Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
Add more filters










Publication year range
1.
Mol Cell ; 84(8): 1475-1495.e18, 2024 Apr 18.
Article in English | MEDLINE | ID: mdl-38521065

ABSTRACT

Transcription and splicing of pre-messenger RNA are closely coordinated, but how this functional coupling is disrupted in human diseases remains unexplored. Using isogenic cell lines, patient samples, and a mutant mouse model, we investigated how cancer-associated mutations in SF3B1 alter transcription. We found that these mutations reduce the elongation rate of RNA polymerase II (RNAPII) along gene bodies and its density at promoters. The elongation defect results from disrupted pre-spliceosome assembly due to impaired protein-protein interactions of mutant SF3B1. The decreased promoter-proximal RNAPII density reduces both chromatin accessibility and H3K4me3 marks at promoters. Through an unbiased screen, we identified epigenetic factors in the Sin3/HDAC/H3K4me pathway, which, when modulated, reverse both transcription and chromatin changes. Our findings reveal how splicing factor mutant states behave functionally as epigenetic disorders through impaired transcription-related changes to the chromatin landscape. We also present a rationale for targeting the Sin3/HDAC complex as a therapeutic strategy.


Subject(s)
Chromatin , Neoplasms , Animals , Humans , Mice , Chromatin/genetics , Mutation , Phosphoproteins/genetics , Phosphoproteins/metabolism , RNA Polymerase II/genetics , RNA Polymerase II/metabolism , RNA Splicing/genetics , RNA Splicing Factors/genetics , RNA Splicing Factors/metabolism
2.
bioRxiv ; 2023 Feb 26.
Article in English | MEDLINE | ID: mdl-36891287

ABSTRACT

Transcription and splicing of pre-messenger RNA are closely coordinated, but how this functional coupling is disrupted in human disease remains unexplored. Here, we investigated the impact of non-synonymous mutations in SF3B1 and U2AF1, two commonly mutated splicing factors in cancer, on transcription. We find that the mutations impair RNA Polymerase II (RNAPII) transcription elongation along gene bodies leading to transcription-replication conflicts, replication stress and altered chromatin organization. This elongation defect is linked to disrupted pre-spliceosome assembly due to impaired association of HTATSF1 with mutant SF3B1. Through an unbiased screen, we identified epigenetic factors in the Sin3/HDAC complex, which, when modulated, normalize transcription defects and their downstream effects. Our findings shed light on the mechanisms by which oncogenic mutant spliceosomes impact chromatin organization through their effects on RNAPII transcription elongation and present a rationale for targeting the Sin3/HDAC complex as a potential therapeutic strategy. HIGHLIGHTS: Oncogenic mutations of SF3B1 and U2AF1 cause a gene-body RNAPII elongation defectRNAPII transcription elongation defect leads to transcription replication conflicts, DNA damage response, and changes to chromatin organization and H3K4me3 marksThe transcription elongation defect is linked to disruption of the early spliceosome formation through impaired interaction of HTATSF1 with mutant SF3B1.Changes to chromatin organization reveal potential therapeutic strategies by targeting the Sin3/HDAC pathway.

3.
NAR Cancer ; 4(2): zcac015, 2022 Jun.
Article in English | MEDLINE | ID: mdl-35528200

ABSTRACT

Musashi 2 (MSI2) is an RNA binding protein (RBP) that regulates asymmetric cell division and cell fate decisions in normal and cancer stem cells. MSI2 appears to repress translation by binding to 3' untranslated regions (3'UTRs) of mRNA, but the identity of functional targets remains unknown. Here, we used individual nucleotide resolution cross-linking and immunoprecipitation (iCLIP) to identify direct RNA binding partners of MSI2 and integrated these data with polysome profiling to obtain insights into MSI2 function. iCLIP revealed specific MSI2 binding to thousands of mRNAs largely in 3'UTRs, but translational differences were restricted to a small fraction of these transcripts, indicating that MSI2 regulation is not triggered by simple binding. Instead, the functional targets identified here were bound at higher density and contain more 'UAG' motifs compared to targets bound nonproductively. To further distinguish direct and indirect targets, MSI2 was acutely depleted. Surprisingly, only 50 transcripts were found to undergo translational induction on acute loss. Using complementary approaches, we determined eukaryotic translation initiation factor 3A (EIF3A) to be an immediate, direct target. We propose that MSI2 downregulation of EIF3A amplifies these effects on translation. Our results also underscore the challenges in defining functional targets of RBPs since mere binding does not imply a discernible functional interaction.

4.
Mol Cell ; 82(6): 1107-1122.e7, 2022 03 17.
Article in English | MEDLINE | ID: mdl-35303483

ABSTRACT

Splicing factor mutations are common among cancers, recently emerging as drivers of myeloid malignancies. U2AF1 carries hotspot mutations in its RNA-binding motifs; however, how they affect splicing and promote cancer remain unclear. The U2AF1/U2AF2 heterodimer is critical for 3' splice site (3'SS) definition. To specifically unmask changes in U2AF1 function in vivo, we developed a crosslinking and immunoprecipitation procedure that detects contacts between U2AF1 and the 3'SS AG at single-nucleotide resolution. Our data reveal that the U2AF1 S34F and Q157R mutants establish new 3'SS contacts at -3 and +1 nucleotides, respectively. These effects compromise U2AF2-RNA interactions, resulting predominantly in intron retention and exon exclusion. Integrating RNA binding, splicing, and turnover data, we predicted that U2AF1 mutations directly affect stress granule components, which was corroborated by single-cell RNA-seq. Remarkably, U2AF1-mutant cell lines and patient-derived MDS/AML blasts displayed a heightened stress granule response, pointing to a novel role for biomolecular condensates in adaptive oncogenic strategies.


Subject(s)
Leukemia, Myeloid, Acute , Myelodysplastic Syndromes , Splicing Factor U2AF , Stress Granules , Humans , Leukemia, Myeloid, Acute/genetics , Mutation , Myelodysplastic Syndromes/genetics , RNA Splice Sites , RNA Splicing/genetics , RNA-Binding Proteins/genetics , Splicing Factor U2AF/genetics , Splicing Factor U2AF/metabolism , Stress Granules/metabolism
5.
G3 (Bethesda) ; 11(2)2021 02 09.
Article in English | MEDLINE | ID: mdl-33609367

ABSTRACT

Small noncoding RNA pathways have been implicated in diverse mechanisms of gene regulation. In Drosophila ovaries, Piwi binds to Piwi-interacting RNAs (piRNAs) of mostly 24-28 nucleotides (nt) and plays an important role in germline stem cell maintenance, transposon repression, and epigenetic regulation. To understand the mechanism underlying these functions, we report the application of the DamID-seq method to identify genome-wide binding sites of Piwi in Drosophila ovaries. Piwi localizes to at least 4535 euchromatic regions that are enriched with piRNA target sites. Surprisingly, the density of Piwi binding to euchromatin is much higher than in heterochromatin. Disrupting the piRNA binding of Piwi results in an overall change of the genomic binding profile, which indicates the role of piRNAs in directing Piwi to specific genomic sites. Most Piwi binding sites were either within or in the vicinity of protein-coding genes, particularly enriched near the transcriptional start and termination sites. The methylation signal near the transcriptional termination sites is significantly reduced when Piwi was mutated to become defective in piRNA binding. These observations indicate that Piwi might directly regulate the expression of many protein-coding genes, especially through regulating the 3' ends of targeted transcripts.


Subject(s)
Drosophila Proteins , Drosophila , Animals , Argonaute Proteins/metabolism , DNA Transposable Elements , Drosophila/genetics , Drosophila Proteins/genetics , Drosophila melanogaster/genetics , Epigenesis, Genetic , Female , Ovary/metabolism , RNA, Small Interfering
6.
Genetics ; 211(1): 201-217, 2019 01.
Article in English | MEDLINE | ID: mdl-30442760

ABSTRACT

Heterochromatin protein 1a (HP1a) is a highly conserved and versatile epigenetic factor that can both silence and activate transcription. However, the function of HP1a in development has been underinvestigated. Here, we report the role of maternal HP1a in producing maternal transcripts that drive early Drosophila embryogenesis. Maternal HP1a upregulates genes involved in translation, mRNA splicing, and cell division, but downregulates genes involved in neurogenesis, organogenesis, and germline development, which all occur later in development. Our study reveals the earliest contribution of HP1a during oogenesis in regulating the production of maternal transcripts that drive early Drosophila embryogenesis.


Subject(s)
Chromosomal Proteins, Non-Histone/genetics , Gene Expression Regulation, Developmental , Maternal Inheritance , Animals , Chromobox Protein Homolog 5 , Chromosomal Proteins, Non-Histone/metabolism , Drosophila , Embryonic Development , Epigenesis, Genetic , Female , Male , Oogenesis , RNA, Messenger/genetics , RNA, Messenger/metabolism
7.
Cell Stem Cell ; 22(2): 235-251.e9, 2018 02 01.
Article in English | MEDLINE | ID: mdl-29337181

ABSTRACT

Polycomb group proteins regulate self-renewal and differentiation in many stem cell systems. When assembled into two canonical complexes, PRC1 and PRC2, they sequentially deposit H3K27me3 and H2AK119ub histone marks and establish repressive chromatin, referred to as Polycomb domains. Non-canonical PRC1 complexes retain RING1/RNF2 E3-ubiquitin ligases but have unique sets of accessory subunits. How these non-canonical complexes recognize and regulate their gene targets remains poorly understood. Here, we show that the BCL6 co-repressor (BCOR), a member of the PRC1.1 complex, is critical for maintaining primed pluripotency in human embryonic stem cells (ESCs). BCOR depletion leads to the erosion of Polycomb domains at key developmental loci and the initiation of differentiation along endoderm and mesoderm lineages. The C terminus of BCOR regulates the assembly and targeting of the PRC1.1 complex, while the N terminus contributes to BCOR-PRC1.1 repressor function. Our findings advance understanding of Polycomb targeting and repression in ESCs and could apply broadly across developmental systems.


Subject(s)
Cell Differentiation , Human Embryonic Stem Cells/cytology , Human Embryonic Stem Cells/metabolism , Multiprotein Complexes/metabolism , Polycomb Repressive Complex 1/metabolism , Proto-Oncogene Proteins/metabolism , Repressor Proteins/metabolism , Chromatin/metabolism , F-Box Proteins/metabolism , Histones/metabolism , Humans , Jumonji Domain-Containing Histone Demethylases/metabolism , Lysine/metabolism , Methylation , Polycomb Repressive Complex 2/metabolism , Promoter Regions, Genetic , Protein Domains , Proto-Oncogene Proteins/chemistry , Repressor Proteins/chemistry
8.
Genes Dev ; 31(13): 1354-1369, 2017 07 01.
Article in English | MEDLINE | ID: mdl-28794184

ABSTRACT

Despite extensive studies on mammalian neurogenesis, its post-transcriptional regulation remains under-explored. Here we report that neural-specific inactivation of two murine post-transcriptional regulators, Pumilio 1 (Pum1) and Pum2, severely reduced the number of neural stem cells (NSCs) in the postnatal dentate gyrus (DG), drastically increased perinatal apoptosis, altered DG cell composition, and impaired learning and memory. Consistently, the mutant DG neurospheres generated fewer NSCs with defects in proliferation, survival, and differentiation, supporting a major role of Pum1 and Pum2 in hippocampal neurogenesis and function. Cross-linking immunoprecipitation revealed that Pum1 and Pum2 bind to thousands of mRNAs, with at least 694 common targets in multiple neurogenic pathways. Depleting Pum1 and/or Pum2 did not change the abundance of most target mRNAs but up-regulated their proteins, indicating that Pum1 and Pum2 regulate the translation of their target mRNAs. Moreover, Pum1 and Pum2 display RNA-dependent interaction with fragile X mental retardation protein (FMRP) and bind to one another's mRNA. This indicates that Pum proteins might form collaborative networks with FMRP and possibly other post-transcriptional regulators to regulate neurogenesis.


Subject(s)
Dentate Gyrus/cytology , Neurogenesis/genetics , RNA-Binding Proteins/genetics , RNA-Binding Proteins/metabolism , Animals , Cell Differentiation/genetics , Cytoplasm/metabolism , Female , Fragile X Mental Retardation Protein/metabolism , Gene Expression Regulation, Developmental , Gene Knockout Techniques , Gene Silencing , Learning Disabilities/genetics , Male , Memory Disorders/genetics , Mice , Neural Stem Cells/cytology , Neural Stem Cells/metabolism , Neurons/cytology , Neurons/metabolism , RNA, Messenger/metabolism , Stem Cells/cytology , Stem Cells/metabolism
9.
EMBO J ; 34(14): 1925-41, 2015 Jul 14.
Article in English | MEDLINE | ID: mdl-26069323

ABSTRACT

The assembly of spliceosomal U snRNPs depends on the coordinated action of PRMT5 and SMN complexes in vivo. These trans-acting factors enable the faithful delivery of seven Sm proteins onto snRNA and the formation of the common core of snRNPs. To gain mechanistic insight into their mode of action, we reconstituted the assembly machinery from recombinant sources. We uncover a stepwise and ordered formation of distinct Sm protein complexes on the PRMT5 complex, which is facilitated by the assembly chaperone pICln. Upon completion, the formed pICln-Sm units are displaced by new pICln-Sm protein substrates and transferred onto the SMN complex. The latter acts as a Brownian machine that couples spontaneous conformational changes driven by thermal energy to prevent mis-assembly and to ensure the transfer of Sm proteins to cognate RNA. Investigation of mutant SMN complexes provided insight into the contribution of individual proteins to these activities. The biochemical reconstitution presented here provides a basis for a detailed molecular dissection of the U snRNP assembly reaction.


Subject(s)
Ribonucleoproteins, Small Nuclear/metabolism , SMN Complex Proteins/metabolism , Animals , DEAD Box Protein 20/genetics , DEAD Box Protein 20/metabolism , Humans , Minor Histocompatibility Antigens , Muscular Atrophy, Spinal/genetics , Mutation , Protein-Arginine N-Methyltransferases/genetics , Protein-Arginine N-Methyltransferases/metabolism , RNA, Small Nuclear/metabolism , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Ribonucleoproteins, Small Nuclear/genetics , SMN Complex Proteins/genetics
10.
Dev Cell ; 32(6): 772-4, 2015 Mar 23.
Article in English | MEDLINE | ID: mdl-25805139

ABSTRACT

Drosophila Piwi was reported by Huang et al. (2013) to be guided by piRNAs to piRNA-complementary sites in the genome, which then recruits heterochromatin protein 1a and histone methyltransferase Su(Var)3-9 to the sites. Among additional findings, Huang et al. (2013) also reported Piwi binding sites in the genome and the reduction of RNA polymerase II in euchromatin but its increase in pericentric regions in piwi mutants. Marinov et al. (2015) disputed the validity of the Huang et al. bioinformatic pipeline that led to the last two claims. Here we report our independent reanalysis of the data using current bioinformatic methods. Our reanalysis agrees with Marinov et al. (2015) that Piwi's genomic targets still remain to be identified but confirms the Huang et al. claim that Piwi influences RNA polymerase II distribution in the genome. This Matters Arising Response addresses the Marinov et al. (2015) Matters Arising, published concurrently in this issue of Developmental Cell.


Subject(s)
Argonaute Proteins/genetics , Chromosomal Proteins, Non-Histone/genetics , DNA-Binding Proteins/genetics , Drosophila Proteins/genetics , Methyltransferases/genetics , RNA Polymerase II/genetics , Animals , Base Sequence , Binding Sites/genetics , Chromatin Immunoprecipitation , Chromobox Protein Homolog 5 , Drosophila melanogaster , Genome , High-Throughput Nucleotide Sequencing , Histone Methyltransferases , Histone-Lysine N-Methyltransferase/genetics , RNA Interference , RNA, Small Interfering/genetics , Sequence Analysis, DNA
11.
ISME J ; 9(6): 1333-51, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25500507

ABSTRACT

Archaea and Bacteria constitute a majority of life systems on Earth but have long been considered inferior to Eukarya in terms of solute tolerance. Whereas the most halophilic prokaryotes are known for an ability to multiply at saturated NaCl (water activity (a(w)) 0.755) some xerophilic fungi can germinate, usually at high-sugar concentrations, at values as low as 0.650-0.605 a(w). Here, we present evidence that halophilic prokayotes can grow down to water activities of <0.755 for Halanaerobium lacusrosei (0.748), Halobacterium strain 004.1 (0.728), Halobacterium sp. NRC-1 and Halococcus morrhuae (0.717), Haloquadratum walsbyi (0.709), Halococcus salifodinae (0.693), Halobacterium noricense (0.687), Natrinema pallidum (0.681) and haloarchaeal strains GN-2 and GN-5 (0.635 a(w)). Furthermore, extrapolation of growth curves (prone to giving conservative estimates) indicated theoretical minima down to 0.611 aw for extreme, obligately halophilic Archaea and Bacteria. These were compared with minima for the most solute-tolerant Bacteria in high-sugar (or other non-saline) media (Mycobacterium spp., Tetragenococcus halophilus, Saccharibacter floricola, Staphylococcus aureus and so on) and eukaryotic microbes in saline (Wallemia spp., Basipetospora halophila, Dunaliella spp. and so on) and high-sugar substrates (for example, Xeromyces bisporus, Zygosaccharomyces rouxii, Aspergillus and Eurotium spp.). We also manipulated the balance of chaotropic and kosmotropic stressors for the extreme, xerophilic fungi Aspergillus penicilloides and X. bisporus and, via this approach, their established water-activity limits for mycelial growth (∼0.65) were reduced to 0.640. Furthermore, extrapolations indicated theoretical limits of 0.632 and 0.636 a(w) for A. penicilloides and X. bisporus, respectively. Collectively, these findings suggest that there is a common water-activity limit that is determined by physicochemical constraints for the three domains of life.


Subject(s)
Aspergillus/metabolism , Bacteria/metabolism , Halobacterium/metabolism , Archaea/metabolism , Artifacts , Ascomycota/metabolism , Carbohydrates/chemistry , Fungi/metabolism , Hydrogen-Ion Concentration , Sodium Chloride/chemistry , Staphylococcus aureus/metabolism , Temperature , Water/physiology , Water Microbiology
12.
PLoS One ; 7(8): e42106, 2012.
Article in English | MEDLINE | ID: mdl-22879910

ABSTRACT

The Epstein-Barr Virus (EBV) -encoded EBNA2 protein, which is essential for the in vitro transformation of B-lymphocytes, interferes with cellular processes by binding to proteins via conserved sequence motifs. Its Arginine-Glycine (RG) repeat element contains either symmetrically or asymmetrically di-methylated arginine residues (SDMA and ADMA, respectively). EBNA2 binds via its SDMA-modified RG-repeat to the survival motor neurons protein (SMN) and via the ADMA-RG-repeat to the NP9 protein of the human endogenous retrovirus K (HERV-K (HML-2) Type 1). The hypothesis of this work was that the methylated RG-repeat mimics an epitope shared with cellular proteins that is used for interaction with target structures. With monoclonal antibodies against the modified RG-repeat, we indeed identified cellular homologues that apparently have the same surface structure as methylated EBNA2. With the SDMA-specific antibodies, we precipitated the Sm protein D3 (SmD3) which, like EBNA2, binds via its SDMA-modified RG-repeat to SMN. With the ADMA-specific antibodies, we precipitated the heterogeneous ribonucleoprotein K (hnRNP K). Specific binding of the ADMA- antibody to hnRNP K was demonstrated using E. coli expressed/ADMA-methylated hnRNP K. In addition, we show that EBNA2 and hnRNP K form a complex in EBV- infected B-cells. Finally, hnRNP K, when co-expressed with EBNA2, strongly enhances viral latent membrane protein 2A (LMP2A) expression by an unknown mechanism as we did not detect a direct association of hnRNP K with DNA-bound EBNA2 in gel shift experiments. Our data support the notion that the methylated surface of EBNA2 mimics the surface structure of cellular proteins to interfere with or co-opt their functional properties.


Subject(s)
Epstein-Barr Virus Nuclear Antigens/metabolism , Heterogeneous-Nuclear Ribonucleoprotein K/metabolism , Viral Matrix Proteins/metabolism , Viral Proteins/metabolism , Antibodies, Monoclonal/immunology , Antibody Specificity/immunology , Arginine/analogs & derivatives , Arginine/metabolism , Cell Line , DNA/metabolism , Epstein-Barr Virus Infections/metabolism , Epstein-Barr Virus Nuclear Antigens/chemistry , Epstein-Barr Virus Nuclear Antigens/immunology , Gene Expression Regulation, Viral , Herpesvirus 4, Human/genetics , Herpesvirus 4, Human/physiology , Humans , Immunoprecipitation , Methylation , Molecular Sequence Data , Multiprotein Complexes/metabolism , Mutant Proteins/metabolism , Promoter Regions, Genetic/genetics , Protein Binding , Protein Transport , Repetitive Sequences, Amino Acid , Transfection , Viral Matrix Proteins/genetics , Viral Proteins/chemistry , Viral Proteins/immunology , snRNP Core Proteins/metabolism
13.
RNA ; 16(8): 1646-59, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20562214

ABSTRACT

Mammalian cleavage factor I (CF I(m)) is composed of two polypeptides of 25 kDa and either a 59 or 68 kDa subunit (CF I(m)25, CF I(m)59, CF I(m)68). It is part of the cleavage and polyadenylation complex responsible for processing the 3' ends of messenger RNA precursors. To investigate post-translational modifications in factors of the 3' processing complex, we systematically searched for enzymes that modify arginines by the addition of methyl groups. Protein arginine methyltransferases (PRMTs) are such enzymes that transfer methyl groups from S-adenosyl methionine to arginine residues within polypeptide chains resulting in mono- or dimethylated arginines. We found that CF I(m)68 and the nuclear poly(A) binding protein 1 (PABPN1) were methylated by HeLa cell extracts in vitro. By fractionation of these extracts followed by mass spectral analysis, we could demonstrate that the catalytic subunit PRMT5, together with its cofactor WD45, could symmetrically dimethylate CF I(m)68, whereas pICln, the third polypeptide of the complex, was stimulatory. As sites of methylation in CF I(m)68 we could exclusively identify arginines in a GGRGRGRF or "GAR" motif that is conserved in vertebrates. Further in vitro assays revealed a second methyltransferase, PRMT1, which modifies CF I(m)68 by asymmetric dimethylation of the GAR motif and also weakly methylates the C-termini of both CF I(m)59 and CF I(m)68. The results suggest that native-as compared with recombinant-protein substrates may contain additional determinants for methylation by specific PRMTs. A possible involvement of CF I(m) methylation in the context of RNA export is discussed.


Subject(s)
Arginine/metabolism , RNA Precursors/metabolism , Animals , Arginine/genetics , Fibrinogen/genetics , Fibrinogen/metabolism , Gene Expression , Mammals/genetics , Mammals/metabolism , Methylation , Protein Processing, Post-Translational , Protein-Arginine N-Methyltransferases , RNA Precursors/genetics
14.
Cell ; 135(3): 497-509, 2008 Oct 31.
Article in English | MEDLINE | ID: mdl-18984161

ABSTRACT

Spliceosomal small nuclear ribonucleoproteins (snRNPs) are essential components of the nuclear pre-mRNA processing machinery. A hallmark of these particles is a ring-shaped core domain generated by the binding of Sm proteins onto snRNA. PRMT5 and SMN complexes mediate the formation of the core domain in vivo. Here, we have elucidated the mechanism of this reaction by both biochemical and structural studies. We show that pICln, a component of the PRMT5 complex, induces the formation of an otherwise unstable higher-order Sm protein unit. In this state, the Sm proteins are kinetically trapped, preventing their association with snRNA. The SMN complex subsequently binds to these Sm protein units, dissociates pICln, and catalyzes ring closure on snRNA. Our data identify pICln as an assembly chaperone and the SMN complex as a catalyst of spliceosomal snRNP formation. The mode of action of this combined chaperone/catalyst system is reminiscent of the mechanism employed by DNA clamp loaders.


Subject(s)
Protein Methyltransferases/chemistry , Ribonucleoproteins, Small Nuclear/metabolism , HeLa Cells , Humans , Models, Biological , Molecular Chaperones/metabolism , Nerve Tissue Proteins/metabolism , Protein Methyltransferases/metabolism , Protein-Arginine N-Methyltransferases , RNA/metabolism , RNA-Binding Proteins/metabolism , Ribonucleoproteins, Small Nuclear/chemistry , Survival of Motor Neuron 1 Protein/metabolism
15.
FEBS Lett ; 582(14): 1997-2003, 2008 Jun 18.
Article in English | MEDLINE | ID: mdl-18348870

ABSTRACT

The assembly of the Sm-class of uridine-rich small nuclear ribonucleoproteins (U snRNPs), albeit spontaneous in vitro, has recently been shown to be dependent on the aid of a large number of assisting factors in vivo. These factors are organized in two interacting units termed survival motor neuron (SMN)- and protein arginine methyltransferase 5 (PRMT5)-complexes, respectively. While the PRMT5-complex acts early in the assembly pathway by activating common proteins of U snRNPs, the SMN-complex functions to join proteins and RNA in a highly ordered, apparently regulated manner. Here, we summarize recent progress in the understanding of this process and discuss the influence exerted by the aforementioned trans-acting factors.


Subject(s)
Cell Nucleus/metabolism , Ribonucleoproteins, Small Nuclear/metabolism , Spliceosomes/metabolism , Humans , Protein Methyltransferases/metabolism , Protein-Arginine N-Methyltransferases
16.
J Biol Chem ; 282(8): 5825-33, 2007 Feb 23.
Article in English | MEDLINE | ID: mdl-17178713

ABSTRACT

Assembly of the Sm-class of U-rich small nuclear ribonucleoprotein particles (U snRNPs) is a process facilitated by the macromolecular survival of motor neuron (SMN) complex. This entity promotes the binding of a set of factors, termed LSm/Sm proteins, onto snRNA to form the core structure of these particles. Nine factors, including the SMN protein, the product of the spinal muscular atrophy (SMA) disease gene, Gemins 2-8 and unrip have been identified as the major components of the SMN complex. So far, however, only little is known about the architecture of this complex and the contribution of individual components to its function. Here, we present a comprehensive interaction map of all core components of the SMN complex based upon in vivo and in vitro methods. Our studies reveal a modular composition of the SMN complex with the three proteins SMN, Gemin8, and Gemin7 in its center. Onto this central building block the other components are bound via multiple interactions. Furthermore, by employing a novel assay, we were able to reconstitute the SMN complex from individual components and confirm the interaction map. Interestingly, SMN protein carrying an SMA-causing mutation was severely impaired in formation of the SMN complex. Finally, we show that the peripheral component Gemin5 contributes an essential activity to the SMN complex, most likely the transfer of Sm proteins onto the U snRNA. Collectively, the data presented here provide a basis for the detailed mechanistic and structural analysis of the assembly machinery of U snRNPs.


Subject(s)
Carrier Proteins/metabolism , Models, Molecular , Motor Neurons/metabolism , Multiprotein Complexes/metabolism , Nuclear Proteins/metabolism , Ribonucleoproteins, Small Nuclear/metabolism , Carrier Proteins/chemistry , Carrier Proteins/genetics , Cell Survival/genetics , Cell-Free System/metabolism , HeLa Cells , Humans , Motor Neurons/chemistry , Multiprotein Complexes/chemistry , Multiprotein Complexes/genetics , Muscular Atrophy, Spinal/genetics , Muscular Atrophy, Spinal/metabolism , Nuclear Proteins/chemistry , Nuclear Proteins/genetics , Ribonucleoproteins, Small Nuclear/chemistry , Ribonucleoproteins, Small Nuclear/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...