Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 4.236
Filter
Add more filters

Publication year range
1.
Cell ; 187(8): 1971-1989.e16, 2024 Apr 11.
Article in English | MEDLINE | ID: mdl-38521060

ABSTRACT

Amyotrophic lateral sclerosis (ALS) and frontotemporal lobar degeneration (FTLD) share many clinical, pathological, and genetic features, but a detailed understanding of their associated transcriptional alterations across vulnerable cortical cell types is lacking. Here, we report a high-resolution, comparative single-cell molecular atlas of the human primary motor and dorsolateral prefrontal cortices and their transcriptional alterations in sporadic and familial ALS and FTLD. By integrating transcriptional and genetic information, we identify known and previously unidentified vulnerable populations in cortical layer 5 and show that ALS- and FTLD-implicated motor and spindle neurons possess a virtually indistinguishable molecular identity. We implicate potential disease mechanisms affecting these cell types as well as non-neuronal drivers of pathogenesis. Finally, we show that neuron loss in cortical layer 5 tracks more closely with transcriptional identity rather than cellular morphology and extends beyond previously reported vulnerable cell types.


Subject(s)
Amyotrophic Lateral Sclerosis , Frontotemporal Lobar Degeneration , Prefrontal Cortex , Animals , Humans , Mice , Amyotrophic Lateral Sclerosis/genetics , Amyotrophic Lateral Sclerosis/metabolism , Amyotrophic Lateral Sclerosis/pathology , Frontotemporal Dementia/genetics , Frontotemporal Lobar Degeneration/genetics , Frontotemporal Lobar Degeneration/metabolism , Frontotemporal Lobar Degeneration/pathology , Gene Expression Profiling , Neurons/metabolism , Prefrontal Cortex/metabolism , Prefrontal Cortex/pathology , Single-Cell Gene Expression Analysis
2.
Cell ; 186(4): 786-802.e28, 2023 02 16.
Article in English | MEDLINE | ID: mdl-36754049

ABSTRACT

Amyotrophic lateral sclerosis (ALS) is a fatal neurodegenerative disease that results from many diverse genetic causes. Although therapeutics specifically targeting known causal mutations may rescue individual types of ALS, these approaches cannot treat most cases since they have unknown genetic etiology. Thus, there is a pressing need for therapeutic strategies that rescue multiple forms of ALS. Here, we show that pharmacological inhibition of PIKFYVE kinase activates an unconventional protein clearance mechanism involving exocytosis of aggregation-prone proteins. Reducing PIKFYVE activity ameliorates ALS pathology and extends survival of animal models and patient-derived motor neurons representing diverse forms of ALS including C9ORF72, TARDBP, FUS, and sporadic. These findings highlight a potential approach for mitigating ALS pathogenesis that does not require stimulating macroautophagy or the ubiquitin-proteosome system.


Subject(s)
Amyotrophic Lateral Sclerosis , Phosphatidylinositol 3-Kinases , Animals , Amyotrophic Lateral Sclerosis/drug therapy , Amyotrophic Lateral Sclerosis/genetics , Amyotrophic Lateral Sclerosis/metabolism , Motor Neurons , Mutation , RNA-Binding Protein FUS/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Disease Models, Animal
3.
Cell ; 183(3): 636-649.e18, 2020 10 29.
Article in English | MEDLINE | ID: mdl-33031745

ABSTRACT

Cytoplasmic accumulation of TDP-43 is a disease hallmark for many cases of amyotrophic lateral sclerosis (ALS), associated with a neuroinflammatory cytokine profile related to upregulation of nuclear factor κB (NF-κB) and type I interferon (IFN) pathways. Here we show that this inflammation is driven by the cytoplasmic DNA sensor cyclic guanosine monophosphate (GMP)-AMP synthase (cGAS) when TDP-43 invades mitochondria and releases DNA via the permeability transition pore. Pharmacologic inhibition or genetic deletion of cGAS and its downstream signaling partner STING prevents upregulation of NF-κB and type I IFN induced by TDP-43 in induced pluripotent stem cell (iPSC)-derived motor neurons and in TDP-43 mutant mice. Finally, we document elevated levels of the specific cGAS signaling metabolite cGAMP in spinal cord samples from patients, which may be a biomarker of mtDNA release and cGAS/STING activation in ALS. Our results identify mtDNA release and cGAS/STING activation as critical determinants of TDP-43-associated pathology and demonstrate the potential for targeting this pathway in ALS.


Subject(s)
Amyotrophic Lateral Sclerosis/metabolism , DNA, Mitochondrial/metabolism , DNA-Binding Proteins/metabolism , Membrane Proteins/metabolism , Mitochondrial Permeability Transition Pore/metabolism , Nucleotidyltransferases/metabolism , Alarmins/metabolism , Amyotrophic Lateral Sclerosis/pathology , Animals , Cytoplasm/metabolism , Disease Models, Animal , Disease Progression , HEK293 Cells , Humans , Induced Pluripotent Stem Cells/metabolism , Inflammation/metabolism , Interferon Type I/metabolism , Mice , Mice, Inbred C57BL , Mitochondria/metabolism , NF-kappa B/metabolism , Nerve Degeneration/pathology , Phosphotransferases (Alcohol Group Acceptor) , Protein Subunits/metabolism , Signal Transduction
4.
Cell ; 179(1): 193-204.e14, 2019 Sep 19.
Article in English | MEDLINE | ID: mdl-31495574

ABSTRACT

Numerous interventions are in clinical development for respiratory syncytial virus (RSV) infection, including small molecules that target viral transcription and replication. These processes are catalyzed by a complex comprising the RNA-dependent RNA polymerase (L) and the tetrameric phosphoprotein (P). RSV P recruits multiple proteins to the polymerase complex and, with the exception of its oligomerization domain, is thought to be intrinsically disordered. Despite their critical roles in RSV transcription and replication, structures of L and P have remained elusive. Here, we describe the 3.2-Å cryo-EM structure of RSV L bound to tetrameric P. The structure reveals a striking tentacular arrangement of P, with each of the four monomers adopting a distinct conformation. The structure also rationalizes inhibitor escape mutants and mutations observed in live-attenuated vaccine candidates. These results provide a framework for determining the molecular underpinnings of RSV replication and transcription and should facilitate the design of effective RSV inhibitors.


Subject(s)
Phosphoproteins/ultrastructure , RNA-Dependent RNA Polymerase/ultrastructure , Respiratory Syncytial Virus Infections/virology , Respiratory Syncytial Virus, Human/enzymology , Viral Proteins/ultrastructure , Acetates/chemistry , Animals , Antiviral Agents/chemistry , Antiviral Agents/therapeutic use , Catalytic Domain , Cryoelectron Microscopy , Deoxycytidine/analogs & derivatives , Deoxycytidine/chemistry , Deoxycytidine/pharmacology , Deoxycytidine/therapeutic use , Hydrogen Bonding , Hydrophobic and Hydrophilic Interactions , Phosphoproteins/chemistry , Phosphoproteins/metabolism , Protein Conformation, alpha-Helical , Protein Interaction Domains and Motifs , Quinolines/chemistry , RNA-Dependent RNA Polymerase/antagonists & inhibitors , RNA-Dependent RNA Polymerase/chemistry , RNA-Dependent RNA Polymerase/metabolism , Respiratory Syncytial Virus Infections/drug therapy , Respiratory Syncytial Virus Vaccines/chemistry , Sf9 Cells , Spodoptera , Viral Proteins/chemistry , Viral Proteins/metabolism , Virus Replication/drug effects
5.
Cell ; 173(4): 958-971.e17, 2018 05 03.
Article in English | MEDLINE | ID: mdl-29628143

ABSTRACT

Defects in nucleocytoplasmic transport have been identified as a key pathogenic event in amyotrophic lateral sclerosis (ALS) and frontotemporal dementia (FTD) mediated by a GGGGCC hexanucleotide repeat expansion in C9ORF72, the most common genetic cause of ALS/FTD. Furthermore, nucleocytoplasmic transport disruption has also been implicated in other neurodegenerative diseases with protein aggregation, suggesting a shared mechanism by which protein stress disrupts nucleocytoplasmic transport. Here, we show that cellular stress disrupts nucleocytoplasmic transport by localizing critical nucleocytoplasmic transport factors into stress granules, RNA/protein complexes that play a crucial role in ALS pathogenesis. Importantly, inhibiting stress granule assembly, such as by knocking down Ataxin-2, suppresses nucleocytoplasmic transport defects as well as neurodegeneration in C9ORF72-mediated ALS/FTD. Our findings identify a link between stress granule assembly and nucleocytoplasmic transport, two fundamental cellular processes implicated in the pathogenesis of C9ORF72-mediated ALS/FTD and other neurodegenerative diseases.


Subject(s)
Active Transport, Cell Nucleus/physiology , Amyotrophic Lateral Sclerosis/pathology , Ataxin-2/metabolism , C9orf72 Protein/genetics , Frontotemporal Dementia/pathology , Active Transport, Cell Nucleus/drug effects , Aged , Amyotrophic Lateral Sclerosis/metabolism , Arsenites/toxicity , Ataxin-2/antagonists & inhibitors , Ataxin-2/genetics , C9orf72 Protein/metabolism , DNA Repeat Expansion/genetics , Female , Frontotemporal Dementia/metabolism , HEK293 Cells , Humans , Male , Membrane Glycoproteins/metabolism , Middle Aged , Nuclear Pore Complex Proteins/metabolism , Oxidative Stress/drug effects , RNA Interference , RNA, Small Interfering/metabolism , Sodium Compounds/toxicity , alpha Karyopherins/antagonists & inhibitors , alpha Karyopherins/genetics , alpha Karyopherins/metabolism , beta Karyopherins/antagonists & inhibitors , beta Karyopherins/genetics , beta Karyopherins/metabolism , ran GTP-Binding Protein/antagonists & inhibitors , ran GTP-Binding Protein/genetics , ran GTP-Binding Protein/metabolism
6.
Cell ; 174(6): 1477-1491.e19, 2018 09 06.
Article in English | MEDLINE | ID: mdl-30146158

ABSTRACT

Aging is a major risk factor for both genetic and sporadic neurodegenerative disorders. However, it is unclear how aging interacts with genetic predispositions to promote neurodegeneration. Here, we investigate how partial loss of function of TBK1, a major genetic cause for amyotrophic lateral sclerosis (ALS) and frontotemporal dementia (FTD) comorbidity, leads to age-dependent neurodegeneration. We show that TBK1 is an endogenous inhibitor of RIPK1 and the embryonic lethality of Tbk1-/- mice is dependent on RIPK1 kinase activity. In aging human brains, another endogenous RIPK1 inhibitor, TAK1, exhibits a marked decrease in expression. We show that in Tbk1+/- mice, the reduced myeloid TAK1 expression promotes all the key hallmarks of ALS/FTD, including neuroinflammation, TDP-43 aggregation, axonal degeneration, neuronal loss, and behavior deficits, which are blocked upon inhibition of RIPK1. Thus, aging facilitates RIPK1 activation by reducing TAK1 expression, which cooperates with genetic risk factors to promote the onset of ALS/FTD.


Subject(s)
Apoptosis , Protein Serine-Threonine Kinases/metabolism , Receptor-Interacting Protein Serine-Threonine Kinases/metabolism , Adult , Aged , Aging , Animals , Apoptosis/drug effects , Axons/metabolism , Behavior, Animal , Brain/cytology , Brain/metabolism , Cells, Cultured , Humans , I-kappa B Kinase/metabolism , Mice , Mice, Knockout , Microglia/cytology , Microglia/drug effects , Microglia/metabolism , Phosphorylation/drug effects , Protein Serine-Threonine Kinases/deficiency , Protein Serine-Threonine Kinases/genetics , Receptor-Interacting Protein Serine-Threonine Kinases/deficiency , Receptor-Interacting Protein Serine-Threonine Kinases/genetics , Spinal Cord/metabolism , Staurosporine/pharmacology , Tumor Necrosis Factor-alpha/pharmacology
7.
Cell ; 172(4): 696-705.e12, 2018 02 08.
Article in English | MEDLINE | ID: mdl-29398115

ABSTRACT

Protein aggregation and dysfunction of the ubiquitin-proteasome system are hallmarks of many neurodegenerative diseases. Here, we address the elusive link between these phenomena by employing cryo-electron tomography to dissect the molecular architecture of protein aggregates within intact neurons at high resolution. We focus on the poly-Gly-Ala (poly-GA) aggregates resulting from aberrant translation of an expanded GGGGCC repeat in C9orf72, the most common genetic cause of amyotrophic lateral sclerosis and frontotemporal dementia. We find that poly-GA aggregates consist of densely packed twisted ribbons that recruit numerous 26S proteasome complexes, while other macromolecules are largely excluded. Proximity to poly-GA ribbons stabilizes a transient substrate-processing conformation of the 26S proteasome, suggesting stalled degradation. Thus, poly-GA aggregates may compromise neuronal proteostasis by driving the accumulation and functional impairment of a large fraction of cellular proteasomes.


Subject(s)
Alanine/analogs & derivatives , C9orf72 Protein , Neurons , Polyglutamic Acid , Proteasome Endopeptidase Complex , Protein Aggregates , Alanine/genetics , Alanine/metabolism , Amyotrophic Lateral Sclerosis/genetics , Amyotrophic Lateral Sclerosis/metabolism , Amyotrophic Lateral Sclerosis/pathology , Animals , C9orf72 Protein/genetics , C9orf72 Protein/metabolism , Frontotemporal Dementia/genetics , Frontotemporal Dementia/metabolism , Frontotemporal Dementia/pathology , HEK293 Cells , Humans , Neurons/metabolism , Neurons/pathology , Polyglutamic Acid/genetics , Polyglutamic Acid/metabolism , Proteasome Endopeptidase Complex/genetics , Proteasome Endopeptidase Complex/metabolism , Protein Biosynthesis , Protein Stability , Protein Structure, Quaternary , Rats , Rats, Sprague-Dawley
8.
Cell ; 173(3): 677-692.e20, 2018 04 19.
Article in English | MEDLINE | ID: mdl-29677512

ABSTRACT

RNA-binding proteins (RBPs) with prion-like domains (PrLDs) phase transition to functional liquids, which can mature into aberrant hydrogels composed of pathological fibrils that underpin fatal neurodegenerative disorders. Several nuclear RBPs with PrLDs, including TDP-43, FUS, hnRNPA1, and hnRNPA2, mislocalize to cytoplasmic inclusions in neurodegenerative disorders, and mutations in their PrLDs can accelerate fibrillization and cause disease. Here, we establish that nuclear-import receptors (NIRs) specifically chaperone and potently disaggregate wild-type and disease-linked RBPs bearing a NLS. Karyopherin-ß2 (also called Transportin-1) engages PY-NLSs to inhibit and reverse FUS, TAF15, EWSR1, hnRNPA1, and hnRNPA2 fibrillization, whereas Importin-α plus Karyopherin-ß1 prevent and reverse TDP-43 fibrillization. Remarkably, Karyopherin-ß2 dissolves phase-separated liquids and aberrant fibrillar hydrogels formed by FUS and hnRNPA1. In vivo, Karyopherin-ß2 prevents RBPs with PY-NLSs accumulating in stress granules, restores nuclear RBP localization and function, and rescues degeneration caused by disease-linked FUS and hnRNPA2. Thus, NIRs therapeutically restore RBP homeostasis and mitigate neurodegeneration.


Subject(s)
Active Transport, Cell Nucleus , Prions/chemistry , RNA-Binding Proteins/chemistry , Receptors, Cytoplasmic and Nuclear/chemistry , Adult , Aged , Animals , Cytoplasm/chemistry , DNA-Binding Proteins/chemistry , Drosophila melanogaster , Female , Green Fluorescent Proteins/chemistry , HEK293 Cells , HeLa Cells , Homeostasis , Humans , Karyopherins/chemistry , Male , Middle Aged , Molecular Chaperones/chemistry , Mutation , Neurodegenerative Diseases/pathology , Protein Domains , RNA-Binding Protein EWS/chemistry , TATA-Binding Protein Associated Factors/chemistry , beta Karyopherins/chemistry
9.
Cell ; 173(3): 706-719.e13, 2018 04 19.
Article in English | MEDLINE | ID: mdl-29677514

ABSTRACT

Cytoplasmic FUS aggregates are a pathological hallmark in a subset of patients with frontotemporal dementia (FTD) or amyotrophic lateral sclerosis (ALS). A key step that is disrupted in these patients is nuclear import of FUS mediated by the import receptor Transportin/Karyopherin-ß2. In ALS-FUS patients, this is caused by mutations in the nuclear localization signal (NLS) of FUS that weaken Transportin binding. In FTD-FUS patients, Transportin is aggregated, and post-translational arginine methylation, which regulates the FUS-Transportin interaction, is lost. Here, we show that Transportin and arginine methylation have a crucial function beyond nuclear import-namely to suppress RGG/RG-driven phase separation and stress granule association of FUS. ALS-associated FUS-NLS mutations weaken the chaperone activity of Transportin and loss of FUS arginine methylation, as seen in FTD-FUS, promote phase separation, and stress granule partitioning of FUS. Our findings reveal two regulatory mechanisms of liquid-phase homeostasis that are disrupted in FUS-associated neurodegeneration.


Subject(s)
Arginine/chemistry , RNA-Binding Protein FUS/chemistry , beta Karyopherins/chemistry , Active Transport, Cell Nucleus , Amino Acid Motifs , Cytoplasm/metabolism , DNA Methylation , DNA, Complementary/metabolism , Densitometry , Frontotemporal Lobar Degeneration/metabolism , HeLa Cells , Homeostasis , Humans , Karyopherins/chemistry , Magnetic Resonance Spectroscopy , Methylation , Molecular Chaperones/chemistry , Mutation , Neurodegenerative Diseases/metabolism , Protein Binding , Protein Domains
10.
Cell ; 170(5): 899-912.e10, 2017 Aug 24.
Article in English | MEDLINE | ID: mdl-28803727

ABSTRACT

Microsatellite repeat expansions in DNA produce pathogenic RNA species that cause dominantly inherited diseases such as myotonic dystrophy type 1 and 2 (DM1/2), Huntington's disease, and C9orf72-linked amyotrophic lateral sclerosis (C9-ALS). Means to target these repetitive RNAs are required for diagnostic and therapeutic purposes. Here, we describe the development of a programmable CRISPR system capable of specifically visualizing and eliminating these toxic RNAs. We observe specific targeting and efficient elimination of microsatellite repeat expansion RNAs both when exogenously expressed and in patient cells. Importantly, RNA-targeting Cas9 (RCas9) reverses hallmark features of disease including elimination of RNA foci among all conditions studied (DM1, DM2, C9-ALS, polyglutamine diseases), reduction of polyglutamine protein products, relocalization of repeat-bound proteins to resemble healthy controls, and efficient reversal of DM1-associated splicing abnormalities in patient myotubes. Finally, we report a truncated RCas9 system compatible with adeno-associated viral packaging. This effort highlights the potential of RCas9 for human therapeutics.


Subject(s)
Clustered Regularly Interspaced Short Palindromic Repeats , Genetic Therapy/methods , Oligonucleotides, Antisense/pharmacology , Animals , COS Cells , Cell Line , Cells, Cultured , Chlorocebus aethiops , Microsatellite Repeats , RNA Splicing , Trinucleotide Repeat Expansion
11.
Cell ; 171(5): 994-1000, 2017 Nov 16.
Article in English | MEDLINE | ID: mdl-29149615

ABSTRACT

Eukaryotic translation is tightly regulated to ensure that protein production occurs at the right time and place. Recent studies on abnormal repeat proteins, especially in age-dependent neurodegenerative diseases caused by nucleotide repeat expansion, have highlighted or identified two forms of unconventional translation initiation: usage of AUG-like sites (near cognates) or repeat-associated non-AUG (RAN) translation. We discuss how repeat proteins may differ due to not just unconventional initiation, but also ribosomal frameshifting and/or imperfect repeat DNA replication, expansion, and repair, and we highlight how research on translation of repeats may uncover insights into the biology of translation and its contribution to disease.


Subject(s)
Neurodegenerative Diseases/genetics , Protein Biosynthesis , Animals , Codon, Initiator , Frameshifting, Ribosomal , Humans , Neurodegenerative Diseases/metabolism , Open Reading Frames , Regulatory Sequences, Ribonucleic Acid , Trinucleotide Repeat Expansion
12.
Genes Dev ; 38(1-2): 11-30, 2024 02 13.
Article in English | MEDLINE | ID: mdl-38182429

ABSTRACT

Amyotrophic lateral sclerosis (ALS) is a debilitating neurodegenerative disease characterized by loss of motor neurons. Human genetic studies have linked mutations in RNA-binding proteins as causative for this disease. The hnRNPA1 protein, a known pre-mRNA splicing factor, is mutated in some ALS patients. Here, two human cell models were generated to investigate how a mutation in the C-terminal low-complexity domain (LCD) of hnRNPA1 can cause splicing changes of thousands of transcripts that collectively are linked to the DNA damage response, cilium organization, and translation. We show that the hnRNPA1 D262V mutant protein binds to new binding sites on differentially spliced transcripts from genes that are linked to ALS. We demonstrate that this ALS-linked hnRNPA1 mutation alters normal RNA-dependent protein-protein interactions. Furthermore, cells expressing this hnRNPA1 mutant exhibit a cell aggregation phenotype, markedly reduced growth rates, changes in stress granule kinetics, and aberrant growth of neuronal processes. This study provides insight into how a single amino acid mutation in a splicing factor can alter RNA splicing networks of genes linked to ALS.


Subject(s)
Amyotrophic Lateral Sclerosis , Heterogeneous-Nuclear Ribonucleoprotein Group A-B , Neurodegenerative Diseases , Humans , Amyotrophic Lateral Sclerosis/genetics , Amyotrophic Lateral Sclerosis/metabolism , Heterogeneous-Nuclear Ribonucleoprotein Group A-B/genetics , Heterogeneous-Nuclear Ribonucleoprotein Group A-B/metabolism , Mutation , RNA Splicing/genetics , RNA Splicing Factors/genetics
13.
Mol Cell ; 83(2): 219-236.e7, 2023 Jan 19.
Article in English | MEDLINE | ID: mdl-36634675

ABSTRACT

RNA methylation at adenosine N6 (m6A) is one of the most common RNA modifications, impacting RNA stability, transport, and translation. Previous studies uncovered RNA destabilization in amyotrophic lateral sclerosis (ALS) models in association with accumulation of the RNA-binding protein TDP43. Here, we show that TDP43 recognizes m6A RNA and that RNA methylation is critical for both TDP43 binding and autoregulation. We also observed extensive RNA hypermethylation in ALS spinal cord, corresponding to methylated TDP43 substrates. Emphasizing the importance of m6A for TDP43 binding and function, we identified several m6A factors that enhance or suppress TDP43-mediated toxicity via single-cell CRISPR-Cas9 in primary neurons. The most promising modifier-the canonical m6A reader YTHDF2-accumulated within ALS spinal neurons, and its knockdown prolonged the survival of human neurons carrying ALS-associated mutations. Collectively, these data show that m6A modifications modulate RNA binding by TDP43 and that m6A is pivotal for TDP43-related neurodegeneration in ALS.


Subject(s)
Amyotrophic Lateral Sclerosis , Frontotemporal Dementia , Humans , Amyotrophic Lateral Sclerosis/pathology , Frontotemporal Dementia/genetics , Frontotemporal Dementia/metabolism , Methylation , Neurons/metabolism , RNA/genetics , RNA/metabolism
14.
Mol Cell ; 83(17): 3188-3204.e7, 2023 09 07.
Article in English | MEDLINE | ID: mdl-37683611

ABSTRACT

Failure to clear damaged mitochondria via mitophagy disrupts physiological function and may initiate damage signaling via inflammatory cascades, although how these pathways intersect remains unclear. We discovered that nuclear factor kappa B (NF-κB) essential regulator NF-κB effector molecule (NEMO) is recruited to damaged mitochondria in a Parkin-dependent manner in a time course similar to recruitment of the structurally related mitophagy adaptor, optineurin (OPTN). Upon recruitment, NEMO partitions into phase-separated condensates distinct from OPTN but colocalizing with p62/SQSTM1. NEMO recruitment, in turn, recruits the active catalytic inhibitor of kappa B kinase (IKK) component phospho-IKKß, initiating NF-κB signaling and the upregulation of inflammatory cytokines. Consistent with a potential neuroinflammatory role, NEMO is recruited to mitochondria in primary astrocytes upon oxidative stress. These findings suggest that damaged, ubiquitinated mitochondria serve as an intracellular platform to initiate innate immune signaling, promoting the formation of activated IKK complexes sufficient to activate NF-κB signaling. We propose that mitophagy and NF-κB signaling are initiated as parallel pathways in response to mitochondrial stress.


Subject(s)
NF-kappa B , Signal Transduction , NF-kappa B/genetics , I-kappa B Kinase/genetics , Protein Serine-Threonine Kinases/genetics , Mitochondria/genetics
15.
Mol Cell ; 79(3): 443-458.e7, 2020 08 06.
Article in English | MEDLINE | ID: mdl-32649883

ABSTRACT

Despite the prominent role of TDP-43 in neurodegeneration, its physiological and pathological functions are not fully understood. Here, we report an unexpected role of TDP-43 in the formation of dynamic, reversible, liquid droplet-like nuclear bodies (NBs) in response to stress. Formation of NBs alleviates TDP-43-mediated cytotoxicity in mammalian cells and fly neurons. Super-resolution microscopy reveals distinct functions of the two RRMs in TDP-43 NB formation. TDP-43 NBs are partially colocalized with nuclear paraspeckles, whose scaffolding lncRNA NEAT1 is dramatically upregulated in stressed neurons. Moreover, increase of NEAT1 promotes TDP-43 liquid-liquid phase separation (LLPS) in vitro. Finally, we discover that the ALS-associated mutation D169G impairs the NEAT1-mediated TDP-43 LLPS and NB assembly, causing excessive cytoplasmic translocation of TDP-43 to form stress granules, which become phosphorylated TDP-43 cytoplasmic foci upon prolonged stress. Together, our findings suggest a stress-mitigating role and mechanism of TDP-43 NBs, whose dysfunction may be involved in ALS pathogenesis.


Subject(s)
Amyotrophic Lateral Sclerosis/genetics , DNA-Binding Proteins/genetics , Intranuclear Inclusion Bodies/metabolism , Neurons/metabolism , RNA, Long Noncoding/genetics , Amyotrophic Lateral Sclerosis/metabolism , Amyotrophic Lateral Sclerosis/pathology , Animals , Animals, Genetically Modified , Arsenites/pharmacology , Cerebral Cortex/drug effects , Cerebral Cortex/metabolism , Cerebral Cortex/ultrastructure , Cytoplasmic Granules/drug effects , Cytoplasmic Granules/metabolism , Cytoplasmic Granules/ultrastructure , DNA-Binding Proteins/metabolism , Disease Models, Animal , Drosophila melanogaster , Gene Expression Regulation , HEK293 Cells , HeLa Cells , Humans , Intranuclear Inclusion Bodies/drug effects , Intranuclear Inclusion Bodies/ultrastructure , Mice , Mutation , Neurons/drug effects , Neurons/ultrastructure , Primary Cell Culture , Protein Transport/drug effects , RNA, Long Noncoding/metabolism , Signal Transduction , Stress, Physiological
16.
Mol Cell ; 80(4): 666-681.e8, 2020 11 19.
Article in English | MEDLINE | ID: mdl-33159856

ABSTRACT

The RNA-binding protein fused in sarcoma (FUS) can form pathogenic inclusions in neurodegenerative diseases like amyotrophic lateral sclerosis (ALS) and frontotemporal lobar dementia (FTLD). Over 70 mutations in Fus are linked to ALS/FTLD. In patients, all Fus mutations are heterozygous, indicating that the mutant drives disease progression despite the presence of wild-type (WT) FUS. Here, we demonstrate that ALS/FTLD-linked FUS mutations in glycine (G) strikingly drive formation of droplets that do not readily interact with WT FUS, whereas arginine (R) mutants form mixed condensates with WT FUS. Remarkably, interactions between WT and G mutants are disfavored at the earliest stages of FUS nucleation. In contrast, R mutants physically interact with the WT FUS such that WT FUS recovers the mutant defects by reducing droplet size and increasing dynamic interactions with RNA. This result suggests disparate molecular mechanisms underlying ALS/FTLD pathogenesis and differing recovery potential depending on the type of mutation.


Subject(s)
Amyotrophic Lateral Sclerosis/pathology , Frontotemporal Dementia/pathology , Glycine/metabolism , Mutation , Neuroblastoma/pathology , RNA-Binding Protein FUS/chemistry , RNA-Binding Protein FUS/metabolism , RNA/metabolism , Amyotrophic Lateral Sclerosis/genetics , Frontotemporal Dementia/genetics , Glycine/chemistry , Glycine/genetics , Humans , Inclusion Bodies , Neuroblastoma/genetics , Neuroblastoma/metabolism , Protein Conformation , RNA/chemistry , RNA/genetics , RNA-Binding Protein FUS/genetics , Tumor Cells, Cultured
17.
Mol Cell ; 79(1): 54-67.e7, 2020 07 02.
Article in English | MEDLINE | ID: mdl-32521226

ABSTRACT

Exposure of cells to heat or oxidative stress causes misfolding of proteins. To avoid toxic protein aggregation, cells have evolved nuclear and cytosolic protein quality control (PQC) systems. In response to proteotoxic stress, cells also limit protein synthesis by triggering transient storage of mRNAs and RNA-binding proteins (RBPs) in cytosolic stress granules (SGs). We demonstrate that the SUMO-targeted ubiquitin ligase (StUbL) pathway, which is part of the nuclear proteostasis network, regulates SG dynamics. We provide evidence that inactivation of SUMO deconjugases under proteotoxic stress initiates SUMO-primed, RNF4-dependent ubiquitylation of RBPs that typically condense into SGs. Impairment of SUMO-primed ubiquitylation drastically delays SG resolution upon stress release. Importantly, the StUbL system regulates compartmentalization of an amyotrophic lateral sclerosis (ALS)-associated FUS mutant in SGs. We propose that the StUbL system functions as surveillance pathway for aggregation-prone RBPs in the nucleus, thereby linking the nuclear and cytosolic axis of proteotoxic stress response.


Subject(s)
Amyotrophic Lateral Sclerosis/pathology , Cell Nucleus/metabolism , Cytoplasmic Granules/metabolism , Nuclear Proteins/metabolism , RNA-Binding Proteins/metabolism , SUMO-1 Protein/metabolism , Transcription Factors/metabolism , Ubiquitin/metabolism , Amyotrophic Lateral Sclerosis/genetics , Cell Nucleus/genetics , HeLa Cells , Heat-Shock Response , Humans , Mutation , Nuclear Proteins/genetics , Proteolysis , RNA-Binding Protein FUS/genetics , RNA-Binding Proteins/genetics , SUMO-1 Protein/genetics , Sumoylation , Transcription Factors/genetics , Ubiquitination
18.
Mol Cell ; 80(5): 876-891.e6, 2020 12 03.
Article in English | MEDLINE | ID: mdl-33217318

ABSTRACT

Stress granules (SGs) are cytoplasmic assemblies of proteins and non-translating mRNAs. Whereas much has been learned about SG formation, a major gap remains in understanding the compositional changes SGs undergo during normal disassembly and under disease conditions. Here, we address this gap by proteomic dissection of the SG temporal disassembly sequence using multi-bait APEX proximity proteomics. We discover 109 novel SG proteins and characterize distinct SG substructures. We reveal dozens of disassembly-engaged proteins (DEPs), some of which play functional roles in SG disassembly, including small ubiquitin-like modifier (SUMO) conjugating enzymes. We further demonstrate that SUMOylation regulates SG disassembly and SG formation. Parallel proteomics with amyotrophic lateral sclerosis (ALS)-associated C9ORF72 dipeptides uncovered attenuated DEP recruitment during SG disassembly and impaired SUMOylation. Accordingly, SUMO activity ameliorated C9ORF72-ALS-related neurodegeneration in Drosophila. By dissecting the SG spatiotemporal proteomic landscape, we provide an in-depth resource for future work on SG function and reveal basic and disease-relevant mechanisms of SG disassembly.


Subject(s)
Amyotrophic Lateral Sclerosis/metabolism , C9orf72 Protein/metabolism , Cytoplasmic Granules/metabolism , Drosophila Proteins/metabolism , Small Ubiquitin-Related Modifier Proteins/metabolism , Sumoylation , Amyotrophic Lateral Sclerosis/genetics , Amyotrophic Lateral Sclerosis/pathology , Animals , C9orf72 Protein/genetics , Cell Line, Tumor , Cytoplasmic Granules/genetics , Cytoplasmic Granules/pathology , Dipeptides/genetics , Dipeptides/metabolism , Drosophila Proteins/genetics , Drosophila melanogaster , Humans , Mice , Proteomics , Small Ubiquitin-Related Modifier Proteins/genetics
19.
Mol Cell ; 77(1): 82-94.e4, 2020 01 02.
Article in English | MEDLINE | ID: mdl-31630970

ABSTRACT

FUS is a nuclear RNA-binding protein, and its cytoplasmic aggregation is a pathogenic signature of amyotrophic lateral sclerosis (ALS) and frontotemporal dementia (FTD). It remains unknown how the FUS-RNA interactions contribute to phase separation and whether its phase behavior is affected by ALS-linked mutations. Here we demonstrate that wild-type FUS binds single-stranded RNA stoichiometrically in a length-dependent manner and that multimers induce highly dynamic interactions with RNA, giving rise to small and fluid condensates. In contrast, mutations in arginine display a severely altered conformation, static binding to RNA, and formation of large condensates, signifying the role of arginine in driving proper RNA interaction. Glycine mutations undergo rapid loss of fluidity, emphasizing the role of glycine in promoting fluidity. Strikingly, the nuclear import receptor Karyopherin-ß2 reverses the mutant defects and recovers the wild-type FUS behavior. We reveal two distinct mechanisms underpinning potentially disparate pathogenic pathways of ALS-linked FUS mutants.


Subject(s)
Amyotrophic Lateral Sclerosis/genetics , Frontotemporal Dementia/genetics , Mutation/genetics , RNA-Binding Protein FUS/genetics , RNA/genetics , Active Transport, Cell Nucleus/genetics , Glycine/genetics , Humans
20.
Genes Dev ; 34(11-12): 785-805, 2020 06 01.
Article in English | MEDLINE | ID: mdl-32381627

ABSTRACT

Dysregulation of the DNA/RNA-binding protein FUS causes certain subtypes of ALS/FTD by largely unknown mechanisms. Recent evidence has shown that FUS toxic gain of function due either to mutations or to increased expression can disrupt critical cellular processes, including mitochondrial functions. Here, we demonstrate that in human cells overexpressing wild-type FUS or expressing mutant derivatives, the protein associates with multiple mRNAs, and these are enriched in mRNAs encoding mitochondrial respiratory chain components. Notably, this sequestration leads to reduced levels of the encoded proteins, which is sufficient to bring about disorganized mitochondrial networks, reduced aerobic respiration and increased reactive oxygen species. We further show that mutant FUS associates with mitochondria and with mRNAs encoded by the mitochondrial genome. Importantly, similar results were also observed in fibroblasts derived from ALS patients with FUS mutations. Finally, we demonstrate that FUS loss of function does not underlie the observed mitochondrial dysfunction, and also provides a mechanism for the preferential sequestration of the respiratory chain complex mRNAs by FUS that does not involve sequence-specific binding. Together, our data reveal that respiratory chain complex mRNA sequestration underlies the mitochondrial defects characteristic of ALS/FTD and contributes to the FUS toxic gain of function linked to this disease spectrum.


Subject(s)
Amyotrophic Lateral Sclerosis/genetics , Amyotrophic Lateral Sclerosis/physiopathology , Gene Expression Regulation/genetics , Mitochondria/pathology , RNA, Messenger/metabolism , RNA-Binding Protein FUS/genetics , RNA-Binding Protein FUS/metabolism , Cell Line , Cell Respiration/genetics , Cells, Cultured , Electron Transport/genetics , Genome, Mitochondrial , Humans , Mitochondria/genetics , Mutation , Protein Aggregation, Pathological/genetics , Protein Binding/genetics
SELECTION OF CITATIONS
SEARCH DETAIL