Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
1.
Brain ; 147(6): 2069-2084, 2024 Jun 03.
Article in English | MEDLINE | ID: mdl-38763511

ABSTRACT

The peroxisomal disease adrenoleukodystrophy (X-ALD) is caused by loss of the transporter of very-long-chain fatty acids (VLCFAs), ABCD1. An excess of VLCFAs disrupts essential homeostatic functions crucial for axonal maintenance, including redox metabolism, glycolysis and mitochondrial respiration. As mitochondrial function and morphology are intertwined, we set out to investigate the role of mitochondrial dynamics in X-ALD models. Using quantitative 3D transmission electron microscopy, we revealed mitochondrial fragmentation in corticospinal axons in Abcd1- mice. In patient fibroblasts, an excess of VLCFAs triggers mitochondrial fragmentation through the redox-dependent phosphorylation of DRP1 (DRP1S616). The blockade of DRP1-driven fission by the peptide P110 effectively preserved mitochondrial morphology. Furthermore, mRNA inhibition of DRP1 not only prevented mitochondrial fragmentation but also protected axonal health in a Caenorhabditis elegans model of X-ALD, underscoring DRP1 as a potential therapeutic target. Elevated levels of circulating cell-free mtDNA in patients' CSF align this leukodystrophy with primary mitochondrial disorders. Our findings underscore the intricate interplay between peroxisomal dysfunction, mitochondrial dynamics and axonal integrity in X-ALD, shedding light on potential avenues for therapeutic intervention.


Subject(s)
ATP Binding Cassette Transporter, Subfamily D, Member 1 , Adrenoleukodystrophy , Dynamins , Mitochondrial Dynamics , Adrenoleukodystrophy/metabolism , Adrenoleukodystrophy/pathology , Adrenoleukodystrophy/genetics , Animals , Mitochondrial Dynamics/physiology , Humans , Mice , Dynamins/metabolism , Dynamins/genetics , ATP Binding Cassette Transporter, Subfamily D, Member 1/genetics , Caenorhabditis elegans , Mitochondria/metabolism , Mitochondria/pathology , Axons/pathology , Axons/metabolism , Fibroblasts/metabolism , Fibroblasts/pathology , Male , DNA, Mitochondrial/genetics , DNA, Mitochondrial/metabolism , Disease Models, Animal , Pyramidal Tracts/pathology , Pyramidal Tracts/metabolism , Peptide Fragments , GTP Phosphohydrolases
2.
Acta Neuropathol ; 147(1): 9, 2024 01 04.
Article in English | MEDLINE | ID: mdl-38175301

ABSTRACT

Nuclear clearance and cytoplasmic accumulations of the RNA-binding protein TDP-43 are pathological hallmarks in almost all patients with amyotrophic lateral sclerosis (ALS) and up to 50% of patients with frontotemporal dementia (FTD) and Alzheimer's disease. In Alzheimer's disease, TDP-43 pathology is predominantly observed in the limbic system and correlates with cognitive decline and reduced hippocampal volume. Disruption of nuclear TDP-43 function leads to abnormal RNA splicing and incorporation of erroneous cryptic exons in numerous transcripts including Stathmin-2 (STMN2, also known as SCG10) and UNC13A, recently reported in tissues from patients with ALS and FTD. Here, we identify both STMN2 and UNC13A cryptic exons in Alzheimer's disease patients, that correlate with TDP-43 pathology burden, but not with amyloid-ß or tau deposits. We also demonstrate that processing of the STMN2 pre-mRNA is more sensitive to TDP-43 loss of function than UNC13A. In addition, full-length RNAs encoding STMN2 and UNC13A are suppressed in large RNA-seq datasets generated from Alzheimer's disease post-mortem brain tissue. Collectively, these results open exciting new avenues to use STMN2 and UNC13A as potential therapeutic targets in a broad range of neurodegenerative conditions with TDP-43 proteinopathy including Alzheimer's disease.


Subject(s)
Alzheimer Disease , Amyotrophic Lateral Sclerosis , Frontotemporal Dementia , Pick Disease of the Brain , Humans , Alzheimer Disease/genetics , DNA-Binding Proteins/genetics , RNA Splicing , RNA, Messenger/genetics , Stathmin/genetics
3.
Nat Neurosci ; 27(1): 34-47, 2024 Jan.
Article in English | MEDLINE | ID: mdl-37996528

ABSTRACT

The mRNA transcript of the human STMN2 gene, encoding for stathmin-2 protein (also called SCG10), is profoundly impacted by TAR DNA-binding protein 43 (TDP-43) loss of function. The latter is a hallmark of several neurodegenerative diseases, including amyotrophic lateral sclerosis (ALS). Using a combination of approaches, including transient antisense oligonucleotide-mediated suppression, sustained shRNA-induced depletion in aging mice, and germline deletion, we show that stathmin-2 has an important role in the establishment and maintenance of neurofilament-dependent axoplasmic organization that is critical for preserving the caliber and conduction velocity of myelinated large-diameter axons. Persistent stathmin-2 loss in adult mice results in pathologies found in ALS, including reduced interneurofilament spacing, axonal caliber collapse that drives tearing within outer myelin layers, diminished conduction velocity, progressive motor and sensory deficits, and muscle denervation. These findings reinforce restoration of stathmin-2 as an attractive therapeutic approach for ALS and other TDP-43-dependent neurodegenerative diseases.


Subject(s)
Amyotrophic Lateral Sclerosis , Animals , Mice , Amyotrophic Lateral Sclerosis/metabolism , Axons/physiology , Denervation , DNA-Binding Proteins/genetics , Intermediate Filaments/metabolism , Intermediate Filaments/pathology , Motor Neurons/metabolism , Stathmin/genetics , Stathmin/metabolism
4.
Biol Chem ; 394(5): 621-9, 2013 May.
Article in English | MEDLINE | ID: mdl-23492556

ABSTRACT

X-linked adrenoleukodystrophy (X-ALD) is a severe inherited neurodegenerative disorder characterized by adrenal insufficiency and graded damage in the nervous system. Loss of function of the peroxisomal ABCD1 fatty-acid transporter, resulting in the accumulation of very long-chain fatty acids in organs and plasma, is the genetic cause. Treatment with a combination of antioxidants halts the axonal degeneration and locomotor impairment displayed by the animal model of X-ALD, and is a proof of concept that oxidative stress contributes to axonal damage. New evidence demonstrates that metabolic failure and the opening of the mitochondrial permeability transition pore orchestrated by cyclophilin D underlies oxidative stress-induced axonal degeneration. Thus, cyclophilin D could serve as a therapeutic target for the treatment of X-ALD and cyclophilin D-dependent neurodegenerative and non-neurodegenerative diseases.


Subject(s)
Adrenoleukodystrophy/drug therapy , Antioxidants/pharmacology , Antioxidants/therapeutic use , Cyclophilins/metabolism , Neurodegenerative Diseases/drug therapy , Neurodegenerative Diseases/metabolism , Adrenoleukodystrophy/genetics , Adrenoleukodystrophy/metabolism , Animals , Clinical Trials, Phase II as Topic , Coenzyme A Ligases/genetics , Coenzyme A Ligases/metabolism , Peptidyl-Prolyl Isomerase F , Humans , Molecular Targeted Therapy , Neurodegenerative Diseases/genetics , Oxidative Stress/drug effects
5.
Brain ; 135(Pt 12): 3584-98, 2012 Dec.
Article in English | MEDLINE | ID: mdl-23250880

ABSTRACT

A common process associated with oxidative stress and severe mitochondrial impairment is the opening of the mitochondrial permeability transition pore, as described in many neurodegenerative diseases. Thus, inhibition of mitochondrial permeability transition pore opening represents a potential target for inhibiting mitochondrial-driven cell death. Among the mitochondrial permeability transition pore components, cyclophilin D is the most studied and has been found increased under pathological conditions. Here, we have used in vitro and in vivo models of X-linked adrenoleukodystrophy to investigate the relationship between the mitochondrial permeability transition pore opening and redox homeostasis. X-linked adrenoleukodystrophy is a neurodegenerative condition caused by loss of function of the peroxisomal ABCD1 transporter, in which oxidative stress plays a pivotal role. In this study, we provide evidence of impaired mitochondrial metabolism in a peroxisomal disease, as fibroblasts in patients with X-linked adrenoleukodystrophy cannot survive when forced to rely on mitochondrial energy production, i.e. on incubation in galactose. Oxidative stress induced under galactose conditions leads to mitochondrial damage in the form of mitochondrial inner membrane potential dissipation, ATP drop and necrotic cell death, together with increased levels of oxidative modifications in cyclophilin D protein. Moreover, we show increased expression levels of cyclophilin D in the affected zones of brains in patients with adrenomyeloneuropathy, in spinal cord of a mouse model of X-linked adrenoleukodystrophy (Abcd1-null mice) and in fibroblasts from patients with X-linked adrenoleukodystrophy. Notably, treatment with antioxidants rescues mitochondrial damage markers in fibroblasts from patients with X-linked adrenoleukodystrophy, including cyclophilin D oxidative modifications, and reverses cyclophilin D induction in vitro and in vivo. These findings provide mechanistic insight into the beneficial effects of antioxidants in neurodegenerative and non-neurodegenerative cyclophilin D-dependent disorders.


Subject(s)
Adrenoleukodystrophy/pathology , Cyclophilins/metabolism , Fibroblasts/ultrastructure , Membrane Potential, Mitochondrial/physiology , Oxidative Stress/physiology , ATP Binding Cassette Transporter 1 , ATP-Binding Cassette Transporters/deficiency , Acetylcysteine/administration & dosage , Adenosine Triphosphate/metabolism , Adrenoleukodystrophy/diet therapy , Age Factors , Analysis of Variance , Animals , Brain/drug effects , Brain/metabolism , Buthionine Sulfoximine/administration & dosage , Cell Death , Chromatin/pathology , Peptidyl-Prolyl Isomerase F , Disease Models, Animal , Enzyme Inhibitors/pharmacology , Fibroblasts/drug effects , Fibroblasts/metabolism , Flow Cytometry , Galactose/toxicity , Gene Expression Regulation/drug effects , Glucose/pharmacology , Glutathione/metabolism , Humans , Mice , Mice, Knockout , Microscopy, Electron, Transmission , Neuroprotective Agents/administration & dosage , Oligopeptides/pharmacology , Oxidative Stress/drug effects , Permeability/drug effects , Reactive Oxygen Species/metabolism , Thioctic Acid/administration & dosage , Time Factors , Tubulin/metabolism , Vitamin E/administration & dosage
6.
Science ; 379(6637): 1140-1149, 2023 03 17.
Article in English | MEDLINE | ID: mdl-36927019

ABSTRACT

Loss of nuclear TDP-43 is a hallmark of neurodegeneration in TDP-43 proteinopathies, including amyotrophic lateral sclerosis (ALS) and frontotemporal dementia (FTD). TDP-43 mislocalization results in cryptic splicing and polyadenylation of pre-messenger RNAs (pre-mRNAs) encoding stathmin-2 (also known as SCG10), a protein that is required for axonal regeneration. We found that TDP-43 binding to a GU-rich region sterically blocked recognition of the cryptic 3' splice site in STMN2 pre-mRNA. Targeting dCasRx or antisense oligonucleotides (ASOs) suppressed cryptic splicing, which restored axonal regeneration and stathmin-2-dependent lysosome trafficking in TDP-43-deficient human motor neurons. In mice that were gene-edited to contain human STMN2 cryptic splice-polyadenylation sequences, ASO injection into cerebral spinal fluid successfully corrected Stmn2 pre-mRNA misprocessing and restored stathmin-2 expression levels independently of TDP-43 binding.


Subject(s)
DNA-Binding Proteins , Gene Editing , Polyadenylation , RNA Splicing , Stathmin , TDP-43 Proteinopathies , Animals , Humans , Mice , DNA-Binding Proteins/metabolism , RNA Precursors/genetics , RNA Precursors/metabolism , Stathmin/genetics , Stathmin/metabolism , TDP-43 Proteinopathies/genetics , TDP-43 Proteinopathies/therapy , RNA Splice Sites , Oligonucleotides, Antisense/genetics , Neuronal Outgrowth
7.
Ann Neurol ; 70(1): 84-92, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21786300

ABSTRACT

OBJECTIVE: Axonal degeneration is a main contributor to disability in progressive neurodegenerative diseases in which oxidative stress is often identified as a pathogenic factor. We aim to demonstrate that antioxidants are able to improve axonal degeneration and locomotor deficits in a mouse model of X-adrenoleukodystrophy (X-ALD). METHODS: X-ALD is a lethal disease caused by loss of function of the ABCD1 peroxisomal transporter of very long chain fatty acids (VLCFA). The mouse model for X-ALD exhibits a late onset neurological phenotype with locomotor disability and axonal degeneration in spinal cord resembling the most common phenotype of the disease, adrenomyeloneuropathy (X-AMN). Recently, we identified oxidative damage as an early event in life, and the excess of VLCFA as a generator of radical oxygen species (ROS) and oxidative damage to proteins in X-ALD. RESULTS: Here, we prove the capability of the antioxidants N-acetyl-cysteine, α-lipoic acid, and α-tocopherol to scavenge VLCFA-dependent ROS generation in vitro. Furthermore, in a preclinical setting, the cocktail of the 3 compounds reversed: (1) oxidative stress and lesions to proteins, (2) immunohistological signs of axonal degeneration, and (3) locomotor impairment in bar cross and treadmill tests. INTERPRETATION: We have established a direct link between oxidative stress and axonal damage in a mouse model of neurodegenerative disease. This conceptual proof of oxidative stress as a major disease-driving factor in X-AMN warrants translation into clinical trials for X-AMN, and invites assessment of antioxidant strategies in axonopathies in which oxidative damage might be a contributing factor.


Subject(s)
Adrenoleukodystrophy/metabolism , Antioxidants/therapeutic use , Axons/metabolism , Disease Models, Animal , Nerve Degeneration/metabolism , Adrenoleukodystrophy/drug therapy , Adrenoleukodystrophy/pathology , Animals , Antioxidants/pharmacology , Axons/drug effects , Axons/pathology , Cells, Cultured , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Nerve Degeneration/drug therapy , Nerve Degeneration/pathology , Oxidative Stress/drug effects , Oxidative Stress/physiology , Random Allocation
8.
Hum Mol Genet ; 17(12): 1762-73, 2008 Jun 15.
Article in English | MEDLINE | ID: mdl-18344354

ABSTRACT

X-linked adrenoleukodystrophy (X-ALD) is a fatal neurodegenerative disorder, characterized by progressive cerebral demyelination cerebral childhood adrenoleukodystrophy (CCALD) or spinal cord neurodegeneration (adrenomyeloneuropathy, AMN), adrenal insufficiency and accumulation of very long-chain fatty acids (VLCFA) in tissues. The disease is caused by mutations in the ABCD1 gene, which encodes a peroxisomal transporter that plays a role in the import of VLCFA or VLCFA-CoA into peroxisomes. The Abcd1 knockout mice develop a spinal cord disease that mimics AMN in adult patients, with late onset at 20 months of age. The mechanisms underlying cerebral demyelination or axonal degeneration in spinal cord are unknown. Here, we present evidence by gas chromatography/mass spectrometry that malonaldehyde-lysine, a consequence of lipoxidative damage to proteins, accumulates in the spinal cord of Abcd1 knockout mice as early as 3.5 months of age. At 12 months, Abcd1- mice accumulate additional proteins modified by oxidative damage arising from metal-catalyzed oxidation and glycoxidation/lipoxidation. While we show that VLCFA excess activates enzymatic antioxidant defenses at the protein expression levels, both in neural tissue, in ex vivo organotypic spinal cord slices from Abcd1- mice, and in human ALD fibroblasts, we also demonstrate that the loss of Abcd1 gene function hampers oxidative stress homeostasis. We find that the alpha-tocopherol analog Trolox is able to reverse oxidative lesions in vitro, thus providing therapeutic hope. These results pave the way for the identification of therapeutic targets that could reverse the deregulated response to oxidative stress in X-ALD.


Subject(s)
Adrenoleukodystrophy/metabolism , Oxidation-Reduction , Spinal Cord/metabolism , Animals , Catalase/metabolism , Chemokine CCL22/genetics , Chemokine CCL22/metabolism , Chromans/pharmacology , Fatty Acids/metabolism , Fibroblasts/metabolism , Humans , In Vitro Techniques , Lysine/metabolism , Malondialdehyde/metabolism , Membrane Potential, Mitochondrial/drug effects , Mice , Mice, Inbred C57BL , Motor Neurons/enzymology , Motor Neurons/metabolism , Oxidative Stress , Superoxide Dismutase
9.
Nat Neurosci ; 22(2): 180-190, 2019 02.
Article in English | MEDLINE | ID: mdl-30643298

ABSTRACT

Amyotrophic lateral sclerosis (ALS) and frontotemporal dementia (FTD) are associated with loss of nuclear transactive response DNA-binding protein 43 (TDP-43). Here we identify that TDP-43 regulates expression of the neuronal growth-associated factor stathmin-2. Lowered TDP-43 levels, which reduce its binding to sites within the first intron of stathmin-2 pre-messenger RNA, uncover a cryptic polyadenylation site whose utilization produces a truncated, non-functional mRNA. Reduced stathmin-2 expression is found in neurons trans-differentiated from patient fibroblasts expressing an ALS-causing TDP-43 mutation, in motor cortex and spinal motor neurons from patients with sporadic ALS and familial ALS with GGGGCC repeat expansion in the C9orf72 gene, and in induced pluripotent stem cell (iPSC)-derived motor neurons depleted of TDP-43. Remarkably, while reduction in TDP-43 is shown to inhibit axonal regeneration of iPSC-derived motor neurons, rescue of stathmin-2 expression restores axonal regenerative capacity. Thus, premature polyadenylation-mediated reduction in stathmin-2 is a hallmark of ALS-FTD that functionally links reduced nuclear TDP-43 function to enhanced neuronal vulnerability.


Subject(s)
DNA-Binding Proteins/metabolism , Membrane Proteins/metabolism , Motor Neurons/metabolism , Nerve Degeneration/metabolism , Amyotrophic Lateral Sclerosis/metabolism , Amyotrophic Lateral Sclerosis/pathology , Female , Humans , Motor Cortex/metabolism , Motor Cortex/pathology , Motor Neurons/pathology , Nerve Degeneration/pathology , Polyadenylation , Spinal Cord/metabolism , Spinal Cord/pathology , Stathmin
10.
Brain Pathol ; 28(5): 611-630, 2018 09.
Article in English | MEDLINE | ID: mdl-29027761

ABSTRACT

Pelizaeus-Merzbacher disease (PMD) is a fatal hypomyelinating disorder characterized by early impairment of motor development, nystagmus, choreoathetotic movements, ataxia and progressive spasticity. PMD is caused by variations in the proteolipid protein gene PLP1, which encodes the two major myelin proteins of the central nervous system, PLP and its spliced isoform DM20, in oligodendrocytes. Large duplications including the entire PLP1 gene are the most frequent causative mutation leading to the classical form of PMD. The Plp1 overexpressing mouse model (PLP-tg66/66 ) develops a phenotype very similar to human PMD, with early and severe motor dysfunction and a dramatic decrease in lifespan. The sequence of cellular events that cause neurodegeneration and ultimately death is poorly understood. In this work, we analyzed patient-derived fibroblasts and spinal cords of the PLP-tg66/66 mouse model, and identified redox imbalance, with altered antioxidant defense and oxidative damage to several enzymes involved in ATP production, such as glycolytic enzymes, creatine kinase and mitochondrial proteins from the Krebs cycle and oxidative phosphorylation. We also evidenced malfunction of the mitochondria compartment with increased ROS production and depolarization in PMD patient's fibroblasts, which was prevented by the antioxidant N-acetyl-cysteine. Finally, we uncovered an impairment of mitochondrial dynamics in patient's fibroblasts which may help explain the ultrastructural abnormalities of mitochondria morphology detected in spinal cords from PLP-tg66/66 mice. Altogether, these results underscore the link between redox and metabolic homeostasis in myelin diseases, provide insight into the pathophysiology of PMD, and may bear implications for tailored pharmacological intervention.


Subject(s)
Mitochondrial Dynamics , Oxidative Stress , Pelizaeus-Merzbacher Disease/metabolism , Animals , Cells, Cultured , Child , Child, Preschool , DNA, Mitochondrial , Fibroblasts/metabolism , Fibroblasts/pathology , Glutamic Acid/metabolism , Humans , Infant , Male , Mice, Inbred C57BL , Mice, Transgenic , Mitochondria/metabolism , Mitochondria/pathology , Mitochondrial Proteins/metabolism , Myelin Proteolipid Protein/genetics , Myelin Proteolipid Protein/metabolism , Pelizaeus-Merzbacher Disease/pathology , RNA, Messenger/metabolism , Spinal Cord/metabolism , Spinal Cord/pathology
11.
Neuron ; 100(4): 816-830.e7, 2018 11 21.
Article in English | MEDLINE | ID: mdl-30344044

ABSTRACT

Through the generation of humanized FUS mice expressing full-length human FUS, we identify that when expressed at near endogenous murine FUS levels, both wild-type and ALS-causing and frontotemporal dementia (FTD)-causing mutations complement the essential function(s) of murine FUS. Replacement of murine FUS with mutant, but not wild-type, human FUS causes stress-mediated induction of chaperones, decreased expression of ion channels and transporters essential for synaptic function, and reduced synaptic activity without loss of nuclear FUS or its cytoplasmic aggregation. Most strikingly, accumulation of mutant human FUS is shown to activate an integrated stress response and to inhibit local, intra-axonal protein synthesis in hippocampal neurons and sciatic nerves. Collectively, our evidence demonstrates that human ALS/FTD-linked mutations in FUS induce a gain of toxicity that includes stress-mediated suppression in intra-axonal translation, synaptic dysfunction, and progressive age-dependent motor and cognitive disease without cytoplasmic aggregation, altered nuclear localization, or aberrant splicing of FUS-bound pre-mRNAs. VIDEO ABSTRACT.


Subject(s)
Amyotrophic Lateral Sclerosis/genetics , Axons/physiology , Frontotemporal Dementia/genetics , Loss of Function Mutation/genetics , Protein Biosynthesis/physiology , RNA-Binding Protein FUS/genetics , Amyotrophic Lateral Sclerosis/metabolism , Amyotrophic Lateral Sclerosis/pathology , Animals , Axons/pathology , Cells, Cultured , Female , Frontotemporal Dementia/metabolism , Frontotemporal Dementia/pathology , Hippocampus/metabolism , Hippocampus/pathology , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Pregnancy , RNA-Binding Protein FUS/biosynthesis
13.
Biochimie ; 98: 143-9, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24076127

ABSTRACT

X-linked adrenoleukodystrophy (X-ALD) is the most frequent inherited monogenic demyelinating disease (minimal incidence 1:17,000). It is often lethal and currently lacks a satisfactory therapy. The disease is caused by loss of function of the ABCD1 gene, a peroxisomal ATP-binding cassette transporter, resulting in the accumulation of VLCFA (very long-chain fatty acids) in organs and plasma. Understanding of the aetiopathogenesis is a prerequisite for the development of novel therapeutic strategies. Functional genomics analysis of an ABCD1 null mouse, a mouse model for adrenomyeloneuropathy, has revealed presymptomatic alterations in several metabolic pathways converging on redox and bioenergetic homeostasis, with failure of mitochondrial OXPHOS disruption and mitochondrial depletion. These defects could be major contributors to the neurodegenerative cascade, as has been reported in several neurodegenerative disorders. Drugs targeting the redox imbalance/mitochondria dysfunction interplay have shown efficacy at halting axonal degeneration and associated disability in the mouse, and thus offer therapeutic hope.


Subject(s)
ATP-Binding Cassette Transporters/deficiency , Adrenoleukodystrophy/physiopathology , Axons/pathology , ATP Binding Cassette Transporter, Subfamily D, Member 1 , Animals , Disease Models, Animal , Energy Metabolism , Fatty Acids/metabolism , Mice , Mice, Knockout , Mitochondria/metabolism , Neurodegenerative Diseases/pathology , Oxidative Stress , Proteasome Endopeptidase Complex/metabolism
14.
Antioxid Redox Signal ; 15(8): 2095-107, 2011 Oct 15.
Article in English | MEDLINE | ID: mdl-21453200

ABSTRACT

AIMS: Chronic metabolic impairment and oxidative stress are associated with the pathogenesis of axonal dysfunction in a growing number of neurodegenerative conditions. To investigate the intertwining of both noxious factors, we have chosen the mouse model of adrenoleukodystrophy (X-ALD), which exhibits axonal degeneration in spinal cords and motor disability. The disease is caused by loss of function of the ABCD1 transporter, involved in the import and degradation of very long-chain fatty acids (VLCFA) in peroxisomes. Oxidative stress due to VLCFA excess appears early in the neurodegenerative cascade. RESULTS: In this study, we demonstrate by redox proteomics that oxidative damage to proteins specifically affects five key enzymes of glycolysis and TCA (Tricarboxylic acid) cycle in spinal cords of Abcd1(-) mice and pyruvate kinase in human X-ALD fibroblasts. We also show that NADH and ATP levels are significantly diminished in these samples, together with decrease of pyruvate kinase activities and GSH levels, and increase of NADPH. INNOVATION: Treating Abcd1(-) mice with the antioxidants N-acetylcysteine and α-lipoic acid (LA) prevents protein oxidation; preserves NADH, NADPH, ATP, and GSH levels; and normalizes pyruvate kinase activity, which implies that oxidative stress provoked by VLCFA results in bioenergetic dysfunction, at a presymptomatic stage. CONCLUSION: Our results provide mechanistic insight into the beneficial effects of antioxidants and enhance the rationale for translation into clinical trials for X-adrenoleukodystrophy.


Subject(s)
Adrenoleukodystrophy/metabolism , Energy Metabolism/physiology , Oxidative Stress/physiology , ATP Binding Cassette Transporter, Subfamily D, Member 1 , ATP-Binding Cassette Transporters/genetics , ATP-Binding Cassette Transporters/metabolism , Adenosine Triphosphate/metabolism , Adrenoleukodystrophy/genetics , Animals , Blotting, Western , Cells, Cultured , Disease Models, Animal , Electrophoresis, Gel, Two-Dimensional , Energy Metabolism/genetics , Glutathione/metabolism , Humans , Male , Mice , Mice, Inbred C57BL , NAD/metabolism , Oxidative Stress/genetics , Proteomics , Pyruvate Kinase/genetics , Pyruvate Kinase/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL