Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 23
Filter
1.
Proc Natl Acad Sci U S A ; 119(45): e2212417119, 2022 Nov 08.
Article in English | MEDLINE | ID: mdl-36322731

ABSTRACT

Mitochondrial dysfunction can be associated with a range of clinical manifestations. Here, we report a family with a complex phenotype including combinations of connective tissue, neurological, and metabolic symptoms that were passed on to all surviving children. Analysis of the maternally inherited mtDNA revealed a novel genotype encompassing the haplogroup J - defining mitochondrial DNA (mtDNA) ND5 m.13708G>A (A458T) variant arising on the mtDNA haplogroup H7A background, an extremely rare combination. Analysis of transmitochondrial cybrids with the 13708A-H7 mtDNA revealed a lower mitochondrial respiration, increased reactive oxygen species production (mROS), and dysregulation of connective tissue gene expression. The mitochondrial dysfunction was exacerbated by histamine, explaining why all eight surviving children inherited the dysfunctional histidine decarboxylase allele (W327X) from the father. Thus, certain combinations of common mtDNA variants can cause mitochondrial dysfunction, mitochondrial dysfunction can affect extracellular matrix gene expression, and histamine-activated mROS production can augment the severity of mitochondrial dysfunction. Most important, we have identified a previously unreported genetic cause of mitochondrial disorder arising from the incompatibility of common, nonpathogenic mtDNA variants.


Subject(s)
DNA, Mitochondrial , Histamine , DNA, Mitochondrial/genetics , DNA, Mitochondrial/metabolism , Haplotypes , Histamine/metabolism , Mitochondria/genetics , Mitochondria/metabolism , Connective Tissue/metabolism
2.
Proc Natl Acad Sci U S A ; 118(6)2021 02 09.
Article in English | MEDLINE | ID: mdl-33536343

ABSTRACT

Autism spectrum disorders (ASDs) are characterized by a deficit in social communication, pathologic repetitive behaviors, restricted interests, and electroencephalogram (EEG) aberrations. While exhaustive analysis of nuclear DNA (nDNA) variation has revealed hundreds of copy number variants (CNVs) and loss-of-function (LOF) mutations, no unifying hypothesis as to the pathophysiology of ASD has yet emerged. Based on biochemical and physiological analyses, it has been hypothesized that ASD may be the result of a systemic mitochondrial deficiency with brain-specific manifestations. This proposal has been supported by recent mitochondrial DNA (mtDNA) analyses identifying both germline and somatic mtDNA variants in ASD. If mitochondrial defects do predispose to ASD, then mice with certain mtDNA mutations should present with autism endophenotypes. To test this prediction, we examined a mouse strain harboring an mtDNA ND6 gene missense mutation (P25L). This mouse manifests impaired social interactions, increased repetitive behaviors and anxiety, EEG alterations, and a decreased seizure threshold, in the absence of reduced hippocampal interneuron numbers. EEG aberrations were most pronounced in the cortex followed by the hippocampus. Aberrations in mitochondrial respiratory function and reactive oxygen species (ROS) levels were also most pronounced in the cortex followed by the hippocampus, but absent in the olfactory bulb. These data demonstrate that mild systemic mitochondrial defects can result in ASD without apparent neuroanatomical defects and that systemic mitochondrial mutations can cause tissue-specific brain defects accompanied by regional neurophysiological alterations.


Subject(s)
Autistic Disorder/genetics , Brain/metabolism , DNA, Mitochondrial/genetics , Mitochondria/genetics , Animals , Autistic Disorder/diagnostic imaging , Autistic Disorder/pathology , Brain/diagnostic imaging , Brain/pathology , DNA Copy Number Variations/genetics , Disease Models, Animal , Electroencephalography , Endophenotypes , Hippocampus/diagnostic imaging , Hippocampus/metabolism , Hippocampus/pathology , Humans , Mice , Mitochondria/pathology , Mutation/genetics , Reactive Oxygen Species/metabolism
3.
Hum Mutat ; 39(1): 69-79, 2018 01.
Article in English | MEDLINE | ID: mdl-29044765

ABSTRACT

Primary coenzyme Q10 (CoQ10 ; MIM# 607426) deficiencies are an emerging group of inherited mitochondrial disorders with heterogonous clinical phenotypes. Over a dozen genes are involved in the biosynthesis of CoQ10 , and mutations in several of these are associated with human disease. However, mutations in COQ5 (MIM# 616359), catalyzing the only C-methylation in the CoQ10 synthetic pathway, have not been implicated in human disease. Here, we report three female siblings of Iraqi-Jewish descent, who had varying degrees of cerebellar ataxia, encephalopathy, generalized tonic-clonic seizures, and cognitive disability. Whole-exome and subsequent whole-genome sequencing identified biallelic duplications in the COQ5 gene, leading to reduced levels of CoQ10 in peripheral white blood cells of all affected individuals and reduced CoQ10 levels in the only muscle tissue available from one affected proband. CoQ10 supplementation led to clinical improvement and increased the concentrations of CoQ10 in blood. This is the first report of primary CoQ10 deficiency caused by loss of function of COQ5, with delineation of the clinical, laboratory, histological, and molecular features, and insights regarding targeted treatment with CoQ10 supplementation.


Subject(s)
Biosynthetic Pathways/genetics , Cerebellar Ataxia/diagnosis , Cerebellar Ataxia/genetics , Methyltransferases/deficiency , Mitochondrial Encephalomyopathies/diagnosis , Mitochondrial Encephalomyopathies/genetics , Mitochondrial Proteins/deficiency , Ubiquinone/analogs & derivatives , Biopsy , Cerebellar Ataxia/diet therapy , Cerebellar Ataxia/metabolism , DNA Copy Number Variations , Dietary Supplements , Electron Transport , Female , Fibroblasts/metabolism , Genetic Association Studies , High-Throughput Nucleotide Sequencing , Humans , Leukocytes/metabolism , Methyltransferases/genetics , Mitochondrial Encephalomyopathies/diet therapy , Mitochondrial Encephalomyopathies/metabolism , Mitochondrial Proteins/genetics , Muscles/pathology , Oxygen Consumption , Pedigree , Polymorphism, Single Nucleotide , Siblings , Ubiquinone/biosynthesis
4.
Brain ; 140(3): 568-581, 2017 03 01.
Article in English | MEDLINE | ID: mdl-28364549

ABSTRACT

Cellular distribution and dynamics of mitochondria are regulated by several motor proteins and a microtubule network. In neurons, mitochondrial trafficking is crucial because of high energy needs and calcium ion buffering along axons to synapses during neurotransmission. The trafficking kinesin proteins (TRAKs) are well characterized for their role in lysosomal and mitochondrial trafficking in cells, especially neurons. Using whole exome sequencing, we identified homozygous truncating variants in TRAK1 (NM_001042646:c.287-2A > C), in six lethal encephalopathic patients from three unrelated families. The pathogenic variant results in aberrant splicing and significantly reduced gene expression at the RNA and protein levels. In comparison with normal cells, TRAK1-deficient fibroblasts showed irregular mitochondrial distribution, altered mitochondrial motility, reduced mitochondrial membrane potential, and diminished mitochondrial respiration. This study confirms the role of TRAK1 in mitochondrial dynamics and constitutes the first report of this gene in association with a severe neurodevelopmental disorder.


Subject(s)
Adaptor Proteins, Vesicular Transport/genetics , Adaptor Proteins, Vesicular Transport/metabolism , Brain Diseases/genetics , Brain Diseases/pathology , Mitochondria/metabolism , Mitochondrial Dynamics/genetics , Brain Diseases/diagnostic imaging , Brain Diseases/mortality , Cells, Cultured , Child, Preschool , Consanguinity , Family Health , Female , Fibroblasts/pathology , Fibroblasts/ultrastructure , Genetic Association Studies , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Humans , Infant , Magnetic Resonance Imaging , Male , Oxygen Consumption/genetics , Protein Transport/genetics , Transfection
5.
Eur J Neurosci ; 53(9): 2943-2945, 2021 05.
Article in English | MEDLINE | ID: mdl-33861478

Subject(s)
Brain
6.
Top Curr Chem ; 366: 97-137, 2015.
Article in English | MEDLINE | ID: mdl-23842869

ABSTRACT

UDP-N-acetylglucosamine 2-epimerase/N-acetylmannosamine kinase is the key enzyme of sialic acid biosynthesis in vertebrates. It catalyzes the first two steps of the cytosolic formation of CMP-N-acetylneuraminic acid from UDP-N-acetylglucosamine. In this review we give an overview of structure, biochemistry, and genetics of the bifunctional enzyme and its complex regulation. Furthermore, we will focus on diseases related to UDP-N-acetylglucosamine 2-epimerase/N-acetylmannosamine kinase.


Subject(s)
Cytidine Monophosphate N-Acetylneuraminic Acid/metabolism , Distal Myopathies/genetics , Genes, Regulator , Multienzyme Complexes/metabolism , Sialic Acid Storage Disease/genetics , Uridine Diphosphate N-Acetylglucosamine/metabolism , Animals , Disease Models, Animal , Distal Myopathies/enzymology , Distal Myopathies/pathology , Gene Expression Regulation , Humans , Mice , Mice, Transgenic , Multienzyme Complexes/chemistry , Multienzyme Complexes/genetics , Mutation , Protein Structure, Quaternary , Sialic Acid Storage Disease/enzymology , Sialic Acid Storage Disease/pathology
7.
J Inherit Metab Dis ; 37(2): 297-308, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24136589

ABSTRACT

BACKGROUND: UDP-GlcNAc 2-epimerase/ManNAc 6-kinase (GNE) is a bifunctional enzyme responsible for the first committed steps in the synthesis of sialic acid, a common terminal monosaccharide in both protein and lipid glycosylation. GNE mutations are responsible for a rare autosomal recessive neuromuscular disorder, GNE myopathy (also called hereditary inclusion body myopathy). The connection between the impairment of sialic acid synthesis and muscle pathology in GNE myopathy remains poorly understood. METHODS: Glycosphingolipid (GSL) analysis was performed by HPLC in multiple models of GNE myopathy, including patients' fibroblasts and plasma, control fibroblasts with inhibited GNE epimerase activity through a novel imino sugar, and tissues of Gne(M712T/M712T) knock-in mice. RESULTS: Not only neutral GSLs, but also sialylated GSLs, were significantly increased compared to controls in all tested models of GNE myopathy. Treatment of GNE myopathy fibroblasts with N-acetylmannosamine (ManNAc), a sialic acid precursor downstream of GNE epimerase activity, ameliorated the increased total GSL concentrations. CONCLUSION: GNE myopathy models have increased total GSL concentrations. ManNAc supplementation results in decrease of GSL levels, linking abnormal increase of total GSLs in GNE myopathy to defects in the sialic acid biosynthetic pathway. These data advocate for further exploring GSL concentrations as an informative biomarker, not only for GNE myopathy, but also for other disorders of sialic acid metabolism.


Subject(s)
Glycosphingolipids/metabolism , Multienzyme Complexes/metabolism , Muscular Diseases/metabolism , Animals , Case-Control Studies , Cells, Cultured , Female , Fibroblasts/metabolism , Glycosphingolipids/blood , Glycosphingolipids/genetics , Hexosamines/blood , Hexosamines/genetics , Hexosamines/metabolism , Humans , Mice , Mice, Inbred C57BL , Multienzyme Complexes/blood , Multienzyme Complexes/genetics , Muscles/metabolism , Muscular Diseases/blood , Muscular Diseases/genetics , Mutation , N-Acetylneuraminic Acid/blood , N-Acetylneuraminic Acid/genetics , N-Acetylneuraminic Acid/metabolism
8.
Leukemia ; 38(7): 1534-1540, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38714877

ABSTRACT

CD19 CAR-T cells have led to durable remissions in patients with refractory B-cell malignancies; nevertheless, most patients eventually relapse in the long term. Many interventions aimed at improving current products have been reported, with a subset of them focusing on a direct or indirect link to the metabolic state of the CAR-T cells. We assessed clinical products from an ongoing clinical trial utilizing CD19-28z CAR-T cells from patients with acute lymphoblastic leukemia. CAR-T clinical products leading to a complete response had significantly higher mitochondrial function (by oxygen consumption rate) irrespective of mitochondrial content. Next, we replaced the carbon source of the media from glucose to galactose to impact cellular metabolism. Galactose-containing media increased mitochondrial activity in CAR-T cells, and improved in in-vitro efficacy, without any consistent phenotypic change in memory profile. Finally, CAR-T cells produced in galactose-based glucose-free media resulted in increased mitochondrial activity. Using an in-vivo model of Nalm6 injected mice, galactose-primed CAR-T cells significantly improved leukemia-free survival compared to standard glucose-cultured CAR-T cells. Our results prove the significance of mitochondrial metabolism on CAR-T cell efficacy and suggest a translational pathway to improve clinical products.


Subject(s)
Galactose , Immunotherapy, Adoptive , Mitochondria , Galactose/metabolism , Animals , Mice , Mitochondria/metabolism , Humans , Immunotherapy, Adoptive/methods , Receptors, Chimeric Antigen/metabolism , Receptors, Chimeric Antigen/immunology , Precursor Cell Lymphoblastic Leukemia-Lymphoma/metabolism , Precursor Cell Lymphoblastic Leukemia-Lymphoma/pathology , Antigens, CD19/metabolism , T-Lymphocytes/metabolism , T-Lymphocytes/immunology , Xenograft Model Antitumor Assays
9.
Am J Pathol ; 180(4): 1431-40, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22322304

ABSTRACT

Pathological glomerular hyposialylation has been implicated in certain unexplained glomerulopathies, including minimal change nephrosis, membranous glomerulonephritis, and IgA nephropathy. We studied our previously established mouse model carrying a homozygous mutation in the key enzyme of sialic acid biosynthesis, N-acetylglucosamine 2-epimerase/N-acetylmannosamine kinase. Mutant mice died before postnatal day 3 (P3) from severe glomerulopathy with podocyte effacement and segmental glomerular basement membrane splitting due to hyposialylation. Administration of the sialic acid precursor N-acetylmannosamine (ManNAc) led to improved sialylation and survival of mutant pups beyond P3. We determined the onset of the glomerulopathy in the embryonic stage. A lectin panel, distinguishing normally sialylated from hyposialylated glycans, used WGA, SNA, PNA, Jacalin, HPA, and VVA, indicating glomerular hyposialylation of predominantly O-linked glycoproteins in mutant mice. The glomerular glycoproteins nephrin and podocalyxin were hyposialylated in this unique murine model. ManNAc treatment appeared to ameliorate the hyposialylation status of mutant mice, indicated by a lectin histochemistry pattern similar to that of wild-type mice, with improved sialylation of both nephrin and podocalyxin, as well as reduced albuminuria compared with untreated mutant mice. These findings suggest application of our lectin panel for categorizing human kidney specimens based on glomerular sialylation status. Moreover, the partial restoration of glomerular architecture in ManNAc-treated mice highlights ManNAc as a potential treatment for humans affected with disorders of glomerular hyposialylation.


Subject(s)
Disease Models, Animal , Kidney Diseases/genetics , Animals , Biomarkers/metabolism , Carbohydrate Epimerases/genetics , Carrier Proteins/genetics , Dietary Supplements , Drug Evaluation, Preclinical/methods , Hexosamines/therapeutic use , Humans , Kidney Diseases/drug therapy , Kidney Diseases/metabolism , Kidney Diseases/pathology , Kidney Glomerulus/embryology , Kidney Glomerulus/metabolism , Kidney Glomerulus/ultrastructure , Membrane Proteins/metabolism , Mice , Mice, Mutant Strains , Microscopy, Electron , Mutation , N-Acetylneuraminic Acid/physiology , Podocytes/metabolism , Podocytes/ultrastructure , Real-Time Polymerase Chain Reaction/methods , Sialoglycoproteins/metabolism
10.
Glycoconj J ; 30(6): 609-18, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23266873

ABSTRACT

The bifunctional enzyme UDP-GlcNAc 2-epimerase/ManNAc kinase (GNE) catalyzes the first two committed steps in sialic acid synthesis. Non-allosteric GNE gene mutations cause the muscular disorder GNE myopathy (also known as hereditary inclusion body myopathy), whose exact pathology remains unknown. Increased knowledge of GNE regulation, including isoform regulation, may help elucidate the pathology of GNE myopathy. While eight mRNA transcripts encoding human GNE isoforms are described, we only identified two mouse Gne mRNA transcripts, encoding mGne1 and mGne2, homologous to human hGNE1 and hGNE2. Orthologs of the other human isoforms were not identified in mice. mGne1 appeared as the ubiquitously expressed, major mouse isoform. The mGne2 encoding transcript is differentially expressed and may act as a tissue-specific regulator of sialylation. mGne2 expression appeared significantly increased the first 2 days of life, possibly reflecting the high sialic acid demand during this period. Tissues of the knock-in Gne p.M712T mouse model had similar mGne transcript expression levels among genotypes, indicating no effect of the mutation on mRNA expression. However, upon treatment of these mice with N-acetylmannosamine (ManNAc, a Gne substrate, sialic acid precursor, and proposed therapy for GNE myopathy), Gne transcript expression, in particular mGne2, increased significantly, likely resulting in increased Gne enzymatic activities. This dual effect of ManNAc supplementation (increased flux through the sialic acid pathway and increased Gne activity) needs to be considered when treating GNE myopathy patients with ManNAc. In addition, the existence and expression of GNE isoforms needs consideration when designing other therapeutic strategies for GNE myopathy.


Subject(s)
Hexosamines/therapeutic use , Multienzyme Complexes/metabolism , Transcription, Genetic , Amino Acid Sequence , Animals , Distal Myopathies/drug therapy , Distal Myopathies/genetics , Isoenzymes/chemistry , Isoenzymes/genetics , Isoenzymes/metabolism , Mice , Mice, Inbred C57BL , Molecular Sequence Data , Multienzyme Complexes/chemistry , Multienzyme Complexes/genetics , Mutation, Missense , Organ Specificity , Protein Structure, Secondary
11.
bioRxiv ; 2023 Sep 25.
Article in English | MEDLINE | ID: mdl-37808778

ABSTRACT

CD19 CAR-T cells have led to durable remissions in patients with refractory B-cell malignancies; nevertheless, most patients eventually relapse in the long term. Many interventions aimed at improving current products have been reported, with a subset of them focusing on a direct or indirect link to the metabolic state of the CAR-T cells. We assessed clinical products from an ongoing clinical trial utilizing CD19-28z CAR-T cells from patients with acute lymphoblastic leukemia. CAR-T clinical products leading to a complete response had significantly higher mitochondrial function (by oxygen consumption rate) irrespective of mitochondrial content. Next, we replaced the carbon source of the media from glucose to galactose to impact cellular metabolism. Galactose-containing media increased mitochondrial activity in CAR-T cells, and improved in vitro efficacy, without any consistent phenotypic change in memory profile. Finally, CAR-T cells produced in galactose-based glucose-free media resulted in increased mitochondrial activity. Using an in vivo model of Nalm6 injected mice, galactose-primed CAR-T cells significantly improved leukemia-free survival compared to standard glucose-cultured CAR-T cells. Our results prove the significance of mitochondrial metabolism on CAR-T cell efficacy and suggest a translational pathway to improve clinical products.

12.
Mol Genet Metab ; 107(4): 748-55, 2012 Dec.
Article in English | MEDLINE | ID: mdl-23122659

ABSTRACT

GNE myopathy, previously termed hereditary inclusion body myopathy (HIBM), is an adult-onset neuromuscular disorder characterized by progressive muscle weakness. The disorder results from biallelic mutations in GNE, encoding UDP-N-acetylglucosamine 2-epimerase/N-acetylmannosamine kinase, the key enzyme of sialic acid synthesis. GNE myopathy, associated with impaired glycan sialylation, has no approved therapy. Here we test potential sialylation-increasing monosaccharides for their effectiveness in prophylaxis (at the embryonic and neonatal stages) and therapy (after the onset of symptoms) by evaluating renal and muscle hyposialylation in a knock-in mouse model (Gne p.M712T) of GNE myopathy. We demonstrate that oral mannosamine (ManN), but not sialic acid (Neu5Ac), mannose (Man), galactose (Gal), or glucosamine (GlcN), administered to pregnant female mice has a similar prophylactic effect on renal hyposialylation, pathology and neonatal survival of mutant offspring, as previously shown for N-acetylmannosamine (ManNAc) therapy. ManN may be converted to ManNAc by a direct, yet unknown, pathway, or may act through another mode of action. The other sugars (Man, Gal, GlcN) may either not cross the placental barrier (Neu5Ac) and/or may not be able to directly increase sialylation. Because GNE myopathy patients will likely require treatment in adulthood after onset of symptoms, we also administered ManNAc (1 or 2g/kg/day for 12 weeks), Neu5Ac (2 g/kg/day for 12 weeks), or ManN (2 g/kg/day for 6 weeks) in drinking water to 6 month old mutant Gne p.M712T mice. All three therapies markedly improved the muscle and renal hyposialylation, as evidenced by lectin histochemistry for overall sialylation status and immunoblotting of specific sialoproteins. These preclinical data strongly support further evaluation of oral ManNAc, Neu5Ac and ManN as therapy for GNE myopathy and conceivably for certain glomerular diseases with hyposialylation.


Subject(s)
Kidney/metabolism , Monosaccharides/administration & dosage , Muscles/metabolism , Myositis, Inclusion Body/congenital , Administration, Oral , Animals , Female , Humans , Kidney/pathology , Kidney/ultrastructure , Mice , Mice, Transgenic , Multienzyme Complexes/genetics , Muscles/pathology , Myositis, Inclusion Body/drug therapy , Myositis, Inclusion Body/genetics , Myositis, Inclusion Body/metabolism , N-Acetylneuraminic Acid/biosynthesis
13.
Mol Metab ; 64: 101560, 2022 10.
Article in English | MEDLINE | ID: mdl-35940554

ABSTRACT

OBJECTIVE: Mitochondrial disorders are often characterized by muscle weakness and fatigue. Null mutations in the heart-muscle adenine nucleotide translocator isoform 1 (ANT1) of both humans and mice cause cardiomyopathy and myopathy associated with exercise intolerance and muscle weakness. Here we decipher the molecular underpinnings of ANT1-deficiency-mediated exercise intolerance. METHODS: This was achieved by correlating exercise physiology, mitochondrial function and metabolomics of mice deficient in ANT1 and comparing this to control mice. RESULTS: We demonstrate a peripheral limitation of skeletal muscle mitochondrial respiration and a reduced complex I respiration in ANT1-deficient mice. Upon exercise, this results in a lack of NAD+ leading to a substrate limitation and stalling of the TCA cycle and mitochondrial respiration, further limiting skeletal muscle mitochondrial respiration. Treatment of ANT1-deficient mice with nicotinamide riboside increased NAD+ levels in skeletal muscle and liver, which increased the exercise capacity and the mitochondrial respiration. CONCLUSION: Increasing NAD+ levels with nicotinamide riboside can alleviate the exercise intolerance associated to ANT1-deficiency, indicating the therapeutic potential of NAD+-stimulating compounds in mitochondrial myopathies.


Subject(s)
Adenine Nucleotide Translocator 1 , Mitochondrial Myopathies , NAD , Niacinamide , Physical Conditioning, Animal , Pyridinium Compounds , Adenine Nucleotide Translocator 1/genetics , Animals , Mice , Mitochondrial Myopathies/genetics , Muscle Weakness , Niacinamide/analogs & derivatives , Niacinamide/pharmacology , Protein Isoforms , Pyridinium Compounds/pharmacology
14.
Biochemistry ; 50(41): 8914-25, 2011 Oct 18.
Article in English | MEDLINE | ID: mdl-21910480

ABSTRACT

UDP-GlcNAc 2-epimerase/ManNAc kinase (GNE) catalyzes the first two committed steps in sialic acid synthesis. In addition to the three previously described human GNE isoforms (hGNE1-hGNE3), our database and polymerase chain reaction analysis yielded five additional human isoforms (hGNE4-hGNE8). hGNE1 is the ubiquitously expressed major isoform, while the hGNE2-hGNE8 isoforms are differentially expressed and may act as tissue-specific regulators of sialylation. hGNE2 and hGNE7 display a 31-residue N-terminal extension compared to hGNE1. On the basis of similarities to kinases and helicases, this extension does not seem to hinder the epimerase enzymatic active site. hGNE3 and hGNE8 contain a 55-residue N-terminal deletion and a 50-residue N-terminal extension compared to hGNE1. The size and secondary structures of these fragments are similar, and modeling predicted that these modifications do not affect the overall fold compared to that of hGNE1. However, the epimerase enzymatic activity of GNE3 and GNE8 is likely absent, because the deleted fragment contains important substrate binding residues in homologous bacterial epimerases. hGNE5-hGNE8 have a 53-residue deletion, which was assigned a role in substrate (UDP-GlcNAc) binding. Deletion of this fragment likely eliminates epimerase enzymatic activity. Our findings imply that GNE is subject to evolutionary mechanisms to improve cellular functions, without increasing the number of genes. Our expression and modeling data contribute to elucidation of the complex functional and regulatory mechanisms of human GNE and may contribute to further elucidating the pathology and treatment strategies of the human GNE-opathies sialuria and hereditary inclusion body myopathy.


Subject(s)
N-Acetylneuraminic Acid/chemistry , Amino Acid Sequence , Carbohydrate Epimerases/chemistry , Catalysis , Catalytic Domain , DNA, Complementary/metabolism , Gene Deletion , Humans , Models, Molecular , Molecular Sequence Data , Polymerase Chain Reaction , Protein Isoforms , Sequence Homology, Amino Acid , Tissue Distribution
15.
Glycobiology ; 20(3): 322-37, 2010 Mar.
Article in English | MEDLINE | ID: mdl-19917666

ABSTRACT

The bifunctional enzyme UDP-GlcNAc 2-epimerase/ ManNAc kinase (GNE/MNK), encoded by the GNE gene, catalyzes the first two committed, rate-limiting steps in the biosynthesis of N-acetylneuraminic acid (sialic acid). GNE/MNK is feedback inhibited by binding of the downstream product, CMP-sialic acid in its allosteric site. GNE mutations can result in two human disorders, hereditary inclusion body myopathy (HIBM) or sialuria. So far, no active site geometry predictions or conformational transitions involved with function are available for mammalian GNE/MNK. The N-terminal GNE domain is homologous to various prokaryotic 2-epimerases, some of which have solved crystallographic structures. The C-terminal MNK domain belongs to the sugar kinases superfamily; its crystallographic structure is solved at 2.84 A and three-dimensional structures have also been reported for several other kinases. In this work, we employed available structural data of GNE/MNK homologs to model the active sites of human GNE/MNK and identify critical amino acid residues responsible for interactions with substrates. In addition, we modeled effects of GNE/MNK missense mutations associated with HIBM or sialuria on helix arrangement, substrate binding, and enzyme action. We found that all reported mutations are associated with the active sites or secondary structure interfaces of GNE/MNK. The Persian-Jewish HIBM founder mutation p.M712T is located at the interface alpha4alpha10 and likely affects GlcNAc, Mg2+, and ATP binding. This work contributes to further understanding of GNE/MNK function and ligand binding, which may assist future studies for therapeutic options that target misfolded GNE/MNK in HIBM and/or sialuria.


Subject(s)
Models, Molecular , Multienzyme Complexes/chemistry , Multienzyme Complexes/genetics , Muscular Diseases/genetics , Mutation , Sialic Acid Storage Disease/genetics , Amino Acid Sequence , Humans , Molecular Sequence Data , Multienzyme Complexes/metabolism , Muscular Diseases/enzymology , Muscular Diseases/metabolism , N-Acetylneuraminic Acid/metabolism , Protein Conformation , Sialic Acid Storage Disease/enzymology
16.
J Hum Genet ; 55(5): 308-13, 2010 May.
Article in English | MEDLINE | ID: mdl-20414253

ABSTRACT

Progressive familial intrahepatic cholestasis (PFIC) is a group of rare heterogeneous autosomal recessive disorders characterized by metabolic defects in biliary proteins involved in the formation and transfer of bile acids in the liver. The genotype-phenotype correlation is not always clear. Mutations in the ATP8B1, BSEP and MDR3 genes have been associated with PFIC1, PFIC2 and PFIC3, respectively. This study sought to characterize the molecular genetic basis for PFIC subtypes in Israel. It was conducted on 14 children with PFIC and their families; 10 with a PFIC1 or PFIC2 phenotype and 4 with a PFIC3 phenotype. Using denaturing high-performance liquid chromatography (DHPLC), five different mutations were identified in four affected families: three novel mutations in BSEP (G19R-g181c, S226L-c803t and G877R-g2755a), one novel mutation in MDR3 (IVS14+6 t/c) and one heterozygous mutation in ATP8B1 (R600W, in a family with the PFIC1/PFIC2 phenotype). The cause of PFIC was identified in 20% of the families tested. These findings indicate the probable involvement of additional genes in PFIC and the need for further studies to determine whether the abnormality lies on the RNA or protein level. A better understanding of the phenotype-genotype correlation in PFIC will lead to improved diagnoses and treatments.


Subject(s)
Cholestasis, Intrahepatic/genetics , Chromatography, High Pressure Liquid/methods , Genetic Association Studies/methods , Mutation , ATP Binding Cassette Transporter, Subfamily B/genetics , ATP Binding Cassette Transporter, Subfamily B, Member 11 , ATP-Binding Cassette Transporters/genetics , Adenosine Triphosphatases/genetics , Base Sequence , Child, Preschool , Cholestasis, Intrahepatic/classification , Cholestasis, Intrahepatic/diagnosis , DNA Mutational Analysis , Family Health , Female , Genetic Testing , Genotype , Humans , Infant , Infant, Newborn , Israel , Male , Pedigree , Retrospective Studies
17.
Sci Signal ; 12(588)2019 07 02.
Article in English | MEDLINE | ID: mdl-31266851

ABSTRACT

Changes in the gut microbiota and the mitochondrial genome are both linked with the development of disease. To investigate why, we examined the gut microbiota of mice harboring various mutations in genes that alter mitochondrial function. These studies revealed that mitochondrial genetic variations altered the composition of the gut microbiota community. In cross-fostering studies, we found that although the initial microbiota community of newborn mice was that obtained from the nursing mother, the microbiota community progressed toward that characteristic of the microbiome of unfostered pups of the same genotype within 2 months. Analysis of the mitochondrial DNA variants associated with altered gut microbiota suggested that microbiome species diversity correlated with host reactive oxygen species (ROS) production. To determine whether the abundance of ROS could alter the gut microbiota, mice were aged, treated with N-acetylcysteine, or engineered to express the ROS scavenger catalase specifically within the mitochondria. All three conditions altered the microbiota from that initially established. Thus, these data suggest that the mitochondrial genotype modulates both ROS production and the species diversity of the gut microbiome, implying that the connection between the gut microbiome and common disease phenotypes might be due to underlying changes in mitochondrial function.


Subject(s)
DNA, Mitochondrial/genetics , Gastrointestinal Microbiome/genetics , Genetic Variation , Mitochondria/genetics , Age Factors , Animals , Bacteria/classification , Bacteria/genetics , Catalase/genetics , Catalase/metabolism , Genotype , Host Microbial Interactions/genetics , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Inbred NZB , Mitochondria/metabolism , NADH Dehydrogenase/genetics , NADH Dehydrogenase/metabolism , Phenotype , Reactive Oxygen Species/metabolism
18.
Kidney Int Rep ; 4(10): 1454-1462, 2019 Oct.
Article in English | MEDLINE | ID: mdl-31701055

ABSTRACT

INTRODUCTION: Sialic acids are important contributors to the polyanionic component of the glomerular filtration barrier, which regulates permeability selectivity. Pathologic glomerular hyposialylation, associated with podocyte effacement, has been implicated in human and mouse glomerulopathies. Oral treatment with N-acetylmannosamine (ManNAc), the uncharged precursor of sialic acid, ameliorates glomerular pathology in different models of glomerular disease. METHODS: Here we explore the sialylation status of kidney biopsies obtained from 27 subjects with various glomerular diseases using lectin histochemistry. RESULTS: We identified severe glomerular hyposialylation in 26% of the biopsies. These preliminary findings suggest that this condition may occur relatively frequently and may be a novel target for therapy. We describe the background, rationale, and design of a phase 1 study to test safety, tolerability, and pharmacokinetics of ManNAc in subjects with primary podocyte diseases. CONCLUSION: We recently demonstrated that ManNAc was safe and well tolerated in a first-in-human phase 1 study in subjects with UDP-N-acetylglucosamine (GlcNAc) 2-epimerase/ManNAc kinase (GNE) myopathy, a disorder of impaired sialic acid synthesis. Using previous preclinical and clinical data, we propose to test ManNAc therapy for subjects with primary glomerular diseases. Even though the exact mechanisms, affected cell types, and pathologic consequences of glomerular hyposialylation need further study, treatment with this physiological monosaccharide could potentially replace or supplement existing glomerular diseases therapies.

19.
Sci Rep ; 8(1): 59, 2018 01 08.
Article in English | MEDLINE | ID: mdl-29311649

ABSTRACT

microRNAs (miRNAs) are critical for neuronal function and their dysregulation is repeatedly observed in neurodegenerative diseases. Here, we implemented high content image analysis for investigating the impact of several miRNAs in mouse primary motor neurons. This survey directed our attention to the neuron-specific miR-124, which controls axonal morphology. By performing next generation sequencing analysis and molecular studies, we characterized novel roles for miR-124 in control of mitochondria localization and function. We further demonstrated that the intermediate filament Vimentin is a key target of miR-124 in this system. Our data establishes a new pathway for control of mitochondria function in motor neurons, revealing the value of a neuron-specific miRNA gene as a mechanism for the re-shaping of otherwise ubiquitously-expressed intermediate filament network, upstream of mitochondria activity and cellular metabolism.


Subject(s)
Gene Expression Regulation , MicroRNAs/genetics , Mitochondria/genetics , Mitochondria/metabolism , Motor Neurons/metabolism , RNA Interference , Vimentin/genetics , Animals , Axons , Cells, Cultured , Computational Biology/methods , Gene Expression Profiling , Gene Ontology , Mice , Molecular Imaging , Transcriptome , Vimentin/metabolism
20.
Biomark Med ; 8(5): 641-52, 2014.
Article in English | MEDLINE | ID: mdl-25123033

ABSTRACT

AIM: The exact pathomechanism of GNE myopathy remains elusive, but likely involves aberrant sialylation. We explored sialylation status of blood-based glycans as potential disease markers. METHODS: We employed immunoblotting, lectin histochemistry and mass spectrometry. RESULTS: GNE myopathy muscle showed hyposialylation of predominantly O-linked glycans. The O-linked glycome of patients' plasma compared with controls showed increased amounts of desialylated Thomsen-Friedenreich (T)-antigen, and/or decreased amounts of its sialylated form, ST-antigen. Importantly, all patients had increased T/ST ratios compared with controls. These ratios were normalized in a patient treated with intravenous immunoglobulins as a source of sialic acid. DISCUSSION:  GNE myopathy clinical trial data will reveal whether T/ST ratios correlate to muscle function.  CONCLUSION: Plasma T/ST ratios are a robust blood-based biomarker for GNE myopathy, and may also help explain the pathology and course of the disease.


Subject(s)
Antigens, Tumor-Associated, Carbohydrate/blood , Antigens, Tumor-Associated, Carbohydrate/metabolism , Multienzyme Complexes/metabolism , Muscular Diseases/blood , Muscular Diseases/enzymology , N-Acetylneuraminic Acid/metabolism , Biomarkers/blood , Biomarkers/metabolism , Humans , Lectins/blood , Neural Cell Adhesion Molecules/blood , Polysaccharides/blood
SELECTION OF CITATIONS
SEARCH DETAIL