Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 68
Filter
1.
Int J Mol Sci ; 25(6)2024 Mar 12.
Article in English | MEDLINE | ID: mdl-38542208

ABSTRACT

Mucopolysaccharidosis type IVA (MPS IVA; Morquio A syndrome) is a rare autosomal recessive lysosomal storage disease (LSD) caused by deficiency of a hydrolase enzyme, N-acetylgalactosamine-6-sulfate sulfatase, and characterized clinically by mainly musculoskeletal manifestations. The mechanisms underlying bone involvement in humans are typically explored using invasive techniques such as bone biopsy, which complicates analysis in humans. We compared bone proteomes using DDA and SWATH-MS in wild-type and MPS IVA knockout mice (UNT) to obtain mechanistic information about the disease. Our findings reveal over 1000 dysregulated proteins in knockout mice, including those implicated in oxidative phosphorylation, oxidative stress (reactive oxygen species), DNA damage, and iron transport, and suggest that lactate dehydrogenase may constitute a useful prognostic and follow-up biomarker. Identifying biomarkers that reflect MPS IVA clinical course, severity, and progression have important implications for disease management.


Subject(s)
Bone Diseases , Cartilage Diseases , Chondroitinsulfatases , Mucopolysaccharidosis IV , Humans , Animals , Mice , Mucopolysaccharidosis IV/metabolism , Chondroitinsulfatases/genetics , Mice, Knockout
2.
Gene ; 780: 145527, 2021 May 15.
Article in English | MEDLINE | ID: mdl-33636292

ABSTRACT

Mucopolysaccharidosis type IVA (MPS IVA) is a lysosomal storage disease produced by the deficiency of the N-acetylgalactosamine-6-sulfate sulfatase (GALNS) enzyme, leading to glycosaminoglycans (GAGs) accumulation. Since currently available treatments remain limited and unspecific, novel therapeutic approaches are essential for the disease treatment. In an attempt to reduce treatment limitations, gene therapy rises as a more effective and specific alternative. We present in this study the delivery assessment of GALNS and sulfatase-modifying factor 1 (SUMF1) genes via HIV-1 derived lentiviral vectors into fibroblasts from MPS IVA patients. After transduction, we determined GALNS enzymatic activity, lysosomal mass change, and autophagy pathway impairment. Additionally, we computationally assessed the effect of mutations over the enzyme-substrate interaction and phenotypic effects. The results showed that the co-transduction of MPS IVA fibroblasts with GALNS and SUMF1 cDNAs led to a significant increase in GALNS enzyme activity and a reduction of lysosomal mass. We show that patient-specific differences in cellular response are directly associated with the set of mutations on each patient. Lastly, we present new evidence supporting autophagy impairment in MPS IVA due to the presence and changes in autophagy proteins in treated MPS IVA fibroblasts. Our results offer new evidence that demonstrate the potential of lentiviral vectors as a strategy to correct GALNS deficiency.


Subject(s)
Chondroitinsulfatases , Fibroblasts/metabolism , Genetic Vectors , HIV-1 , Mucopolysaccharidosis IV , Oxidoreductases Acting on Sulfur Group Donors , Transduction, Genetic , Chondroitinsulfatases/biosynthesis , Chondroitinsulfatases/genetics , Genetic Therapy , HEK293 Cells , Humans , Mucopolysaccharidosis IV/genetics , Mucopolysaccharidosis IV/metabolism , Mucopolysaccharidosis IV/therapy , Oxidoreductases Acting on Sulfur Group Donors/biosynthesis , Oxidoreductases Acting on Sulfur Group Donors/genetics
3.
Int J Mol Sci ; 22(1)2020 Dec 28.
Article in English | MEDLINE | ID: mdl-33379360

ABSTRACT

Mucopolysaccharidosis type IVA (MPS IVA) is a lysosomal storage disease caused by mutations in the N-acetylgalactosamine-6-sulfatase (GALNS) gene. Skeletal dysplasia and the related clinical features of MPS IVA are caused by disruption of the cartilage and its extracellular matrix, leading to a growth imbalance. Enzyme replacement therapy (ERT) with recombinant human GALNS has yielded positive results in activity of daily living and endurance tests. However, no data have demonstrated improvements in bone lesions and bone grow thin MPS IVA after ERT, and there is no correlation between therapeutic efficacy and urine levels of keratan sulfate, which accumulates in MPS IVA patients. Using qualitative and quantitative proteomics approaches, we analyzed leukocyte samples from healthy controls (n = 6) and from untreated (n = 5) and ERT-treated (n = 8, sampled before and after treatment) MPS IVA patients to identify potential biomarkers of disease. Out of 690 proteins identified in leukocytes, we selected a group of proteins that were dysregulated in MPS IVA patients with ERT. From these, we identified four potential protein biomarkers, all of which may influence bone and cartilage metabolism: lactotransferrin, coronin 1A, neutral alpha-glucosidase AB, and vitronectin. Further studies of cartilage and bone alterations in MPS IVA will be required to verify the validity of these proteins as potential biomarkers of MPS IVA.


Subject(s)
Biomarkers/metabolism , Mucopolysaccharidosis IV/metabolism , Proteomics , Adolescent , Adult , Case-Control Studies , Child , Child, Preschool , Down-Regulation , Enzyme Replacement Therapy , Female , Humans , Infant , Leukocytes/metabolism , Male , Mucopolysaccharidosis IV/therapy , Protein Interaction Maps , Young Adult
4.
Glycobiology ; 30(7): 433-445, 2020 07 20.
Article in English | MEDLINE | ID: mdl-31897472

ABSTRACT

Morquio syndrome type A, also known as MPS IVA, is a rare autosomal recessive disorder caused by deficiency of N-acetylgalactosamine-6-sulfatase, a lysosomal hydrolase critical in the degradation of keratan sulfate (KS) and chondroitin sulfate (CS). The CS that accumulates in MPS IVA patients has a disease-specific nonreducing end (NRE) terminating with N-acetyl-D-galactosamine 6-sulfate, which can be specifically quantified after enzymatic depolymerization of CS polysaccharide chains. The abundance of N-acetyl-D-galactosamine 6-sulfate over other possible NRE structures is diagnostic for MPS IVA. Here, we describe an assay for the liberation and measurement of N-acetyl-D-galactosamine 6-sulfate and explore its application to MPS IVA patient samples in pilot studies examining disease detection, effects of age and treatment with enzyme-replacement therapy. This assay complements the existing urinary KS assay by quantifying CS-derived substrates, which represent a distinct biochemical aspect of MPS IVA. A more complete understanding of the disease could help to more definitively detect disease across age ranges and more completely measure the pharmacodynamic efficacy of therapies. Larger studies will be needed to clarify the potential value of this CS-derived substrate to manage disease in MPS IVA patients.


Subject(s)
Chondroitin Sulfates/metabolism , Mucopolysaccharidosis IV/metabolism , Adult , Cells, Cultured , Child , Chondroitin Sulfates/chemistry , Chondroitin Sulfates/urine , Chondroitinsulfatases/metabolism , Enzyme Replacement Therapy , Humans , Mucopolysaccharidosis IV/therapy , Mucopolysaccharidosis IV/urine
5.
Int J Mol Sci ; 20(17)2019 Aug 24.
Article in English | MEDLINE | ID: mdl-31450640

ABSTRACT

Mucopolysaccharidosis IVA (MPS IVA) is caused by a deficiency of the lysosomal enzyme N-acetylgalactosamine-6-sulfate sulfatase (GALNS). Conventional enzyme replacement therapy (ERT) is approved for MPS IVA. However, the fact that the infused enzyme cannot penetrate avascular lesions in cartilage leads to minimal impact on the bone lesion. Moreover, short half-life, high cost, instability, and narrow optimal pH range remain unmet challenges in ERT. Thermostable keratanase, endo-ß-N-acetylglucosaminidase, has a unique character of a wide optimal pH range of pH 5.0-7.0. We hypothesized that this endoglycosidase degrades keratan sulfate (KS) polymer in circulating blood and, therefore, ameliorates the accumulation of KS in multiple tissues. We propose a novel approach, Substrate Degradation Enzyme Therapy (SDET), to treat bone lesion of MPS IVA. We assessed the effect of thermostable keratanase on blood KS level and bone pathology using Galns knock-out MPS IVA mice. After a single administration of 2 U/kg (= 0.2 mg/kg) of the enzyme at 8 weeks of age via intravenous injection, the level of serum KS was significantly decreased to normal range level, and this suppression was maintained for at least 4 weeks. We administered 2 U/kg of the enzyme to MPS IVA mice every fourth week for 12 weeks (total of 3 times) at newborns or 8 weeks of age. After a third injection, serum mono-sulfated KS levels were kept low for 4 weeks, similar to that in control mice, and at 12 weeks, bone pathology was markedly improved when SDET started at newborns, compared with untreated MPS IVA mice. Overall, thermostable keratanase reduces the level of KS in blood and provides a positive impact on cartilage lesions, demonstrating that SDET is a novel therapeutic approach to MPS IVA.


Subject(s)
Enzyme Replacement Therapy , Mucopolysaccharidosis IV/enzymology , Mucopolysaccharidosis IV/therapy , Animals , Biomarkers , Disease Models, Animal , Enzyme Stability , Glycosaminoglycans/metabolism , Glycoside Hydrolases/administration & dosage , Glycoside Hydrolases/chemistry , Glycoside Hydrolases/isolation & purification , Male , Mice , Mice, Knockout , Mucopolysaccharidosis IV/etiology , Mucopolysaccharidosis IV/metabolism , Recombinant Proteins , Substrate Specificity , Temperature , Treatment Outcome
6.
Orphanet J Rare Dis ; 14(1): 137, 2019 06 13.
Article in English | MEDLINE | ID: mdl-31196221

ABSTRACT

INTRODUCTION: Mucopolysaccharidosis (MPS) IVA or Morquio A syndrome is an autosomal recessive lysosomal storage disorder (LSD) caused by deficiency of the N-acetylgalactosamine-6-sulfatase (GALNS) enzyme, which impairs lysosomal degradation of keratan sulphate and chondroitin-6-sulphate. The multiple clinical manifestations of MPS IVA present numerous challenges for management and necessitate the need for individualised treatment. Although treatment guidelines are available, the methodology used to develop this guidance has come under increased scrutiny. This programme was conducted to provide evidence-based, expert-agreed recommendations to optimise management of MPS IVA. METHODS: Twenty six international healthcare professionals across multiple disciplines, with expertise in managing MPS IVA, and three patient advocates formed the Steering Committee (SC) and contributed to the development of this guidance. Representatives from six Patient Advocacy Groups (PAGs) were interviewed to gain insights on patient perspectives. A modified-Delphi methodology was used to demonstrate consensus among a wider group of healthcare professionals with experience managing patients with MPS IVA and the manuscript was evaluated against the validated Appraisal of Guidelines for Research and Evaluation (AGREE II) instrument by three independent reviewers. RESULTS: A total of 87 guidance statements were developed covering five domains: (1) general management principles; (2) recommended routine monitoring and assessments; (3) disease-modifying interventions (enzyme replacement therapy [ERT] and haematopoietic stem cell transplantation [HSCT]); (4) interventions to support respiratory and sleep disorders; (5) anaesthetics and surgical interventions (including spinal, limb, ophthalmic, cardio-thoracic and ear-nose-throat [ENT] surgeries). Consensus was reached on all statements after two rounds of voting. The overall guideline AGREE II assessment score obtained for the development of the guidance was 5.3/7 (where 1 represents the lowest quality and 7 represents the highest quality of guidance). CONCLUSION: This manuscript provides evidence- and consensus-based recommendations for the management of patients with MPS IVA and is for use by healthcare professionals that manage the holistic care of patients with the intention to improve clinical- and patient-reported outcomes and enhance patient quality of life. It is recognised that the guidance provided represents a point in time and further research is required to address current knowledge and evidence gaps.


Subject(s)
Chondroitinsulfatases/metabolism , Mucopolysaccharidosis IV/metabolism , Chondroitinsulfatases/genetics , Enzyme Replacement Therapy/methods , Female , Humans , Hypercapnia/genetics , Hypercapnia/metabolism , Male
7.
Orphanet J Rare Dis ; 14(1): 143, 2019 06 14.
Article in English | MEDLINE | ID: mdl-31200731

ABSTRACT

BACKGROUND: Mucopolysaccharidosis IVA (MPS IVA) is an autosomal recessive lysosomal storage disease due to N-acetylgalactosamine-6-sulfatase (GALNS) deficiency. It results in accumulation of the glycosaminoglycans, keratan sulfate and chondroitin-6-sulfate, leading to skeletal and other systemic impairments. Data on MPS IVA in Asian populations are scarce. METHODS: This is a multicentre descriptive case series of 21 patients comprising all MPS IVA patients in Malaysia. Mutational analysis was performed by PCR and Sanger sequencing of the GALNS gene in 17 patients. RESULTS: The patients (15 females and 6 males) had a mean age (± SD) of 15.5 (± 8.1) years. Mean age at symptom onset was 2.6 (± 2.1) years and at confirmed diagnosis was 6.9 (± 4.5) years. The study cohort included patients from all the main ethnic groups in Malaysia - 57% Malay, 29% Chinese and 14% Indian. Common presenting symptoms included pectus carinatum (57%) and genu valgum (43%). Eight patients (38%) had undergone surgery, most commonly knee surgeries (29%) and cervical spine decompression (24%). Patients had limited endurance with lower mean walking distances with increasing age. GALNS gene analysis identified 18 distinct mutations comprising 13 missense, three nonsense, one small deletion and one splice site mutation. Of these, eight were novel mutations (Tyr133Ser, Glu158Valfs*12, Gly168*, Gly168Val, Trp184*, Leu271Pro, Glu320Lys, Leu508Pro). Mutations in exons 1, 5 and 9 accounted for 51% of the mutant alleles identified. CONCLUSIONS: All the MPS IVA patients in this study had clinical impairments. A better understanding of the natural history and the clinical and genetic spectrum of MPS IVA in this population may assist early diagnosis, improve management and permit timely genetic counselling and prenatal diagnosis.


Subject(s)
Mucopolysaccharidosis IV/genetics , Mucopolysaccharidosis IV/pathology , Adolescent , Adult , Child , Child, Preschool , Chondroitinsulfatases/genetics , Chondroitinsulfatases/metabolism , Cohort Studies , Female , Humans , Malaysia , Male , Mucopolysaccharidosis IV/metabolism , Young Adult
8.
Bioorg Med Chem ; 26(20): 5462-5469, 2018 11 01.
Article in English | MEDLINE | ID: mdl-30270003

ABSTRACT

(5aR)-5a-C-pentyl-4-epi-isofagomine 1 is a powerful inhibitor of lysosomal ß-galactosidase and a remarkable chaperone for mutations associated with GM1-gangliosidosis and Morquio disease type B. We report herein an improved synthesis of this compound and analogs (5a-C-methyl, pentyl, nonyl and phenylethyl derivatives), and a crystal structure of a synthetic intermediate that confirms its configuration resulting from the addition of a Grignard reagent. These compounds were evaluated as glycosidase inhibitors and their potential as chaperones for mutant lysosomal galactosidases determined. Based on these results and on docking studies, the 5-C-pentyl derivative 1 was selected as the optimal structure for further investigations: this compound induces the maturation of mutated ß-galactosidase in fibroblasts of a GM1-gangliosidosis patient and promote the decrease of keratan sulfate and oligosaccharide load in patient cells. Compound 1 is clearly capable of restoring ß-galactosidase activity and of promoting maturation of the protein, which should result in significant clinical benefit. These properties strongly support the development of compound 1 for the treatment of GM1-gangliosidosis and Morquio disease type B patients harboring ß-galactosidase mutations sensitive to pharmacological chaperoning.


Subject(s)
Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacology , Gangliosidosis, GM1/drug therapy , Imino Pyranoses/chemistry , Imino Pyranoses/pharmacology , Mucopolysaccharidosis IV/drug therapy , beta-Galactosidase/antagonists & inhibitors , Drug Discovery , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/therapeutic use , Gangliosidosis, GM1/enzymology , Gangliosidosis, GM1/genetics , Gangliosidosis, GM1/metabolism , Humans , Imino Pyranoses/chemical synthesis , Imino Pyranoses/therapeutic use , Molecular Docking Simulation , Mucopolysaccharidosis IV/enzymology , Mucopolysaccharidosis IV/genetics , Mucopolysaccharidosis IV/metabolism , Mutation/drug effects , Structure-Activity Relationship , beta-Galactosidase/genetics , beta-Galactosidase/metabolism
9.
BMC Med Genet ; 19(1): 183, 2018 10 11.
Article in English | MEDLINE | ID: mdl-30305043

ABSTRACT

BACKGROUND: Mucopolysaccharidosis-IVA (Morquio A disease) is a lysosomal disorder in which the abnormal accumulation of keratan sulfate and chondroitin-6-sulfate is consequent to mutations in the galactosamine-6-sulfatase (GALNS) gene. Since standard DNA sequencing analysis fails to detect about 16% of GALNS mutant alleles, gross DNA rearrangement screening and uniparental disomy evaluation are required to complete the molecular diagnosis. Despite this, the second pathogenic GALNS allele generally remains unidentified in ~ 5% of Morquio-A disease patients. METHODS: In an attempt to bridge the residual gap between clinical and molecular diagnosis, we performed an mRNA-based evaluation of three Morquio-A disease patients in whom the second mutant GALNS allele had not been identified. We also performed sequence analysis of the entire GALNS gene in two patients. RESULTS: Different aberrant GALNS mRNA transcripts were characterized in each patient. Analysis of these transcripts then allowed the identification, in one patient, of a disease-causing deep intronic GALNS mutation. The aberrant mRNA products identified in the other two individuals resulted in partial exon loss. Despite sequencing the entire GALNS gene region in these patients, the identity of a single underlying pathological lesion could not be unequivocally determined. We postulate that a combination of multiple variants, acting in cis, may synergise in terms of their impact on the splicing machinery. CONCLUSIONS: We have identified GALNS variants located within deep intronic regions that have the potential to impact splicing. These findings have prompted us to incorporate mRNA analysis into our diagnostic flow procedure for the molecular analysis of Morquio A disease.


Subject(s)
Chondroitinsulfatases/genetics , Mucopolysaccharidosis IV/genetics , Mutation , RNA Splicing , RNA, Messenger/genetics , Adolescent , Base Sequence , Chondroitinsulfatases/metabolism , DNA Mutational Analysis , Decision Trees , Exons , Female , Genotype , Humans , Introns , Male , Mucopolysaccharidosis IV/diagnosis , Mucopolysaccharidosis IV/metabolism , Mucopolysaccharidosis IV/physiopathology , RNA, Messenger/metabolism
10.
Mol Genet Metab ; 120(1-2): 78-95, 2017.
Article in English | MEDLINE | ID: mdl-27979613

ABSTRACT

Mucopolysaccharidosis IVA (MPS IVA; Morquio A: OMIM 253000) is a lysosomal storage disease with an autosomal recessive trait caused by the deficiency of N-acetylgalactosamine-6-sulfate sulfatase. Deficiency of this enzyme leads to accumulation of specific glycosaminoglycans (GAGs): chondroitin-6-sulfate (C6S) and keratan sulfate (KS). C6S and KS are mainly produced in the cartilage. Therefore, the undegraded substrates are stored primarily in cartilage and in its extracellular matrix (ECM), leading to a direct impact on cartilage and bone development, and successive systemic skeletal dysplasia. Chondrogenesis, the earliest phase of skeletal formation, is maintained by cellular interactions with the ECM, growth and differentiation factors, signaling pathways, and transcription factors in a temporal-spatial manner. In patients with MPS IVA, the cartilage is disrupted at birth as a consequence of abnormal chondrogenesis and/or endochondral ossification. The unique skeletal features are distinguished by a disproportional short stature, odontoid hypoplasia, spinal cord compression, tracheal obstruction, pectus carinatum, kyphoscoliosis, platyspondyly, coxa valga, genu valgum, waddling gait, and laxity of joints. In spite of many descriptions of these unique clinical features, delay of diagnosis still happens. The pathogenesis and treatment of systemic skeletal dysplasia in MPS IVA remains an unmet challenge. In this review article, we comprehensively describe historical aspect, property of GAGs, diagnosis, screening, pathogenesis, and current and future therapies of MPS IVA.


Subject(s)
Cartilage/metabolism , Glycosaminoglycans/metabolism , Mucopolysaccharidosis IV/diagnosis , Mucopolysaccharidosis IV/therapy , Adolescent , Adult , Clinical Trials as Topic , Enzyme Replacement Therapy , Female , Genetic Therapy , Humans , Male , Mucopolysaccharidosis IV/metabolism , Orthopedic Procedures
11.
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
12.
Clin Pharmacokinet ; 53(12): 1137-47, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25234648

ABSTRACT

BACKGROUND AND OBJECTIVES: Morquio A syndrome (mucopolysaccharidosis IVA; MPS IVA) is a lysosomal storage disorder caused by deficiency of N-acetylgalactosamine-6-sulfatase, an enzyme required for degradation of the glycosaminoglycan keratan sulfate. Enzyme replacement therapy with elosulfase alfa provides a potential therapy for Morquio A syndrome. We analyzed the pharmacokinetics and pharmacodynamics of elosulfase alfa in Morquio A patients from a phase III clinical trial. METHODS: In a randomized double-blind study, elosulfase alfa at 2.0 mg/kg was administrated weekly or every other week for 24 weeks. Pharmacokinetic parameters of elosulfase alfa were determined at weeks 0 and 22 by non-compartmental analysis. Safety was assessed throughout the study. The relationship of pharmacokinetic parameters to patient demographics, pharmacodynamic assessments, immunogenicity, and efficacy and safety outcomes were assessed graphically by treatment group. RESULTS: Elosulfase alfa exposure and half-life (t(½)) increased for both dose regimens during the study. There appeared to be no consistent trend between drug clearance (CL) and patient's sex, race, body weight, or age. All patients developed anti-drug antibodies, but no association was noted between total antibody titer and CL. In contrast, positive neutralizing antibody (NAb) status appeared to associate with decreased CL and prolonged t(½) for patients in the cohort dosed weekly. NAb may interfere with receptor-mediated cellular uptake and lead to increased circulation time of elosulfase alfa. CONCLUSION: Despite the association between NAb and decreased drug clearance, neither dosing cohort showed associations between drug exposure and change in urinary keratan sulfate, 6-min walk test distances, or the occurrence of adverse events.


Subject(s)
Chondroitinsulfatases , Enzyme Replacement Therapy , Mucopolysaccharidosis IV , Adolescent , Adult , Antibodies, Neutralizing/blood , Child , Chondroitinsulfatases/blood , Chondroitinsulfatases/pharmacokinetics , Chondroitinsulfatases/pharmacology , Chondroitinsulfatases/therapeutic use , Double-Blind Method , Female , Humans , Male , Middle Aged , Mucopolysaccharidosis IV/drug therapy , Mucopolysaccharidosis IV/immunology , Mucopolysaccharidosis IV/metabolism , Recombinant Proteins/blood , Recombinant Proteins/pharmacokinetics , Recombinant Proteins/pharmacology , Recombinant Proteins/therapeutic use , Young Adult
13.
Cardiovasc Pathol ; 23(3): 145-51, 2014.
Article in English | MEDLINE | ID: mdl-24508139

ABSTRACT

INTRODUCTION: The mucopolysaccharidosis syndromes are a group of lethal inherited disorders affecting multiple organ systems by the progressive deposition of glycosaminoglycan. Advances in treatment such as enzyme replacement and hematopoietic stem cell transplantation have significantly improved the outcome of these disorders. An in-depth understanding of the pathophysiology of heart disease in these disorders is essential since death from cardiac causes continues to be common. Epicardial coronary artery luminal narrowing from myointimal proliferation and glycosaminoglycan deposition is well described in severe mucopolysaccharidosis type I [Hurler syndrome, mucopolysaccharide IH] but poorly understood in other "non-Hurler" phenotypes of these disorders. Given the rarity of these conditions, autopsy specimens are uncommon. METHODS: Tissue from epicardial coronary arteries from autopsies of four patients with non-Hurler mucopolysaccharidosis (attenuated type I, type IIIA, type IIIC, and type VI) who had died after hematopoietic cell transplantation (within 1 month in three cases; after 5 years in the fourth) was examined by light microscopy. RESULTS: Unexpectedly, near-normal coronary arteries were observed in the patient with attenuated mucopolysaccharidosis type I, while the coronaries from patients with type IIIA, IIIC, and VI demonstrated classic histologic features of glycosaminoglycan deposition. The most severe findings were found in the MPS IIIC patient who had 5 years of full donor engraftment after transplantation. CONCLUSIONS: Our current understanding of the cardiac manifestations of the mucopolysaccharidoses fails to explain why near-normal coronary arteries may be observed when abnormalities would be most likely to be expected and, conversely, why significant histopathology is present when it would be least expected. Identification of downstream effects of glycosaminoglycan deposition may identify other metabolites or metabolic pathways that are important in the clinicopathologic manifestations of these diseases. SUMMARY: The mucopolysaccharidosis diseases are a group of inherited disorders affecting multiple organ systems by the progressive deposition of glycosaminoglycan. Severe coronary artery disease is well recognized in severe type I mucopolysaccharidosis (Hurler syndrome), but unexpected coronary artery disease occurs in other, "non-Hurler" mucopolysaccharidoses. Factors responsible for the development of coronary pathology in the mucopolysaccharidoses remain elusive.


Subject(s)
Coronary Artery Disease/pathology , Coronary Vessels/pathology , Mucopolysaccharidosis III/pathology , Mucopolysaccharidosis IV/pathology , Mucopolysaccharidosis I/pathology , Autopsy , Biopsy , Child , Child, Preschool , Coronary Artery Disease/metabolism , Coronary Vessels/chemistry , Fatal Outcome , Female , Glycosaminoglycans/analysis , Hematopoietic Stem Cell Transplantation , Humans , Male , Mucopolysaccharidosis I/metabolism , Mucopolysaccharidosis I/surgery , Mucopolysaccharidosis III/metabolism , Mucopolysaccharidosis III/surgery , Mucopolysaccharidosis IV/metabolism , Mucopolysaccharidosis IV/surgery , Severity of Illness Index , Time Factors , Treatment Outcome , Young Adult
14.
J Dent ; 40(12): 1074-80, 2012 Dec.
Article in English | MEDLINE | ID: mdl-22960538

ABSTRACT

OBJECTIVES: To understand the pathology of the ultrastructure of enamel affected by systemic disorders which disrupt enamel tissue formation in order to give insight into the precise mechanisms of matrix-mediated biomineralization in dental enamel in health and disease. METHODS: Two-dimensional synchrotron X-ray diffraction has been utilized as a sophisticated and useful technique to spatially quantify preferred orientation in mineralized healthy deciduous dental enamel, and the disrupted crystallite organization in enamel affected by a systemic disease affecting bone and dental mineralization (mucopolysaccharidosis Type IVA and Type II are used as examples). The lattice spacing of the hydroxyapatite phase, the crystallite size and aspect ratio, and the quantified preferred orientation of crystallites across whole intact tooth sections, have been determined using synchrotron microdiffraction. RESULTS: Significant differences in mineral crystallite orientation distribution of affected enamel have been observed compared to healthy mineralized tissue. The gradation of enamel crystal orientation seen in healthy tissue is absent in the affected enamel, indicating a continual disruption in the crystallite alignment during mineral formation. CONCLUSIONS: This state of the art technique has the potential to provide a unique insight into the mechanisms leading to deranged enamel formation in a wide range of disease states. CLINICAL RELEVANCE: Characterising crystal orientation patterns and geometry in health and following disruption can be a powerful tool in advancing our overall understanding of mechanisms leading to the tissue phenotypes seen clinically. Findings can be used to inform the appropriate dental management of these tissues and/or to investigate the influence of therapeutic interventions or external stressors which may impact on amelogenesis.


Subject(s)
Dental Enamel/ultrastructure , Tooth, Deciduous/ultrastructure , Algorithms , Anisotropy , Crystallography, X-Ray/methods , Dental Enamel/chemistry , Dentin/chemistry , Dentin/ultrastructure , Durapatite/chemistry , Humans , Incisor/chemistry , Incisor/ultrastructure , Microscopy, Electron, Scanning , Mucopolysaccharidosis II/metabolism , Mucopolysaccharidosis II/pathology , Mucopolysaccharidosis IV/metabolism , Mucopolysaccharidosis IV/pathology , Scattering, Small Angle , Synchrotrons , Tooth Calcification , Tooth, Deciduous/chemistry
15.
Mol Genet Metab ; 105(3): 438-42, 2012 Mar.
Article in English | MEDLINE | ID: mdl-22178352

ABSTRACT

Morquio A syndrome (MPS IVA) is a recessive lysosomal storage disorder (LSD) caused by mutations in the GALNS gene leading to the deficiency of lysosomal enzyme N-acetylgalactosamine-6-sulfate sulfatase (GALNS). Patients show a broad spectrum of phenotypes ranging from classical severe type to mild forms. Classical forms are characterized by severe bone dysplasia and usually normal intelligence. So far, more than 170 unique mutations have been identified in the GALNS gene of MPS IVA patients. We report on a Morquio A patient with a classical phenotype who was found to be homozygous for a missense mutation (c.236 G>A; p.Cys79Tyr) in the GALNS gene. This alteration affects the highly conserved p.Cys79 that is transformed into formylglycine, the catalytic residue of the active site. The mutation was present in the proband's mother, but not in the father, whose paternity was confirmed by microsatellite analysis. In order to test the hypothesis of maternal uniparental disomy (UPD), we investigated the segregation of sixteen microsatellite markers from chromosome 16. The results showed a condition of maternal UPD due to an error in meiosis I. Maternal isodisomy of the 16q24 region led to homozygosity for the GALNS mutant allele, causing the patient's disease. These findings allow to add for the first time the LSD Morquio A syndrome to the list of conditions that can be caused by UPD. The possibility of UPD is relevant when giving genetic counseling to couples since the recurrent risk in future pregnancies is dramatically reduced.


Subject(s)
Chondroitinsulfatases/genetics , Chromosomes, Human, Pair 16/genetics , Mucopolysaccharidosis IV/genetics , Uniparental Disomy , Chromosome Aberrations , Genetic Markers , Humans , Male , Microsatellite Repeats , Mucopolysaccharidosis IV/enzymology , Mucopolysaccharidosis IV/metabolism , Phenotype
16.
Curr Pharm Biotechnol ; 12(6): 931-45, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21506915

ABSTRACT

Mucopolysaccharidosis IVA (MPS IVA), also known as Morquio A, is a rare, autosomal recessive disorder caused by a deficiency of the lysosomal enzyme N-acetylgalatosamine-6-sulfate-sulfatase (GALNS), which catalyzes a step in the catabolism of glycosaminoglycans (GAGs), keratan sulfate (KS) and chondroitin-6-sulfate (C6S). It leads to accumulation of the KS and C6S, mainly in bone and cornea, causing a systemic skeletal chondrodysplasia. MPS IVA has a variable age of onset and variable rate of progression. Common presenting features include elevation of urinary and blood KS, marked short stature, hypoplasia of the odontoid process, pectus carinatum, kyphoscoliosis, genu valgum, laxity of joints and corneal clouding; however there is no central nervous system impairment. Generally, MPS IVA patients with a severe form do not survive beyond the third decade of life whereas those patients with an attenuated form may survive over 70 years. There has been no effective therapy for MPS IVA, and care has been palliative. Enzyme replacement therapy (ERT) and hematopoietic stem cell therapy (HSCT) have emerged as a treatment for mucopolysaccharidoses disorders, including Morquio A disease. This review provides an overview of the clinical manifestations, diagnosis and symptomatic management of patients with MPS IVA and describes potential perspectives of ERT and HSCT. The issue of treating very young patients is also discussed.


Subject(s)
Enzyme Replacement Therapy/methods , Hematopoietic Stem Cell Transplantation/methods , Mucopolysaccharidosis IV/diagnosis , Mucopolysaccharidosis IV/therapy , Animals , Humans , Keratan Sulfate/metabolism , Mucopolysaccharidosis IV/metabolism
17.
PLoS One ; 5(8): e12194, 2010 Aug 16.
Article in English | MEDLINE | ID: mdl-20808938

ABSTRACT

Mucopolysaccharidosis IVA (MPS IVA; Morquio A syndrome) is a lysosomal storage disorder caused by deficiency of N-acetylgalactosamine-6-sulfatase (GALNS), an enzyme that degrades keratan sulfate (KS). Currently no therapy for MPS IVA is available. We produced recombinant human (rh)GALNS as a potential enzyme replacement therapy for MPS IVA. Chinese hamster ovary cells stably overexpressing GALNS and sulfatase modifying factor-1 were used to produce active ( approximately 2 U/mg) and pure (>or=97%) rhGALNS. The recombinant enzyme was phosphorylated and was dose-dependently taken up by mannose-6-phosphate receptor (K(uptake) = 2.5 nM), thereby restoring enzyme activity in MPS IVA fibroblasts. In the absence of an animal model with a skeletal phenotype, we established chondrocytes isolated from two MPS IVA patients as a disease model in vitro. MPS IVA chondrocyte GALNS activity was not detectable and the cells exhibited KS storage up to 11-fold higher than unaffected chondrocytes. MPS IVA chondrocytes internalized rhGALNS into lysosomes, resulting in normalization of enzyme activity and decrease in KS storage. rhGALNS treatment also modulated gene expression, increasing expression of chondrogenic genes Collagen II, Collagen X, Aggrecan and Sox9 and decreasing abnormal expression of Collagen I. Intravenous administration of rhGALNS resulted in biodistribution throughout all layers of the heart valve and the entire thickness of the growth plate in wild-type mice. We show that enzyme replacement therapy with recombinant human GALNS results in clearance of keratan sulfate accumulation, and that such treatment ameliorates aberrant gene expression in human chondrocytes in vitro. Penetration of the therapeutic enzyme throughout poorly vascularized, but clinically relevant tissues, including growth plate cartilage and heart valve, as well as macrophages and hepatocytes in wild-type mouse, further supports development of rhGALNS as enzyme replacement therapy for MPS IVA.


Subject(s)
Cartilage/metabolism , Enzyme Replacement Therapy/methods , Mucopolysaccharidosis IV/drug therapy , Mucopolysaccharidosis IV/metabolism , Animals , Biological Transport , Cattle , Chondrocytes/metabolism , Chondrocytes/pathology , Chondroitinsulfatases/isolation & purification , Chondroitinsulfatases/metabolism , Chondroitinsulfatases/pharmacokinetics , Chondroitinsulfatases/therapeutic use , Female , Gene Expression Regulation/drug effects , Growth Plate/metabolism , Heart Valves/metabolism , Humans , Keratan Sulfate/metabolism , Liver/pathology , Lysosomes/metabolism , Macrophages/metabolism , Mice , Mucopolysaccharidosis IV/pathology , Recombinant Proteins/isolation & purification , Recombinant Proteins/metabolism , Recombinant Proteins/pharmacokinetics , Recombinant Proteins/therapeutic use
18.
Mol Genet Metab ; 100(3): 262-8, 2010 Jul.
Article in English | MEDLINE | ID: mdl-20409738

ABSTRACT

G(M1)-gangliosidosis (GM1) and Morquio B disease (MBD) are rare lysosomal storage disorders caused by mutations in the gene GLB1. Its main gene product, human acid beta-galactosidase (beta-Gal) degrades two functionally important molecules, G(M1)-ganglioside and keratan sulfate in brain and connective tissues, respectively. While GM1 is a severe, phenotypically heterogenous neurodegenerative disorder, MBD is a systemic bone disease without effects on the central nervous system. A MBD-specific mutation, p.W273L, was shown to produce stable beta-Gal precursors, normally transported and processed to mature, intralysosomal beta-Gal. In accordance with the MBD phenotype, elevated residual activity against G(M1)-ganglioside, but strongly reduced affinity towards keratan sulfate was found. Most GM1 alleles, in contrast, were shown to affect precursor stability and intracellular transport. Specific alleles, p.R201C and p.R201H result in misfolded, unstable precursor proteins rapidly degraded by endoplasmic reticulum-associated protein degradation (ERAD). They may therefore be sensitive to stabilization by small molecules which bind at the active site and provide proper conformation. Thus the stabilized protein may escape from ERAD processes, and reach the lysosomes in an active state, as proposed for enzyme enhancement therapy (EET). This paper demonstrates that a novel iminosugar, DLHex-DGJ, has potent effects as competitive inhibitor of human acid beta-galactosidase in vitro, and describes its effects on activity, protein expression, maturation and intracellular transport in vivo in 13 fibroblasts lines with GLB1 mutations. Beside p.R201C and p.R201H, two further alleles, p.C230R and p.G438E, displayed significant sensitivity against DLHex-DGJ, with an increase of catalytic activity, and a normalization of transport and lysosomal processing of beta-Gal precursors.


Subject(s)
1-Deoxynojirimycin/analogs & derivatives , Gangliosidosis, GM1/drug therapy , Mucopolysaccharidosis IV/drug therapy , 1-Deoxynojirimycin/chemistry , 1-Deoxynojirimycin/pharmacokinetics , 1-Deoxynojirimycin/pharmacology , Alleles , Cell Line , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacokinetics , Enzyme Inhibitors/pharmacology , Fibroblasts/drug effects , Fibroblasts/metabolism , Gangliosidosis, GM1/genetics , Gangliosidosis, GM1/metabolism , Humans , Molecular Chaperones/chemistry , Molecular Chaperones/pharmacokinetics , Molecular Chaperones/pharmacology , Mucopolysaccharidosis IV/genetics , Mucopolysaccharidosis IV/metabolism , Mutation , beta-Galactosidase/antagonists & inhibitors , beta-Galactosidase/genetics , beta-Galactosidase/metabolism
19.
J Neurosci ; 27(37): 9928-40, 2007 Sep 12.
Article in English | MEDLINE | ID: mdl-17855607

ABSTRACT

Neurogenetic disorders typically affect cells throughout the brain. Adeno-associated virus (AAV) vector-mediated transfer of a normal cDNA can correct the metabolic defects at the site of injection, but treatment of the entire brain requires widespread delivery of the normal gene and/or protein. Current methods require multiple injections for widespread distribution. However, some AAV vectors can be transported along neuronal pathways associated with the injected region. Thus, targeting widely dispersed systems in the CNS might be a pathway for gene dispersal from a limited number of sites. We tested this hypothesis in the ventral tegmental area (VTA), a region with numerous efferent and afferent projections. A single 1 mul injection resulted in transport of the vector genome to projection sites in distal parts of the brain. When compared with injections into the striatum, the VTA injection resulted in higher enzyme levels in more regions of the brain. The AAV-9 serotype vector was the most widely disseminated, but AAV-Rh.10 and AAV-1 were also transported after VTA injection. The effect on global lesions of a neurogenetic disease was tested in the mouse model of MPS VII (mucopolysaccharidosis VII), a lysosomal storage disorder. Widespread distribution of the vector genome after AAV-9 VTA injection resulted in even further distribution of the enzyme product, by secretion and uptake by surrounding cells, and complete correction of the storage lesions throughout the entire brain. This unprecedented level of correction from a single injection into the developed brain provides a potential strategy to correct a large volume of brain while minimizing the number of injections.


Subject(s)
Adenoviridae/metabolism , Brain/metabolism , Cell Nucleus/metabolism , Genetic Vectors/administration & dosage , Genetic Vectors/metabolism , Mucopolysaccharidosis IV/metabolism , Transduction, Genetic , Adenoviridae/genetics , Animals , Biological Transport/drug effects , Biological Transport/genetics , Brain/drug effects , Cell Nucleus/drug effects , Cell Nucleus/genetics , Genetic Therapy/methods , Genetic Vectors/genetics , Humans , Mice , Mice, Inbred C3H , Microinjections , Mucopolysaccharidosis IV/genetics , Mucopolysaccharidosis IV/therapy
20.
Osteoarthritis Cartilage ; 15(11): 1311-7, 2007 Nov.
Article in English | MEDLINE | ID: mdl-17548214

ABSTRACT

OBJECTIVE: To investigate the gene expression profile and the histological aspects of articular cartilage of patients affected by Morquio syndrome, a lysosomal storage disease characterized by the accumulation of glycosaminoglycans within the cells which result in abnormal formation and growth of the skeletal system. METHOD: Articular cartilage samples were obtained from the femoral condyle of two siblings with Morquio syndrome during surgery performed to treat valgus knee. As controls, four biopsy samples of healthy cartilage were obtained from four different male multiorgan donors. A Real-Time Polymerase Chain reaction (RT-PCR) analysis was performed to evaluate the expression of type I and II collagens and aggrecan mRNAs. Histological and immunohistochemical analyses for some matrix proteins were carried out on paraffin embedded sections. RESULTS: Type I collagen mRNA mean level was higher in the samples of patients with Morquio syndrome compared to controls. Type II collagen and aggrecan mRNAs' mean expression was instead lower. The morphological appearance of the cartilage showed a poorly organized tissue structure with not homogeneously distributed cells that were larger compared to normal chondrocytes due to the presence inside the vacuoles of proteoglycans which were not metabolized. Chondrocytes were negative for collagen II immunostaining while the extracellular matrix was weakly positive. Collagen type I immunostaining was positive at cellular level. Keratan sulfate showed diffuse positivity and chondroitin-6-sulfate was present throughout the cartilaginous thickness. CONCLUSION: In cartilage of patients with Morquio syndrome, a low expression of collagen type II and a high expression of collagen type I both at protein and molecular levels are evidentiated. This finding could give evidence of the reduction in ankle and knee joint movement observable in these patients.


Subject(s)
Cartilage, Articular/metabolism , Chondrocytes/metabolism , Collagen Type II/genetics , Collagen Type I/genetics , Mucopolysaccharidosis IV/genetics , Adult , Aggrecans , Biomarkers/analysis , Chondrocytes/pathology , Collagen Type I/metabolism , Collagen Type II/metabolism , Female , Humans , Immunohistochemistry , Male , Mucopolysaccharidosis IV/metabolism , Mucopolysaccharidosis IV/pathology , RNA, Messenger/analysis , Reverse Transcriptase Polymerase Chain Reaction
SELECTION OF CITATIONS
SEARCH DETAIL
...