Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 28
Filter
Add more filters

Publication year range
1.
Mol Cell ; 84(9): 1637-1650.e10, 2024 May 02.
Article in English | MEDLINE | ID: mdl-38604171

ABSTRACT

Long interspersed element-1 (LINE-1 or L1) comprises 17% of the human genome, continuously generates genetic variations, and causes disease in certain cases. However, the regulation and function of L1 remain poorly understood. Here, we uncover that L1 can enrich RNA polymerase IIs (RNA Pol IIs), express L1 chimeric transcripts, and create contact domain boundaries in human cells. This impact of L1 is restricted by a nuclear matrix protein scaffold attachment factor B (SAFB) that recognizes transcriptionally active L1s by binding L1 transcripts to inhibit RNA Pol II enrichment. Acute inhibition of RNA Pol II transcription abolishes the domain boundaries associated with L1 chimeric transcripts, indicating a transcription-dependent mechanism. Deleting L1 impairs domain boundary formation, and L1 insertions during evolution have introduced species-specific domain boundaries. Our data show that L1 can create RNA Pol II-enriched regions that alter genome organization and that SAFB regulates L1 and RNA Pol II activity to preserve gene regulation.


Subject(s)
Long Interspersed Nucleotide Elements , Matrix Attachment Region Binding Proteins , RNA Polymerase II , Receptors, Estrogen , Transcription, Genetic , Humans , RNA Polymerase II/metabolism , RNA Polymerase II/genetics , Long Interspersed Nucleotide Elements/genetics , Matrix Attachment Region Binding Proteins/metabolism , Matrix Attachment Region Binding Proteins/genetics , Nuclear Matrix-Associated Proteins/metabolism , Nuclear Matrix-Associated Proteins/genetics , Gene Expression Regulation , Protein Binding , HEK293 Cells , Genome, Human
2.
Mol Cell ; 78(2): 289-302.e6, 2020 04 16.
Article in English | MEDLINE | ID: mdl-32302541

ABSTRACT

Microprocessor initiates the processing of microRNAs (miRNAs) from the hairpin regions of primary transcripts (pri-miRNAs). Pri-miRNAs often contain multiple miRNA hairpins, and this clustered arrangement can assist in the processing of otherwise defective hairpins. We find that miR-451, which derives from a hairpin with a suboptimal terminal loop and a suboptimal stem length, accumulates to 40-fold higher levels when clustered with a helper hairpin. This phenomenon tolerates changes in hairpin order, linker lengths, and the identities of the helper hairpin, the recipient hairpin, the linker-sequence, and the RNA polymerase that transcribes the hairpins. It can act reciprocally and need not occur co-transcriptionally. It requires Microprocessor recognition of the helper hairpin and linkage of the two hairpins, yet predominantly manifests after helper-hairpin processing. It also requires enhancer of rudimentary homolog (ERH), which copurifies with Microprocessor and can dimerize and interact with other proteins that can dimerize, suggesting a model in which one Microprocessor recruits another Microprocessor.


Subject(s)
Cell Cycle Proteins/genetics , MicroRNAs/genetics , RNA Polymerase III/genetics , Transcription Factors/genetics , DNA-Directed RNA Polymerases/genetics , Gene Expression Regulation/genetics , Humans , Nucleic Acid Conformation , RNA Processing, Post-Transcriptional/genetics , RNA-Binding Proteins/genetics , Regulatory Sequences, Nucleic Acid/genetics , Transcription, Genetic
3.
Mol Cell ; 78(5): 876-889.e6, 2020 06 04.
Article in English | MEDLINE | ID: mdl-32502422

ABSTRACT

Many microRNAs (miRNAs) are generated from primary transcripts containing multiple clustered stem-loop structures that are thought to be recognized and cleaved by the Microprocessor complex as independent units. Here, we uncover an unexpected mode of processing of the bicistronic miR-15a-16-1 cluster. We find that the primary miR-15a stem-loop is not processed on its own but that the presence of the neighboring primary miR-16-1 stem-loop on the same transcript can compensate for this deficiency in cis. Using a CRISPR/Cas9 screen, we identify SAFB2 (scaffold attachment factor B2) as an essential co-factor in this miR-16-1-assisted pri-miR-15 cleavage and describe SAFB2 as an accessory protein of the Microprocessor. Notably, SAFB2-mediated cleavage expands to other clustered pri-miRNAs, indicating a general mechanism. Together, our study reveals an unrecognized function of SAFB2 in miRNA processing and suggests a scenario in which SAFB2 enables the binding and processing of suboptimal Microprocessor substrates in clustered primary miRNA transcripts.


