Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 1.083
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 ; 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
6.
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
7.
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
8.
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
9.
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
10.
Mol Cell ; 73(6): 1127-1137.e5, 2019 03 21.
Article in English | MEDLINE | ID: mdl-30772175

ABSTRACT

We have previously proposed that selective inheritance, the limited transmission of damaging mtDNA mutations from mother to offspring, is based on replication competition in Drosophila melanogaster. This model, which stems from our observation that wild-type mitochondria propagate much more vigorously in the fly ovary than mitochondria carrying fitness-impairing mutations, implies that germ cells recognize the fitness of individual mitochondria and selectively boost the propagation of healthy ones. Here, we demonstrate that the protein kinase PINK1 preferentially accumulates on mitochondria enriched for a deleterious mtDNA mutation. PINK1 phosphorylates Larp to inhibit protein synthesis on the mitochondrial outer membrane. Impaired local translation on defective mitochondria in turn limits the replication of their mtDNA and hence the transmission of deleterious mutations to the offspring. Our work confirms that selective inheritance occurs at the organelle level during Drosophila oogenesis and provides molecular entry points to test this model in other systems.


Subject(s)
DNA Replication , DNA, Mitochondrial/genetics , Drosophila Proteins/metabolism , Drosophila melanogaster/enzymology , Mitochondria/enzymology , Mitochondrial Membranes/enzymology , Mitochondrial Proteins/biosynthesis , Mutation , Oocytes/enzymology , Protein Serine-Threonine Kinases/metabolism , Animals , Animals, Genetically Modified , DNA, Mitochondrial/biosynthesis , Drosophila Proteins/genetics , Drosophila melanogaster/genetics , Female , Inheritance Patterns , Mitochondria/genetics , Mitochondrial Proteins/genetics , Oogenesis , Organelle Biogenesis , Phosphorylation , Protein Serine-Threonine Kinases/genetics , Protein Stability , Transcription Factors/genetics , Transcription Factors/metabolism
11.
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
12.
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
13.
Proc Natl Acad Sci U S A ; 121(10): e2313540121, 2024 Mar 05.
Article in English | MEDLINE | ID: mdl-38416681

ABSTRACT

Mutations in PTEN-induced putative kinase 1 (PINK1) cause autosomal recessive early-onset Parkinson's disease (PD). PINK1 is a Ser/Thr kinase that regulates mitochondrial quality control by triggering mitophagy mediated by the ubiquitin (Ub) ligase Parkin. Upon mitochondrial damage, PINK1 accumulates on the outer mitochondrial membrane forming a high-molecular-weight complex with the translocase of the outer membrane (TOM). PINK1 then phosphorylates Ub, which enables recruitment and activation of Parkin followed by autophagic clearance of the damaged mitochondrion. Thus, Parkin-dependent mitophagy hinges on the stable accumulation of PINK1 on the TOM complex. Yet, the mechanism linking mitochondrial stressors to PINK1 accumulation and whether the translocases of the inner membrane (TIMs) are also involved remain unclear. Herein, we demonstrate that mitochondrial stress induces the formation of a PINK1-TOM-TIM23 supercomplex in human cultured cell lines, dopamine neurons, and midbrain organoids. Moreover, we show that PINK1 is required to stably tether the TOM to TIM23 complexes in response to stress such that the supercomplex fails to accumulate in cells lacking PINK1. This tethering is dependent on an interaction between the PINK1 N-terminal-C-terminal extension module and the cytosolic domain of the Tom20 subunit of the TOM complex, the disruption of which, by either designer or PD-associated PINK1 mutations, inhibits downstream mitophagy. Together, the findings provide key insight into how PINK1 interfaces with the mitochondrial import machinery, with important implications for the mechanisms of mitochondrial quality control and PD pathogenesis.


Subject(s)
Mitochondrial Precursor Protein Import Complex Proteins , Protein Kinases , Humans , Carrier Proteins/metabolism , Mitochondria/metabolism , Phosphorylation , Protein Kinases/genetics , Protein Kinases/metabolism , Ubiquitin/metabolism , Ubiquitin-Protein Ligases/genetics , Ubiquitin-Protein Ligases/metabolism
14.
EMBO J ; 41(24): e112006, 2022 12 15.
Article in English | MEDLINE | ID: mdl-36398858

ABSTRACT

Mitochondria are increasingly recognized as cellular hubs to orchestrate signaling pathways that regulate metabolism, redox homeostasis, and cell fate decisions. Recent research revealed a role of mitochondria also in innate immune signaling; however, the mechanisms of how mitochondria affect signal transduction are poorly understood. Here, we show that the NF-κB pathway activated by TNF employs mitochondria as a platform for signal amplification and shuttling of activated NF-κB to the nucleus. TNF treatment induces the recruitment of HOIP, the catalytic component of the linear ubiquitin chain assembly complex (LUBAC), and its substrate NEMO to the outer mitochondrial membrane, where M1- and K63-linked ubiquitin chains are generated. NF-κB is locally activated and transported to the nucleus by mitochondria, leading to an increase in mitochondria-nucleus contact sites in a HOIP-dependent manner. Notably, TNF-induced stabilization of the mitochondrial kinase PINK1 furthermore contributes to signal amplification by antagonizing the M1-ubiquitin-specific deubiquitinase OTULIN. Overall, our study reveals a role for mitochondria in amplifying TNF-mediated NF-κB activation, both serving as a signaling platform, as well as a transport mode for activated NF-κB to the nuclear.


Subject(s)
NF-kappa B , Ubiquitin , NF-kappa B/genetics , NF-kappa B/metabolism , Ubiquitin/metabolism , Ubiquitin-Protein Ligases/genetics , Ubiquitin-Protein Ligases/metabolism , Signal Transduction/physiology , Mitochondria/metabolism , Ubiquitination
15.
Mol Cell ; 69(5): 744-756.e6, 2018 03 01.
Article in English | MEDLINE | ID: mdl-29456190

ABSTRACT

Mitochondrial crista structure partitions vital cellular reactions and is precisely regulated by diverse cellular signals. Here, we show that, in Drosophila, mitochondrial cristae undergo dynamic remodeling among distinct subcellular regions and the Parkinson's disease (PD)-linked Ser/Thr kinase PINK1 participates in their regulation. Mitochondria increase crista junctions and numbers in selective subcellular areas, and this remodeling requires PINK1 to phosphorylate the inner mitochondrial membrane protein MIC60/mitofilin, which stabilizes MIC60 oligomerization. Expression of MIC60 restores crista structure and ATP levels of PINK1-null flies and remarkably rescues their behavioral defects and dopaminergic neurodegeneration. In an extension to human relevance, we discover that the PINK1-MIC60 pathway is conserved in human neurons, and expression of several MIC60 coding variants in the mitochondrial targeting sequence found in PD patients in Drosophila impairs crista junction formation and causes locomotion deficits. These findings highlight the importance of maintenance and plasticity of crista junctions to cellular homeostasis in vivo.


Subject(s)
Drosophila Proteins/metabolism , Mitochondrial Membranes/metabolism , Neurons/metabolism , Protein Serine-Threonine Kinases/metabolism , Animals , Drosophila Proteins/genetics , Drosophila melanogaster , Humans , Mitochondrial Membranes/pathology , Mitochondrial Proteins/genetics , Mitochondrial Proteins/metabolism , Muscle Proteins/genetics , Muscle Proteins/metabolism , Neurons/pathology , Parkinson Disease/genetics , Parkinson Disease/metabolism , Parkinson Disease/pathology , Phosphorylation/genetics , Protein Kinases/genetics , Protein Kinases/metabolism , Protein Serine-Threonine Kinases/genetics
16.
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
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 Biol Chem ; 300(4): 107198, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38508312

ABSTRACT

Understanding the mechanisms that govern the stability of functionally crucial proteins is essential for various cellular processes, development, and overall cell viability. Disturbances in protein homeostasis are linked to the pathogenesis of neurodegenerative diseases. PTEN-induced kinase 1 (PINK1), a protein kinase, plays a significant role in mitochondrial quality control and cellular stress response, and its mutated forms lead to early-onset Parkinson's disease. Despite its importance, the specific mechanisms regulating PINK1 protein stability have remained unclear. This study reveals a cytoplasmic interaction between PINK1 and F-box and WD repeat domain-containing 7ß (FBW7ß) in mammalian cells. FBW7ß, a component of the Skp1-Cullin-1-F-box protein complex-type ubiquitin ligase, is instrumental in recognizing substrates. Our findings demonstrate that FBW7ß regulates PINK1 stability through the Skp1-Cullin-1-F-box protein complex and the proteasome pathway. It facilitates the K48-linked polyubiquitination of PINK1, marking it for degradation. When FBW7 is absent, PINK1 accumulates, leading to heightened mitophagy triggered by carbonyl cyanide 3-chlorophenylhydrazone treatment. Moreover, exposure to the toxic compound staurosporine accelerates PINK1 degradation via FBW7ß, correlating with increased cell death. This study unravels the intricate mechanisms controlling PINK1 protein stability and sheds light on the novel role of FBW7ß. These findings deepen our understanding of PINK1-related pathologies and potentially pave the way for therapeutic interventions.


Subject(s)
F-Box-WD Repeat-Containing Protein 7 , Protein Kinases , Proteolysis , Ubiquitination , Humans , F-Box-WD Repeat-Containing Protein 7/metabolism , F-Box-WD Repeat-Containing Protein 7/genetics , HEK293 Cells , Mitophagy , Proteasome Endopeptidase Complex/metabolism , Proteasome Endopeptidase Complex/genetics , Protein Kinases/metabolism , Protein Kinases/genetics , SKP Cullin F-Box Protein Ligases/metabolism , SKP Cullin F-Box Protein Ligases/genetics
20.
Exp Cell Res ; 441(2): 114182, 2024 Aug 15.
Article in English | MEDLINE | ID: mdl-39094903

ABSTRACT

Kawasaki disease (KD) is a systemic vasculitis with an unknown cause that primarily affects children. The objective of this study was to explore the function and underlying mechanism of mitophagy in Mycoplasma pneumoniae (MP)-induced KD. To create MP-induced KD models, Human coronary endothelial cells (HCAECs) and DBA/2 mice were employed and treated with Mp-Lipid-associated membrane proteins (LAMPs). Lactate dehydrogenase (LDH) levels were tested to determine cellular damage or death. The inflammatory cytokines tumor necrosis factor (TNF)--α and interleukin (IL)-6 were measured using the Enzyme-Linked Immunosorbent Assay (ELISA) method. RT-qPCR and Western blotting were used to determine the expression of Intercellular Adhesion Molecule(ICAM)-1, vascular cell adhesion molecule (VCAM)-1, inducible nitric oxide synthase(iNOS), LC3, p62, PINK1(a mitochondrial serine/threonine-protein kinase), and PARKIN(a cytosolic E3-ubiquitin ligase). The adenosine triphosphate (ATP), reactive oxygen species (ROS), and mitochondrial membrane potential(MMP) levels were measured to determine mitochondrial function. Mitophagy was investigated using immunofluorescence and a mitophagy detection test. Autophagosome and mitochondrial morphology were examined using transmission electron microscopy. To identify inflammatory cell infiltration, hematoxylin and eosin staining was utilized. Mp-LAMPs increased the levels of TNF-α, IL-6, ICAM-1, VCAM-1, and iNOS in an HCAEC cell model, along with LDH release. After Mp-LAMPs exposure, there was a rise in LC3 and a reduction in p62. Meanwhile, the expression of PINK1 and Parkin was increased. Cyclosporin A dramatically increased ATP synthesis and MMP in HCAEC cells treated with Mp-LAMPs, while suppressing ROS generation, demonstrating excessive mitophagy-related mitochondrial dysfunction. Additionally, neither body weight nor artery tissue were affected due to PINK1 and Parkin suppression Cyclosporin A in Mp-LAMPs-treated mice. These findings indicated that PINK1/Parkin-mediated mitophagy inhibition may be a therapeutic target for MP-induced KD.


Subject(s)
Mitophagy , Mucocutaneous Lymph Node Syndrome , Mycoplasma pneumoniae , Protein Kinases , Ubiquitin-Protein Ligases , Animals , Mucocutaneous Lymph Node Syndrome/metabolism , Mucocutaneous Lymph Node Syndrome/pathology , Protein Kinases/metabolism , Humans , Mice , Ubiquitin-Protein Ligases/metabolism , Ubiquitin-Protein Ligases/genetics , Mycoplasma pneumoniae/pathogenicity , Mice, Inbred DBA , Endothelial Cells/metabolism , Endothelial Cells/pathology , Pneumonia, Mycoplasma/metabolism , Pneumonia, Mycoplasma/pathology , Pneumonia, Mycoplasma/microbiology , Mitochondria/metabolism , Mitochondria/pathology , Reactive Oxygen Species/metabolism , Membrane Potential, Mitochondrial
SELECTION OF CITATIONS
SEARCH DETAIL