Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 44
Filter
1.
EMBO J ; 40(8): e106283, 2021 04 15.
Article in English | MEDLINE | ID: mdl-33665835

ABSTRACT

Mitochondrial DNA (mtDNA) encodes several key components of respiratory chain complexes that produce cellular energy through oxidative phosphorylation. mtDNA is vulnerable to damage under various physiological stresses, especially oxidative stress. mtDNA damage leads to mitochondrial dysfunction, and dysfunctional mitochondria can be removed by mitophagy, an essential process in cellular homeostasis. However, how damaged mtDNA is selectively cleared from the cell, and how damaged mtDNA triggers mitophagy, remain mostly unknown. Here, we identified a novel mitophagy receptor, ATAD3B, which is specifically expressed in primates. ATAD3B contains a LIR motif that binds to LC3 and promotes oxidative stress-induced mitophagy in a PINK1-independent manner, thus promoting the clearance of damaged mtDNA induced by oxidative stress. Under normal conditions, ATAD3B hetero-oligomerizes with ATAD3A, thus promoting the targeting of the C-terminal region of ATAD3B to the mitochondrial intermembrane space. Oxidative stress-induced mtDNA damage or mtDNA depletion reduces ATAD3B-ATAD3A hetero-oligomerization and leads to exposure of the ATAD3B C-terminus at the mitochondrial outer membrane and subsequent recruitment of LC3 for initiating mitophagy. Furthermore, ATAD3B is little expressed in m.3243A > G mutated cells and MELAS patient fibroblasts showing endogenous oxidative stress, and ATAD3B re-expression promotes the clearance of m.3243A > G mutated mtDNA. Our findings uncover a new pathway to selectively remove damaged mtDNA and reveal that increasing ATAD3B activity is a potential therapeutic approach for mitochondrial diseases.


Subject(s)
ATPases Associated with Diverse Cellular Activities/metabolism , Membrane Proteins/metabolism , Mitochondrial Proteins/metabolism , Mitophagy , Oxidative Stress , ATPases Associated with Diverse Cellular Activities/chemistry , ATPases Associated with Diverse Cellular Activities/genetics , Animals , Cells, Cultured , DNA Damage , DNA, Mitochondrial/genetics , DNA, Mitochondrial/metabolism , HEK293 Cells , HeLa Cells , Humans , Membrane Proteins/chemistry , Membrane Proteins/genetics , Mice , Microtubule-Associated Proteins/metabolism , Mitochondrial Proteins/chemistry , Mitochondrial Proteins/genetics , Protein Binding
2.
Glia ; 2024 Oct 03.
Article in English | MEDLINE | ID: mdl-39360557

ABSTRACT

Histidine dipeptides (HDs) are synthesized in brain oligodendrocytes by carnosine synthase (carns1), but their role is unknown. Using metabolomics and in vivo experiments with both constitutive and oligodendrocyte-selective carns1-KO mouse models, we found that HDs are critical for oligodendrocyte survival and protect against oxidative stress. Carns1-KO mouse models had lower numbers of mature oligodendrocytes, increased lipid peroxidation, and behavioral changes. Cuprizone administration, which increases reactive oxygen species in vivo, resulted in higher oligodendrocyte death, demyelination, axonal alterations, and oxidative damage in the corpus callosum of carns1-KO mice. Gliosis and oxidative damage by cuprizone were prevented by pretreatment with the antioxidant N-acetylcysteine. NADPH levels were increased threefold in the brains of carns1-KO mice as an antioxidant response to oxidative stress through acceleration of the pentose phosphate pathway (PPP). This was due to overexpression of glucose-6-phosphate dehydrogenase, the rate-limiting enzyme of the PPP. Likewise, expression of NAD kinase, the biosynthetic enzyme for NADP+, and NAMPT, which replenishes the NAD+ pool, was higher in carns1-KO mice brains than in controls. Our observations suggest that HDs cell-autonomously protect oligodendrocytes from oxidative stress, with implications for demyelinating diseases.

