Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 101
1.
Nucleic Acids Res ; 50(12): e70, 2022 07 08.
Article En | MEDLINE | ID: mdl-35412634

Discovering rare cancer driver genes is difficult because their mutational frequency is too low for statistical detection by computational methods. EPIMUTESTR is an integrative nearest-neighbor machine learning algorithm that identifies such marginal genes by modeling the fitness of their mutations with the phylogenetic Evolutionary Action (EA) score. Over cohorts of sequenced patients from The Cancer Genome Atlas representing 33 tumor types, EPIMUTESTR detected 214 previously inferred cancer driver genes and 137 new candidates never identified computationally before of which seven genes are supported in the COSMIC Cancer Gene Census. EPIMUTESTR achieved better robustness and specificity than existing methods in a number of benchmark methods and datasets.


Machine Learning , Neoplasms , Humans , Mutation , Neoplasms/genetics , Neoplasms/pathology , Oncogenes , Phylogeny
2.
Genome Res ; 32(5): 916-929, 2022 05.
Article En | MEDLINE | ID: mdl-35301263

Genetic variants drive the evolution of traits and diseases. We previously modeled these variants as small displacements in fitness landscapes and estimated their functional impact by differentiating the evolutionary relationship between genotype and phenotype. Conversely, here we integrate these derivatives to identify genes steering specific traits. Over cancer cohorts, integration identified 460 likely tumor-driving genes. Many have literature and experimental support but had eluded prior genomic searches for positive selection in tumors. Beyond providing cancer insights, these results introduce a general calculus of evolution to quantify the genotype-phenotype relationship and discover genes associated with complex traits and diseases.


Calculi , Neoplasms , Biological Evolution , Genetic Fitness , Genotype , Humans , Models, Genetic , Neoplasms/genetics , Phenotype , Selection, Genetic
3.
Dis Model Mech ; 15(2)2022 02 01.
Article En | MEDLINE | ID: mdl-35174853

Rhabdomyosarcoma (RMS) is the most common soft tissue sarcoma in children, with overall long-term survival rates of ∼65-70%. Thus, additional molecular insights and representative models are critical for identifying and evaluating new treatment modalities. Using MyoD-Cre-mediated introduction of mutant K-RasG12D and perturbations in p53, we developed a novel genetically engineered mouse model (GEMM) for RMS. The anatomic sites of primary RMS development recapitulated human disease, including tumors in the head, neck, extremities and abdomen. We confirmed RMS histology and diagnosis through Hematoxylin and Eosin staining, and positive immunohistochemical staining for desmin, myogenin, and phosphotungstic acid-Hematoxylin. Cell lines from GEMM tumors were established with the ability to engraft in immunocompetent mice with comparable histological and staining features as the primary tumors. Tail vein injection of cell lines had high metastatic potential to the lungs. Transcriptomic analyses of p53R172H/K-RasG12D GEMM-derived tumors showed evidence of high molecular homology with human RMS. Finally, pre-clinical use of these murine RMS lines showed similar therapeutic responsiveness to chemotherapy and targeted therapies as human RMS cell lines.


Rhabdomyosarcoma , Sarcoma , Soft Tissue Neoplasms , Animals , Disease Models, Animal , Humans , Mice , Rhabdomyosarcoma/diagnosis , Rhabdomyosarcoma/drug therapy , Rhabdomyosarcoma/genetics , Sarcoma/metabolism , Soft Tissue Neoplasms/metabolism , Tumor Suppressor Protein p53/genetics
4.
Nat Commun ; 12(1): 937, 2021 02 10.
Article En | MEDLINE | ID: mdl-33568653

The global impact of somatic structural variants (SSVs) on gene expression in pediatric brain tumors has not been thoroughly characterised. Here, using whole-genome and RNA sequencing from 854 tumors of more than 30 different types from the Children's Brain Tumor Tissue Consortium, we report the altered expression of hundreds of genes in association with the presence of nearby SSV breakpoints. SSV-mediated expression changes involve gene fusions, altered cis-regulation, or gene disruption. SSVs considerably extend the numbers of patients with tumors somatically altered for critical pathways, including receptor tyrosine kinases (KRAS, MET, EGFR, NF1), Rb pathway (CDK4), TERT, MYC family (MYC, MYCN, MYB), and HIPPO (NF2). Compared to initial tumors, progressive or recurrent tumors involve a distinct set of SSV-gene associations. High overall SSV burden associates with TP53 mutations, histone H3.3 gene H3F3C mutations, and the transcription of DNA damage response genes. Compared to adult cancers, pediatric brain tumors would involve a different set of genes with SSV-altered cis-regulation. Our comprehensive and pan-histology genomic analyses reveal SSVs to play a major role in shaping the transcriptome of pediatric brain tumors.


Brain Neoplasms/genetics , Gene Rearrangement , Cohort Studies , Gene Expression Regulation, Neoplastic , Genomics , Humans , Mutation , Pediatrics/statistics & numerical data , Sequence Analysis, RNA , Exome Sequencing
5.
Cell Cycle ; 19(4): 479-491, 2020 02.
Article En | MEDLINE | ID: mdl-31959038

The phosphoinositide-3-kinase like kinases (PIKK) such as ATM and ATR play a key role in initiating the cellular DNA damage response (DDR). One key ATM target is the cyclin-dependent kinase inhibitor p27Kip1 that promotes G1 arrest. ATM activates p27Kip1-induced arrest in part through phosphorylation of p27Kip1 at Serine 140. Here we show that this site is dephosphorylated by the type 2C serine/threonine phosphatase, WIP1 (Wildtype p53-Induced Phosphatase-1), encoded by the PPM1D gene. WIP1 has been shown to dephosphorylate numerous ATM target sites in DDR proteins, and its overexpression and/or mutation has often been associated with oncogenesis. We demonstrate that wildtype, but not phosphatase-dead WIP1, efficiently dephosphorylates p27Kip1 Ser140 both in vitro and in cells and that this dephosphorylation is sensitive to the WIP1-specific inhibitor GSK 2830371. Increased expression of wildtype WIP1 reduces stability of p27Kip1 while increased expression of similar amounts of phosphatase-dead WIP1 has no effect on p27Kip1 protein stability. Overexpression of wildtype p27Kip1 reduces cell proliferation and colony forming capability relative to the S140A (constitutively non-phosphorylated) form of p27. Thus, WIP1 plays a significant role in homeostatic modulation of p27Kip1 activity following activation by ATM.


Ataxia Telangiectasia Mutated Proteins/metabolism , Cyclin-Dependent Kinase Inhibitor p27/metabolism , Protein Phosphatase 2C/metabolism , Serine/metabolism , Cell Proliferation , Cyclin-Dependent Kinase Inhibitor p27/genetics , HEK293 Cells , Humans , MCF-7 Cells , Mutation/genetics , Phosphopeptides/metabolism , Phosphorylation , Protein Stability , Reproducibility of Results , Tumor Stem Cell Assay
7.
Cell Rep ; 28(5): 1370-1384.e5, 2019 07 30.
Article En | MEDLINE | ID: mdl-31365877

The TP53 tumor suppressor gene is frequently mutated in human cancers. An analysis of five data platforms in 10,225 patient samples from 32 cancers reported by The Cancer Genome Atlas (TCGA) enables comprehensive assessment of p53 pathway involvement in these cancers. More than 91% of TP53-mutant cancers exhibit second allele loss by mutation, chromosomal deletion, or copy-neutral loss of heterozygosity. TP53 mutations are associated with enhanced chromosomal instability, including increased amplification of oncogenes and deep deletion of tumor suppressor genes. Tumors with TP53 mutations differ from their non-mutated counterparts in RNA, miRNA, and protein expression patterns, with mutant TP53 tumors displaying enhanced expression of cell cycle progression genes and proteins. A mutant TP53 RNA expression signature shows significant correlation with reduced survival in 11 cancer types. Thus, TP53 mutation has profound effects on tumor cell genomic structure, expression, and clinical outlook.


Databases, Nucleic Acid , Gene Expression Regulation, Neoplastic , Loss of Heterozygosity , Neoplasms/genetics , Tumor Suppressor Protein p53/genetics , Humans , MicroRNAs/genetics , MicroRNAs/metabolism , Neoplasms/metabolism , Neoplasms/pathology , RNA, Neoplasm/genetics , RNA, Neoplasm/metabolism , Tumor Suppressor Protein p53/metabolism
8.
Cell Stem Cell ; 23(5): 700-713.e6, 2018 11 01.
Article En | MEDLINE | ID: mdl-30388424

Clonal hematopoiesis (CH), in which stem cell clones dominate blood production, becomes increasingly common with age and can presage malignancy development. The conditions that promote ascendancy of particular clones are unclear. We found that mutations in PPM1D (protein phosphatase Mn2+/Mg2+-dependent 1D), a DNA damage response regulator that is frequently mutated in CH, were present in one-fifth of patients with therapy-related acute myeloid leukemia or myelodysplastic syndrome and strongly correlated with cisplatin exposure. Cell lines with hyperactive PPM1D mutations expand to outcompete normal cells after exposure to cytotoxic DNA damaging agents including cisplatin, and this effect was predominantly mediated by increased resistance to apoptosis. Moreover, heterozygous mutant Ppm1d hematopoietic cells outcompeted their wild-type counterparts in vivo after exposure to cisplatin and doxorubicin, but not during recovery from bone marrow transplantation. These findings establish the clinical relevance of PPM1D mutations in CH and the importance of studying mutation-treatment interactions. VIDEO ABSTRACT.


Antineoplastic Agents/pharmacology , Cisplatin/pharmacology , Clone Cells/drug effects , Doxorubicin/pharmacology , Hematopoiesis/drug effects , Leukemia, Myeloid, Acute/drug therapy , Mutation , Protein Phosphatase 2C/genetics , Aged , Animals , Antineoplastic Agents/chemistry , Cell Proliferation/drug effects , Cisplatin/chemistry , Doxorubicin/chemistry , Drug Screening Assays, Antitumor , Female , HEK293 Cells , Hematopoiesis/genetics , Humans , Leukemia, Myeloid, Acute/metabolism , Leukemia, Myeloid, Acute/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Inbred NOD , Middle Aged , Neoplasms, Experimental/drug therapy , Neoplasms, Experimental/metabolism , Neoplasms, Experimental/pathology , Protein Phosphatase 2C/metabolism
9.
Am J Cancer Res ; 8(9): 1752-1763, 2018.
Article En | MEDLINE | ID: mdl-30323968

Osteosarcoma (OS) is a highly aggressive mesenchymal malignancy and the most common primary bone tumor in the pediatric population. OS frequently presents with or develops distal metastases. Patients with metastatic disease have extremely poor survival rates, thus necessitating improved molecular insights into OS metastatic biology. Utilizing our previously characterized genetically engineered mouse model (GEMM) of metastatic OS, we identified enhanced differential expression of Transglutaminase-2 (TGM2) in metastatic OS. However, the role of TGM2 in sarcoma development and metastatic progression remains largely undefined. To further investigate the role of TGM2 in OS metastasis, we performed both gain- and loss-of-function studies for TGM2 in human and mouse OS cell lines. Our data provide evidence that enhanced expression of TGM2 in metastatic OS contributes to migratory and invasive phenotypes. Besides the effects on metastatic phenotypes, we also observed that TGM2 contributes to OS stem-like properties. In addition, treatment with transglutaminase inhibitors had analogous effects on proliferation and migration to TGM2 knockdown. Finally, in vivo xenograft studies demonstrated that TGM2 functionally alters metastatic potential and survival outcome. Together, these data highlight TGM2 as a pro-metastatic factor in OS and a potential avenue for future therapeutic intervention to inhibit metastatic disease.

10.
Proc Natl Acad Sci U S A ; 115(42): 10666-10671, 2018 10 16.
Article En | MEDLINE | ID: mdl-30266789

Scientific progress depends on formulating testable hypotheses informed by the literature. In many domains, however, this model is strained because the number of research papers exceeds human readability. Here, we developed computational assistance to analyze the biomedical literature by reading PubMed abstracts to suggest new hypotheses. The approach was tested experimentally on the tumor suppressor p53 by ranking its most likely kinases, based on all available abstracts. Many of the best-ranked kinases were found to bind and phosphorylate p53 (P value = 0.005), suggesting six likely p53 kinases so far. One of these, NEK2, was studied in detail. A known mitosis promoter, NEK2 was shown to phosphorylate p53 at Ser315 in vitro and in vivo and to functionally inhibit p53. These bona fide validations of text-based predictions of p53 phosphorylation, and the discovery of an inhibitory p53 kinase of pharmaceutical interest, suggest that automated reasoning using a large body of literature can generate valuable molecular hypotheses and has the potential to accelerate scientific discovery.


Abstracting and Indexing , NIMA-Related Kinases/metabolism , Tumor Suppressor Protein p53/antagonists & inhibitors , Tumor Suppressor Protein p53/metabolism , HCT116 Cells , HEK293 Cells , Humans , NIMA-Related Kinases/genetics , Natural Language Processing , Phosphorylation , PubMed , Tumor Suppressor Protein p53/genetics
11.
Cell Rep ; 23(1): 239-254.e6, 2018 04 03.
Article En | MEDLINE | ID: mdl-29617664

DNA damage repair (DDR) pathways modulate cancer risk, progression, and therapeutic response. We systematically analyzed somatic alterations to provide a comprehensive view of DDR deficiency across 33 cancer types. Mutations with accompanying loss of heterozygosity were observed in over 1/3 of DDR genes, including TP53 and BRCA1/2. Other prevalent alterations included epigenetic silencing of the direct repair genes EXO5, MGMT, and ALKBH3 in ∼20% of samples. Homologous recombination deficiency (HRD) was present at varying frequency in many cancer types, most notably ovarian cancer. However, in contrast to ovarian cancer, HRD was associated with worse outcomes in several other cancers. Protein structure-based analyses allowed us to predict functional consequences of rare, recurrent DDR mutations. A new machine-learning-based classifier developed from gene expression data allowed us to identify alterations that phenocopy deleterious TP53 mutations. These frequent DDR gene alterations in many human cancers have functional consequences that may determine cancer progression and guide therapy.


Genome, Human , Neoplasms/genetics , Recombinational DNA Repair , Cell Line, Tumor , DNA Damage , Gene Silencing , Humans , Loss of Heterozygosity , Machine Learning , Mutation , Neoplasms/classification , Tumor Suppressor Proteins/genetics , Tumor Suppressor Proteins/metabolism
12.
Aging Cell ; 17(1)2018 02.
Article En | MEDLINE | ID: mdl-29047229

Aging is often accompanied by a dramatic increase in cancer susceptibility. To gain insights into how aging affects tumor susceptibility, we generated a conditional mouse model in which oncogenic KrasG12D was activated specifically in lungs of young (3-5 months) and old (19-24 months) mice. Activation of KrasG12D in old mice resulted in shorter survival and development of higher-grade lung tumors. Six weeks after KrasG12D activation, old lung tissues contained higher numbers of adenomas than their young tissue counterparts. Lung tumors in old mice displayed higher proliferation rates, as well as attenuated DNA damage and p53 tumor suppressor responses. Gene expression comparison of lung tumors from young and old mice revealed upregulation of extracellular matrix-related genes in young tumors, indicative of a robust cancer-associated fibroblast response. In old tumors, numerous inflammation-related genes such as Ccl7, IL-1ß, Cxcr6, and IL-15ra were consistently upregulated. Increased numbers of immune cells were localized around the periphery of lung adenomas from old mice. Our experiments indicate that more aggressive lung tumor formation in older KrasG12D mice may be in part the result of subdued tumor suppressor and DNA damage responses, an enhanced inflammatory milieu, and a more accommodating tissue microenvironment.


Aging/physiology , Cell Proliferation/genetics , Cell Transformation, Neoplastic/genetics , Inflammation/genetics , Lung Neoplasms/genetics , Animals , Disease Models, Animal , Genes, ras/genetics , Mice , Mutation/genetics , Signal Transduction/genetics
13.
Int J Cancer ; 141(10): 2062-2075, 2017 11 15.
Article En | MEDLINE | ID: mdl-28748534

Ewing Sarcoma (ES) is a highly aggressive bone tumor with peak incidence in the adolescent population. It has a high propensity to metastasize, which is associated with dismal survival rates of approximately 25%. To further understand mechanisms of metastasis we investigated microRNA regulatory networks in ES. Our studies focused on miR-130b due to our analysis that enhanced expression of this microRNA has clinical relevance in multiple sarcomas, including ES. Our studies provide insights into a novel positive feedback network involving the direct regulation of miR-130b and activation of downstream signaling events contributing toward sarcoma metastasis. Specifically, we demonstrated miR-130b induces proliferation, invasion, and migration in vitro and increased metastatic potential in vivo. Using microarray analysis of ES cells with differential miR-130b expression we identified alterations in downstream signaling cascades including activation of the CDC42 pathway. We identified ARHGAP1, which is a negative regulator of CDC42, as a novel, direct target of miR-130b. In turn, downstream activation of PAK1 activated the JNK and AP-1 cascades and downstream transcriptional targets including IL-8, MMP1 and CCND1. Furthermore, chromatin immunoprecipitation of endogenous AP-1 in ES cells demonstrated direct binding to an upstream consensus binding site within the miR-130b promoter. Finally, small molecule inhibition of PAK1 blocked miR-130b activation of JNK and downstream AP-1 target genes, including primary miR-130b transcripts, and miR-130b oncogenic properties, thus identifying PAK1 as a novel therapeutic target for ES. Taken together, our findings identify and characterize a novel, targetable miR-130b regulatory network that promotes ES metastasis.


Bone Neoplasms/pathology , GTPase-Activating Proteins/metabolism , Gene Expression Regulation, Neoplastic , Liver Neoplasms/secondary , Lung Neoplasms/secondary , MicroRNAs/genetics , Sarcoma, Ewing/pathology , Animals , Apoptosis , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Bone Neoplasms/genetics , Bone Neoplasms/metabolism , Cell Movement , Cell Proliferation , Epithelial-Mesenchymal Transition , Feedback, Physiological , GTPase-Activating Proteins/genetics , Humans , Liver Neoplasms/genetics , Liver Neoplasms/metabolism , Lung Neoplasms/genetics , Lung Neoplasms/metabolism , Mice , Mice, Inbred NOD , Mice, SCID , Neoplasm Staging , Prognosis , Sarcoma, Ewing/genetics , Sarcoma, Ewing/metabolism , Signal Transduction , Transcription Factor AP-1/genetics , Transcription Factor AP-1/metabolism , Tumor Cells, Cultured , Xenograft Model Antitumor Assays , cdc42 GTP-Binding Protein/genetics , cdc42 GTP-Binding Protein/metabolism , p21-Activated Kinases/genetics , p21-Activated Kinases/metabolism
14.
Cancer Res ; 77(6): 1250-1260, 2017 03 15.
Article En | MEDLINE | ID: mdl-28254861

Accurate assessment of TP53 gene status in sporadic tumors and in the germline of individuals at high risk of cancer due to Li-Fraumeni Syndrome (LFS) has important clinical implications for diagnosis, surveillance, and therapy. Genomic data from more than 20,000 cancer genomes provide a wealth of information on cancer gene alterations and have confirmed TP53 as the most commonly mutated gene in human cancer. Analysis of a database of 70,000 TP53 variants reveals that the two newly discovered exons of the gene, exons 9ß and 9γ, generated by alternative splicing, are the targets of inactivating mutation events in breast, liver, and head and neck tumors. Furthermore, germline rearrange-ments in intron 1 of TP53 are associated with LFS and are frequently observed in sporadic osteosarcoma. In this context of constantly growing genomic data, we discuss how screening strategies must be improved when assessing TP53 status in clinical samples. Finally, we discuss how TP53 alterations should be described by using accurate nomenclature to avoid confusion in scientific and clinical reports. Cancer Res; 77(6); 1250-60. ©2017 AACR.


Genetic Variation/genetics , Neoplasms/genetics , Practice Guidelines as Topic/standards , Quality Control , Tumor Suppressor Protein p53/genetics , Humans , Neoplasms/diagnosis , Neoplasms/therapy , Validation Studies as Topic
15.
Cell Cycle ; 16(2): 213-223, 2017 Jan 17.
Article En | MEDLINE | ID: mdl-28027003

The phosphatase Wip1 attenuates the DNA damage response (DDR) by removing phosphorylation marks from a number of DDR proteins (p53, MDM2, Chk1/2, p38). Wip1 also dephosphorylates and inactivates RelA. Notably, LZAP, a putative tumor suppressor, has been linked to dephosphorylation of several of these substrates, including RelA, p38, Chk1, and Chk2. LZAP has no known catalytic activity or functional motifs, suggesting that it exerts its effects through interaction with other proteins. Here we show that LZAP binds Wip1 and stimulates its phosphatase activity. LZAP had been previously shown to bind many Wip1 substrates (RelA, p38, Chk1/2), and our results show that LZAP also binds the previously identified Wip1 substrate, MDM2. This work identifies 2 novel Wip1 substrates, ERK1 and HuR, and demonstrates that HuR is a binding partner of LZAP. Pleasingly, LZAP potentiated Wip1 catalytic activity toward each substrate tested, regardless of whether full-length substrates or phosphopeptides were utilized. Since this effect was observed on ERK1, which does not bind LZAP, as well as for each of 7 peptides tested, we hypothesize that LZAP binding to the substrate is not required for this effect and that LZAP directly binds Wip1 to augment its phosphatase activity.


Nerve Tissue Proteins/metabolism , Phosphoprotein Phosphatases/metabolism , Biocatalysis , Cell Line, Tumor , HEK293 Cells , Humans , Phosphopeptides/metabolism , Phosphorylation , Protein Binding , Substrate Specificity
16.
BMC Cancer ; 16(1): 869, 2016 11 08.
Article En | MEDLINE | ID: mdl-27821163

BACKGROUND: Osteosarcoma (OS), which has a high potential for developing metastatic disease, is the most frequent malignant bone tumor in children and adolescents. Molecular analysis of a metastatic genetically engineered mouse model of osteosarcoma identified enhanced expression of Secreted Frizzled-Related Protein 2 (sFRP2), a putative regulator of Wnt signaling within metastatic tumors. Subsequent analysis correlated increased expression in the human disease, and within highly metastatic OS cells. However, the role of sFRP2 in osteosarcoma development and progression has not been well elucidated. METHODS: Studies using stable gain or loss-of-function alterations of sFRP2 within human and mouse OS cells were performed to assess changes in cell proliferation, migration, and invasive ability in vitro, via both transwell and 3D matrigel assays. In additional, xenograft studies using overexpression of sFRP2 were used to assess effects on in vivo metastatic potential. RESULTS: Functional studies revealed stable overexpression of sFRP2 within localized human and mouse OS cells significantly increased cell migration and invasive ability in vitro and enhanced metastatic potential in vivo. Additional studies exploiting knockdown of sFRP2 within metastatic human and mouse OS cells demonstrated decreased cell migration and invasion ability in vitro, thus corroborating a critical biological phenotype carried out by sFRP2. Interestingly, alterations in sFRP2 expression did not alter OS proliferation rates or primary tumor development. CONCLUSIONS: While future studies further investigating the molecular mechanisms contributing towards this sFRP2-dependent phenotype are needed, our studies clearly provide evidence that aberrant expression of sFRP2 can contribute to the invasive and metastatic potential for osteosarcoma.


Bone Neoplasms/metabolism , Bone Neoplasms/pathology , Membrane Proteins/metabolism , Osteosarcoma/metabolism , Osteosarcoma/pathology , Animals , Bone Neoplasms/genetics , Cell Line, Tumor , Cell Movement , Cell Proliferation , Disease Models, Animal , Female , Gene Expression , Gene Knockdown Techniques , Humans , Membrane Proteins/genetics , Mice , Neoplasm Invasiveness , Neoplasm Metastasis , Osteosarcoma/genetics
17.
Cell Rep ; 14(4): 907-919, 2016 Feb 02.
Article En | MEDLINE | ID: mdl-26804919

The ampulla of Vater is a complex cellular environment from which adenocarcinomas arise to form a group of histopathologically heterogenous tumors. To evaluate the molecular features of these tumors, 98 ampullary adenocarcinomas were evaluated and compared to 44 distal bile duct and 18 duodenal adenocarcinomas. Genomic analyses revealed mutations in the WNT signaling pathway among half of the patients and in all three adenocarcinomas irrespective of their origin and histological morphology. These tumors were characterized by a high frequency of inactivating mutations of ELF3, a high rate of microsatellite instability, and common focal deletions and amplifications, suggesting common attributes in the molecular pathogenesis are at play in these tumors. The high frequency of WNT pathway activating mutation, coupled with small-molecule inhibitors of ß-catenin in clinical trials, suggests future treatment decisions for these patients may be guided by genomic analysis.


Adenocarcinoma/genetics , DNA-Binding Proteins/genetics , Duodenal Neoplasms/genetics , Mutation , Pancreatic Neoplasms/genetics , Proto-Oncogene Proteins c-ets/genetics , Transcription Factors/genetics , Wnt Signaling Pathway , Adenocarcinoma/metabolism , Ampulla of Vater/pathology , Base Sequence , Duodenal Neoplasms/metabolism , Genomic Instability , Humans , Microsatellite Repeats , Molecular Sequence Data , Pancreatic Neoplasms/metabolism
18.
Br J Cancer ; 113(9): 1289-97, 2015 Nov 03.
Article En | MEDLINE | ID: mdl-26528706

BACKGROUND: Osteosarcoma (OS) is the most common bone malignancy in the paediatric population, principally affecting adolescents and young adults. Minimal advancements in patient prognosis have been made over the past two decades because of the poor understanding of disease biology. Runx2, a critical transcription factor in bone development, is frequently amplified and overexpressed in OS. However, the molecular and biological consequences of Runx2 overexpression remain unclear. METHODS: si/shRNA and overexpression technology to alter Runx2 levels in OS cells. In vitro assessment of doxorubicin (doxo)-induced apoptosis and in vivo chemosensitivity studies. Small-molecule inhibitor of c-Myc transcriptional activity was used to assess its role. RESULTS: Loss of Runx2 sensitises cells to doxo-induced apoptosis both in vitro and in vivo. Furthermore, in conjunction with chemotherapy, decreasing Runx2 protein levels activates both the intrinsic and extrinsic apoptotic pathways. Transplanted tumour studies demonstrated that loss of endogenous Runx2 protein expression enhances caspase-3 cleavage and tumour necrosis in response to chemotherapy. Finally, upon doxo-treated Runx2 knockdown OS cells there was evidence of enhanced c-Myc expression and transcriptional activity. Inhibition of c-Myc under these conditions resulted in decreased activation of apoptosis, therefore insinuating a role for c-Myc in dox-induced activation of apoptotic pathways. CONCLUSIONS: Therefore, we have established a novel molecular mechanism by which Runx2 provides a chemoprotective role in OS, indicating that in conjunction to standard chemotherapy, targeting Runx2 may be a new therapeutic strategy for patients with OS.


Apoptosis/genetics , Bone Neoplasms/genetics , Core Binding Factor Alpha 1 Subunit/genetics , Osteosarcoma/genetics , Animals , Apoptosis/drug effects , Bone Neoplasms/drug therapy , Caspase 3/genetics , Cell Line, Tumor , Doxorubicin/pharmacology , Female , Gene Expression Regulation, Neoplastic/drug effects , Gene Expression Regulation, Neoplastic/genetics , Humans , Mice , Mice, Nude , Osteosarcoma/drug therapy , Proto-Oncogene Proteins c-myc/genetics , RNA, Small Interfering/genetics , Transcriptional Activation/drug effects , Transcriptional Activation/genetics
19.
Nat Genet ; 47(12): 1426-34, 2015 Dec.
Article En | MEDLINE | ID: mdl-26551670

Sézary syndrome is a rare leukemic form of cutaneous T cell lymphoma characterized by generalized redness, scaling, itching and increased numbers of circulating atypical T lymphocytes. It is rarely curable, with poor prognosis. Here we present a multiplatform genomic analysis of 37 patients with Sézary syndrome that implicates dysregulation of cell cycle checkpoint and T cell signaling. Frequent somatic alterations were identified in TP53, CARD11, CCR4, PLCG1, CDKN2A, ARID1A, RPS6KA1 and ZEB1. Activating CCR4 and CARD11 mutations were detected in nearly one-third of patients. ZEB1, encoding a transcription repressor essential for T cell differentiation, was deleted in over one-half of patients. IL32 and IL2RG were overexpressed in nearly all cases. Our results demonstrate profound disruption of key signaling pathways in Sézary syndrome and suggest potential targets for new therapies.


Cell Differentiation/genetics , Gene Regulatory Networks , Mutation/genetics , Sezary Syndrome/genetics , Skin Neoplasms/genetics , T-Lymphocytes/metabolism , Case-Control Studies , Exome/genetics , Gene Expression Regulation, Neoplastic , Genetic Drift , Genomics/methods , High-Throughput Nucleotide Sequencing/methods , Humans , Lymphocyte Activation , Prognosis , Sezary Syndrome/mortality , Sezary Syndrome/pathology , Signal Transduction , Skin Neoplasms/mortality , Skin Neoplasms/pathology , Survival Rate , T-Lymphocytes/cytology
20.
Genes Chromosomes Cancer ; 54(12): 796-808, 2015 Dec.
Article En | MEDLINE | ID: mdl-26355645

Osteosarcomas (OSs) are characterized by high levels of genomic instability (GI). To gain insights into the GI and its contribution toward understanding the genetic basis of OS, we characterized 19 primary and 13 metastatic mouse tumors in a genetically engineered novel mouse model of OS by a combination of genomic techniques. Through the bone-specific deletion of the wild-type Trp53 locus or activation of a metastatic-promoting missense R172Hp53 allele, C57BL/6 mice developed either localized or metastatic OS. Subsequent tumors were isolated and primary cultures created from primary bone and/or distal metastatic lesions, for example, lung and liver. These tumors exhibited high levels of GI with complex chromosomal rearrangements, amplifications, and deletions comparable to human OS. The combined genomic approaches identified frequent amplification of chromosome 15D1 and loss of 11B4 by CGH and/or SKY. Both 15D1 and 11B4 have homology with frequently altered chromosomal bands 8q24 and 17p13 in human OS, respectively. Subsequent array CGH, FISH, and qRT-PCR analysis identified coamplification and overexpression of Myc/Pvt1 transcripts from the 15D1 amplicon and loss and decreased expression of the Nlrp1b from 11B4. The Nlrp1 gene is the key mediator of apoptosis and interacts strongly with caspase 2.


Apoptosis Regulatory Proteins/genetics , Bone Neoplasms/genetics , Osteosarcoma/genetics , Proto-Oncogene Proteins c-myc/genetics , RNA, Long Noncoding/genetics , Sarcoma, Experimental/genetics , Tumor Suppressor Protein p53/genetics , Animals , Apoptosis , Apoptosis Regulatory Proteins/metabolism , Bone Neoplasms/pathology , Caspase 2/metabolism , Chromosome Deletion , Gene Amplification , Genetic Loci , Genomic Instability , Homozygote , In Situ Hybridization, Fluorescence , Karyotyping , Mice , Mice, Inbred C57BL , Mice, Transgenic , Mutation, Missense , Neoplasm Metastasis , Oligonucleotide Array Sequence Analysis , Osteoblasts/metabolism , Osteosarcoma/pathology , Primary Cell Culture , Sarcoma, Experimental/pathology , Up-Regulation
...