Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 151
Filter
Add more filters

Country/Region as subject
Publication year range
1.
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
2.
Nat Immunol ; 20(10): 1322-1334, 2019 10.
Article in English | MEDLINE | ID: mdl-31427773

ABSTRACT

We report a new immunodeficiency disorder in mice caused by a viable hypomorphic mutation of Snrnp40, an essential gene encoding a subunit of the U5 small nuclear ribonucleoprotein (snRNP) complex of the spliceosome. Snrnp40 is ubiquitous but strongly expressed in lymphoid tissue. Homozygous mutant mice showed hypersusceptibility to infection by murine cytomegalovirus and multiple defects of lymphoid development, stability and function. Cell-intrinsic defects of hematopoietic stem cell differentiation also affected homozygous mutants. SNRNP40 deficiency in primary hematopoietic stem cells or T cells or the EL4 cell line increased the frequency of splicing errors, mostly intron retention, in several hundred messenger RNAs. Altered expression of proteins associated with immune cell function was also observed in Snrnp40-mutant cells. The immunological consequences of SNRNP40 deficiency presumably result from cumulative, moderate effects on processing of many different mRNA molecules and secondary reductions in the expression of critical immune proteins, yielding a syndromic immune disorder.


Subject(s)
Hematopoietic Stem Cells/physiology , Herpesviridae Infections/immunology , Immunologic Deficiency Syndromes/immunology , Muromegalovirus/physiology , Ribonucleoprotein, U5 Small Nuclear/metabolism , Spliceosomes/metabolism , T-Lymphocytes/physiology , Alleles , Animals , Cell Line , Clustered Regularly Interspaced Short Palindromic Repeats , Disease Susceptibility , Herpesviridae Infections/genetics , Immunologic Deficiency Syndromes/genetics , Lymphopoiesis/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Mutation/genetics , RNA Splicing , Ribonucleoprotein, U5 Small Nuclear/genetics
3.
J Cell Sci ; 136(2)2023 01 15.
Article in English | MEDLINE | ID: mdl-36620952

ABSTRACT

SART3 is a multifunctional protein that acts in several steps of gene expression, including assembly and recycling of the spliceosomal U4/U6 small nuclear ribonucleoprotein particle (snRNP). In this work, we provide evidence that SART3 associates via its N-terminal HAT domain with the 12S U2 snRNP. Further analysis showed that SART3 associates with the post-splicing complex containing U2 and U5 snRNP components. In addition, we observed an interaction between SART3 and the RNA helicase DHX15, which disassembles post-splicing complexes. Based on our data, we propose a model that SART3 associates via its N-terminal HAT domain with the post-splicing complex, where it interacts with U6 snRNA to protect it and to initiate U6 snRNA recycling before a next round of splicing.


Subject(s)
RNA Splicing , Spliceosomes , RNA Splicing/genetics , Spliceosomes/genetics , Spliceosomes/metabolism , RNA, Small Nuclear/genetics , RNA, Small Nuclear/metabolism , Ribonucleoprotein, U4-U6 Small Nuclear/genetics , Ribonucleoprotein, U4-U6 Small Nuclear/metabolism , Ribonucleoprotein, U5 Small Nuclear/genetics , Ribonucleoprotein, U5 Small Nuclear/metabolism , Ribonucleoprotein, U2 Small Nuclear/genetics , Ribonucleoprotein, U2 Small Nuclear/metabolism , Ribonucleoproteins, Small Nuclear/genetics , Ribonucleoproteins, Small Nuclear/metabolism
4.
Proc Natl Acad Sci U S A ; 119(41): e2208415119, 2022 10 11.
Article in English | MEDLINE | ID: mdl-36191209

ABSTRACT

MicroRNAs (miRNAs) play an essential role in plant growth and development, and as such, their biogenesis is fine-tuned via regulation of the core microprocessor components. Here, we report that Arabidopsis AAR2, a homolog of a U5 snRNP assembly factor in yeast and humans, not only acts in splicing but also promotes miRNA biogenesis. AAR2 interacts with the microprocessor component hyponastic leaves 1 (HYL1) in the cytoplasm, nucleus, and dicing bodies. In aar2 mutants, abundance of nonphosphorylated HYL1, the active form of HYL1, and the number of HYL1-labeled dicing bodies are reduced. Primary miRNA (pri-miRNA) accumulation is compromised despite normal promoter activities of MIR genes in aar2 mutants. RNA decay assays show that the aar2-1 mutation leads to faster degradation of pri-miRNAs in a HYL1-dependent manner, which reveals a previously unknown and negative role of HYL1 in miRNA biogenesis. Taken together, our findings reveal a dual role of AAR2 in miRNA biogenesis and pre-messenger RNA splicing.


Subject(s)
Arabidopsis Proteins , Arabidopsis , MicroRNAs , Arabidopsis/genetics , Arabidopsis/metabolism , Arabidopsis Proteins/genetics , Arabidopsis Proteins/metabolism , Eukaryota/genetics , Gene Expression Regulation, Plant , Humans , MicroRNAs/genetics , MicroRNAs/metabolism , RNA Processing, Post-Transcriptional , RNA Splicing Factors/genetics , RNA, Messenger/metabolism , RNA-Binding Proteins/genetics , RNA-Binding Proteins/metabolism , Ribonucleoprotein, U5 Small Nuclear/genetics
5.
Nucleic Acids Res ; 50(20): 11834-11857, 2022 11 11.
Article in English | MEDLINE | ID: mdl-36321655

ABSTRACT

The spliceosome undergoes extensive rearrangements as it assembles onto precursor messenger RNAs. In the earliest assembly step, U1snRNA identifies the 5' splice site. However, U1snRNA leaves the spliceosome relatively early in assembly, and 5' splice site identity is subsequently maintained through interactions with U6snRNA, protein factor PRP8, and other components during the rearrangements that build the catalytic site. Using a forward genetic screen in Caenorhabditis elegans, we have identified suppressors of a locomotion defect caused by a 5'ss mutation. Here we report three new suppressor alleles from this screen, two in PRP8 and one in SNRNP200/BRR2. mRNASeq studies of these suppressor strains indicate that they also affect specific native alternative 5'ss, especially for suppressor PRP8 D1549N. A strong suppressor at the unstructured N-terminus of SNRNP200, N18K, indicates a novel role for this region. By examining distinct changes in the splicing of native genes, examining double mutants between suppressors, comparing these new suppressors to previously identified splicing suppressors from yeast, and mapping conserved suppressor residues onto cryoEM structural models of assembling human spliceosomes, we conclude that there are multiple interactions at multiple stages in spliceosome assembly responsible for maintaining the initial 5'ss identified by U1snRNA for entry into the catalytic core.


Subject(s)
RNA Splice Sites , RNA Splicing Factors , Spliceosomes , Animals , Humans , Caenorhabditis elegans/genetics , Caenorhabditis elegans/metabolism , Caenorhabditis elegans Proteins/genetics , Caenorhabditis elegans Proteins/metabolism , Mutation , Ribonucleoprotein, U5 Small Nuclear/genetics , Ribonucleoprotein, U5 Small Nuclear/metabolism , Ribonucleoproteins, Small Nuclear/genetics , Ribonucleoproteins, Small Nuclear/metabolism , RNA Helicases/genetics , RNA Helicases/metabolism , RNA Precursors/genetics , RNA Precursors/metabolism , RNA Splicing , RNA-Binding Proteins/metabolism , Saccharomyces cerevisiae/genetics , Spliceosomes/genetics , Spliceosomes/metabolism , RNA Splicing Factors/genetics
6.
Hum Mol Genet ; 30(9): 739-757, 2021 05 28.
Article in English | MEDLINE | ID: mdl-33601405

ABSTRACT

EFTUD2 is mutated in patients with mandibulofacial dysostosis with microcephaly (MFDM). We generated a mutant mouse line with conditional mutation in Eftud2 and used Wnt1-Cre2 to delete it in neural crest cells. Homozygous deletion of Eftud2 causes brain and craniofacial malformations, affecting the same precursors as in MFDM patients. RNAseq analysis of embryonic heads revealed a significant increase in exon skipping and increased levels of an alternatively spliced Mdm2 transcript lacking exon 3. Exon skipping in Mdm2 was also increased in O9-1 mouse neural crest cells after siRNA knock-down of Eftud2 and in MFDM patient cells. Moreover, we found increased nuclear P53, higher expression of P53-target genes and increased cell death. Finally, overactivation of the P53 pathway in Eftud2 knockdown cells was attenuated by overexpression of non-spliced Mdm2, and craniofacial development was improved when Eftud2-mutant embryos were treated with Pifithrin-α, an inhibitor of P53. Thus, our work indicates that the P53-pathway can be targeted to prevent craniofacial abnormalities and shows a previously unknown role for alternative splicing of Mdm2 in the etiology of MFDM.


Subject(s)
Ribonucleoprotein, U5 Small Nuclear , Tumor Suppressor Protein p53 , Animals , Homozygote , Humans , Mice , Mutation , Peptide Elongation Factors/genetics , Proto-Oncogene Proteins c-mdm2/genetics , Proto-Oncogene Proteins c-mdm2/metabolism , Ribonucleoprotein, U5 Small Nuclear/genetics , Sequence Deletion , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism
7.
Blood ; 138(21): 2117-2128, 2021 11 25.
Article in English | MEDLINE | ID: mdl-34115847

ABSTRACT

Shwachman-Diamond syndrome (SDS; OMIM #260400) is caused by variants in SBDS (Shwachman-Bodian-Diamond syndrome gene), which encodes a protein that plays an important role in ribosome assembly. Recent reports suggest that recessive variants in EFL1 are also responsible for SDS. However, the precise genetic mechanism that leads to EFL1-induced SDS remains incompletely understood. Here we present 3 unrelated Korean SDS patients who carry biallelic pathogenic variants in EFL1 with biased allele frequencies, resulting from a bone marrow-specific somatic uniparental disomy in chromosome 15. The recombination events generated cells that were homozygous for the relatively milder variant, allowing for the evasion of catastrophic physiologic consequences. However, the milder EFL1 variant was still solely able to impair 80S ribosome assembly and induce SDS features in cell line and animal models. The loss of EFL1 resulted in a pronounced inhibition of terminal oligopyrimidine element-containing ribosomal protein transcript 80S assembly. Therefore, we propose a more accurate pathogenesis mechanism of EFL1 dysfunction that eventually leads to aberrant translational control and ribosomopathy.


Subject(s)
Peptide Elongation Factors/genetics , Ribonucleoprotein, U5 Small Nuclear/genetics , Shwachman-Diamond Syndrome/genetics , Uniparental Disomy/genetics , Adult , Alleles , Animals , Child , Child, Preschool , Female , Humans , Male , Mice, Inbred C57BL , Models, Molecular , Point Mutation
8.
Clin Genet ; 101(2): 255-259, 2022 02.
Article in English | MEDLINE | ID: mdl-34713892

ABSTRACT

The developmental disorder Burn-McKeown Syndrome (BMKS) is characterised by choanal atresia and specific craniofacial features. BMKS is caused by biallelic variants in the pre-messenger RNA splicing factor TXNL4A. Most patients have a loss-of-function variant in trans with a 34-base pair (bp) deletion (type 1 Δ34) in the promoter region. Here, we identified two patients with BMKS. One individual has a TXNL4A c.93_94delCC, p.His32Argfs *21 variant combined with a type 1 Δ34 promoter deletion. The other has an intronic TXNL4A splice site variant (c.258-3C>G) and a type 1 Δ34 promoter deletion. We show the c.258-3C>G variant and a previously reported c.258-2A>G variant, cause skipping of the final exon of TXNL4A in a minigene splicing assay. Furthermore, we identify putative transcription factor binding sites within the 56 bp of the TXNL4A promoter affected by the type 1 and type 2 Δ34 and use dual luciferase assays to identify a 22 bp repeated motif essential for TXNL4A expression within this promoter region. We propose that additional variants affecting critical transcription factor binding nucleotides within the 22 bp repeated motif could be relevant to BMKS aetiology. Finally, our data emphasises the need to analyse the non-coding sequence in individuals where a single likely pathogenic coding variant is identified in an autosomal recessive disorder consistent with the clinical presentation.


Subject(s)
Choanal Atresia/diagnosis , Choanal Atresia/genetics , Deafness/congenital , Genotype , Heart Defects, Congenital/diagnosis , Heart Defects, Congenital/genetics , Mutation , Ribonucleoprotein, U5 Small Nuclear/genetics , Alleles , Binding Sites , Deafness/diagnosis , Deafness/genetics , Facies , Female , Genetic Association Studies , Genetic Predisposition to Disease , Humans , Pedigree , Phenotype , Promoter Regions, Genetic , Protein Binding , RNA Splicing , Ribonucleoprotein, U5 Small Nuclear/metabolism , Transcription Factors/metabolism
9.
Immunol Invest ; 51(5): 1485-1497, 2022 Jul.
Article in English | MEDLINE | ID: mdl-34436958

ABSTRACT

BACKGROUND: The elongation factor Tu GTP-binding domain-containing 2 gene (EFTUD2) participates in antiviral immune responses. However, the association between genetic polymorphisms of EFTUD2 and hepatitis B virus (HBV) infection susceptibility has not been well-studied. We analyzed the relationship between single nucleotide polymorphisms (SNPs) of EFTUD2 and HBV infection susceptibility and clarified the potential function. METHODS: In total, 448 control subjects and 379 patients with chronic HBV infection from Zhangjiagang First People's Hospital (Jiangsu, China) were enrolled. Sequenom iPLEX assay was used to detect genotypes of four SNPs (rs1071682, rs2277617, rs2289674, and rs3809756). Dual-luciferase reporter vectors with wild-type A and mutant-type C alleles of EFTUD2 rs3809756 were transfected into HepG2 cells to explore effects on transcription activity. RESULTS: Only rs3809756 was significantly associated with HBV infection susceptibility (P < .05). The risk of HBV infection was higher in individuals carrying the rs3809756-CC genotype than in those carrying the rs3809756-AA genotype (odds ratio [OR] = 1.945, 95% confidence interval [CI] = 1.129-3.351, P = .017). Subgroup analysis based on the dominant model revealed that rs3809756-AC and rs3809756-CC carriers had a significantly higher risk of HBV infection than rs3809756-AA carriers among patients who were male (OR = 1.732, 95% CI = 1.218-2.464, P = .002), were aged ≥47 years (OR = 1.502, 95% CI = 1.050-2.148, P = .026), or without liver cirrhosis (OR = 1.407, 95% CI = 1.077-1.838, P = .012). In the dual-luciferase reporter assay, the relative luciferase activity of rs3809756-C was significantly lower than that of rs3809756-A (P < .05). CONCLUSION: EFTUD2 rs3809756A>C was associated with HBV infection susceptibility and might be involved in the downregulation of promoter activity.


Subject(s)
Hepatitis B, Chronic , Hepatitis B , Alleles , Case-Control Studies , Female , Gene Frequency , Genetic Predisposition to Disease , Genotype , Guanosine Triphosphate , Hepatitis B/genetics , Hepatitis B virus , Hepatitis B, Chronic/complications , Hepatitis B, Chronic/genetics , Humans , Male , Peptide Elongation Factor Tu/genetics , Peptide Elongation Factors/genetics , Polymorphism, Single Nucleotide , Ribonucleoprotein, U5 Small Nuclear/genetics
10.
Mol Cell ; 56(2): 298-310, 2014 Oct 23.
Article in English | MEDLINE | ID: mdl-25263594

ABSTRACT

BS69 (also called ZMYND11) contains tandemly arranged PHD, BROMO, and PWWP domains, which are chromatin recognition modalities. Here, we show that BS69 selectively recognizes histone variant H3.3 lysine 36 trimethylation (H3.3K36me3) via its chromatin-binding domains. We further identify BS69 association with RNA splicing regulators, including the U5 snRNP components of the spliceosome, such as EFTUD2. Remarkably, RNA sequencing shows that BS69 mainly regulates intron retention (IR), which is the least understood RNA alternative splicing event in mammalian cells. Biochemical and genetic experiments demonstrate that BS69 promotes IR by antagonizing EFTUD2 through physical interactions. We further show that regulation of IR by BS69 also depends on its binding to H3K36me3-decorated chromatin. Taken together, our study identifies an H3.3K36me3-specific reader and a regulator of IR and reveals that BS69 connects histone H3.3K36me3 to regulated RNA splicing, providing significant, important insights into chromatin regulation of pre-mRNA processing.


Subject(s)
Alternative Splicing , Carrier Proteins/metabolism , Chromatin/metabolism , Histones/metabolism , RNA Precursors/genetics , RNA, Messenger/genetics , Base Sequence , Carrier Proteins/genetics , Cell Cycle Proteins , Cell Line, Tumor , Chromatin/genetics , Co-Repressor Proteins , DNA Methylation/genetics , DNA-Binding Proteins , HeLa Cells , Histone-Lysine N-Methyltransferase/genetics , Histone-Lysine N-Methyltransferase/metabolism , Histones/genetics , Humans , Introns/genetics , Lysine/genetics , Lysine/metabolism , Peptide Elongation Factors/antagonists & inhibitors , Peptide Elongation Factors/genetics , Peptide Elongation Factors/metabolism , Protein Binding , Protein Structure, Tertiary , RNA Interference , RNA Processing, Post-Transcriptional/genetics , RNA, Small Interfering , Ribonucleoprotein, U5 Small Nuclear/antagonists & inhibitors , Ribonucleoprotein, U5 Small Nuclear/genetics , Ribonucleoprotein, U5 Small Nuclear/metabolism , Sequence Analysis, RNA , Spliceosomes/genetics
11.
J Clin Lab Anal ; 36(5): e24440, 2022 May.
Article in English | MEDLINE | ID: mdl-35435265

ABSTRACT

BACKGROUND: Mandibulofacial dysostosis with microcephaly (MFDM) is a rare multiple malformation syndrome characterized by malar and mandibular hypoplasia and congenital- or postnatal-onset microcephaly induced by haploinsufficiency of (elongation factor Tu GTP-binding domain-containing 2) EFTUD2. METHODS: We report the case of a 16-month-old boy with MFDM symptoms, including malar and mandibular hypoplasia, microcephaly, micrognathia, midline cleft palate, microtia, auditory canal atresia, severe sensorineural hearing loss, and developmental delay. Whole-exome sequencing (WES) analysis of the patient's family was performed to identify the genetic etiology responsible for this phenotype. RESULTS: We identified a novel de novo missense mutation (c.671G>T, p.Gly224Val) in the EFTUD2. According to the American College of Medical Genetics and Genomics (ACMG) 2015 guidelines, the c.671G>T mutation was classified as likely pathogenic (PS2, PM1, PM2, and PP3). Based on our findings, prenatal diagnosis was performed on the second baby of the proband's parents to exclude the mutation and it was confirmed that the baby did not have the MFDM phenotype after 14 months of follow-up. Furthermore, the zebrafish model confirmed that the EFTUD2 c.671G>T mutation caused a loss of gene function in EFTUD2, and the pathogenicity of the EFTUD2 c.671G>T mutation was classified as pathogenic (PS2, PS3, PM1, and PM2). CONCLUSION: Our results indicate that WES is a useful tool for identifying potentially pathogenic mutations, particularly in rare disorders, and is advantageous for genetic counseling and subsequent prenatal diagnosis. Moreover, the importance of functional assays cannot be underestimated, which could further confirm the pathogenicity of the genetic variants.


Subject(s)
Abnormalities, Multiple , Mandibulofacial Dysostosis , Microcephaly , Abnormalities, Multiple/genetics , Animals , Humans , Mandibulofacial Dysostosis/diagnostic imaging , Mandibulofacial Dysostosis/genetics , Microcephaly/diagnostic imaging , Microcephaly/genetics , Mutation , Mutation, Missense/genetics , Peptide Elongation Factors/genetics , Phenotype , Ribonucleoprotein, U5 Small Nuclear/genetics , Exome Sequencing , Zebrafish/genetics
12.
Proc Natl Acad Sci U S A ; 116(6): 2193-2199, 2019 02 05.
Article in English | MEDLINE | ID: mdl-30674666

ABSTRACT

Pre-mRNA splicing must occur with extremely high fidelity. Spliceosomes assemble onto pre-mRNA guided by specific sequences (5' splice site, 3' splice site, and branchpoint). When splice sites are mutated, as in many hereditary diseases, the spliceosome can aberrantly select nearby pseudo- or "cryptic" splice sites, often resulting in nonfunctional protein. How the spliceosome distinguishes authentic splice sites from cryptic splice sites is poorly understood. We performed a Caenorhabditis elegans genetic screen to find cellular factors that affect the frequency with which the spliceosome uses cryptic splice sites and identified two alleles in core spliceosome component Prp8 that alter cryptic splicing frequency. Subsequent complementary genetic and structural analyses in yeast implicate these alleles in the stability of the spliceosome's catalytic core. However, despite a clear effect on cryptic splicing, high-throughput mRNA sequencing of these prp-8 mutant C. elegans reveals that overall alternative splicing patterns are relatively unchanged. Our data suggest the spliceosome evolved intrinsic mechanisms to reduce the occurrence of cryptic splicing and that these mechanisms are distinct from those that impact alternative splicing.


Subject(s)
Alternative Splicing , RNA Splice Sites , Ribonucleoprotein, U4-U6 Small Nuclear/genetics , Ribonucleoprotein, U5 Small Nuclear/genetics , Saccharomyces cerevisiae Proteins/genetics , Alleles , Amino Acid Sequence , Amino Acid Substitution , Amino Acids , Animals , Caenorhabditis elegans , Conserved Sequence , Gene Frequency , Genetic Loci , Models, Molecular , Protein Conformation , RNA Precursors , Ribonucleoprotein, U4-U6 Small Nuclear/chemistry , Ribonucleoprotein, U5 Small Nuclear/chemistry , Saccharomyces cerevisiae Proteins/chemistry , Spliceosomes
13.
Int J Mol Sci ; 24(1)2022 Dec 31.
Article in English | MEDLINE | ID: mdl-36614170

ABSTRACT

Mammalian orthoreovirus (MRV) is a double-stranded RNA virus from the Reoviridae family that infects a large range of mammals, including humans. Recently, studies have shown that MRV alters cellular alternative splicing (AS) during viral infection. The structural protein µ2 appears to be the main determinant of these AS modifications by decreasing the levels of U5 core components EFTUD2, PRPF8, and SNRNP200 during infection. In the present study, we investigated the mechanism by which µ2 exerts this effect on the U5 components. Our results revealed that µ2 has no impact on steady-state mRNA levels, RNA export, and protein stability of these U5 snRNP proteins. However, polysome profiling and metabolic labeling of newly synthesized proteins revealed that µ2 exerts an inhibitory effect on global translation. Moreover, we showed that µ2 mutants unable to accumulate in the nucleus retain most of the ability to reduce PRPF8 protein levels, indicating that the effect of µ2 on U5 snRNP components mainly occurs in the cytoplasm. Finally, co-expression experiments demonstrated that µ2 suppresses the expression of U5 snRNP proteins in a dose-dependent manner, and that the expression of specific U5 snRNP core components have different sensitivities to µ2's presence. Altogether, these results suggest a novel mechanism by which the µ2 protein reduces the levels of U5 core components through translation inhibition, allowing this viral protein to alter cellular AS during infection.


Subject(s)
Ribonucleoprotein, U5 Small Nuclear , Spliceosomes , Alternative Splicing , Peptide Elongation Factors/metabolism , Ribonucleoprotein, U5 Small Nuclear/genetics , RNA Splicing , RNA, Messenger/genetics , Spliceosomes/metabolism , Viral Proteins/metabolism
14.
Int J Mol Sci ; 23(16)2022 Aug 12.
Article in English | MEDLINE | ID: mdl-36012294

ABSTRACT

Embryos with homozygous mutation of Eftud2 in their neural crest cells (Eftud2ncc-/-) have brain and craniofacial malformations, hyperactivation of the P53-pathway and die before birth. Treatment of Eftud2ncc-/- embryos with pifithrin-α, a P53-inhibitor, partly improved brain and craniofacial development. To uncover if craniofacial malformations and death were indeed due to P53 hyperactivation we generated embryos with homozygous loss of function mutations in both Eftud2 and Trp53 in the neural crest cells. We evaluated the molecular mechanism underlying craniofacial development in pifithrin-α-treated embryos and in Eftud2; Trp53 double homozygous (Eftud2ncc-/-; Trp53ncc-/-) mutant embryos. Eftud2ncc-/- embryos that were treated with pifithrin-α or homozygous mutant for Trp53 in their neural crest cells showed reduced apoptosis in their neural tube and reduced P53-target activity. Furthermore, although the number of SOX10 positive cranial neural crest cells was increased in embryonic day (E) 9.0 Eftud2ncc-/-; Trp53ncc-/- embryos compared to Eftud2ncc-/- mutants, brain and craniofacial development, and survival were not improved in double mutant embryos. Furthermore, mis-splicing of both P53-regulated transcripts, Mdm2 and Foxm1, and a P53-independent transcript, Synj2bp, was increased in the head of Eftud2ncc-/-; Trp53ncc-/- embryos. While levels of Zmat3, a P53- regulated splicing factor, was similar to those of wild-type. Altogether, our data indicate that both P53-regulated and P53-independent pathways contribute to craniofacial malformations and death of Eftud2ncc-/- embryos.


Subject(s)
Craniofacial Abnormalities , Neural Crest , Peptide Elongation Factors , Ribonucleoprotein, U5 Small Nuclear , Animals , Craniofacial Abnormalities/genetics , Craniofacial Abnormalities/metabolism , Gene Deletion , Homozygote , Neural Crest/metabolism , Peptide Elongation Factors/metabolism , Ribonucleoprotein, U5 Small Nuclear/genetics , Ribonucleoprotein, U5 Small Nuclear/metabolism , Tumor Suppressor Protein p53/genetics
15.
Cleft Palate Craniofac J ; 59(11): 1346-1351, 2022 11.
Article in English | MEDLINE | ID: mdl-34714179

ABSTRACT

OBJECTIVE: Facial dysostosis is a group of rare craniofacial congenital disabilities requiring multidisciplinary long-term care. This report presents the phenotypic and genotypic information from South India. DESIGN: The study is a case series. SETTING: This was an international collaborative study involving a tertiary craniofacial clinic and medical genetics unit. PATIENTS, PARTICIPANTS: The participants were 9 families with 17 affected individuals of facial dysostosis. INTERVENTION: Exome analysis focused on known genes associated with acrofacial and mandibulofacial syndromes. MAIN OUTCOME MEASURE: The outcome measure was to report phenotyptic and genetic heterogeneity in affected individuals. RESULTS: A Tessier cleft was seen in 7 (41%), lower eyelid coloboma in 12 (65%), ear anomalies in 10 (59%), uniolateral or bilateral aural atresia in 4 (24%), and deafness in 6 (35%). The facial gestalt of Treacher Collins syndrome (TCS) showed extensive phenotypic variations. Pathogenic variants in TCOF1 (Treacher Collins syndrome) were seen in six families, POLR1A (acrofacial dysostosis, Cincinnati type) and EFTUD2 (mandibulofacial dysostosis with microcephaly) in one each. One family (11.1%) had no detectable variation. Five out of six probands with Treacher Collins syndrome had other affected family members (83.3%), including a non-penetrant mother, identified after sequencing. CONCLUSION: Our report illustrates the molecular heterogeneity of mandibulofacial dysostosis in India.


Subject(s)
Mandibulofacial Dysostosis , Microcephaly , Face , Genotype , Humans , Mandibulofacial Dysostosis/genetics , Microcephaly/genetics , Peptide Elongation Factors/genetics , Ribonucleoprotein, U5 Small Nuclear/genetics , Syndrome
16.
Cleft Palate Craniofac J ; 59(9): 1114-1124, 2022 09.
Article in English | MEDLINE | ID: mdl-34410171

ABSTRACT

OBJECTIVE: Oculoauriculovertebral spectrum (OAVS) is a genetically and clinically heterogeneous disorder that occurs due to a developmental field defect of the first and second pharyngeal arches. Even though recent whole exome sequencing studies (WES) have led to identification of several genes associated with this spectrum in a subset of individuals, complete pathogenesis of OAVS remains unsolved. In this study, molecular genetic etiology of OAVS was systematically investigated. DESIGN/SETTING/PATIENTS: A cohort of 23 Turkish patients with OAVS, referred to Hacettepe University Hospital, Department of Pediatric Genetics from 2008 to 2018, was included in this study. Minimal diagnostic criteria for OAVS were considered as unilateral microtia or hemifacial microsomia with preauricular skin tag. The cohort was clinically reevaluated for craniofacial and extracranial findings. Molecular etiology was investigated using candidate gene sequencing following copy number variant (CNV) analysis. WES was also performed for 2 of the selected patients. RESULTS: Patients in the study cohort presented similar demographic and phenotypic characteristics to previously described patients in the literature except for a higher frequency of bilaterality, cardiac findings, and intellectual disability/developmental delay. CNV analysis revealed a possible genetic etiology for 3 patients (13%). Additional WES in 1 of the 2 patients uncovered a novel heterozygous nonsense variant in Elongation factor Tu GTP-binding domain-containing 2 (EFTUD2) causing mandibulofacial dysostosis with microcephaly (MFDM), which clinically overlaps with OAVS. CONCLUSION: Detailed clinical evaluation for any patient with OAVS is recommended due to a high rate of accompanying systemic findings. We further expand the existing genetic heterogeneity of OAVS by identifying several CNVs and a phenotypically overlapping disorder, MFDM.


Subject(s)
Goldenhar Syndrome , Mandibulofacial Dysostosis , Microcephaly , Child , Cohort Studies , Developmental Disabilities/genetics , Goldenhar Syndrome/genetics , Humans , Mandibulofacial Dysostosis/genetics , Microcephaly/genetics , Peptide Elongation Factors/genetics , Ribonucleoprotein, U5 Small Nuclear/genetics
17.
J Biol Chem ; 295(13): 4093-4100, 2020 03 27.
Article in English | MEDLINE | ID: mdl-32041777

ABSTRACT

Renpenning syndrome belongs to a group of X-linked intellectual disability disorders. The Renpenning syndrome-associated protein PQBP1 (polyglutamine-binding protein 1) is intrinsically disordered, associates with several splicing factors, and is involved in pre-mRNA splicing. PQBP1 uses its C-terminal YxxPxxVL motif for binding to the splicing factor TXNL4A (thioredoxin like 4A), but the biological function of this interaction has yet to be elucidated. In this study, using recombinant protein expression, in vitro binding assays, and immunofluorescence microscopy in HeLa cells, we found that a recently reported X-linked intellectual disability-associated missense mutation, resulting in the PQBP1-P244L variant, disrupts the interaction with TXNL4A. We further show that this interaction is critical for the subcellular location of TXNL4A. In combination with other PQBP1 variants lacking a functional nuclear localization signal required for recognition by the nuclear import receptor karyopherin ß2, we demonstrate that PQBP1 facilitates the nuclear import of TXNL4A via a piggyback mechanism. These findings expand our understanding of the molecular basis of the PQBP1-TXNL4A interaction and of the etiology and pathogenesis of Renpenning syndrome and related disorders.


Subject(s)
Cerebral Palsy/genetics , DNA-Binding Proteins/genetics , Mental Retardation, X-Linked/genetics , Ribonucleoprotein, U5 Small Nuclear/genetics , beta Karyopherins/genetics , Active Transport, Cell Nucleus/genetics , Cerebral Palsy/pathology , Gene Expression Regulation/genetics , HeLa Cells , Humans , Intellectual Disability/genetics , Intellectual Disability/pathology , Mental Retardation, X-Linked/pathology , Mutation, Missense/genetics , Protein Binding/genetics , RNA Precursors/genetics , RNA Splicing/genetics , RNA Splicing Factors/genetics , Signal Transduction/genetics
18.
Hum Mol Genet ; 28(22): 3704-3723, 2019 11 15.
Article in English | MEDLINE | ID: mdl-31304552

ABSTRACT

The craniofacial disorder mandibulofacial dysostosis Guion-Almeida type is caused by haploinsufficiency of the U5 snRNP gene EFTUD2/SNU114. However, it is unclear how reduced expression of this core pre-mRNA splicing factor leads to craniofacial defects. Here we use a CRISPR-Cas9 nickase strategy to generate a human EFTUD2-knockdown cell line and show that reduced expression of EFTUD2 leads to diminished proliferative ability of these cells, increased sensitivity to endoplasmic reticulum (ER) stress and the mis-expression of several genes involved in the ER stress response. RNA-Seq analysis of the EFTUD2-knockdown cell line revealed transcriptome-wide changes in gene expression, with an enrichment for genes associated with processes involved in craniofacial development. Additionally, our RNA-Seq data identified widespread mis-splicing in EFTUD2-knockdown cells. Analysis of the functional and physical characteristics of mis-spliced pre-mRNAs highlighted conserved properties, including length and splice site strengths, of retained introns and skipped exons in our disease model. We also identified enriched processes associated with the affected genes, including cell death, cell and organ morphology and embryonic development. Together, these data support a model in which EFTUD2 haploinsufficiency leads to the mis-splicing of a distinct subset of pre-mRNAs with a widespread effect on gene expression, including altering the expression of ER stress response genes and genes involved in the development of the craniofacial region. The increased burden of unfolded proteins in the ER resulting from mis-splicing would exceed the capacity of the defective ER stress response, inducing apoptosis in cranial neural crest cells that would result in craniofacial abnormalities during development.


Subject(s)
Mandibulofacial Dysostosis/genetics , Peptide Elongation Factors/genetics , Ribonucleoprotein, U5 Small Nuclear/genetics , CRISPR-Cas Systems , Cell Proliferation/genetics , Craniofacial Abnormalities/genetics , Endoplasmic Reticulum Stress/genetics , Exons , Gene Expression/genetics , Gene Expression Regulation, Developmental/genetics , HEK293 Cells , Haploinsufficiency/genetics , Humans , Introns , Mutation , Peptide Elongation Factors/metabolism , Phenotype , RNA Precursors/metabolism , RNA Splicing/genetics , Ribonucleoprotein, U5 Small Nuclear/metabolism , Sequence Analysis, RNA/methods , Spliceosomes/genetics
19.
Mol Genet Genomics ; 296(6): 1263-1278, 2021 Nov.
Article in English | MEDLINE | ID: mdl-34453201

ABSTRACT

Nascent ribosomal 60S subunits undergo the last maturation steps in the cytoplasm. The last one involves removing the anti-association factor eIF6 from the 60S ribosomal surface by the joint action of the Elongation Factor-like 1 (EFL1) GTPase and the SBDS protein. Herein, we studied the evolutionary relationship of the EFL1 and EF-2 protein families and the functional conservation within EFL1 orthologues. Phylogenetic analysis demonstrated that the EFL1 proteins are exclusive of eukaryotes and share an evolutionary origin with the EF-2 and EF-G protein families. EFL1 proteins originated by gene duplication from the EF-2 proteins and specialized in ribosome maturation while the latter retained their function in translation. Some organisms have more than one EFL1 protein resulting from alternative splicing, while others are encoded in different genes originated by gene duplication. However, the function of these alternative EFL1 proteins is still unknown. We performed GTPase activity and complementation assays to study the functional conservation of EFL1 homologs alone and together with their SBDS counterparts. None of the orthologues or cross-species combinations could replace the function of the corresponding yeast EFL1•SBDS binomial. The complementation of SBDS interspecies chimeras indicates that domain 2 is vital for its function together with EFL1 and the 60S subunit. The results suggest a functional species-specificity and possible co-evolution between EFL1, SBDS, and the 60S ribosomal subunit. These findings set the basis for further studies directed to understand the molecular evolution of these proteins and their impact on ribosome biogenesis and disease.


Subject(s)
Peptide Elongation Factor 2/metabolism , Peptide Elongation Factors/genetics , Proteins/genetics , Ribonucleoprotein, U5 Small Nuclear/genetics , Ribosome Subunits, Large, Eukaryotic/metabolism , Ribosomes/metabolism , Alternative Splicing/genetics , Amino Acid Sequence/genetics , Eukaryota/genetics , Evolution, Molecular , Gene Duplication/genetics , Humans , Peptide Elongation Factor 2/genetics , Phylogeny , Sequence Alignment
20.
RNA ; 25(10): 1298-1310, 2019 10.
Article in English | MEDLINE | ID: mdl-31289129

ABSTRACT

There is increasing evidence from yeast to humans that pre-mRNA splicing occurs mainly cotranscriptionally, such that splicing and transcription are functionally coupled. Currently, there is little insight into the contribution of the core transcription elongation machinery to cotranscriptional spliceosome assembly and pre-mRNA splicing. Spt5 is a member of the core transcription elongation machinery and an essential protein, whose absence in budding yeast causes defects in pre-mRNA splicing. To determine how Spt5 affects pre-mRNA splicing, we used the auxin-inducible degron system to conditionally deplete Spt5 in Saccharomyces cerevisiae and assayed effects on cotranscriptional spliceosome assembly and splicing. We show that Spt5 is needed for efficient splicing and for the accumulation of U5 snRNPs at intron-containing genes, and therefore for stable cotranscriptional assembly of spliceosomes. The defect in cotranscriptional spliceosome assembly can explain the relatively mild splicing defect as being a consequence of the failure of cotranscriptional splicing. Coimmunoprecipitation of Spt5 with core spliceosomal proteins and all spliceosomal snRNAs suggests a model whereby Spt5 promotes cotranscriptional pre-mRNA splicing by stabilizing the association of U5 snRNP with spliceosome complexes as they assemble on the nascent transcript. If this phenomenon is conserved in higher eukaryotes, it has the potential to be important for cotranscriptional regulation of alternative splicing.


Subject(s)
Chromosomal Proteins, Non-Histone/physiology , Saccharomyces cerevisiae/metabolism , Spliceosomes , Transcription, Genetic , Transcriptional Elongation Factors/physiology , Chromosomal Proteins, Non-Histone/metabolism , Immunoprecipitation , Protein Binding , RNA Splicing , Ribonucleoprotein, U5 Small Nuclear/genetics , Transcriptional Elongation Factors/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL