Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 494
Filtrar
1.
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
2.
Cell ; 175(5): 1405-1417.e14, 2018 11 15.
Artículo en Inglés | MEDLINE | ID: mdl-30318144

RESUMEN

Programmable control of spatial genome organization is a powerful approach for studying how nuclear structure affects gene regulation and cellular function. Here, we develop a versatile CRISPR-genome organization (CRISPR-GO) system that can efficiently control the spatial positioning of genomic loci relative to specific nuclear compartments, including the nuclear periphery, Cajal bodies, and promyelocytic leukemia (PML) bodies. CRISPR-GO is chemically inducible and reversible, enabling interrogation of real-time dynamics of chromatin interactions with nuclear compartments in living cells. Inducible repositioning of genomic loci to the nuclear periphery allows for dissection of mitosis-dependent and -independent relocalization events and also for interrogation of the relationship between gene position and gene expression. CRISPR-GO mediates rapid de novo formation of Cajal bodies at desired chromatin loci and causes significant repression of endogenous gene expression over long distances (30-600 kb). The CRISPR-GO system offers a programmable platform to investigate large-scale spatial genome organization and function.


Asunto(s)
Sistemas CRISPR-Cas/genética , Edición Génica/métodos , Genoma , Ácido Abscísico/farmacología , Línea Celular Tumoral , Cromatina/genética , Cromatina/metabolismo , Cuerpos Enrollados/genética , Regulación de la Expresión Génica , Sitios Genéticos , Humanos , Hibridación Fluorescente in Situ , Puntos de Control de la Fase S del Ciclo Celular/efectos de los fármacos
3.
Cell ; 166(5): 1188-1197.e9, 2016 Aug 25.
Artículo en Inglés | MEDLINE | ID: mdl-27523609

RESUMEN

Telomerase maintains genome integrity by adding repetitive DNA sequences to the chromosome ends in actively dividing cells, including 90% of all cancer cells. Recruitment of human telomerase to telomeres occurs during S-phase of the cell cycle, but the molecular mechanism of the process is only partially understood. Here, we use CRISPR genome editing and single-molecule imaging to track telomerase trafficking in nuclei of living human cells. We demonstrate that telomerase uses three-dimensional diffusion to search for telomeres, probing each telomere thousands of times each S-phase but only rarely forming a stable association. Both the transient and stable association events depend on the direct interaction of the telomerase protein TERT with the telomeric protein TPP1. Our results reveal that telomerase recruitment to telomeres is driven by dynamic interactions between the rapidly diffusing telomerase and the chromosome end.


Asunto(s)
Telomerasa/metabolismo , Telómero/enzimología , Transporte Activo de Núcleo Celular , Proteínas Bacterianas , Proteína 9 Asociada a CRISPR , Línea Celular , Núcleo Celular/enzimología , Repeticiones Palindrómicas Cortas Agrupadas y Regularmente Espaciadas , Cuerpos Enrollados/enzimología , Endonucleasas , Edición Génica , Genoma Humano , Células HeLa , Humanos , Imagenología Tridimensional , Dominios Proteicos , Fase S , Saccharomyces cerevisiae/enzimología , Saccharomyces cerevisiae/genética , Complejo Shelterina , Telomerasa/química , Telómero/química , Homeostasis del Telómero , Proteínas de Unión a Telómeros/química , Proteínas de Unión a Telómeros/metabolismo
4.
Mol Cell ; 79(1): 115-126.e6, 2020 07 02.
Artículo en Inglés | MEDLINE | ID: mdl-32497497

RESUMEN

Extension of telomeres is a critical step in the immortalization of cancer cells. This complex reaction requires proper spatiotemporal coordination of telomerase and telomeres and remains poorly understood at the cellular level. To understand how cancer cells execute this process, we combine CRISPR genome editing and MS2 RNA tagging to image single molecules of telomerase RNA (hTR). Real-time dynamics and photoactivation experiments of hTR in Cajal bodies (CBs) reveal that hTERT controls the exit of hTR from CBs. Single-molecule tracking of hTR at telomeres shows that TPP1-mediated recruitment results in short telomere-telomerase scanning interactions, and then base pairing between hTR and telomere ssDNA promotes long interactions required for stable telomerase retention. Interestingly, POT1 OB-fold mutations that result in abnormally long telomeres in cancers act by enhancing this retention step. In summary, single-molecule imaging unveils the life cycle of telomerase RNA and provides a framework to reveal how cancer-associated mutations mechanistically drive defects in telomere homeostasis.


Asunto(s)
Cuerpos Enrollados/metabolismo , ADN de Cadena Simple/metabolismo , ARN/metabolismo , Imagen Individual de Molécula/métodos , Telomerasa/metabolismo , Homeostasis del Telómero , Telómero/metabolismo , Repeticiones Palindrómicas Cortas Agrupadas y Regularmente Espaciadas , ADN de Cadena Simple/genética , Edición Génica , Células HeLa , Humanos , Mutación , ARN/genética , Complejo Shelterina , Telomerasa/genética , Telómero/genética , Proteínas de Unión a Telómeros/genética , Proteínas de Unión a Telómeros/metabolismo
5.
Genes Dev ; 34(1-2): 118-131, 2020 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-31831626

RESUMEN

Small RNAs derived from mature tRNAs, referred to as tRNA fragments or "tRFs," are an emerging class of regulatory RNAs with poorly understood functions. We recently identified a role for one specific tRF-5' tRF-Gly-GCC, or tRF-GG-as a repressor of genes associated with the endogenous retroelement MERVL, but the mechanistic basis for this regulation was unknown. Here, we show that tRF-GG plays a role in production of a wide variety of noncoding RNAs-snoRNAs, scaRNAs, and snRNAs-that are dependent on Cajal bodies for stability and activity. Among these noncoding RNAs, regulation of the U7 snRNA by tRF-GG modulates heterochromatin-mediated transcriptional repression of MERVL elements by supporting an adequate supply of histone proteins. Importantly, the effects of inhibiting tRF-GG on histone mRNA levels, on activity of a histone 3' UTR reporter, and ultimately on MERVL regulation could all be suppressed by manipulating U7 RNA levels. We additionally show that the related RNA-binding proteins hnRNPF and hnRNPH bind directly to tRF-GG, and are required for Cajal body biogenesis, positioning these proteins as strong candidates for effectors of tRF-GG function in vivo. Together, our data reveal a conserved mechanism for 5' tRNA fragment control of noncoding RNA biogenesis and, consequently, global chromatin organization.


Asunto(s)
Regulación de la Expresión Génica , Histonas/genética , Histonas/metabolismo , ARN de Transferencia/metabolismo , ARN no Traducido/genética , ARN no Traducido/metabolismo , Animales , Línea Celular , Cuerpos Enrollados/metabolismo , Células Madre Embrionarias Humanas , Humanos , Ratones , Unión Proteica , ARN Nuclear Pequeño/genética , Retroelementos/genética
6.
Cell ; 150(3): 481-94, 2012 Aug 03.
Artículo en Inglés | MEDLINE | ID: mdl-22863003

RESUMEN

Telomere synthesis in cancer cells and stem cells involves trafficking of telomerase to Cajal bodies, and telomerase is thought to be recruited to telomeres through interactions with telomere-binding proteins. Here, we show that the OB-fold domain of the telomere-binding protein TPP1 recruits telomerase to telomeres through an association with the telomerase reverse transcriptase TERT. When tethered away from telomeres and other telomere-binding proteins, the TPP1 OB-fold domain is sufficient to recruit telomerase to a heterologous chromatin locus. Expression of a minimal TPP1 OB-fold inhibits telomere maintenance by blocking access of telomerase to its cognate binding site at telomeres. We identify amino acids required for the TPP1-telomerase interaction, including specific loop residues within the TPP1 OB-fold domain and individual residues within TERT, some of which are mutated in a subset of pulmonary fibrosis patients. These data define a potential interface for telomerase-TPP1 interaction required for telomere maintenance and implicate defective telomerase recruitment in telomerase-related disease.


Asunto(s)
Telomerasa/metabolismo , Proteínas de Unión a Telómeros/química , Proteínas de Unión a Telómeros/metabolismo , Telómero/metabolismo , Secuencia de Aminoácidos , Línea Celular , Línea Celular Tumoral , Cuerpos Enrollados/metabolismo , Humanos , Fibrosis Pulmonar Idiopática/genética , Fibrosis Pulmonar Idiopática/metabolismo , Modelos Moleculares , Datos de Secuencia Molecular , Mutación , Estructura Terciaria de Proteína , Saccharomyces cerevisiae/metabolismo , Alineación de Secuencia , Complejo Shelterina , Telomerasa/química , Telomerasa/genética , Proteínas de Unión a Telómeros/genética
7.
Genes Dev ; 33(13-14): 739-740, 2019 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-31262844

RESUMEN

Box C/D small nucleolar RNAs (snoRNAs) and small Cajal body (CB) RNAs (scaRNAs) form ribonucleoprotein (RNP) complexes to mediate 2'-O-methylation of rRNAs and small nuclear RNAs (snRNAs), respectively. The site of methylation is determined by antisense elements in the box C/D RNAs that are complementary to sequences in target RNAs. However, numerous box C/D RNAs in mammalian cells lack antisense elements to rRNAs or snRNAs; thus, their targets remain unknown. In this issue of Genes & Development, Vitali and Kiss (pp. 741-746) demonstrate that "orphan" nucleolar box C/D snoRNA SNORD97 and CB box C/D scaRNA SCARNA97 contain antisense elements that target the wobble cytidine at position 34 of human elongator tRNAMet(CAT) for 2'-O-methylation (C34m). C34m is jointly mediated by SNORD97 and SCARNA97 despite their apparently different intranuclear locations. Furthermore, the investigators demonstrate that C34m prohibits site-specific cleavage of tRNAMet (CAT) into tRNA fragments (tRFs) by the stress-responsive endoribonuclease angiogenin, thereby uncovering a role for SNORD97 and SCARNA97 in the biogenesis of tRFs, which modulate a diverse set of cellular functions in human health and disease.


Asunto(s)
ARN de Transferencia de Metionina , Ribonucleoproteínas , Animales , Cuerpos Enrollados , Citidina , Humanos , Metilación , ARN Nucleolar Pequeño
8.
Genes Dev ; 33(13-14): 741-746, 2019 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-31171702

RESUMEN

Site-specific 2'-O-ribose methylation of mammalian rRNAs and RNA polymerase II-synthesized spliceosomal small nuclear RNAs (snRNAs) is mediated by small nucleolar and small Cajal body (CB)-specific box C/D ribonucleoprotein particles (RNPs) in the nucleolus and the nucleoplasmic CBs, respectively. Here, we demonstrate that 2'-O-methylation of the C34 wobble cytidine of human elongator tRNAMet(CAT) is achieved by collaboration of a nucleolar and a CB-specific box C/D RNP carrying the SNORD97 and SCARNA97 box C/D 2'-O-methylation guide RNAs. Methylation of C34 prevents site-specific cleavage of tRNAMet(CAT) by the stress-induced endoribonuclease angiogenin, implicating box C/D guide RNPs in controlling stress-responsive production of putative regulatory tRNA fragments.


Asunto(s)
Nucléolo Celular/metabolismo , Cuerpos Enrollados/metabolismo , Citidina/metabolismo , ARN de Transferencia/metabolismo , Ribonucleoproteínas/metabolismo , Línea Celular , Regulación de la Expresión Génica , Células HeLa , Humanos , Metilación , ARN Nucleolar Pequeño/genética , ARN Nucleolar Pequeño/metabolismo , ARN de Transferencia/genética , Ribonucleasa Pancreática/metabolismo , Ribonucleoproteínas/genética , Estrés Fisiológico
9.
Plant Cell ; 35(9): 3214-3235, 2023 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-37202374

RESUMEN

Proper orchestration of the thousands of biochemical processes that are essential to the life of every cell requires highly organized cellular compartmentalization of dedicated microenvironments. There are 2 ways to create this intracellular segregation to optimize cellular function. One way is to create specific organelles, enclosed spaces bounded by lipid membranes that regulate macromolecular flux in and out of the compartment. A second way is via membraneless biomolecular condensates that form due to to liquid-liquid phase separation. Although research on these membraneless condensates has historically been performed using animal and fungal systems, recent studies have explored basic principles governing the assembly, properties, and functions of membraneless compartments in plants. In this review, we discuss how phase separation is involved in a variety of key processes occurring in Cajal bodies (CBs), a type of biomolecular condensate found in nuclei. These processes include RNA metabolism, formation of ribonucleoproteins involved in transcription, RNA splicing, ribosome biogenesis, and telomere maintenance. Besides these primary roles of CBs, we discuss unique plant-specific functions of CBs in RNA-based regulatory pathways such as nonsense-mediated mRNA decay, mRNA retention, and RNA silencing. Finally, we summarize recent progress and discuss the functions of CBs in responses to pathogen attacks and abiotic stresses, responses that may be regulated via mechanisms governed by polyADP-ribosylation. Thus, plant CBs are emerging as highly complex and multifunctional biomolecular condensates that are involved in a surprisingly diverse range of molecular mechanisms that we are just beginning to appreciate.


Asunto(s)
Condensados Biomoleculares , Cuerpos Enrollados , Animales , Cuerpos Enrollados/genética , Cuerpos Enrollados/metabolismo , Núcleo Celular/metabolismo , ARN , Empalme del ARN
10.
Nature ; 581(7807): 209-214, 2020 05.
Artículo en Inglés | MEDLINE | ID: mdl-32405004

RESUMEN

Intracellular bodies such as nucleoli, Cajal bodies and various signalling assemblies represent membraneless organelles, or condensates, that form via liquid-liquid phase separation (LLPS)1,2. Biomolecular interactions-particularly homotypic interactions mediated by self-associating intrinsically disordered protein regions-are thought to underlie the thermodynamic driving forces for LLPS, forming condensates that can facilitate the assembly and processing of biochemically active complexes, such as ribosomal subunits within the nucleolus. Simplified model systems3-6 have led to the concept that a single fixed saturation concentration is a defining feature of endogenous LLPS7-9, and has been suggested as a mechanism for intracellular concentration buffering2,7,8,10. However, the assumption of a fixed saturation concentration remains largely untested within living cells, in which the richly multicomponent nature of condensates could complicate this simple picture. Here we show that heterotypic multicomponent interactions dominate endogenous LLPS, and give rise to nucleoli and other condensates that do not exhibit a fixed saturation concentration. As the concentration of individual components is varied, their partition coefficients change in a manner that can be used to determine the thermodynamic free energies that underlie LLPS. We find that heterotypic interactions among protein and RNA components stabilize various archetypal intracellular condensates-including the nucleolus, Cajal bodies, stress granules and P-bodies-implying that the composition of condensates is finely tuned by the thermodynamics of the underlying biomolecular interaction network. In the context of RNA-processing condensates such as the nucleolus, this manifests in the selective exclusion of fully assembled ribonucleoprotein complexes, providing a thermodynamic basis for vectorial ribosomal RNA flux out of the nucleolus. This methodology is conceptually straightforward and readily implemented, and can be broadly used to extract thermodynamic parameters from microscopy images. These approaches pave the way for a deeper understanding of the thermodynamics of multicomponent intracellular phase behaviour and its interplay with the nonequilibrium activity that is characteristic of endogenous condensates.


Asunto(s)
Espacio Intracelular/química , Espacio Intracelular/metabolismo , Orgánulos/química , Orgánulos/metabolismo , Termodinámica , Proteínas Adaptadoras Transductoras de Señales/deficiencia , Nucléolo Celular/química , Nucléolo Celular/metabolismo , Cuerpos Enrollados/química , Cuerpos Enrollados/metabolismo , Gránulos Citoplasmáticos/química , Gránulos Citoplasmáticos/metabolismo , ADN Helicasas/deficiencia , Células HeLa , Humanos , Proteínas Nucleares/química , Proteínas Nucleares/metabolismo , Nucleofosmina , Transición de Fase , Proteínas de Unión a Poli-ADP-Ribosa/deficiencia , ARN Helicasas/deficiencia , Proteínas con Motivos de Reconocimiento de ARN/deficiencia , ARN Ribosómico/química , ARN Ribosómico/metabolismo , Proteínas de Unión al ARN , Ribosomas/química , Ribosomas/metabolismo
11.
Nucleic Acids Res ; 52(7): 4037-4052, 2024 Apr 24.
Artículo en Inglés | MEDLINE | ID: mdl-38499487

RESUMEN

Here, we identify RBM41 as a novel unique protein component of the minor spliceosome. RBM41 has no previously recognized cellular function but has been identified as a paralog of U11/U12-65K, a known unique component of the U11/U12 di-snRNP. Both proteins use their highly similar C-terminal RRMs to bind to 3'-terminal stem-loops in U12 and U6atac snRNAs with comparable affinity. Our BioID data indicate that the unique N-terminal domain of RBM41 is necessary for its association with complexes containing DHX8, an RNA helicase, which in the major spliceosome drives the release of mature mRNA from the spliceosome. Consistently, we show that RBM41 associates with excised U12-type intron lariats, is present in the U12 mono-snRNP, and is enriched in Cajal bodies, together suggesting that RBM41 functions in the post-splicing steps of the minor spliceosome assembly/disassembly cycle. This contrasts with U11/U12-65K, which uses its N-terminal region to interact with U11 snRNP during intron recognition. Finally, while RBM41 knockout cells are viable, they show alterations in U12-type 3' splice site usage. Together, our results highlight the role of the 3'-terminal stem-loop of U12 snRNA as a dynamic binding platform for the U11/U12-65K and RBM41 proteins, which function at distinct stages of the assembly/disassembly cycle.


