressores de Tumor , Proteínas Proto-Oncogênicas c-myc/genética , Fatores de Transcrição/genética , Transcrição Gênica , Antineoplásicos Fitogênicos/farmacologia , Sequência de Bases , Linhagem Celular Tumoral , Proteínas de Ligação a DNA , Etoposídeo/farmacologia , Regulação Neoplásica da Expressão Gênica , Humanos , Dados de Sequência Molecular , Neoplasias/genética , Neoplasias/metabolismo , Regiões Promotoras Genéticas , Ligação Proteica , Proteínas Proto-Oncogênicas c-myc/metabolismo , Fatores de Transcrição/metabolismo
8.
Am J Surg Pathol ; 35(10): 1531-9, 2011 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-21921781

RESUMO

Tumor necrosis factor-α-inducible protein-2 (TNFAIP2) is a protein upregulated in cultured cells treated with tumor necrosis factor α (TNF), but its expression in normal and neoplastic tissues remains largely unknown. Here, we use standard immunohistochemical techniques to demonstrate that TNFAIP2 is normally expressed by follicular dendritic cells, interdigitating dendritic cells, and macrophages but not by lymphoid cells in secondary lymphoid tissues. Consistent with this expression pattern, we found strong TNFAIP2 staining of tumor cells in 4 of 4 cases (100%) of follicular dendritic cell sarcoma and in 3 of 3 cases (100%) of histiocytic sarcoma. Although TNFAIP2 is not expressed by the small and intermediate-sized neoplastic B cells comprising follicular lymphoma, small lymphocytic lymphoma, mantle cell lymphoma, or marginal zone lymphoma, we observed strong TNFAIP2 staining of the large, neoplastic cells in 31 of 31 cases (100%) of classical Hodgkin lymphoma, in 12 of 12 cases (100%) of nodular lymphocyte-predominant Hodgkin lymphoma, and in 27 of 31 cases (87%) of primary mediastinal (thymic) large B-cell lymphoma. In contrast, TNFAIP2 was expressed by malignant cells in only 2 of 45 cases (4%) of diffuse large B-cell lymphoma, not otherwise specified, in 2 of 18 cases (11%) of Burkitt lymphoma, and in 1 of 19 cases (5%) of anaplastic large cell lymphoma. Further analysis indicates that TNFAIP2, as a single diagnostic marker, is more sensitive (sensitivity=87%) and specific (specificity=96%) than TRAF1, nuclear cRel, or CD23 for distinguishing the malignant B cells of primary mediastinal (thymic) large B-cell lymphoma from those of its morphologic and immunophenotypic mimic, diffuse large B-cell lymphoma, not otherwise specified. Thus, TNFAIP2 may serve as a useful new marker of dendritic and histiocytic sarcomas, the aberrant expression of which in the malignant cells of classical Hodgkin lymphoma and primary mediastinal (thymic) large B-cell lymphoma serves to distinguish these tumors from other large cell lymphomas in routine clinical practice.


Assuntos
Citocinas/metabolismo , Células Dendríticas/patologia , Doença de Hodgkin/diagnóstico , Linfoma Difuso de Grandes Células B/diagnóstico , Neoplasias do Mediastino/diagnóstico , Biomarcadores Tumorais/metabolismo , Células Dendríticas/metabolismo , Diagnóstico Diferencial , Doença de Hodgkin/metabolismo , Humanos , Imuno-Histoquímica , Linfonodos/metabolismo , Linfonodos/patologia , Linfoma Folicular/diagnóstico , Linfoma Difuso de Grandes Células B/metabolismo , Linfoma Imunoblástico de Células Grandes/diagnóstico , Neoplasias do Mediastino/metabolismo , Baço/metabolismo , Baço/patologia
9.
Gynecol Oncol ; 123(1): 5-12, 2011 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-21683992

RESUMO

BACKGROUND: Most high-grade pelvic serous carcinomas (HGPSCs) arise from fallopian tube epithelium (FTE). To date, few markers have been shown to characterize FTE transformation. Stathmin 1 (STMN1) is a candidate oncogene whose activity is influenced by p53, p27Kip1 (p27), and PI3K/Akt pathway activation. As a microtubule destabilizing protein, STMN1 regulates cytoskeletal dynamics, cell cycle progression, mitosis, and cell migration. This study examines the expression of STMN1 and its negative regulator p27 along the morphologic continuum from normal FTE to invasive carcinoma. METHODS: STMN1 and p27 expression were examined by immunohistochemistry (IHC) in benign (n=12) and malignant (n=13) fallopian tubes containing normal epithelium, morphologically benign putative precursor lesions ("p53 signatures"), potential transitional precursor lesions ("proliferative p53 signatures"), tubal intraepithelial carcinoma (TIC), and/or invasive serous carcinoma. STMN1 expression was further assessed in 131 late-stage HGPSCs diagnosed as primary ovarian and in 6 ovarian cancer cell lines by IHC and Western blot, respectively. RESULTS: STMN1 expression was absent in benign FTE and infrequently detected in p53 signatures. However, it was weakly expressed in proliferative p53 signatures and robustly induced upon progression to TIC and invasive carcinoma, typically accompanied by decreased p27 levels. STMN1 was expressed in >80% of high-grade serous ovarian carcinomas and cell lines. CONCLUSIONS: STMN1 is a novel marker of early serous carcinoma that may play a role in FTE tumor initiation. Our data are consistent with a model by which STMN1 overexpression, resulting from loss of p27-mediated regulation, may potentiate aberrant cell proliferation, migration, and/or loss of polarity during early tumorigenesis.


Assuntos
Biomarcadores Tumorais/biossíntese , Neoplasias das Tubas Uterinas/metabolismo , Microtúbulos/metabolismo , Estatmina/biossíntese , Adulto , Idoso , Biomarcadores Tumorais/metabolismo , Linhagem Celular Tumoral , Inibidor de Quinase Dependente de Ciclina p27/metabolismo , Cistadenocarcinoma Seroso/enzimologia , Cistadenocarcinoma Seroso/metabolismo , Neoplasias das Tubas Uterinas/enzimologia , Neoplasias das Tubas Uterinas/patologia , Feminino , Humanos , Pessoa de Meia-Idade , Neoplasias Ovarianas/enzimologia , Neoplasias Ovarianas/metabolismo , Neoplasias Ovarianas/patologia , Fosfatidilinositol 3-Quinases/metabolismo , Transdução de Sinais , Estatmina/metabolismo , Proteína Supressora de Tumor p53/metabolismo
10.
Hum Mutat ; 32(8): 900-11, 2011 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-21462282

RESUMO

Variants in the CDKN2A tumor suppressor are associated with Familial Melanoma (FM), although for many variants the linkage is weak. The effects of missense variants on protein function and pathogenicity are often unclear. Multiple methods (e.g., laboratory, computational, epidemiological) have been developed to analyze whether a missense variant is pathogenic or not. It is not yet clear how to integrate these data types into a strategy for variant classification. We studied 51 CDKN2A missense variants using a cell cycle arrest assay. There was a continuum of results ranging from full wild-type effect through partial activity to complete loss of arrest. A reproducible decrease of 30% of cell cycle arrest activity correlated with FM association. We analyzed missense CDKN2A germline variants using a Bayesian method to combine multiple data types and derive a probability of pathogenicity. When equal to or more than two data types could be evaluated with this method, 22 of 25 FM-associated variants and 8 of 15 variants of uncertain significance were classified as likely pathogenic with >95% probability. The other 10 variants were classified as uncertain (probability 5-95%). For most variants, there were insufficient data to draw a conclusion. The Bayesian model appears to be a sound method of classifying missense variants in cancer susceptibility genes.


Assuntos
Inibidor p16 de Quinase Dependente de Ciclina/genética , Mutação/genética , Polimorfismo de Nucleotídeo Único/genética , Bioensaio , Ciclo Celular/genética , Linhagem Celular Tumoral , Biologia Computacional , Inibidor p16 de Quinase Dependente de Ciclina/metabolismo , Bases de Dados Genéticas , Predisposição Genética para Doença/genética , Humanos , Melanoma/epidemiologia , Melanoma/genética , Melanoma/metabolismo , Melanoma/patologia , Valores de Referência
11.
Mol Cell ; 27(6): 992-1004, 2007 Sep 21.
Artigo em Inglês | MEDLINE | ID: mdl-17889671

RESUMO

The MUC1 heterodimeric transmembrane glycoprotein is aberrantly overexpressed by diverse human carcinomas. Galectin-3 is a beta-galactoside binding protein that has also been associated with the development of human cancers. The present results demonstrate that MUC1 induces galectin-3 expression by a posttranscriptional mechanism. We show that the MUC1 C-terminal subunit is glycosylated on Asn-36 and that this modification is necessary for upregulation of galectin-3. N-glycosylated MUC1-C increases galectin-3 mRNA levels by suppressing expression of the microRNA miR-322 and thereby stabilizing galectin-3 transcripts. The results show that, in turn, galectin-3 binds to MUC1-C at the glycosylated Asn-36 site. The significance of the MUC1-C-galectin-3 interaction is supported by the demonstration that galectin-3 forms a bridge between MUC1 and the epidermal growth factor receptor (EGFR) and that galectin-3 is essential for EGF-mediated interactions between MUC1 and EGFR. These findings indicate that MUC1 and galectin-3 function as part of a miR-322-dependent regulatory loop.


Assuntos
Galectina 3/metabolismo , MicroRNAs/metabolismo , Mucina-1/metabolismo , Proteínas Oncogênicas/metabolismo , Sequência de Aminoácidos , Animais , Asparagina/metabolismo , Células CHO , Linhagem Celular Tumoral , Cricetinae , Cricetulus , Regulação para Baixo/genética , Receptores ErbB/metabolismo , Galectina 3/genética , Glicosilação , Humanos , MicroRNAs/genética , Dados de Sequência Molecular , Mucina-1/química , Ligação Proteica , Estrutura Terciária de Proteína , Subunidades Proteicas/química , Estabilidade de RNA , RNA Mensageiro/metabolismo , Regulação para Cima/genética
12.
Int J Oncol ; 31(3): 671-7, 2007 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-17671696

RESUMO

The mucin (MUC) family consists of secreted and membrane-bound forms. The transmembrane mucin 1 (MUC1) is a heterodimer that is aberrantly overexpressed by diverse human carcinomas and certain hematologic malignancies. The MUC1 N-terminal (MUC1-N) and C-terminal (MUC1-C) subunits are generated by autocleavage within a SEA domain. The MUC1 cytoplasmic domain (MUC1-CD) located downstream of the SEA domain is sufficient for the induction of anchorage-independent growth and tumorigenicity; however, no information is available regarding the origin of these transforming sequences. Previous work demonstrated that, except for the SEA domain, MUC1 has no sequence homology with other membrane-bound mucins. The present results demonstrate that MUC1-CD evolved from repeat regions in the MUC5B secreted mucin. We also show that MUC1 sequences upstream to the SEA domain emerged from MUC5B. These findings indicate that both the MUC1-N and MUC1-C subunits evolved from secreted gel-forming mucins and that the MUC1-CD oncogenic function emerged by diversification after evolution from MUC5B.


Assuntos
Regulação Neoplásica da Expressão Gênica , Mucina-1/genética , Mucina-1/fisiologia , Sequência de Aminoácidos , Animais , Citoplasma/metabolismo , Dimerização , Evolução Molecular , Humanos , Modelos Genéticos , Dados de Sequência Molecular , Mucina-5B , Mucinas/genética , Fosforilação , Filogenia , Estrutura Terciária de Proteína , Proteínas Recombinantes de Fusão
13.
Hum Mutat ; 28(7): 683-93, 2007 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-17370310

RESUMO

The human genome contains frequent single-basepair variants that may or may not cause genetic disease. To characterize benign vs. pathogenic missense variants, numerous computational algorithms have been developed based on comparative sequence and/or protein structure analysis. We compared computational methods that use evolutionary conservation alone, amino acid (AA) change alone, and a combination of conservation and AA change in predicting the consequences of 254 missense variants in the CDKN2A (n = 92), MLH1 (n = 28), MSH2 (n = 14), MECP2 (n = 30), and tyrosinase (TYR) (n = 90) genes. Variants were validated as either neutral or deleterious by curated locus-specific mutation databases and published functional data. All methods that use evolutionary sequence analysis have comparable overall prediction accuracy (72.9-82.0%). Mutations at codons where the AA is absolutely conserved over a sufficient evolutionary distance (about one-third of variants) had a 91.6 to 96.8% likelihood of being deleterious. Three algorithms (SIFT, PolyPhen, and A-GVGD) that differentiate one variant from another at a given codon did not significantly improve predictive value over conservation score alone using the BLOSUM62 matrix. However, when all four methods were in agreement (62.7% of variants), predictive value improved to 88.1%. These results confirm a high predictive value for methods that use evolutionary sequence conservation, with or without considering protein structural change, to predict the clinical consequences of missense variants. The methods can be generalized across genes that cause different types of genetic disease. The results support the clinical use of computational methods as one tool to help interpret missense variants in genes associated with human genetic disease.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/genética , Genes p16 , Proteína 2 de Ligação a Metil-CpG/genética , Monofenol Mono-Oxigenase/genética , Proteína 2 Homóloga a MutS/genética , Mutação de Sentido Incorreto , Proteínas Nucleares/genética , Algoritmos , Evolução Molecular , Humanos , Proteína 1 Homóloga a MutL , Homologia de Sequência de Aminoácidos
14.
Cancer Res ; 66(23): 11187-93, 2006 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-17145863

RESUMO

Malignant melanoma is a common and frequently lethal disease. Current therapeutic interventions have little effect on survival, emphasizing the need for a better understanding of the genetic, epigenetic, and phenotypic changes in melanoma formation and progression. We identified 17 genes that were not previously known to be silenced by methylation in melanoma using a microarray-based screen following treatment of melanoma cell lines with the DNA methylation inhibitor 5-Aza-2'-deoxycytidine. Eight of these genes have not been previously shown to undergo DNA methylation in any form of cancer. Three of the genes, QPCT, CYP1B1, and LXN, are densely methylated in >95% of uncultured melanoma tumor samples. Reexpression of either of two of the silenced genes, HOXB13 and SYK, resulted in reduced colony formation in vitro and diminished tumor formation in vivo, indicating that these genes function as tumor suppressors in melanoma.


Assuntos
Epigênese Genética/genética , Inativação Gênica , Melanoma/patologia , Proteínas Supressoras de Tumor/genética , Animais , Azacitidina/análogos & derivados , Azacitidina/farmacologia , Western Blotting , Linhagem Celular Tumoral , Proliferação de Células , Células Cultivadas , Ilhas de CpG/genética , Metilação de DNA , Decitabina , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Predisposição Genética para Doença/genética , Proteínas de Homeodomínio/genética , Proteínas de Homeodomínio/metabolismo , Humanos , Peptídeos e Proteínas de Sinalização Intracelular/genética , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Masculino , Melanoma/genética , Melanoma/metabolismo , Melanoma Experimental/genética , Melanoma Experimental/patologia , Camundongos , Camundongos Nus , Proteínas Tirosina Quinases/genética , Proteínas Tirosina Quinases/metabolismo , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Quinase Syk , Transfecção , Transplante Heterólogo , Proteínas Supressoras de Tumor/metabolismo
15.
Gene ; 373: 28-34, 2006 May 24.
Artigo em Inglês | MEDLINE | ID: mdl-16500040

RESUMO

The MUC family of mucins consists of secreted and membrane-bound forms. Overexpression of the membrane-bound family members, MUC1 (CA15-3), MUC4 and MUC16 (CA125), is found in diverse human carcinomas. However, despite being classified in the same family, little is known about the genetic origins of the carcinoma-associated mucins. The present results show that MUC1 homologs are restricted to mammalian species. MUC1 has no sequence similarity with the other membrane-bound mucins, except for the presence of a sea urchin sperm protein-enterokinase-agrin (SEA) domain. The results indicate that the MUC1 SEA domain originated from heparin sulfate proteoglycan of basement membrane (HSPG2; perlecan), an inducer of tumor cell growth. MUC4 has no SEA domain, but does have (i) a NIDO domain that evolved from an ancestor common to nidogen, and (ii) AMOP and VWD domains that originated from an ancestor common to the Sushi-domain containing protein. MUC16 contains multiple SEA domains that are found in a chicken gene and were subsequently repeated through duplication events. The SEA domains in MUC16 appear to have evolved from agrin before the divergence of birds and mammals. These findings indicate that MUC1, MUC4 and MUC16 evolved from distinct ancestors and that the membrane-bound mucins consist of different subgroups based on their genetic backgrounds.


Assuntos
Antígenos de Neoplasias/genética , Antígeno Ca-125/genética , Carcinoma/genética , Evolução Molecular , Proteínas de Membrana/genética , Mucinas/genética , Animais , Humanos , Mucina-1 , Mucina-4 , Filogenia , Análise de Sequência de DNA
16.
Gastroenterology ; 129(2): 537-49, 2005 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-16083711

RESUMO

BACKGROUND & AIMS: Germline mutations in mismatch repair genes are associated with hereditary nonpolyposis colorectal cancer. A significant proportion of mutations are nontruncating and associated with a variability of clinical phenotype and microsatellite instability and with occasional presence of residual protein in tumor tissue that suggests impaired functional activity but not total lack of mismatch repair. To address pathogenic significance and mechanism of pathogenicity, we studied the functionality of 31 nontruncating MLH1 mutations found in clinically characterized colorectal cancer families and 3 other variations listed in a mutation database. METHODS: Mutations constructed by site-directed mutagenesis were studied for protein expression/stability, subcellular localization, protein-protein interaction, and repair efficiency. The genetic and biochemical data were correlated with clinical data. Finally, comparative sequence analysis was performed to assess the value of sequence homology as a tool for predicting functional results. RESULTS: Altogether, 22 mutations were pathogenic in more than one assay, 2 variants were impaired in one assay, and 10 variants acted like wild-type protein. Twenty of 34 mutations affected the quantity of MLH1 protein, whereas only 15 mainly amino-terminal mutations were defective in an in vitro repair assay. Comparative sequence analysis correctly predicted functional studies for 82% of variants. CONCLUSIONS: Pathogenic nontruncating alterations in MLH1 may interfere with different biochemical mechanisms but generally more than one. The severe biochemical defects are mirrored by phenotypic characteristics such as early age at onset and high microsatellite instability, whereas variants with no or mild defects in functionality are associated with variable clinical phenotypes.


Assuntos
Pareamento Incorreto de Bases/genética , Neoplasias Colorretais Hereditárias sem Polipose/genética , Predisposição Genética para Doença/epidemiologia , Mutação em Linhagem Germinativa , Proteínas de Neoplasias/genética , Proteínas Nucleares/genética , Proteínas Adaptadoras de Transdução de Sinal , Adulto , Distribuição por Idade , Idoso , Biomarcadores Tumorais/análise , Western Blotting , Proteínas de Transporte , Estudos de Coortes , Neoplasias Colorretais Hereditárias sem Polipose/epidemiologia , Feminino , Regulação Neoplásica da Expressão Gênica , Humanos , Incidência , Masculino , Pessoa de Meia-Idade , Proteína 1 Homóloga a MutL , Linhagem , Fenótipo , Probabilidade , Prognóstico , Medição de Risco , Estudos de Amostragem , Sensibilidade e Especificidade , Índice de Gravidade de Doença , Distribuição por Sexo , Análise de Sobrevida
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA