Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 169
Filtrar
1.
Nat Commun ; 14(1): 7384, 2023 11 15.
Artículo en Inglés | MEDLINE | ID: mdl-37968267

RESUMEN

Spinal muscular atrophy is an autosomal recessive neuromuscular disease caused by mutations in the multifunctional protein Survival of Motor Neuron, or SMN. Within the nucleus, SMN localizes to Cajal bodies, which are associated with nucleoli, nuclear organelles dedicated to the first steps of ribosome biogenesis. The highly organized structure of the nucleolus can be dynamically altered by genotoxic agents. RNAP1, Fibrillarin, and nucleolar DNA are exported to the periphery of the nucleolus after genotoxic stress and, once DNA repair is fully completed, the organization of the nucleolus is restored. We find that SMN is required for the restoration of the nucleolar structure after genotoxic stress. During DNA repair, SMN shuttles from the Cajal bodies to the nucleolus. This shuttling is important for nucleolar homeostasis and relies on the presence of Coilin and the activity of PRMT1.


Asunto(s)
Atrofia Muscular Espinal , Proteínas de Unión al ARN , Humanos , Proteínas de Unión al ARN/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Nucléolo Celular/metabolismo , Atrofia Muscular Espinal/genética , Atrofia Muscular Espinal/metabolismo , Neuronas Motoras/metabolismo , Proteínas del Complejo SMN/metabolismo , Cuerpos Enrollados/metabolismo , Proteína-Arginina N-Metiltransferasas/metabolismo , Proteínas Represoras/metabolismo
2.
Nat Commun ; 14(1): 6580, 2023 10 18.
Artículo en Inglés | MEDLINE | ID: mdl-37852981

RESUMEN

Spliceosomal snRNPs are multicomponent particles that undergo a complex maturation pathway. Human Sm-class snRNAs are generated as 3'-end extended precursors, which are exported to the cytoplasm and assembled together with Sm proteins into core RNPs by the SMN complex. Here, we provide evidence that these pre-snRNA substrates contain compact, evolutionarily conserved secondary structures that overlap with the Sm binding site. These structural motifs in pre-snRNAs are predicted to interfere with Sm core assembly. We model structural rearrangements that lead to an open pre-snRNA conformation compatible with Sm protein interaction. The predicted rearrangement pathway is conserved in Metazoa and requires an external factor that initiates snRNA remodeling. We show that the essential helicase Gemin3, which is a component of the SMN complex, is crucial for snRNA structural rearrangements during snRNP maturation. The SMN complex thus facilitates ATP-driven structural changes in snRNAs that expose the Sm site and enable Sm protein binding.


Asunto(s)
Precursores del ARN , ARN Nuclear Pequeño , Humanos , ARN Nuclear Pequeño/metabolismo , Proteínas del Complejo SMN/metabolismo , Precursores del ARN/metabolismo , Células HeLa , Ribonucleoproteínas Nucleares Pequeñas/metabolismo , Proteínas Nucleares snRNP/genética
3.
Oncogene ; 42(37): 2751-2763, 2023 09.
Artículo en Inglés | MEDLINE | ID: mdl-37573407

RESUMEN

The nuclear factor erythroid 2-like 2 (NFE2L2; NRF2) signaling pathway is frequently deregulated in human cancers. The critical functions of NRF2, other than its transcriptional activation, in cancers remain largely unknown. Here, we uncovered a previously unrecognized role of NRF2 in the regulation of RNA splicing. Global splicing analysis revealed that NRF2 knockdown in non-small cell lung cancer (NSCLC) A549 cells altered 839 alternative splicing (AS) events in 485 genes. Mechanistic studies demonstrated that NRF2 transcriptionally regulated SMN mRNA expression by binding to two antioxidant response elements in the SMN1 promoter. Post-transcriptionally, NRF2 was physically associated with the SMN protein. The Neh2 domain of NRF2, as well as the YG box and the region encoded by exon 7 of SMN, were required for their interaction. NRF2 formed a complex with SMN and Gemin2 in nuclear gems and Cajal bodies. Furthermore, the NRF2-SMN interaction regulated RNA splicing by expressing SMN in NRF2-knockout HeLa cells, reverting some of the altered RNA splicing. Moreover, SMN overexpression was significantly associated with alterations in the NRF2 pathway in patients with lung squamous cell carcinoma from The Cancer Genome Atlas. Taken together, our findings suggest a novel therapeutic strategy for cancers involving an aberrant NRF2 pathway.