Subject(s)
Matrix Attachment Region Binding Proteins/metabolism , MicroRNAs/metabolism , Nuclear Matrix-Associated Proteins/metabolism , Receptors, Estrogen/metabolism , Animals , Cell Line , Cell Nucleus/metabolism , HEK293 Cells , Humans , Inverted Repeat Sequences/genetics , Inverted Repeat Sequences/physiology , Matrix Attachment Region Binding Proteins/genetics , Mice , MicroRNAs/genetics , Nuclear Matrix-Associated Proteins/genetics , Nucleic Acid Conformation , RNA Processing, Post-Transcriptional/genetics , RNA-Binding Proteins/metabolism , Receptors, Estrogen/genetics
4.
Mol Cell ; 77(2): 368-383.e7, 2020 01 16.
Article in English | MEDLINE | ID: mdl-31677973

ABSTRACT

Interphase chromatin is hierarchically organized into higher-order architectures that are essential for gene functions, yet the biomolecules that regulate these 3D architectures remain poorly understood. Here, we show that scaffold attachment factor B (SAFB), a nuclear matrix (NM)-associated protein with RNA-binding functions, modulates chromatin condensation and stabilizes heterochromatin foci in mouse cells. SAFB interacts via its R/G-rich region with heterochromatin-associated repeat transcripts such as major satellite RNAs, which promote the phase separation driven by SAFB. Depletion of SAFB leads to changes in 3D genome organization, including an increase in interchromosomal interactions adjacent to pericentromeric heterochromatin and a decrease in genomic compartmentalization, which could result from the decondensation of pericentromeric heterochromatin. Collectively, we reveal the integrated roles of NM-associated proteins and repeat RNAs in the 3D organization of heterochromatin, which may shed light on the molecular mechanisms of nuclear architecture organization.


Subject(s)
Heterochromatin/genetics , Matrix Attachment Region Binding Proteins/genetics , Nuclear Matrix-Associated Proteins/genetics , RNA, Satellite/genetics , Receptors, Estrogen/genetics , Animals , Cell Line , Chromatin/genetics , Genome/genetics , Humans , Mice
5.
RNA ; 29(10): 1535-1556, 2023 10.
Article in English | MEDLINE | ID: mdl-37468167

ABSTRACT

Scaffold attachment factor B (SAFB) is a conserved RNA-binding protein that is essential for early mammalian development. However, the functions of SAFB in mouse embryonic stem cells (ESCs) have not been characterized. Using RNA immunoprecipitation followed by RNA-seq (RIP-seq), we examined the RNAs associated with SAFB in wild-type and SAFB/SAFB2 double-knockout ESCs. SAFB predominantly associated with introns of protein-coding genes through purine-rich motifs. The transcript most enriched in SAFB association was the lncRNA Malat1, which also contains a purine-rich region in its 5' end. Knockout of SAFB/SAFB2 led to differential expression of approximately 1000 genes associated with multiple biological processes, including apoptosis, cell division, and cell migration. Knockout of SAFB/SAFB2 also led to splicing changes in a set of genes that were largely distinct from those that exhibited changes in expression level. The spliced and nascent transcripts of many genes whose expression levels were positively regulated by SAFB also associated with high levels of SAFB, implying that SAFB binding promotes their expression. Reintroduction of SAFB into double-knockout cells restored gene expression toward wild-type levels, an effect again observable at the level of spliced and nascent transcripts. Proteomics analysis revealed a significant enrichment of nuclear speckle-associated and RS domain-containing proteins among SAFB interactors. Neither Xist nor Polycomb functions were dramatically altered in SAFB/2 knockout ESCs. Our findings suggest that among other potential functions in ESCs, SAFB promotes the expression of certain genes through its ability to bind nascent RNA.


Subject(s)
Mouse Embryonic Stem Cells , RNA , Animals , Mice , Gene Expression , Introns , Mammals , Mice, Knockout
6.
Brief Bioinform ; 22(6)2021 11 05.
Article in English | MEDLINE | ID: mdl-34056657

ABSTRACT

Long non-coding RNAs (lncRNAs) are key regulators of major biological processes and their functional modes are dictated by their subcellular localization. Relative nuclear enrichment of lncRNAs compared to mRNAs is a prevalent phenomenon but the molecular mechanisms governing their nuclear retention in cells remain largely unknown. Here in this study, we harness the recently released eCLIP data for a large number of RNA-binding proteins (RBPs) in K562 and HepG2 cells and utilize multiple bioinformatics methods to comprehensively survey the roles of RBPs in lncRNA nuclear retention. We identify an array of splicing RBPs that bind to nuclear-enriched lincRNAs (large intergenic non-coding RNAs) thus may act as trans-factors regulating their nuclear retention. Further analyses reveal that these RBPs may bind with distinct core motifs, flanking sequence compositions, or secondary structures to drive lincRNA nuclear retention. Moreover, network analyses uncover potential co-regulatory RBP clusters and the physical interaction between HNRNPU and SAFB2 proteins in K562 cells is further experimentally verified. Altogether, our analyses reveal previously unknown factors and mechanisms that govern lincRNA nuclear localization in cells.


Subject(s)
Computational Biology/methods , Models, Biological , RNA Transport , RNA, Long Noncoding/metabolism , RNA-Binding Proteins/metabolism , Binding Sites , Cell Line, Tumor , Cell Nucleus/genetics , Cell Nucleus/metabolism , Humans , Nucleic Acid Conformation , Protein Binding , RNA, Long Noncoding/genetics , RNA-Seq
7.
Proc Natl Acad Sci U S A ; 117(50): 31914-31922, 2020 12 15.
Article in English | MEDLINE | ID: mdl-33257571

ABSTRACT

Inhibiting membrane association of RAS has long been considered a rational approach to anticancer therapy, which led to the development of farnesyltransferase inhibitors (FTIs). However, FTIs proved ineffective against KRAS-driven tumors. To reveal alternative therapeutic strategies, we carried out a genome-wide CRISPR-Cas9 screen designed to identify genes required for KRAS4B membrane association. We identified five enzymes in the prenylation pathway and SAFB, a nuclear protein with both DNA and RNA binding domains. Silencing SAFB led to marked mislocalization of all RAS isoforms as well as RAP1A but not RAB7A, a pattern that phenocopied silencing FNTA, the prenyltransferase α subunit shared by farnesyltransferase and geranylgeranyltransferase type I. We found that SAFB promoted RAS membrane association by controlling FNTA expression. SAFB knockdown decreased GTP loading of RAS, abrogated alternative prenylation, and sensitized RAS-mutant cells to growth inhibition by FTI. Our work establishes the prenylation pathway as paramount in KRAS membrane association, reveals a regulator of prenyltransferase expression, and suggests that reduction in FNTA expression may enhance the efficacy of FTIs.


Subject(s)
Cell Membrane/metabolism , Dimethylallyltranstransferase/metabolism , Matrix Attachment Region Binding Proteins/metabolism , Neoplasms/pathology , Nuclear Matrix-Associated Proteins/metabolism , Proto-Oncogene Proteins p21(ras)/metabolism , Receptors, Estrogen/metabolism , Alkyl and Aryl Transferases/genetics , Alkyl and Aryl Transferases/metabolism , CRISPR-Cas Systems/genetics , Computational Biology , Datasets as Topic , Gene Knockdown Techniques , Humans , Matrix Attachment Region Binding Proteins/genetics , Neoplasms/genetics , Nuclear Matrix-Associated Proteins/genetics , Protein Prenylation , Protein Subunits/metabolism , Proto-Oncogene Proteins p21(ras)/genetics , Receptors, Estrogen/genetics
8.
Basic Res Cardiol ; 116(1): 15, 2021 03 06.
Article in English | MEDLINE | ID: mdl-33675440

ABSTRACT

Long non-coding RNAs (lncRNAs) account for a large proportion of genomic transcripts and are critical regulators in various cardiac diseases. Though lncRNAs have been reported to participate in the process of diverse cardiac diseases, the contribution of lncRNAs in cardiac fibrosis remains to be fully elucidated. Here, we identified a novel anti-fibrotic lncRNA, SAIL (scaffold attachment factor B interacting lncRNA). SAIL was reduced in cardiac fibrotic tissue and activated cardiac fibroblasts. Gain- and loss-of-function studies showed that knockdown of SAIL promoted proliferation and collagen production of cardiac fibroblasts with or without TGF-ß1 (transforming growth factor beta1) treatment, while overexpression of SAIL did the opposite. In mouse cardiac fibrosis induced by myocardial infarction, knockdown of SAIL exacerbated, whereas overexpression of SAIL alleviated cardiac fibrosis. Mechanically, SAIL inhibited the fibrotic process by directly binding with SAFB via 23 conserved nucleotide sequences, which in turn blocked the access of SAFB to RNA pol II (RNA polymerase II) and reduced the transcription of fibrosis-related genes. Intriguingly, the human conserved fragment of SAIL (hSAIL) significantly suppressed the proliferation and collagen production of human cardiac fibroblasts. Our findings demonstrate that SAIL regulates cardiac fibrosis by regulating SAFB-mediated transcription of fibrotic related genes. Both SAIL and SAFB hold the potential to become novel therapeutic targets for cardiac fibrosis.


Subject(s)
Cell Proliferation , Collagen/metabolism , Fibroblasts/metabolism , Myocardial Infarction/metabolism , Myocytes, Cardiac/metabolism , RNA, Long Noncoding/metabolism , Animals , Cells, Cultured , DNA-Binding Proteins/metabolism , Disease Models, Animal , Fibroblasts/pathology , Fibrosis , Male , Mice, Inbred C57BL , Myocardial Infarction/genetics , Myocardial Infarction/pathology , Myocytes, Cardiac/pathology , RNA Polymerase II/metabolism , RNA, Long Noncoding/genetics , RNA-Binding Proteins/metabolism , Transcription, Genetic
9.
Histochem Cell Biol ; 156(3): 239-251, 2021 Sep.
Article in English | MEDLINE | ID: mdl-34129097

ABSTRACT

Estrogen-related receptor (ERR), a member of the nuclear receptor superfamily, consists of three subtypes (α, ß, γ) and has strong homology with estrogen receptor. No endogenous ligands have been identified for ERRs, but they play key roles in metabolic, hormonal, and developmental processes as transcription factors without ligand binding. Although subnuclear dynamics are essential for nuclear events including nuclear receptor-mediated transcriptional regulation, the dynamics of ERRs are poorly understood. Here, we report that ERRs show subcellular kinetic changes in response to diethylstilbestrol (DES), a synthetic estrogen that represses the transactivity of all three ERR subtypes, using live-cell imaging with fluorescent protein labeling. Upon DES treatment, all ERR subtypes formed discrete clusters in the nucleus, with ERRγ also displaying nuclear export. Fluorescence recovery after photobleaching analyses revealed significant reductions in the intranuclear mobility of DES-bound ERRα and ERRß, and a slight reduction in the intranuclear mobility of DES-bound ERRγ. After DES treatment, colocalization of all ERR subtypes with scaffold attachment factor B1 (SAFB1), a nuclear matrix-associated protein, was observed in dot-like subnuclear clusters, suggesting interactions of the ERRs with the nuclear matrix. Consistently, co-immunoprecipitation analyses confirmed enhanced interactions between ERRs and SAFB1 in the presence of DES. SAFB1 was clarified to repress the transactivity of all ERR subtypes through the ERR-response element. These results demonstrate ligand-dependent cluster formation of ERRs in the nucleus that is closely associated with SAFB1-mediated transrepression. Taken together, the present findings provide a new understanding of the pathophysiology regulated by ERR/SAFB1 signaling pathways and their subcellular dynamics.


Subject(s)
Matrix Attachment Region Binding Proteins/metabolism , Nuclear Matrix-Associated Proteins/metabolism , Receptors, Estrogen/metabolism , Animals , Cell Nucleus/metabolism , Cells, Cultured , Chlorocebus aethiops , Humans , Matrix Attachment Region Binding Proteins/analysis , Nuclear Matrix-Associated Proteins/analysis , Receptors, Estrogen/analysis , Signal Transduction , Transcriptional Activation
10.
Int J Mol Sci ; 22(9)2021 May 07.
Article in English | MEDLINE | ID: mdl-34067147

ABSTRACT

Stress resistance mechanisms include upregulation of heat shock proteins (HSPs) and formation of granules. Stress-induced granules are classified into stress granules and nuclear stress bodies (nSBs). The present study examined the involvement of nSB formation in thermal resistance. We used chemical compounds that inhibit heat shock transcription factor 1 (HSF1) and scaffold attachment factor B (SAFB) granule formation and determined their effect on granule formation and HSP expression in HeLa cells. We found that formation of HSF1 and SAFB granules was inhibited by 2,5-hexanediol. We also found that suppression of HSF1 and SAFB granule formation enhanced heat stress-induced apoptosis. In addition, the upregulation of HSP27 and HSP70 during heat stress recovery was suppressed by 2,5-hexanediol. Our results suggested that the formation of HSF1 and SAFB granules was likely to be involved in the upregulation of HSP27 and HSP70 during heat stress recovery. Thus, the formation of HSF1 and SAFB granules was involved in thermal resistance.


Subject(s)
Apoptosis , Heat Shock Transcription Factors/antagonists & inhibitors , Heat-Shock Response , Matrix Attachment Region Binding Proteins/metabolism , Nuclear Matrix-Associated Proteins/metabolism , Receptors, Estrogen/metabolism , Apoptosis/drug effects , Cell Proliferation/drug effects , Cell Survival/drug effects , Cytoplasmic Granules/drug effects , Cytoplasmic Granules/metabolism , Gene Knockdown Techniques , Glycols/pharmacology , HSP27 Heat-Shock Proteins/metabolism , HSP70 Heat-Shock Proteins/metabolism , HeLa Cells , Heat Shock Transcription Factors/metabolism , Heat-Shock Response/drug effects , Humans , Models, Biological , TRPV Cation Channels/antagonists & inhibitors , TRPV Cation Channels/metabolism , Temperature , Up-Regulation/drug effects
11.
J Biol Chem ; 294(14): 5549-5561, 2019 04 05.
Article in English | MEDLINE | ID: mdl-30782847

ABSTRACT

In Sonic hedgehog (SHH) signaling, GLI family zinc finger (GLI)-mediated diverse gene transcription outcomes are strictly regulated and are important for SHH function in both development and disease. However, how the GLI factors differentially regulate transcription in response to variable SHH activities is incompletely understood. Here, using a newly generated, tagged Gli3 knock-in mouse (Gli3TAP ), we performed proteomic analyses and identified the chromatin-associated SAFB-like transcription modulator (SLTM) as a GLI-interacting protein that context-dependently regulates GLI activities. Using immunoprecipitation and immunoblotting, RT-quantitative PCR, and ChIP assays, we show that SLTM interacts with all three GLI proteins and that its cellular levels are regulated by SHH. We also found that SLTM enhances GLI3 binding to chromatin and increases GLI3 repressor (GLI3R) form protein levels. In a GLI3-dependent manner, SLTM promoted the formation of a repressive chromatin environment and functioned as a GLI3 co-repressor. In the absence of GLI3 or in the presence of low GLI3 levels, SLTM co-activated GLI activator (GLIA)-mediated target gene activation and cell differentiation. Moreover, in vivo Sltm deletion generated through CRISPR/Cas9-mediated gene editing caused perinatal lethality and SHH-related abnormal ventral neural tube phenotypes. We conclude that SLTM regulates GLI factor binding to chromatin and contributes to the transcriptional outcomes of SHH signaling via a novel molecular mechanism.


Subject(s)
Hedgehog Proteins/metabolism , Matrix Attachment Region Binding Proteins/metabolism , Nerve Tissue Proteins/metabolism , Nuclear Matrix-Associated Proteins/metabolism , Signal Transduction , Zinc Finger Protein Gli3/metabolism , Animals , CRISPR-Cas Systems , Chromatin , Gene Editing , Gene Knock-In Techniques , Hedgehog Proteins/genetics , Matrix Attachment Region Binding Proteins/genetics , Mice , Mice, Transgenic , Nerve Tissue Proteins/genetics , Nuclear Matrix-Associated Proteins/genetics , Proteomics , Zinc Finger Protein Gli3/genetics
12.
Arch Toxicol ; 92(4): 1581-1591, 2018 Apr.
Article in English | MEDLINE | ID: mdl-29380011

ABSTRACT

Bisphenol A (BPA), one of the most frequently detected emerging pollutants in the environment, has been implicated in adverse effects in male and female reproduction at extremely low concentrations. This study aimed to investigate the effects and potential mechanism of BPA on mouse ovarian follicular development and female germline stem cells (FGSCs). Female CD-1 adult mice were administered gradient concentrations of BPA (12.5, 25, and 50 mg/kg/day) by intraperitoneal injection. We found that the number of atretic ovarian follicles was significantly increased at high BPA concentrations. Additionally, the numbers of primordial follicles, primary follicles, and corpus luteum (CL) were significantly reduced at high BPA concentrations. Interestingly, the number of FGSCs was remarkably reduced in BPA-treated ovaries. Furthermore, the increased apoptotic rate of FGSCs in vitro was triggered by BPA accompanied by increased BPA concentrations. To investigate the mechanism of BPA in ovarian follicular development, 193 differentially expressed proteins were identified in BPA-treated ovaries by the isobaric tags for relative and absolute quantification-coupled 2D liquid chromatography-mass spectrometry technique. A total of 106 proteins were downregulated and 85 proteins were upregulated. Among these proteins, the apoptosis-related protein SAFB-like transcriptional modulator (SLTM) was remarkably upregulated, and this result was consistent with western blotting. Taken together, our results suggest that an ovarian follicular development, especially, the development of primordial follicles, primary follicles, and the CL, is inhibited by high BPA concentrations, and the ovarian follicle atresia is initiated by BPA through upregulated expression of SLTM. Furthermore, BPA induces apoptosis of cultured FGSCs. The effect of BPA on ovarian follicular development and FGSCs, especially the effect on FGSCs, suggests a novel mechanism of how BPA causes female infertility.


Subject(s)
Benzhydryl Compounds/toxicity , Estrogens, Non-Steroidal/toxicity , Oogonial Stem Cells/drug effects , Ovarian Follicle/drug effects , Phenols/toxicity , Animals , Apoptosis/drug effects , DNA-Binding Proteins/genetics , Female , Mice , Oogonial Stem Cells/cytology , Ovarian Follicle/growth & development , Ovarian Follicle/pathology , Proteomics , RNA-Binding Proteins/genetics , Up-Regulation
13.
J Autoimmun ; 76: 101-107, 2017 01.
Article in English | MEDLINE | ID: mdl-27682649

ABSTRACT

OBJECTIVE: To identify and characterize a novel connective tissue disease (CTD)-related autoantibody (autoAb) directed against scaffold attachment factor B (SAFB). METHODS: AutoAb specificity was analyzed using RNA and protein-immunoprecipitation assays. Autoimmune targets were affinity purified using patients' sera and subjected to liquid chromatography mass spectrometry. RESULTS: By immunoprecipitation assay, 10 sera reacted with a protein with a molecular weight of approximately 160 kDa. Liquid chromatography mass spectrometry of the partially purified autoantigen and additional immunoblot-based analyses revealed that the Ab specifically recognized SAFB. Anti-SAFB Abs were detected in 2 of 646 patients with systemic sclerosis (SSc) (0.3%), 1 of 1570 patients with polymyositis/dermatomyositis (0.06%), 4 of 270 patients with interstitial lung disease (ILD) (1.5%), 1 of 43 patients with overlap syndrome (2.3%) and 2 patients with other diseases including primary Raynaud's disease and eosinophilic pneumonia. Five patients with anti-SAFB Abs had Raynaud's phenomenon and 3 had nail fold punctate hemorrhage. Of note, 8 of the 10 patients (80%) suffered from ILD. None of the patients with anti-SAFB Abs had pulmonary arterial hypertension, heart disease, or renal involvement. CONCLUSIONS: Anti-SAFB Ab is a novel CTD-related autoAb possibly associated with ILD.


