Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 36
Filter
Add more filters










Publication year range
2.
J Mol Biol ; 436(14): 168615, 2024 Jul 15.
Article in English | MEDLINE | ID: mdl-38759929

ABSTRACT

Eukaryotic cells are equipped with an intricate proteostasis network (PN), comprising nearly 3,000 components dedicated to preserving proteome integrity and sustaining protein homeostasis. This protective system is particularly important under conditions of external and intrinsic cell stress, where inherently dynamic proteins may unfold and lose functionality. A decline in proteostasis capacity is associated with the aging process, resulting in a reduced folding efficiency of newly synthesized proteins and a deficit in the cellular capacity to degrade misfolded proteins. A critical consequence of PN insufficiency is the accumulation of cytotoxic protein aggregates that underlie various age-related neurodegenerative conditions and other pathologies. By interfering with specific proteostasis components, toxic aggregates place an excessive burden on the PN's ability to maintain proteome integrity. This initiates a feed-forward loop, wherein the generation of misfolded and aggregated proteins ultimately leads to proteostasis collapse and cellular demise.


Subject(s)
Protein Aggregates , Proteostasis , Humans , Animals , Protein Folding , Protein Aggregation, Pathological/metabolism , Neurodegenerative Diseases/metabolism , Neurodegenerative Diseases/pathology , Proteome/metabolism , Aging/metabolism , Proteostasis Deficiencies/metabolism , Proteostasis Deficiencies/pathology
3.
Nat Commun ; 14(1): 560, 2023 02 02.
Article in English | MEDLINE | ID: mdl-36732333

ABSTRACT

Amyloid-like aggregates of the microtubule-associated protein Tau are associated with several neurodegenerative disorders including Alzheimer's disease. The existence of cellular machinery for the removal of such aggregates has remained unclear, as specialized disaggregase chaperones are thought to be absent in mammalian cells. Here we show in cell culture and in neurons that the hexameric ATPase valosin-containing protein (VCP) is recruited to ubiquitylated Tau fibrils, resulting in their efficient disaggregation. Aggregate clearance depends on the functional cooperation of VCP with heat shock 70 kDa protein (Hsp70) and the ubiquitin-proteasome machinery. While inhibition of VCP activity stabilizes large Tau aggregates, disaggregation by VCP generates seeding-active Tau species as byproduct. These findings identify VCP as a core component of the machinery for the removal of neurodegenerative disease aggregates and suggest that its activity can be associated with enhanced aggregate spreading in tauopathies.


Subject(s)
Alzheimer Disease , Neurodegenerative Diseases , Animals , Humans , Valosin Containing Protein/genetics , Valosin Containing Protein/metabolism , Neurodegenerative Diseases/metabolism , Molecular Chaperones/metabolism , Heat-Shock Proteins/metabolism , tau Proteins/genetics , tau Proteins/metabolism , Mammals/metabolism
4.
Trends Biochem Sci ; 48(3): 216-228, 2023 03.
Article in English | MEDLINE | ID: mdl-36280494

ABSTRACT

Aggrephagy describes the selective lysosomal transport and turnover of cytoplasmic protein aggregates by macro-autophagy. In this process, protein aggregates and conglomerates are polyubiquitinated and then sequestered by autophagosomes. Soluble selective autophagy receptors (SARs) are central to aggrephagy and physically bind to both ubiquitin and the autophagy machinery, thus linking the cargo to the forming autophagosomal membrane. Because the accumulation of protein aggregates is associated with cytotoxicity in several diseases, a better molecular understanding of aggrephagy might provide a conceptual framework to develop therapeutic strategies aimed at delaying the onset of these pathologies by preventing the buildup of potentially toxic aggregates. We review recent advances in our knowledge about the mechanism of aggrephagy.


Subject(s)
Autophagy , Protein Aggregates , Sequestosome-1 Protein/metabolism , Autophagosomes , Lysosomes/metabolism
5.
Front Cell Neurosci ; 16: 797588, 2022.
Article in English | MEDLINE | ID: mdl-35496908

ABSTRACT

During adult neurogenesis, neuronal stem cells differentiate into mature neurons that are functionally integrated into the existing network. One hallmark during the late phase of this neurodifferentiation process is the formation of dendritic spines. These morphological specialized structures form the basis of most excitatory synapses in the brain, and are essential for neuronal communication. Additionally, dendritic spines are affected in neurological disorders, such as Alzheimer's disease or schizophrenia. However, the mechanisms underlying spinogenesis, as well as spine pathologies, are poorly understood. Plasticity-related Gene 5 (PRG5), a neuronal transmembrane protein, has previously been linked to spinogenesis in vitro. Here, we analyze endogenous expression of the PRG5 protein in different mouse brain areas, as well as on a subcellular level. We found that native PRG5 is expressed dendritically, and in high abundance in areas characterized by their regenerative capacity, such as the hippocampus and the olfactory bulb. During adult neurogenesis, PRG5 is specifically expressed in a late phase after neuronal cell-fate determination associated with dendritic spine formation. On a subcellular level, we found PRG5 not to be localized at the postsynaptic density, but at the base of the synapse. In addition, we showed that PRG5-induced formation of membrane protrusions is independent from neuronal activity, supporting a possible role in the morphology and stabilization of spines.

6.
EMBO Rep ; 23(6): e53890, 2022 06 07.
Article in English | MEDLINE | ID: mdl-35438230

ABSTRACT

Aggregation of the multifunctional RNA-binding protein TDP-43 defines large subgroups of amyotrophic lateral sclerosis and frontotemporal dementia and correlates with neurodegeneration in both diseases. In disease, characteristic C-terminal fragments of ~25 kDa ("TDP-25") accumulate in cytoplasmic inclusions. Here, we analyze gain-of-function mechanisms of TDP-25 combining cryo-electron tomography, proteomics, and functional assays. In neurons, cytoplasmic TDP-25 inclusions are amorphous, and photobleaching experiments reveal gel-like biophysical properties that are less dynamic than nuclear TDP-43. Compared with full-length TDP-43, the TDP-25 interactome is depleted of low-complexity domain proteins. TDP-25 inclusions are enriched in 26S proteasomes adopting exclusively substrate-processing conformations, suggesting that inclusions sequester proteasomes, which are largely stalled and no longer undergo the cyclic conformational changes required for proteolytic activity. Reporter assays confirm that TDP-25 impairs proteostasis, and this inhibitory function is enhanced by ALS-causing TDP-43 mutations. These findings support a patho-physiological relevance of proteasome dysfunction in ALS/FTD.


Subject(s)
Amyotrophic Lateral Sclerosis , DNA-Binding Proteins , Frontotemporal Dementia , Neurons , Peptide Fragments , Amyotrophic Lateral Sclerosis/genetics , Amyotrophic Lateral Sclerosis/metabolism , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Frontotemporal Dementia/genetics , Frontotemporal Dementia/metabolism , Humans , Inclusion Bodies/metabolism , Neurons/metabolism , Peptide Fragments/genetics , Peptide Fragments/metabolism , Proteasome Endopeptidase Complex/metabolism
7.
Dev Dyn ; 251(4): 714-728, 2022 04.
Article in English | MEDLINE | ID: mdl-34622503

ABSTRACT

BACKGROUND: Plasticity-related genes (Prgs/PRGs) or lipid phosphate phosphatase-related proteins (LPPRs) comprise five known members, which have been linked to neuronal differentiation processes, such as neurite outgrowth, axonal branching, or dendritic spine formation. PRGs are highly brain-specific and belong to the lipid phosphate phosphatases (LPPs) superfamily, which influence lipid metabolism by dephosphorylation of bioactive lipids. PRGs, however, do not possess enzymatic activity, but modify lipid metabolism in a way that is still under investigation. RESULTS: We analyzed mRNA expression levels of all Prgs during mouse brain development, in the hippocampus, neocortex, olfactory bulbs, and cerebellum. We found different spatio-temporal expression patterns for each of the Prgs, and identified a high expression of the uncharacterized Prg4 throughout brain development. Unlike its close family members PRG3 and PRG5, PRG4 did not induce filopodial outgrowth in non-neuronal cell lines, and does not localize to the plasma membrane of filopodia. CONCLUSION: We showed PRG4 to be highly expressed in the developing and the adult brain, suggesting that it is of vital importance for normal brain function. Despite its similarities to other family members, it seems not to be involved in changes of cell morphology; instead, it is more likely to be associated with intracellular signaling.


Subject(s)
Brain , Phosphoric Monoester Hydrolases , Animals , Brain/metabolism , Cell Membrane/metabolism , Hippocampus/metabolism , Mice , Phosphoric Monoester Hydrolases/genetics , Phosphoric Monoester Hydrolases/metabolism , Proteoglycans/metabolism , Pseudopodia
8.
Nat Commun ; 12(1): 4863, 2021 08 11.
Article in English | MEDLINE | ID: mdl-34381050

ABSTRACT

Spreading of aggregate pathology across brain regions acts as a driver of disease progression in Tau-related neurodegeneration, including Alzheimer's disease (AD) and frontotemporal dementia. Aggregate seeds released from affected cells are internalized by naïve cells and induce the prion-like templating of soluble Tau into neurotoxic aggregates. Here we show in a cellular model system and in neurons that Clusterin, an abundant extracellular chaperone, strongly enhances Tau aggregate seeding. Upon interaction with Tau aggregates, Clusterin stabilizes highly potent, soluble seed species. Tau/Clusterin complexes enter recipient cells via endocytosis and compromise the endolysosomal compartment, allowing transfer to the cytosol where they propagate aggregation of endogenous Tau. Thus, upregulation of Clusterin, as observed in AD patients, may enhance Tau seeding and possibly accelerate the spreading of Tau pathology.


Subject(s)
Clusterin/metabolism , Protein Aggregation, Pathological/metabolism , tau Proteins/metabolism , Animals , Clusterin/genetics , Disease Progression , Endocytosis , Humans , Mice , Neurodegenerative Diseases/metabolism , Neurodegenerative Diseases/pathology , Neurons/metabolism , Neurons/pathology , Protein Aggregation, Pathological/pathology , Protein Binding , alpha-Synuclein/metabolism , tau Proteins/genetics
9.
EMBO J ; 40(19): e107260, 2021 10 01.
Article in English | MEDLINE | ID: mdl-34410010

ABSTRACT

The cellular protein quality control machinery is important for preventing protein misfolding and aggregation. Declining protein homeostasis (proteostasis) is believed to play a crucial role in age-related neurodegenerative disorders. However, how neuronal proteostasis capacity changes in different diseases is not yet sufficiently understood, and progress in this area has been hampered by the lack of tools to monitor proteostasis in mammalian models. Here, we have developed reporter mice for in vivo analysis of neuronal proteostasis. The mice express EGFP-fused firefly luciferase (Fluc-EGFP), a conformationally unstable protein that requires chaperones for proper folding, and that reacts to proteotoxic stress by formation of intracellular Fluc-EGFP foci and by reduced luciferase activity. Using these mice, we provide evidence for proteostasis decline in the aging brain. Moreover, we find a marked reaction of the Fluc-EGFP sensor in a mouse model of tauopathy, but not in mouse models of Huntington's disease. Mechanistic investigations in primary neuronal cultures demonstrate that different types of protein aggregates have distinct effects on the cellular protein quality control. Thus, Fluc-EGFP reporter mice enable new insights into proteostasis alterations in different diseases.


Subject(s)
Aging/metabolism , Disease Susceptibility , Genes, Reporter , Mice, Transgenic , Neurons/metabolism , Proteostasis , Aging/genetics , Animals , Cells, Cultured , Disease Models, Animal , Gene Expression , Hippocampus/metabolism , Hippocampus/pathology , Huntington Disease/etiology , Huntington Disease/metabolism , Huntington Disease/pathology , Mice , Neurodegenerative Diseases/etiology , Neurodegenerative Diseases/metabolism , Neurodegenerative Diseases/pathology , Protein Aggregates , Protein Aggregation, Pathological , Protein Folding , Proteostasis Deficiencies/etiology , Proteostasis Deficiencies/metabolism , Proteostasis Deficiencies/pathology , Tauopathies/etiology , Tauopathies/metabolism , Tauopathies/pathology
10.
Elife ; 102021 06 23.
Article in English | MEDLINE | ID: mdl-34161229

ABSTRACT

The most frequent genetic cause of amyotrophic lateral sclerosis and frontotemporal dementia is a G4C2 repeat expansion in the C9orf72 gene. This expansion gives rise to translation of aggregating dipeptide repeat (DPR) proteins, including poly-GA as the most abundant species. However, gain of toxic function effects have been attributed to either the DPRs or the pathological G4C2 RNA. Here, we analyzed in a cellular model the relative toxicity of DPRs and RNA. Cytoplasmic poly-GA aggregates, generated in the absence of G4C2 RNA, interfered with nucleocytoplasmic protein transport, but had little effect on cell viability. In contrast, nuclear poly-GA was more toxic, impairing nucleolar protein quality control and protein biosynthesis. Production of the G4C2 RNA strongly reduced viability independent of DPR translation and caused pronounced inhibition of nuclear mRNA export and protein biogenesis. Thus, while the toxic effects of G4C2 RNA predominate in the cellular model used, DPRs exert additive effects that may contribute to pathology.


Subject(s)
C9orf72 Protein/toxicity , Dipeptides/toxicity , RNA Transport , RNA/metabolism , Humans
11.
Nat Commun ; 12(1): 2110, 2021 04 14.
Article in English | MEDLINE | ID: mdl-33854052

ABSTRACT

The molecular architecture of α-Synuclein (α-Syn) inclusions, pathognomonic of various neurodegenerative disorders, remains unclear. α-Syn inclusions were long thought to consist mainly of α-Syn fibrils, but recent reports pointed to intracellular membranes as the major inclusion component. Here, we use cryo-electron tomography (cryo-ET) to image neuronal α-Syn inclusions in situ at molecular resolution. We show that inclusions seeded by α-Syn aggregates produced recombinantly or purified from patient brain consist of α-Syn fibrils crisscrossing a variety of cellular organelles. Using gold-labeled seeds, we find that aggregate seeding is predominantly mediated by small α-Syn fibrils, from which cytoplasmic fibrils grow unidirectionally. Detailed analysis of membrane interactions revealed that α-Syn fibrils do not contact membranes directly, and that α-Syn does not drive membrane clustering. Altogether, we conclusively demonstrate that neuronal α-Syn inclusions consist of α-Syn fibrils intermixed with membranous organelles, and illuminate the mechanism of aggregate seeding and cellular interaction.


Subject(s)
Inclusion Bodies/metabolism , Multiple System Atrophy/metabolism , Neurons/metabolism , alpha-Synuclein/chemistry , alpha-Synuclein/metabolism , Brain/metabolism , Cryoelectron Microscopy , Humans , Inclusion Bodies/chemistry , Multiple System Atrophy/genetics , Neurons/chemistry , alpha-Synuclein/genetics
12.
Nat Commun ; 11(1): 6271, 2020 12 08.
Article in English | MEDLINE | ID: mdl-33293525

ABSTRACT

Cells adapt to conditions that compromise protein conformational stability by activating various stress response pathways, but the mechanisms used in sensing misfolded proteins remain unclear. Moreover, aggregates of disease proteins often fail to induce a productive stress response. Here, using a yeast model of polyQ protein aggregation, we identified Sis1, an essential Hsp40 co-chaperone of Hsp70, as a critical sensor of proteotoxic stress. At elevated levels, Sis1 prevented the formation of dense polyQ inclusions and directed soluble polyQ oligomers towards the formation of permeable condensates. Hsp70 accumulated in a liquid-like state within this polyQ meshwork, resulting in a potent activation of the HSF1 dependent stress response. Sis1, and the homologous DnaJB6 in mammalian cells, also regulated the magnitude of the cellular heat stress response, suggesting a general role in sensing protein misfolding. Sis1/DnaJB6 functions as a limiting regulator to enable a dynamic stress response and avoid hypersensitivity to environmental changes.


Subject(s)
HSP40 Heat-Shock Proteins/metabolism , Heat-Shock Response , Molecular Chaperones/metabolism , Nerve Tissue Proteins/metabolism , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae/physiology , DNA-Binding Proteins/metabolism , Gene Knockout Techniques , HEK293 Cells , HSP40 Heat-Shock Proteins/genetics , Heat-Shock Proteins/metabolism , Humans , Molecular Chaperones/genetics , Nerve Tissue Proteins/genetics , Peptides/metabolism , Protein Aggregates , Protein Folding , Saccharomyces cerevisiae Proteins/genetics , Transcription Factors/metabolism
13.
EMBO J ; 39(8): e102811, 2020 04 15.
Article in English | MEDLINE | ID: mdl-32175624

ABSTRACT

The C9orf72 repeat expansion causes amyotrophic lateral sclerosis and frontotemporal dementia, but the poor correlation between C9orf72-specific pathology and TDP-43 pathology linked to neurodegeneration hinders targeted therapeutic development. Here, we addressed the role of the aggregating dipeptide repeat proteins resulting from unconventional translation of the repeat in all reading frames. Poly-GA promoted cytoplasmic mislocalization and aggregation of TDP-43 non-cell-autonomously, and anti-GA antibodies ameliorated TDP-43 mislocalization in both donor and receiver cells. Cell-to-cell transmission of poly-GA inhibited proteasome function in neighboring cells. Importantly, proteasome inhibition led to the accumulation of TDP-43 ubiquitinated within the nuclear localization signal (NLS) at lysine 95. Mutagenesis of this ubiquitination site completely blocked poly-GA-dependent mislocalization of TDP-43. Boosting proteasome function with rolipram reduced both poly-GA and TDP-43 aggregation. Our data from cell lines, primary neurons, transgenic mice, and patient tissue suggest that poly-GA promotes TDP-43 aggregation by inhibiting the proteasome cell-autonomously and non-cell-autonomously, which can be prevented by inhibiting poly-GA transmission with antibodies or boosting proteasome activity with rolipram.


Subject(s)
Amyotrophic Lateral Sclerosis/pathology , C9orf72 Protein/metabolism , DNA-Binding Proteins/metabolism , Dipeptides/metabolism , Frontotemporal Dementia/pathology , Active Transport, Cell Nucleus , Amyotrophic Lateral Sclerosis/metabolism , Animals , C9orf72 Protein/genetics , Cytoplasm/metabolism , DNA-Binding Proteins/genetics , Female , Frontotemporal Dementia/metabolism , HeLa Cells , Humans , Male , Mice , Neurons/metabolism , Nuclear Localization Signals , Proteasome Endopeptidase Complex/metabolism , Protein Aggregation, Pathological , Ubiquitin/metabolism
14.
Proc Natl Acad Sci U S A ; 117(8): 4099-4108, 2020 02 25.
Article in English | MEDLINE | ID: mdl-32047030

ABSTRACT

Mammalian cells present a fingerprint of their proteome to the adaptive immune system through the display of endogenous peptides on MHC-I complexes. MHC-I-bound peptides originate from protein degradation by the proteasome, suggesting that stably folded, long-lived proteins could evade monitoring. Here, we investigate the role in antigen presentation of the ribosome-associated quality control (RQC) pathway for the degradation of nascent polypeptides that are encoded by defective messenger RNAs and undergo stalling at the ribosome during translation. We find that degradation of model proteins by RQC results in efficient MHC-I presentation, independent of their intrinsic folding properties. Quantitative profiling of MHC-I peptides in wild-type and RQC-deficient cells by mass spectrometry showed that RQC substantially contributes to the composition of the immunopeptidome. Our results also identify endogenous substrates of the RQC pathway in human cells and provide insight into common principles causing ribosome stalling under physiological conditions.


Subject(s)
Antigen Presentation/physiology , Epitopes/metabolism , Histocompatibility Antigens Class I/physiology , Ribosomes/physiology , Animals , Gene Deletion , Gene Expression Regulation , HeLa Cells , Humans , RNA, Messenger/genetics , RNA, Messenger/metabolism , Ubiquitin-Protein Ligases/metabolism
15.
Cell Stress Chaperones ; 25(3): 407-416, 2020 05.
Article in English | MEDLINE | ID: mdl-32060690

ABSTRACT

The HSP60/HSP10 chaperonin assists folding of proteins in the mitochondrial matrix space by enclosing them in its central cavity. The chaperonin forms part of the mitochondrial protein quality control system. It is essential for cellular survival and mutations in its subunits are associated with rare neurological disorders. Here we present the first survey of interactors of the human mitochondrial HSP60/HSP10 chaperonin. Using a protocol involving metabolic labeling of HEK293 cells, cross-linking, and immunoprecipitation of HSP60, we identified 323 interacting proteins. As expected, the vast majority of these proteins are localized to the mitochondrial matrix space. We find that approximately half of the proteins annotated as mitochondrial matrix proteins interact with the HSP60/HSP10 chaperonin. They cover a broad spectrum of functions and metabolic pathways including the mitochondrial protein synthesis apparatus, the respiratory chain, and mitochondrial protein quality control. Many of the genes encoding HSP60 interactors are annotated as disease genes. There is a correlation between relative cellular abundance and relative abundance in the HSP60 immunoprecipitates. Nineteen abundant matrix proteins occupy more than 60% of the HSP60/HSP10 chaperonin capacity. The reported inventory of interactors can form the basis for interrogating which proteins are especially dependent on the chaperonin.


Subject(s)
Chaperonin 10/metabolism , Chaperonin 60/metabolism , Mitochondrial Proteins/metabolism , HEK293 Cells , Humans , Mitochondria/metabolism
16.
Article in English | MEDLINE | ID: mdl-30833457

ABSTRACT

Cells invest in an extensive network of factors to maintain protein homeostasis (proteostasis) and prevent the accumulation of potentially toxic protein aggregates. This proteostasis network (PN) comprises the machineries for the biogenesis, folding, conformational maintenance, and degradation of proteins with molecular chaperones as central coordinators. Here, we review recent progress in understanding the modular architecture of the PN in mammalian cells and how it is modified during cell differentiation. We discuss the capacity and limitations of the PN in maintaining proteome integrity in the face of proteotoxic stresses, such as aggregate formation in neurodegenerative diseases. Finally, we outline various pharmacological interventions to ameliorate proteostasis imbalance.


Subject(s)
Molecular Chaperones/chemistry , Neurodegenerative Diseases/metabolism , Proteins/chemistry , Proteostasis/physiology , Animals , Cell Differentiation , Homeostasis , Humans , Protein Conformation , Protein Denaturation , Protein Folding , Proteome/metabolism , Thermodynamics
17.
EMBO J ; 38(9)2019 05 02.
Article in English | MEDLINE | ID: mdl-30886048

ABSTRACT

Neurodegenerative diseases are characterized by the accumulation of misfolded proteins in the brain. Insights into protein quality control mechanisms to prevent neuronal dysfunction and cell death are crucial in developing causal therapies. Here, we report that various disease-associated protein aggregates are modified by the linear ubiquitin chain assembly complex (LUBAC). HOIP, the catalytic component of LUBAC, is recruited to misfolded Huntingtin in a p97/VCP-dependent manner, resulting in the assembly of linear polyubiquitin. As a consequence, the interactive surface of misfolded Huntingtin species is shielded from unwanted interactions, for example with the low complexity sequence domain-containing transcription factor Sp1, and proteasomal degradation of misfolded Huntingtin is facilitated. Notably, all three core LUBAC components are transcriptionally regulated by Sp1, linking defective LUBAC expression to Huntington's disease. In support of a protective activity of linear ubiquitination, silencing of OTULIN, a deubiquitinase with unique specificity for linear polyubiquitin, decreases proteotoxicity, whereas silencing of HOIP has the opposite effect. These findings identify linear ubiquitination as a protein quality control mechanism and hence a novel target for disease-modifying strategies in proteinopathies.


Subject(s)
Huntingtin Protein/metabolism , Huntington Disease/metabolism , Polyubiquitin/metabolism , Protein Processing, Post-Translational , Sp1 Transcription Factor/metabolism , Valosin Containing Protein/metabolism , Adult , Aged , Animals , Brain/metabolism , Brain/pathology , Case-Control Studies , Cells, Cultured , Embryo, Mammalian/cytology , Embryo, Mammalian/metabolism , Female , Fibroblasts/cytology , Fibroblasts/metabolism , Humans , Huntingtin Protein/genetics , Huntington Disease/genetics , Huntington Disease/pathology , Male , Mice , Mice, Knockout , Middle Aged , NF-kappa B/genetics , NF-kappa B/metabolism , Neurons/metabolism , Neurons/pathology , Protein Binding , Protein Interaction Domains and Motifs , Signal Transduction , Sp1 Transcription Factor/genetics , Ubiquitination , Valosin Containing Protein/genetics
18.
Nat Rev Mol Cell Biol ; 20(7): 421-435, 2019 07.
Article in English | MEDLINE | ID: mdl-30733602

ABSTRACT

Ageing is a major risk factor for the development of many diseases, prominently including neurodegenerative disorders such as Alzheimer disease and Parkinson disease. A hallmark of many age-related diseases is the dysfunction in protein homeostasis (proteostasis), leading to the accumulation of protein aggregates. In healthy cells, a complex proteostasis network, comprising molecular chaperones and proteolytic machineries and their regulators, operates to ensure the maintenance of proteostasis. These factors coordinate protein synthesis with polypeptide folding, the conservation of protein conformation and protein degradation. However, sustaining proteome balance is a challenging task in the face of various external and endogenous stresses that accumulate during ageing. These stresses lead to the decline of proteostasis network capacity and proteome integrity. The resulting accumulation of misfolded and aggregated proteins affects, in particular, postmitotic cell types such as neurons, manifesting in disease. Recent analyses of proteome-wide changes that occur during ageing inform strategies to improve proteostasis. The possibilities of pharmacological augmentation of the capacity of proteostasis networks hold great promise for delaying the onset of age-related pathologies associated with proteome deterioration and for extending healthspan.


Subject(s)
Alzheimer Disease/metabolism , Molecular Chaperones/metabolism , Parkinson Disease/metabolism , Protein Folding , Proteolysis , Proteostasis , Alzheimer Disease/genetics , Alzheimer Disease/pathology , Animals , Humans , Molecular Chaperones/genetics , Parkinson Disease/genetics , Parkinson Disease/pathology
19.
Proc Natl Acad Sci U S A ; 115(15): E3446-E3453, 2018 04 10.
Article in English | MEDLINE | ID: mdl-29581260

ABSTRACT

Huntington's disease is caused by the expansion of a polyglutamine (polyQ) tract in the N-terminal exon of huntingtin (HttEx1), but the cellular mechanisms leading to neurodegeneration remain poorly understood. Here we present in situ structural studies by cryo-electron tomography of an established yeast model system of polyQ toxicity. We find that expression of polyQ-expanded HttEx1 results in the formation of unstructured inclusion bodies and in some cases fibrillar aggregates. This contrasts with recent findings in mammalian cells, where polyQ inclusions were exclusively fibrillar. In yeast, polyQ toxicity correlates with alterations in mitochondrial and lipid droplet morphology, which do not arise from physical interactions with inclusions or fibrils. Quantitative proteomic analysis shows that polyQ aggregates sequester numerous cellular proteins and cause a major change in proteome composition, most significantly in proteins related to energy metabolism. Thus, our data point to a multifaceted toxic gain-of-function of polyQ aggregates, driven by sequestration of endogenous proteins and mitochondrial and lipid droplet dysfunction.


Subject(s)
Peptides/metabolism , Saccharomyces cerevisiae/metabolism , Humans , Huntington Disease/genetics , Huntington Disease/metabolism , Inclusion Bodies/chemistry , Inclusion Bodies/genetics , Inclusion Bodies/metabolism , Lipid Droplets/chemistry , Lipid Droplets/metabolism , Mitochondria/chemistry , Mitochondria/metabolism , Peptides/chemistry , Peptides/toxicity , Proteomics , Saccharomyces cerevisiae/chemistry , Saccharomyces cerevisiae/drug effects , Saccharomyces cerevisiae Proteins/chemistry , Saccharomyces cerevisiae Proteins/genetics , Saccharomyces cerevisiae Proteins/metabolism
20.
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
SELECTION OF CITATIONS
SEARCH DETAIL