Asunto(s)
ARN Helicasas DEAD-box , Factores de Empalme de ARN , ARN Nuclear Pequeño , Proteínas de Unión al ARN , Ribonucleoproteínas Nucleares Pequeñas , Empalmosomas , Empalmosomas/metabolismo , Empalmosomas/genética , Ribonucleoproteínas Nucleares Pequeñas/metabolismo , Ribonucleoproteínas Nucleares Pequeñas/genética , Ribonucleoproteínas Nucleares Pequeñas/química , Proteínas de Unión al ARN/metabolismo , Proteínas de Unión al ARN/genética , Proteínas de Unión al ARN/química , Humanos , ARN Nuclear Pequeño/metabolismo , ARN Nuclear Pequeño/genética , ARN Nuclear Pequeño/química , ARN Helicasas DEAD-box/metabolismo , ARN Helicasas DEAD-box/genética , Empalme del ARN , Intrones/genética , Células HeLa , Unión Proteica , Cuerpos Enrollados/metabolismo , Células HEK293
12.
Plant Cell ; 34(6): 2404-2423, 2022 05 24.
Artículo en Inglés | MEDLINE | ID: mdl-35294035

RESUMEN

Gene regulation ensures that the appropriate genes are expressed at the proper time. Nuclear retention of incompletely spliced or mature mRNAs is emerging as a novel, previously underappreciated layer of posttranscriptional regulation. Studies on this phenomenon indicated that it exerts a significant influence on the regulation of gene expression by regulating export and translation delay, which allows the synthesis of specific proteins in response to a stimulus or at strictly controlled time points, for example, during cell differentiation or development. Here, we show that transcription in microsporocytes of European larch (Larix decidua) occurs in a pulsatile manner during prophase of the first meiotic division. Transcriptional activity was then silenced after each pulse. However, the transcripts synthesized were not exported immediately to the cytoplasm but were retained in the nucleoplasm and Cajal bodies (CBs). In contrast to the nucleoplasm, we did not detect mature transcripts in CBs, which only stored nonfully spliced transcripts with retained introns. Notably, the retained introns were spliced at precisely defined times, and fully mature mRNAs were released into the cytoplasm for translation. As similar processes have been observed during spermatogenesis in animals, our results illustrate an evolutionarily conserved mechanism of gene expression regulation during generative cells development in Eukaryota.


Asunto(s)
Larix , Animales , Cuerpos Enrollados/genética , Cuerpos Enrollados/metabolismo , Larix/genética , Larix/metabolismo , Meiosis , Profase , Precursores del ARN/genética , Precursores del ARN/metabolismo , ARN Mensajero/genética , ARN Mensajero/metabolismo
13.
J Cell Sci ; 135(8)2022 04 15.
Artículo en Inglés | MEDLINE | ID: mdl-35356988

RESUMEN

Coilin is a conserved protein essential for integrity of nuclear membrane-less inclusions called Cajal bodies. Here, we report an amino acid substitution (p.K496E) found in a widely-used human EGFP-coilin construct that has a dominant-negative effect on Cajal body formation. We show that this coilin-K496E variant fails to rescue Cajal bodies in cells lacking endogenous coilin, whereas the wild-type construct restores Cajal bodies in mouse and human coilin-knockout cells. In cells containing endogenous coilin, both the wild-type and K496E variant proteins accumulate in Cajal bodies. However, high-level overexpression of coilin-K496E causes Cajal body disintegration. Thus, a mutation in the C-terminal region of human coilin can disrupt Cajal body assembly. Caution should be used when interpreting data from coilin plasmids that are derived from this variant (currently deposited at Addgene).


Asunto(s)
Cuerpos Enrollados , Mutación Puntual , Animales , Cuerpos Enrollados/genética , Células HeLa , Humanos , Ratones , Mutación/genética , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Mutación Puntual/genética
14.
RNA ; 28(1): 52-57, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-34772787

RESUMEN

Macroscopic membraneless organelles containing RNA such as the nucleoli, germ granules, and the Cajal body have been known for decades. These biomolecular condensates are liquid-like bodies that can be formed by a phase transition. Recent evidence has revealed the presence of similar microscopic condensates associated with the transcription of genes. This brief article summarizes thoughts about the importance of condensates in the regulation of transcription and how RNA molecules, as components of such condensates, control the synthesis of RNA. Models and experimental data suggest that RNAs from enhancers facilitate the formation of a condensate that stabilizes the binding of transcription factors and accounts for a burst of transcription at the promoter. Termination of this burst is pictured as a nonequilibrium feedback loop where additional RNA destabilizes the condensate.


Asunto(s)
Condensados Biomoleculares/química , ADN/química , Proteínas de Unión al ARN/química , ARN/química , Factores de Transcripción/química , Transcripción Genética , Sitios de Unión , Condensados Biomoleculares/metabolismo , Compartimento Celular , Nucléolo Celular/química , Nucléolo Celular/metabolismo , Cuerpos Enrollados/química , Cuerpos Enrollados/metabolismo , ADN/metabolismo , Células Eucariotas/química , Células Eucariotas/metabolismo , Retroalimentación Fisiológica , Gránulos de Ribonucleoproteína de Células Germinales/química , Gránulos de Ribonucleoproteína de Células Germinales/metabolismo , Humanos , Sustancias Macromoleculares/química , Sustancias Macromoleculares/metabolismo , Regiones Promotoras Genéticas , Unión Proteica , ARN/metabolismo , Proteínas de Unión al ARN/metabolismo , Factores de Transcripción/metabolismo
15.
Nucleic Acids Res ; 50(21): 12444-12461, 2022 11 28.
Artículo en Inglés | MEDLINE | ID: mdl-36454007

RESUMEN

RNA polymerase II (RNAPII) transcribes small nuclear RNA (snRNA) genes in close proximity to Cajal bodies, subnuclear compartments that depend on the SUMO isopeptidase USPL1 for their assembly. We show here that overexpression of USPL1 as well as of another nuclear SUMO isopeptidase, SENP6, alters snRNA 3'-end cleavage, a process carried out by the Integrator complex. Beyond its role in snRNA biogenesis, this complex is responsible for regulating the expression of different RNAPII transcripts. While several subunits of the complex are SUMO conjugation substrates, we found that the SUMOylation of the INTS11 subunit is regulated by USPL1 and SENP6. We defined Lys381, Lys462 and Lys475 as bona fide SUMO attachment sites on INTS11 and observed that SUMOylation of this protein modulates its subcellular localization and is required for Integrator activity. Moreover, while an INTS11 SUMOylation-deficient mutant is still capable of interacting with INTS4 and INTS9, its interaction with other subunits of the complex is affected. These findings point to a regulatory role for SUMO conjugation on Integrator activity and suggest the involvement of INTS11 SUMOylation in the assembly of the complex. Furthermore, this work adds Integrator-dependent RNA processing to the growing list of cellular processes regulated by SUMO conjugation.


Asunto(s)
ARN Nuclear Pequeño , Sumoilación , ARN Nuclear Pequeño/genética , ARN Nuclear Pequeño/metabolismo , Cuerpos Enrollados/metabolismo , Núcleo Celular/genética , Núcleo Celular/metabolismo , ARN Polimerasa II/genética , ARN Polimerasa II/metabolismo
16.
Neurobiol Dis ; 183: 106172, 2023 07.
Artículo en Inglés | MEDLINE | ID: mdl-37257665

RESUMEN

Distal hereditary neuropathies and neuro motor diseases are complex neurological phenotypes associated with pathogenic variants in a large number of genes, but in some the origin is unknown. Recently, rare pathogenic variants of the human VRK1 gene have been associated with these neurological phenotypes. All VRK1 pathogenic variants are recessive, and their clinical presentation occurs in either homozygous or compound heterozygous patients. The pathogenic VRK1 gene pathogenic variants are located in three clusters within the protein sequence. The main, and initial, shared clinical phenotype among VRK1 pathogenic variants is a distal progressive loss of motor and/or sensory function, which includes diseases such as spinal muscular atrophy, Charcot-Marie-Tooth, amyotrophic lateral sclerosis and hereditary spastic paraplegia. In most cases, symptoms start early in infancy, or in utero, and are slowly progressive. Additional neurological symptoms vary among non-related patients, probably because of their different VRK1 variants and their genetic background. The underlying common pathogenic mechanism, by its functional impairment, is a likely consequence of the roles that the VRK1 protein plays in the regulation on the stability and assembly of Cajal bodies, which affect RNA maturation and processing, neuronal migration of RNPs along axons, and DNA-damage responses. Alterations of these processes are associated with several neuro sensory or motor syndromes. The clinical heterogeneity of the neurological phenotypes associated with VRK1 is a likely consequence of the protein complexes in which VRK1 is integrated, which include several proteins known to be associated with Cajal bodies and DNA damage responses. Several hereditary distal neurological diseases are a consequence of pathogenic variants in genes that alter these cellular functions. We conclude that VRK1-related distal hereditary neuropathies and motor neuron diseases represent a novel subgroup of Cajal body related neurological syndromes.


Asunto(s)
Enfermedad de Charcot-Marie-Tooth , Enfermedad de la Neurona Motora , Humanos , Cuerpos Enrollados/metabolismo , Síndrome , Mutación , Enfermedad de la Neurona Motora/metabolismo , Péptidos y Proteínas de Señalización Intracelular/genética , Enfermedad de Charcot-Marie-Tooth/genética
17.
J Cell Sci ; 134(12)2021 06 15.
Artículo en Inglés | MEDLINE | ID: mdl-34137440

RESUMEN

Hypoxia is a severe stressor to cellular homeostasis. At the cellular level, low oxygen triggers the transcription of a variety of genes supporting cell survival and oxygen homeostasis mediated by transcription factors, such as hypoxia-inducible factors (HIFs). Among many determinants dictating cell responses to hypoxia and HIFs are microRNAs (miRNAs). Cajal bodies (CBs), subnuclear structures involved in ribonucleoprotein biogenesis, have been recently proven to contribute to miRNA processing and biogenesis but have not been studied under hypoxia. Here, we show, for the first time, a hypoxia-dependent increase in CB number in WI-38 primary fibroblasts, which normally have very few CBs. Additionally, the CB marker protein coilin is upregulated in hypoxic WI-38 cells. However, the hypoxic coilin upregulation was not seen in transformed cell lines. Furthermore, we found that coilin is needed for the hypoxic induction of a well-known hypoxia-induced miRNA (hypoxamiR), miR-210, as well as for the hypoxia-induced alternative splicing of the miR-210 host gene, MIR210HG. These findings provide a new link in the physiological understanding of coilin, CBs and miRNA dysregulation in hypoxic pathology.


Asunto(s)
MicroARNs , Empalme Alternativo/genética , Hipoxia de la Célula , Cuerpos Enrollados/genética , Cuerpos Enrollados/metabolismo , Humanos , MicroARNs/genética , MicroARNs/metabolismo , Factores de Transcripción/metabolismo
18.
RNA ; 27(11): 1363-1373, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-34385348

RESUMEN

In eukaryotes, rRNAs and spliceosomal snRNAs are heavily modified post-transcriptionally. Pseudouridylation and 2'-O-methylation are the most abundant types of RNA modifications. They are mediated by modification guide RNAs, also known as small nucleolar (sno)RNAs and small Cajal body-specific (sca)RNAs. We used yeast and vertebrate cells to test guide activities predicted for a number of snoRNAs, based on their regions of complementarity with rRNAs. We showed that human SNORA24 is a genuine guide RNA for 18S-Ψ609, despite some noncanonical base-pairing with its target. At the same time, we found quite a few snoRNAs that have the ability to base-pair with rRNAs and can induce predicted modifications in artificial substrate RNAs, but do not modify the same target sequence within endogenous rRNA molecules. Furthermore, certain fragments of rRNAs can be modified by the endogenous yeast modification machinery when inserted into an artificial backbone RNA, even though the same sequences are not modified in endogenous yeast rRNAs. In Xenopus cells, a guide RNA generated from scaRNA, but not from snoRNA, could induce an additional pseudouridylation of U2 snRNA at position 60; both guide RNAs were equally active on a U2 snRNA-specific substrate in yeast cells. Thus, post-transcriptional modification of functionally important RNAs, such as rRNAs and snRNAs, is highly regulated and more complex than simply strong base-pairing between a guide RNA and substrate RNA. We discuss possible regulatory roles for these unexpected modifications.


Asunto(s)
Cuerpos Enrollados/metabolismo , Seudouridina/metabolismo , Procesamiento Postranscripcional del ARN , ARN Guía de Kinetoplastida/metabolismo , ARN Nucleolar Pequeño/metabolismo , Empalmosomas/metabolismo , Secuencia de Bases , Cuerpos Enrollados/genética , Humanos , Metilación , ARN Guía de Kinetoplastida/genética , ARN Ribosómico/genética , ARN Ribosómico/metabolismo , ARN Nucleolar Pequeño/química , ARN Nucleolar Pequeño/genética , Empalmosomas/genética
19.
Nucleic Acids Res ; 49(11): 6437-6455, 2021 06 21.
Artículo en Inglés | MEDLINE | ID: mdl-34096600

RESUMEN

The biogenesis of small uridine-rich nuclear ribonucleoproteins (UsnRNPs) depends on the methylation of Sm proteins catalyzed by the methylosome and the subsequent action of the SMN complex, which assembles the heptameric Sm protein ring onto small nuclear RNAs (snRNAs). In this sophisticated process, the methylosome subunit pICln (chloride conductance regulatory protein) is attributed to an exceptional key position as an 'assembly chaperone' by building up a stable precursor Sm protein ring structure. Here, we show that-apart from its autophagic role-the Ser/Thr kinase ULK1 (Uncoordinated [unc-51] Like Kinase 1) functions as a novel key regulator in UsnRNP biogenesis by phosphorylation of the C-terminus of pICln. As a consequence, phosphorylated pICln is no longer capable to hold up the precursor Sm ring structure. Consequently, inhibition of ULK1 results in a reduction of efficient UsnRNP core assembly. Thus ULK1, depending on its complex formation, exerts different functions in autophagy or snRNP biosynthesis.


Asunto(s)
Homólogo de la Proteína 1 Relacionada con la Autofagia/metabolismo , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Ribonucleoproteínas Nucleares Pequeñas/biosíntesis , Homólogo de la Proteína 1 Relacionada con la Autofagia/antagonistas & inhibidores , Homólogo de la Proteína 1 Relacionada con la Autofagia/fisiología , Línea Celular , Cuerpos Enrollados , Humanos , Péptidos y Proteínas de Señalización Intracelular/antagonistas & inhibidores , Péptidos y Proteínas de Señalización Intracelular/fisiología , Canales Iónicos/metabolismo , Fosforilación , Proteína-Arginina N-Metiltransferasas/metabolismo
20.
Nucleic Acids Res ; 49(10): 5845-5866, 2021 06 04.
Artículo en Inglés | MEDLINE | ID: mdl-34023904

RESUMEN

Splicing is catalyzed by the spliceosome, a compositionally dynamic complex assembled stepwise on pre-mRNA. We reveal links between splicing machinery components and the intrinsically disordered ciliopathy protein SANS. Pathogenic mutations in SANS/USH1G lead to Usher syndrome-the most common cause of deaf-blindness. Previously, SANS was shown to function only in the cytosol and primary cilia. Here, we have uncovered molecular links between SANS and pre-mRNA splicing catalyzed by the spliceosome in the nucleus. We show that SANS is found in Cajal bodies and nuclear speckles, where it interacts with components of spliceosomal sub-complexes such as SF3B1 and the large splicing cofactor SON but also with PRPFs and snRNAs related to the tri-snRNP complex. SANS is required for the transfer of tri-snRNPs between Cajal bodies and nuclear speckles for spliceosome assembly and may also participate in snRNP recycling back to Cajal bodies. SANS depletion alters the kinetics of spliceosome assembly, leading to accumulation of complex A. SANS deficiency and USH1G pathogenic mutations affects splicing of genes related to cell proliferation and human Usher syndrome. Thus, we provide the first evidence that splicing dysregulation may participate in the pathophysiology of Usher syndrome.


Asunto(s)
Empalme Alternativo/genética , Proteínas del Tejido Nervioso/metabolismo , Precursores del ARN/metabolismo , Ribonucleoproteínas Nucleares Pequeñas/metabolismo , Empalmosomas/metabolismo , Síndromes de Usher/metabolismo , Núcleo Celular/metabolismo , Proliferación Celular/genética , Cuerpos Enrollados/metabolismo , Proteínas de Unión al ADN/metabolismo , Proteínas del Ojo/metabolismo , Técnicas de Silenciamiento del Gen , Células HEK293 , Humanos , Inmunohistoquímica , Hibridación Fluorescente in Situ , Espectrometría de Masas , Microscopía Electrónica de Transmisión , Antígenos de Histocompatibilidad Menor/metabolismo , Proteínas del Tejido Nervioso/genética , Fosfoproteínas/metabolismo , Proteómica , Precursores del ARN/genética , Factores de Empalme de ARN/metabolismo , ARN Nuclear Pequeño/genética , ARN Nuclear Pequeño/metabolismo , Ribonucleoproteína Nuclear Pequeña U4-U6/metabolismo , Ribonucleoproteínas Nucleares Pequeñas/genética , Empalmosomas/genética , Factores de Transcripción/metabolismo , Síndromes de Usher/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA