Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 23
Filter
1.
Genes Dev ; 34(3-4): 166-178, 2020 02 01.
Article in English | MEDLINE | ID: mdl-31919188

ABSTRACT

Oocytes are indispensable for mammalian life. Thus, it is important to understand how mature oocytes are generated. As a critical stage of oocytes development, meiosis has been extensively studied, yet how chromatin remodeling contributes to this process is largely unknown. Here, we demonstrate that the ATP-dependent chromatin remodeling factor Snf2h (also known as Smarca5) plays a critical role in regulating meiotic cell cycle progression. Females with oocyte-specific depletion of Snf2h are infertile and oocytes lacking Snf2h fail to undergo meiotic resumption. Mechanistically, depletion of Snf2h results in dysregulation of meiosis-related genes, which causes failure of maturation-promoting factor (MPF) activation. ATAC-seq analysis in oocytes revealed that Snf2h regulates transcription of key meiotic genes, such as Prkar2b, by increasing its promoter chromatin accessibility. Thus, our studies not only demonstrate the importance of Snf2h in oocyte meiotic resumption, but also reveal the mechanism underlying how a chromatin remodeling factor can regulate oocyte meiosis.


Subject(s)
Adenosine Triphosphatases/genetics , Adenosine Triphosphatases/metabolism , Chromatin Assembly and Disassembly/genetics , Chromosomal Proteins, Non-Histone/genetics , Chromosomal Proteins, Non-Histone/metabolism , Maturation-Promoting Factor/genetics , Meiosis/genetics , Oogenesis/genetics , Animals , Female , Gene Expression Regulation, Developmental , Gene Knockout Techniques , Mesothelin , Mice , Oocytes/cytology , Transcriptome
2.
Cancer Cell Int ; 21(1): 315, 2021 Jun 19.
Article in English | MEDLINE | ID: mdl-34147108

ABSTRACT

RSF1, remodelling and spacing factor 1, is an important interphase centromere protein and is overexpressed in many types of cancers and correlated with poor overall survival. RSF1 has functions mainly in maintaining chromosome stability, facilitating DNA repair, maintaining the protein homeostasis of RSF1 and suppressing the transcription of some oncogenes when RSF1 protein is expressed at an optimal level; however, RSF1 overexpression facilitates drug resistance and cell cycle checkpoint inhibition to prompt cancer proliferation and survival. The RSF1 expression level and gene background are crucial for RSF1 functions, which may explain why RSF1 has different functions in different cancer types. This review summarizes the functional domains of RSF1, the overexpression status of RSF1 and SNF2H in cancer based on the TCGA and GTEX databases, the cancer-related functions of RSF1 in interacting with H2Aub, HDAC1, CENP-A, PLK1, ATM, CENP-S, SNF2H, HBX, BubR1, cyclin E1, CBP and NF-κB and the potential clinical value of RSF1, which will lay a theoretical foundation for the structural biology study of RSF1 and application of RSF1 inhibitors, truncated RSF1 proteins and SNF2H inhibitors in the treatment of RSF1-overexpressing tumours.

3.
Int J Mol Sci ; 21(6)2020 Mar 18.
Article in English | MEDLINE | ID: mdl-32197313

ABSTRACT

ISWI chromatin remodeling ATPase SMARCA5 (SNF2H) is a well-known factor for its role in regulation of DNA access via nucleosome sliding and assembly. SMARCA5 transcriptionally inhibits the myeloid master regulator PU.1. Upregulation of SMARCA5 was previously observed in CD34+ hematopoietic progenitors of acute myeloid leukemia (AML) patients. Since high levels of SMARCA5 are necessary for intensive cell proliferation and cell cycle progression of developing hematopoietic stem and progenitor cells in mice, we reasoned that removal of SMARCA5 enzymatic activity could affect the cycling or undifferentiated state of leukemic progenitor-like clones. Indeed, we observed that CRISPR/cas9-mediated SMARCA5 knockout in AML cell lines (S5KO) inhibited the cell cycle progression. We also observed that the SMARCA5 deletion induced karyorrhexis and nuclear budding as well as increased the ploidy, indicating its role in mitotic division of AML cells. The cytogenetic analysis of S5KO cells revealed the premature chromatid separation. We conclude that deleting SMARCA5 in AML blocks leukemic proliferation and chromatid cohesion.


Subject(s)
Adenosine Triphosphatases/deficiency , Cell Proliferation , Chromatids , Chromosomal Proteins, Non-Histone/deficiency , Gene Knockout Techniques , Leukemia, Myeloid, Acute , Neoplasm Proteins , Adenosine Triphosphatases/metabolism , Cell Line, Tumor , Chromatids/genetics , Chromatids/metabolism , Chromosomal Proteins, Non-Histone/metabolism , Female , Humans , K562 Cells , Leukemia, Myeloid, Acute/enzymology , Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/pathology , Male , Neoplasm Proteins/deficiency , Neoplasm Proteins/metabolism
4.
J Biol Chem ; 293(34): 13176-13190, 2018 08 24.
Article in English | MEDLINE | ID: mdl-29959226

ABSTRACT

Genes are transcribed in irregular pulses of activity termed transcriptional bursts. Cellular differentiation requires coordinated gene expression; however, it is unknown whether the burst fraction (i.e. the number of active phases of transcription) or size/intensity (the number of RNA molecules produced within a burst) changes during cell differentiation. In the ocular lens, the positions of lens fiber cells correlate precisely with their differentiation status, and the most advanced cells degrade their nuclei. Here, we examined the transcriptional parameters of the ß-actin and lens differentiation-specific α-, ß-, and γ-crystallin genes by RNA fluorescent in situ hybridization (FISH) in the lenses of embryonic day (E) E12.5, E14.5, and E16.5 mouse embryos and newborns. We found that cellular differentiation dramatically alters the burst fraction in synchronized waves across the lens fiber cell compartment with less dramatic changes in burst intensity. Surprisingly, we observed nascent transcription of multiple genes in nuclei just before nuclear destruction. Nuclear condensation was accompanied by transfer of nuclear proteins, including histone and nonhistone proteins, to the cytoplasm. Although lens-specific deletion of the chromatin remodeler SWI/SNF-related matrix-associated actin-dependent regulator of chromatin subfamily A member 5 (Smarca5/Snf2h) interfered with denucleation, persisting nuclei remained transcriptionally competent and exhibited changes in both burst intensity and fraction depending on the gene examined. Our results uncover the mechanisms of nascent transcriptional control during differentiation and chromatin remodeling, confirm the burst fraction as the major factor adjusting gene expression levels, and reveal transcriptional competence of fiber cell nuclei even as they approach disintegration.


Subject(s)
Cell Differentiation , Cell Nucleus/physiology , Embryo, Mammalian/cytology , Gene Expression Regulation, Developmental , Lens, Crystalline/cytology , Organogenesis , Transcription, Genetic , Animals , Animals, Newborn , Chromatin Assembly and Disassembly , Embryo, Mammalian/physiology , Female , Lens, Crystalline/physiology , Mice
5.
Development ; 143(11): 1937-47, 2016 06 01.
Article in English | MEDLINE | ID: mdl-27246713

ABSTRACT

Ocular lens morphogenesis is a model for investigating mechanisms of cellular differentiation, spatial and temporal gene expression control, and chromatin regulation. Brg1 (Smarca4) and Snf2h (Smarca5) are catalytic subunits of distinct ATP-dependent chromatin remodeling complexes implicated in transcriptional regulation. Previous studies have shown that Brg1 regulates both lens fiber cell differentiation and organized degradation of their nuclei (denucleation). Here, we employed a conditional Snf2h(flox) mouse model to probe the cellular and molecular mechanisms of lens formation. Depletion of Snf2h induces premature and expanded differentiation of lens precursor cells forming the lens vesicle, implicating Snf2h as a key regulator of lens vesicle polarity through spatial control of Prox1, Jag1, p27(Kip1) (Cdkn1b) and p57(Kip2) (Cdkn1c) gene expression. The abnormal Snf2h(-/-) fiber cells also retain their nuclei. RNA profiling of Snf2h(-/) (-) and Brg1(-/-) eyes revealed differences in multiple transcripts, including prominent downregulation of those encoding Hsf4 and DNase IIß, which are implicated in the denucleation process. In summary, our data suggest that Snf2h is essential for the establishment of lens vesicle polarity, partitioning of prospective lens epithelial and fiber cell compartments, lens fiber cell differentiation, and lens fiber cell nuclear degradation.


Subject(s)
Adenosine Triphosphatases/metabolism , Cell Differentiation , Cell Nucleus/metabolism , Chromatin Assembly and Disassembly , Chromosomal Proteins, Non-Histone/metabolism , Embryo, Mammalian/metabolism , Lens, Crystalline/cytology , Lens, Crystalline/embryology , Animals , Autophagy , Cell Compartmentation , Cell Cycle , DNA Helicases/metabolism , DNA-Binding Proteins/metabolism , Epithelial Cells/cytology , Epithelial Cells/metabolism , Female , Gene Expression Regulation, Developmental , Heat Shock Transcription Factors , Mice, Knockout , Mitophagy , Models, Biological , Mutation/genetics , Nuclear Proteins/metabolism , PAX6 Transcription Factor/metabolism , Transcription Factors/metabolism , Transcriptome/genetics
6.
J Neuroinflammation ; 16(1): 135, 2019 Jul 04.
Article in English | MEDLINE | ID: mdl-31272467

ABSTRACT

BACKGROUND: Conditional ablation of the Smarca5 gene in mice severely impairs the postnatal growth of the cerebellum and causes an ataxic phenotype. Comparative gene expression studies indicated that complement-related proteins were upregulated in the cerebellum of Smarca5 mutant mice. Complement proteins play critical roles within innate immune signaling pathways and, in the brain, are produced by glial cells under both normal and pathological conditions. The C3 complement protein-derived signaling peptide, C3a, has been implicated in contributing to both tissue damage and repair in conditions such as multiple sclerosis and stroke. Here, we investigated whether C3a receptor (C3aR) signaling promoted damage or repair in the developing cerebellum of Smarca5 mutant mice. METHODS: Brain and cerebellum lysates from single Smarca5 conditional knockout (Smarca5 cKO) mice, C3aR1 KO mice, or double mutant mice were used for qRT-PCR and immunoblotting to assess the contribution of C3aR to the Smarca5 cKO brain pathology. Immunohistochemistry was used to characterize alterations to astroglia and phagocyte cells in the developing cerebellum of each of the genotypes. RESULTS: C3aR signaling was observed to limit gliosis and promote granule neuron survival during postnatal cerebellar development. In Smarca5 cKO mice, disorganized astroglia with increased GFAP expression develops concurrently with cerebellar granule neuron loss and phagocyte invasion over the first 10 days following birth. Potential ligand precursors of C3aR-VGF and C3-were found to have upregulated expression and/or altered processing during this time. Phagocytes (microglia and macrophages) in both the control and Smarca5 mutant mice were the only cells observed to express C3aR. Loss of C3aR in the Smarca5 cKO cerebellum resulted in increased numbers of apoptotic cells and early phagocyte invasion into the external granule cell layer, as well as an exacerbated disorganization of the Bergmann glia. The loss of C3aR expression also attenuated an increase in the expression of the efferocytosis-related protein, MerTK, whose transcript was upregulated ~ 2.5-fold in the Smarca5 mutant cerebellum at P10. CONCLUSIONS: This data indicates that C3aR can play an important role in limiting astrogliosis and regulating phagocyte phenotypes following developmental cell loss in the brain.


Subject(s)
Cerebellum/metabolism , Gliosis/metabolism , Neurodevelopmental Disorders/metabolism , Receptors, G-Protein-Coupled/deficiency , Signal Transduction/physiology , Adenosine Triphosphatases/deficiency , Adenosine Triphosphatases/genetics , Amino Acid Sequence , Animals , Cerebellum/pathology , Chromosomal Proteins, Non-Histone/deficiency , Chromosomal Proteins, Non-Histone/genetics , Gliosis/genetics , Gliosis/pathology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Neurodevelopmental Disorders/genetics , Neurodevelopmental Disorders/pathology , Receptors, G-Protein-Coupled/genetics
7.
Mol Cell Neurosci ; 87: 55-64, 2018 03.
Article in English | MEDLINE | ID: mdl-29249292

ABSTRACT

The mammalian ISWI (Imitation Switch) genes SMARCA1 and SMARCA5 encode the ATP-dependent chromatin remodeling proteins SNF2L and SNF2H. The ISWI proteins interact with BAZ (bromodomain adjacent to PHD zinc finger) domain containing proteins to generate eight distinct remodeling complexes. ISWI complex-mediated nucleosome positioning within genes and gene regulatory elements is proving important for the transition from a committed progenitor state to a differentiated cell state. Genetic studies have implicated the involvement of many ATP-dependent chromatin remodeling proteins in neurodevelopmental disorders (NDDs), including SMARCA1. Here we review the characterization of mice inactivated for ISWI and their interacting proteins, as it pertains to brain development and disease. A better understanding of chromatin dynamics during neural development is a prerequisite to understanding disease pathologies and the development of therapeutics for these complex disorders.


Subject(s)
Adenosine Triphosphatases/metabolism , Brain/growth & development , Chromatin/genetics , Neurodevelopmental Disorders/genetics , Transcription Factors/metabolism , Adenosine Triphosphatases/genetics , Animals , Cell Nucleus/metabolism , Chromatin Assembly and Disassembly/genetics , Humans , Neurodevelopmental Disorders/metabolism , Transcription Factors/genetics
8.
Adv Exp Med Biol ; 1042: 61-78, 2017.
Article in English | MEDLINE | ID: mdl-29357053

ABSTRACT

DNA replication is a fundamental process required for the accurate and timely duplication of chromosomes. During late mitosis to G1 phase, the MCM2-7 complex is loaded onto chromatin in a manner dependent on ORC, CDC6, and Cdt1, and chromatin becomes licensed for replication. Although every eukaryotic organism shares common features in replication control, there are also some differences among species. For example, in higher eukaryotic cells including human cells, no strict sequence specificity has been observed for replication origins, unlike budding yeast or bacterial replication origins. Therefore, elements other than beyond DNA sequences are important for regulating replication. For example, the stability and precise positioning of nucleosomes affects replication control. However, little is known about how nucleosome structure is regulated when replication licensing occurs. During the last decade, histone acetylation enzyme HBO1, chromatin remodeler SNF2H, and histone chaperone GRWD1 have been identified as chromatin-handling factors involved in the promotion of replication licensing. In this review, we discuss how the rearrangement of nucleosome formation by these factors affects replication licensing.


Subject(s)
Chromatin Assembly and Disassembly/physiology , Chromatin/metabolism , DNA Replication/physiology , Eukaryotic Cells/metabolism , Minichromosome Maintenance Proteins/metabolism , Replication Origin , Animals , Cell Cycle Proteins/metabolism , Chromatin/genetics , Chromatin Assembly and Disassembly/genetics , Eukaryotic Cells/physiology , G1 Phase/genetics , Histones/metabolism , Humans , Mammals/genetics
9.
J Biol Chem ; 288(44): 31458-67, 2013 Nov 01.
Article in English | MEDLINE | ID: mdl-24047897

ABSTRACT

Both DNA and chromatin need to be duplicated during each cell division cycle. Replication happens in the context of defects in the DNA template and other forms of replication stress that present challenges to both genetic and epigenetic inheritance. The replication machinery is highly regulated by replication stress responses to accomplish this goal. To identify important replication and stress response proteins, we combined isolation of proteins on nascent DNA (iPOND) with quantitative mass spectrometry. We identified 290 proteins enriched on newly replicated DNA at active, stalled, and collapsed replication forks. Approximately 16% of these proteins are known replication or DNA damage response proteins. Genetic analysis indicates that several of the newly identified proteins are needed to facilitate DNA replication, especially under stressed conditions. Our data provide a useful resource for investigators studying DNA replication and the replication stress response and validate the use of iPOND combined with mass spectrometry as a discovery tool.


Subject(s)
DNA Damage , DNA Replication , DNA-Binding Proteins/chemistry , DNA/metabolism , Mass Spectrometry/methods , DNA/biosynthesis , DNA-Binding Proteins/metabolism , Humans
10.
Cells ; 12(7)2023 03 28.
Article in English | MEDLINE | ID: mdl-37048108

ABSTRACT

Chromatin remodeling complexes are required for many distinct nuclear processes such as transcription, DNA replication, and DNA repair. However, the contribution of these complexes to the development of complex tissues within an organism is poorly characterized. Imitation switch (ISWI) proteins are among the most evolutionarily conserved ATP-dependent chromatin remodeling factors and are represented by yeast Isw1/Isw2, and their vertebrate counterparts Snf2h (Smarca5) and Snf2l (Smarca1). In this study, we focused on the role of the Snf2h gene during the development of the mammalian retina. We show that Snf2h is expressed in both retinal progenitors and post-mitotic retinal cells. Using Snf2h conditional knockout mice (Snf2h cKO), we found that when Snf2h is deleted, the laminar structure of the adult retina is not retained, the overall thickness of the retina is significantly reduced compared with controls, and the outer nuclear layer (ONL) is completely missing. The depletion of Snf2h did not influence the ability of retinal progenitors to generate all the differentiated retinal cell types. Instead, the Snf2h function is critical for the proliferation of retinal progenitor cells. Cells lacking Snf2h have a defective S-phase, leading to the entire cell division process impairments. Although all retinal cell types appear to be specified in the absence of the Snf2h function, cell-cycle defects and concomitantly increased apoptosis in Snf2h cKO result in abnormal retina lamination, complete destruction of the photoreceptor layer, and consequently, a physiologically non-functional retina.


Subject(s)
Adenosine Triphosphatases , Chromatin Assembly and Disassembly , Chromatin , Chromosomal Proteins, Non-Histone , Animals , Mice , Cell Nucleus/metabolism , Cell Proliferation , Chromatin/metabolism , Mice, Knockout , Retina , Chromosomal Proteins, Non-Histone/metabolism , Adenosine Triphosphatases/metabolism
11.
bioRxiv ; 2023 Jun 20.
Article in English | MEDLINE | ID: mdl-37546986

ABSTRACT

Single-particle cryo-EM is widely used to determine enzyme-nucleosome complex structures. However, cryo-EM sample preparation remains challenging and inconsistent due to complex denaturation at the air-water interface (AWI). To address this issue, we developed graphene-oxide-coated EM grids functionalized with either single-stranded DNA (ssDNA) or thiol-poly(acrylic acid-co-styrene) (TAASTY) co-polymer. These grids protect complexes between the chromatin remodeler SNF2h and nucleosomes from the AWI and facilitated collection of high-quality micrographs of intact SNF2h-nucleosome complexes in the absence of crosslinking. The data yields maps ranging from 2.3 to 3 Å in resolution. 3D variability analysis reveals nucleotide-state linked conformational changes in SNF2h bound to a nucleosome. In addition, the analysis provides structural evidence for asymmetric coordination between two SNF2h protomers acting on the same nucleosome. We envision these grids will enable similar detailed structural analyses for other enzyme-nucleosome complexes and possibly other protein-nucleic acid complexes in general.

12.
Cells ; 11(5)2022 02 25.
Article in English | MEDLINE | ID: mdl-35269430

ABSTRACT

Smarca5, an ATPase of the ISWI class of chromatin remodelers, is a key regulator of chromatin structure, cell cycle and DNA repair. Smarca5 is deregulated in leukemia and breast, lung and gastric cancers. However, its role in oncogenesis is not well understood. Chromatin remodelers often play dosage-dependent roles in cancer. We therefore investigated the epigenomic and phenotypic impact of controlled stepwise attenuation of Smarca5 function in the context of primary cell transformation, a process relevant to tumor formation. Upon conditional single- or double-allele Smarca5 deletion, the cells underwent both accelerated growth arrest and senescence entry and displayed gradually increased sensitivity to genotoxic insults. These phenotypic characteristics were explained by specific remodeling of the chromatin structure and the transcriptome in primary cells prior to the immortalization onset. These molecular programs implicated Smarca5 requirement in DNA damage repair, telomere maintenance, cell cycle progression and in restricting apoptosis and cellular senescence. Consistent with the molecular programs, we demonstrate for the first time that Smarca5-deficient primary cells exhibit dramatically decreased capacity to bypass senescence and immortalize, an indispensable step during cell transformation and cancer development. Thus, Smarca5 plays a crucial role in key homeostatic processes and sustains cancer-promoting molecular programs and cellular phenotypes.


Subject(s)
Chromatin , Neoplasms , Adenosine Triphosphatases/metabolism , Chromatin Assembly and Disassembly , DNA Damage , DNA Repair
13.
Front Mol Neurosci ; 12: 243, 2019.
Article in English | MEDLINE | ID: mdl-31680852

ABSTRACT

Alterations in the homeostasis of either cortical progenitor pool, namely the apically located radial glial (RG) cells or the basal intermediate progenitors (IPCs) can severely impair cortical neuron production. Such changes are reflected by microcephaly and are often associated with cognitive defects. Genes encoding epigenetic regulators are a frequent cause of intellectual disability and many have been shown to regulate progenitor cell growth, including our inactivation of the Smarca1 gene encoding Snf2l, which is one of two ISWI mammalian orthologs. Loss of the Snf2l protein resulted in dysregulation of Foxg1 and IPC proliferation leading to macrocephaly. Here we show that inactivation of the closely related Smarca5 gene encoding the Snf2h chromatin remodeler is necessary for embryonic IPC expansion and subsequent specification of callosal projection neurons. Telencephalon-specific Smarca5 cKO embryos have impaired cell cycle kinetics and increased cell death, resulting in fewer Tbr2+ and FoxG1+ IPCs by mid-neurogenesis. These deficits give rise to adult mice with a dramatic reduction in Satb2+ upper layer neurons, and partial agenesis of the corpus callosum. Mice survive into adulthood but molecularly display reduced expression of the clustered protocadherin genes that may further contribute to altered dendritic arborization and a hyperactive behavioral phenotype. Our studies provide novel insight into the developmental function of Snf2h-dependent chromatin remodeling processes during brain development.

14.
Elife ; 82019 05 16.
Article in English | MEDLINE | ID: mdl-31094676

ABSTRACT

The acidic patch is a functionally important epitope on each face of the nucleosome that affects chromatin remodeling. Although related by 2-fold symmetry of the nucleosome, each acidic patch is uniquely positioned relative to a bound remodeler. An open question is whether remodelers are distinctly responsive to each acidic patch. Previously we reported a method for homogeneously producing asymmetric nucleosomes with distinct H2A/H2B dimers (Levendosky et al., 2016). Here, we use this methodology to show that the Chd1 remodeler from Saccharomyces cerevisiae and ISWI remodelers from human and Drosophila have distinct spatial requirements for the acidic patch. Unlike Chd1, which is equally affected by entry- and exit-side mutations, ISWI remodelers strongly depend on the entry-side acidic patch. Remarkably, asymmetry in the two acidic patches stimulates ISWI to slide mononucleosomes off DNA ends, overriding the remodeler's preference to shift the histone core toward longer flanking DNA.


Subject(s)
Adenosine Triphosphatases/metabolism , Chromatin Assembly and Disassembly , Chromosomal Proteins, Non-Histone/metabolism , DNA-Binding Proteins/metabolism , Nucleosomes/metabolism , Saccharomyces cerevisiae Proteins/metabolism , Transcription Factors/metabolism , Animals , Drosophila , Humans , Nucleosomes/chemistry , Saccharomyces cerevisiae
15.
Mol Cells ; 41(2): 127-133, 2018 Feb 28.
Article in English | MEDLINE | ID: mdl-29385673

ABSTRACT

Chromatin remodeling factors are involved in many cellular processes such as transcription, replication, and DNA damage response by regulating chromatin structure. As one of chromatin remodeling factors, remodeling and spacing factor 1 (RSF1) is recruited at double strand break (DSB) sites and regulates ataxia telangiectasia mutated (ATM) -dependent checkpoint pathway upon DNA damage for the efficient repair. RSF1 is overexpressed in a variety of cancers, but regulation of RSF1 levels remains largely unknown. Here, we showed that protein levels of RSF1 chromatin remodeler are temporally upregulated in response to different DNA damage agents without changing the RSF1 mRNA level. In the absence of SNF2h, a binding partner of RSF1, the RSF1 protein level was significantly diminished. Intriguingly, the level of RSF1-3SA mutant lacking ATM-mediated phosphorylation sites significantly increased, and upregulation of RSF1 levels under DNA damage was not observed in cells overexpressing ATM kinase. Furthermore, failure in the regulation of RSF1 level caused a significant reduction in DNA repair, whereas reconstitution of RSF1, but not of RSF1-3SA mutants, restored DSB repair. Our findings reveal that temporal regulation of RSF1 levels at its post-translational modification by SNF2h and ATM is essential for efficient DNA repair.


Subject(s)
DNA Damage , Nuclear Proteins/metabolism , Protein Processing, Post-Translational , Trans-Activators/metabolism , Adenosine Triphosphatases/genetics , Adenosine Triphosphatases/metabolism , Ataxia Telangiectasia Mutated Proteins/metabolism , Blotting, Western , Cell Line, Tumor , Chromatin Assembly and Disassembly , Chromosomal Proteins, Non-Histone/genetics , Chromosomal Proteins, Non-Histone/metabolism , DNA Breaks, Double-Stranded , DNA Repair , HEK293 Cells , Humans , MCF-7 Cells , Nuclear Proteins/genetics , Phosphorylation , Protein Binding , Trans-Activators/genetics
16.
Cell Rep ; 23(6): 1853-1866, 2018 05 08.
Article in English | MEDLINE | ID: mdl-29742439

ABSTRACT

Embryonic stem cells (ESCs) display an abbreviated cell cycle, resulting in a short doubling time and rapid proliferation. The histone variant H2A.X is critical for proliferation of stem cells, although mechanistic insights have remained obscure. Here, we show that H2A.X defines the rate of mouse ESC proliferation independently of the DNA damage response pathway, and it associates with three major chromatin-modifying complexes. Our functional and biochemical analyses demonstrate that H2A.X-associated factors mediate the H2A.X-dependent effect on ESC proliferation and involve the nucleolar remodeling complex (NoRC). A specific H2A.X deposition at rDNA promoters determines the chromatin recruitment of the NoRC, histone modifications, the rRNA transcription, and the rate of proliferation. Collectively, our results suggest that NoRC assembly by H2A.X deposition at rRNA promoters silences transcription, and this represents an important regulatory component for ESC proliferation.


Subject(s)
Cell Nucleolus/metabolism , Histones/metabolism , Mouse Embryonic Stem Cells/cytology , Mouse Embryonic Stem Cells/metabolism , Multiprotein Complexes/metabolism , Promoter Regions, Genetic/genetics , Animals , Cell Proliferation , Chromatin Assembly and Disassembly , Gene Silencing , Heterochromatin/metabolism , Mice , Phosphorylation , Protein Binding , Protein Interaction Mapping
17.
Oncol Lett ; 12(2): 1329-1336, 2016 Aug.
Article in English | MEDLINE | ID: mdl-27446433

ABSTRACT

Hepatocellular carcinoma (HCC) is one of the most common cancers worldwide and has an extremely poor prognosis. Surgical resection is always inapplicable to HCC patients diagnosed at an advanced tumor stage. The mechanisms underlying HCC cell proliferation remain obscure. In the present study, SWItch/sucrose nonfermentable catalytic subunit SNF2 (SNF2H) expression was tested in HCC tissues and Wnt/ß-catenin pathway activation upon overexpression of SNF2H or knockdown of SNF2H expression was investigated in cultured HCC cells. It was demonstrated that SNF2H is a vital factor for HCC growth. The SNF2H expression level is increased in HCC tissues compared with paratumoral liver tissues. SNF2H promotes HCC cell proliferation and colony formation ability in vitro. SNF2H may increase the protein level of ß-catenin and enhance its nuclear accumulation in HCC cells, thereby leading to the activation of the Wnt/ß-catenin signaling pathway. In conclusion, the present results indicate that SNF2H plays a vital role in HCC cell growth, suggesting that SNF2H may be a promising therapeutic target for HCC treatment.

18.
DNA Repair (Amst) ; 43: 69-77, 2016 07.
Article in English | MEDLINE | ID: mdl-27268481

ABSTRACT

The protein XRCC1 has no inherent enzymatic activity, and is believed to function in base excision repair as a dedicated scaffold component that coordinates other DNA repair factors. Repair foci clearly represent the recruitment and accumulation of DNA repair factors at sites of damage; however, uncertainties remain regarding their organization in the context of nuclear architecture and their biological significance. Here we identified the chromatin remodeling factor SNF2H/SMARCA5 as a novel binding partner of XRCC1, with their interaction dependent on the casein kinase 2-mediated constitutive phosphorylation of XRCC1. The proficiency of repairing H2O2-induced damage was strongly impaired by SNF2H knock-down, and similar impairment was observed with knock-down of both XRCC1 and SNF2H simultaneously, suggesting their role in a common repair pathway. Most SNF2H exists in the nuclear matrix fraction, forming salt extraction-resistant foci-like structures in unchallenged nuclei. Remarkably, damage-induced formation of both PAR and XRCC1 foci depended on SNF2H, and the PAR and XRCC1 foci co-localized with the SNF2H foci. We propose a model in which a base excision repair complex containing damaged chromatin is recruited to specific locations in the nuclear matrix for repair, with this recruitment mediated by XRCC1-SNF2H interaction.


Subject(s)
Adenosine Triphosphatases/genetics , Casein Kinase II/genetics , Chromosomal Proteins, Non-Histone/genetics , DNA Repair , DNA-Binding Proteins/genetics , DNA/genetics , Hydrogen Peroxide/pharmacology , Adenosine Triphosphatases/antagonists & inhibitors , Adenosine Triphosphatases/metabolism , Casein Kinase II/metabolism , Cell Nucleus/drug effects , Cell Nucleus/metabolism , Chromatin/chemistry , Chromatin/drug effects , Chromatin/metabolism , Chromosomal Proteins, Non-Histone/antagonists & inhibitors , Chromosomal Proteins, Non-Histone/metabolism , DNA/metabolism , DNA Damage , DNA-Binding Proteins/antagonists & inhibitors , DNA-Binding Proteins/metabolism , Gene Expression , Gene Library , HeLa Cells , Humans , Phosphorylation , Protein Binding , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Two-Hybrid System Techniques , X-ray Repair Cross Complementing Protein 1
19.
Cell Rep ; 17(3): 862-875, 2016 10 11.
Article in English | MEDLINE | ID: mdl-27732860

ABSTRACT

Exercise has been argued to enhance cognitive function and slow progressive neurodegenerative disease. Although exercise promotes neurogenesis, oligodendrogenesis and adaptive myelination are also significant contributors to brain repair and brain health. Nonetheless, the molecular details underlying these effects remain poorly understood. Conditional ablation of the Snf2h gene impairs cerebellar development producing mice with poor motor function, progressive ataxia, and death between postnatal days 25-45. Here, we show that voluntary running induced an endogenous brain repair mechanism that resulted in a striking increase in hindbrain myelination and the long-term survival of Snf2h cKO mice. Further experiments identified the VGF growth factor as a major driver underlying this effect. VGF neuropeptides promote oligodendrogenesis in vitro, whereas Snf2h cKO mice treated with full-length VGF-encoding adenoviruses removed the requirement of exercise for survival. Together, these results suggest that VGF delivery could represent a therapeutic strategy for cerebellar ataxia and other pathologies of the CNS.


Subject(s)
Adenosine Triphosphatases/deficiency , Ataxia/metabolism , Chromosomal Proteins, Non-Histone/deficiency , Longevity , Neurogenesis , Neuropeptides/metabolism , Oligodendroglia/metabolism , Physical Conditioning, Animal , Adenosine Triphosphatases/metabolism , Adenoviridae/metabolism , Animals , Ataxia/pathology , Ataxia/physiopathology , Cell Differentiation , Cell Proliferation , Cells, Cultured , Cerebellum/metabolism , Cerebellum/pathology , Cerebellum/physiopathology , Cerebellum/ultrastructure , Chromosomal Proteins, Non-Histone/metabolism , Dendrites/metabolism , Dendrites/ultrastructure , Mice, Inbred C57BL , Mice, Knockout , Motor Activity , Myelin Sheath/metabolism , Oligodendroglia/pathology , Rhombencephalon/metabolism , Rhombencephalon/pathology , Rhombencephalon/physiopathology , Rhombencephalon/ultrastructure , Sequence Analysis, RNA , Signal Transduction
20.
Genes (Basel) ; 6(3): 592-606, 2015 Jul 14.
Article in English | MEDLINE | ID: mdl-26184323

ABSTRACT

Rapid progress in the study on the association of histone modifications with chromatin remodeling factors has broadened our understanding of chromatin dynamics in DNA transactions. In DNA double-strand break (DSB) repair, the well-known mark of histones is the phosphorylation of the H2A variant, H2AX, which has been used as a surrogate marker of DSBs. The ubiquitylation of histone H2B by RNF20 E3 ligase was recently found to be a DNA damage-induced histone modification. This modification is required for DSB repair and regulated by a distinctive pathway from that of histone H2AX phosphorylation. Moreover, the connection between H2B ubiquitylation and the chromatin remodeling activity of SNF2H has been elucidated. In this review, we summarize the current knowledge of RNF20-mediated processes and the molecular link to H2AX-mediated processes during DSB repair.

SELECTION OF CITATIONS
SEARCH DETAIL