51
|
Wilkin J, Kerr NC, Byrd KW, Ward JC, Iannaccone A. Characterization of a Case of Pigmentary Retinopathy in Sanfilippo Syndrome Type IIIA Associated with Compound Heterozygous Mutations in the SGSH Gene. Ophthalmic Genet 2015; 37:217-27. [PMID: 26331342 DOI: 10.3109/13816810.2015.1028647] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
PURPOSE To report longitudinal phenotypic findings in a patient with Sanfilippo syndrome type IIIA, harboring SGSH mutations, one of which is novel. METHODS Heparan-N-sulfatidase enzyme function testing in skin fibroblasts and white blood cells and SGSH gene sequencing were obtained. Clinical office examinations, examinations under anesthesia, electroretinogram, spectral domain optical coherence tomography (SD-OCT), and fundus photography were performed over a 5-year period. RESULTS Fundus examination revealed a progressive breadcrumb-like pigmentary retinopathy with perifoveal pigmentary involvement. SD-OCT showed loss of normal neuroretinal lamination and cystic macular changes responsive to treatment with carbonic anhydrase inhibitors. Electroretinography exhibited complex characteristics indicative of a generalized retinal rod > cone dysfunction with significant ON > OFF postreceptoral response compromise. Sequencing revealed compound heterozygous mutations in the SGSH gene, the novel c.88G > C (p.A30P) change and a second, previously reported one (c.734G > A, p.R245H). CONCLUSIONS We have identified ocular features of a patient with Sanfilippo syndrome type IIIA harboring a novel SGHS mutation that were not previously known to occur in this disease - namely, a progressive retinopathy with distinctive features, cystic macular changes responsive to carbonic anhydrase inhibitors, and complex electroretinographic abnormalities consistent with postreceptoral dysfunction. SD-OCT imaging revealed retinal lamination changes consistent with previously reported histologic studies. Both the SD-OCT and the electroretinogram changes appear attributable to intraretinal deposition of heparan sulfate.
Collapse
Affiliation(s)
- Justin Wilkin
- a Hamilton Eye Institute, Department of Ophthalmology and
| | - Natalie C Kerr
- a Hamilton Eye Institute, Department of Ophthalmology and
| | - Kathryn W Byrd
- a Hamilton Eye Institute, Department of Ophthalmology and
| | - Jewell C Ward
- b Medical Genetics Division, Department of Pediatrics , University of Tennessee Health Science Center , Memphis , TN , USA
| | | |
Collapse
|
52
|
Andrade F, Aldámiz-Echevarría L, Llarena M, Couce ML. Sanfilippo syndrome: Overall review. Pediatr Int 2015; 57:331-8. [PMID: 25851924 DOI: 10.1111/ped.12636] [Citation(s) in RCA: 72] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/02/2014] [Revised: 12/03/2014] [Accepted: 02/13/2015] [Indexed: 12/17/2022]
Abstract
Mucopolysaccharidosis type III (MPS III, Sanfilippo syndrome) is a lysosomal storage disorder, caused by a deficiency in one of the four enzymes involved in the catabolism of glycosaminoglycan heparan sulfate. It is characterized by progressive cognitive decline and severe hyperactivity, with relatively mild somatic features. This review focuses on clinical features, diagnosis, treatment, and follow-up of MPS III, and provides information about supplementary tests and differential diagnosis. Given that few reviews of MPS III have been published, several studies were compiled to establish diagnostic recommendations. Quantitative urinary glycosaminoglycan analysis is strongly recommended, and measurement of disaccharides, heparin cofactor II-thrombin complex and gangliosides is also used. Enzyme activity of the different enzymes in blood serum, leukocytes or fibroblasts, and mutational analysis for SGSH, NAGLU, HGSNAT or GNS genes are required to confirm diagnosis and differentiate four subtypes of MPS III. Although there is no global consensus for treatment, enzyme replacement therapy and gene therapy can provide appropriate results. In this regard, recent publications on treatment and follow-up are discussed.
Collapse
Affiliation(s)
- Fernando Andrade
- Division of Metabolism, BioCruces Health Research Institute, CIBER de Enfermedades Raras (CIBERER), Barakaldo, Spain
| | - Luis Aldámiz-Echevarría
- Division of Metabolism, BioCruces Health Research Institute, CIBER de Enfermedades Raras (CIBERER), Barakaldo, Spain
| | - Marta Llarena
- Division of Metabolism, BioCruces Health Research Institute, CIBER de Enfermedades Raras (CIBERER), Barakaldo, Spain
| | - María Luz Couce
- Unit of Diagnosis and Treatment of Congenital Metabolic Diseases, Neonatology Service, Department of Pediatrics, CIBER de Enfermedades Raras (CIBERER), IDIS Clinic University Hospital of Santiago de Compostela, Coruña, Spain
| |
Collapse
|
53
|
Ugrinov KG, Freed SD, Thomas CL, Lee SW. A multiparametric computational algorithm for comprehensive assessment of genetic mutations in mucopolysaccharidosis type IIIA (Sanfilippo syndrome). PLoS One 2015; 10:e0121511. [PMID: 25807448 PMCID: PMC4373678 DOI: 10.1371/journal.pone.0121511] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2014] [Accepted: 02/12/2015] [Indexed: 12/22/2022] Open
Abstract
Mucopolysaccharidosis type IIIA (MPS-IIIA, Sanfilippo syndrome) is a Lysosomal Storage Disease caused by cellular deficiency of N-sulfoglucosamine sulfohydrolase (SGSH). Given the large heterogeneity of genetic mutations responsible for the disease, a comprehensive understanding of the mechanisms by which these mutations affect enzyme function is needed to guide effective therapies. We developed a multiparametric computational algorithm to assess how patient genetic mutations in SGSH affect overall enzyme biogenesis, stability, and function. 107 patient mutations for the SGSH gene were obtained from the Human Gene Mutation Database representing all of the clinical mutations documented for Sanfilippo syndrome. We assessed each mutation individually using ten distinct parameters to give a comprehensive predictive score of the stability and misfolding capacity of the SGSH enzyme resulting from each of these mutations. The predictive score generated by our multiparametric algorithm yielded a standardized quantitative assessment of the severity of a given SGSH genetic mutation toward overall enzyme activity. Application of our algorithm has identified SGSH mutations in which enzymatic malfunction of the gene product is specifically due to impairments in protein folding. These scores provide an assessment of the degree to which a particular mutation could be treated using approaches such as chaperone therapies. Our multiparametric protein biogenesis algorithm advances a key understanding in the overall biochemical mechanism underlying Sanfilippo syndrome. Importantly, the design of our multiparametric algorithm can be tailored to many other diseases of genetic heterogeneity for which protein misfolding phenotypes may constitute a major component of disease manifestation.
Collapse
Affiliation(s)
- Krastyu G Ugrinov
- Department of Biological Sciences, University of Notre Dame, Notre Dame, Indiana, 46556, United States of America; Center for Rare and Neglected Diseases, University of Notre Dame, Notre Dame, Indiana, 46556, United States of America
| | - Stefan D Freed
- Department of Biological Sciences, University of Notre Dame, Notre Dame, Indiana, 46556, United States of America; Center for Rare and Neglected Diseases, University of Notre Dame, Notre Dame, Indiana, 46556, United States of America
| | - Clayton L Thomas
- Department of Biological Sciences, University of Notre Dame, Notre Dame, Indiana, 46556, United States of America; Center for Rare and Neglected Diseases, University of Notre Dame, Notre Dame, Indiana, 46556, United States of America
| | - Shaun W Lee
- Department of Biological Sciences, University of Notre Dame, Notre Dame, Indiana, 46556, United States of America; Center for Rare and Neglected Diseases, University of Notre Dame, Notre Dame, Indiana, 46556, United States of America
| |
Collapse
|
54
|
Cross EM, Grant S, Jones S, Bigger BW, Wraith JE, Mahon LV, Lomax M, Hare DJ. An investigation of the middle and late behavioural phenotypes of Mucopolysaccharidosis Type-III. J Neurodev Disord 2014; 6:46. [PMID: 25657821 PMCID: PMC4318134 DOI: 10.1186/1866-1955-6-46] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/13/2013] [Accepted: 12/10/2014] [Indexed: 12/29/2022] Open
Abstract
Background Mucopolysaccharidosis type-III (MPS III) is an autosomal recessive lysosomal
storage disorder. It causes progressive physical and cognitive decline and has
been linked to increased incidences of behavioural problems. Methods Data on the behaviour and adaptive skills of 20 children with MPS III and 25
children with intellectual disability (ID) (17 included in analysis) were gathered
via parental report questionnaire. The frequencies of different types of behaviour
displayed by children with MPS III and children with ID were compared across two
age categories. Results The total frequency of challenging behaviours displayed by children aged 2–9
years with MPS III and ID was not significantly different. Behaviours associated
with hyperactivity, orality, unusual body movements and inattention were seen
significantly more frequently in 2–9 year olds with MPS III than in those with ID.
