Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 24
Filter
Add more filters










Publication year range
1.
Neurol Genet ; 9(2): e200058, 2023 Apr.
Article in English | MEDLINE | ID: mdl-37090936

ABSTRACT

Background and Objectives: Coenzyme Q10 (CoQ10)-deficient cerebellar ataxia can be due to pathogenic variants in genes encoding for CoQ10 biosynthetic proteins or associated with defects in protein unrelated to its biosynthesis. Diagnosis is crucial because patients may respond favorably to CoQ10 supplementation. The aim of this study was to identify through whole-exome sequencing (WES) the pathogenic variants, and assess CoQ10 levels, in fibroblasts from patients with undiagnosed cerebellar ataxia referred to investigate CoQ10 deficiency. Methods: WES was performed on genomic DNA extracted from 16 patients. Sequencing data were filtered using a virtual panel of genes associated with CoQ10 deficiency and/or cerebellar ataxia. CoQ10 levels were measured by high-performance liquid chromatography in 14 patient-derived fibroblasts. Results: A definite genetic etiology was identified in 8 samples of 16 (diagnostic yield = 50%). The identified genetic causes were pathogenic variants of the genes COQ8A (ADCK3) (n = 3 samples), ATP1A3 (n = 2), PLA2G6 (n = 1), SPG7 (n = 1), and MFSD8 (n = 1). Five novel mutations were found (COQ8A n = 3, PLA2G6 n = 1, and MFSD8 n = 1). CoQ10 levels were significantly decreased in 3/14 fibroblast samples (21.4%), 1 carrying compound heterozygous COQ8A pathogenic variants, 1 harboring a homozygous pathogenic SPG7 variant, and 1 with an unknown molecular defect. Discussion: This work confirms the importance of COQ8A gene mutations as a frequent genetic cause of cerebellar ataxia and CoQ10 deficiency and suggests SPG7 mutations as a novel cause of secondary CoQ10 deficiency.

2.
Ann Neurol ; 90(4): 640-652, 2021 10.
Article in English | MEDLINE | ID: mdl-34338329

ABSTRACT

OBJECTIVE: Autosomal recessive human thymidine kinase 2 (TK2) mutations cause TK2 deficiency, which typically manifests as a progressive and fatal mitochondrial myopathy in infants and children. Treatment with pyrimidine deoxynucleosides deoxycytidine and thymidine ameliorates mitochondrial defects and extends the lifespan of Tk2 knock-in mouse (Tk2KI ) and compassionate use deoxynucleoside therapy in TK2 deficient patients have shown promising indications of efficacy. To augment therapy for Tk2 deficiency, we assessed gene therapy alone and in combination with deoxynucleoside therapy in Tk2KI mice. METHODS: We generated pAAVsc CB6 PI vectors containing human TK2 cDNA (TK2). Adeno-associated virus (AAV)-TK2 was administered to Tk2KI , which were serially assessed for weight, motor functions, and survival as well as biochemical functions in tissues. AAV-TK2 treated mice were further treated with deoxynucleosides. RESULTS: AAV9 delivery of human TK2 cDNA to Tk2KI mice efficiently rescued Tk2 activity in all the tissues tested except the kidneys, delayed disease onset, and increased lifespan. Sequential treatment of Tk2KI mice with AAV9 first followed by AAV2 at different ages allowed us to reduce the viral dose while further prolonging the lifespan. Furthermore, addition of deoxycytidine and deoxythymidine supplementation to AAV9 + AAV2 treated Tk2KI mice dramatically improved mtDNA copy numbers in the liver and kidneys, animal growth, and lifespan. INTERPRETATION: Our data indicate that AAV-TK2 gene therapy as well as combination deoxynucleoside and gene therapies is more effective in Tk2KI mice than pharmacological alone. Thus, combination of gene therapy with substrate enhancement is a promising therapeutic approach for TK2 deficiency and potentially other metabolic disorders. ANN NEUROL 2021;90:640-652.


Subject(s)
Genetic Therapy , Mitochondria/metabolism , Mitochondrial Myopathies/therapy , Thymidine Kinase/deficiency , Animals , Compassionate Use Trials , DNA, Mitochondrial/genetics , Humans , Mice , Mitochondria/genetics , Mitochondrial Myopathies/genetics , Mutation/genetics , Thymidine/genetics , Thymidine/metabolism , Thymidine Kinase/genetics
3.
Autophagy ; 17(8): 1889-1906, 2021 08.
Article in English | MEDLINE | ID: mdl-32686621

ABSTRACT

SETX (senataxin) is an RNA/DNA helicase that has been implicated in transcriptional regulation and the DNA damage response through resolution of R-loop structures. Mutations in SETX result in either of two distinct neurodegenerative disorders. SETX dominant mutations result in a juvenile form of amyotrophic lateral sclerosis (ALS) called ALS4, whereas recessive mutations are responsible for ataxia called ataxia with oculomotor apraxia type 2 (AOA2). How mutations in the same protein can lead to different phenotypes is still unclear. To elucidate AOA2 disease mechanisms, we first examined gene expression changes following SETX depletion. We observed the effects on both transcription and RNA processing, but surprisingly observed decreased R-loop accumulation in SETX-depleted cells. Importantly, we discovered a strong connection between SETX and the macroautophagy/autophagy pathway, reflecting a direct effect on transcription of autophagy genes. We show that SETX depletion inhibits the progression of autophagy, leading to an accumulation of ubiquitinated proteins, decreased ability to clear protein aggregates, as well as mitochondrial defects. Analysis of AOA2 patient fibroblasts also revealed a perturbation of the autophagy pathway. Our work has thus identified a novel function for SETX in the regulation of autophagy, whose modulation may have a therapeutic impact for AOA2.Abbreviations: 3'READS: 3' region extraction and deep sequencing; ACTB: actin beta; ALS4: amyotrophic lateral sclerosis type 4; AOA2: ataxia with oculomotor apraxia type 2; APA: alternative polyadenylation; AS: alternative splicing; ATG7: autophagy-related 7; ATP6V0D2: ATPase H+ transporting V0 subunit D2; BAF: bafilomycin A1; BECN1: beclin 1; ChIP: chromatin IP; Chloro: chloroquine; CPT: camptothecin; DDR: DNA damage response; DNMT1: DNA methyltransferase 1; DRIP: DNA/RNA IP; DSBs: double strand breaks; EBs: embryoid bodies; FTD: frontotemporal dementia; GABARAP: GABA type A receptor-associated protein; GO: gene ontology; HR: homologous recombination; HTT: huntingtin; IF: immunofluorescence; IP: immunoprecipitation; iPSCs: induced pluripotent stem cells; KD: knockdown; MAP1LC3/LC3: microtubule associated protein 1 light chain 3; MN: motor neuron; MTORC1: mechanistic target of rapamycin kinase complex 1; PASS: PolyA Site Supporting; PFA: paraformaldehyde; RNAPII: RNA polymerase II; SCA: spinocerebellar ataxia; SETX: senataxin; SMA: spinal muscular atrophy; SMN1: survival of motor neuron 1, telomeric; SQSTM1/p62: sequestosome 1; TFEB: transcription factor EB; TSS: transcription start site; TTS: transcription termination site; ULK1: unc-51 like autophagy activating kinase 1; WB: western blot; WIPI2: WD repeat domain, phosphoinositide interacting 2; XRN2: 5'-3' exoribonuclease 2.


Subject(s)
Amyotrophic Lateral Sclerosis/metabolism , Autophagy/physiology , DNA Helicases/metabolism , Multifunctional Enzymes/metabolism , RNA Helicases/metabolism , Gene Expression Regulation/genetics , Humans , Motor Neurons/metabolism
4.
EBioMedicine ; 46: 356-367, 2019 Aug.
Article in English | MEDLINE | ID: mdl-31383553

ABSTRACT

BACKGROUND: TK2 is a nuclear gene encoding the mitochondrial matrix protein thymidine kinase 2 (TK2), a critical enzyme in the mitochondrial nucleotide salvage pathway. Deficiency of TK2 activity causes mitochondrial DNA (mtDNA) depletion, which in humans manifests predominantly as a mitochondrial myopathy with onset typically in infancy and childhood. We previously showed that oral treatment of the Tk2 H126N knock-in mouse model (Tk2-/-) with the TK2 substrates, deoxycytidine (dCtd) and thymidine (dThd), delayed disease onset and prolonged median survival by 3-fold. Nevertheless, dCtd + dThd treated Tk2-/- mice showed mtDNA depletion in brain as early as postnatal day 13 and in virtually all other tissues at age 29 days. METHODS: To enhance mechanistic understanding and efficacy of dCtd + dThd therapy, we studied the bioavailability of dCtd and dThd in various tissues as well as levels of the cytosolic enzymes, TK1 and dCK that convert the deoxynucleosides into dCMP and dTMP. FINDINGS: Parenteral treatment relative to oral treatment produced higher levels of dCtd and dThd and improved mtDNA levels in liver and heart, but did not ameliorate molecular defects in brain or prolong survival. Down-regulation of TK1 correlated with temporal- and tissue-specificity of response to dCtd + dThd. Finally, we observed in human infant and adult muscle expression of TK1 and dCK, which account for the long-term efficacy to dCtd + dThd therapy in TK2 deficient patients. INTERPRETATIONS: These data indicate that the cytosolic pyrimidine salvage pathway enzymes TK1 and dCK are critical for therapeutic efficacy of deoxynucleoside therapy for Tk2 deficiency. FUND: National Institutes of Health P01HD32062.


Subject(s)
Deoxyribonucleosides/pharmacology , Thymidine Kinase/deficiency , Animals , Biological Availability , Blood-Brain Barrier/metabolism , DNA, Mitochondrial , Deoxyribonucleosides/pharmacokinetics , Disease Models, Animal , Enzyme Activation/drug effects , Humans , Mice , Mice, Knockout , Mice, Transgenic , Mitochondria/genetics , Mitochondria/metabolism , Muscle, Skeletal/metabolism , Organ Specificity , Oxidative Phosphorylation , Phenotype , Thymidine Kinase/genetics , Thymidine Kinase/metabolism
5.
Biochim Biophys Acta Mol Basis Dis ; 1864(11): 3708-3722, 2018 11.
Article in English | MEDLINE | ID: mdl-30251690

