Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 4.804
Filter
1.
Cell ; 187(14): 3619-3637.e27, 2024 Jul 11.
Article in English | MEDLINE | ID: mdl-38851188

ABSTRACT

Mitochondrial dynamics play a critical role in cell fate decisions and in controlling mtDNA levels and distribution. However, the molecular mechanisms linking mitochondrial membrane remodeling and quality control to mtDNA copy number (CN) regulation remain elusive. Here, we demonstrate that the inner mitochondrial membrane (IMM) protein mitochondrial fission process 1 (MTFP1) negatively regulates IMM fusion. Moreover, manipulation of mitochondrial fusion through the regulation of MTFP1 levels results in mtDNA CN modulation. Mechanistically, we found that MTFP1 inhibits mitochondrial fusion to isolate and exclude damaged IMM subdomains from the rest of the network. Subsequently, peripheral fission ensures their segregation into small MTFP1-enriched mitochondria (SMEM) that are targeted for degradation in an autophagic-dependent manner. Remarkably, MTFP1-dependent IMM quality control is essential for basal nucleoid recycling and therefore to maintain adequate mtDNA levels within the cell.


Subject(s)
DNA, Mitochondrial , Mitochondria , Mitochondrial Dynamics , Mitochondrial Membranes , Mitochondrial Proteins , DNA, Mitochondrial/metabolism , DNA, Mitochondrial/genetics , Mitochondrial Proteins/metabolism , Humans , Mitochondrial Membranes/metabolism , Mitochondria/metabolism , Animals , HeLa Cells , Mice , Membrane Proteins/metabolism , Membrane Proteins/genetics , Autophagy
2.
Cell ; 184(17): 4464-4479.e19, 2021 08 19.
Article in English | MEDLINE | ID: mdl-34384544

ABSTRACT

Emerging evidence supports that mitochondrial dysfunction contributes to systemic lupus erythematosus (SLE) pathogenesis. Here we show that programmed mitochondrial removal, a hallmark of mammalian erythropoiesis, is defective in SLE. Specifically, we demonstrate that during human erythroid cell maturation, a hypoxia-inducible factor (HIF)-mediated metabolic switch is responsible for the activation of the ubiquitin-proteasome system (UPS), which precedes and is necessary for the autophagic removal of mitochondria. A defect in this pathway leads to accumulation of red blood cells (RBCs) carrying mitochondria (Mito+ RBCs) in SLE patients and in correlation with disease activity. Antibody-mediated internalization of Mito+ RBCs induces type I interferon (IFN) production through activation of cGAS in macrophages. Accordingly, SLE patients carrying both Mito+ RBCs and opsonizing antibodies display the highest levels of blood IFN-stimulated gene (ISG) signatures, a distinctive feature of SLE.


Subject(s)
Interferon Type I/metabolism , Lupus Erythematosus, Systemic/metabolism , Mitochondria/metabolism , Myeloid Cells/metabolism , Adolescent , Basic Helix-Loop-Helix Transcription Factors/metabolism , Child , Child, Preschool , Erythroblasts/metabolism , Erythroblasts/ultrastructure , Erythrocytes/metabolism , Erythropoiesis , Humans , Mitophagy , Proteasome Endopeptidase Complex/metabolism , Ubiquitin/metabolism
3.
Cell ; 181(5): 1176-1187.e16, 2020 05 28.
Article in English | MEDLINE | ID: mdl-32437660

ABSTRACT

Dysfunctional mitochondria accumulate in many human diseases. Accordingly, mitophagy, which removes these mitochondria through lysosomal degradation, is attracting broad attention. Due to uncertainties in the operational principles of conventional mitophagy probes, however, the specificity and quantitativeness of their readouts are disputable. Thorough investigation of the behaviors and fates of fluorescent proteins inside and outside lysosomes enabled us to develop an indicator for mitophagy, mito-SRAI. Through strict control of its mitochondrial targeting, we were able to monitor mitophagy in fixed biological samples more reproducibly than before. Large-scale image-based high-throughput screening led to the discovery of a hit compound that induces selective mitophagy of damaged mitochondria. In a mouse model of Parkinsons disease, we found that dopaminergic neurons selectively failed to execute mitophagy that promoted their survival within lesions. These results show that mito-SRAI is an essential tool for quantitative studies of mitochondrial quality control.


Subject(s)
Fluorescence Resonance Energy Transfer/methods , Lysosomes/metabolism , Mitophagy/physiology , Animals , Autophagy/physiology , Fluorescent Antibody Technique/methods , Fluorescent Dyes/chemistry , Humans , Lysosomes/physiology , Male , Mice , Mice, Inbred C57BL , Mitochondria/metabolism , Mitophagy/genetics
4.
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
5.
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
6.
Immunity ; 57(6): 1289-1305.e9, 2024 Jun 11.
Article in English | MEDLINE | ID: mdl-38772366

ABSTRACT

Adipose tissue group 2 innate lymphoid cells (ILC2s) help maintain metabolic homeostasis by sustaining type 2 immunity and promoting adipose beiging. Although impairment of the ILC2 compartment contributes to obesity-associated insulin resistance, the underlying mechanisms have not been elucidated. Here, we found that ILC2s in obese mice and humans exhibited impaired liver kinase B1 (LKB1) activation. Genetic ablation of LKB1 disrupted ILC2 mitochondrial metabolism and suppressed ILC2 responses, resulting in exacerbated insulin resistance. Mechanistically, LKB1 deficiency induced aberrant PD-1 expression through activation of NFAT, which in turn enhanced mitophagy by suppressing Bcl-xL expression. Blockade of PD-1 restored the normal functions of ILC2s and reversed obesity-induced insulin resistance in mice. Collectively, these data present the LKB1-PD-1 axis as a promising therapeutic target for the treatment of metabolic disease.


Subject(s)
Adipose Tissue , Homeostasis , Insulin Resistance , Lymphocytes , Mitochondria , Obesity , Programmed Cell Death 1 Receptor , Protein Serine-Threonine Kinases , Animals , Insulin Resistance/immunology , Programmed Cell Death 1 Receptor/metabolism , Mice , Protein Serine-Threonine Kinases/metabolism , Protein Serine-Threonine Kinases/genetics , Mitochondria/metabolism , Humans , Adipose Tissue/metabolism , Adipose Tissue/immunology , Obesity/immunology , Obesity/metabolism , Lymphocytes/immunology , Lymphocytes/metabolism , AMP-Activated Protein Kinases/metabolism , Mice, Inbred C57BL , Mice, Knockout , Immunity, Innate , Male , Mitophagy/immunology , AMP-Activated Protein Kinase Kinases
7.
Cell ; 174(1): 88-101.e16, 2018 06 28.
Article in English | MEDLINE | ID: mdl-29909986

ABSTRACT

In colorectal cancer patients, a high density of cytotoxic CD8+ T cells in tumors is associated with better prognosis. Using a Stat3 loss-of-function approach in two wnt/ß-catenin-dependent autochthonous models of sporadic intestinal tumorigenesis, we unravel a complex intracellular process in intestinal epithelial cells (IECs) that controls the induction of a CD8+ T cell based adaptive immune response. Elevated mitophagy in IECs causes iron(II)-accumulation in epithelial lysosomes, in turn, triggering lysosomal membrane permeabilization. Subsequent release of proteases into the cytoplasm augments MHC class I presentation and activation of CD8+ T cells via cross-dressing of dendritic cells. Thus, our findings highlight a so-far-unrecognized link between mitochondrial function, lysosomal integrity, and MHC class I presentation in IECs and suggest that therapies triggering mitophagy or inducing LMP in IECs may prove successful in shifting the balance toward anti-tumor immunity in colorectal cancer.


Subject(s)
Adaptive Immunity , Mitophagy , Adaptive Immunity/drug effects , Animals , Azoxymethane/toxicity , CD8-Positive T-Lymphocytes/cytology , CD8-Positive T-Lymphocytes/drug effects , CD8-Positive T-Lymphocytes/metabolism , Cell Membrane Permeability , Colorectal Neoplasms/mortality , Colorectal Neoplasms/pathology , Cytokines/metabolism , Dendritic Cells/cytology , Dendritic Cells/immunology , Dendritic Cells/metabolism , Female , Ferrous Compounds/metabolism , Humans , Interferon-gamma/metabolism , Interferon-gamma/pharmacology , Intestinal Mucosa/cytology , Intestinal Mucosa/drug effects , Intestinal Mucosa/metabolism , Lysosomes/metabolism , Male , Mice , Mice, Knockout , Mitophagy/drug effects , STAT3 Transcription Factor/genetics , STAT3 Transcription Factor/metabolism , Survival Rate
8.
Annu Rev Cell Dev Biol ; 35: 477-500, 2019 10 06.
Article in English | MEDLINE | ID: mdl-31340124

ABSTRACT

Autophagy is the major cellular pathway to degrade dysfunctional organelles and protein aggregates. Autophagy is particularly important in neurons, which are terminally differentiated cells that must last the lifetime of the organism. There are both constitutive and stress-induced pathways for autophagy in neurons, which catalyze the turnover of aged or damaged mitochondria, endoplasmic reticulum, other cellular organelles, and aggregated proteins. These pathways are required in neurodevelopment as well as in the maintenance of neuronal homeostasis. Here we review the core components of the pathway for autophagosome biogenesis, as well as the cell biology of bulk and selective autophagy in neurons. Finally, we discuss the role of autophagy in neuronal development, homeostasis, and aging and the links between deficits in autophagy and neurodegeneration.


Subject(s)
Autophagy/genetics , Endoplasmic Reticulum/metabolism , Mitochondria/metabolism , Neurons/metabolism , Animals , Autophagosomes/metabolism , Autophagy/physiology , Axons/metabolism , Dendrites/metabolism , Endoplasmic Reticulum/physiology , Homeostasis/genetics , Homeostasis/physiology , Humans , Memory/physiology , Mitochondria/enzymology , Mitochondria/genetics , Mitochondria/pathology , Neurodegenerative Diseases/genetics , Neurodegenerative Diseases/metabolism , Neurons/cytology , Proteasome Endopeptidase Complex/metabolism , Synapses/metabolism , Synapses/physiology , Ubiquitination/genetics , Ubiquitination/physiology , Unfolded Protein Response/genetics , Unfolded Protein Response/physiology
9.
Annu Rev Cell Dev Biol ; 35: 453-475, 2019 10 06.
Article in English | MEDLINE | ID: mdl-31283377

ABSTRACT

Macroautophagy is an intracellular degradation system that delivers diverse cytoplasmic materials to lysosomes via autophagosomes. Recent advances have enabled identification of several selective autophagy substrates and receptors, greatly expanding our understanding of the cellular functions of autophagy. In this review, we describe the diverse cellular functions of macroautophagy, including its essential contribution to metabolic adaptation and cellular homeostasis. We also discuss emerging findings on the mechanisms and functions of various types of selective autophagy.


Subject(s)
Autophagosomes/metabolism , Autophagy/genetics , Endoplasmic Reticulum/metabolism , Lysosomes/metabolism , Mitochondria/metabolism , Animals , Autophagosomes/enzymology , Autophagosomes/microbiology , Autophagy/physiology , Endoplasmic Reticulum/physiology , Homeostasis/genetics , Homeostasis/physiology , Humans , Lysosomes/pathology , Mitochondria/pathology , Nutrients/deficiency , Nutrients/metabolism , Peroxisomes/metabolism , Peroxisomes/physiology
10.
Annu Rev Biochem ; 86: 193-224, 2017 06 20.
Article in English | MEDLINE | ID: mdl-28460188

ABSTRACT

Autophagy and the ubiquitin-proteasome system are the two major quality control pathways responsible for cellular homeostasis. As such, they provide protection against age-associated changes and a plethora of human diseases. Ubiquitination is utilized as a degradation signal by both systems, albeit in different ways, to mark cargoes for proteasomal and lysosomal degradation. Both systems intersect and communicate at multiple points to coordinate their actions in proteostasis and organelle homeostasis. This review summarizes molecular details of how proteasome and autophagy pathways are functionally interconnected in cells and indicates common principles and nodes of communication that can be therapeutically exploited.


Subject(s)
Autophagy/genetics , Kelch-Like ECH-Associated Protein 1/metabolism , NF-E2-Related Factor 2/metabolism , Proteasome Endopeptidase Complex/metabolism , Sequestosome-1 Protein/metabolism , Ubiquitin/metabolism , Eukaryotic Cells/cytology , Eukaryotic Cells/metabolism , Gene Expression Regulation , Homeostasis , Humans , Kelch-Like ECH-Associated Protein 1/chemistry , Kelch-Like ECH-Associated Protein 1/genetics , Mitophagy/genetics , Molecular Chaperones/genetics , Molecular Chaperones/metabolism , NF-E2-Related Factor 2/chemistry , NF-E2-Related Factor 2/genetics , Protein Conformation , Proteolysis , Sequestosome-1 Protein/chemistry , Sequestosome-1 Protein/genetics , Signal Transduction , Ubiquitin/genetics , Ubiquitination
11.
Cell ; 168(1-2): 224-238.e10, 2017 Jan 12.
Article in English | MEDLINE | ID: mdl-28017329

ABSTRACT

The removal of unwanted or damaged mitochondria by autophagy, a process called mitophagy, is essential for key events in development, cellular homeostasis, tumor suppression, and prevention of neurodegeneration and aging. However, the precise mechanisms of mitophagy remain uncertain. Here, we identify the inner mitochondrial membrane protein, prohibitin 2 (PHB2), as a crucial mitophagy receptor involved in targeting mitochondria for autophagic degradation. PHB2 binds the autophagosomal membrane-associated protein LC3 through an LC3-interaction region (LIR) domain upon mitochondrial depolarization and proteasome-dependent outer membrane rupture. PHB2 is required for Parkin-induced mitophagy in mammalian cells and for the clearance of paternal mitochondria after embryonic fertilization in C. elegans. Our findings pinpoint a conserved mechanism of eukaryotic mitophagy and demonstrate a function of prohibitin 2 that may underlie its roles in physiology, aging, and disease.


Subject(s)
Caenorhabditis elegans/metabolism , Mitochondrial Membranes/metabolism , Repressor Proteins/metabolism , Aging/metabolism , Animals , Autophagosomes/metabolism , Caenorhabditis elegans Proteins/metabolism , Embryo, Nonmammalian/metabolism , Membrane Proteins/metabolism , Prohibitins
12.
Mol Cell ; 84(6): 1090-1100.e6, 2024 Mar 21.
Article in English | MEDLINE | ID: mdl-38340717

ABSTRACT

To maintain mitochondrial homeostasis, damaged or excessive mitochondria are culled in coordination with the physiological state of the cell. The integrated stress response (ISR) is a signaling network that recognizes diverse cellular stresses, including mitochondrial dysfunction. Because the four ISR branches converge to common outputs, it is unclear whether mitochondrial stress detected by this network can regulate mitophagy, the autophagic degradation of mitochondria. Using a whole-genome screen, we show that the heme-regulated inhibitor (HRI) branch of the ISR selectively induces mitophagy. Activation of the HRI branch results in mitochondrial localization of phosphorylated eukaryotic initiation factor 2, which we show is sufficient to induce mitophagy. The HRI mitophagy pathway operates in parallel with the mitophagy pathway controlled by the Parkinson's disease related genes PINK1 and PARKIN and is mechanistically distinct. Therefore, HRI repurposes machinery that is normally used for translational initiation to trigger mitophagy in response to mitochondrial damage.


Subject(s)
Mitophagy , Protein Kinases , Mitophagy/physiology , Protein Kinases/genetics , Protein Kinases/metabolism , Autophagy/genetics , Ubiquitin-Protein Ligases/genetics , Ubiquitin-Protein Ligases/metabolism , Protein Processing, Post-Translational , Signal Transduction
13.
Mol Cell ; 84(2): 327-344.e9, 2024 Jan 18.
Article in English | MEDLINE | ID: mdl-38151018

ABSTRACT

Mitophagy mediated by BNIP3 and NIX critically regulates mitochondrial mass. Cellular BNIP3 and NIX levels are tightly controlled by SCFFBXL4-mediated ubiquitination to prevent excessive mitochondrial loss and lethal disease. Here, we report that knockout of PPTC7, a mitochondrial matrix protein, hyperactivates BNIP3-/NIX-mediated mitophagy and causes perinatal lethality that is rescued by NIX knockout in mice. Biochemically, the PPTC7 precursor is trapped by BNIP3 and NIX to the mitochondrial outer membrane, where PPTC7 scaffolds assembly of a substrate-PPTC7-SCFFBXL4 holocomplex to degrade BNIP3 and NIX, forming a homeostatic regulatory loop. PPTC7 possesses an unusually weak mitochondrial targeting sequence to facilitate its outer membrane retention and mitophagy control. Starvation upregulates PPPTC7 expression in mouse liver to repress mitophagy, which critically maintains hepatic mitochondrial mass, bioenergetics, and gluconeogenesis. Collectively, PPTC7 functions as a mitophagy sensor that integrates homeostatic and physiological signals to dynamically control BNIP3 and NIX degradation, thereby maintaining mitochondrial mass and cellular homeostasis.


Subject(s)
Membrane Proteins , Mitochondrial Membranes , Mitochondrial Proteins , Mitophagy , Protein Phosphatase 2C , Proteolysis , Animals , Mice , Membrane Proteins/metabolism , Mitochondria/genetics , Mitochondria/metabolism , Mitochondrial Membranes/metabolism , Mitochondrial Proteins/metabolism , Mitophagy/genetics , Protein Phosphatase 2C/metabolism
14.
Immunity ; 55(11): 2059-2073.e8, 2022 11 08.
Article in English | MEDLINE | ID: mdl-36351375

ABSTRACT

T memory stem cells (TSCM) display increased self-renewal and prolonged survival capabilities, thus preventing T cell exhaustion and promoting effective anti-tumor T cell responses. TSCM cells can be expanded by Urolithin A (UA), which is produced by the commensal gut microbiome from foods rich in ellagitannins and is known to improve mitochondrial health. Oral UA administration to tumor-bearing mice conferred strong anti-tumor CD8+ T cell immunity, whereas ex vivo UA pre-treated T cells displayed improved anti-tumor function upon adoptive cell transfer. UA-induced TSCM formation depended on Pink1-mediated mitophagy triggering cytosolic release of the mitochondrial phosphatase Pgam5. Cytosolic Pgam5 dephosphorylated ß-catenin, which drove Wnt signaling and compensatory mitochondrial biogenesis. Collectively, we unravel a critical signaling pathway linking mitophagy to TSCM formation and suggest that the well-tolerated metabolic compound UA represents an attractive option to improve immune therapy.


Subject(s)
Coumarins , Mitophagy , Mice , Animals , Coumarins/pharmacology , Wnt Signaling Pathway , Stem Cells , Immunologic Memory
15.
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
16.
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.

17.
Mol Cell ; 83(12): 2045-2058.e9, 2023 Jun 15.
Article in English | MEDLINE | ID: mdl-37192628

ABSTRACT

Mitophagy plays an important role in mitochondrial homeostasis by selective degradation of mitochondria. During mitophagy, mitochondria should be fragmented to allow engulfment within autophagosomes, whose capacity is exceeded by the typical mitochondria mass. However, the known mitochondrial fission factors, dynamin-related proteins Dnm1 in yeasts and DNM1L/Drp1 in mammals, are dispensable for mitophagy. Here, we identify Atg44 as a mitochondrial fission factor that is essential for mitophagy in yeasts, and we therefore term Atg44 and its orthologous proteins mitofissin. In mitofissin-deficient cells, a part of the mitochondria is recognized by the mitophagy machinery as cargo but cannot be enwrapped by the autophagosome precursor, the phagophore, due to a lack of mitochondrial fission. Furthermore, we show that mitofissin directly binds to lipid membranes and brings about lipid membrane fragility to facilitate membrane fission. Taken together, we propose that mitofissin acts directly on lipid membranes to drive mitochondrial fission required for mitophagy.


Subject(s)
Autophagy , Mitophagy , Animals , Mitochondrial Dynamics , Mitochondrial Proteins/genetics , Mitochondrial Proteins/metabolism , Mitochondria/genetics , Mitochondria/metabolism , Dynamins/genetics , Dynamins/metabolism , Lipids , Mammals/metabolism
18.
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
19.
Physiol Rev ; 103(4): 2349-2422, 2023 10 01.
Article in English | MEDLINE | ID: mdl-37021870

ABSTRACT

Mitochondria are well known as organelles responsible for the maintenance of cellular bioenergetics through the production of ATP. Although oxidative phosphorylation may be their most important function, mitochondria are also integral for the synthesis of metabolic precursors, calcium regulation, the production of reactive oxygen species, immune signaling, and apoptosis. Considering the breadth of their responsibilities, mitochondria are fundamental for cellular metabolism and homeostasis. Appreciating this significance, translational medicine has begun to investigate how mitochondrial dysfunction can represent a harbinger of disease. In this review, we provide a detailed overview of mitochondrial metabolism, cellular bioenergetics, mitochondrial dynamics, autophagy, mitochondrial damage-associated molecular patterns, mitochondria-mediated cell death pathways, and how mitochondrial dysfunction at any of these levels is associated with disease pathogenesis. Mitochondria-dependent pathways may thereby represent an attractive therapeutic target for ameliorating human disease.


Subject(s)
Aging , Mitochondria , Humans , Aging/metabolism , Mitochondria/metabolism , Autophagy , Apoptosis , Reactive Oxygen Species/metabolism
20.
Annu Rev Genet ; 56: 17-39, 2022 11 30.
Article in English | MEDLINE | ID: mdl-35679620

ABSTRACT

Autophagy, a lysosome-mediated degradation process evolutionarily conserved from yeast to mammals, is essential for maintaining cellular homeostasis and combating diverse cellular stresses. Autophagy involves de novo synthesis of a double-membrane autophagosome, sequestration of selected cellular contents, and subsequent delivery of sequestrated contents to the vacuole (in yeasts and plants) or to lysosomes (in animal cells) for degradation and recycling. Genetic studies in unicellular and multicellular model organisms have systematically revealed the molecular machinery, regulation, and function of autophagy in physiological settings. I review genetic studies in model organisms-from yeast to worm to fly-that enable us to not only identify autophagy genes, including ATG genes and the metazoan-specific EPG genes, but also uncover variants of autophagy in developmental contexts, novel regulatory mechanisms, and signaling events involved in mediating systemic autophagy response. Genetic analysis also helps us understand the liquid-liquid phase separation and transition that control autophagic degradation of protein aggregates. The emerging role of autophagy in zebrafish tissue regeneration is also discussed.


Subject(s)
Saccharomyces cerevisiae , Zebrafish , Animals , Autophagy/genetics , Lysosomes , Signal Transduction/genetics , Mammals
SELECTION OF CITATIONS
SEARCH DETAIL