Subject(s)
Autoantibodies/immunology , Autoantigens/immunology , Lung Diseases, Interstitial/immunology , Matrix Attachment Region Binding Proteins/immunology , Nuclear Matrix-Associated Proteins/immunology , Receptors, Estrogen/immunology , Aged , Biomarkers , Case-Control Studies , Connective Tissue Diseases/diagnosis , Connective Tissue Diseases/immunology , Enzyme-Linked Immunosorbent Assay , Female , Humans , Lung Diseases, Interstitial/diagnosis , Male , Middle Aged , Phenotype
14.
Elife ; 132024 May 09.
Article in English | MEDLINE | ID: mdl-38722021

ABSTRACT

Neural stem cells (NSCs) are multipotent and correct fate determination is crucial to guarantee brain formation and homeostasis. How NSCs are instructed to generate neuronal or glial progeny is not well understood. Here, we addressed how murine adult hippocampal NSC fate is regulated and described how scaffold attachment factor B (SAFB) blocks oligodendrocyte production to enable neuron generation. We found that SAFB prevents NSC expression of the transcription factor nuclear factor I/B (NFIB) by binding to sequences in the Nfib mRNA and enhancing Drosha-dependent cleavage of the transcripts. We show that increasing SAFB expression prevents oligodendrocyte production by multipotent adult NSCs, and conditional deletion of Safb increases NFIB expression and oligodendrocyte formation in the adult hippocampus. Our results provide novel insights into a mechanism that controls Drosha functions for selective regulation of NSC fate by modulating the post-transcriptional destabilization of Nfib mRNA in a lineage-specific manner.


Subject(s)
Hippocampus , NFI Transcription Factors , Neural Stem Cells , Nuclear Matrix-Associated Proteins , RNA, Messenger , Animals , Mice , Cell Differentiation , Hippocampus/metabolism , Hippocampus/cytology , Neural Stem Cells/metabolism , NFI Transcription Factors/metabolism , NFI Transcription Factors/genetics , Nuclear Matrix-Associated Proteins/metabolism , Oligodendroglia/metabolism , Ribonuclease III/metabolism , Ribonuclease III/genetics , RNA Stability , RNA, Messenger/metabolism , RNA, Messenger/genetics
15.
Biol Open ; 12(6)2023 06 15.
Article in English | MEDLINE | ID: mdl-37283223

ABSTRACT

The Polycomb Repressive Complex 2 (PRC2) is a conserved enzyme that tri-methylates Lysine 27 on Histone 3 (H3K27me3) to promote gene silencing. PRC2 is remarkably responsive to the expression of certain long noncoding RNAs (lncRNAs). In the most notable example, PRC2 is recruited to the X-chromosome shortly after expression of the lncRNA Xist begins during X-chromosome inactivation. However, the mechanisms by which lncRNAs recruit PRC2 to chromatin are not yet clear. We report that a broadly used rabbit monoclonal antibody raised against human EZH2, a catalytic subunit of PRC2, cross-reacts with an RNA-binding protein called Scaffold Attachment Factor B (SAFB) in mouse embryonic stem cells (ESCs) under buffer conditions that are commonly used for chromatin immunoprecipitation (ChIP). Knockout of EZH2 in ESCs demonstrated that the antibody is specific for EZH2 by western blot (no cross-reactivity). Likewise, comparison to previously published datasets confirmed that the antibody recovers PRC2-bound sites by ChIP-Seq. However, RNA-IP from formaldehyde-crosslinked ESCs using ChIP wash conditions recovers distinct peaks of RNA association that co-localize with peaks of SAFB and whose enrichment disappears upon knockout of SAFB but not EZH2. IP and mass spectrometry-based proteomics in wild-type and EZH2 knockout ESCs confirm that the EZH2 antibody recovers SAFB in an EZH2-independent manner. Our data highlight the importance of orthogonal assays when studying interactions between chromatin-modifying enzymes and RNA.


Subject(s)
Matrix Attachment Region Binding Proteins , RNA, Long Noncoding , Humans , Animals , Mice , Enhancer of Zeste Homolog 2 Protein/genetics , Enhancer of Zeste Homolog 2 Protein/metabolism , RNA, Long Noncoding/genetics , Antibodies, Monoclonal/genetics , Antibodies, Monoclonal/metabolism , Mice, Knockout , Polycomb Repressive Complex 2/genetics , Polycomb Repressive Complex 2/metabolism , Chromatin , RNA-Binding Proteins/genetics , Receptors, Estrogen/genetics , Receptors, Estrogen/metabolism , Nuclear Matrix-Associated Proteins/genetics , Nuclear Matrix-Associated Proteins/metabolism , Matrix Attachment Region Binding Proteins/genetics , Matrix Attachment Region Binding Proteins/metabolism
16.
Mol Biotechnol ; 65(9): 1465-1475, 2023 Sep.
Article in English | MEDLINE | ID: mdl-36652182

ABSTRACT

Aberrant scaffold attachment factor-B2 (SAFB2) expression is associated with several malignant tumors. In this study, we investigated how SAFB2 worked in the process of breast cancer as well as the underlying mechanism. Quantitative real-time polymerase chain reaction (qRT-PCR) and western blotting analysis were used to investigate the expression of SAFB2 and nuclear factor of activated T cells 5 (NFAT5). Cellular proliferative ability was detected with cell counting kit 8 (CCK8), colony formation and 5-Ethynyl-2'-deoxyuridine (EdU) staining assays. Cell apoptosis was measured via flow cytometry and western blotting analysis. Wound healing, transwell assays, and western blotting analysis were executed to estimate cell migration and invasion. The relationship between SAFB2 and NFAT5 was verified by RNA immunoprecipitation (RIP) assay and NFAT5 mRNA stability was examined with actinomycin (Act) D assay. Western blotting analysis also tested the expression of Wnt/ß-catenin signaling-associated proteins. As a result, SAFB2 was downregulated in breast cancer cell lines, while NFAT5 was highly expressed in most breast cancer cell lines. Overexpression of SAFB2 suppressed the proliferation, migration, and invasion while exacerbated the apoptosis of breast cancer cells. SAFB2 interacted with NFAT5 mRNA and declined the stability of NFAT5 mRNA. Overexpression of NFAT5 counteracted anti-proliferative, anti-metastatic and pro-apoptotic effects of SAFB2 in breast cancer cells. Mechanistically, SAFB2 overexpression inhibited the Wnt/ß-catenin signaling pathway, while this effect was partially eliminated by NFAT5. Collectively, SAFB2 hindered breast cancer development and inactivated Wnt/ß-catenin signaling via regulation of NFAT5, suggesting that SAFB2 might be a promising therapeutic target for breast cancer.


Subject(s)
Breast Neoplasms , Matrix Attachment Region Binding Proteins , Humans , Female , Wnt Signaling Pathway/genetics , beta Catenin/genetics , beta Catenin/metabolism , Breast Neoplasms/drug therapy , Breast Neoplasms/genetics , MCF-7 Cells , RNA, Messenger/genetics , Cell Proliferation/genetics , Cell Line, Tumor , Cell Movement/genetics , Gene Expression Regulation, Neoplastic , Transcription Factors/genetics , Transcription Factors/metabolism , Receptors, Estrogen/genetics , Receptors, Estrogen/metabolism , Nuclear Matrix-Associated Proteins/genetics , Nuclear Matrix-Associated Proteins/metabolism , Matrix Attachment Region Binding Proteins/genetics , Matrix Attachment Region Binding Proteins/metabolism
17.
Redox Biol ; 58: 102545, 2022 12.
Article in English | MEDLINE | ID: mdl-36427398

ABSTRACT

The cellular response to hypoxia, in addition to HIF-dependent transcriptional reprogramming, also involves less characterized transcription-independent processes, such as alternative splicing of the VEGFA transcript leading to the production of the proangiogenic VEGF form. We now show that this event depends on reorganization of the splicing machinery, triggered after short-term hypoxia by ROS production and intranuclear redistribution of the nucleoskeletal proteins SAFB1/2. Exposure to low oxygen causes fast dissociation of SAFB1/2 from the nuclear matrix, which is reversible, inhibited by antioxidant treatment, and also observed under normoxia when the mitochondrial electron transport chain is blocked. This is accompanied by altered interactions between SAFB1/2 and the splicing machinery, translocation of kinase SRPK1 to the cytoplasm, and dephosphorylation of RS-splicing factors. Depletion of SAFB1/2 under normoxia phenocopies the hypoxic and ROS-mediated switch in VEGF mRNA splicing. These data suggest that ROS-dependent remodeling of the nuclear architecture can promote production of splicing variants that facilitate adaptation to hypoxia.


Subject(s)
Matrix Attachment Region Binding Proteins , Nuclear Matrix-Associated Proteins , Humans , Nuclear Matrix-Associated Proteins/genetics , Nuclear Matrix-Associated Proteins/metabolism , Matrix Attachment Region Binding Proteins/genetics , Matrix Attachment Region Binding Proteins/metabolism , Reactive Oxygen Species/metabolism , Vascular Endothelial Growth Factor A/genetics , Vascular Endothelial Growth Factor A/metabolism , Receptors, Estrogen/genetics , Receptors, Estrogen/metabolism , Nuclear Matrix/metabolism , Hypoxia/genetics , Hypoxia/metabolism , RNA, Messenger/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Cell Hypoxia/genetics , Protein Serine-Threonine Kinases
18.
Aging (Albany NY) ; 14(23): 9679-9698, 2022 12 02.
Article in English | MEDLINE | ID: mdl-36462498

ABSTRACT

The most hostile form of urologic cancer, clear cell renal cell carcinoma (ccRCC), has a high fatality rate and poor prognosis due to tumor metastasis at initial presentation. The complex process driving ccRCC metastasis is still unknown, though. In this study, we demonstrate that Spindle and kinetochore-associated protein 1 (SKA1) expression is significantly upregulated in ccRCC tissues and associated with aggressive clinicopathologic characteristics. Functionally, SKA1 knockdown on ccRCC cells reduced cancer cell motility both in vivo and in vitro research. These bioactivities of SKA1 may be brought on by its specific interaction with scaffold attachment factor B, according to the proposed mechanism (SAFB), which could further depress the transcription of dual specificity phosphatase 6 (DUSP6). Our findings may provide a new way of researching SKA1-regulated tumor metastasis, and indicate that SKA1 is a prospective therapeutic target for renal carcinoma.


Subject(s)
Carcinoma, Renal Cell , Carcinoma , Kidney Neoplasms , Matrix Attachment Region Binding Proteins , Humans , Carcinoma, Renal Cell/genetics , Carcinoma, Renal Cell/pathology , Dual Specificity Phosphatase 6 , Kidney Neoplasms/genetics , Kidney Neoplasms/pathology , Cell Line, Tumor , Cell Proliferation/genetics , Gene Expression Regulation, Neoplastic , Neoplasm Metastasis , Cell Movement/genetics , Receptors, Estrogen , Nuclear Matrix-Associated Proteins
19.
Am J Clin Exp Urol ; 9(4): 337-349, 2021.
Article in English | MEDLINE | ID: mdl-34541032

ABSTRACT

The nuclear matrix protein Scaffold Attachment Factor B1 (SAFB1, SAFB) can act in prostate cancer (PCa) as an androgen receptor (AR) co-repressor that functions through epigenetic silencing of AR targets, such as prostate specific antigen (PSA, KLK3). Genomic profiling of SAFB1-silenced PCa cells indicated that SAFB1 may play a role in modulating intracrine androgen levels through the regulation of UDP-glucuronosyltransferase (UGT) genes, which inactivate steroid hormones. Gene silencing of SAFB1 resulted in increased levels of free dihydrotesterosterone (DHT), and increased resistance to the AR inhibitor enzalutamide. SAFB1 silencing suppressed expression of the UDP-glucuronosyltransferase family 2 member B15 gene (UGT2B15) and the closely related UGT2B17 gene, which encode proteins that irreversibly inactivate testosterone (T) and DHT. Analysis of human data indicated that genomic loss at the SAFB locus, or down-regulation of expression of the SAFB gene, is associated with aggressive PCa. These findings identify SAFB1 as an important regulator of androgen catabolism in PCa and suggest that loss or inactivation of this protein may promote AR activity by retention of active androgen in tumor cells.

20.
Brain Pathol ; 30(6): 1041-1055, 2020 11.
Article in English | MEDLINE | ID: mdl-32580238

ABSTRACT

SAFB1 is a DNA and RNA binding protein that is highly expressed in the cerebellum and hippocampus and is involved in the processing of coding and non-coding RNAs, splicing and dendritic function. We analyzed SAFB1 expression in the post-mortem brain tissue of spinocerebellar ataxia (SCA), Huntington's disease (HD), Multiple sclerosis (MS), Parkinson's disease patients and controls. In SCA cases, the expression of SAFB1 in the nucleus was increased and there was abnormal and extensive expression in the cytoplasm where it co-localized with the markers of Purkinje cell injury. Significantly, no SAFB1 expression was found in the cerebellar neurons of the dentate nucleus in control or MS patients; however, in SCA patients, SAFB1 expression was increased significantly in both the nucleus and cytoplasm of dentate neurons. In HD, we found that SAFB1 expression was increased in the nucleus and cytoplasm of striatal neurons; however, there was no SAFB1 staining in the striatal neurons of controls. In PD substantia nigra, we did not see any changes in neuronal SAFB1 expression. iCLIP analysis found that SAFB1 crosslink sites within ATXN1 RNA were adjacent to the start and within the glutamine repeat sequence. Further investigation found increased binding of SAFB1 to pathogenic ATXN1-85Q mRNA. These novel data strongly suggest SAFB1 contributes to the etiology of SCA and Huntington's chorea and that it may be a pathological marker of polyglutamine repeat expansion diseases.


Subject(s)
Brain/metabolism , Huntington Disease/metabolism , Matrix Attachment Region Binding Proteins/metabolism , Neurons/metabolism , Nuclear Matrix-Associated Proteins/metabolism , Receptors, Estrogen/metabolism , Spinocerebellar Ataxias/metabolism , Aged , Aged, 80 and over , Brain/pathology , Cerebellum/metabolism , Cerebellum/pathology , Corpus Striatum/metabolism , Corpus Striatum/pathology , Female , Humans , Huntington Disease/pathology , Male , Middle Aged , Multiple Sclerosis/metabolism , Multiple Sclerosis/pathology , Neurons/pathology , Parkinson Disease/metabolism , Parkinson Disease/pathology , Purkinje Cells/metabolism , Purkinje Cells/pathology , Spinocerebellar Ataxias/pathology
SELECTION OF CITATIONS
SEARCH DETAIL