Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 14 de 14
1.
EMBO Mol Med ; 15(3): e14837, 2023 03 08.
Article En | MEDLINE | ID: mdl-36789546

Multiple sulfatase deficiency (MSD, MIM #272200) results from pathogenic variants in the SUMF1 gene that impair proper function of the formylglycine-generating enzyme (FGE). FGE is essential for the posttranslational activation of cellular sulfatases. MSD patients display reduced or absent sulfatase activities and, as a result, clinical signs of single sulfatase disorders in a unique combination. Up to date therapeutic options for MSD are limited and mostly palliative. We performed a screen of FDA-approved drugs using immortalized MSD patient fibroblasts. Recovery of arylsulfatase A activity served as the primary readout. Subsequent analysis confirmed that treatment of primary MSD fibroblasts with tazarotene and bexarotene, two retinoids, led to a correction of MSD pathophysiology. Upon treatment, sulfatase activities increased in a dose- and time-dependent manner, reduced glycosaminoglycan content decreased and lysosomal position and size normalized. Treatment of MSD patient derived induced pluripotent stem cells (iPSC) differentiated into neuronal progenitor cells (NPC) resulted in a positive treatment response. Tazarotene and bexarotene act to ultimately increase the stability of FGE variants. The results lay the basis for future research on the development of a first therapeutic option for MSD patients.


Multiple Sulfatase Deficiency Disease , Humans , Multiple Sulfatase Deficiency Disease/diagnosis , Multiple Sulfatase Deficiency Disease/genetics , Multiple Sulfatase Deficiency Disease/pathology , Bexarotene , Drug Evaluation, Preclinical , Sulfatases/genetics , Oxidoreductases Acting on Sulfur Group Donors
2.
J Inherit Metab Dis ; 43(6): 1298-1309, 2020 11.
Article En | MEDLINE | ID: mdl-32749716

Multiple sulfatase deficiency (MSD) is an ultra-rare neurodegenerative disorder caused by pathogenic variants in SUMF1. This gene encodes formylglycine-generating enzyme (FGE), a protein required for sulfatase activation. The clinical course of MSD results from additive effect of each sulfatase deficiency, including metachromatic leukodystrophy (MLD), several mucopolysaccharidoses (MPS II, IIIA, IIID, IIIE, IVA, VI), chondrodysplasia punctata, and X-linked ichthyosis. While it is known that affected individuals demonstrate a complex and severe phenotype, the genotype-phenotype relationship and detailed clinical course is unknown. We report on 35 cases enrolled in our retrospective natural history study, n = 32 with detailed histories. Neurologic function was longitudinally assessed with retrospective scales. Biochemical and computational modeling of novel SUMF1 variants was performed. Genotypes were classified based on predicted functional change, and each individual was assigned a genotype severity score. The median age at symptom onset was 0.25 years; median age at diagnosis was 2.7 years; and median age at death was 13 years. All individuals demonstrated developmental delay, and only a subset of individuals attained ambulation and verbal communication. All subjects experienced an accumulating systemic symptom burden. Earlier age at symptom onset and severe variant pathogenicity correlated with poor neurologic outcomes. Using retrospective deep phenotyping and detailed variant analysis, we defined the natural history of MSD. We found that attenuated cases can be distinguished from severe cases by age of onset, attainment of ambulation, and genotype. Results from this study can help inform prognosis and facilitate future study design.


Leukodystrophy, Metachromatic/genetics , Mucopolysaccharidoses/genetics , Multiple Sulfatase Deficiency Disease/genetics , Oxidoreductases Acting on Sulfur Group Donors/genetics , Adolescent , Child , Child, Preschool , Female , Genotype , Glycine/analogs & derivatives , Glycine/genetics , Glycine/metabolism , Humans , Infant , Internationality , Leukodystrophy, Metachromatic/pathology , Male , Mucopolysaccharidoses/pathology , Multiple Sulfatase Deficiency Disease/pathology , Mutation , Phenotype , Rare Diseases , Retrospective Studies , Sulfatases/deficiency , Sulfatases/genetics
3.
J Inherit Metab Dis ; 43(6): 1288-1297, 2020 11.
Article En | MEDLINE | ID: mdl-32621519

Multiple Sulfatase Deficiency (MSD, MIM#272200) is an ultra-rare lysosomal storage disorder arising from mutations in the SUMF1 gene, which encodes the formylglycine-generating enzyme (FGE). FGE is necessary for the activation of sulfatases, a family of enzymes that are involved in the degradation of sulfated substrates such as glycosaminoglycans and sulfolipids. SUMF1 mutations lead to functionally impaired FGE and individuals with MSD demonstrate clinical signs of single sulfatase deficiencies, including metachromatic leukodystrophy (MLD) and several mucopolysaccharidosis (MPS) subtypes. Comprehensive information related to the natural history of MSD is missing. We completed a systematic literature review and a meta-analysis on data from published cases reporting on MSD. As available from these reports, we extracted clinical, genetic, biochemical, and brain imaging information. We identified 75 publications with data on 143 MSD patients with a total of 53 unique SUMF1 mutations. The mean survival was 13 years (95% CI 9.8-16.2 years). Seventy-five clinical signs and 11 key clusters of signs were identified. The most frequently affected organs systems were the nervous, skeletal, and integumentary systems. The most frequent MRI features were abnormal myelination and cerebral atrophy. Individuals with later onset MSD signs and survived longer than those with signs at birth. Less severe mutations, low disease burden and achievement of independent walking positively correlated with longer survival. Despite the limitations of our approach, we were able to define clinical characteristics and disease outcomes in MSD. This work will provide the foundation of natural disease history data needed for future clinical trial design.


Leukodystrophy, Metachromatic/genetics , Mucopolysaccharidoses/genetics , Multiple Sulfatase Deficiency Disease/genetics , Oxidoreductases Acting on Sulfur Group Donors/genetics , Glycine/analogs & derivatives , Glycine/genetics , Glycine/metabolism , Humans , Leukodystrophy, Metachromatic/pathology , Mucopolysaccharidoses/pathology , Multiple Sulfatase Deficiency Disease/pathology , Protein Processing, Post-Translational/genetics , Sulfatases/deficiency , Sulfatases/genetics
4.
Int J Mol Sci ; 21(10)2020 May 13.
Article En | MEDLINE | ID: mdl-32414121

Multiple sulfatase deficiency (MSD, MIM #272200) is an ultra-rare disease comprising pathophysiology and clinical features of mucopolysaccharidosis, sphingolipidosis and other sulfatase deficiencies. MSD is caused by impaired posttranslational activation of sulfatases through the formylglycine generating enzyme (FGE) encoded by the sulfatase modifying factor 1 (SUMF1) gene, which is mutated in MSD. FGE is a highly conserved, non-redundant ER protein that activates all cellular sulfatases by oxidizing a conserved cysteine in the active site of sulfatases that is necessary for full catalytic activity. SUMF1 mutations result in unstable, degradation-prone FGE that demonstrates reduced or absent catalytic activity, leading to decreased activity of all sulfatases. As the majority of sulfatases are localized to the lysosome, loss of sulfatase activity induces lysosomal storage of glycosaminoglycans and sulfatides and subsequent cellular pathology. MSD patients combine clinical features of all single sulfatase deficiencies in a systemic disease. Disease severity classifications distinguish cases based on age of onset and disease progression. A genotype- phenotype correlation has been proposed, biomarkers like excreted storage material and residual sulfatase activities do not correlate well with disease severity. The diagnosis of MSD is based on reduced sulfatase activities and detection of mutations in SUMF1. No therapy exists for MSD yet. This review summarizes the unique FGE/ sulfatase physiology, pathophysiology and clinical aspects in patients and their care and outlines future perspectives in MSD.


Mucopolysaccharidoses/genetics , Multiple Sulfatase Deficiency Disease/genetics , Oxidoreductases Acting on Sulfur Group Donors/genetics , Sphingolipidoses/genetics , Glycine/analogs & derivatives , Glycine/genetics , Glycine/metabolism , Humans , Mucopolysaccharidoses/pathology , Multiple Sulfatase Deficiency Disease/pathology , Mutation/genetics , Protein Processing, Post-Translational/genetics , Sphingolipidoses/pathology , Sulfatases/deficiency , Sulfatases/genetics
5.
Mol Genet Genomic Med ; 8(9): e1167, 2020 09.
Article En | MEDLINE | ID: mdl-32048457

BACKGROUND: Multiple sulfatase deficiency (MSD, MIM #272200) is an ultrarare congenital disorder caused by SUMF1 mutation and often misdiagnosed due to its complex clinical presentation. Impeded by a lack of natural history, knowledge gained from individual case studies forms the source for a reliable diagnosis and consultation of patients and parents. METHODS: We collected clinical records as well as genetic and metabolic test results from two MSD patients. The functional properties of a novel SUMF1 variant were analyzed after expression in a cell culture model. RESULTS: We report on two MSD patients-the first neonatal type reported in Israel-both presenting with this most severe manifestation of MSD. Our patients showed uniform clinical symptoms with persistent pulmonary hypertension, hypotonia, and dysmorphism at birth. Both patients were homozygous for the same novel SUMF1 mutation (c.1043C>T, p.A348V). Functional analysis revealed that the SUMF1-encoded variant of formylglycine-generating enzyme is highly instable and lacks catalytic function. CONCLUSION: The obtained results confirm genotype-phenotype correlation in MSD, expand the spectrum of clinical presentation and are relevant for diagnosis including the extremely rare neonatal severe type of MSD.


Multiple Sulfatase Deficiency Disease/genetics , Mutation, Missense , Oxidoreductases Acting on Sulfur Group Donors/genetics , Phenotype , Cell Line, Tumor , Child, Preschool , Enzyme Stability , Homozygote , Humans , Infant , Male , Multiple Sulfatase Deficiency Disease/pathology , Oxidoreductases Acting on Sulfur Group Donors/metabolism
6.
Mol Genet Metab ; 123(3): 337-346, 2018 03.
Article En | MEDLINE | ID: mdl-29397290

Multiple sulfatase deficiency (MSD) is an ultra-rare neurodegenerative disorder that results in defective sulfatase post-translational modification. Sulfatases in the body are activated by a unique protein, formylglycine-generating enzyme (FGE) that is encoded by SUMF1. When FGE is absent or insufficient, all 17 known human sulfatases are affected, including the enzymes associated with metachromatic leukodystrophy (MLD), several mucopolysaccharidoses (MPS II, IIIA, IIID, IVA, VI), chondrodysplasia punctata, and X-linked ichthyosis. As such, individuals demonstrate a complex and severe clinical phenotype that has not been fully characterized to date. In this report, we describe two individuals with distinct clinical presentations of MSD. Also, we detail a comprehensive systems-based approach to the management of individuals with MSD, from the initial diagnostic evaluation to unique multisystem issues and potential management options. As there have been no natural history studies to date, the recommendations within this report are based on published studies and consensus opinion and underscore the need for future research on evidence-based outcomes to improve management of children with MSD.


Consensus , Multiple Sulfatase Deficiency Disease/therapy , Rare Diseases/therapy , Sulfatases/metabolism , Brain/diagnostic imaging , Brain/metabolism , Brain/pathology , Child, Preschool , Female , Humans , Male , Multiple Sulfatase Deficiency Disease/diagnosis , Multiple Sulfatase Deficiency Disease/etiology , Multiple Sulfatase Deficiency Disease/pathology , Mutation , Oxidoreductases Acting on Sulfur Group Donors , Protein Processing, Post-Translational/genetics , Rare Diseases/diagnosis , Rare Diseases/etiology , Sulfatases/deficiency
7.
Eur J Hum Genet ; 21(9): 1020-3, 2013 Sep.
Article En | MEDLINE | ID: mdl-23321616

Multiple sulfatase deficiency (MSD) is a rare inborn error of metabolism affecting posttranslational activation of sulfatases by the formylglycine generating enzyme (FGE). Due to mutations in the encoding SUMF1 gene, FGE's catalytic capacity is impaired resulting in reduced cellular sulfatase activities. Both, FGE protein stability and residual activity determine disease severity and have previously been correlated with the clinical MSD phenotype. Here, we report a patient with a late infantile severe course of disease. The patient is compound heterozygous for two so far undescribed SUMF1 mutations, c.156delC (p.C52fsX57) and c.390A>T (p.E130D). In patient fibroblasts, mRNA of the frameshift allele is undetectable. In contrast, the allele encoding FGE-E130D is expressed. FGE-E130D correctly localizes to the endoplasmic reticulum and has a very high residual molecular activity in vitro (55% of wildtype FGE); however, it is rapidly degraded. Thus, despite substantial residual enzyme activity, protein instability determines disease severity, which highlights that potential MSD treatment approaches should target protein folding and stabilization mechanisms.


Multiple Sulfatase Deficiency Disease/diagnosis , Sulfatases/genetics , Cell Line, Tumor , Child, Preschool , Enzyme Stability/genetics , Fatal Outcome , Female , Humans , Molecular Diagnostic Techniques , Multiple Sulfatase Deficiency Disease/genetics , Multiple Sulfatase Deficiency Disease/pathology , Oxidoreductases Acting on Sulfur Group Donors , Sulfatases/metabolism
8.
Proc Natl Acad Sci U S A ; 109(35): E2334-42, 2012 Aug 28.
Article En | MEDLINE | ID: mdl-22826245

The role of astrocytes in neurodegenerative processes is increasingly appreciated. Here we investigated the contribution of astrocytes to neurodegeneration in multiple sulfatase deficiency (MSD), a severe lysosomal storage disorder caused by mutations in the sulfatase modifying factor 1 (SUMF1) gene. Using Cre/Lox mouse models, we found that astrocyte-specific deletion of Sumf1 in vivo induced severe lysosomal storage and autophagy dysfunction with consequential cytoplasmic accumulation of autophagic substrates. Lysosomal storage in astrocytes was sufficient to induce degeneration of cortical neurons in vivo. Furthermore, in an ex vivo coculture assay, we observed that Sumf1(-/-) astrocytes failed to support the survival and function of wild-type cortical neurons, suggesting a non-cell autonomous mechanism for neurodegeneration. Compared with the astrocyte-specific deletion of Sumf1, the concomitant removal of Sumf1 in both neurons and glia in vivo induced a widespread neuronal loss and robust neuroinflammation. Finally, behavioral analysis of mice with astrocyte-specific deletion of Sumf1 compared with mice with Sumf1 deletion in both astrocytes and neurons allowed us to link a subset of neurological manifestations of MSD to astrocyte dysfunction. This study indicates that astrocytes are integral components of the neuropathology in MSD and that modulation of astrocyte function may impact disease course.


Astrocytes/pathology , Multiple Sulfatase Deficiency Disease/pathology , Nerve Degeneration/pathology , Neurons/pathology , Sulfatases/genetics , Animals , Cell Communication/physiology , Cell Survival/physiology , Cells, Cultured , Cerebellum/pathology , Cerebral Cortex/pathology , Lysosomal Storage Diseases, Nervous System/genetics , Lysosomal Storage Diseases, Nervous System/pathology , Lysosomes/pathology , Lysosomes/ultrastructure , Mice , Mice, Inbred C57BL , Mice, Mutant Strains , Microscopy, Electron , Multiple Sulfatase Deficiency Disease/genetics , Neurons/ultrastructure , Oxidoreductases Acting on Sulfur Group Donors , Purkinje Cells/pathology , Purkinje Cells/ultrastructure
9.
Hum Mol Genet ; 21(8): 1770-81, 2012 Apr 15.
Article En | MEDLINE | ID: mdl-22215441

Dysfunctional mitochondria are a well-known disease hallmark. The accumulation of aberrant mitochondria can alter cell homeostasis, thus resulting in tissue degeneration. Lysosomal storage disorders (LSDs) are a group of inherited diseases characterized by the buildup of undegraded material inside the lysosomes that leads to autophagic-lysosomal dysfunction. In LSDs, autophagic stress has been associated to mitochondrial accumulation and dysfunction. However, the mechanisms underlying mitochondrial aberrations and how these are involved in tissue pathogenesis remain largely unexplored. In normal conditions, mitochondrial clearance occurs by mitophagy, a selective form of autophagy, which relies on a parkin-mediated mitochondrial priming and subsequent sequestration by autophagosomes. Here, we performed a detailed analysis of key steps of mitophagy in a mouse model of multiple sulfatase deficiency (MSD), a severe type of LSD characterized by both neurological and systemic involvement. We demonstrated that in MSD liver reduced parkin levels resulted in inefficient mitochondrial priming, thus contributing to the accumulation of giant mitochondria that are located outside autophagic vesicles ultimately leading to cytochrome c release and apoptotic cell death. Morphological and functional changes were also observed in mitochondria from MSD brain but these were not directly associated with neuronal cell loss, suggesting a secondary contribution of mitochondria to neurodegeneration. Together, these data shed new light on the mechanisms underlying mitochondrial dysfunction in LSDs and on their tissue-specific differential contribution to the pathogenesis of this group of metabolic disorders.


Autophagy , Mitochondria, Liver/metabolism , Mitochondria, Liver/ultrastructure , Multiple Sulfatase Deficiency Disease/metabolism , Multiple Sulfatase Deficiency Disease/pathology , Phagosomes/metabolism , Ubiquitin-Protein Ligases/metabolism , Animals , Apoptosis , Brain/pathology , Cytochromes c/metabolism , Disease Models, Animal , Liver/pathology , Mice , Mitochondria/metabolism , Mitochondria/ultrastructure , Multiple Sulfatase Deficiency Disease/genetics , Ubiquitination
10.
Dev Cell ; 21(3): 421-30, 2011 Sep 13.
Article En | MEDLINE | ID: mdl-21889421

Lysosomes are cellular organelles primarily involved in degradation and recycling processes. During lysosomal exocytosis, a Ca²âº-regulated process, lysosomes are docked to the cell surface and fuse with the plasma membrane (PM), emptying their content outside the cell. This process has an important role in secretion and PM repair. Here we show that the transcription factor EB (TFEB) regulates lysosomal exocytosis. TFEB increases the pool of lysosomes in the proximity of the PM and promotes their fusion with PM by raising intracellular Ca²âº levels through the activation of the lysosomal Ca²âº channel MCOLN1. Induction of lysosomal exocytosis by TFEB overexpression rescued pathologic storage and restored normal cellular morphology both in vitro and in vivo in lysosomal storage diseases (LSDs). Our data indicate that lysosomal exocytosis may directly modulate cellular clearance and suggest an alternative therapeutic strategy for disorders associated with intracellular storage.


Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/metabolism , Exocytosis/genetics , Lysosomes/metabolism , TRPM Cation Channels/genetics , Transcriptional Activation , Animals , Apoptosis , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/genetics , COS Cells , Calcium/metabolism , Cell Membrane/physiology , Chlorocebus aethiops , Disease Models, Animal , HeLa Cells , Humans , Lysosomes/genetics , Membrane Fusion , Mice , Multiple Sulfatase Deficiency Disease/genetics , Multiple Sulfatase Deficiency Disease/metabolism , Multiple Sulfatase Deficiency Disease/pathology , Transient Receptor Potential Channels , Up-Regulation/drug effects
11.
Eur J Immunol ; 39(10): 2748-54, 2009 Oct.
Article En | MEDLINE | ID: mdl-19637231

A defect in invariant NKT (iNKT) cell selection was hypothesized in lysosomal storage disorders (LSD). Accumulation of glycosphingolipids (GSL) in LSD could influence lipid loading and/or presentation causing entrapment of endogenous ligand(s) within storage bodies or competition of the selecting ligand(s) by stored lipids for CD1d binding. However, when we analyzed the iNKT cell compartment in newly tested LSD animal models that accumulate GSL, glycoaminoglycans or both, we observed a defective iNKT cell selection only in animals affected by multiple sulfatase deficiency, in which a generalized aberrant T-cell development, rather than a pure iNKT defect, was present. Mice with single lysosomal enzyme deficiencies had normal iNKT cell development. Thus, GSL/glycoaminoglycans storage and lysosomal engulfment are not sufficient for affecting iNKT cell development. Rather, lipid ligand(s) or storage compounds, which are affected in those LSD lacking mature iNKT cells, might indeed be relevant for iNKT cell selection.


Cell Differentiation/immunology , Lysosomal Storage Diseases/immunology , Natural Killer T-Cells/immunology , Animals , Cell Count , Cell Differentiation/drug effects , Disease Models, Animal , Enzyme Inhibitors/pharmacology , Female , Leukodystrophy, Globoid Cell/genetics , Leukodystrophy, Globoid Cell/immunology , Leukodystrophy, Globoid Cell/pathology , Leukodystrophy, Metachromatic/genetics , Leukodystrophy, Metachromatic/immunology , Liver/immunology , Liver/pathology , Lymphocytes/pathology , Lysosomal Storage Diseases/genetics , Male , Mice , Mice, Inbred C57BL , Mice, Inbred Strains , Mice, Knockout , Mucopolysaccharidosis I/genetics , Mucopolysaccharidosis I/immunology , Multiple Sulfatase Deficiency Disease/genetics , Multiple Sulfatase Deficiency Disease/immunology , Multiple Sulfatase Deficiency Disease/pathology , Natural Killer T-Cells/pathology , Pyrroles/pharmacology , Receptor, Fibroblast Growth Factor, Type 1/antagonists & inhibitors , Sandhoff Disease/genetics , Sandhoff Disease/immunology , Sandhoff Disease/pathology , Spleen/immunology , Spleen/pathology , Thymus Gland/immunology , Thymus Gland/pathology
12.
Biochim Biophys Acta ; 1793(4): 710-25, 2009 Apr.
Article En | MEDLINE | ID: mdl-19124046

Multiple sulfatase deficiency (MSD), mucolipidosis (ML) II/III and Niemann-Pick type C1 (NPC1) disease are rare but fatal lysosomal storage disorders caused by the genetic defect of non-lysosomal proteins. The NPC1 protein mainly localizes to late endosomes and is essential for cholesterol redistribution from endocytosed LDL to cellular membranes. NPC1 deficiency leads to lysosomal accumulation of a broad range of lipids. The precise functional mechanism of this membrane protein, however, remains puzzling. ML II, also termed I cell disease, and the less severe ML III result from deficiencies of the Golgi enzyme N-acetylglucosamine 1-phosphotransferase leading to a global defect of lysosome biogenesis. In patient cells, newly synthesized lysosomal proteins are not equipped with the critical lysosomal trafficking marker mannose 6-phosphate, thus escaping from lysosomal sorting at the trans Golgi network. MSD affects the entire sulfatase family, at least seven members of which are lysosomal enzymes that are specifically involved in the degradation of sulfated glycosaminoglycans, sulfolipids or other sulfated molecules. The combined deficiencies of all sulfatases result from a defective post-translational modification by the ER-localized formylglycine-generating enzyme (FGE), which oxidizes a specific cysteine residue to formylglycine, the catalytic residue enabling a unique mechanism of sulfate ester hydrolysis. This review gives an update on the molecular bases of these enigmatic diseases, which have been challenging researchers since many decades and so far led to a number of surprising findings that give deeper insight into both the cell biology and the pathobiochemistry underlying these complex disorders. In case of MSD, considerable progress has been made in recent years towards an understanding of disease-causing FGE mutations. First approaches to link molecular parameters with clinical manifestation have been described and even therapeutical options have been addressed. Further, the discovery of FGE as an essential sulfatase activating enzyme has considerable impact on enzyme replacement or gene therapy of lysosomal storage disorders caused by single sulfatase deficiencies.


Mucolipidoses/pathology , Multiple Sulfatase Deficiency Disease/pathology , Niemann-Pick Disease, Type C/pathology , Proteins/metabolism , Biological Transport , Humans , Mucolipidoses/classification , Multiple Sulfatase Deficiency Disease/enzymology , Multiple Sulfatase Deficiency Disease/genetics , Multiple Sulfatase Deficiency Disease/therapy , Protein Processing, Post-Translational
13.
Brain Dev ; 30(5): 374-7, 2008 May.
Article En | MEDLINE | ID: mdl-18509892

Multiple sulfatase deficiency (MSD) is an inherited lysosomal storage disease that affects post-translational activation of all of the sulfatases. Since biochemical and clinical findings are variable, the diagnosis is difficult in most of the cases. Missense, nonsense, microdeletion and splicing mutations in SUMF1 gene were found in all of the MSD patients analyzed. Here, we present clinical findings of two consanguineous patients with multiple sulfatase deficiency. They were found to be homozygous for a novel missense mutation c.739G > C causing a p.G247R amino acid substitution in the SUMF1 protein.


Family Health , Multiple Sulfatase Deficiency Disease/genetics , Mutation/genetics , Sulfatases/genetics , Arginine/genetics , Atrophy/pathology , Cerebral Cortex/pathology , Child, Preschool , Female , Glycine/genetics , Humans , Infant , Multiple Sulfatase Deficiency Disease/pathology , Oxidoreductases Acting on Sulfur Group Donors , Turkey
14.
Hum Mol Genet ; 17(1): 119-29, 2008 Jan 01.
Article En | MEDLINE | ID: mdl-17913701

Most lysosomal storage disorders (LSDs) are caused by deficiencies of lysosomal hydrolases. While LSDs were among the first inherited diseases for which the underlying biochemical defects were identified, the mechanisms from enzyme deficiency to cell death are poorly understood. Here we show that lysosomal storage impairs autophagic delivery of bulk cytosolic contents to lysosomes. By studying the mouse models of two LSDs associated with severe neurodegeneration, multiple sulfatase deficiency (MSD) and mucopolysaccharidosis type IIIA (MPSIIIA), we observed an accumulation of autophagosomes resulting from defective autophagosome-lysosome fusion. An impairment of the autophagic pathway was demonstrated by the inefficient degradation of exogenous aggregate-prone proteins (i.e. expanded huntingtin and mutated alpha-synuclein) in cells from LSD mice. This impairment resulted in massive accumulation of polyubiquitinated proteins and of dysfunctional mitochondria which are the putative mediators of cell death. These data identify LSDs as 'autophagy disorders' and suggest the presence of common mechanisms in the pathogenesis of these and other neurodegenerative diseases.


Autophagy/physiology , Lysosomal Storage Diseases/pathology , Animals , Autophagy/genetics , Base Sequence , Cells, Cultured , DNA Primers/genetics , Humans , Lysosomal Storage Diseases/genetics , Lysosomal Storage Diseases/physiopathology , Lysosomal Storage Diseases, Nervous System/genetics , Lysosomal Storage Diseases, Nervous System/pathology , Lysosomal Storage Diseases, Nervous System/physiopathology , Lysosomes/pathology , Membrane Fusion , Mice , Microtubule-Associated Proteins/genetics , Microtubule-Associated Proteins/metabolism , Mitochondria/pathology , Mucopolysaccharidosis III/genetics , Mucopolysaccharidosis III/pathology , Mucopolysaccharidosis III/physiopathology , Multiple Sulfatase Deficiency Disease/genetics , Multiple Sulfatase Deficiency Disease/pathology , Multiple Sulfatase Deficiency Disease/physiopathology , Nerve Degeneration/genetics , Nerve Degeneration/pathology , Nerve Degeneration/physiopathology , Phagosomes/pathology , Transfection , Ubiquitination
...