Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 29
Filter
1.
Nature ; 615(7952): 499-506, 2023 03.
Article in English | MEDLINE | ID: mdl-36890229

ABSTRACT

Mutations in fumarate hydratase (FH) cause hereditary leiomyomatosis and renal cell carcinoma1. Loss of FH in the kidney elicits several oncogenic signalling cascades through the accumulation of the oncometabolite fumarate2. However, although the long-term consequences of FH loss have been described, the acute response has not so far been investigated. Here we generated an inducible mouse model to study the chronology of FH loss in the kidney. We show that loss of FH leads to early alterations of mitochondrial morphology and the release of mitochondrial DNA (mtDNA) into the cytosol, where it triggers the activation of the cyclic GMP-AMP synthase (cGAS)-stimulator of interferon genes (STING)-TANK-binding kinase 1 (TBK1) pathway and stimulates an inflammatory response that is also partially dependent on retinoic-acid-inducible gene I (RIG-I). Mechanistically, we show that this phenotype is mediated by fumarate and occurs selectively through mitochondrial-derived vesicles in a manner that depends on sorting nexin 9 (SNX9). These results reveal that increased levels of intracellular fumarate induce a remodelling of the mitochondrial network and the generation of mitochondrial-derived vesicles, which allows the release of mtDNAin the cytosol and subsequent activation of the innate immune response.


Subject(s)
DNA, Mitochondrial , Fumarates , Immunity, Innate , Mitochondria , Animals , Mice , DNA, Mitochondrial/metabolism , Fumarate Hydratase/genetics , Fumarate Hydratase/metabolism , Fumarates/metabolism , Mitochondria/enzymology , Mitochondria/genetics , Mitochondria/metabolism , Mitochondria/pathology , Kidney/enzymology , Kidney/metabolism , Kidney/pathology , Cytosol/metabolism
2.
Mol Cell ; 81(19): 4059-4075.e11, 2021 10 07.
Article in English | MEDLINE | ID: mdl-34437837

ABSTRACT

DDX3X is a ubiquitously expressed RNA helicase involved in multiple stages of RNA biogenesis. DDX3X is frequently mutated in Burkitt lymphoma, but the functional basis for this is unknown. Here, we show that loss-of-function DDX3X mutations are also enriched in MYC-translocated diffuse large B cell lymphoma and reveal functional cooperation between mutant DDX3X and MYC. DDX3X promotes the translation of mRNA encoding components of the core translational machinery, thereby driving global protein synthesis. Loss-of-function DDX3X mutations moderate MYC-driven global protein synthesis, thereby buffering MYC-induced proteotoxic stress during early lymphomagenesis. Established lymphoma cells restore full protein synthetic capacity by aberrant expression of DDX3Y, a Y chromosome homolog, the expression of which is normally restricted to the testis. These findings show that DDX3X loss of function can buffer MYC-driven proteotoxic stress and highlight the capacity of male B cell lymphomas to then compensate for this loss by ectopic DDX3Y expression.


Subject(s)
B-Lymphocytes/enzymology , DEAD-box RNA Helicases/metabolism , Lymphoma, B-Cell/enzymology , Minor Histocompatibility Antigens/metabolism , Neoplasm Proteins/biosynthesis , Proto-Oncogene Proteins c-myc/metabolism , Adolescent , Adult , Aged , Aged, 80 and over , Animals , B-Lymphocytes/pathology , Cell Line, Tumor , Child , Child, Preschool , DEAD-box RNA Helicases/genetics , Endoplasmic Reticulum Stress , Female , Gene Expression Regulation, Enzymologic , Gene Expression Regulation, Neoplastic , Humans , Loss of Function Mutation , Lymphoma, B-Cell/genetics , Lymphoma, B-Cell/pathology , Male , Mice, Transgenic , Middle Aged , Minor Histocompatibility Antigens/genetics , Neoplasm Proteins/genetics , Protein Biosynthesis , Proteome , Proteostasis , Proto-Oncogene Proteins c-myc/genetics , Young Adult
3.
Nature ; 606(7916): 999-1006, 2022 06.
Article in English | MEDLINE | ID: mdl-35676472

ABSTRACT

Large-scale human genetic data1-3 have shown that cancer mutations display strong tissue-selectivity, but how this selectivity arises remains unclear. Here, using experimental models, functional genomics and analyses of patient samples, we demonstrate that the lineage transcription factor paired box 8 (PAX8) is required for oncogenic signalling by two common genetic alterations that cause clear cell renal cell carcinoma (ccRCC) in humans: the germline variant rs7948643 at 11q13.3 and somatic inactivation of the von Hippel-Lindau tumour suppressor (VHL)4-6. VHL loss, which is observed in about 90% of ccRCCs, can lead to hypoxia-inducible factor 2α (HIF2A) stabilization6,7. We show that HIF2A is preferentially recruited to PAX8-bound transcriptional enhancers, including a pro-tumorigenic cyclin D1 (CCND1) enhancer that is controlled by PAX8 and HIF2A. The ccRCC-protective allele C at rs7948643 inhibits PAX8 binding at this enhancer and downstream activation of CCND1 expression. Co-option of a PAX8-dependent physiological programme that supports the proliferation of normal renal epithelial cells is also required for MYC expression from the ccRCC metastasis-associated amplicons at 8q21.3-q24.3 (ref. 8). These results demonstrate that transcriptional lineage factors are essential for oncogenic signalling and that they mediate tissue-specific cancer risk associated with somatic and inherited genetic variants.


Subject(s)
Carcinogenesis , Kidney Neoplasms , PAX8 Transcription Factor , Signal Transduction , Alleles , Basic Helix-Loop-Helix Transcription Factors/metabolism , Carcinogenesis/genetics , Carcinoma, Renal Cell/metabolism , Carcinoma, Renal Cell/pathology , Cyclin D1/genetics , Gene Expression Regulation, Neoplastic , Humans , Kidney/metabolism , Kidney/pathology , Kidney Neoplasms/metabolism , Kidney Neoplasms/pathology , Mutation , PAX8 Transcription Factor/genetics , PAX8 Transcription Factor/metabolism , Proto-Oncogene Proteins c-myc/genetics , Von Hippel-Lindau Tumor Suppressor Protein/genetics
4.
Genes Dev ; 31(13): 1339-1353, 2017 07 01.
Article in English | MEDLINE | ID: mdl-28790158

ABSTRACT

Lung adenocarcinoma accounts for ∼40% of lung cancers, the leading cause of cancer-related death worldwide, and current therapies provide only limited survival benefit. Approximately half of lung adenocarcinomas harbor mutations in TP53 (p53), making these mutants appealing targets for lung cancer therapy. As mutant p53 remains untargetable, mutant p53-dependent phenotypes represent alternative targeting opportunities, but the prevalence and therapeutic relevance of such effects (gain of function and dominant-negative activity) in lung adenocarcinoma are unclear. Through transcriptional and functional analysis of murine KrasG12D -p53null , -p53R172H (conformational), and -p53R270H (contact) mutant lung tumors, we identified genotype-independent and genotype-dependent therapeutic sensitivities. Unexpectedly, we found that wild-type p53 exerts a dominant tumor-suppressive effect on mutant tumors, as all genotypes were similarly sensitive to its restoration in vivo. These data show that the potential of p53 targeted therapies is comparable across all p53-deficient genotypes and may explain the high incidence of p53 loss of heterozygosity in mutant tumors. In contrast, mutant p53 gain of function and their associated vulnerabilities can vary according to mutation type. Notably, we identified a p53R270H -specific sensitivity to simvastatin in lung tumors, and the transcriptional signature that underlies this sensitivity was also present in human lung tumors, indicating that this therapeutic approach may be clinically relevant.


Subject(s)
Adenocarcinoma/drug therapy , Adenocarcinoma/genetics , Lung Neoplasms/drug therapy , Lung Neoplasms/genetics , Simvastatin/therapeutic use , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism , Adenocarcinoma of Lung , Animals , Antineoplastic Agents/pharmacology , Cell Cycle Checkpoints/genetics , Cell Death/genetics , Cell Line, Tumor , Disease Models, Animal , Drug Resistance, Neoplasm/genetics , Gene Expression Regulation, Neoplastic/drug effects , Gene Expression Regulation, Neoplastic/genetics , Gene Expression Regulation, Neoplastic/radiation effects , Genotype , Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacology , Imidazoles/pharmacology , Mice , Molecular Targeted Therapy , Mutation , Piperazines/pharmacology , Simvastatin/pharmacology
5.
Blood ; 138(11): 959-964, 2021 09 16.
Article in English | MEDLINE | ID: mdl-33988691

ABSTRACT

Serum and glucocorticoid-regulated kinase 1 (SGK1) is one of the most frequently mutated genes in diffuse large B-cell lymphoma (DLBCL). However, little is known about its function or the consequence of its mutation. The frequent finding of truncating mutations has led to the widespread assumption that these represent loss-of-function variants and, accordingly, that SGK1 must act as a tumor suppressor. In this study, instead, the most common SGK1 mutations led to production of aberrantly spliced messenger RNA neoisoforms in which translation is initiated from downstream methionines. The resulting N-terminal truncated protein isoforms showed increased expression related to the exclusion of an N-terminal degradation domain. However, they retained a functional kinase domain, the overexpression of which rendered cells resistant to AKT inhibition, in part because of increased phosphorylation of GSK3B. These findings challenge the prevailing assumption that SGK1 is a tumor-suppressor gene in DLBCL and provide the impetus to explore further the pharmacological inhibition of SGK1 as a therapeutic strategy for DLBCL.


Subject(s)
Immediate-Early Proteins/genetics , Lymphoma, Large B-Cell, Diffuse/genetics , Protein Serine-Threonine Kinases/genetics , Proto-Oncogene Proteins c-akt/metabolism , Cells, Cultured , Enzyme Stability , Humans , Immediate-Early Proteins/chemistry , Immediate-Early Proteins/metabolism , Lymphoma, Large B-Cell, Diffuse/metabolism , Phosphorylation , Protein Domains , Protein Isoforms/chemistry , Protein Isoforms/genetics , Protein Isoforms/metabolism , Protein Serine-Threonine Kinases/chemistry , Protein Serine-Threonine Kinases/metabolism
6.
Mol Cell ; 47(2): 203-14, 2012 Jul 27.
Article in English | MEDLINE | ID: mdl-22795131

ABSTRACT

The expansion of repressive epigenetic marks has been implicated in heterochromatin formation during embryonic development, but the general applicability of this mechanism is unclear. Here we show that nuclear rearrangement of repressive histone marks H3K9me3 and H3K27me3 into nonoverlapping structural layers characterizes senescence-associated heterochromatic foci (SAHF) formation in human fibroblasts. However, the global landscape of these repressive marks remains unchanged upon SAHF formation, suggesting that in somatic cells, heterochromatin can be formed through the spatial repositioning of pre-existing repressively marked histones. This model is reinforced by the correlation of presenescent replication timing with both the subsequent layered structure of SAHFs and the global landscape of the repressive marks, allowing us to integrate microscopic and genomic information. Furthermore, modulation of SAHF structure does not affect the occupancy of these repressive marks, nor vice versa. These experiments reveal that high-order heterochromatin formation and epigenetic remodeling of the genome can be discrete events.


Subject(s)
Chromatin/chemistry , Heterochromatin/chemistry , Histones/metabolism , Bromodeoxyuridine/pharmacology , Cellular Senescence , Chromosomes/ultrastructure , Epigenesis, Genetic , Fibroblasts/cytology , Gene Expression Regulation, Developmental , Gene Silencing , Genome , Genome-Wide Association Study , Histones/chemistry , Humans , Laser Scanning Cytometry/methods , Microscopy, Fluorescence/methods
7.
Nature ; 486(7403): 346-52, 2012 Apr 18.
Article in English | MEDLINE | ID: mdl-22522925

ABSTRACT

The elucidation of breast cancer subgroups and their molecular drivers requires integrated views of the genome and transcriptome from representative numbers of patients. We present an integrated analysis of copy number and gene expression in a discovery and validation set of 997 and 995 primary breast tumours, respectively, with long-term clinical follow-up. Inherited variants (copy number variants and single nucleotide polymorphisms) and acquired somatic copy number aberrations (CNAs) were associated with expression in ~40% of genes, with the landscape dominated by cis- and trans-acting CNAs. By delineating expression outlier genes driven in cis by CNAs, we identified putative cancer genes, including deletions in PPP2R2A, MTAP and MAP2K4. Unsupervised analysis of paired DNA­RNA profiles revealed novel subgroups with distinct clinical outcomes, which reproduced in the validation cohort. These include a high-risk, oestrogen-receptor-positive 11q13/14 cis-acting subgroup and a favourable prognosis subgroup devoid of CNAs. Trans-acting aberration hotspots were found to modulate subgroup-specific gene networks, including a TCR deletion-mediated adaptive immune response in the 'CNA-devoid' subgroup and a basal-specific chromosome 5 deletion-associated mitotic network. Our results provide a novel molecular stratification of the breast cancer population, derived from the impact of somatic CNAs on the transcriptome.


