Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 31
Filter
1.
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
2.
Hum Mol Genet ; 24(3): 714-26, 2015 Feb 01.
Article in English | MEDLINE | ID: mdl-25274776

ABSTRACT

A member of the four-and-a-half-LIM (FHL) domain protein family, FHL1, is highly expressed in human adult skeletal and cardiac muscle. Mutations in FHL1 have been associated with diverse X-linked muscle diseases: scapuloperoneal (SP) myopathy, reducing body myopathy, X-linked myopathy with postural muscle atrophy, rigid spine syndrome (RSS) and Emery-Dreifuss muscular dystrophy. In 2008, we identified a missense mutation in the second LIM domain of FHL1 (c.365 G>C, p.W122S) in a family with SP myopathy. We generated a knock-in mouse model harboring the c.365 G>C Fhl1 mutation and investigated the effects of this mutation at three time points (3-5 months, 7-10 months and 18-20 months) in hemizygous male and heterozygous female mice. Survival was comparable in mutant and wild-type animals. We observed decreased forelimb strength and exercise capacity in adult hemizygous male mice starting from 7 to 10 months of age. Western blot analysis showed absence of Fhl1 in muscle at later stages. Thus, adult hemizygous male, but not heterozygous female, mice showed a slowly progressive phenotype similar to human patients with late-onset muscle weakness. In contrast to SP myopathy patients with the FHL1 W122S mutation, mutant mice did not manifest cytoplasmic inclusions (reducing bodies) in muscle. Because muscle weakness was evident prior to loss of Fhl1 protein and without reducing bodies, our findings indicate that loss of function is responsible for the myopathy in the Fhl1 W122S knock-in mice.


Subject(s)
Forelimb/pathology , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/metabolism , LIM Domain Proteins/genetics , LIM Domain Proteins/metabolism , Muscle Proteins/genetics , Muscle Proteins/metabolism , Muscle, Skeletal/pathology , Muscular Dystrophy, Emery-Dreifuss/pathology , Myocardium/pathology , Age of Onset , Animals , Disease Models, Animal , Female , Gene Knock-In Techniques , Hemizygote , Heterozygote , Humans , Male , Mice , Mice, Inbred C57BL , Muscular Dystrophy, Emery-Dreifuss/epidemiology , Muscular Dystrophy, Emery-Dreifuss/genetics , Muscular Dystrophy, Emery-Dreifuss/metabolism , Mutation, Missense
3.
Nature ; 478(7367): 127-31, 2011 Oct 05.
Article in English | MEDLINE | ID: mdl-21979053

ABSTRACT

Fukuyama muscular dystrophy (FCMD; MIM253800), one of the most common autosomal recessive disorders in Japan, was the first human disease found to result from ancestral insertion of a SINE-VNTR-Alu (SVA) retrotransposon into a causative gene. In FCMD, the SVA insertion occurs in the 3' untranslated region (UTR) of the fukutin gene. The pathogenic mechanism for FCMD is unknown, and no effective clinical treatments exist. Here we show that aberrant messenger RNA (mRNA) splicing, induced by SVA exon-trapping, underlies the molecular pathogenesis of FCMD. Quantitative mRNA analysis pinpointed a region that was missing from transcripts in patients with FCMD. This region spans part of the 3' end of the fukutin coding region, a proximal part of the 3' UTR and the SVA insertion. Correspondingly, fukutin mRNA transcripts in patients with FCMD and SVA knock-in model mice were shorter than the expected length. Sequence analysis revealed an abnormal splicing event, provoked by a strong acceptor site in SVA and a rare alternative donor site in fukutin exon 10. The resulting product truncates the fukutin carboxy (C) terminus and adds 129 amino acids encoded by the SVA. Introduction of antisense oligonucleotides (AONs) targeting the splice acceptor, the predicted exonic splicing enhancer and the intronic splicing enhancer prevented pathogenic exon-trapping by SVA in cells of patients with FCMD and model mice, rescuing normal fukutin mRNA expression and protein production. AON treatment also restored fukutin functions, including O-glycosylation of α-dystroglycan (α-DG) and laminin binding by α-DG. Moreover, we observe exon-trapping in other SVA insertions associated with disease (hypercholesterolemia, neutral lipid storage disease) and human-specific SVA insertion in a novel gene. Thus, although splicing into SVA is known, we have discovered in human disease a role for SVA-mediated exon-trapping and demonstrated the promise of splicing modulation therapy as the first radical clinical treatment for FCMD and other SVA-mediated diseases.


Subject(s)
Alternative Splicing/genetics , Exons/genetics , Retroelements/genetics , Walker-Warburg Syndrome/genetics , Walker-Warburg Syndrome/pathology , 3' Untranslated Regions/genetics , Alternative Splicing/drug effects , Animals , Disease Models, Animal , Dystroglycans/metabolism , Gene Knock-In Techniques , Glycosylation , Humans , Introns/genetics , Japan , Laminin/metabolism , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mice , Molecular Sequence Data , Mutagenesis, Insertional/drug effects , Mutagenesis, Insertional/genetics , Oligonucleotides, Antisense/genetics , Oligonucleotides, Antisense/pharmacology , Oligonucleotides, Antisense/therapeutic use , RNA Isoforms/genetics , RNA Splice Sites/genetics , Walker-Warburg Syndrome/therapy
4.
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
5.
Ann Neurol ; 74(6): 914-9, 2013 Dec.
Article in English | MEDLINE | ID: mdl-23798481

ABSTRACT

Glycogen storage diseases are important causes of myopathy and cardiomyopathy. We describe 10 patients from 8 families with childhood or juvenile onset of myopathy, 8 of whom also had rapidly progressive cardiomyopathy, requiring heart transplant in 4. The patients were homozygous or compound heterozygous for missense or truncating mutations in RBCK1, which encodes for a ubiquitin ligase, and had extensive polyglucosan accumulation in skeletal muscle and in the heart in cases of cardiomyopathy. We conclude that RBCK1 deficiency is a frequent cause of polyglucosan storage myopathy associated with progressive muscle weakness and cardiomyopathy.


Subject(s)
Glycogen Storage Disease/enzymology , Glycogen Storage Disease/genetics , Muscular Diseases/enzymology , Muscular Diseases/genetics , Nervous System Diseases/enzymology , Nervous System Diseases/genetics , Transcription Factors/deficiency , Ubiquitin/genetics , Adolescent , Adult , Cardiomyopathies/enzymology , Cardiomyopathies/etiology , Cardiomyopathies/genetics , Female , Genome, Human , Glycogen Storage Disease/complications , Humans , Male , Middle Aged , Muscle Weakness/enzymology , Muscle Weakness/etiology , Muscle Weakness/genetics , Muscular Diseases/etiology , Mutation, Missense/genetics , Nervous System Diseases/complications , Ubiquitin-Protein Ligases , Young Adult
6.
JCI Insight ; 9(8)2024 Mar 14.
Article in English | MEDLINE | ID: mdl-38483541

ABSTRACT

Glioblastoma (GBM) remains an incurable disease, requiring more effective therapies. Through interrogation of publicly available CRISPR and RNAi library screens, we identified the α-ketoglutarate dehydrogenase (OGDH) gene, which encodes an enzyme that is part of the tricarboxylic acid (TCA) cycle, as essential for GBM growth. Moreover, by combining transcriptome and metabolite screening analyses, we discovered that loss of function of OGDH by the clinically validated drug compound CPI-613 was synthetically lethal with Bcl-xL inhibition (genetically and through the clinically validated BH3 mimetic, ABT263) in patient-derived xenografts as well neurosphere GBM cultures. CPI-613-mediated energy deprivation drove an integrated stress response with an upregulation of the BH3-only domain protein, Noxa, in an ATF4-dependent manner, as demonstrated by genetic loss-of-function experiments. Consistently, silencing of Noxa attenuated cell death induced by CPI-613 in model systems of GBM. In patient-derived xenograft models of GBM in mice, the combination treatment of ABT263 and CPI-613 suppressed tumor growth and extended animal survival more potently than each compound on its own. Therefore, combined inhibition of Bcl-xL along with disruption of the TCA cycle might be a treatment strategy for GBM.


Subject(s)
Aniline Compounds , Caprylates , Glioblastoma , Ketoglutarate Dehydrogenase Complex , Sulfides , Sulfonamides , Synthetic Lethal Mutations , Xenograft Model Antitumor Assays , bcl-X Protein , Animals , Humans , Mice , Activating Transcription Factor 4/metabolism , Activating Transcription Factor 4/genetics , Aniline Compounds/pharmacology , bcl-X Protein/metabolism , bcl-X Protein/genetics , Brain Neoplasms/pathology , Brain Neoplasms/metabolism , Brain Neoplasms/genetics , Brain Neoplasms/drug therapy , Cell Line, Tumor , Citric Acid Cycle/drug effects , Glioblastoma/pathology , Glioblastoma/genetics , Glioblastoma/metabolism , Glioblastoma/drug therapy , Ketoglutarate Dehydrogenase Complex/metabolism , Ketoglutarate Dehydrogenase Complex/genetics , Ketoglutarate Dehydrogenase Complex/antagonists & inhibitors , Proto-Oncogene Proteins c-bcl-2/metabolism , Proto-Oncogene Proteins c-bcl-2/genetics , Sulfonamides/pharmacology
7.
Hum Mol Genet ; 20(1): 155-64, 2011 Jan 01.
Article in English | MEDLINE | ID: mdl-20940150

ABSTRACT

Deficiency of thymidine kinase 2 (TK2) is a frequent cause of isolated myopathy or encephalomyopathy in children with mitochondrial DNA (mtDNA) depletion. To determine the bases of disease onset, organ specificity and severity of TK2 deficiency, we have carefully characterized Tk2 H126N knockin mice (Tk2-/-). Although normal until postnatal day 8, Tk2-/- mice rapidly develop fatal encephalomyopathy between postnatal days 10 and 13. We have observed that wild-type Tk2 activity is constant in the second week of life, while Tk1 activity decreases significantly between postnatal days 8 and 13. The down-regulation of Tk1 activity unmasks Tk2 deficiency in Tk2-/- mice and correlates with the onset of mtDNA depletion in the brain and the heart. Resistance to pathology in Tk2 mutant organs depends on compensatory mechanisms to the reduced mtDNA level. Our analyses at postnatal day 13 have revealed that Tk2-/- heart significantly increases mitochondrial transcript levels relative to the mtDNA content. This transcriptional compensation allows the heart to maintain normal levels of mtDNA-encoded proteins. The up-regulation in mitochondrial transcripts is not due to increased expression of the master mitochondrial biogenesis regulators peroxisome proliferator-activated receptor-gamma coactivator 1 alpha and nuclear respiratory factors 1 and 2, or to enhanced expression of the mitochondrial transcription factors A, B1 or B2. Instead, Tk2-/- heart compensates for mtDNA depletion by down-regulating the expression of the mitochondrial transcriptional terminator transcription factor 3 (MTERF3). Understanding the molecular mechanisms that allow Tk2 mutant organs to be spared may help design therapies for Tk2 deficiency.


Subject(s)
Mitochondrial Encephalomyopathies/enzymology , Mitochondrial Proteins/genetics , Muscular Diseases/enzymology , Thymidine Kinase/deficiency , Thymidine Kinase/genetics , Transcription Factors/genetics , Age of Onset , Animals , Brain/enzymology , Brain/pathology , DNA, Mitochondrial/genetics , Disease Models, Animal , Down-Regulation/genetics , Gene Expression Regulation , Gene Knock-In Techniques , Heart , Mice , Mitochondrial Encephalomyopathies/genetics , Muscular Diseases/genetics , Organ Specificity/genetics , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha , Trans-Activators/genetics , Up-Regulation/genetics
8.
Ann Neurol ; 72(3): 433-41, 2012 Sep.
Article in English | MEDLINE | ID: mdl-23034915

ABSTRACT

OBJECTIVE: Adult polyglucosan body disease (APBD) is an autosomal recessive leukodystrophy characterized by neurogenic bladder, progressive spastic gait, and peripheral neuropathy. Polyglucosan bodies accumulate in the central and peripheral nervous systems and are often associated with glycogen branching enzyme (GBE) deficiency. To improve clinical diagnosis and enable future evaluation of therapeutic strategies, we conducted a multinational study of the natural history and imaging features of APBD. METHODS: We gathered clinical, biochemical, and molecular findings in 50 APBD patients with GBE deficiency from Israel, the United States, France, and the Netherlands. Brain and spine magnetic resonance images were reviewed in 44 patients. RESULTS: The most common clinical findings were neurogenic bladder (100%), spastic paraplegia with vibration loss (90%), and axonal neuropathy (90%). The median age was 51 years for the onset of neurogenic bladder symptoms, 63 years for wheelchair dependence, and 70 years for death. As the disease progressed, mild cognitive decline may have affected up to half of the patients. Neuroimaging showed hyperintense white matter abnormalities on T2 and fluid attenuated inversion recovery sequences predominantly in the periventricular regions, the posterior limb of the internal capsule, the external capsule, and the pyramidal tracts and medial lemniscus of the pons and medulla. Atrophy of the medulla and spine was universal. p.Y329S was the most common GBE1 mutation, present as a single heterozygous (28%) or homozygous (48%) mutation. INTERPRETATION: APBD with GBE deficiency, with occasional exceptions, is a clinically homogenous disorder that should be suspected in patients with adult onset leukodystrophy or spastic paraplegia with early onset of urinary symptoms and spinal atrophy.


Subject(s)
Glycogen Storage Disease , Magnetic Resonance Imaging , Nervous System Diseases , 1,4-alpha-Glucan Branching Enzyme/genetics , 1,4-alpha-Glucan Branching Enzyme/metabolism , Adult , Aged , Cerebral Cortex/pathology , Female , France , Glycogen Storage Disease/genetics , Glycogen Storage Disease/pathology , Glycogen Storage Disease/physiopathology , Humans , Israel , Kaplan-Meier Estimate , Male , Middle Aged , Mutation/genetics , Nervous System Diseases/genetics , Nervous System Diseases/pathology , Nervous System Diseases/physiopathology , Netherlands , Neurologic Examination , Spinal Cord/pathology , United States
9.
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
10.
Hum Mol Genet ; 17(16): 2433-40, 2008 Aug 15.
Article in English | MEDLINE | ID: mdl-18467430

ABSTRACT

Mitochondrial DNA (mtDNA) depletion syndrome (MDS), an autosomal recessive condition, is characterized by variable organ involvement with decreased mtDNA copy number and activities of respiratory chain enzymes in affected tissues. MtDNA depletion has been associated with mutations in nine autosomal genes, including thymidine kinase (TK2), which encodes a ubiquitous mitochondrial protein. To study the pathogenesis of TK2-deficiency, we generated mice harboring an H126N Tk2 mutation. Homozygous Tk2 mutant (Tk2(-/-)) mice developed rapidly progressive weakness after age 10 days and died between ages 2 and 3 weeks. Tk2(-/-) animals showed Tk2 deficiency, unbalanced dNTP pools, mtDNA depletion and defects of respiratory chain enzymes containing mtDNA-encoded subunits that were most prominent in the central nervous system. Histopathology revealed an encephalomyelopathy with prominent vacuolar changes in the anterior horn of the spinal cord. The H126N TK2 mouse is the first knock-in animal model of human MDS and demonstrates that the severity of TK2 deficiency in tissues may determine the organ-specific phenotype.


Subject(s)
Deoxyribonucleotides/metabolism , Mitochondria/enzymology , Mitochondria/genetics , Mitochondrial Diseases/enzymology , Mutation, Missense , Thymidine Kinase/deficiency , Animals , Deoxyribonucleotides/genetics , Electron Transport Complex I/genetics , Electron Transport Complex I/metabolism , Electron Transport Complex IV/genetics , Electron Transport Complex IV/metabolism , Female , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Mitochondria/metabolism , Mitochondrial Diseases/genetics , Mitochondrial Diseases/metabolism , Mitochondrial Diseases/physiopathology , Mutagenesis, Insertional , Organ Specificity , Thymidine Kinase/genetics
11.
Genomics ; 91(5): 458-66, 2008 May.
Article in English | MEDLINE | ID: mdl-18358695

ABSTRACT

Polysaccharide storage myopathy (PSSM) is a novel glycogenosis in horses characterized by abnormal glycogen accumulation in skeletal muscle and muscle damage with exertion. It is unlike glycogen storage diseases resulting from known defects in glycogenolysis, glycolysis, and glycogen synthesis that have been described in humans and domestic animals. A genome-wide association identified GYS1, encoding skeletal muscle glycogen synthase (GS), as a candidate gene for PSSM. DNA sequence analysis revealed a mutation resulting in an arginine-to-histidine substitution in a highly conserved region of GS. Functional analysis demonstrated an elevated GS activity in PSSM horses, and haplotype analysis and allele age estimation demonstrated that this mutation is identical by descent among horse breeds. This is the first report of a gain-of-function mutation in GYS1 resulting in a glycogenosis.


Subject(s)
Glycogen Storage Disease/veterinary , Glycogen Synthase/genetics , Glycogen Synthase/metabolism , Horse Diseases/enzymology , Horse Diseases/genetics , Muscle, Skeletal/enzymology , Amino Acid Sequence , Amino Acid Substitution , Animals , Gene Frequency , Genome , Glycogen Storage Disease/enzymology , Glycogen Storage Disease/genetics , Haplotypes , Horses , Microsatellite Repeats , Molecular Sequence Data , Muscle, Skeletal/metabolism , Mutation, Missense , Polymorphism, Single Nucleotide
12.
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
13.
Arch Neurol ; 65(1): 121-4, 2008 Jan.
Article in English | MEDLINE | ID: mdl-18195149

ABSTRACT

BACKGROUND: Alpers disease is commonly associated with polymerase gamma deficiency and usually affects infants or young children. OBJECTIVE: To report a juvenile case of Alpers disease due to mutations in the polymerase gamma gene (POLG1). DESIGN: Clinical, pathologic, biochemical, and molecular analysis. SETTING: Tertiary care university hospital and academic institutions. PATIENT: A 17-year-old adolescent girl with intractable epilepsy and liver disease. MAIN OUTCOME MEASURES: Clinical course and pathologic, biochemical, and molecular features. RESULTS: Biochemical and pathologic evidence suggested a respiratory chain defect, which was confirmed by enzyme analysis of the liver. Mutational analysis of POLG1 showed 2 novel mutations: T851A and R1047W. CONCLUSION: The POLG1 mutations can cause juvenile and childhood Alpers disease.


Subject(s)
DNA-Directed DNA Polymerase/genetics , Diffuse Cerebral Sclerosis of Schilder/genetics , Adolescent , Anticonvulsants/therapeutic use , Brain/pathology , DNA Mutational Analysis , DNA Polymerase gamma , Diffuse Cerebral Sclerosis of Schilder/pathology , Diffuse Cerebral Sclerosis of Schilder/therapy , Drug Resistance , Electroencephalography , Electron Transport/genetics , Electron Transport/physiology , Exons/genetics , Family , Female , Heterozygote , Humans , Liver Diseases/genetics , Liver Diseases/pathology , Magnetic Resonance Imaging , Mutation , Seizures/etiology , Seizures/pathology
14.
J Child Neurol ; 22(7): 858-62, 2007 Jul.
Article in English | MEDLINE | ID: mdl-17715279

ABSTRACT

A 14-year-old boy had exercise intolerance, weakness, ataxia, and lactic acidosis. Because his muscle biopsy showed a mosaic pattern of fibers staining intensely with the succinate dehydrogenase reaction but not at all with the cytochrome c oxidase reaction, we sequenced his mitochondrial DNA and found a novel mutation (C14680A) in the gene for tRNAGlu. The mutation was present in accessible tissues from the asymptomatic mother but not from a brother with Asperger syndrome. These data expand the clinical heterogeneity of mutations in this mitochondrial gene.


Subject(s)
Cytochrome-c Oxidase Deficiency/genetics , DNA, Mitochondrial/genetics , Mitochondrial Encephalomyopathies/genetics , Muscle, Skeletal/metabolism , RNA, Transfer, Glu/genetics , Adolescent , Electron Transport Complex IV/metabolism , Humans , Male , Mitochondrial Encephalomyopathies/metabolism , Mitochondrial Encephalomyopathies/pathology , Muscle, Skeletal/pathology , Mutation , Polymorphism, Single Nucleotide , RNA, Transfer, Glu/metabolism
16.
Neuromuscul Disord ; 14(4): 253-60, 2004 Apr.
Article in English | MEDLINE | ID: mdl-15019703

ABSTRACT

Glycogen storage disease type IV or Andersen disease is an autosomal recessive disorder due to deficiency of glycogen branching enzyme. Typically, glycogen storage disease type IV presents with rapidly progressive liver cirrhosis and death in childhood. Variants include a cardiopathic form of childhood, a relatively benign myopathic form of young adults, and a late-onset neurodegenerative disorder (adult polyglucosan body disease). A severe neuromuscular variant resembling Werdnig-Hoffmann disease has also been described in two patients. The objective was to describe two additional infants with the neuromuscular variant and novel mutations in the GBE1 gene. Branching enzyme assay, Western blot, RT-PCR and sequencing were performed in muscle biopsies from both patients. The cDNA of patient 1 was subcloned and sequenced to define the mutations. Muscle biopsies showed accumulation of periodic acid Schiff-positive, diastase-resistant storage material in both patients and increased lysosomal enzyme activity in patient 1. Branching enzyme activity in muscle was negligible in both patients, and Western blot showed decreased branching enzyme protein. Patient 1 had two single base pair deletions, one in exon 10 (1238delT) and the other in exon 12 (1467delC), and each parent was heterozygous for one of the deletions. Patient 2 had a large homozygous deletion that spanned 627 bp and included exons 8-12. Patient 1, who died at 41 days, had neurophysiological and neuropathological features of Spinal Muscular Atrophy. Patient 2, who died at 5(1/2) weeks, had a predominantly myopathic process. The infantile neuromuscular form of glycogen storage disease type IV is considered extremely rare, but our encountering two patients in close succession suggests that the disease may be underdiagnosed.


Subject(s)
Glycogen Storage Disease Type IV , Muscle, Skeletal/pathology , Neuromuscular Diseases , 1,4-alpha-Glucan Branching Enzyme/metabolism , Aminosalicylic Acid/metabolism , Biopsy/methods , Blotting, Western/methods , Brain Stem/enzymology , Brain Stem/pathology , DNA Mutational Analysis/methods , Exons , Female , Glycogen Storage Disease Type IV/complications , Glycogen Storage Disease Type IV/genetics , Glycogen Storage Disease Type IV/metabolism , Humans , Infant , Intramolecular Oxidoreductases , Lysosomes/enzymology , Microscopy, Electron/methods , Muscle, Skeletal/enzymology , Muscle, Skeletal/metabolism , Muscle, Skeletal/ultrastructure , Mutation , Neuromuscular Diseases/enzymology , Neuromuscular Diseases/pathology , Prostaglandin-E Synthases , RNA, Messenger/biosynthesis , Reverse Transcriptase Polymerase Chain Reaction/methods , Spinal Muscular Atrophies of Childhood/complications , Spinal Muscular Atrophies of Childhood/pathology , Trans-Activators/genetics
17.
JAMA Neurol ; 71(1): 41-7, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24248152

ABSTRACT

IMPORTANCE: The neuromuscular presentation of glycogen branching enzyme deficiency includes a severe infantile form and a late-onset variant known as adult polyglucosan body disease. Herein, we describe 2 patients with adult acute onset of fluctuating neurological signs and brain magnetic resonance imaging lesions simulating multiple sclerosis. A better definition of this new clinical entity is needed to facilitate diagnosis. OBJECTIVES: To describe the clinical presentation and progression of a new intermediate variant of glycogen branching enzyme deficiency and to discuss genotype-phenotype correlations. DESIGN, SETTING, AND PARTICIPANTS: Clinical, biochemical, morphological, and molecular study of 2 patients followed up for 6 years and 8 years at academic medical centers. The participants were 2 patients of non-Ashkenazi descent with adult acute onset of neurological signs initially diagnosed as multiple sclerosis. MAIN OUTCOMES AND MEASURES: Clinical course, muscle and nerve morphology, longitudinal study of brain magnetic resonance imaging, and glycogen branching enzyme activity and GBE1 molecular analysis. RESULTS: Molecular analysis showed that one patient was homozygous (c.1544G>A) and the other patient was compound heterozygous (c.1544G>A and c.1961-1962delCA) for GBE1 mutations. Residual glycogen branching enzyme activity was 16% and 30% of normal in leukocytes. Both patients manifested acute episodes of transient neurological symptoms, and neurological impairment was mild at age 45 years and 53 years. Brain magnetic resonance imaging revealed nonprogressive white matter lesions and spinocerebellar atrophy similar to typical adult polyglucosan body disease. CONCLUSIONS AND RELEVANCE: GBE1 mutations can cause an early adult-onset relapsing-remitting form of polyglucosan body disease distinct from adult polyglucosan body disease in several ways, including younger age at onset, history of infantile liver involvement, and subacute and remitting course simulating multiple sclerosis. This should orient neurologists toward the correct diagnosis.


Subject(s)
Glycogen Debranching Enzyme System/genetics , Glycogen Storage Disease Type IV/genetics , Glycogen Storage Disease Type IV/pathology , Acute Disease , Age of Onset , Disease Progression , Female , Follow-Up Studies , Genetic Carrier Screening , Glycogen Storage Disease Type IV/enzymology , Homozygote , Humans , Leukoencephalopathies/diagnosis , Leukoencephalopathies/genetics , Leukoencephalopathies/pathology , Magnetic Resonance Imaging , Male , Middle Aged , Spinocerebellar Ataxias/diagnosis , Spinocerebellar Ataxias/genetics , Spinocerebellar Ataxias/pathology
18.
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
19.
J Child Neurol ; 29(10): NP105-10, 2014 Oct.
Article in English | MEDLINE | ID: mdl-24284231

ABSTRACT

We report an unusual case of Leigh syndrome due to the m.10191T>C mutation in the complex I gene MT-ND3. This mutation has been associated with a spectrum of clinical phenotypes ranging from infant lethality to adult onset. Despite infantile onset and severe symptoms, our patient has survived to early adulthood because of a strict dietary regimen and parental care. This patient is an extreme example of the frequently prolonged course of Leigh syndrome due to this particular mutation.


Subject(s)
Electron Transport Complex I/genetics , Leigh Disease/genetics , Adult , Female , Humans , Leigh Disease/physiopathology , Leigh Disease/therapy , Mutation , Phenotype
20.
J Child Neurol ; 28(2): 264-8, 2013 Feb.
Article in English | MEDLINE | ID: mdl-22532554

ABSTRACT

Prenatal diagnosis of disorders due to mitochondrial DNA (mtDNA) tRNA gene mutations is problematic. Experience in families harboring the protein-coding ATPase 6 m.8993T>G mutation suggests that the mutant load is homogeneous in different tissues, thus allowing prenatal diagnosis. We have encountered a novel protein-coding gene mutation, m.10198C>T in MT-ND3. A baby girl homoplasmic for this mutation died at 3 months after severe psychomotor regression and respiratory arrest. The mother had no detectable mutation in accessible tissues. The product of a second pregnancy showed only wild-type mt genomes in amniocytes, chorionic villi, and biopsied fetal muscle. This second girl is now 18 months old and healthy. Our observations support the concept that the pathogenic mutation in this patient appeared de novo and that fetal muscle biopsy is a useful aide in prenatal diagnosis.


Subject(s)
Fetal Growth Retardation , Mitochondrial Proton-Translocating ATPases/genetics , Mutation/genetics , Prenatal Diagnosis , Biopsy , Cytochromes c/metabolism , DNA Mutational Analysis , DNA, Mitochondrial , Family Health , Female , Fetal Growth Retardation/diagnosis , Fetal Growth Retardation/genetics , Fetal Growth Retardation/physiopathology , Humans , Infant , Male , Muscle, Skeletal/metabolism , Muscle, Skeletal/pathology , Pregnancy
SELECTION OF CITATIONS
SEARCH DETAIL