Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 4 de 4
Filter
Add more filters










Database
Type of study
Language
Publication year range
1.
Nucleic Acids Res ; 52(7): 4037-4052, 2024 Apr 24.
Article in English | MEDLINE | ID: mdl-38499487

ABSTRACT

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.


Subject(s)
DEAD-box RNA Helicases , RNA Splicing Factors , RNA, Small Nuclear , RNA-Binding Proteins , Ribonucleoproteins, Small Nuclear , Spliceosomes , Spliceosomes/metabolism , Spliceosomes/genetics , Ribonucleoproteins, Small Nuclear/metabolism , Ribonucleoproteins, Small Nuclear/genetics , Ribonucleoproteins, Small Nuclear/chemistry , RNA-Binding Proteins/metabolism , RNA-Binding Proteins/genetics , RNA-Binding Proteins/chemistry , Humans , RNA, Small Nuclear/metabolism , RNA, Small Nuclear/genetics , RNA, Small Nuclear/chemistry , DEAD-box RNA Helicases/metabolism , DEAD-box RNA Helicases/genetics , RNA Splicing , Introns/genetics , HeLa Cells , Protein Binding , Coiled Bodies/metabolism , HEK293 Cells
2.
Nucleic Acids Res ; 49(5): 2835-2847, 2021 03 18.
Article in English | MEDLINE | ID: mdl-33577674

ABSTRACT

Disruption of minor spliceosome functions underlies several genetic diseases with mutations in the minor spliceosome-specific small nuclear RNAs (snRNAs) and proteins. Here, we define the molecular outcome of the U12 snRNA mutation (84C>U) resulting in an early-onset form of cerebellar ataxia. To understand the molecular consequences of the U12 snRNA mutation, we created cell lines harboring the 84C>T mutation in the U12 snRNA gene (RNU12). We show that the 84C>U mutation leads to accelerated decay of the snRNA, resulting in significantly reduced steady-state U12 snRNA levels. Additionally, the mutation leads to accumulation of 3'-truncated forms of U12 snRNA, which have undergone the cytoplasmic steps of snRNP biogenesis. Our data suggests that the 84C>U-mutant snRNA is targeted for decay following reimport into the nucleus, and that the U12 snRNA fragments are decay intermediates that result from the stalling of a 3'-to-5' exonuclease. Finally, we show that several other single-nucleotide variants in the 3' stem-loop of U12 snRNA that are segregating in the human population are also highly destabilizing. This suggests that the 3' stem-loop is important for the overall stability of the U12 snRNA and that additional disease-causing mutations are likely to exist in this region.


Subject(s)
Cerebellar Ataxia/genetics , RNA, Small Nuclear/chemistry , RNA, Small Nuclear/genetics , HEK293 Cells , HeLa Cells , Humans , Mutation , Point Mutation , RNA Stability , RNA, Small Nuclear/metabolism
3.
Semin Cell Dev Biol ; 79: 103-112, 2018 07.
Article in English | MEDLINE | ID: mdl-28965864

ABSTRACT

The U12-dependent (minor) spliceosome excises a rare group of introns that are characterized by a highly conserved 5' splice site and branch point sequence. Several new congenital or somatic diseases have recently been associated with mutations in components of the minor spliceosome. A common theme in these diseases is the detection of elevated levels of transcripts containing U12-type introns, of which a subset is associated with other splicing defects. Here we review the present understanding of minor spliceosome diseases, particularly those associated with the specific components of the minor spliceosome. We also present a model for interpreting the molecular-level consequences of the different diseases.


Subject(s)
Disease/genetics , RNA Precursors/genetics , RNA Splicing , Ribonucleoproteins, Small Nuclear/genetics , Spliceosomes/genetics , Animals , Base Sequence , Humans , Mutation , RNA, Messenger/genetics
4.
RNA ; 24(3): 396-409, 2018 03.
Article in English | MEDLINE | ID: mdl-29255062

ABSTRACT

Mutations in the components of the minor spliceosome underlie several human diseases. A subset of patients with isolated growth hormone deficiency (IGHD) harbors mutations in the RNPC3 gene, which encodes the minor spliceosome-specific U11/U12-65K protein. Although a previous study showed that IGHD patient cells have defects in U12-type intron recognition, the biochemical effects of these mutations on the 65K protein have not been characterized. Here, we show that a proline-to-threonine missense mutation (P474T) and a nonsense mutation (R502X) in the C-terminal RNA recognition motif (C-RRM) of the 65K protein impair the binding of 65K to U12 and U6atac snRNAs. We further show that the nonsense allele is targeted to the nonsense-mediated decay (NMD) pathway, but in an isoform-specific manner, with the nuclear-retained 65K long-3'UTR isoform escaping the NMD pathway. In contrast, the missense P474T mutation leads, in addition to the RNA-binding defect, to a partial defect in the folding of the C-RRM and reduced stability of the full-length protein, thus reducing the formation of U11/U12 di-snRNP complexes. We propose that both the C-RRM folding defect and NMD-mediated decrease in the levels of the U11/U12-65K protein reduce formation of the U12-type intron recognition complex and missplicing of a subset of minor introns leading to pituitary hypoplasia and a subsequent defect in growth hormone secretion.


Subject(s)
Dwarfism, Pituitary/genetics , Models, Molecular , Nonsense Mediated mRNA Decay , Nuclear Proteins/genetics , RNA, Small Nuclear/genetics , RNA-Binding Proteins/genetics , Ribonucleoproteins, Small Nuclear/genetics , Spliceosomes , Codon, Nonsense , Dwarfism, Pituitary/metabolism , HeLa Cells , Humans , Introns/genetics , Mutation, Missense , Nuclear Proteins/chemistry , Proline , RNA, Small Nuclear/chemistry , RNA-Binding Motifs , RNA-Binding Proteins/chemistry , Ribonucleoproteins, Small Nuclear/chemistry , Threonine
SELECTION OF CITATIONS
SEARCH DETAIL
...