Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
1.
Histopathology ; 83(3): 426-434, 2023 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-37195579

RESUMO

INTRODUCTION: Adamantinoma-like Ewing sarcoma (ALES) is a rare aggressive malignancy occasionally diagnosed in the thyroid gland. ALES shows basaloid cytomorphology, expresses keratins, p63, p40, frequently CD99, and harbours the t(11;22) EWSR1::FLI1 translocation. There is debate on whether ALES resembles more sarcoma or carcinoma. METHODS: We performed RNA sequencing from two ALES cases and compared findings with skeletal Ewing's sarcomas and nonneoplastic thyroid tissue. ALES was investigated by in situ hybridization (ISH) for high-risk human papillomavirus (HPV) DNA and immunohistochemistry for the following antigens: keratin 7, keratin 20, keratin 5, keratins (AE1/AE3 and CAM5.2), CD45, CD20, CD5, CD99, chromogranin, synaptophysin, calcitonin, thyroglobulin, PAX8, TTF1, S100, p40, p63, p16, NUT, desmin, ER, FLI1, INI1, and myogenin. RESULTS: An uncommon EWSR1::FLI transcript with retained EWSR1 exon 8 was detected in both ALES cases. Regulators of EWSR1::FLI1 splicing (HNRNPH1, SUPT6H, SF3B1) necessary for production of a functional fusion oncoprotein, as well as 53 genes (including TNNT1, NKX2.2) activated downstream to the EWSR1::FLI1 cascade, were overexpressed. Eighty-six genes were uniquely overexpressed in ALES, most of which were related to squamous differentiation. Immunohistochemically, ALES strongly expressed keratins 5, AE1/AE3 and CAM5.2, p63, p40, p16, and focally CD99. INI1 was retained. The remaining immunostains and HPV DNA ISH were negative. CONCLUSION: Comparative transcriptomic profiling reveals overlapping features of ALES with skeletal Ewing's sarcoma and an epithelial carcinoma, as evidenced by immunohistochemical expression of keratin 5, p63, p40, CD99, the transcriptome profile, and detection of EWSR1::FLI1 fusion transcript by RNA sequencing.


Assuntos
Adamantinoma , Carcinoma , Infecções por Papillomavirus , Sarcoma de Ewing , Humanos , Sarcoma de Ewing/diagnóstico , Sarcoma de Ewing/genética , Adamantinoma/diagnóstico , Adamantinoma/genética , Adamantinoma/química , Glândula Tireoide/patologia , Transcriptoma , Queratina-5/metabolismo , Proteína EWS de Ligação a RNA/genética , Proteína EWS de Ligação a RNA/metabolismo , Fatores de Transcrição/genética , Proteínas de Fusão Oncogênica/genética , Proteínas de Fusão Oncogênica/metabolismo
2.
Mod Pathol ; 33(10): 1889-1895, 2020 10.
Artigo em Inglês | MEDLINE | ID: mdl-32415263

RESUMO

Epithelial marker expression and/or epithelial differentiation, as well as "anomalous" expression of keratins, are features of some soft tissue tumors. Recently, we have encountered an unusual mesenchymal tumor composed of bland, distinctly eosinophilic, keratin-positive epithelial cells, which were almost entirely obscured by xanthogranulomatous inflammation. Six cases were identified (5 F, 1 M; 16-62 years (median 21 years)) arising in soft tissue (n = 4) and bone (n = 2) and ranging in size from 2 to 7 cm. The tumors were generally circumscribed, with a fibrous capsule containing lymphoid aggregates, and consisted in large part of a sheet-like proliferation of foamy histiocytes, Touton-type and osteoclast-type giant cells, and chronic inflammatory cells. Closer inspection, however, disclosed a distinct population of uniform, cytologically bland mononuclear cells with brightly eosinophilic cytoplasm arranged singly and in small nests and cords. Overt squamous and/or glandular differentiation was absent. By immunohistochemistry, these cells were diffusely positive with the OSCAR and AE1/AE3 keratin antibodies, and focally positive for high-molecular weight keratins; endothelial and myoid markers were negative and SMARCB1 was retained. RNA-seq identified a PLEKHM1 variant of undetermined significance in one case, likely related to this patient's underlying osteopetrosis. Follow-up to date has been benign. In summary, we have identified a novel tumor of soft tissue and bone with a predilection for young females, provisionally termed "xanthogranulomatous epithelial tumor". These unusual lesions do not appear to arise from adnexa, or represent known keratin-positive soft tissue tumors, and the origin of their constituent epithelial cells is obscure. The natural history of this distinctive lesion appears indolent, although study of additional cases and longer term follow-up are needed.


Assuntos
Neoplasias Ósseas/patologia , Neoplasias de Tecido Conjuntivo e de Tecidos Moles/patologia , Neoplasias de Tecidos Moles/patologia , Adolescente , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Adulto Jovem
3.
PLoS Comput Biol ; 11(12): e1004465, 2015 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-26699225

RESUMO

High-throughput mRNA sequencing (RNA-Seq) is widely used for transcript quantification of gene isoforms. Since RNA-Seq data alone is often not sufficient to accurately identify the read origins from the isoforms for quantification, we propose to explore protein domain-domain interactions as prior knowledge for integrative analysis with RNA-Seq data. We introduce a Network-based method for RNA-Seq-based Transcript Quantification (Net-RSTQ) to integrate protein domain-domain interaction network with short read alignments for transcript abundance estimation. Based on our observation that the abundances of the neighboring isoforms by domain-domain interactions in the network are positively correlated, Net-RSTQ models the expression of the neighboring transcripts as Dirichlet priors on the likelihood of the observed read alignments against the transcripts in one gene. The transcript abundances of all the genes are then jointly estimated with alternating optimization of multiple EM problems. In simulation Net-RSTQ effectively improved isoform transcript quantifications when isoform co-expressions correlate with their interactions. qRT-PCR results on 25 multi-isoform genes in a stem cell line, an ovarian cancer cell line, and a breast cancer cell line also showed that Net-RSTQ estimated more consistent isoform proportions with RNA-Seq data. In the experiments on the RNA-Seq data in The Cancer Genome Atlas (TCGA), the transcript abundances estimated by Net-RSTQ are more informative for patient sample classification of ovarian cancer, breast cancer and lung cancer. All experimental results collectively support that Net-RSTQ is a promising approach for isoform quantification. Net-RSTQ toolbox is available at http://compbio.cs.umn.edu/Net-RSTQ/.


Assuntos
Algoritmos , Sequenciamento de Nucleotídeos em Larga Escala/métodos , Proteínas de Neoplasias/genética , RNA Neoplásico/genética , Software , Transcriptoma/genética , Sequência de Bases , Humanos , Dados de Sequência Molecular , Neoplasias , Mapeamento de Interação de Proteínas/métodos , Isoformas de Proteínas/genética , Transdução de Sinais/genética
4.
J Natl Cancer Inst ; 113(9): 1246-1257, 2021 09 04.
Artigo em Inglês | MEDLINE | ID: mdl-33555293

RESUMO

BACKGROUND: TP53 mutations occur in more than 50% of cancers. We sought to determine the effect of the intragenic P72R single nucleotide polymorphism (SNP; rs1042522) on the oncogenic properties of mutant p53. METHODS: P72R allelic selection in tumors was determined from genotype calls and a Gaussian distributed mixture model. The SNP effect on mutant p53 was determined in p53-negative cancer cell lines. RNA-sequencing, chromatin immunoprecipitation, and survival analysis were performed to describe the SNP effect. All statistical tests were 2-sided. RESULTS: Among 409 patients with germline heterozygous P72R SNP who harbored somatic mutations in TP53, we observed a selection bias against missense TP53 mutants encoding the P72 SNP (P = 1.64 x 10-13). Exogenously expressed hotspot p53 mutants with the P72 SNP were negatively selected in cancer cells. Gene expression analyses showed the enrichment of p53 pathway genes and inflammatory genes in cancer cells transduced with mutants encoding P72 SNP. Immune gene signature is enriched in patients harboring missense TP53 mutations with homozygous P72 SNP. These patients have improved overall survival as compared with those with the R72 SNP (P = .04). CONCLUSION: This is the largest study demonstrating a selection against the P72 SNP. Missense p53 mutants with the P72 SNP retain partial wild-type tumor-suppressive functions, which may explain the selection bias against P72 SNP across cancer types. Ovarian cancer patients with the P72 SNP have a better prognosis than with the R72 SNP. Our study describes a previously unknown role through which the rs1042522 SNP modifies tumor suppressor activities of mutant p53 in patients.


Assuntos
Neoplasias Ovarianas , Proteína Supressora de Tumor p53 , Alelos , Feminino , Genes p53 , Humanos , Neoplasias Ovarianas/genética , Polimorfismo de Nucleotídeo Único , Proteína Supressora de Tumor p53/genética , Proteína Supressora de Tumor p53/metabolismo
5.
J Neuropathol Exp Neurol ; 80(9): 821-829, 2021 09 27.
Artigo em Inglês | MEDLINE | ID: mdl-34363682

RESUMO

Polymorphous low-grade neuroepithelial tumor of the young (PLNTY) is a recently described epileptogenic tumor characterized by oligodendroglioma-like components, aberrant CD34 expression, and frequent mitogen-activated protein kinase (MAPK) pathway activation. We molecularly profiled 13 cases with diagnostic histopathological features of PLNTY (10 female; median age, 16 years; range, 5-52). Patients frequently presented with seizures (9 of 12 with available history) and temporal lobe tumors (9 of 13). MAPK pathway activating alterations were identified in all 13 cases. Fusions were present in the 7 youngest patients: FGFR2-CTNNA3 (n = 2), FGFR2-KIAA1598 (FGFR2-SHTN1) (n = 1), FGFR2-INA (n = 1), FGFR2-MPRIP (n = 1), QKI-NTRK2 (n = 1), and KIAA1549-BRAF (n = 1). BRAF V600E mutation was present in 6 patients (17 years or older). Two fusion-positive cases additionally harbored TP53/RB1 abnormalities suggesting biallelic inactivation. Copy number changes predominantly involving whole chromosomes were observed in all 10 evaluated cases, with losses of chromosome 10q occurring with FGFR2-KIAA1598 (SHTN1)/CTNNA3 fusions. The KIAA1549-BRAF and QKI-NTRK2 fusions were associated respectively with a 7q34 deletion and 9q21 duplication. This study shows that despite its name, PLNTY also occurs in older adults, who frequently show BRAF V600E mutation. It also expands the spectrum of the MAPK pathway activating alterations associated with PLNTY and demonstrates recurrent chromosomal copy number changes consistent with chromosomal instability.


Assuntos
Glicoproteínas de Membrana/metabolismo , Quinases de Proteína Quinase Ativadas por Mitógeno/metabolismo , Neoplasias Neuroepiteliomatosas/metabolismo , Receptor trkB/metabolismo , Convulsões/metabolismo , Adulto , Aneuploidia , Cromossomos Humanos Par 9/metabolismo , Feminino , Fusão Gênica/fisiologia , Humanos , Recidiva , Convulsões/genética , Fatores de Transcrição/metabolismo
6.
J Mol Diagn ; 23(5): 555-564, 2021 05.
Artigo em Inglês | MEDLINE | ID: mdl-33549857

RESUMO

Tumor mutation burden (TMB) is an emerging biomarker of immunotherapy response. RNA sequencing in FFPE tissue samples was used for determining TMB in microsatellite-stable (MSS) and microsatellite instability-high (MSI-H) tumors in patients with colorectal or endometrial cancer. Tissue from tumors and paired normal tissue from 46 MSI-H and 12 MSS cases were included. Of the MSI-H tumors, 29 had defective DNA mismatch-repair mutations, and 17 had MLH1 promoter hypermethylation. TMB was measured using the expressed somatic nucleotide variants (eTMB). A method of accurate measurement of eTMB was developed that removes FFPE-derived artifacts by leveraging mutation signatures. There was a significant difference in the median eTMB values observed between MSI-H and MSS cases: 27.3 versus 6.7 mutations/megabase (mut/Mb) (P = 3.5 × 10-9). Among tumors with defective DNA-mismatch repair, those with mismatch-repair mutations had a significantly higher median eTMB than those with hypermethylation: 28.1 versus 17.5 mut/Mb (P = 0.037). Multivariate analysis showed that MSI status, tumor type (endometrial or colorectal), and age were significantly associated with eTMB. Additionally, using whole-exome sequencing in a subset of these patients, it was determined that DNA TMB correlated well with eTMB (Spearman correlation coefficient, 0.83). These results demonstrate that RNA sequencing can be used for measuring eTMB in FFPE tumor specimens.


Assuntos
Biomarcadores Tumorais/genética , Neoplasias Colorretais/patologia , Reparo de Erro de Pareamento de DNA/genética , Neoplasias do Endométrio/patologia , Mutação , RNA-Seq/métodos , Adulto , Idoso , Idoso de 80 Anos ou mais , Neoplasias Colorretais/genética , Neoplasias do Endométrio/genética , Feminino , Seguimentos , Humanos , Masculino , Pessoa de Meia-Idade , Prognóstico , Estudos Retrospectivos
7.
J Cell Biol ; 165(4): 459-64, 2004 May 24.
Artigo em Inglês | MEDLINE | ID: mdl-15159415

RESUMO

53BP1 participates early in the DNA damage response and is involved in cell cycle checkpoint control. Moreover, the phenotype of mice and cells deficient in 53BP1 suggests a defect in DNA repair (Ward et al., 2003b). Therefore, we asked whether or not 53BP1 would be required for the efficient repair of DNA double strand breaks. Our data indicate that homologous recombination by gene conversion does not depend on 53BP1. Moreover, 53BP1-deficient mice support normal V(D)J recombination, indicating that 53BP1 is not required for "classic" nonhomologous end joining. However, class switch recombination is severely impaired in the absence of 53BP1, suggesting that 53BP1 facilitates DNA end joining in a way that is not required or redundant for the efficient closing of RAG-induced strand breaks. These findings are similar to those observed in mice or cells deficient in the tumor suppressors ATM and H2AX, further suggesting that the functions of ATM, H2AX, and 53BP1 are closely linked.


Assuntos
Proteínas de Transporte/metabolismo , Dano ao DNA/genética , Reparo do DNA/genética , DNA/metabolismo , Peptídeos e Proteínas de Sinalização Intracelular , Fosfoproteínas , Recombinação Genética/genética , Animais , Proteínas Mutadas de Ataxia Telangiectasia , Proteínas de Transporte/genética , Proteínas de Ciclo Celular , Linhagem Celular , DNA/genética , Proteínas de Ligação a DNA , Regulação da Expressão Gênica/genética , Rearranjo Gênico/genética , Histonas/deficiência , Histonas/genética , Camundongos , Proteínas Serina-Treonina Quinases/deficiência , Proteínas Serina-Treonina Quinases/genética , Homologia de Sequência do Ácido Nucleico , Proteínas Supressoras de Tumor
8.
Commun Biol ; 2: 335, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31508509

RESUMO

Treatment of cancer with poly (ADP-ribose) polymerase (PARP) inhibitors is currently limited to cells defective in the homologous recombination (HR) pathway. Identification of genetic targets that induce or mimic HR deficiencies will extend the clinical utility of PARP inhibitors. Here we perform a CRISPR/Cas9-based genome-scale loss-of-function screen, using the sensitivity of PARP inhibitor olaparib as a surrogate. We identify C12orf5, encoding TP53 induced glycolysis and apoptosis regulator (TIGAR), as a modifier of PARP inhibitor response. We show that TIGAR is amplified in several cancer types, and higher expression of TIGAR associates with poor overall survival in ovarian cancer. TIGAR knockdown enhances sensitivity to olaparib in cancer cells via downregulation of BRCA1 and the Fanconi anemia pathway and increases senescence of these cells by affecting metabolic pathways and increasing the cytotoxic effects of olaparib. Our results indicate TIGAR should be explored as a therapeutic target for treating cancer and extending the use of PARP inhibitors.


Assuntos
Proteínas Reguladoras de Apoptose/genética , Sistemas CRISPR-Cas , Resistencia a Medicamentos Antineoplásicos/genética , Monoéster Fosfórico Hidrolases/genética , Inibidores de Poli(ADP-Ribose) Polimerases/farmacologia , Antineoplásicos/farmacologia , Biomarcadores , Senescência Celular , Dano ao DNA , Técnicas de Silenciamento de Genes , Estudo de Associação Genômica Ampla , Humanos , Ftalazinas/farmacologia , Piperazinas/farmacologia , Transdução de Sinais
9.
Sci Rep ; 9(1): 11002, 2019 07 29.
Artigo em Inglês | MEDLINE | ID: mdl-31358864

RESUMO

In recent years, multiple studies including ours have reported on the mechanism of resistance towards valosin-containing protein (VCP) inhibitors. While all these studies reported target alterations via mutations in VCP as the primary mechanism of resistance, discrepancies persist to date regarding the zygosity of these mutations responsible for the resistance. In addition, the extent to which resistant cells harbor additional mutations in other genes is not well described. In this study, we performed global transcript analysis of the parental and previously reported VCP inhibitor (CB-5083) resistant cells and found additional mutations in the resistant cells. However, our CRISPR-Cas9 gene editing studies indicate that specific mutations in VCP are sufficient to produce resistance to CB-5083 suggesting the importance of on-target mutations in VCP for resistance. Strikingly, our analysis indicates a preexisting heterozygous frameshift mutation at codon 616 (N616fs*) in one of the VCP alleles in HCT116 cells, and we showed that this mutant allele is subjected to the nonsense-mediated decay (NMD). Accordingly, we identified a heterozygous mutation at codon 526 (L526S) in genomic DNA sequencing but a homozygous L526S mutation in complementary DNA sequencing in our independently generated CB-5083 resistant HCT116 cells, implying that the L526S mutation occurs in the allele that does not harbor the frameshift N616fs* mutation. Our results suggest the NMD as a possible mechanism for achieving the homozygosity of VCP mutant responsible for the resistance to VCP inhibitors while resolving the discrepancies among previous studies. Our results also underscore the importance of performing simultaneous genomic and complementary DNA sequencing when attributing mutational effects on the functionality particularly for an oligomer protein like VCP.


Assuntos
Inibidores Enzimáticos/farmacologia , Indóis/farmacologia , Pirimidinas/farmacologia , Proteína com Valosina/antagonistas & inibidores , Proteína com Valosina/genética , Linhagem Celular , Resistência a Medicamentos/genética , Mutação da Fase de Leitura , Células HCT116 , Células HEK293 , Heterozigoto , Humanos , Mutação , Proteína com Valosina/metabolismo
10.
Mol Cell Biol ; 25(22): 10079-86, 2005 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-16260621

RESUMO

p53 binding protein 1 (53BP1) is a putative DNA damage sensor that accumulates at sites of double-strand breaks (DSBs) in a manner dependent on histone H2AX. Here we show that the loss of one or both copies of 53BP1 greatly accelerates lymphomagenesis in a p53-null background, suggesting that 53BP1 and p53 cooperate in tumor suppression. A subset of 53BP1-/- p53-/- lymphomas, like those in H2AX-/- p53-/- mice, were diploid and harbored clonal translocations involving antigen receptor loci, indicating misrepair of DSBs during V(D)J recombination as one cause of oncogenic transformation. Loss of a single 53BP1 allele compromised genomic stability and DSB repair, which could explain the susceptibility of 53BP1+/- mice to tumorigenesis. In addition to structural aberrations, there were high rates of chromosomal missegregation and accumulation of aneuploid cells in 53BP1-/- p53+/+ and 53BP1-/- p53-/- tumors as well as in primary 53BP1-/- splenocytes. We conclude that 53BP1 functions as a dosage-dependent caretaker that promotes genomic stability by a mechanism that preserves chromosome structure and number.


Assuntos
Peptídeos e Proteínas de Sinalização Intracelular/fisiologia , Neoplasias/genética , Fosfoproteínas/fisiologia , Proteína Supressora de Tumor p53/fisiologia , Alelos , Motivos de Aminoácidos , Animais , Sítios de Ligação , Western Blotting , Células Cultivadas , Centrossomo/ultraestrutura , Aberrações Cromossômicas , Cromossomos/ultraestrutura , Cruzamentos Genéticos , Reparo do DNA , Feminino , Genes Supressores de Tumor , Predisposição Genética para Doença , Hibridização in Situ Fluorescente , Cariotipagem , Linfoma/metabolismo , Masculino , Camundongos , Camundongos Transgênicos , Microscopia de Fluorescência , Neoplasias/patologia , Receptores de Antígenos/metabolismo , Recombinação Genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Baço/citologia , Fatores de Tempo , Translocação Genética , Proteína Supressora de Tumor p53/metabolismo , Proteína 1 de Ligação à Proteína Supressora de Tumor p53
11.
Mol Cell Biol ; 23(7): 2556-63, 2003 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-12640136

RESUMO

53BP1 is a p53 binding protein of unknown function that binds to the central DNA-binding domain of p53. It relocates to the sites of DNA strand breaks in response to DNA damage and is a putative substrate of the ataxia telangiectasia-mutated (ATM) kinase. To study the biological role of 53BP1, we disrupted the 53BP1 gene in the mouse. We show that, similar to ATM(-/-) mice, 53BP1-deficient mice were growth retarded, immune deficient, radiation sensitive, and cancer prone. 53BP1(-/-) cells show a slight S-phase checkpoint defect and prolonged G(2)/M arrest after treatment with ionizing radiation. Moreover, 53BP1(-/-) cells feature a defective DNA damage response with impaired Chk2 activation. These data indicate that 53BP1 acts downstream of ATM and upstream of Chk2 in the DNA damage response pathway and is involved in tumor suppression.


Assuntos
Proteínas de Transporte/metabolismo , Dano ao DNA , Transtornos do Crescimento/genética , Peptídeos e Proteínas de Sinalização Intracelular , Linfoma/genética , Síndromes Neoplásicas Hereditárias/genética , Fosfoproteínas , Proteínas Supressoras de Tumor/metabolismo , Animais , Proteínas de Transporte/genética , Ciclo Celular/fisiologia , Proteínas Cromossômicas não Histona , Dano ao DNA/fisiologia , Proteínas de Ligação a DNA , Marcação de Genes , Transtornos do Crescimento/complicações , Linfoma/complicações , Linfoma/patologia , Camundongos , Camundongos Knockout , Síndromes Neoplásicas Hereditárias/complicações , Fenótipo , Ligação Proteica/fisiologia , Tolerância a Radiação/genética , Proteína 1 de Ligação à Proteína Supressora de Tumor p53
13.
Sci Rep ; 5: 12335, 2015 Jul 23.
Artigo em Inglês | MEDLINE | ID: mdl-26202458

RESUMO

Current genomic studies are limited by the availability of fresh tissue samples. Here, we show that Illumina RNA sequencing of formalin-fixed diagnostic tumor samples produces gene expression that is strongly correlated with matched frozen tumor samples (r > 0.89). In addition, sequence variations identified from FFPE RNA show 99.67% concordance with that from exome sequencing of matched frozen tumor samples. Because FFPE is a routine diagnostic sample preparation, the feasibility results reported here will facilitate the setup of large-scale research and clinical studies in medical genomics that are currently limited by the availability of fresh frozen samples.


Assuntos
Análise Mutacional de DNA/métodos , Perfilação da Expressão Gênica/métodos , Genes Neoplásicos/genética , Neoplasias Ovarianas/genética , RNA Neoplásico/genética , Análise de Sequência de RNA/métodos , Sequência de Bases , Feminino , Fixadores/química , Formaldeído/química , Predisposição Genética para Doença/genética , Humanos , Dados de Sequência Molecular , Mutação/genética , Neoplasias Ovarianas/diagnóstico , Reprodutibilidade dos Testes , Sensibilidade e Especificidade , Preservação de Tecido/métodos
14.
Oncotarget ; 5(22): 11365-80, 2014 Nov 30.
Artigo em Inglês | MEDLINE | ID: mdl-25426548

RESUMO

FoxM1 is an oncogenic Forkhead transcription factor that is overexpressed in ovarian cancer. However, the mechanisms by which FoxM1 is deregulated in ovarian cancer and the extent to which FoxM1 can be targeted in ovarian cancer have not been reported previously. In this study, we showed that MDM2 inhibitor Nutlin-3 upregulated p53 protein and downregulated FoxM1 expression in several cancer cell lines with wild type TP53 but not in cell lines with mutant TP53. FoxM1 downregulation was partially blocked by cycloheximide or actinomycin D, and pulse-chase studies indicate Nutlin-3 enhances FoxM1 mRNA decay. Knockdown of p53 using shRNAs abrogated the FoxM1 downregulation by Nutlin-3, indicating a p53-dependent mechanism. FoxM1 inhibitor, thiostrepton, induces apoptosis in cancer cell lines and enhances sensitivity to cisplatin in these cells. Thiostrepton downregulates FoxM1 expression in several cancer cell lines and enhances sensitivity to carboplatin in vivo. Finally, FoxM1 expression is elevated in nearly all (48/49) ovarian tumors, indicating that thiostrepton target gene is highly expressed in ovarian cancer. In summary, the present study provides novel evidence that both amorphic and neomorphic mutations in TP53 contribute to FoxM1 overexpression and that FoxM1 may be targeted for therapeutic benefits in cancers.


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica/farmacologia , Carboplatina/farmacologia , Fatores de Transcrição Forkhead/biossíntese , Neoplasias Ovarianas/tratamento farmacológico , Tioestreptona/farmacologia , Proteína Supressora de Tumor p53/genética , Animais , Apoptose/efeitos dos fármacos , Carboplatina/administração & dosagem , Linhagem Celular Tumoral , Sinergismo Farmacológico , Feminino , Proteína Forkhead Box M1 , Células HEK293 , Humanos , Imidazóis/farmacologia , Camundongos , Camundongos Nus , Terapia de Alvo Molecular , Mutação , Neoplasias Ovarianas/genética , Neoplasias Ovarianas/metabolismo , Piperazinas/farmacologia , Tioestreptona/administração & dosagem , Ensaios Antitumorais Modelo de Xenoenxerto
15.
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
16.
Cell ; 131(5): 901-14, 2007 Nov 30.
Artigo em Inglês | MEDLINE | ID: mdl-18001825

RESUMO

DNA-damage signaling utilizes a multitude of posttranslational modifiers as molecular switches to regulate cell-cycle checkpoints, DNA repair, cellular senescence, and apoptosis. Here we show that RNF8, a FHA/RING domain-containing protein, plays a critical role in the early DNA-damage response. We have solved the X-ray crystal structure of the FHA domain structure at 1.35 A. We have shown that RNF8 facilitates the accumulation of checkpoint mediator proteins BRCA1 and 53BP1 to the damaged chromatin, on one hand through the phospho-dependent FHA domain-mediated binding of RNF8 to MDC1, on the other hand via its role in ubiquitylating H2AX and possibly other substrates at damage sites. Moreover, RNF8-depleted cells displayed a defective G2/M checkpoint and increased IR sensitivity. Together, our study implicates RNF8 as a novel DNA-damage-responsive protein that integrates protein phosphorylation and ubiquitylation signaling and plays a critical role in the cellular response to genotoxic stress.


Assuntos
Proteínas de Ciclo Celular/metabolismo , Dano ao DNA/fisiologia , Proteínas de Ligação a DNA/fisiologia , Histonas/metabolismo , Ubiquitinação , Proteínas Adaptadoras de Transdução de Sinal , Motivos de Aminoácidos , Proteína BRCA1/metabolismo , Sítios de Ligação , Ciclo Celular/efeitos da radiação , Células Cultivadas , Proteínas de Ligação a DNA/química , Proteínas de Ligação a DNA/metabolismo , Histonas/fisiologia , Humanos , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Modelos Biológicos , Modelos Moleculares , Proteínas Nucleares/fisiologia , Fosforilação , Ligação Proteica , Estrutura Terciária de Proteína , Transdução de Sinais , Transativadores/fisiologia , Proteína 1 de Ligação à Proteína Supressora de Tumor p53 , Ubiquitina-Proteína Ligases , Ubiquitinação/fisiologia
17.
J Biol Chem ; 281(50): 38472-7, 2006 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-17043355

RESUMO

53BP1 plays an important role in cellular response to DNA damage. It is thought to be the mammalian homologue of budding yeast Rad9 and/or fission yeast Crb2. Rad9/Crb2 are bona fide checkpoint proteins whose activation requires their corresponding C-terminal tandem BRCT (BRCA1 C-terminal) motifs, which mediate their oligomerization and phosphorylation at multiple sites following DNA damage. Here we show that the function of human 53BP1 similarly depends on its oligomerization and phosphorylation at multiple sites but in a BRCT domain-independent manner. Moreover, unlike its proposed yeast counterparts, human 53BP1 only has limited checkpoint functions but rather acts as an adaptor in the repair of DNA double strand breaks. This difference in function may reflect the higher complexity of the DNA damage response network in metazoa including the evolution of other BRCT domain-containing proteins that may have functions redundant or overlapping with those of 53BP1.


Assuntos
Reparo do DNA/fisiologia , Peptídeos e Proteínas de Sinalização Intracelular/fisiologia , Fosfoproteínas/fisiologia , Linhagem Celular , Humanos , Imunoprecipitação , Peptídeos e Proteínas de Sinalização Intracelular/química , Peptídeos e Proteínas de Sinalização Intracelular/genética , Mutagênese Sítio-Dirigida , Fosfoproteínas/química , Fosfoproteínas/genética , Proteína 1 de Ligação à Proteína Supressora de Tumor p53
18.
J Biol Chem ; 279(11): 9677-80, 2004 Mar 12.
Artigo em Inglês | MEDLINE | ID: mdl-14742437

RESUMO

Ataxia-telangiectasia-mutated and Rad3-related (ATR) plays an essential role in the maintenance of genome integrity and cell viability. The kinase is activated in response to DNA damage and initiates a checkpoint signaling cascade by phosphorylating a number of downstream substrates including Chk1. Unlike ataxia-telangiectasia-mutated (ATM), which appears to be mainly activated by DNA double-strand breaks, ATR can be activated by a variety of DNA damaging agents. However, it is still unclear what triggers ATR activation in response to such diverse DNA lesions. One model proposes that ATR can directly recognize DNA lesions, while other recent data suggest that ATR is activated by a common single-stranded DNA (ssDNA) intermediate generated during DNA repair. In this study, we show that UV lesions do not directly activate ATR in vivo. In addition, ssDNA lesions created during the repair of UV damage are also not sufficient to activate the ATR-dependent pathway. ATR activation is only observed in replicating cells indicating that replication stress is required to trigger the ATR-mediated checkpoint cascade in response to UV irradiation. Interestingly, H2AX appears to be required for the accumulation of ATR at stalled replication forks. Together our data suggest that ssDNA at arrested replication forks recruits ATR and initiates ATR-mediated phosphorylation of H2AX and Chk1. Phosphorylated H2AX might further facilitate ATR activation by stabilizing ATR at the sites of arrested replication forks.


Assuntos
Raios Ultravioleta , Proteínas de Ciclo Celular , Linhagem Celular Tumoral , Separação Celular , Sobrevivência Celular , Quinase 1 do Ponto de Checagem , Dano ao DNA , Reparo do DNA , Replicação do DNA , DNA de Cadeia Simples , Citometria de Fluxo , Células HeLa , Humanos , Immunoblotting , Microscopia de Fluorescência , Fosforilação , Proteínas Quinases/metabolismo , Transdução de Sinais , Estresse Fisiológico , Fatores de Tempo
19.
J Biol Chem ; 278(22): 19579-82, 2003 May 30.
Artigo em Inglês | MEDLINE | ID: mdl-12697768

RESUMO

53BP1 participates in the cellular response to DNA damage. Like many proteins involved in the DNA damage response, 53BP1 becomes hyperphosphorylated after radiation and colocalizes with phosphorylated H2AX in megabase regions surrounding the sites of DNA strand breaks. However, it is not yet clear whether the phosphorylation status of 53BP1 determines its localization or vice versa. In this study we mapped a region upstream of the 53BP1 C terminus that is required and sufficient for the recruitment of 53BP1 to these DNA break areas. In vitro assays revealed that this region binds to phosphorylated but not unphosphorylated H2AX. Moreover, using H2AX-deficient cells reconstituted with wild-type or a phosphorylation-deficient mutant of H2AX, we have shown that phosphorylation of H2AX at serine 140 is critical for efficient 53BP1 foci formation, implying that a direct interaction between 53BP1 and phosphorylated H2AX is required for the accumulation of 53BP1 at DNA break sites. On the other hand, radiation-induced phosphorylation of the 53BP1 N terminus by the ATM (ataxia-telangiectasia mutated) kinase is not essential for 53BP1 foci formation and takes place independently of 53BP1 redistribution. Thus, these two damage-induced events, hyperphosphorylation and relocalization of 53BP1, occur independently in the cell.


Assuntos
Proteínas de Transporte/metabolismo , Dano ao DNA , DNA/metabolismo , Histonas/metabolismo , Peptídeos e Proteínas de Sinalização Intracelular , Fosfoproteínas , Proteínas de Transporte/genética , Linhagem Celular , Mutagênese Sítio-Dirigida , Fosforilação , Ligação Proteica
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa