Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 37
Filter
1.
Cell Genom ; 3(10): 100402, 2023 Oct 11.
Article in English | MEDLINE | ID: mdl-37868040

ABSTRACT

Neuroblastoma is a pediatric solid tumor characterized by strong clinical heterogeneity. Although clinical risk-defining genomic alterations exist in neuroblastomas, the mutational processes involved in their generation remain largely unclear. By examining the topography and mutational signatures derived from all variant classes, we identified co-occurring mutational footprints, which we termed mutational scenarios. We demonstrate that clinical neuroblastoma heterogeneity is associated with differences in the mutational processes driving these scenarios, linking risk-defining pathognomonic variants to distinct molecular processes. Whereas high-risk MYCN-amplified neuroblastomas were characterized by signs of replication slippage and stress, homologous recombination-associated signatures defined high-risk non-MYCN-amplified patients. Non-high-risk neuroblastomas were marked by footprints of chromosome mis-segregation and TOP1 mutational activity. Furthermore, analysis of subclonal mutations uncovered differential activity of these processes through neuroblastoma evolution. Thus, clinical heterogeneity of neuroblastoma patients can be linked to differences in the mutational processes that are active in their tumors.

3.
Nat Neurosci ; 23(7): 842-853, 2020 07.
Article in English | MEDLINE | ID: mdl-32424282

ABSTRACT

Many immunotherapies act by enhancing the ability of cytotoxic T cells to kill tumor cells. Killing depends on T cell recognition of antigens presented by class I major histocompatibility complex (MHC-I) proteins on tumor cells. In this study, we showed that medulloblastomas lacking the p53 tumor suppressor do not express surface MHC-I and are therefore resistant to immune rejection. Mechanistically, this is because p53 regulates expression of the peptide transporter Tap1 and the aminopeptidase Erap1, which are required for MHC-I trafficking to the cell surface. In vitro, tumor necrosis factor (TNF) or lymphotoxin-ß receptor agonist can rescue expression of Erap1, Tap1 and MHC-I on p53-mutant tumor cells. In vivo, low doses of TNF prolong survival and synergize with immune checkpoint inhibitors to promote tumor rejection. These studies identified p53 as a key regulator of immune evasion and suggest that TNF could be used to enhance sensitivity of tumors to immunotherapy.


Subject(s)
Cerebellar Neoplasms/immunology , Medulloblastoma/immunology , Tumor Escape/immunology , Tumor Necrosis Factor-alpha/immunology , Tumor Suppressor Protein p53/immunology , Animals , Cerebellar Neoplasms/genetics , Cerebellar Neoplasms/metabolism , Medulloblastoma/genetics , Medulloblastoma/metabolism , Mice , Mice, Inbred C57BL , Mice, Inbred NOD , Mice, SCID , Neoplasm Transplantation , Tumor Necrosis Factor-alpha/metabolism , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism
5.
J Gerontol A Biol Sci Med Sci ; 75(6): 1068-1072, 2020 05 22.
Article in English | MEDLINE | ID: mdl-31504207

ABSTRACT

The genetic basis of healthy aging and longevity remains largely unexplained. One hypothesis as to why long-lived individuals do not appear to have a lower number of common-complex disease variants, is that despite carrying risk variants, they express disease-linked alleles at a lower level than the wild-type alleles. Allele-specific abundance (ASA) is the different transcript abundance of the two haplotypes of a diploid individual. We sequenced the transcriptomes of four healthy centenarians and four mid-life controls. CIBERSORT was used to estimate blood cell fractions: neutrophils were the most abundant source of RNA, followed by CD8+ T cells, resting NK cells, and monocytes. ASA variants were more common in noncoding than coding regions. Centenarians and controls had a comparable distribution of ASA variants by predicted effect, and we did not observe an overall bias in expression toward major or minor alleles. Immune pathways were most highly represented among the gene set that showed ASA. Although we found evidence of ASA in disease-associated genes and transcription factors, we did not observe any differences in the pattern of expression between centenarians and controls in this small pilot study.


Subject(s)
Aged, 80 and over , Alleles , Healthy Aging/genetics , Transcriptome/genetics , Diploidy , Female , Gene Expression Profiling , Genes/genetics , Genetic Predisposition to Disease/genetics , Genetic Variation/genetics , Haplotypes/genetics , Humans , Male , Pilot Projects
6.
Nat Genet ; 52(1): 29-34, 2020 01.
Article in English | MEDLINE | ID: mdl-31844324

ABSTRACT

Extrachromosomal circularization of DNA is an important genomic feature in cancer. However, the structure, composition and genome-wide frequency of extrachromosomal circular DNA have not yet been profiled extensively. Here, we combine genomic and transcriptomic approaches to describe the landscape of extrachromosomal circular DNA in neuroblastoma, a tumor arising in childhood from primitive cells of the sympathetic nervous system. Our analysis identifies and characterizes a wide catalog of somatically acquired and undescribed extrachromosomal circular DNAs. Moreover, we find that extrachromosomal circular DNAs are an unanticipated major source of somatic rearrangements, contributing to oncogenic remodeling through chimeric circularization and reintegration of circular DNA into the linear genome. Cancer-causing lesions can emerge out of circle-derived rearrangements and are associated with adverse clinical outcome. It is highly probable that circle-derived rearrangements represent an ongoing mutagenic process. Thus, extrachromosomal circular DNAs represent a multihit mutagenic process, with important functional and clinical implications for the origins of genomic remodeling in cancer.


Subject(s)
Carcinogenesis/pathology , DNA, Circular/genetics , Extrachromosomal Inheritance/genetics , Gene Rearrangement , Genome, Human , Neuroblastoma/pathology , Oncogenes/genetics , Recombination, Genetic , Humans , Neuroblastoma/genetics , Tumor Cells, Cultured
7.
Nat Commun ; 9(1): 4439, 2018 10 25.
Article in English | MEDLINE | ID: mdl-30361512

ABSTRACT

We conducted an RNA sequencing study to identify novel gene fusions in 80 discovery dataset tumors collected from young patients with diffuse gastric cancer (DGC). Twenty-five in-frame fusions are associated with DGC, three of which (CLDN18-ARHGAP26, CTNND1-ARHGAP26, and ANXA2-MYO9A) are recurrent in 384 DGCs based on RT-PCR. All three fusions contain a RhoGAP domain in their 3' partner genes. Patients with one of these three fusions have a significantly worse prognosis than those without. Ectopic expression of CLDN18-ARHGAP26 promotes the migration and invasion capacities of DGC cells. Parallel targeted RNA sequencing analysis additionally identifies TACC2-PPAPDC1A as a recurrent and poor prognostic in-frame fusion. Overall, PPAPDC1A fusions and in-frame fusions containing a RhoGAP domain clearly define the aggressive subset (7.5%) of DGCs, and their prognostic impact is greater than, and independent of, chromosomal instability and CDH1 mutations. Our study may provide novel genomic insights guiding future strategies for managing DGCs.


Subject(s)
GTPase-Activating Proteins/chemistry , Oncogene Proteins, Fusion/genetics , Phosphatidate Phosphatase/genetics , Stomach Neoplasms/pathology , Adult , Base Sequence , Cell Aggregation , Cell Line, Tumor , Cell Proliferation , Female , Humans , Male , Middle Aged , Prognosis , Protein Domains , Sequence Analysis, RNA , Young Adult
8.
NPJ Precis Oncol ; 2(1): 8, 2018.
Article in English | MEDLINE | ID: mdl-29872726

ABSTRACT

Eccrine porocarcinomas (EPs) are rare malignant tumours of the intraepidermic sweat gland duct and most often arise from benign eccrine poromas. Some recurrent somatic genomic events have been identified in these malignancies, but very little is known about the complexity of their molecular pathophysiology. We describe the whole genome and whole transcriptome genomic profiling of a metastatic EP in a 66-year-old male patient with a previous history of localized porocarcinoma of the scalp. Whole genome and whole transcriptome genomic profiling was performed on the metastatic EP. Whole genome sequencing was performed on blood-derived DNA in order to allow a comparison between germline and somatic events. We found somatic copy losses of several tumour suppressor genes including APC, PTEN and CDKN2A, CDKN2B and CDKN1A. We identified a somatic hemizygous CDKN2A pathogenic splice site variant. De novo transcriptome assembly revealed abnormal splicing of CDKN2A p14ARF and p16INK4a. Elevated expression of oncogenes EGFR and NOTCH1 was noted and no somatic mutations were found in these genes. Wnt pathway somatic alterations were also observed. In conclusion, our results suggest that the molecular pathophysiology of malignant EP features high complexity and subtle interactions of multiple key genes. Cell cycle dysregulation and CDKN2A loss of function was found to be a new potential driver in EP tumourigenesis. Moreover, the combination of somatic copy number variants and abnormal gene expression perhaps partly related to epigenetic mechanisms, all likely contribute to the development of this rare malignancy in our patient.

9.
Clin Cancer Res ; 23(24): 7521-7530, 2017 Dec 15.
Article in English | MEDLINE | ID: mdl-29246904

ABSTRACT

Purpose: Recent studies have identified mutation signatures of homologous recombination deficiency (HRD) in over 20% of breast cancers, as well as pancreatic, ovarian, and gastric cancers. There is an urgent need to understand the clinical implications of HRD signatures. Whereas BRCA1/2 mutations confer sensitivity to platinum-based chemotherapies, it is not yet clear whether mutation signatures can independently predict platinum response.Experimental Design: In this observational study, we sequenced tumor whole genomes (100× depth) and matched normals (60×) of 93 advanced-stage breast cancers (33 platinum-treated). We computed a published metric called HRDetect, independently trained to predict BRCA1/2 status, and assessed its capacity to predict outcomes on platinum-based chemotherapies. Clinical endpoints were overall survival (OS), total duration on platinum-based therapy (TDT), and radiographic evidence of clinical improvement (CI).Results: HRDetect predicted BRCA1/2 status with an area under the curve (AUC) of 0.94 and optimal threshold of 0.7. Elevated HRDetect was also significantly associated with CI on platinum-based therapy (AUC = 0.89; P = 0.006) with the same optimal threshold, even after adjusting for BRCA1/2 mutation status and treatment timing. HRDetect scores over 0.7 were associated with a 3-month extended median TDT (P = 0.0003) and 1.3-year extended median OS (P = 0.04).Conclusions: Our findings not only independently validate HRDetect, but also provide the first evidence of its association with platinum response in advanced breast cancer. We demonstrate that HRD mutation signatures may offer clinically relevant information independently of BRCA1/2 mutation status and hope this work will guide the development of clinical trials. Clin Cancer Res; 23(24); 7521-30. ©2017 AACR.


Subject(s)
BRCA1 Protein/genetics , BRCA2 Protein/genetics , Homologous Recombination/genetics , Triple Negative Breast Neoplasms/genetics , Disease-Free Survival , Female , Homologous Recombination/drug effects , Humans , Middle Aged , Mutation , Neoplasm Staging , Platinum/administration & dosage , Treatment Outcome , Triple Negative Breast Neoplasms/drug therapy , Triple Negative Breast Neoplasms/pathology , Whole Genome Sequencing
10.
Gastroenterology ; 153(2): 536-549.e26, 2017 08.
Article in English | MEDLINE | ID: mdl-28522256

ABSTRACT

BACKGROUND & AIMS: Early-onset gastric cancer, which develops in patients younger than most gastric cancers, is usually detected at advanced stages, has diffuse histologic features, and occurs more frequently in women. We investigated somatic genomic alterations associated with the unique characteristics of sporadic diffuse gastric cancers (DGCs) from younger patients. METHODS: We conducted whole exome and RNA sequence analyses of 80 resected DGC samples from patients 45 years old or younger in Korea. Patients with pathogenic germline mutations in CDH1, TP53, and ATM were excluded from the onset of this analysis, given our focus on somatic alterations. We used MutSig2CV to evaluate the significance of mutated genes. We recruited 29 additional early-onset Korean DGC samples and performed SNP6.0 array and targeted sequencing analyses of these 109 early-onset DGC samples (54.1% female, median age, 38 years). We compared the SNP6.0 array and targeted sequencing data of the 109 early-onset DGC samples with those from diffuse-type stomach tumor samples collected from 115 patients in Korea who were 46 years or older (late onset) at the time of diagnosis (controls; 29.6% female, median age, 67 years). We compared patient survival times among tumors from different subgroups and with different somatic mutations. We performed gene silencing of RHOA or CDH1 in DGC cells with small interfering RNAs for cell-based assays. RESULTS: We identified somatic mutations in the following genes in a significant number of early-onset DGCs: the cadherin 1 gene (CDH1), TP53, ARID1A, KRAS, PIK3CA, ERBB3, TGFBR1, FBXW7, RHOA, and MAP2K1. None of 109 early-onset DGC cases had pathogenic germline CDH1 mutations. A higher proportion of early-onset DGCs had mutations in CDH1 (42.2%) or TGFBR1 (7.3%) compared with control DGCs (17.4% and 0.9%, respectively) (P < .001 and P = .014 for CDH1 and TGFBR1, respectively). In contrast, a smaller proportion of early-onset DGCs contained mutations in RHOA (9.2%) than control DGCs (19.1%) (P = .033). Late-onset DGCs in The Cancer Genome Atlas also contained less frequent mutations in CDH1 and TGFBR1 and more frequent RHOA mutations, compared with early-onset DGCs. Early-onset DGCs from women contained significantly more mutations in CDH1 or TGFBR1 than early-onset DGCs from men. CDH1 alterations, but not RHOA mutations, were associated with shorter survival times in patients with early-onset DGCs (hazard ratio, 3.4; 95% confidence interval, 1.5-7.7). RHOA activity was reduced by an R5W substitution-the RHOA mutation most frequently detected in early-onset DGCs. Silencing of CDH1, but not RHOA, increased migratory activity of DGC cells. CONCLUSIONS: In an integrative genomic analysis, we found higher proportions of early-onset DGCs to contain somatic mutations in CDH1 or TGFBR1 compared with late-onset DGCs. However, a smaller proportion of early-onset DGCs contained somatic mutations in RHOA than late-onset DGCs. CDH1 alterations, but not RHOA mutations, were associated with shorter survival times of patients, which might account for the aggressive clinical course of early-onset gastric cancer. Female predominance in early-onset gastric cancer may be related to relatively high rates of somatic CDH1 and TGFBR1 mutations in this population.


Subject(s)
Age of Onset , Cadherins/genetics , Protein Serine-Threonine Kinases/genetics , Receptors, Transforming Growth Factor beta/genetics , Stomach Neoplasms/genetics , rhoA GTP-Binding Protein/genetics , Adult , Antigens, CD , Female , Gene Frequency , Genome-Wide Association Study , Germ-Line Mutation , Humans , Male , Middle Aged , Receptor, Transforming Growth Factor-beta Type I , Republic of Korea , Sex Factors , Young Adult
11.
Nat Genet ; 49(5): 780-788, 2017 May.
Article in English | MEDLINE | ID: mdl-28394352

ABSTRACT

Spatial heterogeneity of transcriptional and genetic markers between physically isolated biopsies of a single tumor poses major barriers to the identification of biomarkers and the development of targeted therapies that will be effective against the entire tumor. We analyzed the spatial heterogeneity of multiregional biopsies from 35 patients, using a combination of transcriptomic and genomic profiles. Medulloblastomas (MBs), but not high-grade gliomas (HGGs), demonstrated spatially homogeneous transcriptomes, which allowed for accurate subgrouping of tumors from a single biopsy. Conversely, somatic mutations that affect genes suitable for targeted therapeutics demonstrated high levels of spatial heterogeneity in MB, malignant glioma, and renal cell carcinoma (RCC). Actionable targets found in a single MB biopsy were seldom clonal across the entire tumor, which brings the efficacy of monotherapies against a single target into question. Clinical trials of targeted therapies for MB should first ensure the spatially ubiquitous nature of the target mutation.


Subject(s)
Cerebellar Neoplasms/genetics , Gene Expression Regulation, Neoplastic , Medulloblastoma/genetics , Transcriptome , Adult , Aged , Aged, 80 and over , Cerebellar Neoplasms/pathology , Child , Child, Preschool , Cluster Analysis , DNA Copy Number Variations , Female , Gene Expression Profiling/methods , Genetic Heterogeneity , Genome-Wide Association Study , Humans , INDEL Mutation , Male , Medulloblastoma/pathology , Middle Aged , Mutation , Polymorphism, Single Nucleotide , Principal Component Analysis , Reverse Transcriptase Polymerase Chain Reaction
13.
Oncotarget ; 7(18): 25930-48, 2016 May 03.
Article in English | MEDLINE | ID: mdl-27036018

ABSTRACT

Ovarian cancer presents as an aggressive, advanced stage cancer with widespread metastases that depend primarily on multicellular spheroids in the peritoneal fluid. To identify new druggable pathways related to metastatic progression and spheroid formation, we integrated microRNA and mRNA sequencing data from 293 tumors from The Cancer Genome Atlas (TCGA) ovarian cancer cohort. We identified miR-509-3p as a clinically significant microRNA that is more abundant in patients with favorable survival in both the TCGA cohort (P = 2.3E-3), and, by in situ hybridization (ISH), in an independent cohort of 157 tumors (P < 1.0E-3). We found that miR-509-3p attenuated migration and disrupted multi-cellular spheroids in HEYA8, OVCAR8, SKOV3, OVCAR3, OVCAR4 and OVCAR5 cell lines. Consistent with disrupted spheroid formation, in TCGA data miR-509-3p's most strongly anti-correlated predicted targets were enriched in components of the extracellular matrix (ECM). We validated the Hippo pathway effector YAP1 as a direct miR-509-3p target. We showed that siRNA to YAP1 replicated 90% of miR-509-3p-mediated migration attenuation in OVCAR8, which contained high levels of YAP1 protein, but not in the other cell lines, in which levels of this protein were moderate to low. Our data suggest that the miR-509-3p/YAP1 axis may be a new druggable target in cancers with high YAP1, and we propose that therapeutically targeting the miR-509-3p/YAP1/ECM axis may disrupt early steps in multi-cellular spheroid formation, and so inhibit metastasis in epithelial ovarian cancer and potentially in other cancers.


Subject(s)
Cell Movement/genetics , Gene Expression Regulation, Neoplastic/genetics , MicroRNAs/genetics , Neoplasms, Glandular and Epithelial/pathology , Ovarian Neoplasms/pathology , Adaptor Proteins, Signal Transducing/biosynthesis , Biomarkers, Tumor/analysis , Carcinoma, Ovarian Epithelial , Cell Line, Tumor , Female , Humans , Kaplan-Meier Estimate , Neoplasms, Glandular and Epithelial/genetics , Neoplasms, Glandular and Epithelial/mortality , Ovarian Neoplasms/genetics , Ovarian Neoplasms/mortality , Phosphoproteins/biosynthesis , Spheroids, Cellular/pathology , Transcription Factors , YAP-Signaling Proteins
14.
Cancer Cell ; 29(3): 394-406, 2016 Mar 14.
Article in English | MEDLINE | ID: mdl-26977886

ABSTRACT

Malignant rhabdoid tumors (MRTs) are rare lethal tumors of childhood that most commonly occur in the kidney and brain. MRTs are driven by SMARCB1 loss, but the molecular consequences of SMARCB1 loss in extra-cranial tumors have not been comprehensively described and genomic resources for analyses of extra-cranial MRT are limited. To provide such data, we used whole-genome sequencing, whole-genome bisulfite sequencing, whole transcriptome (RNA-seq) and microRNA sequencing (miRNA-seq), and histone modification profiling to characterize extra-cranial MRTs. Our analyses revealed gene expression and methylation subgroups and focused on dysregulated pathways, including those involved in neural crest development.


Subject(s)
Gene Expression Regulation, Developmental/genetics , Rhabdoid Tumor/genetics , Cell Line, Tumor , Chromosomal Proteins, Non-Histone/genetics , DNA Methylation/genetics , DNA-Binding Proteins/genetics , Gene Expression/genetics , Histones/genetics , Humans , MicroRNAs/genetics , SMARCB1 Protein , Transcription Factors/genetics , Transcriptome/genetics
15.
Nature ; 529(7586): 351-7, 2016 Jan 21.
Article in English | MEDLINE | ID: mdl-26760213

ABSTRACT

The development of targeted anti-cancer therapies through the study of cancer genomes is intended to increase survival rates and decrease treatment-related toxicity. We treated a transposon-driven, functional genomic mouse model of medulloblastoma with 'humanized' in vivo therapy (microneurosurgical tumour resection followed by multi-fractionated, image-guided radiotherapy). Genetic events in recurrent murine medulloblastoma exhibit a very poor overlap with those in matched murine diagnostic samples (<5%). Whole-genome sequencing of 33 pairs of human diagnostic and post-therapy medulloblastomas demonstrated substantial genetic divergence of the dominant clone after therapy (<12% diagnostic events were retained at recurrence). In both mice and humans, the dominant clone at recurrence arose through clonal selection of a pre-existing minor clone present at diagnosis. Targeted therapy is unlikely to be effective in the absence of the target, therefore our results offer a simple, proximal, and remediable explanation for the failure of prior clinical trials of targeted therapy.


Subject(s)
Cerebellar Neoplasms/therapy , Clone Cells/drug effects , Clone Cells/metabolism , Medulloblastoma/therapy , Neoplasm Recurrence, Local/genetics , Neoplasm Recurrence, Local/pathology , Selection, Genetic/drug effects , Animals , Cerebellar Neoplasms/genetics , Cerebellar Neoplasms/pathology , Cerebellar Neoplasms/radiotherapy , Cerebellar Neoplasms/surgery , Clone Cells/pathology , Craniospinal Irradiation , DNA Mutational Analysis , Disease Models, Animal , Drosophila melanogaster/cytology , Drosophila melanogaster/genetics , Female , Genome, Human/genetics , Humans , Male , Medulloblastoma/genetics , Medulloblastoma/pathology , Medulloblastoma/radiotherapy , Medulloblastoma/surgery , Mice , Molecular Targeted Therapy/methods , Neoplasm Recurrence, Local/therapy , Radiotherapy, Image-Guided , Signal Transduction , Xenograft Model Antitumor Assays
16.
Gerontology ; 62(1): 58-62, 2015.
Article in English | MEDLINE | ID: mdl-26066993

ABSTRACT

BACKGROUND: It is not understood whether long-term good health is promoted by the absence of disease risk variants, the presence of protective variants, or both. We characterized the exomes of two exceptionally healthy centenarian brothers aged 106 and 109 years who had never been diagnosed with cancer, cardiovascular disease, diabetes, Alzheimer's disease, or major pulmonary disease. OBJECTIVE: The aim of this study was to gain insight into whether exceptional health and longevity are a result of carrying fewer disease-associated variants than typical individuals. METHODS: We compared the number of disease-associated alleles, and the proportion of alleles predicted to be functionally damaging, between the centenarian brothers and published population data. Mitochondrial sequence reads were extracted from the exome data in order to analyze mitochondrial variants. RESULTS: The brothers carry a similar number of common disease-associated variants and predicted damaging variants compared to reference groups. They did not carry any high-penetrance clinically actionable variants. They carry mitochondrial haplogroup T, and one brother has a single heteroplasmic variant. CONCLUSION: Although our small sample size does not allow for definitive conclusions, a healthy aging and longevity phenotype is not necessarily due to a decreased burden of common disease-associated variants. Instead, it may be rare 'positive' variants that play a role in this desirable phenotype.


Subject(s)
Aging , Disease/genetics , Longevity , Aged, 80 and over , Alleles , Apolipoproteins B/genetics , Calcium Channels/genetics , Calcium Channels, L-Type , DNA, Mitochondrial/genetics , Exome , Genes, BRCA2 , Genes, p53/genetics , Genetic Predisposition to Disease , Genetic Variation , Genotype , Humans , Male , Phenotype , Polymorphism, Single Nucleotide , Sequence Analysis, DNA , Siblings
17.
Cell Rep ; 9(1): 261-271, 2014 Oct 09.
Article in English | MEDLINE | ID: mdl-25263553

ABSTRACT

Cell fate acquisition is heavily influenced by direct interactions between master regulators and tissue-specific enhancers. However, it remains unclear how lineage-specifying transcription factors, which are often expressed in both progenitor and mature cell populations, influence cell differentiation. Using in vivo mouse liver development as a model, we identified thousands of enhancers that are bound by the master regulators HNF4A and FOXA2 in a differentiation-dependent manner, subject to chromatin remodeling, and associated with differentially expressed target genes. Enhancers exclusively occupied in the embryo were found to be responsive to developmentally regulated TEAD2 and coactivator YAP1. Our data suggest that Hippo signaling may affect hepatocyte differentiation by influencing HNF4A and FOXA2 interactions with temporal enhancers. In summary, transcription factor-enhancer interactions are not only tissue specific but also differentiation dependent, which is an important consideration for researchers studying cancer biology or mammalian development and/or using transformed cell lines.


Subject(s)
Hepatocyte Nuclear Factor 3-beta/metabolism , Hepatocyte Nuclear Factor 4/metabolism , Hepatocytes/cytology , Hepatocytes/metabolism , Protein Serine-Threonine Kinases/metabolism , Animals , Cell Differentiation/physiology , Female , Gene Expression , Hepatocyte Nuclear Factor 3-beta/genetics , Hepatocyte Nuclear Factor 4/genetics , Hippo Signaling Pathway , Mice , Mice, Inbred C57BL , Mice, Transgenic , Protein Serine-Threonine Kinases/genetics , Signal Transduction
18.
PLoS One ; 9(7): e102398, 2014.
Article in English | MEDLINE | ID: mdl-25062255

ABSTRACT

JAGuaR is an alignment protocol for RNA-seq reads that uses an extended reference to increase alignment sensitivity. It uses BWA to align reads to the genome and reference transcript models (including annotated exon-exon junctions) specifically allowing for the possibility of a single read spanning multiple exons. Reads aligned to the transcript models are then re-mapped on to genomic coordinates, transforming alignments that span multiple exons into large-gapped alignments on the genome. While JAGuaR does not detect novel junctions, we demonstrate how JAGuaR generates fast and accurate transcriptome alignments, which allows for both sensitive and specific SNV calling.


Subject(s)
RNA/genetics , Sequence Alignment/methods , Sequence Analysis, RNA/methods , Software , Algorithms , Animals , Base Sequence , Exons , Gene Expression Profiling/methods , High-Throughput Nucleotide Sequencing , Humans , RNA Splicing/genetics
19.
BMC Genomics ; 14: 550, 2013 Aug 14.
Article in English | MEDLINE | ID: mdl-23941359

ABSTRACT

BACKGROUND: Chimeric transcripts, including partial and internal tandem duplications (PTDs, ITDs) and gene fusions, are important in the detection, prognosis, and treatment of human cancers. RESULTS: We describe Barnacle, a production-grade analysis tool that detects such chimeras in de novo assemblies of RNA-seq data, and supports prioritizing them for review and validation by reporting the relative coverage of co-occurring chimeric and wild-type transcripts. We demonstrate applications in large-scale disease studies, by identifying PTDs in MLL, ITDs in FLT3, and reciprocal fusions between PML and RARA, in two deeply sequenced acute myeloid leukemia (AML) RNA-seq datasets. CONCLUSIONS: Our analyses of real and simulated data sets show that, with appropriate filter settings, Barnacle makes highly specific predictions for three types of chimeric transcripts that are important in a range of cancers: PTDs, ITDs, and fusions. High specificity makes manual review and validation efficient, which is necessary in large-scale disease studies. Characterizing an extended range of chimera types will help generate insights into progression, treatment, and outcomes for complex diseases.


Subject(s)
Gene Duplication/genetics , Gene Expression Profiling/methods , Gene Fusion/genetics , Genomics , Breast Neoplasms/genetics , Exons/genetics , Humans , Leukemia, Myeloid, Acute/genetics , Molecular Sequence Annotation , RNA, Messenger/genetics , Statistics as Topic
20.
J Pathol ; 230(3): 249-60, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23616356

ABSTRACT

Parathyroid carcinoma is a rare endocrine malignancy with an estimated incidence of less than 1 per million population. Excessive secretion of parathyroid hormone, extremely high serum calcium level, and the deleterious effects of hypercalcaemia are the clinical manifestations of the disease. Up to 60% of patients develop multiple disease recurrences and although long-term survival is possible with palliative surgery, permanent remission is rarely achieved. Molecular drivers of sporadic parathyroid carcinoma have remained largely unknown. Previous studies, mostly based on familial cases of the disease, suggested potential roles for the tumour suppressor MEN1 and proto-oncogene RET in benign parathyroid tumourigenesis, while the tumour suppressor HRPT2 and proto-oncogene CCND1 may also act as drivers in parathyroid cancer. Here, we report the complete genomic analysis of a sporadic and recurring parathyroid carcinoma. Mutational landscapes of the primary and recurrent tumour specimens were analysed using high-throughput sequencing technologies. Such molecular profiling allowed for identification of somatic mutations never previously identified in this malignancy. These included single nucleotide point mutations in well-characterized cancer genes such as mTOR, MLL2, CDKN2C, and PIK3CA. Comparison of acquired mutations in patient-matched primary and recurrent tumours revealed loss of PIK3CA activating mutation during the evolution of the tumour from the primary to the recurrence. Structural variations leading to gene fusions and regions of copy loss and gain were identified at a single-base resolution. Loss of the short arm of chromosome 1, along with somatic missense and truncating mutations in CDKN2C and THRAP3, respectively, provides new evidence for the potential role of these genes as tumour suppressors in parathyroid cancer. The key somatic mutations identified in this study can serve as novel diagnostic markers as well as therapeutic targets.


Subject(s)
Biomarkers, Tumor/genetics , Gene Expression Profiling , Genomics , Neoplasm Recurrence, Local/genetics , Parathyroid Neoplasms/genetics , Adult , Base Sequence , Calcium/blood , Cell Transformation, Neoplastic , Class I Phosphatidylinositol 3-Kinases , Cyclin-Dependent Kinase Inhibitor p18/genetics , DNA, Neoplasm/chemistry , DNA, Neoplasm/genetics , DNA-Binding Proteins/genetics , Gene Dosage , Gene Fusion , Humans , Male , Molecular Sequence Data , Mutation , Neoplasm Proteins/genetics , Neoplasm Recurrence, Local/pathology , Neoplasm Recurrence, Local/surgery , Parathyroid Hormone/metabolism , Parathyroid Neoplasms/pathology , Parathyroid Neoplasms/surgery , Phosphatidylinositol 3-Kinases/genetics , Polymorphism, Single Nucleotide , Proto-Oncogene Mas , RNA, Neoplasm/genetics , TOR Serine-Threonine Kinases/genetics , Transcription Factors/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...