Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 29
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.
Genome Res ; 33(8): 1409-1423, 2023 08.
Article in English | MEDLINE | ID: mdl-37730438

ABSTRACT

Krüppel-associated box (KRAB) domain-containing zinc finger proteins (KZFPs) are one of the largest groups of transcription factors encoded by tetrapods, with 378 members in human alone. KZFP genes are often grouped in clusters reflecting amplification by gene and segment duplication since the gene family first emerged more than 400 million years ago. Previous work has revealed that many KZFPs recognize transposable element (TE)-embedded sequences as genomic targets, and that KZFPs facilitate the co-option of the regulatory potential of TEs for the benefit of the host. Here, we present a comprehensive survey of the genetic features and genomic targets of human KZFPs, notably completing past analyses by adding data on close to a hundred family members. General principles emerge from our study of the TE-KZFP regulatory system, which point to multipronged evolutionary mechanisms underlaid by highly complex and combinatorial modes of action with strong influences on human speciation.


Subject(s)
Transcription Factors , Zinc Fingers , Humans , Zinc Fingers/genetics , Transcription Factors/genetics , Biological Evolution , DNA Transposable Elements/genetics , Genomics
3.
Development ; 147(2)2020 01 23.
Article in English | MEDLINE | ID: mdl-31806660

ABSTRACT

Some of the earliest transcripts produced in fertilized human and mouse oocytes code for DUX, a double homeodomain protein that promotes embryonic genome activation (EGA). Deleting Dux by genome editing at the one- to two-cell stage in the mouse impairs EGA and blastocyst maturation. Here, we demonstrate that mice carrying homozygous Dux deletions display markedly reduced expression of DUX target genes and defects in both pre- and post-implantation development, with, notably, a disruption of the pace of the first few cell divisions and significant rates of late embryonic mortality. However, some Dux-/- embryos give rise to viable pups, indicating that DUX is important but not strictly essential for embryogenesis.


Subject(s)
Gene Expression Regulation, Developmental , Genome , Homeodomain Proteins/metabolism , Zygote/metabolism , Animals , Crosses, Genetic , Down-Regulation/genetics , Embryo, Mammalian/metabolism , Embryonic Development/genetics , Female , Genotype , Homeodomain Proteins/genetics , Male , Mice , Mice, Transgenic , Mouse Embryonic Stem Cells/metabolism
4.
Cell ; 135(6): 1118-29, 2008 Dec 12.
Article in English | MEDLINE | ID: mdl-19062086

ABSTRACT

Bone marrow hematopoietic stem cells (HSCs) are crucial to maintain lifelong production of all blood cells. Although HSCs divide infrequently, it is thought that the entire HSC pool turns over every few weeks, suggesting that HSCs regularly enter and exit cell cycle. Here, we combine flow cytometry with label-retaining assays (BrdU and histone H2B-GFP) to identify a population of dormant mouse HSCs (d-HSCs) within the lin(-)Sca1+cKit+CD150+CD48(-)CD34(-) population. Computational modeling suggests that d-HSCs divide about every 145 days, or five times per lifetime. d-HSCs harbor the vast majority of multilineage long-term self-renewal activity. While they form a silent reservoir of the most potent HSCs during homeostasis, they are efficiently activated to self-renew in response to bone marrow injury or G-CSF stimulation. After re-establishment of homeostasis, activated HSCs return to dormancy, suggesting that HSCs are not stochastically entering the cell cycle but reversibly switch from dormancy to self-renewal under conditions of hematopoietic stress.


Subject(s)
Adult Stem Cells/cytology , Hematopoietic Stem Cells/cytology , Adult Stem Cells/physiology , Animals , Antigens, Differentiation/metabolism , Bone Marrow/physiology , Bromouracil/analogs & derivatives , Fluorouracil/metabolism , Green Fluorescent Proteins , Hematopoietic Stem Cells/physiology , Homeostasis , Mice , Mice, Transgenic , Uridine/analogs & derivatives , Uridine/metabolism
5.
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
6.
Mol Cell ; 44(3): 361-72, 2011 Nov 04.
Article in English | MEDLINE | ID: mdl-22055183

ABSTRACT

The maintenance of H3K9 and DNA methylation at imprinting control regions (ICRs) during early embryogenesis is key to the regulation of imprinted genes. Here, we reveal that ZFP57, its cofactor KAP1, and associated effectors bind selectively to the H3K9me3-bearing, DNA-methylated allele of ICRs in ES cells. KAP1 deletion induces a loss of heterochromatin marks at ICRs, whereas deleting ZFP57 or DNMTs leads to ICR DNA demethylation. Accordingly, we find that ZFP57 and KAP1 associated with DNMTs and hemimethylated DNA-binding NP95. Finally, we identify the methylated TGCCGC hexanucleotide as the motif that is recognized by ZFP57 in all ICRs and in several tens of additional loci, several of which are at least ZFP57-dependently methylated in ES cells. These results significantly advance our understanding of imprinting and suggest a general mechanism for the protection of specific loci against the wave of DNA demethylation that affects the mammalian genome during early embryogenesis.


Subject(s)
Chromatin Assembly and Disassembly , DNA Methylation , Embryonic Stem Cells/metabolism , Gene Expression Regulation, Developmental , Genomic Imprinting , Nuclear Proteins/metabolism , Nucleotide Motifs , Repressor Proteins/metabolism , Animals , Base Sequence , Binding Sites , CCAAT-Enhancer-Binding Proteins , Cell Line , Chromosomal Proteins, Non-Histone/metabolism , DNA Modification Methylases/metabolism , Gene Knockout Techniques , Histone-Lysine N-Methyltransferase , Mice , Mice, Inbred C57BL , Molecular Sequence Data , Nuclear Proteins/genetics , Protein Methyltransferases/metabolism , Repressor Proteins/genetics , Tripartite Motif-Containing Protein 28 , Ubiquitin-Protein Ligases
7.
Hepatology ; 66(1): 235-251, 2017 07.
Article in English | MEDLINE | ID: mdl-28370258

ABSTRACT

Hepatocellular carcinoma (HCC) represents the fifth-most common form of cancer worldwide and carries a high mortality rate attributed to lack of effective treatment. Males are 8 times more likely to develop HCC than females, an effect largely driven by sex hormones, albeit through still poorly understood mechanisms. We previously identified TRIM28 (tripartite protein 28), a scaffold protein capable of recruiting a number of chromatin modifiers, as a crucial mediator of sexual dimorphism in the liver. Trim28hep-/- mice display sex-specific transcriptional deregulation of a wide range of bile and steroid metabolism genes and development of liver adenomas in males. We now demonstrate that obesity and aging precipitate alterations of TRIM28-dependent transcriptional dynamics, leading to a metabolic infection state responsible for highly penetrant male-restricted hepatic carcinogenesis. Molecular analyses implicate aberrant androgen receptor stimulation, biliary acid disturbances, and altered responses to gut microbiota in the pathogenesis of Trim28hep-/- -associated HCC. Correspondingly, androgen deprivation markedly attenuates the frequency and severity of tumors, and raising animals under axenic conditions completely abrogates their abnormal phenotype, even upon high-fat diet challenge. CONCLUSION: This work underpins how discrete polyphenic traits in epigenetically metastable conditions can contribute to a cancer-prone state and more broadly provides new evidence linking hormonal imbalances, metabolic disturbances, gut microbiota, and cancer. (Hepatology 2017;66:235-251).


Subject(s)
Carcinogenesis/pathology , Carcinoma, Hepatocellular/genetics , Genomic Instability , Liver Neoplasms/genetics , Repressor Proteins/genetics , Aging/genetics , Animals , Carcinoma, Hepatocellular/pathology , Diet, High-Fat , Disease Models, Animal , Epigenomics/methods , Female , Liver Neoplasms/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Oxidative Stress , Phenotype , Random Allocation , Risk Assessment , Risk Factors , Tripartite Motif-Containing Protein 28
8.
Development ; 140(3): 519-29, 2013 Feb 01.
Article in English | MEDLINE | ID: mdl-23293284

ABSTRACT

Endogenous retroviruses (ERVs) undergo de novo DNA methylation during the first few days of mammalian embryogenesis, although the factors that control the targeting of this process are largely unknown. We asked whether KAP1 (KRAB-associated protein 1) is involved in this mechanism because of its previously defined role in maintaining the silencing of ERVs through the histone methyltransferase ESET and histone H3 lysine 9 trimethylation. Here, we demonstrate that introduced ERV sequences are sufficient to direct rapid de novo methylation of a flanked promoter in embryonic stem (ES) cells. This mechanism requires the presence of an ERV sequence-recognizing KRAB zinc-finger protein (ZFP) and both KAP1 and ESET. Furthermore, this process can also take place on a strong cellular promoter and leads to methylation signatures that are subsequently maintained in vivo throughout embryogenesis. Finally, we show that methylation of ERVs residing in the genome is affected by knockout of KAP1 in early embryos. KRAB-ZFPs, KAP1 and ESET are thus likely to be responsible for the early embryonic instatement of stable epigenetic marks at ERV-containing loci.


Subject(s)
DNA Methylation , DNA, Viral/metabolism , Endogenous Retroviruses/metabolism , Histone-Lysine N-Methyltransferase/metabolism , Nuclear Proteins/metabolism , Repressor Proteins/metabolism , Animals , Animals, Genetically Modified , DNA, Viral/genetics , Embryo, Mammalian/cytology , Embryo, Mammalian/metabolism , Embryo, Mammalian/virology , Embryonic Stem Cells/cytology , Embryonic Stem Cells/metabolism , Embryonic Stem Cells/virology , Endogenous Retroviruses/enzymology , Endogenous Retroviruses/genetics , Gene Expression Regulation, Developmental , Gene Knockout Techniques , Gene Silencing , Gene Transfer Techniques , Genetic Vectors/genetics , Genetic Vectors/metabolism , HEK293 Cells , Histone-Lysine N-Methyltransferase/genetics , Histones/genetics , Histones/metabolism , Humans , Lentivirus/genetics , Lentivirus/metabolism , Mice , Nuclear Proteins/genetics , Promoter Regions, Genetic , Repressor Proteins/genetics , Transcriptome , Transfection , Tripartite Motif-Containing Protein 28
9.
Nature ; 458(7240): 904-8, 2009 Apr 16.
Article in English | MEDLINE | ID: mdl-19212321

ABSTRACT

Maintenance of the blood system is dependent on dormant haematopoietic stem cells (HSCs) with long-term self-renewal capacity. After injury these cells are induced to proliferate to quickly re-establish homeostasis. The signalling molecules promoting the exit of HSCs out of the dormant stage remain largely unknown. Here we show that in response to treatment of mice with interferon-alpha (IFNalpha), HSCs efficiently exit G(0) and enter an active cell cycle. HSCs respond to IFNalpha treatment by the increased phosphorylation of STAT1 and PKB/Akt (also known as AKT1), the expression of IFNalpha target genes, and the upregulation of stem cell antigen-1 (Sca-1, also known as LY6A). HSCs lacking the IFNalpha/beta receptor (IFNAR), STAT1 (ref. 3) or Sca-1 (ref. 4) are insensitive to IFNalpha stimulation, demonstrating that STAT1 and Sca-1 mediate IFNalpha-induced HSC proliferation. Although dormant HSCs are resistant to the anti-proliferative chemotherapeutic agent 5-fluoro-uracil, HSCs pre-treated (primed) with IFNalpha and thus induced to proliferate are efficiently eliminated by 5-fluoro-uracil exposure in vivo. Conversely, HSCs chronically activated by IFNalpha are functionally compromised and are rapidly out-competed by non-activatable Ifnar(-/-) cells in competitive repopulation assays. Whereas chronic activation of the IFNalpha pathway in HSCs impairs their function, acute IFNalpha treatment promotes the proliferation of dormant HSCs in vivo. These data may help to clarify the so far unexplained clinical effects of IFNalpha on leukaemic cells, and raise the possibility for new applications of type I interferons to target cancer stem cells.


Subject(s)
Cell Cycle/drug effects , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/drug effects , Interferon-alpha/pharmacology , Animals , Antigens, Ly/metabolism , Cell Count , Cell Proliferation/drug effects , Fluorouracil/pharmacology , Membrane Proteins/deficiency , Membrane Proteins/metabolism , Mice , Mice, Inbred C57BL , Phosphorylation/drug effects , Receptor, Interferon alpha-beta/deficiency , Receptor, Interferon alpha-beta/metabolism , STAT1 Transcription Factor/deficiency , STAT1 Transcription Factor/metabolism , Signal Transduction/drug effects , Up-Regulation/drug effects
10.
Blood ; 119(20): 4675-85, 2012 May 17.
Article in English | MEDLINE | ID: mdl-22452978

ABSTRACT

Chromatin remodeling is fundamental for B-cell differentiation. In the present study, we explored the role of KAP1, the cofactor of KRAB-ZFP transcriptional repressors, in this process. B-lymphoid-specific Kap1-KO mice displayed reduced numbers of mature B cells, lower steady-state levels of Abs, and accelerated rates of decay of neutralizing Abs after viral immunization. Transcriptome analyses of Kap1-deleted B splenocytes revealed an up-regulation of PTEN, the enzymatic counteractor of PIK3 signaling, and of genes encoding DNA-damage response factors, cell-cycle regulators, and chemokine receptors. ChIP/seq studies established that KAP1 bound at or close to several of these genes and controlled chromatin status at their promoters. Genome wide, KAP1 binding sites lacked active B cell-specific enhancers and were enriched in repressive histone marks, further supporting a role for this molecule in gene silencing in vivo. Likely responsible for tethering KAP1 to at least some of these targets, a discrete subset of KRAB-ZFPs is enriched in B lymphocytes. Our results therefore reveal the role of KRAB/KAP1-mediated epigenetic regulation in B-cell development and homeostasis.


Subject(s)
B-Lymphocytes/physiology , Cell Differentiation/genetics , Lymphocytes/physiology , Nuclear Proteins/physiology , Repressor Proteins/physiology , Animals , Antibody Formation/genetics , Antibody Formation/immunology , B-Lymphocytes/immunology , B-Lymphocytes/metabolism , Bacterial Proteins/genetics , Cell Differentiation/immunology , Cell Differentiation/physiology , Chromatin/metabolism , Epigenesis, Genetic/genetics , Epigenesis, Genetic/immunology , Epigenesis, Genetic/physiology , Gene Expression Profiling , Gene Expression Regulation , Gene Regulatory Networks/genetics , Gene Regulatory Networks/physiology , Luminescent Proteins/genetics , Lymphocyte Count , Lymphocytes/immunology , Lymphocytes/metabolism , Mice , Mice, Inbred C57BL , Mice, Transgenic , Microarray Analysis , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Repressor Proteins/genetics , Repressor Proteins/metabolism , Tripartite Motif-Containing Protein 28
11.
Sci Adv ; 10(37): eado1662, 2024 Sep 13.
Article in English | MEDLINE | ID: mdl-39270011

ABSTRACT

Long known as the site of ribosome biogenesis, the nucleolus is increasingly recognized for its role in shaping three-dimensional (3D) genome organization. Still, the mechanisms governing the targeting of selected regions of the genome to nucleolus-associated domains (NADs) remain enigmatic. Here, we reveal the essential role of ZNF274, a SCAN-bearing member of the Krüppel-associated box (KRAB)-containing zinc finger protein (KZFP) family, in sequestering lineage-specific gene clusters within NADs. Ablation of ZNF274 triggers transcriptional activation across entire genomic neighborhoods-encompassing, among others, protocadherin and KZFP-encoding genes-with loss of repressive chromatin marks, altered the 3D genome architecture and de novo CTCF binding. Mechanistically, ZNF274 anchors target DNA sequences at the nucleolus and facilitates their compartmentalization via a previously uncharted function of the SCAN domain. Our findings illuminate the mechanisms underlying NAD organization and suggest that perinucleolar entrapment into repressive hubs constrains the activation of tandemly arrayed genes to enable selective expression and modulate cell differentiation programs during development.


Subject(s)
Cell Nucleolus , Multigene Family , Cell Nucleolus/metabolism , Cell Nucleolus/genetics , Animals , Humans , Mice , CCCTC-Binding Factor/metabolism , CCCTC-Binding Factor/genetics , Chromatin/metabolism , Chromatin/genetics , Cell Lineage/genetics , Zinc Fingers/genetics , Cell Differentiation/genetics , Protein Binding
12.
Cancer Res ; 84(6): 808-826, 2024 Mar 15.
Article in English | MEDLINE | ID: mdl-38345497

ABSTRACT

Heterochromatin loss and genetic instability enhance cancer progression by favoring clonal diversity, yet uncontrolled replicative stress leads to mitotic catastrophe and inflammatory responses that promote immune rejection. KRAB domain-containing zinc finger proteins (KZFP) contribute to heterochromatin maintenance at transposable elements (TE). Here, we identified an association of upregulation of a cluster of primate-specific KZFPs with poor prognosis, increased copy-number alterations, and changes in the tumor microenvironment in diffuse large B-cell lymphoma (DLBCL). Depleting two of these KZFPs targeting evolutionarily recent TEs, ZNF587 and ZNF417, impaired the proliferation of cells derived from DLBCL and several other tumor types. ZNF587 and ZNF417 depletion led to heterochromatin redistribution, replicative stress, and cGAS-STING-mediated induction of an interferon/inflammatory response, which enhanced susceptibility to macrophage-mediated phagocytosis and increased surface expression of HLA-I, together with presentation of a neoimmunopeptidome. Thus, cancer cells can exploit KZFPs to dampen TE-originating surveillance mechanisms, which likely facilitates clonal expansion, diversification, and immune evasion. SIGNIFICANCE: Upregulation of a cluster of primate-specific KRAB zinc finger proteins in cancer cells prevents replicative stress and inflammation by regulating heterochromatin maintenance, which could facilitate the development of improved biomarkers and treatments.


Subject(s)
Heterochromatin , Neoplasms , Animals , Heterochromatin/genetics , Zinc Fingers/genetics , DNA Transposable Elements , Primates/genetics , Inflammation/genetics , Neoplasms/genetics
13.
J Biol Chem ; 287(30): 25361-9, 2012 Jul 20.
Article in English | MEDLINE | ID: mdl-22605343

ABSTRACT

The study of chromatin and its regulators is key to understanding and manipulating transcription. We previously exploited the Krüppel-associated box (KRAB) transcriptional repressor domain, present in hundreds of vertebrate-specific zinc finger proteins, to assess the effect of its binding to gene bodies. These experiments revealed that the ectopic and doxycycline (dox)-controlled tet repressor KRAB fusion protein (tTRKRAB) can induce reversible and long-range silencing of cellular promoters. Here, we extend this system to in vivo applications and use tTRKRAB to achieve externally controllable repression of an endogenous mouse locus. We employed lentiviral-mediated transgenesis with promoterless TetO-containing gene traps to engineer a mouse line where the endogenous kinesin family member 2A (Kif2A) promoter drives a YFP reporter gene. When these mice were crossed to animals expressing the TetO-binding tTRKRAB repressor, this regulator was recruited to the Kif2A locus, and YFP expression was reduced. This effect was reversed when dox was given to embryos or adult mice, demonstrating that the cellular Kif2A promoter was only silenced upon repressor binding. Molecular analyses confirmed that tTRKRAB induced transcriptional repression through the spread of H3K9me3-containing heterochromatin, without DNA methylation of the trapped Kif2A promoter. Therefore, we demonstrate that targeting of tTRKRAB to a gene body in vivo results in reversible transcriptional repression through the spreading of facultative heterochromatin. This finding not only sheds light on KRAB-mediated transcriptional processes, but also suggests approaches for the externally controllable and reversible modulation of chromatin and transcription in vivo.


Subject(s)
Carrier Proteins/metabolism , Chromatin Assembly and Disassembly/physiology , Genetic Loci/physiology , Nuclear Proteins/metabolism , Promoter Regions, Genetic/physiology , Repressor Proteins/metabolism , Transcription, Genetic/physiology , Animals , Carrier Proteins/genetics , Heterochromatin/genetics , Heterochromatin/metabolism , Kinesins/biosynthesis , Kinesins/genetics , Mice , Mice, Transgenic , Nuclear Proteins/genetics , Protein Structure, Tertiary , Repressor Proteins/biosynthesis , Repressor Proteins/genetics
14.
Hepatology ; 56(4): 1279-90, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22684873

ABSTRACT

UNLABELLED: The liver is characterized by sexually dimorphic gene expression translating into sex-specific differences in lipid, drug, steroid hormone, and xenobiotic metabolism, with distinct responses of males and females to environmental challenges. Here, we investigated the role of the Krüppel-associated box (KRAB)-associated protein 1 (KAP1) epigenetic regulator in this process. Liver-specific KAP1 knockout (KO) led to strikingly sexually dimorphic phenotypic disturbances, including male-predominant steatosis and hepatic tumors with up-regulation of protein kinase B and extracellular signal-related kinases 1/2 mitogen-activated protein kinase signaling. This correlated with the sex-specific transcriptional dysregulation of a wide range of metabolic genes, notably those involved in retinol and sex hormone processing as well as in detoxification. Furthermore, chromatin immunoprecipitation followed by deep sequencing indicated that a number of dysregulated genes are direct targets of the KRAB/KAP1 repression system. Those genes include sexually dimorphic cytochrome P 450 Cyp2d9, glutathione S-transferase π, Cyp2a, Cyp2b, and Cyp3a gene clusters. Additionally, we identified a male-restricted KAP1-binding site in the fat-specific protein 27 gene, correlating with its male-predominant up-regulation upon Kap1 deletion, suggesting that the latter might be an important trigger in the development of male-specific hepatosteatosis and secondary tumorigenesis. CONCLUSION: This work reveals KRAB/KAP1-mediated transcriptional regulation as a central event in metabolic control hormones, drugs, and xenobiotics in the liver and further links disturbances in these processes with hepatic carcinogenesis.


Subject(s)
Adenoma/genetics , Cell Transformation, Neoplastic/genetics , Fatty Liver/genetics , Genetic Predisposition to Disease , Liver Neoplasms/genetics , Nuclear Proteins/metabolism , Repressor Proteins/metabolism , Adenoma/pathology , Animals , Biopsy, Needle , Cell Transformation, Neoplastic/pathology , DNA-Binding Proteins/genetics , Disease Models, Animal , Fatty Liver/pathology , Female , Gene Expression Regulation , Immunohistochemistry , Liver Neoplasms/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Random Allocation , Sensitivity and Specificity , Sex Factors , Tripartite Motif-Containing Protein 28 , Zinc Fingers/genetics
15.
FASEB J ; 26(11): 4561-75, 2012 Nov.
Article in English | MEDLINE | ID: mdl-22872677

ABSTRACT

Chromatin remodeling at specific genomic loci controls lymphoid differentiation. Here, we investigated the role played in this process by Kruppel-associated box (KRAB)-associated protein 1 (KAP1), the universal cofactor of KRAB-zinc finger proteins (ZFPs), a tetrapod-restricted family of transcriptional repressors. T-cell-specific Kap1-deleted mice displayed a significant expansion of immature thymocytes, imbalances in CD4(+)/CD8(+) cell ratios, and altered responses to TCR and TGFß stimulation when compared to littermate KAP1 control mice. Transcriptome and chromatin studies revealed that KAP1 binds T-cell-specific cis-acting regulatory elements marked by the H3K9me3 repressive mark and enriched in Ikaros/NuRD complexes. Also, KAP1 directly controls the expression of several genes involved in TCR and cytokine signaling. Among these, regulation of FoxO1 seems to play a major role in this system. Likely responsible for tethering KAP1 to at least part of its genomic targets, a small number of KRAB-ZFPs are selectively expressed in T-lymphoid cells. These results reveal the so far unsuspected yet important role of KAP1-mediated epigenetic regulation in T-lymphocyte differentiation and activation.


Subject(s)
Gene Expression Regulation/physiology , Nuclear Proteins/metabolism , Repressor Proteins/metabolism , T-Lymphocytes/physiology , Animals , Binding Sites , CD4-Positive T-Lymphocytes/cytology , CD4-Positive T-Lymphocytes/physiology , CD8-Positive T-Lymphocytes/cytology , CD8-Positive T-Lymphocytes/physiology , DNA/genetics , DNA/metabolism , Epigenesis, Genetic , Mice , Mice, Knockout , Nuclear Proteins/genetics , Oligonucleotide Array Sequence Analysis , Phenotype , Phylogeny , Protein Binding , RNA/genetics , RNA/metabolism , Repressor Proteins/genetics , T-Lymphocytes/cytology , Transcriptome , Tripartite Motif-Containing Protein 28
16.
Nat Commun ; 13(1): 7178, 2022 11 23.
Article in English | MEDLINE | ID: mdl-36418324

ABSTRACT

The human genome contains more than 4.5 million inserts derived from transposable elements (TEs), the result of recurrent waves of invasion and internal propagation throughout evolution. For new TE copies to be inherited, they must become integrated in the genome of the germline or pre-implantation embryo, which requires that their source TE be expressed at these stages. Accordingly, many TEs harbor DNA binding sites for the pluripotency factors OCT4, NANOG, SOX2, and KLFs and are transiently expressed during embryonic genome activation. Here, we describe how many primate-restricted TEs have additional binding sites for lineage-specific transcription factors driving their expression during human gastrulation and later steps of fetal development. These TE integrants serve as lineage-specific enhancers fostering the transcription, amongst other targets, of KRAB-zinc finger proteins (KZFPs) of comparable evolutionary age, which in turn corral the activity of TE-embedded regulatory sequences in a similarly lineage-restricted fashion. Thus, TEs and their KZFP controllers play broad roles in shaping transcriptional networks during early human development.


Subject(s)
DNA Transposable Elements , Gene Regulatory Networks , Animals , Humans , DNA Transposable Elements/genetics , Primates/genetics , Transcription Factors/genetics , Transcription Factors/metabolism , Genome, Human
17.
Nat Commun ; 13(1): 4913, 2022 08 20.
Article in English | MEDLINE | ID: mdl-35987910

ABSTRACT

The treatment of colorectal cancer (CRC) is an unmet medical need in absence of early diagnosis. Here, upon characterizing cancer-specific transposable element-driven transpochimeric gene transcripts (TcGTs) produced by this tumor in the SYSCOL cohort, we find that expression of the hominid-restricted retrogene POU5F1B through aberrant activation of a primate-specific endogenous retroviral promoter is a strong negative prognostic biomarker. Correlating this observation, we demonstrate that POU5F1B fosters the proliferation and metastatic potential of CRC cells. We further determine that POU5F1B, in spite of its phylogenetic relationship with the POU5F1/OCT4 transcription factor, is a membrane-enriched protein that associates with protein kinases and known targets or interactors as well as with cytoskeleton-related molecules, and induces intracellular signaling events and the release of trans-acting factors involved in cell growth and cell adhesion. As POU5F1B is an apparently non-essential gene only lowly expressed in normal tissues, and as POU5F1B-containing TcGTs are detected in other tumors besides CRC, our data provide interesting leads for the development of cancer therapies.


Subject(s)
Colorectal Neoplasms , Genes, Homeobox , Homeodomain Proteins , Animals , Cell Line, Tumor , Cell Movement/genetics , Cell Proliferation/genetics , Colorectal Neoplasms/pathology , Gene Expression Regulation, Neoplastic , Humans , Neoplasm Metastasis , Phylogeny
18.
Stem Cells ; 28(8): 1390-8, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20641037

ABSTRACT

Hematopoietic stem cells (HSC) are probably the best understood somatic stem cells and often serve as a paradigm for other stem cells. Nevertheless, most current techniques to genetically manipulate them in vivo are either constitutive and/or induced in settings of hematopoietic stress such as after irradiation. Here, we present a conditional expression system that allows for externally controllable transgenesis and knockdown in resident HSCs, based on a lentiviral vector containing a tet-O sequence and a transgenic mouse line expressing a doxycyclin-regulated tTR-KRAB repressor protein. HSCs harvested from tTR-KRAB mice are transduced with the lentiviral vector containing a cDNA (i.e., Green Fluorescent Protein (GFP)) and/or shRNA (i.e., p53) of interest and then transplanted into lethally irradiated recipients. While the vector is effectively repressed by tTR-KRAB during homing and engraftment, robust GFP/shp53 expression is induced on doxycyclin treatment in HSCs and their progeny. Doxycylin-controllable transcription is maintained on serial transplantation, indicating that repopulating HSCs are stably modified by this approach. In summary, this easy to implement conditional system provides inducible and reversible overexpression or knock down of genes in resident HSCs in vivo using a drug devoid of toxic or activating effects.


Subject(s)
Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/metabolism , RNA, Small Interfering/physiology , Animals , Apoptosis/drug effects , Apoptosis/genetics , Cell Cycle/drug effects , Cell Cycle/genetics , Cell Proliferation , Cells, Cultured , Doxycycline/pharmacology , Female , Flow Cytometry , Gene Expression/drug effects , Gene Expression/genetics , Genetic Vectors/genetics , Hematopoietic Stem Cell Transplantation , Hematopoietic Stem Cells/drug effects , Humans , Lentivirus/genetics , Mice , Mice, Inbred C57BL , Mice, Transgenic , RNA, Small Interfering/genetics , Reverse Transcriptase Polymerase Chain Reaction , Tumor Suppressor Protein p53/genetics
19.
Hepatology ; 48(4): 1302-11, 2008 Oct.
Article in English | MEDLINE | ID: mdl-18798339

ABSTRACT

UNLABELLED: In the adult liver, 1,4-bis[2-(3,5-dichloropyridyloxy)]benzene (TCPOBOP), an agonist of the constitutive androstane receptor (CAR, NR1I3), produces rapid hepatomegaly in the absence of injury. In this study, we identify c-Myc as a gene induced by CAR and demonstrate that TCPOBOP-induced proliferation of hepatocytes depends on c-Myc function. Moreover, the TCPOBOP-induced cell cycle program (Cdc2, cyclins, MCM proteins, Cdc20, and genes implicated in the spindle assembly checkpoint) is severely impaired in c-Myc mutant livers. Strikingly, many of these genes overlap with a program controlled by the forkhead transcription factor FoxM1, known to control progression through S-phase and mitosis. Indeed, FoxM1 is also induced by TCPOBOP. Moreover, we show that c-Myc binds to the FoxM1 promoter in a TCPOBOP-dependent manner, suggesting a CAR --> c-Myc --> FoxM1 pathway downstream of TCPOBOP. CONCLUSION: Collectively, this study identifies c-Myc and FoxM1 mediated proliferative programs as key mediators of TCPOBOP-CAR induced direct liver hyperplasia.


Subject(s)
Forkhead Transcription Factors/metabolism , Liver/metabolism , Liver/pathology , Proto-Oncogene Proteins c-myc/metabolism , Receptors, Cytoplasmic and Nuclear/metabolism , Transcription Factors/metabolism , Animals , Cell Proliferation , Constitutive Androstane Receptor , Disease Models, Animal , Forkhead Box Protein M1 , Hepatocytes/metabolism , Hepatocytes/pathology , Hyperplasia/chemically induced , Hyperplasia/metabolism , Hyperplasia/pathology , Liver/physiopathology , Mice , Mice, Transgenic , Proto-Oncogene Proteins c-myc/genetics , Pyridines , Receptors, Cytoplasmic and Nuclear/agonists , Signal Transduction , Transcription Factors/agonists
SELECTION OF CITATIONS
SEARCH DETAIL