Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 1.251
Filter
Add more filters

Publication year range
1.
Cell ; 186(19): 4172-4188.e18, 2023 09 14.
Article in English | MEDLINE | ID: mdl-37633267

ABSTRACT

Selective clearance of organelles, including endoplasmic reticulum (ER) and mitochondria, by autophagy plays an important role in cell health. Here, we describe a developmentally programmed selective ER clearance by autophagy. We show that Parkinson's disease-associated PINK1, as well as Atl, Rtnl1, and Trp1 receptors, regulate ER clearance by autophagy. The E3 ubiquitin ligase Parkin functions downstream of PINK1 and is required for mitochondrial clearance while having the opposite function in ER clearance. By contrast, Keap1 and the E3 ubiquitin ligase Cullin3 function downstream of PINK1 to regulate ER clearance by influencing Rtnl1 and Atl. PINK1 regulates a change in Keap1 localization and Keap1-dependent ubiquitylation of the ER-phagy receptor Rtnl1 to facilitate ER clearance. Thus, PINK1 regulates the selective clearance of ER and mitochondria by influencing the balance of Keap1- and Parkin-dependent ubiquitylation of substrates that determine which organelle is removed by autophagy.


Subject(s)
Endoplasmic Reticulum , NF-E2-Related Factor 2 , Endoplasmic Reticulum/metabolism , Kelch-Like ECH-Associated Protein 1 , Protein Kinases , Ubiquitin-Protein Ligases , Drosophila melanogaster , Animals
2.
Annu Rev Cell Dev Biol ; 36: 237-264, 2020 10 06.
Article in English | MEDLINE | ID: mdl-32749865

ABSTRACT

Parkinson's disease (PD) is a leading cause of neurodegeneration that is defined by the selective loss of dopaminergic neurons and the accumulation of protein aggregates called Lewy bodies (LBs). The unequivocal identification of Mendelian inherited mutations in 13 genes in PD has provided transforming insights into the pathogenesis of this disease. The mechanistic analysis of several PD genes, including α-synuclein (α-syn), leucine-rich repeat kinase 2 (LRRK2), PTEN-induced kinase 1 (PINK1), and Parkin, has revealed central roles for protein aggregation, mitochondrial damage, and defects in endolysosomal trafficking in PD neurodegeneration. In this review, we outline recent advances in our understanding of these gene pathways with a focus on the emergent role of Rab (Ras analog in brain) GTPases and vesicular trafficking as a common mechanism that underpins how mutations in PD genes lead to neuronal loss. These advances have led to previously distinct genes such as vacuolar protein-sorting-associated protein 35 (VPS35) and LRRK2 being implicated in a common signaling pathway. A greater understanding of these common nodes of vesicular trafficking will be crucial for linking other PD genes and improving patient stratification in clinical trials underway against α-syn and LRRK2 targets.


Subject(s)
Parkinson Disease/metabolism , Animals , Autophagy , Cytoplasmic Vesicles/metabolism , Humans , Mitochondria/metabolism , Parkinson Disease/genetics , Protein Aggregates , Protein Transport
3.
Mol Cell ; 83(10): 1693-1709.e9, 2023 05 18.
Article in English | MEDLINE | ID: mdl-37207627

ABSTRACT

Cargo sequestration is a fundamental step of selective autophagy in which cells generate a double-membrane structure termed an "autophagosome" on the surface of cargoes. NDP52, TAX1BP1, and p62 bind FIP200, which recruits the ULK1/2 complex to initiate autophagosome formation on cargoes. How OPTN initiates autophagosome formation during selective autophagy remains unknown despite its importance in neurodegeneration. Here, we uncover an unconventional path of PINK1/Parkin mitophagy initiation by OPTN that does not begin with FIP200 binding or require the ULK1/2 kinases. Using gene-edited cell lines and in vitro reconstitutions, we show that OPTN utilizes the kinase TBK1, which binds directly to the class III phosphatidylinositol 3-kinase complex I to initiate mitophagy. During NDP52 mitophagy initiation, TBK1 is functionally redundant with ULK1/2, classifying TBK1's role as a selective autophagy-initiating kinase. Overall, this work reveals that OPTN mitophagy initiation is mechanistically distinct and highlights the mechanistic plasticity of selective autophagy pathways.


Subject(s)
Mitophagy , Ubiquitin-Protein Ligases , Ubiquitin-Protein Ligases/genetics , Ubiquitin-Protein Ligases/metabolism , Autophagosomes/metabolism , Apoptosis Regulatory Proteins , Protein Kinases/genetics , Protein Kinases/metabolism , Autophagy
4.
Mol Cell ; 83(19): 3404-3420, 2023 Oct 05.
Article in English | MEDLINE | ID: mdl-37708893

ABSTRACT

Mitochondria are central hubs of cellular metabolism that also play key roles in signaling and disease. It is therefore fundamentally important that mitochondrial quality and activity are tightly regulated. Mitochondrial degradation pathways contribute to quality control of mitochondrial networks and can also regulate the metabolic profile of mitochondria to ensure cellular homeostasis. Here, we cover the many and varied ways in which cells degrade or remove their unwanted mitochondria, ranging from mitophagy to mitochondrial extrusion. The molecular signals driving these varied pathways are discussed, including the cellular and physiological contexts under which the different degradation pathways are engaged.

5.
Mol Cell ; 83(17): 3188-3204.e7, 2023 09 07.
Article in English | MEDLINE | ID: mdl-37683611

ABSTRACT

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


Subject(s)
NF-kappa B , Signal Transduction , NF-kappa B/genetics , I-kappa B Kinase/genetics , Protein Serine-Threonine Kinases/genetics , Mitochondria/genetics
6.
Mol Cell ; 82(1): 44-59.e6, 2022 01 06.
Article in English | MEDLINE | ID: mdl-34875213

ABSTRACT

Mutations in PINK1 cause autosomal-recessive Parkinson's disease. Mitochondrial damage results in PINK1 import arrest on the translocase of the outer mitochondrial membrane (TOM) complex, resulting in the activation of its ubiquitin kinase activity by autophosphorylation and initiation of Parkin-dependent mitochondrial clearance. Herein, we report crystal structures of the entire cytosolic domain of insect PINK1. Our structures reveal a dimeric autophosphorylation complex targeting phosphorylation at the invariant Ser205 (human Ser228). The dimer interface requires insert 2, which is unique to PINK1. The structures also reveal how an N-terminal helix binds to the C-terminal extension and provide insights into stabilization of PINK1 on the core TOM complex.


Subject(s)
Insect Proteins/metabolism , Mitochondria/enzymology , Mitochondrial Precursor Protein Import Complex Proteins/metabolism , Protein Kinases/metabolism , Tribolium/enzymology , Animals , Cell Line, Tumor , Enzyme Activation , Enzyme Stability , Humans , Insect Proteins/genetics , Kinetics , Mitochondria/genetics , Mitochondrial Precursor Protein Import Complex Proteins/genetics , Molecular Docking Simulation , Mutation , Phosphorylation , Protein Interaction Domains and Motifs , Protein Kinases/genetics , Structure-Activity Relationship , Tribolium/genetics
7.
Physiol Rev ; 102(4): 1721-1755, 2022 10 01.
Article in English | MEDLINE | ID: mdl-35466694

ABSTRACT

As a central hub for cellular metabolism and intracellular signaling, the mitochondrion is a pivotal organelle, dysfunction of which has been linked to several human diseases including neurodegenerative disorders and in particular Parkinson's disease. An inherent challenge that mitochondria face is the continuous exposure to diverse stresses that increase their likelihood of dysregulation. In response, eukaryotic cells have evolved sophisticated quality control mechanisms to monitor, identify, repair, and/or eliminate abnormal or misfolded proteins within the mitochondrion and/or the dysfunctional mitochondrion itself. Chaperones identify unstable or otherwise abnormal conformations in mitochondrial proteins and can promote their refolding to recover their correct conformation and stability. However, if repair is not possible, the abnormal protein is selectively degraded to prevent potentially damaging interactions with other proteins or its oligomerization into toxic multimeric complexes. The autophagic-lysosomal system and the ubiquitin-proteasome system mediate the selective and targeted degradation of such abnormal or misfolded protein species. Mitophagy (a specific kind of autophagy) mediates the selective elimination of dysfunctional mitochondria, to prevent the deleterious effects of the dysfunctional organelles within the cell. Despite our increasing understanding of the molecular responses toward dysfunctional mitochondria, many key aspects remain relatively poorly understood. Here, we review the emerging mechanisms of mitochondrial quality control including quality control strategies coupled to mitochondrial import mechanisms. In addition, we review the molecular mechanisms regulating mitophagy, with an emphasis on the regulation of PINK1/Parkin-mediated mitophagy in cellular physiology and in the context of Parkinson's disease cell biology.


Subject(s)
Parkinson Disease , Autophagy , Humans , Mitochondria/metabolism , Mitophagy/physiology , Parkinson Disease/metabolism , Protein Kinases/metabolism , Protein Kinases/pharmacology
8.
Mol Cell ; 81(9): 2013-2030.e9, 2021 05 06.
Article in English | MEDLINE | ID: mdl-33773106

ABSTRACT

The sequestration of damaged mitochondria within double-membrane structures termed autophagosomes is a key step of PINK1/Parkin mitophagy. The ATG4 family of proteases are thought to regulate autophagosome formation exclusively by processing the ubiquitin-like ATG8 family (LC3/GABARAPs). We discover that human ATG4s promote autophagosome formation independently of their protease activity and of ATG8 family processing. ATG4 proximity networks reveal a role for ATG4s and their proximity partners, including the immune-disease protein LRBA, in ATG9A vesicle trafficking to mitochondria. Artificial intelligence-directed 3D electron microscopy of phagophores shows that ATG4s promote phagophore-ER contacts during the lipid-transfer phase of autophagosome formation. We also show that ATG8 removal during autophagosome maturation does not depend on ATG4 activity. Instead, ATG4s can disassemble ATG8-protein conjugates, revealing a role for ATG4s as deubiquitinating-like enzymes. These findings establish non-canonical roles of the ATG4 family beyond the ATG8 lipidation axis and provide an AI-driven framework for rapid 3D electron microscopy.


Subject(s)
Apoptosis Regulatory Proteins/metabolism , Autophagosomes/metabolism , Autophagy-Related Proteins/metabolism , Cysteine Endopeptidases/metabolism , Lipid Metabolism , Microtubule-Associated Proteins/metabolism , Mitochondria/metabolism , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/metabolism , Apoptosis Regulatory Proteins/genetics , Artificial Intelligence , Autophagosomes/genetics , Autophagosomes/ultrastructure , Autophagy-Related Protein 8 Family/genetics , Autophagy-Related Protein 8 Family/metabolism , Autophagy-Related Proteins/genetics , Cysteine Endopeptidases/genetics , HEK293 Cells , HeLa Cells , Humans , Imaging, Three-Dimensional , Membrane Proteins/genetics , Membrane Proteins/metabolism , Microscopy, Electron, Transmission , Microtubule-Associated Proteins/genetics , Mitochondria/genetics , Mitochondria/ultrastructure , Mitophagy , Protein Kinases/genetics , Protein Kinases/metabolism , Protein Transport , Signal Transduction , Ubiquitin-Protein Ligases/genetics , Ubiquitin-Protein Ligases/metabolism , Vesicular Transport Proteins/genetics , Vesicular Transport Proteins/metabolism
9.
EMBO J ; 43(5): 754-779, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38287189

ABSTRACT

Tank-binding kinase 1 (TBK1) is a Ser/Thr kinase that is involved in many intracellular processes, such as innate immunity, cell cycle, and apoptosis. TBK1 is also important for phosphorylating the autophagy adaptors that mediate the selective autophagic removal of damaged mitochondria. However, the mechanism by which PINK1-Parkin-mediated mitophagy activates TBK1 remains largely unknown. Here, we show that the autophagy adaptor optineurin (OPTN) provides a unique platform for TBK1 activation. Both the OPTN-ubiquitin and the OPTN-pre-autophagosomal structure (PAS) interaction axes facilitate assembly of the OPTN-TBK1 complex at a contact sites between damaged mitochondria and the autophagosome formation sites. At this assembly point, a positive feedback loop for TBK1 activation is initiated that accelerates hetero-autophosphorylation of the protein. Expression of monobodies engineered here to bind OPTN impaired OPTN accumulation at contact sites, as well as the subsequent activation of TBK1, thereby inhibiting mitochondrial degradation. Taken together, these data show that a positive and reciprocal relationship between OPTN and TBK1 initiates autophagosome biogenesis on damaged mitochondria.


Subject(s)
Cell Cycle Proteins , Membrane Transport Proteins , Mitochondrial Membranes , Mitophagy , Humans , Autophagy/physiology , Cell Cycle Proteins/metabolism , HeLa Cells , Membrane Transport Proteins/metabolism , Mitochondria/metabolism , Mitochondrial Membranes/metabolism , Protein Serine-Threonine Kinases/metabolism
10.
Mol Cell ; 77(5): 1124-1142.e10, 2020 03 05.
Article in English | MEDLINE | ID: mdl-32142685

ABSTRACT

The ubiquitin ligase Parkin, protein kinase PINK1, USP30 deubiquitylase, and p97 segregase function together to regulate turnover of damaged mitochondria via mitophagy, but our mechanistic understanding in neurons is limited. Here, we combine induced neurons (iNeurons) derived from embryonic stem cells with quantitative proteomics to reveal the dynamics and specificity of Parkin-dependent ubiquitylation under endogenous expression conditions. Targets showing elevated ubiquitylation in USP30-/- iNeurons are concentrated in components of the mitochondrial translocon, and the ubiquitylation kinetics of the vast majority of Parkin targets are unaffected, correlating with a modest kinetic acceleration in accumulation of pS65-Ub and mitophagic flux upon mitochondrial depolarization without USP30. Basally, ubiquitylated translocon import substrates accumulate, suggesting a quality control function for USP30. p97 was dispensable for Parkin ligase activity in iNeurons. This work provides an unprecedented quantitative landscape of the Parkin-modified ubiquitylome in iNeurons and reveals the underlying specificity of central regulatory elements in the pathway.


Subject(s)
Human Embryonic Stem Cells/enzymology , Mitochondria/enzymology , Mitochondrial Proteins/metabolism , Mitophagy , Neural Stem Cells/enzymology , Neurogenesis , Neurons/enzymology , Thiolester Hydrolases/metabolism , Ubiquitin-Protein Ligases/metabolism , HeLa Cells , Human Embryonic Stem Cells/pathology , Humans , Kinetics , Mitochondria/genetics , Mitochondria/pathology , Mitochondrial Proteins/genetics , Neural Stem Cells/pathology , Neurons/pathology , Phosphorylation , Protein Kinases/genetics , Protein Kinases/metabolism , Proteomics , Signal Transduction , Thiolester Hydrolases/genetics , Ubiquitin-Protein Ligases/genetics , Ubiquitination , Valosin Containing Protein/genetics , Valosin Containing Protein/metabolism
11.
Mol Cell ; 80(4): 607-620.e12, 2020 11 19.
Article in English | MEDLINE | ID: mdl-33113344

ABSTRACT

Aberrant mitophagy has been implicated in a broad spectrum of disorders. PINK1, Parkin, and ubiquitin have pivotal roles in priming mitophagy. However, the entire regulatory landscape and the precise control mechanisms of mitophagy remain to be elucidated. Here, we uncover fundamental mitophagy regulation involving PINK1 and a non-canonical role of the mitochondrial Tu translation elongation factor (TUFm). The mitochondrion-cytosol dual-localized TUFm interacts with PINK1 biochemically and genetically, which is an evolutionarily conserved Parkin-independent route toward mitophagy. A PINK1-dependent TUFm phosphoswitch at Ser222 determines conversion from activating to suppressing mitophagy. PINK1 modulates differential translocation of TUFm because p-S222-TUFm is restricted predominantly to the cytosol, where it inhibits mitophagy by impeding Atg5-Atg12 formation. The self-antagonizing feature of PINK1/TUFm is critical for the robustness of mitophagy regulation, achieved by the unique kinetic parameters of p-S222-TUFm, p-S65-ubiquitin, and their common kinase PINK1. Our findings provide new mechanistic insights into mitophagy and mitophagy-associated disorders.


Subject(s)
Drosophila melanogaster/growth & development , Mitochondria/pathology , Mitochondrial Proteins/metabolism , Mitophagy , Peptide Elongation Factor Tu/metabolism , Protein Kinases/metabolism , Animals , Cytosol/metabolism , Drosophila melanogaster/genetics , Drosophila melanogaster/metabolism , Female , HeLa Cells , Humans , Male , Mitochondria/genetics , Mitochondria/metabolism , Mitochondrial Proteins/genetics , Peptide Elongation Factor Tu/genetics , Phosphorylation , Protein Interaction Domains and Motifs , Protein Kinases/genetics , Protein Transport , Ubiquitin-Protein Ligases/genetics , Ubiquitin-Protein Ligases/metabolism
12.
Mol Cell ; 74(2): 347-362.e6, 2019 04 18.
Article in English | MEDLINE | ID: mdl-30853401

ABSTRACT

Selective autophagy recycles damaged organelles and clears intracellular pathogens to prevent their aberrant accumulation. How ULK1 kinase is targeted and activated during selective autophagic events remains to be elucidated. In this study, we used chemically inducible dimerization (CID) assays in tandem with CRISPR KO lines to systematically analyze the molecular basis of selective autophagosome biogenesis. We demonstrate that ectopic placement of NDP52 on mitochondria or peroxisomes is sufficient to initiate selective autophagy by focally localizing and activating the ULK1 complex. The capability of NDP52 to induce mitophagy is dependent on its interaction with the FIP200/ULK1 complex, which is facilitated by TBK1. Ectopically tethering ULK1 to cargo bypasses the requirement for autophagy receptors and TBK1. Focal activation of ULK1 occurs independently of AMPK and mTOR. Our findings provide a parsimonious model of selective autophagy, which highlights the coordination of ULK1 complex localization by autophagy receptors and TBK1 as principal drivers of targeted autophagosome biogenesis.


Subject(s)
Autophagy-Related Protein-1 Homolog/genetics , Autophagy/genetics , Nuclear Proteins/genetics , Protein Serine-Threonine Kinases/genetics , AMP-Activated Protein Kinase Kinases , Autophagy-Related Proteins , Clustered Regularly Interspaced Short Palindromic Repeats/genetics , HeLa Cells , Humans , Mitochondria/chemistry , Mitochondria/genetics , Multiprotein Complexes/chemistry , Multiprotein Complexes/genetics , Peroxisomes/chemistry , Peroxisomes/genetics , Phosphorylation , Protein Kinases/genetics , Protein Multimerization , Protein-Tyrosine Kinases/chemistry , Protein-Tyrosine Kinases/genetics , Signal Transduction/genetics , TOR Serine-Threonine Kinases/genetics
13.
Mol Cell ; 75(4): 835-848.e8, 2019 08 22.
Article in English | MEDLINE | ID: mdl-31378462

ABSTRACT

Mitochondrial dysfunction and proteostasis failure frequently coexist as hallmarks of neurodegenerative disease. How these pathologies are related is not well understood. Here, we describe a phenomenon termed MISTERMINATE (mitochondrial-stress-induced translational termination impairment and protein carboxyl terminal extension), which mechanistically links mitochondrial dysfunction with proteostasis failure. We show that mitochondrial dysfunction impairs translational termination of nuclear-encoded mitochondrial mRNAs, including complex-I 30kD subunit (C-I30) mRNA, occurring on the mitochondrial surface in Drosophila and mammalian cells. Ribosomes stalled at the normal stop codon continue to add to the C terminus of C-I30 certain amino acids non-coded by mRNA template. C-terminally extended C-I30 is toxic when assembled into C-I and forms aggregates in the cytosol. Enhancing co-translational quality control prevents C-I30 C-terminal extension and rescues mitochondrial and neuromuscular degeneration in a Parkinson's disease model. These findings emphasize the importance of efficient translation termination and reveal unexpected link between mitochondrial health and proteome homeostasis mediated by MISTERMINATE.


Subject(s)
Codon, Terminator , Drosophila Proteins/metabolism , Mitochondria/metabolism , Mitochondrial Diseases/metabolism , Mitochondrial Proteins/metabolism , Proteostasis Deficiencies/metabolism , Animals , Drosophila Proteins/genetics , Drosophila melanogaster , HeLa Cells , Humans , Mitochondria/genetics , Mitochondria/pathology , Mitochondrial Diseases/genetics , Mitochondrial Diseases/pathology , Mitochondrial Proteins/genetics , Proteostasis Deficiencies/genetics , Proteostasis Deficiencies/pathology , RNA, Mitochondrial/genetics , RNA, Mitochondrial/metabolism
14.
Proc Natl Acad Sci U S A ; 121(32): e2403114121, 2024 Aug 06.
Article in English | MEDLINE | ID: mdl-39078678

ABSTRACT

Parkin is an E3 ubiquitin ligase implicated in early-onset forms of Parkinson's disease. It catalyzes a transthiolation reaction by accepting ubiquitin (Ub) from an E2 conjugating enzyme, forming a short-lived thioester intermediate, and transfers Ub to mitochondrial membrane substrates to signal mitophagy. A major impediment to the development of Parkinsonism therapeutics is the lack of structural and mechanistic detail for the essential, short-lived transthiolation intermediate. It is not known how Ub is recognized by the catalytic Rcat domain in parkin that enables Ub transfer from an E2~Ub conjugate to the catalytic site and the structure of the transthiolation complex is undetermined. Here, we capture the catalytic intermediate for the Rcat domain of parkin in complex with ubiquitin (Rcat-Ub) and determine its structure using NMR-based chemical shift perturbation experiments. We show that a previously unidentified α-helical region near the Rcat domain is unmasked as a recognition motif for Ub and guides the C-terminus of Ub toward the parkin catalytic site. Further, we apply a combination of guided AlphaFold modeling, chemical cross-linking, and single turnover assays to establish and validate a model of full-length parkin in complex with UbcH7, its donor Ub, and phosphoubiquitin, trapped in the process of transthiolation. Identification of this catalytic intermediate and orientation of Ub with respect to the Rcat domain provides important structural insights into Ub transfer by this E3 ligase and explains how the previously enigmatic Parkinson's pathogenic mutation T415N alters parkin activity.


Subject(s)
Ubiquitin-Protein Ligases , Ubiquitination , Ubiquitin-Protein Ligases/metabolism , Ubiquitin-Protein Ligases/genetics , Humans , Catalytic Domain , Ubiquitin/metabolism , Ubiquitin-Conjugating Enzymes/metabolism , Ubiquitin-Conjugating Enzymes/genetics , Parkinson Disease/metabolism , Parkinson Disease/genetics , Models, Molecular
15.
Mol Cell ; 70(2): 211-227.e8, 2018 04 19.
Article in English | MEDLINE | ID: mdl-29656925

ABSTRACT

Flux through kinase and ubiquitin-driven signaling systems depends on the modification kinetics, stoichiometry, primary site specificity, and target abundance within the pathway, yet we rarely understand these parameters and their spatial organization within cells. Here we develop temporal digital snapshots of ubiquitin signaling on the mitochondrial outer membrane in embryonic stem cell-derived neurons, and we model HeLa cell systems upon activation of the PINK1 kinase and PARKIN ubiquitin ligase by proteomic counting of ubiquitylation and phosphorylation events. We define the kinetics and site specificity of PARKIN-dependent target ubiquitylation, and we demonstrate the power of this approach to quantify pathway modulators and to mechanistically define the role of PARKIN UBL phosphorylation in pathway activation in induced neurons. Finally, through modulation of pS65-Ub on mitochondria, we demonstrate that Ub hyper-phosphorylation is inhibitory to mitophagy receptor recruitment, indicating that pS65-Ub stoichiometry in vivo is optimized to coordinate PARKIN recruitment via pS65-Ub and mitophagy receptors via unphosphorylated chains.


Subject(s)
Human Embryonic Stem Cells/enzymology , Mitochondrial Membranes/enzymology , Neural Stem Cells/enzymology , Neurogenesis , Neurons/enzymology , Proteomics/methods , Ubiquitin-Protein Ligases/metabolism , Enzyme Activation , HeLa Cells , Humans , Kinetics , Mitophagy , Phenotype , Phosphorylation , Protein Kinases/genetics , Protein Kinases/metabolism , Signal Transduction , Ubiquitin-Protein Ligases/chemistry , Ubiquitin-Protein Ligases/genetics , Ubiquitination , Voltage-Dependent Anion Channel 1/genetics , Voltage-Dependent Anion Channel 1/metabolism
16.
Mol Cell ; 70(6): 1008-1024.e6, 2018 06 21.
Article in English | MEDLINE | ID: mdl-29932898

ABSTRACT

UBE2M and UBE2F are two family members of neddylation E2 conjugating enzyme that, together with E3s, activate CRLs (Cullin-RING Ligases) by catalyzing cullin neddylation. However, whether and how two E2s cross-talk with each other are largely unknown. Here, we report that UBE2M is a stress-inducible gene subjected to cis-transactivation by HIF-1 and AP1, and MLN4924, a small molecule inhibitor of E1 NEDD8-activating enzyme (NAE), upregulates UBE2M via blocking degradation of HIF-1α and c-JUN. UBE2M is a dual E2 for targeted ubiquitylation and degradation of UBE2F, acting as a neddylation E2 to activate CUL3-Keap1 E3 under physiological conditions but as a ubiquitylation E2 for Parkin-DJ-1 E3 under stressed conditions. UBE2M-induced UBE2F degradation leads to CRL5 inactivation and subsequent NOXA accumulation to suppress the growth of lung cancer cells. Collectively, our study establishes a negative regulatory axis between two neddylation E2s with UBE2M ubiquitylating UBE2F, and two CRLs with CRL3 inactivating CRL5.


Subject(s)
Ubiquitin-Conjugating Enzymes/metabolism , Animals , Cell Line , Cell Line, Tumor , Cullin Proteins/metabolism , Cyclopentanes/pharmacology , Female , HEK293 Cells , Humans , Mice , Mice, Inbred BALB C , Mice, Nude , Pyrimidines/pharmacology , Stress, Physiological/physiology , Ubiquitin-Activating Enzymes/antagonists & inhibitors , Ubiquitin-Activating Enzymes/metabolism , Ubiquitin-Conjugating Enzymes/biosynthesis , Ubiquitin-Protein Ligases/metabolism , Ubiquitination , Ubiquitins/metabolism
17.
Trends Biochem Sci ; 46(4): 329-343, 2021 04.
Article in English | MEDLINE | ID: mdl-33323315

ABSTRACT

Mitochondrial dysfunction has been associated with neurodegeneration in Parkinson's disease (PD) for over 30 years. Despite this, the role of mitochondrial dysfunction as an initiator, propagator, or bystander remains undetermined. The discovery of the role of the PD familial genes PTEN-induced putative kinase 1 (PINK1) and parkin (PRKN) in mediating mitochondrial degradation (mitophagy) reaffirmed the importance of this process in PD aetiology. Recently, progress has been made in understanding the upstream and downstream regulators of canonical PINK1/parkin-mediated mitophagy, alongside noncanonical PINK1/parkin mitophagy, in response to mitochondrial damage. Progress has also been made in understanding the role of PD-associated genes, such as SNCA, LRRK2, and CHCHD2, in mitochondrial dysfunction and their overlap with sporadic PD (sPD), opening opportunities for therapeutically targeting mitochondria in PD.


Subject(s)
Mitochondria/pathology , Mitophagy , Parkinson Disease , DNA-Binding Proteins , Humans , Leucine-Rich Repeat Serine-Threonine Protein Kinase-2 , Parkinson Disease/drug therapy , Protein Kinases , Transcription Factors , Ubiquitin-Protein Ligases , alpha-Synuclein
18.
J Biol Chem ; 300(8): 107543, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38992440

ABSTRACT

The pathogenesis of Parkinson's disease (PD) has been associated with mitochondrial dysfunction. Given that the PINK1/Parkin pathway governs mitochondrial quality control by inducing mitophagy to remove damaged mitochondria, therapeutic approaches to activate PINK1/Parkin-mediated mitophagy have the potential in the treatment of PD. Here, we have identified a new small molecule, BL-918, as an inducer of mitophagy via activating the PINK1/Parkin pathway. BL-918 triggers PINK1 accumulation and Parkin mitochondrial translocation to initiate PINK1/Parkin-mediated mitophagy. We found that mitochondrial membrane potential and mitochondrial permeability transition pore were involved in BL-918-induced PINK1/Parkin pathway activation. Moreover, we showed that BL-918 mitigated PD progression in MPTP-induced PD mice in a PINK1-dependent manner. Our results unravel a new activator of the PINK1/Parkin signaling pathway and provide a potential strategy for the treatment of PD and other diseases with dysfunctional mitochondria.


Subject(s)
Mitochondria , Mitophagy , Parkinson Disease , Protein Kinases , Signal Transduction , Ubiquitin-Protein Ligases , Ubiquitin-Protein Ligases/metabolism , Ubiquitin-Protein Ligases/genetics , Protein Kinases/metabolism , Protein Kinases/genetics , Animals , Mice , Mitophagy/drug effects , Humans , Parkinson Disease/metabolism , Parkinson Disease/pathology , Parkinson Disease/genetics , Mitochondria/metabolism , Membrane Potential, Mitochondrial/drug effects , Mice, Inbred C57BL , Male , Disease Progression , Phenylacetates
19.
J Cell Sci ; 136(13)2023 07 01.
Article in English | MEDLINE | ID: mdl-37313742

ABSTRACT

MTP18 (also known as MTFP1), an inner mitochondrial membrane protein, plays a vital role in maintaining mitochondrial morphology by regulating mitochondrial fission. Here, we found that MTP18 functions as a mitophagy receptor that targets dysfunctional mitochondria into autophagosomes for elimination. Interestingly, MTP18 interacts with members of the LC3 (also known as MAP1LC3) family through its LC3-interacting region (LIR) to induce mitochondrial autophagy. Mutation in the LIR motif (mLIR) inhibited that interaction, thus suppressing mitophagy. Moreover, Parkin or PINK1 deficiency abrogated mitophagy in MTP18-overexpressing human oral cancer-derived FaDu cells. Upon exposure to the mitochondrial oxidative phosphorylation uncoupler CCCP, MTP18[mLIR]-FaDu cells showed decreased TOM20 levels without affecting COX IV levels. Conversely, loss of Parkin or PINK1 resulted in inhibition of TOM20 and COX IV degradation in MTP18[mLIR]-FaDu cells exposed to CCCP, establishing Parkin-mediated proteasomal degradation of outer mitochondrial membrane as essential for effective mitophagy. We also found that MTP18 provides a survival advantage to oral cancer cells exposed to cellular stress and that inhibition of MTP18-dependent mitophagy induced cell death in oral cancer cells. These findings demonstrate that MTP18 is a novel mitophagy receptor and that MTP18-dependent mitophagy has pathophysiologic implications for oral cancer progression, indicating inhibition of MTP18-mitophagy could thus be a promising cancer therapy strategy.


Subject(s)
Mitochondrial Membranes , Mouth Neoplasms , Humans , Apoptosis/genetics , Carbonyl Cyanide m-Chlorophenyl Hydrazone/metabolism , Carbonyl Cyanide m-Chlorophenyl Hydrazone/pharmacology , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mitochondrial Dynamics , Mitochondrial Membranes/metabolism , Mitophagy/genetics , Mouth Neoplasms/genetics , Mouth Neoplasms/metabolism , Protein Kinases/genetics , Protein Kinases/metabolism , Ubiquitin-Protein Ligases/genetics , Ubiquitin-Protein Ligases/metabolism
20.
EMBO Rep ; 24(9): e55859, 2023 09 06.
Article in English | MEDLINE | ID: mdl-37501540

ABSTRACT

Amyotrophic lateral sclerosis (ALS) and frontotemporal dementia (FTD) are two aging-related neurodegenerative diseases that share common key features, including aggregation of pathogenic proteins, dysfunction of mitochondria, and impairment of autophagy. Mutations in ubiquilin 2 (UBQLN2), a shuttle protein in the ubiquitin-proteasome system (UPS), can cause ALS/FTD, but the mechanism underlying UBQLN2-mediated pathogenesis is still uncertain. Recent studies indicate that mitophagy, a selective form of autophagy which is crucial for mitochondrial quality control, is tightly associated with neurodegenerative diseases including Alzheimer's disease, Parkinson's disease, and ALS. In this study, we show that after Parkin-dependent ubiquitination of damaged mitochondria, UBQLN2 is recruited to poly-ubiquitinated mitochondria through the UBA domain. UBQLN2 cooperates with the chaperone HSP70 to promote UPS-driven degradation of outer mitochondrial membrane (OMM) proteins. The resulting rupture of the OMM triggers the autophagosomal recognition of the inner mitochondrial membrane receptor PHB2. UBQLN2 is required for Parkin-mediated mitophagy and neuronal survival upon mitochondrial damage, and the ALS/FTD pathogenic mutations in UBQLN2 impair mitophagy in primary cultured neurons. Taken together, our findings link dysfunctional mitophagy to UBQLN2-mediated neurodegeneration.


Subject(s)
Amyotrophic Lateral Sclerosis , Frontotemporal Dementia , Neurodegenerative Diseases , Humans , Mitochondrial Membranes/metabolism , Amyotrophic Lateral Sclerosis/genetics , Mitophagy , Frontotemporal Dementia/genetics , Adaptor Proteins, Signal Transducing/genetics , Autophagy-Related Proteins/genetics , Proteasome Endopeptidase Complex/metabolism , Ubiquitin/metabolism , Neurodegenerative Diseases/metabolism , Transcription Factors/metabolism , Ubiquitin-Protein Ligases/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL