Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 51
Filter
1.
Cell Death Dis ; 15(6): 405, 2024 Jun 10.
Article in English | MEDLINE | ID: mdl-38858390

ABSTRACT

Genetic mutations causing primary mitochondrial disease (i.e those compromising oxidative phosphorylation [OxPhos]) resulting in reduced bioenergetic output display great variability in their clinical features, but the reason for this is unknown. We hypothesized that disruption of the communication between endoplasmic reticulum (ER) and mitochondria at mitochondria-associated ER membranes (MAM) might play a role in this variability. To test this, we assayed MAM function and ER-mitochondrial communication in OxPhos-deficient cells, including cybrids from patients with selected pathogenic mtDNA mutations. Our results show that each of the various mutations studied indeed altered MAM functions, but notably, each disorder presented with a different MAM "signature". We also found that mitochondrial membrane potential is a key driver of ER-mitochondrial connectivity. Moreover, our findings demonstrate that disruption in ER-mitochondrial communication has consequences for cell survivability that go well beyond that of reduced ATP output. The findings of a "MAM-OxPhos" axis, the role of mitochondrial membrane potential in controlling this process, and the contribution of MAM dysfunction to cell death, reveal a new relationship between mitochondria and the rest of the cell, as well as providing new insights into the diagnosis and treatment of these devastating disorders.


Subject(s)
Endoplasmic Reticulum , Membrane Potential, Mitochondrial , Mitochondria , Mitochondrial Diseases , Oxidative Phosphorylation , Humans , Endoplasmic Reticulum/metabolism , Mitochondria/metabolism , Mitochondrial Diseases/metabolism , Mitochondrial Diseases/genetics , Mitochondrial Diseases/pathology , Mutation/genetics , DNA, Mitochondrial/metabolism , DNA, Mitochondrial/genetics
2.
Mol Genet Metab ; 138(3): 107525, 2023 03.
Article in English | MEDLINE | ID: mdl-36796138

ABSTRACT

Glycogen storage disease type IV (GSD IV) is an ultra-rare autosomal recessive disorder caused by pathogenic variants in GBE1 which results in reduced or deficient glycogen branching enzyme activity. Consequently, glycogen synthesis is impaired and leads to accumulation of poorly branched glycogen known as polyglucosan. GSD IV is characterized by a remarkable degree of phenotypic heterogeneity with presentations in utero, during infancy, early childhood, adolescence, or middle to late adulthood. The clinical continuum encompasses hepatic, cardiac, muscular, and neurologic manifestations that range in severity. The adult-onset form of GSD IV, referred to as adult polyglucosan body disease (APBD), is a neurodegenerative disease characterized by neurogenic bladder, spastic paraparesis, and peripheral neuropathy. There are currently no consensus guidelines for the diagnosis and management of these patients, resulting in high rates of misdiagnosis, delayed diagnosis, and lack of standardized clinical care. To address this, a group of experts from the United States developed a set of recommendations for the diagnosis and management of all clinical phenotypes of GSD IV, including APBD, to support clinicians and caregivers who provide long-term care for individuals with GSD IV. The educational resource includes practical steps to confirm a GSD IV diagnosis and best practices for medical management, including (a) imaging of the liver, heart, skeletal muscle, brain, and spine, (b) functional and neuromusculoskeletal assessments, (c) laboratory investigations, (d) liver and heart transplantation, and (e) long-term follow-up care. Remaining knowledge gaps are detailed to emphasize areas for improvement and future research.


Subject(s)
Glycogen Storage Disease Type IV , Glycogen Storage Disease , Neurodegenerative Diseases , Child, Preschool , Humans , Glycogen Storage Disease Type IV/diagnosis , Glycogen Storage Disease Type IV/genetics , Glycogen Storage Disease Type IV/therapy , Glycogen Storage Disease/diagnosis , Glycogen Storage Disease/genetics , Glycogen Storage Disease/therapy , Glycogen
3.
Life Sci Alliance ; 6(2)2023 02.
Article in English | MEDLINE | ID: mdl-36450448

ABSTRACT

Mitotic kinase Aurora A (AURKA) diverges from other kinases in its multiple active conformations that may explain its interphase roles and the limited efficacy of drugs targeting the kinase pocket. Regulation of AURKA activity by the cell is critically dependent on destruction mediated by the anaphase-promoting complex (APC/CFZR1) during mitotic exit and G1 phase and requires an atypical N-terminal degron in AURKA called the "A-box" in addition to a reported canonical D-box degron in the C-terminus. Here, we find that the reported C-terminal D-box of AURKA does not act as a degron and instead mediates essential structural features of the protein. In living cells, the N-terminal intrinsically disordered region of AURKA containing the A-box is sufficient to confer FZR1-dependent mitotic degradation. Both in silico and in cellulo assays predict the QRVL short linear interacting motif of the A-box to be a phospho-regulated D-box. We propose that degradation of full-length AURKA also depends on an intact C-terminal domain because of critical conformational parameters permissive for both activity and mitotic degradation of AURKA.


Subject(s)
Aurora Kinase A , Biological Assay , Humans , Aurora Kinase A/genetics , Cell Nucleus , Cdh1 Proteins
4.
Ann Clin Transl Neurol ; 7(11): 2186-2198, 2020 11.
Article in English | MEDLINE | ID: mdl-33034425

ABSTRACT

OBJECTIVE: Adult polyglucosan body disease (APBD) is an adult-onset neurological variant of glycogen storage disease type IV. APBD is caused by recessive mutations in the glycogen branching enzyme gene, and the consequent accumulation of poorly branched glycogen aggregates called polyglucosan bodies in the nervous system. There are presently no treatments for APBD. Here, we test whether downregulation of glycogen synthesis is therapeutic in a mouse model of the disease. METHODS: We characterized the effects of knocking out two pro-glycogenic proteins in an APBD mouse model. APBD mice were crossed with mice deficient in glycogen synthase (GYS1), or mice deficient in protein phosphatase 1 regulatory subunit 3C (PPP1R3C), a protein involved in the activation of GYS1. Phenotypic and histological parameters were analyzed and glycogen was quantified. RESULTS: APBD mice deficient in GYS1 or PPP1R3C demonstrated improvements in life span, morphology, and behavioral assays of neuromuscular function. Histological analysis revealed a reduction in polyglucosan body accumulation and of astro- and micro-gliosis in the brains of GYS1- and PPP1R3C-deficient APBD mice. Brain glycogen quantification confirmed the reduction in abnormal glycogen accumulation. Analysis of skeletal muscle, heart, and liver found that GYS1 deficiency reduced polyglucosan body accumulation in all three tissues and PPP1R3C knockout reduced skeletal muscle polyglucosan bodies. INTERPRETATION: GYS1 and PPP1R3C are effective therapeutic targets in the APBD mouse model. These findings represent a critical step toward the development of a treatment for APBD and potentially other glycogen storage disease type IV patients.


Subject(s)
Glycogen Storage Disease/metabolism , Glycogen Synthase/deficiency , Intracellular Signaling Peptides and Proteins/deficiency , Nervous System Diseases/metabolism , Animals , Behavior, Animal/physiology , Disease Models, Animal , Glycogen Storage Disease/physiopathology , Glycogen Storage Disease/therapy , Mice , Mice, Knockout , Nervous System Diseases/physiopathology , Nervous System Diseases/therapy
5.
Oncogene ; 39(37): 6009-6023, 2020 09.
Article in English | MEDLINE | ID: mdl-32772043

ABSTRACT

Aurora B kinase plays essential roles in mitosis. Its protein levels increase before the onset of mitosis and sharply decrease during mitosis exit. The latter decrease is due to a balance between the actions of the E3 ubiquitin ligase anaphase-promoting complex or cyclosome (activated by the Cdh1 adapter), and the deubiquitinating enzyme USP35. Aurora B also executes important functions in interphase. Abnormal modulation of Aurora B in interphase leads to cell cycle defects often linked to aberrant chromosomal condensation and segregation. Very little is however known about how Aurora B levels are regulated in interphase. Here we found that USP13-associates with and stabilizes Aurora B in cells, especially before their entry into mitosis. In order for USP13 to exert its stabilizing effect on Aurora B, their association is promoted by the Aurora B-mediated phosphorylation of USP13 at Serine 114. We also present evidence that USP13 instigates Aurora B deubiquitination and/or protect it from degradation in a non-catalytic manner. In addition, we report that genetic or chemical modulation of the cellular levels/activity of USP13 affects unperturbed cell-cycle progression. Overall our study unveils the molecular and cellular connections of the USP13-Aurora B axis, which potentially participates in the rewiring of the cell cycle happening in cancer cells.


Subject(s)
Aurora Kinase B/metabolism , Cell Cycle/genetics , Endopeptidases/genetics , Cell Cycle Checkpoints/genetics , Cell Line, Tumor , Disease Progression , Endopeptidases/metabolism , Enzyme Stability , Gene Expression , Gene Knockdown Techniques , Humans , Phosphorylation , Protein Binding , Serine/metabolism , Ubiquitin-Specific Proteases
6.
J Cell Sci ; 133(12)2020 06 16.
Article in English | MEDLINE | ID: mdl-32393600

ABSTRACT

Activity of AURKA is controlled through multiple mechanisms including phosphorylation, ubiquitin-mediated degradation and allosteric interaction with TPX2. Activity peaks at mitosis, before AURKA is degraded during and after mitotic exit in a process strictly dependent on the APC/C coactivator FZR1. We used FZR1 knockout cells (FZR1KO) and a novel FRET-based AURKA biosensor to investigate how AURKA activity is regulated in the absence of destruction. We found that AURKA activity in FZR1KO cells dropped at mitotic exit as rapidly as in parental cells, despite absence of AURKA destruction. Unexpectedly, TPX2 was degraded normally in FZR1KO cells. Overexpression of an N-terminal TPX2 fragment sufficient for AURKA binding, but that is not degraded at mitotic exit, caused delay in AURKA inactivation. We conclude that inactivation of AURKA at mitotic exit is determined not by AURKA degradation but by degradation of TPX2 and therefore is dependent on CDC20 rather than FZR1. The biosensor revealed that FZR1 instead suppresses AURKA activity in interphase and is critically required for assembly of the interphase mitochondrial network after mitosis.This article has an associated First Person interview with the first authors of the paper.


Subject(s)
Aurora Kinase A , Cell Cycle Proteins , Anaphase-Promoting Complex-Cyclosome , Aurora Kinase A/genetics , Cell Cycle Proteins/genetics , Interphase , Mitosis/genetics , Ubiquitin-Protein Ligase Complexes
7.
Niger J Clin Pract ; 23(4): 489-497, 2020 Apr.
Article in English | MEDLINE | ID: mdl-32246655

ABSTRACT

OBJECTIVE: This prospective study aimed to evaluate the clinical performance of different restorative materials in primary molars with class II carious lesions. MATERIALS AND METHODS: A total of 160 class II carious lesions (with radiographic involvement of the outer half of dentin) in 30 patients were randomly divided into four groups and restored with a glass ionomer restorative system (Equia™), two different bulk-fill composites (Sonicfill™ and X-tra fil™), and a nanohybrid composite (Filtek Z550™). The restorations were clinically and radiographically evaluated at the baseline, and 3, 6, and 12 months according to the modified United States Public Health Service criteria. Statistical analyses were performed using Pearson's Chi-square and McNemar tests. RESULTS: After 1 year, 134 restorations were evaluated in 26 patients. Equia was statistically less successful than the other restorative materials in marginal adaptation and retention criteria (P < 0.05). However, no material was found to be superior to the others over the study period in marginal discoloration, color matching, secondary caries, anatomical form, and postoperative sensitivity (P > 0.05). CONCLUSION: The bulk-fill and conventional composites exhibited good clinical performance, and Equia exhibited minor changes over the 1-year trial period.


Subject(s)
Acrylic Resins , Composite Resins/therapeutic use , Dental Restoration, Permanent/methods , Glass Ionomer Cements/therapeutic use , Silicon Dioxide , Adaptation, Physiological , Biometry , Child , Color , Dental Marginal Adaptation , Dental Materials , Dentin , Female , Follow-Up Studies , Humans , Male , Molar , Postoperative Period , Prospective Studies , Tooth, Deciduous , Treatment Outcome
8.
Hum Mol Genet ; 28(11): 1782-1800, 2019 06 01.
Article in English | MEDLINE | ID: mdl-30649465

ABSTRACT

Charcot-Marie-Tooth disease (CMT) type 2A is a form of peripheral neuropathy, due almost exclusively to dominant mutations in the nuclear gene encoding the mitochondrial protein mitofusin-2 (MFN2). However, there is no understanding of the relationship of clinical phenotype to genotype. MFN2 has two functions: it promotes inter-mitochondrial fusion and mediates endoplasmic reticulum (ER)-mitochondrial tethering at mitochondria-associated ER membranes (MAM). MAM regulates a number of key cellular functions, including lipid and calcium homeostasis, and mitochondrial behavior. To date, no studies have been performed to address whether mutations in MFN2 in CMT2A patient cells affect MAM function, which might provide insight into pathogenesis. Using fibroblasts from three CMT2AMFN2 patients with different mutations in MFN2, we found that some, but not all, examined aspects of ER-mitochondrial connectivity and of MAM function were indeed altered, and correlated with disease severity. Notably, however, respiratory chain function in those cells was unimpaired. Our results suggest that CMT2AMFN2 is a MAM-related disorder but is not a respiratory chain-deficiency disease. The alterations in MAM function described here could also provide insight into the pathogenesis of other forms of CMT.


Subject(s)
Charcot-Marie-Tooth Disease/genetics , Endoplasmic Reticulum/genetics , GTP Phosphohydrolases/genetics , Mitochondria/genetics , Mitochondrial Proteins/genetics , Adult , Charcot-Marie-Tooth Disease/metabolism , Charcot-Marie-Tooth Disease/pathology , Endoplasmic Reticulum/metabolism , Energy Metabolism/genetics , Female , Fibroblasts/metabolism , Genotype , Humans , Male , Middle Aged , Mitochondria/metabolism , Mitochondrial Dynamics/genetics , Mitochondrial Membranes/metabolism , Mutation , Oxidative Phosphorylation , Severity of Illness Index
9.
JCI Insight ; 3(17)2018 09 06.
Article in English | MEDLINE | ID: mdl-30185673

ABSTRACT

Adult polyglucosan body disease (APBD) is a late-onset disease caused by intracellular accumulation of polyglucosan bodies, formed due to glycogen-branching enzyme (GBE) deficiency. To find a treatment for APBD, we screened 1,700 FDA-approved compounds in fibroblasts derived from APBD-modeling GBE1-knockin mice. Capitalizing on fluorescent periodic acid-Schiff reagent, which interacts with polyglucosans in the cell, this screen discovered that the flavoring agent guaiacol can lower polyglucosans, a result also confirmed in APBD patient fibroblasts. Biochemical assays showed that guaiacol lowers basal and glucose 6-phosphate-stimulated glycogen synthase (GYS) activity. Guaiacol also increased inactivating GYS1 phosphorylation and phosphorylation of the master activator of catabolism, AMP-dependent protein kinase. Guaiacol treatment in the APBD mouse model rescued grip strength and shorter lifespan. These treatments had no adverse effects except making the mice slightly hyperglycemic, possibly due to the reduced liver glycogen levels. In addition, treatment corrected penile prolapse in aged GBE1-knockin mice. Guaiacol's curative effects can be explained by its reduction of polyglucosans in peripheral nerve, liver, and heart, despite a short half-life of up to 60 minutes in most tissues. Our results form the basis to use guaiacol as a treatment and prepare for the clinical trials in APBD.


Subject(s)
Glucans/metabolism , Glycogen Storage Disease/drug therapy , Guaiacol/pharmacology , Nervous System Diseases/drug therapy , Animals , Disease Models, Animal , Dual-Specificity Phosphatases/genetics , Fibroblasts , Glucose/metabolism , Glycogen/metabolism , Glycogen Synthase/drug effects , Glycogen Synthase/metabolism , Heart , Kinetics , Liver , Mice , Mice, Inbred C57BL , Mice, Knockout , Peripheral Nerves/metabolism , Phosphorylation , Protein Tyrosine Phosphatases, Non-Receptor , Ubiquitin-Protein Ligases/genetics
10.
Neurology ; 91(11): e1077-e1082, 2018 09 11.
Article in English | MEDLINE | ID: mdl-30111548

ABSTRACT

OBJECTIVE: To study the variable clinical picture and exercise tolerance of patients with phosphoglycerate kinase (PGK) 1 deficiency and how it relates to residual PGK enzyme activity. METHODS: In this case series study, we evaluated 7 boys and men from 5 families with PGK1 deficiency. Five had pure muscle symptoms, while 2 also had mild intellectual disability with or without anemia. Muscle glycolytic and oxidative capacities were evaluated by an ischemic forearm exercise test and by cycle ergometry. RESULTS: Enzyme levels of PGK were 4% to 9% of normal in red cells and 5% to10% in muscle in pure myopathy patients and 2.6% in both muscle and red cells in the 2 patients with multisystem involvement. Patients with pure myopathy had greater increases in lactate with ischemic exercise (2-3 mmol/L) vs the 2 multisystem-affected patients (<1 mmol/L). Myopathy patients had higher oxidative capacity in cycle exercise vs multisystem affected patients (≈30 vs ≈15 mL/kg per minute). One multisystem-affected patient developed frank myoglobinuria after the short exercise test. CONCLUSIONS: This case series study of PGK1 deficiency suggests that the level of impaired glycolysis in PGK deficiency is a major determinant of phenotype. Lower glycolytic capacity in PGK1 deficiency seems to result in multisystem involvement and increased susceptibility to exertional rhabdomyolysis.


Subject(s)
Exercise Tolerance/physiology , Genetic Diseases, X-Linked/enzymology , Genetic Diseases, X-Linked/physiopathology , Metabolism, Inborn Errors/enzymology , Metabolism, Inborn Errors/physiopathology , Phosphoglycerate Kinase/deficiency , Phosphoglycerate Kinase/metabolism , Ergometry , Exercise Test , Genetic Diseases, X-Linked/complications , Genetic Diseases, X-Linked/diagnosis , Humans , Intellectual Disability/blood , Intellectual Disability/complications , Intellectual Disability/enzymology , Intellectual Disability/physiopathology , Lactic Acid/blood , Male , Metabolism, Inborn Errors/complications , Metabolism, Inborn Errors/diagnosis , Muscle, Skeletal/metabolism , Muscular Diseases/blood , Muscular Diseases/complications , Muscular Diseases/enzymology , Muscular Diseases/physiopathology , Phenotype , Phosphoglycerate Kinase/blood
11.
J Inherit Metab Dis ; 41(5): 877-883, 2018 09.
Article in English | MEDLINE | ID: mdl-29110179

ABSTRACT

BACKGROUND: Adult polyglucosan body disease (APBD) is a progressive neurometabolic disorder caused by a deficiency of glycogen branching enzyme. We tested the efficacy of triheptanoin as a therapy for patients with APBD based on the hypothesis that decreased glycogen degradation leads to brain energy deficit. METHODS AND RESULTS: This was a two-site, randomized crossover trial of 23 patients (age 35-73 years; 63% men) who received triheptanoin or vegetable oil as placebo. The trial took place over 1 year and was followed by a 4-year open-label phase. Generalized linear mixed models were used to analyze this study. At baseline, using the 6-min walk test, patients could walk a mean of 389 ± 164 m (range 95-672; n = 19), highlighting the great clinical heterogeneity of our cohort. The overall mean difference between patients on triheptanoin versus placebo was 6 m; 95% confidence interval (CI) -11 to 22; p = 0.50. Motion capture gait analysis, gait quality, and stair climbing showed no consistent direction of change. All secondary endpoints were statistically nonsignificant after false discovery rate adjustment. Triheptanoin was safe and generally well tolerated. During the open-label phase of the study, the most affected patients at baseline kept deteriorating while mildly disabled patients remained notably stable up to 4 years. CONCLUSIONS: We cannot conclude that triheptanoin was effective in the treatment of APBD over a 6-month period, but we found it had a good safety profile. This study also emphasizes the difficulty of conducting trials in very rare diseases presenting with a wide clinical heterogeneity. ClinicalTrials.gov Identifier: NCT00947960.


Subject(s)
Glycogen Storage Disease/drug therapy , Nervous System Diseases/drug therapy , Triglycerides/therapeutic use , Walking , Adult , Aged , Cross-Over Studies , Disability Evaluation , Double-Blind Method , Female , Humans , Male , Middle Aged , Regression Analysis , Treatment Outcome , Walk Test
12.
Child Neurol Open ; 3: 2329048X15627937, 2016.
Article in English | MEDLINE | ID: mdl-28503604

ABSTRACT

Dystonia is often associated with the symmetrical basal ganglia lesions of Leigh syndrome. However, it has also been associated with mitochondrial ND mutations, with or without Leber hereditary optic neuropathy. The m.14459G>A mutation in ND6 causes dystonia with or without familial Leber hereditary optic neuropathy. We report heteroplasmic 14459G>A mutations in 2 unrelated children with nonmaternally inherited generalized dystonia and showing bilateral magnetic resonance imaging lesions in nucleus pallidus and putamen. Both children have reached their teenage years, and they are intellectually active, despite their motor problems.

13.
Neuromuscul Disord ; 26(1): 16-20, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26652229

ABSTRACT

Five Sardinian patients presented in their 5th or 6th decade with progressive limb girdle muscle weakness but their muscle biopsies showed vacuolar myopathy. The more or less abundant subsarcolemmal and intermyofibrillar vacuoles showed intense, partially α-amylase resistant, PAS-positive deposits consistent with polyglucosan. The recent description of late-onset polyglucosan myopathy has prompted us to find new genetic defects in the gene (GYG1) encoding glycogenin-1, the crucial primer enzyme of glycogen synthesis in muscle. We found a single homozygous intronic mutation harbored by five patients, who, except for two siblings, appear to be unrelated but all five live in central or south Sardinian villages.


Subject(s)
Glucans/genetics , Glucosyltransferases/genetics , Glycogen Storage Disease/genetics , Glycoproteins/genetics , Mutation/genetics , Nervous System Diseases/genetics , Adult , Aged , DNA Mutational Analysis , Female , Homozygote , Humans , Magnetic Resonance Imaging , Male , Middle Aged , Muscle, Skeletal/pathology , Muscle, Skeletal/ultrastructure
14.
Hum Mol Genet ; 24(23): 6801-10, 2015 Dec 01.
Article in English | MEDLINE | ID: mdl-26385640

ABSTRACT

Glycogen storage disease type IV (GSD IV) is a rare autosomal recessive disorder caused by deficiency of the glycogen-branching enzyme (GBE). The diagnostic hallmark of the disease is the accumulation of a poorly branched form of glycogen known as polyglucosan (PG). The disease is clinically heterogeneous, with variable tissue involvement and age at onset. Complete loss of enzyme activity is lethal in utero or in infancy and affects primarily the muscle and the liver. However, residual enzyme activity as low as 5-20% leads to juvenile or adult onset of a disorder that primarily affects the central and peripheral nervous system and muscles and in the latter is termed adult polyglucosan body disease (APBD). Here, we describe a mouse model of GSD IV that reflects this spectrum of disease. Homologous recombination was used to knock in the most common GBE1 mutation p.Y329S c.986A > C found in APBD patients of Ashkenazi Jewish decent. Mice homozygous for this allele (Gbe1(ys/ys)) exhibit a phenotype similar to APBD, with widespread accumulation of PG. Adult mice exhibit progressive neuromuscular dysfunction and die prematurely. While the onset of symptoms is limited to adult mice, PG accumulates in tissues of newborn mice but is initially absent from the cerebral cortex and heart muscle. Thus, PG is well tolerated in most tissues, but the eventual accumulation in neurons and their axons causes neuropathy that leads to hind limb spasticity and premature death. This mouse model mimics the pathology and pathophysiologic features of human adult-onset branching enzyme deficiency.


Subject(s)
Disease Models, Animal , Glycogen Debranching Enzyme System/genetics , Glycogen Storage Disease Type IV/metabolism , Mutation , Animals , Central Nervous System/metabolism , Central Nervous System/physiopathology , Gene Knock-In Techniques , Glycogen Storage Disease/genetics , Glycogen Storage Disease/metabolism , Glycogen Storage Disease/physiopathology , Glycogen Storage Disease Type IV/genetics , Glycogen Storage Disease Type IV/physiopathology , Mice , Muscle, Striated/metabolism , Muscle, Striated/physiopathology , Nervous System Diseases/genetics , Nervous System Diseases/metabolism , Nervous System Diseases/physiopathology , Peripheral Nervous System/metabolism , Peripheral Nervous System/physiopathology , Phenotype
15.
JAMA Neurol ; 72(4): 441-5, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25665141

ABSTRACT

IMPORTANCE: We describe a deep intronic mutation in adult polyglucosan body disease. Similar mechanisms can also explain manifesting heterozygous cases in other inborn metabolic diseases. OBJECTIVE: To explain the genetic change consistently associated with manifesting heterozygous patients with adult polyglucosan body disease. DESIGN, SETTING, AND PARTICIPANTS: This retrospective study took place from November 8, 2012, to November 7, 2014. We studied 35 typical patients with adult polyglucosan body disease, of whom 16 were heterozygous for the well-known c.986A>C mutation in the glycogen branching enzyme gene (GBE1) but harbored no other known mutation in 16 exons. MAIN OUTCOMES AND MEASURES: All 16 manifesting heterozygous patients had lower glycogen branching activity compared with homozygous patients, which showed inactivation of the apparently normal allele. We studied the messenger ribonucleic acid (mRNA) structure and the genetic change due to the elusive second mutation. RESULTS: When we reverse transcribed and sequenced the mRNA of GBE1, we found that all manifesting heterozygous patients had the c.986A>C mutant mRNA and complete lack of mRNA encoded by the second allele. We identified a deep intronic mutation in this allele, GBE1-IVS15+5289_5297delGTGTGGTGGinsTGTTTTTTACATGACAGGT, which acts as a gene trap, creating an ectopic last exon. The mRNA transcript from this allele missed the exon 16 and 3'UTR and encoded abnormal GBE causing further decrease of enzyme activity from 18% to 8%. CONCLUSIONS AND RELEVANCE: We identified the deep intronic mutation, which acts as a gene trap. This second-most common adult polyglucosan body disease mutation explains another founder effect in all Ashkenazi-Jewish cases.


Subject(s)
1,4-alpha-Glucan Branching Enzyme/genetics , Glycogen Debranching Enzyme System/genetics , Glycogen Storage Disease/genetics , Mutation/genetics , Nervous System Diseases/genetics , Adult , Alleles , Base Sequence , Heterozygote , Homozygote , Humans , Introns , Retrospective Studies
16.
Tumour Biol ; 36(5): 3511-20, 2015 May.
Article in English | MEDLINE | ID: mdl-25539763

ABSTRACT

MicroRNA (miRNA) deregulation is associated with various cancers. Among an expanding list of cancer-related miRNAs, deregulation of miR-125b has been well documented in many cancers including breast. Based on current knowledge, miR-125b is considered to be a tumor suppressor in breast cancers. While important messenger RNA (mRNA) targets have been defined for miR-125b, here, we aimed to further investigate direct/indirect consequences of miR-125b expression in breast cancer cells by using a transcriptome approach. Upon miR-125b expression, a total of 138 cancer-related genes were found to be differentially expressed in breast cancer cells. While only a few of these were predicted to be direct mRNA targets, majority of the gene expression changes were potentially downstream and indirect effects of miR-125b expression. Among these, activated leukocyte antigen molecule (ALCAM) mRNA and protein levels were found to be highly significantly increased upon miR-125b expression. Given the tumor suppressor role of miR-125b in our model system, upon silencing of ALCAM expression, cell proliferation rate re-increased in miR-125b-expressing cells. While ALCAM's possible context-dependent roles are not clear in breast cancer, a diverse expression pattern of ALCAM mRNA was detected in a panel of breast cancer patient samples. Differentially expressed/regulated cancer-related genes upon miR-125b expression along with the significant increase of ALCAM are of future interest to understand how deregulated expression of miR-125b may have a tumor suppressor role in breast and other cancers.


Subject(s)
Antigens, CD/biosynthesis , Breast Neoplasms/genetics , Cell Adhesion Molecules, Neuronal/biosynthesis , Fetal Proteins/biosynthesis , MicroRNAs/biosynthesis , Antigens, CD/genetics , Breast Neoplasms/pathology , Cell Adhesion Molecules, Neuronal/genetics , Female , Fetal Proteins/genetics , Gene Expression Regulation, Neoplastic/genetics , Humans , MCF-7 Cells , MicroRNAs/genetics , RNA, Messenger/biosynthesis
17.
Ann Neurol ; 76(6): 891-8, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25272951

ABSTRACT

We describe a slowly progressive myopathy in 7 unrelated adult patients with storage of polyglucosan in muscle fibers. Genetic investigation revealed homozygous or compound heterozygous deleterious variants in the glycogenin-1 gene (GYG1). Most patients showed depletion of glycogenin-1 in skeletal muscle, whereas 1 showed presence of glycogenin-1 lacking the C-terminal that normally binds glycogen synthase. Our results indicate that either depletion of glycogenin-1 or impaired interaction with glycogen synthase underlies this new form of glycogen storage disease that differs from a previously reported patient with GYG1 mutations who showed profound glycogen depletion in skeletal muscle and accumulation of glycogenin-1.


Subject(s)
Glucosyltransferases/deficiency , Glycogen Storage Disease/diagnosis , Glycogen Storage Disease/metabolism , Glycoproteins/deficiency , Muscle, Skeletal/metabolism , Adult , Aged , Female , Glucosyltransferases/genetics , Glycogen Storage Disease/genetics , Glycogen Synthase/metabolism , Glycoproteins/genetics , Humans , Male , Middle Aged
18.
Muscle Nerve ; 50(2): 292-5, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24711008

ABSTRACT

INTRODUCTION: A 61-year-old woman with a 5-year history of progressive muscle weakness and atrophy had a muscle biopsy characterized by a combination of dystrophic features (necrotic fibers and endomysial fibrosis) and mitochondrial alterations [ragged-red, cytochrome c oxidase (COX)-negative fibers]. METHODS: Sequencing of the whole mtDNA, assessment of the mutation load in muscle and accessible nonmuscle tissues, and single fiber polymerase chain reaction. RESULTS: Muscle mitochondrial DNA (mtDNA) sequencing revealed a novel heteroplasmic mutation (m.4403G>A) in the gene (MTTM) that encodes tRNA(Met). The mutation was not present in accessible nonmuscle tissues from the patient or 2 asymptomatic sisters. CONCLUSIONS: The clinical features and muscle morphology in this patient are very similar to those described in a previous patient with a different mutation, also in MTTM, which suggests that mutations in this gene confer a distinctive "dystrophic" quality. This may be a diagnostic clue in patients with isolated mitochondrial myopathy.


Subject(s)
Dystonia/genetics , Mitochondrial Myopathies/genetics , Mutation/genetics , RNA, Transfer/genetics , Dystonia/complications , Female , Humans , Middle Aged , Mitochondrial Myopathies/complications
19.
Am J Hum Genet ; 93(5): 906-14, 2013 Nov 07.
Article in English | MEDLINE | ID: mdl-24119684

ABSTRACT

We used exome sequencing to identify mutations in sideroflexin 4 (SFXN4) in two children with mitochondrial disease (the more severe case also presented with macrocytic anemia). SFXN4 is an uncharacterized mitochondrial protein that localizes to the mitochondrial inner membrane. sfxn4 knockdown in zebrafish recapitulated the mitochondrial respiratory defect observed in both individuals and the macrocytic anemia with megaloblastic features of the more severe case. In vitro and in vivo complementation studies with fibroblasts from the affected individuals and zebrafish demonstrated the requirement of SFXN4 for mitochondrial respiratory homeostasis and erythropoiesis. Our findings establish mutations in SFXN4 as a cause of mitochondriopathy and macrocytic anemia.


Subject(s)
Anemia, Macrocytic/genetics , Membrane Proteins/genetics , Mitochondrial Diseases/genetics , Adolescent , Animals , Child , Erythropoiesis/genetics , Exome , Female , Gene Knockdown Techniques , Humans , Mitochondrial Proteins/genetics , Mutation , Zebrafish/genetics
20.
J Child Neurol ; 28(2): 236-42, 2013 Feb.
Article in English | MEDLINE | ID: mdl-22638077

ABSTRACT

Mutations in the mitochondrial DNA cytochrome b gene (MTCYB) have been commonly associated with isolated mitochondrial myopathy and exercise intolerance, rarely with multisystem disorders, and only once with a parkinsonism/mitochondrial encephalomyopathy, lactic acidosis, and strokelike episodes (MELAS) overlap syndrome. Here, we describe a novel mutation (m.14864 T>C) in MTCYB in a 15-year-old girl with a clinical history of migraines, epilepsy, sensorimotor neuropathy, and strokelike episodes, a clinical picture reminiscent of MELAS.  The mutation, which changes a highly conserved cysteine to arginine at amino acid position 40 of cytochrome b, was heteroplasmic in muscle, blood, fibroblasts, and urinary sediment from the patient but absent in accessible tissues from her asymptomatic mother. This case demonstrates that MTCYB must be included in the already long list of mitochondrial DNA genes that have been associated with the MELAS phenotype.


Subject(s)
Acidosis, Lactic/complications , Cytochromes b/genetics , Mitochondrial Encephalomyopathies/genetics , Mutation/genetics , Stroke/complications , Child , Female , Humans , Magnetic Resonance Imaging , Mitochondrial Encephalomyopathies/complications , Mitochondrial Encephalomyopathies/diagnosis , Parietal Lobe/pathology
SELECTION OF CITATIONS
SEARCH DETAIL