Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 37
Filter
Add more filters










Publication year range
1.
PLoS Genet ; 20(2): e1011137, 2024 Feb.
Article in English | MEDLINE | ID: mdl-38335241

ABSTRACT

Lipid dyshomeostasis has been implicated in a variety of diseases ranging from obesity to neurodegenerative disorders such as Neurodegeneration with Brain Iron Accumulation (NBIA). Here, we uncover the physiological role of Nazo, the Drosophila melanogaster homolog of the NBIA-mutated protein-c19orf12, whose function has been elusive. Ablation of Drosophila c19orf12 homologs leads to dysregulation of multiple lipid metabolism genes. nazo mutants exhibit markedly reduced gut lipid droplet and whole-body triglyceride contents. Consequently, they are sensitive to starvation and oxidative stress. Nazo is required for maintaining normal levels of Perilipin-2, an inhibitor of the lipase-Brummer. Concurrent knockdown of Brummer or overexpression of Perilipin-2 rescues the nazo phenotype, suggesting that this defect, at least in part, may arise from diminished Perilipin-2 on lipid droplets leading to aberrant Brummer-mediated lipolysis. Our findings potentially provide novel insights into the role of c19orf12 as a possible link between lipid dyshomeostasis and neurodegeneration, particularly in the context of NBIA.


Subject(s)
Drosophila melanogaster , Drosophila , Animals , Drosophila/genetics , Drosophila/metabolism , Drosophila melanogaster/genetics , Perilipin-2 , Homeostasis/genetics , Triglycerides/genetics , Triglycerides/metabolism , Lipids
2.
Proc Natl Acad Sci U S A ; 119(38): e2204083119, 2022 09 20.
Article in English | MEDLINE | ID: mdl-36095197

ABSTRACT

Mammalian target of rapamycin (mTOR) is a highly conserved eukaryotic protein kinase that coordinates cell growth and metabolism, and plays a critical role in cancer, immunity, and aging. It remains unclear how mTOR signaling in individual tissues contributes to whole-organism processes because mTOR inhibitors, like the natural product rapamycin, are administered systemically and target multiple tissues simultaneously. We developed a chemical-genetic system, termed selecTOR, that restricts the activity of a rapamycin analog to specific cell populations through targeted expression of a mutant FKBP12 protein. This analog has reduced affinity for its obligate binding partner FKBP12, which reduces its ability to inhibit mTOR in wild-type cells and tissues. Expression of the mutant FKBP12, which contains an expanded binding pocket, rescues the activity of this rapamycin analog. Using this system, we show that selective mTOR inhibition can be achieved in Saccharomyces cerevisiae and human cells, and we validate the utility of our system in an intact metazoan model organism by identifying the tissues responsible for a rapamycin-induced developmental delay in Drosophila.


Subject(s)
Protein Kinase Inhibitors , Sirolimus , TOR Serine-Threonine Kinases , Humans , Organ Specificity , Phosphorylation , Protein Kinase Inhibitors/pharmacology , Signal Transduction , Sirolimus/analogs & derivatives , Sirolimus/pharmacology , TOR Serine-Threonine Kinases/antagonists & inhibitors , TOR Serine-Threonine Kinases/genetics , Tacrolimus Binding Protein 1A/genetics , Tacrolimus Binding Protein 1A/metabolism
3.
PLoS Genet ; 17(2): e1008859, 2021 02.
Article in English | MEDLINE | ID: mdl-33539341

ABSTRACT

Abnormal protein aggregation within neurons is a key pathologic feature of Parkinson's disease (PD). The spread of brain protein aggregates is associated with clinical disease progression, but how this occurs remains unclear. Mutations in glucosidase, beta acid 1 (GBA), which encodes glucocerebrosidase (GCase), are the most penetrant common genetic risk factor for PD and dementia with Lewy bodies and associate with faster disease progression. To explore how GBA mutations influence pathogenesis, we previously created a Drosophila model of GBA deficiency (Gba1b) that manifests neurodegeneration and accelerated protein aggregation. Proteomic analysis of Gba1b mutants revealed dysregulation of proteins involved in extracellular vesicle (EV) biology, and we found altered protein composition of EVs from Gba1b mutants. Accordingly, we hypothesized that GBA may influence pathogenic protein aggregate spread via EVs. We found that accumulation of ubiquitinated proteins and Ref(2)P, Drosophila homologue of mammalian p62, were reduced in muscle and brain tissue of Gba1b flies by ectopic expression of wildtype GCase in muscle. Neuronal GCase expression also rescued protein aggregation both cell-autonomously in brain and non-cell-autonomously in muscle. Muscle-specific GBA expression reduced the elevated levels of EV-intrinsic proteins and Ref(2)P found in EVs from Gba1b flies. Perturbing EV biogenesis through neutral sphingomyelinase (nSMase), an enzyme important for EV release and ceramide metabolism, enhanced protein aggregation when knocked down in muscle, but did not modify Gba1b mutant protein aggregation when knocked down in neurons. Lipidomic analysis of nSMase knockdown on ceramide and glucosylceramide levels suggested that Gba1b mutant protein aggregation may depend on relative depletion of specific ceramide species often enriched in EVs. Finally, we identified ectopically expressed GCase within isolated EVs. Together, our findings suggest that GCase deficiency promotes accelerated protein aggregate spread between cells and tissues via dysregulated EVs, and EV-mediated trafficking of GCase may partially account for the reduction in aggregate spread.


Subject(s)
Drosophila melanogaster/metabolism , Extracellular Vesicles/metabolism , Glucosylceramidase/metabolism , Neurons/metabolism , Parkinson Disease/metabolism , Protein Aggregation, Pathological/metabolism , Animals , Biological Transport , Brain/metabolism , Ceramides/metabolism , DNA-Binding Proteins/metabolism , Disease Models, Animal , Drosophila Proteins/metabolism , Drosophila melanogaster/genetics , Gene Knockdown Techniques , Glucosylceramidase/deficiency , Glucosylceramidase/genetics , Glucosylceramides/metabolism , Lipidomics , Muscles/metabolism , Mutation , Parkinson Disease/genetics , Parkinson Disease/pathology , Protein Aggregation, Pathological/genetics , Proteome/genetics , Proteome/metabolism , RNA Interference
4.
J Gerontol A Biol Sci Med Sci ; 76(10): 1734-1739, 2021 09 13.
Article in English | MEDLINE | ID: mdl-33453098

ABSTRACT

The accumulation of protein aggregates and dysfunctional organelles as organisms age has led to the hypothesis that aging involves general breakdown of protein quality control. We tested this hypothesis using a proteomic and informatic approach in the fruit fly Drosophila melanogaster. Turnover of most proteins was markedly slower in old flies. However, ribosomal and proteasomal proteins maintained high turnover rates, suggesting that the observed slowdowns in protein turnover might not be due to a global failure of quality control. As protein turnover reflects the balance of protein synthesis and degradation, we investigated whether decreases in synthesis or decreases in degradation would best explain the observed slowdowns in protein turnover. We found that while many individual proteins in old flies showed slower turnover due to decreased degradation, an approximately equal number showed slower turnover due to decreased synthesis, and enrichment analyses revealed that translation machinery itself was less abundant. Mitochondrial complex I subunits and glycolytic enzymes were decreased in abundance as well, and proteins involved in glutamine-dependent anaplerosis were increased, suggesting that old flies modify energy production to limit oxidative damage. Together, our findings suggest that age-related proteostasis changes in Drosophila represent a coordinated adaptation rather than a system collapse.


Subject(s)
Drosophila Proteins , Drosophila , Aging , Animals , Drosophila Proteins/metabolism , Drosophila melanogaster/metabolism , Proteins/metabolism , Proteomics , Proteostasis
5.
PLoS Genet ; 16(10): e1009118, 2020 10.
Article in English | MEDLINE | ID: mdl-33075064

ABSTRACT

The m-AAA proteases play a critical role in the proteostasis of inner mitochondrial membrane proteins, and mutations in the genes encoding these proteases cause severe incurable neurological diseases. To further explore the biological role of the m-AAA proteases and the pathological consequences of their deficiency, we used a genetic approach in the fruit fly Drosophila melanogaster to inactivate the ATPase family gene 3-like 2 (AFG3L2) gene, which encodes a critical component of the m-AAA proteases. We found that null alleles of Drosophila AFG3L2 die early in development, but partial inactivation of AFG3L2 using RNAi allowed survival to the late pupal and adult stages of development. Flies with partial inactivation of AFG3L2 exhibited behavioral defects, neurodegeneration, accumulation of unfolded mitochondrial proteins, and diminished respiratory chain (RC) activity. Further work revealed that the reduced RC activity was primarily a consequence of severely diminished mitochondrial transcription and translation. These defects were accompanied by activation of the mitochondrial unfolded protein response (mito-UPR) and autophagy. Overexpression of mito-UPR components partially rescued the AFG3L2-deficient phenotypes, indicating that protein aggregation partly accounts for the defects of AFG3L2-deficient animals. Our work suggests that strategies designed to activate mitochondrial stress pathways and mitochondrial gene expression could be therapeutic in the diseases caused by mutations in AFG3L2.


Subject(s)
ATP-Dependent Proteases/genetics , ATPases Associated with Diverse Cellular Activities/genetics , Electron Transport/genetics , Mitochondria/genetics , Animals , Drosophila melanogaster/genetics , Drosophila melanogaster/growth & development , Gene Expression Regulation, Developmental/genetics , Humans , Microscopy, Electron, Transmission , Mitochondria/ultrastructure , Mitochondrial Diseases/genetics , Mitochondrial Diseases/pathology , Peptide Hydrolases , Pupa/genetics , Pupa/growth & development , RNA Interference , Ribosomes/genetics
6.
Nat Commun ; 10(1): 3280, 2019 07 23.
Article in English | MEDLINE | ID: mdl-31337756

ABSTRACT

Somatic mutations in the mitochondrial genome (mtDNA) have been linked to multiple disease conditions and to ageing itself. In Drosophila, knock-in of a proofreading deficient mtDNA polymerase (POLG) generates high levels of somatic point mutations and also small indels, but surprisingly limited impact on organismal longevity or fitness. Here we describe a new mtDNA mutator model based on a mitochondrially-targeted cytidine deaminase, APOBEC1. mito-APOBEC1 acts as a potent mutagen which exclusively induces C:G>T:A transitions with no indels or mtDNA depletion. In these flies, the presence of multiple non-synonymous substitutions, even at modest heteroplasmy, disrupts mitochondrial function and dramatically impacts organismal fitness. A detailed analysis of the mutation profile in the POLG and mito-APOBEC1 models reveals that mutation type (quality) rather than quantity is a critical factor in impacting organismal fitness. The specificity for transition mutations and the severe phenotypes make mito-APOBEC1 an excellent mtDNA mutator model for ageing research.


Subject(s)
APOBEC-1 Deaminase/physiology , DNA, Mitochondrial/chemistry , Drosophila/genetics , APOBEC-1 Deaminase/genetics , APOBEC-1 Deaminase/metabolism , Animals , Drosophila/physiology , Mitochondria/metabolism , Mitochondria/physiology , Models, Genetic , Mutation , Organisms, Genetically Modified
7.
Autophagy ; 15(9): 1592-1605, 2019 09.
Article in English | MEDLINE | ID: mdl-30865561

ABSTRACT

The destruction of mitochondria through macroautophagy (autophagy) has been recognised as a major route of mitochondrial protein degradation since its discovery more than 50 years ago, but fundamental questions remain unanswered. First, how much mitochondrial protein turnover occurs through auto-phagy? Mitochondrial proteins are also degraded by nonautophagic mechanisms, and the proportion of mitochondrial protein turnover that occurs through autophagy is still unknown. Second, does auto-phagy degrade mitochondrial proteins uniformly or selectively? Autophagy was originally thought to degrade all mitochondrial proteins at the same rate, but recent work suggests that mitochondrial autophagy may be protein selective. To investigate these questions, we used a proteomics-based approach in the fruit fly Drosophila melanogaster, comparing mitochondrial protein turnover rates in autophagy-deficient Atg7 mutants and controls. We found that ~35% of mitochondrial protein turnover occurred via autophagy. Similar analyses using parkin mutants revealed that parkin-dependent mitophagy accounted for ~25% of mitochondrial protein turnover, suggesting that most mitochondrial autophagy specifically eliminates dysfunctional mitochondria. We also found that our results were incompatible with uniform autophagic turnover of mitochondrial proteins and consistent with protein-selective autophagy. In particular, the autophagic turnover rates of individual mitochondrial proteins varied widely, and only a small amount of the variation could be attributed to tissue differences in mitochondrial composition and autophagy rate. Furthermore, analyses comparing autophagy-deficient and control human fibroblasts revealed diverse autophagy-dependent turnover rates even in homogeneous cells. In summary, our work indicates that autophagy acts selectively on mitochondrial proteins, and that most mitochondrial protein turnover occurs through non-autophagic processes. Abbreviations:Atg5: Autophagy-related 5 (Drosophila); ATG5: autophagy related 5 (human); Atg7: Autophagy-related 7 (Drosophila); ATG7: autophagy related 7 (human); DNA: deoxyribonucleic acid; ER: endoplasmic reticulum; GFP: green fluorescent protein; MS: mass spectrometry; park: parkin (Drosophila); Pink1: PTEN-induced putative kinase 1 (Drosophila); PINK1: PTEN-induced kinase 1 (human); PRKN: parkin RBR E3 ubiquitin protein ligase (human); RNA: ribonucleic acid; SD: standard deviation; Ub: ubiquitin/ubiquitinated; WT: wild-type; YME1L: YME1 like ATPase (Drosophila); YME1L1: YME1 like 1 ATPase (human).


Subject(s)
Autophagy-Related Protein 7/metabolism , Drosophila Proteins/metabolism , Drosophila melanogaster/metabolism , Mitochondria/metabolism , Mitochondrial Proteins/metabolism , Mitophagy/genetics , Proteome/metabolism , Ubiquitin-Protein Ligases/metabolism , Animals , Autophagy-Related Protein 5/metabolism , Autophagy-Related Protein 7/genetics , Drosophila Proteins/genetics , Fibroblasts/metabolism , Humans , Models, Genetic , Organ Specificity/genetics , Proteolysis , Proteome/genetics , Ubiquitin-Protein Ligases/genetics
8.
Cell Death Dis ; 9(12): 1168, 2018 12 05.
Article in English | MEDLINE | ID: mdl-30518747

ABSTRACT

The mitochondrial Unfolded Protein Response (UPRmt) pathway confers protection from misfolded and aggregated proteins by activating factors that promote protein folding and degradation. Our recent work on Lon protease, a member of the mitochondrial ATPase Associated with diverse cellular Activities (AAA+) family of mitochondrial resident proteases, suggests that mitochondrial translational inhibition may also be a feature of the UPRmt pathway.


Subject(s)
ATPases Associated with Diverse Cellular Activities/genetics , Drosophila melanogaster/genetics , Mitochondria/genetics , Mitochondrial Proteins/genetics , Protease La/genetics , Unfolded Protein Response , ATPases Associated with Diverse Cellular Activities/metabolism , Animals , Caenorhabditis elegans/genetics , Caenorhabditis elegans/metabolism , Drosophila melanogaster/metabolism , Mitochondria/metabolism , Mitochondrial Proteins/metabolism , Protease La/metabolism , Protein Folding , Proteostasis/genetics
9.
PLoS Genet ; 14(11): e1007805, 2018 11.
Article in English | MEDLINE | ID: mdl-30452458

ABSTRACT

Mitochondrial DNA (mtDNA) mutations cause severe maternally inherited syndromes and the accumulation of somatic mtDNA mutations is implicated in aging and common diseases. However, the mechanisms that influence the frequency and pathogenicity of mtDNA mutations are poorly understood. To address this matter, we created a Drosophila mtDNA mutator strain expressing a proofreading-deficient form of the mitochondrial DNA polymerase. Mutator flies have a dramatically increased somatic mtDNA mutation frequency that correlates with the dosage of the proofreading-deficient polymerase. Mutator flies also exhibit mitochondrial dysfunction, shortened lifespan, a progressive locomotor deficit, and loss of dopaminergic neurons. Surprisingly, the frequency of nonsynonymous, pathogenic, and conserved-site mutations in mutator flies exceeded predictions of a neutral mutational model, indicating the existence of a positive selection mechanism that favors deleterious mtDNA variants. We propose from these findings that deleterious mtDNA mutations are overrepresented because they selectively evade quality control surveillance or because they are amplified through compensatory mitochondrial biogenesis.


Subject(s)
DNA Polymerase gamma/genetics , DNA Polymerase gamma/metabolism , DNA, Mitochondrial/genetics , Drosophila Proteins/genetics , Drosophila Proteins/metabolism , Drosophila melanogaster/enzymology , Drosophila melanogaster/genetics , Point Mutation , Aging/genetics , Aging/metabolism , Animals , Animals, Genetically Modified , DNA Replication/genetics , Dopaminergic Neurons/metabolism , Dopaminergic Neurons/pathology , Drosophila melanogaster/cytology , Genes, Insect , Longevity/genetics , Mitochondria/enzymology , Mitochondria/genetics , Motor Activity/genetics , Organelle Biogenesis
10.
Cell Death Discov ; 4: 51, 2018.
Article in English | MEDLINE | ID: mdl-30374414

ABSTRACT

Mitochondrial dysfunction is a frequent participant in common diseases and a principal suspect in aging. To combat mitochondrial dysfunction, eukaryotes have evolved a large repertoire of quality control mechanisms. One such mechanism involves the selective degradation of damaged or misfolded mitochondrial proteins by mitochondrial resident proteases, including proteases of the ATPase Associated with diverse cellular Activities (AAA+) family. The importance of the AAA+ family of mitochondrial proteases is exemplified by the fact that mutations that impair their functions cause a variety of human diseases, yet our knowledge of the cellular responses to their inactivation is limited. To address this matter, we created and characterized flies with complete or partial inactivation of the Drosophila matrix-localized AAA+ protease Lon. We found that a Lon null allele confers early larval lethality and that severely reducing Lon expression using RNAi results in shortened lifespan, locomotor impairment, and respiratory defects specific to respiratory chain complexes that contain mitochondrially encoded subunits. The respiratory chain defects of Lon knockdown (Lon KD ) flies appeared to result from severely reduced translation of mitochondrially encoded genes. This translational defect was not a consequence of reduced mitochondrial transcription, as evidenced by the fact that mitochondrial transcripts were elevated in abundance in Lon KD flies. Rather, the translational defect of Lon KD flies appeared to be derived from sequestration of mitochondrially encoded transcripts in highly dense ribonucleoparticles. The translational defect of Lon KD flies was also accompanied by a substantial increase in unfolded mitochondrial proteins. Together, our findings suggest that the accumulation of unfolded mitochondrial proteins triggers a stress response that culminates in the inhibition of mitochondrial translation. Our work provides a foundation to explore the underlying molecular mechanisms.

11.
PLoS Genet ; 14(9): e1007694, 2018 09.
Article in English | MEDLINE | ID: mdl-30256786

ABSTRACT

Mutations in the glucosylceramidase beta (GBA) gene are strongly associated with neurodegenerative diseases marked by protein aggregation. GBA encodes the lysosomal enzyme glucocerebrosidase, which breaks down glucosylceramide. A common explanation for the link between GBA mutations and protein aggregation is that lysosomal accumulation of glucosylceramide causes impaired autophagy. We tested this hypothesis directly by measuring protein turnover and abundance in Drosophila mutants with deletions in the GBA ortholog Gba1b. Proteomic analyses revealed that known autophagy substrates, which had severely impaired turnover in autophagy-deficient Atg7 mutants, showed little to no overall slowing of turnover or increase in abundance in Gba1b mutants. Likewise, Gba1b mutants did not have the marked impairment of mitochondrial protein turnover seen in mitophagy-deficient parkin mutants. Proteasome activity, microautophagy, and endocytic degradation also appeared unaffected in Gba1b mutants. However, we found striking changes in the turnover and abundance of proteins associated with extracellular vesicles (EVs), which have been proposed as vehicles for the spread of protein aggregates in neurodegenerative disease. These changes were specific to Gba1b mutants and did not represent an acceleration of normal aging. Western blotting of isolated EVs confirmed the increased abundance of EV proteins in Gba1b mutants, and nanoparticle tracking analysis revealed that Gba1b mutants had six times as many EVs as controls. Genetic perturbations of EV production in Gba1b mutants suppressed protein aggregation, demonstrating that the increase in EV abundance contributed to the accumulation of protein aggregates. Together, our findings indicate that glucocerebrosidase deficiency causes pathogenic changes in EV metabolism and may promote the spread of protein aggregates through extracellular vesicles.


Subject(s)
Drosophila Proteins/genetics , Extracellular Vesicles/pathology , Glucosylceramidase/deficiency , Parkinson Disease/pathology , Protein Aggregation, Pathological/pathology , Animals , Animals, Genetically Modified , Autophagy/genetics , Autophagy-Related Protein 7/genetics , Disease Models, Animal , Drosophila , Female , Glucosylceramidase/genetics , Humans , Male , Mutation , Parkinson Disease/genetics , Protein Aggregation, Pathological/genetics , Proteomics
12.
Cell Death Dis ; 9(3): 304, 2018 02 21.
Article in English | MEDLINE | ID: mdl-29467464

ABSTRACT

The progressive accumulation of dysfunctional mitochondria is implicated in aging and in common diseases of the elderly. To oppose this occurrence, organisms employ a variety of strategies, including the selective degradation of oxidatively damaged and misfolded mitochondrial proteins. Genetic studies in yeast indicate that the ATPase Associated with diverse cellular Activities (AAA+) family of mitochondrial proteases account for a substantial fraction of this protein degradation, but their metazoan counterparts have been little studied, despite the fact that mutations in the genes encoding these proteases cause a variety of human diseases. To begin to explore the biological roles of the metazoan mitochondrial AAA+ protease family, we have created a CRISPR/Cas9 allele of the Drosophila homolog of SPG7, which encodes an inner membrane-localized AAA+ protease known as paraplegin. Drosophila SPG7 mutants exhibited shortened lifespan, progressive locomotor defects, sensitivity to chemical and environmental stress, and muscular and neuronal degeneration. Ultrastructural examination of photoreceptor neurons indicated that the neurodegenerative phenotype of SPG7 mutants initiates at the synaptic terminal. A variety of mitochondrial defects accompanied the degenerative phenotypes of SPG7 mutants, including altered axonal transport of mitochondria, accumulation of electron-dense material in the matrix of flight muscle mitochondria, reduced activities of respiratory chain complexes I and II, and severely swollen and dysmorphic mitochondria in the synaptic terminals of photoreceptors. Drosophila SPG7 mutants recapitulate key features of human diseases caused by mutations in SPG7, and thus provide a foundation for the identification of Drosophila paraplegin substrates and strategies that could be used to ameliorate the symptoms of these diseases.


Subject(s)
ATPases Associated with Diverse Cellular Activities/metabolism , Drosophila Proteins/metabolism , Drosophila melanogaster/metabolism , Longevity , Metalloendopeptidases/deficiency , Mitochondria/pathology , Muscles/pathology , Nerve Degeneration/pathology , ATPases Associated with Diverse Cellular Activities/genetics , Animals , Axons/pathology , Behavior, Animal , Drosophila Proteins/genetics , Drosophila melanogaster/ultrastructure , Electron Transport , Larva , Metalloendopeptidases/metabolism , Mitochondria/metabolism , Mitochondria/ultrastructure , Mutation/genetics , Nerve Degeneration/metabolism , Sequence Homology, Amino Acid , Synapses/pathology
13.
Curr Opin Genet Dev ; 44: 47-53, 2017 Jun.
Article in English | MEDLINE | ID: mdl-28213158

ABSTRACT

Mitochondria are essential organelles that provide cellular energy and buffer cytoplasmic calcium. At the same time they produce damaging reactive oxygen species and sequester pro-apoptotic factors. Hence, eukaryotes have evolved exquisite homeostatic processes that maintain mitochondrial integrity, or ultimately remove damaged organelles. This subject has garnered intense interest recently following the discovery that two Parkinson's disease genes, PINK1 and parkin, regulate mitochondrial degradation (mitophagy). The molecular details of PINK1/Parkin-induced mitophagy are emerging but much of our insight derives from work using cultured cells and potent mitochondrial toxins, raising questions about the physiological significance of these findings. Here we review the evidence supporting PINK1/Parkin mitophagy in vivo and its causative role in neurodegeneration, and outline outstanding questions for future investigations.


Subject(s)
Nerve Degeneration/genetics , Parkinson Disease/genetics , Protein Kinases/genetics , Ubiquitin-Protein Ligases/genetics , Calcium/metabolism , Humans , Mitochondria/genetics , Mitochondria/pathology , Mitophagy/genetics , Nerve Degeneration/pathology , Parkinson Disease/metabolism , Parkinson Disease/pathology , Reactive Oxygen Species/metabolism
14.
Elife ; 52016 08 02.
Article in English | MEDLINE | ID: mdl-27481326

ABSTRACT

Due to their strict maternal inheritance in most animals and plants, mitochondrial genomes are predicted to accumulate mutations that are beneficial or neutral in females but harmful in males. Although a few male-harming mtDNA mutations have been identified, consistent with this 'Mother's Curse', their effect on females has been largely unexplored. Here, we identify COII(G177S), a mtDNA hypomorph of cytochrome oxidase II, which specifically impairs male fertility due to defects in sperm development and function without impairing other male or female functions. COII(G177S) represents one of the clearest examples of a 'male-harming' mtDNA mutation in animals and suggest that the hypomorphic mtDNA mutations like COII(G177S) might specifically impair male gametogenesis. Intriguingly, some D. melanogaster nuclear genetic backgrounds can fully rescue COII(G177S) -associated sterility, consistent with previously proposed models that nuclear genomes can regulate the phenotypic manifestation of mtDNA mutations.


Subject(s)
DNA, Mitochondrial/genetics , Drosophila melanogaster/physiology , Electron Transport Complex IV/genetics , Infertility/genetics , Mitochondrial Proteins/genetics , Mutation, Missense , Animals , Drosophila melanogaster/genetics , Male
15.
PLoS Genet ; 12(3): e1005944, 2016 Mar.
Article in English | MEDLINE | ID: mdl-27019408

ABSTRACT

Mutations in the glucosidase, beta, acid (GBA1) gene cause Gaucher's disease, and are the most common genetic risk factor for Parkinson's disease (PD) and dementia with Lewy bodies (DLB) excluding variants of low penetrance. Because α-synuclein-containing neuronal aggregates are a defining feature of PD and DLB, it is widely believed that mutations in GBA1 act by enhancing α-synuclein toxicity. To explore this hypothesis, we deleted the Drosophila GBA1 homolog, dGBA1b, and compared the phenotypes of dGBA1b mutants in the presence and absence of α-synuclein expression. Homozygous dGBA1b mutants exhibit shortened lifespan, locomotor and memory deficits, neurodegeneration, and dramatically increased accumulation of ubiquitinated protein aggregates that are normally degraded through an autophagic mechanism. Ectopic expression of human α-synuclein in dGBA1b mutants resulted in a mild enhancement of dopaminergic neuron loss and increased α-synuclein aggregation relative to controls. However, α-synuclein expression did not substantially enhance other dGBA1b mutant phenotypes. Our findings indicate that dGBA1b plays an important role in the metabolism of protein aggregates, but that the deleterious consequences of mutations in dGBA1b are largely independent of α-synuclein. Future work with dGBA1b mutants should reveal the mechanism by which mutations in dGBA1b lead to accumulation of protein aggregates, and the potential influence of this protein aggregation on neuronal integrity.


Subject(s)
Gaucher Disease/genetics , Glucosylceramidase/genetics , Nerve Degeneration/genetics , Parkinson Disease/genetics , alpha-Synuclein/genetics , Animals , Dopaminergic Neurons/metabolism , Dopaminergic Neurons/pathology , Drosophila melanogaster , Gaucher Disease/metabolism , Gaucher Disease/pathology , Glucosylceramidase/metabolism , Humans , Lysosomes/genetics , Lysosomes/pathology , Nerve Degeneration/pathology , Parkinson Disease/metabolism , Parkinson Disease/pathology , Phenotype , Protein Aggregation, Pathological
16.
Dis Model Mech ; 7(10): 1165-74, 2014 Oct.
Article in English | MEDLINE | ID: mdl-25085991

ABSTRACT

Mutations affecting mitochondrial complex I, a multi-subunit assembly that couples electron transfer to proton pumping, are the most frequent cause of heritable mitochondrial diseases. However, the mechanisms by which complex I dysfunction results in disease remain unclear. Here, we describe a Drosophila model of complex I deficiency caused by a homoplasmic mutation in the mitochondrial-DNA-encoded NADH dehydrogenase subunit 2 (ND2) gene. We show that ND2 mutants exhibit phenotypes that resemble symptoms of mitochondrial disease, including shortened lifespan, progressive neurodegeneration, diminished neural mitochondrial membrane potential and lower levels of neural ATP. Our biochemical studies of ND2 mutants reveal that complex I is unable to efficiently couple electron transfer to proton pumping. Thus, our study provides evidence that the ND2 subunit participates directly in the proton pumping mechanism of complex I. Together, our findings support the model that diminished respiratory chain activity, and consequent energy deficiency, are responsible for the pathogenesis of complex-I-associated neurodegeneration.


Subject(s)
Disease Models, Animal , Electron Transport Complex I/genetics , Mitochondrial Diseases/etiology , Mutation , Proton Pumps/metabolism , Animals , Drosophila , Electron Transport , Mitochondrial Diseases/enzymology , Mitochondrial Diseases/metabolism , Oxidative Phosphorylation , Reactive Oxygen Species/metabolism
17.
PLoS Genet ; 10(5): e1004279, 2014.
Article in English | MEDLINE | ID: mdl-24874806

ABSTRACT

Loss-of-function mutations in PINK1, which encodes a mitochondrially targeted serine/threonine kinase, result in an early-onset heritable form of Parkinson's disease. Previous work has shown that PINK1 is constitutively degraded in healthy cells, but selectively accumulates on the surface of depolarized mitochondria, thereby initiating their autophagic degradation. Although PINK1 is known to be a cleavage target of several mitochondrial proteases, whether these proteases account for the constitutive degradation of PINK1 in healthy mitochondria remains unclear. To explore the mechanism by which PINK1 is degraded, we performed a screen for mitochondrial proteases that influence PINK1 abundance in the fruit fly Drosophila melanogaster. We found that genetic perturbations targeting the matrix-localized protease Lon caused dramatic accumulation of processed PINK1 species in several mitochondrial compartments, including the matrix. Knockdown of Lon did not decrease mitochondrial membrane potential or trigger activation of the mitochondrial unfolded protein stress response (UPRmt), indicating that PINK1 accumulation in Lon-deficient animals is not a secondary consequence of mitochondrial depolarization or the UPRmt. Moreover, the influence of Lon on PINK1 abundance was highly specific, as Lon inactivation had little or no effect on the abundance of other mitochondrial proteins. Further studies indicated that the processed forms of PINK1 that accumulate upon Lon inactivation are capable of activating the PINK1-Parkin pathway in vivo. Our findings thus suggest that Lon plays an essential role in regulating the PINK1-Parkin pathway by promoting the degradation of PINK1 in the matrix of healthy mitochondria.


Subject(s)
Drosophila Proteins/genetics , Mitochondria/genetics , Parkinson Disease/genetics , Protein Serine-Threonine Kinases/genetics , Ubiquitin-Protein Ligases/metabolism , Animals , Drosophila Proteins/metabolism , Drosophila melanogaster , Membrane Potential, Mitochondrial/genetics , Mitochondria/pathology , Mutation , Parkinson Disease/metabolism , Parkinson Disease/pathology , Protease La/metabolism , Protein Serine-Threonine Kinases/metabolism , Proteolysis , Ubiquitin-Protein Ligases/genetics , Unfolded Protein Response/genetics
18.
PLoS Genet ; 10(2): e1003974, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24516391

ABSTRACT

The accumulation of somatic mitochondrial DNA (mtDNA) mutations is implicated in aging and common diseases of the elderly, including cancer and neurodegenerative disease. However, the mechanisms that influence the frequency of somatic mtDNA mutations are poorly understood. To develop a simple invertebrate model system to address this matter, we used the Random Mutation Capture (RMC) assay to characterize the age-dependent frequency and distribution of mtDNA mutations in the fruit fly Drosophila melanogaster. Because oxidative stress is a major suspect in the age-dependent accumulation of somatic mtDNA mutations, we also used the RMC assay to explore the influence of oxidative stress on the somatic mtDNA mutation frequency. We found that many of the features associated with mtDNA mutations in vertebrates are conserved in Drosophila, including a comparable somatic mtDNA mutation frequency (∼10(-5)), an increased frequency of mtDNA mutations with age, and a prevalence of transition mutations. Only a small fraction of the mtDNA mutations detected in young or old animals were G∶C to T∶A transversions, a signature of oxidative damage, and loss-of-function mutations in the mitochondrial superoxide dismutase, Sod2, had no detectable influence on the somatic mtDNA mutation frequency. Moreover, a loss-of-function mutation in Ogg1, which encodes a DNA repair enzyme that removes oxidatively damaged deoxyguanosine residues (8-hydroxy-2'-deoxyguanosine), did not significantly influence the somatic mtDNA mutation frequency of Sod2 mutants. Together, these findings indicate that oxidative stress is not a major cause of somatic mtDNA mutations. Our data instead suggests that somatic mtDNA mutations arise primarily from errors that occur during mtDNA replication. Further studies using Drosophila should aid in the identification of factors that influence the frequency of somatic mtDNA mutations.


Subject(s)
Aging/genetics , DNA, Mitochondrial/genetics , Mutation/genetics , Oxidative Stress , Aging/pathology , Animals , DNA Glycosylases/genetics , DNA Repair/genetics , Drosophila Proteins/genetics , Drosophila melanogaster , Humans , Mitochondria/genetics , Mitochondria/pathology , Models, Animal , Mutation Rate , Reactive Oxygen Species/metabolism , Superoxide Dismutase/genetics
19.
Proc Natl Acad Sci U S A ; 110(16): 6400-5, 2013 Apr 16.
Article in English | MEDLINE | ID: mdl-23509287

ABSTRACT

The accumulation of damaged mitochondria has been proposed as a key factor in aging and the pathogenesis of many common age-related diseases, including Parkinson disease (PD). Recently, in vitro studies of the PD-related proteins Parkin and PINK1 have found that these factors act in a common pathway to promote the selective autophagic degradation of damaged mitochondria (mitophagy). However, whether Parkin and PINK1 promote mitophagy under normal physiological conditions in vivo is unknown. To address this question, we used a proteomic approach in Drosophila to compare the rates of mitochondrial protein turnover in parkin mutants, PINK1 mutants, and control flies. We found that parkin null mutants showed a significant overall slowing of mitochondrial protein turnover, similar to but less severe than the slowing seen in autophagy-deficient Atg7 mutants, consistent with the model that Parkin acts upstream of Atg7 to promote mitophagy. By contrast, the turnover of many mitochondrial respiratory chain (RC) subunits showed greater impairment in parkin than Atg7 mutants, and RC turnover was also selectively impaired in PINK1 mutants. Our findings show that the PINK1-Parkin pathway promotes mitophagy in vivo and, unexpectedly, also promotes selective turnover of mitochondrial RC subunits. Failure to degrade damaged RC proteins could account for the RC deficits seen in many PD patients and may play an important role in PD pathogenesis.


Subject(s)
Drosophila Proteins/metabolism , Electron Transport/physiology , Mitochondrial Proteins/metabolism , Mitophagy/physiology , Parkinson Disease/etiology , Protein Serine-Threonine Kinases/metabolism , Signal Transduction/physiology , Ubiquitin-Protein Ligases/metabolism , Animals , Autophagy-Related Protein 7 , Brain/metabolism , Drosophila , Half-Life , Isotope Labeling , Mass Spectrometry , Mice , Parkinson Disease/metabolism
20.
J Cell Biol ; 191(7): 1225-7, 2010 Dec 27.
Article in English | MEDLINE | ID: mdl-21187326

ABSTRACT

The PINK1-Parkin pathway plays a critical role in mitochondrial quality control by selectively targeting damaged mitochondria for autophagy. In this issue, Tanaka et al. (2010. J. Cell Biol. doi: 10.1083/jcb.201007013) demonstrate that the AAA-type ATPase p97 acts downstream of PINK1 and Parkin to segregate fusion-incompetent mitochondria for turnover. p97 acts by targeting the mitochondrial fusion-promoting factor mitofusin for degradation through an endoplasmic reticulum-associated degradation (ERAD)-like mechanism.


Subject(s)
Adenosine Triphosphatases/metabolism , Autophagy/physiology , Mitochondria/physiology , Nuclear Proteins/metabolism , Proteasome Endopeptidase Complex/metabolism , Protein Kinases/metabolism , Ubiquitin-Protein Ligases/metabolism , Animals , Autophagy/drug effects , Carbonyl Cyanide m-Chlorophenyl Hydrazone/pharmacology , Dynamins , Endoplasmic Reticulum/metabolism , GTP Phosphohydrolases/genetics , GTP Phosphohydrolases/metabolism , Humans , Membrane Fusion/drug effects , Membrane Fusion/physiology , Membrane Proteins/metabolism , Membrane Transport Proteins/metabolism , Mice , Microtubule-Associated Proteins/genetics , Mitochondria/drug effects , Mitochondrial Membrane Transport Proteins , Mitochondrial Proteins/genetics , Mitochondrial Proteins/metabolism , Models, Biological , Ubiquitination/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...