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










Publication year range
1.
Cell Death Dis ; 15(3): 203, 2024 Mar 11.
Article in English | MEDLINE | ID: mdl-38467609

ABSTRACT

Organelles form membrane contact sites between each other, allowing for the transfer of molecules and signals. Mitochondria-endoplasmic reticulum (ER) contact sites (MERCS) are cellular subdomains characterized by close apposition of mitochondria and ER membranes. They have been implicated in many diseases, including neurodegenerative, metabolic, and cardiac diseases. Although MERCS have been extensively studied, much remains to be explored. To uncover novel regulators of MERCS, we conducted a genome-wide, flow cytometry-based screen using an engineered MERCS reporter cell line. We found 410 genes whose downregulation promotes MERCS and 230 genes whose downregulation decreases MERCS. From these, 29 genes were selected from each population for arrayed screening and 25 were validated from the high population and 13 from the low population. GET4 and BAG6 were highlighted as the top 2 genes that upon suppression increased MERCS from both the pooled and arrayed screens, and these were subjected to further investigation. Multiple microscopy analyses confirmed that loss of GET4 or BAG6 increased MERCS. GET4 and BAG6 were also observed to interact with the known MERCS proteins, inositol 1,4,5-trisphosphate receptors (IP3R) and glucose-regulated protein 75 (GRP75). In addition, we found that loss of GET4 increased mitochondrial calcium uptake upon ER-Ca2+ release and mitochondrial respiration. Finally, we show that loss of GET4 rescues motor ability, improves lifespan and prevents neurodegeneration in a Drosophila model of Alzheimer's disease (Aß42Arc). Together, these results suggest that GET4 is involved in decreasing MERCS and that its loss is neuroprotective.


Subject(s)
CRISPR-Cas Systems , Mitochondria Associated Membranes , CRISPR-Cas Systems/genetics , Mitochondria/genetics , Mitochondria/metabolism , Mitochondrial Membranes/metabolism , Endoplasmic Reticulum/genetics , Endoplasmic Reticulum/metabolism , Calcium/metabolism
2.
Sci Rep ; 13(1): 20616, 2023 11 23.
Article in English | MEDLINE | ID: mdl-37996473

ABSTRACT

Biological pathways between alcohol consumption and alcohol liver disease (ALD) are not fully understood. We selected genes with known effect on (1) alcohol consumption, (2) liver function, and (3) gene expression. Expression of the orthologs of these genes in Caenorhabditis elegans and Drosophila melanogaster was suppressed using mutations and/or RNA interference (RNAi). In humans, association analysis, pathway analysis, and Mendelian randomization analysis were performed to identify metabolic changes due to alcohol consumption. In C. elegans, we found a reduction in locomotion rate after exposure to ethanol for RNAi knockdown of ACTR1B and MAPT. In Drosophila, we observed (1) a change in sedative effect of ethanol for RNAi knockdown of WDPCP, TENM2, GPN1, ARPC1B, and SCN8A, (2) a reduction in ethanol consumption for RNAi knockdown of TENM2, (3) a reduction in triradylglycerols (TAG) levels for RNAi knockdown of WDPCP, TENM2, and GPN1. In human, we observed (1) a link between alcohol consumption and several metabolites including TAG, (2) an enrichment of the candidate (alcohol-associated) metabolites within the linoleic acid (LNA) and alpha-linolenic acid (ALA) metabolism pathways, (3) a causal link between gene expression of WDPCP to liver fibrosis and liver cirrhosis. Our results imply that WDPCP might be involved in ALD.


Subject(s)
Caenorhabditis elegans , Drosophila melanogaster , Lipid Metabolism , Liver Diseases, Alcoholic , Animals , Humans , Alcohol Drinking/genetics , Caenorhabditis elegans/genetics , Drosophila melanogaster/genetics , Ethanol/metabolism , Lipid Metabolism/genetics , Liver/metabolism , Liver Cirrhosis/pathology , Liver Diseases, Alcoholic/metabolism
3.
Biol Direct ; 18(1): 43, 2023 08 01.
Article in English | MEDLINE | ID: mdl-37528429

ABSTRACT

Antipsychotic drugs are the mainstay of treatment for schizophrenia and provide adjunct therapies for other prevalent psychiatric conditions, including bipolar disorder and major depressive disorder. However, they also induce debilitating extrapyramidal syndromes (EPS), such as Parkinsonism, in a significant minority of patients. The majority of antipsychotic drugs function as dopamine receptor antagonists in the brain while the most recent 'third'-generation, such as aripiprazole, act as partial agonists. Despite showing good clinical efficacy, these newer agents are still associated with EPS in ~ 5 to 15% of patients. However, it is not fully understood how these movement disorders develop. Here, we combine clinically-relevant drug concentrations with mutliscale model systems to show that aripiprazole and its primary active metabolite induce mitochondrial toxicity inducing robust declines in cellular ATP and viability. Aripiprazole, brexpiprazole and cariprazine were shown to directly inhibit respiratory complex I through its ubiquinone-binding channel. Importantly, all three drugs induced mitochondrial toxicity in primary embryonic mouse neurons, with greater bioenergetic inhibition in ventral midbrain neurons than forebrain neurons. Finally, chronic feeding with aripiprazole resulted in structural damage to mitochondria in the brain and thoracic muscle of adult Drosophila melanogaster consistent with locomotor dysfunction. Taken together, we show that antipsychotic drugs acting as partial dopamine receptor agonists exhibit off-target mitochondrial liabilities targeting complex I.


Subject(s)
Antipsychotic Agents , Depressive Disorder, Major , Animals , Mice , Aripiprazole/pharmacology , Aripiprazole/therapeutic use , Antipsychotic Agents/pharmacology , Antipsychotic Agents/therapeutic use , Depressive Disorder, Major/drug therapy , Drosophila melanogaster , Electron Transport
4.
Dis Model Mech ; 16(4)2023 04 01.
Article in English | MEDLINE | ID: mdl-37083954

ABSTRACT

Eukaryotic Tribbles proteins are pseudoenzymes that regulate multiple aspects of intracellular signalling. Both Drosophila melanogaster and mammalian members of this family of pseudokinases act as negative regulators of insulin signalling. Mammalian tribbles pseudokinase (TRIB) genes have also been linked to insulin resistance and type 2 diabetes mellitus. Type 2 diabetes mellitus is associated with increased body weight, sleep problems and increased long-term mortality. Here, we investigated how manipulating the expression of Tribbles impacts body weight, sleep and mortality. We showed that the overexpression of Drosophila tribbles (trbl) in the fly fat body reduces both body weight and lifespan in adult flies without affecting food intake. Furthermore, it decreases the levels of Drosophila insulin-like peptide 2 (DILP2; ILP2) and increases night-time sleep. The three genes encoding TRIBs of mammals, TRIB1, TRIB2 and TRIB3, show both common and unique features. As the three human TRIB genes share features with Drosophila trbl, we further explored the links between TRIB genetic variants and both body weight and sleep in the human population. We identified associations between the polymorphisms and expression levels of the pseudokinases and markers of body weight and sleep duration. We conclude that Tribbles pseudokinases are involved in the control of body weight, lifespan and sleep.


Subject(s)
Diabetes Mellitus, Type 2 , Neuropeptides , Animals , Humans , Body Weight , Calcium-Calmodulin-Dependent Protein Kinases/genetics , Calcium-Calmodulin-Dependent Protein Kinases/metabolism , Cell Cycle Proteins/genetics , Drosophila melanogaster/genetics , Drosophila melanogaster/metabolism , Insulin/metabolism , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/metabolism , Mammals/metabolism , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Sleep Duration , Up-Regulation/genetics
5.
Cell Death Dis ; 14(3): 206, 2023 03 22.
Article in English | MEDLINE | ID: mdl-36949073

ABSTRACT

Parkinson's disease (PD) is characterised by selective death of dopaminergic (DA) neurons in the midbrain and motor function impairment. Gastrointestinal issues often precede motor deficits in PD, indicating that the gut-brain axis is involved in the pathogenesis of this disease. The features of PD include both mitochondrial dysfunction and activation of the unfolded protein response (UPR) in the endoplasmic reticulum (ER). PINK1 is a mitochondrial kinase involved in the recycling of defective mitochondria, and PINK1 mutations cause early-onset PD. Like PD patients, pink1 mutant Drosophila show degeneration of DA neurons and intestinal dysfunction. These mutant flies also lack vital proteins due to sustained activation of the kinase R-like endoplasmic reticulum kinase (dPerk), a kinase that induces the UPR. Here, we investigated the role of dPerk in intestinal dysfunction. We showed that intestinal expression of dPerk impairs mitochondrial function, induces cell death, and decreases lifespan. We found that suppressing dPerk in the intestine of pink1-mutant flies rescues intestinal cell death and is neuroprotective. We conclude that in a fly model of PD, blocking gut-brain transmission of UPR-mediated toxicity, is neuroprotective.


Subject(s)
Drosophila Proteins , Parkinson Disease , Animals , Drosophila/metabolism , Drosophila melanogaster/genetics , Drosophila melanogaster/metabolism , Drosophila Proteins/genetics , Drosophila Proteins/metabolism , Parkinson Disease/metabolism , Protein Serine-Threonine Kinases/metabolism , Unfolded Protein Response
6.
Geroscience ; 44(4): 1961-1974, 2022 08.
Article in English | MEDLINE | ID: mdl-35355221

ABSTRACT

Mitochondrial reactive oxygen species (mtROS) are cellular messengers essential for cellular homeostasis. In response to stress, reverse electron transport (RET) through respiratory complex I generates high levels of mtROS. Suppression of ROS production via RET (ROS-RET) reduces survival under stress, while activation of ROS-RET extends lifespan in basal conditions. Here, we demonstrate that ROS-RET signalling requires increased electron entry and uninterrupted electron flow through the electron transport chain (ETC). We find that in old fruit flies, ROS-RET is abolished when electron flux is decreased and that their mitochondria produce consistently high levels of mtROS. Finally, we demonstrate that in young flies, limiting electron exit, but not entry, from the ETC phenocopies mtROS generation observed in old individuals. Our results elucidate the mechanism by which ROS signalling is lost during ageing.


Subject(s)
Diptera , Electrons , Animals , Reactive Oxygen Species , Electron Transport , Aging
7.
Nat Aging ; 2(4): 317-331, 2022 04.
Article in English | MEDLINE | ID: mdl-37117744

ABSTRACT

The innate immune response mounts a defense against foreign invaders and declines with age. An inappropriate induction of this response can cause diseases. Previous studies showed that mitochondria can be repurposed to promote inflammatory signaling. Damaged mitochondria can also trigger inflammation and promote diseases. Mutations in pink1, a gene required for mitochondrial health, cause Parkinson's disease, and Drosophila melanogaster pink1 mutants accumulate damaged mitochondria. Here, we show that defective mitochondria in pink1 mutants activate Relish targets and demonstrate that inflammatory signaling causes age-dependent intestinal dysfunction in pink1-mutant flies. These effects result in the death of intestinal cells, metabolic reprogramming and neurotoxicity. We found that Relish signaling is activated downstream of a pathway stimulated by cytosolic DNA. Suppression of Relish in the intestinal midgut of pink1-mutant flies restores mitochondrial function and is neuroprotective. We thus conclude that gut-brain communication modulates neurotoxicity in a fly model of Parkinson's disease through a mechanism involving mitochondrial dysfunction.


Subject(s)
Drosophila Proteins , Gastrointestinal Diseases , Intestinal Diseases , Parkinson Disease , Animals , Drosophila/metabolism , Drosophila melanogaster/genetics , Drosophila Proteins/genetics , Protein Serine-Threonine Kinases/genetics , Parkinson Disease/genetics
9.
Cell Death Dis ; 12(7): 651, 2021 06 25.
Article in English | MEDLINE | ID: mdl-34172715

ABSTRACT

Alzheimer's disease is the most common age-related neurodegenerative disorder. Familial forms of Alzheimer's disease associated with the accumulation of a toxic form of amyloid-ß (Aß) peptides are linked to mitochondrial impairment. The coenzyme nicotinamide adenine dinucleotide (NAD+) is essential for both mitochondrial bioenergetics and nuclear DNA repair through NAD+-consuming poly (ADP-ribose) polymerases (PARPs). Here we analysed the metabolomic changes in flies overexpressing Aß and showed a decrease of metabolites associated with nicotinate and nicotinamide metabolism, which is critical for mitochondrial function in neurons. We show that increasing the bioavailability of NAD+ protects against Aß toxicity. Pharmacological supplementation using NAM, a form of vitamin B that acts as a precursor for NAD+ or a genetic mutation of PARP rescues mitochondrial defects, protects neurons against degeneration and reduces behavioural impairments in a fly model of Alzheimer's disease. Next, we looked at links between PARP polymorphisms and vitamin B intake in patients with Alzheimer's disease. We show that polymorphisms in the human PARP1 gene or the intake of vitamin B are associated with a decrease in the risk and severity of Alzheimer's disease. We suggest that enhancing the availability of NAD+ by either vitamin B supplements or the inhibition of NAD+-dependent enzymes such as PARPs are potential therapies for Alzheimer's disease.


Subject(s)
Alzheimer Disease/genetics , Drosophila Proteins/genetics , Mitochondria/genetics , Mutation , NAD/metabolism , Neurons/enzymology , Poly (ADP-Ribose) Polymerase-1/genetics , Alzheimer Disease/drug therapy , Alzheimer Disease/enzymology , Alzheimer Disease/pathology , Amyloid beta-Peptides/genetics , Amyloid beta-Peptides/metabolism , Animals , Animals, Genetically Modified , Behavior, Animal , Disease Models, Animal , Drosophila Proteins/metabolism , Drosophila melanogaster/genetics , Humans , Metabolome , Metabolomics , Mitochondria/drug effects , Mitochondria/enzymology , Mitochondria/ultrastructure , Motor Activity , Nerve Degeneration , Neurons/drug effects , Neurons/pathology , Niacinamide/pharmacology , Poly (ADP-Ribose) Polymerase-1/metabolism , Polymorphism, Single Nucleotide
10.
Int J Mol Sci ; 22(9)2021 Apr 27.
Article in English | MEDLINE | ID: mdl-33925631

ABSTRACT

In Drosophila, endoplasmic reticulum (ER) stress activates the protein kinase R-like endoplasmic reticulum kinase (dPerk). dPerk can also be activated by defective mitochondria in fly models of Parkinson's disease caused by mutations in pink1 or parkin. The Perk branch of the unfolded protein response (UPR) has emerged as a major toxic process in neurodegenerative disorders causing a chronic reduction in vital proteins and neuronal death. In this study, we combined microarray analysis and quantitative proteomics analysis in adult flies overexpressing dPerk to investigate the relationship between the transcriptional and translational response to dPerk activation. We identified tribbles and Heat shock protein 22 as two novel Drosophila activating transcription factor 4 (dAtf4) regulated transcripts. Using a combined bioinformatics tool kit, we demonstrated that the activation of dPerk leads to translational repression of mitochondrial proteins associated with glutathione and nucleotide metabolism, calcium signalling and iron-sulphur cluster biosynthesis. Further efforts to enhance these translationally repressed dPerk targets might offer protection against Perk toxicity.


Subject(s)
eIF-2 Kinase/genetics , eIF-2 Kinase/metabolism , Activating Transcription Factor 4/metabolism , Animals , Cell Cycle Proteins/metabolism , Computational Biology/methods , Drosophila Proteins/metabolism , Drosophila melanogaster , Endoplasmic Reticulum/pathology , Endoplasmic Reticulum Stress , Eukaryotic Initiation Factor-2/metabolism , Heat-Shock Proteins/metabolism , Mitochondria/metabolism , Neurodegenerative Diseases/metabolism , Phosphorylation , Protein Processing, Post-Translational , Protein Serine-Threonine Kinases/metabolism , Proteomics/methods , Signal Transduction , Transcription Factors/metabolism , Transcriptome , Ubiquitin-Protein Ligases/metabolism , Unfolded Protein Response/genetics , Unfolded Protein Response/physiology
11.
Redox Biol ; 32: 101450, 2020 05.
Article in English | MEDLINE | ID: mdl-32146156

ABSTRACT

Reactive Oxygen Species (ROS) are essential cellular messengers required for cellular homeostasis and regulate the lifespan of several animal species. The main site of ROS production is the mitochondrion, and within it, respiratory complex I (CI) is the main ROS generator. ROS produced by CI trigger several physiological responses that are essential for the survival of neurons, cardiomyocytes and macrophages. Here, we show that CI produces ROS when electrons flow in either the forward (Forward Electron Transport, FET) or reverse direction (Reverse Electron Transport, RET). We demonstrate that ROS production via RET (ROS-RET) is activated under thermal stress conditions and that interruption of ROS-RET production, through ectopic expression of the alternative oxidase AOX, attenuates the activation of pro-survival pathways in response to stress. Accordingly, we find that both suppressing ROS-RET signalling or decreasing levels of mitochondrial H2O2 by overexpressing mitochondrial catalase (mtCAT), reduces survival dramatically in flies under stress. Our results uncover a specific ROS signalling pathway where hydrogen peroxide (H2O2) generated by CI via RET is required to activate adaptive mechanisms, maximising survival under stress conditions.


Subject(s)
Drosophila melanogaster , Electron Transport Complex I , Animals , Drosophila melanogaster/genetics , Drosophila melanogaster/metabolism , Electron Transport , Electron Transport Complex I/genetics , Electron Transport Complex I/metabolism , Hydrogen Peroxide , Reactive Oxygen Species/metabolism
12.
Biol Open ; 9(1)2020 01 14.
Article in English | MEDLINE | ID: mdl-31822473

ABSTRACT

Eukaryotic cells are complex systems containing internal compartments with specialised functions. Among these compartments, the endoplasmic reticulum (ER) plays a major role in processing proteins for modification and delivery to other organelles, whereas mitochondria generate energy in the form of ATP. Mitochondria and the ER form physical interactions, defined as mitochondria-ER contact sites (MERCs) to exchange metabolites such as calcium ions (Ca2+) and lipids. Sites of contact between mitochondria and the ER can regulate biological processes such as ATP generation and mitochondrial division. The interactions between mitochondria and the ER are dynamic and respond to the metabolic state of cells. Changes in MERCs have been linked to metabolic pathologies such as diabetes, neurodegenerative diseases and sleep disruption. Here we explored the consequences of increasing contacts between mitochondria and the ER in flies using a synthetic linker. We showed that enhancing MERCs increases locomotion and extends lifespan. We also showed that, in a Drosophila model of Alzheimer's disease linked to toxic amyloid beta (Aß), linker expression can suppress motor impairment and extend lifespan. We conclude that strategies for increasing contacts between mitochondria and the ER may improve symptoms of diseases associated with mitochondria dysfunction. A video abstract for this article is available at https://youtu.be/_YWA4oKZkes.This article has an associated First Person interview with the first author of the paper.


Subject(s)
Alzheimer Disease/metabolism , Endoplasmic Reticulum/metabolism , Mitochondria/metabolism , Alzheimer Disease/etiology , Alzheimer Disease/pathology , Animals , Biological Transport , Calcium/metabolism , Disease Models, Animal , Disease Susceptibility , Drosophila , Endoplasmic Reticulum/ultrastructure , Fluorescent Antibody Technique , Gene Expression , Genes, Reporter , Humans , Locomotion , Mitochondria/ultrastructure , Reactive Oxygen Species/metabolism , Signal Transduction
13.
Cell Death Dis ; 10(4): 288, 2019 03 25.
Article in English | MEDLINE | ID: mdl-30911005

ABSTRACT

Mutations in the mitochondrial GTPase mitofusin 2 (MFN2) cause Charcot-Marie-Tooth disease type 2 (CMT2A), a form of peripheral neuropathy that compromises axonal function. Mitofusins promote mitochondrial fusion and regulate mitochondrial dynamics. They are also reported to be involved in forming contacts between mitochondria and the endoplasmic reticulum. The fruit fly, Drosophila melanogaster, is a powerful tool to model human neurodegenerative diseases, including CMT2A. Here, we have downregulated the expression of the Drosophila mitofusin (dMfn RNAi) in adult flies and showed that this activates mitochondrial retrograde signalling and is associated with an upregulation of genes involved in folic acid (FA) metabolism. Additionally, we demonstrated that pharmacological and genetic interventions designed to increase the FA metabolism pathway suppresses the phenotype of the dMfn RNAi flies. We conclude that strategies to increase FA metabolism may ameliorate diseases, such as peripheral neuropathies, that are associated with loss of mitochondrial function. A video abstract for this article is available at  https://youtu.be/fs1G-QRo6xI .


Subject(s)
Down-Regulation/genetics , Drosophila Proteins/genetics , Drosophila Proteins/metabolism , Drosophila/metabolism , Folic Acid/metabolism , Membrane Proteins/genetics , Membrane Proteins/metabolism , Activating Transcription Factor 4/metabolism , Animals , Axonal Transport/genetics , Charcot-Marie-Tooth Disease/metabolism , Disease Models, Animal , Folic Acid/genetics , Locomotion/genetics , Male , Mitochondria/metabolism , Phenotype , RNA Interference , Reactive Oxygen Species/metabolism
14.
Cell Death Differ ; 26(9): 1861, 2019 Sep.
Article in English | MEDLINE | ID: mdl-30546072

ABSTRACT

Following publication of the article, Dr. Roberta Tufi of the Mitochondrial Biology Unit at the University of Cambridge was concerned to note that her own contribution to the study during her postdoc in Leicester at the MRC Toxicology Unit had not been acknowledged. Specifically, the data in Fig. 1 (panels a, b, and d) were produced though her work.

16.
Cell Rep ; 19(9): 1739-1749, 2017 05 30.
Article in English | MEDLINE | ID: mdl-28564594

ABSTRACT

Motor neurons (MNs) and astrocytes (ACs) are implicated in the pathogenesis of amyotrophic lateral sclerosis (ALS), but their interaction and the sequence of molecular events leading to MN death remain unresolved. Here, we optimized directed differentiation of induced pluripotent stem cells (iPSCs) into highly enriched (> 85%) functional populations of spinal cord MNs and ACs. We identify significantly increased cytoplasmic TDP-43 and ER stress as primary pathogenic events in patient-specific valosin-containing protein (VCP)-mutant MNs, with secondary mitochondrial dysfunction and oxidative stress. Cumulatively, these cellular stresses result in synaptic pathology and cell death in VCP-mutant MNs. We additionally identify a cell-autonomous VCP-mutant AC survival phenotype, which is not attributable to the same molecular pathology occurring in VCP-mutant MNs. Finally, through iterative co-culture experiments, we uncover non-cell-autonomous effects of VCP-mutant ACs on both control and mutant MNs. This work elucidates molecular events and cellular interplay that could guide future therapeutic strategies in ALS.


Subject(s)
Amyotrophic Lateral Sclerosis/metabolism , Amyotrophic Lateral Sclerosis/pathology , Astrocytes/pathology , Models, Biological , Motor Neurons/pathology , Valosin Containing Protein/metabolism , Cell Survival , DNA-Binding Proteins/metabolism , Endoplasmic Reticulum/metabolism , Endoplasmic Reticulum/ultrastructure , Endoplasmic Reticulum Stress , Humans , Induced Pluripotent Stem Cells/metabolism , Membrane Potential, Mitochondrial , Mitochondria/metabolism , Mitochondria/ultrastructure , Mutation/genetics , Nerve Degeneration/pathology , Neurogenesis , Oxidative Stress , Phenotype , Synapses/pathology
17.
Biol Open ; 6(2): 141-147, 2017 Feb 15.
Article in English | MEDLINE | ID: mdl-28011627

ABSTRACT

Familial forms of Parkinson's disease (PD) caused by mutations in PINK1 are linked to mitochondrial impairment. Defective mitochondria are also found in Drosophila models of PD with pink1 mutations. The co-enzyme nicotinamide adenine dinucleotide (NAD+) is essential for both generating energy in mitochondria and nuclear DNA repair through NAD+-consuming poly(ADP-ribose) polymerases (PARPs). We found alterations in NAD+ salvage metabolism in Drosophila pink1 mutants and showed that a diet supplemented with the NAD+ precursor nicotinamide rescued mitochondrial defects and protected neurons from degeneration. Additionally, a mutation of Parp improved mitochondrial function and was neuroprotective in the pink1 mutants. We conclude that enhancing the availability of NAD+ by either the use of a diet supplemented with NAD+ precursors or the inhibition of NAD+-dependent enzymes, such as PARPs, which compete with mitochondria for NAD+, is a viable approach to preventing neurotoxicity associated with mitochondrial defects.

18.
Nat Cell Biol ; 16(2): 157-66, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24441527

ABSTRACT

Mutations in PINK1 cause early-onset Parkinson's disease (PD). Studies in Drosophila melanogaster have highlighted mitochondrial dysfunction on loss of Pink1 as a central mechanism of PD pathogenesis. Here we show that global analysis of transcriptional changes in Drosophila pink1 mutants reveals an upregulation of genes involved in nucleotide metabolism, critical for neuronal mitochondrial DNA synthesis. These key transcriptional changes were also detected in brains of PD patients harbouring PINK1 mutations. We demonstrate that genetic enhancement of the nucleotide salvage pathway in neurons of pink1 mutant flies rescues mitochondrial impairment. In addition, pharmacological approaches enhancing nucleotide pools reduce mitochondrial dysfunction caused by Pink1 deficiency. We conclude that loss of Pink1 evokes the activation of a previously unidentified metabolic reprogramming pathway to increase nucleotide pools and promote mitochondrial biogenesis. We propose that targeting strategies enhancing nucleotide synthesis pathways may reverse mitochondrial dysfunction and rescue neurodegeneration in PD and, potentially, other diseases linked to mitochondrial impairment.


Subject(s)
Disease Models, Animal , Drosophila Proteins/physiology , Mitochondria/physiology , Mutation , Nucleotides/metabolism , Parkinson Disease/physiopathology , Protein Serine-Threonine Kinases/physiology , Animals , DNA, Mitochondrial/biosynthesis , Drosophila Proteins/genetics , Drosophila melanogaster , Parkinson Disease/genetics , Parkinson Disease/metabolism , Protein Serine-Threonine Kinases/genetics
19.
Hum Mol Genet ; 20(5): 867-79, 2011 Mar 01.
Article in English | MEDLINE | ID: mdl-21138942

ABSTRACT

Mutations in PTEN-induced kinase 1 (PINK1) cause early onset autosomal recessive Parkinson's disease (PD). PINK1 is a 63 kDa protein kinase, which exerts a neuroprotective function and is known to localize to mitochondria. Upon entry into the organelle, PINK1 is cleaved to produce a ∼53 kDa protein (ΔN-PINK1). In this paper, we show that PINK1 is cleaved between amino acids Ala-103 and Phe-104 to generate ΔN-PINK1. We demonstrate that a reduced ability to cleave PINK1, and the consequent accumulation of full-length protein, results in mitochondrial abnormalities reminiscent of those observed in PINK1 knockout cells, including disruption of the mitochondrial network and a reduction in mitochondrial mass. Notably, we assessed three N-terminal PD-associated PINK1 mutations located close to the cleavage site and, while these do not prevent PINK1 cleavage, they alter the ratio of full-length to ΔN-PINK1 protein in cells, resulting in an altered mitochondrial phenotype. Finally, we show that PINK1 interacts with the mitochondrial protease presenilin-associated rhomboid-like protein (PARL) and that loss of PARL results in aberrant PINK1 cleavage in mammalian cells. These combined results suggest that PINK1 cleavage is important for basal mitochondrial health and that PARL cleaves PINK1 to produce the ΔN-PINK1 fragment.


Subject(s)
Metalloproteases/metabolism , Mitochondria/enzymology , Mitochondrial Proteins/metabolism , Protein Kinases/chemistry , Protein Kinases/metabolism , Amino Acid Sequence , Cell Line , Humans , Metalloproteases/genetics , Mitochondria/chemistry , Mitochondria/metabolism , Mitochondrial Proteins/genetics , Molecular Sequence Data , Mutation , Parkinson Disease/enzymology , Parkinson Disease/genetics , Parkinson Disease/metabolism , Parkinsonian Disorders , Protein Binding , Protein Kinases/genetics , Protein Processing, Post-Translational , Sequence Alignment
20.
Proc Natl Acad Sci U S A ; 106(29): 11978-83, 2009 Jul 21.
Article in English | MEDLINE | ID: mdl-19587239

ABSTRACT

Intracellular signal transduction networks involving protein kinases are important modulators of cell survival and cell death in multicellular organisms. Functional compromise of these networks has been linked to aberrant apoptosis in diseases such as cancer. To identify novel kinase regulators of cell death, we conducted an RNAi-based screen to identify modulators of the intrinsic apoptosis pathway. Using this approach, we identified MAP4K3 as a novel apoptosis inducer. Here, we present evidence that this pro-apoptotic kinase orchestrates activation of BAX via the concerted posttranscriptional modulation of PUMA, BAD, and BIM. Additionally, we found decreased levels of this kinase in pancreatic cancer samples, suggesting a tumor suppressor role for MAP4K3 in pancreatic tumorigenesis.


Subject(s)
Gene Expression Regulation , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins c-bcl-2/metabolism , Transcription, Genetic , Amino Acid Sequence , Apoptosis Regulatory Proteins/genetics , Apoptosis Regulatory Proteins/metabolism , Bcl-2-Like Protein 11 , Cell Death , Cell Line , DNA Damage , Humans , JNK Mitogen-Activated Protein Kinases/metabolism , Mechanistic Target of Rapamycin Complex 1 , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mitochondria/enzymology , Models, Biological , Molecular Sequence Data , Multiprotein Complexes , Pancreatic Neoplasms/enzymology , Pancreatic Neoplasms/pathology , Protein Serine-Threonine Kinases/antagonists & inhibitors , Protein Serine-Threonine Kinases/chemistry , Protein Serine-Threonine Kinases/genetics , Proteins , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/metabolism , RNA Interference , Signal Transduction , TOR Serine-Threonine Kinases , Transcription Factors/metabolism , bcl-Associated Death Protein/genetics , bcl-Associated Death Protein/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL