Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 185
1.
Cells ; 12(13)2023 07 05.
Article En | MEDLINE | ID: mdl-37443816

The main approach used in the current therapy of mucopolysaccharidosis (MPS) is to reduce the levels of glycosaminoglycans (GAGs) in cells, the deposits considered to be the main cause of the disease. Previous studies have revealed significant differences in the expression of genes encoding proteins involved in many processes, like those related to actin filaments, in MPS cells. Since the regulation of actin filaments is essential for the intracellular transport of specific molecules, the process which may affect the course of MPSs, the aim of this study was to evaluate the changes that occur in the actin cytoskeleton and focal adhesion in cells derived from patients with this disease, as well as in the MPS I mouse model, and to assess whether they could be potential therapeutic targets for different MPS types. Western-blotting, flow cytometry and transcriptomic analyses were employed to address these issues. The levels of the key proteins involved in the studied processes, before and after specific treatment, were assessed. We have also analyzed transcripts whose levels were significantly altered in MPS cells. We identified genes whose expressions were changed in the majority of MPS types and those with particularly highly altered expression. For the first time, significant changes in the expression of genes involved in the actin cytoskeleton structure/functions were revealed which may be considered as an additional element in the pathogenesis of MPSs. Our results suggest the possibility of using the actin cytoskeleton as a potential target in therapeutic approaches for this disease.


Mucopolysaccharidoses , Mucopolysaccharidosis I , Animals , Mice , Focal Adhesions/metabolism , Polymerization , Mucopolysaccharidoses/therapy , Mucopolysaccharidosis I/therapy , Mucopolysaccharidosis I/metabolism , Actin Cytoskeleton/metabolism
2.
Eur J Med Chem ; 247: 115005, 2023 Feb 05.
Article En | MEDLINE | ID: mdl-36563498

Synthesis of a series of l-iduronic acid (IdoA)- and imino-IdoA-typed C-glycosides for modulating α-l-iduronidase (IDUA) activity is described. In an enzyme inhibition study, IdoA-typed C-glycosides were more potent than imino-IdoA analogs, with the most potent IdoA-typed C-glycoside 27c showing an IC50 value of 1 µM. On the other hand, co-treatment of 12 with rh-α-IDUA in mucopolysaccharidosis type I (MPS I) fibroblasts exhibited a nearly 3-fold increase of the IDUA activity, resulting in a clear reduction of the accumulated heparan sulfate (HS) compared to the exogenous enzyme treatment alone. This is the first report of small molecules facilitating IDUA stabilization, enhancing enzyme activity, and reducing accumulated HS in MPS I cell-based assays, which reveals that small molecules as rh-α-IDUA stabilizers to improve enzyme replacement therapy (ERT) efficacy toward MPS I is feasible and promising.


Mucopolysaccharidosis I , Humans , Mucopolysaccharidosis I/drug therapy , Mucopolysaccharidosis I/metabolism , Iduronidase/pharmacology , Iduronidase/metabolism , Heparitin Sulfate/pharmacology , Fibroblasts/metabolism , Glycosides
3.
Biochem Biophys Res Commun ; 636(Pt 1): 147-154, 2022 12 25.
Article En | MEDLINE | ID: mdl-36332477

Mucopolysaccharidosis type I Hurler syndrome (MPS IH) is a severe lysosomal storage disorder caused by alpha-l-iduronidase (IDUA) deficiency. Premature truncation mutations (PTC) are the most common (50%-70%) type of IDUA mutations and correlate with MPS IH. Nonsense suppression therapy is a therapeutic approach that aims to induce stop codon readthrough. The different ability of gentamicin to bind mutant mRNA in readthrough is determined by nucleotide sequence (PTC context: UGA > UAG > UAA) and inserted amino acid including the nucleotide position +4 of the PTC, as well as the mRNA secondary structure. We used COS-7 cells to investigate the functional characteristics of p.Q500X and p.R619X, IDUA variants and the effects of gentamicin in inducing stop codon readthrough of seven IDUA variants including p.Q500X, p.R619X, p.Q70X, p.E299X, p.W312X, p.Q380X, and p.W402X. Moreover, we performed prediction of RNA secondary structure using the online tool RNAfold. We found that cells treated with gentamicin showed significantly enhanced full-length IDUA expression and restored IDUA activity, in a dose-dependent manner, only in cells expressing cDNA with W312X, Q380X, W402X, and R619X. Among the readthrough-responsive variants, we observed UGA PTC in W312X, W402X and R619X; and UAG PTC with C at nucleotide +4 in Q380X. Changes of RNA secondary structure were noted only in mutants with readthrough-responsive variants including W312X, Q380X, W402X, and R619X. Additional preclinical studies of selected PTCs with potential readthrough, using drugs with less oto-nephrotoxicity, in patient's skin fibroblasts and animal model are necessary for the premise of personalized medicine.


Iduronidase , Mucopolysaccharidosis I , Chlorocebus aethiops , Animals , Iduronidase/genetics , Codon, Nonsense/genetics , Gentamicins/pharmacology , Codon, Terminator/genetics , COS Cells , Mucopolysaccharidosis I/drug therapy , Mucopolysaccharidosis I/genetics , Mucopolysaccharidosis I/metabolism , Mutation , RNA, Messenger/metabolism , Nucleotides/therapeutic use
4.
J Mol Med (Berl) ; 100(8): 1223-1235, 2022 08.
Article En | MEDLINE | ID: mdl-35857082

ABSTARCT: Suppressing translation termination at premature termination codons (PTCs), termed readthrough, is a potential therapy for genetic diseases caused by nonsense mutations. Ataluren is a compound that has shown promise for clinical use as a readthrough agent. However, some reports suggest that ataluren is ineffective at suppressing PTCs. To further evaluate the effectiveness of ataluren as a readthrough agent, we examined its ability to suppress PTCs in a variety of previously untested models. Using NanoLuc readthrough reporters expressed in two different cell types, we found that ataluren stimulated a significant level of readthrough. We also explored the ability of ataluren to suppress a nonsense mutation associated with Mucopolysaccharidosis I-Hurler (MPS I-H), a genetic disease that is caused by a deficiency of α-L-iduronidase that leads to lysosomal accumulation of glycosaminoglycans (GAGs). Using mouse embryonic fibroblasts (MEFs) derived from Idua-W402X mice, we found that ataluren partially rescued α-L-iduronidase function and significantly reduced GAG accumulation relative to controls. Two-week oral administration of ataluren to Idua-W402X mice led to significant GAG reductions in most tissues compared to controls. Together, these data reveal important details concerning the efficiency of ataluren as a readthrough agent and the mechanisms that govern its ability to suppress PTCs. KEY MESSAGES: Ataluren promotes readthrough of PTCs in a wide variety of contexts. Ataluren reduces glycosaminoglyan storage in MPS I-H cell and mouse models. Ataluren has a bell-shaped dose-response curve and a narrow effective range.


Iduronidase , Mucopolysaccharidosis I , Animals , Codon, Nonsense/metabolism , Fibroblasts/metabolism , Iduronidase/genetics , Iduronidase/metabolism , Iduronidase/therapeutic use , Luciferases , Mice , Mucopolysaccharidosis I/drug therapy , Mucopolysaccharidosis I/genetics , Mucopolysaccharidosis I/metabolism , Oxadiazoles
5.
Eur J Cell Biol ; 101(3): 151232, 2022.
Article En | MEDLINE | ID: mdl-35537249

Mucopolysaccharidoses (MPS) are inherited metabolic diseases caused by storage of glycosaminoglycans (GAGs), however, various modulations of the course of these diseases were identified recently due to impairment of different cellular processes. Here, using transcriptomic analyses in cells derived from patients suffering from eleven types of MPS, we demonstrated that expression of dozens to hundreds of genes coding for proteins involved in signal transduction processes is significantly changed in MPS cell relative to controls. When studying membrane estrogen receptor 1 (GPER1) and oxytocin receptor (OXTR) in more detail, we unexpectedly found formation of aggregates of GPER1 in MPS I, and those of OXTR in both MPS I and MPS II cells. The presence of these aggregates did not correlate with levels of expression of GPER1 and OXTR genes and levels of corresponding gene products. On the other hand, the aggregates disappeared in cells treated with enzymes which are otherwise deficient in MPS I and MPS II, causing efficient degradation of GAGs. We demonstrated that GPER1 and OXTR aggregates might be formed due to interactions with GAGs rather than arising from changes of levels of these proteins in cells.


Mucopolysaccharidosis II , Mucopolysaccharidosis I , Receptors, Estrogen , Receptors, G-Protein-Coupled , Receptors, Oxytocin , Glycosaminoglycans/metabolism , Humans , Mucopolysaccharidosis I/genetics , Mucopolysaccharidosis I/metabolism , Mucopolysaccharidosis II/genetics , Mucopolysaccharidosis II/metabolism , Receptors, Estrogen/genetics , Receptors, Estrogen/metabolism , Receptors, G-Protein-Coupled/genetics , Receptors, G-Protein-Coupled/metabolism , Receptors, Oxytocin/genetics , Receptors, Oxytocin/metabolism , Signal Transduction
6.
Article En | MEDLINE | ID: mdl-35619307

BACKGROUND: Mucopolysaccharidosis-1H (Hurler syndrome, MPS-1H) is the most severe form of a lysosomal storage disorder (LSD) caused by variants in IDUA, encoding alpha- L-iduronidase (IDUA). MPS-1H is also associated with various degrees of skeletal defects due to the accumulation of partially degraded glycosaminoglycans (GAGs) in the lysosomes of connective tissue cells. The efficacy of hematopoietic stem cell transplantation (HSCT) and enzymatic replacement therapy (ERT) on MPS-1H skeletal manifestations is still considered unsatisfactory. CASE PRESENTATION: We report the case of a young girl, who manifested significant changes in bone remodeling markers and osteoclastogenesis potential after HSCT combined with ERT. She received ERT and underwent two HSCTs. The skeletal alterations at the time of diagnosis showed a trend toward improvement of both mobility and radiological features after HSCT. We observed the highest levels of Receptor activator of nuclear factor-kappa-Β ligand (RANKL) and RANK/osteoprotegerin (OPG) ratio at diagnosis and during ERT, consistently with spontaneous osteoclastogenesis. Conversely, after the successful HSCT with ongoing ERT, the highest levels of osteocalcin were observed and all markers of bone formation and resorption improved. CONCLUSION: The combination therapy of ERT and HSCT was effective in reducing osteoclast activity and increasing osteoblast activity, and these changes were according to the child's bone phenotype, IDUA activity, and Glycosaminoglycan (GAG) trends. These results represent one of the few pieces of human evidence in this context.


Hematopoietic Stem Cell Transplantation , Lysosomal Storage Diseases , Mucopolysaccharidosis I , Child , Female , Humans , Mucopolysaccharidosis I/diagnosis , Mucopolysaccharidosis I/metabolism , Mucopolysaccharidosis I/therapy , Hematopoietic Stem Cell Transplantation/methods , Enzyme Replacement Therapy/methods , Bone Remodeling
7.
N Engl J Med ; 385(21): 1929-1940, 2021 11 18.
Article En | MEDLINE | ID: mdl-34788506

BACKGROUND: Allogeneic hematopoietic stem-cell transplantation is the standard of care for Hurler syndrome (mucopolysaccharidosis type I, Hurler variant [MPSIH]). However, this treatment is only partially curative and is associated with complications. METHODS: We are conducting an ongoing study involving eight children with MPSIH. At enrollment, the children lacked a suitable allogeneic donor and had a Developmental Quotient or Intelligence Quotient score above 70 (i.e., none had moderate or severe cognitive impairment). The children received autologous hematopoietic stem and progenitor cells (HSPCs) transduced ex vivo with an α-L-iduronidase (IDUA)-encoding lentiviral vector after myeloablative conditioning. Safety and correction of blood IDUA activity up to supraphysiologic levels were the primary end points. Clearance of lysosomal storage material as well as skeletal and neurophysiological development were assessed as secondary and exploratory end points. The planned duration of the study is 5 years. RESULTS: We now report interim results. The children's mean (±SD) age at the time of HSPC gene therapy was 1.9±0.5 years. At a median follow-up of 2.10 years, the procedure had a safety profile similar to that known for autologous hematopoietic stem-cell transplantation. All the patients showed prompt and sustained engraftment of gene-corrected cells and had supraphysiologic blood IDUA activity within a month, which was maintained up to the latest follow-up. Urinary glycosaminoglycan (GAG) excretion decreased steeply, reaching normal levels at 12 months in four of five patients who could be evaluated. Previously undetectable levels of IDUA activity in the cerebrospinal fluid became detectable after gene therapy and were associated with local clearance of GAGs. Patients showed stable cognitive performance, stable motor skills corresponding to continued motor development, improved or stable findings on magnetic resonance imaging of the brain and spine, reduced joint stiffness, and normal growth in line with World Health Organization growth charts. CONCLUSIONS: The delivery of HSPC gene therapy in patients with MPSIH resulted in extensive metabolic correction in peripheral tissues and the central nervous system. (Funded by Fondazione Telethon and others; ClinicalTrials.gov number, NCT03488394; EudraCT number, 2017-002430-23.).


Genetic Therapy , Hematopoietic Stem Cell Transplantation , Iduronidase/metabolism , Mucopolysaccharidosis I/therapy , Child, Preschool , Female , Follow-Up Studies , Genetic Vectors , Glycosaminoglycans/urine , Humans , Iduronidase/deficiency , Iduronidase/genetics , Infant , Lentivirus , Male , Mucopolysaccharidosis I/metabolism , Mutation , Stem Cell Transplantation , Transplantation, Autologous
8.
Glycobiology ; 31(10): 1319-1329, 2021 11 18.
Article En | MEDLINE | ID: mdl-34192316

Mucopolysaccharidosis type I (MPS-I) is a rare lysosomal storage disorder caused by deficiency of the enzyme alpha-L-iduronidase, which removes iduronic acid in both chondroitin/dermatan sulfate (CS/DS) and heparan sulfate (HS) and thereby contributes to the catabolism of glycosaminoglycans (GAGs). To ameliorate this genetic defect, the patients are currently treated by enzyme replacement and bone marrow transplantation, which have a number of drawbacks. This study was designed to develop an alternative treatment by inhibition of iduronic acid formation. By screening the Prestwick drug library, we identified ebselen as a potent inhibitor of enzymes that produce iduronic acid in CS/DS and HS. Ebselen efficiently inhibited iduronic acid formation during CS/DS synthesis in cultured fibroblasts. Treatment of MPS-I fibroblasts with ebselen not only reduced accumulation of CS/DS but also promoted GAG degradation. In early Xenopus embryos, this drug phenocopied the effect of downregulation of DS-epimerase 1, the main enzyme responsible for iduronic production in CS/DS, suggesting that ebselen inhibits iduronic acid production in vivo. However, ebselen failed to ameliorate the CS/DS and GAG burden in MPS-I mice. Nevertheless, the results propose a potential of iduronic acid substrate reduction therapy for MPS-I patients.


Fibroblasts/drug effects , Glycosaminoglycans/antagonists & inhibitors , Iduronic Acid/antagonists & inhibitors , Isoindoles/pharmacology , Mucopolysaccharidosis I/drug therapy , Organoselenium Compounds/pharmacology , Dose-Response Relationship, Drug , Fibroblasts/metabolism , Fibroblasts/pathology , Glycosaminoglycans/metabolism , HEK293 Cells , Humans , Iduronic Acid/metabolism , Isoindoles/chemistry , Molecular Structure , Mucopolysaccharidosis I/metabolism , Mucopolysaccharidosis I/pathology , Organoselenium Compounds/chemistry , Structure-Activity Relationship
9.
Acta Histochem ; 123(2): 151678, 2021 Feb.
Article En | MEDLINE | ID: mdl-33434858

Mucopolysaccharidosis type I (MPS I) is a lysosomal storage disorder characterized by alpha-L-iduronidase (IDUA) deficiency, an enzyme responsible for glycosaminoglycan degradation. Musculoskeletal impairment is an important component of the morbidity related to the disease, as it has a major impact on patients' quality of life. To understand how this disease affects bone structure, morphological, biomechanical and histological analyses of femurs from 3- and 6-month-old wild type (Idua +/+) and MPS I knockout mice (Idua -/-) were performed. Femurs from 3-month-old Idua -/- mice were found to be smaller and less resistant to fracture when compared to their age matched controls. In addition, at this age, the femurs presented important alterations in articular cartilage, trabecular bone architecture, and deposition of type I and III collagen. At 6 months of age, femurs from Idua -/- mice were more resistant to fracture than those from Idua +/+. Our results suggest that the abnormalities observed in bone matrix and articular cartilage in 3-month-old Idua -/- animals caused bone tissue to be less flexible and more likely to fracture, whereas in 6-month-old Idua -/- group the ability to withstand more load before fracturing than wild type animals is possibly due to changes in the bone matrix.


Iduronidase/metabolism , Mucopolysaccharidosis I/metabolism , Mucopolysaccharidosis I/pathology , Animals , Biomechanical Phenomena/physiology , Collagen/metabolism , Disease Models, Animal , Female , Femur/enzymology , Femur/metabolism , Femur/pathology , Iduronidase/genetics , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mucopolysaccharidosis I/enzymology
10.
Mol Biol Rep ; 48(1): 363-370, 2021 Jan.
Article En | MEDLINE | ID: mdl-33319323

Mucopolysaccharidosis type I (MPS I) is a lysosomal storage disease caused by a mutation in the IDUA gene, which codes α-L-iduronidase (IDUA), a lysosomal hydrolase that degrades two glycosaminoglycans (GAGs): heparan sulfate (HS) and dermatan sulfate (DS). GAGs are macromolecules found mainly in the extracellular matrix and have important signaling and structural roles which are essential to the maintenance of cell and tissue physiology. Nondegraded GAGs accumulate in various cell types, which characterizes MPS I as a multisystemic progressive disease. Many tissues and vital organs have been described in MPS I models, but there is a lack of studies focused on their effects on the reproductive tract. Our previous studies indicated lower sperm production and morphological damage in the epididymis and accessory glands in male MPS I mice, despite their ability to copulate and to impregnate females. Our aim was to improve the testicular characterization of the MPS I model, with a specific focus on ultrastructural observation of the different cell types that compose the seminiferous tubules and interstitium. We investigated the testicular morphology of 6-month-old male C57BL/6 wild-type (Idua+/+) and MPS I (Idua-/-) mice. We found vacuolated cells widely present in the interstitium and important signs of damage in myoid, Sertoli and Leydig cells. In the cytoplasmic region of Sertoli cells, we found an increased number of vesicles with substrates under digestion and a decreased number of electron-dense vesicles similar to lysosomes, suggesting an impaired flux of substrate degradation. Conclusions: Idua exerts an important role in the morphological maintenance of the seminiferous tubules and the testicular interstitium, which may influence the quality of spermatogenesis, having a greater effect with the progression of the disease.


Glycosaminoglycans/genetics , Lysosomal Storage Diseases/genetics , Mucopolysaccharidosis I/genetics , Sertoli Cells/pathology , Animals , Disease Models, Animal , Disease Progression , Humans , Interstitial Cells of Cajal/metabolism , Interstitial Cells of Cajal/pathology , Leydig Cells/metabolism , Leydig Cells/pathology , Lysosomal Storage Diseases/metabolism , Lysosomal Storage Diseases/pathology , Male , Mice , Mucopolysaccharidosis I/metabolism , Mucopolysaccharidosis I/pathology , Mutation/genetics , Sertoli Cells/metabolism , Spermatozoa/metabolism , Spermatozoa/pathology
11.
Cells ; 9(12)2020 12 03.
Article En | MEDLINE | ID: mdl-33287330

Background: Mucopolysaccharidosis type I-Hurler (MPS1-H) is a severe genetic lysosomal storage disorder due to loss-of-function mutations in the IDUA gene. The subsequent complete deficiency of alpha l-iduronidase enzyme is directly responsible of a progressive accumulation of glycosaminoglycans (GAG) in lysosomes which affects the functions of many tissues. Consequently, MPS1 is characterized by systemic symptoms (multiorgan dysfunction) including respiratory and cardiac dysfunctions, skeletal abnormalities and early fatal neurodegeneration. Methods: To understand mechanisms underlying MPS1 neuropathology, we generated induced pluripotent stem cells (iPSC) from a MPS1-H patient with loss-of-function mutations in both IDUA alleles. To avoid variability due to different genetic background of iPSC, we established an isogenic control iPSC line by rescuing IDUA expression by a lentivectoral approach. Results: Marked differences between MPS1-H and IDUA-corrected isogenic controls were observed upon neural differentiation. A scratch assay revealed a strong migration defect of MPS1-H cells. Also, there was a massive impact of IDUA deficiency on gene expression (340 genes with an FDR <0.05). Conclusions: Our results demonstrate a hitherto unknown connection between lysosomal degradation, gene expression and neural motility, which might account at least in part for the phenotype of MPS1-H patients.


Cell Movement/genetics , Induced Pluripotent Stem Cells/metabolism , Mucopolysaccharidosis I/metabolism , Neurons/metabolism , Cell Differentiation/genetics , Cells, Cultured , Gene Expression/genetics , Glycosaminoglycans/genetics , Glycosaminoglycans/metabolism , Humans , Iduronidase/genetics , Iduronidase/metabolism , Lysosomes/genetics , Lysosomes/metabolism , Mucopolysaccharidosis I/genetics , Mutation/genetics , Phenotype
12.
Mol Genet Metab ; 131(1-2): 197-205, 2020.
Article En | MEDLINE | ID: mdl-32739280

The cause of neurodegeneration in MPS mouse models is the focus of much debate and what the underlying cause of disease pathology in MPS mice is. The timing of development of pathology and when this can be reversed or impacted is the key to developing suitable therapies in MPS. This study is the first of its kind to correlate the biochemical changes with the functional outcome as assessed using non-invasive behaviour testing across multiple mucopolysaccharidosis (MPS) mouse models. In the MPS brain, the primary lysosomal enzyme dysfunction leads to accumulation of primary glycosaminoglycans (GAGs) with gangliosides (GM2 and GM3) being the major secondary storage products. With a focus on the neuropathology, a time course experiment was conducted in MPS I, MPS IIIA, MPS VII (severe and attenuated models) in order to understand the relative timing and level of GAG and ganglioside accumulation and how this correlates to behaviour deficits. Time course analysis from 1 to 6 months of age was conducted on brain samples to assess primary GAG (uronic acid), ß-hexosaminidase enzyme activity and levels of GM2 and GM3 gangliosides. This was compared to a battery of non-invasive behaviour tests including open field, inverted grid, rotarod and water cross maze were assessed to determine effects on motor function, activity and learning ability. The results show that the GAG and ganglioside accumulation begins prior to the onset of detectable changes in learning ability and behaviour. Interestingly, the highest levels of GAG and ganglioside accumulation was observed in the MPS IIIA mouse despite having 3% residual enzyme activity. Deficits in motor function were clearly observed in the severe Gusmps/mps, which were significantly delayed in the attenuated Gustm(L175F)Sly model despite their minimal increase in detectable enzyme activity. This suggests that genotype and residual enzyme activity are not indicative of severity of disease pathology in MPS disease and there exists a window when there are considerable storage products without detectable functional deficits which may allow an alteration to occur with therapy.


Brain/metabolism , Glucuronidase/genetics , Mucopolysaccharidosis III/metabolism , Mucopolysaccharidosis I/metabolism , Mucopolysaccharidosis VII/metabolism , Animals , Brain/pathology , Disease Models, Animal , G(M2) Ganglioside/genetics , G(M2) Ganglioside/metabolism , G(M3) Ganglioside/genetics , G(M3) Ganglioside/metabolism , Glycosaminoglycans/genetics , Glycosaminoglycans/metabolism , Heparitin Sulfate/metabolism , Humans , Male , Maze Learning/physiology , Mice , Mucopolysaccharidosis I/genetics , Mucopolysaccharidosis I/pathology , Mucopolysaccharidosis III/genetics , Mucopolysaccharidosis III/pathology , Mucopolysaccharidosis VII/genetics , Mucopolysaccharidosis VII/pathology
13.
Mol Ther ; 28(10): 2161-2176, 2020 10 07.
Article En | MEDLINE | ID: mdl-32610100

During brain maturation, cation-independent mannose-6-phosphate receptor (CI-MPR), a key transporter for lysosomal hydrolases, decreases significantly on the blood-brain barrier (BBB). Such a phenomenon leads to poor brain penetration of therapeutic enzymes and subsequent failure in reversing neurological complications in patients with neuropathic lysosomal storage diseases (nLSDs), such as Hurler syndrome (severe form of mucopolysaccharidosis type I [MPS I]). In this study, we discover that upregulation of microRNA-143 (miR-143) contributes to the decline of CI-MPR on the BBB during development. Gain- and loss-of-function studies showed that miR-143 inhibits CI-MPR expression and its transport function in human endothelial cells in vitro. Genetic removal of miR-143 in MPS I mice enhances CI-MPR expression and improves enzyme transport across the BBB, leading to brain metabolic correction, pathology normalization, and correction of neurological functional deficits 5 months after peripheral protein delivery at clinically relevant levels that derived from erythroid/megakaryocytic cells via hematopoietic stem cell-mediated gene therapy, when otherwise no improvement was observed in MPS I mice at a parallel setting. These studies not only uncover a novel role of miR-143 as an important modulator for the developmental decline of CI-MPR on the BBB, but they also demonstrate the functional significance of depleting miR-143 for "rescuing" BBB-anchored CI-MPR on advancing CNS treatment for nLSDs.


Blood-Brain Barrier/metabolism , Central Nervous System/metabolism , Lysosomes/metabolism , MicroRNAs/genetics , Mucopolysaccharidosis I/genetics , Mucopolysaccharidosis I/metabolism , Animals , Central Nervous System/pathology , Disease Models, Animal , Endothelial Cells/metabolism , Gene Expression Regulation , Gene Transfer Techniques , Genetic Therapy , Hematopoietic Stem Cells/metabolism , Humans , Mice , Mucopolysaccharidosis I/therapy , Protein Transport , RNA Interference , Transduction, Genetic
14.
Lipids ; 55(6): 627-637, 2020 11.
Article En | MEDLINE | ID: mdl-32537944

Mucopolysaccharidosis type I (MPS I) is a lysosomal disease with progressive central nervous system involvement. This study examined the lipid, cholesterol, and myelin basic protein composition of white matter in the corpus callosum of MPS I mice. We studied 50 week-old, male MPS I mice and littermate, heterozygote controls (n = 12 per group). Male MPS I mice showed lower phosphatidylcholine and ether-linked phosphatidylcholine quantities than controls (p < 0.05). Twenty-two phospholipid or ceramide species showed significant differences in percent of total. Regarding specific lipid species, MPS I mice exhibited lower quantities of sphingomyelin 18:1, phosphatidylserine 38:3, and hexosylceramide d18:1(22:1) mH2 O than controls. Principal components analyses of polar, ceramide, and hexosylceramide lipids, respectively, showed some separation of MPS I and control mice. We found no significant differences in myelin gene expression, myelin basic protein, or total cholesterol in the MPS I mice versus heterozygous controls. There was a trend toward lower proteolipid protein-1 levels in MPS I mice (p = 0.06). MPS I mice show subtle changes in white matter composition, with an unknown impact on pathogenesis in this model.


Corpus Callosum/chemistry , Lipids/analysis , Lipids/chemistry , Mucopolysaccharidosis I/pathology , Myelin Sheath/chemistry , Animals , Case-Control Studies , Cholesterol/analysis , Cholesterol/metabolism , Corpus Callosum/pathology , Female , Gene Expression , Male , Mice, Inbred C57BL , Mice, Knockout , Mucopolysaccharidosis I/metabolism , Myelin Basic Protein/analysis , Myelin Basic Protein/metabolism , Myelin Sheath/genetics , Myelin Sheath/pathology
15.
Mol Ther ; 28(6): 1442-1454, 2020 06 03.
Article En | MEDLINE | ID: mdl-32278382

Our previous study delivered zinc finger nucleases to treat mice with mucopolysaccharidosis type I (MPS I), resulting in a phase I/II clinical trial (ClinicalTrials.gov: NCT02702115). However, in the clinical trial, the efficacy needs to be improved due to the low transgene expression level. To this end, we designed a proprietary system (PS) gene editing approach with CRISPR to insert a promoterless α-l-iduronidase (IDUA) cDNA sequence into the albumin locus of hepatocytes. In this study, adeno-associated virus 8 (AAV8) vectors delivering the PS gene editing system were injected into neonatal and adult MPS I mice. IDUA enzyme activity in the brain significantly increased, while storage levels were normalized. Neurobehavioral tests showed that treated mice had better memory and learning ability. Also, histological analysis showed efficacy reflected by the absence of foam cells in the liver and vacuolation in neuronal cells. No vector-associated toxicity or increased tumorigenesis risk was observed. Moreover, no off-target effects were detected through the unbiased genome-wide unbiased identification of double-stranded breaks enabled by sequencing (GUIDE-seq) analysis. In summary, these results showed the safety and efficacy of the PS in treating MPS I and paved the way for clinical studies. Additionally, as a therapeutic platform, the PS has the potential to treat other lysosomal diseases.


Gene Editing/methods , Gene Expression , Genetic Therapy , Iduronidase/genetics , Mucopolysaccharidosis I/genetics , Mucopolysaccharidosis I/therapy , Transgenes , Animals , Brain/metabolism , CRISPR-Cas Systems , Clustered Regularly Interspaced Short Palindromic Repeats , Dependovirus/genetics , Disease Models, Animal , Enzyme Activation , Gene Dosage , Gene Order , Gene Transfer Techniques , Genetic Therapy/adverse effects , Genetic Therapy/methods , Genetic Vectors/genetics , Humans , Liver/metabolism , Liver/pathology , Mice , Mucopolysaccharidosis I/metabolism , RNA, Guide, Kinetoplastida , Treatment Outcome
16.
J Mol Histol ; 51(2): 137-145, 2020 Apr.
Article En | MEDLINE | ID: mdl-32162173

Mucopolysaccharidosis type I (MPS I) is a genetic disease caused by a deficiency of the lysosomal hydrolase α-L-iduronidase (IDUA). IDUA degrades two types of glycosaminoglycans (GAGs): heparan and dermatan sulfates, important components of extracellular matrix, with signaling and structural functions. The accumulation of GAGs results in progressive physiological impairments in a variety of tissues, making MPS I a complex and multisystemic disease. Due the advent of therapeutic strategies which have increased patients' life expectancy, our group have been investigating the effect of IDUA deficiency on the reproductive system. In the present study, we aimed to characterize some of the accessory glands of the male reproductive tract in an MPS I mouse model. We used 6-month-old Idua+/+ and Idua-/- male mice to evaluate the histology of the seminal vesicles and prostate. Interstitial deposits of GAGs and collagen fibers were also observed. Seminal vesicles were smaller in the Idua-/- group, regardless of the normal staining pattern of the epithelial cells, marked with antiandrogen receptor. The prostate of Idua-/- mice presented necrotic acini and increased deposition of collagen fibers in the interstitium. All glands presented evident deposits of GAGs in the extracellular matrix, especially inside vacuolated interstitial cells. We concluded that, at this stage of the disease, the prostate is the most damaged accessory gland and may therefore, be the first to manifest functional impairments during disease progression.


Genitalia, Male/pathology , Mucopolysaccharidosis I/pathology , Animals , Biomarkers , Biopsy , Disease Models, Animal , Genitalia, Male/metabolism , Iduronidase/deficiency , Immunohistochemistry , Male , Mice , Mice, Knockout , Mucopolysaccharidosis I/etiology , Mucopolysaccharidosis I/metabolism , Prostate/metabolism , Prostate/pathology , Seminal Vesicles/metabolism , Seminal Vesicles/pathology
17.
Int J Mol Sci ; 21(4)2020 Feb 20.
Article En | MEDLINE | ID: mdl-32093427

Mucopolysaccharidosis type I (MPS I) is caused by genetic deficiency of α-l-iduronidase and impairment of lysosomal catabolism of heparan sulfate and dermatan sulfate. In the brain, these substrates accumulate in the lysosomes of neurons and glial cells, leading to neuroinflammation and neurodegeneration. Their storage also affects lysosomal homeostasis-inducing activity of several lysosomal proteases including cathepsin B (CATB). In the central nervous system, increased CATB activity has been associated with the deposition of amyloid plaques due to an alternative pro-amyloidogenic processing of the amyloid precursor protein (APP), suggesting a potential role of this enzyme in the neuropathology of MPS I. In this study, we report elevated levels of protein expression and activity of CATB in cortex tissues of 6-month-old MPS I (Idua -/- mice. Besides, increased CATB leakage from lysosomes to the cytoplasm of Idua -/- cortical pyramidal neurons was indicative of damaged lysosomal membranes. The increased CATB activity coincided with an elevated level of the 16-kDa C-terminal APP fragment, which together with unchanged levels of ß-secretase 1 was suggestive for the role of this enzyme in the amyloidogenic APP processing. Neuronal accumulation of Thioflavin-S-positive misfolded protein aggregates and drastically increased levels of neuroinflammatory glial fibrillary acidic protein (GFAP)-positive astrocytes and CD11b-positive activated microglia were observed in Idua -/- cortex by confocal fluorescent microscopy. Together, our results point to the existence of a novel CATB-associated alternative amyloidogenic pathway in MPS I brain induced by lysosomal storage and potentially leading to neurodegeneration.


Amyloid beta-Protein Precursor/metabolism , Cathepsin B/metabolism , Cerebral Cortex/metabolism , Mucopolysaccharidosis I/metabolism , Pyramidal Cells/metabolism , Amyloid beta-Protein Precursor/genetics , Animals , Astrocytes/metabolism , Astrocytes/pathology , Cathepsin B/genetics , Cerebral Cortex/pathology , Glial Fibrillary Acidic Protein/genetics , Glial Fibrillary Acidic Protein/metabolism , Lysosomes/metabolism , Lysosomes/pathology , Mice , Mice, Knockout , Mucopolysaccharidosis I/genetics , Mucopolysaccharidosis I/pathology , Pyramidal Cells/pathology
18.
Reprod Fertil Dev ; 32(3): 304-312, 2020 Feb.
Article En | MEDLINE | ID: mdl-31679559

Mucopolysaccharidosis type I (MPS I) is a lysosomal storage disease caused by a deficiency of the lysosomal hydrolase, α-L-iduronidase (IDUA). IDUA degrades heparan and dermatan sulfates, two types of glycosaminoglycan (GAG), important signalling and structural molecules of the extracellular matrix. Because many cell types store GAGs, MPS I has been investigated in human and animal models. Enzyme replacement therapy is available for MPS I patients and has improved their life expectancy, allowing them to achieve reproductive age. The aim of this study was to evaluate epididymal and sperm morphology and function in a murine model of MPS I. We used C57BL Idua+/+ and Idua-/- adult male mice (6 months old) to investigate epididymal morphology, sperm ultrastructure, GAG characterisation and mating competence. Epithelial GAG storage, especially in the cauda epididymidis, was seen in Idua-/- mice. Regardless of the morphologic change and GAG storage found in the cauda epididymis, sperm morphology and motility were normal, similar to wild types. In the interstitium, vacuolated cells were found in addition to deposits of GAGs. Mating was not impaired in Idua-/- males and litter sizes were similar between groups. At the time point of the disease evaluated, the deficiency in IDUA affected the morphology of the epididymis in male Idua-/- mice, whereas sperm appearance and motility and the male's capacity to mate and impregnate females were preserved.


Collagen/metabolism , Epididymis/metabolism , Glycosaminoglycans/metabolism , Mucopolysaccharidosis I/metabolism , Sperm Motility , Spermatozoa/metabolism , Animals , Cell Survival , Disease Models, Animal , Epididymis/ultrastructure , Fertilization , Iduronidase/deficiency , Iduronidase/genetics , Male , Mice, Inbred C57BL , Mice, Knockout , Mucopolysaccharidosis I/genetics , Mucopolysaccharidosis I/pathology , Spermatozoa/ultrastructure
19.
World J Pediatr Congenit Heart Surg ; 11(4): NP22-NP24, 2020 Jul.
Article En | MEDLINE | ID: mdl-28421916

Mucopolysaccharidosis type I is a genetic disorder with impaired glycosaminoglycan degradation. Cardiac pathologic involvement in this subset of patients is predominantly valvular heart disease. Valvular heart disease seen in these patients will most likely require surgical intervention in their lifetime. Only a limited amount of reports are dedicated to the cardiac surgical management of mucopolysaccharidoses. We present the case of a 32-year-old female with Hurler-Scheie syndrome who required multiple valve replacements due to progression of valvular dysfunction and decline in the quality of life. Multidisciplinary evaluation and discussion early are crucial for quality of life optimization in this cohort of patients.


Heart Valve Diseases/etiology , Heart Valve Prosthesis Implantation/methods , Mitral Valve/surgery , Mucopolysaccharidosis I/complications , Adult , Echocardiography , Female , Heart Valve Diseases/diagnosis , Heart Valve Diseases/surgery , Humans , Mitral Valve/diagnostic imaging , Mucopolysaccharidosis I/diagnosis , Mucopolysaccharidosis I/metabolism
20.
Sci Rep ; 9(1): 14105, 2019 Oct 01.
Article En | MEDLINE | ID: mdl-31575939

Allogeneic hematopoietic cell transplantation (HCT) benefits children with Hurler syndrome (MPS-IH). However, survivors remain burdened by substantial MPS-IH related residual disease. We studied the feasibility, safety and biochemical impact of augmentative recombinant intravenous enzyme replacement therapy (IV-ERT) post transplantation. Ten children with MPS-IH and ≥2 years from successful HCT underwent IV-ERT for 2 years' duration. Patients were monitored for anti-drug antibody (ADA) development, including inhibitory capacity and changes in urinary excretion of glycosaminoglycans (uGAG). Three patients demonstrated low-level ADA at baseline, though all children tolerated IV-ERT well. Eight patients developed ADA over the 2-year study, with 3 (38%) meeting criteria for an inhibitory ADA response. The aggregate cohort experienced a reduction in uGAG from baseline to study end, which was enhanced in children with low or no ADA response. Conversely, children with inhibitory ADA showed increase in uGAG over time. IV-ERT in previously transplanted children with MPS-IH appears safe and can reduce uGAG, although this is reversed by the presence of inhibitory ADA. These data show a biochemical change after initiation of post-HCT IV-ERT, but the occurrence of ADA and inhibitory antibodies are a concern and should be monitored in future efficacy trials. This trial was registered at www.clinicaltrials.gov , NCT01173016, 07/30/2010.


Iduronidase/therapeutic use , Mucopolysaccharidosis I/surgery , Neoplasm, Residual/drug therapy , Administration, Intravenous/methods , Adolescent , Antibodies/metabolism , Child , Child, Preschool , Enzyme Replacement Therapy/methods , Female , Glycosaminoglycans/metabolism , Glycosaminoglycans/urine , Hematopoietic Stem Cell Transplantation/adverse effects , Humans , Leukocytes/drug effects , Leukocytes/metabolism , Male , Mucopolysaccharidosis I/metabolism , Neoplasm, Residual/metabolism , Survivors , Transplants/drug effects
...