3.
Mov Disord ; 37(7): 1346-1359, 2022 07.
Article in English | MEDLINE | ID: mdl-35579450

ABSTRACT

Neurodegenerative proteinopathies are defined as a class of neurodegenerative disorders, with either genetic or sporadic age-related onset, characterized by the pathological accumulation of aggregated protein deposits. These mainly include Alzheimer's disease (AD), Parkinson's disease (PD), amyotrophic lateral sclerosis (ALS), Huntington's disease (HD) as well as frontotemporal lobar degeneration (FTLD). The deposition of abnormal protein aggregates in the brain of patients affected by these disorders is thought to play a causative role in neuronal loss and disease progression. On that account, the idea of improving the clearance of pathological protein aggregates has taken hold as a potential therapeutic strategy. Among the possible approaches to pursue for reducing disease protein accumulation, there is the stimulation of the main protein degradation machineries of eukaryotic cells: the ubiquitin proteasomal system (UPS) and autophagy lysosomal pathway (ALP). Of note, several clinical trials testing the efficacy of either UPS- or ALP-active compounds are currently ongoing. Here, we discuss the main gaps and controversies emerging from experimental studies and clinical trials assessing the therapeutic efficacy of modulators of either the UPS or ALP in neurodegenerative proteinopathies, to gather whether they may constitute a real gateway from these disorders. © 2022 International Parkinson and Movement Disorder Society.


Subject(s)
Amyotrophic Lateral Sclerosis , Frontotemporal Lobar Degeneration , Humans , Protein Aggregates , Proteins/metabolism , Proteolysis , Ubiquitin/metabolism
4.
Proc Natl Acad Sci U S A ; 116(41): 20736-20742, 2019 10 08.
Article in English | MEDLINE | ID: mdl-31548413

ABSTRACT

Astrocytes express the 3-phosphoglycerate dehydrogenase (Phgdh) enzyme required for the synthesis of l-serine from glucose. Astrocytic l-serine was proposed to regulate NMDAR activity by shuttling to neurons to sustain d-serine production, but this hypothesis remains untested. We now report that inhibition of astrocytic Phgdh suppressed the de novo synthesis of l-and d-serine and reduced the NMDAR synaptic potentials and long-term potentiation (LTP) at the Schaffer collaterals-CA1 synapse. Likewise, enzymatic removal of extracellular l-serine impaired LTP, supporting an l-serine shuttle mechanism between glia and neurons in generating the NMDAR coagonist d-serine. Moreover, deletion of serine racemase (SR) in glutamatergic neurons abrogated d-serine synthesis to the same extent as Phgdh inhibition, suggesting that neurons are the predominant source of the newly synthesized d-serine. We also found that the synaptic NMDAR activation in adult SR-knockout (KO) mice requires Phgdh-derived glycine, despite the sharp decline in the postnatal glycine levels as a result of the emergence of the glycine cleavage system. Unexpectedly, we also discovered that glycine regulates d-serine metabolism by a dual mechanism. The first consists of tonic inhibition of SR by intracellular glycine observed in vitro, primary cultures, and in vivo microdialysis. The second involves a transient glycine-induce d-serine release through the Asc-1 transporter, an effect abolished in Asc-1 KO mice and diminished by deleting SR in glutamatergic neurons. Our observations suggest that glycine is a multifaceted regulator of d-serine metabolism and implicate both d-serine and glycine in mediating NMDAR synaptic activation at the mature hippocampus through a Phgdh-dependent shuttle mechanism.


Subject(s)
Astrocytes/metabolism , Glycine/metabolism , Phosphoglycerate Dehydrogenase/metabolism , Racemases and Epimerases/physiology , Receptors, N-Methyl-D-Aspartate/metabolism , Serine/metabolism , Synapses/physiology , Animals , Astrocytes/cytology , Hippocampus/cytology , Hippocampus/metabolism , Long-Term Potentiation , Mice , Mice, Inbred C57BL , Mice, Knockout , Neurons/cytology , Neurons/metabolism , Phosphoglycerate Dehydrogenase/genetics , Receptors, N-Methyl-D-Aspartate/genetics
5.
Hum Mol Genet ; 28(23): 3982-3996, 2019 12 01.
Article in English | MEDLINE | ID: mdl-31626293

ABSTRACT

Mutations in LRRK2 cause autosomal dominant and sporadic Parkinson's disease, but the mechanisms involved in LRRK2 toxicity in PD are yet to be fully understood. We found that LRRK2 translocates to the nucleus by binding to seven in absentia homolog (SIAH-1), and in the nucleus it directly interacts with lamin A/C, independent of its kinase activity. LRRK2 knockdown caused nuclear lamina abnormalities and nuclear disruption. LRRK2 disease mutations mostly abolish the interaction with lamin A/C and, similar to LRRK2 knockdown, cause disorganization of lamin A/C and leakage of nuclear proteins. Dopaminergic neurons of LRRK2 G2019S transgenic and LRRK2 -/- mice display decreased circularity of the nuclear lamina and leakage of the nuclear protein 53BP1 to the cytosol. Dopaminergic nigral and cortical neurons of both LRRK2 G2019S and idiopathic PD patients exhibit abnormalities of the nuclear lamina. Our data indicate that LRRK2 plays an essential role in maintaining nuclear envelope integrity. Disruption of this function by disease mutations suggests a novel phosphorylation-independent loss-of-function mechanism that may synergize with other neurotoxic effects caused by LRRK2 mutations.


Subject(s)
Leucine-Rich Repeat Serine-Threonine Protein Kinase-2/genetics , Leucine-Rich Repeat Serine-Threonine Protein Kinase-2/metabolism , Nuclear Envelope/metabolism , Parkinson Disease/genetics , Animals , Cells, Cultured , Dopaminergic Neurons/cytology , Dopaminergic Neurons/metabolism , Dopaminergic Neurons/pathology , HEK293 Cells , Humans , Lamin Type A/metabolism , Loss of Function Mutation , Mice , Parkinson Disease/metabolism , Parkinson Disease/pathology , Phosphorylation , Rats , Tumor Suppressor p53-Binding Protein 1/metabolism
6.
Proc Natl Acad Sci U S A ; 114(50): 13176-13181, 2017 12 12.
Article in English | MEDLINE | ID: mdl-29180403

ABSTRACT

α-Synuclein accumulation is a pathological hallmark of Parkinson's disease (PD). Ubiquitinated α-synuclein is targeted to proteasomal or lysosomal degradation. Here, we identify SUMOylation as a major mechanism that counteracts ubiquitination by different E3 ubiquitin ligases and regulates α-synuclein degradation. We report that PIAS2 promotes SUMOylation of α-synuclein, leading to a decrease in α-synuclein ubiquitination by SIAH and Nedd4 ubiquitin ligases, and causing its accumulation and aggregation into inclusions. This was associated with an increase in α-synuclein release from the cells. A SUMO E1 inhibitor, ginkgolic acid, decreases α-synuclein levels by relieving the inhibition exerted on α-synuclein proteasomal degradation. α-Synuclein disease mutants are more SUMOylated compared with the wild-type protein, and this is associated with increased aggregation and inclusion formation. We detected a marked increase in PIAS2 expression along with SUMOylated α-synuclein in PD brains, providing a causal mechanism underlying the up-regulation of α-synuclein SUMOylation in the disease. We also found a significant proportion of Lewy bodies in nigral neurons containing SUMO1 and PIAS2. Our observations suggest that SUMOylation of α-synuclein by PIAS2 promotes α-synuclein aggregation by two mutually reinforcing mechanisms. First, it has a direct proaggregatory effect on α-synuclein. Second, SUMOylation facilitates α-synuclein aggregation by blocking its ubiquitin-dependent degradation pathways and promoting its accumulation. Therefore, inhibitors of α-synuclein SUMOylation provide a strategy to reduce α-synuclein levels and possibly aggregation in PD.


Subject(s)
Parkinson Disease/metabolism , Proteolysis , Sumoylation , alpha-Synuclein/metabolism , Animals , Cells, Cultured , HEK293 Cells , Humans , Neurons/drug effects , Neurons/metabolism , Protein Inhibitors of Activated STAT/genetics , Protein Inhibitors of Activated STAT/metabolism , Rats, Sprague-Dawley , Salicylates/pharmacology , Substantia Nigra/metabolism
7.
J Neuroinflammation ; 16(1): 153, 2019 Jul 22.
Article in English | MEDLINE | ID: mdl-31331333

ABSTRACT

This article describes pathogenic concepts and factors, in particular glycolipid abnormalities, that create cell dysfunction and synaptic loss in neurodegenerative diseases. By phenocopying lysosomal storage disorders, such as Gaucher disease and related disorders, age- and dose-dependent changes in glycolipid cell metabolism can lead to Parkinson's disease and related dementias. Recent results show that perturbation of sphingolipid metabolism can precede or is a part of abnormal protein handling in both genetic and idiopathic Parkinson's disease and Lewy body dementia. In aging and genetic predisposition with lipid disturbance, α-synuclein's normal vesicular and synaptic role may be detrimentally shifted toward accommodating and binding such lipids. Specific neuronal glycolipid, protein, and vesicular interactions create potential pathophysiology that is amplified by astroglial and microglial immune mechanisms resulting in neurodegeneration. This perspective provides a new logic for therapeutic interventions that do not focus on protein aggregation, but rather provides a guide to the complex biology and the common sequence of events that lead to age-dependent neurodegenerative disorders.


Subject(s)
Brain/pathology , Inflammation/pathology , Nerve Degeneration/pathology , Neurons/pathology , Parkinson Disease/pathology , Animals , Brain/immunology , Brain/metabolism , Humans , Inflammation/immunology , Inflammation/metabolism , Nerve Degeneration/immunology , Nerve Degeneration/metabolism , Neurons/immunology , Neurons/metabolism , Parkinson Disease/immunology , Parkinson Disease/metabolism , alpha-Synuclein/metabolism , tau Proteins/metabolism
8.
Hum Mol Genet ; 25(16): 3476-3490, 2016 08 15.
Article in English | MEDLINE | ID: mdl-27334109

ABSTRACT

PTEN-induced putative kinase 1 (PINK1) and parkin are mutated in familial forms of Parkinson's disease and are important in promoting the mitophagy of damaged mitochondria. In this study, we showed that synphilin-1 interacted with PINK1 and was recruited to the mitochondria. Once in the mitochondria, it promoted PINK1-dependent mitophagy, as revealed by Atg5 knockdown experiments and the recruitment of LC3 and Lamp1 to the mitochondria. PINK1-synphilin-1 mitophagy did not depend on PINK1-mediated phosphorylation of synphilin-1 and occurred in the absence of parkin. Synphilin-1 itself caused depolarization of the mitochondria and increased the amount of uncleaved PINK1 at the organelle. Furthermore, synphilin-1 recruited seven in absentia homolog (SIAH)-1 to the mitochondria where it promoted mitochondrial protein ubiquitination and subsequent mitophagy. Mitophagy via this pathway was impaired by synphilin-1 knockdown or by the use of a synphilin-1 mutant that is unable to recruit SIAH-1 to the mitochondria. Likewise, knockdown of SIAH-1 or the use of a catalytically inactive SIAH-1 mutant abrogated mitophagy. PINK1 disease mutants failed to recruit synphilin-1 and did not activate mitophagy, indicating that PINK1-synphilin-1-SIAH-1 represents a new parkin-independent mitophagy pathway. Drugs that activate this pathway will provide a novel strategy to promote the clearance of damaged mitochondria in Parkinson's disease.


Subject(s)
Carrier Proteins/genetics , Mitophagy/genetics , Nerve Tissue Proteins/genetics , Nuclear Proteins/genetics , Parkinson Disease/genetics , Protein Kinases/genetics , Ubiquitin-Protein Ligases/genetics , Autophagy-Related Protein 5/genetics , Carrier Proteins/metabolism , Humans , Mitochondria/genetics , Mitochondria/pathology , Nerve Tissue Proteins/metabolism , Nuclear Proteins/metabolism , Parkinson Disease/metabolism , Parkinson Disease/pathology , Phosphorylation , Protein Binding , Protein Kinases/metabolism , Signal Transduction , Ubiquitin , Ubiquitin-Protein Ligases/metabolism , Ubiquitination
9.
Hum Mol Genet ; 25(4): 672-80, 2016 Feb 15.
Article in English | MEDLINE | ID: mdl-26744328

ABSTRACT

Mutations in leucine-rich repeat kinase 2 (LRRK2) cause autosomal-dominant Parkinsonism with pleomorphic pathology including deposits of aggregated protein and neuronal degeneration. The pathogenesis of LRRK2-linked Parkinson's disease (PD) is not fully understood. Here, using co-immunoprecipitation, we found that LRRK2 interacted with synphilin-1 (SP1), a cytoplasmic protein that interacts with α-synuclein and has implications in PD pathogenesis. LRRK2 interacted with the N-terminus of SP1 whereas SP1 predominantly interacted with the C-terminus of LRRK2, including kinase domain. Co-expression of SP1 with LRRK2 increased LRRK2-induced cytoplasmic aggregation in cultured cells. Moreover, SP1 also attenuated mutant LRRK2-induced toxicity and reduced LRRK2 kinase activity in cultured cells. Knockdown of SP1 by siRNA enhanced LRRK2 neuronal toxicity. In vivo Drosophila studies, co-expression of SP1 and mutant G2019S-LRRK2 in double transgenic Drosophila increased survival and improved locomotor activity. Expression of SP1 protects against G2019S-LRRK2-induced dopamine neuron loss and reduced LRRK2 phosphorylation in double transgenic fly brains. Our findings demonstrate that SP1 attenuates mutant LRRK2-induced PD-like phenotypes and plays a neural protective role.


Subject(s)
Carrier Proteins/metabolism , Leucine-Rich Repeat Serine-Threonine Protein Kinase-2/metabolism , Nerve Tissue Proteins/metabolism , Parkinson Disease/metabolism , Animals , Animals, Genetically Modified , Carrier Proteins/genetics , Cell Line, Tumor , Disease Models, Animal , Dopamine/metabolism , Drosophila , Gene Knockdown Techniques , HEK293 Cells , Humans , Leucine-Rich Repeat Serine-Threonine Protein Kinase-2/genetics , Mice , Mutation , Nerve Degeneration/genetics , Nerve Tissue Proteins/genetics , Neurons/enzymology , Neurons/metabolism , Neurons/pathology , Parkinson Disease/enzymology , Parkinson Disease/genetics , Parkinson Disease/pathology , Phosphorylation , Protein Interaction Domains and Motifs , alpha-Synuclein/genetics , alpha-Synuclein/metabolism
10.
Brain ; 140(5): 1399-1419, 2017 May 01.
Article in English | MEDLINE | ID: mdl-28398476

ABSTRACT

α-Synuclein misfolding and aggregation is a hallmark in Parkinson's disease and in several other neurodegenerative diseases known as synucleinopathies. The toxic properties of α-synuclein are conserved from yeast to man, but the precise underpinnings of the cellular pathologies associated are still elusive, complicating the development of effective therapeutic strategies. Combining molecular genetics with target-based approaches, we established that glycation, an unavoidable age-associated post-translational modification, enhanced α-synuclein toxicity in vitro and in vivo, in Drosophila and in mice. Glycation affected primarily the N-terminal region of α-synuclein, reducing membrane binding, impaired the clearance of α-synuclein, and promoted the accumulation of toxic oligomers that impaired neuronal synaptic transmission. Strikingly, using glycation inhibitors, we demonstrated that normal clearance of α-synuclein was re-established, aggregation was reduced, and motor phenotypes in Drosophila were alleviated. Altogether, our study demonstrates glycation constitutes a novel drug target that can be explored in synucleinopathies as well as in other neurodegenerative conditions.


Subject(s)
Neurodegenerative Diseases/metabolism , Protein Aggregation, Pathological/metabolism , alpha-Synuclein/metabolism , alpha-Synuclein/toxicity , Aging/metabolism , Animals , Cell Differentiation/drug effects , Cell Survival/drug effects , Cell Survival/physiology , Cells, Cultured , Disease Models, Animal , Drosophila , Enzyme Inhibitors/pharmacology , Female , Glycosylation/drug effects , Hippocampus/drug effects , Hippocampus/physiology , Humans , Induced Pluripotent Stem Cells/drug effects , Induced Pluripotent Stem Cells/physiology , Male , Mice , Mice, Transgenic , Protein Processing, Post-Translational , Pyruvaldehyde/pharmacology , Rats , Yeasts/drug effects , Yeasts/physiology , alpha-Synuclein/drug effects , alpha-Synuclein/physiology
11.
EMBO Rep ; 16(5): 590-8, 2015 May.
Article in English | MEDLINE | ID: mdl-25755256

ABSTRACT

Asc-1 (SLC7A10) is an amino acid transporter whose deletion causes neurological abnormalities and early postnatal death in mice. Using metabolomics and behavioral and electrophysiological methods, we demonstrate that Asc-1 knockout mice display a marked decrease in glycine levels in the brain and spinal cord along with impairment of glycinergic inhibitory transmission, and a hyperekplexia-like phenotype that is rescued by replenishing brain glycine. Asc-1 works as a glycine and L-serine transporter, and its transport activity is required for the subsequent conversion of L-serine into glycine in vivo. Asc-1 is a novel regulator of glycine metabolism and a candidate for hyperekplexia disorders.


Subject(s)
Amino Acid Transport System y+/metabolism , Brain/metabolism , Glycine/metabolism , Synaptic Transmission , Amino Acid Transport System y+/genetics , Animals , Biological Transport , Genotype , Hypoglossal Nerve/cytology , Metabolome , Metabolomics/methods , Mice , Mice, Knockout , Mutation , Neurons/metabolism , Phenotype , Receptors, Glycine/genetics , Receptors, Glycine/metabolism , Serine/metabolism , Synaptic Transmission/genetics
14.
J Biol Chem ; 290(22): 13862-74, 2015 May 29.
Article in English | MEDLINE | ID: mdl-25861987

ABSTRACT

Mutations in PARKIN (PARK2), an ubiquitin ligase, cause early onset Parkinson disease. Parkin was shown to bind, ubiquitinate, and target depolarized mitochondria for destruction by autophagy. This process, mitophagy, is considered crucial for maintaining mitochondrial integrity and suppressing Parkinsonism. Here, we report that under moderate mitochondrial stress, parkin does not translocate to mitochondria to induce mitophagy; rather, it stimulates mitochondrial connectivity. Mitochondrial stress-induced fusion requires PINK1 (PARK6), mitofusins, and parkin ubiquitin ligase activity. Upon exposure to mitochondrial toxins, parkin binds α-synuclein (PARK1), and in conjunction with the ubiquitin-conjugating enzyme Ubc13, stimulates K63-linked ubiquitination. Importantly, α-synuclein inactivation phenocopies parkin overexpression and suppresses stress-induced mitochondria fission, whereas Ubc13 inactivation abrogates parkin-dependent mitochondrial fusion. The convergence of parkin, PINK1, and α-synuclein on mitochondrial dynamics uncovers a common function of these PARK genes in the mitochondrial stress response and provides a potential physiological basis for the prevalence of α-synuclein pathology in Parkinson disease.


Subject(s)
Gene Expression Regulation , Mitochondria/metabolism , Protein Kinases/metabolism , Ubiquitin-Protein Ligases/metabolism , alpha-Synuclein/metabolism , Animals , Carbonyl Cyanide m-Chlorophenyl Hydrazone/chemistry , Female , Fibroblasts/metabolism , Gene Silencing , HeLa Cells , Humans , Male , Mice , Mice, Knockout , Microscopy, Confocal , Mitophagy , Mutation , Neurons/metabolism , Parkinson Disease/metabolism , Phosphorylation , Ubiquitin/chemistry
15.
Hum Mol Genet ; 22(10): 2083-96, 2013 May 15.
Article in English | MEDLINE | ID: mdl-23393160

ABSTRACT

Parkin E3 ubiquitin-ligase activity and its role in mitochondria homeostasis are thought to play a role in Parkinson's disease (PD). We now report that AF-6 is a novel parkin interacting protein that modulates parkin ubiquitin-ligase activity and mitochondrial roles. Parkin interacts with the AF-6 PDZ region through its C-terminus. This leads to ubiquitination of cytosolic AF-6 and its degradation by the proteasome. On the other hand, endogenous AF-6 robustly increases parkin translocation and ubiquitin-ligase activity at the mitochondria. Mitochondrial AF-6 is not a parkin substrate, but rather co-localizes with parkin and enhances mitochondria degradation through PINK1/parkin-mediated mitophagy. On the other hand, several parkin and PINK1 juvenile disease-mutants are insensitive to AF-6 effects. AF-6 is present in Lewy bodies and its soluble levels are strikingly decreased in the caudate/putamen and substantia nigra of sporadic PD patients, suggesting that decreased AF-6 levels may contribute to the accumulation of dysfunctional mitochondria in the disease. The identification of AF-6 as a positive modulator of parkin translocation to the mitochondria sheds light on the mechanisms involved in PD and underscores AF-6 as a novel target for future therapeutics.


Subject(s)
Kinesins/metabolism , Mitochondria/metabolism , Mutation , Myosins/metabolism , Parkinson Disease/metabolism , Protein Kinases/metabolism , Ubiquitin-Protein Ligases/metabolism , Caudate Nucleus/metabolism , Caudate Nucleus/pathology , HEK293 Cells , Humans , Kinesins/genetics , Mitochondria/genetics , Mitochondria/pathology , Myosins/genetics , Parkinson Disease/genetics , Parkinson Disease/pathology , Protein Kinases/genetics , Protein Transport/genetics , Proteolysis , Substantia Nigra/metabolism , Substantia Nigra/pathology , Ubiquitin-Protein Ligases/genetics , Ubiquitination/genetics
16.
Proc Natl Acad Sci U S A ; 108(46): 18666-71, 2011 Nov 15.
Article in English | MEDLINE | ID: mdl-22065755

ABSTRACT

α-Synuclein is central to the pathogenesis of Parkinson disease (PD). Mutations as well as accumulation of α-synuclein promote the death of dopaminergic neurons and the formation of Lewy bodies. α-Synuclein is monoubiquitinated by SIAH, but the regulation and roles of monoubiquitination in α-synuclein biology are poorly understood. We now report that the deubiquitinase USP9X interacts in vivo with and deubiquitinates α-synuclein. USP9X levels are significantly lower in cytosolic fractions of PD substantia nigra and Diffuse Lewy Body disease (DLBD) cortices compared to controls. This was associated to lower deubiquitinase activity toward monoubiquitinated α-synuclein in DLBD cortical extracts. A fraction of USP9X seems to be aggregated in PD and DLBD, as USP9X immunoreactivity is detected in Lewy bodies. Knockdown of USP9X expression promotes accumulation of monoubiquitinated α-synuclein species and enhances the formation of toxic α-synuclein inclusions upon proteolytic inhibition. On the other hand, by manipulating USP9X expression levels in the absence of proteolytic impairment, we demonstrate that monoubiquitination controls the partition of α-synuclein between different protein degradation systems. Deubiquitinated α-synuclein is mostly degraded by autophagy, while monoubiquitinated α-synuclein is preferentially degraded by the proteasome. Moreover, monoubiquitination promotes the degradation of α-synuclein, whereas deubiquitination leads to its accumulation, suggesting that the degradation of deubiquitinated α-synuclein by the autophagy pathway is less efficient than the proteasomal one. Lower levels of cytosolic USP9X and deubiquitinase activity in α-synucleinopathies may contribute to the accumulation and aggregation of monoubiquitinated α-synuclein in Lewy bodies. Our data indicate that monoubiquitination is a key determinant of α-synuclein fate.


Subject(s)
Gene Expression Regulation , Ubiquitin Thiolesterase/chemistry , Ubiquitin/chemistry , alpha-Synuclein/chemistry , Autophagy , Cell Line, Tumor , Cerebral Cortex/metabolism , Dopaminergic Neurons/metabolism , Humans , Lewy Bodies/metabolism , Lewy Body Disease/metabolism , Proteasome Endopeptidase Complex/metabolism , Proteolysis , RNA, Small Interfering/metabolism
17.
Nat Commun ; 15(1): 168, 2024 Jan 02.
Article in English | MEDLINE | ID: mdl-38168065

ABSTRACT

Endoplasmic reticulum (ER)-mitochondria contacts are critical for the regulation of lipid transport, synthesis, and metabolism. However, the molecular mechanism and physiological function of endoplasmic reticulum-mitochondrial contacts remain unclear. Here, we show that Mic19, a key subunit of MICOS (mitochondrial contact site and cristae organizing system) complex, regulates ER-mitochondria contacts by the EMC2-SLC25A46-Mic19 axis. Mic19 liver specific knockout (LKO) leads to the reduction of ER-mitochondrial contacts, mitochondrial lipid metabolism disorder, disorganization of mitochondrial cristae and mitochondrial unfolded protein stress response in mouse hepatocytes, impairing liver mitochondrial fatty acid ß-oxidation and lipid metabolism, which may spontaneously trigger nonalcoholic steatohepatitis (NASH) and liver fibrosis in mice. Whereas, the re-expression of Mic19 in Mic19 LKO hepatocytes blocks the development of liver disease in mice. In addition, Mic19 overexpression suppresses MCD-induced fatty liver disease. Thus, our findings uncover the EMC2-SLC25A46-Mic19 axis as a pathway regulating ER-mitochondria contacts, and reveal that impairment of ER-mitochondria contacts may be a mechanism associated with the development of NASH and liver fibrosis.


Subject(s)
Lipid Metabolism , Non-alcoholic Fatty Liver Disease , Mice , Animals , Lipid Metabolism/genetics , Non-alcoholic Fatty Liver Disease/metabolism , Endoplasmic Reticulum Stress , Liver/metabolism , Mitochondria/metabolism , Liver Cirrhosis/pathology , Endoplasmic Reticulum/metabolism
18.
Nat Commun ; 14(1): 4105, 2023 07 11.
Article in English | MEDLINE | ID: mdl-37433770

ABSTRACT

Mitochondria are the key organelles for sensing oxygen, which is consumed by oxidative phosphorylation to generate ATP. Lysosomes contain hydrolytic enzymes that degrade misfolded proteins and damaged organelles to maintain cellular homeostasis. Mitochondria physically and functionally interact with lysosomes to regulate cellular metabolism. However, the mode and biological functions of mitochondria-lysosome communication remain largely unknown. Here, we show that hypoxia remodels normal tubular mitochondria into megamitochondria by inducing broad inter-mitochondria contacts and subsequent fusion. Importantly, under hypoxia, mitochondria-lysosome contacts are promoted, and certain lysosomes are engulfed by megamitochondria, in a process we term megamitochondria engulfing lysosome (MMEL). Both megamitochondria and mature lysosomes are required for MMEL. Moreover, the STX17-SNAP29-VAMP7 complex contributes to mitochondria-lysosome contacts and MMEL under hypoxia. Intriguingly, MMEL mediates a mode of mitochondrial degradation, which we termed mitochondrial self-digestion (MSD). Moreover, MSD increases mitochondrial ROS production. Our results reveal a mode of crosstalk between mitochondria and lysosomes and uncover an additional pathway for mitochondrial degradation.


Subject(s)
Lysosomes , Mitochondria , Humans , Hypoxia , Oxygen , Digestion
19.
Aging Cell ; 21(12): e13731, 2022 12.
Article in English | MEDLINE | ID: mdl-36307912

ABSTRACT

Parkinson's disease (PD) is characterized by degeneration of neurons, particularly dopaminergic neurons in the substantia nigra. PD brains show accumulation of α-synuclein in Lewy bodies and accumulation of dysfunctional mitochondria. However, the mechanisms leading to mitochondrial pathology in sporadic PD are poorly understood. PINK1 is a key for mitophagy activation and recycling of unfit mitochondria. The activation of mitophagy depends on the accumulation of uncleaved PINK1 at the outer mitochondrial membrane and activation of a cascade of protein ubiquitination at the surface of the organelle. We have now found that SIAH3, a member of the SIAH proteins but lacking ubiquitin-ligase activity, is increased in PD brains and cerebrospinal fluid and in neurons treated with α-synuclein preformed fibrils (α-SynPFF). We also observed that SIAH3 is aggregated together with PINK1 in the mitochondria of PD brains. SIAH3 directly interacts with PINK1, leading to their intra-mitochondrial aggregation in cells and neurons and triggering a cascade of toxicity with PINK1 inactivation along with mitochondrial depolarization and neuronal death. We also found that SIAH1 interacts with PINK1 and promotes ubiquitination and proteasomal degradation of PINK1. Similar to the dimerization of SIAH1/SIAH2, SIAH3 interacts with SIAH1, promoting its translocation to mitochondria and preventing its ubiquitin-ligase activity toward PINK1. Our results support the notion that the increase in SIAH3 and intra-mitochondrial aggregation of SIAH3-PINK1 may mediate α-synuclein pathology by promoting proteotoxicity and preventing the elimination of dysfunctional mitochondria. We consider it possible that PINK1 activity is decreased in sporadic PD, which impedes proper mitochondrial renewal in the disease.


Subject(s)
Parkinson Disease , alpha-Synuclein , Humans , alpha-Synuclein/metabolism , Parkinson Disease/metabolism , Protein Kinases/metabolism , Mitophagy , Ubiquitin-Protein Ligases/genetics , Ubiquitin-Protein Ligases/metabolism , Ubiquitin
20.
Neurogenetics ; 11(2): 203-15, 2010 May.
Article in English | MEDLINE | ID: mdl-19730898

ABSTRACT

Parkinson's disease (PD) is a common neurodegenerative disorder characterized by the loss of dopaminergic neurons and the presence of Lewy bodies. Alpha-synuclein and its interactor synphilin-1 are major components of these inclusions. Rare mutations in the alpha-synuclein and synphilin-1 genes have been implicated in the pathogenesis of PD; however, the normal function of these proteins is far from being completely elucidated. We, thus, searched for novel synphilin-1-interacting proteins and deciphered periphilin as new interactor. Periphilin isoforms are involved in multiple cellular functions in vivo, and the protein is broadly expressed during embryogenesis and in the adult brain. We show that periphilin displays an overlapping expression pattern with synphilin-1 in cellular and animal models and in Lewy bodies of PD patients. Functional studies demonstrate that periphilin, as previously shown for synphilin-1, displays an antiapoptotic function by reducing caspase-3 activity. Searching for mutations in the periphilin gene, we detected a K69E substitution in two patients of a PD family. Taken together, these findings support for the first time an involvement of periphilin in PD.


Subject(s)
Antigens, Neoplasm/metabolism , Carrier Proteins/metabolism , Nerve Tissue Proteins/metabolism , Nuclear Proteins/metabolism , Parkinson Disease/metabolism , Amino Acid Sequence , Animals , Antigens, Neoplasm/genetics , Carrier Proteins/genetics , Cell Line , DNA Mutational Analysis , Humans , Lewy Bodies/metabolism , Male , Mice , Mice, Transgenic , Middle Aged , Molecular Sequence Data , Mutation , Nerve Tissue Proteins/genetics , Nuclear Proteins/genetics , Parkinson Disease/genetics , Sequence Alignment , Two-Hybrid System Techniques
SELECTION OF CITATIONS
SEARCH DETAIL