Children aged 10–15 years with MPS III showed significantly fewer problem
behaviours than a contrasting group with ID. The frequency of challenging
behaviours displayed by children with MPS III and their adaptive skills was found
to decrease with age. Conclusions Behaviours relating to hyperactivity, orality, unusual body movements and
inattention are part of the behavioural phenotype of the middle phase of MPS III.
The late phase of MPS III is associated with low rates of problem behaviour and
loss of adaptive skills. Therefore, families with a child with MPS III may benefit
from a different type of clinical service when the child is aged 2–9 years, than
when aged 10–15 years.
Collapse
Affiliation(s)
- Elaine M Cross
- Section for Clinical and Health Psychology, School of Psychological Sciences, University of Manchester, Zochonis Building, Brunswick Street, Manchester, M13 9PL UK
| | - Sheena Grant
- Section for Clinical and Health Psychology, School of Psychological Sciences, University of Manchester, Zochonis Building, Brunswick Street, Manchester, M13 9PL UK
| | - Simon Jones
- Department of Genetic Medicine, St Mary's Hospital, Manchester, UK
| | - Brian W Bigger
- Stem Cell & Neurotherapies, Faculty of Medical and Human Sciences, University of Manchester, Manchester, UK
| | - James E Wraith
- Section for Clinical and Health Psychology, School of Psychological Sciences, University of Manchester, Zochonis Building, Brunswick Street, Manchester, M13 9PL UK
| | - Louise V Mahon
- Section for Clinical and Health Psychology, School of Psychological Sciences, University of Manchester, Zochonis Building, Brunswick Street, Manchester, M13 9PL UK
| | - Michelle Lomax
- Section for Clinical and Health Psychology, School of Psychological Sciences, University of Manchester, Zochonis Building, Brunswick Street, Manchester, M13 9PL UK
| | - Dougal J Hare
- Section for Clinical and Health Psychology, School of Psychological Sciences, University of Manchester, Zochonis Building, Brunswick Street, Manchester, M13 9PL UK
| |
Collapse
|
55
|
Ribera A, Haurigot V, Garcia M, Marcó S, Motas S, Villacampa P, Maggioni L, León X, Molas M, Sánchez V, Muñoz S, Leborgne C, Moll X, Pumarola M, Mingozzi F, Ruberte J, Añor S, Bosch F. Biochemical, histological and functional correction of mucopolysaccharidosis type IIIB by intra-cerebrospinal fluid gene therapy. Hum Mol Genet 2014; 24:2078-95. [PMID: 25524704 DOI: 10.1093/hmg/ddu727] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
Gene therapy is an attractive tool for the treatment of monogenic disorders, in particular for lysosomal storage diseases (LSD) caused by deficiencies in secretable lysosomal enzymes in which neither full restoration of normal enzymatic activity nor transduction of all affected cells are necessary. However, some LSD such as Mucopolysaccharidosis Type IIIB (MPSIIIB) are challenging because the disease's main target organ is the brain and enzymes do not efficiently cross the blood-brain barrier even if present at very high concentration in circulation. To overcome these limitations, we delivered AAV9 vectors encoding for α-N-acetylglucosaminidase (NAGLU) to the Cerebrospinal Fluid (CSF) of MPSIIIB mice with the disease already detectable at biochemical, histological and functional level. Restoration of enzymatic activity in Central Nervous System (CNS) resulted in normalization of glycosaminoglycan content and lysosomal physiology, resolved neuroinflammation and restored the pattern of gene expression in brain similar to that of healthy animals. Additionally, transduction of the liver due to passage of vectors to the circulation led to whole-body disease correction. Treated animals also showed reversal of behavioural deficits and extended lifespan. Importantly, when the levels of enzymatic activity were monitored in the CSF of dogs following administration of canine NAGLU-coding vectors to animals that were either naïve or had pre-existing immunity against AAV9, similar levels of activity were achieved, suggesting that CNS efficacy would not be compromised in patients seropositive for AAV9. Our studies provide a strong rationale for the clinical development of this novel therapeutic approach as the treatment for MPSIIIB.
Collapse
Affiliation(s)
- Albert Ribera
- Center of Animal Biotechnology and Gene Therapy, Department of Biochemistry and Molecular Biology
| | - Virginia Haurigot
- Center of Animal Biotechnology and Gene Therapy, Department of Biochemistry and Molecular Biology
| | - Miguel Garcia
- Center of Animal Biotechnology and Gene Therapy, Department of Biochemistry and Molecular Biology
| | - Sara Marcó
- Center of Animal Biotechnology and Gene Therapy, Department of Biochemistry and Molecular Biology
| | - Sandra Motas
- Center of Animal Biotechnology and Gene Therapy, Department of Biochemistry and Molecular Biology
| | - Pilar Villacampa
- Center of Animal Biotechnology and Gene Therapy, Department of Biochemistry and Molecular Biology
| | - Luca Maggioni
- Center of Animal Biotechnology and Gene Therapy, Department of Biochemistry and Molecular Biology
| | - Xavier León
- Center of Animal Biotechnology and Gene Therapy, Department of Biochemistry and Molecular Biology
| | - Maria Molas
- Center of Animal Biotechnology and Gene Therapy, Department of Biochemistry and Molecular Biology
| | - Víctor Sánchez
- Center of Animal Biotechnology and Gene Therapy, Department of Biochemistry and Molecular Biology
| | - Sergio Muñoz
- Center of Animal Biotechnology and Gene Therapy, Department of Biochemistry and Molecular Biology
| | | | - Xavier Moll
- Department of Animal Medicine and Surgery and
| | - Martí Pumarola
- Center of Animal Biotechnology and Gene Therapy, Department of Animal Medicine and Surgery and
| | - Federico Mingozzi
- Généthon, 91000 Evry, France and University Pierre and Marie Curie, 75005 Paris, France
| | - Jesús Ruberte
- Center of Animal Biotechnology and Gene Therapy, Department of Animal Health and Anatomy, School of Veterinary Medicine, Universitat Autònoma de Barcelona, 08193 Bellaterra, Spain
| | - Sònia Añor
- Department of Animal Medicine and Surgery and
| | - Fatima Bosch
- Center of Animal Biotechnology and Gene Therapy, Department of Biochemistry and Molecular Biology,
| |
Collapse
|
56
|
Krawiec P, Pac-Kożuchowska E, Mełges B, Mroczkowska-Juchkiewicz A, Skomra S, Pawłowska-Kamieniak A, Kominek K. From hypertransaminasemia to mucopolysaccharidosis IIIA. Ital J Pediatr 2014; 40:97. [PMID: 25439061 PMCID: PMC4260237 DOI: 10.1186/s13052-014-0097-z] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/26/2014] [Accepted: 11/17/2014] [Indexed: 01/16/2023] Open
Abstract
UNLABELLED ᅟ: Mucopolysaccharidosis type III (MPS III; Sanfilippo syndrome) is a metabolic disorder characterized by the deficiency of a lysosomal enzyme catalyzing the catabolic pathway of heparan sulphate. MPS III presents with progressive mental deterioration, speech delay and behavioural problems with subtle somatic features, which can often lead to misdiagnosis with idiopathic developmental/speech delay, attention deficit/hyperactivity disorder or autism. We report a case of a 5-year-old boy with developmental delay and behaviour problems admitted to the Department of Paediatrics due to chronic hypertransaminasemia. The patient developed normally until the age of 2 years when he was referred to a paediatric neurologist for suspected motor and speech delay. Liver function tests were unexpectedly found elevated at the age of 3.5 years. Physical examination revealed obesity, mildly coarse facial features and stocky hands. He showed mental retardation and mild motor delay. The clinical picture strongly suggested mucopolysaccharidosis. The diagnosis of MPS IIIA was confirmed by decreased activity of heparan N-sulfatase in leucocytes. CONCLUSION We strongly recommend screening for MPS III in children with severe behavioural abnormalities with hyperactivity, psychomotor or speech deterioration and failure to achieve early developmental milestones particularly with facial dysmorphism.
Collapse
Affiliation(s)
- Paulina Krawiec
- Department of Paediatrics, Medical University of Lublin, Racławickie 1, 20-059, Lublin, Poland. .,Department of Paediatrics, Children's University Hospital in Lublin, Gębali 6, 20-093, Lublin, Poland.
| | - Elżbieta Pac-Kożuchowska
- Department of Paediatrics, Medical University of Lublin, Racławickie 1, 20-059, Lublin, Poland. .,Department of Paediatrics, Children's University Hospital in Lublin, Gębali 6, 20-093, Lublin, Poland.
| | - Beata Mełges
- Department of Paediatrics, Medical University of Lublin, Racławickie 1, 20-059, Lublin, Poland. .,Department of Paediatrics, Children's University Hospital in Lublin, Gębali 6, 20-093, Lublin, Poland.
| | - Agnieszka Mroczkowska-Juchkiewicz
- Department of Paediatrics, Medical University of Lublin, Racławickie 1, 20-059, Lublin, Poland. .,Department of Paediatrics, Children's University Hospital in Lublin, Gębali 6, 20-093, Lublin, Poland.
| | - Stanisław Skomra
- Department of Paediatrics, Children's University Hospital in Lublin, Gębali 6, 20-093, Lublin, Poland.
| | - Agnieszka Pawłowska-Kamieniak
- Department of Paediatrics, Medical University of Lublin, Racławickie 1, 20-059, Lublin, Poland. .,Department of Paediatrics, Children's University Hospital in Lublin, Gębali 6, 20-093, Lublin, Poland.
| | - Katarzyna Kominek
- Department of Paediatrics, Medical University of Lublin, Racławickie 1, 20-059, Lublin, Poland. .,Department of Paediatrics, Children's University Hospital in Lublin, Gębali 6, 20-093, Lublin, Poland.
| |
Collapse
|
57
|
Kowalewski B, Heimann P, Ortkras T, Lüllmann-Rauch R, Sawada T, Walkley SU, Dierks T, Damme M. Ataxia is the major neuropathological finding in arylsulfatase G-deficient mice: similarities and dissimilarities to Sanfilippo disease (mucopolysaccharidosis type III). Hum Mol Genet 2014; 24:1856-68. [PMID: 25452429 DOI: 10.1093/hmg/ddu603] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
Deficiency of arylsulfatase G (ARSG) leads to a lysosomal storage disease in mice resembling biochemical and pathological features of the mucopolysaccharidoses and particularly features of mucopolysaccharidosis type III (Sanfilippo syndrome). Here we show that Arsg KO mice share common neuropathological findings with other Sanfilippo syndrome models and patients, but they can be clearly distinguished by the limitation of most phenotypic alterations to the cerebellum, presenting with ataxia as the major neurological finding. We determined in detail the expression of ARSG in the central nervous system and observed highest expression in perivascular macrophages (which are characterized by abundant vacuolization in Arsg KO mice) and oligodendrocytes. To gain insight into possible mechanisms leading to ataxia, the pathology in older adult mice (>12 months) was investigated in detail. This study revealed massive loss of Purkinje cells and gliosis in the cerebellum, and secondary accumulation of glycolipids like GM2 and GM3 gangliosides and unesterified cholesterol in surviving Purkinje cells, as well as neurons of some other brain regions. The abundant presence of ubiquitin and p62-positive aggregates in degenerating Purkinje cells coupled with the absence of significant defects in macroautophagy is consistent with lysosomal membrane permeabilization playing a role in the pathogenesis of Arsg-deficient mice and presumably Sanfilippo disease in general. Our data delineating the phenotype of mucopolysaccharidosis IIIE in a mouse KO model should help in the identification of possible human cases of this disease.
Collapse
Affiliation(s)
- Björn Kowalewski
- Department of Chemistry, Biochemistry I, Bielefeld University, 33615 Bielefeld, Germany
| | - Peter Heimann
- Molecular Neurobiology, Faculty of Biology, Bielefeld University, 33615 Bielefeld, Germany
| | - Theresa Ortkras
- Department of Chemistry, Biochemistry I, Bielefeld University, 33615 Bielefeld, Germany
| | | | - Tomo Sawada
- The Dominick P. Purpura Department of Neuroscience, Rose F. Kennedy Intellectual and Developmental Disabilities Research Center, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Steven U Walkley
- The Dominick P. Purpura Department of Neuroscience, Rose F. Kennedy Intellectual and Developmental Disabilities Research Center, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Thomas Dierks
- Department of Chemistry, Biochemistry I, Bielefeld University, 33615 Bielefeld, Germany
| | - Markus Damme
- Department of Chemistry, Biochemistry I, Bielefeld University, 33615 Bielefeld, Germany,
| |
Collapse
|
58
|
Ribeiro EM, Brusius-Facchin AC, Leistner-Segal S, da Silva CAB, Schwartz IV. Cardiac disease as the presenting feature of mucopolysaccharidosis type IIIA: A case report. Mol Genet Metab Rep 2014; 1:422-424. [PMID: 27896117 PMCID: PMC5121363 DOI: 10.1016/j.ymgmr.2014.09.003] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2014] [Revised: 09/01/2014] [Accepted: 09/01/2014] [Indexed: 11/29/2022] Open
Abstract
Severe cardiac involvement is a common feature of mucopolysaccharidoses (MPS), but occurs only rarely in MPS III (Sanfilippo syndrome). We report herein a case of MPS III-A having cardiac involvement as its first manifestation. Analysis of the SGSH gene showed homozygosity for the novel mutation p.G80V. We propose that MPS disorders, including MPS III-A, should be included in the differential diagnosis of every case of cardiomyopathy presenting during the first year of life.
Collapse
Affiliation(s)
- Erlane Marques Ribeiro
- Postgraduate Program in Health Science, Universidade Federal do Rio Grande do Norte, Caixa Postal 1524, Campus Universitário Lagoa Nova, 59078-970 Natal, RN, Brazil; Hospital Infantil Albert Sabin, Secretaria de Saúde do Estado do Ceará, Rua Tertuliano Sales, 544, 60410-790 Fortaleza, CE, Brazil
| | - Ana Carolina Brusius-Facchin
- Medical Genetics Service, Hospital de Clínicas de Porto Alegre, Rua Ramiro Barcelos, 2350, 90035-903 Porto Alegre, RS, Brazil
| | - Sandra Leistner-Segal
- Medical Genetics Service, Hospital de Clínicas de Porto Alegre, Rua Ramiro Barcelos, 2350, 90035-903 Porto Alegre, RS, Brazil; Postgraduate Program in Medicine: Medical Sciences, Universidade Federal do Rio Grande do Sul, Av. Paulo Gama, 110. Bairro Farroupilha, 90040-060 Porto Alegre, RS, Brazil
| | - Carlos Antônio Bruno da Silva
- Postgraduate Program in Health Science, Universidade Federal do Rio Grande do Norte, Caixa Postal 1524, Campus Universitário Lagoa Nova, 59078-970 Natal, RN, Brazil
| | - Ida Vanessa Schwartz
- Medical Genetics Service, Hospital de Clínicas de Porto Alegre, Rua Ramiro Barcelos, 2350, 90035-903 Porto Alegre, RS, Brazil; Department of Genetics, Universidade Federal do Rio Grande do Sul, Av. Paulo Gama, 110. Bairro Farroupilha, 90040-060 Porto Alegre, RS, Brazil
| |
Collapse
|
59
|
Bodamer OA, Giugliani R, Wood T. The laboratory diagnosis of mucopolysaccharidosis III (Sanfilippo syndrome): A changing landscape. Mol Genet Metab 2014; 113:34-41. [PMID: 25127543 DOI: 10.1016/j.ymgme.2014.07.013] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/05/2014] [Revised: 07/11/2014] [Accepted: 07/11/2014] [Indexed: 12/24/2022]
Abstract
Mucopolysaccharidosis type III (MPS III) is characterized by progressive neurological deterioration, behavioral abnormalities, a relatively mild somatic phenotype, and early mortality. Because of the paucity of somatic manifestations and the rarity of the disease, early diagnosis is often difficult. Therapy targeting the underlying disease pathophysiology may offer the greatest clinical benefit when started prior to the onset of significant neurologic sequelae. Here we review current practices in the laboratory diagnosis of MPS III in order to facilitate earlier patient identification and diagnosis. When clinical suspicion of MPS III arises, the first step is to order a quantitative assay that screens urine for the presence of glycosaminoglycan biomarkers using a spectrophotometric compound (e.g., dimethylmethylene blue). We recommend testing all patients with developmental delay and/or behavioral abnormalities as part of the diagnostic work-up because quantitative urine screening is inexpensive and non-invasive. Semi-quantitative urine screening assays using cationic dyes on filter paper (e.g., spot tests) have relatively high rates of false-positives and false-negatives and are obsolete. Of note, a negative urinary glycosaminoglycan assay does not necessarily rule out MPS because, in some patients, an overlap in excretion levels with healthy controls may occur. All urine samples that test positive for glycosaminoglycans with a quantitative assay should be confirmed by electrophoresis, thin layer chromatography, or tandem mass spectrometry, which further improves the sensitivity and specificity. The gold standard for diagnosis remains the enzyme activity assay in cultured skin fibroblasts, leukocytes, plasma, or serum, which can be used as a first-line diagnostic test in some regions. Molecular genetic analysis should be offered to all families of patients to allow genetic counseling for informed family planning. For a small number of variants, genotype-phenotype correlations are available and can offer prognostic value. Prenatal testing via enzyme activity assay in chorionic villi or amniotic fluid cells is available at a limited number of centers worldwide, but whenever possible, a molecular genetic analysis is preferred for prenatal diagnosis. To conclude, we discuss the development of newborn screening assays in dried blood spots and high-throughput methods for sequencing the protein-coding regions of the genome (whole exome sequencing) and their relevance to future changes in the MPS III diagnostic landscape.
