Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 27
Filter
Add more filters










Publication year range
1.
PLoS Pathog ; 20(4): e1012172, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38662769

ABSTRACT

The implementation of antiretroviral therapy (ART) has effectively restricted the transmission of Human Immunodeficiency Virus (HIV) and improved overall clinical outcomes. However, a complete cure for HIV remains out of reach, as the virus persists in a stable pool of infected cell reservoir that is resistant to therapy and thus a main barrier towards complete elimination of viral infection. While the mechanisms by which host proteins govern viral gene expression and latency are well-studied, the emerging regulatory functions of non-coding RNAs (ncRNA) in the context of T cell activation, HIV gene expression and viral latency have not yet been thoroughly explored. Here, we report the identification of the Cytoskeleton Regulator (CYTOR) long non-coding RNA (lncRNA) as an activator of HIV gene expression that is upregulated following T cell stimulation. Functional studies show that CYTOR suppresses viral latency by directly binding to the HIV promoter and associating with the cellular positive transcription elongation factor (P-TEFb) to activate viral gene expression. CYTOR also plays a global role in regulating cellular gene expression, including those involved in controlling actin dynamics. Depletion of CYTOR expression reduces cytoplasmic actin polymerization in response to T cell activation. In addition, treating HIV-infected cells with pharmacological inhibitors of actin polymerization reduces HIV gene expression. We conclude that both direct and indirect effects of CYTOR regulate HIV gene expression.


Subject(s)
Gene Expression Regulation, Viral , HIV Infections , HIV-1 , RNA, Long Noncoding , Virus Latency , Humans , HIV Infections/virology , HIV Infections/genetics , HIV-1/genetics , HIV-1/physiology , Jurkat Cells , Lymphocyte Activation , Promoter Regions, Genetic , RNA, Long Noncoding/genetics
2.
Biopolymers ; 115(3): e23576, 2024 May.
Article in English | MEDLINE | ID: mdl-38511874

ABSTRACT

EWSR1 (Ewing Sarcoma Related protein 1) is an RNA binding protein that is ubiquitously expressed across cell lines and involved in multiple parts of RNA processing, such as transcription, splicing, and mRNA transport. EWSR1 has also been implicated in cellular mechanisms to control formation of R-loops, a three-stranded nucleic acid structure consisting of a DNA:RNA hybrid and a displaced single-stranded DNA strand. Unscheduled R-loops result in genomic and transcription stress. Loss of function of EWSR1 functions commonly found in Ewing Sarcoma correlates with high abundance of R-loops. In this study, we investigated the mechanism for EWSR1 to recognize an R-loop structure specifically. Using electrophoretic mobility shift assays (EMSA), we detected the high affinity binding of EWSR1 to substrates representing components found in R-loops. EWSR1 specificity could be isolated to the DNA fork region, which transitions between double- and single-stranded DNA. Our data suggests that the Zinc-finger domain (ZnF) with flanking arginine and glycine rich (RGG) domains provide high affinity binding, while the RNA recognition motif (RRM) with its RGG domains offer improved specificity. This model offers a rational for EWSR1 specificity to encompass a wide range in contexts due to the DNA forks always found with R-loops.


Subject(s)
DNA , R-Loop Structures , RNA-Binding Protein EWS , RNA-Binding Protein EWS/metabolism , RNA-Binding Protein EWS/chemistry , RNA-Binding Protein EWS/genetics , Humans , DNA/metabolism , DNA/chemistry , Protein Binding , Sarcoma, Ewing/metabolism , Sarcoma, Ewing/genetics , Zinc Fingers , RNA-Binding Proteins/metabolism , RNA-Binding Proteins/chemistry , RNA-Binding Proteins/genetics , Electrophoretic Mobility Shift Assay
3.
bioRxiv ; 2024 Jan 21.
Article in English | MEDLINE | ID: mdl-38293191

ABSTRACT

EWSR1 (Ewing Sarcoma Related protein 1) is an RNA binding protein that is ubiquitously expressed across cell lines and involved in multiple parts of RNA processing, such as transcription, splicing, and mRNA transport. EWSR1 has also been implicated in cellular mechanisms to control formation of R-loops, a three-stranded nucleic acid structure consisting of a DNA:RNA hybrid and a displaced single-stranded DNA strand. Unscheduled R-loops result in genomic and transcription stress. Loss of function of EWSR1 functions commonly found in Ewing Sarcoma correlates with high abundance of R-loops. In this study, we investigated the mechanism for EWSR1 to recognize an R-loop structure specifically. Using electrophoretic mobility shift assays (EMSA), we detected the high affinity binding of EWSR1 to substrates representing components found in R-loops. EWSR1 specificity could be isolated to the DNA fork region, which transitions between double- and single-stranded DNA. Our data suggests that the Zinc-finger domain (ZnF) with flanking arginine and glycine rich (RGG) domains provide high affinity binding, while the RNA recognition motif (RRM) with its RGG domains offer improved specificity. This model offers a rational for EWSR1 specificity to encompass a wide range in contexts due to the DNA forks always found with R-loops.

4.
J Biol Chem ; 299(10): 105237, 2023 Oct.
Article in English | MEDLINE | ID: mdl-37690693

ABSTRACT

The protein FUS (FUSed in sarcoma) is a metazoan RNA-binding protein that influences RNA production by all three nuclear polymerases. FUS also binds nascent transcripts, RNA processing factors, RNA polymerases, and transcription machinery. Here, we explored the role of FUS binding interactions for activity during transcription. In vitro run-off transcription assays revealed FUS-enhanced RNA produced by a non-eukaryote polymerase. The activity also reduced the formation of R-loops between RNA products and their DNA template. Analysis by domain mutation and deletion indicated RNA-binding was required for activity. We interpret that FUS binds and sequesters nascent transcripts to prevent R-loops from forming with nearby DNA. DRIP-seq analysis showed that a knockdown of FUS increased R-loop enrichment near expressed genes. Prevention of R-loops by FUS binding to nascent transcripts has the potential to affect transcription by any RNA polymerase, highlighting the broad impact FUS can have on RNA metabolism in cells and disease.


Subject(s)
DNA , R-Loop Structures , RNA-Binding Protein FUS , RNA , DNA/metabolism , R-Loop Structures/genetics , RNA/metabolism , RNA-Binding Protein FUS/metabolism , Protein Binding , Humans , DNA-Directed RNA Polymerases/metabolism , HEK293 Cells
5.
Elife ; 102021 05 26.
Article in English | MEDLINE | ID: mdl-34060470

ABSTRACT

Traumatic brain injury (TBI) is a predisposing factor for many neurodegenerative diseases, including amyotrophic lateral sclerosis (ALS), Alzheimer's disease (AD), Parkinson's disease (PD), and chronic traumatic encephalopathy (CTE). Although defects in nucleocytoplasmic transport (NCT) is reported ALS and other neurodegenerative diseases, whether defects in NCT occur in TBI remains unknown. We performed proteomic analysis on Drosophila exposed to repeated TBI and identified resultant alterations in several novel molecular pathways. TBI upregulated nuclear pore complex (NPC) and nucleocytoplasmic transport (NCT) proteins as well as alter nucleoporin stability. Traumatic injury disrupted RanGAP1 and NPC protein distribution in flies and a rat model and led to coaggregation of NPC components and TDP-43. In addition, trauma-mediated NCT defects and lethality are rescued by nuclear export inhibitors. Importantly, genetic upregulation of nucleoporins in vivo and in vitro triggered TDP-43 cytoplasmic mislocalization, aggregation, and altered solubility and reduced motor function and lifespan of animals. We also found NUP62 pathology and elevated NUP62 concentrations in postmortem brain tissues of patients with mild or severe CTE as well as co-localization of NUP62 and TDP-43 in CTE. These findings indicate that TBI leads to NCT defects, which potentially mediate the TDP-43 pathology in CTE.


Subject(s)
Active Transport, Cell Nucleus , Brain Injuries, Traumatic/metabolism , Brain/metabolism , DNA-Binding Proteins/metabolism , Drosophila Proteins/metabolism , Drosophila melanogaster/metabolism , Nuclear Pore/metabolism , TDP-43 Proteinopathies/metabolism , Animals , Animals, Genetically Modified , Brain/pathology , Brain Injuries, Traumatic/genetics , Brain Injuries, Traumatic/pathology , Case-Control Studies , DNA-Binding Proteins/genetics , Disease Models, Animal , Drosophila Proteins/genetics , Drosophila melanogaster/genetics , GTPase-Activating Proteins/genetics , GTPase-Activating Proteins/metabolism , HEK293 Cells , Humans , Longevity , Male , Membrane Glycoproteins/metabolism , Motor Activity , Nuclear Pore/genetics , Nuclear Pore/pathology , Nuclear Pore Complex Proteins/metabolism , Protein Aggregates , Protein Aggregation, Pathological , Rats, Sprague-Dawley , TDP-43 Proteinopathies/genetics , TDP-43 Proteinopathies/pathology
6.
RNA ; 2021 May 25.
Article in English | MEDLINE | ID: mdl-34035145

ABSTRACT

Ewing sarcoma is driven by fusion proteins containing a low complexity (LC) domain that is intrinsically disordered and a powerful transcriptional regulator. The most common fusion protein found in Ewing sarcoma, EWS-FLI1, takes its LC domain from the RNA-binding protein EWSR1 (Ewing Sarcoma RNA-binding protein 1) and a DNA-binding domain from the transcription factor FLI1 (Friend Leukemia Virus Integration 1). EWS-FLI1 can bind RNA polymerase II (RNA Pol II) and self-assemble through its low-complexity (LC) domain. The ability of RNA-binding proteins like EWSR1 to self-assemble or phase separate in cells has raised questions about the contribution of this process to EWS-FLI1 activity. We examined EWSR1 and EWS-FLI1 activity in Ewing sarcoma cells by siRNA-mediated knockdown and RNA-seq analysis. More transcripts were affected by the EWSR1 knockdown than expected and these included many EWS-FLI1 regulated genes. We reevaluated physical interactions between EWS-FLI1, EWSR1, and RNA Pol II, and employed a cross-linking based strategy to investigate protein assemblies associated with the proteins. The LC domain of EWS-FLI1 was required for the assemblies observed to form in cells. These results offer new insights into a protein assembly that may enable EWS-FLI1 to bind its wide network of protein partners and contribute to regulation of gene expression in Ewing sarcoma.

7.
Curr Protoc ; 1(3): e35, 2021 Mar.
Article in English | MEDLINE | ID: mdl-33740275

ABSTRACT

Recent advancements in detection methods have made protein condensates, also called granules, a major area of study, but tools to characterize these assemblies need continued development to keep up with evolving paradigms. We have optimized a protocol to separate condensates from cells using chemical cross-linking followed by size-exclusion chromatography (SEC). After SEC fractionation, the samples can be characterized by a variety of approaches including enzyme-linked immunosorbent assay, dynamic light scattering, electron microscopy, and mass spectrometry. The protocol described here has been optimized for cultured mammalian cells and E. coli expressing recombinant proteins. Since the lysates are fractionated by size, this protocol can be modified to study other large protein assemblies, including the nuclear pore complex, and for other tissues or organisms. © 2021 Wiley Periodicals LLC. Basic Protocol 1: SEC separation of cross-linked mammalian cell lysates Alternate Protocol: Preparation of non-crosslinked mammalian cells Basic Protocol 2: SEC separation of E. coli lysate Support Protocol 1: Detecting protein of interest by ELISA Support Protocol 2: TCA precipitation of SEC fractions.


Subject(s)
Escherichia coli , Proteins , Animals , Chromatography, Gel , Dynamic Light Scattering , Mass Spectrometry
8.
Protein Sci ; 29(8): 1784-1793, 2020 08.
Article in English | MEDLINE | ID: mdl-32483864

ABSTRACT

Chemical modification of proteins has been crucial in engineering protein-based therapies, targeted biopharmaceutics, molecular probes, and biomaterials. Here, we explore the use of a conjugation-based approach to sense alternative conformational states in proteins. Tyrosine has both hydrophobic and hydrophilic qualities, thus allowing it to be positioned at protein surfaces, or binding interfaces, or to be buried within a protein. Tyrosine can be conjugated with 4-phenyl-3H-1,2,4-triazole-3,5(4H)-dione (PTAD). We hypothesized that individual protein conformations could be distinguished by labeling tyrosine residues in the protein with PTAD. We conjugated tyrosine residues in a well-folded protein, bovine serum albumin (BSA), and quantified labeled tyrosine with liquid chromatography with tandem mass spectrometry. We applied this approach to alternative conformations of BSA produced in the presence of urea. The amount of PTAD labeling was found to relate to the depth of each tyrosine relative to the protein surface. This study demonstrates a new use of tyrosine conjugation using PTAD as an analytic tool able to distinguish the conformational states of a protein.


Subject(s)
Serum Albumin, Bovine/chemistry , Triazoles/chemistry , Animals , Cattle , Chromatography, Liquid , Protein Domains , Tandem Mass Spectrometry , Tyrosine/chemistry
9.
J Proteome Res ; 19(1): 360-370, 2020 01 03.
Article in English | MEDLINE | ID: mdl-31693373

ABSTRACT

The RNA-binding proteins TDP-43 and FUS are tied as the third leading known genetic cause for amyotrophic lateral sclerosis (ALS), and TDP-43 proteopathies are found in nearly all ALS patients. Both the natural function and contribution to pathology for TDP-43 remain unclear. The intersection of functions between TDP-43 and FUS can focus attention for those natural functions mostly likely to be relevant to disease. Here, we compare the role played by TDP-43 and FUS, maintaining chromatin stability for dividing HEK293T cells. We also determine and compare the interactomes of TDP-43 and FUS, quantitating changes in those before and after DNA damage. Finally, selected interactions with known importance to DNA damage repair were validated by co-immunoprecipitation assays. This study uncovered TDP-43 and FUS binding to several factors important to DNA repair mechanisms that can be replication-dependent, -independent, or both. These results provide further evidence that TDP-43 has an important role in DNA stability and provide new ways that TDP-43 can bind to the machinery that guards DNA integrity in cells.


Subject(s)
DNA Damage , DNA-Binding Proteins/metabolism , RNA-Binding Protein FUS/metabolism , DNA Repair , DNA-Binding Proteins/genetics , HEK293 Cells , Humans , Immunoprecipitation , Protein Interaction Maps , RNA-Binding Protein FUS/genetics
10.
Biochim Biophys Acta Gene Regul Mech ; 1862(10): 194434, 2019 10.
Article in English | MEDLINE | ID: mdl-31655156

ABSTRACT

The 43-kDa transactive response DNA-binding protein (TDP-43) is an example of an RNA-binding protein that regulates RNA metabolism at multiple levels from transcription and splicing to translation. Its role in post-transcriptional RNA processing has been a primary focus of recent research, but its role in regulating transcription has been studied for only a few human genes. We characterized the effects of TDP-43 on transcription genome-wide and found that TDP-43 broadly affects transcription of protein-coding and noncoding RNA genes. Among protein-coding genes, the effects of TDP-43 were greatest for genes <30 thousand base pairs in length. Surprisingly, we found that the loss of TDP-43 resulted in increased evidence for transcription activity near repetitive Alu elements found within expressed genes. The highest densities of affected Alu elements were found in the shorter genes, whose transcription was most affected by TDP-43. Thus, in addition to its role in post-transcriptional RNA processing, TDP-43 plays a critical role in maintaining the transcriptional stability of protein-coding genes and transposable DNA elements.


Subject(s)
Alu Elements/genetics , DNA-Binding Proteins/genetics , Open Reading Frames/genetics , Transcription, Genetic , Genome, Human/genetics , Humans , RNA Splicing/genetics , RNA-Binding Proteins/genetics , Retroelements/genetics , Transcription Factors/genetics , Transcriptional Activation/genetics
11.
Retrovirology ; 16(1): 16, 2019 06 25.
Article in English | MEDLINE | ID: mdl-31238957

ABSTRACT

BACKGROUND: The human immunodeficiency virus (HIV) cell reservoir is currently a main obstacle towards complete eradication of the virus. This infected pool is refractory to anti-viral therapy and harbors integrated proviruses that are transcriptionally repressed but replication competent. As transcription silencing is key for establishing the HIV reservoir, significant efforts have been made to understand the mechanism that regulate HIV gene transcription, and the role of the elongation machinery in promoting this step. However, while the role of the super elongation complex (SEC) in enhancing transcription activation of HIV is well established, the function of SEC in modulating viral latency is less defined and its cell partners are yet to be identified. RESULTS: In this study we identify fused in sarcoma (FUS) as a partner of AFF4 in cells. FUS inhibits the activation of HIV transcription by AFF4 and ELL2, and silences overall HIV gene transcription. Concordantly, depletion of FUS elevates the occupancy of AFF4 and Cdk9 on the viral promoter and activates HIV gene transcription. Live cell imaging demonstrates that FUS co-localizes with AFF4 within nuclear punctuated condensates, which are disrupted upon treating cells with aliphatic alcohol. In HIV infected cells, knockout of FUS delays the gradual entry of HIV into latency, and similarly promotes viral activation in a T cell latency model that is treated with JQ1. Finally, effects of FUS on HIV gene transcription are also exhibited genome wide, where FUS mainly occupies gene promoters at transcription starting sites, while its knockdown leads to an increase in AFF4 and Cdk9 occupancy on gene promoters of FUS affected genes. CONCLUSIONS: Towards eliminating the HIV infected reservoir, understanding the mechanisms by which the virus persists in the face of therapy is important. Our observations show that FUS regulates both HIV and global gene transcription and modulates viral latency, thus can potentially serve as a target for future therapy that sets to reactivate HIV from its latent state.


Subject(s)
HIV-1/genetics , Proviruses/genetics , RNA-Binding Protein FUS/metabolism , Transcription, Genetic , Transcriptional Elongation Factors/genetics , Virus Latency/genetics , Cyclin-Dependent Kinase 9 , Disease Reservoirs/virology , Gene Silencing , HEK293 Cells , HIV Infections/virology , HIV-1/physiology , Humans , Jurkat Cells , Promoter Regions, Genetic , T-Lymphocytes/virology , Virus Activation
12.
Biochemistry ; 57(51): 7021-7032, 2018 12 26.
Article in English | MEDLINE | ID: mdl-30488693

ABSTRACT

Purified recombinant FUsed in Sarcoma (FUS) assembles into an oligomeric state in an RNA-dependent manner to form large condensates. FUS condensates bind and concentrate the C-terminal domain of RNA polymerase II (RNA Pol II). We asked whether a granule in cells contained FUS and RNA Pol II as suggested by the binding of FUS condensates to the polymerase. We developed cross-linking protocols to recover protein particles containing FUS from cells and separated them by size exclusion chromatography. We found a significant fraction of RNA Pol II in large granules containing FUS with diameters of >50 nm or twice that of the RNA Pol II holoenzyme. Inhibition of transcription prevented the polymerase from associating with the granules. Altogether, we found physical evidence of granules containing FUS and RNA Pol II in cells that possess properties comparable to those of in vitro FUS condensates.


Subject(s)
RNA Polymerase II/metabolism , RNA-Binding Protein FUS/metabolism , Cell Nucleus/metabolism , Cell Nucleus/ultrastructure , Cross-Linking Reagents , HEK293 Cells , Humans , Microscopy, Electron, Transmission , Models, Biological , Particle Size , Protein Interaction Domains and Motifs , RNA Polymerase II/chemistry , RNA Polymerase II/genetics , RNA-Binding Protein FUS/chemistry , RNA-Binding Protein FUS/genetics , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Transcription, Genetic
13.
Cell Rep ; 21(1): 110-125, 2017 Oct 03.
Article in English | MEDLINE | ID: mdl-28978466

ABSTRACT

Amyotrophic lateral sclerosis (ALS) is a synaptopathy accompanied by the presence of cytoplasmic aggregates containing TDP-43, an RNA-binding protein linked to ∼97% of ALS cases. Using a Drosophila model of ALS, we show that TDP-43 overexpression (OE) in motor neurons results in decreased expression of the Hsc70-4 chaperone at the neuromuscular junction (NMJ). Mechanistically, mutant TDP-43 sequesters hsc70-4 mRNA and impairs its translation. Expression of the Hsc70-4 ortholog, HSPA8, is also reduced in primary motor neurons and NMJs of mice expressing mutant TDP-43. Electrophysiology, imaging, and genetic interaction experiments reveal TDP-43-dependent defects in synaptic vesicle endocytosis. These deficits can be partially restored by OE of Hsc70-4, cysteine-string protein (Csp), or dynamin. This suggests that TDP-43 toxicity results in part from impaired activity of the synaptic CSP/Hsc70 chaperone complex impacting dynamin function. Finally, Hsc70-4/HSPA8 expression is also post-transcriptionally reduced in fly and human induced pluripotent stem cell (iPSC) C9orf72 models, suggesting a common disease pathomechanism.


Subject(s)
Amyotrophic Lateral Sclerosis/genetics , DNA-Binding Proteins/genetics , HSC70 Heat-Shock Proteins/genetics , RNA, Messenger/genetics , Synaptic Vesicles/metabolism , Amyotrophic Lateral Sclerosis/metabolism , Amyotrophic Lateral Sclerosis/pathology , Animals , C9orf72 Protein/genetics , C9orf72 Protein/metabolism , DNA-Binding Proteins/metabolism , Disease Models, Animal , Drosophila melanogaster/genetics , Drosophila melanogaster/metabolism , Dynamins/genetics , Dynamins/metabolism , Endocytosis , Gene Expression Regulation , HSC70 Heat-Shock Proteins/metabolism , HSP40 Heat-Shock Proteins/genetics , HSP40 Heat-Shock Proteins/metabolism , Humans , Induced Pluripotent Stem Cells/cytology , Induced Pluripotent Stem Cells/metabolism , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mice , Motor Neurons/metabolism , Motor Neurons/pathology , Neuromuscular Junction/metabolism , Neuromuscular Junction/pathology , Protein Aggregates , Protein Biosynthesis , RNA, Messenger/metabolism , Signal Transduction , Synaptic Transmission , Synaptic Vesicles/pathology
14.
Nucleic Acids Res ; 45(13): 7984-7996, 2017 Jul 27.
Article in English | MEDLINE | ID: mdl-28575444

ABSTRACT

RGG/RG domains are the second most common RNA binding domain in the human genome, yet their RNA-binding properties remain poorly understood. Here, we report a detailed analysis of the RNA binding characteristics of intrinsically disordered RGG/RG domains from Fused in Sarcoma (FUS), FMRP and hnRNPU. For FUS, previous studies defined RNA binding as mediated by its well-folded domains; however, we show that RGG/RG domains are the primary mediators of binding. RGG/RG domains coupled to adjacent folded domains can achieve affinities approaching that of full-length FUS. Analysis of RGG/RG domains from FUS, FMRP and hnRNPU against a spectrum of contrasting RNAs reveals that each display degenerate binding specificity, while still displaying different degrees of preference for RNA.


Subject(s)
Intrinsically Disordered Proteins/metabolism , RNA/metabolism , Animals , Fragile X Mental Retardation Protein/chemistry , Fragile X Mental Retardation Protein/metabolism , G-Quadruplexes , HEK293 Cells , Heterogeneous-Nuclear Ribonucleoprotein U/chemistry , Heterogeneous-Nuclear Ribonucleoprotein U/metabolism , Humans , Intrinsically Disordered Proteins/chemistry , Mice , Models, Biological , Protein Binding , Protein Domains , RNA/chemistry , RNA-Binding Protein FUS/chemistry , RNA-Binding Protein FUS/metabolism , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , Static Electricity
15.
Nucleic Acids Res ; 43(15): 7535-43, 2015 Sep 03.
Article in English | MEDLINE | ID: mdl-26150427

ABSTRACT

FUS, a nuclear RNA-binding protein, plays multiple roles in RNA processing. Five specific FUS-binding RNA sequence/structure motifs have been proposed, but their affinities for FUS have not been directly compared. Here we find that human FUS binds all these sequences with Kd (app) values spanning a 10-fold range. Furthermore, some RNAs that do not contain any of these motifs bind FUS with similar affinity. FUS binds RNA in a length-dependent manner, consistent with a substantial non-specific component to binding. Finally, investigation of FUS binding to different nucleic acids shows that it binds single-stranded DNA with three-fold lower affinity than ssRNA of the same length and sequence, while binding to double-stranded nucleic acids is weaker. We conclude that FUS has quite general nucleic acid-binding activity, with the various proposed RNA motifs being neither necessary for FUS binding nor sufficient to explain its diverse binding partners.


Subject(s)
RNA-Binding Protein FUS/metabolism , RNA/metabolism , DNA/metabolism , DNA, Single-Stranded/metabolism , Humans , Nucleotide Motifs , Protein Binding , RNA/chemistry
16.
Annu Rev Biochem ; 84: 355-79, 2015.
Article in English | MEDLINE | ID: mdl-25494299

ABSTRACT

Members of the FET protein family, consisting of FUS, EWSR1, and TAF15, bind to RNA and contribute to the control of transcription, RNA processing, and the cytoplasmic fates of messenger RNAs in metazoa. FET proteins can also bind DNA, which may be important in transcription and DNA damage responses. FET proteins are of medical interest because chromosomal rearrangements of their genes promote various sarcomas and because point mutations in FUS or TAF15 can cause neurodegenerative diseases such as amyotrophic lateral sclerosis and frontotemporal lobar dementia. Recent results suggest that both the normal and pathological effects of FET proteins are modulated by low-complexity or prion-like domains, which can form higher-order assemblies with novel interaction properties. Herein, we review FET proteins with an emphasis on how the biochemical properties of FET proteins may relate to their biological functions and to pathogenesis.


Subject(s)
RNA-Binding Protein FUS/metabolism , RNA-Binding Proteins/metabolism , TATA-Binding Protein Associated Factors/metabolism , Active Transport, Cell Nucleus , Animals , DNA Repair , Humans , Neoplasms/metabolism , Neurodegenerative Diseases/metabolism , RNA Processing, Post-Transcriptional , RNA-Binding Protein FUS/chemistry , RNA-Binding Proteins/chemistry , TATA-Binding Protein Associated Factors/chemistry , Transcription, Genetic
17.
Mol Biol Cell ; 25(17): 2571-8, 2014 Sep 01.
Article in English | MEDLINE | ID: mdl-25009283

ABSTRACT

Mutations in the RNA-binding protein FUS have been shown to cause the neurodegenerative disease amyotrophic lateral sclerosis (ALS). We investigate whether mutant FUS protein in ALS patient-derived fibroblasts affects normal FUS functions in the nucleus. We investigated fibroblasts from two ALS patients possessing different FUS mutations and a normal control. Fibroblasts from these patients have their nuclear FUS protein trapped in SDS-resistant aggregates. Genome-wide analysis reveals an inappropriate accumulation of Ser-2 phosphorylation on RNA polymerase II (RNA Pol II) near the transcription start sites of 625 genes for ALS patient cells and after small interfering RNA (siRNA) knockdown of FUS in normal fibroblasts. Furthermore, both the presence of mutant FUS protein and siRNA knockdown of wild-type FUS correlate with altered distribution of RNA Pol II within fibroblast nuclei. A loss of FUS function in orchestrating Ser-2 phosphorylation of the CTD of RNA Pol II is detectable in ALS patient-derived fibroblasts expressing mutant FUS protein, even when the FUS protein remains largely nuclear. A likely explanation for this loss of function is the aggregation of FUS protein in nuclei. Thus our results suggest a specific mechanism by which mutant FUS can have biological consequences other than by the formation of cytoplasmic aggregates.


Subject(s)
Amyotrophic Lateral Sclerosis/metabolism , Cell Nucleus/metabolism , RNA-Binding Protein FUS/metabolism , Amyotrophic Lateral Sclerosis/genetics , Cells, Cultured , Fibroblasts/metabolism , Humans , Phosphorylation , Protein Aggregates , RNA Interference , RNA Polymerase II/metabolism , RNA-Binding Protein FUS/antagonists & inhibitors , RNA-Binding Protein FUS/genetics , Transcription Initiation Site
18.
Cell Rep ; 5(4): 918-25, 2013 Nov 27.
Article in English | MEDLINE | ID: mdl-24268778

ABSTRACT

The abundant nuclear RNA binding protein FUS binds the C-terminal domain (CTD) of RNA polymerase II in an RNA-dependent manner, affecting Ser2 phosphorylation and transcription. Here, we examine the mechanism of this process and find that RNA binding nucleates the formation of higher-order FUS ribonucleoprotein assemblies that bind the CTD. Both the low-complexity domain and the arginine-glycine rich domain of FUS contribute to assembly. The assemblies appear fibrous by electron microscopy and have characteristics of ß zipper structures. These results support the emerging view that the pathologic protein aggregation seen in neurodegenerative diseases such as amyotrophic lateral sclerosis may occur via the exaggeration of functionally important assemblies of RNA binding proteins.


Subject(s)
RNA Polymerase II/metabolism , RNA-Binding Protein FUS/metabolism , RNA/genetics , Amyotrophic Lateral Sclerosis/genetics , Cell Line , DNA (Cytosine-5-)-Methyltransferases/genetics , HEK293 Cells , Humans , Promoter Regions, Genetic , Protein Binding , Protein Structure, Tertiary , RNA-Binding Protein FUS/biosynthesis , RNA-Binding Protein FUS/genetics , Ribonucleoproteins/biosynthesis , Ribonucleoproteins/genetics , Ribonucleoproteins/metabolism , Transcription, Genetic , DNA Methyltransferase 3B
19.
Genes Dev ; 26(24): 2690-5, 2012 Dec 15.
Article in English | MEDLINE | ID: mdl-23249733

ABSTRACT

Mutations in the RNA-binding protein FUS (fused in sarcoma)/TLS have been shown to cause the neurodegenerative disease amyotrophic lateral sclerosis (ALS), but the normal role of FUS is incompletely understood. We found that FUS binds the C-terminal domain (CTD) of RNA polymerase II (RNAP2) and prevents inappropriate hyperphosphorylation of Ser2 in the RNAP2 CTD at thousands of human genes. The loss of FUS leads to RNAP2 accumulation at the transcription start site and a shift in mRNA isoform expression toward early polyadenylation sites. Thus, in addition to its role in alternative RNA splicing, FUS has a general function in orchestrating CTD phosphorylation during RNAP2 transcription.


Subject(s)
RNA Polymerase II/metabolism , RNA-Binding Protein FUS/metabolism , Transcription, Genetic/physiology , Cell Line , Gene Expression Regulation , HEK293 Cells , HeLa Cells , Humans , Immunoglobulin G/metabolism , Phosphorylation , Protein Binding , Protein Structure, Tertiary , Serine/metabolism , Transcription Initiation Site
20.
EMBO J ; 30(16): 3242-58, 2011 Jul 29.
Article in English | MEDLINE | ID: mdl-21804531

ABSTRACT

Mammalian target of rapamycin (mTOR) complex 1 (mTORC1) is an important, highly conserved, regulator of cell growth. Ancient among the signals that regulate mTORC1 are nutrients. Amino acids direct mTORC1 to the surface of the late endosome/lysosome, where mTORC1 becomes receptive to other inputs. However, the interplay between endosomes and mTORC1 is poorly understood. Here, we report the discovery of a network that links mTORC1 to a critical component of the late endosome/lysosome, the V-ATPase. In an unbiased screen, we found that mTORC1 regulated the expression of, among other lysosomal genes, the V-ATPases. mTORC1 regulates V-ATPase expression both in cells and in mice. V-ATPase regulation by mTORC1 involves a transcription factor translocated in renal cancer, TFEB. TFEB is required for the expression of a large subset of mTORC1 responsive genes. mTORC1 coordinately regulates TFEB phosphorylation and nuclear localization and in a manner dependent on both TFEB and V-ATPases, mTORC1 promotes endocytosis. These data uncover a regulatory network linking an oncogenic transcription factor that is a master regulator of lysosomal biogenesis, TFEB, to mTORC1 and endocytosis.


Subject(s)
Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/physiology , Endocytosis/physiology , Protein Processing, Post-Translational , Proteins/physiology , Vacuolar Proton-Translocating ATPases/physiology , Amino Acid Motifs , Animals , Cell Line, Transformed/drug effects , Cell Line, Transformed/metabolism , Dactinomycin/pharmacology , Endocytosis/drug effects , Enzyme Induction/drug effects , Fibroblasts/drug effects , Fibroblasts/metabolism , Gene Expression Regulation , Lysosomes/enzymology , MAP Kinase Signaling System/drug effects , Mechanistic Target of Rapamycin Complex 1 , Mice , Multiprotein Complexes , Phosphorylation , Protein Kinase Inhibitors/pharmacology , Protein Transport/drug effects , Protein Transport/physiology , Signal Transduction/physiology , Sirolimus/pharmacology , TOR Serine-Threonine Kinases , Tuberous Sclerosis Complex 1 Protein , Tuberous Sclerosis Complex 2 Protein , Tumor Suppressor Proteins/physiology , Vacuolar Proton-Translocating ATPases/biosynthesis , Vacuolar Proton-Translocating ATPases/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...