Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 136
Filter
1.
Mol Cell ; 84(6): 995-997, 2024 Mar 21.
Article in English | MEDLINE | ID: mdl-38518749

ABSTRACT

Chakrabarty et al.1 demonstrate that phospho-EIF2α (pEIF2α), the translation initiation factor that mediates the integrated stress response (ISR), is necessary and sufficient for the autophagic degradation of mitochondria following the addition of mitochondrial stressors.


Subject(s)
Mitochondria , Stress, Physiological , Phosphorylation , Mitochondria/metabolism , Eukaryotic Initiation Factor-2/genetics , Eukaryotic Initiation Factor-2/metabolism
2.
bioRxiv ; 2023 Oct 16.
Article in English | MEDLINE | ID: mdl-37873486

ABSTRACT

STING activation by cyclic dinucleotides in mammals induces interferon- and NFκB -related gene expression, and the lipidation of LC3B at Golgi membranes. While mechanisms of the interferon response are well understood, the mechanisms of NFκB activation mediated by STING remain unclear. We report that STING activation induces K63- and M1-linked/linear ubiquitin chain formation at LC3B-associated Golgi membranes. Loss of the LUBAC E3 ubiquitin ligase prevents formation of linear, but not K63-linked ubiquitin chains or STING activation and inhibits STING-induced NFκB and IRF3-mediated signaling in monocytic THP1 cells. The proton channel activity of STING is also important for both K63 and linear ubiquitin chain formation, and NFκB- and interferon-related gene expression. Thus, LUBAC synthesis of linear ubiquitin chains regulates STING-mediated innate immune signaling.

3.
EMBO J ; 42(22): e113491, 2023 Nov 15.
Article in English | MEDLINE | ID: mdl-37621214

ABSTRACT

Nix is a membrane-anchored outer mitochondrial protein that induces mitophagy. While Nix has an LC3-interacting (LIR) motif that binds to ATG8 proteins, it also contains a minimal essential region (MER) that induces mitophagy through an unknown mechanism. We used chemically induced dimerization (CID) to probe the mechanism of Nix-mediated mitophagy and found that both the LIR and MER are required for robust mitophagy. We find that the Nix MER interacts with the autophagy effector WIPI2 and recruits WIPI2 to mitochondria. The Nix LIR motif is also required for robust mitophagy and converts a homogeneous WIPI2 distribution on the surface of the mitochondria into puncta, even in the absence of ATG8s. Together, this work reveals unanticipated mechanisms in Nix-induced mitophagy and the elusive role of the MER, while also describing an interesting example of autophagy induction that acts downstream of the canonical initiation complexes.


Subject(s)
Autophagy , Mitophagy , Mitochondria/metabolism , Membrane Proteins/genetics , Membrane Proteins/metabolism , Autophagy-Related Protein 8 Family/metabolism , Mitochondrial Proteins/genetics , Mitochondrial Proteins/metabolism
4.
PLoS Genet ; 19(7): e1010828, 2023 07.
Article in English | MEDLINE | ID: mdl-37440574

ABSTRACT

The early pathogenesis and underlying molecular causes of motor neuron degeneration in Parkinson's Disease (PD) remains unresolved. In the model organism Drosophila melanogaster, loss of the early-onset PD gene parkin (the ortholog of human PRKN) results in impaired climbing ability, damage to the indirect flight muscles, and mitochondrial fragmentation with swelling. These stressed mitochondria have been proposed to activate innate immune pathways through release of damage associated molecular patterns (DAMPs). Parkin-mediated mitophagy is hypothesized to suppress mitochondrial damage and subsequent activation of the cGAS/STING innate immunity pathway, but the relevance of this interaction in the fly remains unresolved. Using a combination of genetics, immunoassays, and RNA sequencing, we investigated a potential role for STING in the onset of parkin-null phenotypes. Our findings demonstrate that loss of Drosophila STING in flies rescues the thorax muscle defects and the climbing ability of parkin-/- mutants. Loss of STING also suppresses the disrupted mitochondrial morphology in parkin-/- flight muscles, suggesting unexpected feedback of STING on mitochondria integrity or activation of a compensatory mitochondrial pathway. In the animals lacking both parkin and sting, PINK1 is activated and cell death pathways are suppressed. These findings support a unique, non-canonical role for Drosophila STING in the cellular and organismal response to mitochondria stress.


Subject(s)
Drosophila Proteins , Parkinson Disease , Animals , Humans , Drosophila melanogaster/genetics , Drosophila Proteins/genetics , Mitochondria/genetics , Ubiquitin-Protein Ligases/genetics , Drosophila/metabolism , Muscles/metabolism , Parkinson Disease/genetics , Protein Serine-Threonine Kinases/genetics
5.
Mol Cell ; 83(6): 927-941.e8, 2023 03 16.
Article in English | MEDLINE | ID: mdl-36898370

ABSTRACT

Mitophagy is a form of selective autophagy that disposes of superfluous and potentially damage-inducing organelles in a tightly controlled manner. While the machinery involved in mitophagy induction is well known, the regulation of the components is less clear. Here, we demonstrate that TNIP1 knockout in HeLa cells accelerates mitophagy rates and that ectopic TNIP1 negatively regulates the rate of mitophagy. These functions of TNIP1 depend on an evolutionarily conserved LIR motif as well as an AHD3 domain, which are required for binding to the LC3/GABARAP family of proteins and the autophagy receptor TAX1BP1, respectively. We further show that phosphorylation appears to regulate its association with the ULK1 complex member FIP200, allowing TNIP1 to compete with autophagy receptors, which provides a molecular rationale for its inhibitory function during mitophagy. Taken together, our findings describe TNIP1 as a negative regulator of mitophagy that acts at the early steps of autophagosome biogenesis.


Subject(s)
Autophagy-Related Proteins , Autophagy , Mitophagy , Humans , Apoptosis Regulatory Proteins/genetics , Apoptosis Regulatory Proteins/metabolism , Autophagy/genetics , Autophagy-Related Protein 8 Family/metabolism , DNA-Binding Proteins/metabolism , HeLa Cells , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/metabolism , Mitophagy/genetics , Neoplasm Proteins/metabolism
6.
Nat Rev Mol Cell Biol ; 24(3): 167-185, 2023 03.
Article in English | MEDLINE | ID: mdl-36302887

ABSTRACT

Autophagy is a process that targets various intracellular elements for degradation. Autophagy can be non-selective - associated with the indiscriminate engulfment of cytosolic components - occurring in response to nutrient starvation and is commonly referred to as bulk autophagy. By contrast, selective autophagy degrades specific targets, such as damaged organelles (mitophagy, lysophagy, ER-phagy, ribophagy), aggregated proteins (aggrephagy) or invading bacteria (xenophagy), thereby being importantly involved in cellular quality control. Hence, not surprisingly, aberrant selective autophagy has been associated with various human pathologies, prominently including neurodegeneration and infection. In recent years, considerable progress has been made in understanding mechanisms governing selective cargo engulfment in mammals, including the identification of ubiquitin-dependent selective autophagy receptors such as p62, NBR1, OPTN and NDP52, which can bind cargo and ubiquitin simultaneously to initiate pathways leading to autophagy initiation and membrane recruitment. This progress opens the prospects for enhancing selective autophagy pathways to boost cellular quality control capabilities and alleviate pathology.


Subject(s)
Macroautophagy , Proteins , Animals , Humans , Proteins/metabolism , Autophagy , Ubiquitin/metabolism , Mammals/metabolism
8.
J Cell Biol ; 220(5)2021 05 03.
Article in English | MEDLINE | ID: mdl-33891012

ABSTRACT

The VPS13 gene family consists of VPS13A-D in mammals. Although all four genes have been linked to human diseases, their cellular functions are poorly understood, particularly those of VPS13D. We generated and characterized knockouts of each VPS13 gene in HeLa cells. Among the individual knockouts, only VPS13D-KO cells exhibit abnormal mitochondrial morphology. Additionally, VPS13D loss leads to either partial or complete peroxisome loss in several transformed cell lines and in fibroblasts derived from a VPS13D mutation-carrying patient with recessive spinocerebellar ataxia. Our data show that VPS13D regulates peroxisome biogenesis.


Subject(s)
Peroxisomes/genetics , Peroxisomes/metabolism , Proteins/genetics , Proteins/metabolism , HEK293 Cells , HeLa Cells , Humans , Mitochondria/genetics , Mitochondria/metabolism , Mutation/genetics
9.
J Cell Biol ; 220(2)2021 02 01.
Article in English | MEDLINE | ID: mdl-33464298

ABSTRACT

Genome-wide CRISPR screens have transformed our ability to systematically interrogate human gene function, but are currently limited to a subset of cellular phenotypes. We report a novel pooled screening approach for a wider range of cellular and subtle subcellular phenotypes. Machine learning and convolutional neural network models are trained on the subcellular phenotype to be queried. Genome-wide screening then utilizes cells stably expressing dCas9-KRAB (CRISPRi), photoactivatable fluorescent protein (PA-mCherry), and a lentiviral guide RNA (gRNA) pool. Cells are screened by using microscopy and classified by artificial intelligence (AI) algorithms, which precisely identify the genetically altered phenotype. Cells with the phenotype of interest are photoactivated and isolated via flow cytometry, and the gRNAs are identified by sequencing. A proof-of-concept screen accurately identified PINK1 as essential for Parkin recruitment to mitochondria. A genome-wide screen identified factors mediating TFEB relocation from the nucleus to the cytosol upon prolonged starvation. Twenty-one of the 64 hits called by the neural network model were independently validated, revealing new effectors of TFEB subcellular localization. This approach, AI-photoswitchable screening (AI-PS), offers a novel screening platform capable of classifying a broad range of mammalian subcellular morphologies, an approach largely unattainable with current methodologies at genome-wide scale.


Subject(s)
CRISPR-Cas Systems/genetics , Genetic Testing , Genome , Imaging, Three-Dimensional , Artificial Intelligence , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/metabolism , Cell Line, Tumor , Cell Nucleus/metabolism , Cyclic AMP Response Element-Binding Protein A/metabolism , Deep Learning , Green Fluorescent Proteins , HEK293 Cells , Humans , Models, Biological , Neural Networks, Computer , Phenotype , Reproducibility of Results , Single-Cell Analysis , Support Vector Machine , Ubiquitin-Protein Ligases/metabolism , RNA, Guide, CRISPR-Cas Systems
10.
Cell Death Differ ; 28(2): 439-454, 2021 02.
Article in English | MEDLINE | ID: mdl-33208890

ABSTRACT

Ubiquitin signaling is a sequence of events driving the fate of a protein based on the type of ubiquitin modifications attached. In the case of neurodegenerative diseases, ubiquitin signaling is mainly associated with degradation signals to process aberrant proteins, which form aggregates often fatal for the brain cells. This signaling is often perturbed by the aggregates themselves and leads to the accumulation of toxic aggregates and inclusion bodies that are deleterious due to a toxic gain of function. Decrease in quality control pathways is often seen with age and is a critical onset for the development of neurodegeneration. Many aggregates are now thought to propagate in a prion-like manner, where mutated proteins acting like seeds are transitioning from cell to cell, converting normal proteins to toxic aggregates. Modulation of ubiquitin signaling, by stimulating ubiquitin ligase activation, is a potential therapeutic strategy to treat patients with neurodegeneration diseases.


Subject(s)
Autophagy/physiology , Neurodegenerative Diseases/metabolism , Neurodegenerative Diseases/physiopathology , Signal Transduction , Ubiquitin/metabolism , Animals , Humans , Proteasome Endopeptidase Complex/metabolism , Protein Folding , Ubiquitin-Protein Ligases/metabolism , Ubiquitination
11.
Mol Cell ; 80(5): 779-795.e10, 2020 12 03.
Article in English | MEDLINE | ID: mdl-33207181

ABSTRACT

Protein aggregates disrupt cellular homeostasis, causing toxicity linked to neurodegeneration. Selective autophagic elimination of aggregates is critical to protein quality control, but how aggregates are selectively targeted for degradation is unclear. We compared the requirements for autophagy receptor proteins: OPTN, NBR1, p62, NDP52, and TAX1BP1 in clearance of proteotoxic aggregates. Endogenous TAX1BP1 is recruited to and required for the clearance of stress-induced aggregates, whereas ectopic expression of TAX1BP1 increases clearance through autophagy, promoting viability of human induced pluripotent stem cell-derived neurons. In contrast, TAX1BP1 depletion sensitizes cells to several forms of aggregate-induced proteotoxicity. Furthermore, TAX1BP1 is more specifically expressed in the brain compared to other autophagy receptor proteins. In vivo, loss of TAX1BP1 results in accumulation of high molecular weight ubiquitin conjugates and premature lipofuscin accumulation in brains of young TAX1BP1 knockout mice. TAX1BP1 mediates clearance of a broad range of cytotoxic proteins indicating therapeutic potential in neurodegenerative diseases.


Subject(s)
Apoptosis Regulatory Proteins/deficiency , Autophagy , Brain/metabolism , Intracellular Signaling Peptides and Proteins/deficiency , Neoplasm Proteins/deficiency , Neurodegenerative Diseases/metabolism , Protein Aggregation, Pathological/metabolism , Animals , Apoptosis Regulatory Proteins/metabolism , Brain/pathology , Female , HEK293 Cells , HeLa Cells , Humans , Intracellular Signaling Peptides and Proteins/metabolism , Lipofuscin/genetics , Lipofuscin/metabolism , Male , Mice , Mice, Knockout , Neoplasm Proteins/metabolism , Neurodegenerative Diseases/genetics , Neurodegenerative Diseases/pathology , Protein Aggregation, Pathological/genetics , Protein Aggregation, Pathological/pathology , Rats , Rats, Sprague-Dawley , Ubiquitin/genetics , Ubiquitin/metabolism
12.
J Cell Biol ; 219(12)2020 12 07.
Article in English | MEDLINE | ID: mdl-33201170

ABSTRACT

Following the detection of cytosolic double-stranded DNA from viral or bacterial infection in mammalian cells, cyclic dinucleotide activation of STING induces interferon ß expression to initiate innate immune defenses. STING activation also induces LC3B lipidation, a classical but equivocal marker of autophagy, that promotes a cell-autonomous antiviral response that arose before evolution of the interferon pathway. We report that STING activation induces LC3B lipidation onto single-membrane perinuclear vesicles mediated by ATG16L1 via its WD40 domain, bypassing the requirement of canonical upstream autophagy machinery. This process is blocked by bafilomycin A1 that binds and inhibits the vacuolar ATPase (V-ATPase) and by SopF, a bacterial effector that catalytically modifies the V-ATPase to inhibit LC3B lipidation via ATG16L1. These results indicate that activation of the cGAS-STING pathway induces V-ATPase-dependent LC3B lipidation that may mediate cell-autonomous host defense, an unanticipated mechanism that is distinct from LC3B lipidation onto double-membrane autophagosomes.


Subject(s)
Autophagy-Related Proteins/metabolism , Cell-Derived Microparticles/metabolism , Lipoylation , Membrane Proteins/metabolism , Microtubule-Associated Proteins/metabolism , Vacuolar Proton-Translocating ATPases/metabolism , Animals , Autophagy , Autophagy-Related Proteins/genetics , Cell-Derived Microparticles/genetics , HeLa Cells , Humans , Membrane Proteins/genetics , Mice , Microtubule-Associated Proteins/genetics , Nucleotidyltransferases/genetics , Nucleotidyltransferases/metabolism , Protein Domains , Signal Transduction , Vacuolar Proton-Translocating ATPases/genetics
13.
Annu Rev Cell Dev Biol ; 36: 265-289, 2020 10 06.
Article in English | MEDLINE | ID: mdl-33021820

ABSTRACT

Maintaining mitochondrial health is essential for the survival and function of eukaryotic organisms. Misfunctioning mitochondria activate stress-responsive pathways to restore mitochondrial network homeostasis, remove damaged or toxic proteins, and eliminate damaged organelles via selective autophagy of mitochondria, a process termed mitophagy. Failure of these quality control pathways is implicated in the pathogenesis of Parkinson's disease and other neurodegenerative diseases. Impairment of mitochondrial quality control has been demonstrated to activate innate immune pathways, including inflammasome-mediated signaling and the antiviral cyclic GMP-AMP synthase (cGAS)/stimulator of interferon genes (STING)-regulated interferon response. Immune system malfunction is a common hallmark in many neurodegenerative diseases; however, whether inflammation suppresses or exacerbates disease pathology is still unclear. The goal of this review is to provide a historical overview of the field, describe mechanisms of mitochondrial quality control, and highlight recent advances on the emerging role of mitochondria in innate immunity and inflammation.


Subject(s)
Immunity, Innate , Mitochondria/metabolism , Animals , Apoptosis , DNA, Mitochondrial/genetics , Humans , Membrane Potential, Mitochondrial , Mitochondrial Proteins/metabolism
14.
Brain ; 143(10): 3041-3051, 2020 10 01.
Article in English | MEDLINE | ID: mdl-33029617

ABSTRACT

There is increasing evidence for a role of inflammation in Parkinson's disease. Recent research in murine models suggests that parkin and PINK1 deficiency leads to impaired mitophagy, which causes the release of mitochondrial DNA (mtDNA), thereby triggering inflammation. Specifically, the CGAS (cyclic GMP-AMP synthase)-STING (stimulator of interferon genes) pathway mitigates activation of the innate immune system, quantifiable as increased interleukin-6 (IL6) levels. However, the role of IL6 and circulating cell-free mtDNA in unaffected and affected individuals harbouring mutations in PRKN/PINK1 and idiopathic Parkinson's disease patients remain elusive. We investigated IL6, C-reactive protein, and circulating cell-free mtDNA in serum of 245 participants in two cohorts from tertiary movement disorder centres. We performed a hypothesis-driven rank-based statistical approach adjusting for multiple testing. We detected (i) elevated IL6 levels in patients with biallelic PRKN/PINK1 mutations compared to healthy control subjects in a German cohort, supporting the concept of a role for inflammation in PRKN/PINK1-linked Parkinson's disease. In addition, the comparison of patients with biallelic and heterozygous mutations in PRKN/PINK1 suggests a gene dosage effect. The differences in IL6 levels were validated in a second independent Italian cohort; (ii) a correlation between IL6 levels and disease duration in carriers of PRKN/PINK1 mutations, while no such association was observed for idiopathic Parkinson's disease patients. These results highlight the potential of IL6 as progression marker in Parkinson's disease due to PRKN/PINK1 mutations; (iii) increased circulating cell-free mtDNA serum levels in both patients with biallelic or with heterozygous PRKN/PINK1 mutations compared to idiopathic Parkinson's disease, which is in line with previous findings in murine models. By contrast, circulating cell-free mtDNA concentrations in unaffected heterozygous carriers of PRKN/PINK1 mutations were comparable to control levels; and (iv) that circulating cell-free mtDNA levels have good predictive potential to discriminate between idiopathic Parkinson's disease and Parkinson's disease linked to heterozygous PRKN/PINK1 mutations, providing functional evidence for a role of heterozygous mutations in PRKN or PINK1 as Parkinson's disease risk factor. Taken together, our study further implicates inflammation due to impaired mitophagy and subsequent mtDNA release in the pathogenesis of PRKN/PINK1-linked Parkinson's disease. In individuals carrying mutations in PRKN/PINK1, IL6 and circulating cell-free mtDNA levels may serve as markers of Parkinson's disease state and progression, respectively. Finally, our study suggests that targeting the immune system with anti-inflammatory medication holds the potential to influence the disease course of Parkinson's disease, at least in this subset of patients.


Subject(s)
DNA, Mitochondrial/blood , Interleukin-6/blood , Parkinsonian Disorders/blood , Parkinsonian Disorders/genetics , Protein Kinases/genetics , Ubiquitin-Protein Ligases/genetics , Adult , Aged , Biomarkers/blood , Cross-Sectional Studies , Female , Humans , Inflammation/blood , Inflammation/genetics , Male , Middle Aged , Retrospective Studies
15.
Autophagy ; 16(11): 2105-2107, 2020 11.
Article in English | MEDLINE | ID: mdl-32892694

ABSTRACT

PINK1 and PRKN, proteins mutated in Parkinson disease, selectively amplify ubiquitin signals on damaged mitochondria for elimination via mitophagy. Because all five macroautophagy/autophagy receptors in mammals possess domains binding to ubiquitin and Atg8-family proteins, they were thought to recruit Atg8-family protein labeled phagophores from a cytosolic pool. However, our recent findings show that, in addition to Atg8-family protein binding, two of the receptors CALCOCO2 and OPTN interact with RB1CC1 and ATG9A, respectively, indicating that two different axes, CALCOCO2-RB1CC1 and OPTN-ATG9A, can initiate de novo biogenesis of autophagic membranes on ubiquitin-coated damaged mitochondria. These results explain the critical roles of the autophagy receptors CALCOCO2 and OPTN in mitochondrial degradation, and their abilities to simultaneously bind multiple autophagy core proteins propose a new function, i.e. a scaffold to build multivalent interactions for the orchestrated assembly of autophagy proteins near the ubiquitinated cargo. ABBREVIATIONS: ATG: autophagy-related; CALCOCO2/NDP52: calcium binding and coiled-coil domain 2; CRABP2: cellular retinoic acid binding protein 2; LIR: MAP1LC3/LC3-interacting region; MAP1LC3: microtubule associated protein 1 light chain 3; NBR1: NBR1 autophagy cargo receptor; OPTN: optineurin; PINK1: PTEN induced kinase 1; PRKN: parkin RBR E3 ubiquitin protein ligase; RB1CC1/FIP200: RB1 inducible coiled-coil 1; SNIPER: specific and nongenetic IAP-dependent protein eraser; SQSTM1/p62: sequestosome 1; ULK: unc-51 like autophagy activating kinase.


Subject(s)
Autophagy-Related Proteins/metabolism , Autophagy/physiology , Mitophagy/physiology , Ubiquitin-Protein Ligases/metabolism , Animals , Cell Cycle Proteins/metabolism , Humans , Membrane Transport Proteins/metabolism , Mitochondria/metabolism
16.
J Cell Biol ; 219(7)2020 07 06.
Article in English | MEDLINE | ID: mdl-32516362

ABSTRACT

The autophagy-initiating human ULK complex consists of the kinase ULK1/2, FIP200, ATG13, and ATG101. Hydrogen-deuterium exchange mass spectrometry was used to map their mutual interactions. The N-terminal 640 residues (NTD) of FIP200 interact with the C-terminal IDR of ATG13. Mutations in these regions abolish their interaction. Negative stain EM and multiangle light scattering showed that FIP200 is a dimer, while a single molecule each of the other subunits is present. The FIP200NTD is flexible in the absence of ATG13, but in its presence adopts the shape of the letter C ∼20 nm across. The ULK1 EAT domain interacts loosely with the NTD dimer, while the ATG13:ATG101 HORMA dimer does not contact the NTD. Cryo-EM of the NTD dimer revealed a structural similarity to the scaffold domain of TBK1, suggesting an evolutionary similarity between the autophagy-initiating TBK1 kinase and the ULK1 kinase complex.


Subject(s)
Autophagy-Related Protein-1 Homolog/chemistry , Autophagy-Related Proteins/chemistry , Autophagy/genetics , Intracellular Signaling Peptides and Proteins/chemistry , Protein Serine-Threonine Kinases/chemistry , Vesicular Transport Proteins/chemistry , Amino Acid Sequence , Autophagy-Related Protein-1 Homolog/genetics , Autophagy-Related Protein-1 Homolog/metabolism , Autophagy-Related Proteins/genetics , Autophagy-Related Proteins/metabolism , Binding Sites , Cloning, Molecular , Cryoelectron Microscopy , Deuterium Exchange Measurement , Gene Expression , Gene Expression Regulation , Genetic Vectors/chemistry , Genetic Vectors/metabolism , HEK293 Cells , Humans , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/metabolism , Mutation , Protein Binding , Protein Interaction Domains and Motifs , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Sequence Alignment , Sequence Homology, Amino Acid , Signal Transduction , Vesicular Transport Proteins/genetics , Vesicular Transport Proteins/metabolism
17.
Cell Rep ; 29(1): 225-235.e5, 2019 10 01.
Article in English | MEDLINE | ID: mdl-31577952

ABSTRACT

PINK1 and Parkin are established mediators of mitophagy, the selective removal of damaged mitochondria by autophagy. PINK1 and Parkin have been proposed to act as tumor suppressors, as loss-of-function mutations are correlated with enhanced tumorigenesis. However, it is unclear how PINK1 and Parkin act in coordination during mitophagy to influence the cell cycle. Here we show that PINK1 and Parkin genetically interact with proteins involved in cell cycle regulation, and loss of PINK1 and Parkin accelerates cell growth. PINK1- and Parkin-mediated activation of TBK1 at the mitochondria during mitophagy leads to a block in mitosis due to the sequestration of TBK1 from its physiological role at centrosomes during mitosis. Our study supports a diverse role for the far-reaching, regulatory effects of mitochondrial quality control in cellular homeostasis and demonstrates that the PINK1/Parkin pathway genetically interacts with the cell cycle, providing a framework for understanding the molecular basis linking PINK1 and Parkin to mitosis.


Subject(s)
Cell Cycle/genetics , Mitochondria/genetics , Mitosis/genetics , Protein Kinases/genetics , Protein Serine-Threonine Kinases/genetics , Ubiquitin-Protein Ligases/genetics , Autophagy/genetics , Cell Line, Tumor , Cell Proliferation/genetics , HCT116 Cells , HEK293 Cells , HeLa Cells , Homeostasis/genetics , Humans , Mitophagy/genetics
18.
Science ; 365(6454)2019 08 16.
Article in English | MEDLINE | ID: mdl-31416937

ABSTRACT

Mitochondria are organelles with their own genome that arose from α-proteobacteria living within single-celled Archaea more than a billion years ago. This step of endosymbiosis offered tremendous opportunities for energy production and metabolism and allowed the evolution of fungi, plants, and animals. However, less appreciated are the downsides of this endosymbiosis. Coordinating gene expression between the mitochondrial genomes and the nuclear genome is imprecise and can lead to proteotoxic stress. The clonal reproduction of mitochondrial DNA requires workarounds to avoid mutational meltdown. In metazoans that developed innate immune pathways to thwart bacterial and viral infections, mitochondrial components can cross-react with pathogen sensors and invoke inflammation. Here, I focus on the numerous and elegant quality control processes that compensate for or mitigate these challenges of endosymbiosis.


Subject(s)
Host Microbial Interactions/immunology , Immunity, Innate , Mitochondria/immunology , Symbiosis/immunology , Animals , DNA, Mitochondrial/immunology , Humans , Mitochondria/genetics , Mitophagy/immunology
19.
J Biol Chem ; 294(30): 11498-11512, 2019 07 26.
Article in English | MEDLINE | ID: mdl-31177092

ABSTRACT

Neurolastin is a dynamin family GTPase that also contains a RING domain and exhibits both GTPase and E3 ligase activities. It is specifically expressed in the brain and is important for synaptic transmission, as neurolastin knockout animals have fewer dendritic spines and exhibit a reduction in functional synapses. Our initial study of neurolastin revealed that it is membrane-associated and partially co-localizes with endosomes. Using various biochemical and cell-culture approaches, we now show that neurolastin also localizes to mitochondria in HeLa cells, cultured neurons, and brain tissue. We found that the mitochondrial localization of neurolastin depends upon an N-terminal mitochondrial targeting sequence and that neurolastin is imported into the mitochondrial intermembrane space. Although neurolastin was only partially mitochondrially localized at steady state, it displayed increased translocation to mitochondria in response to neuronal stress and mitochondrial fragmentation. Interestingly, inactivation or deletion of neurolastin's RING domain also increased its mitochondrial localization. Using EM, we observed that neurolastin knockout animals have smaller but more numerous mitochondria in cerebellar Purkinje neurons, indicating that neurolastin regulates mitochondrial morphology. We conclude that the brain-specific dynamin GTPase neurolastin exhibits stress-responsive localization to mitochondria and is required for proper mitochondrial morphology.


Subject(s)
Dynamins/metabolism , Mitochondria/metabolism , Purkinje Cells/metabolism , Animals , Cells, Cultured , Dynamins/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Mitochondria/enzymology , Mutation , Protein Transport
20.
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
SELECTION OF CITATIONS
SEARCH DETAIL
...