Collapse
Affiliation(s)
- Olaf A Bodamer
- Division of Clinical and Translational Genetics, Dr. John T. MacDonald Foundation, Department of Human Genetics, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Roberto Giugliani
- Department of Genetics/UFRGS, Medical Genetics Service/HCPA and INAGEMP, Porto Alegre, RS, Brazil
| | - Tim Wood
- Metabolic Laboratory, Greenwood Genetic Center, Greenwood, SC, USA.
| |
Collapse
|
60
|
Steele R, Siden H, Cadell S, Davies B, Andrews G, Feichtinger L, Singh M, Spicer S, Goez H, Davies D, Rapoport A, Vadeboncoeur C, Liben S, Gregoire MC, Schwantes S, Friedrichsdorf SJ. Charting the territory: symptoms and functional assessment in children with progressive, non-curable conditions. Arch Dis Child 2014; 99:754-62. [PMID: 24833792 DOI: 10.1136/archdischild-2013-305246] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Abstract
BACKGROUND Children with progressive, non-curable genetic, metabolic, or neurological conditions require specialised care to enhance their quality of life. Prevention and relief of physical symptoms for these children needs to begin at diagnosis, yet, little is known about their patterns of symptoms and functional abilities. AIM To describe these children's symptoms, as well as how the children's condition affects them physically. DESIGN Cross-sectional, baseline results from an observational, longitudinal study, Charting the Territory, that followed 275 children and their families. SETTING/PARTICIPANTS Seven tertiary care children's hospitals in Canada, 2 in the USA. Families were eligible based on the child's condition. A total of 275 children from 258 families participated. RESULTS The 3 most common symptoms in these children were pain, sleep problems, and feeding difficulties; on average, they had 3.2 symptoms of concern. There was a pattern of under-reporting of children's symptoms for clinicians compared with parents. Regardless of use of associated medications, pain, feeding and constipation symptoms were often frequent and distressing. Children with a G/J tube had a higher total number of symptoms, and respiratory problems, pain, feeding difficulties and constipation were more likely to occur. They also tended to have frequent and distressing symptoms, and to need extensive mobility modifications which, in turn, were associated with higher numbers of symptoms. CONCLUSIONS These children experience multiple symptoms that have been previously documented individually, but not collectively. Effective interventions are needed to reduce their symptom burden. Future longitudinal analyses will examine which disease-modifying interventions improve, or do not improve, symptom burden.
Collapse
Affiliation(s)
- Rose Steele
- School of Nursing, Faculty of Health, York University, Toronto, Ontario, Canada
| | - Harold Siden
- Department of Paediatrics, University of British Columbia; Canuck Place Children's Hospice, Vancouver, British Columbia, Canada Developmental Neuroscience and Child Health, Child & Family Research Institute, Vancouver, British Columbia, Canada
| | - Susan Cadell
- School of Social Work, Renison University College-University of Waterloo, Waterloo, Ontario, Canada
| | - Betty Davies
- School of Nursing, University of Victoria, Victoria, British Columbia, Canada
| | - Gail Andrews
- Developmental Neuroscience and Child Health, Child & Family Research Institute, Vancouver, British Columbia, Canada
| | - Leanne Feichtinger
- Developmental Neuroscience and Child Health, Child & Family Research Institute, Vancouver, British Columbia, Canada
| | - Mina Singh
- School of Nursing, Faculty of Health, York University, Toronto, Ontario, Canada
| | | | | | | | | | | | | | | | | | | |
Collapse
|
61
|
Tardieu M, Zérah M, Husson B, de Bournonville S, Deiva K, Adamsbaum C, Vincent F, Hocquemiller M, Broissand C, Furlan V, Ballabio A, Fraldi A, Crystal RG, Baugnon T, Roujeau T, Heard JM, Danos O. Intracerebral administration of adeno-associated viral vector serotype rh.10 carrying human SGSH and SUMF1 cDNAs in children with mucopolysaccharidosis type IIIA disease: results of a phase I/II trial. Hum Gene Ther 2014; 25:506-16. [PMID: 24524415 DOI: 10.1089/hum.2013.238] [Citation(s) in RCA: 189] [Impact Index Per Article: 18.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
Mucopolysaccharidosis type IIIA is a severe degenerative disease caused by an autosomal recessive defect of a gene encoding a lysosomal heparan-N-sulfamidase, the N-sulfoglycosamine sulfohydrolase (SGSH), the catalytic site of which is activated by a sulfatase-modifying factor (SUMF1). Four children (Patients 1-3, aged between 5.5 and 6 years; Patient 4 aged 2 years 8 months) received intracerebral injections of an adeno-associated viral vector serotype rh.10-SGSH-IRES-SUMF1 vector in a phase I/II clinical trial. All children were able to walk, but their cognitive abilities were abnormal and had declined (Patients 1-3). Patients 1-3 presented with brain atrophy. The therapeutic vector was delivered in a frameless stereotaxic device, at a dose of 7.2×10(11) viral genomes/patient simultaneously via 12 needles as deposits of 60 μl over a period of 2 hr. The vector was delivered bilaterally to the white matter anterior, medial, and posterior to the basal ganglia. Immunosuppressive treatment (mycophenolate mofetil and tacrolimus) was initiated 15 days before surgery and maintained for 8 weeks (mycophenolate mofetil) or throughout follow-up (tacrolimus, with progressive dose reduction) to prevent elimination of transduced cells. Safety data collected from inclusion, during the neurosurgery period and over the year of follow-up, showed good tolerance, absence of adverse events related to the injected product, no increase in the number of infectious events, and no biological sign of toxicity related to immunosuppressive drugs. Efficacy analysis was necessarily preliminary in this phase I/II trial on four children, in the absence of validated surrogate markers. Brain atrophy evaluated by magnetic resonance imaging seemed to be stable in Patients 1 and 3 but tended to increase in Patients 2 and 4. Neuropsychological evaluations suggested a possible although moderate improvement in behavior, attention, and sleep in Patients 1-3. The youngest patient was the most likely to display neurocognitive benefit.
Collapse
Affiliation(s)
- Marc Tardieu
- 1 Pediatric Neurology, Assistance Publique-Hôpitaux de Paris, Hôpitaux Universitaires Paris-Sud , Le Kremlin Bucêtre 94275, France
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
62
|
Buhrman D, Thakkar K, Poe M, Escolar ML. Natural history of Sanfilippo syndrome type A. J Inherit Metab Dis 2014; 37:431-7. [PMID: 24271936 DOI: 10.1007/s10545-013-9661-8] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/04/2013] [Revised: 10/18/2013] [Accepted: 10/23/2013] [Indexed: 01/05/2023]
Abstract
OBJECTIVE To describe the natural history of Sanfilippo syndrome type A. METHODS We performed a retrospective review of 46 children (21 boys, 25 girls) with Sanfilippo syndrome type A evaluated between January 2000 and April 2013. Assessments included neurodevelopmental evaluations, audiologic testing, and assessment of growth, adaptive behavior, cognitive behavior, motor function, and speech/language skills. Only the baseline evaluation was included for patients who received hematopoietic stem cell transplantation. RESULTS Median age at diagnosis was 35 months, with a median delay between initial symptoms to diagnosis of 24 months. The most common initial symptoms were speech/language delay (48%), dysmorphology (22%), and hearing loss (20%). Early behavioral problems included perseverative chewing and difficulty with toilet training. All children developed sleep difficulties and behavioral changes (e.g., hyperactivity, aggression). More than 93% of the children experienced somatic symptoms such as hepatomegaly (67%), abnormal dentition (39%), enlarged tongue (37%), coarse facial features (76%), and protuberant abdomen (43%). Kaplan-Meier analysis showed a 60% probability of surviving past 17 years of age. CONCLUSIONS Sanfilippo type A is characterized by severe hearing loss and speech delay, followed by a rapid decline in cognitive skills by 3 years of age. Significant somatic disease occurs in more than half of patients. Behavioral difficulties presented between 2 and 4 years of age during a rapid period of cognitive decline. Gross motor abilities are maintained during this period, which results in an active child with impaired cognition. Sleep difficulties are concurrent with the period of cognitive degeneration. There is currently an unacceptable delay in diagnosis, highlighting the need to increase awareness of this disease among clinicians.