Subject(s)
Breast Neoplasms/genetics , Breast Neoplasms/pathology , DNA Copy Number Variations/genetics , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Genome, Human/genetics , Breast Neoplasms/classification , Breast Neoplasms/diagnosis , Female , Gene Regulatory Networks/genetics , Genes, Neoplasm/genetics , Genomics , Humans , Kaplan-Meier Estimate , MAP Kinase Kinase 4/genetics , Polymorphism, Single Nucleotide/genetics , Prognosis , Protein Phosphatase 2/genetics , Treatment Outcome
8.
PLoS Genet ; 11(3): e1005053, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25790137

ABSTRACT

The downstream functions of the DNA binding tumor suppressor p53 vary depending on the cellular context, and persistent p53 activation has recently been implicated in tumor suppression and senescence. However, genome-wide information about p53-target gene regulation has been derived mostly from acute genotoxic conditions. Using ChIP-seq and expression data, we have found distinct p53 binding profiles between acutely activated (through DNA damage) and chronically activated (in senescent or pro-apoptotic conditions) p53. Compared to the classical 'acute' p53 binding profile, 'chronic' p53 peaks were closely associated with CpG-islands. Furthermore, the chronic CpG-island binding of p53 conferred distinct expression patterns between senescent and pro-apoptotic conditions. Using the p53 targets seen in the chronic conditions together with external high-throughput datasets, we have built p53 networks that revealed extensive self-regulatory 'p53 hubs' where p53 and many p53 targets can physically interact with each other. Integrating these results with public clinical datasets identified the cancer-associated lipogenic enzyme, SCD, which we found to be directly repressed by p53 through the CpG-island promoter, providing a mechanistic link between p53 and the 'lipogenic phenotype', a hallmark of cancer. Our data reveal distinct phenotype associations of chronic p53 targets that underlie specific gene regulatory mechanisms.


Subject(s)
DNA Methylation/genetics , DNA-Binding Proteins/genetics , Protein Interaction Maps/genetics , Tumor Suppressor Protein p53/genetics , Aging/genetics , Apoptosis/genetics , Cell Line , CpG Islands/genetics , DNA Damage/genetics , DNA-Binding Proteins/metabolism , Gene Expression Regulation , Genes, Tumor Suppressor , Humans , Phenotype , Stearoyl-CoA Desaturase/genetics , Stearoyl-CoA Desaturase/metabolism , Tumor Suppressor Protein p53/metabolism
9.
Breast Cancer Res ; 18(1): 70, 2016 07 01.
Article in English | MEDLINE | ID: mdl-27368372

ABSTRACT

BACKGROUND: High mitotic activity is associated with the genesis and progression of many cancers. Small molecule inhibitors of mitotic apparatus proteins are now being developed and evaluated clinically as anticancer agents. With clinical trials of several of these experimental compounds underway, it is important to understand the molecular mechanisms that determine high mitotic activity, identify tumor subtypes that carry molecular aberrations that confer high mitotic activity, and to develop molecular markers that distinguish which tumors will be most responsive to mitotic apparatus inhibitors. METHODS: We identified a coordinately regulated mitotic apparatus network by analyzing gene expression profiles for 53 malignant and non-malignant human breast cancer cell lines and two separate primary breast tumor datasets. We defined the mitotic network activity index (MNAI) as the sum of the transcriptional levels of the 54 coordinately regulated mitotic apparatus genes. The effect of those genes on cell growth was evaluated by small interfering RNA (siRNA). RESULTS: High MNAI was enriched in basal-like breast tumors and was associated with reduced survival duration and preferential sensitivity to inhibitors of the mitotic apparatus proteins, polo-like kinase, centromere associated protein E and aurora kinase designated GSK462364, GSK923295 and GSK1070916, respectively. Co-amplification of regions of chromosomes 8q24, 10p15-p12, 12p13, and 17q24-q25 was associated with the transcriptional upregulation of this network of 54 mitotic apparatus genes, and we identify transcription factors that localize to these regions and putatively regulate mitotic activity. Knockdown of the mitotic network by siRNA identified 22 genes that might be considered as additional therapeutic targets for this clinically relevant patient subgroup. CONCLUSIONS: We define a molecular signature which may guide therapeutic approaches for tumors with high mitotic network activity.


Subject(s)
Breast Neoplasms/genetics , Gene Expression Regulation, Neoplastic , Gene Regulatory Networks/genetics , Genome, Human/genetics , Mitosis/drug effects , Aurora Kinases/antagonists & inhibitors , Aurora Kinases/genetics , Aurora Kinases/metabolism , Breast Neoplasms/drug therapy , Breast Neoplasms/metabolism , Cell Cycle Proteins/antagonists & inhibitors , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Proliferation/genetics , Chromosomal Proteins, Non-Histone/antagonists & inhibitors , Chromosomal Proteins, Non-Histone/genetics , Chromosomal Proteins, Non-Histone/metabolism , Female , Gene Amplification , Gene Expression Profiling/methods , Gene Regulatory Networks/drug effects , Humans , Kaplan-Meier Estimate , Mitosis/genetics , Prognosis , Protein Serine-Threonine Kinases/antagonists & inhibitors , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins/antagonists & inhibitors , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/metabolism , RNA Interference , Small Molecule Libraries/pharmacology , Treatment Outcome , Polo-Like Kinase 1
10.
J Immunol ; 192(9): 4425-35, 2014 May 01.
Article in English | MEDLINE | ID: mdl-24696235

ABSTRACT

Circulating levels of a soluble type I IFNR are elevated in diseases, such as chronic inflammation, infections, and cancer, but whether it functions as an antagonist, agonist, or transporter is unknown. In this study, we elucidate the in vivo importance of the soluble type I IFNAR, soluble (s)IFNAR2a, which is generated by alternative splicing of the Ifnar2 gene. A transgenic mouse model was established to mimic the 10-15-fold elevated expression of sIFNAR2a observed in some human diseases. We generated transgenic mouse lines, designated SolOX, in which the transgene mRNA and protein-expression patterns mirrored the expression patterns of the endogenous gene. SolOX were demonstrated to be more susceptible to LPS-mediated septic shock, a disease model in which type I IFN plays a crucial role. This effect was independent of "classical" proinflammatory cytokines, such as TNF-α and IL-6, whose levels were unchanged. Because the increased levels of sIFNAR2a did not affect the kinetics of the increased interferonemia, this soluble receptor does not potentiate its ligand signaling by improving IFN pharmacokinetics. Mechanistically, increased levels of sIFNAR2a are likely to facilitate IFN signaling, as demonstrated in spleen cells overexpressing sIFNAR2a, which displayed quicker, higher, and more sustained activation of STAT1 and STAT3. Thus, the soluble IFNR is an important agonist of endogenous IFN actions in pathophysiological processes and also is likely to modulate the therapeutic efficacy of clinically administered IFNs.


Subject(s)
Interferon Type I/immunology , Receptor, Interferon alpha-beta/immunology , Shock, Septic/immunology , Signal Transduction/immunology , Animals , Disease Models, Animal , Enzyme-Linked Immunosorbent Assay , Flow Cytometry , Immunoblotting , Immunophenotyping , Inflammation/immunology , Inflammation/metabolism , Interferon Type I/metabolism , Mice , Mice, Transgenic , Real-Time Polymerase Chain Reaction , Receptor, Interferon alpha-beta/metabolism , Shock, Septic/metabolism , Toll-Like Receptor 4/metabolism
11.
Biol Methods Protoc ; 9(1): bpae028, 2024.
Article in English | MEDLINE | ID: mdl-38903861

ABSTRACT

Cancer, a collection of more than two hundred different diseases, remains a leading cause of morbidity and mortality worldwide. Usually detected at the advanced stages of disease, metastatic cancer accounts for 90% of cancer-associated deaths. Therefore, the early detection of cancer, combined with current therapies, would have a significant impact on survival and treatment of various cancer types. Epigenetic changes such as DNA methylation are some of the early events underlying carcinogenesis. Here, we report on an interpretable machine learning model that can classify 13 cancer types as well as non-cancer tissue samples using only DNA methylome data, with 98.2% accuracy. We utilize the features identified by this model to develop EMethylNET, a robust model consisting of an XGBoost model that provides information to a deep neural network that can generalize to independent data sets. We also demonstrate that the methylation-associated genomic loci detected by the classifier are associated with genes, pathways and networks involved in cancer, providing insights into the epigenomic regulation of carcinogenesis.

12.
Blood ; 118(2): 298-308, 2011 Jul 14.
Article in English | MEDLINE | ID: mdl-21411754

ABSTRACT

Many viruses have developed mechanisms to evade the IFN response. Here, HIV-1 was shown to induce a distinct subset of IFN-stimulated genes (ISGs) in monocyte-derived dendritic cells (DCs), without detectable type I or II IFN. These ISGs all contained an IFN regulatory factor 1 (IRF-1) binding site in their promoters, and their expression was shown to be driven by IRF-1, indicating this subset was induced directly by viral infection by IRF-1. IRF-1 and -7 protein expression was enriched in HIV p24 antigen-positive DCs. A HIV deletion mutant with the IRF-1 binding site deleted from the long terminal repeat showed reduced growth kinetics. Early and persistent induction of IRF-1 was coupled with sequential transient up-regulation of its 2 inhibitors, IRF-8, followed by IRF-2, suggesting a mechanism for IFN inhibition. HIV-1 mutants with Vpr deleted induced IFN, showing that Vpr is inhibitory. However, HIV IFN inhibition was mediated by failure of IRF-3 activation rather than by its degradation, as in T cells. In contrast, herpes simplex virus type 2 markedly induced IFNß and a broader range of ISGs to higher levels, supporting the hypothesis that HIV-1 specifically manipulates the induction of IFN and ISGs to enhance its noncytopathic replication in DCs.


Subject(s)
Dendritic Cells/virology , HIV Infections/genetics , HIV Infections/immunology , HIV-1/physiology , Interferon Regulatory Factor-1/physiology , Interferon Type I/metabolism , Cells, Cultured , Dendritic Cells/metabolism , Dendritic Cells/pathology , Down-Regulation/immunology , Gene Expression Profiling , Gene Expression Regulation/immunology , HIV Infections/metabolism , HIV Infections/pathology , Humans , Interferon Regulatory Factor-1/genetics , Interferon Regulatory Factor-1/metabolism , Microarray Analysis , Promoter Regions, Genetic , Sequence Analysis, DNA , Signal Transduction/immunology , Signal Transduction/physiology , Up-Regulation/genetics , Up-Regulation/immunology
14.
Breast Cancer Res ; 14(2): R63, 2012 Apr 18.
Article in English | MEDLINE | ID: mdl-22513257

ABSTRACT

INTRODUCTION: Cis-acting regulatory single nucleotide polymorphisms (SNPs) at specific loci may modulate penetrance of germline mutations at the same loci by introducing different levels of expression of the wild-type allele. We have previously reported that BRCA2 shows differential allelic expression and we hypothesize that the known variable penetrance of BRCA2 mutations might be associated with this mechanism. METHODS: We combined haplotype analysis and differential allelic expression of BRCA2 in breast tissue to identify expression haplotypes and candidate cis-regulatory variants. These candidate variants underwent selection based on in silico predictions for regulatory potential and disruption of transcription factor binding, and were functionally analyzed in vitro and in vivo in normal and breast cancer cell lines. SNPs tagging the expression haplotypes were correlated with the total expression of several genes in breast tissue measured by Taqman and microarray technologies. The effect of the expression haplotypes on breast cancer risk in BRCA2 mutation carriers was investigated in 2,754 carriers. RESULTS: We identified common haplotypes associated with differences in the levels of BRCA2 expression in human breast cells. We characterized three cis-regulatory SNPs located at the promoter and two intronic regulatory elements which affect the binding of the transcription factors C/EBPα, HMGA1, D-binding protein (DBP) and ZF5. We showed that the expression haplotypes also correlated with changes in the expression of other genes in normal breast. Furthermore, there was suggestive evidence that the minor allele of SNP rs4942440, which is associated with higher BRCA2 expression, is also associated with a reduced risk of breast cancer (per-allele hazard ratio (HR) = 0.85, 95% confidence interval (CI) = 0.72 to 1.00, P-trend = 0.048). CONCLUSIONS: Our work provides further insights into the role of cis-regulatory variation in the penetrance of disease-causing mutations. We identified small-effect genetic variants associated with allelic expression differences in BRCA2 which could possibly affect the risk in mutation carriers through altering expression levels of the wild-type allele.


Subject(s)
BRCA2 Protein/genetics , Gene Expression Regulation , Regulatory Sequences, Nucleic Acid , Female , Gene Expression Regulation, Neoplastic , Genetic Predisposition to Disease , Haplotypes , Heterozygote , Humans , Polymorphism, Single Nucleotide , Promoter Regions, Genetic , Reference Values , Transcription Factors/genetics
15.
Nucleic Acids Res ; 38(3): e17, 2010 Jan.
Article in English | MEDLINE | ID: mdl-19923232

ABSTRACT

Illumina BeadArrays are among the most popular and reliable platforms for gene expression profiling. However, little external scrutiny has been given to the design, selection and annotation of BeadArray probes, which is a fundamental issue in data quality and interpretation. Here we present a pipeline for the complete genomic and transcriptomic re-annotation of Illumina probe sequences, also applicable to other platforms, with its output available through a Web interface and incorporated into Bioconductor packages. We have identified several problems with the design of individual probes and we show the benefits of probe re-annotation on the analysis of BeadArray gene expression data sets. We discuss the importance of aspects such as probe coverage of individual transcripts, alternative messenger RNA splicing, single-nucleotide polymorphisms, repeat sequences, RNA degradation biases and probes targeting genomic regions with no known transcription. We conclude that many of the Illumina probes have unreliable original annotation and that our re-annotation allows analyses to focus on the good quality probes, which form the majority, and also to expand the scope of biological information that can be extracted.


Subject(s)
Gene Expression Profiling/methods , Oligonucleotide Array Sequence Analysis/methods , Oligonucleotide Probes/chemistry , Alternative Splicing , Base Pair Mismatch , Humans , Polymorphism, Single Nucleotide , Repetitive Sequences, Nucleic Acid , Software
16.
Sci Adv ; 8(39): eabn9828, 2022 Sep 30.
Article in English | MEDLINE | ID: mdl-36170366

ABSTRACT

Current gold standard diagnostic strategies are unable to accurately differentiate malignant from benign small renal masses preoperatively; consequently, 20% of patients undergo unnecessary surgery. Devising a more confident presurgical diagnosis is key to improving treatment decision-making. We therefore developed MethylBoostER, a machine learning model leveraging DNA methylation data from 1228 tissue samples, to classify pathological subtypes of renal tumors (benign oncocytoma, clear cell, papillary, and chromophobe RCC) and normal kidney. The prediction accuracy in the testing set was 0.960, with class-wise ROC AUCs >0.988 for all classes. External validation was performed on >500 samples from four independent datasets, achieving AUCs >0.89 for all classes and average accuracies of 0.824, 0.703, 0.875, and 0.894 for the four datasets. Furthermore, consistent classification of multiregion samples (N = 185) from the same patient demonstrates that methylation heterogeneity does not limit model applicability. Following further clinical studies, MethylBoostER could facilitate a more confident presurgical diagnosis to guide treatment decision-making in the future.

17.
Nat Aging ; 2(1): 31-45, 2022 01.
Article in English | MEDLINE | ID: mdl-37118356

ABSTRACT

Senescence is a fate-determined state, accompanied by reorganization of heterochromatin. Although lineage-appropriate genes can be temporarily repressed through facultative heterochromatin, stable silencing of lineage-inappropriate genes often involves the constitutive heterochromatic mark, histone H3 lysine 9 trimethylation (H3K9me3). The fate of these heterochromatic genes during senescence is unclear. In the present study, we show that a small number of lineage-inappropriate genes, exemplified by the LCE2 skin genes, are derepressed during senescence from H3K9me3 regions in fibroblasts. DNA FISH experiments reveal that these gene loci, which are condensed at the nuclear periphery in proliferative cells, are decompacted during senescence. Decompaction of the locus is not sufficient for LCE2 expression, which requires p53 and C/EBPß signaling. NLRP3, which is predominantly expressed in macrophages from an open topologically associated domain (TAD), is also derepressed in senescent fibroblasts due to the local disruption of the H3K9me3-rich TAD that contains it. NLRP3 has been implicated in the amplification of inflammatory cytokine signaling in senescence and aging, highlighting the functional relevance of gene induction from 'permissive' H3K9me3 regions in senescent cells.


Subject(s)
Heterochromatin , Histones , Heterochromatin/genetics , Histones/genetics , NLR Family, Pyrin Domain-Containing 3 Protein/genetics , Cellular Senescence/genetics , Gene Expression
18.
Nucleic Acids Res ; 37(Database issue): D852-7, 2009 Jan.
Article in English | MEDLINE | ID: mdl-18996892

ABSTRACT

INTERFEROME is an open access database of types I, II and III Interferon regulated genes (http://www.interferome.org) collected from analysing expression data sets of cells treated with IFNs. This database of interferon regulated genes integrates information from high-throughput experiments with annotation, ontology, orthologue sequences from 37 species, tissue expression patterns and gene regulatory information to enable a detailed investigation of the molecular mechanisms underlying IFN biology. INTERFEROME fulfils a need in infection, immunity, development and cancer research by providing computational tools to assist in identifying interferon signatures in gene lists generated by high-throughput expression technologies, and their potential molecular and biological consequences.


Subject(s)
Databases, Genetic , Gene Expression Regulation , Interferons/pharmacology , Animals , Gene Expression Profiling , Humans , Mice , Oligonucleotide Array Sequence Analysis , Promoter Regions, Genetic , Proteomics
19.
EMBO Mol Med ; 13(2): e12105, 2021 02 05.
Article in English | MEDLINE | ID: mdl-33369245

ABSTRACT

Lamins are crucial proteins for nuclear functionality. Here, we provide new evidence showing that increased lamin B1 levels contribute to the pathophysiology of Huntington's disease (HD), a CAG repeat-associated neurodegenerative disorder. Through fluorescence-activated nuclear suspension imaging, we show that nucleus from striatal medium-sized spiny and CA1 hippocampal neurons display increased lamin B1 levels, in correlation with altered nuclear morphology and nucleocytoplasmic transport disruption. Moreover, ChIP-sequencing analysis shows an alteration of lamin-associated chromatin domains in hippocampal nuclei, accompanied by changes in chromatin accessibility and transcriptional dysregulation. Supporting lamin B1 alterations as a causal role in mutant huntingtin-mediated neurodegeneration, pharmacological normalization of lamin B1 levels in the hippocampus of the R6/1 mouse model of HD by betulinic acid administration restored nuclear homeostasis and prevented motor and cognitive dysfunction. Collectively, our work points increased lamin B1 levels as a new pathogenic mechanism in HD and provides a novel target for its intervention.


Subject(s)
Huntington Disease , Animals , Corpus Striatum , Huntington Disease/genetics , Lamin Type B/genetics , Mice , Neurons
20.
Nat Commun ; 11(1): 6049, 2020 11 27.
Article in English | MEDLINE | ID: mdl-33247104

ABSTRACT

Senescence is a state of stable proliferative arrest, generally accompanied by the senescence-associated secretory phenotype, which modulates tissue homeostasis. Enhancer-promoter interactions, facilitated by chromatin loops, play a key role in gene regulation but their relevance in senescence remains elusive. Here, we use Hi-C to show that oncogenic RAS-induced senescence in human diploid fibroblasts is accompanied by extensive enhancer-promoter rewiring, which is closely connected with dynamic cohesin binding to the genome. We find de novo cohesin peaks often at the 3' end of a subset of active genes. RAS-induced de novo cohesin peaks are transcription-dependent and enriched for senescence-associated genes, exemplified by IL1B, where de novo cohesin binding is involved in new loop formation. Similar IL1B induction with de novo cohesin appearance and new loop formation are observed in terminally differentiated macrophages, but not TNFα-treated cells. These results suggest that RAS-induced senescence represents a cell fate determination-like process characterised by a unique gene expression profile and 3D genome folding signature, mediated in part through cohesin redistribution on chromatin.


Subject(s)
Cell Cycle Proteins/metabolism , Cellular Senescence/genetics , Chromatin/metabolism , Chromosomal Proteins, Non-Histone/metabolism , Transcription, Genetic , CCCTC-Binding Factor/metabolism , Cell Differentiation/genetics , Cell Line , Enhancer Elements, Genetic/genetics , Gene Expression Regulation , Genetic Loci , Genome , Humans , Interleukin-1/genetics , Macrophages/cytology , Promoter Regions, Genetic , Protein Binding/drug effects , Tumor Necrosis Factor-alpha/pharmacology , ras Proteins/metabolism , Cohesins
SELECTION OF CITATIONS
SEARCH DETAIL