Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
1.
Biochim Biophys Acta Biomembr ; 1862(12): 183336, 2020 12 01.
Article in English | MEDLINE | ID: mdl-32389669

ABSTRACT

Disorders caused by defects in lysosomal membrane transporters form a distinct subgroup of lysosomal storage disorders (LSDs). To date, defects in only 10 lysosomal membrane transporters have been associated with inherited disorders. The clinical presentations of these diseases resemble the phenotypes of other LSDs; they are heterogeneous and often present in children with neurodegenerative manifestations. However, for pathomechanistic and therapeutic studies, lysosomal membrane transport defects should be distinguished from LSDs caused by defective hydrolytic enzymes. The involved proteins differ in function, localization, and lysosomal targeting, and the diseases themselves differ in their stored material and therapeutic approaches. We provide an overview of the small group of disorders of lysosomal membrane transporters, emphasizing discovery, pathomechanism, clinical features, diagnostic methods and therapeutic aspects. We discuss common aspects of lysosomal membrane transporter defects that can provide the basis for preclinical research into these disorders.


Subject(s)
Lysosomal Storage Diseases/pathology , Lysosomes/metabolism , Membrane Transport Proteins/metabolism , Amino Acid Transport Systems, Neutral/genetics , Amino Acid Transport Systems, Neutral/metabolism , Cystinosis/genetics , Cystinosis/pathology , Histiocytosis/genetics , Histiocytosis/pathology , Humans , Lysosomal Storage Diseases/genetics , Membrane Transport Proteins/genetics , Nucleoside Transport Proteins/genetics , Nucleoside Transport Proteins/metabolism , Organic Anion Transporters/genetics , Organic Anion Transporters/metabolism , Sialic Acid Storage Disease/genetics , Sialic Acid Storage Disease/pathology , Symporters/genetics , Symporters/metabolism
2.
Prenat Diagn ; 40(5): 605-611, 2020 04.
Article in English | MEDLINE | ID: mdl-32003481

ABSTRACT

OBJECTIVES: There are many causes of fetal effusions, including the rare lysosomal storage diseases (LSDs). Vacuolated lymphocytes (VLs) are found in the blood of infants with LSDs, and their presence in fetal effusion could increase the risk of underlying LSD. METHODS: Between 2006 and 2018, all fetal effusions samples from 43 fetal multidisciplinary centers were referred to a single laboratory. Cells were counted, and, if observed, VLs were categorized and counted. Screening for LSDs was performed by metabolite analyses on amniotic fluid supernatant. The diagnosis of an LSD was confirmed by measuring the activity of the corresponding enzyme and/or mutation analysis. RESULTS: Our laboratory received 614 ascitic fluids and 280 pleural fluids sampled between 22 and 33 weeks of gestation. The final diagnosis was LSD in 16 cases (1.8%). VLs were reported in all these 16 cases, in a mix of lymphocytes with and without vacuoles. Vacuoles in VLs varied in size and number. In most cases, VLs were easy to recognize, with numerous, large, round, well-defined vacuoles, but in three cases of LSDs, VLs were atypical. CONCLUSION: The finding of VLs in fetal effusions is an inexpensive first-line test that may help to prioritize biochemical and genetic tests for LSDs.


Subject(s)
Ascites/pathology , Lymphocytes/pathology , Lysosomal Storage Diseases/pathology , Pleural Effusion/pathology , Vacuoles/pathology , Ascitic Fluid/pathology , Female , Gangliosidosis, GM1/diagnosis , Gangliosidosis, GM1/pathology , Humans , Lysosomal Storage Diseases/diagnosis , Mucolipidoses/diagnosis , Mucolipidoses/pathology , Mucopolysaccharidosis VII/diagnosis , Mucopolysaccharidosis VII/pathology , Niemann-Pick Disease, Type C/diagnosis , Niemann-Pick Disease, Type C/pathology , Pregnancy , Prenatal Diagnosis , Sensitivity and Specificity , Sialic Acid Storage Disease/diagnosis , Sialic Acid Storage Disease/pathology
3.
Genet Med ; 21(2): 347-352, 2019 02.
Article in English | MEDLINE | ID: mdl-29875421

ABSTRACT

PURPOSE: Quantitative definition of the natural history of free sialic acid storage disease (SASD, OMIM 604369), an orphan disorder due to the deficiency of the proton-driven carrier SLC17A5. METHODS: Analysis of published cases with SASD (N = 116) respecting STROBE criteria. MAIN OUTCOME PARAMETERS: survival and diagnostic delay. Phenotype, phenotype-biomarker associations, and geographical patient distribution were explored. RESULTS: Median age at disease onset was 0.17 years. Median age at diagnosis was 3 years with a median diagnostic delay of 2.5 years. Median survival was 11 years. The biochemical phenotype clearly predicted the disease course: patients with a urinary free sialic acid excretion below 6.37-fold or an intracellular free sialic acid storage in fibroblasts below 7.37-fold of the mean of normal survived longer than patients with biochemical values above these thresholds. Cluster analysis of disease features suggested a continuous phenotypic spectrum. Patient distribution was panethnic. CONCLUSION: Combination of neurologic symptoms, visceromegaly, and dysmorphic features and/or nonimmune hydrops fetalis should prompt specific tests for SASD, reducing diagnostic delay. The present quantitative data inform clinical studies and may stimulate and accelerate development of specific therapies. Biomarker-phenotype association is particularly important for both counseling parents and study design.


Subject(s)
Sialic Acid Storage Disease , Age of Onset , Biomarkers , Child , Child, Preschool , Cohort Studies , Cross-Sectional Studies , Delayed Diagnosis , Female , Humans , Infant , Male , N-Acetylneuraminic Acid/urine , Phenotype , Pregnancy , Prenatal Diagnosis , Sialic Acid Storage Disease/diagnosis , Sialic Acid Storage Disease/epidemiology , Sialic Acid Storage Disease/pathology , Survival Analysis
4.
Clin Chim Acta ; 482: 199-202, 2018 Jul.
Article in English | MEDLINE | ID: mdl-29654786

ABSTRACT

Nonimmune hydrops fetalis is the most severe clinical manifestation of lysosomal storage diseases (LSDs). Around 14 different LSDs have been accounted for as 1-15% of the cause of nonimmune hydrops fetalis. We report a Korean infant affected by an extremely rare but severe form of sialic acid storage disease. The patient presented with nonimmune hydrops fetalis, dysmorphic facial features, hepatosplenomegaly, and dysostosis multiplex and died at 39 days of age due to persistent pulmonary hypertension. LSD was suspected based on the presence of diffuse vacuolation of syncytiotrophoblast, villous stromal cells, and intermediate trophoblast in placental biopsy. Increased excretion of urinary free sialic acid was detected by liquid chromatography-tandem mass spectrometry. The patient was compound heterozygous of the c.908G>A (p.Trp303Ter) and the splicing mutation c.1259+5G>T (IVS9+5 G>T) in the SLC17A5 gene.


Subject(s)
Hydrops Fetalis , Lysosomal Storage Diseases/complications , Sialic Acid Storage Disease/pathology , Alternative Splicing/genetics , Humans , Hypertension, Pulmonary/mortality , Infant , Infant, Newborn , Lysosomal Storage Diseases/pathology , Mutation , Organic Anion Transporters/genetics , Polymorphism, Single Nucleotide , Republic of Korea , Sialic Acid Storage Disease/genetics , Symporters/genetics
5.
Exp Neurol ; 291: 106-119, 2017 05.
Article in English | MEDLINE | ID: mdl-28189729

ABSTRACT

Slc17a5-/- mice represent an animal model for the infantile form of sialic acid storage disease (SASD). We analyzed genetic and histological time-course expression of myelin and oligodendrocyte (OL) lineage markers in different parts of the CNS, and related this to postnatal neurobehavioral development in these mice. Sialin-deficient mice display a distinct spatiotemporal pattern of sialic acid storage, CNS hypomyelination and leukoencephalopathy. Whereas few genes are differentially expressed in the perinatal stage (p0), microarray analysis revealed increased differential gene expression in later postnatal stages (p10-p18). This included progressive upregulation of neuroinflammatory genes, as well as continuous down-regulation of genes that encode myelin constituents and typical OL lineage markers. Age-related histopathological analysis indicates that initial myelination occurs normally in hindbrain regions, but progression to more frontal areas is affected in Slc17a5-/- mice. This course of progressive leukoencephalopathy and CNS hypomyelination delays neurobehavioral development in sialin-deficient mice. Slc17a5-/- mice successfully achieve early neurobehavioral milestones, but exhibit progressive delay of later-stage sensory and motor milestones. The present findings may contribute to further understanding of the processes of CNS myelination as well as help to develop therapeutic strategies for SASD and other myelination disorders.


Subject(s)
Brain/pathology , Gene Expression Regulation, Developmental/genetics , Leukoencephalopathies , Mental Disorders/etiology , Organic Anion Transporters/deficiency , Sialic Acid Storage Disease , Symporters/deficiency , Age Factors , Animals , Animals, Newborn , Brain/metabolism , Developmental Disabilities/etiology , Developmental Disabilities/genetics , Disease Models, Animal , Glial Fibrillary Acidic Protein/metabolism , Intermediate Filaments/metabolism , Leukoencephalopathies/complications , Leukoencephalopathies/etiology , Leukoencephalopathies/genetics , Lysosomal-Associated Membrane Protein 1/metabolism , Mice , Mice, Inbred C57BL , Mice, Transgenic , Organic Anion Transporters/genetics , Sialic Acid Storage Disease/complications , Sialic Acid Storage Disease/genetics , Sialic Acid Storage Disease/pathology , Symporters/genetics
6.
Chembiochem ; 18(13): 1188-1193, 2017 07 04.
Article in English | MEDLINE | ID: mdl-27966821

ABSTRACT

Sialuria is a rare autosomal dominant disorder of mammalian metabolism, caused by defective feedback inhibition of the UDP-N-acetylglucosamine-2-epimerase N-acetylmannosamine kinase (GNE), the key enzyme of sialic acid biosynthesis. Sialuria is characterized by overproduction of free sialic acid in the cell cytoplasm. Patients exhibit vastly increased urinary excretion of sialic acid and show differently pronounced developmental delays. The physiopathology of sialuria is not well understood. Here we established a transgenic mouse line that expresses GNE containing the sialuria mutation R263L, in order to investigate the influence of an altered sialic acid concentration on the organism. The transgenic mice that expressed the mutated RNA excreted up to 400 times more N-acetylneuraminic acid than wild-type mice. Additionally, we found higher sialic acid concentration in the brain cytoplasm. Analyzing the (poly)sialylation of neural cell adhesion molecule (NCAM) revealed increased polysialylation in brains of transgenic mice compared to wild-type. However, we found only minor changes in membrane-bound sialylation in various organs but, surprisingly, a significant increase in surface sialylation on leukocytes. Our results suggest that the intracellular sialic acid concentration regulates polysialylation on NCAM in vivo; this could play a role in the manifestation of the developmental delays in sialuria patients.


Subject(s)
Leukocytes/metabolism , Multienzyme Complexes/genetics , N-Acetylneuraminic Acid/urine , Neural Cell Adhesion Molecules/metabolism , Protein Processing, Post-Translational , Sialic Acid Storage Disease/metabolism , Age Factors , Animals , Brain/metabolism , Disease Models, Animal , Feedback, Physiological , Humans , Leukocytes/pathology , Liver/metabolism , Mice , Mice, Transgenic , Multienzyme Complexes/deficiency , Mutation , Neural Cell Adhesion Molecules/chemistry , Neural Cell Adhesion Molecules/genetics , Organ Specificity , Sialic Acid Storage Disease/genetics , Sialic Acid Storage Disease/pathology
7.
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
8.
Mol Genet Metab ; 109(1): 77-85, 2013 May.
Article in English | MEDLINE | ID: mdl-23465695

ABSTRACT

Infection of Spodoptera frugiperda (Sf9) cells by baculovirus (BV) is well established for transgene expression of soluble proteins, but few correctly folded transmembrane proteins have been so produced. We here report the use of the BV/Sf9 (BVES) method for the expression and transfer, via microvesicles, of the exclusive lysosomal exporters for cystine and sialic acid, human cystinosin and sialin. These proteins and their mRNA are released into the culture medium as very low-density microvesicles (~1.05 g/ml), which do not label for lysobisphosphatidic acid. The presence of the human transgene proteins in the vesicles was confirmed by western blotting and confirmed and quantified by mass spectrometry. Addition of vesicles to cultures of human fibroblast lines deficient in either cystinosin or sialin produced a progressive depletion of stored lysosomal cystine or sialic acid, respectively. The depletion effect was slow (T1/2 ~48 h), saturable (down to ~40% of initial after 4 days) and stable (>one week). Surprisingly, BV infection of Spodoptera appeared to induce expression and release into microvesicles of the insect orthologue of cystinosin, but not of sialin. We conclude that BVES is an effective method to express and transfer functional transmembrane proteins so as to study their properties in mammalian cells, and has a generic potential for transport protein replacement therapy.


Subject(s)
Amino Acid Transport Systems, Neutral/metabolism , Organic Anion Transporters/metabolism , Sialic Acid Storage Disease/genetics , Sialic Acid Storage Disease/therapy , Symporters/metabolism , Amino Acid Transport Systems, Neutral/genetics , Animals , Baculoviridae , Cell Line , Gene Transfer Techniques , Humans , In Vitro Techniques , Lysosomes/genetics , Lysosomes/metabolism , Lysosomes/pathology , Membrane Proteins/genetics , Membrane Proteins/metabolism , Microvessels/metabolism , Organic Anion Transporters/genetics , Promoter Regions, Genetic , Sialic Acid Storage Disease/pathology , Spodoptera/cytology , Symporters/genetics
9.
Semin Neurol ; 32(1): 51-4, 2012 Feb.
Article in English | MEDLINE | ID: mdl-22422206
10.
J Inherit Metab Dis ; 34(5): 1069-73, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21617927

ABSTRACT

Sialic acid storage disease (SASD) is an inborn error resulting from defects in the lysosomal membrane protein sialin. The SASD phenotypical spectrum ranges from a severe presentation, infantile sialic acid storage disease (ISSD) which may present as hydrops fetalis, to a relatively mild form, Salla disease. Screening for SASD is performed by determination of free sialic acid (FSA) in urine or amniotic fluid supernatant (AFS). Subsequent diagnosis of SASD is performed by quantification of FSA in cultured fibroblasts and by mutation analysis of the sialin gene, SLC17A5. We describe simple quantitative procedures to determine FSA as well as conjugated sialic acid in AFS, and FSA in cultured fibroblasts, using isotope dilution ((13)C(3)-sialic acid) and multiple reaction monitoring LC-ESI-MS/MS. The whole procedure can be performed in 2-4 h. Reference values in AFS were 0-8.2 µmol/L for 15-25 weeks of gestation and 3.2-12.0 µmol/L for 26-38 weeks of gestation. In AFS samples from five fetuses affected with ISSD FSA was 23.9-58.9 µmol/L demonstrating that this method is able to discriminate ISSD pregnancies from normal ones. The method was also validated for determination of FSA in fibroblast homogenates. FSA in SASD fibroblasts (ISSD; 20-154 nmol/mg protein, intermediate SASD; 12.9-15.1 nmol/mg, Salla disease; 5.9-7.4 nmol/mg) was clearly elevated compared to normal controls (0.3-2.2 nmol/mg). In conclusion, we report simple quantitative procedures to determine FSA in AFS and cultured fibroblasts improving both prenatal diagnostic efficacy for ISSD as well as confirmatory testing in cultured fibroblasts following initial screening in urine or AFS.


Subject(s)
Fibroblasts/cytology , Fibroblasts/pathology , Prenatal Diagnosis/methods , Sialic Acid Storage Disease/diagnosis , Tandem Mass Spectrometry/methods , Amniotic Fluid/chemistry , Calibration , Cells, Cultured , Chromatography, Liquid/methods , Chromatography, Liquid/standards , Female , Humans , Pregnancy , Pregnancy Trimester, Second/metabolism , Pregnancy Trimester, Second/urine , Pregnancy Trimester, Third/metabolism , Pregnancy Trimester, Third/urine , Prenatal Diagnosis/instrumentation , Prenatal Diagnosis/standards , Reproducibility of Results , Sialic Acid Storage Disease/pathology , Tandem Mass Spectrometry/standards , Urinalysis/methods
13.
J Child Neurol ; 20(10): 814-6, 2005 Oct.
Article in English | MEDLINE | ID: mdl-16417876

ABSTRACT

The objective of this article is to describe the clinical, radiographic, and molecular genetic features of a new intermediate form of free sialic storage disease. Free sialic storage disease is a rare autosomal recessive lysosomal disorder that results from mutations in SLC17A5, a gene that codes for sialin, a lysosomal membrane sialic acid transporting protein. Infantile sialic acid storage disease has a severe phenotype, and Salla disease (Finnish variant) is generally milder in phenotype; intermediate forms have also been described. There have been few reports of magnetic resonance imaging (MRI) in the sialic acid storage disorders; leukodystrophy has been the characteristic finding, along with hypoplasia of the corpus callosum. An 8-month-old non-Finnish child presented with hypotonia and global developmental delay. Serial MRIs with magnetic resonance spectroscopy at 9 and 16 months revealed severe hypomyelination and hypogenesis of the corpus callosum. There was mild elevation of urinary sialic acid (4.5 times above normal). Electron microscopy of a skin biopsy showed lysosomal enlargement with oligosaccharide storage, and confirmatory molecular genetic testing revealed compound heterozygosity for two new SLC17A5 mutations. Free sialic storage disease of the intermediate type is an important part of the differential diagnosis of a hypotonic, delayed child with abnormal white matter on MRI. Intermediate types of free sialic acid overlap in phenotype with infantile sialic acid storage disease and the milder Salla disease and thus might be more difficult to identify clinically; the lack of Finnish ethnicity should not preclude testing for this probably under-recognized disorder. White-matter abnormalities appear to be characteristic of the entire phenotypic spectrum.


Subject(s)
Brain/pathology , Sialic Acid Storage Disease/genetics , Sialic Acid Storage Disease/pathology , Humans , Infant , Magnetic Resonance Imaging , Male , Microscopy, Electron , N-Acetylneuraminic Acid/urine , Phenotype , Severity of Illness Index , Skin/pathology
15.
Neurobiol Dis ; 15(2): 251-61, 2004 Mar.
Article in English | MEDLINE | ID: mdl-15006695

ABSTRACT

SLC17A5 encodes a lysosomal membrane protein, sialin, which transports sialic acid from lysosomes. Mutations in sialin result in neurodegenerative sialic acid storage disorders, Salla disease (SD) and infantile sialic acid storage disease (ISSD). Here we analyzed sialin in mouse central nervous system (CNS) and primary cortical and hippocampal neurons and glia. In the CNS, sialin was predominantly expressed in neurons, especially in the proliferative zone of the prospective neocortex and the hippocampus in developing brain. In nonneuronal cells and primary glial cell cultures, mouse sialin was localized into lysosomes but interestingly, in primary neuronal cultures sialin was not targeted into lysosomes but rather revealed a punctate staining along the neuronal processes and was also seen in the plasma membrane. These data demonstrate a nonlysosomal localization of sialin in neurons and would imply a role for sialin in the secretory processes of neuronal cells.


Subject(s)
Brain/embryology , Brain/metabolism , Cell Membrane/metabolism , Lysosomes/metabolism , Neurons/metabolism , Organic Anion Transporters/metabolism , Symporters/metabolism , Animals , Biomarkers , Brain/ultrastructure , Cell Differentiation/physiology , Cell Membrane/ultrastructure , Cerebral Cortex/cytology , Cerebral Cortex/embryology , Cerebral Cortex/metabolism , Fetus , HeLa Cells , Hippocampus/embryology , Hippocampus/metabolism , Hippocampus/ultrastructure , Humans , Immunohistochemistry , Mice , N-Acetylneuraminic Acid/metabolism , Neurites/metabolism , Neurites/ultrastructure , Neuroglia/cytology , Neuroglia/metabolism , Neurons/ultrastructure , Presynaptic Terminals/metabolism , Presynaptic Terminals/ultrastructure , Sialic Acid Storage Disease/genetics , Sialic Acid Storage Disease/metabolism , Sialic Acid Storage Disease/pathology , Sialic Acid Storage Disease/physiopathology
16.
J Hum Genet ; 49(12): 656-663, 2004.
Article in English | MEDLINE | ID: mdl-15635485

ABSTRACT

We report the first Japanese case of Salla disease. A 5-year-old male patient developed unique proteinuria with other clinical manifestations, including coarse facies, dysostosis multiplex, mild mitral valve regurgitation, umbilical and inguinal herniation, and mild developmental delay. Pathological analysis of biopsied kidney tissues showed marked vacuolation of podocytes, mesangial cells, capillary endothelial cells, and tubular cells. Biochemical studies involving thin-layer chromatography and mass spectrometry revealed increased excretion of free sialic acid (N-acetylneuraminic acid) into the patient's urine. Immuno- and lectin staining of the patient's cells demonstrated the accumulation of sialyl and asialyl glycoconjugates in lysosomes and late endosomes. A defect in sialyl glycoconjugate metabolism is thought to have occurred in the patient's cells, besides impairment of the lysosomal transport of free sialic acid residues. A renal disorder should be considered as an important manifestation, not only in infantile free sialic acid storage disease but also in Salla disease.


Subject(s)
Endothelial Cells/pathology , Kidney Diseases/pathology , Kidney Diseases/urine , Kidney Tubules/pathology , Lysosomes/pathology , N-Acetylneuraminic Acid/urine , Sialic Acid Storage Disease/pathology , Sialic Acid Storage Disease/urine , Biological Transport , Child, Preschool , Endothelial Cells/metabolism , Humans , Japan , Kidney Diseases/complications , Kidney Tubules/metabolism , Lysosomes/metabolism , Male , Sialic Acid Storage Disease/complications
SELECTION OF CITATIONS
SEARCH DETAIL
...