ABSTRACT

Nephrotic syndrome (NS), a frequent chronic kidney disease in children and young adults, is the most common phenotype associated with primary coenzyme Q10 (CoQ10) deficiency and is very responsive to CoQ10 supplementation, although the pathomechanism is not clear. Here, using a mouse model of CoQ deficiency-associated NS, we show that long-term oral CoQ10 supplementation prevents kidney failure by rescuing defects of sulfides oxidation and ameliorating oxidative stress, despite only incomplete normalization of kidney CoQ levels and lack of rescue of CoQ-dependent respiratory enzymes activities. Liver and kidney lipidomics, and urine metabolomics analyses, did not show CoQ metabolites. To further demonstrate that sulfides metabolism defects cause oxidative stress in CoQ deficiency, we show that silencing of sulfide quinone oxido-reductase (SQOR) in wild-type HeLa cells leads to similar increases of reactive oxygen species (ROS) observed in HeLa cells depleted of the CoQ biosynthesis regulatory protein COQ8A. While CoQ10 supplementation of COQ8A depleted cells decreases ROS and increases SQOR protein levels, knock-down of SQOR prevents CoQ10 antioxidant effects. We conclude that kidney failure in CoQ deficiency-associated NS is caused by oxidative stress mediated by impaired sulfides oxidation and propose that CoQ supplementation does not significantly increase the kidney pool of CoQ bound to the respiratory supercomplexes, but rather enhances the free pool of CoQ, which stabilizes SQOR protein levels rescuing oxidative stress.


Subject(s)
Antioxidants/pharmacology , Ataxia/drug therapy , Hydrogen Sulfide/metabolism , Mitochondrial Diseases/drug therapy , Muscle Weakness/drug therapy , Nephrotic Syndrome/drug therapy , Ubiquinone/analogs & derivatives , Ubiquinone/deficiency , Alkyl and Aryl Transferases/genetics , Animals , Antioxidants/therapeutic use , Ataxia/complications , Ataxia/metabolism , Disease Models, Animal , HeLa Cells , Humans , Kidney/metabolism , Kidney/pathology , Metabolic Networks and Pathways/drug effects , Mice , Mice, Transgenic , Mitochondria/drug effects , Mitochondria/metabolism , Mitochondrial Diseases/complications , Mitochondrial Diseases/metabolism , Muscle Weakness/complications , Muscle Weakness/metabolism , Nephrotic Syndrome/etiology , Nephrotic Syndrome/metabolism , Nephrotic Syndrome/pathology , Oxidation-Reduction/drug effects , Oxidative Stress/drug effects , Oxidoreductases Acting on Sulfur Group Donors/genetics , Oxidoreductases Acting on Sulfur Group Donors/metabolism , Reactive Oxygen Species/metabolism , Ubiquinone/metabolism , Ubiquinone/pharmacology , Ubiquinone/therapeutic use
6.
Hum Mol Genet ; 27(19): 3305-3312, 2018 10 01.
Article in English | MEDLINE | ID: mdl-29917077

ABSTRACT

Leigh syndrome is a frequent, heterogeneous pediatric presentation of mitochondrial oxidative phosphorylation (OXPHOS) disease, manifesting with psychomotor retardation and necrotizing lesions in brain deep gray matter. OXPHOS occurs at the inner mitochondrial membrane through the integrated activity of five protein complexes, of which complex V (CV) functions in a dimeric form to directly generate adenosine triphosphate (ATP). Mutations in several different structural CV subunits cause Leigh syndrome; however, dimerization defects have not been associated with human disease. We report four Leigh syndrome subjects from three unrelated Ashkenazi Jewish families harboring a homozygous splice-site mutation (c.87 + 1G>C) in a novel CV subunit disease gene, USMG5. The Ashkenazi population allele frequency is 0.57%. This mutation produces two USMG5 transcripts, wild-type and lacking exon 3. Fibroblasts from two Leigh syndrome probands had reduced wild-type USMG5 mRNA expression and undetectable protein. The mutation did not alter monomeric CV expression, but reduced both CV dimer expression and ATP synthesis rate. Rescue with wild-type USMG5 cDNA in proband fibroblasts restored USMG5 protein, increased CV dimerization and enhanced ATP production rate. These data demonstrate that a recurrent USMG5 splice-site founder mutation in the Ashkenazi Jewish population causes autosomal recessive Leigh syndrome by reduction of CV dimerization and ATP synthesis.


Subject(s)
Leigh Disease/genetics , Mitochondria/genetics , Mitochondrial Diseases/genetics , Mitochondrial Proton-Translocating ATPases/genetics , Adenosine Triphosphate/biosynthesis , Child , Child, Preschool , Dimerization , Exons/genetics , Founder Effect , Gene Frequency , Haplotypes , Humans , Infant , Infant, Newborn , Jews/genetics , Leigh Disease/metabolism , Leigh Disease/pathology , Male , Mitochondria/metabolism , Mitochondria/pathology , Mitochondrial Diseases/metabolism , Mitochondrial Diseases/pathology , Mutation , Oxidative Phosphorylation , RNA Splice Sites/genetics , Exome Sequencing
7.
Ann Neurol ; 81(5): 641-652, 2017 May.
Article in English | MEDLINE | ID: mdl-28318037

ABSTRACT

OBJECTIVE: Thymidine kinase 2 (TK2), a critical enzyme in the mitochondrial pyrimidine salvage pathway, is essential for mitochondrial DNA (mtDNA) maintenance. Mutations in the nuclear gene, TK2, cause TK2 deficiency, which manifests predominantly in children as myopathy with mtDNA depletion. Molecular bypass therapy with the TK2 products, deoxycytidine monophosphate (dCMP) and deoxythymidine monophosphate (dTMP), prolongs the life span of Tk2-deficient (Tk2-/- ) mice by 2- to 3-fold. Because we observed rapid catabolism of the deoxynucleoside monophosphates to deoxythymidine (dT) and deoxycytidine (dC), we hypothesized that: (1) deoxynucleosides might be the major active agents and (2) inhibition of deoxycytidine deamination might enhance dTMP+dCMP therapy. METHODS: To test these hypotheses, we assessed two therapies in Tk2-/- mice: (1) dT+dC and (2) coadministration of the deaminase inhibitor, tetrahydrouridine (THU), with dTMP+dCMP. RESULTS: We observed that dC+dT delayed disease onset, prolonged life span of Tk2-deficient mice and restored mtDNA copy number as well as respiratory chain enzyme activities and levels. In contrast, dCMP+dTMP+THU therapy decreased life span of Tk2-/- animals compared to dCMP+dTMP. INTERPRETATION: Our studies demonstrate that deoxynucleoside substrate enhancement is a novel therapy, which may ameliorate TK2 deficiency in patients. Ann Neurol 2017;81:641-652.


Subject(s)
Antimetabolites/pharmacology , Deoxycytidine Monophosphate/pharmacology , Metabolism, Inborn Errors/drug therapy , Mitochondrial Diseases/drug therapy , Tetrahydrouridine/pharmacology , Thymidine Kinase/deficiency , Thymidine/pharmacology , Animals , Antimetabolites/administration & dosage , DNA, Mitochondrial/drug effects , Deoxycytidine Monophosphate/administration & dosage , Disease Models, Animal , Drug Therapy, Combination , Metabolism, Inborn Errors/enzymology , Mice , Mice, Transgenic , Mitochondrial Diseases/enzymology , Tetrahydrouridine/administration & dosage , Thymidine/administration & dosage
8.
EMBO Mol Med ; 9(1): 96-111, 2017 01.
Article in English | MEDLINE | ID: mdl-27856618

ABSTRACT

Coenzyme Q (CoQ) is an electron acceptor for sulfide-quinone reductase (SQR), the first enzyme of the hydrogen sulfide oxidation pathway. Here, we show that lack of CoQ in human skin fibroblasts causes impairment of hydrogen sulfide oxidation, proportional to the residual levels of CoQ. Biochemical and molecular abnormalities are rescued by CoQ supplementation in vitro and recapitulated by pharmacological inhibition of CoQ biosynthesis in skin fibroblasts and ADCK3 depletion in HeLa cells. Kidneys of Pdss2kd/kd mice, which only have ~15% residual CoQ concentrations and are clinically affected, showed (i) reduced protein levels of SQR and downstream enzymes, (ii) accumulation of hydrogen sulfides, and (iii) glutathione depletion. These abnormalities were not present in brain, which maintains ~30% residual CoQ and is clinically unaffected. In Pdss2kd/kd mice, we also observed low levels of plasma and urine thiosulfate and increased blood C4-C6 acylcarnitines. We propose that impairment of the sulfide oxidation pathway induced by decreased levels of CoQ causes accumulation of sulfides and consequent inhibition of short-chain acyl-CoA dehydrogenase and glutathione depletion, which contributes to increased oxidative stress and kidney failure.


Subject(s)
Ataxia/physiopathology , Mitochondrial Diseases/physiopathology , Muscle Weakness/physiopathology , Sulfides/metabolism , Ubiquinone/deficiency , Alkyl and Aryl Transferases/deficiency , Animals , Cells, Cultured , Fibroblasts/metabolism , Humans , Mice , Mice, Knockout , Oxidation-Reduction , Quinone Reductases/analysis
9.
J Neuropathol Exp Neurol ; 75(7): 663-72, 2016 07.
Article in English | MEDLINE | ID: mdl-27235405

ABSTRACT

In familial and sporadic multiple system atrophy (MSA) patients, deficiency of coenzyme Q10 (CoQ10) has been associated with mutations in COQ2, which encodes the second enzyme in the CoQ10 biosynthetic pathway. Cerebellar ataxia is the most common presentation of CoQ10 deficiency, suggesting that the cerebellum might be selectively vulnerable to low levels of CoQ10 To investigate whether CoQ10 deficiency represents a common feature in the brains of MSA patients independent of the presence of COQ2 mutations, we studied CoQ10 levels in postmortem brains of 12 MSA, 9 Parkinson disease (PD), 9 essential tremor (ET) patients, and 12 controls. We also assessed mitochondrial respiratory chain enzyme activities, oxidative stress, mitochondrial mass, and levels of enzymes involved in CoQ biosynthesis. Our studies revealed CoQ10 deficiency in MSA cerebellum, which was associated with impaired CoQ biosynthesis and increased oxidative stress in the absence of COQ2 mutations. The levels of CoQ10 in the cerebella of ET and PD patients were comparable or higher than in controls. These findings suggest that CoQ10 deficiency may contribute to the pathogenesis of MSA. Because no disease modifying therapies are currently available, increasing CoQ10 levels by supplementation or upregulation of its biosynthesis may represent a novel treatment strategy for MSA patients.


Subject(s)
Ataxia/metabolism , Cerebellum/metabolism , Mitochondrial Diseases/metabolism , Multiple System Atrophy/metabolism , Muscle Weakness/metabolism , Ubiquinone/analogs & derivatives , Ubiquinone/deficiency , Aged , Aged, 80 and over , Ataxia/complications , Ataxia/pathology , Case-Control Studies , Cerebellum/pathology , Female , Humans , Male , Middle Aged , Mitochondrial Diseases/complications , Mitochondrial Diseases/pathology , Multiple System Atrophy/complications , Multiple System Atrophy/pathology , Muscle Weakness/complications , Muscle Weakness/pathology , Oxidative Stress/physiology , Ubiquinone/metabolism
10.
Hum Mol Genet ; 24(16): 4516-29, 2015 Aug 15.
Article in English | MEDLINE | ID: mdl-25976310

ABSTRACT

Ataxia oculomotor apraxia type 1 (AOA1) is an autosomal recessive disease caused by mutations in APTX, which encodes the DNA strand-break repair protein aprataxin (APTX). CoQ10 deficiency has been identified in fibroblasts and muscle of AOA1 patients carrying the common W279X mutation, and aprataxin has been localized to mitochondria in neuroblastoma cells, where it enhances preservation of mitochondrial function. In this study, we show that aprataxin deficiency impairs mitochondrial function, independent of its role in mitochondrial DNA repair. The bioenergetics defect in AOA1-mutant fibroblasts and APTX-depleted Hela cells is caused by decreased expression of SDHA and genes encoding CoQ biosynthetic enzymes, in association with reductions of APE1, NRF1 and NRF2. The biochemical and molecular abnormalities in APTX-depleted cells are recapitulated by knockdown of APE1 in Hela cells and are rescued by overexpression of NRF1/2. Importantly, pharmacological upregulation of NRF1 alone by 5-aminoimidazone-4-carboxamide ribonucleotide does not rescue the phenotype, which, in contrast, is reversed by the upregulation of NRF2 by rosiglitazone. Accordingly, we propose that the lack of aprataxin causes reduction of the pathway APE1/NRF1/NRF2 and their target genes. Our findings demonstrate a critical role of APTX in transcription regulation of mitochondrial function and the pathogenesis of AOA1 via a novel pathomechanistic pathway, which may be relevant to other neurodegenerative diseases.


Subject(s)
DNA-(Apurinic or Apyrimidinic Site) Lyase/biosynthesis , DNA-Binding Proteins/deficiency , Down-Regulation , Fibroblasts/metabolism , Mitochondria/metabolism , NF-E2-Related Factor 2/biosynthesis , Nuclear Proteins/deficiency , Nuclear Respiratory Factor 1/biosynthesis , Signal Transduction , Ataxia/genetics , Ataxia/metabolism , Ataxia/pathology , DNA-(Apurinic or Apyrimidinic Site) Lyase/genetics , DNA-Binding Proteins/genetics , Female , Fibroblasts/pathology , Genetic Diseases, Inborn/genetics , Genetic Diseases, Inborn/metabolism , Genetic Diseases, Inborn/pathology , Humans , Male , Mitochondria/pathology , NF-E2-Related Factor 2/genetics , Nuclear Proteins/genetics , Nuclear Respiratory Factor 1/genetics
11.
Ann Neurol ; 76(4): 620-4, 2014 Oct.
Article in English | MEDLINE | ID: mdl-25090982

ABSTRACT

Energy metabolism could influence amyotrophic lateral sclerosis (ALS) and progressive lateral sclerosis (PLS) pathogenesis and the response to therapy. We developed a novel assay to simultaneously assess mitochondrial content and membrane potential in patients' skin fibroblasts. In ALS and PLS fibroblasts, membrane potential was increased and mitochondrial content decreased, relative to healthy controls. In ALS higher mitochondrial membrane potential correlated with age at diagnosis, and in PLS it correlated with disease severity. These unprecedented findings in ALS and PLS fibroblasts could shed new light onto disease pathogenesis and help in developing biomarkers to predict disease evolution and the individual response to therapy in motor neuron diseases.


Subject(s)
Amyotrophic Lateral Sclerosis/pathology , Energy Metabolism/physiology , Fibroblasts/pathology , Motor Neuron Disease/pathology , Skin/pathology , Adult , Aged , Aldehydes , Biomarkers , Humans , Male , Membrane Potential, Mitochondrial/physiology , Middle Aged , Rhodamines/metabolism
12.
EMBO Mol Med ; 6(8): 1016-27, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24968719

ABSTRACT

Autosomal recessive mutations in the thymidine kinase 2 gene (TK2) cause mitochondrial DNA depletion, multiple deletions, or both due to loss of TK2 enzyme activity and ensuing unbalanced deoxynucleotide triphosphate (dNTP) pools. To bypass Tk2 deficiency, we administered deoxycytidine and deoxythymidine monophosphates (dCMP+dTMP) to the Tk2 H126N (Tk2(-/-)) knock-in mouse model from postnatal day 4, when mutant mice are phenotypically normal, but biochemically affected. Assessment of 13-day-old Tk2(-/-) mice treated with dCMP+dTMP 200 mg/kg/day each (Tk2(-/-200dCMP/) (dTMP)) demonstrated that in mutant animals, the compounds raise dTTP concentrations, increase levels of mtDNA, ameliorate defects of mitochondrial respiratory chain enzymes, and significantly prolong their lifespan (34 days with treatment versus 13 days untreated). A second trial of dCMP+dTMP each at 400 mg/kg/day showed even greater phenotypic and biochemical improvements. In conclusion, dCMP/dTMP supplementation is the first effective pharmacologic treatment for Tk2 deficiency.


Subject(s)
Mitochondrial Diseases/drug therapy , Thymidine Kinase/deficiency , Thymidine Monophosphate/therapeutic use , Animals , Deoxycytidine Monophosphate/therapeutic use , Gene Knock-In Techniques , Mice , Survival Analysis , Treatment Outcome
13.
FASEB J ; 27(2): 612-21, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23150520

ABSTRACT

Primary human CoQ(10) deficiencies are clinically heterogeneous diseases caused by mutations in PDSS2 and other genes required for CoQ(10) biosynthesis. Our in vitro studies of PDSS2 mutant fibroblasts, with <20% CoQ(10) of control cells, revealed reduced activity of CoQ(10)-dependent complex II+III and ATP synthesis, without amplification of reactive oxygen species (ROS), markers of oxidative damage, or antioxidant defenses. In contrast, COQ2 and ADCK3 mutant fibroblasts, with 30-50% CoQ(10) of controls, showed milder bioenergetic defects but significantly increased ROS and oxidation of lipids and proteins. We hypothesized that absence of oxidative stress markers and cell death in PDSS2 mutant fibroblasts were due to the extreme severity of CoQ(10) deficiency. Here, we have investigated in vivo effects of Pdss2 deficiency in affected and unaffected organs of CBA/Pdss2(kd/kd) mice at presymptomatic, phenotypic-onset, and end-stages of the disease. Although Pdss2 mutant mice manifest widespread CoQ(9) deficiency and mitochondrial respiratory chain abnormalities, only affected organs show increased ROS production, oxidative stress, mitochondrial DNA depletion, and reduced citrate synthase activity, an index of mitochondrial mass. Our data indicate that kidney-specific loss of mitochondria triggered by oxidative stress may be the cause of renal failure in Pdss2(kd/kd) mice.


Subject(s)
Alkyl and Aryl Transferases/deficiency , Alkyl and Aryl Transferases/genetics , Mitochondria/metabolism , Ubiquinone/deficiency , Adenosine Diphosphate/metabolism , Adenosine Triphosphate/metabolism , Animals , DNA, Mitochondrial/genetics , DNA, Mitochondrial/metabolism , Electron Transport , Fibroblasts/metabolism , Humans , Kidney/metabolism , Kidney/pathology , Mice , Mice, Inbred CBA , Mice, Mutant Strains , Oxidative Stress , Tissue Distribution
14.
Am J Hum Genet ; 91(4): 729-36, 2012 Oct 05.
Article in English | MEDLINE | ID: mdl-23022099

ABSTRACT

Defects of mitochondrial protein synthesis are clinically and genetically heterogeneous. We previously described a male infant who was born to consanguineous parents and who presented with severe congenital encephalopathy, peripheral neuropathy, myopathy, and lactic acidosis associated with deficiencies of multiple mitochondrial respiratory-chain enzymes and defective mitochondrial translation. In this work, we have characterized four additional affected family members, performed homozygosity mapping, and identified a homozygous splicing mutation in the splice donor site of exon 2 (c.504+1G>A) of RMND1 (required for meiotic nuclear division-1) in the affected individuals. Fibroblasts from affected individuals expressed two aberrant transcripts and had decreased wild-type mRNA and deficiencies of mitochondrial respiratory-chain enzymes. The RMND1 mutation caused haploinsufficiency that was rescued by overexpression of the wild-type transcript in mutant fibroblasts; this overexpression increased the levels and activities of mitochondrial respiratory-chain proteins. Knockdown of RMND1 via shRNA recapitulated the biochemical defect of the mutant fibroblasts, further supporting a loss-of-function pathomechanism in this disease. RMND1 belongs to the sif2 family, an evolutionary conserved group of proteins that share the DUF155 domain, have unknown function, and have never been associated with human disease. We documented that the protein localizes to mitochondria in mammalian and yeast cells. Further studies are necessary for understanding the function of this protein in mitochondrial protein translation.


Subject(s)
Cell Cycle Proteins/genetics , Mitochondria/genetics , Mitochondrial Encephalomyopathies/genetics , Mitochondrial Proteins/genetics , Mutation , Protein Biosynthesis , Consanguinity , DNA, Mitochondrial/genetics , Exons , Fibroblasts/metabolism , Genetic Predisposition to Disease , Homozygote , Humans , Infant, Newborn , Male , Mitochondrial Encephalomyopathies/metabolism , RNA Splice Sites/genetics , RNA Splicing/genetics , RNA, Messenger/genetics
15.
Arch Neurol ; 69(8): 978-83, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22490322

ABSTRACT

Coenzyme Q(10) (CoQ(10)) deficiency has been associated with 5 major clinical phenotypes: encephalomyopathy, severe infantile multisystemic disease, nephropathy, cerebellar ataxia, and isolated myopathy. Primary CoQ(10) deficiency is due to defects in CoQ(10) biosynthesis, while secondary forms are due to other causes. A review of 149 cases, including our cohort of 76 patients, confirms that CoQ(10) deficiency is a clinically and genetically heterogeneous syndrome that mainly begins in childhood and predominantly manifests as cerebellar ataxia. Coenzyme Q(10) measurement in muscle is the gold standard for diagnosis. Identification of CoQ(10) deficiency is important because the condition frequently responds to treatment. Causative mutations have been identified in a small proportion of patients.


Subject(s)
Genetic Heterogeneity , Ubiquinone/analogs & derivatives , Animals , Cerebellar Ataxia/genetics , Cerebellar Ataxia/therapy , Cohort Studies , Humans , Ubiquinone/biosynthesis , Ubiquinone/deficiency , Ubiquinone/therapeutic use
16.
PLoS One ; 7(2): e30606, 2012.
Article in English | MEDLINE | ID: mdl-22359546

ABSTRACT

Coenzyme Q(10) (CoQ(10)) is a potent lipophilic antioxidant in cell membranes and a carrier of electrons in the mitochondrial respiratory chain. We previously characterized the effects of varying severities of CoQ(10) deficiency on ROS production and mitochondrial bioenergetics in cells harboring genetic defects of CoQ(10) biosynthesis. We observed a unimodal distribution of ROS production with CoQ(10) deficiency: cells with <20% of CoQ(10) and 50-70% of CoQ(10) did not generate excess ROS while cells with 30-45% of CoQ(10) showed increased ROS production and lipid peroxidation. Because our previous studies were limited to a small number of mutant cell lines with heterogeneous molecular defects, here, we treated 5 control and 2 mildly CoQ(10) deficient fibroblasts with varying doses of 4-nitrobenzoate (4-NB), an analog of 4-hydroxybenzoate (4-HB) and inhibitor of 4-para-hydroxybenzoate:polyprenyl transferase (COQ2) to induce a range of CoQ(10) deficiencies. Our results support the concept that the degree of CoQ(10) deficiency in cells dictates the extent of ATP synthesis defects and ROS production and that 40-50% residual CoQ(10) produces maximal oxidative stress and cell death.


Subject(s)
Fibroblasts/drug effects , Nitrobenzoates/pharmacology , Ubiquinone/analogs & derivatives , Adenosine Triphosphate/biosynthesis , Cell Death , Dose-Response Relationship, Drug , Humans , Oxidative Stress , Reactive Oxygen Species , Ubiquinone/antagonists & inhibitors , Ubiquinone/biosynthesis
17.
Brain ; 134(Pt 11): 3326-32, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21933806

ABSTRACT

Mitochondrial neurogastrointestinal encephalomyopathy is a rare multisystemic autosomic recessive disorder characterized by: onset typically before the age of 30 years; ptosis; progressive external ophthalmoplegia; gastrointestinal dysmotility; cachexia; peripheral neuropathy; and leucoencephalopathy. The disease is caused by mutations in the TYMP gene encoding thymidine phosphorylasethymine phosphorylase. Anecdotal reports suggest that allogeneic haematopoetic stem cell transplantation may be beneficial for mitochondrial neurogastrointestinal encephalomyopathy, but is associated with a high mortality. After selecting patients who fulfilled the clinical criteria for mitochondrial neurogastrointestinal encephalomyopathy and had severe thymidine phosphorylase deficiency in the buffy coat (<10% of normal activity), we reviewed their medical records and laboratory studies. We identified 102 patients (50 females) with mitochondrial neurogastrointestinal encephalomyopathy and an average age of 32.4 years (range 11-59 years). We found 20 novel TYMP mutations. The average age-at-onset was 17.9 years (range 5 months to 35 years); however, the majority of patients reported the first symptoms before the age of 12 years. The patient distribution suggests a relatively high prevalence in Europeans, while the mutation distribution suggests founder effects for a few mutations, such as c.866A>G in Europe and c.518T>G in the Dominican Republic, that could guide genetic screening in each location. Although the sequence of clinical manifestations in the disease varied, half of the patients initially had gastrointestinal symptoms. We confirmed anecdotal reports of intra- and inter-familial clinical variability and absence of genotype-phenotype correlation in the disease, suggesting genetic modifiers, environmental factors or both contribute to disease manifestations. Acute medical events such as infections often provoked worsening of symptoms, suggesting that careful monitoring and early treatment of intercurrent illnesses may be beneficial. We observed endocrine/exocrine pancreatic insufficiency, which had not previously been reported. Kaplan-Meier analysis revealed significant mortality between the ages of 20 and 40 years due to infectious or metabolic complications. Despite increasing awareness of this illness, a high proportion of patients had been misdiagnosed. Early and accurate diagnosis of mitochondrial neurogastrointestinal encephalomyopathy, together with timely treatment of acute intercurrent illnesses, may retard disease progression and increase the number of patients eligible for allogeneic haematopoetic stem cell transplantation.


Subject(s)
Gastrointestinal Motility/genetics , Intestinal Pseudo-Obstruction/diagnosis , Intestinal Pseudo-Obstruction/genetics , Mitochondrial Encephalomyopathies/diagnosis , Mitochondrial Encephalomyopathies/genetics , Thymidine Phosphorylase/genetics , Adolescent , Adult , Age of Onset , Child , Child, Preschool , Diagnostic Errors , Disease Progression , Gastrointestinal Diseases/genetics , Gastrointestinal Diseases/physiopathology , Humans , Infant , Intestinal Pseudo-Obstruction/physiopathology , Middle Aged , Mitochondrial Encephalomyopathies/physiopathology , Muscle, Skeletal/physiopathology , Muscular Dystrophy, Oculopharyngeal , Mutation , Ophthalmoplegia/congenital
18.
Hum Mol Genet ; 18(4): 714-22, 2009 Feb 15.
Article in English | MEDLINE | ID: mdl-19028666

ABSTRACT

Replication and repair of DNA require equilibrated pools of deoxynucleoside triphosphate precursors. This concept has been proven by in vitro studies over many years, but in vivo models are required to demonstrate its relevance to multicellular organisms and to human diseases. Accordingly, we have generated thymidine phosphorylase (TP) and uridine phosphorylase (UP) double knockout (TP(-/-)UP(-/-)) mice, which show severe TP deficiency, increased thymidine and deoxyuridine in tissues and elevated mitochondrial deoxythymidine triphosphate. As consequences of the nucleotide pool imbalances, brains of mutant mice developed partial depletion of mtDNA, deficiencies of respiratory chain complexes and encephalopathy. These findings largely account for the pathogenesis of mitochondrial neurogastrointestinal encephalopathy (MNGIE), the first inherited human disorder of nucleoside metabolism associated with somatic DNA instability.


Subject(s)
DNA, Mitochondrial/chemistry , Deoxyribonucleotides/metabolism , Genomic Instability , Purine-Pyrimidine Metabolism, Inborn Errors/metabolism , Thymidine Phosphorylase/deficiency , Uridine Phosphorylase/metabolism , Animals , Brain/metabolism , DNA, Mitochondrial/genetics , DNA, Mitochondrial/metabolism , Electron Transport Complex I/genetics , Electron Transport Complex I/metabolism , Humans , Mice , Mice, Inbred C57BL , Mice, Knockout , Molecular Sequence Data , Purine-Pyrimidine Metabolism, Inborn Errors/genetics , Thymidine Phosphorylase/genetics , Uridine Phosphorylase/genetics
19.
FEBS Lett ; 581(18): 3410-4, 2007 Jul 24.
Article in English | MEDLINE | ID: mdl-17612528

ABSTRACT

Mitochondrial neurogastrointestinal encephalomyopathy (MNGIE) is an autosomal recessive disease due to ECGF1 gene mutations causing thymidine phosphorylase (TP) deficiency. Analysis of post-mortem samples of five MNGIE patients and two controls, revealed TP activity in all control tissues, but not in MNGIE samples. Converse to TP activity, thymidine and deoxyuridine were absent in control samples, but present in all tissues of MNGIE patients. Concentrations of both nucleosides in the tissues were generally higher than those observed in plasma of MNGIE patients. Our observations indicate that in the absence of TP activity, tissues accumulate nucleosides, which are excreted into plasma.


Subject(s)
Deoxyuridine/metabolism , Gastrointestinal Diseases/metabolism , Mitochondrial Encephalomyopathies/metabolism , Thymidine/metabolism , Adolescent , Adult , Autopsy , Biopsy , Deoxyuridine/analogs & derivatives , Female , Gastrointestinal Diseases/genetics , Gastrointestinal Diseases/pathology , Humans , Infant , Male , Mitochondrial Encephalomyopathies/genetics , Mitochondrial Encephalomyopathies/pathology , Oxygen/metabolism , Thymidine Phosphorylase/metabolism
20.
Ann Neurol ; 58(4): 649-52, 2005 Oct.
Article in English | MEDLINE | ID: mdl-16178026

ABSTRACT

Mitochondrial neurogastrointestinal encephalomyopathy (MNGIE) is caused by mutations in the gene encoding thymidine phosphorylase (TP). All MNGIE patients have had severe loss of TP function and prominent plasma accumulations of the TP substrates thymidine (dThd) and deoxyuridine (dUrd). Here, we report for the first time to our knowledge three MNGIE patients with later onset, milder phenotype, and less severe TP dysfunction, compared with typical MNGIE patients. This report demonstrates a direct relationship between the biochemical defects and clinical phenotypes in MNGIE and supports the notion that reduction of dThd and dUrd accumulation or TP replacement could be useful therapy for MNGIE.


Subject(s)
Mitochondrial Encephalomyopathies/enzymology , Thymidine Phosphorylase/metabolism , Arginine/metabolism , Chromatography, High Pressure Liquid/methods , Conserved Sequence , Female , Glycine/metabolism , Humans , Leucine/metabolism , Male , Middle Aged , Mitochondrial Encephalomyopathies/genetics , Polymorphism, Restriction Fragment Length , Thymidine Phosphorylase/genetics , Valine/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...