Collapse
Affiliation(s)
- Dakota Buhrman
- University of Pittsburgh and Children's Hospital of Pittsburgh of UPMC, 4401 Penn Ave, Pittsburgh, PA, 15213, USA
| | | | | | | |
Collapse
|
63
|
Mahon LV, Lomax M, Grant S, Cross E, Hare DJ, Wraith JE, Jones S, Bigger B, Langford-Smith K, Canal M. Assessment of sleep in children with mucopolysaccharidosis type III. PLoS One 2014; 9:e84128. [PMID: 24504123 PMCID: PMC3913580 DOI: 10.1371/journal.pone.0084128] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2013] [Accepted: 11/12/2013] [Indexed: 01/06/2023] Open
Abstract
Sleep disturbances are prevalent in mucopolysaccharidosis Type III (MPS III), yet there is a lack of objective, ecologically valid evidence detailing sleep quantity, quality or circadian system. Eight children with MPS III and eight age-matched typically developing children wore an actigraph for 7–10 days/nights. Saliva samples were collected at three time-points on two separate days, to permit analysis of endogenous melatonin levels. Parents completed a sleep questionnaire and a daily sleep diary. Actigraphic data revealed that children with MPS III had significantly longer sleep onset latencies and greater daytime sleep compared to controls, but night-time sleep duration did not differ between groups. In the MPS III group, sleep efficiency declined, and sleep onset latency increased, with age. Questionnaire responses showed that MPS III patients had significantly more sleep difficulties in all domains compared to controls. Melatonin concentrations showed an alteration in the circadian system in MPS III, which suggests that treatment for sleep problems should attempt to synchronise the sleep-wake cycle to a more regular pattern. Actigraphy was tolerated by children and this monitoring device can be recommended as a measure of treatment success in research and clinical practice.
Collapse
Affiliation(s)
- Louise Victoria Mahon
- Division of Clinical Psychology, University of Manchester, Manchester, United Kingdom
- * E-mail:
| | - Michelle Lomax
- Division of Clinical Psychology, University of Manchester, Manchester, United Kingdom
| | - Sheena Grant
- Division of Clinical Psychology, University of Manchester, Manchester, United Kingdom
| | - Elaine Cross
- Division of Clinical Psychology, University of Manchester, Manchester, United Kingdom
| | - Dougal Julian Hare
- Division of Clinical Psychology, University of Manchester, Manchester, United Kingdom
| | - James Ed Wraith
- Genetic Medicine, St. Mary’s Hospital, Manchester, United Kingdom
| | - Simon Jones
- Genetic Medicine, St. Mary’s Hospital, Manchester, United Kingdom
| | - Brian Bigger
- Faculty of Medical and Human Sciences, University of Manchester, Manchester, United Kingdom
| | - Kia Langford-Smith
- Faculty of Medical and Human Sciences, University of Manchester, Manchester, United Kingdom
| | - Maria Canal
- Faculty of Life Sciences, University of Manchester, Manchester, United Kingdom
| |
Collapse
|
64
|
Haurigot V, Bosch F. Toward a gene therapy for neurological and somatic MPSIIIA. ACTA ACUST UNITED AC 2013; 1:e27209. [PMID: 25003015 PMCID: PMC3927492 DOI: 10.4161/rdis.27209] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2013] [Revised: 11/06/2013] [Accepted: 11/14/2013] [Indexed: 01/13/2023]
Abstract
Mucopolysaccharidosis Type IIIA (MPSIIIA) represents an unmet medical need. MPSIIIA shares with many other lysosomal storage disorders (LSD) the characteristic of being a severe neurodegenerative disease accompanied by mild somatic involvement. Thus, the main target organ for the development of new treatments is the central nervous system (CNS), but overall clinical efficacy would be greatly enhanced by simultaneous correction of peripheral disease. We have recently developed a novel treatment for MPSIIIA based on the delivery to the cerebrospinal fluid of serotype 9 adeno-associated virus (AAV9)-derived vectors. This gene therapy strategy corrected both CNS and somatic pathology in animal models through widespread transduction of CNS, peripheral nervous system (PNS), and liver. The work set the grounds for the clinical translation of the approach to treat MPSIIIA in humans. Here we discuss some important considerations that further support the applicability of this treatment to MPSIIIA and other LSD with CNS and somatic involvement.
Collapse
Affiliation(s)
- Virginia Haurigot
- Center of Animal Biotechnology and Gene Therapy (CBATEG) and Department of Biochemistry and Molecular Biology; School of Veterinary Medicine; Universitat Autònoma de Barcelona; Barcelona, Spain
| | - Fatima Bosch
- Center of Animal Biotechnology and Gene Therapy (CBATEG) and Department of Biochemistry and Molecular Biology; School of Veterinary Medicine; Universitat Autònoma de Barcelona; Barcelona, Spain
| |
Collapse
|
65
|
Delgadillo V, O'Callaghan MDM, Gort L, Coll MJ, Pineda M. Natural history of Sanfilippo syndrome in Spain. Orphanet J Rare Dis 2013; 8:189. [PMID: 24314109 PMCID: PMC3879199 DOI: 10.1186/1750-1172-8-189] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2013] [Accepted: 12/01/2013] [Indexed: 01/15/2023] Open
Abstract
Background Mucopolysaccharidosis type III (MPS III), or Sanfilippo syndrome, is caused by a deficiency in one of the four enzymes involved in the lysosomal degradation of heparan sulphate. Four MPS III types have been recognized, characterized by a large phenotypic heterogeneity. This is the first Spanish study describing the natural history of Sanfilippo patients (MPSIIIA, MPSIIIB and MPSIIIC), representing an essential step for understanding patient prognosis and for the establishment and application of future therapies. Methods This retrospective study aimed to establish the natural history of MPS III in Spain based on an extensive chronological data survey involving physicians and parents of 55 Spanish MPSIII patients. In addition to clinical description we report biochemical and molecular analysis already performed in the majority of cases. Results The most frequent subtype was MPS IIIA (62%). Symptoms before diagnosis were speech delay in 85%, followed by coarse facial features in 78%, and hyperactivity in 65% of cases at a mean age of 3 years old. The median age at clinical and biochemical diagnosis for each MPS III subtype were as follows: IIIA 4.4 years (1.2 – 16 years), IIIB 3.1 years (1–29 years), and IIIC 6.3 years (3.4-22 years). 45% of patients developed epilepsy at a median age of 8.7 (2.5 – 37) years old. Age of death for MPS IIIA patients was 15 years (11.5 – 26 years). Molecular analysis of our cohort reveals, as alluded to above, a great allelic heterogeneity in the three subtypes without clear genotype-phenotype correlations in most cases. Conclusion MPS IIIA is the most frequent subtype in Spanish Sanfilippo patients. Diagnosing physicians should consider Sanfilippo syndrome in children with non-specific speech delay, behavioural abnormalities, and/or mild dysmorphic features. We stress the importance of establishing early diagnosis procedures as soon as possible so as to be able to determine future short-term enzymatic or gene therapy treatments that can change the prognosis of the disease.
Collapse
|
66
|
Garbuzova-Davis S, Mirtyl S, Sallot SA, Hernandez-Ontiveros DG, Haller E, Sanberg PR. Blood-brain barrier impairment in MPS III patients. BMC Neurol 2013; 13:174. [PMID: 24225396 PMCID: PMC3835134 DOI: 10.1186/1471-2377-13-174] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2013] [Accepted: 11/07/2013] [Indexed: 12/30/2022] Open
Abstract
Background Mucopolysaccharidosis type III (MPS III) is an autosomal recessive disorder caused by deficiency of a specific enzyme leading to heparan sulfate (HS) accumulation within cells and to eventual progressive cerebral and systemic organ abnormalities. Different enzyme deficiencies comprise the MPS III subcategories (A, B, C, D). Since neuropathological manifestations are common to all MPS III types, determining blood-brain barrier (BBB) condition may be critical to understand potential additional disease mechanisms. Methods We investigated BBB integrity in various brain structures of post-mortem tissues from an eleven year old Caucasian female with MPS III A and from a twenty four year old Caucasian female with MPS III D. Control tissues were obtained post-mortem from three Caucasians without neurological deficits: a twelve year old male, a twenty four year old female, and a twenty seven year old female. BBB capillary ultrastructure (electron microscopy) and capillary functional integrity (IgG leakage, tight junction proteins, and lysosomal accumulation within endothelium) were examined. Results Compromised BBB integrity was found in both MPS III cases. Major study findings were: (1) capillary endothelial and pericyte cell damage; (2) mucopolysaccharide bodies in a majority of endothelial cells and pericytes rupturing cell membranes; (3) severe extracellular edema; (4) IgG microvascular leakage and reductions of occludin and claudin-5 with variations between MPS III types; (5) extensive lysosomal accumulation in capillary endothelium. Conclusions These new findings of BBB structural and functional impairment, although from only two cases, MPS III A and III D, may have implications for disease pathogenesis and should be considered in treatment development for MPS III.
Collapse
Affiliation(s)
- Svitlana Garbuzova-Davis
- Center of Excellence for Aging & Brain Repair, University of South Florida, Morsani College of Medicine, 12901 Bruce B, Downs Blvd, Tampa, FL 33612, USA.
| | | | | | | | | | | |
Collapse
|
67
|
Delaney KA, Rudser KR, Yund BD, Whitley CB, Haslett PAJ, Shapiro EG. Methods of neurodevelopmental assessment in children with neurodegenerative disease: Sanfilippo syndrome. JIMD Rep 2013; 13:129-37. [PMID: 24190801 DOI: 10.1007/8904_2013_269] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/25/2013] [Revised: 08/07/2013] [Accepted: 09/25/2013] [Indexed: 12/13/2022] Open
Abstract
OBJECTIVES (1) Develop a methodology for obtaining reliable cognitive and developmental data in children with neurodegenerative disease and cognitive impairment and in turn monitor disease state and treatment outcomes. (2) Demonstrate validity of age-equivalent scores. METHODS We present guidelines for obtaining accurate test scores in low-functioning and behaviorally disruptive pediatric patients, followed by a method validation study: (1) using disease-specific protocols to assess salient aspects of the known phenotype, (2) selecting appropriate tests, (3) managing behavior, and (4) using age-equivalent scores on standardized tools. We used the Bayley Scales of Infant Development-III or Kaufman Assessment Battery for Children-II with a group of 25 children with mucopolysaccharidosis type IIIA (MPS IIIA or Sanfilippo syndrome type A) with dementia. To demonstrate concurrent validity, we used the Vineland Adaptive Behavior Scales-II, comparing parent-reported age-equivalent scores (AEs) with those of the cognitive measures. RESULTS We were successful in obtaining cognitive age-equivalents for 25 patients with MPS IIIA including those with severe behavioral disruption and a correlation of 0.95 was obtained comparing scores on the parent measure with cognitive age-equivalents validating the age-equivalent approach. CONCLUSION An approach to the assessment of severely impaired children including those with behavioral disruption was implemented and is applicable to children with other severe neurological diseases. This approach will enhance the assessment of disease progression and monitoring of treatment outcome in clinical trials.
Collapse
Affiliation(s)
- Kathleen A Delaney
- Department of Pediatrics, University of Minnesota, Minneapolis, MN, USA,
| | | | | | | | | | | |
Collapse
|
68
|
Meijer OLM, van Vlies N, Wijburg FA. Treatment of mucopolysaccharidosis type III (Sanfilippo syndrome). Expert Opin Orphan Drugs 2013. [DOI: 10.1517/21678707.2013.830069] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Affiliation(s)
- Olga LM Meijer
- University of Amsterdam, Academic Medical Centre, Department of Pediatrics and Amsterdam Lysosome Centre ‘Sphinx', Meibergdreef 9, 1105 AZ, Amsterdam, The Netherlands ;
| | - Naomi van Vlies
- University of Amsterdam, Academic Medical Centre, Department of Pediatrics and Amsterdam Lysosome Centre ‘Sphinx', Meibergdreef 9, 1105 AZ, Amsterdam, The Netherlands ;
- University of Amsterdam, Academic Medical Centre, Department of Clinical Chemistry and Pediatrics, Lab Genetic Metabolic Diseases, Meibergdreef 9, 1105 AZ, Amsterdam, The Netherlands
| | - Frits A Wijburg
- University of Amsterdam, Academic Medical Centre, Department of Pediatrics and Amsterdam Lysosome Centre ‘Sphinx', Meibergdreef 9, 1105 AZ, Amsterdam, The Netherlands ;
| |
Collapse
|
69
|
Kim KH, Dodsworth C, Paras A, Burton BK. High dose genistein aglycone therapy is safe in patients with mucopolysaccharidoses involving the central nervous system. Mol Genet Metab 2013; 109:382-5. [PMID: 23845234 DOI: 10.1016/j.ymgme.2013.06.012] [Citation(s) in RCA: 50] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/08/2013] [Revised: 06/14/2013] [Accepted: 06/15/2013] [Indexed: 01/02/2023]
Abstract
Genistein (4,5,7-trihydroexyisoflavone), a soy derived isoflavone, has been proposed as a substrate reduction therapy in patients with mucopolysaccharidoses (MPS) disorders with central nervous system involvement based on studies in cultured fibroblasts demonstrating that this agent inhibits glycosaminoglycan synthesis. Several studies have reported treatment of MPS III patients with low dose genistein (5-15mg/kg/day) with no serious adverse effects and variable neurocognitive outcomes. Mice with MPS IIIB treated with high dose (160mg/kg/day) genistein exhibited a significant decrease in heparan sulfate accumulation and neuroinflammation in the brain and improvement of the behavioral phenotype. No study to date has been performed using high dose genistein treatment in MPS patients. We initiated an open label study to assess the safety of high dose genistein treatment in MPS patients with neurological impairment. Twenty-two eligible patients were treated at least 12months with pure synthetic genistein at a dose of 150mg/kg/day. Safety labs, urine GAG levels, clinical status and history of adverse events were obtained every 3months and physical examination was performed by single examiner at least every 12months. While neurocognitive level was not a primary endpoint, participants were asked to obtain annual neurocognitive testing if available and a 9 point disability scale (FPSS) was recorded at each study visit. In the course of 12months of treatment, we observed no serious adverse events that were unexplained by the underlying condition and no severe adverse events that were felt to be potentially related to the genistein therapy. Two male subjects developed Tanner II breast development not present at baseline which could be related to the mild estrogenic effects of genistein. We observed no consistent decline in urine GAG levels; however, urinary GAG excretion was erratic. After 12months, the FPSS remained unchanged in 16 patients and declined by 1 point in 2 patients. Based on the results obtained so far, high dose oral genistein therapy appears to be safe in MPS patients and additional testing in a larger randomized placebo controlled trial is needed to further assess safety and efficacy.
Collapse
Affiliation(s)
- Katherine H Kim
- Ann & Robert H. Lurie Children's Hospital of Chicago, 225 E. Chicago Ave, Chicago, IL 60611, USA.
| | | | | | | |
Collapse
|
70
|
Haurigot V, Marcó S, Ribera A, Garcia M, Ruzo A, Villacampa P, Ayuso E, Añor S, Andaluz A, Pineda M, García-Fructuoso G, Molas M, Maggioni L, Muñoz S, Motas S, Ruberte J, Mingozzi F, Pumarola M, Bosch F. Whole body correction of mucopolysaccharidosis IIIA by intracerebrospinal fluid gene therapy. J Clin Invest 2013; 123:3254-3271. [PMID: 23863627 DOI: 10.1172/jci66778] [Citation(s) in RCA: 164] [Impact Index Per Article: 14.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2012] [Accepted: 04/25/2013] [Indexed: 01/12/2023] Open
Abstract
For most lysosomal storage diseases (LSDs) affecting the CNS, there is currently no cure. The BBB, which limits the bioavailability of drugs administered systemically, and the short half-life of lysosomal enzymes, hamper the development of effective therapies. Mucopolysaccharidosis type IIIA (MPS IIIA) is an autosomic recessive LSD caused by a deficiency in sulfamidase, a sulfatase involved in the stepwise degradation of glycosaminoglycan (GAG) heparan sulfate. Here, we demonstrate that intracerebrospinal fluid (intra-CSF) administration of serotype 9 adenoassociated viral vectors (AAV9s) encoding sulfamidase corrects both CNS and somatic pathology in MPS IIIA mice. Following vector administration, enzymatic activity increased throughout the brain and in serum, leading to whole body correction of GAG accumulation and lysosomal pathology, normalization of behavioral deficits, and prolonged survival. To test this strategy in a larger animal, we treated beagle dogs using intracisternal or intracerebroventricular delivery. Administration of sulfamidase-encoding AAV9 resulted in transgenic expression throughout the CNS and liver and increased sulfamidase activity in CSF. High-titer serum antibodies against AAV9 only partially blocked CSF-mediated gene transfer to the brains of dogs. Consistently, anti-AAV antibody titers were lower in CSF than in serum collected from healthy and MPS IIIA-affected children. These results support the clinical translation of this approach for the treatment of MPS IIIA and other LSDs with CNS involvement.
Collapse
|
71
|
Myeloid/Microglial driven autologous hematopoietic stem cell gene therapy corrects a neuronopathic lysosomal disease. Mol Ther 2013; 21:1938-49. [PMID: 23748415 PMCID: PMC3808137 DOI: 10.1038/mt.2013.141] [Citation(s) in RCA: 82] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2013] [Accepted: 05/30/2013] [Indexed: 02/07/2023] Open
Abstract
Mucopolysaccharidosis type IIIA (MPSIIIA) is a lysosomal storage disorder caused by mutations in N-sulfoglucosamine sulfohydrolase (SGSH), resulting in heparan sulfate (HS) accumulation and progressive neurodegeneration. There are no treatments. We previously demonstrated improved neuropathology in MPSIIIA mice using lentiviral vectors (LVs) overexpressing SGSH in wild-type (WT) hematopoietic stem cell (HSC) transplants (HSCTs), achieved via donor monocyte/microglial engraftment in the brain. However, neurological disease was not corrected using LVs in autologous MPSIIIA HSCTs. To improve brain expression via monocyte/microglial specificity, LVs expressing enhanced green fluorescent protein (eGFP) under ubiquitous phosphoglycerate kinase (PGK) or myeloid-specific promoters were compared in transplanted HSCs. LV-CD11b-GFP gave significantly higher monocyte/B-cell eGFP expression than LV-PGK-GFP or LV-CD18-GFP after 6 months. Subsequently, autologous MPSIIIA HSCs were transduced with either LV-PGK-coSGSH or LV-CD11b-coSGSH vectors expressing codon-optimized SGSH and transplanted into MPSIIIA mice. Eight months after HSCT, LV-PGK-coSGSH vectors produced bone marrow SGSH (576% normal activity) similar to LV-CD11b-coSGSH (473%), but LV-CD11b-coSGSH had significantly higher brain expression (11 versus 7%), demonstrating improved brain specificity. LV-CD11b-coSGSH normalized MPSIIIA behavior, brain HS, GM2 ganglioside, and neuroinflammation to WT levels, whereas LV-PGK-coSGSH partly corrected neuropathology but not behavior. We demonstrate compelling evidence of neurological disease correction using autologous myeloid driven lentiviral-HSC gene therapy in MPSIIIA mice.
Collapse
|
72
|
Wijburg FA, Węgrzyn G, Burton BK, Tylki‐Szymańska A. Mucopolysaccharidosis type III (Sanfilippo syndrome) and misdiagnosis of idiopathic developmental delay, attention deficit/hyperactivity disorder or autism spectrum disorder. Acta Paediatr 2013; 102:462-70. [PMID: 23336697 PMCID: PMC3654162 DOI: 10.1111/apa.12169] [Citation(s) in RCA: 89] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/21/2012] [Revised: 12/20/2012] [Accepted: 01/16/2013] [Indexed: 12/18/2022]
Abstract
Mucopolysaccharidosis III is a rare genetic disease characterized by progressive cognitive decline and severe hyperactivity that does not respond to stimulants. Somatic features are relatively mild. Patients are often initially misdiagnosed as having idiopathic developmental delay, attention deficit/hyperactivity disorder and/or autism spectrum disorders, putting them at risk for unnecessary testing and treatments. Conclusion: Children with developmental or speech delay, especially those with a characteristic somatic feature or behavioural abnormalities, should be screened for MPS III.
Collapse
Affiliation(s)
- Frits A Wijburg
- Department of Paediatrics Academic Medical Centre Amsterdam The Netherlands
| | - Grzegorz Węgrzyn
- Department of Molecular Biology University of Gdańsk Gdańsk Poland
| | - Barbara K Burton
- Division of Genetics, Birth Defects and Metabolism Ann & Robert H. Lurie Children's Hospital of Chicago Chicago IL USA
| | - Anna Tylki‐Szymańska
- Department of Metabolic Diseases The Children's Memorial Health Institute Warsaw Poland
| |
Collapse
|
73
|
Grant S, Cross E, Wraith JE, Jones S, Mahon L, Lomax M, Bigger B, Hare D. Parental social support, coping strategies, resilience factors, stress, anxiety and depression levels in parents of children with MPS III (Sanfilippo syndrome) or children with intellectual disabilities (ID). J Inherit Metab Dis 2013; 36:281-91. [PMID: 23151683 DOI: 10.1007/s10545-012-9558-y] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/30/2012] [Revised: 10/14/2012] [Accepted: 10/23/2012] [Indexed: 10/27/2022]
Abstract
Mucopolysaccharidosis type III (MPS III, Sanfilippo syndrome) is a lysosomal storage disorder, caused by a deficiency in one of four enzymes involved in the catabolism of the glycosaminoglycan heparan sulphate. It is a degenerative disorder, with a progressive decline in children's intellectual and physical functioning. There is currently no cure for the disorder. To date there is a paucity of research on how this disorder impacts parents psychological functioning. Specifically, research in the area has failed to employ adequate control groups to assess if the impact of this disorder on parents psychological functioning differs from parenting a child with intellectual disability (ID). The current study examined child behaviour and parental psychological functioning in 23 parents of children with MPS III and 23 parents of children with ID. Parents completed postal questionnaires about their child's behaviour and abilities and their own psychological functioning. Parents of children with MPS III reported fewer behavioural difficulties as their child aged, more severe level of intellectual disability, and similar levels of perceived social support, coping techniques, stress, anxiety and depression levels as parents of children with ID. Both groups of parents scored above the clinical cut off for anxiety and depression. Parents of children with MPS III rated themselves as significantly less future-orientated and goal directed than parents of children with ID. Services should develop support packages for parents of children with MPS III that incorporate an understanding of the unique stressors and current-difficulty approach of this population. Future research should examine gender differences between parental psychological functioning, using mixed qualitative and quantitative approaches, and utilise matched developmental level and typically developing control groups.
Collapse
Affiliation(s)
- Sheena Grant
- Division of Clinical Psychology, School of Psychological Sciences, University of Manchester Clin. Psy. D. Programme, Brunswick Street, Manchester, UK.
| | | | | | | | | | | | | | | |
Collapse
|
74
|
Cross EM, Hare DJ. Behavioural phenotypes of the mucopolysaccharide disorders: a systematic literature review of cognitive, motor, social, linguistic and behavioural presentation in the MPS disorders. J Inherit Metab Dis 2013; 36:189-200. [PMID: 23385295 DOI: 10.1007/s10545-012-9572-0] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/25/2012] [Revised: 11/25/2012] [Accepted: 11/29/2012] [Indexed: 11/24/2022]
Abstract
BACKGROUND The mucopolysaccharide disorders (MPS) are a group of recessively inherited metabolic disorders resulting in progressive physical and cognitive decline. MATERIALS AND METHODS MEDLINE, PsycINFO and Embase databases were searched, alongside manual screening, to identify relevant literature. Papers were included in the review if they were published in a peer reviewed journal and conducted empirical research into cognitive, motor, social or linguistic development or behaviour in one or more MPS disorders. RESULTS Twenty-five papers were reviewed. Two papers used methodology of a sufficiently high standard to demonstrate a behavioural phenotype; both found sleep disturbance to be part of the phenotype of MPS III. Fearfulness and sleep disturbance were frequently observed in people with MPS I and II. Cognitive and motor impairment and decline, and challenging behaviour were highly prevalent in the severe form of MPS II. Cognitive decline and severe behavioural problems relating to aggression, hyperactivity, orality, unusual affect and temper tantrums were seen in MPS III. CONCLUSIONS Sleep disturbance is part of the behavioural phenotype of MPS III, and challenging behaviour is highly prevalent in MPS II and MPS III, therefore the efficacy of behavioural interventions for these populations should be investigated. Further research into the behaviour and adaptive skills of children with MPS III and MPS IV is required.
Collapse
Affiliation(s)
- E M Cross
- Department of Clinical Psychology, University of Manchester, Manchester, UK.
| | | |
Collapse
|
75
|
Hematopoietic stem cell and gene therapy corrects primary neuropathology and behavior in mucopolysaccharidosis IIIA mice. Mol Ther 2012; 20:1610-21. [PMID: 22547151 DOI: 10.1038/mt.2012.82] [Citation(s) in RCA: 80] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Mucopolysaccharidosis IIIA (MPS IIIA or Sanfilippo disease) is a neurodegenerative disorder caused by a deficiency in the lysosomal enzyme sulfamidase (SGSH), catabolizing heparan sulfate (HS). Affected children present with severe behavioral abnormalities, sleep disturbances, and progressive neurodegeneration, leading to death in their second decade. MPS I, a similar neurodegenerative disease accumulating HS, is treated successfully with hematopoietic stem cell transplantation (HSCT) but this treatment is ineffectual for MPS IIIA. We compared HSCT in MPS IIIA mice using wild-type donor cells transduced ex vivo with lentiviral vector-expressing SGSH (LV-WT-HSCT) versus wild-type donor cell transplant (WT-HSCT) or lentiviral-SGSH transduced MPS IIIA cells (LV-IIIA-HSCT). LV-WT-HSCT results in 10% of normal brain enzyme activity, near normalization of brain HS and GM2 gangliosides, significant improvements in neuroinflammation and behavioral correction. Both WT-HSCT and LV-IIIA-HSCT mediated improvements in GM2 gangliosides and neuroinflammation but were less effective at reducing HS or in ameliorating abnormal HS sulfation and had no significant effect on behavior. This suggests that HS may have a more significant role in neuropathology than neuroinflammation or GM2 gangliosides. These data provide compelling evidence for the efficacy of gene therapy in conjunction with WT-HSCT for neurological correction of MPS IIIA where conventional transplant is ineffectual.
