Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 431
Filter
1.
Cell Cycle ; 20(5-6): 465-479, 2021.
Article in English | MEDLINE | ID: mdl-33590780

ABSTRACT

In eukaryotic cells, changes in chromatin accessibility are necessary for chromatin to maintain its highly dynamic nature at different times during the cell cycle. Histone chaperones interact with histones and regulate chromatin dynamics. Facilitates chromatin transcription (FACT) is an important histone chaperone that plays crucial roles during various cellular processes. Here, we analyze the structural characteristics of FACT, discuss how FACT regulates nucleosome/chromatin reorganization and summarize possible functions of FACT in transcription, replication, and DNA repair. The possible involvement of FACT in cell fate determination is also discussed.Abbreviations: FACT: facilitates chromatin transcription, Spt16: suppressor of Ty16, SSRP1: structure-specific recognition protein-1, NTD: N-terminal domain, DD: dimerization domain, MD: middle domain, CTD: C-terminus domain, IDD: internal intrinsically disordered domain, HMG: high mobility group, CID: C-terminal intrinsically disordered domain, Nhp6: non-histone chromosomal protein 6, RNAPII: RNA polymerase II, CK2: casein kinase 2, AID: acidic inner disorder, PIC: pre-initiation complex, IR: ionizing radiation, DDSB: DNA double-strand break, PARlation: poly ADP-ribosylation, BER: base-excision repair, UVSSA: UV-stimulated scaffold protein A, HR: homologous recombination, CAF-1: chromatin assembly factor 1, Asf1: anti-silencing factor 1, Rtt106: regulator of Ty1 transposition protein 106, H3K56ac: H3K56 acetylation, KD: knock down, SETD2: SET domain containing 2, H3K36me3: trimethylation of lysine36 in histone H3, H2Bub: H2B ubiquitination, iPSCs: induced pluripotent stem cells, ESC: embryonic stem cell, H3K4me3: trimethylation of lysine 4 on histone H3 protein subunit, CHD1: chromodomain protein.


Subject(s)
Chromatin/chemistry , Chromatin/physiology , DNA Repair/physiology , DNA-Binding Proteins/chemistry , DNA-Binding Proteins/physiology , High Mobility Group Proteins/chemistry , High Mobility Group Proteins/physiology , Transcriptional Elongation Factors/chemistry , Transcriptional Elongation Factors/physiology , Animals , Histone Chaperones/chemistry , Histone Chaperones/physiology , Humans , Nucleosomes/chemistry , Nucleosomes/physiology , Protein Binding/physiology , Protein Structure, Secondary , Transcription Factors/chemistry , Transcription Factors/physiology
2.
Nucleic Acids Res ; 48(18): 10211-10225, 2020 10 09.
Article in English | MEDLINE | ID: mdl-32894293

ABSTRACT

Endogenous retroviruses (ERVs) were usually silenced by various histone modifications on histone H3 variants and respective histone chaperones in embryonic stem cells (ESCs). However, it is still unknown whether chaperones of other histones could repress ERVs. Here, we show that H2A/H2B histone chaperone FACT plays a critical role in silencing ERVs and ERV-derived cryptic promoters in ESCs. Loss of FACT component Ssrp1 activated MERVL whereas the re-introduction of Ssrp1 rescued the phenotype. Additionally, Ssrp1 interacted with MERVL and suppressed cryptic transcription of MERVL-fused genes. Remarkably, Ssrp1 interacted with and recruited H2B deubiquitinase Usp7 to Ssrp1 target genes. Suppression of Usp7 caused similar phenotypes as loss of Ssrp1. Furthermore, Usp7 acted by deubiquitinating H2Bub and thereby repressed the expression of MERVL-fused genes. Taken together, our study uncovers a unique mechanism by which FACT complex silences ERVs and ERV-derived cryptic promoters in ESCs.


Subject(s)
DNA-Binding Proteins/physiology , Endogenous Retroviruses/genetics , High Mobility Group Proteins/physiology , Histone Chaperones/physiology , Promoter Regions, Genetic , Transcription Factors/physiology , Animals , Cell Line , Gene Expression Regulation , Histones/metabolism , Mice , Mouse Embryonic Stem Cells , Ubiquitin-Specific Peptidase 7/physiology
3.
Elife ; 92020 04 23.
Article in English | MEDLINE | ID: mdl-32324136

ABSTRACT

Germ cells are vulnerable to stress. Therefore, how organisms protect their future progeny from damage in a fluctuating environment is a fundamental question in biology. We show that in Caenorhabditis elegans, serotonin released by maternal neurons during stress ensures the viability and stress resilience of future offspring. Serotonin acts through a signal transduction pathway conserved between C. elegans and mammalian cells to enable the transcription factor HSF1 to alter chromatin in soon-to-be fertilized germ cells by recruiting the histone chaperone FACT, displacing histones, and initiating protective gene expression. Without serotonin release by maternal neurons, FACT is not recruited by HSF1 in germ cells, transcription occurs but is delayed, and progeny of stressed C. elegans mothers fail to complete development. These studies uncover a novel mechanism by which stress sensing by neurons is coupled to transcription response times of germ cells to protect future offspring.


Subject(s)
Embryonic Development/physiology , Germ Cells/physiology , Neurons/physiology , Serotonin/physiology , Stress, Physiological/physiology , Animals , Caenorhabditis elegans/physiology , Cyclic AMP-Dependent Protein Kinases/physiology , DNA-Binding Proteins/physiology , Female , Gene Expression Regulation, Developmental , Heat Shock Transcription Factors/physiology , High Mobility Group Proteins/physiology , Hot Temperature , Humans , Mice , Mice, Inbred C57BL , Signal Transduction/physiology , Transcriptional Elongation Factors/physiology
4.
Int J Mol Sci ; 21(2)2020 Jan 10.
Article in English | MEDLINE | ID: mdl-31936777

ABSTRACT

Chromatin plays a key role in regulating gene expression programs necessary for the orderly progress of development and for preventing changes in cell identity that can lead to disease. The high mobility group N (HMGN) is a family of nucleosome binding proteins that preferentially binds to chromatin regulatory sites including enhancers and promoters. HMGN proteins are ubiquitously expressed in all vertebrate cells potentially affecting chromatin function and epigenetic regulation in multiple cell types. Here, we review studies aimed at elucidating the biological function of HMGN proteins, focusing on their possible role in vertebrate development and the etiology of disease. The data indicate that changes in HMGN levels lead to cell type-specific phenotypes, suggesting that HMGN optimize epigenetic processes necessary for maintaining cell identity and for proper execution of specific cellular functions. This manuscript contains tables that can be used as a comprehensive resource for all the English written manuscripts describing research aimed at elucidating the biological function of the HMGN protein family.


Subject(s)
Epigenesis, Genetic , Gene Expression Regulation, Developmental , High Mobility Group Proteins/physiology , Animals , Chromatin , Disease , HMGN Proteins , High Mobility Group Proteins/classification , High Mobility Group Proteins/genetics , Humans , Mice , Promoter Regions, Genetic
5.
Gut ; 69(2): 329-342, 2020 02.
Article in English | MEDLINE | ID: mdl-31439637

ABSTRACT

OBJECTIVE: Facilitates Chromatin Transcription (FACT) complex is a histone chaperone participating in DNA repair-related and transcription-related chromatin dynamics. In this study, we investigated its oncogenic functions, underlying mechanisms and therapeutic implications in human hepatocellular carcinoma (HCC). DESIGN: We obtained HCC and its corresponding non-tumorous liver samples from 16 patients and identified FACT complex as the most upregulated histone chaperone by RNA-Seq. We further used CRISPR-based gene activation and knockout systems to demonstrate the functions of FACT complex in HCC growth and metastasis. Functional roles and mechanistic insights of FACT complex in oxidative stress response were investigated by ChIP assay, flow cytometry, gene expression assays and 4sU-DRB transcription elongation assay. Therapeutic effect of FACT complex inhibitor, Curaxin, was tested in both in vitro and in vivo models. RESULTS: We showed that FACT complex was remarkably upregulated in HCC and contributed to HCC progression. Importantly, we unprecedentedly revealed an indispensable role of FACT complex in NRF2-driven oxidative stress response. Oxidative stress prevented NRF2 and FACT complex from KEAP1-mediated protein ubiquitination and degradation. Stabilised NRF2 and FACT complex form a positive feedback loop; NRF2 transcriptionally activates the FACT complex, while FACT complex promotes the transcription elongation of NRF2 and its downstream antioxidant genes through facilitating rapid nucleosome disassembly for the passage of RNA polymerase. Therapeutically, Curaxin effectively suppressed HCC growth and sensitised HCC cell to sorafenib. CONCLUSION: In conclusion, our findings demonstrated that FACT complex is essential for the expeditious HCC oxidative stress response and is a potential therapeutic target for HCC treatment.


Subject(s)
Carcinoma, Hepatocellular/physiopathology , DNA-Binding Proteins/physiology , High Mobility Group Proteins/physiology , Histone Chaperones/physiology , Liver Neoplasms/physiopathology , Oxidative Stress/physiology , Transcriptional Elongation Factors/physiology , Animals , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Carbazoles/pharmacology , Carbazoles/therapeutic use , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/pathology , Carcinoma, Hepatocellular/prevention & control , Cell Cycle Proteins/deficiency , Cell Cycle Proteins/genetics , Cell Cycle Proteins/physiology , Cell Line, Tumor , Cell Movement/genetics , Cell Movement/physiology , Cell Proliferation/genetics , Cell Proliferation/physiology , DNA-Binding Proteins/antagonists & inhibitors , DNA-Binding Proteins/biosynthesis , DNA-Binding Proteins/genetics , Disease Progression , Gene Expression Regulation, Neoplastic/physiology , Gene Knockout Techniques/methods , High Mobility Group Proteins/antagonists & inhibitors , High Mobility Group Proteins/biosynthesis , High Mobility Group Proteins/genetics , Humans , Liver Neoplasms/genetics , Liver Neoplasms/pathology , Liver Neoplasms, Experimental/genetics , Liver Neoplasms, Experimental/pathology , Liver Neoplasms, Experimental/physiopathology , Liver Neoplasms, Experimental/prevention & control , Mice, Inbred BALB C , Mice, Nude , Oxidative Stress/genetics , Sorafenib/pharmacology , Sorafenib/therapeutic use , Transcription Factors/deficiency , Transcription Factors/genetics , Transcription Factors/physiology , Transcriptional Elongation Factors/antagonists & inhibitors , Transcriptional Elongation Factors/biosynthesis , Transcriptional Elongation Factors/genetics , Up-Regulation/physiology , Xenograft Model Antitumor Assays
6.
Nat Rev Immunol ; 19(11): 665-674, 2019 11.
Article in English | MEDLINE | ID: mdl-31570879

ABSTRACT

'T cell exhaustion' is a broad term that has been used to describe the response of T cells to chronic antigen stimulation, first in the setting of chronic viral infection but more recently in response to tumours. Understanding the features of and pathways to exhaustion has crucial implications for the success of checkpoint blockade and adoptive T cell transfer therapies. In this Viewpoint article, 18 experts in the field tell us what exhaustion means to them, ranging from complete lack of effector function to altered functionality to prevent immunopathology, with potential differences between cancer and chronic infection. Their responses highlight the dichotomy between terminally differentiated exhausted T cells that are TCF1- and the self-renewing TCF1+ population from which they derive. These TCF1+ cells are considered by some to have stem cell-like properties akin to memory T cell populations, but the developmental relationships are unclear at present. Recent studies have also highlighted an important role for the transcriptional regulator TOX in driving the epigenetic enforcement of exhaustion, but key questions remain about the potential to reverse the epigenetic programme of exhaustion and how this might affect the persistence of T cell populations.


Subject(s)
T-Lymphocytes/immunology , Animals , Hepatocyte Nuclear Factor 1-alpha/physiology , High Mobility Group Proteins/physiology , Humans , Infections/immunology , Neoplasms/immunology , Programmed Cell Death 1 Receptor/physiology
7.
Sci Rep ; 9(1): 10183, 2019 07 15.
Article in English | MEDLINE | ID: mdl-31308435

ABSTRACT

Facilitates chromatin transcription (FACT) is a histone chaperone, which accomplishes both nucleosome assembly and disassembly. Our combined cryo-electron microscopy (EM) and native mass spectrometry (MS) studies revealed novel key steps of nucleosome reorganization conducted by a Mid domain and its adjacent acidic AID segment of human FACT. We determined three cryo-EM structures of respective octasomes complexed with the Mid-AID and AID regions, and a hexasome alone. We discovered extensive contacts between a FACT region and histones H2A, H2B, and H3, suggesting that FACT is competent to direct functional replacement of a nucleosomal DNA end by its phosphorylated AID segment (pAID). Mutational assays revealed that the aromatic and phosphorylated residues within pAID are essential for octasome binding. The EM structure of the hexasome, generated by the addition of Mid-pAID or pAID, indicated that the dissociation of H2A-H2B dimer causes significant alteration from the canonical path of the nucleosomal DNA.


Subject(s)
Chromatin Assembly and Disassembly/physiology , DNA-Binding Proteins/metabolism , High Mobility Group Proteins/metabolism , Nucleosomes/metabolism , Transcriptional Elongation Factors/metabolism , Chromatin/chemistry , Cryoelectron Microscopy/methods , DNA/chemistry , DNA-Binding Proteins/physiology , High Mobility Group Proteins/physiology , Histones/metabolism , Histones/physiology , Humans , Mass Spectrometry/methods , Models, Molecular , Molecular Chaperones/metabolism , Nucleosomes/physiology , Protein Binding/physiology , Transcriptional Elongation Factors/physiology
9.
Dev Cell ; 49(5): 748-763.e7, 2019 06 03.
Article in English | MEDLINE | ID: mdl-31105007

ABSTRACT

Oxygen (O2) is both an indispensable metabolic substrate and a regulatory signal that controls the activity of Hypoxia-Inducible Factor 1α (Hif1a), a mediator of the cellular adaptation to low O2 tension (hypoxia). Hypoxic cells require Hif1a to survive. Additionally, Hif1a is an inhibitor of mitochondrial respiration. Hence, we hypothesized that enhancing mitochondrial respiration is detrimental to the survival of hypoxic cells in vivo. We tested this hypothesis in the fetal growth plate, which is hypoxic. Our findings show that mitochondrial respiration is dispensable for survival of growth plate chondrocytes. Furthermore, its impairment prevents the extreme hypoxia and the massive chondrocyte death observed in growth plates lacking Hif1a. Consequently, augmenting mitochondrial respiration affects the survival of hypoxic chondrocytes by, at least in part, increasing intracellular hypoxia. We thus propose that partial suppression of mitochondrial respiration is crucial during development to protect the tissues that are physiologically hypoxic from lethal intracellular anoxia.


Subject(s)
Chondrocytes/physiology , Fetal Development/physiology , Growth Plate/physiology , Hypoxia/physiopathology , Mitochondria/physiology , Animals , Basic Helix-Loop-Helix Transcription Factors/physiology , Cell Respiration , Cell Survival , Chondrocytes/cytology , DNA-Binding Proteins/physiology , Female , High Mobility Group Proteins/physiology , Homeodomain Proteins/physiology , Hypoxia-Inducible Factor 1, alpha Subunit/physiology , Male , Mice , Mice, Inbred C57BL
10.
Cell Mol Life Sci ; 76(8): 1529-1539, 2019 Apr.
Article in English | MEDLINE | ID: mdl-30683982

ABSTRACT

HMG box protein 1 (HBP1) is a transcription factor and a potent cell cycle inhibitor in normal and cancer cells. HBP1 activates or represses the expression of different cell cycle genes (such as CDKN2A, CDKN1A, and CCND1) through direct DNA binding, cofactor recruitment, chromatin remodeling, or neutralization of other transcription factors. Among these are LEF1, TCF4, and MYC in the WNT/beta-catenin pathway. HBP1 also contributes to oncogenic RAS-induced senescence and terminal cell differentiation. Collectively, these activities suggest a tumor suppressor function. However, HBP1 is not listed among frequently mutated cancer driver genes. Nevertheless, HBP1 expression is lower in several tumor types relative to matched normal tissues. Several micro-RNAs, such as miR-155, miR-17-92, and miR-29a, dampen HBP1 expression in cancer cells of various origins. The phosphatidylinositol-3 kinase (PI3K)/AKT pathway also inhibits HBP1 transcription by preventing FOXO binding to the HBP1 promoter. In addition, AKT directly phosphorylates HBP1, thereby inhibiting its transcriptional activity. Taken together, these findings place HBP1 at the center of a network of micro-RNAs and oncoproteins that control cell proliferation. In this review, we discuss our current understanding of HBP1 function in human physiology and diseases.


Subject(s)
Cell Cycle Checkpoints , Gene Expression Regulation, Neoplastic , High Mobility Group Proteins/physiology , Neoplasms/genetics , Neoplasms/pathology , Repressor Proteins/physiology , Animals , Cell Line, Tumor , High Mobility Group Proteins/genetics , Humans , Mice, Transgenic , Primary Cell Culture , Protein Processing, Post-Translational , Repressor Proteins/genetics , Transcriptional Activation
11.
Mol Cell Biochem ; 454(1-2): 139-152, 2019 Apr.
Article in English | MEDLINE | ID: mdl-30353496

ABSTRACT

Heart failure (HF) is a functional lack of myocardial performance due to a loss of molecular control over increases in calcium and ROS, resulting in proteolytic degradative advances and cardiac remodeling. Mitochondria are the molecular powerhouse of cells, shifting the sphere of cardiomyocyte stability and performance. Functional mitochondria rely on the molecular abilities of safety factors such as TFAM to maintain physiological parameters. Mitochondrial transcription factor A (TFAM) creates a mitochondrial nucleoid structure around mtDNA, protecting it from mutation, inhibiting NFAT (ROS activator/hypertrophic stimulator), and transcriptionally activates Serca2a to decrease calcium mishandling. Calpain1 and MMP9 are proteolytic degratory factors that play a major role in cardiomyocyte decline in HF. Current literature depicts major decreases in TFAM as HF progresses. We aim to assess TFAM function against Calpain1 and MMP9 proteolytic activity and its role in cardiac remodeling. To this date, no publication has surfaced describing the effects of aortic banding (AB) as a surgical HF model in TFAM-TG mice. HF models were created via AB in TFAM transgenic (TFAM-TG) and C57BLJ-6 (WT) mice. Eight weeks post AB, functional analysis revealed a successful banding procedure, resulting in cardiac hypertrophy as observed via echocardiography. Pulse wave and color doppler show increased aortic flow rates as well as turbulent flow at the banding site. Preliminary results of cardiac tissue immuno-histochemistry of HF-control mice show decreased TFAM and compensatory increases in Serca2a fluorescent expression, along with increased Calpain1 and MMP9 expression. Protein, RNA, and IHC analysis will further assess TFAM-TG results post-banding. Echocardiography shows more cardiac stability and functionality in HF-induced TFAM-TG mice than the control counterpart. These findings complement our published in vitro results. Overall, this suggests that TFAM has molecular therapeutic potential to reduce protease expression.


Subject(s)
DNA-Binding Proteins/physiology , Gene Expression Regulation , Heart Failure/metabolism , High Mobility Group Proteins/physiology , Animals , Calpain/genetics , Cardiomegaly , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Disease Models, Animal , Heart Failure/genetics , Heart Failure/physiopathology , High Mobility Group Proteins/genetics , High Mobility Group Proteins/metabolism , Matrix Metalloproteinase 9/genetics , Mice , Mice, Transgenic , Myocardium , Sarcoplasmic Reticulum Calcium-Transporting ATPases/genetics , Up-Regulation
12.
Dev Biol ; 439(2): 92-101, 2018 07 15.
Article in English | MEDLINE | ID: mdl-29684311

ABSTRACT

During development, the embryo transitions from a metabolism favoring glycolysis to a metabolism favoring mitochondrial respiration. How metabolic shifts regulate developmental processes, or how developmental processes regulate metabolic shifts, remains unclear. To test the requirement of mitochondrial function in developing endoderm-derived tissues, we genetically inactivated the mitochondrial transcription factor, Tfam, using the Shh-Cre driver. Tfam mutants did not survive postnatally, exhibiting defects in lung development. In the developing intestine, TFAM-loss was tolerated until late fetal development, during which the process of villus elongation was compromised. While progenitor cell populations appeared unperturbed, markers of enterocyte maturation were diminished and villi were blunted. Loss of TFAM was also tested in the adult intestinal epithelium, where enterocyte maturation was similarly dependent upon the mitochondrial transcription factor. While progenitor cells in the transit amplifying zone of the adult intestine remained proliferative, intestinal stem cell renewal was dependent upon TFAM, as indicated by molecular profiling and intestinal organoid formation assays. Taken together, these studies point to critical roles for the mitochondrial regulator TFAM for multiple aspects of intestinal development and maturation, and highlight the importance of mitochondrial regulators in tissue development and homeostasis.


Subject(s)
DNA-Binding Proteins/metabolism , DNA-Binding Proteins/physiology , High Mobility Group Proteins/metabolism , High Mobility Group Proteins/physiology , Intestinal Mucosa/metabolism , Animals , Cell Culture Techniques/methods , Cell Self Renewal/physiology , DNA-Binding Proteins/genetics , Fetus/metabolism , Gene Expression Regulation/genetics , Glycolysis/genetics , Glycolysis/physiology , High Mobility Group Proteins/genetics , Intestinal Mucosa/embryology , Intestinal Mucosa/growth & development , Mice , Mitochondria/metabolism , Mitochondrial Proteins/genetics , Organogenesis/genetics , Organogenesis/physiology , Organoids/metabolism , Stem Cells/metabolism , Transcription Factors/metabolism
13.
J Virol ; 91(7)2017 04 01.
Article in English | MEDLINE | ID: mdl-28122976

ABSTRACT

All retroviruses need to integrate a DNA copy of their genome into the host chromatin. Cellular proteins regulating and targeting lentiviral and gammaretroviral integration in infected cells have been discovered, but the factors that mediate alpharetroviral avian leukosis virus (ALV) integration are unknown. In this study, we have identified the FACT protein complex, which consists of SSRP1 and Spt16, as a principal cellular binding partner of ALV integrase (IN). Biochemical experiments with purified recombinant proteins show that SSRP1 and Spt16 are able to individually bind ALV IN, but only the FACT complex effectively stimulates ALV integration activity in vitro Likewise, in infected cells, the FACT complex promotes ALV integration activity, with proviral integration frequency varying directly with cellular expression levels of the FACT complex. An increase in 2-long-terminal-repeat (2-LTR) circles in the depleted FACT complex cell line indicates that this complex regulates the ALV life cycle at the level of integration. This regulation is shown to be specific to ALV, as disruption of the FACT complex did not inhibit either lentiviral or gammaretroviral integration in infected cells.IMPORTANCE The majority of human gene therapy approaches utilize HIV-1- or murine leukemia virus (MLV)-based vectors, which preferentially integrate near genes and regulatory regions; thus, insertional mutagenesis is a substantial risk. In contrast, ALV integrates more randomly throughout the genome, which decreases the risks of deleterious integration. Understanding how ALV integration is regulated could facilitate the development of ALV-based vectors for use in human gene therapy. Here we show that the FACT complex directly binds and regulates ALV integration efficiency in vitro and in infected cells.


Subject(s)
Avian Leukosis Virus/genetics , Cell Cycle Proteins/physiology , DNA, Viral/physiology , DNA-Binding Proteins/physiology , High Mobility Group Proteins/physiology , Transcription Factors/physiology , Transcriptional Elongation Factors/physiology , Amino Acid Sequence , Animals , Avian Leukosis Virus/enzymology , Chick Embryo , Conserved Sequence , HEK293 Cells , Humans , Integrases/physiology , Protein Binding , Protein Interaction Domains and Motifs , Virus Integration
14.
PLoS One ; 12(1): e0170576, 2017.
Article in English | MEDLINE | ID: mdl-28107452

ABSTRACT

Fetal germ cell development is tightly regulated by the somatic cell environment, and is characterised by cell cycle states that differ between XY and XX gonads. In the testis, gonocytes enter G1/G0 arrest from 12.5 days post coitum (dpc) in mice and maintain cell cycle arrest until after birth. Failure to correctly maintain G1/G0 arrest can result in loss of germ cells or, conversely, germ cell tumours. High mobility group box containing transcription factor 1 (HBP1) is a transcription factor that was previously identified in fetal male germ cells at the time of embryonic cell cycle arrest. In somatic cells, HBP1 is classified as a tumour suppressor protein, known to regulate proliferation and senescence. We therefore investigated the possible role of HBP1 in the initiation and maintenance of fetal germ cell G1/G0 arrest using the mouse model. We identified two splice variants of Hbp1, both of which are expressed in XY and XX fetal gonads, but only one of which is localised to the nucleus in in vitro assays. To investigate Hbp1 loss of function, we used embryonic stem (ES) cells carrying a Genetrap mutation for Hbp1 to generate mice lacking Hbp1 function. We found that Hbp1-genetrap mouse mutant germ cells proliferated correctly throughout development, and adult males were viable and fertile. Multiple Hbp1-LacZ reporter mouse lines were generated, unexpectedly revealing Hbp1 embryonic expression in hair follicles, eye and limbs. Lastly, in a model of defective germ cell G1/G0 arrest, the Rb1-knockout model, we found no evidence for Hbp1 mis-regulation, suggesting that the reported RB1-HBP1 interaction is not critical in the germline, despite co-expression.


Subject(s)
Fertility/genetics , Germ Cells/physiology , High Mobility Group Proteins/physiology , Repressor Proteins/physiology , Animals , Cell Line , Germ Cells/metabolism , In Situ Hybridization , Male , Mice/embryology , Mice, Inbred C57BL , Mice, Knockout , Promoter Regions, Genetic/genetics , Real-Time Polymerase Chain Reaction
15.
Biochim Biophys Acta ; 1859(3): 462-7, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26455954

ABSTRACT

The dynamic interaction of nucleosome binding proteins with their chromatin targets is an important element in regulating the structure and function of chromatin. Histone H1 variants and High Mobility Group (HMG) proteins are ubiquitously expressed in all vertebrate cells, bind dynamically to chromatin, and are known to affect chromatin condensation and the ability of regulatory factors to access their genomic binding sites. Here, we review the studies that focus on the interactions between H1 and HMGs and highlight the functional consequences of the interplay between these architectural chromatin binding proteins. H1 and HMG proteins are mobile molecules that bind to nucleosomes as members of a dynamic protein network. All HMGs compete with H1 for chromatin binding sites, in a dose dependent fashion, but each HMG family has specific effects on the interaction of H1 with chromatin. The interplay between H1 and HMGs affects chromatin organization and plays a role in epigenetic regulation.


Subject(s)
Chromatin/chemistry , High Mobility Group Proteins/physiology , Histones/physiology , Animals , Carrier Proteins/physiology , Humans
16.
Mediators Inflamm ; 2015: 243868, 2015.
Article in English | MEDLINE | ID: mdl-26556952

ABSTRACT

TOX, an evolutionarily conserved member of the HMG-box family of proteins, is essential for the development of various cells of both the innate and adaptive immune system. TOX is required for the development of CD4(+) T lineage cells in the thymus, including natural killer T and T regulatory cells, as well as development of natural killer cells and fetal lymphoid tissue inducer cells, the latter required for lymph node organogenesis. Recently, we have identified a broader role for TOX in the innate immune system, demonstrating that this nuclear protein is required for generation of bone marrow progenitors that have potential to give rise to all innate lymphoid cells. Innate lymphoid cells, classified according to transcription factor expression and cytokine secretion profiles, derive from common lymphoid progenitors in the bone marrow and require Notch signals for their development. We discuss here the role of TOX in specifying CLP toward an innate lymphoid cell fate and hypothesize a possible role for TOX in regulating Notch gene targets during innate lymphoid cell development.


Subject(s)
High Mobility Group Proteins/physiology , Immunity, Innate , Lymphocytes/immunology , Basic-Leucine Zipper Transcription Factors/physiology , Cell Lineage , Humans , Killer Cells, Natural/immunology
17.
J Biol Chem ; 290(45): 27297-27310, 2015 Nov 06.
Article in English | MEDLINE | ID: mdl-26378236

ABSTRACT

Our functional genomic RNAi screens have identified the protein components of the FACT (facilitates chromatin transcription) complex, SUPT16H and SSRP1, as top host factors that negatively regulate HIV-1 replication. FACT interacts specifically with histones H2A/H2B to affect assembly and disassembly of nucleosomes, as well as transcription elongation. We further investigated the suppressive role of FACT proteins in HIV-1 transcription. First, depletion of SUPT16H or SSRP1 protein enhances Tat-mediated HIV-1 LTR (long terminal repeat) promoter activity. Second, HIV-1 Tat interacts with SUPT16H but not SSRP1 protein. However, both SUPT16H and SSRP1 are recruited to LTR promoter. Third, the presence of SUPT16H interferes with the association of Cyclin T1 (CCNT1), a subunit of P-TEFb, with the Tat-LTR axis. Removing inhibitory mechanisms to permit HIV-1 transcription is an initial and key regulatory step to reverse post-integrated latent HIV-1 proviruses for purging of reservoir cells. We therefore evaluated the role of FACT proteins in HIV-1 latency and reactivation. Depletion of SUPT16H or SSRP1 protein affects both HIV-1 transcriptional initiation and elongation and spontaneously reverses latent HIV-1 in U1/HIV and J-LAT cells. Similar effects were observed with a primary CD4+ T cell model of HIV-1 latency. FACT proteins also interfere with HTLV-1 Tax-LTR-mediated transcription and viral latency, indicating that they may act as general transcriptional suppressors for retroviruses. We conclude that FACT proteins SUPT16H and SSRP1 play a key role in suppressing HIV-1 transcription and promoting viral latency, which may serve as promising gene targets for developing novel HIV-1 latency-reversing agents.


Subject(s)
Cell Cycle Proteins/physiology , DNA-Binding Proteins/physiology , HIV-1/physiology , High Mobility Group Proteins/physiology , Human T-lymphotropic virus 1/physiology , Transcription Factors/physiology , Transcriptional Elongation Factors/physiology , Virus Latency/physiology , CD4-Positive T-Lymphocytes/physiology , CD4-Positive T-Lymphocytes/virology , Cell Cycle Proteins/antagonists & inhibitors , Cell Cycle Proteins/genetics , Cell Line , Cyclin T/physiology , DNA-Binding Proteins/antagonists & inhibitors , DNA-Binding Proteins/genetics , HEK293 Cells , HIV Long Terminal Repeat , HIV-1/genetics , High Mobility Group Proteins/antagonists & inhibitors , High Mobility Group Proteins/genetics , Host-Pathogen Interactions/genetics , Host-Pathogen Interactions/physiology , Human T-lymphotropic virus 1/genetics , Humans , Models, Biological , Positive Transcriptional Elongation Factor B/physiology , Promoter Regions, Genetic , RNA Interference , Transcription Factors/antagonists & inhibitors , Transcription Factors/genetics , Transcriptional Elongation Factors/antagonists & inhibitors , Transcriptional Elongation Factors/genetics , Virus Latency/genetics
18.
DNA Repair (Amst) ; 36: 122-136, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26411874

ABSTRACT

It has been almost a decade since the last review appeared comparing and contrasting the influences that the different families of High Mobility Group proteins (HMGA, HMGB and HMGN) have on the various DNA repair pathways in mammalian cells. During that time considerable progress has been made in our understanding of how these non-histone proteins modulate the efficiency of DNA repair by all of the major cellular pathways: nucleotide excision repair, base excision repair, double-stand break repair and mismatch repair. Although there are often similar and over-lapping biological activities shared by all HMG proteins, members of each of the different families appear to have a somewhat 'individualistic' impact on various DNA repair pathways. This review will focus on what is currently known about the roles that different HMG proteins play in DNA repair processes and discuss possible future research areas in this rapidly evolving field.


Subject(s)
Chromatin Assembly and Disassembly , Chromatin/chemistry , DNA Repair , DNA/metabolism , High Mobility Group Proteins/physiology , Animals , Humans , Mammals/genetics , Mammals/metabolism
19.
Nucleic Acids Res ; 43(12): 5759-70, 2015 Jul 13.
Article in English | MEDLINE | ID: mdl-25979266

ABSTRACT

DNA is packaged into condensed chromatin fibers by association with histones and architectural proteins such as high mobility group (HMGB) proteins. However, this DNA packaging reduces accessibility of enzymes that act on DNA, such as proteins that process DNA after double strand breaks (DSBs). Chromatin remodeling overcomes this barrier. We show here that the Saccharomyces cerevisiae HMGB protein HMO1 stabilizes chromatin as evidenced by faster chromatin remodeling in its absence. HMO1 was evicted along with core histones during repair of DSBs, and chromatin remodeling events such as histone H2A phosphorylation and H3 eviction were faster in absence of HMO1. The facilitated chromatin remodeling in turn correlated with more efficient DNA resection and recruitment of repair proteins; for example, inward translocation of the DNA-end-binding protein Ku was faster in absence of HMO1. This chromatin stabilization requires the lysine-rich C-terminal extension of HMO1 as truncation of the HMO1 C-terminal tail phenocopies hmo1 deletion. Since this is reminiscent of the need for the basic C-terminal domain of mammalian histone H1 in chromatin compaction, we speculate that HMO1 promotes chromatin stability by DNA bending and compaction imposed by its lysine-rich domain and that it must be evicted along with core histones for efficient DSB repair.


Subject(s)
Chromatin Assembly and Disassembly , Chromatin/metabolism , DNA Breaks, Double-Stranded , DNA Repair , High Mobility Group Proteins/physiology , Saccharomyces cerevisiae Proteins/physiology , Antigens, Nuclear/metabolism , DNA-Binding Proteins/metabolism , Gene Deletion , Genetic Loci , High Mobility Group Proteins/analysis , High Mobility Group Proteins/genetics , Histones/metabolism , Ku Autoantigen , Phosphorylation , Rad51 Recombinase/metabolism , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism , Saccharomyces cerevisiae Proteins/analysis , Saccharomyces cerevisiae Proteins/genetics , Saccharomyces cerevisiae Proteins/metabolism , Sequence Deletion
20.
Oncogene ; 34(41): 5264-76, 2015 Oct 08.
Article in English | MEDLINE | ID: mdl-25639869

ABSTRACT

HMG20A is a high mobility group (HMG) domain containing protein homologous to HMG20B, a core subunit of the Lys-specific demethylase 1/REST co-repressor 1 (LSD1-CoREST) histone demethylase complex. Here, we show that HMG20A can replace HMG20B and, therefore, they are mutually exclusive subunits of the complex. Both proteins interact through a coiled-coil domain with BHC80, another subunit of the LSD1-CoREST complex. To investigate the functional differences between the two proteins, we performed transcriptomic analysis of HMG20A- and HMG20B-depleted cells. Analysis of the misregulated genes in HMG20A-knockdown cells evidenced a high proportion of genes related to the epithelial-to-mesenchymal transition (EMT) process. EMT occurs during embryonic development or during the course of malignant cancer progression and consists in the dynamic and reversible transitions between epithelial and mesenchymal phenotypes. We show that HMG20A together with LSD1 are required for SNAI1-dependent repression of epithelial genes and for (transforming growth factor ß) TGF-ß-triggered EMT. Importantly, HMG20A-depleted cells displayed reduced binding of LSD1 to epithelial gene promoters and increased methylation of lysine 4 of histone H3, suggesting a role of HMG20A in recruiting or in stabilizing the complex at the chromatin. SNAI1 and the TGF-ß-related transcription factor SMAD4 were found to be associated with the LSD1-CoREST complex containing HMG20A. Furthermore, we show that HMG20A-depleted cells displayed reduced motility and invasion activity. Finally, we show that expression of HMG20A correlates positively with mesenchymal markers and negatively with epithelial markers in human tumor samples. Taken together, our data demonstrate that HMG20A is essential for the mesenchymal phenotype.


Subject(s)
Epithelial-Mesenchymal Transition , High Mobility Group Proteins/physiology , Transcription Factors/metabolism , Cell Line, Tumor , Cell Movement , Gene Expression Regulation, Neoplastic , Gene Ontology , HEK293 Cells , Histone Demethylases/metabolism , Humans , Neoplasm Invasiveness , Neoplasms/genetics , Neoplasms/metabolism , Smad4 Protein/metabolism , Snail Family Transcription Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...