Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22
Filter
1.
Nature ; 547(7661): 55-60, 2017 07 06.
Article in English | MEDLINE | ID: mdl-28658208

ABSTRACT

Genomic analysis of tumours has led to the identification of hundreds of cancer genes on the basis of the presence of mutations in protein-coding regions. By contrast, much less is known about cancer-causing mutations in non-coding regions. Here we perform deep sequencing in 360 primary breast cancers and develop computational methods to identify significantly mutated promoters. Clear signals are found in the promoters of three genes. FOXA1, a known driver of hormone-receptor positive breast cancer, harbours a mutational hotspot in its promoter leading to overexpression through increased E2F binding. RMRP and NEAT1, two non-coding RNA genes, carry mutations that affect protein binding to their promoters and alter expression levels. Our study shows that promoter regions harbour recurrent mutations in cancer with functional consequences and that the mutations occur at similar frequencies as in coding regions. Power analyses indicate that more such regions remain to be discovered through deep sequencing of adequately sized cohorts of patients.


Subject(s)
Breast Neoplasms/genetics , Gene Expression Regulation, Neoplastic/genetics , Mutation , Promoter Regions, Genetic/genetics , Cohort Studies , E2F Transcription Factors/metabolism , Exome/genetics , Hepatocyte Nuclear Factor 3-alpha/genetics , Hepatocyte Nuclear Factor 3-alpha/metabolism , High-Throughput Nucleotide Sequencing , Humans , Protein Binding/genetics , RNA, Long Noncoding/genetics , Receptors, Estrogen/antagonists & inhibitors
2.
Proc Natl Acad Sci U S A ; 111(34): 12450-5, 2014 Aug 26.
Article in English | MEDLINE | ID: mdl-25107291

ABSTRACT

Congenital diaphragmatic hernia (CDH) is a common and severe birth defect. Despite its clinical significance, the genetic and developmental pathways underlying this disorder are incompletely understood. In this study, we report a catalog of variants detected by a whole exome sequencing study on 275 individuals with CDH. Predicted pathogenic variants in genes previously identified in either humans or mice with diaphragm defects are enriched in our CDH cohort compared with 120 size-matched random gene sets. This enrichment was absent in control populations. Variants in these critical genes can be found in up to 30.9% of individuals with CDH. In addition, we filtered variants by using genes derived from regions of recurrent copy number variations in CDH, expression profiles of the developing diaphragm, protein interaction networks expanded from the known CDH-causing genes, and prioritized genes with ultrarare and highly disruptive variants, in 11.3% of CDH patients. These strategies have identified several high priority genes and developmental pathways that likely contribute to the CDH phenotype. These data are valuable for comparison of candidate genes generated from whole exome sequencing of other CDH cohorts or multiplex kindreds and provide ideal candidates for further functional studies. Furthermore, we propose that these genes and pathways will enhance our understanding of the heterogeneous molecular etiology of CDH.


Subject(s)
Hernias, Diaphragmatic, Congenital/etiology , Hernias, Diaphragmatic, Congenital/genetics , Animals , Cohort Studies , Computational Biology , DNA Copy Number Variations , Diaphragm/embryology , Exome , Genetic Variation , Hernias, Diaphragmatic, Congenital/embryology , Humans , Mice , Protein Interaction Maps
3.
Proc Natl Acad Sci U S A ; 109(8): 2978-83, 2012 Feb 21.
Article in English | MEDLINE | ID: mdl-22315423

ABSTRACT

Congenital diaphragmatic hernia (CDH) is a common (1 in 3,000 live births) major congenital malformation that results in significant morbidity and mortality. The discovery of CDH loci using standard genetic approaches has been hindered by its genetic heterogeneity. We hypothesized that gene expression profiling of developing embryonic diaphragms would help identify genes likely to be associated with diaphragm defects. We generated a time series of whole-transcriptome expression profiles from laser captured embryonic mouse diaphragms at embryonic day (E)11.5 and E12.5 when experimental perturbations lead to CDH phenotypes, and E16.5 when the diaphragm is fully formed. Gene sets defining biologically relevant pathways and temporal expression trends were identified by using a series of bioinformatic algorithms. These developmental sets were then compared with a manually curated list of genes previously shown to cause diaphragm defects in humans and in mouse models. Our integrative filtering strategy identified 27 candidates for CDH. We examined the diaphragms of knockout mice for one of the candidate genes, pre-B-cell leukemia transcription factor 1 (Pbx1), and identified a range of previously undetected diaphragmatic defects. Our study demonstrates the utility of genetic characterization of normal development as an integral part of a disease gene identification and prioritization strategy for CDH, an approach that can be extended to other diseases and developmental anomalies.


Subject(s)
Embryo, Mammalian/metabolism , Embryo, Mammalian/pathology , Genetic Association Studies , Hernias, Diaphragmatic, Congenital , Transcriptome/genetics , Animals , Diaphragm/embryology , Diaphragm/metabolism , Diaphragm/pathology , Gene Expression Regulation, Developmental , Hernia, Diaphragmatic/genetics , Hernia, Diaphragmatic/pathology , Homeodomain Proteins/metabolism , Lasers , Mesoderm/embryology , Mesoderm/metabolism , Mesoderm/pathology , Mice , Mice, Knockout , Models, Biological , Pre-B-Cell Leukemia Transcription Factor 1 , Signal Transduction/genetics , Time Factors , Transcription Factors/deficiency , Transcription Factors/metabolism , Transcription, Genetic
4.
Am J Med Genet A ; 158A(12): 3148-58, 2012 Dec.
Article in English | MEDLINE | ID: mdl-23165946

ABSTRACT

Chromosome 8p23.1 is a common hotspot associated with major congenital malformations, including congenital diaphragmatic hernia (CDH) and cardiac defects. We present findings from high-resolution arrays in patients who carry a loss (n = 18) or a gain (n = 1) of sub-band 8p23.1. We confirm a region involved in both diaphragmatic and heart malformations. Results from a novel CNVConnect algorithm, prioritizing protein-protein interactions between products of genes in the 8p23.1 hotspot and products of previously known CDH causing genes, implicated GATA4, NEIL2, and SOX7 in diaphragmatic defects. Sequence analysis of these genes in 226 chromosomally normal CDH patients, as well as in a small number of deletion 8p23.1 patients, showed rare unreported variants in the coding region; these may be contributing to the diaphragmatic phenotype. We also demonstrated that two of these three genes were expressed in the E11.5-12.5 primordial mouse diaphragm, the developmental stage at which CDH is thought to occur. This combination of bioinformatics and expression studies can be applied to other chromosomal hotspots, as well as private microdeletions or microduplications, to identify causative genes and their interaction networks.


Subject(s)
Hernias, Diaphragmatic, Congenital , Animals , Chromosome Deletion , Chromosomes, Human, Pair 8/genetics , Chromosomes, Human, Pair 8/metabolism , DNA/blood , DNA/genetics , DNA Glycosylases/genetics , DNA-(Apurinic or Apyrimidinic Site) Lyase/genetics , Female , GATA4 Transcription Factor/genetics , Heart Defects, Congenital/blood , Heart Defects, Congenital/genetics , Heart Defects, Congenital/metabolism , Hernia, Diaphragmatic/blood , Hernia, Diaphragmatic/genetics , Hernia, Diaphragmatic/metabolism , Humans , Karyotyping , Mice , Mice, Inbred C57BL , Phenotype , Pregnancy , Protein Interaction Maps , SOXF Transcription Factors/genetics
5.
Tetrahedron Lett ; 53(14): 1701-1704, 2012 Apr 04.
Article in English | MEDLINE | ID: mdl-22753995

ABSTRACT

The hydroxyalkylation reaction has been used to condense benzocrown ethers with various aldehydes and ketones. The condensation reactions are catalyzed by triflic or sulfuric acid. The products from the reactions are bis(benzocrown ethers) and they are formed in good yields (42-98%, 13 examples).

6.
Org Biomol Chem ; 9(12): 4545-9, 2011 Jun 21.
Article in English | MEDLINE | ID: mdl-21547324

ABSTRACT

A series of trifluoromethyl-substituted arenes were studied in their reactions with Brønsted superacids. The products from these reactions suggest the formation of reactive electrophiles, such as carbocations, acylium cations or equivalent electrophilic species. As such, Friedel-Crafts-type reactions occur between these species and arene nucleophiles. NMR studies were done, and the results suggest the formation of an acyl group from the trifluoromethyl groups in the superacid.


Subject(s)
Chemistry, Pharmaceutical/methods , Fluorides/chemistry , Mesylates/chemistry , Toluene/analogs & derivatives , Acylation , Benzoic Acid/chemistry , Catalysis , Cations , Magnetic Resonance Spectroscopy , Toluene/chemistry
8.
Clin Cancer Res ; 25(18): 5561-5571, 2019 09 15.
Article in English | MEDLINE | ID: mdl-31253631

ABSTRACT

PURPOSE: Molecular properties associated with complete response or acquired resistance to concurrent chemotherapy and radiotherapy (CRT) are incompletely characterized.Experimental Design: We performed integrated whole-exome/transcriptome sequencing and immune infiltrate analysis on rectal adenocarcinoma tumors prior to neoadjuvant CRT (pre-CRT) and at time of resection (post-CRT) in 17 patients [8 complete/partial responders, 9 nonresponders (NR)]. RESULTS: CRT was not associated with increased tumor mutational burden or neoantigen load and did not alter the distribution of established somatic tumor mutations in rectal cancer. Concurrent KRAS/TP53 mutations (KP) associated with NR tumors and were enriched for an epithelial-mesenchymal transition transcriptional program. Furthermore, NR was associated with reduced CD4/CD8 T-cell infiltrates and a post-CRT M2 macrophage phenotype. Absence of any local tumor recurrences, KP/NR status predicted worse progression-free survival, suggesting that local immune escape during or after CRT with specific genomic features contributes to distant progression. CONCLUSIONS: Overall, while CRT did not impact genomic profiles, CRT impacted the tumor immune microenvironment, particularly in resistant cases.


Subject(s)
Biomarkers, Tumor , Drug Resistance, Neoplasm , Radiation Tolerance , Rectal Neoplasms/etiology , Antigens, Neoplasm/immunology , Chemoradiotherapy, Adjuvant , Drug Resistance, Neoplasm/genetics , Gene Expression Profiling , Humans , Immunohistochemistry , Mutation , Proto-Oncogene Proteins p21(ras)/genetics , Proto-Oncogene Proteins p21(ras)/metabolism , Radiation Tolerance/genetics , Rectal Neoplasms/metabolism , Rectal Neoplasms/therapy , Transcriptome , Tumor Microenvironment/genetics , Tumor Microenvironment/immunology , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism , Exome Sequencing
9.
Clin Cancer Res ; 25(16): 5135-5142, 2019 08 15.
Article in English | MEDLINE | ID: mdl-31164371

ABSTRACT

PURPOSE: Leiomyosarcoma and liposarcoma are common subtypes of soft tissue sarcoma (STS). Patients with metastatic leiomyosarcoma or dedifferentiated liposarcoma (DDLPS) typically have worse outcomes compared with localized leiomyosarcoma or well-differentiated liposarcoma (WDLPS). A better understanding of genetic changes between primary/metastatic leiomyosarcoma and between WDLPS/DDLPS may provide insight into their genetic evolution. EXPERIMENTAL DESIGN: We interrogated whole-exome sequencing (WES) from "trios" of normal tissue, primary tumor, and metastatic tumor from individual patients with leiomyosarcoma (n = 9), and trios of normal tissue, well-differentiated tumor, and dedifferentiated tumor from individual patients with liposarcoma (n = 19). Specifically, we performed mutational, copy number, and tumor evolution analyses on these cohorts and compared patterns among leiomyosarcoma and liposarcoma trios. RESULTS: Leiomyosarcoma cases harbored shared drivers through a typical parent/child relationship where the metastatic tumor was derived from the primary tumor. In contrast, while all liposarcoma cases shared the characteristic focal chromosome 12 amplicon, most paired liposarcoma cases did not share additional mutations, suggesting a divergent evolutionary pattern from a common precursor. No highly recurrent genomic alterations from WES were identified that could be implicated as driving the progression of disease in either sarcoma subtype. CONCLUSIONS: From a genomic perspective, leiomyosarcoma metastases contain genetic alterations that are also found in primary tumors. WDLPS and DDLPS, however, appear to divergently evolve from a common precursor harboring 12q amplification, rather than as a transformation to a higher-grade tumor. Further efforts to identify specific drivers of these distinct evolutionary patterns may inform future translational and clinical research in STS.


Subject(s)
Cell Transformation, Neoplastic/genetics , Genetic Predisposition to Disease , Genomics , Leiomyosarcoma/genetics , Leiomyosarcoma/pathology , Liposarcoma/genetics , Liposarcoma/pathology , Adult , Aged , Biopsy , Female , Gene Expression Profiling , Genomics/methods , Humans , Male , Middle Aged , Neoplasm Metastasis , Neoplasm Staging , Retrospective Studies , Exome Sequencing
10.
Nat Med ; 25(12): 1916-1927, 2019 12.
Article in English | MEDLINE | ID: mdl-31792460

ABSTRACT

Immune-checkpoint blockade (ICB) has demonstrated efficacy in many tumor types, but predictors of responsiveness to anti-PD1 ICB are incompletely characterized. In this study, we analyzed a clinically annotated cohort of patients with melanoma (n = 144) treated with anti-PD1 ICB, with whole-exome and whole-transcriptome sequencing of pre-treatment tumors. We found that tumor mutational burden as a predictor of response was confounded by melanoma subtype, whereas multiple novel genomic and transcriptomic features predicted selective response, including features associated with MHC-I and MHC-II antigen presentation. Furthermore, previous anti-CTLA4 ICB exposure was associated with different predictors of response compared to tumors that were naive to ICB, suggesting selective immune effects of previous exposure to anti-CTLA4 ICB. Finally, we developed parsimonious models integrating clinical, genomic and transcriptomic features to predict intrinsic resistance to anti-PD1 ICB in individual tumors, with validation in smaller independent cohorts limited by the availability of comprehensive data. Broadly, we present a framework to discover predictive features and build models of ICB therapeutic response.


Subject(s)
CTLA-4 Antigen/immunology , Melanoma/drug therapy , Melanoma/genetics , Programmed Cell Death 1 Receptor/immunology , Antibodies, Monoclonal, Humanized/administration & dosage , Antigen Presentation/genetics , Antigen Presentation/immunology , CTLA-4 Antigen/antagonists & inhibitors , Drug Resistance, Neoplasm/genetics , Female , Humans , Male , Melanoma/immunology , Melanoma/pathology , Mutation/genetics , Neoplasm Metastasis , Nivolumab/administration & dosage , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Transcriptome/genetics , Transcriptome/immunology , Exome Sequencing
11.
Elife ; 82019 03 12.
Article in English | MEDLINE | ID: mdl-30860482

ABSTRACT

Renal medullary carcinoma (RMC) is a rare and deadly kidney cancer in patients of African descent with sickle cell trait. We have developed faithful patient-derived RMC models and using whole-genome sequencing, we identified loss-of-function intronic fusion events in one SMARCB1 allele with concurrent loss of the other allele. Biochemical and functional characterization of these models revealed that RMC requires the loss of SMARCB1 for survival. Through integration of RNAi and CRISPR-Cas9 loss-of-function genetic screens and a small-molecule screen, we found that the ubiquitin-proteasome system (UPS) was essential in RMC. Inhibition of the UPS caused a G2/M arrest due to constitutive accumulation of cyclin B1. These observations extend across cancers that harbor SMARCB1 loss, which also require expression of the E2 ubiquitin-conjugating enzyme, UBE2C. Our studies identify a synthetic lethal relationship between SMARCB1-deficient cancers and reliance on the UPS which provides the foundation for a mechanism-informed clinical trial with proteasome inhibitors.


Subject(s)
Carcinoma, Medullary/genetics , Kidney Neoplasms/genetics , Proteasome Endopeptidase Complex/genetics , Proteasome Inhibitors/pharmacology , SMARCB1 Protein/genetics , Alleles , Animals , CRISPR-Cas Systems , Carcinoma, Medullary/drug therapy , Cell Cycle , Cell Line, Tumor , Exome , Female , Humans , In Situ Hybridization, Fluorescence , Kidney/metabolism , Kidney Neoplasms/drug therapy , Mice , Mice, Nude , Mutation , Neoplasm Transplantation , RNA Interference , Sequence Analysis, RNA , Ubiquitin/chemistry , Whole Genome Sequencing
12.
Nat Commun ; 10(1): 2400, 2019 06 03.
Article in English | MEDLINE | ID: mdl-31160565

ABSTRACT

BET-bromodomain inhibition (BETi) has shown pre-clinical promise for MYC-amplified medulloblastoma. However, the mechanisms for its action, and ultimately for resistance, have not been fully defined. Here, using a combination of expression profiling, genome-scale CRISPR/Cas9-mediated loss of function and ORF/cDNA driven rescue screens, and cell-based models of spontaneous resistance, we identify bHLH/homeobox transcription factors and cell-cycle regulators as key genes mediating BETi's response and resistance. Cells that acquire drug tolerance exhibit a more neuronally differentiated cell-state and expression of lineage-specific bHLH/homeobox transcription factors. However, they do not terminally differentiate, maintain expression of CCND2, and continue to cycle through S-phase. Moreover, CDK4/CDK6 inhibition delays acquisition of resistance. Therefore, our data provide insights about the mechanisms underlying BETi effects and the appearance of resistance and support the therapeutic use of combined cell-cycle inhibitors with BETi in MYC-amplified medulloblastoma.


Subject(s)
Azepines/pharmacology , Cell Cycle/drug effects , Cerebellar Neoplasms/drug therapy , Medulloblastoma/drug therapy , Neurogenesis/drug effects , Proteins/antagonists & inhibitors , Triazoles/pharmacology , Animals , Basic Helix-Loop-Helix Transcription Factors/drug effects , Basic Helix-Loop-Helix Transcription Factors/metabolism , CRISPR-Cas Systems , Cell Cycle Proteins/drug effects , Cell Cycle Proteins/metabolism , Cell Line, Tumor , Cell Lineage , Cerebellar Neoplasms/genetics , Cyclin D2/drug effects , Cyclin D2/metabolism , Cyclin-Dependent Kinase 4/antagonists & inhibitors , Cyclin-Dependent Kinase 6/antagonists & inhibitors , Drug Resistance, Neoplasm , Gene Expression Profiling , Humans , Medulloblastoma/genetics , Mice , Neural Stem Cells/drug effects , Neural Stem Cells/metabolism , Proto-Oncogene Proteins c-myc/genetics , S Phase/drug effects
13.
Blood Adv ; 2(24): 3608-3617, 2018 12 26.
Article in English | MEDLINE | ID: mdl-30567725

ABSTRACT

Novel treatment strategies are needed for older patients with acute myeloid leukemia (AML). This randomized phase 2 trial compared the efficacy and safety of 20 mg/m2 of IV decitabine on days 1 to 10 alone (arm A) with those of 1.3 mg/m2 of subcutaneous bortezomib (arm B) on days 1, 4, 8, and 11 for up to 4 10-day cycles followed by monthly 5-day cycles. Previously untreated AML patients age ≥60 years (excluding those with FLT3 mutations and favorable-risk cytogenetics) without restrictions in performance status (PS) or organ function were eligible. Median age was 72.4 years (range, 60.5-92.3 years); 31 patients (19%) had baseline PS ≥2, 35 (22%) had an antecedent hematological disorder, 58 had (39%) adverse cytogenetics, and 7 (5%) and 23 (14%) had abnormal cardiac or renal function. There were no statistically significant differences in overall survival (OS) or responses between the 2 treatment arms. The overall response rate (complete remission + complete remission with incomplete blood count recovery) was 39% (n = 64), with median OS of 9.3 months. Nineteen responders (31%) underwent allogeneic stem cell transplantation. The most common adverse event was febrile neutropenia, and there were no unexpected toxicities. Adding bortezomib to decitabine did not improve outcomes, but responses were better than those in previous trials using 5-day decitabine cycles. This trial was registered at www.clinicaltrials.gov as #NCT01420926.


Subject(s)
Antineoplastic Agents/therapeutic use , Bortezomib/therapeutic use , Decitabine/therapeutic use , Leukemia, Myeloid, Acute/drug therapy , Aged , Aged, 80 and over , Disease-Free Survival , Drug Administration Schedule , Drug Therapy, Combination , Female , Humans , Kaplan-Meier Estimate , Leukemia, Myeloid, Acute/mortality , Male , Middle Aged , Mutation , Repressor Proteins/genetics , Treatment Outcome , Tumor Suppressor Protein p53/genetics
14.
Science ; 359(6377): 801-806, 2018 Feb 16.
Article in English | MEDLINE | ID: mdl-29301960

ABSTRACT

Immune checkpoint inhibitors targeting the programmed cell death 1 receptor (PD-1) improve survival in a subset of patients with clear cell renal cell carcinoma (ccRCC). To identify genomic alterations in ccRCC that correlate with response to anti-PD-1 monotherapy, we performed whole-exome sequencing of metastatic ccRCC from 35 patients. We found that clinical benefit was associated with loss-of-function mutations in the PBRM1 gene (P = 0.012), which encodes a subunit of the PBAF switch-sucrose nonfermentable (SWI/SNF) chromatin remodeling complex. We confirmed this finding in an independent validation cohort of 63 ccRCC patients treated with PD-1 or PD-L1 (PD-1 ligand) blockade therapy alone or in combination with anti-CTLA-4 (cytotoxic T lymphocyte-associated protein 4) therapies (P = 0.0071). Gene-expression analysis of PBAF-deficient ccRCC cell lines and PBRM1-deficient tumors revealed altered transcriptional output in JAK-STAT (Janus kinase-signal transducers and activators of transcription), hypoxia, and immune signaling pathways. PBRM1 loss in ccRCC may alter global tumor-cell expression profiles to influence responsiveness to immune checkpoint therapy.


Subject(s)
B7-H1 Antigen/antagonists & inhibitors , Carcinoma, Renal Cell/genetics , Carcinoma, Renal Cell/therapy , Immunotherapy/methods , Kidney Neoplasms/genetics , Kidney Neoplasms/therapy , Programmed Cell Death 1 Receptor/antagonists & inhibitors , CTLA-4 Antigen/antagonists & inhibitors , Chromosomal Proteins, Non-Histone/genetics , Cohort Studies , Exome/genetics , Gene Expression Profiling , Genomics , Humans , Mutation , Transcription Factors/genetics
16.
Nat Med ; 24(8): 1292, 2018 Aug.
Article in English | MEDLINE | ID: mdl-29955181

ABSTRACT

In the version of this article originally published, some text above the "Tri-nucleotide sequence motifs" label in Fig. 2a appeared incorrectly. The text was garbled and should have appeared as nucleotide codes.Additionally, the labels on the bars in Fig. 2c were not italicized in the original publication. These are gene symbols, and they should have been italicized.The colored labels above the graphs in Fig. 4b were also erroneously not italicized. These labels represent gene names and loci, and they should have been italicized.

17.
Nat Genet ; 50(9): 1271-1281, 2018 09.
Article in English | MEDLINE | ID: mdl-30150660

ABSTRACT

Tumor mutational burden correlates with response to immune checkpoint blockade in multiple solid tumors, although in microsatellite-stable tumors this association is of uncertain clinical utility. Here we uniformly analyzed whole-exome sequencing (WES) of 249 tumors and matched normal tissue from patients with clinically annotated outcomes to immune checkpoint therapy, including radiographic response, across multiple cancer types to examine additional tumor genomic features that contribute to selective response. Our analyses identified genomic correlates of response beyond mutational burden, including somatic events in individual driver genes, certain global mutational signatures, and specific HLA-restricted neoantigens. However, these features were often interrelated, highlighting the complexity of identifying genetic driver events that generate an immunoresponsive tumor environment. This study lays a path forward in analyzing large clinical cohorts in an integrated and multifaceted manner to enhance the ability to discover clinically meaningful predictive features of response to immune checkpoint blockade.


Subject(s)
Microsatellite Repeats , Neoplasms/genetics , Neoplasms/immunology , Cohort Studies , Exome , Genomics/methods , Humans , Immunity/genetics , Mutation
18.
Nat Med ; 24(5): 679-690, 2018 05.
Article in English | MEDLINE | ID: mdl-29713087

ABSTRACT

Diffuse large B cell lymphoma (DLBCL), the most common lymphoid malignancy in adults, is a clinically and genetically heterogeneous disease that is further classified into transcriptionally defined activated B cell (ABC) and germinal center B cell (GCB) subtypes. We carried out a comprehensive genetic analysis of 304 primary DLBCLs and identified low-frequency alterations, captured recurrent mutations, somatic copy number alterations, and structural variants, and defined coordinate signatures in patients with available outcome data. We integrated these genetic drivers using consensus clustering and identified five robust DLBCL subsets, including a previously unrecognized group of low-risk ABC-DLBCLs of extrafollicular/marginal zone origin; two distinct subsets of GCB-DLBCLs with different outcomes and targetable alterations; and an ABC/GCB-independent group with biallelic inactivation of TP53, CDKN2A loss, and associated genomic instability. The genetic features of the newly characterized subsets, their mutational signatures, and the temporal ordering of identified alterations provide new insights into DLBCL pathogenesis. The coordinate genetic signatures also predict outcome independent of the clinical International Prognostic Index and suggest new combination treatment strategies. More broadly, our results provide a roadmap for an actionable DLBCL classification.


Subject(s)
Lymphoma, Large B-Cell, Diffuse/genetics , Lymphoma, Large B-Cell, Diffuse/pathology , DNA Copy Number Variations/genetics , Gene Rearrangement/genetics , Genes, Neoplasm , Genetic Heterogeneity , Humans , Mutation/genetics , Mutation Rate , Treatment Outcome
19.
PLoS One ; 11(2): e0149425, 2016.
Article in English | MEDLINE | ID: mdl-26891231

ABSTRACT

Congenital diaphragmatic hernia is associated with pulmonary hypoplasia and respiratory distress, which result in high mortality and morbidity. Although several transgenic mouse models of lung hypoplasia exist, the role of miRNAs in this phenotype is incompletely characterized. In this study, we assessed microRNA expression levels during the pseudoglandular to canalicular phase transition of normal human fetal lung development. At this critical time, when the distal respiratory portion of the airways begins to form, microarray analysis showed that the most significantly differentially expressed miRNA was miR-449a. Prediction algorithms determined that N-myc is a target of miR-449a and identified the likely miR-449a:N-myc binding sites, confirmed by luciferase assays and targeted mutagenesis. Functional ex vivo knock-down in organ cultures of murine embryonic lungs, as well as in ovo overexpression in avian embryonic lungs, suggested a role for miR-449a in distal epithelial proliferation. Finally, miR-449a expression was found to be abnormal in rare pulmonary specimens of human fetuses with Congenital Diaphragmatic Hernia in the pseudoglandular or canalicular phase. This study confirms the conserved role of miR-449a for proper pulmonary organogenesis, supporting the delicate balance between expansion of progenitor cells and their terminal differentiation, and proposes the potential involvement of this miRNA in human pulmonary hypoplasia.


Subject(s)
Lung/embryology , Lung/metabolism , MicroRNAs/genetics , Organogenesis/genetics , 3' Untranslated Regions , Animals , Base Sequence , Binding Sites , Cell Differentiation/genetics , Cell Proliferation , Chickens , Gene Expression Regulation, Developmental , Humans , Lung/pathology , Mice , MicroRNAs/chemistry , Oncogene Proteins/chemistry , Oncogene Proteins/genetics , RNA Interference , RNA, Messenger/chemistry , RNA, Messenger/genetics , Transcription, Genetic
20.
Nat Genet ; 48(3): 273-82, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26829751

ABSTRACT

Angiocentric gliomas are pediatric low-grade gliomas (PLGGs) without known recurrent genetic drivers. We performed genomic analysis of new and published data from 249 PLGGs, including 19 angiocentric gliomas. We identified MYB-QKI fusions as a specific and single candidate driver event in angiocentric gliomas. In vitro and in vivo functional studies show that MYB-QKI rearrangements promote tumorigenesis through three mechanisms: MYB activation by truncation, enhancer translocation driving aberrant MYB-QKI expression and hemizygous loss of the tumor suppressor QKI. To our knowledge, this represents the first example of a single driver rearrangement simultaneously transforming cells via three genetic and epigenetic mechanisms in a tumor.


Subject(s)
Glioma/genetics , Oncogene Proteins v-myb/genetics , Oncogene Proteins, Fusion/genetics , RNA-Binding Proteins/genetics , Carcinogenesis/genetics , Cell Line, Tumor , Child , Comparative Genomic Hybridization , Exome/genetics , Gene Expression Regulation, Neoplastic , Gene Rearrangement , Glioma/pathology , High-Throughput Nucleotide Sequencing , Humans , Mutation , Oncogene Proteins v-myb/biosynthesis , Oncogene Proteins, Fusion/biosynthesis , RNA-Binding Proteins/biosynthesis
SELECTION OF CITATIONS
SEARCH DETAIL