Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 35
Filter
1.
Proc Natl Acad Sci U S A ; 120(9): e2102569120, 2023 02 28.
Article in English | MEDLINE | ID: mdl-36802443

ABSTRACT

In the human genome, about 750 genes contain one intron excised by the minor spliceosome. This spliceosome comprises its own set of snRNAs, among which U4atac. Its noncoding gene, RNU4ATAC, has been found mutated in Taybi-Linder (TALS/microcephalic osteodysplastic primordial dwarfism type 1), Roifman (RFMN), and Lowry-Wood (LWS) syndromes. These rare developmental disorders, whose physiopathological mechanisms remain unsolved, associate ante- and post-natal growth retardation, microcephaly, skeletal dysplasia, intellectual disability, retinal dystrophy, and immunodeficiency. Here, we report bi-allelic RNU4ATAC mutations in five patients presenting with traits suggestive of the Joubert syndrome (JBTS), a well-characterized ciliopathy. These patients also present with traits typical of TALS/RFMN/LWS, thus widening the clinical spectrum of RNU4ATAC-associated disorders and indicating ciliary dysfunction as a mechanism downstream of minor splicing defects. Intriguingly, all five patients carry the n.16G>A mutation, in the Stem II domain, either at the homozygous or compound heterozygous state. A gene ontology term enrichment analysis on minor intron-containing genes reveals that the cilium assembly process is over-represented, with no less than 86 cilium-related genes containing at least one minor intron, among which there are 23 ciliopathy-related genes. The link between RNU4ATAC mutations and ciliopathy traits is supported by alterations of primary cilium function in TALS and JBTS-like patient fibroblasts, as well as by u4atac zebrafish model, which exhibits ciliopathy-related phenotypes and ciliary defects. These phenotypes could be rescued by WT but not by pathogenic variants-carrying human U4atac. Altogether, our data indicate that alteration of cilium biogenesis is part of the physiopathological mechanisms of TALS/RFMN/LWS, secondarily to defects of minor intron splicing.


Subject(s)
Ciliopathies , Spliceosomes , Female , Animals , Humans , Spliceosomes/genetics , RNA, Small Nuclear/genetics , Zebrafish/genetics , Fetal Growth Retardation/genetics , Mutation , Ciliopathies/genetics
2.
Nucleic Acids Res ; 51(2): 712-727, 2023 01 25.
Article in English | MEDLINE | ID: mdl-36537210

ABSTRACT

Various genetic diseases associated with microcephaly and developmental defects are due to pathogenic variants in the U4atac small nuclear RNA (snRNA), a component of the minor spliceosome essential for the removal of U12-type introns from eukaryotic mRNAs. While it has been shown that a few RNU4ATAC mutations result in impaired binding of essential protein components, the molecular defects of the vast majority of variants are still unknown. Here, we used lymphoblastoid cells derived from RNU4ATAC compound heterozygous (g.108_126del;g.111G>A) twin patients with MOPD1 phenotypes to analyze the molecular consequences of the mutations on small nuclear ribonucleoproteins (snRNPs) formation and on splicing. We found that the U4atac108_126del mutant is unstable and that the U4atac111G>A mutant as well as the minor di- and tri-snRNPs are present at reduced levels. Our results also reveal the existence of 3'-extended snRNA transcripts in patients' cells. Moreover, we show that the mutant cells have alterations in splicing of INTS7 and INTS10 minor introns, contain lower levels of the INTS7 and INTS10 proteins and display changes in the assembly of Integrator subunits. Altogether, our results show that compound heterozygous g.108_126del;g.111G>A mutations induce splicing defects and affect the homeostasis and function of the Integrator complex.


Subject(s)
Ribonucleoproteins, Small Nuclear , Spliceosomes , Spliceosomes/genetics , Spliceosomes/metabolism , Ribonucleoproteins, Small Nuclear/genetics , Mutation , Introns/genetics , RNA Splicing/genetics , RNA, Small Nuclear/metabolism , Homeostasis/genetics
3.
RNA ; 28(3): 303-319, 2022 03.
Article in English | MEDLINE | ID: mdl-34893560

ABSTRACT

Spinal muscular atrophy (SMA) is a devastating neurodegenerative disease caused by reduced amounts of the ubiquitously expressed Survival of Motor Neuron (SMN) protein. In agreement with its crucial role in the biogenesis of spliceosomal snRNPs, SMN-deficiency is correlated to numerous splicing alterations in patient cells and various tissues of SMA mouse models. Among the snRNPs whose assembly is impacted by SMN-deficiency, those involved in the minor spliceosome are particularly affected. Importantly, splicing of several, but not all U12-dependent introns has been shown to be affected in different SMA models. Here, we have investigated the molecular determinants of this differential splicing in spinal cords from SMA mice. We show that the branchpoint sequence (BPS) is a key element controlling splicing efficiency of minor introns. Unexpectedly, splicing of several minor introns with suboptimal BPS is not affected in SMA mice. Using in vitro splicing experiments and oligonucleotides targeting minor or major snRNAs, we show for the first time that splicing of these introns involves both the minor and major machineries. Our results strongly suggest that splicing of a subset of minor introns is not affected in SMA mice because components of the major spliceosome compensate for the loss of minor splicing activity.


Subject(s)
Muscular Atrophy, Spinal/genetics , RNA Splicing , Spliceosomes/metabolism , Animals , HeLa Cells , Humans , Introns , Mice , Muscular Atrophy, Spinal/metabolism , RNA Splice Sites , Ribonucleoproteins, Small Nuclear/metabolism
4.
Clin Genet ; 2024 Jun 04.
Article in English | MEDLINE | ID: mdl-38837402

ABSTRACT

Microcephalic osteodysplastic primordial dwarfism type I (MOPDI) is a very rare and severe autosomal recessive disorder characterized by marked intrauterine growth retardation, skeletal dysplasia, microcephaly and brain malformations. MOPDI is caused by biallelic mutations in RNU4ATAC, a non-coding gene involved in U12-type splicing of 1% of the introns in the genome, which are recognized by their specific splicing consensus sequences. Here, we describe a unique observation of immunodeficiency in twin sisters with mild MOPDI, who harbor a novel n.108_126del mutation, encompassing part of the U4atac snRNA 3' stem-loop and Sm protein binding site, and the previously reported n.111G>A mutation. Interestingly, both twin sisters show mild B-cell anomalies, including low naive B-cell counts and increased memory B-cell and plasmablasts counts, suggesting partial and transitory blockage of B-cell maturation and/or excessive activation of naive B-cells. Hence, the localization of a mutation in stem II of U4atac snRNA, as observed in another RNU4ATAC-opathy with immunodeficiency, that is, Roifman syndrome (RFMN), is not required for the occurrence of an immune deficiency. Finally, we emphasize the importance of considering immunodeficiency in MOPDI management to reduce the risk of serious infectious episodes.

5.
Nucleic Acids Res ; 49(13): 7207-7223, 2021 07 21.
Article in English | MEDLINE | ID: mdl-33754639

ABSTRACT

The macromolecular SMN complex facilitates the formation of Sm-class ribonucleoproteins involved in mRNA processing (UsnRNPs). While biochemical studies have revealed key activities of the SMN complex, its structural investigation is lagging behind. Here we report on the identification and structural determination of the SMN complex from the lower eukaryote Schizosaccharomyces pombe, consisting of SMN, Gemin2, 6, 7, 8 and Sm proteins. The core of the SMN complex is formed by several copies of SMN tethered through its C-terminal alpha-helices arranged with alternating polarity. This creates a central platform onto which Gemin8 binds and recruits Gemins 6 and 7. The N-terminal parts of the SMN molecules extrude via flexible linkers from the core and enable binding of Gemin2 and Sm proteins. Our data identify the SMN complex as a multivalent hub where Sm proteins are collected in its periphery to allow their joining with UsnRNA.


Subject(s)
SMN Complex Proteins/chemistry , Schizosaccharomyces pombe Proteins/chemistry , Carrier Proteins/chemistry , Crystallography, X-Ray , Humans , Models, Molecular , Muscular Atrophy, Spinal/genetics , Mutation , Nuclear Proteins/chemistry , Protein Binding , SMN Complex Proteins/metabolism , Scattering, Small Angle , Schizosaccharomyces pombe Proteins/metabolism , Structural Homology, Protein , X-Ray Diffraction
6.
RNA ; 23(6): 899-909, 2017 06.
Article in English | MEDLINE | ID: mdl-28258160

ABSTRACT

Spinal muscular atrophy (SMA) is caused by mutations and/or deletions of the survival motor neuron gene (SMN1). Besides its function in the biogenesis of spliceosomal snRNPs, SMN might possess a motor neuron specific role and could function in the transport of axonal mRNAs and in the modulation of local protein translation. Accordingly, SMN colocalizes with axonal mRNAs of differentiated NSC-34 motor neuron-like cells. We recently showed that SMN depletion gives rise to a decrease in the axonal transport of the mRNAs encoding Annexin A2 (Anxa2). In this work, we have characterized the structural features of the Anxa2 mRNA required for its axonal targeting by SMN. We found that a G-rich motif located near the 3'UTR is essential for axonal localization of the Anxa2 transcript. We also show that mutations in the motif sequence abolish targeting of Anxa2 reporter mRNAs in axon-like structures of differentiated NSC-34 cells. Finally, localization of both wild-type and mutated Anxa2 reporters is restricted to the cell body in SMN-depleted cells. Altogether, our studies show that this G-motif represents a novel and essential determinant for axonal localization of the Anxa2 mRNA mediated by the SMN complex.


Subject(s)
Annexin A2/genetics , Annexin A2/metabolism , Axons/metabolism , Nucleotide Motifs , RNA, Messenger , Survival of Motor Neuron 1 Protein/metabolism , 3' Untranslated Regions , Animals , Annexin A2/chemistry , Base Sequence , Cell Line , G-Quadruplexes , Gene Expression , Genes, Reporter , Humans , Mice , Motor Neurons/metabolism , Protein Binding , Protein Transport , RNA Transport
7.
Neurobiol Dis ; 94: 245-58, 2016 Oct.
Article in English | MEDLINE | ID: mdl-27388936

ABSTRACT

The neuromuscular disorder, spinal muscular atrophy (SMA), results from insufficient levels of the survival motor neuron (SMN) protein. Together with Gemins 2-8 and Unrip, SMN forms the large macromolecular SMN-Gemins complex, which is known to be indispensable for chaperoning the assembly of spliceosomal small nuclear ribonucleoproteins (snRNPs). It remains unclear whether disruption of this function is responsible for the selective neuromuscular degeneration in SMA. In the present study, we first show that loss of wmd, the Drosophila Unrip orthologue, has a negative impact on the motor system. However, due to lack of a functional relationship between wmd/Unrip and Gemin3, it is likely that Unrip joined the SMN-Gemins complex only recently in evolution. Second, we uncover that disruption of either Tgs1 or pICln, two cardinal players in snRNP biogenesis, results in viability and motor phenotypes that closely resemble those previously uncovered on loss of the constituent members of the SMN-Gemins complex. Interestingly, overexpression of both factors leads to motor dysfunction in Drosophila, a situation analogous to that of Gemin2. Toxicity is conserved in the yeast S. pombe where pICln overexpression induces a surplus of Sm proteins in the cytoplasm, indicating that a block in snRNP biogenesis is partly responsible for this phenotype. Importantly, we show a strong functional relationship and a physical interaction between Gemin3 and either Tgs1 or pICln. We propose that snRNP biogenesis is the pathway connecting the SMN-Gemins complex to a functional neuromuscular system, and its disturbance most likely leads to the motor dysfunction that is typical in SMA.


Subject(s)
Drosophila Proteins/metabolism , Motor Neurons/metabolism , Ribonucleoproteins, Small Nuclear/metabolism , SMN Complex Proteins/metabolism , Animals , Cytoplasm/metabolism , Drosophila Proteins/genetics , Drosophila melanogaster , Phenotype
8.
EMBO J ; 30(11): 2205-18, 2011 Jun 01.
Article in English | MEDLINE | ID: mdl-21522132

ABSTRACT

Transport of C/D snoRNPs to nucleoli involves nuclear export factors. In particular, CRM1 binds nascent snoRNPs, but its precise role remains unknown. We show here that both CRM1 and nucleocytoplasmic trafficking are required to transport snoRNPs to nucleoli, but the snoRNPs do not transit through the cytoplasm. Instead, CRM1 controls the composition of nucleoplasmic pre-snoRNP complexes. We observed that Tgs1 long form (Tgs1 LF), the long isoform of the cap hypermethylase, contains a leucine-rich nuclear export signal, shuttles in a CRM1-dependent manner, and binds to the nucleolar localization signal (NoLS) of the core snoRNP protein Nop58. In vitro data indicate that CRM1 binds Tgs1 LF and promotes its dissociation from Nop58 NoLS, and immunoprecipitation experiments from cells indicate that the association of Tgs1 LF with snoRNPs increases upon CRM1 inhibition. Thus, CRM1 appears to promote nucleolar transport of snoRNPs by removing Tgs1 LF from the Nop58 NoLS. Microarray/IP data show that this occurs on most snoRNPs, from both C/D and H/ACA families, and on the telomerase RNA. Hence, CRM1 provides a general molecular link between nuclear events and nucleocytoplasmic trafficking.


Subject(s)
Cell Nucleus/metabolism , Karyopherins/metabolism , RNA, Small Nucleolar/metabolism , Receptors, Cytoplasmic and Nuclear/metabolism , Ribonucleoproteins, Small Nucleolar/metabolism , Active Transport, Cell Nucleus , Cell Line , Humans , Methyltransferases/metabolism , Nuclear Proteins/metabolism , Protein Binding , Exportin 1 Protein
9.
RNA ; 19(12): 1755-66, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24152552

ABSTRACT

Spinal muscular atrophy is a neuromuscular disease resulting from mutations in the SMN1 gene, which encodes the survival motor neuron (SMN) protein. SMN is part of a large complex that is essential for the biogenesis of spliceosomal small nuclear RNPs. SMN also colocalizes with mRNAs in granules that are actively transported in neuronal processes, supporting the hypothesis that SMN is involved in axonal trafficking of mRNPs. Here, we have performed a genome-wide analysis of RNAs present in complexes containing the SMN protein and identified more than 200 mRNAs associated with SMN in differentiated NSC-34 motor neuron-like cells. Remarkably, ~30% are described to localize in axons of different neuron types. In situ hybridization and immuno-fluorescence experiments performed on several candidates indicate that these mRNAs colocalize with the SMN protein in neurites and axons of differentiated NSC-34 cells. Moreover, they localize in cell processes in an SMN-dependent manner. Thus, low SMN levels might result in localization deficiencies of mRNAs required for axonogenesis.


Subject(s)
Neurites/metabolism , RNA, Messenger/metabolism , Survival of Motor Neuron 1 Protein/metabolism , Animals , Annexin A2/genetics , Annexin A2/metabolism , Cell Line , Electron Transport Complex IV/genetics , Electron Transport Complex IV/metabolism , Gene Knockdown Techniques , Genome , Mice , Motor Neurons/metabolism , Neuromuscular Junction/physiology , Oligonucleotide Array Sequence Analysis , RNA Transport , RNA, Messenger/genetics , RNA, Small Interfering/genetics , Selenoprotein W/genetics , Selenoprotein W/metabolism , Survival of Motor Neuron 1 Protein/genetics
10.
Nucleic Acids Res ; 41(2): 1255-72, 2013 Jan.
Article in English | MEDLINE | ID: mdl-23221635

ABSTRACT

Spinal muscular atrophy is a severe motor neuron disease caused by reduced levels of the ubiquitous Survival of MotoNeurons (SMN) protein. SMN is part of a complex that is essential for spliceosomal UsnRNP biogenesis. Signal recognition particle (SRP) is a ribonucleoprotein particle crucial for co-translational targeting of secretory and membrane proteins to the endoplasmic reticulum. SRP biogenesis is a nucleo-cytoplasmic multistep process in which the protein components, except SRP54, assemble with 7S RNA in the nucleolus. Then, SRP54 is incorporated after export of the pre-particle into the cytoplasm. The assembly factors necessary for SRP biogenesis remain to be identified. Here, we show that 7S RNA binds to purified SMN complexes in vitro and that SMN complexes associate with SRP in cellular extracts. We identified the RNA determinants required. Moreover, we report a specific reduction of 7S RNA levels in the spinal cord of SMN-deficient mice, and in a Schizosaccharomyces pombe strain carrying a temperature-degron allele of SMN. Additionally, microinjected antibodies directed against SMN or Gemin2 interfere with the association of SRP54 with 7S RNA in Xenopus laevis oocytes. Our data show that reduced levels of the SMN protein lead to defect in SRP steady-state level and describe the SMN complex as the first identified cellular factor required for SRP biogenesis.


Subject(s)
RNA, Small Cytoplasmic/metabolism , SMN Complex Proteins/metabolism , Signal Recognition Particle/metabolism , Alleles , Animals , Antibodies/pharmacology , Base Sequence , Cytoplasm/metabolism , HeLa Cells , Humans , Mice , Molecular Sequence Data , Muscular Atrophy, Spinal/metabolism , Mutation , RNA, Small Cytoplasmic/chemistry , RNA, Small Nuclear/metabolism , SMN Complex Proteins/antagonists & inhibitors , SMN Complex Proteins/immunology , Schizosaccharomyces/genetics , Signal Recognition Particle/chemistry , Spinal Cord/metabolism , Xenopus laevis
11.
EMBO J ; 29(11): 1817-29, 2010 Jun 02.
Article in English | MEDLINE | ID: mdl-20400941

ABSTRACT

Spinal muscular atrophy results from deletions or mutations in the survival of motor neuron (SMN1) gene. The SMN protein has an essential role in the biogenesis of spliceosomal snRNPs, but the link between a defect in this process and specific splicing inhibition of pre-mRNAs has not been established. In this study, we report the construction of a temperature-degron (td) allele of the Schizosaccharomyces pombe SMN protein and show that its depletion at 37 degrees C affects splicing and formation of U1, U2, U4 and U5 snRNPs, but not of U6 and U3 ribonucleoproteins. The function of the tdSMN allele in snRNP assembly is already perturbed at 25 degrees C, suggesting a deleterious effect of the tag at this temperature. Using a genome-wide approach, we report that introns react unequally to lower levels of snRNPs in tdSMN cells and that increasing the length of the polypyrimidine tract can improve the splicing efficiency of some, but not all, affected introns. Altogether, our results suggest that the defects observed in tdSMN fission yeast cells mimic splicing deficits observed in SMN-deficient metazoan cells.


Subject(s)
Genes, Fungal , RNA Precursors/metabolism , RNA Splicing , Ribonucleoproteins, Small Nuclear/metabolism , Schizosaccharomyces pombe Proteins/genetics , Schizosaccharomyces/genetics , Spliceosomes/metabolism , Survival of Motor Neuron 1 Protein/genetics , Alleles , Cell Nucleus/genetics , Cell Nucleus/metabolism , Humans , Introns , Motor Neurons/metabolism , Muscular Atrophy, Spinal/genetics , Muscular Atrophy, Spinal/metabolism , Mutation , RNA Precursors/genetics , Ribonucleoproteins/genetics , Ribonucleoproteins/metabolism , Ribonucleoproteins, Small Nuclear/genetics , Spliceosomes/genetics , snRNP Core Proteins
12.
RNA ; 18(1): 31-6, 2012 Jan.
Article in English | MEDLINE | ID: mdl-22124016

ABSTRACT

The survival of motor neuron (SMN) protein plays an important role in the biogenesis of spliceosomal snRNPs and is one factor required for the integrity of nuclear Cajal bodies (CBs). CBs are enriched in small CB-specific (sca) RNAs, which guide the formation of pseudouridylated and 2'-O-methylated residues in the snRNAs. Because SMN-deficient cells lack typical CBs, we asked whether the modification of internal residues of major and minor snRNAs is defective in these cells. We mapped modified nucleotides in the major U2 and the minor U4atac and U12 snRNAs. Using both radioactive and fluorescent primer extension approaches, we found that modification of major and minor spliceosomal snRNAs is normal in SMN-deficient cells. Our experiments also revealed a previously undetected pseudouridine at position 60 in human U2 and 2'-O-methylation of A1, A2, and G19 in human U4atac. These results confirm, and extend to minor snRNAs, previous experiments showing that scaRNPs can function in the absence of typical CBs. Furthermore, they show that the differential splicing defects in SMN-deficient cells are not due to failure of post-transcriptional modification of either major or minor snRNAs.


Subject(s)
RNA Processing, Post-Transcriptional/genetics , RNA, Small Nuclear/metabolism , Survival of Motor Neuron 1 Protein/genetics , Coiled Bodies/chemistry , Coiled Bodies/metabolism , HeLa Cells , Humans , Methylation , RNA, Small Nuclear/chemistry , Spliceosomes/genetics , Spliceosomes/metabolism
13.
Hum Mol Genet ; 20(4): 641-8, 2011 Feb 15.
Article in English | MEDLINE | ID: mdl-21098506

ABSTRACT

The survival of motor neuron (SMN) protein is essential for cytoplasmic assembly of spliceosomal snRNPs. Although the normal proportion of endogenous snRNAs is unevenly altered in spinal muscular atrophy (SMA) tissues, the biogenesis of individual snRNPs is not dramatically affected in SMN-deficient cells. The SMN protein is also required for normal Cajal body (CB) formation, but the functional consequences of CB disruption upon SMN deficiency have not yet been analyzed at the level of macromolecular snRNPs assembly. Here, we show that the SMN protein is required for tri-snRNPs formation and that the level of the minor U4atac/U6atac/U5 tri-snRNPs is dramatically decreased in lymphoblasts derived from a patient suffering from a severe form of SMA. We found also that splicing of some, but not all, minor introns is inhibited in these cells, demonstrating links between SMN deficiency and differential alterations of splicing events mediated by the minor spliceosome. Our results suggest that SMA might result from the inefficient splicing of one or only a few pre-mRNAs carrying minor introns and coding for proteins required for motor neurons function and/or organization.


Subject(s)
Introns/genetics , Lymphocytes/pathology , RNA Splicing/genetics , RNA, Small Nuclear/genetics , Ribonucleoproteins, Small Nuclear , Spinal Muscular Atrophies of Childhood/pathology , Spliceosomes/pathology , Cell Survival/genetics , Coiled Bodies/pathology , Gene Expression Profiling , Gene Expression Regulation , Gene Knockout Techniques , Humans , Ribonucleoproteins, Small Nuclear/genetics , Ribonucleoproteins, Small Nuclear/metabolism , Spinal Muscular Atrophies of Childhood/genetics
14.
iScience ; 23(1): 100809, 2020 Jan 24.
Article in English | MEDLINE | ID: mdl-31927482

ABSTRACT

Spinal muscular atrophy (SMA) is a devastating motor neuron disorder caused by mutations in the survival motor neuron (SMN) gene. It remains unclear how SMN deficiency leads to the loss of motor neurons. By screening Schizosaccharomyces pombe, we found that the growth defect of an SMN mutant can be alleviated by deletion of the actin-capping protein subunit gene acp1+. We show that SMN mutated cells have splicing defects in the profilin gene, which thus directly hinder actin cytoskeleton homeostasis including endocytosis and cytokinesis. We conclude that deletion of acp1+ in an SMN mutant background compensates for actin cytoskeleton alterations by restoring redistribution of actin monomers between different types of cellular actin networks. Our data reveal a direct correlation between an impaired function of SMN in snRNP assembly and defects in actin dynamics. They also point to important common features in the pathogenic mechanism of SMA and ALS.

15.
J Neurosci ; 28(51): 13793-804, 2008 Dec 17.
Article in English | MEDLINE | ID: mdl-19091970

ABSTRACT

Intracellular mRNA transport and local translation play a key role in neuronal physiology. Translationally repressed mRNAs are transported as a part of ribonucleoprotein (RNP) particles to distant dendritic sites, but the properties of different RNP particles and mechanisms of their repression and transport remain largely unknown. Here, we describe a new class of RNP-particles, the dendritic P-body-like structures (dlPbodies), which are present in the soma and dendrites of mammalian neurons and have both similarities and differences to P-bodies of non-neuronal cells. These structures stain positively for a number of P-body and microRNP components, a microRNA-repressed mRNA and some translational repressors. They appear more heterogeneous than P-bodies of HeLa cells, and they rarely contain the exonuclease Xrn1 but are positive for rRNA. These particles show motorized movements along dendrites and relocalize to distant sites in response to synaptic activation. Furthermore, Dcp1a is stably associated with dlP-bodies in unstimulated cells, but exchanges rapidly on neuronal activation, concomitantly with the loss of Ago2 from dlP-bodies. Thus, dlP-bodies may regulate local translation by storing repressed mRNPs in unstimulated cells, and releasing them on synaptic activation.


Subject(s)
Dendrites/physiology , Dendrites/ultrastructure , MicroRNAs/metabolism , Neurons/ultrastructure , Ribonucleoproteins/physiology , Animals , Argonaute Proteins , Biological Transport/physiology , Cells, Cultured , Dendrites/drug effects , Endoribonucleases/genetics , Eukaryotic Initiation Factor-2/genetics , Excitatory Amino Acid Agonists/pharmacology , Green Fluorescent Proteins/genetics , HeLa Cells , Hippocampus/cytology , Humans , Hypothalamus/cytology , Neurons/drug effects , Neurons/physiology , Particle Size , RNA, Ribosomal/metabolism , Rats , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Synaptic Transmission/drug effects , Synaptic Transmission/physiology , Trans-Activators/genetics , Transfection
16.
Mol Biol Cell ; 17(7): 3221-31, 2006 Jul.
Article in English | MEDLINE | ID: mdl-16687569

ABSTRACT

Cajal bodies (CBs) have been implicated in the nuclear phase of the biogenesis of spliceosomal U small nuclear ribonucleoproteins (U snRNPs). Here, we have investigated the distribution of the CB marker protein coilin, U snRNPs, and proteins present in C/D box small nucleolar (sno)RNPs in cells depleted of hTGS1, SMN, or PHAX. Knockdown of any of these three proteins by RNAi interferes with U snRNP maturation before the reentry of U snRNA Sm cores into the nucleus. Strikingly, CBs are lost in the absence of hTGS1, SMN, or PHAX and coilin is dispersed in the nucleoplasm into numerous small foci. This indicates that the integrity of canonical CBs is dependent on ongoing U snRNP biogenesis. Spliceosomal U snRNPs show no detectable concentration in nuclear foci and do not colocalize with coilin in cells lacking hTGS1, SMN, or PHAX. In contrast, C/D box snoRNP components concentrate into nuclear foci that partially colocalize with coilin after inhibition of U snRNP maturation. We demonstrate by siRNA-mediated depletion that coilin is required for the condensation of U snRNPs, but not C/D box snoRNP components, into nucleoplasmic foci, and also for merging these factors into canonical CBs. Altogether, our data suggest that CBs have a modular structure with distinct domains for spliceosomal U snRNPs and snoRNPs.


Subject(s)
Coiled Bodies/metabolism , Coiled Bodies/ultrastructure , Nuclear Proteins/analysis , Ribonucleoproteins, Small Nuclear/analysis , Ribonucleoproteins, Small Nuclear/biosynthesis , Cell Nucleus/chemistry , Cell Nucleus/ultrastructure , Coiled Bodies/chemistry , Cyclic AMP Response Element-Binding Protein/antagonists & inhibitors , Cyclic AMP Response Element-Binding Protein/genetics , HeLa Cells , Humans , Methylation , Methyltransferases/antagonists & inhibitors , Methyltransferases/genetics , Mutation , Nerve Tissue Proteins/antagonists & inhibitors , Nerve Tissue Proteins/genetics , Nuclear Proteins/antagonists & inhibitors , Nuclear Proteins/genetics , Nucleocytoplasmic Transport Proteins/antagonists & inhibitors , Nucleocytoplasmic Transport Proteins/genetics , Phosphoproteins/antagonists & inhibitors , Phosphoproteins/genetics , RNA Interference , RNA, Small Interfering/genetics , RNA, Small Interfering/pharmacology , RNA-Binding Proteins/antagonists & inhibitors , RNA-Binding Proteins/genetics , Ribonucleoprotein, U4-U6 Small Nuclear/analysis , SMN Complex Proteins , Spliceosomes/metabolism , Spliceosomes/ultrastructure
17.
Sci Rep ; 9(1): 18666, 2019 12 10.
Article in English | MEDLINE | ID: mdl-31822699

ABSTRACT

The predominant motor neuron disease in infants and adults is spinal muscular atrophy (SMA) and amyotrophic lateral sclerosis (ALS), respectively. SMA is caused by insufficient levels of the Survival Motor Neuron (SMN) protein, which operates as part of the multiprotein SMN complex that includes the DEAD-box RNA helicase Gemin3/DDX20/DP103. C9orf72, SOD1, TDP-43 and FUS are ranked as the four major genes causing familial ALS. Accumulating evidence has revealed a surprising molecular overlap between SMA and ALS. Here, we ask the question of whether Drosophila can also be exploited to study shared pathogenic pathways. Focusing on motor behaviour, muscle mass and survival, we show that disruption of either TBPH/TDP-43 or Caz/FUS enhance defects associated with Gemin3 loss-of-function. Gemin3-associated neuromuscular junction overgrowth was however suppressed. Sod1 depletion had a modifying effect in late adulthood. We also show that Gemin3 self-interacts and Gem3ΔN, a helicase domain deletion mutant, retains the ability to interact with its wild-type counterpart. Importantly, mutant:wild-type dimers are favoured more than wild-type:wild-type dimers. In addition to reinforcing the link between SMA and ALS, further exploration of mechanistic overlaps is now possible in a genetically tractable model organism. Notably, Gemin3 can be elevated to a candidate for modifying motor neuron degeneration.


Subject(s)
DEAD-box RNA Helicases/metabolism , DNA-Binding Proteins/metabolism , Drosophila Proteins/metabolism , RNA-Binding Proteins/metabolism , Superoxide Dismutase/metabolism , Transcription Factor TFIID/metabolism , Alleles , Animals , C9orf72 Protein/genetics , C9orf72 Protein/metabolism , DEAD-box RNA Helicases/genetics , DNA-Binding Proteins/genetics , Drosophila Proteins/genetics , Drosophila melanogaster/genetics , Drosophila melanogaster/metabolism , Female , Flight, Animal , Genotype , Humans , Male , Phenotype , RNA, Small Interfering/metabolism , RNA-Binding Proteins/genetics , Superoxide Dismutase/genetics , Superoxide Dismutase-1/genetics , Superoxide Dismutase-1/metabolism , Transcription Factor TFIID/genetics
18.
Nucleic Acids Res ; 34(10): 2925-32, 2006.
Article in English | MEDLINE | ID: mdl-16738131

ABSTRACT

Neuronal degeneration in spinal muscular atrophy (SMA) is caused by reduced expression of the survival of motor neuron (SMN) protein. The SMN protein is ubiquitously expressed and is present both in the cytoplasm and in the nucleus where it localizes in Cajal bodies. The SMN complex plays an essential role for the biogenesis of spliceosomal U-snRNPs. In this article, we have used an RNA interference approach in order to analyse the effects of SMN depletion on snRNP assembly in HeLa cells. Although snRNP profiles are not perturbed in SMN-depleted cells, we found that SMN depletion gives rise to cytoplasmic accumulation of a GFP-SmB reporter protein. We also demonstrate that the SMN protein depletion induces defects in Cajal body formation with coilin being localized in multiple nuclear foci and in nucleolus instead of canonical Cajal bodies. Interestingly, the coilin containing foci do not contain snRNPs but appear to co-localize with U85 scaRNA. Because Cajal bodies represent the location in which snRNPs undergo 2'-O-methylation and pseudouridylation, our results raise the possibility that SMN depletion might give rise to a defect in the snRNA modification process.


Subject(s)
Coiled Bodies/chemistry , Cyclic AMP Response Element-Binding Protein/antagonists & inhibitors , Nerve Tissue Proteins/antagonists & inhibitors , RNA-Binding Proteins/antagonists & inhibitors , Ribonucleoproteins, Small Nuclear/metabolism , Autoantigens/analysis , Cyclic AMP Response Element-Binding Protein/genetics , Cytoplasm/chemistry , HeLa Cells , Humans , Nerve Tissue Proteins/genetics , Nuclear Proteins/analysis , RNA Interference , RNA, Small Nuclear/analysis , RNA-Binding Proteins/genetics , Ribonucleoproteins, Small Nuclear/analysis , SMN Complex Proteins , snRNP Core Proteins , RNA, Small Untranslated
19.
Mol Cell Biol ; 24(18): 7976-86, 2004 Sep.
Article in English | MEDLINE | ID: mdl-15340060

ABSTRACT

Nucleolar morphogenesis is a poorly defined process. Here we report that the Saccharomyces cerevisiae nucleolar trimethyl guanosine synthase I (Tgs1p), which specifically selects the m(7)G cap structure of snRNAs and snoRNAs for m(2,2,7)G conversion, is required not only for efficient pre-mRNA splicing but also for pre-rRNA processing and small ribosomal subunit synthesis. Mutational analysis indicates that the requirement for Tgs1p in pre-mRNA splicing, but not its involvement in ribosome synthesis, is dependent upon its function in cap trimethylation. In addition, we report that cells lacking Tgs1p showed a striking and unexpected loss of nucleolar structural organization. Tgs1p is not a core component of the snoRNP proteins; however, in vitro, the protein interacts with the KKD/E domain present at the carboxyl-terminal ends of several snoRNP proteins. Strains expressing versions of the snoRNPs lacking the KKD/E domain were also defective for nucleolar morphology and showed a loss of nucleolar compaction. We propose that the transient and functional interactions of Tgs1p with the abundant snoRNPs, through presumed interactions with the KKD/E domain of the snoRNP proteins, contribute substantially to the coalescence of nucleolar components. This conclusion is compatible with a model of self-organization for nucleolar assembly.


Subject(s)
Cell Nucleolus/metabolism , Cell Nucleolus/ultrastructure , Methyltransferases/metabolism , RNA Caps/metabolism , RNA, Fungal/metabolism , Base Sequence , Genes, Fungal , Methyltransferases/genetics , Microscopy, Electron , Protein Structure, Tertiary , RNA Processing, Post-Transcriptional , Ribonucleoproteins, Small Nucleolar/chemistry , Ribonucleoproteins, Small Nucleolar/genetics , Ribonucleoproteins, Small Nucleolar/metabolism , Ribosomes/metabolism , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism , Saccharomyces cerevisiae/ultrastructure , Saccharomyces cerevisiae Proteins/chemistry , Saccharomyces cerevisiae Proteins/genetics , Saccharomyces cerevisiae Proteins/metabolism , Sequence Deletion
20.
Mol Biol Cell ; 13(7): 2436-47, 2002 Jul.
Article in English | MEDLINE | ID: mdl-12134081

ABSTRACT

Members of the highly conserved serine/arginine-rich (SR) protein family are nuclear factors involved in splicing of metazoan mRNA precursors. In mammals, two nuclear import receptors, transportin (TRN)-SR1 and TRN-SR2, are responsible for targeting SR proteins to the nucleus. Distinctive features in the nuclear localization signal between Drosophila and mammalian SR proteins prompted us to examine the mechanism by which Drosophila SR proteins and their antagonist repressor splicing factor 1 (RSF1) are imported into nucleus. Herein, we report the identification and characterization of a Drosophila importin beta-family protein (dTRN-SR), homologous to TRN-SR2, that specifically interacts with both SR proteins and RSF1. dTRN-SR has a broad localization in the cytoplasm and the nucleus, whereas an N-terminal deletion mutant colocalizes with SR proteins in nuclear speckles. Far Western experiments established that the RS domain of SR proteins and the GRS domain of RSF1 are required for the direct interaction with dTRN-SR, an interaction that can be modulated by phosphorylation. Using the yeast model system in which nuclear import of Drosophila SR proteins and RSF1 is impaired, we demonstrate that complementation with dTRN-SR is sufficient to target these proteins to the nucleus. Together, the results imply that the mechanism by which SR proteins are imported to the nucleus is conserved between Drosophila and humans.


Subject(s)
Active Transport, Cell Nucleus/physiology , Drosophila Proteins/metabolism , Drosophila melanogaster/metabolism , RNA-Binding Proteins/metabolism , Repressor Proteins/metabolism , beta Karyopherins/metabolism , Amino Acid Sequence , Animals , Drosophila Proteins/chemistry , Drosophila Proteins/genetics , Drosophila melanogaster/cytology , HeLa Cells , Humans , Microscopy, Fluorescence , Molecular Sequence Data , Nuclear Proteins/metabolism , Protein Binding , Protein Splicing , Protein Structure, Tertiary , RNA Splicing Factors , RNA-Binding Proteins/genetics , Receptors, Cytoplasmic and Nuclear/genetics , Receptors, Cytoplasmic and Nuclear/metabolism , Recombinant Fusion Proteins/metabolism , Repressor Proteins/genetics , Sequence Alignment , Yeasts/genetics , Yeasts/metabolism , beta Karyopherins/chemistry , beta Karyopherins/genetics
SELECTION OF CITATIONS
SEARCH DETAIL