Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 2.238
Filter
Add more filters

Publication year range
1.
Annu Rev Cell Dev Biol ; 40(1): 265-281, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39356809

ABSTRACT

Antibodies that gain specificity by a large insert encoding for an extra domain were described for the first time in 2016. In malaria-exposed individuals, an exon deriving from the leukocyte-associated immunoglobulin-like 1 (LAIR1) gene integrated via a copy-and-paste insertion into the immunoglobulin heavy chain encoding region. A few years later, a second example was identified, namely a dual exon integration from the leukocyte immunoglobulin-like receptor B1 (LILRB1) gene that is located in close proximity to LAIR1. A dedicated high-throughput characterization of chimeric immunoglobulin heavy chain transcripts unraveled, that insertions from distant genomic regions (including mitochondrial DNA) can contribute to human antibody diversity. This review describes the modalities of insert-containing antibodies. The role of known DNA mobility aspects, such as genomic translocation, gene conversion, and DNA fragility, is discussed in the context of insert-antibody generation. Finally, the review covers why insert antibodies were omitted from the past repertoire analyses and how insert antibodies can contribute to protective immunity or an autoreactive response.


Subject(s)
Exons , V(D)J Recombination , Humans , V(D)J Recombination/genetics , Exons/genetics , Animals , Antibodies/immunology , Antibodies/genetics , Receptors, Immunologic/genetics , Receptors, Immunologic/metabolism , Receptors, Immunologic/immunology , Antibody Diversity/genetics
2.
Annu Rev Biochem ; 89: 333-358, 2020 06 20.
Article in English | MEDLINE | ID: mdl-31815536

ABSTRACT

Splicing of the precursor messenger RNA, involving intron removal and exon ligation, is mediated by the spliceosome. Together with biochemical and genetic investigations of the past four decades, structural studies of the intact spliceosome at atomic resolution since 2015 have led to mechanistic delineation of RNA splicing with remarkable insights. The spliceosome is proven to be a protein-orchestrated metalloribozyme. Conserved elements of small nuclear RNA (snRNA) constitute the splicing active site with two catalytic metal ions and recognize three conserved intron elements through duplex formation, which are delivered into the splicing active site for branching and exon ligation. The protein components of the spliceosome stabilize the conformation of the snRNA, drive spliceosome remodeling, orchestrate the movement of the RNA elements, and facilitate the splicing reaction. The overall organization of the spliceosome and the configuration of the splicing active site are strictly conserved between human and yeast.


Subject(s)
RNA Splicing Factors/genetics , RNA Splicing , RNA-Binding Proteins/genetics , Ribonucleoprotein, U4-U6 Small Nuclear/genetics , Ribonucleoprotein, U5 Small Nuclear/genetics , Saccharomyces cerevisiae Proteins/genetics , Saccharomyces cerevisiae/genetics , Spliceosomes/metabolism , Catalytic Domain , Conserved Sequence , Exons , Humans , Introns , Models, Molecular , Nucleic Acid Conformation , Protein Structure, Secondary , RNA Helicases/chemistry , RNA Helicases/genetics , RNA Helicases/metabolism , RNA Precursors/chemistry , RNA Precursors/genetics , RNA Precursors/metabolism , RNA Splicing Factors/chemistry , RNA Splicing Factors/metabolism , RNA, Small Nuclear/chemistry , RNA, Small Nuclear/genetics , RNA, Small Nuclear/metabolism , RNA-Binding Proteins/chemistry , RNA-Binding Proteins/metabolism , Ribonucleoprotein, U4-U6 Small Nuclear/chemistry , Ribonucleoprotein, U4-U6 Small Nuclear/metabolism , Ribonucleoprotein, U5 Small Nuclear/chemistry , Ribonucleoprotein, U5 Small Nuclear/metabolism , Saccharomyces cerevisiae/metabolism , Saccharomyces cerevisiae Proteins/chemistry , Saccharomyces cerevisiae Proteins/metabolism , Spliceosomes/genetics , Spliceosomes/ultrastructure
3.
Cell ; 169(5): 918-929.e14, 2017 May 18.
Article in English | MEDLINE | ID: mdl-28502770

