Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 44
Filter
1.
Mol Genet Metab ; 131(1-2): 197-205, 2020.
Article in English | MEDLINE | ID: mdl-32739280

ABSTRACT

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.


Subject(s)
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
2.
Bone ; 128: 115042, 2019 11.
Article in English | MEDLINE | ID: mdl-31442675

ABSTRACT

Mucopolysaccharidosis (MPS) VII is a lysosomal storage disorder characterized by deficient activity of ß-glucuronidase, leading to progressive accumulation of incompletely degraded heparan, dermatan, and chondroitin sulfate glycosaminoglycans (GAGs). Patients with MPS VII exhibit progressive skeletal deformity including kyphoscoliosis and joint dysplasia, which decrease quality of life and increase mortality. Previously, using the naturally-occurring canine model, we demonstrated that one of the earliest skeletal abnormalities to manifest in MPS VII is failed initiation of secondary ossification in vertebrae and long bones at the requisite postnatal developmental stage. The objective of this study was to obtain global insights into the molecular mechanisms underlying this failed initiation of secondary ossification. Epiphyseal tissue was isolated postmortem from the vertebrae of control and MPS VII-affected dogs at 9 and 14 days-of-age (n = 5 for each group). Differences in global gene expression across this developmental window for both cohorts were measured using whole-transcriptome sequencing (RNA-Seq). Principal Component Analysis revealed clustering of samples within each group, indicating clear effects of both age and disease state. At 9 days-of-age, 1375 genes were significantly differentially expressed between MPS VII and control, and by 14 days-of-age, this increased to 4719 genes. A targeted analysis focused on signaling pathways important in the regulation of endochondral ossification was performed, and a subset of gene expression differences were validated using qPCR. Osteoactivin (GPNMB) was the top upregulated gene in MPS VII at both ages. In control samples, temporal changes in gene expression from 9 to 14 days-of-age were consistent with chondrocyte maturation, cartilage resorption, and osteogenesis. In MPS VII samples, however, elements of key osteogenic pathways such as Wnt/ß-catenin and BMP signaling were not upregulated during this same developmental window suggesting that important bone formation pathways are not activated. In conclusion, this study represents an important step towards identifying therapeutic targets and biomarkers for bone disease in MPS VII patients during postnatal growth.


Subject(s)
Bone Diseases/metabolism , Mucopolysaccharidosis VII/metabolism , Osteogenesis/physiology , Animals , Cell Differentiation/genetics , Cell Differentiation/physiology , Chondrocytes/metabolism , Dogs , Matrix Metalloproteinases/genetics , Matrix Metalloproteinases/metabolism , Mutation, Missense/genetics , Osteogenesis/genetics , Principal Component Analysis , Exome Sequencing
3.
Clin Pharmacokinet ; 58(5): 673-683, 2019 05.
Article in English | MEDLINE | ID: mdl-30467742

ABSTRACT

INTRODUCTION: Mucopolysaccharidosis type VII (MPS VII, Sly Syndrome) is a progressive, debilitating, ultra-rare lysosomal storage disorder caused by the deficiency of ß-glucuronidase (GUS), an enzyme required for breakdown of glycosaminoglycans (GAGs). Vestronidase alfa, a recombinant human GUS, is an enzyme replacement therapy approved in the US and EU for the treatment of MPS VII. METHODS: The pharmacokinetics (PK) and pharmacodynamics (PD) of vestronidase alfa were evaluated in 23 adult and pediatric subjects with MPS VII enrolled in phase I-III clinical trials to optimize the clinical dosing regimen of vestronidase alfa. The serum concentration-time profiles were adequately described by a two-compartment population PK model incorporating subjects' body weight as the only significant covariate. RESULTS: Model-based simulations predicted a substantially decreased time duration of serum exposures exceeding the level of Kuptake (the in vitro determined vestronidase alfa concentration corresponding to 50% maximum rate of cellular uptake) for 4 or 8 mg/kg once every 4 weeks dosing, compared with 4 mg/kg once every other week (QOW) dosing by intravenous infusion, suggesting that given the same total monthly dose, the QOW dosing frequency should result in more efficient delivery to the GUS-deficient tissue cells, and therefore superior treatment efficacy. A standard inhibitory maximal effect model reasonably explained the observed pharmacological PD responses of reduction in urinary GAGs from pretreatment baseline, which appeared to have reached the plateau of maximal effect at the 4 mg/kg QOW dose. CONCLUSION: The modeling results, together with the clinical evidence of safety and efficacy, supported the recommended 4 mg/kg QOW dosing regimen of vestronidase alfa for pediatric and adult patients with MPS VII. CLINICAL TRIAL REGISTRATION: NCT01856218, NCT02418455, NCT02230566.


