Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 98
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Cell ; 154(3): 583-95, 2013 Aug 01.
Article in English | MEDLINE | ID: mdl-23911323

ABSTRACT

Intron retention (IR) is widely recognized as a consequence of mis-splicing that leads to failed excision of intronic sequences from pre-messenger RNAs. Our bioinformatic analyses of transcriptomic and proteomic data of normal white blood cell differentiation reveal IR as a physiological mechanism of gene expression control. IR regulates the expression of 86 functionally related genes, including those that determine the nuclear shape that is unique to granulocytes. Retention of introns in specific genes is associated with downregulation of splicing factors and higher GC content. IR, conserved between human and mouse, led to reduced mRNA and protein levels by triggering the nonsense-mediated decay (NMD) pathway. In contrast to the prevalent view that NMD is limited to mRNAs encoding aberrant proteins, our data establish that IR coupled with NMD is a conserved mechanism in normal granulopoiesis. Physiological IR may provide an energetically favorable level of dynamic gene expression control prior to sustained gene translation.


Subject(s)
Granulocytes/metabolism , Hematopoiesis , RNA Splicing , Algorithms , Animals , Base Composition , Cell Nucleus/metabolism , Down-Regulation , Granulocytes/cytology , Humans , Introns , Lamin Type B/genetics , Mice , Mice, Inbred C57BL , Nonsense Mediated mRNA Decay
2.
Blood ; 143(16): 1586-1598, 2024 Apr 18.
Article in English | MEDLINE | ID: mdl-38211335

ABSTRACT

ABSTRACT: Maintenance of quiescence and DNA replication dynamics are 2 paradoxical requirements for the distinct states of dormant and active hematopoietic stem cells (HSCs), which are required to preserve the stem cell reservoir and replenish the blood cell system in response to hematopoietic stress, respectively. Here, we show that key self-renewal factors, ß-catenin or Hoxa9, largely dispensable for HSC integrity, in fact, have dual functions in maintaining quiescence and enabling efficient DNA replication fork dynamics to preserve the functionality of hematopoietic stem and progenitor cells (HSPCs). Although ß-catenin or Hoxa9 single knockout (KO) exhibited mostly normal hematopoiesis, their coinactivation led to severe hematopoietic defects stemmed from aberrant cell cycle, DNA replication, and damage in HSPCs. Mechanistically, ß-catenin and Hoxa9 function in a compensatory manner to sustain key transcriptional programs that converge on the pivotal downstream target and epigenetic modifying enzyme, Prmt1, which protects the quiescent state and ensures an adequate supply of DNA replication and repair factors to maintain robust replication fork dynamics. Inactivation of Prmt1 phenocopied both cellular and molecular phenotypes of ß-catenin/Hoxa9 combined KO, which at the same time could also be partially rescued by Prmt1 expression. The discovery of the highly resilient ß-catenin/Hoxa9/Prmt1 axis in protecting both quiescence and DNA replication dynamics essential for HSCs at different key states provides not only novel mechanistic insights into their intricate regulation but also a potential tractable target for therapeutic intervention.


Subject(s)
Hematopoietic Stem Cells , beta Catenin , beta Catenin/metabolism , Hematopoietic Stem Cells/metabolism , Cell Cycle , Cell Division , DNA Replication
3.
Nucleic Acids Res ; 52(D1): D1131-D1137, 2024 Jan 05.
Article in English | MEDLINE | ID: mdl-37870453

ABSTRACT

The BloodChIP Xtra database (http://bloodchipXtra.vafaeelab.com/) facilitates genome-wide exploration and visualization of transcription factor (TF) occupancy and chromatin configuration in rare primary human hematopoietic stem (HSC-MPP) and progenitor (CMP, GMP, MEP) cells and acute myeloid leukemia (AML) cell lines (KG-1, ME-1, Kasumi1, TSU-1621-MT), along with chromatin accessibility and gene expression data from these and primary patient AMLs. BloodChIP Xtra features significantly more datasets than our earlier database BloodChIP (two primary cell types and two cell lines). Improved methodologies for determining TF occupancy and chromatin accessibility have led to increased availability of data for rare primary cell types across the spectrum of healthy and AML hematopoiesis. However, there is a continuing need for these data to be integrated in an easily accessible manner for gene-based queries and use in downstream applications. Here, we provide a user-friendly database based around genome-wide binding profiles of key hematopoietic TFs and histone marks in healthy stem/progenitor cell types. These are compared with binding profiles and chromatin accessibility derived from primary and cell line AML and integrated with expression data from corresponding cell types. All queries can be exported to construct TF-gene and protein-protein networks and evaluate the association of genes with specific cellular processes.


Subject(s)
Binding Sites , Gene Expression Profiling , Leukemia, Myeloid, Acute , Humans , Chromatin/genetics , Gene Expression Regulation , Leukemia, Myeloid, Acute/genetics , Transcription Factors/genetics , Transcription Factors/metabolism
4.
Blood ; 142(17): 1448-1462, 2023 10 26.
Article in English | MEDLINE | ID: mdl-37595278

ABSTRACT

Hematopoietic stem and progenitor cells (HSPCs) rely on a complex interplay among transcription factors (TFs) to regulate differentiation into mature blood cells. A heptad of TFs (FLI1, ERG, GATA2, RUNX1, TAL1, LYL1, LMO2) bind regulatory elements in bulk CD34+ HSPCs. However, whether specific heptad-TF combinations have distinct roles in regulating hematopoietic differentiation remains unknown. We mapped genome-wide chromatin contacts (HiC, H3K27ac, HiChIP), chromatin modifications (H3K4me3, H3K27ac, H3K27me3) and 10 TF binding profiles (heptad, PU.1, CTCF, STAG2) in HSPC subsets (stem/multipotent progenitors plus common myeloid, granulocyte macrophage, and megakaryocyte erythrocyte progenitors) and found TF occupancy and enhancer-promoter interactions varied significantly across cell types and were associated with cell-type-specific gene expression. Distinct regulatory elements were enriched with specific heptad-TF combinations, including stem-cell-specific elements with ERG, and myeloid- and erythroid-specific elements with combinations of FLI1, RUNX1, GATA2, TAL1, LYL1, and LMO2. Furthermore, heptad-occupied regions in HSPCs were subsequently bound by lineage-defining TFs, including PU.1 and GATA1, suggesting that heptad factors may prime regulatory elements for use in mature cell types. We also found that enhancers with cell-type-specific heptad occupancy shared a common grammar with respect to TF binding motifs, suggesting that combinatorial binding of TF complexes was at least partially regulated by features encoded in DNA sequence motifs. Taken together, this study comprehensively characterizes the gene regulatory landscape in rare subpopulations of human HSPCs. The accompanying data sets should serve as a valuable resource for understanding adult hematopoiesis and a framework for analyzing aberrant regulatory networks in leukemic cells.


Subject(s)
Core Binding Factor Alpha 2 Subunit , Hematopoietic Stem Cells , Humans , Core Binding Factor Alpha 2 Subunit/genetics , Core Binding Factor Alpha 2 Subunit/metabolism , Hematopoietic Stem Cells/metabolism , Gene Expression Regulation , Hematopoiesis/genetics , Chromatin/metabolism
5.
Haematologica ; 109(8): 2445-2458, 2024 08 01.
Article in English | MEDLINE | ID: mdl-38356460

ABSTRACT

ETV6::ACSL6 represents a rare genetic aberration in hematopoietic neoplasms and is often associated with severe eosinophilia, which confers an unfavorable prognosis requiring additional anti-inflammatory treatment. However, since the translocation is unlikely to produce a fusion protein, the mechanism of ETV6::ACSL6 action remains unclear. Here, we performed multi-omics analyses of primary leukemia cells and patient-derived xenografts from an acute lymphoblastic leukemia (ALL) patient with ETV6::ACSL6 translocation. We identified a super-enhancer located within the ETV6 gene locus, and revealed translocation and activation of the super-enhancer associated with the ETV6::ACSL6 fusion. The translocated super-enhancer exhibited intense interactions with genomic regions adjacent to and distal from the breakpoint at chromosomes 5 and 12, including genes coding inflammatory factors such as IL-3. This led to modulations in DNA methylation, histone modifications, and chromatin structures, triggering transcription of inflammatory factors leading to eosinophilia. Furthermore, the bromodomain and extraterminal domain (BET) inhibitor synergized with standard-of-care drugs for ALL, effectively reducing IL-3 expression and inhibiting ETV6::ACSL6 ALL growth in vitro and in vivo. Overall, our study revealed for the first time a cis-regulatory mechanism of super-enhancer translocation in ETV6::ACSL6ALL, leading to an ALL-accompanying clinical syndrome. These findings may stimulate novel treatment approaches for this challenging ALL subtype.


Subject(s)
ETS Translocation Variant 6 Protein , Enhancer Elements, Genetic , Eosinophilia , Interleukin-3 , Oncogene Proteins, Fusion , Precursor Cell Lymphoblastic Leukemia-Lymphoma , Proto-Oncogene Proteins c-ets , Repressor Proteins , Translocation, Genetic , Animals , Humans , Mice , Eosinophilia/genetics , Eosinophilia/metabolism , Eosinophilia/pathology , Gene Expression Regulation, Leukemic , Interleukin-3/genetics , Interleukin-3/metabolism , Oncogene Proteins, Fusion/genetics , Oncogene Proteins, Fusion/metabolism , Precursor Cell Lymphoblastic Leukemia-Lymphoma/genetics , Precursor Cell Lymphoblastic Leukemia-Lymphoma/pathology , Precursor Cell Lymphoblastic Leukemia-Lymphoma/metabolism , Proto-Oncogene Proteins c-ets/genetics , Proto-Oncogene Proteins c-ets/metabolism , Repressor Proteins/genetics , Repressor Proteins/metabolism
6.
Blood ; 138(16): 1441-1455, 2021 10 21.
Article in English | MEDLINE | ID: mdl-34075404

ABSTRACT

Changes in gene regulation and expression govern orderly transitions from hematopoietic stem cells to terminally differentiated blood cell types. These transitions are disrupted during leukemic transformation, but knowledge of the gene regulatory changes underpinning this process is elusive. We hypothesized that identifying core gene regulatory networks in healthy hematopoietic and leukemic cells could provide insights into network alterations that perturb cell state transitions. A heptad of transcription factors (LYL1, TAL1, LMO2, FLI1, ERG, GATA2, and RUNX1) bind key hematopoietic genes in human CD34+ hematopoietic stem and progenitor cells (HSPCs) and have prognostic significance in acute myeloid leukemia (AML). These factors also form a densely interconnected circuit by binding combinatorially at their own, and each other's, regulatory elements. However, their mutual regulation during normal hematopoiesis and in AML cells, and how perturbation of their expression levels influences cell fate decisions remains unclear. In this study, we integrated bulk and single-cell data and found that the fully connected heptad circuit identified in healthy HSPCs persists, with only minor alterations in AML, and that chromatin accessibility at key heptad regulatory elements was predictive of cell identity in both healthy progenitors and leukemic cells. The heptad factors GATA2, TAL1, and ERG formed an integrated subcircuit that regulates stem cell-to-erythroid transition in both healthy and leukemic cells. Components of this triad could be manipulated to facilitate erythroid transition providing a proof of concept that such regulatory circuits can be harnessed to promote specific cell-type transitions and overcome dysregulated hematopoiesis.


Subject(s)
GATA2 Transcription Factor/genetics , Gene Expression Regulation, Leukemic , Leukemia, Myeloid, Acute/genetics , T-Cell Acute Lymphocytic Leukemia Protein 1/genetics , Erythroid Cells/metabolism , Erythroid Cells/pathology , Gene Regulatory Networks , Hematopoiesis , Humans , Leukemia, Myeloid, Acute/pathology , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , Transcriptional Regulator ERG/genetics
7.
FASEB J ; 36(5): e22296, 2022 05.
Article in English | MEDLINE | ID: mdl-35363392

ABSTRACT

Metabolic reprogramming is a hallmark of cancer characterized by global changes in metabolite levels. However, compared with the study of gene expression, profiling of metabolites in cancer samples remains relatively understudied. We obtained metabolomic profiling and gene expression data from 454 human solid cancer cell lines across 24 cancer types from the Cancer Cell Line Encyclopedia (CCLE) database, to evaluate the feasibility of inferring metabolite levels from gene expression data. For each metabolite, we trained multivariable LASSO regression models to identify gene sets that are most predictive of the level of each metabolite profiled. Even when accounting for cell culture conditions or cell lineage in the model, few metabolites could be accurately predicted. In some cases, the inclusion of the upstream and downstream metabolites improved prediction accuracy, suggesting that gene expression is a poor predictor of steady-state metabolite levels. Our analysis uncovered a single robust relationship between the expression of nicotinamide N-methyltransferase (NNMT) and 1-methylnicotinamide (MNA), however, this relationship could only be validated in cancer samples with high purity, as NNMT is not expressed in immune cells. Together, we have trained models that use gene expression profiles to predict the level of individual metabolites. Our analysis suggests that inferring metabolite levels based on the expression of genes is generally challenging in cancer.


Subject(s)
Metabolomics , Neoplasms , Databases, Factual , Gene Expression , Humans , Neoplasms/genetics
8.
PLoS Genet ; 16(2): e1008572, 2020 02.
Article in English | MEDLINE | ID: mdl-32012149

ABSTRACT

Cancer genomes with mutations in the exonuclease domain of Polymerase Epsilon (POLE) present with an extraordinarily high somatic mutation burden. In vitro studies have shown that distinct POLE mutants exhibit different polymerase activity. Yet, genome-wide mutation patterns and driver mutation formation arising from different POLE mutants remains unclear. Here, we curated somatic mutation calls from 7,345 colorectal cancer samples from published studies and publicly available databases. These include 44 POLE mutant samples including 9 with whole genome sequencing data available. The POLE mutant samples were categorized based on the specific POLE mutation present. Mutation spectrum, associations of somatic mutations with epigenomics features and co-occurrence with specific driver mutations were examined across different POLE mutants. We found that different POLE mutants exhibit distinct mutation spectrum with significantly higher relative frequency of C>T mutations in POLE V411L mutants. Our analysis showed that this increase frequency in C>T mutations is not dependent on DNA methylation and not associated with other genomic features and is thus specifically due to DNA sequence context alone. Notably, we found strong association of the TP53 R213* mutation specifically with POLE P286R mutants. This truncation mutation occurs within the TT[C>T]GA context. For C>T mutations, this sequence context is significantly more likely to be mutated in POLE P286R mutants compared with other POLE exonuclease domain mutants. This study refines our understanding of DNA polymerase fidelity and underscores genome-wide mutation spectrum and specific cancer driver mutation formation observed in POLE mutant cancers.


Subject(s)
Carcinogenesis/genetics , Colorectal Neoplasms/genetics , DNA Polymerase II/metabolism , Poly-ADP-Ribose Binding Proteins/metabolism , Protein Domains/genetics , Tumor Suppressor Protein p53/genetics , CpG Islands/genetics , Cytosine/metabolism , DNA Methylation/genetics , DNA Mutational Analysis/statistics & numerical data , DNA Polymerase II/genetics , Databases, Genetic/statistics & numerical data , Datasets as Topic , Epigenesis, Genetic , Humans , Mutation , Poly-ADP-Ribose Binding Proteins/genetics , Whole Genome Sequencing/statistics & numerical data
9.
Nature ; 532(7598): 259-63, 2016 Apr 14.
Article in English | MEDLINE | ID: mdl-27075100

ABSTRACT

Promoters are DNA sequences that have an essential role in controlling gene expression. While recent whole cancer genome analyses have identified numerous hotspots of somatic point mutations within promoters, many have not yet been shown to perturb gene expression or drive cancer development. As such, positive selection alone may not adequately explain the frequency of promoter point mutations in cancer genomes. Here we show that increased mutation density at gene promoters can be linked to promoter activity and differential nucleotide excision repair (NER). By analysing 1,161 human cancer genomes across 14 cancer types, we find evidence for increased local density of somatic point mutations within the centres of DNase I-hypersensitive sites (DHSs) in gene promoters. Mutated DHSs were strongly associated with transcription initiation activity, in which active promoters but not enhancers of equal DNase I hypersensitivity were most mutated relative to their flanking regions. Notably, analysis of genome-wide maps of NER shows that NER is impaired within the DHS centre of active gene promoters, while XPC-deficient skin cancers do not show increased promoter mutation density, pinpointing differential NER as the underlying cause of these mutation hotspots. Consistent with this finding, we observe that melanomas with an ultraviolet-induced DNA damage mutation signature show greatest enrichment of promoter mutations, whereas cancers that are not highly dependent on NER, such as colon cancer, show no sign of such enrichment. Taken together, our analysis has uncovered the presence of a previously unknown mechanism linking transcription initiation and NER as a major contributor of somatic point mutation hotspots at active gene promoters in cancer genomes.


Subject(s)
DNA Repair/genetics , Genome, Human/genetics , Mutagenesis/genetics , Mutation Rate , Neoplasms/genetics , Promoter Regions, Genetic/genetics , Transcription Initiation, Genetic , Colonic Neoplasms/genetics , DNA Damage/genetics , DNA Repair/radiation effects , Deoxyribonuclease I/metabolism , Enhancer Elements, Genetic/genetics , Gene Expression Regulation, Neoplastic/genetics , Humans , Lung Neoplasms/genetics , Melanoma/genetics , Point Mutation/genetics , Ultraviolet Rays
10.
Br J Haematol ; 193(4): 841-844, 2021 05.
Article in English | MEDLINE | ID: mdl-33481245

ABSTRACT

Clonal haematopoiesis of indeterminant potential (CHIP) increases in frequency with age. The effect of CHIP on the mobilization of autologous CD34+ peripheral blood stem cells (PBSC) has not been reported. This study uses a DNA-based targeted candidate gene approach to identify the presence of somatic mutations in ASXL1, DNMT3A, JAK2, SF3B1, TET2 and TP53 in CD34+ haematopoietic progenitor cell-apheresis products of 96 patients who undergo PBSC mobilization for autologous stem cell transplantation (ASCT). Variants were identified in a significantly greater proportion of patients who experience poor CD34+ PBSC mobilization. A DNA-based targeted candidate gene array is able to predict poor CD34+ PBSC mobilization and may be deployed pre-emptively to minimize mobilization and graft failures.


Subject(s)
Clonal Hematopoiesis/genetics , Hematopoietic Stem Cell Mobilization , Mutation , Oligonucleotide Array Sequence Analysis , Peripheral Blood Stem Cell Transplantation , Peripheral Blood Stem Cells , Adult , Age Factors , Aged , Autografts , Female , Humans , Male , Middle Aged
11.
Expert Rev Proteomics ; 18(3): 221-232, 2021 03.
Article in English | MEDLINE | ID: mdl-33877947

ABSTRACT

Introduction: Cancer cell lines (CCLs) have been a major resource for cancer research. Over the past couple of decades, they have been instrumental in omic profiling method development and as model systems to generate new knowledge in cell and cancer biology. More recently, with the increasing amount of genomic, transcriptomic and proteomic data being generated in hundreds of CCLs, there is growing potential for integrative proteogenomic data analyses to be performed.Areas covered: In this review, we first describe the most commonly used proteome profiling methods in CCLs. We then discuss how these proteomics data can be integrated with genomics data for proteogenomics analyses. Finally, we highlight some of the recent biological discoveries that have arisen from proteogenomics analyses of CCLs.Expert opinion: Protegeonomics analyses of CCLs have so far enabled the discovery of novel proteins and proteoforms. It has also improved our understanding of biological processes including post-transcriptional regulation of protein abundance and the presentation of antigens by major histocompatibility complex alleles. With proteomics data to be generated in hundreds to thousands of CCLs in coming years, there will be further potential for large-scale proteogenomics analyses and data integration with the phenotypically well-characterized CCLs.


Subject(s)
Neoplasms , Proteogenomics , Cell Line , Genomics , Humans , Neoplasms/genetics , Proteomics
12.
Rev Endocr Metab Disord ; 22(4): 1121-1136, 2021 12.
Article in English | MEDLINE | ID: mdl-34272645

ABSTRACT

Developed as an antidiabetic drug, recent evidence suggests that several sodium-glucose co-transporter 2 inhibitors (SGLT2i), especially canagliflozin and dapagliflozin, may exhibit in vitro and in vivo anticancer activities in selected cancer types, including an inhibition of tumor growth and induction of cell death. When used in combination with chemotherapy or radiotherapy, SGLT2i may offer possible synergistic effects in enhancing their treatment efficacy while alleviating associated side effects. Potential mechanisms include a reduction of glucose uptake into cancer cells, systemic glucose restriction, modulation of multiple signaling pathways, and regulation of different gene and protein expression. Furthermore, preliminary clinical findings have reported potential anticancer properties of canagliflozin and dapagliflozin in patients with liver and colon cancers respectively, with reference to decreases in their tumor marker levels. Given its general tolerability and routine use in diabetes management, SGLT2i may be a good candidate for drug repurposing in cancer treatment and as adjunct to conventional therapies. While current evidence reveals that only certain SGLT2i appear to be effective against selected cancer types, further studies are needed to explore the antitumor abilities of each SGLT2i in various cancers. Moreover, clinical trials are called for to evaluate the safety and feasibility of introducing SGLT2i in the treatment regimen of patients with specific cancers, and to identify the preferred route of drug administration for targeted delivery to selected tumor sites.


Subject(s)
Diabetes Mellitus, Type 2 , Neoplasms , Sodium-Glucose Transporter 2 Inhibitors , Symporters , Diabetes Mellitus, Type 2/drug therapy , Drug Repositioning , Glucose , Humans , Neoplasms/drug therapy , Sodium/therapeutic use , Sodium-Glucose Transporter 2 Inhibitors/adverse effects , Symporters/therapeutic use
13.
PLoS Genet ; 14(11): e1007779, 2018 11.
Article in English | MEDLINE | ID: mdl-30412573

ABSTRACT

Driver mutations are the genetic variants responsible for oncogenesis, but how specific somatic mutational events arise in cells remains poorly understood. Mutational signatures derive from the frequency of mutated trinucleotides in a given cancer sample, and they provide an avenue for investigating the underlying mutational processes that operate in cancer. Here we analyse somatic mutations from 7,815 cancer exomes from The Cancer Genome Atlas (TCGA) across 26 cancer types. We curate a list of 50 known cancer driver mutations by analysing recurrence in our cohort and annotations of known cancer-associated genes from the Cancer Gene Census, IntOGen database and Cancer Genome Interpreter. We then use these datasets to perform binary univariate logistic regression and establish the statistical relationship between individual driver mutations and known mutational signatures across different cancer types. Our analysis led to the identification of 39 significant associations between driver mutations and mutational signatures (P < 0.004, with a false discovery rate of < 5%). We first validate our methodology by establishing statistical links for known and novel associations between driver mutations and the mutational signature arising from Polymerase Epsilon proofreading deficiency. We then examine associations between driver mutations and mutational signatures for AID/APOBEC enzyme activity and deficient mismatch repair. We also identify negative associations (odds ratio < 1) between mutational signatures and driver mutations, and here we examine the role of aging and cigarette smoke mutagenesis in the generation of driver mutations in IDH1 and KRAS in brain cancers and lung adenocarcinomas respectively. Our study provides statistical foundations for hypothesised links between otherwise independent biological processes and we uncover previously unexplored relationships between driver mutations and mutagenic processes during cancer development. These associations give insights into how cancers acquire advantageous mutations and can provide direction to guide further mechanistic studies into cancer pathogenesis.


Subject(s)
Exome , Mutation , Neoplasms/genetics , Brain Neoplasms/genetics , Carcinogenesis/genetics , Cohort Studies , Colorectal Neoplasms/genetics , DNA Mutational Analysis , DNA Replication/genetics , DNA, Neoplasm/genetics , Databases, Genetic , Female , Genome, Human , Humans , Logistic Models , Male , Models, Genetic , Mutagenesis , Neoplastic Syndromes, Hereditary/genetics , Oncogenes , Proto-Oncogene Proteins B-raf/genetics , Exome Sequencing
14.
Gut ; 69(12): 2165-2179, 2020 12.
Article in English | MEDLINE | ID: mdl-32217638

ABSTRACT

OBJECTIVE: Sporadic early-onset colorectal cancer (EOCRC) has bad prognosis, yet is poorly represented by cell line models. We examine the key mutational and transcriptomic alterations in an organoid biobank enriched in EOCRCs. DESIGN: We established paired cancer (n=32) and normal organoids (n=18) from 20 patients enriched in microsatellite-stable EOCRC. Exome and transcriptome analysis was performed. RESULTS: We observed a striking diversity of molecular phenotypes, including PTPRK-RSPO3 fusions. Transcriptionally, RSPO fusion organoids resembled normal colon organoids and were distinct from APC mutant organoids, with high BMP2 and low PTK7 expression. Single cell transcriptome analysis confirmed the similarity between RSPO fusion organoids and normal organoids, with a propensity for maturation on Wnt withdrawal, whereas the APC mutant organoids were locked in progenitor stages. CRISPR/Cas9 engineered mutation of APC in normal human colon organoids led to upregulation of PTK7 protein and suppression of BMP2, but less so with an engineered RNF43 mutation. The frequent co-occurrence of RSPO fusions with SMAD4 or BMPR1A mutation was confirmed in TCGA database searches. RNF43 mutation was found in organoid from a leukaemia survivor with a novel mutational signature; and organoids with POLE proofreading mutation displayed ultramutation. The cancer organoid genomes were stable over long culture periods, while normal human colon organoids tended to be subject to clonal dominance over time. CONCLUSIONS: These organoid models enriched in EOCRCs with linked genomic data fill a gap in existing CRC models and reveal distinct genetic profiles and novel pathway cooperativity.


Subject(s)
Colorectal Neoplasms/genetics , Genetic Profile , Organoids/pathology , Adenomatous Polyposis Coli Protein/genetics , Bone Morphogenetic Protein 2/genetics , Bone Morphogenetic Protein Receptors, Type I/genetics , CRISPR-Cas Systems , Cell Adhesion Molecules/genetics , Gene Expression Profiling , Gene Fusion , Humans , Models, Genetic , Mutation , Receptor Protein-Tyrosine Kinases/genetics , Receptor-Like Protein Tyrosine Phosphatases, Class 2/genetics , Smad4 Protein/genetics , Thrombospondins/genetics , Tissue Banks , Ubiquitin-Protein Ligases/genetics , Up-Regulation , Exome Sequencing
15.
Nucleic Acids Res ; 45(13): 7786-7795, 2017 Jul 27.
Article in English | MEDLINE | ID: mdl-28531315

ABSTRACT

Methylated cytosines (5mCs) are frequently mutated in the genome. However, no studies have yet comprehensively analysed mutation-methylation associations across cancer types. Here we analyse 916 cancer genomes, together with tissue type-specific methylation and replication timing data. We describe a strong mutation-methylation association across colorectal cancer subtypes, most interestingly in samples with microsatellite instability (MSI) or Polymerase epsilon (POLE) exonuclease domain mutations. By analysing genomic regions with differential mismatch repair (MMR) efficiency, we suggest a possible role for MMR in the correction of 5mC deamination events, potentially accounting for the high rate of 5mC mutation accumulation in MSI tumours. Additionally, we propose that mutant POLE asserts a mutator phenotype specifically at 5mCs, and we find coding mutation hotspots in POLE-mutant cancers at highly-methylated CpGs in the tumour-suppressor genes APC and TP53. Finally, using multivariable regression models, we demonstrate that different cancers exhibit distinct mutation-methylation associations, with DNA repair influencing such associations in certain cancer genomes. Taken together, we find differential associations with methylation that are vital for accurately predicting expected mutation loads across cancer types. Our findings reveal links between methylation and common mutation and repair processes, with these mechanisms defining a key part of the mutational landscape of cancer genomes.


Subject(s)
DNA Methylation/genetics , DNA Replication/genetics , Genes, Neoplasm , Mutation , Neoplasms/genetics , Neoplasms/metabolism , 5-Methylcytosine/metabolism , Colorectal Neoplasms/genetics , Colorectal Neoplasms/metabolism , CpG Islands , DNA Mismatch Repair , DNA Polymerase II/genetics , DNA Polymerase II/metabolism , DNA Repair , Genes, APC , Genes, p53 , Genome, Human , Humans , Microsatellite Instability , Poly-ADP-Ribose Binding Proteins
16.
Proc Natl Acad Sci U S A ; 113(16): E2306-15, 2016 Apr 19.
Article in English | MEDLINE | ID: mdl-27044077

ABSTRACT

Current approaches in tissue engineering are geared toward generating tissue-specific stem cells. Given the complexity and heterogeneity of tissues, this approach has its limitations. An alternate approach is to induce terminally differentiated cells to dedifferentiate into multipotent proliferative cells with the capacity to regenerate all components of a damaged tissue, a phenomenon used by salamanders to regenerate limbs. 5-Azacytidine (AZA) is a nucleoside analog that is used to treat preleukemic and leukemic blood disorders. AZA is also known to induce cell plasticity. We hypothesized that AZA-induced cell plasticity occurs via a transient multipotent cell state and that concomitant exposure to a receptive growth factor might result in the expansion of a plastic and proliferative population of cells. To this end, we treated lineage-committed cells with AZA and screened a number of different growth factors with known activity in mesenchyme-derived tissues. Here, we report that transient treatment with AZA in combination with platelet-derived growth factor-AB converts primary somatic cells into tissue-regenerative multipotent stem (iMS) cells. iMS cells possess a distinct transcriptome, are immunosuppressive, and demonstrate long-term self-renewal, serial clonogenicity, and multigerm layer differentiation potential. Importantly, unlike mesenchymal stem cells, iMS cells contribute directly to in vivo tissue regeneration in a context-dependent manner and, unlike embryonic or pluripotent stem cells, do not form teratomas. Taken together, this vector-free method of generating iMS cells from primary terminally differentiated cells has significant scope for application in tissue regeneration.


Subject(s)
Azacitidine/pharmacology , Cellular Reprogramming , Induced Pluripotent Stem Cells/metabolism , Mesenchymal Stem Cells/metabolism , Platelet-Derived Growth Factor/pharmacology , Animals , Cells, Cultured , Induced Pluripotent Stem Cells/cytology , Mesenchymal Stem Cells/cytology , Mice , Mice, Transgenic , Organ Specificity/physiology
17.
Nucleic Acids Res ; 44(22): 10644-10661, 2016 12 15.
Article in English | MEDLINE | ID: mdl-27604872

ABSTRACT

Aberrant stem cell-like gene regulatory networks are a feature of leukaemogenesis. The ETS-related gene (ERG), an important regulator of normal haematopoiesis, is also highly expressed in T-ALL and acute myeloid leukaemia (AML). However, the transcriptional regulation of ERG in leukaemic cells remains poorly understood. In order to discover transcriptional regulators of ERG, we employed a quantitative mass spectrometry-based method to identify factors binding the 321 bp ERG +85 stem cell enhancer region in MOLT-4 T-ALL and KG-1 AML cells. Using this approach, we identified a number of known binders of the +85 enhancer in leukaemic cells along with previously unknown binders, including ETV6 and IKZF1. We confirmed that ETV6 and IKZF1 were also bound at the +85 enhancer in both leukaemic cells and in healthy human CD34+ haematopoietic stem and progenitor cells. Knockdown experiments confirmed that ETV6 and IKZF1 are transcriptional regulators not just of ERG, but also of a number of genes regulated by a densely interconnected network of seven transcription factors. At last, we show that ETV6 and IKZF1 expression levels are positively correlated with expression of a number of heptad genes in AML and high expression of all nine genes confers poorer overall prognosis.


Subject(s)
Ikaros Transcription Factor/physiology , Proto-Oncogene Proteins c-ets/physiology , Repressor Proteins/physiology , Transcription, Genetic , Base Sequence , Binding Sites , Cell Line, Tumor , Consensus Sequence , Enhancer Elements, Genetic , Gene Expression Regulation, Leukemic , Gene Regulatory Networks , Humans , Kaplan-Meier Estimate , Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/metabolism , Leukemia, Myeloid, Acute/mortality , Prognosis , Proportional Hazards Models , Protein Binding , Proteome , Proteomics , Transcriptional Regulator ERG/physiology , ETS Translocation Variant 6 Protein
18.
Blood ; 125(2): 273-83, 2015 Jan 08.
Article in English | MEDLINE | ID: mdl-25336632

ABSTRACT

Glucocorticoids are critical components of combination chemotherapy regimens in pediatric acute lymphoblastic leukemia (ALL). The proapoptotic BIM protein is an important mediator of glucocorticoid-induced apoptosis in normal and malignant lymphocytes, whereas the antiapoptotic BCL2 confers resistance. The signaling pathways regulating BIM and BCL2 expression in glucocorticoid-treated lymphoid cells remain unclear. In this study, pediatric ALL patient-derived xenografts (PDXs) inherently sensitive or resistant to glucocorticoids were exposed to dexamethasone in vivo. Microarray analysis showed that KLF13 and MYB gene expression changes were significantly greater in dexamethasone-sensitive than -resistant PDXs. Chromatin immunoprecipitation (ChIP) analysis detected glucocorticoid receptor (GR) binding at the KLF13 promoter to trigger KLF13 expression only in sensitive PDXs. Next, KLF13 bound to the MYB promoter, deactivating MYB expression only in sensitive PDXs. Sustained MYB expression in resistant PDXs resulted in maintenance of BCL2 expression and inhibition of apoptosis. ChIP sequencing analysis revealed a novel GR binding site in a BIM intronic region (IGR) that was engaged only in dexamethasone-sensitive PDXs. The absence of GR binding at the BIM IGR was associated with BIM silencing and dexamethasone resistance. This study has identified novel mechanisms of opposing BCL2 and BIM gene regulation that control glucocorticoid-induced apoptosis in pediatric ALL cells in vivo.


Subject(s)
Apoptosis Regulatory Proteins/genetics , Apoptosis/genetics , Membrane Proteins/genetics , Precursor Cell Lymphoblastic Leukemia-Lymphoma/genetics , Proto-Oncogene Proteins c-bcl-2/genetics , Proto-Oncogene Proteins/genetics , Receptors, Glucocorticoid/genetics , Animals , Antineoplastic Agents, Hormonal/pharmacology , Apoptosis/drug effects , Apoptosis/physiology , Apoptosis Regulatory Proteins/biosynthesis , Bcl-2-Like Protein 11 , Blotting, Western , Chromatin Immunoprecipitation , Dexamethasone/pharmacology , Drug Resistance, Neoplasm/genetics , Enzyme-Linked Immunosorbent Assay , Gene Expression Regulation, Neoplastic/physiology , Humans , Membrane Proteins/biosynthesis , Mice , Mice, Inbred NOD , Mice, SCID , Oligonucleotide Array Sequence Analysis , Precursor Cell Lymphoblastic Leukemia-Lymphoma/metabolism , Proto-Oncogene Proteins/biosynthesis , Proto-Oncogene Proteins c-bcl-2/biosynthesis , Real-Time Polymerase Chain Reaction , Receptors, Glucocorticoid/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction/drug effects , Xenograft Model Antitumor Assays
19.
Blood ; 123(13): 2000-7, 2014 Mar 27.
Article in English | MEDLINE | ID: mdl-24523239

ABSTRACT

Most proteins in nature are chemically modified after they are made to control how, when, and where they function. The 3 core features of proteins are posttranslationally modified: amino acid side chains can be modified, peptide bonds can be cleaved or isomerized, and disulfide bonds can be cleaved. Cleavage of peptide bonds is a major mechanism of protein control in the circulation, as exemplified by activation of the blood coagulation and complement zymogens. Cleavage of disulfide bonds is emerging as another important mechanism of protein control in the circulation. Recent advances in our understanding of control of soluble blood proteins and blood cell receptors by functional disulfide bonds is discussed as is how these bonds are being identified and studied.


Subject(s)
Allosteric Regulation/physiology , Blood Proteins/chemistry , Blood Proteins/metabolism , Disulfides/chemistry , Angiotensinogen/chemistry , Angiotensinogen/metabolism , Animals , Disulfides/metabolism , Humans , Hydrogen Bonding , Interleukin Receptor Common gamma Subunit/chemistry , Interleukin Receptor Common gamma Subunit/metabolism , Plasminogen/chemistry , Plasminogen/metabolism , beta 2-Glycoprotein I/chemistry , beta 2-Glycoprotein I/metabolism
20.
Nucleic Acids Res ; 42(Database issue): D172-7, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24185696

ABSTRACT

The BloodChIP database (http://www.med.unsw.edu.au/CRCWeb.nsf/page/BloodChIP) supports exploration and visualization of combinatorial transcription factor (TF) binding at a particular locus in human CD34-positive and other normal and leukaemic cells or retrieval of target gene sets for user-defined combinations of TFs across one or more cell types. Increasing numbers of genome-wide TF binding profiles are being added to public repositories, and this trend is likely to continue. For the power of these data sets to be fully harnessed by experimental scientists, there is a need for these data to be placed in context and easily accessible for downstream applications. To this end, we have built a user-friendly database that has at its core the genome-wide binding profiles of seven key haematopoietic TFs in human stem/progenitor cells. These binding profiles are compared with binding profiles in normal differentiated and leukaemic cells. We have integrated these TF binding profiles with chromatin marks and expression data in normal and leukaemic cell fractions. All queries can be exported into external sites to construct TF-gene and protein-protein networks and to evaluate the association of genes with cellular processes and tissue expression.


Subject(s)
Blood Cells/metabolism , Databases, Genetic , Regulatory Elements, Transcriptional , Transcription Factors/metabolism , Binding Sites , Chromatin Immunoprecipitation , DNA/chemistry , DNA/metabolism , Gene Expression Profiling , Genome, Human , Hematopoietic Stem Cells/metabolism , Humans , Internet , Software , Transcription Factors/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL