Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters










Database
Language
Publication year range
1.
Semin Cell Dev Biol ; 156: 176-189, 2024 03 15.
Article in English | MEDLINE | ID: mdl-37268555

ABSTRACT

In recent years, there has been an emphasis on the role of phase-separated biomolecular condensates, especially stress granules, in neurodegenerative diseases such as amyotrophic lateral sclerosis (ALS). This is largely due to several ALS-associated mutations occurring in genes involved in stress granule assembly and observations that pathological inclusions detected in ALS patient neurons contain stress granule proteins, including the ALS-linked proteins TDP-43 and FUS. However, protein components of stress granules are also found in numerous other phase-separated biomolecular condensates under physiological conditions which are inadequately discussed in the context of ALS. In this review, we look beyond stress granules and describe the roles of TDP-43 and FUS in physiological condensates occurring in the nucleus and neurites, such as the nucleolus, Cajal bodies, paraspeckles and neuronal RNA transport granules. We also discuss the consequences of ALS-linked mutations in TDP-43 and FUS on their ability to phase separate into these stress-independent biomolecular condensates and perform their respective functions. Importantly, biomolecular condensates sequester multiple overlapping protein and RNA components, and their dysregulation could contribute to the observed pleiotropic effects of both sporadic and familial ALS on RNA metabolism.


Subject(s)
Amyotrophic Lateral Sclerosis , Neurodegenerative Diseases , Humans , Amyotrophic Lateral Sclerosis/metabolism , Biomolecular Condensates , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Neurodegenerative Diseases/metabolism , Mutation/genetics , RNA
2.
Nucleic Acids Res ; 50(13): 7623-7636, 2022 07 22.
Article in English | MEDLINE | ID: mdl-35801873

ABSTRACT

Processing bodies (P-bodies) are ribonucleoprotein granules that contain mRNAs, RNA-binding proteins and effectors of mRNA turnover. While P-bodies have been reported to contain translationally repressed mRNAs, a causative role for P-bodies in regulating mRNA decay has yet to be established. Enhancer of decapping protein 4 (EDC4) is a core P-body component that interacts with multiple mRNA decay factors, including the mRNA decapping (DCP2) and decay (XRN1) enzymes. EDC4 also associates with the RNA endonuclease MARF1, an interaction that antagonizes the decay of MARF1-targeted mRNAs. How EDC4 interacts with MARF1 and how it represses MARF1 activity is unclear. In this study, we show that human MARF1 and XRN1 interact with EDC4 using analogous conserved short linear motifs in a mutually exclusive manner. While the EDC4-MARF1 interaction is required for EDC4 to inhibit MARF1 activity, our data indicate that the interaction with EDC4 alone is not sufficient. Importantly, we show that P-body architecture plays a critical role in antagonizing MARF1-mediated mRNA decay. Taken together, our study suggests that P-bodies can directly regulate mRNA turnover by sequestering an mRNA decay enzyme and preventing it from interfacing with and degrading targeted mRNAs.


Subject(s)
Cell Cycle Proteins/metabolism , Endoribonucleases/metabolism , RNA Stability , Endoribonucleases/genetics , Exoribonucleases/metabolism , Humans , Microtubule-Associated Proteins/metabolism , Proteins/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA-Binding Proteins/genetics , RNA-Binding Proteins/metabolism
3.
Brain ; 144(11): 3461-3476, 2021 12 16.
Article in English | MEDLINE | ID: mdl-34115105

ABSTRACT

TDP-43 nuclear depletion and concurrent cytoplasmic accumulation in vulnerable neurons is a hallmark feature of progressive neurodegenerative proteinopathies such as amyotrophic lateral sclerosis (ALS) and frontotemporal dementia (FTD). Cellular stress signalling and stress granule dynamics are now recognized to play a role in ALS/FTD pathogenesis. Defective stress granule assembly is associated with increased cellular vulnerability and death. Ras-GAP SH3-domain-binding protein 1 (G3BP1) is a critical stress granule assembly factor. Here, we define that TDP-43 stabilizes G3BP1 transcripts via direct binding of a highly conserved cis regulatory element within the 3' untranslated region. Moreover, we show in vitro and in vivo that nuclear TDP-43 depletion is sufficient to reduce G3BP1 protein levels. Finally, we establish that G3BP1 transcripts are reduced in ALS/FTD patient neurons bearing TDP-43 cytoplasmic inclusions/nuclear depletion. Thus, our data indicate that, in ALS/FTD, there is a compromised stress granule response in disease-affected neurons due to impaired G3BP1 mRNA stability caused by TDP-43 nuclear depletion. These data implicate TDP-43 and G3BP1 loss of function as contributors to disease.


Subject(s)
Amyotrophic Lateral Sclerosis/metabolism , DNA Helicases/metabolism , DNA-Binding Proteins/metabolism , Frontotemporal Dementia/metabolism , Neurons/metabolism , Poly-ADP-Ribose Binding Proteins/metabolism , RNA Helicases/metabolism , RNA Recognition Motif Proteins/metabolism , Amyotrophic Lateral Sclerosis/pathology , Cells, Cultured , Frontotemporal Dementia/pathology , Humans , Neurons/pathology , RNA, Messenger
4.
Methods Mol Biol ; 2209: 333-345, 2021.
Article in English | MEDLINE | ID: mdl-33201479

ABSTRACT

Experiments in cell cultures have been useful for investigating a number of RNA endonucleases. However, endonuclease decay intermediates are often challenging to study in cellulo, as decay intermediates are rapidly degraded by exoribonucleases. Thus, cell-free assays have been critical for assessing endonuclease kinetics. Here, we describe such an in vitro assay to analyze endoribonuclease activity using recombinant proteins and end-radiolabeled RNA oligonucleotides. Specifically, we detail a protocol for assaying the endoribonuclease activity and kinetics of the human MARF1 protein.


Subject(s)
Cell Cycle Proteins/chemistry , Cell-Free System , Endoribonucleases/chemistry , Enzyme Assays/methods , Humans , Kinetics , RNA, Messenger/metabolism , Recombinant Proteins/chemistry
5.
Adv Exp Med Biol ; 1203: 149-164, 2019.
Article in English | MEDLINE | ID: mdl-31811634

ABSTRACT

Most eukaryotic mRNAs maintain a 5' cap structure and 3' poly(A) tail, cis-acting elements that are often separated by thousands of nucleotides. Nevertheless, multiple paradigms exist where mRNA 5' and 3' termini interact with each other in order to regulate mRNA translation and turnover. mRNAs recruit translation initiation factors to their termini, which in turn physically interact with each other. This physical bridging of the mRNA termini is known as the "closed loop" model, with years of genetic and biochemical evidence supporting the functional synergy between the 5' cap and 3' poly(A) tail to enhance mRNA translation initiation. However, a number of examples exist of "non-canonical" 5'-3' communication for cellular and viral RNAs that lack 5' cap structures and/or poly(A) tails. Moreover, in several contexts, mRNA 5'-3' communication can function to repress translation. Overall, we detail how various mRNA 5'-3' interactions play important roles in posttranscriptional regulation, wherein depending on the protein factors involved can result in translational stimulation or repression.


Subject(s)
Protein Biosynthesis , RNA, Messenger , Eukaryotic Cells , Gene Expression Regulation , Protein Biosynthesis/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA, Viral/metabolism
6.
Nucleic Acids Res ; 46(22): 12008-12021, 2018 12 14.
Article in English | MEDLINE | ID: mdl-30364987

ABSTRACT

Meiosis arrest female 1 (MARF1) is a cytoplasmic RNA binding protein that is essential for meiotic progression of mouse oocytes, in part by limiting retrotransposon expression. MARF1 is also expressed in somatic cells and tissues; however, its mechanism of action has yet to be investigated. Human MARF1 contains a NYN-like domain, two RRMs and eight LOTUS domains. Here we provide evidence that MARF1 post-transcriptionally silences targeted mRNAs. MARF1 physically interacts with the DCP1:DCP2 mRNA decapping complex but not with deadenylation machineries. Importantly, we provide a 1.7 Å resolution crystal structure of the human MARF1 NYN domain, which we demonstrate is a bona fide endoribonuclease, the activity of which is essential for the repression of MARF1-targeted mRNAs. Thus, MARF1 post-transcriptionally represses gene expression by serving as both an endoribonuclease and as a platform that recruits the DCP1:DCP2 decapping complex to targeted mRNAs.


Subject(s)
Cell Cycle Proteins/metabolism , Endoribonucleases/metabolism , RNA Interference , Trans-Activators/metabolism , Amino Acid Sequence , Animals , Binding Sites , Cell Cycle Proteins/chemistry , Cell Cycle Proteins/genetics , Crystallography, X-Ray , Endoribonucleases/chemistry , Endoribonucleases/genetics , Escherichia coli/genetics , Escherichia coli/metabolism , Gene Expression , HEK293 Cells , HeLa Cells , Humans , Mice , Models, Molecular , Protein Binding , Protein Conformation, alpha-Helical , Protein Conformation, beta-Strand , Protein Interaction Domains and Motifs , Protein Isoforms/chemistry , Protein Isoforms/genetics , Protein Isoforms/metabolism , RNA Cleavage , RNA, Messenger/chemistry , RNA, Messenger/genetics , RNA, Messenger/metabolism , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Sequence Homology, Amino Acid , Trans-Activators/chemistry , Trans-Activators/genetics
7.
EMBO J ; 37(7)2018 04 03.
Article in English | MEDLINE | ID: mdl-29510985

ABSTRACT

The LSM domain-containing protein LSM14/Rap55 plays a role in mRNA decapping, translational repression, and RNA granule (P-body) assembly. How LSM14 interacts with the mRNA silencing machinery, including the eIF4E-binding protein 4E-T and the DEAD-box helicase DDX6, is poorly understood. Here we report the crystal structure of the LSM domain of LSM14 bound to a highly conserved C-terminal fragment of 4E-T. The 4E-T C-terminus forms a bi-partite motif that wraps around the N-terminal LSM domain of LSM14. We also determined the crystal structure of LSM14 bound to the C-terminal RecA-like domain of DDX6. LSM14 binds DDX6 via a unique non-contiguous motif with distinct directionality as compared to other DDX6-interacting proteins. Together with mutational and proteomic studies, the LSM14-DDX6 structure reveals that LSM14 has adopted a divergent mode of binding DDX6 in order to support the formation of mRNA silencing complexes and P-body assembly.


Subject(s)
DEAD-box RNA Helicases/chemistry , DEAD-box RNA Helicases/metabolism , Protein Interaction Domains and Motifs , Proto-Oncogene Proteins/chemistry , Proto-Oncogene Proteins/metabolism , RNA Interference/physiology , RNA, Messenger/metabolism , Ribonucleoproteins/chemistry , Ribonucleoproteins/metabolism , Amino Acid Sequence , Animals , Binding Sites , Caenorhabditis elegans , Crystallography, X-Ray , DEAD-box RNA Helicases/genetics , Drosophila melanogaster , Eukaryotic Initiation Factor-4E/metabolism , HeLa Cells , Humans , Hydrophobic and Hydrophilic Interactions , Models, Molecular , Mutation , Protein Binding , Protein Conformation , Protein Structure, Secondary , Proteins/chemistry , Proteins/metabolism , Proteomics , Proto-Oncogene Proteins/genetics , Rec A Recombinases/chemistry , Recombinant Proteins/chemistry , Ribonucleoproteins/genetics , Sequence Alignment
8.
Oncotarget ; 8(41): 69328-69350, 2017 Sep 19.
Article in English | MEDLINE | ID: mdl-29050207

ABSTRACT

A yeast culture grown in a nutrient-rich medium initially containing 2% glucose is not limited in calorie supply. When yeast cells cultured in this medium consume glucose, they undergo cell cycle arrest at a checkpoint in late G1 and differentiate into quiescent and non-quiescent cell populations. Studies of such differentiation have provided insights into mechanisms of yeast chronological aging under conditions of excessive calorie intake. Caloric restriction is an aging-delaying dietary intervention. Here, we assessed how caloric restriction influences the differentiation of chronologically aging yeast cultures into quiescent and non-quiescent cells, and how it affects their properties. We found that caloric restriction extends yeast chronological lifespan via a mechanism linking cellular aging to cell cycle regulation, maintenance of quiescence, entry into a non-quiescent state and survival in this state. Our findings suggest that caloric restriction delays yeast chronological aging by causing specific changes in the following: 1) a checkpoint in G1 for cell cycle arrest and entry into a quiescent state; 2) a growth phase in which high-density quiescent cells are committed to become low-density quiescent cells; 3) the differentiation of low-density quiescent cells into low-density non-quiescent cells; and 4) the conversion of high-density quiescent cells into high-density non-quiescent cells.

9.
Cell Rep ; 11(9): 1425-36, 2015 Jun 09.
Article in English | MEDLINE | ID: mdl-26027925

ABSTRACT

Eukaryotic mRNA degradation often initiates with the recruitment of the CCR4-NOT deadenylase complex and decay factors to the mRNA 3' terminus. How the 3'-proximal decay machinery interacts with the 5'-terminal cap structure in order to engender mRNA decapping and 5'-3' degradation is unclear. Human 4E-T is an eIF4E-binding protein that has been reported to promote mRNA decay, albeit via an unknown mechanism. Here, we show that 4E-T is a component of the mRNA decay machinery and interacts with factors including DDX6, LSM14, and the LSM1-7-PAT1 complex. We also provide evidence that 4E-T associates with, and enhances the decay of, mRNAs targeted by the CCR4-NOT deadenylase complex, including microRNA targets. Importantly, we demonstrate that 4E-T must interact with eIF4E to engender mRNA decay. Taken together, our data support a model where 4E-T promotes mRNA turnover by physically linking the 3'-terminal mRNA decay machinery to the 5' cap via its interaction with eIF4E.


Subject(s)
Nucleocytoplasmic Transport Proteins/metabolism , RNA Interference/physiology , RNA Stability/physiology , RNA, Messenger/metabolism , HeLa Cells , Humans , Immunoprecipitation , RNA, Small Interfering , Transfection
SELECTION OF CITATIONS
SEARCH DETAIL