Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 89
Filter
1.
Nucleic Acids Res ; 52(8): 4483-4501, 2024 May 08.
Article in English | MEDLINE | ID: mdl-38587191

ABSTRACT

Messenger RNA precursors (pre-mRNA) generally undergo 3' end processing by cleavage and polyadenylation (CPA), which is specified by a polyadenylation site (PAS) and adjacent RNA sequences and regulated by a large variety of core and auxiliary CPA factors. To date, most of the human CPA factors have been discovered through biochemical and proteomic studies. However, genetic identification of the human CPA factors has been hampered by the lack of a reliable genome-wide screening method. We describe here a dual fluorescence readthrough reporter system with a PAS inserted between two fluorescent reporters. This system enables measurement of the efficiency of 3' end processing in living cells. Using this system in combination with a human genome-wide CRISPR/Cas9 library, we conducted a screen for CPA factors. The screens identified most components of the known core CPA complexes and other known CPA factors. The screens also identified CCNK/CDK12 as a potential core CPA factor, and RPRD1B as a CPA factor that binds RNA and regulates the release of RNA polymerase II at the 3' ends of genes. Thus, this dual fluorescence reporter coupled with CRISPR/Cas9 screens reliably identifies bona fide CPA factors and provides a platform for investigating the requirements for CPA in various contexts.


Subject(s)
CRISPR-Cas Systems , Genes, Reporter , RNA Precursors , mRNA Cleavage and Polyadenylation Factors , Humans , Cyclin-Dependent Kinases/metabolism , Cyclin-Dependent Kinases/genetics , Genome, Human , HEK293 Cells , mRNA Cleavage and Polyadenylation Factors/metabolism , mRNA Cleavage and Polyadenylation Factors/genetics , Polyadenylation , RNA Cleavage , RNA Polymerase II/metabolism , RNA Precursors/metabolism , RNA Precursors/genetics
2.
PLoS Pathog ; 19(7): e1011477, 2023 07.
Article in English | MEDLINE | ID: mdl-37410772

ABSTRACT

SUMO modifications regulate the function of many proteins and are important in controlling herpesvirus infections. We performed a site-specific proteomic analysis of SUMO1- and SUMO2-modified proteins in Epstein-Barr virus (EBV) latent and lytic infection to identify proteins that change in SUMO modification status in response to EBV reactivation. Major changes were identified in all three components of the TRIM24/TRIM28/TRIM33 complex, with TRIM24 being rapidly degraded and TRIM33 being phosphorylated and SUMOylated in response to EBV lytic infection. Further experiments revealed TRIM24 and TRIM33 repress expression of the EBV BZLF1 lytic switch gene, suppressing EBV reactivation. However, BZLF1 was shown to interact with TRIM24 and TRIM33, resulting in disruption of TRIM24/TRIM28/TRIM33 complexes, degradation of TRIM24 and modification followed by degradation of TRIM33. Therefore, we have identified TRIM24 and TRIM33 as cellular antiviral defence factors against EBV lytic infection and established the mechanism by which BZLF1 disables this defence.


Subject(s)
Epstein-Barr Virus Infections , Humans , Herpesvirus 4, Human/genetics , Trans-Activators/genetics , Trans-Activators/metabolism , Small Ubiquitin-Related Modifier Proteins/metabolism , Proteomics , Virus Activation , Virus Latency , Transcription Factors/metabolism , Carrier Proteins
3.
Bio Protoc ; 13(11): e4688, 2023 Jun 05.
Article in English | MEDLINE | ID: mdl-37323634

ABSTRACT

Individual nucleotide resolution UV cross-linking and immunoprecipitation followed by high-throughput sequencing (iCLIP-seq) is a powerful technique that is used to identify RNA-binding proteins' (RBP) binding sites on target RNAs and to characterize the molecular basis of posttranscriptional regulatory pathways. Several variants of CLIP have been developed to improve its efficiency and simplify the protocol [e.g., iCLIP2 and enhanced CLIP (eCLIP)]. We have recently reported that transcription factor SP1 functions in the regulation of alternative cleavage and polyadenylation through direct RNA binding. We utilized a modified iCLIP method to identify RNA-binding sites for SP1 and several of the cleavage and polyadenylation complex subunits, including CFIm25, CPSF7, CPSF100, CPSF2, and Fip1. Our revised protocol takes advantage of several features of the eCLIP procedure and also improves on certain steps of the original iCLIP method, including optimization of circularization of cDNA. Herein, we describe a step-by-step procedure for our revised iCLIP-seq protocol, that we designate as iCLIP-1.5, and provide alternative approaches for certain difficult-to-CLIP proteins. Key features Identification of RNA-binding sites of RNA-binding proteins (RBPs) at nucleotide resolution. iCLIP-seq provides precise positional and quantitative information on the RNA-binding sites of RBPs in living cells. iCLIP facilitates the identification of sequence motifs recognized by RBPs. Allows quantitative analysis of genome-wide changes in protein-RNA interactions. Revised iCLIP-1.5 protocol is more efficient and highly robust; it provides higher coverage even for low-input samples. Graphical overview.

4.
Epigenetics Chromatin ; 16(1): 10, 2023 04 07.
Article in English | MEDLINE | ID: mdl-37024975

ABSTRACT

BACKGROUND: Eukaryotic cells can rapidly adjust their transcriptional profile in response to molecular needs. Such dynamic regulation is, in part, achieved through epigenetic modifications and selective incorporation of histone variants into chromatin. H3.3 is the ancestral H3 variant with key roles in regulating chromatin states and transcription. Although H3.3 has been well studied in metazoans, information regarding the assembly of H3.3 onto chromatin and its possible role in transcription regulation remain poorly documented outside of Opisthokonts. RESULTS: We used the nuclear dimorphic ciliate protozoan, Tetrahymena thermophila, to investigate the dynamics of H3 variant function in evolutionarily divergent eukaryotes. Functional proteomics and immunofluorescence analyses of H3.1 and H3.3 revealed a highly conserved role for Nrp1 and Asf1 histone chaperones in nuclear influx of histones. Cac2, a putative subunit of H3.1 deposition complex CAF1, is not required for growth, whereas the expression of the putative ortholog of the H3.3-specific chaperone Hir1 is essential in Tetrahymena. Our results indicate that Cac2 and Hir1 have distinct localization patterns during different stages of the Tetrahymena life cycle and suggest that Cac2 might be dispensable for chromatin assembly. ChIP-seq experiments in growing Tetrahymena show H3.3 enrichment over the promoters, gene bodies, and transcription termination sites of highly transcribed genes. H3.3 knockout followed by RNA-seq reveals large-scale transcriptional alterations in functionally important genes. CONCLUSION: Our results provide an evolutionary perspective on H3.3's conserved role in maintaining the transcriptional landscape of cells and on the emergence of specialized chromatin assembly pathways.


Subject(s)
Gene Expression Regulation , Histones , Histones/genetics , Histones/metabolism , Chromatin/genetics , Chromatin/metabolism , Transcription, Genetic , Cell Nucleus/metabolism
5.
Nat Commun ; 13(1): 5453, 2022 09 16.
Article in English | MEDLINE | ID: mdl-36114190

ABSTRACT

Survival of motor neuron (SMN) functions in diverse biological pathways via recognition of symmetric dimethylarginine (Rme2s) on proteins by its Tudor domain, and deficiency of SMN leads to spinal muscular atrophy. Here we report a potent and selective antagonist with a 4-iminopyridine scaffold targeting the Tudor domain of SMN. Our structural and mutagenesis studies indicate that both the aromatic ring and imino groups of compound 1 contribute to its selective binding to SMN. Various on-target engagement assays support that compound 1 specifically recognizes SMN in a cellular context and prevents the interaction of SMN with the R1810me2s of RNA polymerase II subunit POLR2A, resulting in transcription termination and R-loop accumulation mimicking SMN depletion. Thus, in addition to the antisense, RNAi and CRISPR/Cas9 techniques, potent SMN antagonists could be used as an efficient tool to understand the biological functions of SMN.


Subject(s)
RNA Polymerase II , SMN Complex Proteins , Humans , Motor Neurons/metabolism , Muscular Atrophy, Spinal/metabolism , RNA Polymerase II/drug effects , RNA Polymerase II/metabolism , SMN Complex Proteins/antagonists & inhibitors , SMN Complex Proteins/drug effects , SMN Complex Proteins/metabolism
6.
Mol Cell ; 82(17): 3135-3150.e9, 2022 09 01.
Article in English | MEDLINE | ID: mdl-35914531

ABSTRACT

Alternative polyadenylation (APA) enhances gene regulatory potential by increasing the diversity of mRNA transcripts. 3' UTR shortening through APA correlates with enhanced cellular proliferation and is a widespread phenomenon in tumor cells. Here, we show that the ubiquitously expressed transcription factor Sp1 binds RNA in vivo and is a common repressor of distal poly(A) site usage. RNA sequencing identified 2,344 genes (36% of the total mapped mRNA transcripts) with lengthened 3' UTRs upon Sp1 depletion. Sp1 preferentially binds the 3' UTRs of such lengthened transcripts and inhibits cleavage at distal sites by interacting with the subunits of the core cleavage and polyadenylation (CPA) machinery. The 3' UTR lengths of Sp1 target genes in breast cancer patient RNA-seq data correlate with Sp1 expression levels, implicating Sp1-mediated APA regulation in modulating tumorigenic properties. Taken together, our findings provide insights into the mechanism for dynamic APA regulation by unraveling a previously unknown function of the DNA-binding transcription factor Sp1.


Subject(s)
Poly A , Polyadenylation , 3' Untranslated Regions , Humans , Poly A/metabolism , RNA, Messenger/metabolism , Sp1 Transcription Factor/genetics , Sp1 Transcription Factor/metabolism , Zinc/metabolism
7.
Mol Cell ; 82(16): 2982-2999.e14, 2022 08 18.
Article in English | MEDLINE | ID: mdl-35914530

ABSTRACT

Alternative splicing (AS) is a critical regulatory layer; yet, factors controlling functionally coordinated splicing programs during developmental transitions are poorly understood. Here, we employ a screening strategy to identify factors controlling dynamic splicing events important for mammalian neurogenesis. Among previously unknown regulators, Rbm38 acts widely to negatively control neural AS, in part through interactions mediated by the established repressor of splicing, Ptbp1. Puf60, a ubiquitous factor, is surprisingly found to promote neural splicing patterns. This activity requires a conserved, neural-differential exon that remodels Puf60 co-factor interactions. Ablation of this exon rewires distinct AS networks in embryonic stem cells and at different stages of mouse neurogenesis. Single-cell transcriptome analyses further reveal distinct roles for Rbm38 and Puf60 isoforms in establishing neuronal identity. Our results describe important roles for previously unknown regulators of neurogenesis and establish how an alternative exon in a widely expressed splicing factor orchestrates temporal control over cell differentiation.


Subject(s)
Neurogenesis , RNA Splicing , Alternative Splicing , Animals , Exons/genetics , Mammals , Mice , Neurogenesis/genetics , Neurons , RNA-Binding Proteins/genetics
8.
J Biol Chem ; 298(9): 102277, 2022 09.
Article in English | MEDLINE | ID: mdl-35863436

ABSTRACT

La-related protein 1 (LARP1) has been identified as a key translational inhibitor of terminal oligopyrimidine (TOP) mRNAs downstream of the nutrient sensing protein kinase complex, mTORC1. LARP1 exerts this inhibitory effect on TOP mRNA translation by binding to the mRNA cap and the adjacent 5'TOP motif, resulting in the displacement of the cap-binding protein eIF4E from TOP mRNAs. However, the involvement of additional signaling pathway in regulating LARP1-mediated inhibition of TOP mRNA translation is largely unexplored. In the present study, we identify a second nutrient sensing kinase GCN2 that converges on LARP1 to control TOP mRNA translation. Using chromatin-immunoprecipitation followed by massive parallel sequencing (ChIP-seq) analysis of activating transcription factor 4 (ATF4), an effector of GCN2 in nutrient stress conditions, in WT and GCN2 KO mouse embryonic fibroblasts, we determined that LARP1 is a GCN2-dependent transcriptional target of ATF4. Moreover, we identified GCN1, a GCN2 activator, participates in a complex with LARP1 on stalled ribosomes, suggesting a role for GCN1 in LARP1-mediated translation inhibition in response to ribosome stalling. Therefore, our data suggest that the GCN2 pathway controls LARP1 activity via two mechanisms: ATF4-dependent transcriptional induction of LARP1 mRNA and GCN1-mediated recruitment of LARP1 to stalled ribosomes.


Subject(s)
Amino Acids , Protein Biosynthesis , Protein Serine-Threonine Kinases , RNA 5' Terminal Oligopyrimidine Sequence , RNA, Messenger , RNA-Binding Proteins , Activating Transcription Factor 4/genetics , Activating Transcription Factor 4/metabolism , Amino Acids/metabolism , Animals , Cell Culture Techniques , Chromatin Immunoprecipitation , Eukaryotic Initiation Factor-4E/metabolism , Fibroblasts , Mechanistic Target of Rapamycin Complex 1/metabolism , Mice , Mice, Knockout , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA-Binding Proteins/genetics , RNA-Binding Proteins/metabolism
9.
Nat Chem Biol ; 18(1): 56-63, 2022 01.
Article in English | MEDLINE | ID: mdl-34782742

ABSTRACT

Nuclear receptor-binding SET domain-containing 2 (NSD2) is the primary enzyme responsible for the dimethylation of lysine 36 of histone 3 (H3K36), a mark associated with active gene transcription and intergenic DNA methylation. In addition to a methyltransferase domain, NSD2 harbors two proline-tryptophan-tryptophan-proline (PWWP) domains and five plant homeodomains (PHDs) believed to serve as chromatin reading modules. Here, we report a chemical probe targeting the N-terminal PWWP (PWWP1) domain of NSD2. UNC6934 occupies the canonical H3K36me2-binding pocket of PWWP1, antagonizes PWWP1 interaction with nucleosomal H3K36me2 and selectively engages endogenous NSD2 in cells. UNC6934 induces accumulation of endogenous NSD2 in the nucleolus, phenocopying the localization defects of NSD2 protein isoforms lacking PWWP1 that result from translocations prevalent in multiple myeloma (MM). Mutations of other NSD2 chromatin reader domains also increase NSD2 nucleolar localization and enhance the effect of UNC6934. This chemical probe and the accompanying negative control UNC7145 will be useful tools in defining NSD2 biology.


Subject(s)
Cell Nucleolus/metabolism , Histone-Lysine N-Methyltransferase/metabolism , Molecular Probes/chemistry , Protein Domains , Repressor Proteins/metabolism , Methylation , Multiple Myeloma/metabolism , Nucleosomes/metabolism
10.
iScience ; 25(1): 103562, 2022 Jan 21.
Article in English | MEDLINE | ID: mdl-34901782

ABSTRACT

Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) nucleocapsid (N) protein is essential for viral replication, making it a promising target for antiviral drug and vaccine development. SARS-CoV-2 infected patients exhibit an uncoordinated immune response; however, the underlying mechanistic details of this imbalance remain obscure. Here, starting from a functional proteomics workflow, we cataloged the protein-protein interactions of SARS-CoV-2 proteins, including an evolutionarily conserved specific interaction of N with the stress granule resident proteins G3BP1 and G3BP2. N localizes to stress granules and sequesters G3BPs away from their typical interaction partners, thus attenuating stress granule formation. We found that N binds directly to host mRNAs in cells, with a preference for 3' UTRs, and modulates target mRNA stability. We show that the N protein rewires the G3BP1 mRNA-binding profile and suppresses the physiological stress response of host cells, which may explain the imbalanced immune response observed in SARS-CoV-2 infected patients.

11.
Nucleic Acids Res ; 49(11): 6196-6212, 2021 06 21.
Article in English | MEDLINE | ID: mdl-34086947

ABSTRACT

Retinoblastoma-binding proteins 4 and 7 (RBBP4 and RBBP7) are two highly homologous human histone chaperones. They function in epigenetic regulation as subunits of multiple chromatin-related complexes and have been implicated in numerous cancers. Due to their overlapping functions, our understanding of RBBP4 and 7, particularly outside of Opisthokonts, has remained limited. Here, we report that in the ciliate protozoan Tetrahymena thermophila a single orthologue of human RBBP4 and 7 proteins, RebL1, physically interacts with histone H4 and functions in multiple epigenetic regulatory pathways. Functional proteomics identified conserved functional links for Tetrahymena RebL1 protein as well as human RBBP4 and 7. We found that putative subunits of multiple chromatin-related complexes including CAF1, Hat1, Rpd3, and MuvB, co-purified with RebL1 during Tetrahymena growth and conjugation. Iterative proteomics analyses revealed that the cell cycle regulatory MuvB-complex in Tetrahymena is composed of at least five subunits including evolutionarily conserved Lin54, Lin9 and RebL1 proteins. Genome-wide analyses indicated that RebL1 and Lin54 (Anqa1) bind within genic and intergenic regions. Moreover, Anqa1 targets primarily promoter regions suggesting a role for Tetrahymena MuvB in transcription regulation. RebL1 depletion inhibited cellular growth and reduced the expression levels of Anqa1 and Lin9. Consistent with observations in glioblastoma tumors, RebL1 depletion suppressed DNA repair protein Rad51 in Tetrahymena, thus underscoring the evolutionarily conserved functions of RBBP4/7 proteins. Our results suggest the essentiality of RebL1 functions in multiple epigenetic regulatory complexes in which it impacts transcription regulation and cellular viability.


Subject(s)
Histone Chaperones/metabolism , Protozoan Proteins/metabolism , Tetrahymena thermophila/metabolism , Amino Acid Sequence , Bacterial Proteins/metabolism , Biological Evolution , Conserved Sequence , DNA/metabolism , DNA-Binding Proteins/metabolism , Epigenesis, Genetic , Gene Expression , HEK293 Cells , Histone Chaperones/chemistry , Histone Chaperones/physiology , Histones/metabolism , Humans , Neoplasms/metabolism , Neoplasms/mortality , Oncogenes , Protozoan Proteins/chemistry , Protozoan Proteins/physiology , Retinoblastoma-Binding Protein 4/metabolism , Retinoblastoma-Binding Protein 7/metabolism , Tetrahymena thermophila/genetics , Tetrahymena thermophila/growth & development
12.
Nature ; 585(7824): 298-302, 2020 09.
Article in English | MEDLINE | ID: mdl-32669707

ABSTRACT

Proteins are manufactured by ribosomes-macromolecular complexes of protein and RNA molecules that are assembled within major nuclear compartments called nucleoli1,2. Existing models suggest that RNA polymerases I and III (Pol I and Pol III) are the only enzymes that directly mediate the expression of the ribosomal RNA (rRNA) components of ribosomes. Here we show, however, that RNA polymerase II (Pol II) inside human nucleoli operates near genes encoding rRNAs to drive their expression. Pol II, assisted by the neurodegeneration-associated enzyme senataxin, generates a shield comprising triplex nucleic acid structures known as R-loops at intergenic spacers flanking nucleolar rRNA genes. The shield prevents Pol I from producing sense intergenic noncoding RNAs (sincRNAs) that can disrupt nucleolar organization and rRNA expression. These disruptive sincRNAs can be unleashed by Pol II inhibition, senataxin loss, Ewing sarcoma or locus-associated R-loop repression through an experimental system involving the proteins RNaseH1, eGFP and dCas9 (which we refer to as 'red laser'). We reveal a nucleolar Pol-II-dependent mechanism that drives ribosome biogenesis, identify disease-associated disruption of nucleoli by noncoding RNAs, and establish locus-targeted R-loop modulation. Our findings revise theories of labour division between the major RNA polymerases, and identify nucleolar Pol II as a major factor in protein synthesis and nuclear organization, with potential implications for health and disease.


Subject(s)
Cell Nucleolus/enzymology , Cell Nucleolus/genetics , DNA, Ribosomal/genetics , RNA Polymerase II/metabolism , RNA, Untranslated/biosynthesis , RNA, Untranslated/genetics , Ribosomes/metabolism , CRISPR-Associated Protein 9/genetics , CRISPR-Associated Protein 9/metabolism , Cell Line, Tumor , Cell Nucleolus/physiology , DNA Helicases/metabolism , DNA, Intergenic/genetics , Humans , Multifunctional Enzymes/metabolism , Protein Biosynthesis , R-Loop Structures , RNA Helicases/metabolism , RNA Polymerase I/antagonists & inhibitors , RNA Polymerase I/metabolism , Ribonuclease H/metabolism , Ribosomes/chemistry , Ribosomes/genetics , Sarcoma, Ewing/genetics , Sarcoma, Ewing/pathology
13.
Cell Rep ; 31(8): 107693, 2020 05 26.
Article in English | MEDLINE | ID: mdl-32460013

ABSTRACT

The mammalian mRNA nuclear export process is thought to terminate at the cytoplasmic face of the nuclear pore complex through ribonucleoprotein remodeling. We conduct a stringent affinity-purification mass-spectrometry-based screen of the physical interactions of human RNA-binding E3 ubiquitin ligases. The resulting protein-interaction network reveals interactions between the RNA-binding E3 ubiquitin ligase MKRN2 and GLE1, a DEAD-box helicase activator implicated in mRNA export termination. We assess MKRN2 epistasis with GLE1 in a zebrafish model. Morpholino-mediated knockdown or CRISPR/Cas9-based knockout of MKRN2 partially rescue retinal developmental defects seen upon GLE1 depletion, consistent with a functional association between GLE1 and MKRN2. Using ribonomic approaches, we show that MKRN2 binds selectively to the 3' UTR of a diverse subset of mRNAs and that nuclear export of MKRN2-associated mRNAs is enhanced upon knockdown of MKRN2. Taken together, we suggest that MKRN2 interacts with GLE1 to selectively regulate mRNA nuclear export and retinal development.


Subject(s)
Mass Spectrometry/methods , RNA, Messenger/metabolism , RNA-Binding Proteins/metabolism , Retina/physiopathology , Ribonucleoproteins/metabolism , Zebrafish Proteins/metabolism , Animals , Humans , Zebrafish
14.
Nucleic Acids Res ; 48(6): 2897-2911, 2020 04 06.
Article in English | MEDLINE | ID: mdl-31960028

ABSTRACT

The Nrd1-Nab3-Sen1 (NNS) complex integrates molecular cues to direct termination of noncoding transcription in budding yeast. NNS is positively regulated by histone methylation as well as through Nrd1 binding to the initiating form of RNA PolII. These cues collaborate with Nrd1 and Nab3 binding to target RNA sequences in nascent transcripts through their RRM RNA recognition motifs. In this study, we identify nine lysine residues distributed amongst Nrd1, Nab3 and Sen1 that are methylated, suggesting novel molecular inputs for NNS regulation. We identify mono-methylation of one these residues (Nab3-K363me1) as being partly dependent on the H3K4 methyltransferase, Set1, a known regulator of NNS function. Moreover, the accumulation of Nab3-K363me1 is essentially abolished in strains lacking SET3, a SET domain containing protein that is positively regulated by H3K4 methylation. Nab3-K363 resides within its RRM and physically contacts target RNA. Mutation of Nab3-K363 to arginine (Nab3-K363R) decreases RNA binding of the Nab3 RRM in vitro and causes transcription termination defects and slow growth. These findings identify SET3 as a potential contextual regulator of Nab3 function through its role in methylation of Nab3-K363. Consistent with this hypothesis, we report that SET3 exhibits genetic activation of NAB3 that is observed in a sensitized context.


Subject(s)
Histone Deacetylases/metabolism , Histone-Lysine N-Methyltransferase/metabolism , Lysine/metabolism , Nuclear Proteins/chemistry , Nuclear Proteins/metabolism , RNA Recognition Motif , RNA-Binding Proteins/chemistry , RNA-Binding Proteins/metabolism , RNA/metabolism , Saccharomyces cerevisiae Proteins/chemistry , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae/metabolism , Methylation , Protein Binding , Structure-Activity Relationship
15.
Mol Cell ; 65(3): 539-553.e7, 2017 Feb 02.
Article in English | MEDLINE | ID: mdl-28157508

ABSTRACT

Networks of coordinated alternative splicing (AS) events play critical roles in development and disease. However, a comprehensive knowledge of the factors that regulate these networks is lacking. We describe a high-throughput system for systematically linking trans-acting factors to endogenous RNA regulatory events. Using this system, we identify hundreds of factors associated with diverse regulatory layers that positively or negatively control AS events linked to cell fate. Remarkably, more than one-third of the regulators are transcription factors. Further analyses of the zinc finger protein Zfp871 and BTB/POZ domain transcription factor Nacc1, which regulate neural and stem cell AS programs, respectively, reveal roles in controlling the expression of specific splicing regulators. Surprisingly, these proteins also appear to regulate target AS programs via binding RNA. Our results thus uncover a large "missing cache" of splicing regulators among annotated transcription factors, some of which dually regulate AS through direct and indirect mechanisms.


Subject(s)
Alternative Splicing , Gene Regulatory Networks , Sequence Analysis, RNA/methods , Transcription Factors/metabolism , Animals , Cell Line , Embryonic Stem Cells/cytology , Embryonic Stem Cells/metabolism , HEK293 Cells , Humans , Mice , Neurons/cytology , Neurons/metabolism , RNA, Messenger/genetics
16.
Genome Res ; 26(12): 1742-1752, 2016 12.
Article in English | MEDLINE | ID: mdl-27852650

ABSTRACT

C2H2 zinc finger proteins represent the largest and most enigmatic class of human transcription factors. Their C2H2-ZF arrays are highly variable, indicating that most will have unique DNA binding motifs. However, most of the binding motifs have not been directly determined. In addition, little is known about whether or how these proteins regulate transcription. Most of the ∼700 human C2H2-ZF proteins also contain at least one KRAB, SCAN, BTB, or SET domain, suggesting that they may have common interacting partners and/or effector functions. Here, we report a multifaceted functional analysis of 131 human C2H2-ZF proteins, encompassing DNA binding sites, interacting proteins, and transcriptional response to genetic perturbation. We confirm the expected diversity in DNA binding motifs and genomic binding sites, and provide motif models for 78 previously uncharacterized C2H2-ZF proteins, most of which are unique. Surprisingly, the diversity in protein-protein interactions is nearly as high as diversity in DNA binding motifs: Most C2H2-ZF proteins interact with a unique spectrum of co-activators and co-repressors. Thus, multiparameter diversification likely underlies the evolutionary success of this large class of human proteins.


Subject(s)
DNA/metabolism , Transcription Factors/chemistry , Transcription Factors/genetics , Transcription Factors/metabolism , Binding Sites , CYS2-HIS2 Zinc Fingers , Evolution, Molecular , Gene Expression Regulation , HEK293 Cells , Humans , Protein Binding , Protein Interaction Maps , Sequence Analysis, DNA , Sequence Analysis, RNA
17.
Stem Cell Reports ; 7(3): 454-470, 2016 09 13.
Article in English | MEDLINE | ID: mdl-27546533

ABSTRACT

Proliferating progenitor cells undergo changes in competence to give rise to post-mitotic progeny of specialized function. These cell-fate transitions typically involve dynamic regulation of gene expression by histone methyltransferase (HMT) complexes. However, the composition, roles, and regulation of these assemblies in regulating cell-fate decisions in vivo are poorly understood. Using unbiased affinity purification and mass spectrometry, we identified the uncharacterized C2H2-like zinc finger protein ZNF644 as a G9a/GLP-interacting protein and co-regulator of histone methylation. In zebrafish, functional characterization of ZNF644 orthologs, znf644a and znf644b, revealed complementary roles in regulating G9a/H3K9me2-mediated gene silencing during neurogenesis. The non-overlapping requirements for znf644a and znf644b during retinal differentiation demarcate critical aspects of retinal differentiation programs regulated by differential G9a-ZNF644 associations, such as transitioning proliferating progenitor cells toward differentiation. Collectively, our data point to ZNF644 as a critical co-regulator of G9a/H3K9me2-mediated gene silencing during neuronal differentiation.


Subject(s)
Gene Expression Regulation, Developmental , Histocompatibility Antigens/metabolism , Histone-Lysine N-Methyltransferase/metabolism , Neural Stem Cells/cytology , Neural Stem Cells/metabolism , Neurogenesis/genetics , Transcription Factors/metabolism , Animals , Binding Sites , Biomarkers , Cell Differentiation , Cell Proliferation , Cell Survival/genetics , Gene Silencing , Histocompatibility Antigens/genetics , Histone-Lysine N-Methyltransferase/genetics , Histones/metabolism , Humans , Methylation , Neurons/cytology , Neurons/metabolism , Phenotype , Protein Binding , Protein Interaction Mapping , Protein Interaction Maps , Retina/metabolism , Transcription Factors/genetics , Zebrafish
18.
Proc Natl Acad Sci U S A ; 113(13): 3527-32, 2016 Mar 29.
Article in English | MEDLINE | ID: mdl-26976582

ABSTRACT

Skp1-Cul1-F-box (SCF) E3 ligases play key roles in multiple cellular processes through ubiquitination and subsequent degradation of substrate proteins. Although Skp1 and Cul1 are invariant components of all SCF complexes, the 69 different human F-box proteins are variable substrate binding modules that determine specificity. SCF E3 ligases are activated in many cancers and inhibitors could have therapeutic potential. Here, we used phage display to develop specific ubiquitin-based inhibitors against two F-box proteins, Fbw7 and Fbw11. Unexpectedly, the ubiquitin variants bind at the interface of Skp1 and F-box proteins and inhibit ligase activity by preventing Cul1 binding to the same surface. Using structure-based design and phage display, we modified the initial inhibitors to generate broad-spectrum inhibitors that targeted many SCF ligases, or conversely, a highly specific inhibitor that discriminated between even the close homologs Fbw11 and Fbw1. We propose that most F-box proteins can be targeted by this approach for basic research and for potential cancer therapies.


Subject(s)
Cullin Proteins/metabolism , SKP Cullin F-Box Protein Ligases/antagonists & inhibitors , Ubiquitins/pharmacology , Amino Acid Sequence , Binding Sites , Cell Cycle Proteins/antagonists & inhibitors , Cell Cycle Proteins/chemistry , Cell Cycle Proteins/genetics , Cullin Proteins/chemistry , Drug Design , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacology , F-Box Proteins/antagonists & inhibitors , F-Box Proteins/chemistry , F-Box Proteins/genetics , F-Box-WD Repeat-Containing Protein 7 , Genetic Variation , Humans , Models, Molecular , Molecular Sequence Data , Peptide Library , Protein Engineering , Protein Interaction Domains and Motifs , SKP Cullin F-Box Protein Ligases/chemistry , SKP Cullin F-Box Protein Ligases/genetics , Sequence Homology, Amino Acid , Ubiquitin-Protein Ligases/antagonists & inhibitors , Ubiquitin-Protein Ligases/chemistry , Ubiquitin-Protein Ligases/genetics , Ubiquitins/chemistry , Ubiquitins/genetics , beta-Transducin Repeat-Containing Proteins/antagonists & inhibitors , beta-Transducin Repeat-Containing Proteins/chemistry , beta-Transducin Repeat-Containing Proteins/genetics
19.
Nature ; 529(7584): 48-53, 2016 Jan 07.
Article in English | MEDLINE | ID: mdl-26700805

ABSTRACT

The carboxy-terminal domain (CTD) of the RNA polymerase II (RNAP II) subunit POLR2A is a platform for modifications specifying the recruitment of factors that regulate transcription, mRNA processing, and chromatin remodelling. Here we show that a CTD arginine residue (R1810 in human) that is conserved across vertebrates is symmetrically dimethylated (me2s). This R1810me2s modification requires protein arginine methyltransferase 5 (PRMT5) and recruits the Tudor domain of the survival of motor neuron (SMN, also known as GEMIN1) protein, which is mutated in spinal muscular atrophy. SMN interacts with senataxin, which is sometimes mutated in ataxia oculomotor apraxia type 2 and amyotrophic lateral sclerosis. Because POLR2A R1810me2s and SMN, like senataxin, are required for resolving RNA-DNA hybrids created by RNA polymerase II that form R-loops in transcription termination regions, we propose that R1810me2s, SMN, and senataxin are components of an R-loop resolution pathway. Defects in this pathway can influence transcription termination and may contribute to neurodegenerative disorders.


Subject(s)
Arginine/metabolism , RNA Polymerase II/chemistry , RNA Polymerase II/metabolism , Survival of Motor Neuron 1 Protein/metabolism , Transcription Termination, Genetic , Cell Line , DNA Damage , DNA Helicases , Humans , Methylation , Multifunctional Enzymes , Neurodegenerative Diseases/genetics , Protein Binding , Protein Structure, Tertiary , Protein-Arginine N-Methyltransferases/genetics , Protein-Arginine N-Methyltransferases/metabolism , RNA Helicases/genetics , RNA Helicases/metabolism , Survival of Motor Neuron 1 Protein/genetics , Transcription Elongation, Genetic
20.
PLoS One ; 10(10): e0139695, 2015.
Article in English | MEDLINE | ID: mdl-26437229

ABSTRACT

We developed and optimized a high-throughput project workflow to generate renewable recombinant antibodies to human proteins involved in epigenetic signalling. Three different strategies to produce phage display compatible protein antigens in bacterial systems were compared, and we found that in vivo biotinylation through the use of an Avi tag was the most productive method. Phage display selections were performed on 265 in vivo biotinylated antigen domains. High-affinity Fabs (<20nM) were obtained for 196. We constructed and optimized a new expression vector to produce in vivo biotinylated Fabs in E. coli. This increased average yields up to 10-fold, with an average yield of 4 mg/L. For 118 antigens, we identified Fabs that could immunoprecipitate their full-length endogenous targets from mammalian cell lysates. One Fab for each antigen was converted to a recombinant IgG and produced in mammalian cells, with an average yield of 15 mg/L. In summary, we have optimized each step of the pipeline to produce recombinant antibodies, significantly increasing both efficiency and yield, and also showed that these Fabs and IgGs can be generally useful for chromatin immunoprecipitation (ChIP) protocols.


Subject(s)
Antibody Formation/physiology , Antigens/immunology , Immunoglobulin Fab Fragments/immunology , Recombinant Proteins/immunology , Cloning, Molecular , Humans , Peptide Library
SELECTION OF CITATIONS
SEARCH DETAIL
...