Asunto(s)
Carcinoma de Pulmón de Células no Pequeñas , Neoplasias Pulmonares , Atrofia Muscular Espinal , Humanos , Células HeLa , Carcinoma de Pulmón de Células no Pequeñas/genética , Carcinoma de Pulmón de Células no Pequeñas/metabolismo , Factor 2 Relacionado con NF-E2/genética , Factor 2 Relacionado con NF-E2/metabolismo , Proteínas del Complejo SMN/genética , Proteínas del Complejo SMN/metabolismo , Proteínas de Unión al ARN/genética , Atrofia Muscular Espinal/genética , Atrofia Muscular Espinal/metabolismo , Atrofia Muscular Espinal/terapia , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/metabolismo , Neuronas Motoras/metabolismo , Empalme del ARN/genética , Proteína de Unión a Elemento de Respuesta al AMP Cíclico/metabolismo
4.
Trends Biochem Sci ; 48(8): 689-698, 2023 08.
Artículo en Inglés | MEDLINE | ID: mdl-37156649

RESUMEN

Biomolecular condensates (BMCs) can facilitate or inhibit diverse cellular functions. BMC formation is driven by noncovalent protein-protein, protein-RNA, and RNA-RNA interactions. Here, we focus on Tudor domain-containing proteins - such as survival motor neuron protein (SMN) - that contribute to BMC formation by binding to dimethylarginine (DMA) modifications on protein ligands. SMN is present in RNA-rich BMCs, and its absence causes spinal muscular atrophy (SMA). SMN's Tudor domain forms cytoplasmic and nuclear BMCs, but its DMA ligands are largely unknown, highlighting open questions about the function of SMN. Moreover, DMA modification can alter intramolecular interactions and affect protein localization. Despite these emerging functions, the lack of direct methods of DMA detection remains an obstacle to understanding Tudor-DMA interactions in cells.


Asunto(s)
Proteínas de Unión al ARN , ARN , Ligandos , Proteínas de Unión al ARN/metabolismo , Proteínas del Complejo SMN/metabolismo
5.
Int J Mol Sci ; 24(3)2023 Jan 23.
Artículo en Inglés | MEDLINE | ID: mdl-36768569

RESUMEN

In the cell, RNA exists and functions in a complex with RNA binding proteins (RBPs) that regulate each step of the RNA life cycle from transcription to degradation. Central to this regulation is the role of several molecular chaperones that ensure the correct interactions between RNA and proteins, while aiding the biogenesis of large RNA-protein complexes (ribonucleoproteins or RNPs). Accurate formation of RNPs is fundamentally important to cellular development and function, and its impairment often leads to disease. The survival motor neuron (SMN) protein exemplifies this biological paradigm. SMN is part of a multi-protein complex essential for the biogenesis of various RNPs that function in RNA metabolism. Mutations leading to SMN deficiency cause the neurodegenerative disease spinal muscular atrophy (SMA). A fundamental question in SMA biology is how selective motor system dysfunction results from reduced levels of the ubiquitously expressed SMN protein. Recent clarification of the central role of the SMN complex in RNA metabolism and a thorough characterization of animal models of SMA have significantly advanced our knowledge of the molecular basis of the disease. Here we review the expanding role of SMN in the regulation of gene expression through its multiple functions in RNP biogenesis. We discuss developments in our understanding of SMN activity as a molecular chaperone of RNPs and how disruption of SMN-dependent RNA pathways can contribute to the SMA phenotype.


Asunto(s)
Atrofia Muscular Espinal , Enfermedades Neurodegenerativas , Animales , Neuronas Motoras/metabolismo , Enfermedades Neurodegenerativas/genética , Enfermedades Neurodegenerativas/metabolismo , Proteínas de Unión al ARN/genética , Proteínas de Unión al ARN/metabolismo , Ribonucleoproteínas/genética , Proteínas del Complejo SMN/metabolismo , Atrofia Muscular Espinal/genética , Atrofia Muscular Espinal/metabolismo , ARN/genética , ARN/metabolismo , Chaperonas Moleculares/genética , Chaperonas Moleculares/metabolismo , Proteína 1 para la Supervivencia de la Neurona Motora/genética , Proteína 1 para la Supervivencia de la Neurona Motora/metabolismo
6.
Biomolecules ; 12(10)2022 Oct 20.
Artículo en Inglés | MEDLINE | ID: mdl-36291733

RESUMEN

Survival motor neuron (SMN) is an essential and ubiquitously expressed protein that participates in several aspects of RNA metabolism. SMN deficiency causes a devastating motor neuron disease called spinal muscular atrophy (SMA). SMN forms the core of a protein complex localized at the cytoplasm and nuclear gems and that catalyzes spliceosomal snRNP particle synthesis. In cultured motor neurons, SMN is also present in dendrites and axons, and forms part of the ribonucleoprotein transport granules implicated in mRNA trafficking and local translation. Nevertheless, the distribution, regulation, and role of SMN at the axons and presynaptic motor terminals in vivo are still unclear. By using conventional confocal microscopy and STED super-resolution nanoscopy, we found that SMN appears in the form of granules distributed along motor axons at nerve terminals. Our fluorescence in situ hybridization and electron microscopy studies also confirmed the presence of ß-actin mRNA, ribosomes, and polysomes in the presynaptic motor terminal, key elements of the protein synthesis machinery involved in local translation in this compartment. SMN granules co-localize with the microtubule-associated protein 1B (MAP1B) and neurofilaments, suggesting that the cytoskeleton participates in transporting and positioning the granules. We also found that, while SMN granules are physiologically downregulated at the presynaptic element during the period of postnatal maturation in wild-type (non-transgenic) mice, they accumulate in areas of neurofilament aggregation in SMA mice, suggesting that the high expression of SMN at the NMJ, together with the cytoskeletal defects, contribute to impairing the bi-directional traffic of proteins and organelles between the axon and the presynaptic terminal.


Asunto(s)
Filamentos Intermedios , Atrofia Muscular Espinal , Animales , Ratones , Actinas/metabolismo , Modelos Animales de Enfermedad , Hibridación Fluorescente in Situ , Filamentos Intermedios/metabolismo , Neuronas Motoras/metabolismo , Atrofia Muscular Espinal/genética , Atrofia Muscular Espinal/metabolismo , Ribonucleoproteínas/metabolismo , Ribonucleoproteínas Nucleares Pequeñas/metabolismo , ARN Mensajero/genética , ARN Mensajero/metabolismo , Proteínas del Complejo SMN/genética , Proteínas del Complejo SMN/metabolismo
7.
Nat Commun ; 13(1): 5166, 2022 09 02.
Artículo en Inglés | MEDLINE | ID: mdl-36056043

RESUMEN

Gemin5 in the Survival Motor Neuron (SMN) complex serves as the RNA-binding protein to deliver small nuclear RNAs (snRNAs) to the small nuclear ribonucleoprotein Sm complex via its N-terminal WD40 domain. Additionally, the C-terminal region plays an important role in regulating RNA translation by directly binding to viral RNAs and cellular mRNAs. Here, we present the three-dimensional structure of the Gemin5 C-terminal region, which adopts a homodecamer architecture comprised of a dimer of pentamers. By structural analysis, mutagenesis, and RNA-binding assays, we find that the intact pentamer/decamer is critical for the Gemin5 C-terminal region to bind cognate RNA ligands and to regulate mRNA translation. The Gemin5 high-order architecture is assembled via pentamerization, allowing binding to RNA ligands in a coordinated manner. We propose a model depicting the regulatory role of Gemin5 in selective RNA binding and translation. Therefore, our work provides insights into the SMN complex-independent function of Gemin5.


Asunto(s)
ARN Nuclear Pequeño , Ribonucleoproteínas Nucleares Pequeñas , Ligandos , Unión Proteica , ARN Mensajero/genética , ARN Mensajero/metabolismo , ARN Nuclear Pequeño/metabolismo , Ribonucleoproteínas Nucleares Pequeñas/metabolismo , Proteínas del Complejo SMN/metabolismo
8.
Nat Commun ; 13(1): 5453, 2022 09 16.
Artículo en Inglés | MEDLINE | ID: mdl-36114190

RESUMEN

Survival of motor neuron (SMN) functions in diverse biological pathways via recognition of symmetric dimethylarginine (Rme2s) on proteins by its Tudor domain, and deficiency of SMN leads to spinal muscular atrophy. Here we report a potent and selective antagonist with a 4-iminopyridine scaffold targeting the Tudor domain of SMN. Our structural and mutagenesis studies indicate that both the aromatic ring and imino groups of compound 1 contribute to its selective binding to SMN. Various on-target engagement assays support that compound 1 specifically recognizes SMN in a cellular context and prevents the interaction of SMN with the R1810me2s of RNA polymerase II subunit POLR2A, resulting in transcription termination and R-loop accumulation mimicking SMN depletion. Thus, in addition to the antisense, RNAi and CRISPR/Cas9 techniques, potent SMN antagonists could be used as an efficient tool to understand the biological functions of SMN.


Asunto(s)
ARN Polimerasa II , Proteínas del Complejo SMN , Humanos , Neuronas Motoras/metabolismo , Atrofia Muscular Espinal/metabolismo , ARN Polimerasa II/efectos de los fármacos , ARN Polimerasa II/metabolismo , Proteínas del Complejo SMN/antagonistas & inhibidores , Proteínas del Complejo SMN/efectos de los fármacos , Proteínas del Complejo SMN/metabolismo
9.
Cell Rep ; 40(9): 111288, 2022 08 30.
Artículo en Inglés | MEDLINE | ID: mdl-36044849

RESUMEN

Insulin expression is primarily restricted to the pancreatic ß cells, which are physically or functionally depleted in diabetes. Identifying targetable pathways repressing insulin in non-ß cells, particularly in the developmentally related glucagon-secreting α cells, is an important aim of regenerative medicine. Here, we perform an RNA interference screen in a murine α cell line to identify silencers of insulin expression. We discover that knockdown of the splicing factor Smndc1 triggers a global repression of α cell gene-expression programs in favor of increased ß cell markers. Mechanistically, Smndc1 knockdown upregulates the ß cell transcription factor Pdx1 by modulating the activities of the BAF and Atrx chromatin remodeling complexes. SMNDC1's repressive role is conserved in human pancreatic islets, its loss triggering enhanced insulin secretion and PDX1 expression. Our study identifies Smndc1 as a key factor connecting splicing and chromatin remodeling to the control of insulin expression in human and mouse islet cells.


Asunto(s)
Ensamble y Desensamble de Cromatina , Células Secretoras de Glucagón , Células Secretoras de Insulina , Islotes Pancreáticos , Factores de Empalme de ARN , Empalme del ARN , Proteínas del Complejo SMN , Animales , Células Secretoras de Glucagón/metabolismo , Humanos , Insulina/metabolismo , Secreción de Insulina , Células Secretoras de Insulina/metabolismo , Islotes Pancreáticos/metabolismo , Ratones , Empalme del ARN/genética , Factores de Empalme de ARN/metabolismo , Proteínas del Complejo SMN/metabolismo , Factores de Transcripción/metabolismo
10.
Life Sci Alliance ; 5(7)2022 07.
Artículo en Inglés | MEDLINE | ID: mdl-35393353

RESUMEN

Dysfunction of RNA-binding proteins is often linked to a wide range of human disease, particularly with neurological conditions. Gemin5 is a member of the survival of the motor neurons (SMN) complex, a ribosome-binding protein and a translation reprogramming factor. Recently, pathogenic mutations in Gemin5 have been reported, but the functional consequences of these variants remain elusive. Here, we report functional and structural deficiencies associated with compound heterozygosity variants within the Gemin5 gene found in patients with neurodevelopmental disorders. These clinical variants are located in key domains of Gemin5, the tetratricopeptide repeat (TPR)-like dimerization module and the noncanonical RNA-binding site 1 (RBS1). We show that the TPR-like variants disrupt protein dimerization, whereas the RBS1 variant confers protein instability. All mutants are defective in the interaction with protein networks involved in translation and RNA-driven pathways. Importantly, the TPR-like variants fail to associate with native ribosomes, hampering its involvement in translation control and establishing a functional difference with the wild-type protein. Our study provides insights into the molecular basis of disease associated with malfunction of the Gemin5 protein.


Asunto(s)
Enfermedades del Sistema Nervioso , Proteínas de Unión al ARN , Ribosomas , Humanos , Enfermedades del Sistema Nervioso/genética , Enfermedades del Sistema Nervioso/metabolismo , ARN/genética , Proteínas de Unión al ARN/genética , Proteínas de Unión al ARN/metabolismo , Ribosomas/genética , Ribosomas/metabolismo , Proteínas del Complejo SMN/genética , Proteínas del Complejo SMN/metabolismo
11.
Neuroscience ; 491: 32-42, 2022 05 21.
Artículo en Inglés | MEDLINE | ID: mdl-35314252

RESUMEN

Increasing evidence points to the involvement of cell types other than motor neurons in both amyotrophic lateral sclerosis (ALS) and spinal muscular atrophy (SMA), the predominant motor neuron disease in adults and infants, respectively. The contribution of glia to ALS pathophysiology is well documented. Studies have since focused on evaluating the contribution of glia in SMA. Here, we made use of the Drosophila model to ask whether the survival motor neuron (Smn) protein, the causative factor for SMA, is required selectively in glia. We show that the specific loss of Smn function in glia during development reduced survival to adulthood but did not affect motoric performance or neuromuscular junction (NMJ) morphology in flies. In contrast, gain rather than loss of ALS-linked TDP-43, FUS or C9orf72 function in glia induced significant defects in motor behaviour in addition to reduced survival. Furthermore, glia-specific gain of TDP-43 function caused both NMJ defects and muscle atrophy. Smn together with Gemins 2-8 and Unrip, form the Smn complex which is indispensable for the assembly of spliceosomal small nuclear ribonucleoproteins (snRNPs). We show that glial-selective perturbation of Smn complex components or disruption of key snRNP biogenesis factors pICln and Tgs1, induce deleterious effects on adult fly viability but, similar to Smn reduction, had no negative effect on neuromuscular function. Our findings suggest that the role of Smn in snRNP biogenesis as part of the Smn complex is required in glia for the survival of the organism, underscoring the importance of glial cells in SMA disease formation.


Asunto(s)
Atrofia Muscular Espinal , Envejecimiento , Animales , Proteínas de Unión al ADN/metabolismo , Drosophila/metabolismo , Neuronas Motoras/fisiología , Neuroglía/metabolismo , Ribonucleoproteínas Nucleares Pequeñas/metabolismo , Proteínas del Complejo SMN/metabolismo , Proteína 1 para la Supervivencia de la Neurona Motora/metabolismo
12.
Cells ; 10(11)2021 11 04.
Artículo en Inglés | MEDLINE | ID: mdl-34831238

RESUMEN

Increasing evidence points to the Survival Motor Neuron (SMN) protein as a key determinant of translation pathway. Besides its role in RNA processing and sorting, several works support a critical implication of SMN in ribosome biogenesis. We previously showed that SMN binds ribosomal proteins (RPs) as well as their encoding transcripts, ensuring an appropriate level of locally synthesized RPs. SMN impacts the translation machinery in both neural and non-neural cells, in agreement with the concept that SMN is an essential protein in all cell types. Here, we further assessed the relationship between SMN and translation-related factors in immortalized human fibroblasts. We focused on SMN-nucleolin interaction, keeping in mind that nucleolin is an RNA-binding protein, highly abundant within the nucleolus, that exhibits a central role in ribosomes production. Nucleolin may also affects translation network by binding the mammalian target of rapamycin (mTOR) mRNA and promoting its local synthesis. In this regard, for the first time we provided evidence that SMN protein itself associates with mTOR transcript. Collectively, we found that: (1) SMN coexists with nucleolin-mTOR mRNA complexes at subcellular level; (2) SMN deficiency impairs nucleolar compartmentalization of nucleolin, and (3) this event correlates with the nuclear retention of mTOR mRNA. These findings suggest that SMN may regulate not only structural components of translation machinery, but also their upstream regulating factors.


Asunto(s)
Fosfoproteínas/metabolismo , Proteínas de Unión al ARN/metabolismo , Proteínas del Complejo SMN/metabolismo , Serina-Treonina Quinasas TOR/genética , Línea Celular , Nucléolo Celular/metabolismo , Técnicas de Silenciamiento del Gen , Humanos , Unión Proteica , Biosíntesis de Proteínas , ARN Mensajero/genética , ARN Mensajero/metabolismo , Proteínas del Complejo SMN/deficiencia , Fracciones Subcelulares/metabolismo , Serina-Treonina Quinasas TOR/metabolismo , Nucleolina
13.
Nat Commun ; 12(1): 5040, 2021 08 19.
Artículo en Inglés | MEDLINE | ID: mdl-34413305

RESUMEN

SMN is a ubiquitously expressed protein and is essential for life. SMN deficiency causes the neurodegenerative disease spinal muscular atrophy (SMA), the leading genetic cause of infant mortality. SMN interacts with itself and other proteins to form a complex that functions in the assembly of ribonucleoproteins. SMN is modified by SUMO (Small Ubiquitin-like Modifier), but whether sumoylation is required for the functions of SMN that are relevant to SMA pathogenesis is not known. Here, we show that inactivation of a SUMO-interacting motif (SIM) alters SMN sub-cellular distribution, the integrity of its complex, and its function in small nuclear ribonucleoproteins biogenesis. Expression of a SIM-inactivated mutant of SMN in a mouse model of SMA slightly extends survival rate with limited and transient correction of motor deficits. Remarkably, although SIM-inactivated SMN attenuates motor neuron loss and improves neuromuscular junction synapses, it fails to prevent the loss of sensory-motor synapses. These findings suggest that sumoylation is important for proper assembly and function of the SMN complex and that loss of this post-translational modification impairs the ability of SMN to correct selective deficits in the sensory-motor circuit of SMA mice.


Asunto(s)
Neuronas Motoras/metabolismo , Atrofia Muscular Espinal/patología , Enfermedades Neurodegenerativas/patología , Ribonucleoproteínas Nucleares Pequeñas/metabolismo , Proteínas del Complejo SMN/metabolismo , Sumoilación , Sinapsis/metabolismo , Animales , Animales Modificados Genéticamente , Células Cultivadas , Modelos Animales de Enfermedad , Humanos , Ratones , Neuronas Motoras/patología , Atrofia Muscular Espinal/metabolismo , Enfermedades Neurodegenerativas/metabolismo , Sinapsis/patología , Pez Cebra
14.
RNA Biol ; 18(sup1): 496-506, 2021 10 15.
Artículo en Inglés | MEDLINE | ID: mdl-34424823

RESUMEN

Gemin5 is a multifaceted RNA-binding protein that comprises distinct structural domains, including a WD40 and TPR-like for which the X-ray structure is known. In addition, the protein contains a non-canonical RNA-binding domain (RBS1) towards the C-terminus. To understand the RNA binding features of the RBS1 domain, we have characterized its structural characteristics by solution NMR linked to RNA-binding activity. Here we show that a short version of the RBS1 domain that retains the ability to interact with RNA is predominantly unfolded even in the presence of RNA. Furthermore, an exhaustive mutational analysis indicates the presence of an evolutionarily conserved motif enriched in R, S, W, and H residues, necessary to promote RNA-binding via π-π interactions. The combined results of NMR and RNA-binding on wild-type and mutant proteins highlight the importance of aromatic and arginine residues for RNA recognition by RBS1, revealing that the net charge and the π-amino acid density of this region of Gemin5 are key factors for RNA recognition.


Asunto(s)
Arginina/metabolismo , Motivos de Unión al ARN , ARN/química , ARN/metabolismo , Proteínas del Complejo SMN/química , Proteínas del Complejo SMN/metabolismo , Triptófano/metabolismo , Secuencia de Aminoácidos , Arginina/química , Arginina/genética , Sitios de Unión , Humanos , Modelos Moleculares , Unión Proteica , ARN/genética , Proteínas del Complejo SMN/genética , Homología de Secuencia , Triptófano/química , Triptófano/genética
15.
Cell Rep ; 35(12): 109277, 2021 06 22.
Artículo en Inglés | MEDLINE | ID: mdl-34161763

RESUMEN

The activity of the SMN complex in promoting the assembly of pre-mRNA processing UsnRNPs correlates with condensation of the complex in nuclear Cajal bodies. While mechanistic details of its activity have been elucidated, the molecular basis for condensation remains unclear. High SMN complex phosphorylation suggests extensive regulation. Here, we report on systematic siRNA-based screening for modulators of the capacity of SMN to condense in Cajal bodies and identify mTOR and ribosomal protein S6 kinase ß-1 as key regulators. Proteomic analysis reveals TOR-dependent phosphorylations in SMN complex subunits. Using stably expressed or optogenetically controlled phospho mutants, we demonstrate that serine 49 and 63 phosphorylation of human SMN controls the capacity of the complex to condense in Cajal bodies via liquid-liquid phase separation. Our findings link SMN complex condensation and UsnRNP biogenesis to cellular energy levels and suggest modulation of TOR signaling as a rational concept for therapy of the SMN-linked neuromuscular disorder spinal muscular atrophy.


Asunto(s)
Ribonucleoproteínas Nucleares Pequeñas/biosíntesis , Proteínas del Complejo SMN/metabolismo , Transducción de Señal , Serina-Treonina Quinasas TOR/metabolismo , Núcleo Celular/metabolismo , Células HeLa , Humanos , Mutación/genética , Fosforilación , Fosfoserina/metabolismo , Multimerización de Proteína , Proteómica , Reproducibilidad de los Resultados , Ribonucleoproteínas Nucleares Pequeñas/metabolismo , Proteínas Quinasas S6 Ribosómicas 70-kDa/metabolismo
16.
Cell ; 184(14): 3612-3625.e17, 2021 07 08.
Artículo en Inglés | MEDLINE | ID: mdl-34115980

RESUMEN

Biomolecular condensation is a widespread mechanism of cellular compartmentalization. Because the "survival of motor neuron protein" (SMN) is implicated in the formation of three different membraneless organelles (MLOs), we hypothesized that SMN promotes condensation. Unexpectedly, we found that SMN's globular tudor domain was sufficient for dimerization-induced condensation in vivo, whereas its two intrinsically disordered regions (IDRs) were not. Binding to dimethylarginine (DMA) modified protein ligands was required for condensate formation by the tudor domains in SMN and at least seven other fly and human proteins. Remarkably, asymmetric versus symmetric DMA determined whether two distinct nuclear MLOs-gems and Cajal bodies-were separate or "docked" to one another. This substructure depended on the presence of either asymmetric or symmetric DMA as visualized with sub-diffraction microscopy. Thus, DMA-tudor interaction modules-combinations of tudor domains bound to their DMA ligand(s)-represent versatile yet specific regulators of MLO assembly, composition, and morphology.


Asunto(s)
Arginina/análogos & derivados , Condensados Biomoleculares/metabolismo , Proteínas del Complejo SMN/química , Proteínas del Complejo SMN/metabolismo , Animales , Arginina/metabolismo , Núcleo Celular/metabolismo , Cuerpos Enrollados/metabolismo , Drosophila melanogaster/metabolismo , Células HEK293 , Células HeLa , Humanos , Ligandos , Metilación , Ratones , Modelos Biológicos , Células 3T3 NIH , Unión Proteica , Dominios Proteicos , Multimerización de Proteína , Ribonucleoproteínas Nucleares Pequeñas/metabolismo
17.
Nat Commun ; 12(1): 2558, 2021 05 07.
Artículo en Inglés | MEDLINE | ID: mdl-33963192

RESUMEN

GEMIN5, an RNA-binding protein is essential for assembly of the survival motor neuron (SMN) protein complex and facilitates the formation of small nuclear ribonucleoproteins (snRNPs), the building blocks of spliceosomes. Here, we have identified 30 affected individuals from 22 unrelated families presenting with developmental delay, hypotonia, and cerebellar ataxia harboring biallelic variants in the GEMIN5 gene. Mutations in GEMIN5 perturb the subcellular distribution, stability, and expression of GEMIN5 protein and its interacting partners in patient iPSC-derived neurons, suggesting a potential loss-of-function mechanism. GEMIN5 mutations result in disruption of snRNP complex assembly formation in patient iPSC neurons. Furthermore, knock down of rigor mortis, the fly homolog of human GEMIN5, leads to developmental defects, motor dysfunction, and a reduced lifespan. Interestingly, we observed that GEMIN5 variants disrupt a distinct set of transcripts and pathways as compared to SMA patient neurons, suggesting different molecular pathomechanisms. These findings collectively provide evidence that pathogenic variants in GEMIN5 perturb physiological functions and result in a neurodevelopmental delay and ataxia syndrome.


Asunto(s)
Regulación del Desarrollo de la Expresión Génica/genética , Células Madre Pluripotentes Inducidas/metabolismo , Trastornos del Neurodesarrollo/metabolismo , Neuronas/metabolismo , Ribonucleoproteínas Nucleares Pequeñas/metabolismo , Proteínas del Complejo SMN/genética , Alelos , Secuencia de Aminoácidos , Animales , Preescolar , Discapacidades del Desarrollo/genética , Drosophila/genética , Drosophila/crecimiento & desarrollo , Femenino , Técnicas de Silenciamiento del Gen , Ontología de Genes , Células HEK293 , Humanos , Mutación con Pérdida de Función , Masculino , Hipotonía Muscular/genética , Disinergia Cerebelosa Mioclónica/genética , Trastornos del Neurodesarrollo/diagnóstico por imagen , Trastornos del Neurodesarrollo/genética , Trastornos del Neurodesarrollo/fisiopatología , Linaje , Polimorfismo de Nucleótido Simple , RNA-Seq , Ribonucleoproteínas Nucleares Pequeñas/genética , Rigor Mortis/genética , Proteínas del Complejo SMN/metabolismo
18.
Nucleic Acids Res ; 49(13): 7207-7223, 2021 07 21.
Artículo en Inglés | MEDLINE | ID: mdl-33754639

RESUMEN

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.


Asunto(s)
Proteínas del Complejo SMN/química , Proteínas de Schizosaccharomyces pombe/química , Proteínas Portadoras/química , Cristalografía por Rayos X , Humanos , Modelos Moleculares , Atrofia Muscular Espinal/genética , Mutación , Proteínas Nucleares/química , Unión Proteica , Proteínas del Complejo SMN/metabolismo , Dispersión del Ángulo Pequeño , Proteínas de Schizosaccharomyces pombe/metabolismo , Homología Estructural de Proteína , Difracción de Rayos X
19.
Nat Commun ; 12(1): 1278, 2021 02 24.
Artículo en Inglés | MEDLINE | ID: mdl-33627647

RESUMEN

Gene expression requires tight coordination of the molecular machineries that mediate transcription and splicing. While the interplay between transcription kinetics and spliceosome fidelity has been investigated before, less is known about mechanisms regulating the assembly of the spliceosomal machinery in response to transcription changes. Here, we report an association of the Smn complex, which mediates spliceosomal snRNP biogenesis, with the 7SK complex involved in transcriptional regulation. We found that Smn interacts with the 7SK core components Larp7 and Mepce and specifically associates with 7SK subcomplexes containing hnRNP R. The association between Smn and 7SK complexes is enhanced upon transcriptional inhibition leading to reduced production of snRNPs. Taken together, our findings reveal a functional association of Smn and 7SK complexes that is governed by global changes in transcription. Thus, in addition to its canonical nuclear role in transcriptional regulation, 7SK has cytosolic functions in fine-tuning spliceosome production according to transcriptional demand.


Asunto(s)
ARN Largo no Codificante/metabolismo , Ribonucleoproteínas Nucleares Pequeñas/metabolismo , Proteínas del Complejo SMN/metabolismo , Animales , Células Cultivadas , Células HEK293 , Células HeLa , Humanos , Ratones , Neuronas Motoras/metabolismo , ARN Largo no Codificante/genética , Ribonucleoproteínas Nucleares Pequeñas/genética , Proteínas del Complejo SMN/genética , Espectrometría de Masas en Tándem , Transcripción Genética/genética
20.
Int J Biochem Cell Biol ; 132: 105919, 2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-33422691

RESUMEN

The AAA-ATPase NVL2 associates with an RNA helicase MTR4 and the nuclear RNA exosome in the course of ribosome biogenesis. In our proteomic screen, we had identified a ribosome biogenesis factor WDR74 as a MTR4-interacting partner, whose dissociation is stimulated by the ATP hydrolysis of NVL2. In this study, we report the identification of splicing factor 30 (SPF30), another MTR4-interacting protein with a similar regulatory mechanism. SPF30 is a pre-mRNA splicing factor harboring a Tudor domain in its central region, which regulates various cellular events by binding to dimethylarginine-modified proteins. The interaction between SPF30 and the exosome core is mediated by MTR4 and RRP6, a catalytic component of the nuclear exosome. The N- and C-terminal regions, but not the Tudor domain, of SPF30 are involved in the association with MTR4 and the exosome. The knockdown of SPF30 caused subtle delay in the 12S pre-rRNA processing to mature 5.8S rRNA, even though no obvious effect was observed on the ribosome subunit profile in the cells. Shotgun proteomic analysis to search for SPF30-interacting proteins indicated its role in ribosome biogenesis, pre-mRNA splicing, and box C/D snoRNA biogenesis. These results suggest that SPF30 collaborates with the MTR4-exosome machinery to play a functional role in multiple RNA metabolic pathways, some of which may be regulated by the ATP hydrolysis of NVL2.


Asunto(s)
ATPasas Asociadas con Actividades Celulares Diversas/metabolismo , Exosomas/genética , ARN Helicasas/metabolismo , Factores de Empalme de ARN/química , Factores de Empalme de ARN/metabolismo , Estabilidad del ARN , Proteínas del Complejo SMN/química , Proteínas del Complejo SMN/metabolismo , Humanos , Unión Proteica , Dominios Proteicos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...