Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 28
Filter
Add more filters










Publication year range
1.
FEBS Lett ; 596(22): 2873-2888, 2022 11.
Article in English | MEDLINE | ID: mdl-36056653

ABSTRACT

The Golgi pH regulator (GPHR) is essential for maintaining the function and morphology of the Golgi apparatus through the regulation of luminal acidic pH. Abnormal morphology of the Golgi apparatus is associated with neurodegenerative diseases. Here, we found that knockout of GPHR in the mouse brain led to morphological changes in the Golgi apparatus and neurodegeneration, which included brain atrophy, neuronal cell death, and gliosis. Furthermore, in the GPHR knockout mouse brain, transcriptional activity of sterol regulatory element-binding protein 2 (SREBP2) decreased, resulting in a reduction in cholesterol levels. GPHR-deficient cells exhibited suppressed neurite outgrowth, which was recovered by exogenous expression of the active form of SREBP2. Our results show that GPHR-mediated luminal acidification of the Golgi apparatus maintains proper cholesterol levels and, thereby, neuronal morphology.


Subject(s)
Golgi Apparatus , Sterol Regulatory Element Binding Protein 2 , Animals , Mice , Golgi Apparatus/metabolism , Sterol Regulatory Element Binding Protein 2/genetics , Sterol Regulatory Element Binding Protein 2/metabolism , Mice, Knockout , Cholesterol/metabolism , Hydrogen-Ion Concentration , Brain/metabolism
2.
J Biol Chem ; 297(6): 101405, 2021 12.
Article in English | MEDLINE | ID: mdl-34774801

ABSTRACT

Several amyotrophic lateral sclerosis (ALS)-related proteins such as FUS, TDP-43, and hnRNPA1 demonstrate liquid-liquid phase separation, and their disease-related mutations correlate with a transition of their liquid droplet form into aggregates. Missense mutations in SQSTM1/p62, which have been identified throughout the gene, are associated with ALS, frontotemporal degeneration (FTD), and Paget's disease of bone. SQSTM1/p62 protein forms liquid droplets through interaction with ubiquitinated proteins, and these droplets serve as a platform for autophagosome formation and the antioxidative stress response via the LC3-interacting region (LIR) and KEAP1-interacting region (KIR) of p62, respectively. However, it remains unclear whether ALS/FTD-related p62 mutations in the LIR and KIR disrupt liquid droplet formation leading to defects in autophagy, the stress response, or both. To evaluate the effects of ALS/FTD-related p62 mutations in the LIR and KIR on a major oxidative stress system, the Keap1-Nrf2 pathway, as well as on autophagic turnover, we developed systems to monitor each of these with high sensitivity. These methods such as intracellular protein-protein interaction assay, doxycycline-inducible gene expression system, and gene expression into primary cultured cells with recombinant adenovirus revealed that some mutants, but not all, caused reduced NRF2 activation and delayed autophagic cargo turnover. In contrast, while all p62 mutants demonstrated sufficient ability to form liquid droplets, all of these droplets also exhibited reduced inner fluidity. These results indicate that like other ALS-related mutant proteins, p62 missense mutations result in a primary defect in ALS/FTD via a qualitative change in p62 liquid droplet fluidity.


Subject(s)
Amyotrophic Lateral Sclerosis/metabolism , Sequestosome-1 Protein/metabolism , Amyotrophic Lateral Sclerosis/genetics , Animals , HEK293 Cells , Humans , Kelch-Like ECH-Associated Protein 1/genetics , Kelch-Like ECH-Associated Protein 1/metabolism , Mice , Mutation, Missense , NF-E2-Related Factor 2/genetics , NF-E2-Related Factor 2/metabolism , Sequestosome-1 Protein/genetics
3.
Nat Commun ; 12(1): 16, 2021 01 04.
Article in English | MEDLINE | ID: mdl-33397898

ABSTRACT

Autophagy contributes to the selective degradation of liquid droplets, including the P-Granule, Ape1-complex and p62/SQSTM1-body, although the molecular mechanisms and physiological relevance of selective degradation remain unclear. In this report, we describe the properties of endogenous p62-bodies, the effect of autophagosome biogenesis on these bodies, and the in vivo significance of their turnover. p62-bodies are low-liquidity gels containing ubiquitin and core autophagy-related proteins. Multiple autophagosomes form on the p62-gels, and the interaction of autophagosome-localizing Atg8-proteins with p62 directs autophagosome formation toward the p62-gel. Keap1 also reversibly translocates to the p62-gels in a p62-binding dependent fashion to activate the transcription factor Nrf2. Mice deficient for Atg8-interaction-dependent selective autophagy show that impaired turnover of p62-gels leads to Nrf2 hyperactivation in vivo. These results indicate that p62-gels are not simple substrates for autophagy but serve as platforms for both autophagosome formation and anti-oxidative stress.


Subject(s)
Autophagosomes/metabolism , Oxidative Stress , Sequestosome-1 Protein/metabolism , Animals , Apoptosis Regulatory Proteins/metabolism , Autophagosomes/ultrastructure , Autophagy , Cell Line , Gels , Hepatocytes/metabolism , Hepatocytes/ultrastructure , Humans , Kelch-Like ECH-Associated Protein 1/metabolism , Liver/injuries , Liver/pathology , Mice, Inbred C57BL , Microtubule-Associated Proteins/metabolism , NF-E2-Related Factor 2/metabolism , Protein Binding , Unilamellar Liposomes
4.
EMBO Rep ; 21(3): e48902, 2020 03 04.
Article in English | MEDLINE | ID: mdl-31916398

ABSTRACT

p62/SQSTM1 is a multivalent protein that has the ability to cause liquid-liquid phase separation and serves as a receptor protein that participates in cargo isolation during selective autophagy. This protein is also involved in the non-canonical activation of the Keap1-Nrf2 system, a major oxidative stress response pathway. Here, we show a role of neighbor of BRCA1 gene 1 (NBR1), an autophagy receptor structurally similar to p62/SQSTM1, in p62-liquid droplet formation and Keap1-Nrf2 pathway activation. Overexpression of NBR1 blocks selective degradation of p62/SQSTM1 through autophagy and promotes the accumulation and phosphorylation of p62/SQSTM1 in liquid-like bodies, which is required for the activation of Nrf2. NBR1 is induced in response to oxidative stress, which triggers p62-mediated Nrf2 activation. Conversely, loss of Nbr1 suppresses not only the formation of p62/SQSTM1-liquid droplets, but also of p62-dependent Nrf2 activation during oxidative stress. Taken together, our results show that NBR1 mediates p62/SQSTM1-liquid droplet formation to activate the Keap1-Nrf2 pathway.


Subject(s)
NF-E2-Related Factor 2 , Signal Transduction , Animals , Autophagy , Kelch-Like ECH-Associated Protein 1/genetics , Kelch-Like ECH-Associated Protein 1/metabolism , Mice , NF-E2-Related Factor 2/genetics , NF-E2-Related Factor 2/metabolism , Sequestosome-1 Protein/genetics , Sequestosome-1 Protein/metabolism
5.
Life Sci Alliance ; 3(1)2020 01.
Article in English | MEDLINE | ID: mdl-31879337

ABSTRACT

Lipid droplets (LDs) are dynamic organelles that store neutral lipids during times of energy excess, such as after a meal. LDs serve as an energy reservoir during fasting and have a buffering capacity that prevents lipotoxicity. Autophagy and the autophagic machinery have been proposed to play a role in LD biogenesis, but the underlying molecular mechanism remains unclear. Here, we show that when nuclear receptor co-repressor 1 (NCoR1), which inhibits the transactivation of nuclear receptors, accumulates because of autophagy suppression, LDs decrease in size and number. Ablation of ATG7, a gene essential for autophagy, suppressed the expression of gene targets of liver X receptor α, a nuclear receptor responsible for fatty acid and triglyceride synthesis in an NCoR1-dependent manner. LD accumulation in response to fasting and after hepatectomy was hampered by the suppression of autophagy. These results suggest that autophagy controls physiological hepatosteatosis by fine-tuning NCoR1 protein levels.


Subject(s)
Autophagy-Related Protein 7/genetics , Autophagy/genetics , Fatty Liver/metabolism , Nuclear Receptor Co-Repressor 1/metabolism , Animals , Fasting/metabolism , Fatty Acids/biosynthesis , Gene Knockout Techniques , Hep G2 Cells , Humans , Lipid Droplets/metabolism , Lipogenesis/genetics , Liver X Receptors/genetics , Liver X Receptors/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Nuclear Receptor Co-Repressor 1/genetics , Transfection , Triglycerides/biosynthesis
6.
eNeuro ; 6(3)2019.
Article in English | MEDLINE | ID: mdl-31118204

ABSTRACT

The Golgi apparatus plays an indispensable role in posttranslational modification and transport of proteins to their target destinations. Although it is well established that the Golgi apparatus requires an acidic luminal pH for optimal activity, morphological and functional abnormalities at the neuronal circuit level because of perturbations in Golgi pH are not fully understood. In addition, morphological alteration of the Golgi apparatus is associated with several neurodegenerative diseases, including Parkinson's disease, Alzheimer's disease, and amyotrophic lateral sclerosis. Here, we used anatomical and electrophysiological approaches to characterize morphological and functional abnormalities of neuronal circuits in Golgi pH regulator (GPHR) conditional knock-out mice. Purkinje cells (PCs) from the mutant mice exhibited vesiculation and fragmentation of the Golgi apparatus, followed by axonal degeneration and progressive cell loss. Morphological analysis provided evidence for the disruption of basket cell (BC) terminals around PC soma, and electrophysiological recordings showed selective loss of large amplitude responses, suggesting BC terminal disassembly. In addition, the innervation of mutant PCs was altered such that climbing fiber (CF) terminals abnormally synapsed on the somatic spines of mutant PCs in the mature cerebellum. The combined results describe an essential role for luminal acidification of the Golgi apparatus in maintaining proper neuronal morphology and neuronal circuitry.


Subject(s)
Cerebellum/metabolism , Cerebellum/ultrastructure , Golgi Apparatus/ultrastructure , Neuronal Plasticity , Neurons/ultrastructure , Receptors, G-Protein-Coupled/metabolism , Animals , Cerebellar Ataxia/metabolism , Cerebellar Ataxia/pathology , Disease Models, Animal , Female , Golgi Apparatus/metabolism , Hydrogen-Ion Concentration , Male , Mice, Knockout , Neural Pathways/metabolism , Neural Pathways/ultrastructure , Neurons/metabolism , Primary Cell Culture , Purkinje Cells/metabolism , Purkinje Cells/ultrastructure
7.
Nat Microbiol ; 2: 17066, 2017 May 08.
Article in English | MEDLINE | ID: mdl-28481361

ABSTRACT

Ubiquitination of invading Salmonella Typhimurium triggers autophagy of cytosolic bacteria and restricts their spread in epithelial cells. Ubiquitin (Ub) chains recruit autophagy receptors such as p62/SQSTM1, NDP52/CALCOCO and optineurin (OPTN), which initiate the formation of double-membrane autophagosomal structures and lysosomal destruction in a process known as xenophagy. Besides this, the functional consequences and mechanistic regulation of differentially linked Ub chains at the host-Salmonella interface have remained unexplored. Here, we show, for the first time, that distinct Ub chains on cytosolic S. Typhimurium serve as a platform triggering further signalling cascades. By using single-molecule localization microscopy, we visualized the balance and nanoscale distribution pattern of linear (M1-linked) Ub chain formation at the surface of cytosolic S. Typhimurium. In addition, we identified the deubiquitinase OTULIN as central regulator of these M1-linked Ub chains on the bacterial coat. OTULIN depletion leads to enhanced formation of linear Ub chains, resulting in local recruitment of NEMO, activation of IKKα/IKKß and ultimately NF-κB, which in turn promotes secretion of pro-inflammatory cytokines and restricts bacterial proliferation. Our results establish a role for the linear Ub coat around cytosolic S. Typhimurium as the local NF-κB signalling platform and provide insights into the function of OTULIN in NF-κB activation during bacterial pathogenesis.


Subject(s)
Cytosol/microbiology , Endopeptidases/metabolism , NF-kappa B/metabolism , Salmonella typhimurium/metabolism , Signal Transduction , Ubiquitination , Autophagy , Cell Proliferation , Cytosol/metabolism , Endopeptidases/genetics , Epithelial Cells/microbiology , HEK293 Cells , HeLa Cells , Host-Pathogen Interactions/genetics , Host-Pathogen Interactions/physiology , Humans , I-kappa B Kinase/genetics , I-kappa B Kinase/metabolism , NF-kappa B/genetics , Salmonella typhimurium/pathogenicity , Ubiquitin/metabolism
8.
Nat Commun ; 6: 6116, 2015 Jan 20.
Article in English | MEDLINE | ID: mdl-25600778

ABSTRACT

Ubiquitin is known to be one of the most soluble and stably folded intracellular proteins, but it is often found in inclusion bodies associated with various diseases including neurodegenerative disorders and cancer. To gain insight into this contradictory behaviour, we have examined the physicochemical properties of ubiquitin and its polymeric chains that lead to aggregate formation. We find that the folding stability of ubiquitin chains unexpectedly decreases with increasing chain length, resulting in the formation of amyloid-like fibrils. Furthermore, when expressed in cells, polyubiquitin chains covalently linked to EGFP also form aggregates depending on chain length. Notably, these aggregates are selectively degraded by autophagy. We propose a novel model in which the physical and chemical instability of polyubiquitin chains drives the formation of fibrils, which then serve as an initiation signal for autophagy.


Subject(s)
Polyubiquitin/chemistry , Polyubiquitin/metabolism , Animals , Autophagy , Calorimetry, Differential Scanning , Circular Dichroism , Escherichia coli/metabolism , Immunohistochemistry , Mice , Microscopy, Electron, Transmission , Polyubiquitin/ultrastructure , Sf9 Cells , Spectrometry, Fluorescence , Ubiquitin/chemistry , Ubiquitin/metabolism
9.
Mol Cell ; 56(2): 261-274, 2014 Oct 23.
Article in English | MEDLINE | ID: mdl-25219498

ABSTRACT

Biological roles for UFM1, a ubiquitin-like protein, are largely unknown, and therefore we screened for targets of ufmylation. Here we show that ufmylation of the nuclear receptor coactivator ASC1 is a key step for ERα transactivation in response to 17ß-estradiol (E2). In the absence of E2, the UFM1-specific protease UfSP2 was bound to ASC1, which maintains ASC1 in a nonufmylated state. In the presence of E2, ERα bound ASC1 and displaced UfSP2, leading to ASC1 ufmylation. Polyufmylation of ASC1 enhanced association of p300, SRC1, and ASC1 at promoters of ERα target genes. ASC1 overexpression or UfSP2 knockdown promoted ERα-mediated tumor formation in vivo, which could be abrogated by treatment with the anti-breast cancer drug tamoxifen. In contrast, expression of ufmylation-deficient ASC1 mutant or knockdown of the UFM1-activating E1 enzyme UBA5 prevented tumor growth. These findings establish a role for ASC1 ufmylation in breast cancer development by promoting ERα transactivation.


Subject(s)
Amino Acid Transport System y+/metabolism , Breast Neoplasms/pathology , Estrogen Receptor alpha/metabolism , Proteins/chemistry , Amino Acid Transport System y+/chemistry , Amino Acid Transport System y+/genetics , Animals , Breast Neoplasms/metabolism , Carrier Proteins/metabolism , Cell Line, Tumor , Cysteine Endopeptidases/metabolism , E1A-Associated p300 Protein/genetics , Enzyme Activation/genetics , Estradiol/genetics , Estradiol/metabolism , Estrogen Antagonists/pharmacology , Estrogen Receptor alpha/genetics , Female , HEK293 Cells , Humans , MCF-7 Cells , Mice , Mice, Nude , Nuclear Receptor Coactivator 1/genetics , Promoter Regions, Genetic/genetics , Protein Binding/genetics , Proteins/metabolism , Tamoxifen/pharmacology , Transcriptional Activation , Ubiquitin/metabolism , Ubiquitin-Activating Enzymes/genetics , Ubiquitin-Protein Ligases/metabolism
10.
J Biol Chem ; 289(36): 24944-55, 2014 Sep 05.
Article in English | MEDLINE | ID: mdl-25049227

ABSTRACT

The ubiquitin-proteasome system and autophagy are crucially important for proteostasis in cells. These pathways are interdependent, and dysfunction in either pathway causes accumulation of ubiquitin-positive aggregates, a hallmark of human pathological conditions. To elucidate in vivo compensatory action(s) against proteasomal dysfunction, we developed mice with reduced proteasome activity in their livers. The mutant mice exhibited severe liver damage, accompanied by formation of aggregates positive for ubiquitin and p62/Sqstm1, an adaptor protein for both selective autophagy and the anti-oxidative Keap1-Nrf2 pathway. These aggregates were selectively entrapped by autophagosomes, and pathological features of livers with impaired proteasome activity were exacerbated by simultaneous suppression of autophagy. In contrast, concomitant loss of p62/Sqstm1 had no apparent effect on the liver pathology though p62/Sqstm1 was indispensable for the aggregates formation. Furthermore, defective proteasome function led to transcriptional activation of the Nrf2, which served as a physiological adaptation. Our in vivo data suggest that cells contain networks of cellular defense mechanisms against defective proteostasis.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Autophagy , Cytoskeletal Proteins/metabolism , NF-E2-Related Factor 2/metabolism , Proteasome Endopeptidase Complex/metabolism , Signal Transduction , Adaptor Proteins, Signal Transducing/genetics , Animals , Cytoskeletal Proteins/genetics , Heat-Shock Proteins/genetics , Heat-Shock Proteins/metabolism , Immunoblotting , Kelch-Like ECH-Associated Protein 1 , Liver/metabolism , Liver/pathology , Liver/ultrastructure , Mice, Knockout , Mice, Transgenic , Microscopy, Confocal , Microscopy, Immunoelectron , NF-E2-Related Factor 2/genetics , Phagosomes/genetics , Phagosomes/metabolism , Phosphorylation , Proteasome Endopeptidase Complex/genetics , Sequestosome-1 Protein , Time Factors , Ubiquitin/metabolism
11.
Mol Cell Biol ; 34(9): 1695-706, 2014 May.
Article in English | MEDLINE | ID: mdl-24591649

ABSTRACT

Recent findings have suggested that the autophagic isolation membrane (IM) might originate from a domain of the endoplasmic reticulum (ER) called the omegasome. However, the morphological relationships between ER, omegasome, and IM remain unclear. In the present study, we found that hybrid structures composed of a double FYVE domain-containing protein 1 (DFCP1)-positive omegasome and the IM accumulated in Atg3-deficient mouse embryonic fibroblasts (MEFs). Moreover, correlative light and electron microscopy and immunoelectron microscopy revealed that green fluorescent protein (GFP)-tagged DFCP1 was localized on tubular or vesicular elements adjacent to the IM rims. Through detailed morphological analyses, including optimization of a fixation method and electron tomography, we observed a cluster of thin tubular structures between the IM edges and ER, part of which were continuous with IM and/or ER. The formation of these thin tubular clusters was observed in several cell lines and MEFs deficient for Atg5, Atg7, or Atg16L1 but not in FIP200-deficient cells, suggesting that they were relevant to the earlier events in autophagosome formation. Taken together, our findings indicate that these tubular profiles represent a part of the omegasome that links the ER with the IM.


Subject(s)
Autophagy , Endoplasmic Reticulum/ultrastructure , Fibroblasts/cytology , Animals , Autophagy-Related Proteins , Cell Line , Electron Microscope Tomography , Endoplasmic Reticulum/metabolism , Fibroblasts/metabolism , Fibroblasts/ultrastructure , Gene Deletion , Membrane Proteins/analysis , Mice , Microscopy, Immunoelectron , Ubiquitin-Conjugating Enzymes/genetics
12.
Biochem Biophys Res Commun ; 446(1): 309-15, 2014 Mar 28.
Article in English | MEDLINE | ID: mdl-24582747

ABSTRACT

Autophagy is a unique intracellular protein degradation system accompanied by autophagosome formation. Besides its important role through bulk degradation in supplying nutrients, this system has an ability to degrade certain proteins, organelles, and invading bacteria selectively to maintain cellular homeostasis. In yeasts, Atg8p plays key roles in both autophagosome formation and selective autophagy based on its membrane fusion property and interaction with autophagy adaptors/specific substrates. In contrast to the single Atg8p in yeast, mammals have 6 homologs of Atg8p comprising LC3 and GABARAP families. However, it is not clear these two families have different or similar functions. The aim of this study was to determine the separate roles of LC3 and GABARAP families in basal/constitutive and/or selective autophagy. While the combined knockdown of LC3 and GABARAP families caused a defect in long-lived protein degradation through lysosomes, knockdown of each had no effect on the degradation. Meanwhile, knockdown of LC3B but not GABARAPs resulted in significant accumulation of p62/Sqstm1, one of the selective substrate for autophagy. Our results suggest that while mammalian Atg8 homologs are functionally redundant with regard to autophagosome formation, selective autophagy is regulated by specific Atg8 homologs.


Subject(s)
Adaptor Proteins, Signal Transducing/physiology , Autophagy/physiology , Microtubule-Associated Proteins/physiology , Adaptor Proteins, Signal Transducing/chemistry , Adaptor Proteins, Signal Transducing/genetics , Animals , Apoptosis Regulatory Proteins , Autophagy/genetics , Autophagy-Related Protein 8 Family , Cells, Cultured , HEK293 Cells , Heat-Shock Proteins/chemistry , Heat-Shock Proteins/genetics , Heat-Shock Proteins/physiology , Humans , Mice , Microfilament Proteins/chemistry , Microfilament Proteins/genetics , Microfilament Proteins/physiology , Microtubule-Associated Proteins/chemistry , Microtubule-Associated Proteins/genetics , Models, Molecular , RNA, Small Interfering/genetics , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Sequestosome-1 Protein , Static Electricity
13.
Mol Cell ; 51(5): 618-31, 2013 Sep 12.
Article in English | MEDLINE | ID: mdl-24011591

ABSTRACT

The Keap1-Nrf2 system and autophagy are both involved in the oxidative-stress response, metabolic pathways, and innate immunity, and dysregulation of these processes is associated with pathogenic processes. However, the interplay between these two pathways remains largely unknown. Here, we show that phosphorylation of the autophagy-adaptor protein p62 markedly increases p62's binding affinity for Keap1, an adaptor of the Cul3-ubiquitin E3 ligase complex responsible for degrading Nrf2. Thus, p62 phosphorylation induces expression of cytoprotective Nrf2 targets. p62 is assembled on selective autophagic cargos such as ubiquitinated organelles and subsequently phosphorylated in an mTORC1-dependent manner, implying coupling of the Keap1-Nrf2 system to autophagy. Furthermore, persistent activation of Nrf2 through accumulation of phosphorylated p62 contributes to the growth of human hepatocellular carcinomas (HCCs). These results demonstrate that selective autophagy and the Keap1-Nrf2 pathway are interdependent, and that inhibitors of the interaction between phosphorylated p62 and Keap1 have potential as therapeutic agents against human HCC.


Subject(s)
Adaptor Proteins, Signal Transducing/chemistry , Adaptor Proteins, Signal Transducing/metabolism , Autophagy/physiology , Cytoskeletal Proteins/chemistry , Cytoskeletal Proteins/metabolism , NF-E2-Related Factor 2/metabolism , Adenoma/metabolism , Adenoma/pathology , Amino Acid Sequence , Animals , Carcinoma, Hepatocellular/metabolism , Carcinoma, Hepatocellular/pathology , Crystallography, X-Ray , Heat-Shock Proteins/chemistry , Heat-Shock Proteins/metabolism , Kelch-Like ECH-Associated Protein 1 , Liver Neoplasms/metabolism , Liver Neoplasms/pathology , Mechanistic Target of Rapamycin Complex 1 , Mice , Molecular Sequence Data , Multiprotein Complexes/metabolism , Phosphorylation , Sequestosome-1 Protein , TOR Serine-Threonine Kinases/metabolism
14.
J Biol Chem ; 286(36): 31864-74, 2011 Sep 09.
Article in English | MEDLINE | ID: mdl-21715324

ABSTRACT

p62/SQSTM1/A170 is a multimodular protein that is found in ubiquitin-positive inclusions associated with neurodegenerative diseases. Recent findings indicate that p62 mediates the interaction between ubiquitinated proteins and autophagosomes, leading these proteins to be degraded via the autophagy-lysosomal pathway. This ubiquitin-mediated selective autophagy is thought to begin with recognition of the ubiquitinated proteins by the C-terminal ubiquitin-associated (UBA) domain of p62. We present here the crystal structure of the UBA domain of mouse p62 and the solution structure of its ubiquitin-bound form. The p62 UBA domain adopts a novel dimeric structure in crystals, which is distinctive from those of other UBA domains. NMR analyses reveal that in solution the domain exists in equilibrium between the dimer and monomer forms, and binding ubiquitin shifts the equilibrium toward the monomer to form a 1:1 complex between the UBA domain and ubiquitin. The dimer-to-monomer transition is associated with a structural change of the very C-terminal end of the p62 UBA domain, although the UBA fold itself is essentially maintained. Our data illustrate that dimerization and ubiquitin binding of the p62 UBA domain are incompatible with each other. These observations reveal an autoinhibitory mechanism in the p62 UBA domain and suggest that autoinhibition plays a role in the function of p62.


Subject(s)
Adaptor Proteins, Signal Transducing/chemistry , Heat-Shock Proteins/chemistry , Protein Interaction Domains and Motifs , Ubiquitin/metabolism , Adaptor Proteins, Signal Transducing/antagonists & inhibitors , Animals , Crystallography, X-Ray , Heat-Shock Proteins/antagonists & inhibitors , Magnetic Resonance Spectroscopy , Mice , Phase Transition , Protein Multimerization , Sequestosome-1 Protein , Ubiquitin/chemistry
15.
Nat Commun ; 2: 181, 2011 Feb 08.
Article in English | MEDLINE | ID: mdl-21304510

ABSTRACT

Post-translational protein modifications are systems designed to expand restricted genomic information through functional conversion of target molecules. Ubiquitin-like post-translational modifiers regulate numerous cellular events through their covalent linkages to target protein(s) by an enzymatic cascade analogous to ubiquitylation consisting of E1 (activating), E2 (conjugating) and E3 (ligating) enzymes. In this study, we report the essential role of Uba5, a specific activating enzyme for the ubiquitin-like modifier, Ufm1, in erythroid development. Mice lacking Uba5 exhibited severe anaemia, followed by death in utero. Although Uba5 was dispensable for the production of erythropoietin, its genetic loss led to impaired development of megakaryocyte and erythroid progenitors from common myeloid progenitors. Intriguingly, transgenic expression of Uba5 in the erythroid lineage rescued the Uba5-deficient embryos from anaemia and prolonged their survival, demonstrating the importance of Uba5 in cell-autonomous erythroid differentiation. Our results suggest that one of the ubiquitin-like protein modification systems, the Ufm1 system, is involved in the regulation of haematopoiesis.


Subject(s)
Cell Differentiation/physiology , Erythroid Cells , Hematopoiesis/physiology , Protein Processing, Post-Translational/physiology , Proteins/metabolism , Ubiquitin-Activating Enzymes/metabolism , Animals , Cell Line , Colony-Forming Units Assay , DNA Primers/genetics , Flow Cytometry , Humans , Immunoblotting , Immunoprecipitation , Mice , Mice, Transgenic , Reverse Transcriptase Polymerase Chain Reaction , Ubiquitin-Activating Enzymes/genetics
16.
Genes Cells ; 15(8): 887-900, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20604804

ABSTRACT

PINK1 and Parkin were first identified as the causal genes responsible for familial forms of early-onset Parkinson's disease (PD), a prevalent neurodegenerative disorder. PINK1 encodes a mitochondrial serine/threonine protein kinase, whereas Parkin encodes an ubiquitin-protein ligase. PINK1 and Parkin cooperate to maintain mitochondrial integrity; however, the detailed molecular mechanism of how Parkin-catalyzed ubiquitylation results in mitochondrial integrity remains an enigma. In this study, we show that Parkin-catalyzed K63-linked polyubiquitylation of depolarized mitochondria resulted in ubiquitylated mitochondria being transported along microtubules to cluster in the perinuclear region, which was interfered by pathogenic mutations of Parkin. In addition, p62/SQSTM1 (hereafter referred to as p62) was recruited to depolarized mitochondria after Parkin-directed ubiquitylation. Intriguingly, deletion of p62 in mouse embryonic fibroblasts resulted in a gross loss of mitochondrial perinuclear clustering but did not hinder mitochondrial degradation. Thus, p62 is required for ubiquitylation-dependent clustering of damaged mitochondria, which resembles p62-mediated 'aggresome' formation of misfolded/unfolded proteins after ubiquitylation.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Cell Nucleus/metabolism , Mitochondria/metabolism , Ubiquitin-Protein Ligases/metabolism , Adaptor Proteins, Signal Transducing/deficiency , Adaptor Proteins, Signal Transducing/genetics , Biocatalysis , Cells, Cultured , DNA, Mitochondrial/genetics , HeLa Cells , Humans , Protein Folding , Sequestosome-1 Protein , Ubiquitin/metabolism , Ubiquitin-Protein Ligases/genetics , Ubiquitination
17.
J Cell Biol ; 189(2): 211-21, 2010 Apr 19.
Article in English | MEDLINE | ID: mdl-20404107

ABSTRACT

Parkinson's disease (PD) is a prevalent neurodegenerative disorder. Recent identification of genes linked to familial forms of PD such as Parkin and PINK1 (PTEN-induced putative kinase 1) has revealed that ubiquitylation and mitochondrial integrity are key factors in disease pathogenesis. However, the exact mechanism underlying the functional interplay between Parkin-catalyzed ubiquitylation and PINK1-regulated mitochondrial quality control remains an enigma. In this study, we show that PINK1 is rapidly and constitutively degraded under steady-state conditions in a mitochondrial membrane potential-dependent manner and that a loss in mitochondrial membrane potential stabilizes PINK1 mitochondrial accumulation. Furthermore, PINK1 recruits Parkin from the cytoplasm to mitochondria with low membrane potential to initiate the autophagic degradation of damaged mitochondria. Interestingly, the ubiquitin ligase activity of Parkin is repressed in the cytoplasm under steady-state conditions; however, PINK1-dependent mitochondrial localization liberates the latent enzymatic activity of Parkin. Some pathogenic mutations of PINK1 and Parkin interfere with the aforementioned events, suggesting an etiological importance. These results provide crucial insight into the pathogenic mechanisms of PD.


Subject(s)
Membrane Potential, Mitochondrial/physiology , Mitochondria/pathology , Mitochondria/physiology , Protein Kinases/metabolism , Ubiquitin-Protein Ligases/metabolism , Animals , Biomarkers/metabolism , Carbonyl Cyanide m-Chlorophenyl Hydrazone/pharmacology , Dimethyl Sulfoxide/pharmacology , Fibroblasts/cytology , Fibroblasts/metabolism , Fluorescent Dyes/metabolism , HeLa Cells , Humans , Mice , Mice, Knockout , Mitochondria/drug effects , Mitochondrial Proteins/genetics , Mitochondrial Proteins/metabolism , Parkinson Disease/metabolism , Parkinson Disease/pathology , Protein Kinases/genetics , Receptors, Cytoplasmic and Nuclear/genetics , Receptors, Cytoplasmic and Nuclear/metabolism , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Solvents/pharmacology , Ubiquitin-Protein Ligases/genetics , Uncoupling Agents/pharmacology
18.
Nat Cell Biol ; 12(3): 213-23, 2010 Mar.
Article in English | MEDLINE | ID: mdl-20173742

ABSTRACT

Impaired selective turnover of p62 by autophagy causes severe liver injury accompanied by the formation of p62-positive inclusions and upregulation of detoxifying enzymes. These phenotypes correspond closely to the pathological conditions seen in human liver diseases, including alcoholic hepatitis and hepatocellular carcinoma. However, the molecular mechanisms and pathophysiological processes in these events are still unknown. Here we report the identification of a novel regulatory mechanism by p62 of the transcription factor Nrf2, whose target genes include antioxidant proteins and detoxification enzymes. p62 interacts with the Nrf2-binding site on Keap1, a component of Cullin-3-type ubiquitin ligase for Nrf2. Thus, an overproduction of p62 or a deficiency in autophagy competes with the interaction between Nrf2 and Keap1, resulting in stabilization of Nrf2 and transcriptional activation of Nrf2 target genes. Our findings indicate that the pathological process associated with p62 accumulation results in hyperactivation of Nrf2 and delineates unexpected roles of selective autophagy in controlling the transcription of cellular defence enzyme genes.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Autophagy/physiology , Cytoskeletal Proteins/metabolism , Heat-Shock Proteins/metabolism , NF-E2-Related Factor 2/metabolism , Oxidative Stress/physiology , Signal Transduction/physiology , Adaptor Proteins, Signal Transducing/chemistry , Adaptor Proteins, Signal Transducing/genetics , Animals , Autophagy-Related Protein 7 , Binding, Competitive/physiology , Calorimetry , Cell Line , Crystallography, X-Ray , Cytoskeletal Proteins/chemistry , Cytoskeletal Proteins/genetics , Gene Expression/genetics , Heat-Shock Proteins/chemistry , Heat-Shock Proteins/genetics , Hepatocytes/metabolism , Hepatocytes/pathology , Humans , Inclusion Bodies/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Kelch-Like ECH-Associated Protein 1 , Liver/metabolism , Liver/pathology , Liver/physiopathology , Mice , Mice, Knockout , Mice, Transgenic , Microtubule-Associated Proteins/genetics , Models, Biological , Models, Molecular , Mutation/physiology , NF-E2-Related Factor 2/genetics , Organ Size/genetics , Protein Binding/physiology , Protein Interaction Domains and Motifs/physiology , Protein Interaction Mapping , Sequestosome-1 Protein , Transfection
19.
J Biol Chem ; 285(8): 5417-27, 2010 Feb 19.
Article in English | MEDLINE | ID: mdl-20018847

ABSTRACT

The ubiquitin fold modifier 1 (Ufm1) is the most recently discovered ubiquitin-like modifier whose conjugation (ufmylation) system is conserved in multicellular organisms. Ufm1 is known to covalently attach with cellular protein(s) via a specific E1-activating enzyme (Uba5) and an E2-conjugating enzyme (Ufc1), but its E3-ligating enzyme(s) as well as the target protein(s) remain unknown. Herein, we report both a novel E3 ligase for Ufm1, designated Ufl1, and an Ufm1-specific substrate ligated by Ufl1, C20orf116. Ufm1 was covalently conjugated with C20orf116. Although Ufl1 has no obvious sequence homology to any other known E3s for ubiquitin and ubiquitin-like modifiers, the C20orf116 x Ufm1 formation was greatly accelerated by Ufl1. The C20orf116 x Ufm1 conjugate was cleaved by Ufm1-specific proteases, implying the reversibility of ufmylation. The conjugation was abundant in the liver and lungs of Ufm1-transgenic mice, fractionated into membrane fraction, and impaired in Uba5 knock-out cells. Intriguingly, immunological analysis revealed localizations of Ufl1 and C20orf116 mainly to the endoplasmic reticulum. Our results provide novel insights into the Ufm1 system involved in cellular regulation of multicellular organisms.


Subject(s)
Carrier Proteins/metabolism , Endoplasmic Reticulum/metabolism , Proteins/metabolism , Ubiquitin-Protein Ligases/metabolism , Ubiquitin/metabolism , Animals , Carrier Proteins/genetics , Endoplasmic Reticulum/genetics , Humans , Mice , Mice, Knockout , Proteins/genetics , Ubiquitin/genetics , Ubiquitin-Activating Enzymes/genetics , Ubiquitin-Activating Enzymes/metabolism , Ubiquitin-Protein Ligases/genetics
20.
Mol Cell ; 33(4): 505-16, 2009 Feb 27.
Article in English | MEDLINE | ID: mdl-19250911

ABSTRACT

Autophagy is a catabolic process where cytosolic cellular components are delivered to the lysosome for degradation. Recent studies have indicated the existence of specific receptors, such as p62, which link ubiquitinated targets to autophagosomal degradation pathways. Here we show that NBR1 (neighbor of BRCA1 gene 1) is an autophagy receptor containing LC3- and ubiquitin (Ub)-binding domains. NBR1 is recruited to Ub-positive protein aggregates and degraded by autophagy depending on an LC3-interacting region (LIR) and LC3 family modifiers. Although NBR1 and p62 interact and form oligomers, they can function independently, as shown by autophagosomal clearance of NBR1 in p62-deficient cells. NBR1 was localized to Ub-positive inclusions in patients with liver dysfunction, and depletion of NBR1 abolished the formation of Ub-positive p62 bodies upon puromycin treatment of cells. We propose that NBR1 and p62 act as receptors for selective autophagosomal degradation of ubiquitinated targets.


Subject(s)
Autophagy , Proteins/metabolism , Ubiquitin/metabolism , Adaptor Proteins, Signal Transducing/metabolism , Animals , Binding Sites , Cells, Cultured , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , HeLa Cells , Humans , Intracellular Signaling Peptides and Proteins , Mice , Microscopy, Confocal , Microtubule-Associated Proteins/genetics , Microtubule-Associated Proteins/metabolism , Proteins/analysis , Sequestosome-1 Protein , Substrate Specificity
SELECTION OF CITATIONS
SEARCH DETAIL
...