Subject(s)
Glucuronidase/pharmacokinetics , Models, Biological , Mucopolysaccharidosis VII/metabolism , Adolescent , Adult , Child , Child, Preschool , Computer Simulation , Cross-Over Studies , Enzyme Replacement Therapy , Female , Glucuronidase/administration & dosage , Glucuronidase/blood , Glycosaminoglycans/urine , Humans , Infant , Male , Mucopolysaccharidosis VII/blood , Mucopolysaccharidosis VII/drug therapy , Young Adult
4.
Sci Rep ; 8(1): 16644, 2018 11 09.
Article in English | MEDLINE | ID: mdl-30413728

ABSTRACT

Mucopolysaccharidosis type VII (MPS VII) is a lysosomal storage disease caused by deficient ß-glucuronidase (ß-gluc) activity. Significantly reduced ß-gluc activity leads to accumulation of glycosaminoglycans (GAGs) in many tissues, including the brain. Numerous combinations of mutations in GUSB (the gene that codes for ß-gluc) cause a range of neurological features that make disease prognosis and treatment challenging. Currently, there is little understanding of the molecular basis for MPS VII brain anomalies. To identify a neuronal phenotype that could be used to complement genetic analyses, we generated two iPSC clones derived from skin fibroblasts of an MPS VII patient. We found that MPS VII neurons exhibited reduced ß-gluc activity and showed previously established disease-associated phenotypes, including GAGs accumulation, expanded endocytic compartments, accumulation of lipofuscin granules, more autophagosomes, and altered lysosome function. Addition of recombinant ß-gluc to MPS VII neurons, which mimics enzyme replacement therapy, restored disease-associated phenotypes to levels similar to the healthy control. MPS VII neural cells cultured as 3D neurospheroids showed upregulated GFAP gene expression, which was associated with astrocyte reactivity, and downregulation of GABAergic neuron markers. Spontaneous calcium imaging analysis of MPS VII neurospheroids showed reduced neuronal activity and altered network connectivity in patient-derived neurospheroids compared to a healthy control. These results demonstrate the interplay between reduced ß-gluc activity, GAG accumulation and alterations in neuronal activity, and provide a human experimental model for elucidating the bases of MPS VII-associated cognitive defects.


Subject(s)
Glycosaminoglycans/metabolism , Induced Pluripotent Stem Cells/pathology , Lysosomes/pathology , Mucopolysaccharidosis VII/pathology , Neural Pathways , Neurons/pathology , Stem Cells/pathology , Case-Control Studies , Cell Differentiation , Cells, Cultured , Humans , Induced Pluripotent Stem Cells/metabolism , Lysosomes/metabolism , Mucopolysaccharidosis VII/metabolism , Neurons/metabolism , Stem Cells/metabolism
5.
Genet Med ; 19(9): 983-988, 2017 09.
Article in English | MEDLINE | ID: mdl-28383542

ABSTRACT

PURPOSE: The main purpose of the study was to provide quantitative data regarding survival and diagnostic delay. Mucopolysaccharidosis (MPS) type VII (OMIM 253220) is a progressive neurometabolic disorder caused by deficiency of the lysosomal enzyme ß-glucuronidase (GUS). Hard clinical end points have not been quantitatedMethods:We quantitatively analyzed published cases with MPS VII (N = 53/88 with sufficient data). Main outcome measures were onset of disease and survival. The role of biomarkers such as GUS residual enzyme activity and levels of storage material assessed as urinary excretion of glucosaminoglycans (GAG) as potential predictors of clinical outcomes were investigated. The analysis was conducted according to STROBE criteria. RESULTS: Median survival of the postnatally diagnosed population was up to 360 months . Median age of disease onset was the first day of life; median age at diagnosis was 11 months. Hydrops fetalis was frequent. Patients with residual GUS activity in fibroblasts more than 1.4% or urinary GAG excretion less than 602% of normal survived longer than patients with GUS enzyme activity below or GAG excretion above these thresholds. CONCLUSION: MPS VII has its disease onset prenatally. In the absence of a prenatal diagnosis, most cases are clinically apparent at birth. Our data corroborate a phenotype-biomarker association in MPS VII. The survival data characterize the natural history with important implications for therapeutic studies.Genet Med advance online publication 06 April 2017.


Subject(s)
Mucopolysaccharidosis VII/diagnosis , Mucopolysaccharidosis VII/epidemiology , Age Factors , Age of Onset , Biomarkers , Cross-Sectional Studies , Female , Genetic Testing , Glucuronidase/metabolism , Humans , Male , Mucopolysaccharidosis VII/etiology , Mucopolysaccharidosis VII/metabolism , Phenotype , Prenatal Diagnosis , Symptom Assessment
6.
Prenat Diagn ; 37(5): 435-439, 2017 May.
Article in English | MEDLINE | ID: mdl-28207930

ABSTRACT

OBJECTIVE: The aim of this study was to quantify glycosaminoglycans (GAGs) in amniotic fluid (AF) from an MPS VII fetus compared with age-matched fetuses obtained from normal pregnancies. METHOD: Disaccharides were measured by liquid chromatography tandem mass spectrometry, compared to age-matched controls. Enzyme assay was performed in AF supernatant or cultured amniocytes. GUSB was analyzed by next generation sequencing using Ion Torrent Personal Genome Machine with a customized panel. RESULTS: No activity of ß-glucuronidase was detected in fetal cells. The pregnancy was spontaneously terminated in the third trimester. Genetic studies identified a homozygous mutation of p.N379D (c.1135A > G) in the GUSB gene. Liquid chromatography tandem mass spectrometry showed that chondroitin sulfate, dermatan sulfate, heparan sulfate, and keratan sulfate levels were markedly increased in the MPS VII AF, compared to those in age-matched control AF (dermatan sulfate, heparan sulfate, and chondroitin-6-sulfate more than 10 × than age-matched controls; chondroitin-4-sulfate and keratan sulfate more than 3 times higher). CONCLUSION: This is the first report of specific GAG analysis in AF from an MPS VII fetus, indicating that GAG elevation in AF occurs by 21 weeks of gestation and could be an additional tool for prenatal diagnosis of MPS VII and potentially other MPS types. © 2017 John Wiley & Sons, Ltd.


Subject(s)
Amniotic Fluid/metabolism , Fetal Diseases/metabolism , Fetus/metabolism , Glycosaminoglycans/metabolism , Mucopolysaccharidosis VII/metabolism , Adult , Case-Control Studies , Female , Fetal Diseases/pathology , Fetus/pathology , Humans , Mucopolysaccharidosis VII/embryology , Mucopolysaccharidosis VII/pathology , Pregnancy , Up-Regulation
7.
Int J Mol Sci ; 17(12)2016 Nov 29.
Article in English | MEDLINE | ID: mdl-27916847

ABSTRACT

The prevalence of aortic root dilatation (ARD) in mucopolysaccharidosis (MPS) is not well documented. We investigated aortic root measurements in 34 MPS patients at the Children's Hospital of Orange County (CHOC). The diagnosis, treatment status, age, gender, height, weight and aortic root parameters (aortic valve annulus (AVA), sinuses of Valsalva (SoV), and sinotubular junction (STJ)) were extracted by retrospective chart review and echocardiographic measurements. Descriptive statistics, ANOVA, and paired post-hoc t-tests were used to summarize the aortic dimensions. Exact binomial 95% confidence intervals (CIs) were constructed for ARD, defined as a z-score greater than 2 at the SoV. The patient age ranged from 3.4-25.9 years (mean 13.3 ± 6.1), the height from 0.87-1.62 meters (mean 1.24 ± 0.21), and the weight from 14.1-84.5 kg (mean 34.4 ± 18.0). The prevalence of dilation at the AVA was 41% (14/34; 95% CI: 25%-59%); at the SoV was 35% (12/34; 95% CI: 20%-54%); and at the STJ was 30% (9/30; 95% CI: 15%-49%). The highest prevalence of ARD was in MPS IVa (87.5%). There was no significant difference between mean z-scores of MPS patients who received treatment with hematopoietic stem cell transplantation (HSCT) or enzyme replacement therapy (ERT) vs. untreated MPS patients at the AVA (z = 1.9 ± 2.5 vs. z = 1.5 ± 2.4; p = 0.62), SoV (z = 1.2 ± 1.6 vs. z = 1.3 ± 2.2; p = 0.79), or STJ (z = 1.0 ± 1.8 vs. z = 1.2 ± 1.6; p = 0.83). The prevalence of ARD was 35% in our cohort of MPS I-VII patients. Thus, we recommend screening for ARD on a routine basis in this patient population.


Subject(s)
Aortic Diseases/diagnosis , Dilatation, Pathologic/diagnosis , Mucopolysaccharidosis III/pathology , Mucopolysaccharidosis II/pathology , Mucopolysaccharidosis IV/pathology , Mucopolysaccharidosis I/pathology , Mucopolysaccharidosis VII/pathology , Mucopolysaccharidosis VI/pathology , Adolescent , Adult , Aorta/metabolism , Aorta/pathology , Aortic Diseases/drug therapy , Aortic Diseases/therapy , Child , Dilatation, Pathologic/drug therapy , Dilatation, Pathologic/therapy , Female , Humans , Male , Mucopolysaccharidosis I/metabolism , Mucopolysaccharidosis II/metabolism , Mucopolysaccharidosis III/metabolism , Mucopolysaccharidosis IV/metabolism , Mucopolysaccharidosis VI/metabolism , Mucopolysaccharidosis VII/metabolism , Retrospective Studies , Young Adult
8.
Mol Genet Metab ; 118(1): 41-54, 2016 May.
Article in English | MEDLINE | ID: mdl-27053151

ABSTRACT

Mucopolysaccharidosis type VII (MPS VII) is a lysosomal storage disease caused by the deficiency of ß-glucuronidase. In this study, we compared the changes relative to normal littermates in the proteome and transcriptome of the hippocampus in the C57Bl/6 mouse model of MPS VII, which has well-documented histopathological and neurodegenerative changes. A completely different set of significant changes between normal and MPS VII littermates were found in each assay. Nevertheless, the functional annotation terms generated by the two methods showed agreement in many of the processes, which also corresponded to known pathology associated with the disease. Additionally, assay-specific changes were found, which in the proteomic analysis included mitochondria, energy generation, and cytoskeletal differences in the mutant, while the transcriptome differences included immune, vesicular, and extracellular matrix changes. In addition, the transcriptomic changes in the mutant hippocampus were concordant with those in a MPS VII mouse caused by the same mutation but on a different background inbred strain.


Subject(s)
Gene Expression Profiling/methods , Hippocampus/metabolism , Mucopolysaccharidosis VII/genetics , Mucopolysaccharidosis VII/metabolism , Proteomics/methods , Animals , Gene Expression Regulation , Gene Regulatory Networks , Mice , Mice, Inbred C57BL , Molecular Sequence Annotation , Oligonucleotide Array Sequence Analysis/methods , Tandem Mass Spectrometry
9.
J Med Genet ; 53(6): 403-18, 2016 06.
Article in English | MEDLINE | ID: mdl-26908836

ABSTRACT

BACKGROUND: Mucopolysaccharidosis VII (MPS VII) is an ultra-rare disease characterised by the deficiency of ß-glucuronidase (GUS). Patients' phenotypes vary from severe forms with hydrops fetalis, skeletal dysplasia and mental retardation to milder forms with fewer manifestations and mild skeletal abnormalities. Accurate assessments on the frequency and clinical characteristics of the disease have been scarce. The aim of this study was to collect such data. METHODS: We have conducted a survey of physicians to document the medical history of patients with MPS VII. The survey included anonymous information on patient demographics, family history, mode of diagnosis, age of onset, signs and symptoms, severity, management, clinical features and natural progression of the disease. RESULTS: We collected information on 56 patients from 11 countries. Patients with MPS VII were classified based on their phenotype into three different groups: (1) neonatal non-immune hydrops fetalis (NIHF) (n=10), (2) Infantile or adolescent form with history of hydrops fetalis (n=13) and (3) Infantile or adolescent form without known hydrops fetalis (n=33). Thirteen patients with MPS VII who had the infantile form with history of hydrops fetalis and survived childhood, had a wide range of clinical manifestations from mild to severe. Five patients underwent bone marrow transplantation and one patient underwent enzyme replacement therapy with recombinant human GUS. CONCLUSIONS: MPS VII is a pan-ethnic inherited lysosomal storage disease with considerable phenotypical heterogeneity. Most patients have short stature, skeletal dysplasia, hepatosplenomegaly, hernias, cardiac involvement, pulmonary insufficiency and cognitive impairment. In these respects it resembles MPS I and MPS II. In MPS VII, however, one unique and distinguishing clinical feature is the unexpectedly high proportion of patients (41%) that had a history of NIHF. Presence of NIHF does not, by itself, predict the eventual severity of the clinical course, if the patient survives infancy.


Subject(s)
Mucopolysaccharidosis VII/pathology , Adolescent , Adult , Child , Child, Preschool , Female , Glucuronidase/metabolism , Humans , Infant , Lysosomal Storage Diseases/metabolism , Lysosomal Storage Diseases/pathology , Male , Mucopolysaccharidosis VII/metabolism , Phenotype , Surveys and Questionnaires , Young Adult
10.
Mol Ther ; 24(2): 206-216, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26447927

ABSTRACT

Mucopolysaccharidosis VII (MPS VII) is a lysosomal storage disease arising from mutations in ß-d-glucuronidase (GUSB), which results in glycosaminoglycan (GAG) accumulation and a variety of clinical manifestations including neurological disease. Herein, MPS VII dogs were injected intravenously (i.v.) and/or intrathecally (i.t.) via the cisterna magna with AAV9 or AAVrh10 vectors carrying the canine GUSB cDNA. Although i.v. injection alone at 3 days of age resulted in normal cerebrospinal fluid (CSF) GUSB activity, brain tissue homogenates had only ~1 to 6% normal GUSB activity and continued to have elevated GAG storage. In contrast, i.t. injection at 3 weeks of age resulted in CSF GUSB activity 44-fold normal while brain tissue homogenates had >100% normal GUSB activity and reduced GAGs compared with untreated dogs. Markers for secondary storage and inflammation were eliminated in i.t.-treated dogs and reduced in i.v.-treated dogs compared with untreated dogs. Given that i.t.-treated dogs expressed higher levels of GUSB in the CNS tissues compared to those treated i.v., we conclude that i.t. injection of AAV9 or AAVrh10 vectors is more effective than i.v. injection alone in the large animal model of MPS VII.


Subject(s)
Central Nervous System Diseases/therapy , Genetic Therapy/methods , Glucuronidase/genetics , Mucopolysaccharidosis VII/therapy , Animals , Animals, Newborn , Central Nervous System Diseases/genetics , Central Nervous System Diseases/metabolism , Dependovirus/genetics , Disease Models, Animal , Dogs , Genetic Vectors/administration & dosage , Glucuronidase/cerebrospinal fluid , Glycosaminoglycans/metabolism , Injections, Intravenous , Injections, Spinal , Male , Mucopolysaccharidosis VII/complications , Mucopolysaccharidosis VII/genetics , Mucopolysaccharidosis VII/metabolism
11.
J Neuropathol Exp Neurol ; 73(1): 39-49, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24335527

ABSTRACT

High-resolution microscopic magnetic resonance imaging (µMRI) and diffusion tensor imaging (DTI) were performed to characterize brain structural abnormalities in a mouse model of mucopolysaccharidosis type VII (MPS VII). Microscopic magnetic resonance imaging demonstrated a decrease in the volume of anterior commissure and corpus callosum and a slight increase in the volume of the hippocampus in MPS VII versus wild-type mice. Diffusion tensor imaging indices were analyzed in gray and white matter. In vivo and ex vivo DTI demonstrated significantly reduced fractional anisotropy in the anterior commissure, corpus callosum, external capsule, and hippocampus in MPS VII versus control brains. Significantly increased mean diffusivity was also found in the anterior commissure and corpus callosum from ex vivo DTI. Significantly reduced linear anisotropy was observed from the hippocampus from in vivo DTI, whereas significantly decreased planar anisotropy and spherical anisotropy were observed in the external capsule from only ex vivo DTI. There were corresponding morphologic differences in the brains of MPS VII mice by hematoxylin and eosin staining. Luxol fast blue staining demonstrated less intense staining of the corpus callosum and external capsule; myelin abnormalities in the corpus callosum were also demonstrated quantitatively in toluidine blue-stained sections and confirmed by electron microscopy. These results demonstrate the potential for µMRI and DTI for quantitative assessment of brain pathology in murine models of brain diseases.


Subject(s)
Brain/metabolism , Brain/pathology , Diffusion Tensor Imaging/methods , Disease Models, Animal , Mucopolysaccharidosis VII/metabolism , Mucopolysaccharidosis VII/pathology , Animals , Brain/ultrastructure , Mice , Mice, Inbred C3H , Microscopy, Electron
12.
Osteoarthritis Cartilage ; 21(12): 1813-23, 2013 Dec.
Article in English | MEDLINE | ID: mdl-23954699

ABSTRACT

BACKGROUND: Mucopolysaccharidoses (MPSs) are rare genetic diseases caused by a deficient activity of one of the lysosomal enzymes involved in the glycosaminoglycan (GAG) breakdown pathway. These metabolic blocks lead to the accumulation of GAGs in various organs and tissues, resulting in a multisystemic clinical picture. The pathological GAG accumulation begins a cascade of interrelated responses: metabolic, inflammatory and immunological with systemic effects. Metabolic inflammation, secondary to GAG storage, is a significant cause of osteoarticular symptoms in MPS disorders. OBJECTIVE AND METHOD: The aim of this review is to present recent progress in the understanding of the role of inflammatory and immune processes in the pathophysiology of osteoarticular symptoms in MPS disorders and potential therapeutic interventions based on published reports in MPS patients and studies in animal models. RESULTS AND CONCLUSIONS: The immune and skeletal systems have a number of shared regulatory molecules and many relationships between bone disorders and aberrant immune responses in MPS can be explained by osteoimmunology. The treatment options currently available are not sufficiently effective in the prevention, inhibition and treatment of osteoarticular symptoms in MPS disease. A lot can be learnt from interactions between skeletal and immune systems in autoimmune diseases such as rheumatoid arthritis (RA) and similarities between RA and MPS point to the possibility of using the experience with RA in the treatment of MPS in the future. The use of different anti-inflammatory drugs requires further study, but it seems to be an important direction for new therapeutic options for MPS patients.


Subject(s)
Bone Diseases/immunology , Joint Diseases/immunology , Mucopolysaccharidoses/immunology , Bone Diseases/etiology , Bone Diseases/metabolism , Cartilage, Articular/immunology , Cartilage, Articular/metabolism , Dysostoses/etiology , Dysostoses/immunology , Dysostoses/metabolism , Glycosaminoglycans/immunology , Glycosaminoglycans/metabolism , Humans , Joint Diseases/etiology , Joint Diseases/metabolism , Mucopolysaccharidoses/complications , Mucopolysaccharidoses/metabolism , Mucopolysaccharidosis I/complications , Mucopolysaccharidosis I/immunology , Mucopolysaccharidosis I/metabolism , Mucopolysaccharidosis II/complications , Mucopolysaccharidosis II/immunology , Mucopolysaccharidosis II/metabolism , Mucopolysaccharidosis VI/complications , Mucopolysaccharidosis VI/immunology , Mucopolysaccharidosis VI/metabolism , Mucopolysaccharidosis VII/complications , Mucopolysaccharidosis VII/immunology , Mucopolysaccharidosis VII/metabolism , Synovitis/etiology , Synovitis/immunology , Synovitis/metabolism
13.
Mol Genet Metab ; 110(3): 319-28, 2013 Nov.
Article in English | MEDLINE | ID: mdl-23856419

ABSTRACT

Mucopolysaccharidosis VII (MPS VII) is due to the deficient activity of ß-glucuronidase (GUSB) and results in the accumulation of glycosaminoglycans (GAGs) in lysosomes and multisystemic disease with cardiovascular manifestations. The goal here was to determine the pathogenesis of mitral valve (MV) disease in MPS VII dogs. Untreated MPS VII dogs had a marked reduction in the histochemical signal for structurally-intact collagen in the MV at 6 months of age, when mitral regurgitation had developed. Electron microscopy demonstrated that collagen fibrils were of normal diameter, but failed to align into large parallel arrays. mRNA analysis demonstrated a modest reduction in the expression of genes that encode collagen or collagen-associated proteins such as the proteoglycan decorin which helps collagen fibrils assemble, and a marked increase for genes that encode proteases such as cathepsins. Indeed, enzyme activity for cathepsin B (CtsB) was 19-fold normal. MPS VII dogs that received neonatal intravenous injection of a gamma retroviral vector had an improved signal for structurally-intact collagen, and reduced CtsB activity relative to that seen in untreated MPS VII dogs. We conclude that MR in untreated MPS VII dogs was likely due to abnormalities in MV collagen structure. This could be due to upregulation of enzymes that degrade collagen or collagen-associated proteins, to the accumulation of GAGs that compete with proteoglycans such as decorin for binding to collagen, or to other causes. Further delineation of the etiology of abnormal collagen structure may lead to treatments that improve biomechanical properties of the MV and other tissues.


Subject(s)
Heart Valve Diseases/etiology , Mitral Valve/pathology , Mucopolysaccharidosis VII/complications , Animals , Chordae Tendineae/metabolism , Collagen/metabolism , Dogs , Extracellular Matrix/genetics , Extracellular Matrix/metabolism , Female , Glycosaminoglycans/metabolism , Heart Valve Diseases/metabolism , Male , Mitral Valve/metabolism , Mucopolysaccharidosis VII/metabolism , Peptide Hydrolases/genetics , Peptide Hydrolases/metabolism , Signal Transduction
14.
PLoS One ; 7(3): e32419, 2012.
Article in English | MEDLINE | ID: mdl-22403656

ABSTRACT

The characteristic neurological feature of many neurogenetic diseases is intellectual disability. Although specific neuropathological features have been described, the mechanisms by which specific gene defects lead to cognitive impairment remain obscure. To gain insight into abnormal functions occurring secondary to a single gene defect, whole transcriptome analysis was used to identify molecular and cellular pathways that are dysregulated in the brain in a mouse model of a lysosomal storage disorder (LSD) (mucopolysaccharidosis [MPS] VII). We assayed multiple anatomical regions separately, in a large cohort of normal and diseased mice, which greatly increased the number of significant changes that could be detected compared to past studies in LSD models. We found that patterns of aberrant gene expression and involvement of multiple molecular and cellular systems varied significantly between brain regions. A number of changes revealed unexpected system and process alterations, such as up-regulation of the immune system with few inflammatory changes (a significant difference from the closely related MPS IIIb model), down-regulation of major oligodendrocyte genes even though white matter changes are not a feature histopathologically, and a plethora of developmental gene changes. The involvement of multiple neural systems indicates that the mechanisms of neuropathology in this type of disease are much broader than previously appreciated. In addition, the variation in gene dysregulation between brain regions indicates that different neuropathologic mechanisms may predominate within different regions of a diseased brain caused by a single gene mutation.


Subject(s)
Brain/metabolism , Brain/pathology , Mucopolysaccharidosis VII/genetics , Mucopolysaccharidosis VII/pathology , Transcriptome , Animals , Brain/cytology , Brain/immunology , Cell Adhesion/genetics , Cell Cycle/genetics , Cell Nucleus/genetics , Circadian Rhythm/genetics , Extracellular Matrix/metabolism , Female , Gliosis/genetics , Gliosis/metabolism , Gliosis/pathology , Ion Channels/metabolism , Male , Mice , Microglia/metabolism , Microglia/pathology , Mucopolysaccharidosis VII/immunology , Mucopolysaccharidosis VII/metabolism , Myelin Sheath/physiology , Neurons/metabolism , Neurons/pathology , Olfactory Bulb/pathology , Signal Transduction/genetics
15.
Invest Ophthalmol Vis Sci ; 52(9): 6720-8, 2011 Aug 24.
Article in English | MEDLINE | ID: mdl-21743015

ABSTRACT

PURPOSE: Deficiencies in enzymes involved in proteoglycan (PG) turnover underlie a number of rare mucopolysaccharidoses (MPS), investigations of which can considerably aid understanding of the roles of PGs in corneal matrix biology. Here, the authors analyze novel pathologic changes in MPS VII (Sly syndrome) to determine the nature of PG-collagen associations in stromal ultrastructure. METHODS: Transmission electron microscopy and electron tomography were used to investigate PG-collagen architectures and interactions in a cornea obtained at keratoplasty from a 22-year-old man with MPS VII, which was caused by a compound heterozygous mutation in the GUSB gene. RESULTS: Transmission electron microscopy showed atypical morphology of the epithelial basement membrane and Bowman's layer in MPS VII. Keratocytes were packed with cytoplasmic vacuoles containing abnormal glycosaminoglycan (GAG) material, and collagen fibrils were thinner than in normal cornea and varied considerably throughout anterior (14-32 nm), mid (13-42 nm), and posterior (17-39 nm) regions of the MPS VII stroma. PGs viewed in three dimensions were striking in appearance in that they were significantly larger than PGs in normal cornea and formed highly extended linkages with multiple collagen fibrils. CONCLUSIONS: Cellular changes in the MPS VII cornea resemble those in other MPS. However, the wide range of collagen fibril diameters throughout the stroma and the extensive matrix presence of supranormal-sized PG structures appear to be unique features of this disorder. The findings suggest that the accumulation of stromal chondroitin-, dermatan-, and heparan-sulfate glycosaminoglycans in the absence of ß-glucuronidase-mediated degradation can modulate collagen fibrillogenesis.


Subject(s)
Collagen/metabolism , Cornea/metabolism , Cornea/pathology , Mucopolysaccharidosis VII/metabolism , Mucopolysaccharidosis VII/pathology , Proteoglycans/metabolism , Basement Membrane/metabolism , Basement Membrane/pathology , Basement Membrane/ultrastructure , Cornea/ultrastructure , Corneal Transplantation , Electron Microscope Tomography , Extracellular Matrix/metabolism , Extracellular Matrix/pathology , Extracellular Matrix/ultrastructure , Humans , Male , Microscopy, Electron, Transmission , Mucopolysaccharidosis VII/surgery , Multiprotein Complexes/metabolism , Young Adult
16.
Adv Drug Deliv Rev ; 62(12): 1167-74, 2010 Sep 30.
Article in English | MEDLINE | ID: mdl-20920540

ABSTRACT

Mesenchymal stem cells (MSC) are a promising tool for cell therapy, either through direct contribution to the repair of bone, tendon and cartilage or as an adjunct therapy through protein production and immune mediation. They are an attractive vehicle for cellular therapies due to a variety of cell intrinsic and environmentally responsive properties. Following transplantation, MSC are capable of systemic migration, are not prone to tumor formation, and appear to tolerize the immune response across donor mismatch. These attributes combine to allow MSC to reside in many different tissue types without disrupting the local microenvironment and, in some cases, responding to the local environment with appropriate protein secretion. We describe work done by our group and others in using human MSC for the sustained in vivo production of supraphysiological levels of cytokines for the support of cotransplanted hematopoietic stem cells and enzymes that are deficient in animal models of lysosomal storage disorders such as MPSVII. In addition, the use of MSC engineered to secrete protein products has been reviewed in several fields of tissue injury repair, including but not limited to revascularization after myocardial infarction, regeneration of intervertebral disc defects and spine therapy, repair of stroke, therapy for epilepsy, skeletal tissue repair, chondrogenesis/knee and joint repair, and neurodegenerative diseases. Genetically engineered MSC have thus proven safe and efficacious in numerous animal models of disease modification and tissue repair and are poised to be tested in human clinical trials. The potential for these interesting cells to secrete endogenous or transgene products in a sustained and long-term manner is highly promising and is discussed in the current review.


Subject(s)
Cytokines/metabolism , Drug Delivery Systems , Genetic Engineering , Immunologic Factors/metabolism , Mesenchymal Stem Cell Transplantation , Mesenchymal Stem Cells , Clinical Trials as Topic , Cytokines/genetics , Hematopoietic Stem Cell Transplantation , Humans , Immunologic Factors/genetics , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/physiology , Mucopolysaccharidosis VII/genetics , Mucopolysaccharidosis VII/metabolism , Mucopolysaccharidosis VII/therapy , Regeneration
17.
Proc Natl Acad Sci U S A ; 107(17): 7886-91, 2010 Apr 27.
Article in English | MEDLINE | ID: mdl-20385825

ABSTRACT

Most lysosomal storage diseases (LSDs) are life-threatening genetic diseases. The pathogenesis of these diseases is poorly understood. Induced pluripotent stem (iPS) cell technology offers new opportunities for both mechanistic studies and development of stem cell- based therapies. Here we report the generation of disease-specific iPS cells from mouse models of Fabry disease, globoid cell leukodystrophy (GLD), and mucopolysaccharidosis VII (MPSVII). These mouse model-derived iPS cells showed defects in disease-specific enzyme activities and significant accumulation of substrates for these enzymes. In the lineage-directed differentiation studies, Fabry-iPS and GLD-iPS cells were efficiently differentiated into disease-relevant cell types, such as cardiomyocytes and neural stem cells, which might be useful in mechanistic and therapeutic studies. Notably, MPSVII-iPS cells demonstrated a markedly impaired ability to form embryoid bodies (EBs) in vitro. MPSVII-EBs exibited elevated levels of hyaluronan and its receptor CD44, and markedly reduced expression levels of E-cadherin and cell-proliferating marker. Partial correction of enzyme deficiency in MSPVII-iPS cells led to improved EB formation and reversal of aberrant protein expression. These data indicate a potential mechanism for the partial lethality of MPSVII mice in utero, and suggest a possible abnormality of embryonic development in MPSVII patients. Thus, our study demonstrates the unique promise of iPS cells for studying the pathogenesis and treatment of LSDs.


Subject(s)
Cell Line , Cell- and Tissue-Based Therapy/methods , Fabry Disease/physiopathology , Induced Pluripotent Stem Cells/cytology , Leukodystrophy, Globoid Cell/physiopathology , Mucopolysaccharidosis VII/physiopathology , Alkaline Phosphatase , Animals , Blotting, Western , Cadherins/metabolism , Cell Differentiation/physiology , Chromatography, High Pressure Liquid , Chromatography, Thin Layer , Fabry Disease/metabolism , Fabry Disease/therapy , Hyaluronan Receptors/metabolism , Hyaluronic Acid/metabolism , Immunohistochemistry , Induced Pluripotent Stem Cells/metabolism , Leukodystrophy, Globoid Cell/metabolism , Leukodystrophy, Globoid Cell/therapy , Mice , Mucopolysaccharidosis VII/metabolism , Mucopolysaccharidosis VII/therapy , Reverse Transcriptase Polymerase Chain Reaction
18.
Fetal Pediatr Pathol ; 28(1): 1-8, 2009.
Article in English | MEDLINE | ID: mdl-19116811

ABSTRACT

Type VII mucopolysaccharidosis is a very rare recessive lysosomal storage disease. We diagnosed a type VII MPS in a case of severe fetal hydrops after pregnancy termination at 23 weeks of gestation. The diagnosis was suspected on histopathological examination by the presence of foam cells in many viscera and foamy placental Hofbauer cells. Enzyme assay on cultured amniotic cells showed a markedly deficient beta-glucuronidase activity, thus confirming the diagnosis. This report shows the importance of a precise necropsy diagnosis in nonimmune hydrops because of putative implications for genetic counseling and prenatal diagnosis in subsequent pregnancies.


Subject(s)
Fetus/pathology , Hydrops Fetalis/pathology , Mucopolysaccharidosis VII/pathology , Abortion, Induced , Adult , Female , Humans , Hydrops Fetalis/etiology , Hydrops Fetalis/metabolism , Male , Mucopolysaccharidosis VII/complications , Mucopolysaccharidosis VII/metabolism , Placenta/pathology , Pregnancy , Prenatal Diagnosis
19.
Mol Genet Metab ; 94(2): 178-89, 2008 Jun.
Article in English | MEDLINE | ID: mdl-18359257

ABSTRACT

We have tested an acidic oligopeptide-based targeting system for delivery of enzymes to tissues, especially bone and brain, in a murine mucopolysaccharidosis type VII (MPS VII) model. This strategy is based upon tagging a short peptide consisting of acidic amino acids (AAA) to N terminus of human beta-glucuronidase (GUS). The pharmacokinetics, biodistribution, and the pathological effect on MPS VII mouse after 12 weekly infusions were determined for recombinant human untagged and tagged GUS. The tagged GUS was taken up by MPS VII fibroblasts in a mannose 6-phosphate receptor-dependent manner. Intravenously injected AAA-tagged enzyme had five times more prolonged blood clearance compared with the untagged enzyme. The tagged enzyme was delivered effectively to bone, bone marrow, and brain in MPS VII mice and was effective in reversing the storage pathology. The storage in osteoblasts was cleared similarly with both enzyme types. However, cartilage showed a little response to any of the enzymes. The tagged enzyme reduced storage in cortical neurons, hippocampus, and glia cells. A highly sensitive method of tandem mass spectrometry on serum indicated that the concentration of serum dermatan sulfate and heparan sulfate in mice treated with the tagged enzyme decreased more than the untagged enzyme. These preclinical studies suggest that this AAA-based targeting system may enhance enzyme-replacement therapy.


Subject(s)
Amino Acids, Acidic/therapeutic use , Glucuronidase/metabolism , Mucopolysaccharidosis VII/drug therapy , Amino Acids, Acidic/genetics , Animals , CHO Cells , Cell Line , Cricetinae , Cricetulus , Disease Models, Animal , Gene Targeting , Glucuronidase/administration & dosage , Glucuronidase/genetics , Humans , Lysosomes/enzymology , Mice , Mice, Transgenic , Mucopolysaccharidosis VII/enzymology , Mucopolysaccharidosis VII/genetics , Mucopolysaccharidosis VII/metabolism , Peptides/genetics , Recombinant Fusion Proteins/administration & dosage , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Recombinant Fusion Proteins/therapeutic use
20.
J Inherit Metab Dis ; 30(2): 227-38, 2007 Apr.
Article in English | MEDLINE | ID: mdl-17308887

ABSTRACT

Mucopolysaccharidosis type VII (MPS VII) is a lysosomal storage disease caused by beta-glucuronidase (GUSB) deficiency. This disease exhibits a broad spectrum of clinical signs including skeletal dysplasia, retinal degeneration, cognitive deficits and hearing impairment. Sustained, high-level expression of GUSB significantly improves the clinical course of the disease in the murine model of MPS VII. Low levels of enzyme expression (1-5% of normal) can significantly reduce the biochemical and histopathological manifestations of MPS VII. However, it has not been clear from previous studies whether persistent, low levels of circulating GUSB lead to significant improvements in the clinical presentation of this disease. We generated a rAAV2 vector that mediates persistent, low-level GUSB expression in the liver. Liver and serum levels of GUSB were maintained at approximately 5% and approximately 2.5% of normal, respectively, while other tissue ranged from background levels to 0.9%. This level of activity significantly reduced the secondary elevations of alpha-galactosidase and the levels of glycosaminoglycans in multiple tissues. Interestingly, this level of GUSB was also sufficient to reduce lysosomal storage in neurons in the brain. Although there were small but statistically significant improvements in retinal function, auditory function, skeletal dysplasia, and reproduction in rAAV-treated MPS VII mice, the clinical deficits were still profound and there was no improvement in lifespan. These data suggest that circulating levels of GUSB greater than 2.5% will be required to achieve substantial clinical improvements in MPS VII.


Subject(s)
Gene Transfer Techniques , Glucuronidase/genetics , Mucopolysaccharidosis VII/physiopathology , Animals , Bone Diseases, Developmental/etiology , Bone Diseases, Developmental/pathology , Dependovirus/genetics , Disease Models, Animal , Genetic Vectors , Glucuronidase/blood , Glucuronidase/metabolism , Glycosaminoglycans/metabolism , Hearing , Liver/enzymology , Longevity , Lysosomes/ultrastructure , Mice , Mucopolysaccharidosis VII/complications , Mucopolysaccharidosis VII/metabolism , Mucopolysaccharidosis VII/pathology , Reproduction , Retina/physiopathology , Tissue Distribution , alpha-Galactosidase/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...