ABSTRACT

Mechanistic understanding of pre-mRNA splicing requires detailed structural information on various states of the spliceosome. Here we report the cryo electron microscopy (cryo-EM) structure of the human spliceosome just before exon ligation (the C∗ complex) at an average resolution of 3.76 Å. The splicing factor Prp17 stabilizes the active site conformation. The step II factor Slu7 adopts an extended conformation, binds Prp8 and Cwc22, and is poised for selection of the 3'-splice site. Remarkably, the intron lariat traverses through a positively charged central channel of RBM22; this unusual organization suggests mechanisms of intron recruitment, confinement, and release. The protein PRKRIP1 forms a 100-Å α helix linking the distant U2 snRNP to the catalytic center. A 35-residue fragment of the ATPase/helicase Prp22 latches onto Prp8, and the quaternary exon junction complex (EJC) recognizes upstream 5'-exon sequences and associates with Cwc22 and the GTPase Snu114. These structural features reveal important mechanistic insights into exon ligation.


Subject(s)
RNA Precursors/metabolism , Spliceosomes/chemistry , Spliceosomes/ultrastructure , Base Sequence , Cryoelectron Microscopy , DEAD-box RNA Helicases/metabolism , Exons , Humans , Introns , Models, Molecular , RNA Splicing , RNA Splicing Factors/metabolism , RNA-Binding Proteins/metabolism , Saccharomyces cerevisiae/chemistry , Spliceosomes/metabolism
4.
Cell ; 168(5): 843-855.e13, 2017 02 23.
Article in English | MEDLINE | ID: mdl-28215706

ABSTRACT

The transcription-related DNA damage response was analyzed on a genome-wide scale with great spatial and temporal resolution. Upon UV irradiation, a slowdown of transcript elongation and restriction of gene activity to the promoter-proximal ∼25 kb is observed. This is associated with a shift from expression of long mRNAs to shorter isoforms, incorporating alternative last exons (ALEs) that are more proximal to the transcription start site. Notably, this includes a shift from a protein-coding ASCC3 mRNA to a shorter ALE isoform of which the RNA, rather than an encoded protein, is critical for the eventual recovery of transcription. The non-coding ASCC3 isoform counteracts the function of the protein-coding isoform, indicating crosstalk between them. Thus, the ASCC3 gene expresses both coding and non-coding transcript isoforms with opposite effects on transcription recovery after UV-induced DNA damage.


Subject(s)
Alternative Splicing/radiation effects , DNA Helicases/genetics , RNA, Untranslated/genetics , Transcription, Genetic , Ultraviolet Rays , Cell Line , Exons , Humans , RNA Polymerase II/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Transcription Elongation, Genetic/radiation effects , Transcription Initiation, Genetic/radiation effects
5.
Cell ; 171(7): 1589-1598.e8, 2017 Dec 14.
Article in English | MEDLINE | ID: mdl-29153833

ABSTRACT

Removal of an intron from a pre-mRNA by the spliceosome results in the ligation of two exons in the post-catalytic spliceosome (known as the P complex). Here, we present a cryo-EM structure of the P complex from Saccharomyces cerevisiae at an average resolution of 3.6 Å. The ligated exon is held in the active site through RNA-RNA contacts. Three bases at the 3' end of the 5' exon remain anchored to loop I of U5 small nuclear RNA, and the conserved AG nucleotides of the 3'-splice site (3'SS) are specifically recognized by the invariant adenine of the branch point sequence, the guanine base at the 5' end of the 5'SS, and an adenine base of U6 snRNA. The 3'SS is stabilized through an interaction with the 1585-loop of Prp8. The P complex structure provides a view on splice junction formation critical for understanding the complete splicing cycle.


Subject(s)
Saccharomyces cerevisiae/chemistry , Spliceosomes/chemistry , Cryoelectron Microscopy , Humans , Models, Molecular , RNA Splicing , Saccharomyces cerevisiae/metabolism , Spliceosomes/metabolism
6.
Mol Cell ; 84(13): 2553-2572.e19, 2024 Jul 11.
Article in English | MEDLINE | ID: mdl-38917794

ABSTRACT

CRISPR-Cas technology has transformed functional genomics, yet understanding of how individual exons differentially shape cellular phenotypes remains limited. Here, we optimized and conducted massively parallel exon deletion and splice-site mutation screens in human cell lines to identify exons that regulate cellular fitness. Fitness-promoting exons are prevalent in essential and highly expressed genes and commonly overlap with protein domains and interaction interfaces. Conversely, fitness-suppressing exons are enriched in nonessential genes, exhibiting lower inclusion levels, and overlap with intrinsically disordered regions and disease-associated mutations. In-depth mechanistic investigation of the screen-hit TAF5 alternative exon-8 revealed that its inclusion is required for assembly of the TFIID general transcription initiation complex, thereby regulating global gene expression output. Collectively, our orthogonal exon perturbation screens established a comprehensive repository of phenotypically important exons and uncovered regulatory mechanisms governing cellular fitness and gene expression.


Subject(s)
Exons , Humans , Exons/genetics , CRISPR-Cas Systems , Transcription Factor TFIID/genetics , Transcription Factor TFIID/metabolism , Genetic Fitness , HEK293 Cells , TATA-Binding Protein Associated Factors/genetics , TATA-Binding Protein Associated Factors/metabolism , RNA Splice Sites , Mutation , Gene Expression Regulation , Alternative Splicing
7.
Mol Cell ; 84(14): 2634-2647.e9, 2024 Jul 25.
Article in English | MEDLINE | ID: mdl-38964321

ABSTRACT

DNA repair is directly performed by hundreds of core factors and indirectly regulated by thousands of others. We massively expanded a CRISPR inhibition and Cas9-editing screening system to discover factors indirectly modulating homology-directed repair (HDR) in the context of ∼18,000 individual gene knockdowns. We focused on CCAR1, a poorly understood gene that we found the depletion of reduced both HDR and interstrand crosslink repair, phenocopying the loss of the Fanconi anemia pathway. CCAR1 loss abrogated FANCA protein without substantial reduction in the level of its mRNA or that of other FA genes. We instead found that CCAR1 prevents inclusion of a poison exon in FANCA. Transcriptomic analysis revealed that the CCAR1 splicing modulatory activity is not limited to FANCA, and it instead regulates widespread changes in alternative splicing that would damage coding sequences in mouse and human cells. CCAR1 therefore has an unanticipated function as a splicing fidelity factor.


Subject(s)
Alternative Splicing , Fanconi Anemia Complementation Group A Protein , Humans , Animals , Mice , Fanconi Anemia Complementation Group A Protein/genetics , Fanconi Anemia Complementation Group A Protein/metabolism , Recombinational DNA Repair , Fanconi Anemia/genetics , Fanconi Anemia/metabolism , HEK293 Cells , Exons , CRISPR-Cas Systems , DNA Repair , HeLa Cells , DNA Damage
8.
Mol Cell ; 84(14): 2618-2633.e10, 2024 Jul 25.
Article in English | MEDLINE | ID: mdl-39025073

ABSTRACT

The twenty-three Fanconi anemia (FA) proteins cooperate in the FA/BRCA pathway to repair DNA interstrand cross-links (ICLs). The cell division cycle and apoptosis regulator 1 (CCAR1) protein is also a regulator of ICL repair, though its possible function in the FA/BRCA pathway remains unknown. Here, we demonstrate that CCAR1 plays a unique upstream role in the FA/BRCA pathway and is required for FANCA protein expression in human cells. Interestingly, CCAR1 co-immunoprecipitates with FANCA pre-mRNA and is required for FANCA mRNA processing. Loss of CCAR1 results in retention of a poison exon in the FANCA transcript, thereby leading to reduced FANCA protein expression. A unique domain of CCAR1, the EF hand domain, is required for interaction with the U2AF heterodimer of the spliceosome and for excision of the poison exon. Taken together, CCAR1 is a splicing modulator required for normal splicing of the FANCA mRNA and other mRNAs involved in various cellular pathways.


Subject(s)
Apoptosis Regulatory Proteins , Cell Cycle Proteins , Fanconi Anemia Complementation Group A Protein , Fanconi Anemia , RNA Splicing , Splicing Factor U2AF , Humans , BRCA1 Protein/metabolism , BRCA1 Protein/genetics , BRCA2 Protein/metabolism , BRCA2 Protein/genetics , DNA Repair , Endodeoxyribonucleases , Exons , Fanconi Anemia/genetics , Fanconi Anemia/metabolism , Fanconi Anemia Complementation Group A Protein/genetics , Fanconi Anemia Complementation Group A Protein/metabolism , HEK293 Cells , HeLa Cells , Protein Binding , RNA Precursors/metabolism , RNA Precursors/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , Signal Transduction , Spliceosomes/metabolism , Spliceosomes/genetics , Splicing Factor U2AF/metabolism , Splicing Factor U2AF/genetics , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Apoptosis Regulatory Proteins/genetics , Apoptosis Regulatory Proteins/metabolism
9.
Mol Cell ; 84(13): 2573-2589.e5, 2024 Jul 11.
Article in English | MEDLINE | ID: mdl-38917795

ABSTRACT

Efficient targeted control of splicing is a major goal of functional genomics and therapeutic applications. Guide (g)RNA-directed, deactivated (d)Cas CRISPR enzymes fused to splicing effectors represent a promising strategy due to the flexibility of these systems. However, efficient, specific, and generalizable activation of endogenous exons using this approach has not been previously reported. By screening over 300 dCasRx-splicing factor fusion proteins tethered to splicing reporters, we identify dCasRx-RBM25 as a potent activator of exons. Moreover, dCasRx-RBM25 efficiently activates the splicing of ∼90% of targeted endogenous alternative exons and displays high on-target specificity. Using gRNA arrays for combinatorial targeting, we demonstrate that dCasRx-RBM25 enables multiplexed activation and repression of exons. Using this feature, the targeting of neural-regulated exons in Ptpb1 and Puf60 in embryonic stem cells reveals combinatorial effects on downstream alternative splicing events controlled by these factors. Collectively, our results enable versatile, combinatorial exon-resolution functional assays and splicing-directed therapeutic applications.


Subject(s)
Alternative Splicing , CRISPR-Cas Systems , Exons , RNA Splicing Factors , RNA-Binding Proteins , Humans , RNA-Binding Proteins/genetics , RNA-Binding Proteins/metabolism , HEK293 Cells , RNA Splicing Factors/genetics , RNA Splicing Factors/metabolism , RNA, Guide, CRISPR-Cas Systems/genetics , RNA, Guide, CRISPR-Cas Systems/metabolism , Animals , Mice
10.
Mol Cell ; 83(2): 237-251.e7, 2023 Jan 19.
Article in English | MEDLINE | ID: mdl-36599352

ABSTRACT

N6-methyladenosine (m6A), a widespread destabilizing mark on mRNA, is non-uniformly distributed across the transcriptome, yet the basis for its selective deposition is unknown. Here, we propose that m6A deposition is not selective. Instead, it is exclusion based: m6A consensus motifs are methylated by default, unless they are within a window of ∼100 nt from a splice junction. A simple model which we extensively validate, relying exclusively on presence of m6A motifs and exon-intron architecture, allows in silico recapitulation of experimentally measured m6A profiles. We provide evidence that exclusion from splice junctions is mediated by the exon junction complex (EJC), potentially via physical occlusion, and that previously observed associations between exon-intron architecture and mRNA decay are mechanistically mediated via m6A. Our findings establish a mechanism coupling nuclear mRNA splicing and packaging with the covalent installation of m6A, in turn controlling cytoplasmic decay.


Subject(s)
RNA Splicing , Transcriptome , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA Stability , Exons/genetics
11.
Mol Cell ; 83(24): 4479-4493.e6, 2023 Dec 21.
Article in English | MEDLINE | ID: mdl-38096826

ABSTRACT

4.5SH RNA is a highly abundant, small rodent-specific noncoding RNA that localizes to nuclear speckles enriched in pre-mRNA-splicing regulators. To investigate the physiological functions of 4.5SH RNA, we have created mutant mice that lack the expression of 4.5SH RNA. The mutant mice exhibited embryonic lethality, suggesting that 4.5SH RNA is an essential species-specific noncoding RNA in mice. RNA-sequencing analyses revealed that 4.5SH RNA protects the transcriptome from abnormal exonizations of the antisense insertions of the retrotransposon SINE B1 (asB1), which would otherwise introduce deleterious premature stop codons or frameshift mutations. Mechanistically, 4.5SH RNA base pairs with complementary asB1-containing exons via the target recognition region and recruits effector proteins including Hnrnpm via its 5' stem loop region. The modular organization of 4.5SH RNA allows us to engineer a programmable splicing regulator to induce the skipping of target exons of interest. Our results also suggest the general existence of splicing regulatory noncoding RNAs.


Subject(s)
RNA Splicing , RNA, Small Untranslated , Mice , Animals , RNA Splicing/genetics , Exons/genetics , Retroelements/genetics , Codon, Nonsense , Alternative Splicing
12.
Mol Cell ; 83(21): 3801-3817.e8, 2023 Nov 02.
Article in English | MEDLINE | ID: mdl-37922872

ABSTRACT

Histones shape chromatin structure and the epigenetic landscape. H1, the most diverse histone in the human genome, has 11 variants. Due to the high structural similarity between the H1s, their unique functions in transferring information from the chromatin to mRNA-processing machineries have remained elusive. Here, we generated human cell lines lacking up to five H1 subtypes, allowing us to characterize the genomic binding profiles of six H1 variants. Most H1s bind to specific sites, and binding depends on multiple factors, including GC content. The highly expressed H1.2 has a high affinity for exons, whereas H1.3 binds intronic sequences. H1s are major splicing regulators, especially of exon skipping and intron retention events, through their effects on the elongation of RNA polymerase II (RNAPII). Thus, H1 variants determine splicing fate by modulating RNAPII elongation.


Subject(s)
Histones , RNA Polymerase II , Humans , Histones/genetics , Histones/metabolism , RNA Polymerase II/genetics , RNA Polymerase II/metabolism , RNA Splicing , Transcription, Genetic , Chromatin/genetics , Alternative Splicing
13.
Mol Cell ; 83(15): 2653-2672.e15, 2023 08 03.
Article in English | MEDLINE | ID: mdl-37506698

ABSTRACT

Splicing of pre-mRNAs critically contributes to gene regulation and proteome expansion in eukaryotes, but our understanding of the recognition and pairing of splice sites during spliceosome assembly lacks detail. Here, we identify the multidomain RNA-binding protein FUBP1 as a key splicing factor that binds to a hitherto unknown cis-regulatory motif. By collecting NMR, structural, and in vivo interaction data, we demonstrate that FUBP1 stabilizes U2AF2 and SF1, key components at the 3' splice site, through multivalent binding interfaces located within its disordered regions. Transcriptional profiling and kinetic modeling reveal that FUBP1 is required for efficient splicing of long introns, which is impaired in cancer patients harboring FUBP1 mutations. Notably, FUBP1 interacts with numerous U1 snRNP-associated proteins, suggesting a unique role for FUBP1 in splice site bridging for long introns. We propose a compelling model for 3' splice site recognition of long introns, which represent 80% of all human introns.


Subject(s)
RNA Splice Sites , RNA Splicing , Humans , RNA Splice Sites/genetics , Introns/genetics , RNA Splicing Factors/genetics , RNA Splicing Factors/metabolism , RNA-Binding Proteins/genetics , RNA-Binding Proteins/metabolism , RNA Precursors/genetics , RNA Precursors/metabolism , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism
14.
Mol Cell ; 83(24): 4509-4523.e11, 2023 Dec 21.
Article in English | MEDLINE | ID: mdl-38134885

ABSTRACT

The cytoplasm is highly compartmentalized, but the extent and consequences of subcytoplasmic mRNA localization in non-polarized cells are largely unknown. We determined mRNA enrichment in TIS granules (TGs) and the rough endoplasmic reticulum (ER) through particle sorting and isolated cytosolic mRNAs by digitonin extraction. When focusing on genes that encode non-membrane proteins, we observed that 52% have transcripts enriched in specific compartments. Compartment enrichment correlates with a combinatorial code based on mRNA length, exon length, and 3' UTR-bound RNA-binding proteins. Compartment-biased mRNAs differ in the functional classes of their encoded proteins: TG-enriched mRNAs encode low-abundance proteins with strong enrichment of transcription factors, whereas ER-enriched mRNAs encode large and highly expressed proteins. Compartment localization is an important determinant of mRNA and protein abundance, which is supported by reporter experiments showing that redirecting cytosolic mRNAs to the ER increases their protein expression. In summary, the cytoplasm is functionally compartmentalized by local translation environments.


Subject(s)
Endoplasmic Reticulum , Proteins , Endoplasmic Reticulum/genetics , Endoplasmic Reticulum/metabolism , Proteins/metabolism , Cytosol/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Protein Transport , Protein Biosynthesis
15.
Annu Rev Biochem ; 84: 291-323, 2015.
Article in English | MEDLINE | ID: mdl-25784052

ABSTRACT

Precursor messenger RNA (pre-mRNA) splicing is a critical step in the posttranscriptional regulation of gene expression, providing significant expansion of the functional proteome of eukaryotic organisms with limited gene numbers. Split eukaryotic genes contain intervening sequences or introns disrupting protein-coding exons, and intron removal occurs by repeated assembly of a large and highly dynamic ribonucleoprotein complex termed the spliceosome, which is composed of five small nuclear ribonucleoprotein particles, U1, U2, U4/U6, and U5. Biochemical studies over the past 10 years have allowed the isolation as well as compositional, functional, and structural analysis of splicing complexes at distinct stages along the spliceosome cycle. The average human gene contains eight exons and seven introns, producing an average of three or more alternatively spliced mRNA isoforms. Recent high-throughput sequencing studies indicate that 100% of human genes produce at least two alternative mRNA isoforms. Mechanisms of alternative splicing include RNA-protein interactions of splicing factors with regulatory sites termed silencers or enhancers, RNA-RNA base-pairing interactions, or chromatin-based effects that can change or determine splicing patterns. Disease-causing mutations can often occur in splice sites near intron borders or in exonic or intronic RNA regulatory silencer or enhancer elements, as well as in genes that encode splicing factors. Together, these studies provide mechanistic insights into how spliceosome assembly, dynamics, and catalysis occur; how alternative splicing is regulated and evolves; and how splicing can be disrupted by cis- and trans-acting mutations leading to disease states. These findings make the spliceosome an attractive new target for small-molecule, antisense, and genome-editing therapeutic interventions.


Subject(s)
Alternative Splicing , Gene Expression Regulation , RNA Precursors/genetics , Animals , Disease/genetics , Humans , Mutation , RNA Splicing , RNA, Catalytic/metabolism , Ribonucleoproteins, Small Nuclear/metabolism , Spliceosomes/chemistry , Spliceosomes/drug effects
16.
Annu Rev Genet ; 56: 315-337, 2022 11 30.
Article in English | MEDLINE | ID: mdl-36055647

ABSTRACT

Animal species present relatively high levels of gene conservation, and yet they display a great variety of cell type and tissue phenotypes. These diverse phenotypes are mainly specified through differential gene usage, which relies on several mechanisms. Two of the most relevant mechanisms are regulated gene transcription, usually referred to as gene expression (rGE), and regulated alternative splicing (rAS). Several works have addressed how either rGE or rAS contributes to phenotypic diversity throughout evolution, but a back-to-back comparison between the two molecular mechanisms, specifically highlighting both their common regulatory principles and unique properties, is still missing. In this review, we propose an innovative framework for the unified comparison between rGE and rAS from different perspectives: the three-dimensional (3D)-evo space. We use the 3D-evo space to comprehensively (a) review the molecular basis of rGE and rAS (i.e., the molecular axis), (b) depict the tissue-specific phenotypes they contribute to (i.e., the tissue axis), and (c) describe the determinants that drive the evolution of rGE and rAS programs (i.e., the evolution axis). Finally, we unify the perspectives emerging from the three axes by discussing general trends and specific examples of rGE and rAS tissue program evolution.


Subject(s)
Alternative Splicing , Animals , Alternative Splicing/genetics , Phenotype , Gene Expression
17.
Mol Cell ; 82(5): 1021-1034.e8, 2022 03 03.
Article in English | MEDLINE | ID: mdl-35182478

ABSTRACT

How the splicing machinery defines exons or introns as the spliced unit has remained a puzzle for 30 years. Here, we demonstrate that peripheral and central regions of the nucleus harbor genes with two distinct exon-intron GC content architectures that differ in the splicing outcome. Genes with low GC content exons, flanked by long introns with lower GC content, are localized in the periphery, and the exons are defined as the spliced unit. Alternative splicing of these genes results in exon skipping. In contrast, the nuclear center contains genes with a high GC content in the exons and short flanking introns. Most splicing of these genes occurs via intron definition, and aberrant splicing leads to intron retention. We demonstrate that the nuclear periphery and center generate different environments for the regulation of alternative splicing and that two sets of splicing factors form discrete regulatory subnetworks for the two gene architectures. Our study connects 3D genome organization and splicing, thus demonstrating that exon and intron definition modes of splicing occur in different nuclear regions.


Subject(s)
Alternative Splicing , RNA Splicing , Base Composition , Exons/genetics , Introns/genetics
18.
Mol Cell ; 82(15): 2769-2778.e4, 2022 08 04.
Article in English | MEDLINE | ID: mdl-35705093

ABSTRACT

Pre-mRNA splicing involves two sequential reactions: branching and exon ligation. The C complex after branching undergoes remodeling to become the C∗ complex, which executes exon ligation. Here, we report cryo-EM structures of two intermediate human spliceosomal complexes, pre-C∗-I and pre-C∗-II, both at 3.6 Å. In both structures, the 3' splice site is already docked into the active site, the ensuing 3' exon sequences are anchored on PRP8, and the step II factor FAM192A contacts the duplex between U2 snRNA and the branch site. In the transition of pre-C∗-I to pre-C∗-II, the step II factors Cactin, FAM32A, PRKRIP1, and SLU7 are recruited. Notably, the RNA helicase PRP22 is positioned quite differently in the pre-C∗-I, pre-C∗-II, and C∗ complexes, suggesting a role in 3' exon binding and proofreading. Together with information on human C and C∗ complexes, our studies recapitulate a molecular choreography of the C-to-C∗ transition, revealing mechanistic insights into exon ligation.


Subject(s)
Saccharomyces cerevisiae Proteins , Spliceosomes , Exons/genetics , Humans , RNA Precursors/metabolism , RNA Splice Sites , RNA Splicing , RNA Splicing Factors/genetics , RNA Splicing Factors/metabolism , RNA, Small Nuclear/genetics , Saccharomyces cerevisiae Proteins/metabolism , Spliceosomes/metabolism
19.
Mol Cell ; 82(24): 4681-4699.e8, 2022 12 15.
Article in English | MEDLINE | ID: mdl-36435176

ABSTRACT

Long introns with short exons in vertebrate genes are thought to require spliceosome assembly across exons (exon definition), rather than introns, thereby requiring transcription of an exon to splice an upstream intron. Here, we developed CoLa-seq (co-transcriptional lariat sequencing) to investigate the timing and determinants of co-transcriptional splicing genome wide. Unexpectedly, 90% of all introns, including long introns, can splice before transcription of a downstream exon, indicating that exon definition is not obligatory for most human introns. Still, splicing timing varies dramatically across introns, and various genetic elements determine this variation. Strong U2AF2 binding to the polypyrimidine tract predicts early splicing, explaining exon definition-independent splicing. Together, our findings question the essentiality of exon definition and reveal features beyond intron and exon length that are determinative for splicing timing.


Subject(s)
Alternative Splicing , RNA Splicing , Humans , Base Sequence , Introns/genetics , Exons/genetics
20.
Mol Cell ; 82(15): 2779-2796.e10, 2022 08 04.
Article in English | MEDLINE | ID: mdl-35675814

ABSTRACT

Despite a long appreciation for the role of nonsense-mediated mRNA decay (NMD) in destroying faulty, disease-causing mRNAs and maintaining normal, physiologic mRNA abundance, additional effectors that regulate NMD activity in mammalian cells continue to be identified. Here, we describe a haploid-cell genetic screen for NMD effectors that has unexpectedly identified 13 proteins constituting the AKT signaling pathway. We show that AKT supersedes UPF2 in exon-junction complexes (EJCs) that are devoid of RNPS1 but contain CASC3, defining an unanticipated insulin-stimulated EJC. Without altering UPF1 RNA binding or ATPase activity, AKT-mediated phosphorylation of the UPF1 CH domain at T151 augments UPF1 helicase activity, which is critical for NMD and also decreases the dependence of helicase activity on ATP. We demonstrate that upregulation of AKT signaling contributes to the hyperactivation of NMD that typifies Fragile X syndrome, as exemplified using FMR1-KO neural stem cells derived from induced pluripotent stem cells.


Subject(s)
Nonsense Mediated mRNA Decay , Proto-Oncogene Proteins c-akt , Animals , Codon, Nonsense/genetics , Exons/genetics , Mammals/metabolism , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/metabolism , RNA Helicases/genetics , RNA Helicases/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Trans-Activators/genetics , Trans-Activators/metabolism , Transcription Factors/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL