Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters










Publication year range
1.
Cell Rep ; 43(8): 114558, 2024 Jul 30.
Article in English | MEDLINE | ID: mdl-39088321

ABSTRACT

Chromatin priming promotes cell-type-specific gene expression, lineage differentiation, and development. The mechanism of chromatin priming has not been fully understood. Here, we report that mouse hematopoietic stem and progenitor cells (HSPCs) lacking the Baf155 subunit of the BAF (BRG1/BRM-associated factor) chromatin remodeling complex produce a significantly reduced number of mature blood cells, leading to a failure of hematopoietic regeneration upon transplantation and 5-fluorouracil (5-FU) injury. Baf155-deficient HSPCs generate particularly fewer neutrophils, B cells, and CD8+ T cells at homeostasis, supporting a more immune-suppressive tumor microenvironment and enhanced tumor growth. Single-nucleus multiomics analysis reveals that Baf155-deficient HSPCs fail to establish accessible chromatin in selected regions that are enriched for putative enhancers and binding motifs of hematopoietic lineage transcription factors. Our study provides a fundamental mechanistic understanding of the role of Baf155 in hematopoietic lineage chromatin priming and the functional consequences of Baf155 deficiency in regeneration and tumor immunity.

2.
Dev Dyn ; 253(1): 157-172, 2024 Jan.
Article in English | MEDLINE | ID: mdl-37083132

ABSTRACT

BACKGROUND: Essential patterning processes transform the heart tube into a compartmentalized organ with distinct chambers separated by an atrioventricular canal (AVC). This transition involves the refinement of expression of genes that are first found broadly throughout the heart tube and then become restricted to the AVC. Despite the importance of cardiac patterning, we do not fully understand the mechanisms that limit gene expression to the AVC. RESULTS: We show that the zebrafish gene smarcc1a, encoding a BAF chromatin remodeling complex subunit homologous to mammalian BAF155, is critical for cardiac patterning. In smarcc1a mutants, myocardial differentiation and heart tube assembly appear to proceed normally. Subsequently, the smarcc1a mutant heart fails to exhibit refinement of gene expression patterns to the AVC, and the persistence of broad gene expression is accompanied by failure of chamber expansion. In addition to their cardiac defects, smarcc1a mutants lack pectoral fins, indicating similarity to tbx5a mutants. However, comparison of smarcc1a and tbx5a mutants suggests that perturbation of tbx5a function is not sufficient to cause the smarcc1a mutant phenotype. CONCLUSIONS: Our data indicate an important role for Smarcc1a-containing chromatin remodeling complexes in regulating the changes in gene expression and morphology that distinguish the AVC from the cardiac chambers.


Subject(s)
Endocardial Cushions , Zebrafish , Animals , Zebrafish/genetics , Zebrafish/metabolism , Chromatin Assembly and Disassembly , Zebrafish Proteins/metabolism , Heart , Transcription Factors/genetics , Transcription Factors/metabolism , Gene Expression Regulation, Developmental , Mammals/metabolism
3.
Front Cell Dev Biol ; 9: 619538, 2021.
Article in English | MEDLINE | ID: mdl-34336815

ABSTRACT

Oligodendrocytes are responsible for axon myelination in the brain and spinal cord. Generation of oligodendrocytes entails highly regulated multistage neurodevelopmental events, including proliferation, differentiation and maturation. The chromatin remodeling BAF (mSWI/SNF) complex is a notable regulator of neural development. In our previous studies, we determined the indispensability of the BAF complex scaffolding subunits BAF155 and BAF170 for neurogenesis, whereas their role in gliogenesis is unknown. Here, we show that the expression of BAF155 and BAF170 is essential for the genesis of oligodendrocytes during brain development. We report that the ablation of BAF155 and BAF170 in the dorsal telencephalic (dTel) neural progenitors or in oligodendrocyte-producing progenitors in the ventral telencephalon (vTel) in double-conditional knockout (dcKO) mouse mutants, perturbed the process of oligodendrogenesis. Molecular marker and cell cycle analyses revealed impairment of oligodendrocyte precursor specification and proliferation, as well as overt depletion of oligodendrocytes pool in dcKO mutants. Our findings unveil a central role of BAF155 and BAF170 in oligodendrogenesis, and thus substantiate the involvement of the BAF complex in the production of oligodendrocytes in the forebrain.

4.
Cell Rep ; 33(7): 108395, 2020 11 17.
Article in English | MEDLINE | ID: mdl-33207205

ABSTRACT

The mammalian SWitch/Sucrose Non-Fermentable (SWI/SNF) chromatin-remodeling BAF (BRG1/BRM-associated factor) complex plays an essential role in developmental and pathological processes. We show that the deletion of Baf155, which encodes a subunit of the BAF complex, in the Tie2(+) lineage (Baf155 (CKO) leads to defects in yolk sac myeloid and definitive erythroid (EryD) lineage differentiation from erythromyeloid progenitors (EMPs). The chromatin of myeloid gene loci in Baf155 CKO EMPs is mostly inaccessible and enriched mainly by the ETS binding motif. BAF155 interacts with PU.1 and is recruited to PU.1 target gene loci together with p300 and KDM6a. Treatment of Baf155 CKO embryos with GSK126, an H3K27me2/3 methyltransferase EZH2 inhibitor, rescues myeloid lineage gene expression. This study uncovers indispensable BAF-mediated chromatin remodeling of myeloid gene loci at the EMP stage. Future studies exploiting epigenetics in the generation and application of EMP derivatives for tissue repair, regeneration, and disease are warranted.


Subject(s)
Cell Lineage/physiology , Chromatin Assembly and Disassembly/physiology , Transcription Factors/metabolism , Animals , Cell Differentiation/physiology , Chromatin/metabolism , Chromatin Assembly and Disassembly/genetics , Chromosomal Proteins, Non-Histone/metabolism , DNA Helicases/metabolism , DNA-Binding Proteins/metabolism , Epigenesis, Genetic/genetics , Erythroid Cells/metabolism , Female , HEK293 Cells , Humans , Male , Mice , Mice, Inbred C57BL , Mouse Embryonic Stem Cells/metabolism , Myeloid Cells/metabolism , Nuclear Proteins/metabolism , Proto-Oncogene Proteins/metabolism , Trans-Activators/metabolism
5.
Int J Mol Sci ; 21(7)2020 Apr 01.
Article in English | MEDLINE | ID: mdl-32244797

ABSTRACT

Human SNF5 and BAF155 constitute the core subunit of multi-protein SWI/SNF chromatin-remodeling complexes that are required for ATP-dependent nucleosome mobility and transcriptional control. Human SNF5 (hSNF5) utilizes its repeat 1 (RPT1) domain to associate with the SWIRM domain of BAF155. Here, we employed X-ray crystallography, nuclear magnetic resonance (NMR) spectroscopy, and various biophysical methods in order to investigate the detailed binding mechanism between hSNF5 and BAF155. Multi-angle light scattering data clearly indicate that hSNF5171-258 and BAF155SWIRM are both monomeric in solution and they form a heterodimer. NMR data and crystal structure of the hSNF5171-258/BAF155SWIRM complex further reveal a unique binding interface, which involves a coil-to-helix transition upon protein binding. The newly formed αN helix of hSNF5171-258 interacts with the ß2-α1 loop of hSNF5 via hydrogen bonds and it also displays a hydrophobic interaction with BAF155SWIRM. Therefore, the N-terminal region of hSNF5171-258 plays an important role in tumorigenesis and our data will provide a structural clue for the pathogenesis of Rhabdoid tumors and malignant melanomas that originate from mutations in the N-terminal loop region of hSNF5.


Subject(s)
Chromatin Assembly and Disassembly/genetics , Mutation , Nucleosomes/genetics , SMARCB1 Protein/genetics , Transcription Factors/genetics , Binding Sites/genetics , Circular Dichroism , Crystallography, X-Ray , Gene Expression Regulation , Humans , Magnetic Resonance Spectroscopy , Melanoma/genetics , Melanoma/metabolism , Melanoma/pathology , Nucleosomes/metabolism , Protein Binding , Rhabdoid Tumor/genetics , Rhabdoid Tumor/metabolism , Rhabdoid Tumor/pathology , SMARCB1 Protein/chemistry , SMARCB1 Protein/metabolism , Transcription Factors/chemistry , Transcription Factors/metabolism
6.
Cancer Cell ; 37(2): 157-167.e6, 2020 02 10.
Article in English | MEDLINE | ID: mdl-32004442

ABSTRACT

In response to DNA double-strand breaks, MAD2L2-containing shieldin complex plays a critical role in the choice between homologous recombination (HR) and non-homologous end-joining (NHEJ)-mediated repair. Here we show that EZH2 inhibition upregulates MAD2L2 and sensitizes HR-proficient epithelial ovarian cancer (EOC) to poly(adenosine diphosphate-ribose) polymerase (PARP) inhibitor in a CARM1-dependent manner. CARM1 promotes MAD2L2 silencing by driving the switch from the SWI/SNF complex to EZH2 through methylating the BAF155 subunit of the SWI/SNF complex on the MAD2L2 promoter. EZH2 inhibition upregulates MAD2L2 to decrease DNA end resection, which increases NHEJ and chromosomal abnormalities, ultimately causing mitotic catastrophe in PARP inhibitor treated HR-proficient cells. Significantly, EZH2 inhibitor sensitizes CARM1-high, but not CARM-low, EOCs to PARP inhibitors in both orthotopic and patient-derived xenografts.


Subject(s)
Enhancer of Zeste Homolog 2 Protein/antagonists & inhibitors , Homologous Recombination/drug effects , Ovarian Neoplasms/drug therapy , Poly(ADP-ribose) Polymerase Inhibitors/pharmacology , Antineoplastic Agents/therapeutic use , DNA Breaks, Double-Stranded/drug effects , DNA End-Joining Repair/drug effects , Enzyme Inhibitors/therapeutic use , Female , Humans , Ovarian Neoplasms/genetics , Protein-Arginine N-Methyltransferases/drug effects , Recombinational DNA Repair/drug effects
7.
Emerg Microbes Infect ; 8(1): 1393-1405, 2019.
Article in English | MEDLINE | ID: mdl-31533543

ABSTRACT

HBx is a short-lived protein whose rapid turnover is mainly regulated by ubiquitin-dependent proteasomal degradation pathways. Our prior work identified BAF155 to be one of the HBx binding partners. Since BAF155 has been shown to stabilize other members of the SWI/SNF chromatin remodelling complex by attenuating their proteasomal degradation, we proposed that BAF155 might also contribute to stabilizing HBx protein in a proteasome-dependent manner. Here we report that BAF155 protected hepatitis B virus X protein (HBx) from ubiquitin-independent proteasomal degradation by competing with the 20S proteasome subunit PSMA7 to bind to HBx. BAF155 was found to directly interact with HBx via binding of its SANT domain to the HBx region between amino acid residues 81 and 120. Expression of either full-length BAF155 or SANT domain increased HBx protein levels whereas siRNA-mediated knockdown of endogenous BAF155 reduced HBx protein levels. Increased HBx stability and steady-state level by BAF155 were attributable to inhibition of ubiquitin-independent and PSMA7-mediated protein degradation. Consequently, overexpression of BAF155 enhanced the transcriptional transactivation function of HBx, activated protooncogene expression and inhibited hepatoma cell clonogenicity. These results suggest that BAF155 plays important roles in ubiquitin-independent degradation of HBx, which may be related to the pathogenesis and carcinogenesis of HBV-associated HCC.


Subject(s)
Carcinoma, Hepatocellular/virology , Liver Neoplasms/virology , Proteasome Endopeptidase Complex/metabolism , Trans-Activators/metabolism , Transcription Factors/genetics , Ubiquitin/metabolism , Cell Line , Chromatin Assembly and Disassembly , Hep G2 Cells , Hepatitis B/complications , Hepatitis B virus/genetics , Humans , Proteasome Endopeptidase Complex/genetics , Trans-Activators/genetics , Viral Regulatory and Accessory Proteins , Virus Replication
8.
Mol Neurobiol ; 56(11): 7305-7320, 2019 Nov.
Article in English | MEDLINE | ID: mdl-31020615

ABSTRACT

Chromatin remodeling factor BAF155 is an important regulator of many biological processes. As a core and scaffold subunit of the BAF (SWI/SNF-like) complex, BAF155 is capable of regulating the stability and function of the BAF complex. The spatiotemporal expression of BAF155 during embryogenesis is essential for various aspects of organogenesis, particularly in the brain development. However, our understanding of the mechanisms that regulate the expression and function of BAF155 is limited. Here, we report that RBM15, a subunit of the m6A methyltransferase complex, interacts with BAF155 mRNA and mediates BAF155 mRNA degradation through the mRNA methylation machinery. Ablation of endogenous RBM15 expression in cultured neuronal cells and in the developing cortex augmented the expression of BAF155. Conversely, RBM15 overexpression decreased BAF155 mRNA and protein levels, and perturbed BAF155 functions in vivo, including repression of BAF155-dependent transcriptional activity and delamination of apical radial glial progenitors as a hallmark of basal radial glial progenitor genesis. Furthermore, we demonstrated that the regulation of BAF155 by RBM15 depends on the activity of the mRNA methylation complex core catalytic subunit METTL3. Altogether, our findings reveal a new regulatory avenue that elucidates how BAF complex subunit stoichiometry and functional modulation are achieved in mammalian cells.


Subject(s)
Cerebral Cortex/embryology , Cerebral Cortex/metabolism , Chromatin Assembly and Disassembly , RNA-Binding Proteins/metabolism , RNA/metabolism , Adenosine/analogs & derivatives , Adenosine/metabolism , Adherens Junctions/metabolism , Animals , Cell Line , Humans , Methylation , Methyltransferases/metabolism , Mice , Models, Biological , Neuroglia/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA-Binding Proteins/genetics , Transcription Factors
9.
Mol Reprod Dev ; 84(12): 1238-1249, 2017 12.
Article in English | MEDLINE | ID: mdl-29024220

ABSTRACT

In vitro embryo production is an established method for both humans and animals, but is fraught with inferior development and health issues in offspring born after in vitro fertilization procedures. Analysis of epigenetic changes caused by exposure to in vitro conditions should shed light on potential sources of these phenotypes. Using immunocytochemistry, we investigated the localization and relative abundance of components associated with the SWI/SNF (Switch/Sucrose non-fermentable) chromatin-remodeling complex-including BAF155, BAF170, BAF180, BAF53A, BAF57, BAF60A, BAF45D, ARID1A, ARID1B, ARID2, SNF5, and BRD7-in oocytes and in in vitro-produced and in vivo-derived porcine embryos. Differences in the localization of BAF155, BAF170, BAF60A, and ARID1B among these sources indicate that improper timing of chromatin remodeling and cellular differentiation might occur in early preimplantation embryos produced and cultured in vitro.


Subject(s)
Blastocyst/metabolism , Chromatin Assembly and Disassembly/physiology , Gene Expression Regulation, Developmental/physiology , Multiprotein Complexes/biosynthesis , Animals , Blastocyst/cytology , Swine
10.
J Mol Biol ; 429(11): 1650-1660, 2017 06 02.
Article in English | MEDLINE | ID: mdl-28438634

ABSTRACT

Mammalian BAF complexes are a subfamily of SWI/SNF ATP-dependent chromatin remodelers that dynamically modulate chromatin structure to regulate fundamental cellular processes including gene transcription, cell cycle control, and DNA damage response. So far, many distinct BAF complexes have been identified with polymorphic assemblies of up to 15 subunits from 29 genes. The evolutionarily conserved BRG1/BRM, BAF47, and BAF155/BAF170 form a stable complex that carries out essential chromatin remodeling activity and therefore have been regarded as the core components of BAF complex. Here, we first confirmed that SWIRM domain of BAF155 is responsible for its interaction with BAF47 and then narrowed down the SWIRM-binding region in BAF47 to the Repeat 1 (RPT1) domain. We further presented the high-resolution crystal structure of SWIRM/RPT1 complex. Extensive mutagenesis experiments together with isothermal titration calorimetry and NMR titrations were performed to corroborate the interactions observed in crystal structure. Overall, we demonstrated that BAF155 SWIRM is a modular domain involved in BAF47 interaction, which is functionally distinct from other characterized SWIRM domains that possess DNA binding activity.


Subject(s)
SMARCB1 Protein/chemistry , SMARCB1 Protein/metabolism , Transcription Factors/chemistry , Transcription Factors/metabolism , Calorimetry , Crystallography, X-Ray , DNA Mutational Analysis , Humans , Magnetic Resonance Spectroscopy , Models, Molecular , Protein Binding , Protein Conformation , Protein Interaction Mapping , SMARCB1 Protein/genetics
11.
Biosci Rep ; 36(3)2016 07.
Article in English | MEDLINE | ID: mdl-27190130

ABSTRACT

Recent experimental evidence increasingly shows that the dysregulation of cellular bioenergetics is associated with a wide array of common human diseases, including cancer, neurological diseases and diabetes. Respiration provides a vital source of cellular energy for most eukaryotic cells, particularly high energy demanding cells. However, the understanding of how respiration is globally regulated is very limited. Interestingly, recent evidence suggests that Swi3 is an important regulator of respiration genes in yeast. In this report, we performed an array of biochemical and genetic experiments and computational analysis to directly evaluate the function of Swi3 and its human homologues in regulating respiration. First, we showed, by computational analysis and measurements of oxygen consumption and promoter activities, that Swi3, not Swi2, regulates genes encoding functions involved in respiration and oxygen consumption. Biochemical analysis showed that the levels of mitochondrial respiratory chain complexes were substantially increased in Δswi3 cells, compared with the parent cells. Additionally, our data showed that Swi3 strongly affects haem/oxygen-dependent activation of respiration gene promoters whereas Swi2 affects only the basal, haem-independent activities of these promoters. We found that increased expression of aerobic expression genes is correlated with increased oxygen consumption and growth rates in Δswi3 cells in air. Furthermore, using computational analysis and RNAi knockdown, we showed that the mammalian Swi3 BAF155 and BAF170 regulate respiration in HeLa cells. Together, these experimental and computational data demonstrated that Swi3 and its mammalian homologues are key regulators in regulating respiration.


Subject(s)
Nuclear Proteins/genetics , Respiration/genetics , Saccharomyces cerevisiae Proteins/genetics , Transcription Factors/genetics , Adenosine Triphosphatases , Amino Acid Sequence/genetics , Animals , Chromatin/genetics , DNA-Binding Proteins/genetics , Energy Metabolism/genetics , HeLa Cells , Humans , Oxygen Consumption/genetics , Promoter Regions, Genetic , Saccharomyces cerevisiae/genetics , Sequence Homology, Amino Acid
12.
Dev Neurobiol ; 74(5): 483-97, 2014 May.
Article in English | MEDLINE | ID: mdl-24170322

ABSTRACT

Failure of embryonic neural tube closure results in the second most common class of birth defects known as neural tube defects (NTDs). While NTDs are likely the result of complex multigenic dysfunction, it is not known whether polymorphisms in epigenetic regulators may be risk factors for NTDs. Here we characterized Baf155(msp3) , a unique ENU-induced allele in mice. Homozygous Baf155(mps3) embryos exhibit highly penetrant exencephaly, allowing us to investigate the roles of an assembled, but malfunctional BAF chromatin remodeling complex in vivo at the time of neural tube closure. Evidence of defects in proliferation and apoptosis were found within the neural tube. RNA-Seq analysis revealed that surprisingly few genes showed altered expression in Baf155 mutant neural tissue, given the broad epigenetic role of the BAF complex, but included genes involved in neural development and cell survival. Moreover, gene expression changes between individual mutants were variable even though the NTD was consistently observed. This suggests that inconsistent gene regulation contributes to failed neural tube closure. These results shed light on the role of the BAF complex in the process of neural tube closure and highlight the importance of studying missense alleles to understand epigenetic regulation during critical phases of development.


Subject(s)
Mutation, Missense , Neural Tube Defects/genetics , Neural Tube Defects/pathology , Transcription Factors/genetics , Alleles , Animals , Blotting, Western , Cell Death/genetics , Cell Death/physiology , Gene Expression , Genotyping Techniques , Immunoprecipitation , Mice, Inbred C57BL , Mice, Inbred Strains , Neural Stem Cells/pathology , Neural Stem Cells/physiology , Neural Tube/metabolism , Neural Tube/pathology , Neural Tube Defects/physiopathology , Neurogenesis/genetics , Neurogenesis/physiology , Phenotype
13.
Immune Network ; : 224-228, 2004.
Article in English | WPRIM (Western Pacific) | ID: wpr-13654

ABSTRACT

BACKGROUND: The expression of BRG1 associated factors (BAF) 155 and BAF 170 in response to IFN-gamma or TNF-alpha was studied in astrocytoma cell lines and primary astrocytes. BAFs are complexed with BRG1 and are also associated with activated glucocorticoid for glucocorticoid trans-activation. METHODS: IFN-gamma was pretreated for 18 hrs and cells were incubated with IL-1 or TNF-alpha for 72 hrs or 96 hrs with different concentrations of steroid. Cell death was measured by LDH assay. BAF expression was assayed by RT-PCR. RESULTS: IFN-gamma increased cell death by dexamethasone in LN215 cells but not in LN319 cells. The IFN-gamma increased the expression of BAF 155 and BAF 170 in adult astrocytes and LN215 cells, but IFN-gamma decreased the expression of BAF 155/170 in LN319 cells. The effect of IFN-gamma on the expression of BAF was not as clear in fetal astrocytes as it was in adult astrocytes. CONCLUSION: Our results suggest cytokines produced during immune reaction or immunotherapy may modulate steroid susceptibility of astrocytes and astrocytoma cells by influencing the expression of BAFs.


Subject(s)
Adult , Humans , Astrocytes , Astrocytoma , Cell Death , Cell Line , Cytokines , Dexamethasone , Immunotherapy , Interleukin-1 , Tumor Necrosis Factor-alpha
SELECTION OF CITATIONS
SEARCH DETAIL