Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 39
Filter
Add more filters










Publication year range
1.
Elife ; 132024 Jul 03.
Article in English | MEDLINE | ID: mdl-38899618

ABSTRACT

The autophagy-lysosome pathway plays an indispensable role in the protein quality control by degrading abnormal organelles and proteins including α-synuclein (αSyn) associated with the pathogenesis of Parkinson's disease (PD). However, the activation of this pathway is mainly by targeting lysosomal enzymic activity. Here, we focused on the autophagosome-lysosome fusion process around the microtubule-organizing center (MTOC) regulated by lysosomal positioning. Through high-throughput chemical screening, we identified 6 out of 1200 clinically approved drugs enabling the lysosomes to accumulate around the MTOC with autophagy flux enhancement. We further demonstrated that these compounds induce the lysosomal clustering through a JIP4-TRPML1-dependent mechanism. Among them, the lysosomal-clustering compound albendazole promoted the autophagy-dependent degradation of Triton-X-insoluble, proteasome inhibitor-induced aggregates. In a cellular PD model, albendazole boosted insoluble αSyn degradation. Our results revealed that lysosomal clustering can facilitate the breakdown of protein aggregates, suggesting that lysosome-clustering compounds may offer a promising therapeutic strategy against neurodegenerative diseases characterized by the presence of aggregate-prone proteins.


Subject(s)
Autophagy , Lysosomes , Parkinson Disease , Lysosomes/drug effects , Lysosomes/metabolism , Parkinson Disease/metabolism , Parkinson Disease/drug therapy , Parkinson Disease/pathology , Autophagy/drug effects , Humans , alpha-Synuclein/metabolism , Albendazole/pharmacology , Microtubule-Organizing Center/metabolism , Autophagosomes/metabolism , Autophagosomes/drug effects
2.
Development ; 151(4)2024 Feb 15.
Article in English | MEDLINE | ID: mdl-38265146

ABSTRACT

Lysosomes are intracellular organelles responsible for degrading diverse macromolecules delivered from several pathways, including the endo-lysosomal and autophagic pathways. Recent reports have suggested that lysosomes are essential for regulating neural stem cells in developing, adult and aged brains. However, the activity of these lysosomes has yet to be monitored in these brain tissues. Here, we report the development of a new probe to measure lysosomal protein degradation in brain tissue by immunostaining. Our results indicate that lysosomal protein degradation fluctuates in neural stem cells of the hippocampal dentate gyrus, depending on age and brain disorders. Neural stem cells increase their lysosomal activity during hippocampal development in the dentate gyrus, but aging and aging-related disease reduce lysosomal activity. In addition, physical exercise increases lysosomal activity in neural stem cells and astrocytes in the dentate gyrus. We therefore propose that three different stages of lysosomal activity exist: the state of increase during development, the stable state during adulthood and the state of reduction due to damage caused by either age or disease.


Subject(s)
Dentate Gyrus , Neural Stem Cells , Animals , Mice , Dentate Gyrus/metabolism , Proteolysis , Neural Stem Cells/metabolism , Astrocytes/metabolism , Lysosomes/metabolism
3.
Biochim Biophys Acta Gene Regul Mech ; 1866(2): 194932, 2023 06.
Article in English | MEDLINE | ID: mdl-36997115

ABSTRACT

The spatiotemporal sequestration of misfolded proteins is a mechanism by which cells counterbalance proteome homeostasis upon exposure to various stress stimuli. Chronic inhibition of proteasomes results in a large, juxtanuclear, membrane-less inclusion, known as the aggresome. Although the molecular mechanisms driving its formation, clearance, and pathophysiological implications are continuously being uncovered, the biophysical aspects of aggresomes remain largely uncharacterized. Using fluorescence recovery after photobleaching and liquid droplet disruption assays, we found that the aggresomes are a homogeneously blended condensates with liquid-like properties similar to droplets formed via liquid-liquid phase separation. However, unlike fluidic liquid droplets, aggresomes have more viscosity and hydrogel-like characteristics. We also observed that the inhibition of aggresome formation using microtubule-disrupting agents resulted in less soluble and smaller cytoplasmic speckles, which was associated with marked cytotoxicity. Therefore, the aggresome appears to be cytoprotective and serves as a temporal reservoir for dysfunctional proteasomes and substrates that need to be degraded. Our results suggest that the aggresome assembles through distinct and potentially sequential processes of energy-dependent retrograde transportation and spontaneous condensation into a hydrogel.


Subject(s)
Hydrogels , Proteasome Endopeptidase Complex , Proteasome Endopeptidase Complex/metabolism , Hydrogels/metabolism , Proteins/metabolism , Inclusion Bodies/metabolism , Microtubules/metabolism
4.
Sci Rep ; 13(1): 4680, 2023 03 28.
Article in English | MEDLINE | ID: mdl-36977730

ABSTRACT

Proteostasis regulates protein folding and degradation; its maintenance is essential for resistance to stress and aging. The loss of proteostasis is associated with many age-related diseases. Within the cell, molecular chaperones facilitate the refolding of misfolded proteins into their bioactive forms, thus preventing undesirable interactions and aggregation. Although the mechanisms of intracellular protein degradation pathways for intracellular misfolded proteins have been extensively studied, the protein degradation pathway for extracellular proteins remain poorly understood. In this study, we identified several misfolded proteins that are substrates for alpha 2-macroglobulin (α2M), an extracellular chaperone. We also established a lysosomal internalization assay for α2M, which revealed that α2M mediates the lysosomal degradation of extracellular misfolded proteins. Comparative analyses of α2M and clusterin, another extracellular chaperone, indicated that α2M preferentially targets aggregation-prone proteins. Thus, we present the degradation pathway of α2M, which interacts with aggregation-prone proteins for lysosomal degradation via selective internalization.


Subject(s)
Pregnancy-Associated alpha 2-Macroglobulins , Female , Pregnancy , Humans , Pregnancy-Associated alpha 2-Macroglobulins/metabolism , Protein Folding , Proteostasis , Proteolysis , Transcription Factors/metabolism , Lysosomes/metabolism
5.
Mol Biol Cell ; 34(4): ar29, 2023 04 01.
Article in English | MEDLINE | ID: mdl-36735498

ABSTRACT

The endoplasmic reticulum (ER) is a major cell compartment where protein synthesis, folding, and posttranslational modifications occur with assistance from a wide variety of chaperones and enzymes. Quality control systems selectively eliminate abnormal proteins that accumulate inside the ER due to cellular stresses. ER-phagy, that is, selective autophagy of the ER, is a mechanism that maintains or reestablishes cellular and ER-specific homeostasis through removal of abnormal proteins. However, how ER luminal proteins are recognized by the ER-phagy machinery remains unclear. Here, we applied the aggregation-prone protein, six-repeated islet amyloid polypeptide (6xIAPP), as a model ER-phagy substrate and found that cell cycle progression 1 (CCPG1), which is an ER-phagy receptor, efficiently mediates its degradation via ER-phagy. We also identified prolyl 3-hydroxylase family member 4 (P3H4) as an endogenous cargo of CCPG1-dependent ER-phagy. The ER luminal region of CCPG1 contains several highly conserved regions that we refer to as cargo-interacting regions (CIRs); these interact directly with specific luminal cargos for ER-phagy. Notably, 6xIAPP and P3H4 interact directly with different CIRs. These findings indicate that CCPG1 is a bispecific ER-phagy receptor for ER luminal proteins and the autophagosomal membrane that contributes to the efficient removal of aberrant ER-resident proteins through ER-phagy.


Subject(s)
Autophagy , Endoplasmic Reticulum Stress , Carrier Proteins/metabolism , Endoplasmic Reticulum/metabolism , Homeostasis , Proteins/metabolism , Cell Cycle Proteins/metabolism
6.
Cell Death Discov ; 8(1): 37, 2022 Jan 25.
Article in English | MEDLINE | ID: mdl-35079001

ABSTRACT

Autophagy plays important role in the intracellular protein quality control system by degrading abnormal organelles and proteins, including large protein complexes such as ribosomes. The eukaryotic chaperonin tailless complex polypeptide 1 (TCP1) ring complex (TRiC), also called chaperonin-containing TCP1 (CCT), is a 1-MDa hetero-oligomer complex comprising 16 subunits that facilitates the folding of ~10% of the cellular proteome that contains actin. However, the quality control mechanism of TRiC remains unclear. To monitor the autophagic degradation of TRiC, we generated TCP1α-RFP-GFP knock-in HeLa cells using a CRISPR/Cas9-knock-in system with an RFP-GFP donor vector. We analyzed the autophagic degradation of TRiC under several stress conditions and found that treatment with actin (de)polymerization inhibitors increased the lysosomal degradation of TRiC, which was localized in lysosomes and suppressed by deficiency of autophagy-related genes. Furthermore, we found that treatment with actin (de)polymerization inhibitors increased the association between TRiC and unfolded actin, suggesting that TRiC was inactivated. Moreover, unfolded actin mutants were degraded by autophagy. Taken together, our results indicate that autophagy eliminates inactivated TRiC, serving as a quality control system.

7.
STAR Protoc ; 2(4): 100975, 2021 12 17.
Article in English | MEDLINE | ID: mdl-34877548

ABSTRACT

Endocytic internalization of extracellular proteins plays roles in signaling, nutrient uptake, immunity, and extracellular protein quality control. However, there are few protocols for analyzing the lysosomal degradation of extracellular protein. Here, we purified secreted proteins fused with pH-sensitive GFP and acid- and protease-resistant RFP from mammalian cells and describe an internalization assay for mammalian cells. This protocol enables quantification of cellular uptake and lysosomal degradation of protein-of-interest (POI) via cell biological and biochemical analyses. For full details on the use and execution of this protocol, please refer to Itakura et al. (2020).


Subject(s)
Flow Cytometry/methods , Immunoblotting/methods , Lysosomes , Proteins , Antibodies, Monoclonal , Endocytosis/physiology , HEK293 Cells , Humans , Luminescent Proteins , Lysosomes/chemistry , Lysosomes/metabolism , Proteins/analysis , Proteins/chemistry , Proteins/metabolism , Recombinant Proteins
8.
J Biol Chem ; 297(5): 101279, 2021 11.
Article in English | MEDLINE | ID: mdl-34624312

ABSTRACT

Mitochondria are essential organelles that carry out a number of pivotal metabolic processes and maintain cellular homeostasis. Mitochondrial dysfunction caused by various stresses is associated with many diseases such as type 2 diabetes, obesity, cancer, heart failure, neurodegenerative disorders, and aging. Therefore, it is important to understand the stimuli that induce mitochondrial stress. However, broad analysis of mitochondrial stress has not been carried out to date. Here, we present a set of fluorescent tools, called mito-Pain (mitochondrial PINK1 accumulation index), which enable the labeling of stressed mitochondria. Mito-Pain uses PTEN-induced putative kinase 1 (PINK1) stabilization on mitochondria and quantifies mitochondrial stress levels by comparison with PINK1-GFP, which is stabilized under mitochondrial stress, and RFP-Omp25, which is constitutively localized on mitochondria. To identify compounds that induce mitochondrial stress, we screened a library of 3374 compounds using mito-Pain and identified 57 compounds as mitochondrial stress inducers. Furthermore, we classified each compound into several categories based on mitochondrial response: depolarization, mitochondrial morphology, or Parkin recruitment. Parkin recruitment to mitochondria was often associated with mitochondrial depolarization and aggregation, suggesting that Parkin is recruited to heavily damaged mitochondria. In addition, many of the compounds led to various mitochondrial morphological changes, including fragmentation, aggregation, elongation, and swelling, with or without Parkin recruitment or mitochondrial depolarization. We also found that several compounds induced an ectopic response of Parkin, leading to the formation of cytosolic puncta dependent on PINK1. Thus, mito-Pain enables the detection of stressed mitochondria under a wide variety of conditions and provides insights into mitochondrial quality control systems.


Subject(s)
Fluorescence , Fluorescent Dyes/chemistry , Mitochondria/enzymology , Mitochondrial Proteins/metabolism , Protein Kinases/metabolism , Small Molecule Libraries/chemistry , Stress, Physiological , Animals , COS Cells , Chlorocebus aethiops , Enzyme Stability , HEK293 Cells , HeLa Cells , Humans , Mitochondria/genetics , Mitochondrial Proteins/genetics , Protein Kinases/genetics
9.
J Cell Biol ; 219(3)2020 03 02.
Article in English | MEDLINE | ID: mdl-32211892

ABSTRACT

The accumulation of aberrant proteins leads to various neurodegenerative disorders. Mammalian cells contain several intracellular protein degradation systems, including autophagy and proteasomal systems, that selectively remove aberrant intracellular proteins. Although mammals contain not only intracellular but also extracellular proteins, the mechanism underlying the quality control of aberrant extracellular proteins is poorly understood. Here, using a novel quantitative fluorescence assay and genome-wide CRISPR screening, we identified the receptor-mediated degradation pathway by which misfolded extracellular proteins are selectively captured by the extracellular chaperone Clusterin and undergo endocytosis via the cell surface heparan sulfate (HS) receptor. Biochemical analyses revealed that positively charged residues on Clusterin electrostatically interact with negatively charged HS. Furthermore, the Clusterin-HS pathway facilitates the degradation of amyloid ß peptide and diverse leaked cytosolic proteins in extracellular space. Our results identify a novel protein quality control system for preserving extracellular proteostasis and highlight its role in preventing diseases associated with aberrant extracellular proteins.


Subject(s)
Amyloid beta-Peptides/metabolism , Cell Membrane/metabolism , Clusterin/metabolism , Endocytosis , Heparitin Sulfate/metabolism , Intrinsically Disordered Proteins/metabolism , A549 Cells , Amyloid beta-Peptides/chemistry , Clusterin/genetics , HCT116 Cells , HEK293 Cells , HeLa Cells , Humans , Intrinsically Disordered Proteins/chemistry , Lysosomes/metabolism , Protein Folding , Proteolysis , Proteostasis , Surface Properties , Time Factors
10.
FEBS Lett ; 594(10): 1586-1595, 2020 05.
Article in English | MEDLINE | ID: mdl-31997355

ABSTRACT

Autophagy is an intracellular process that regulates the degradation of cytosolic proteins and organelles. Dying cells often accumulate autophagosomes. However, the mechanisms by which necroptotic stimulation induces autophagosomes are not defined. Here, we demonstrate that the activation of necroptosis with TNF-α plus the cell-permeable pan-caspase inhibitor Z-VAD induces LC3-II and LC3 puncta, markers of autophagosomes, via the receptor-interacting protein kinase 3 (RIPK3) in intestinal epithelial cells. Surprisingly, necroptotic stimulation reduces autophagic activity, as evidenced by enlarged puncta of the autophagic substrate SQSTM1/p62 and its increased colocalization with LC3. However, necroptotic stimulation does not induce the lysosomal-associated membrane protein 1 (LAMP1) nor syntaxin 17, which mediates autophagosome-lysosome fusion, to colocalize with LC3. These data indicate that necroptosis attenuates autophagic flux before the lysosome fusion step. Our findings may provide insights into human diseases involving necroptosis.


Subject(s)
Autophagy , Epithelial Cells/cytology , Epithelial Cells/enzymology , Intestines/cytology , Necroptosis , Receptor-Interacting Protein Serine-Threonine Kinases/metabolism , Autophagosomes/drug effects , Autophagosomes/metabolism , Autophagy/drug effects , Cell Line, Tumor , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Humans , Lysosomes/drug effects , Lysosomes/metabolism , Microtubule-Associated Proteins/metabolism , Necroptosis/drug effects , Oligopeptides/pharmacology , Sequestosome-1 Protein/metabolism , Tumor Necrosis Factor-alpha/pharmacology
11.
Sci Rep ; 9(1): 11635, 2019 08 12.
Article in English | MEDLINE | ID: mdl-31406169

ABSTRACT

Lysosomes are largely responsible for significant degradation of intracellular and extracellular proteins via the secretory pathway, autophagy, and endocytosis. Therefore, dysregulation of lysosomal homeostasis influences diverse cellular functions. However, a straightforward and quantitative method to measure the integrity of the lysosomal pathway has not been developed. Here, we report the plasmid-based lysosomal-METRIQ (MEasurement of protein Transporting integrity by RatIo Quantification) probe that enables simple quantification of lysosomal integrity by lysosomal green and cytosolic red fluorescent proteins using a flow cytometer. In cultured cells, the lysosomal-METRIQ probe detected not only suppression of the lysosomal pathway but also upregulation of lysosomal activity such as lysosomal biogenesis. To identify factors involved in lysosomal homeostasis, we carried out compound screening and found that the cyclin-dependent kinase (CDK) inhibitors kenpaullone and purvalanol A induce synthesis of cathepsin D and an increase in the number of lysosomes. Subsequent studies revealed that CDK5 maintains lysosomal homeostasis independently of cell cycle arrest. Our results suggest that the lysosomal-METRIQ probe is an effective and efficient tool for measuring lysosomal activity in mammalian cells.


Subject(s)
Cyclin-Dependent Kinase 5/metabolism , Endocytosis/physiology , Luminescent Proteins/chemistry , Lysosomes/metabolism , Molecular Probes/chemistry , Benzazepines/pharmacology , Cyclin-Dependent Kinase 5/antagonists & inhibitors , Cyclin-Dependent Kinase 5/genetics , Endocytosis/drug effects , Flow Cytometry , Gene Knockdown Techniques , HEK293 Cells , HeLa Cells , Humans , Indoles/pharmacology , Luminescent Agents/chemistry , Luminescent Proteins/genetics , Lysosomes/drug effects , Microscopy, Confocal/methods , Microscopy, Fluorescence/methods , Molecular Probes/genetics , Plasmids/genetics , Purines/pharmacology
12.
Genes Cells ; 24(8): 546-558, 2019 Aug.
Article in English | MEDLINE | ID: mdl-31145520

ABSTRACT

The telomere protects the ends of eukaryotic linear chromosomes, and its shortening or erosion is recognized as DNA damage, leading to loss of proliferation activity and, thus, cellular senescence at the population level. Here, using a GFP-based DNA damage checkpoint marker suited for single-cell observation of Saccharomyces cerevisiae cells, we correlated the checkpoint status of telomere-shortened cells with their behavior. We show that some cells possessing short telomeres retain proliferation capacity even after the DNA damage checkpoint is activated. At the presenescent stage, the activation of the checkpoint causes cell cycle delay, but does not induce permanent cell cycle arrest, eventually leading to the expansion of cell size that is characteristic of cellular senescence. Moreover, the proliferation capacity of checkpoint-activated cells is not dependent on homologous recombination or the checkpoint adaptation pathway. The retention of proliferation capacity is specific to the telomere-derived DNA damage response, suggesting that damaged telomeres differ functionally from other types of DNA damage. Our data establish the role of the presenescent stage in telomere shortening-induced senescence, which proceeds gradually and is associated with a variety of changes, including altered cell morphology and metabolism.


Subject(s)
DNA Damage , Saccharomyces cerevisiae/genetics , Telomerase/genetics , Adaptation, Biological , Cell Cycle/genetics , Gene Expression , Genes, Reporter , Saccharomyces cerevisiae/metabolism , Saccharomyces cerevisiae Proteins/genetics , Saccharomyces cerevisiae Proteins/metabolism , Telomerase/metabolism , Telomere Shortening
13.
Exp Cell Res ; 376(2): 168-180, 2019 03 15.
Article in English | MEDLINE | ID: mdl-30742807

ABSTRACT

Ras-activated ERK pathway (Raf-MEK-ERK phosphorylation cascade) regulates a variety of cellular responses including cell proliferation, differentiation, survival, and apoptosis. DA-Raf1 (DA-Raf) is a splicing variant of A-Raf and contains the Ras-binding domain but lacks the kinase domain. Accordingly, DA-Raf antagonizes the Ras-ERK pathway in a dominant-negative manner. Here we show that DA-Raf plays essential roles in skeletal myocyte differentiation including myoblast fusion and in apoptosis, which are suppressed by the Ras-ERK pathway. Expression of DA-Raf was highly induced in C2C12 skeletal myocytes in a low serum concentration of differentiation condition and in NIH3T3 fibroblasts under a serum starvation apoptosis-inducing condition. Stable knockdown of DA-Raf resulted in suppression of muscle-specific gene expression, myoblast fusion, and apoptosis. In contrast, exogenous overexpression of DA-Raf prominently caused apoptosis. DA-Raf induces apoptosis by preventing ERK-RSK-mediated inhibitory phosphorylation of Bad. Although it has been reported that apoptosis triggers myoblast fusion, DA-Raf-induced apoptosis was not involved in myoblast fusion in C2C12 cells. These results imply that suppression of the Ras-ERK pathway by DA-Raf is essential for both myocyte differentiation including myoblast fusion and apoptosis but that apoptosis is not a prerequisite for myoblast fusion.


Subject(s)
Cell Differentiation/physiology , MAP Kinase Signaling System/physiology , Muscle Fibers, Skeletal/cytology , Proto-Oncogene Proteins A-raf/physiology , Animals , Apoptosis , Cell Fusion , Cell Line , HEK293 Cells , Humans , Mice , NIH 3T3 Cells , ras Proteins/metabolism
14.
Development ; 145(4)2018 02 23.
Article in English | MEDLINE | ID: mdl-29475974

ABSTRACT

Although autophagy is classically viewed as a non-selective degradation system, recent studies have revealed that various forms of selective autophagy also play crucial physiological roles. However, the induction of selective autophagy is not well understood. In this study, we established a forced selective autophagy system using a fusion of an autophagy adaptor and a substrate-binding protein. In both mammalian cells and fertilized mouse embryos, efficient forced lipophagy was induced by expression of a fusion of p62 (Sqstm1) and a lipid droplet (LD)-binding domain. In mouse embryos, induction of forced lipophagy caused a reduction in LD size and number, and decreased the triglyceride level throughout embryonic development, resulting in developmental retardation. Furthermore, lipophagy-induced embryos could eliminate excess LDs and were tolerant of lipotoxicity. Thus, by inducing forced lipophagy, expression of the p62 fusion protein generated LD-depleted cells, revealing an unexpected role of LD during preimplantation development.


Subject(s)
Autophagy/physiology , Embryonic Development/physiology , Lipid Droplets/metabolism , Lipid Metabolism/physiology , Animals , Cell Culture Techniques , Flow Cytometry , Immunoblotting , Lipolysis/physiology , Mice , Microscopy, Fluorescence , Perilipin-3/metabolism
15.
Mol Biol Cell ; 29(4): 510-522, 2018 02 15.
Article in English | MEDLINE | ID: mdl-29237820

ABSTRACT

Target of rapamycin complex 1 (TORC1) is a central cellular signaling coordinator that allows eukaryotic cells to adapt to the environment. In the budding yeast, Saccharomyces cerevisiae, TORC1 senses nitrogen and various stressors and modulates proteosynthesis, nitrogen uptake and metabolism, stress responses, and autophagy. There is some indication that TORC1 may regulate these downstream pathways individually. However, the potential mechanisms for such differential regulation are unknown. Here we show that the serine/threonine protein kinase Sch9 branch of TORC1 signaling depends specifically on the integrity of the vacuolar membrane, and this dependency originates in changes in Sch9 localization reflected by phosphatidylinositol 3,5-bisphosphate. Moreover, oxidative stress induces the delocalization of Sch9 from vacuoles, contributing to the persistent inhibition of the Sch9 branch after stress. Thus, our results establish that regulation of the vacuolar localization of Sch9 serves as a selective switch for the Sch9 branch in divergent TORC1 signaling. We propose that the Sch9 branch integrates the intrinsic activity of TORC1 kinase and vacuolar status, which is monitored by the phospholipids of the vacuolar membrane, into the regulation of macromolecular synthesis.


Subject(s)
Protein Serine-Threonine Kinases/metabolism , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae/genetics , Signal Transduction , Transcription Factors/metabolism , Vacuoles/metabolism , Gene Expression Regulation, Fungal , Gene Regulatory Networks , Microscopy, Fluorescence , Oxidative Stress/genetics , Protein Serine-Threonine Kinases/genetics , Saccharomyces cerevisiae/growth & development , Saccharomyces cerevisiae/metabolism , Saccharomyces cerevisiae Proteins/genetics , Transcription Factors/genetics
16.
Bio Protoc ; 7(15)2017 Aug 05.
Article in English | MEDLINE | ID: mdl-29075655

ABSTRACT

This protocol describes a method for purifying glycosylated FLAG-tagged secreted proteins with disulfide bonds from mammalian cells. The purified products can be used for various applications, such as ligand binding assays.

17.
FEBS Lett ; 591(9): 1199-1211, 2017 05.
Article in English | MEDLINE | ID: mdl-28369861

ABSTRACT

Basal autophagy plays an essential role as a protein quality control system. Although it has been demonstrated that the loss of autophagy results in the accumulation of ubiquitin-positive aggregates and the development of neurodegenerative diseases, the precise autophagy substrate(s) remain unclear. Here, we determined whether ubiquitinated proteins are direct substrates for basal autophagy using a fluorescent ratiometric probe for ubiquitin. We show that the degradation of polyubiquitinated proteins is not dependent on basal autophagy. Although ubiquitin-positive aggregates are observed in autophagy knockout cultured cells, the aggregates consist of soluble and mobile polyubiquitinated proteins, which are trapped by p62 without an increase in the total amount of ubiquitinated proteins. These results suggest that ubiquitinated proteins are not major targets for basal autophagy.


Subject(s)
Autophagy , Polyubiquitin/metabolism , Ubiquitinated Proteins/metabolism , Ubiquitination , Adaptor Proteins, Vesicular Transport/genetics , Adaptor Proteins, Vesicular Transport/metabolism , Autophagy-Related Proteins/genetics , Autophagy-Related Proteins/metabolism , Fluorescence Recovery After Photobleaching , HEK293 Cells , HeLa Cells , Humans , Immunoblotting , Microscopy, Fluorescence , Proteolysis , RNA Interference , Sequestosome-1 Protein/genetics , Sequestosome-1 Protein/metabolism , Ubiquitinated Proteins/genetics
18.
Nature ; 542(7639): 43-48, 2017 02 02.
Article in English | MEDLINE | ID: mdl-28099418

ABSTRACT

Interleukin-17 (IL-17) is a major pro-inflammatory cytokine: it mediates responses to pathogens or tissue damage, and drives autoimmune diseases. Little is known about its role in the nervous system. Here we show that IL-17 has neuromodulator-like properties in Caenorhabditis elegans. IL-17 can act directly on neurons to alter their response properties and contribution to behaviour. Using unbiased genetic screens, we delineate an IL-17 signalling pathway and show that it acts in the RMG hub interneurons. Disrupting IL-17 signalling reduces RMG responsiveness to input from oxygen sensors, and renders sustained escape from 21% oxygen transient and contingent on additional stimuli. Over-activating IL-17 receptors abnormally heightens responses to 21% oxygen in RMG neurons and whole animals. IL-17 deficiency can be bypassed by optogenetic stimulation of RMG. Inducing IL-17 expression in adults can rescue mutant defects within 6 h. These findings reveal a non-immunological role of IL-17 modulating circuit function and behaviour.


Subject(s)
Caenorhabditis elegans Proteins/metabolism , Caenorhabditis elegans/cytology , Caenorhabditis elegans/physiology , Interleukin-17/metabolism , Sensation/physiology , Animals , Behavior, Animal/drug effects , Behavior, Animal/physiology , Caenorhabditis elegans/drug effects , HEK293 Cells , Humans , Interneurons/drug effects , Interneurons/metabolism , Oxygen/metabolism , Oxygen/pharmacology , Receptors, Interleukin-17/metabolism , Sensation/drug effects , Signal Transduction/drug effects
19.
Dev Cell ; 39(1): 116-130, 2016 10 10.
Article in English | MEDLINE | ID: mdl-27693508

ABSTRACT

Autophagy is a cytoplasmic degradation system that is important for starvation adaptation and cellular quality control. Previously, we reported that Atg5-null mice are neonatal lethal; however, the exact cause of their death remains unknown. Here, we show that restoration of ATG5 in the brain is sufficient to rescue Atg5-null mice from neonatal lethality. This suggests that neuronal dysfunction, including suckling failure, is the primary cause of the death of Atg5-null neonates, which would further be accelerated by nutrient insufficiency due to a systemic failure in autophagy. The rescued Atg5-null mouse model, as a resource, allows us to investigate the physiological roles of autophagy in the whole body after the neonatal period. These rescued mice demonstrate previously unappreciated abnormalities such as hypogonadism and iron-deficiency anemia. These observations provide new insights into the physiological roles of the autophagy factor ATG5.


Subject(s)
Autophagy-Related Protein 5/deficiency , Neurons/metabolism , Anemia/genetics , Anemia/pathology , Animals , Animals, Newborn , Autophagy-Related Protein 5/metabolism , Brain/metabolism , Gene Expression Regulation, Developmental , Gonadotropins/metabolism , Green Fluorescent Proteins/metabolism , Iron/metabolism , Iron Deficiencies , Male , Mice, Knockout , Organ Specificity , Phosphopyruvate Hydratase/genetics , Promoter Regions, Genetic/genetics , Spermatogenesis , Testosterone/metabolism , Ubiquitinated Proteins/metabolism , Ubiquitination
20.
Mol Cell ; 63(1): 21-33, 2016 07 07.
Article in English | MEDLINE | ID: mdl-27345149

ABSTRACT

We investigated how mitochondrial membrane proteins remain soluble in the cytosol until their delivery to mitochondria or degradation at the proteasome. We show that Ubiquilin family proteins bind transmembrane domains in the cytosol to prevent aggregation and temporarily allow opportunities for membrane targeting. Over time, Ubiquilins recruit an E3 ligase to ubiquitinate bound clients. The attached ubiquitin engages Ubiquilin's UBA domain, normally bound to an intramolecular UBL domain, and stabilizes the Ubiquilin-client complex. This conformational change precludes additional chances at membrane targeting for the client, while simultaneously freeing Ubiquilin's UBL domain for targeting to the proteasome. Loss of Ubiquilins by genetic ablation or sequestration in polyglutamine aggregates leads to accumulation of non-inserted mitochondrial membrane protein precursors. These findings define Ubiquilins as a family of chaperones for cytosolically exposed transmembrane domains and explain how they use ubiquitin to triage clients for degradation via coordinated intra- and intermolecular interactions.


Subject(s)
Carrier Proteins/metabolism , Cell Cycle Proteins/metabolism , Membrane Proteins/metabolism , Mitochondria/metabolism , Mitochondrial Membranes/metabolism , Molecular Chaperones/metabolism , Proteolysis , Ubiquitins/metabolism , Adaptor Proteins, Signal Transducing , Autophagy-Related Proteins , CRISPR-Cas Systems , Carrier Proteins/chemistry , Carrier Proteins/genetics , Cell Cycle Proteins/chemistry , Cell Cycle Proteins/genetics , Cytosol/metabolism , HEK293 Cells , HeLa Cells , Humans , Molecular Chaperones/chemistry , Molecular Chaperones/genetics , Peptides/metabolism , Protein Aggregates , Protein Interaction Domains and Motifs , RNA Interference , Structure-Activity Relationship , Transfection , Ubiquitin-Protein Ligases/metabolism , Ubiquitination , Ubiquitins/chemistry , Ubiquitins/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...