Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters










Publication year range
1.
J Mol Biol ; 435(24): 168364, 2023 12 15.
Article in English | MEDLINE | ID: mdl-37952770

ABSTRACT

Demixing of proteins and nucleic acids into condensed liquid phases is rapidly emerging as a ubiquitous mechanism underlying the complex spatiotemporal organisation of molecules within the cell. Long disordered regions of low sequence complexity (LCRs) are a common feature of proteins that form liquid-like microscopic biomolecular condensates. In particular, RNA-binding proteins with prion-like regions have emerged as key drivers of liquid demixing to form condensates such as nucleoli, paraspeckles and stress granules. Splicing factor proline- and glutamine-rich (SFPQ) is an RNA- and DNA-binding protein essential for DNA repair and paraspeckle formation. SFPQ contains two LCRs of different length and composition. Here, we show that the shorter C-terminal LCR of SFPQ is the main region responsible for the condensation of SFPQ in vitro and in the cell nucleus. In contrast, we find that the longer N-terminal prion-like LCR of SFPQ attenuates condensation of the full-length protein, suggesting a more regulatory role in preventing aberrant condensate formation in the cell. The compositions of these respective LCRs are discussed with reference to current literature. Our data add nuance to the emerging understanding of biomolecular condensation, by providing the first example of a common multifunctional nucleic acid-binding protein with an extensive prion-like region that serves to regulate rather than drive condensate formation.


Subject(s)
Biomolecular Condensates , Prions , RNA-Binding Proteins/metabolism , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , RNA , Prions/genetics , Prions/metabolism
2.
EMBO Rep ; 24(2): e54977, 2023 02 06.
Article in English | MEDLINE | ID: mdl-36416237

ABSTRACT

High-risk neuroblastoma patients have poor survival rates and require better therapeutic options. High expression of a multifunctional DNA and RNA-binding protein, NONO, in neuroblastoma is associated with poor patient outcome; however, there is little understanding of the mechanism of NONO-dependent oncogenic gene regulatory activity in neuroblastoma. Here, we used cell imaging, biochemical and genome-wide molecular analysis to reveal complex NONO-dependent regulation of gene expression. NONO forms RNA- and DNA-tethered condensates throughout the nucleus and undergoes phase separation in vitro, modulated by nucleic acid binding. CLIP analyses show that NONO mainly binds to the 5' end of pre-mRNAs and modulates pre-mRNA processing, dependent on its RNA-binding activity. NONO regulates super-enhancer-associated genes, including HAND2 and GATA2. Abrogating NONO RNA binding, or phase separation activity, results in decreased expression of HAND2 and GATA2. Thus, future development of agents that target RNA-binding activity of NONO may have therapeutic potential in this cancer context.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors , DNA-Binding Proteins , Neuroblastoma , Humans , DNA/metabolism , DNA-Binding Proteins/genetics , GATA2 Transcription Factor/genetics , GATA2 Transcription Factor/metabolism , Neuroblastoma/genetics , Neuroblastoma/metabolism , RNA Processing, Post-Transcriptional , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA-Binding Proteins/genetics , Transcription Factors/metabolism , Basic Helix-Loop-Helix Transcription Factors/metabolism
3.
J Biol Chem ; 298(11): 102563, 2022 11.
Article in English | MEDLINE | ID: mdl-36209820

ABSTRACT

RNA-binding proteins of the DBHS (Drosophila Behavior Human Splicing) family, NONO, SFPQ, and PSPC1 have numerous roles in genome stability and transcriptional and posttranscriptional regulation. Critical to DBHS activity is their recruitment to distinct subnuclear locations, for example, paraspeckle condensates, where DBHS proteins bind to the long noncoding RNA NEAT1 in the first essential step in paraspeckle formation. To carry out their diverse roles, DBHS proteins form homodimers and heterodimers, but how this dimerization influences DBHS localization and function is unknown. Here, we present an inducible GFP-NONO stable cell line and use it for live-cell 3D-structured illumination microscopy, revealing paraspeckles with dynamic, twisted elongated structures. Using siRNA knockdowns, we show these labeled paraspeckles consist of GFP-NONO/endogenous SFPQ dimers and that GFP-NONO localization to paraspeckles depends on endogenous SFPQ. Using purified proteins, we confirm that partner swapping between NONO and SFPQ occurs readily in vitro. Crystallographic analysis of the NONO-SFPQ heterodimer reveals conformational differences to the other DBHS dimer structures, which may contribute to partner preference, RNA specificity, and subnuclear localization. Thus overall, our study suggests heterodimer partner availability is crucial for NONO subnuclear distribution and helps explain the complexity of both DBHS protein and paraspeckle dynamics through imaging and structural approaches.


Subject(s)
Paraspeckles , RNA, Long Noncoding , Humans , Dimerization , RNA-Binding Proteins/metabolism , Gene Expression Regulation , RNA, Long Noncoding/genetics
4.
Biochimie ; 190: 124-131, 2021 Nov.
Article in English | MEDLINE | ID: mdl-34329720

ABSTRACT

Paraspeckles are RNA-protein structures within the nucleus of mammalian cells, capable of orchestrating various biochemical processes. An overexpression of the architectural component of paraspeckles, a long non-coding RNA called NEAT1 (Nuclear Enriched Abundant Transcript 1), has been linked to a variety of cancers and is often associated with poor patient prognosis. Thus, there is an accumulating interest in the role of paraspeckles in carcinogenesis, however there is a limited understanding of how NEAT1 expression is regulated. Here, we demonstrate that both nuclear G-quadruplex (G4) and paraspeckle formation are significantly increased in a human breast cancer cell line compared to non-tumorigenic breast cells. Moreover, we identified and characterized G4-forming sequences within the NEAT1 promoter and demonstrate stabilization of G4 DNA with a G4-stabilizing small molecule results in a significant alteration in both paraspeckle formation and NEAT1 expression. This G4-mediated alteration of NEAT1 at both the transcriptional and post-transcriptional levels was evident in U2OS osteosarcoma cells, MCF-7 breast adenocarcinoma and MDA-MB-231 triple negative breast cancer cells.


Subject(s)
G-Quadruplexes , Neoplasms/genetics , Neoplasms/metabolism , Paraspeckles/genetics , Paraspeckles/metabolism , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Cell Line, Tumor , Humans , RNA, Long Noncoding/chemistry , RNA, Long Noncoding/genetics , RNA, Long Noncoding/metabolism
5.
Cell Rep ; 25(7): 1912-1923.e9, 2018 11 13.
Article in English | MEDLINE | ID: mdl-30428357

ABSTRACT

We and others have recently reported that the SMC protein Smchd1 is a regulator of chromosome conformation. Smchd1 is critical for the structure of the inactive X chromosome and at autosomal targets such as the Hox genes. However, it is unknown how Smchd1 is recruited to these sites. Here, we report that Smchd1 localizes to the inactive X via the Xist-HnrnpK-PRC1 (polycomb repressive complex 1) pathway. Contrary to previous reports, Smchd1 does not bind Xist or other RNA molecules with any specificity. Rather, the localization of Smchd1 to the inactive X is H2AK119ub dependent. Following perturbation of this interaction, Smchd1 is destabilized, which has consequences for gene silencing genome-wide. Our work adds Smchd1 to the PRC1 silencing pathway for X chromosome inactivation.


Subject(s)
Chromosomal Proteins, Non-Histone/metabolism , Heterogeneous-Nuclear Ribonucleoprotein K/metabolism , Polycomb Repressive Complex 1/metabolism , RNA, Long Noncoding/metabolism , X Chromosome Inactivation/genetics , Animals , Base Sequence , Cell Differentiation , Female , Genome , Histones/metabolism , Lysine/metabolism , Mice , Mouse Embryonic Stem Cells/cytology , Mouse Embryonic Stem Cells/metabolism , Oligonucleotides/metabolism , Protein Transport
6.
Mol Cell ; 70(6): 1038-1053.e7, 2018 06 21.
Article in English | MEDLINE | ID: mdl-29932899

ABSTRACT

A class of long noncoding RNAs (lncRNAs) has architectural functions in nuclear body construction; however, specific RNA domains dictating their architectural functions remain uninvestigated. Here, we identified the domains of the architectural NEAT1 lncRNA that construct paraspeckles. Systematic deletion of NEAT1 portions using CRISPR/Cas9 in haploid cells revealed modular domains of NEAT1 important for RNA stability, isoform switching, and paraspeckle assembly. The middle domain, containing functionally redundant subdomains, was responsible for paraspeckle assembly. Artificial tethering of the NONO protein to a NEAT1_2 mutant lacking the functional subdomains rescued paraspeckle assembly, and this required the NOPS dimerization domain of NONO. Paraspeckles exhibit phase-separated properties including susceptibility to 1,6-hexanediol treatment. RNA fragments of the NEAT1_2 subdomains preferentially bound NONO/SFPQ, leading to phase-separated aggregates in vitro. Thus, we demonstrate that the enrichment of NONO dimers on the redundant NEAT1_2 subdomains initiates construction of phase-separated paraspeckles, providing mechanistic insights into lncRNA-based nuclear body formation.


Subject(s)
RNA, Long Noncoding/genetics , RNA, Long Noncoding/metabolism , Base Sequence , CRISPR-Cas Systems , Cell Nucleus/metabolism , HeLa Cells , Humans , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Protein Domains , RNA Recognition Motif Proteins/genetics , RNA Recognition Motif Proteins/metabolism , RNA Stability , Transcription Factors/metabolism
7.
J Cell Biol ; 210(4): 529-39, 2015 Aug 17.
Article in English | MEDLINE | ID: mdl-26283796

ABSTRACT

Prion-like domains (PLDs) are low complexity sequences found in RNA binding proteins associated with the neurodegenerative disorder amyotrophic lateral sclerosis. Recently, PLDs have been implicated in mediating gene regulation via liquid-phase transitions that drive ribonucleoprotein granule assembly. In this paper, we report many PLDs in proteins associated with paraspeckles, subnuclear bodies that form around long noncoding RNA. We mapped the interactome network of paraspeckle proteins, finding enrichment of PLDs. We show that one protein, RBM14, connects key paraspeckle subcomplexes via interactions mediated by its PLD. We further show that the RBM14 PLD, as well as the PLD of another essential paraspeckle protein, FUS, is required to rescue paraspeckle formation in cells in which their endogenous counterpart has been knocked down. Similar to FUS, the RBM14 PLD also forms hydrogels with amyloid-like properties. These results suggest a role for PLD-mediated liquid-phase transitions in paraspeckle formation, highlighting this nuclear body as an excellent model system for understanding the perturbation of such processes in neurodegeneration.


Subject(s)
Cell Nucleus/metabolism , Intracellular Signaling Peptides and Proteins/physiology , Prions/chemistry , RNA-Binding Proteins/chemistry , Amyloidogenic Proteins/chemistry , HeLa Cells , Humans , Hydrogels/chemistry , Intracellular Signaling Peptides and Proteins/chemistry , Prions/metabolism , Protein Binding , Protein Interaction Maps , RNA-Binding Proteins/metabolism
8.
Proc Natl Acad Sci U S A ; 110(16): 6536-41, 2013 Apr 16.
Article in English | MEDLINE | ID: mdl-23550157

ABSTRACT

The cytoplasmic RNA-induced silencing complex (RISC) contains dsRNA binding proteins, including protein kinase RNA activator (PACT), transactivation response RNA binding protein (TRBP), and Dicer, that process pre-microRNAs into mature microRNAs (miRNAs) that target specific mRNA species for regulation. There is increasing evidence for important functional interactions between the miRNA and nuclear receptor (NR) signaling networks, with recent data showing that estrogen, acting through the estrogen receptor, can modulate initial aspects of nuclear miRNA processing. Here, we show that the cytoplasmic RISC proteins PACT, TRBP, and Dicer are steroid receptor RNA activator (SRA) binding NR coregulators that target steroid-responsive promoters and regulate NR activity and downstream gene expression. Furthermore, each of the RISC proteins, together with Argonaute 2, associates with SRA and specific pre-microRNAs in both the nucleus and cytoplasm, providing evidence for links between NR-mediated transcription and some of the factors involved in miRNA processing.


Subject(s)
Carrier Proteins/metabolism , DEAD-box RNA Helicases/metabolism , Gene Expression Regulation/genetics , MicroRNAs/metabolism , RNA-Binding Proteins/metabolism , RNA-Induced Silencing Complex/metabolism , Ribonuclease III/metabolism , Blotting, Western , Cell Fractionation , Chromatin Immunoprecipitation , Cloning, Molecular , HEK293 Cells , HeLa Cells , Humans , Luciferases , MCF-7 Cells , Plasmids/genetics , RNA Interference , Reverse Transcriptase Polymerase Chain Reaction , Transfection , Two-Hybrid System Techniques
9.
J Biol Chem ; 281(50): 38791-800, 2006 Dec 15.
Article in English | MEDLINE | ID: mdl-17008314

ABSTRACT

Myeloid leukemia factor 1 (MLF1) is an oncoprotein associated with hemopoietic lineage commitment and acute myeloid leukemia. Here we show that Mlf1 associated with a novel binding partner, Mlf1-associated nuclear protein (Manp), a new heterogeneous nuclear ribonucleoprotein (hnRNP) family member, related to hnRNP-U. Manp localized exclusively in the nucleus and could redirect Mlf1 from the cytoplasm into the nucleus. The nuclear content of Mlf1 was also regulated by 14-3-3 binding to a canonical 14-3-3 binding motif within the N terminus of Mlf1. Significantly Mlf1 contains a functional nuclear export signal and localized primarily to the nuclei of hemopoietic cells. Mlf1 was capable of binding DNA, and microarray analysis revealed that it affected the expression of several genes, including transcription factors. In summary, this study reveals that Mlf1 translocates between nucleus and cytoplasm, associates with a novel hnRNP, and influences gene expression.


Subject(s)
Heterogeneous-Nuclear Ribonucleoprotein U/metabolism , Proteins/metabolism , Amino Acid Sequence , Animals , Base Sequence , COS Cells , Cell Cycle Proteins , Cell Nucleus/metabolism , Chlorocebus aethiops , DNA, Complementary , DNA-Binding Proteins , Molecular Sequence Data , Protein Binding , Proteins/chemistry , Proteins/genetics
10.
Oncogene ; 23(29): 5105-9, 2004 Jun 24.
Article in English | MEDLINE | ID: mdl-15122318

ABSTRACT

Myeloid leukemia factor 1 (MLF1) is a novel oncoprotein involved in translocations associated with acute myeloid leukemia (AML), especially erythroleukemias. In this study, we demonstrate that ectopic expression of Mlf1 prevented J2E erythroleukemic cells from undergoing biological and morphological maturation in response to erythropoietin (Epo). We show that Mlf1 inhibited Epo-induced cell cycle exit and suppressed a rise in the cell cycle inhibitor p27(Kip1). Unlike differentiating J2E cells, Mlf1-expressing cells did not downregulate Cul1 and Skp2, components of the ubiquitin E3 ligase complex SCF(Skp2) involved in the proteasomal degradation of p27(Kip1). In contrast, Mlf1 did not interfere with increases in p27(Kip1) and terminal differentiation initiated by thyroid hormone withdrawal from erythroid cells, or cytokine-stimulated maturation of myeloid cells. These data demonstrate that Mlf1 interferes with an Epo-responsive pathway involving p27(Kip1) accumulation, which inhibits cell cycle arrest essential for erythroid terminal differentiation.


Subject(s)
Cell Cycle , Erythropoietin/pharmacology , Proteins/pharmacology , Animals , Cell Cycle Proteins , Cell Differentiation/drug effects , DNA-Binding Proteins , Mice , Proliferating Cell Nuclear Antigen/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...