Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 63
Filter
Add more filters










Publication year range
1.
RNA ; 29(11): 1673-1690, 2023 11.
Article in English | MEDLINE | ID: mdl-37562960

ABSTRACT

U7 snRNP is a multisubunit endonuclease required for 3' end processing of metazoan replication-dependent histone pre-mRNAs. In contrast to the spliceosomal snRNPs, U7 snRNP lacks the Sm subunits D1 and D2 and instead contains two related proteins, Lsm10 and Lsm11. The remaining five subunits of the U7 heptameric Sm ring, SmE, F, G, B, and D3, are shared with the spliceosomal snRNPs. The pathway that assembles the unique ring of U7 snRNP is unknown. Here, we show that a heterodimer of Lsm10 and Lsm11 tightly interacts with the methylosome, a complex of the arginine methyltransferase PRMT5, MEP50, and pICln known to methylate arginines in the carboxy-terminal regions of the Sm proteins B, D1, and D3 during the spliceosomal Sm ring assembly. Both biochemical and cryo-EM structural studies demonstrate that the interaction is mediated by PRMT5, which binds and methylates two arginine residues in the amino-terminal region of Lsm11. Surprisingly, PRMT5 also methylates an amino-terminal arginine in SmE, a subunit that does not undergo this type of modification during the biogenesis of the spliceosomal snRNPs. An intriguing possibility is that the unique methylation pattern of Lsm11 and SmE plays a vital role in the assembly of the U7 snRNP.


Subject(s)
Ribonucleoprotein, U7 Small Nuclear , Ribonucleoproteins, Small Nuclear , Animals , Ribonucleoprotein, U7 Small Nuclear/chemistry , Methylation , Ribonucleoproteins, Small Nuclear/metabolism , Histones/metabolism , Arginine/chemistry
2.
Cell Rep ; 40(12): 111393, 2022 09 20.
Article in English | MEDLINE | ID: mdl-36130491

ABSTRACT

The neuromuscular junction (NMJ) is an essential synapse whose loss is a key hallmark of the neurodegenerative disease spinal muscular atrophy (SMA). Here, we show that activity of the SMA-determining SMN protein in the assembly of U7 small nuclear ribonucleoprotein (snRNP)-which functions in the 3'-end processing of replication-dependent histone mRNAs-is required for NMJ integrity. Co-expression of U7-specific Lsm10 and Lsm11 proteins selectively enhances U7 snRNP assembly, corrects histone mRNA processing defects, and rescues key structural and functional abnormalities of neuromuscular pathology in SMA mice-including NMJ denervation, decreased synaptic transmission, and skeletal muscle atrophy. Furthermore, U7 snRNP dysfunction drives selective loss of the synaptic organizing protein Agrin at NMJs innervating vulnerable muscles of SMA mice. These findings reveal a direct contribution of U7 snRNP dysfunction to neuromuscular pathology in SMA and suggest a role for histone gene regulation in maintaining functional synaptic connections between motor neurons and muscles.


Subject(s)
Muscular Atrophy, Spinal , Neurodegenerative Diseases , Agrin/metabolism , Animals , Histones/metabolism , Mice , Muscular Atrophy, Spinal/metabolism , Neurodegenerative Diseases/metabolism , Neuromuscular Junction/metabolism , RNA, Messenger/metabolism , Ribonucleoprotein, U7 Small Nuclear/chemistry , Ribonucleoprotein, U7 Small Nuclear/metabolism
3.
Biochem Soc Trans ; 49(5): 2229-2240, 2021 11 01.
Article in English | MEDLINE | ID: mdl-34351387

ABSTRACT

In animal cells, replication-dependent histone mRNAs end with a highly conserved stem-loop structure followed by a 4- to 5-nucleotide single-stranded tail. This unique 3' end distinguishes replication-dependent histone mRNAs from all other eukaryotic mRNAs, which end with a poly(A) tail produced by the canonical 3'-end processing mechanism of cleavage and polyadenylation. The pioneering studies of Max Birnstiel's group demonstrated nearly 40 years ago that the unique 3' end of animal replication-dependent histone mRNAs is generated by a distinct processing mechanism, whereby histone mRNA precursors are cleaved downstream of the stem-loop, but this cleavage is not followed by polyadenylation. The key role is played by the U7 snRNP, a complex of a ∼60 nucleotide U7 snRNA and many proteins. Some of these proteins, including the enzymatic component CPSF73, are shared with the canonical cleavage and polyadenylation machinery, justifying the view that the two metazoan pre-mRNA 3'-end processing mechanisms have a common evolutionary origin. The studies on U7 snRNP culminated in the recent breakthrough of reconstituting an entirely recombinant human machinery that is capable of accurately cleaving histone pre-mRNAs, and determining its structure in complex with a pre-mRNA substrate (with 13 proteins and two RNAs) that is poised for the cleavage reaction. The structure uncovered an unanticipated network of interactions within the U7 snRNP and a remarkable mechanism of activating catalytically dormant CPSF73 for the cleavage. This work provides a conceptual framework for understanding other eukaryotic 3'-end processing machineries.


Subject(s)
Histones/genetics , RNA, Messenger/metabolism , RNA, Small Nuclear/genetics , Animals , Humans , Hydrolysis , Recombinant Proteins/metabolism , Ribonucleoprotein, U7 Small Nuclear/metabolism
4.
RNA ; 27(10): 1148-1154, 2021 10.
Article in English | MEDLINE | ID: mdl-34230059

ABSTRACT

CPSF73 is the endonuclease that catalyzes the cleavage reaction for 3'-end processing of mRNA precursors (pre-mRNAs) in two distinct machineries, a canonical machinery for the majority of pre-mRNAs and a U7 snRNP (U7 machinery) for replication-dependent histone pre-mRNAs in animal cells. CPSF73 also possesses 5'-3' exonuclease activity in the U7 machinery, degrading the downstream cleavage product after the endonucleolytic cleavage. Recent studies show that CPSF73 is a potential target for developing anticancer, antimalarial, and antiprotozoal drugs, spurring interest in identifying new small-molecule inhibitors against this enzyme. CPSF73 nuclease activity has so far been demonstrated using a gel-based end-point assay, using radiolabeled or fluorescently labeled RNA substrates. By taking advantage of unique properties of the U7 machinery, we have developed a novel, real-time fluorescence assay for the nuclease activity of CPSF73. This assay is facile and high-throughput, and should also be helpful for the discovery of new CPSF73 inhibitors.


Subject(s)
Biological Assay , Cleavage And Polyadenylation Specificity Factor/metabolism , Histones/metabolism , RNA 3' End Processing , RNA Precursors/metabolism , Ribonucleoprotein, U7 Small Nuclear/metabolism , Cell-Free System , Cleavage And Polyadenylation Specificity Factor/chemistry , Cleavage And Polyadenylation Specificity Factor/genetics , Enzyme Assays , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacology , Fluorescence , Fluorescent Dyes/chemistry , Histones/chemistry , Histones/genetics , Humans , Models, Molecular , Phenylalanine/analogs & derivatives , Phenylalanine/chemistry , Phenylalanine/pharmacology , Piperazines/chemistry , Piperazines/pharmacology , Proteolysis , RNA Precursors/chemistry , RNA Precursors/genetics , Rhodamines/chemistry , Ribonucleoprotein, U7 Small Nuclear/chemistry , Ribonucleoprotein, U7 Small Nuclear/genetics , Small Molecule Libraries/chemistry , Small Molecule Libraries/pharmacology
5.
Methods Enzymol ; 655: 291-324, 2021.
Article in English | MEDLINE | ID: mdl-34183127

ABSTRACT

In animal cells, replication-dependent histone pre-mRNAs are processed at the 3'-end by an endonucleolytic cleavage carried out by the U7 snRNP, a machinery that contains the U7 snRNA and many protein subunits. Studies on the composition of this machinery and understanding of its role in 3'-end processing were greatly facilitated by the development of an in vitro system utilizing nuclear extracts from mammalian cells 35 years ago and later from Drosophila cells. Most recently, recombinant expression and purification of the components of the machinery have enabled the full reconstitution of an active machinery and its complex with a model pre-mRNA substrate, using 13 proteins and 2 RNAs, and the determination of the structure of this active machinery. This chapter presents protocols for preparing nuclear extracts containing endogenous processing machinery, for assembling semi-recombinant and fully reconstituted machineries, and for histone pre-mRNA 3'-end processing assays with these samples.


Subject(s)
Histones , RNA Precursors , Animals , Drosophila/metabolism , Histones/genetics , Histones/metabolism , Humans , RNA Precursors/genetics , RNA Precursors/metabolism , RNA Processing, Post-Transcriptional , Ribonucleoprotein, U7 Small Nuclear/genetics , Ribonucleoprotein, U7 Small Nuclear/metabolism
6.
Sci Rep ; 11(1): 11868, 2021 06 04.
Article in English | MEDLINE | ID: mdl-34088960

ABSTRACT

Genes encoding replication-dependent histones lack introns, and the mRNAs produced are a unique class of RNA polymerase II transcripts in eukaryotic cells that do not end in a polyadenylated tail. Mature mRNAs are thus formed by a single endonucleolytic cleavage that releases the pre-mRNA from the DNA and is the only processing event necessary. U7 snRNP is one of the key factors that determines the cleavage site within the 3'UTR of replication-dependent histone pre-mRNAs. We have previously showed that the FUS protein interacts with U7 snRNA/snRNP and regulates the expression of histone genes by stimulating transcription and 3' end maturation. Mutations in the FUS gene first identified in patients with amyotrophic lateral sclerosis (ALS) lead to the accumulation of the FUS protein in cytoplasmic inclusions. Here, we report that mutations in FUS lead to disruption of the transcriptional activity of FUS and mislocalization of U7 snRNA/snRNP in cytoplasmic aggregates in cellular models and primary neurons. As a consequence, decreased transcriptional efficiency and aberrant 3' end processing of histone pre-mRNAs were observed. This study highlights for the first time the deregulation of replication-dependent histone gene expression and its involvement in ALS.


Subject(s)
Amyotrophic Lateral Sclerosis/genetics , Gene Expression Regulation , Histones/metabolism , Mutation , RNA-Binding Protein FUS/genetics , Ribonucleoprotein, U7 Small Nuclear/genetics , 3' Untranslated Regions , Cell Line, Tumor , Cell Nucleus/metabolism , Cytoplasm/metabolism , Gene Expression Profiling , HeLa Cells , Humans , In Situ Hybridization, Fluorescence , Neurosciences , Plasmids/metabolism , RNA, Small Nuclear/genetics , Ribonucleoproteins, Small Nuclear/genetics
7.
Mol Biol Cell ; 32(9): 942-955, 2021 04 19.
Article in English | MEDLINE | ID: mdl-33788585

ABSTRACT

The histone locus body (HLB) is an evolutionarily conserved nuclear body that regulates the transcription and processing of replication-dependent (RD) histone mRNAs, which are the only eukaryotic mRNAs lacking a poly-A tail. Many nuclear bodies contain distinct domains, but how internal organization is related to nuclear body function is not fully understood. Here, we demonstrate using structured illumination microscopy that Drosophila HLBs have a "core-shell" organization in which the internal core contains transcriptionally active RD histone genes. The N-terminus of Mxc, which contains a domain required for Mxc oligomerization, HLB assembly, and RD histone gene expression, is enriched in the HLB core. In contrast, the C-terminus of Mxc is enriched in the HLB outer shell as is FLASH, a component of the active U7 snRNP that cotranscriptionally cleaves RD histone pre-mRNA. Consistent with these results, we show biochemically that FLASH binds directly to the Mxc C-terminal region. In the rapid S-M nuclear cycles of syncytial blastoderm Drosophila embryos, the HLB disassembles at mitosis and reassembles the core-shell arrangement as histone gene transcription is activated immediately after mitosis. Thus, the core-shell organization is coupled to zygotic histone gene transcription, revealing a link between HLB internal organization and RD histone gene expression.


Subject(s)
Cell Nucleus Structures/metabolism , Histones/metabolism , Microscopy/methods , Animals , Carrier Proteins/metabolism , Cell Nucleus/metabolism , Cell Nucleus Structures/genetics , Drosophila Proteins/metabolism , Drosophila melanogaster/metabolism , Mitosis , RNA Precursors/metabolism , RNA Processing, Post-Transcriptional , RNA, Messenger/metabolism , Regulatory Elements, Transcriptional/genetics , Ribonucleoprotein, U7 Small Nuclear/metabolism , Tumor Suppressor Proteins/metabolism , Zygote/metabolism
8.
Nat Genet ; 52(12): 1364-1372, 2020 12.
Article in English | MEDLINE | ID: mdl-33230297

ABSTRACT

Inappropriate stimulation or defective negative regulation of the type I interferon response can lead to autoinflammation. In genetically uncharacterized cases of the type I interferonopathy Aicardi-Goutières syndrome, we identified biallelic mutations in LSM11 and RNU7-1, which encode components of the replication-dependent histone pre-mRNA-processing complex. Mutations were associated with the misprocessing of canonical histone transcripts and a disturbance of linker histone stoichiometry. Additionally, we observed an altered distribution of nuclear cyclic guanosine monophosphate-adenosine monophosphate synthase (cGAS) and enhanced interferon signaling mediated by the cGAS-stimulator of interferon genes (STING) pathway in patient-derived fibroblasts. Finally, we established that chromatin without linker histone stimulates cyclic guanosine monophosphate-adenosine monophosphate (cGAMP) production in vitro more efficiently. We conclude that nuclear histones, as key constituents of chromatin, are essential in suppressing the immunogenicity of self-DNA.


Subject(s)
Chromatin/metabolism , Histones/metabolism , Interferon Type I/biosynthesis , RNA Precursors/metabolism , RNA-Binding Proteins/genetics , Ribonucleoprotein, U7 Small Nuclear/genetics , Autoimmune Diseases of the Nervous System/genetics , Autoimmune Diseases of the Nervous System/immunology , Cell Line , DNA/immunology , Gene Expression Regulation/genetics , Gene Expression Regulation/immunology , HCT116 Cells , HEK293 Cells , Hereditary Autoinflammatory Diseases/genetics , Hereditary Autoinflammatory Diseases/immunology , Humans , Membrane Proteins/metabolism , Nervous System Malformations/genetics , Nervous System Malformations/immunology , Nucleotides, Cyclic/biosynthesis , Nucleotidyltransferases/metabolism
9.
RNA ; 26(10): 1345-1359, 2020 10.
Article in English | MEDLINE | ID: mdl-32554553

ABSTRACT

Metazoan replication-dependent histone pre-mRNAs are cleaved at the 3' end by U7 snRNP, an RNA-guided endonuclease that contains U7 snRNA, seven proteins of the Sm ring, FLASH, and four polyadenylation factors: symplekin, CPSF73, CPSF100, and CstF64. A fully recombinant U7 snRNP was recently reconstituted from all 13 components for functional and structural studies and shown to accurately cleave histone pre-mRNAs. Here, we analyzed the activity of recombinant U7 snRNP in more detail. We demonstrate that in addition to cleaving histone pre-mRNAs endonucleolytically, reconstituted U7 snRNP acts as a 5'-3' exonuclease that degrades the downstream product generated from histone pre-mRNAs as a result of the endonucleolytic cleavage. Surprisingly, recombinant U7 snRNP also acts as an endonuclease on single-stranded DNA substrates. All these activities depend on the ability of U7 snRNA to base-pair with the substrate and on the presence of the amino-terminal domain (NTD) of symplekin in either cis or trans, and are abolished by mutations within the catalytic center of CPSF73, or by binding of the NTD to the SSU72 phosphatase of RNA polymerase II. Altogether, our results demonstrate that recombinant U7 snRNP functionally mimics its endogenous counterpart and provide evidence that CPSF73 is both an endonuclease and a 5'-3' exonuclease, consistent with the activity of other members of the ß-CASP family. Our results also raise the intriguing possibility that CPSF73 may be involved in some aspects of DNA metabolism in vivo.


Subject(s)
Cleavage And Polyadenylation Specificity Factor/genetics , Endonucleases/genetics , Exonucleases/genetics , RNA, Small Nuclear/genetics , Ribonucleoprotein, U7 Small Nuclear/genetics , Animals , Histones/genetics , Mice , RNA Precursors/genetics , RNA Processing, Post-Transcriptional/genetics
10.
Science ; 367(6478): 700-703, 2020 02 07.
Article in English | MEDLINE | ID: mdl-32029631

ABSTRACT

The 3'-end processing machinery for metazoan replication-dependent histone precursor messenger RNAs (pre-mRNAs) contains the U7 small nuclear ribonucleoprotein and shares the key cleavage module with the canonical cleavage and polyadenylation machinery. We reconstituted an active human histone pre-mRNA processing machinery using 13 recombinant proteins and two RNAs and determined its structure by cryo-electron microscopy. The overall structure is highly asymmetrical and resembles an amphora with one long handle. We captured the pre-mRNA in the active site of the endonuclease, the 73-kilodalton subunit of the cleavage and polyadenylation specificity factor, poised for cleavage. The endonuclease and the entire cleavage module undergo extensive rearrangements for activation, triggered through the recognition of the duplex between the authentic pre-mRNA and U7 small nuclear RNA (snRNA). Our study also has notable implications for understanding canonical and snRNA 3'-end processing.


Subject(s)
Cleavage And Polyadenylation Specificity Factor/chemistry , Histones/genetics , RNA Cleavage , RNA Precursors/metabolism , Catalytic Domain , Cryoelectron Microscopy , Humans , Polyadenylation , RNA, Small Nuclear/metabolism , Recombinant Proteins , Ribonucleoprotein, U7 Small Nuclear/chemistry
11.
Nucleic Acids Res ; 48(3): 1508-1530, 2020 02 20.
Article in English | MEDLINE | ID: mdl-31819999

ABSTRACT

In animal cells, replication-dependent histone pre-mRNAs are cleaved at the 3' end by U7 snRNP consisting of two core components: a ∼60-nucleotide U7 snRNA and a ring of seven proteins, with Lsm10 and Lsm11 replacing the spliceosomal SmD1 and SmD2. Lsm11 interacts with FLASH and together they recruit the endonuclease CPSF73 and other polyadenylation factors, forming catalytically active holo U7 snRNP. Here, we assembled core U7 snRNP bound to FLASH from recombinant components and analyzed its appearance by electron microscopy and ability to support histone pre-mRNA processing in the presence of polyadenylation factors from nuclear extracts. We demonstrate that semi-recombinant holo U7 snRNP reconstituted in this manner has the same composition and functional properties as endogenous U7 snRNP, and accurately cleaves histone pre-mRNAs in a reconstituted in vitro processing reaction. We also demonstrate that the U7-specific Sm ring assembles efficiently in vitro on a spliceosomal Sm site but the engineered U7 snRNP is functionally impaired. This approach offers a unique opportunity to study the importance of various regions in the Sm proteins and U7 snRNA in 3' end processing of histone pre-mRNAs.


Subject(s)
Apoptosis Regulatory Proteins/genetics , Calcium-Binding Proteins/genetics , RNA-Binding Proteins/genetics , Ribonucleoprotein, U7 Small Nuclear/genetics , Ribonucleoproteins, Small Nuclear/genetics , Amino Acid Sequence/genetics , Animals , Cell Nucleus/genetics , Drosophila/genetics , Histones/genetics , Humans , Mice , Protein Binding/genetics , RNA Precursors/genetics , Spliceosomes/genetics , mRNA Cleavage and Polyadenylation Factors/genetics
12.
EMBO J ; 38(9)2019 05 02.
Article in English | MEDLINE | ID: mdl-30858280

ABSTRACT

The RNA-binding protein ALYREF plays key roles in nuclear export and also 3'-end processing of polyadenylated mRNAs, but whether such regulation also extends to non-polyadenylated RNAs is unknown. Replication-dependent (RD)-histone mRNAs are not polyadenylated, but instead end in a stem-loop (SL) structure. Here, we demonstrate that ALYREF prevalently binds a region next to the SL on RD-histone mRNAs. SL-binding protein (SLBP) directly interacts with ALYREF and promotes its recruitment. ALYREF promotes histone pre-mRNA 3'-end processing by facilitating U7-snRNP recruitment through physical interaction with the U7-snRNP-specific component Lsm11. Furthermore, ALYREF, together with other components of the TREX complex, enhances histone mRNA export. Moreover, we show that 3'-end processing promotes ALYREF recruitment and histone mRNA export. Together, our results point to an important role of ALYREF in coordinating 3'-end processing and nuclear export of non-polyadenylated mRNAs.


Subject(s)
Histones/metabolism , Nuclear Proteins/metabolism , RNA Processing, Post-Transcriptional , RNA Transport , RNA, Messenger/metabolism , RNA-Binding Proteins/metabolism , Ribonucleoprotein, U7 Small Nuclear/metabolism , Transcription Factors/metabolism , Active Transport, Cell Nucleus , Exodeoxyribonucleases/genetics , Exodeoxyribonucleases/metabolism , Histones/genetics , Humans , Nuclear Proteins/genetics , Nucleocytoplasmic Transport Proteins/genetics , Nucleocytoplasmic Transport Proteins/metabolism , Phosphoproteins/genetics , Phosphoproteins/metabolism , RNA, Messenger/genetics , RNA-Binding Proteins/genetics , Ribonucleoprotein, U7 Small Nuclear/genetics , Transcription Factors/genetics
13.
BMC Mol Biol ; 19(1): 9, 2018 07 27.
Article in English | MEDLINE | ID: mdl-30053800

ABSTRACT

BACKGROUND: Core canonical histones are required in the S phase of the cell cycle to pack newly synthetized DNA, therefore the expression of their genes is highly activated during DNA replication. In mammalian cells, this increment is achieved by both enhanced transcription and 3' end processing. In this paper, we described positive cofactor 4 (PC4) as a protein that contributes to the regulation of replication-dependent histone gene expression. RESULTS: We showed that PC4 influences RNA polymerase II recruitment to histone gene loci in a cell cycle-dependent manner. The most important effect was observed in S phase where PC4 knockdown leads to the elevated level of RNA polymerase II on histone genes, which corresponds to the increased total level of those gene transcripts. The opposite effect was caused by PC4 overexpression. Moreover, we found that PC4 has a negative effect on the unique 3' end processing of histone pre-mRNAs that can be based on the interaction of PC4 with U7 snRNP and CstF64. Interestingly, this effect does not depend on the cell cycle. CONCLUSIONS: We conclude that PC4 might repress RNA polymerase II recruitment and transcription of replication-dependent histone genes in order to maintain the very delicate balance between histone gene expression and DNA synthesis. It guards the cell from excess of histones in S phase. Moreover, PC4 might promote the interaction of cleavage and polyadenylation complex with histone pre-mRNAs, that might impede with the recruitment of histone cleavage complex. This in turn decreases the 3' end processing efficiency of histone gene transcripts.


Subject(s)
DNA Replication , DNA-Binding Proteins/metabolism , Histones/genetics , RNA Polymerase II/metabolism , Transcription Factors/metabolism , Cell Cycle , Cleavage Stimulation Factor/metabolism , Gene Expression Regulation , HEK293 Cells , HeLa Cells , Humans , RNA 3' End Processing , Ribonucleoprotein, U7 Small Nuclear/metabolism
14.
Nucleic Acids Res ; 46(9): 4752-4770, 2018 05 18.
Article in English | MEDLINE | ID: mdl-29529248

ABSTRACT

3' end cleavage of metazoan replication-dependent histone pre-mRNAs requires the multi-subunit holo-U7 snRNP and the stem-loop binding protein (SLBP). The exact composition of the U7 snRNP and details of SLBP function in processing remain unclear. To identify components of the U7 snRNP in an unbiased manner, we developed a novel approach for purifying processing complexes from Drosophila and mouse nuclear extracts. In this method, catalytically active processing complexes are assembled in vitro on a cleavage-resistant histone pre-mRNA containing biotin and a photo-sensitive linker, and eluted from streptavidin beads by UV irradiation for direct analysis by mass spectrometry. In the purified processing complexes, Drosophila and mouse U7 snRNP have a remarkably similar composition, always being associated with CPSF73, CPSF100, symplekin and CstF64. Many other proteins previously implicated in the U7-dependent processing are not present. Drosophila U7 snRNP bound to histone pre-mRNA in the absence of SLBP contains the same subset of polyadenylation factors but is catalytically inactive and addition of recombinant SLBP is sufficient to trigger cleavage. This result suggests that Drosophila SLBP promotes a structural rearrangement of the processing complex, resulting in juxtaposition of the CPSF73 endonuclease with the cleavage site in the pre-mRNA substrate.


Subject(s)
Histones/genetics , RNA 3' End Processing , RNA Precursors/metabolism , RNA, Messenger/metabolism , Ribonucleoprotein, U7 Small Nuclear/chemistry , Ribonucleoprotein, U7 Small Nuclear/metabolism , Animals , Biocatalysis , Biotin , Drosophila Proteins/isolation & purification , Histones/metabolism , Mass Spectrometry , Mice , Nucleotides/chemistry , RNA Cleavage , RNA Precursors/chemistry , RNA, Messenger/chemistry , Ribonucleoprotein, U7 Small Nuclear/isolation & purification , Tumor Cells, Cultured , Ultraviolet Rays
15.
RNA ; 23(6): 938-951, 2017 06.
Article in English | MEDLINE | ID: mdl-28289156

ABSTRACT

Cleavage of histone pre-mRNAs at the 3' end requires stem-loop binding protein (SLBP) and U7 snRNP that consists of U7 snRNA and a unique Sm ring containing two U7-specific proteins: Lsm10 and Lsm11. Lsm11 interacts with FLASH and together they bring a subset of polyadenylation factors to U7 snRNP, including the CPSF73 endonuclease that cleaves histone pre-mRNA. SLBP binds to a conserved stem-loop structure upstream of the cleavage site and acts by promoting an interaction between the U7 snRNP and a sequence element located downstream from the cleavage site. We show that both human and Drosophila SLBPs stabilize U7 snRNP on histone pre-mRNA via two regions that are not directly involved in recognizing the stem-loop structure: helix B of the RNA binding domain and the C-terminal region that follows the RNA binding domain. Stabilization of U7 snRNP binding to histone pre-mRNA by SLBP requires FLASH but not the polyadenylation factors. Thus, FLASH plays two roles in 3' end processing of histone pre-mRNAs: It interacts with Lsm11 to form a docking platform for the polyadenylation factors, and it cooperates with SLBP to recruit U7 snRNP to histone pre-mRNA.


Subject(s)
Apoptosis Regulatory Proteins/metabolism , Calcium-Binding Proteins/metabolism , Histones/genetics , RNA Precursors/genetics , RNA Precursors/metabolism , Ribonucleoprotein, U7 Small Nuclear/metabolism , Amino Acid Sequence , Animals , Cell Line , Drosophila , Drosophila Proteins/chemistry , Drosophila Proteins/genetics , Drosophila Proteins/metabolism , Humans , Mice , Models, Biological , Models, Molecular , Multiprotein Complexes/metabolism , Mutation , Nuclear Proteins/metabolism , Protein Binding , Protein Conformation , Protein Interaction Domains and Motifs , RNA Precursors/chemistry , RNA-Binding Proteins/chemistry , RNA-Binding Proteins/genetics , RNA-Binding Proteins/metabolism , mRNA Cleavage and Polyadenylation Factors/metabolism
16.
RNA Biol ; 14(6): 726-738, 2017 06 03.
Article in English | MEDLINE | ID: mdl-28059623

ABSTRACT

Metazoan replication-dependent (RD) histone genes encode the only known cellular mRNAs that are not polyadenylated. These mRNAs end instead in a conserved stem-loop, which is formed by an endonucleolytic cleavage of the pre-mRNA. The genes for all 5 histone proteins are clustered in all metazoans and coordinately regulated with high levels of expression during S phase. Production of histone mRNAs occurs in a nuclear body called the Histone Locus Body (HLB), a subdomain of the nucleus defined by a concentration of factors necessary for histone gene transcription and pre-mRNA processing. These factors include the scaffolding protein NPAT, essential for histone gene transcription, and FLASH and U7 snRNP, both essential for histone pre-mRNA processing. Histone gene expression is activated by Cyclin E/Cdk2-mediated phosphorylation of NPAT at the G1-S transition. The concentration of factors within the HLB couples transcription with pre-mRNA processing, enhancing the efficiency of histone mRNA biosynthesis.


Subject(s)
Cell Cycle/genetics , Gene Expression Regulation , Genetic Loci , Histones/genetics , Histones/metabolism , Animals , Coiled Bodies/genetics , Coiled Bodies/metabolism , Humans , Protein Binding , RNA Precursors/genetics , RNA Precursors/metabolism , RNA Processing, Post-Transcriptional , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA, Small Nuclear/genetics , RNA, Small Nuclear/metabolism , Ribonucleoprotein, U7 Small Nuclear/metabolism
17.
RNA Biol ; 14(6): 752-760, 2017 06 03.
Article in English | MEDLINE | ID: mdl-27858508

ABSTRACT

Nuclear bodies are cellular compartments that lack lipid bilayers and harbor specific RNAs and proteins. Recent proposals that nuclear bodies form through liquid-liquid phase separation leave the question of how different nuclear bodies maintain their distinct identities unanswered. Here we investigate Cajal bodies (CBs), histone locus bodies (HLBs) and nucleoli - involved in assembly of the splicing machinery, histone mRNA 3' end processing, and rRNA processing, respectively - in the embryos of the zebrafish, Danio rerio. We take advantage of the transcriptional silence of the 1-cell embryo and follow nuclear body appearance as zygotic transcription becomes activated. CBs are present from fertilization onwards, while HLB and nucleolar components formed foci several hours later when histone genes and rDNA became active. HLB formation was blocked by transcription inhibition, suggesting nascent histone transcripts recruit HLB components like U7 snRNP. Surprisingly, we found that U7 base-pairing with nascent histone transcripts was not required for localization to HLBs. Rather, the type of Sm ring assembled on U7 determined its targeting to HLBs or CBs; the spliceosomal Sm ring targeted snRNAs to CBs while the specialized U7 Sm-ring localized to HLBs, demonstrating the contribution of protein constituents to the distinction among nuclear bodies. Thus, nucleolar, HLB, and CB components can mix in early embryogenesis when transcription is naturally or artificially silenced. These data support a model in which transcription of specific gene loci nucleates nuclear body components with high specificity and fidelity to perform distinct regulatory functions.


Subject(s)
Cell Nucleolus/metabolism , Coiled Bodies/metabolism , Histones/metabolism , Transcriptional Activation , Zebrafish/embryology , Zebrafish/physiology , Animals , Cell Nucleolus/genetics , Chromosomal Proteins, Non-Histone/metabolism , Coiled Bodies/genetics , DNA, Ribosomal/genetics , Embryonic Development/genetics , Histones/genetics , Models, Biological , RNA Splicing , RNA, Small Nuclear/genetics , RNA, Small Nuclear/metabolism , Ribonucleoprotein, U7 Small Nuclear/metabolism , Ribonucleoproteins, Small Nuclear , Spliceosomes , Zygote/metabolism
18.
J Cell Biol ; 213(5): 509-11, 2016 06 06.
Article in English | MEDLINE | ID: mdl-27241912

ABSTRACT

The classic archetypal function of nuclear bodies is to accelerate specific reactions within their crowded space. In this issue, Tatomer et al. (2016. J. Cell Biol http://dx.doi.org/10.1083/jcb.201504043) provide the first direct evidence that the histone locus body acts to concentrate key factors required for the proper processing of histone pre-mRNAs.


Subject(s)
Intranuclear Inclusion Bodies/metabolism , Animals , Drosophila melanogaster/metabolism , Histones/metabolism , Humans , Models, Biological , RNA/metabolism , Ribonucleoprotein, U7 Small Nuclear/metabolism
19.
J Cell Biol ; 213(5): 557-70, 2016 06 06.
Article in English | MEDLINE | ID: mdl-27241916

ABSTRACT

The histone locus body (HLB) assembles at replication-dependent histone genes and concentrates factors required for histone messenger RNA (mRNA) biosynthesis. FLASH (Flice-associated huge protein) and U7 small nuclear RNP (snRNP) are HLB components that participate in 3' processing of the nonpolyadenylated histone mRNAs by recruiting the endonuclease CPSF-73 to histone pre-mRNA. Using transgenes to complement a FLASH mutant, we show that distinct domains of FLASH involved in U7 snRNP binding, histone pre-mRNA cleavage, and HLB localization are all required for proper FLASH function in vivo. By genetically manipulating HLB composition using mutations in FLASH, mutations in the HLB assembly factor Mxc, or depletion of the variant histone H2aV, we find that failure to concentrate FLASH and/or U7 snRNP in the HLB impairs histone pre-mRNA processing. This failure results in accumulation of small amounts of polyadenylated histone mRNA and nascent read-through transcripts at the histone locus. Thus, the HLB concentrates FLASH and U7 snRNP, promoting efficient histone mRNA biosynthesis and coupling 3' end processing with transcription termination.


Subject(s)
Drosophila melanogaster/genetics , Genetic Loci , Histones/genetics , RNA Precursors/genetics , RNA Processing, Post-Transcriptional/genetics , Amino Acid Sequence , Animals , Carrier Proteins/chemistry , Carrier Proteins/metabolism , Drosophila Proteins/chemistry , Drosophila Proteins/metabolism , Histones/metabolism , In Situ Hybridization, Fluorescence , Models, Biological , Mutant Proteins/chemistry , Mutant Proteins/metabolism , Mutation/genetics , Phenotype , RNA Precursors/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Ribonucleoprotein, U7 Small Nuclear/metabolism , Transgenes
20.
Nucleic Acids Res ; 43(20): 9711-28, 2015 Nov 16.
Article in English | MEDLINE | ID: mdl-26250115

ABSTRACT

Replication-dependent histone genes are up-regulated during the G1/S phase transition to meet the requirement for histones to package the newly synthesized DNA. In mammalian cells, this increment is achieved by enhanced transcription and 3' end processing. The non-polyadenylated histone mRNA 3' ends are generated by a unique mechanism involving the U7 small ribonucleoprotein (U7 snRNP). By using affinity purification methods to enrich U7 snRNA, we identified FUS/TLS as a novel U7 snRNP interacting protein. Both U7 snRNA and histone transcripts can be precipitated by FUS antibodies predominantly in the S phase of the cell cycle. Moreover, FUS depletion leads to decreased levels of correctly processed histone mRNAs and increased levels of extended transcripts. Interestingly, FUS antibodies also co-immunoprecipitate histone transcriptional activator NPAT and transcriptional repressor hnRNP UL1 in different phases of the cell cycle. We further show that FUS binds to histone genes in S phase, promotes the recruitment of RNA polymerase II and is important for the activity of histone gene promoters. Thus, FUS may serve as a linking factor that positively regulates histone gene transcription and 3' end processing by interacting with the U7 snRNP and other factors involved in replication-dependent histone gene expression.


Subject(s)
DNA Replication , Gene Expression Regulation , Histones/genetics , RNA-Binding Protein FUS/metabolism , Ribonucleoprotein, U7 Small Nuclear/metabolism , Transcription, Genetic , Cell Cycle , Cell Cycle Proteins/metabolism , HEK293 Cells , HeLa Cells , Heterogeneous-Nuclear Ribonucleoproteins/metabolism , Histones/biosynthesis , Humans , Nuclear Proteins/metabolism , Promoter Regions, Genetic , RNA, Small Nuclear/metabolism , Transcription Factors/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...