Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
1.
Genes Dev ; 33(1-2): 49-54, 2019 01 01.
Article in English | MEDLINE | ID: mdl-30602440

ABSTRACT

Genomic imprinting is an epigenetic process regulated by germline-derived DNA methylation, causing parental origin-specific monoallelic gene expression. Zinc finger protein 57 (ZFP57) is critical for maintenance of this epigenetic memory during post-fertilization reprogramming, yet incomplete penetrance of ZFP57 mutations in humans and mice suggests additional effectors. We reveal that ZNF445/ZFP445, which we trace to the origins of imprinting, binds imprinting control regions (ICRs) in mice and humans. In mice, ZFP445 and ZFP57 act together, maintaining all but one ICR in vivo, whereas earlier embryonic expression of ZNF445 and its intolerance to loss-of-function mutations indicate greater importance in the maintenance of human imprints.


Subject(s)
DNA Methylation/genetics , Genomic Imprinting/genetics , Kruppel-Like Transcription Factors/metabolism , Transcription Factors/metabolism , Animals , Cells, Cultured , Conserved Sequence , Embryonic Stem Cells , HEK293 Cells , Humans , Kruppel-Like Transcription Factors/genetics , Mice , Mice, Inbred C57BL , Repressor Proteins , Transcription Factors/genetics
2.
Nat Methods ; 19(7): 803-811, 2022 07.
Article in English | MEDLINE | ID: mdl-35710609

ABSTRACT

The laboratory mouse ranks among the most important experimental systems for biomedical research and molecular reference maps of such models are essential informational tools. Here, we present a quantitative draft of the mouse proteome and phosphoproteome constructed from 41 healthy tissues and several lines of analyses exemplify which insights can be gleaned from the data. For instance, tissue- and cell-type resolved profiles provide protein evidence for the expression of 17,000 genes, thousands of isoforms and 50,000 phosphorylation sites in vivo. Proteogenomic comparison of mouse, human and Arabidopsis reveal common and distinct mechanisms of gene expression regulation and, despite many similarities, numerous differentially abundant orthologs that likely serve species-specific functions. We leverage the mouse proteome by integrating phenotypic drug (n > 400) and radiation response data with the proteomes of 66 pancreatic ductal adenocarcinoma (PDAC) cell lines to reveal molecular markers for sensitivity and resistance. This unique atlas complements other molecular resources for the mouse and can be explored online via ProteomicsDB and PACiFIC.


Subject(s)
Arabidopsis , Carcinoma, Pancreatic Ductal , Pancreatic Neoplasms , Animals , Arabidopsis/genetics , Carcinoma, Pancreatic Ductal/metabolism , Mass Spectrometry , Mice , Pancreatic Neoplasms/genetics , Proteome/analysis
3.
EMBO J ; 38(18): e101220, 2019 09 16.
Article in English | MEDLINE | ID: mdl-31403225

ABSTRACT

Krüppel-associated box (KRAB)-containing zinc finger proteins (KZFPs) are encoded in the hundreds by the genomes of higher vertebrates, and many act with the heterochromatin-inducing KAP1 as repressors of transposable elements (TEs) during early embryogenesis. Yet, their widespread expression in adult tissues and enrichment at other genetic loci indicate additional roles. Here, we characterized the protein interactome of 101 of the ~350 human KZFPs. Consistent with their targeting of TEs, most KZFPs conserved up to placental mammals essentially recruit KAP1 and associated effectors. In contrast, a subset of more ancient KZFPs rather interacts with factors related to functions such as genome architecture or RNA processing. Nevertheless, KZFPs from coelacanth, our most distant KZFP-encoding relative, bind the cognate KAP1. These results support a hypothetical model whereby KZFPs first emerged as TE-controlling repressors, were continuously renewed by turnover of their hosts' TE loads, and occasionally produced derivatives that escaped this evolutionary flushing by development and exaptation of novel functions.


Subject(s)
Placenta/metabolism , Repressor Proteins/metabolism , Tripartite Motif-Containing Protein 28/metabolism , Animals , DNA Transposable Elements , Evolution, Molecular , Female , Fish Proteins/metabolism , Fishes/metabolism , HEK293 Cells , Humans , Pregnancy , Protein Interaction Maps , Repressor Proteins/chemistry , Zinc Fingers
4.
Genes Dev ; 28(13): 1397-409, 2014 Jul 01.
Article in English | MEDLINE | ID: mdl-24939876

ABSTRACT

Mobile elements are important evolutionary forces that challenge genomic integrity. Long interspersed element-1 (L1, also known as LINE-1) is the only autonomous transposon still active in the human genome. It displays an unusual pattern of evolution, with, at any given time, a single active L1 lineage amplifying to thousands of copies before getting replaced by a new lineage, likely under pressure of host restriction factors, which act notably by silencing L1 expression during early embryogenesis. Here, we demonstrate that in human embryonic stem (hES) cells, KAP1 (KRAB [Krüppel-associated box domain]-associated protein 1), the master cofactor of KRAB-containing zinc finger proteins (KRAB-ZFPs) previously implicated in the restriction of endogenous retroviruses, represses a discrete subset of L1 lineages predicted to have entered the ancestral genome between 26.8 million and 7.6 million years ago. In mice, we documented a similar chronologically conditioned pattern, albeit with a much contracted time scale. We could further identify an L1-binding KRAB-ZFP, suggesting that this rapidly evolving protein family is more globally responsible for L1 recognition. KAP1 knockdown in hES cells induced the expression of KAP1-bound L1 elements, but their younger, human-specific counterparts (L1Hs) were unaffected. Instead, they were stimulated by depleting DNA methyltransferases, consistent with recent evidence demonstrating that the PIWI-piRNA (PIWI-interacting RNA) pathway regulates L1Hs in hES cells. Altogether, these data indicate that the early embryonic control of L1 is an evolutionarily dynamic process and support a model in which newly emerged lineages are first suppressed by DNA methylation-inducing small RNA-based mechanisms before KAP1-recruiting protein repressors are selected.


Subject(s)
Gene Expression Regulation , Long Interspersed Nucleotide Elements/genetics , Animals , DNA (Cytosine-5-)-Methyltransferases/metabolism , DNA Methylation , Embryonic Stem Cells , Evolution, Molecular , Humans , Mice , Repressor Proteins/genetics , Tripartite Motif-Containing Protein 28
5.
Gut ; 69(8): 1472-1482, 2020 08.
Article in English | MEDLINE | ID: mdl-32001555

ABSTRACT

OBJECTIVE: Pancreatic ductal adenocarcinoma (PDAC) still carries a dismal prognosis with an overall 5-year survival rate of 9%. Conventional combination chemotherapies are a clear advance in the treatment of PDAC; however, subtypes of the disease exist, which exhibit extensive resistance to such therapies. Genomic MYC amplifications represent a distinct subset of PDAC with an aggressive tumour biology. It is clear that hyperactivation of MYC generates dependencies that can be exploited therapeutically. The aim of the study was to find and to target MYC-associated dependencies. DESIGN: We analysed human PDAC gene expression datasets. Results were corroborated by the analysis of the small ubiquitin-like modifier (SUMO) pathway in a large PDAC cohort using immunohistochemistry. A SUMO inhibitor was used and characterised using human and murine two-dimensional, organoid and in vivo models of PDAC. RESULTS: We observed that MYC is connected to the SUMOylation machinery in PDAC. Components of the SUMO pathway characterise a PDAC subtype with a dismal prognosis and we provide evidence that hyperactivation of MYC is connected to an increased sensitivity to pharmacological SUMO inhibition. CONCLUSION: SUMO inhibitor-based therapies should be further developed for an aggressive PDAC subtype.


Subject(s)
Carcinoma, Pancreatic Ductal/genetics , Pancreatic Neoplasms/genetics , Proto-Oncogene Proteins c-myc/genetics , Small Ubiquitin-Related Modifier Proteins/genetics , Small Ubiquitin-Related Modifier Proteins/metabolism , Aged , Animals , Apoptosis , Carcinoma, Pancreatic Ductal/drug therapy , Cell Line, Tumor , Cell Proliferation , Enzyme Inhibitors/pharmacology , Esters/pharmacology , Female , Gene Amplification , Gene Expression , Humans , Male , Mice , Middle Aged , Neoplasm Transplantation , Organoids/metabolism , Pancreatic Neoplasms/drug therapy , Prognosis , Proto-Oncogene Proteins c-myc/metabolism , Pyrazoles/pharmacology , Pyrimidines/pharmacology , SUMO-1 Protein/genetics , SUMO-1 Protein/metabolism , Sulfonic Acids , Sumoylation/drug effects , Sumoylation/genetics , Transcriptome/drug effects , Ubiquitin-Activating Enzymes/antagonists & inhibitors , Ubiquitin-Activating Enzymes/genetics , Ubiquitin-Conjugating Enzymes/genetics , Ubiquitins/genetics , Ubiquitins/metabolism
6.
Nucleic Acids Res ; 46(17): 8788-8802, 2018 09 28.
Article in English | MEDLINE | ID: mdl-29955894

ABSTRACT

During cell division, maintenance of chromatin features from the parental genome requires their proper establishment on its newly synthetized copy. The loss of epigenetic marks within heterochromatin, typically enriched in repetitive elements, endangers genome stability and permits chromosomal rearrangements via recombination. However, how histone modifications associated with heterochromatin are maintained across mitosis remains poorly understood. KAP1 is known to act as a scaffold for a repressor complex that mediates local heterochromatin formation, and was previously demonstrated to play an important role during DNA repair. Accordingly, we investigated a putative role for this protein in the replication of heterochromatic regions. We first found that KAP1 associates with several DNA replication factors including PCNA, MCM3 and MCM6. We then observed that these interactions are promoted by KAP1 phosphorylation on serine 473 during S phase. Finally, we could demonstrate that KAP1 forms a complex with PCNA and the histone-lysine methyltransferase Suv39h1 to reinstate heterochromatin after DNA replication.


Subject(s)
Chromatin Assembly and Disassembly/genetics , DNA Replication/physiology , Heterochromatin/metabolism , Tripartite Motif-Containing Protein 28/physiology , Animals , Cells, Cultured , HEK293 Cells , HeLa Cells , Humans , K562 Cells , Methyltransferases/metabolism , Mice , NIH 3T3 Cells , Phosphorylation , Proliferating Cell Nuclear Antigen/metabolism , Protein Processing, Post-Translational , Repressor Proteins/metabolism
7.
Mol Ther ; 21(9): 1695-704, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23760447

ABSTRACT

Preclinical and clinical studies showed that autologous transplantation of epidermis derived from genetically modified epithelial stem cells (EpSCs) leads to long-term correction of inherited skin adhesion defects. These studies were based on potentially genotoxic retroviral vectors. We developed an alternative gene transfer strategy aimed at targeting a "safe harbor" locus, the adeno-associated virus integration site 1 (AAVS1), by zinc-finger nuclease (ZFN)-induced homologous recombination (HR). Delivery of AAVS1-specific ZFNs and a GFP-expressing HR cassette by integration-defective lentiviral (LV) vectors (IDLVs) or adenoviral (Ad) vectors resulted in targeted gene addition with an efficiency of > 20% in a human keratinocyte cell line, > 10% in immortalized keratinocytes, and < 1% in primary keratinocytes. Deep sequencing of the AAVS1 locus showed that ZFN-induced double-strand breaks are mostly repaired by nonhomologous end joining (NHEJ) in primary cells, indicating that poor induction of the HR-dependent DNA repair pathway may be a significant limitation for targeted gene integration. Skin equivalents derived from unselected keratinocyte cultures coinfected with a GFP-IDLV and a ZFN-Ad vector were grafted onto immunodeficient mice. GFP-positive clones were observed in all grafts up to 18 weeks post-transplantation. By histological and molecular analysis, we were able to demonstrate highly efficient targeting of the AAVS1 locus in human repopulating EpSCs.


Subject(s)
Dependovirus/genetics , Endonucleases/genetics , Gene Targeting , Homologous Recombination , Keratinocytes/metabolism , Stem Cells/metabolism , Virus Integration , Animals , Cell Line , Cell Transplantation , Cells, Cultured , Dependovirus/metabolism , Endonucleases/metabolism , Genetic Vectors , Humans , Mice , Transduction, Genetic , Zinc Fingers
8.
Mol Oncol ; 16(5): 1091-1118, 2022 03.
Article in English | MEDLINE | ID: mdl-34748271

ABSTRACT

Aberrant activation of the phosphoinositide 3-kinase (PI3K)/AKT/mTOR and Ras/mitogen-activated protein kinase (MAPK) pathways is a hallmark of hepatocarcinogenesis. In a subset of hepatocellular carcinomas (HCCs), PI3K/AKT/mTOR signaling dysregulation depends on phosphatidylinositol-4,5-bisphosphate 3-kinase, catalytic subunit alpha (PIK3CA) mutations, while RAS/MAPK activation is partly attributed to promoter methylation of the tumor suppressor Ras association domain-containing protein 1 (RASSF1A). To evaluate a possible cocarcinogenic effect of PIK3CA activation and RASSF1A knockout, plasmids expressing oncogenic forms of PIK3CA (E545K or H1047R mutants) were delivered to the liver of RASSF1A knockout and wild-type mice by hydrodynamic tail vein injection combined with sleeping beauty-mediated somatic integration. Transfection of either PIK3CA E545K or H1047R mutants sufficed to induce HCCs in mice irrespective of RASSF1A mutational background. The related tumors displayed a lipogenic phenotype with upregulation of fatty acid synthase and stearoyl-CoA desaturase-1 (SCD1). Galectin-1, which was commonly upregulated in preneoplastic lesions and tumors, emerged as a regulator of SCD1. Co-inhibitory treatment with PIK3CA inhibitors and the galectin-1 inhibitor OTX008 resulted in synergistic cytotoxicity in human HCC cell lines, suggesting novel therapeutic venues.


Subject(s)
Carcinoma, Hepatocellular , Liver Neoplasms , Animals , Carcinogenesis , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/pathology , Cell Line, Tumor , Class I Phosphatidylinositol 3-Kinases/genetics , Class I Phosphatidylinositol 3-Kinases/metabolism , Galectin 1/genetics , Humans , Liver Neoplasms/genetics , Liver Neoplasms/pathology , Mice , Mutation , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , TOR Serine-Threonine Kinases/metabolism , Up-Regulation
9.
Nat Cancer ; 3(3): 318-336, 2022 03.
Article in English | MEDLINE | ID: mdl-35122074

ABSTRACT

KRAS-mutant pancreatic ductal adenocarcinoma (PDAC) is highly immunosuppressive and resistant to targeted and immunotherapies. Among the different PDAC subtypes, basal-like mesenchymal PDAC, which is driven by allelic imbalance, increased gene dosage and subsequent high expression levels of oncogenic KRAS, shows the most aggressive phenotype and strongest therapy resistance. In the present study, we performed a systematic high-throughput combination drug screen and identified a synergistic interaction between the MEK inhibitor trametinib and the multi-kinase inhibitor nintedanib, which targets KRAS-directed oncogenic signaling in mesenchymal PDAC. This combination treatment induces cell-cycle arrest and cell death, and initiates a context-dependent remodeling of the immunosuppressive cancer cell secretome. Using a combination of single-cell RNA-sequencing, CRISPR screens and immunophenotyping, we show that this combination therapy promotes intratumor infiltration of cytotoxic and effector T cells, which sensitizes mesenchymal PDAC to PD-L1 immune checkpoint inhibition. Overall, our results open new avenues to target this aggressive and therapy-refractory mesenchymal PDAC subtype.


Subject(s)
Adenocarcinoma , Carcinoma, Pancreatic Ductal , Pancreatic Neoplasms , Adenocarcinoma/drug therapy , Carcinoma, Pancreatic Ductal/drug therapy , Humans , Immune Checkpoint Inhibitors , Pancreatic Neoplasms/drug therapy , Tumor Microenvironment
10.
Epigenetics Chromatin ; 11(1): 7, 2018 02 26.
Article in English | MEDLINE | ID: mdl-29482634

ABSTRACT

BACKGROUND: The KZFP/KAP1 (KRAB zinc finger proteins/KRAB-associated protein 1) system plays a central role in repressing transposable elements (TEs) and maintaining parent-of-origin DNA methylation at imprinting control regions (ICRs) during the wave of genome-wide reprogramming that precedes implantation. In naïve murine embryonic stem cells (mESCs), the genome is maintained highly hypomethylated by a combination of TET-mediated active demethylation and lack of de novo methylation, yet KAP1 is tethered by sequence-specific KZFPs to ICRs and TEs where it recruits histone and DNA methyltransferases to impose heterochromatin formation and DNA methylation. RESULTS: Here, upon removing either KAP1 or the cognate KZFP, we observed rapid TET2-dependent accumulation of 5hmC at both ICRs and TEs. In the absence of the KZFP/KAP1 complex, ICRs lost heterochromatic histone marks and underwent both active and passive DNA demethylation. For KAP1-bound TEs, 5mC hydroxylation correlated with transcriptional reactivation. Using RNA-seq, we further compared the expression profiles of TEs upon Kap1 removal in wild-type, Dnmt and Tet triple knockout mESCs. While we found that KAP1 represents the main effector of TEs repression in all three settings, we could additionally identify specific groups of TEs further controlled by DNA methylation. Furthermore, we observed that in the absence of TET proteins, activation upon Kap1 depletion was blunted for some TE integrants and increased for others. CONCLUSIONS: Our results indicate that the KZFP/KAP1 complex maintains heterochromatin and DNA methylation at ICRs and TEs in naïve embryonic stem cells partly by protecting these loci from TET-mediated demethylation. Our study further unveils an unsuspected level of complexity in the transcriptional control of the endovirome by demonstrating often integrant-specific differential influences of histone-based heterochromatin modifications, DNA methylation and 5mC oxidation in regulating TEs expression.


Subject(s)
DNA-Binding Proteins/metabolism , Mouse Embryonic Stem Cells/cytology , Retroelements , Tripartite Motif-Containing Protein 28/genetics , Animals , DNA Methylation , DNA-Binding Proteins/genetics , Dioxygenases , Gene Deletion , Gene Knockout Techniques , Genomic Imprinting , Histones/metabolism , Mice , Proto-Oncogene Proteins/metabolism , Sequence Analysis, RNA , Transcriptional Activation , Tripartite Motif-Containing Protein 28/metabolism , Zinc Fingers
11.
Nat Genet ; 49(6): 941-945, 2017 Jun.
Article in English | MEDLINE | ID: mdl-28459456

ABSTRACT

In animal embryos, transcription is mostly silent for several cell divisions, until the release of the first major wave of embryonic transcripts through so-called zygotic genome activation (ZGA). Maternally provided ZGA-triggering factors have been identified in Drosophila melanogaster and Danio rerio, but their mammalian homologs are still undefined. Here, we provide evidence that the DUX family of transcription factors is essential to this process in mice and potentially in humans. First, human DUX4 and mouse Dux are both expressed before ZGA in their respective species. Second, both orthologous proteins bind the promoters of ZGA-associated genes and activate their transcription. Third, Dux knockout in mouse embryonic stem cells (mESCs) prevents the cells from cycling through a 2-cell-like state. Finally, zygotic depletion of Dux leads to impaired early embryonic development and defective ZGA. We conclude that DUX-family proteins are key inducers of zygotic genome activation in placental mammals.


Subject(s)
Homeodomain Proteins/genetics , Zygote , Animals , Binding Sites , Cells, Cultured , Chromatin Immunoprecipitation , Dogs , Embryonic Stem Cells/physiology , Evolution, Molecular , Gene Expression Regulation, Developmental , Homeodomain Proteins/metabolism , Humans , Mice, Knockout , Mice, Transgenic , Myoblasts/cytology , Myoblasts/physiology , Promoter Regions, Genetic , Retroelements , Transcription Factors/genetics
12.
Dev Cell ; 36(6): 611-23, 2016 Mar 21.
Article in English | MEDLINE | ID: mdl-27003935

ABSTRACT

KRAB-containing zinc finger proteins (KRAB-ZFPs) are early embryonic controllers of transposable elements (TEs), which they repress with their cofactor KAP1 through histone and DNA methylation, a process thought to result in irreversible silencing. Using a target-centered functional screen, we matched murine TEs with their cognate KRAB-ZFP. We found the paralogs ZFP932 and Gm15446 to bind overlapping but distinguishable subsets of ERVK (endogenous retrovirus K), repress these elements in embryonic stem cells, and regulate secondarily the expression of neighboring genes. Most importantly, we uncovered that these KRAB-ZFPs and KAP1 control TEs in adult tissues, in cell culture and in vivo, where they partner up to modulate cellular genes. Therefore, TEs and KRAB-ZFPs establish transcriptional networks that likely regulate not only development but also many physiological events. Given the high degree of species specificity of TEs and KRAB-ZFPs, these results have important implications for understanding the biology of higher vertebrates, including humans.


Subject(s)
DNA Transposable Elements/genetics , Repressor Proteins/genetics , Repressor Proteins/metabolism , Zinc Fingers/genetics , Amino Acid Sequence , Animals , Cell Differentiation , Embryonic Stem Cells/metabolism , Endogenous Retroviruses/genetics , Endogenous Retroviruses/metabolism , Gene Expression Regulation , Humans , Mice , Mice, Knockout , Molecular Sequence Data , Nuclear Proteins/deficiency , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Repressor Proteins/deficiency , Tripartite Motif-Containing Protein 28
SELECTION OF CITATIONS
SEARCH DETAIL