Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 38
1.
Sci Adv ; 10(13): eadk5386, 2024 Mar 29.
Article En | MEDLINE | ID: mdl-38536927

While pancreatic ductal adenocarcinomas (PDACs) are addicted to KRAS-activating mutations, inhibitors of downstream KRAS effectors, such as the MEK1/2 kinase inhibitor trametinib, are devoid of therapeutic effects. However, the extensive rewiring of regulatory circuits driven by the attenuation of the KRAS pathway may induce vulnerabilities of therapeutic relevance. An in-depth molecular analysis of the transcriptional and epigenomic alterations occurring in PDAC cells in the initial hours after MEK1/2 inhibition by trametinib unveiled the induction of endogenous retroviruses (ERVs) escaping epigenetic silencing, leading to the production of double-stranded RNAs and the increased expression of interferon (IFN) genes. We tracked ERV activation to the early induction of the transcription factor ELF3, which extensively bound and activated nonsilenced retroelements and synergized with IRF1 (interferon regulatory factor 1) in the activation of IFNs and IFN-stimulated genes. Trametinib-induced viral mimicry in PDAC may be exploited in the rational design of combination therapies in immuno-oncology.


Carcinoma, Pancreatic Ductal , Endogenous Retroviruses , Pancreatic Neoplasms , Humans , Endogenous Retroviruses/genetics , Signal Transduction , Proto-Oncogene Proteins p21(ras)/genetics , Proto-Oncogene Proteins p21(ras)/metabolism , Pancreatic Neoplasms/drug therapy , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/metabolism , Carcinoma, Pancreatic Ductal/drug therapy , Carcinoma, Pancreatic Ductal/genetics , Carcinoma, Pancreatic Ductal/metabolism
2.
Mol Cell ; 84(5): 967-980.e10, 2024 Mar 07.
Article En | MEDLINE | ID: mdl-38242130

Histone-modifying enzymes depend on the availability of cofactors, with acetyl-coenzyme A (CoA) being required for histone acetyltransferase (HAT) activity. The discovery that mitochondrial acyl-CoA-producing enzymes translocate to the nucleus suggests that high concentrations of locally synthesized metabolites may impact acylation of histones and other nuclear substrates, thereby controlling gene expression. Here, we show that 2-ketoacid dehydrogenases are stably associated with the Mediator complex, thus providing a local supply of acetyl-CoA and increasing the generation of hyper-acetylated histone tails. Nitric oxide (NO), which is produced in large amounts in lipopolysaccharide-stimulated macrophages, inhibited the activity of Mediator-associated 2-ketoacid dehydrogenases. Elevation of NO levels and the disruption of Mediator complex integrity both affected de novo histone acetylation within a shared set of genomic regions. Our findings indicate that the local supply of acetyl-CoA generated by 2-ketoacid dehydrogenases bound to Mediator is required to maximize acetylation of histone tails at sites of elevated HAT activity.


Histones , Nitric Oxide , Histones/genetics , Histones/metabolism , Acetyl Coenzyme A/metabolism , Acetylation , Nitric Oxide/metabolism , Mediator Complex/metabolism , Oxidoreductases/metabolism
3.
Front Endocrinol (Lausanne) ; 14: 1272646, 2023.
Article En | MEDLINE | ID: mdl-37842307

Inflammation-dependent changes in gene expression programs in innate immune cells, such as macrophages, involve extensive reprogramming of metabolism. This reprogramming is essential for the production of metabolites required for chromatin modifications, such as acetyl-CoA, and regulate their usage and availability impacting the macrophage epigenome. One of the most transcriptionally induced proinflammatory mediator is nitric oxide (NO), which has been shown to inhibit key metabolic enzymes involved in the production of these metabolites. Recent evidence indicates that NO inhibits mitochondrial enzymes such as pyruvate dehydrogenase (PDH) in macrophages induced by inflammatory stimulus. PDH is involved in the production of acetyl-CoA, which is essential for chromatin modifications in the nucleus, such as histone acetylation. In addition, acetyl-CoA levels in inflamed macrophages are regulated by ATP citrate lyase (ACLY) and citrate transporter SLC25A1. Interestingly, acetyl-CoA producing enzymes, such as PDH and ACLY, have also been reported to be present in the nucleus and to support the local generation of cofactors such as acetyl-CoA. Here, we will discuss the mechanisms involved in the regulation of acetyl-CoA production by metabolic enzymes, their inhibition by prolonged exposure to inflammation stimuli, their involvement in dynamic inflammatory expression changes and how these emerging findings could have significant implications for the design of novel therapeutic approaches.


ATP Citrate (pro-S)-Lyase , Epigenesis, Genetic , Humans , Acetyl Coenzyme A/metabolism , ATP Citrate (pro-S)-Lyase/genetics , ATP Citrate (pro-S)-Lyase/metabolism , Inflammation/genetics , Acyltransferases/genetics , Chromatin
4.
Biomedicines ; 11(10)2023 Sep 23.
Article En | MEDLINE | ID: mdl-37892987

Inflammation impacts human hematopoiesis across physiologic and pathologic conditions, as signals derived from the bone marrow microenvironment, such as pro-inflammatory cytokines and chemokines, have been shown to alter hematopoietic stem cell (HSCs) homeostasis. Dysregulated inflammation can skew HSC fate-related decisions, leading to aberrant hematopoiesis and potentially contributing to the pathogenesis of hematological disorders such as myelodysplastic syndromes (MDS). Recently, emerging studies have used single-cell sequencing and muti-omic approaches to investigate HSC cellular heterogeneity and gene expression in normal hematopoiesis as well as in myeloid malignancies. This review summarizes recent reports mechanistically dissecting the role of inflammatory signaling and innate immune response activation due to MDS progression. Furthermore, we highlight the growing importance of using multi-omic techniques, such as single-cell profiling and deconvolution methods, to unravel MDSs' heterogeneity. These approaches have provided valuable insights into the patterns of clonal evolution that drive MDS progression and have elucidated the impact of inflammation on the composition of the bone marrow immune microenvironment in MDS.

5.
EMBO Rep ; 22(9): e53251, 2021 09 06.
Article En | MEDLINE | ID: mdl-34328708

Macrophages react to microbial and endogenous danger signals by activating a broad panel of effector and homeostatic responses. Such responses entail rapid and stimulus-specific changes in gene expression programs accompanied by extensive rewiring of metabolism, with alterations in chromatin modifications providing one layer of integration of transcriptional and metabolic regulation. A systematic and mechanistic understanding of the mutual influences between signal-induced metabolic changes and gene expression is still lacking. Here, we discuss current evidence, controversies, knowledge gaps, and future areas of investigation on how metabolic and transcriptional changes are dynamically integrated during macrophage activation. The cross-talk between metabolism and inflammatory gene expression is in part accounted for by alterations in the production, usage, and availability of metabolic intermediates that impact the macrophage epigenome. In addition, stimulus-inducible gene expression changes alter the production of inflammatory mediators, such as nitric oxide, that in turn modulate the activity of metabolic enzymes thus determining complex regulatory loops. Critical issues remain to be understood, notably whether and how metabolic rewiring can bring about gene-specific (as opposed to global) expression changes.


Inflammation , Macrophage Activation , Gene Expression , Gene Expression Regulation , Humans , Inflammation/genetics , Macrophage Activation/genetics , Macrophages
6.
Nat Struct Mol Biol ; 28(4): 337-346, 2021 04.
Article En | MEDLINE | ID: mdl-33767452

Interactions between the splicing machinery and RNA polymerase II increase protein-coding gene transcription. Similarly, exons and splicing signals of enhancer-generated long noncoding RNAs (elncRNAs) augment enhancer activity. However, elncRNAs are inefficiently spliced, suggesting that, compared with protein-coding genes, they contain qualitatively different exons with a limited ability to drive splicing. We show here that the inefficiently spliced first exons of elncRNAs as well as promoter-antisense long noncoding RNAs (pa-lncRNAs) in human and mouse cells trigger a transcription termination checkpoint that requires WDR82, an RNA polymerase II-binding protein, and its RNA-binding partner of previously unknown function, ZC3H4. We propose that the first exons of elncRNAs and pa-lncRNAs are an intrinsic component of a regulatory mechanism that, on the one hand, maximizes the activity of these cis-regulatory elements by recruiting the splicing machinery and, on the other, contains elements that suppress pervasive extragenic transcription.


Chromosomal Proteins, Non-Histone/genetics , DNA-Binding Proteins/genetics , RNA Polymerase II/ultrastructure , RNA, Long Noncoding/genetics , Transcription, Genetic , Alternative Splicing/genetics , Animals , Chromosomal Proteins, Non-Histone/ultrastructure , DNA-Binding Proteins/ultrastructure , Exons/genetics , Humans , Mice , Promoter Regions, Genetic/genetics , RNA Polymerase II/genetics , RNA Splicing/genetics , RNA, Antisense/genetics , RNA, Antisense/ultrastructure , RNA, Long Noncoding/ultrastructure , RNA, Messenger/genetics , Regulatory Sequences, Nucleic Acid/genetics
7.
J Cell Biol ; 220(3)2021 03 01.
Article En | MEDLINE | ID: mdl-33566069

The commitment of mesenchymal stem cells to preadipocytes is stimulated by hormonal induction. Preadipocytes induced to differentiate repress protein synthesis, remodel their cytoskeleton, and increase mitochondrial function to support anabolic pathways. These changes enable differentiation into mature adipocytes. Our understanding of the factors that coordinately regulate the early events of adipocyte differentiation remains incomplete. Here, by using multipronged approaches, we have identified zinc finger CCCH-type containing 10 (Zc3h10) as a critical regulator of the early stages of adipogenesis. Zc3h10 depletion in preadipocytes resulted in increased protein translation and impaired filamentous (F)-actin remodeling, with the latter detrimental effect leading to mitochondrial and metabolic dysfunction. These defects negatively affected differentiation to mature adipocytes. In contrast, Zc3h10 overexpression yielded mature adipocytes with remarkably increased lipid droplet size. Overall, our study establishes Zc3h10 as a fundamental proadipogenic transcription factor that represses protein synthesis and promotes F-actin/mitochondria dynamics to ensure proper energy metabolism and favor lipid accumulation.


Actins/metabolism , Adipogenesis , Mitochondria/metabolism , Protein Biosynthesis , RNA-Binding Proteins/metabolism , Actin Cytoskeleton/metabolism , Adipocytes/metabolism , Adipogenesis/genetics , Adipose Tissue, White/metabolism , Animals , Cell Line , Citric Acid Cycle , Cyclic AMP-Dependent Protein Kinases/metabolism , Energy Metabolism/genetics , Gene Expression Regulation , Lipid Metabolism/genetics , Male , Metabolome , Mice, Inbred C57BL , Mitochondrial Dynamics , RNA Precursors/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA-Binding Proteins/genetics , Transcriptome/genetics , rho GTP-Binding Proteins/metabolism
8.
PLoS Genet ; 14(5): e1007380, 2018 05.
Article En | MEDLINE | ID: mdl-29734330

Chronic inflammation promotes oncogenic transformation and tumor progression. Many inflammatory agents also generate a toxic microenvironment, implying that adaptive mechanisms must be deployed for cells to survive and undergo transformation in such unfavorable contexts. A paradigmatic case is represented by cancers occurring in pediatric patients with genetic defects of hepatocyte phosphatidylcholine transporters and in the corresponding mouse model (Mdr2-/- mice), in which impaired bile salt emulsification leads to chronic hepatocyte damage and inflammation, eventually resulting in oncogenic transformation. By combining genomics and metabolomics, we found that the transition from inflammation to cancer in Mdr2-/- mice was linked to the sustained transcriptional activation of metabolic detoxification systems and transporters by the Constitutive Androstane Receptor (CAR), a hepatocyte-specific nuclear receptor. Activation of CAR-dependent gene expression programs coincided with reduced content of toxic bile acids in cancer nodules relative to inflamed livers. Treatment of Mdr2-/- mice with a CAR inhibitor blocked cancer progression and caused a partial regression of existing tumors. These results indicate that the acquisition of resistance to endo- or xeno-biotic toxicity is critical for cancers that develop in toxic microenvironments.


Bile Acids and Salts/metabolism , Cell Transformation, Neoplastic/genetics , Inactivation, Metabolic/genetics , Liver/metabolism , ATP Binding Cassette Transporter, Subfamily B/genetics , ATP Binding Cassette Transporter, Subfamily B/metabolism , Androstanols/pharmacology , Animals , Cell Transformation, Neoplastic/metabolism , Constitutive Androstane Receptor , Gene Expression Profiling/methods , Gene Ontology , Hepatitis/genetics , Hepatitis/metabolism , Liver Neoplasms/genetics , Liver Neoplasms/metabolism , Mice, Knockout , Receptors, Cytoplasmic and Nuclear/antagonists & inhibitors , Receptors, Cytoplasmic and Nuclear/genetics , Signal Transduction/genetics , Transcriptional Activation/drug effects , ATP-Binding Cassette Sub-Family B Member 4
9.
Genome Res ; 28(5): 639-653, 2018 05.
Article En | MEDLINE | ID: mdl-29643204

The retrotransposon Long Interspersed Element 1 (LINE-1 or L1) is a continuing source of germline and somatic mutagenesis in mammals. Deregulated L1 activity is a hallmark of cancer, and L1 mutagenesis has been described in numerous human malignancies. We previously employed retrotransposon capture sequencing (RC-seq) to analyze hepatocellular carcinoma (HCC) samples from patients infected with hepatitis B or hepatitis C virus and identified L1 variants responsible for activating oncogenic pathways. Here, we have applied RC-seq and whole-genome sequencing (WGS) to an Abcb4 (Mdr2)-/- mouse model of hepatic carcinogenesis and demonstrated for the first time that L1 mobilization occurs in murine tumors. In 12 HCC nodules obtained from 10 animals, we validated four somatic L1 insertions by PCR and capillary sequencing, including TF subfamily elements, and one GF subfamily example. One of the TF insertions carried a 3' transduction, allowing us to identify its donor L1 and to demonstrate that this full-length TF element retained retrotransposition capacity in cultured cancer cells. Using RC-seq, we also identified eight tumor-specific L1 insertions from 25 HCC patients with a history of alcohol abuse. Finally, we used RC-seq and WGS to identify three tumor-specific L1 insertions among 10 intra-hepatic cholangiocarcinoma (ICC) patients, including one insertion traced to a donor L1 on Chromosome 22 known to be highly active in other cancers. This study reveals L1 mobilization as a common feature of hepatocarcinogenesis in mammals, demonstrating that the phenomenon is not restricted to human viral HCC etiologies and is encountered in murine liver tumors.


Carcinoma, Hepatocellular/genetics , Liver Neoplasms/genetics , Long Interspersed Nucleotide Elements/genetics , Retroelements/genetics , ATP Binding Cassette Transporter, Subfamily B/genetics , Adult , Aged , Aged, 80 and over , Animals , Cell Transformation, Neoplastic/genetics , Female , Humans , Liver/metabolism , Liver/pathology , Male , Mammals/genetics , Mice, Knockout , Middle Aged , Mutagenesis, Insertional , ATP-Binding Cassette Sub-Family B Member 4
10.
EMBO Rep ; 19(4)2018 04.
Article En | MEDLINE | ID: mdl-29507079

Mitochondria are the energy-generating hubs of the cell. In spite of considerable advances, our understanding of the factors that regulate the molecular circuits that govern mitochondrial function remains incomplete. Using a genome-wide functional screen, we identify the poorly characterized protein Zinc finger CCCH-type containing 10 (Zc3h10) as regulator of mitochondrial physiology. We show that Zc3h10 is upregulated during physiological mitochondriogenesis as it occurs during the differentiation of myoblasts into myotubes. Zc3h10 overexpression boosts mitochondrial function and promotes myoblast differentiation, while the depletion of Zc3h10 results in impaired myoblast differentiation, mitochondrial dysfunction, reduced expression of electron transport chain (ETC) subunits, and blunted TCA cycle flux. Notably, we have identified a loss-of-function mutation of Zc3h10 in humans (Tyr105 to Cys105) that is associated with increased body mass index, fat mass, fasting glucose, and triglycerides. Isolated peripheral blood mononuclear cells from individuals homozygotic for Cys105 display reduced oxygen consumption rate, diminished expression of some ETC subunits, and decreased levels of some TCA cycle metabolites, which all together derive in mitochondrial dysfunction. Taken together, our study identifies Zc3h10 as a novel mitochondrial regulator.


Carrier Proteins/metabolism , Mitochondria/metabolism , Aged , Animals , Carrier Proteins/genetics , Cell Differentiation , Cell Line , Citric Acid Cycle , Computational Biology/methods , Energy Metabolism , Female , Gene Expression , Gene Expression Profiling , Gene Silencing , Humans , Male , Mice , Mitochondria/genetics , Muscle Fibers, Skeletal/metabolism , Muscle, Skeletal/metabolism , Mutation , Myoblasts/cytology , Myoblasts/metabolism , Proteome , Proteomics/methods
11.
Transcription ; 9(3): 177-181, 2018.
Article En | MEDLINE | ID: mdl-28992423

Cell type-specific and housekeeping enhancers and promoters collectively control the transcriptional output of mammalian cells. Recent data clarify how DNA sequence features on the one hand control functional coupling of promoters with selected enhancers, and on the other impart high level of activity to a broad range of regulatory elements.


Enhancer Elements, Genetic , Promoter Regions, Genetic , Transcription, Genetic , Animals , Chromatin/genetics , DNA/genetics , Genes, Essential , Humans
12.
Genes Dev ; 31(4): 399-412, 2017 02 15.
Article En | MEDLINE | ID: mdl-28275002

Enhancers and promoters that control the transcriptional output of terminally differentiated cells include cell type-specific and broadly active housekeeping elements. Whether the high constitutive activity of these two groups of cis-regulatory elements relies on entirely distinct or instead also on shared regulators is unknown. By dissecting the cis-regulatory repertoire of macrophages, we found that the ELF subfamily of ETS proteins selectively bound within 60 base pairs (bp) from the transcription start sites of highly active housekeeping genes. ELFs also bound constitutively active, but not poised, macrophage-specific enhancers and promoters. The role of ELFs in promoting high-level constitutive transcription was suggested by multiple evidence: ELF sites enabled robust transcriptional activation by endogenous and minimal synthetic promoters, ELF recruitment was stabilized by the transcriptional machinery, and ELF proteins mediated recruitment of transcriptional and chromatin regulators to core promoters. These data suggest that the co-optation of a limited number of highly active transcription factors represents a broadly adopted strategy to equip both cell type-specific and housekeeping cis-regulatory elements with the ability to efficiently promote transcription.


Gene Expression Regulation/genetics , Genes, Essential/genetics , Macrophages/physiology , Transcription Factors/metabolism , Animals , Chromatin/metabolism , Enhancer Elements, Genetic/genetics , Mice , Mice, Inbred C57BL , Promoter Regions, Genetic/genetics , Protein Binding , Protein Transport , Transcription Factors/genetics
13.
Nat Immunol ; 18(5): 530-540, 2017 05.
Article En | MEDLINE | ID: mdl-28288101

Stimulation of macrophages with interferon-γ (IFN-γ) and interleukin 4 (IL-4) triggers distinct and opposing activation programs. During mixed infections or cancer, macrophages are often exposed to both cytokines, but how these two programs influence each other remains unclear. We found that IFN-γ and IL-4 mutually inhibited the epigenomic and transcriptional changes induced by each cytokine alone. Computational and functional analyses revealed the genomic bases for gene-specific cross-repression. For instance, while binding motifs for the transcription factors STAT1 and IRF1 were associated with robust and IL-4-resistant responses to IFN-γ, their coexistence with binding sites for auxiliary transcription factors such as AP-1 generated vulnerability to IL-4-mediated inhibition. These data provide a core mechanistic framework for the integration of signals that control macrophage activation in complex environmental conditions.


Cell Differentiation , Epigenesis, Genetic , Macrophages/physiology , Proto-Oncogene Proteins c-myc/metabolism , Transcriptional Activation , Animals , Cell Line , Gene Expression Regulation , Humans , Interferon Regulatory Factor-1/genetics , Interferon Regulatory Factor-1/metabolism , Interferon-gamma/metabolism , Interleukin-4/metabolism , Mice , Mice, Inbred Strains , Proto-Oncogene Proteins c-myc/genetics , RNA, Small Interfering/genetics , STAT1 Transcription Factor/genetics , STAT1 Transcription Factor/metabolism , Signal Transduction , Transcription Factor AP-1/metabolism
14.
Hepatology ; 65(5): 1708-1719, 2017 05.
Article En | MEDLINE | ID: mdl-27859418

The ST18 gene has been proposed to act either as a tumor suppressor or as an oncogene in different human cancers, but direct evidence for its role in tumorigenesis has been lacking thus far. Here, we demonstrate that ST18 is critical for tumor progression and maintenance in a mouse model of liver cancer, based on oncogenic transformation and adoptive transfer of primary precursor cells (hepatoblasts). ST18 messenger RNA (mRNA) and protein were detectable neither in normal liver nor in cultured hepatoblasts, but were readily expressed after subcutaneous engraftment and tumor growth. ST18 expression in liver cells was induced by inflammatory cues, including acute or chronic inflammation in vivo, as well as coculture with macrophages in vitro. Knocking down the ST18 mRNA in transplanted hepatoblasts delayed tumor progression. Induction of ST18 knockdown in pre-established tumors caused rapid tumor involution associated with pervasive morphological changes, proliferative arrest, and apoptosis in tumor cells, as well as depletion of tumor-associated macrophages, vascular ectasia, and hemorrhage. Reciprocally, systemic depletion of macrophages in recipient animals had very similar phenotypic consequences, impairing either tumor development or maintenance, and suppressing ST18 expression in hepatoblasts. Finally, RNA sequencing of ST18-depleted tumors before involution revealed down-regulation of inflammatory response genes, pointing to the suppression of nuclear factor kappa B-dependent transcription. CONCLUSION: ST18 expression in epithelial cells is induced by tumor-associated macrophages, contributing to the reciprocal feed-forward loop between both cell types in liver tumorigenesis. Our findings warrant the exploration of means to interfere with ST18-dependent epithelium-macrophage interactions in a therapeutic setting. (Hepatology 2017;65:1708-1719).


Carcinoma, Hepatocellular/etiology , Liver Neoplasms, Experimental/etiology , Transcription Factors/metabolism , Animals , Carcinoma, Hepatocellular/metabolism , Liver Neoplasms, Experimental/metabolism , Mice, Inbred C57BL
15.
Mol Cell ; 60(3): 460-74, 2015 Nov 05.
Article En | MEDLINE | ID: mdl-26593720

Upon recruitment to active enhancers and promoters, RNA polymerase II (Pol II) generates short non-coding transcripts of unclear function. The mechanisms that control the length and the amount of ncRNAs generated by cis-regulatory elements are largely unknown. Here, we show that the adaptor protein WDR82 and its associated complexes actively limit such non-coding transcription. WDR82 targets the SET1 H3K4 methyltransferases and the nuclear protein phosphatase 1 (PP1) complexes to the initiating Pol II. WDR82 and PP1 also interact with components of the transcriptional termination and RNA processing machineries. Depletion of WDR82, SET1, or the PP1 subunit required for its nuclear import caused distinct but overlapping transcription termination defects at highly expressed genes and active enhancers and promoters, thus enabling the increased synthesis of unusually long ncRNAs. These data indicate that transcription initiated from cis-regulatory elements is tightly coordinated with termination mechanisms that impose the synthesis of short RNAs.


Cell Nucleus/metabolism , Enhancer Elements, Genetic/physiology , Promoter Regions, Genetic/physiology , RNA Polymerase II/metabolism , RNA, Untranslated/biosynthesis , Transcription Termination, Genetic/physiology , Active Transport, Cell Nucleus/physiology , Animals , Cell Nucleus/genetics , Chromosomal Proteins, Non-Histone/genetics , Chromosomal Proteins, Non-Histone/metabolism , Histone-Lysine N-Methyltransferase/genetics , Histone-Lysine N-Methyltransferase/metabolism , Mice , Phosphoprotein Phosphatases/genetics , Phosphoprotein Phosphatases/metabolism , RNA Polymerase II/genetics , RNA, Untranslated/genetics
16.
Genome Res ; 25(12): 1812-24, 2015 Dec.
Article En | MEDLINE | ID: mdl-26510915

An increasing number of noncoding RNAs (ncRNAs) have been implicated in various human diseases including cancer; however, the ncRNA transcriptome of hepatocellular carcinoma (HCC) is largely unexplored. We used CAGE to map transcription start sites across various types of human and mouse HCCs with emphasis on ncRNAs distant from protein-coding genes. Here, we report that retroviral LTR promoters, expressed in healthy tissues such as testis and placenta but not liver, are widely activated in liver tumors. Despite HCC heterogeneity, a subset of LTR-derived ncRNAs were more than 10-fold up-regulated in the vast majority of samples. HCCs with a high LTR activity mostly had a viral etiology, were less differentiated, and showed higher risk of recurrence. ChIP-seq data show that MYC and MAX are associated with ncRNA deregulation. Globally, CAGE enabled us to build a mammalian promoter map for HCC, which uncovers a new layer of complexity in HCC genomics.


Carcinoma, Hepatocellular/etiology , Gene Expression Profiling , Liver Neoplasms/etiology , Promoter Regions, Genetic , RNA, Untranslated/genetics , Terminal Repeat Sequences , Transcription Initiation Site , ATP Binding Cassette Transporter, Subfamily B/genetics , Animals , Carcinoma, Hepatocellular/pathology , Cell Line, Tumor , Cell Transformation, Neoplastic/genetics , Cell Transformation, Viral , Computational Biology/methods , Disease Models, Animal , Disease Progression , Gene Expression Profiling/methods , Gene Expression Regulation, Neoplastic , Humans , Liver Neoplasms/pathology , Mice , Mice, Knockout , Protein Binding , Transcription Factors/metabolism , Transcriptome , ATP-Binding Cassette Sub-Family B Member 4
18.
Genes Dev ; 29(4): 394-408, 2015 Feb 15.
Article En | MEDLINE | ID: mdl-25637355

The transcription factor (TF) interferon regulatory factor 8 (IRF8) controls both developmental and inflammatory stimulus-inducible genes in macrophages, but the mechanisms underlying these two different functions are largely unknown. One possibility is that these different roles are linked to the ability of IRF8 to bind alternative DNA sequences. We found that IRF8 is recruited to distinct sets of DNA consensus sequences before and after lipopolysaccharide (LPS) stimulation. In resting cells, IRF8 was mainly bound to composite sites together with the master regulator of myeloid development PU.1. Basal IRF8-PU.1 binding maintained the expression of a broad panel of genes essential for macrophage functions (such as microbial recognition and response to purines) and contributed to basal expression of many LPS-inducible genes. After LPS stimulation, increased expression of IRF8, other IRFs, and AP-1 family TFs enabled IRF8 binding to thousands of additional regions containing low-affinity multimerized IRF sites and composite IRF-AP-1 sites, which were not premarked by PU.1 and did not contribute to the basal IRF8 cistrome. While constitutively expressed IRF8-dependent genes contained only sites mediating basal IRF8/PU.1 recruitment, inducible IRF8-dependent genes contained variable combinations of constitutive and inducible sites. Overall, these data show at the genome scale how the same TF can be linked to constitutive and inducible gene regulation via distinct combinations of alternative DNA-binding sites.


Gene Expression Regulation , Interferon Regulatory Factors/metabolism , Macrophages/metabolism , Animals , Cells, Cultured , Interferon Regulatory Factors/genetics , Lipopolysaccharides/pharmacology , Macrophages/cytology , Macrophages/drug effects , Mice , Mice, Inbred C57BL , Protein Binding
19.
Nat Commun ; 5: 3850, 2014 May 13.
Article En | MEDLINE | ID: mdl-24819516

Hepatocellular carcinoma (HCC) is almost invariably associated with an underlying inflammatory state, whose direct contribution to the acquisition of critical genomic changes is unclear. Here we map acquired genomic alterations in human and mouse HCCs induced by defects in hepatocyte biliary transporters, which expose hepatocytes to bile salts and cause chronic inflammation that develops into cancer. In both human and mouse cancer genomes, we find few somatic point mutations with no impairment of cancer genes, but massive gene amplification and rearrangements. This genomic landscape differs from that of virus- and alcohol-associated liver cancer. Copy-number gains preferentially occur at late stages of cancer development and frequently target the MAPK signalling pathway, and in particular direct regulators of JNK. The pharmacological inhibition of JNK retards cancer progression in the mouse. Our study demonstrates that intrahepatic cholestasis leading to hepatocyte exposure to bile acids and inflammation promotes cancer through genomic modifications that can be distinguished from those determined by other aetiological factors.


Carcinoma, Hepatocellular/genetics , Cholestasis, Intrahepatic/genetics , DNA Copy Number Variations/genetics , Gene Amplification , Hepatocytes/metabolism , Liver Neoplasms, Experimental/genetics , Liver Neoplasms/genetics , MAP Kinase Signaling System/genetics , ATP Binding Cassette Transporter, Subfamily B/genetics , ATP Binding Cassette Transporter, Subfamily B, Member 11 , ATP-Binding Cassette Transporters/deficiency , ATP-Binding Cassette Transporters/genetics , Animals , Child , Child, Preschool , Female , Humans , Infant , Male , Mice , Mice, Knockout , ATP-Binding Cassette Sub-Family B Member 4
20.
Mol Cell ; 54(5): 844-857, 2014 Jun 05.
Article En | MEDLINE | ID: mdl-24813947

Transcription factors (TFs) preferentially bind sites contained in regions of computationally predicted high nucleosomal occupancy, suggesting that nucleosomes are gatekeepers of TF binding sites. However, because of their complexity mammalian genomes contain millions of randomly occurring, unbound TF consensus binding sites. We hypothesized that the information controlling nucleosome assembly may coincide with the information that enables TFs to bind cis-regulatory elements while ignoring randomly occurring sites. Hence, nucleosomes would selectively mask genomic sites that can be contacted by TFs and thus be potentially functional. The hematopoietic pioneer TF Pu.1 maintained nucleosome depletion at macrophage-specific enhancers that displayed a broad range of nucleosome occupancy in other cell types and in reconstituted chromatin. We identified a minimal set of DNA sequence and shape features that accurately predicted both Pu.1 binding and nucleosome occupancy genome-wide. These data reveal a basic organizational principle of mammalian cis-regulatory elements whereby TF recruitment and nucleosome deposition are controlled by overlapping DNA sequence features.


Enhancer Elements, Genetic , Nucleosomes/genetics , Proto-Oncogene Proteins/metabolism , Trans-Activators/metabolism , Animals , Base Sequence , Binding Sites , Cells, Cultured , Consensus Sequence , Gene Expression Regulation , Gene Knockdown Techniques , Humans , Mice , Models, Genetic , Nucleosomes/metabolism , Proto-Oncogene Proteins/genetics , RNA, Small Interfering/genetics , Sequence Analysis, DNA , Support Vector Machine , Trans-Activators/genetics
...