Collapse
|
76
|
Evaluation of miglustat treatment in patients with type III mucopolysaccharidosis: a randomized, double-blind, placebo-controlled study. J Pediatr 2011; 159:838-844.e1. [PMID: 21658716 DOI: 10.1016/j.jpeds.2011.04.040] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/05/2010] [Revised: 02/17/2011] [Accepted: 04/21/2011] [Indexed: 01/06/2023]
Abstract
OBJECTIVE To evaluate the efficacy and safety of oral miglustat treatment in patients with mucopolysaccharidosis type III. The primary outcome was efficacy with improvement or stabilization in at least two domains of Vineland Adaptative Behavior Scales at 6 months. The secondary outcome measured the evolution of other cognitive tests at 12 months. The safety and tolerability were assessed throughout the study. STUDY DESIGN This was a randomized, double-blind, placebo-controlled, monocenter, institutional, phase IIb to III study. In case of efficacy at 6 months, the study would go on for another 6 months on an open design with all patients receiving miglustat. In the absence of efficacy at 6 months, the trial had to be continued for 6 more months with the initial design. RESULTS After 6 months, efficacy was not superior in patients with miglustat. The independent review board confirmed continuing the study until 12 months. CONCLUSION Miglustat treatment was not associated with any improvement/stabilization in behavior problems in patients with mucopolysaccharidosis type III. Miglustat has an acceptable safety profile. However, the study has confirmed that miglustat is able to pass through the blood-brain barrier without significantly decreasing ganglioside levels.
Collapse
|
77
|
Female mucopolysaccharidosis IIIA mice exhibit hyperactivity and a reduced sense of danger in the open field test. PLoS One 2011; 6:e25717. [PMID: 22028789 PMCID: PMC3196509 DOI: 10.1371/journal.pone.0025717] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2011] [Accepted: 09/08/2011] [Indexed: 01/12/2023] Open
Abstract
Reliable behavioural tests in animal models of neurodegenerative diseases allow us to study the natural history of disease and evaluate the efficacy of novel therapies. Mucopolysaccharidosis IIIA (MPS IIIA or Sanfilippo A), is a severe, neurodegenerative lysosomal storage disorder caused by a deficiency in the heparan sulphate catabolising enzyme, sulfamidase. Undegraded heparan sulphate accumulates, resulting in lysosomal enlargement and cellular dysfunction. Patients suffer a progressive loss of motor and cognitive function with severe behavioural manifestations and premature death. There is currently no treatment. A spontaneously occurring mouse model of the disease has been described, that has approximately 3% of normal enzyme activity levels. Behavioural phenotyping of the MPS IIIA mouse has been previously reported, but the results are conflicting and variable, even after full backcrossing to the C57BL/6 background. Therefore we have independently backcrossed the MPS IIIA model onto the C57BL/6J background and evaluated the behaviour of male and female MPS IIIA mice at 4, 6 and 8 months of age using the open field test, elevated plus maze, inverted screen and horizontal bar crossing at the same circadian time point. Using a 60 minute open field, we have demonstrated that female MPS IIIA mice are hyperactive, have a longer path length, display rapid exploratory behaviour and spend less time immobile than WT mice. Female MPS IIIA mice also display a reduced sense of danger and spend more time in the centre of the open field. There were no significant differences found between male WT and MPS IIIA mice and no differences in neuromuscular strength were seen with either sex. The altered natural history of behaviour that we observe in the MPS IIIA mouse will allow more accurate evaluation of novel therapeutics for MPS IIIA and potentially other neurodegenerative disorders.
Collapse
|
78
|
Abstract
Autism is an etiologically and clinically heterogeneous group of disorders, diagnosed solely by the complex behavioral phenotype. On the basis of the high-heritability index, geneticists are confident that autism will be the first behavioral disorder for which the genetic basis can be well established. Although it was initially assumed that major genome-wide and candidate gene association studies would lead most directly to common autism genes, progress has been slow. Rather, most discoveries have come from studies of known genetic disorders associated with the behavioral phenotype. New technology, especially array chromosomal genomic hybridization, has both increased the identification of putative autism genes and raised to approximately 25%, the percentage of children for whom an autism-related genetic change can be identified. Incorporating clinical geneticists into the diagnostic and autism research arenas is vital to the field. Interpreting this new technology and deciphering autism's genetic montage require the skill set of the clinical geneticist including knowing how to acquire and interpret family pedigrees, how to analyze complex morphologic, neurologic, and medical phenotypes, sorting out heterogeneity, developing rational genetic models, and designing studies. The current emphasis on deciphering autism spectrum disorders has accelerated the field of neuroscience and demonstrated the necessity of multidisciplinary research that must include clinical geneticists both in the clinics and in the design and implementation of basic, clinical, and translational research.
Collapse
|
79
|
Langford-Smith A, Malinowska M, Langford-Smith KJ, Wegrzyn G, Jones S, Wynn R, Wraith JE, Wilkinson FL, Bigger BW. Hyperactive behaviour in the mouse model of mucopolysaccharidosis IIIB in the open field and home cage environments. GENES BRAIN AND BEHAVIOR 2011; 10:673-82. [DOI: 10.1111/j.1601-183x.2011.00706.x] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
|
80
|
Valstar MJ, Marchal JP, Grootenhuis M, Colland V, Wijburg FA. Cognitive development in patients with Mucopolysaccharidosis type III (Sanfilippo syndrome). Orphanet J Rare Dis 2011; 6:43. [PMID: 21689409 PMCID: PMC3130633 DOI: 10.1186/1750-1172-6-43] [Citation(s) in RCA: 80] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2011] [Accepted: 06/20/2011] [Indexed: 12/30/2022] Open
Abstract
Background Mucopolysaccharidosis type III (MPS III, Sanfilippo syndrome) is a lysosomal storage disorder caused by a deficiency of one of the enzymes involved in the degradation of heparan sulfate. MPS III is characterized by progressive mental deterioration resulting in severe dementia. A number of potentially disease-modifying therapies are studied. As preservation of cognitive function is the ultimate goal of treatment, assessment of cognitive development will be essential in order to evaluate treatment efficacy. However, no large scale studies on cognitive levels in MPS III patients, using formal psychometric tests, have been reported. Methods We aimed to assess cognitive development in all 73 living patients with MPS III in the Netherlands. Results Cognitive development could be assessed in 69 patients. In 39 of them developmental level was estimated > 3 months and formal psychometric testing was attempted. A remarkable variation in the intellectual disability was detected. Conclusions Despite special challenges encountered, testing failed in only three patients. The observed broad variation in intellectual disability, should be taken into account when designing therapeutic trials.
Collapse
Affiliation(s)
- Marlies J Valstar
- Department of Pediatrics and Amsterdam Lysosome Center Sphinx, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | | | | | | | | |
Collapse
|
81
|
Muschol N, Pohl S, Meyer A, Gal A, Ullrich K, Braulke T. Residual activity and proteasomal degradation of p.Ser298Pro sulfamidase identified in patients with a mild clinical phenotype of Sanfilippo A syndrome. Am J Med Genet A 2011; 155A:1634-9. [DOI: 10.1002/ajmg.a.34053] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2010] [Accepted: 03/24/2011] [Indexed: 11/06/2022]
|
82
|
Héron B, Mikaeloff Y, Froissart R, Caridade G, Maire I, Caillaud C, Levade T, Chabrol B, Feillet F, Ogier H, Valayannopoulos V, Michelakakis H, Zafeiriou D, Lavery L, Wraith E, Danos O, Heard JM, Tardieu M. Incidence and natural history of mucopolysaccharidosis type III in France and comparison with United Kingdom and Greece. Am J Med Genet A 2011; 155A:58-68. [PMID: 21204211 DOI: 10.1002/ajmg.a.33779] [Citation(s) in RCA: 115] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Sanfilippo syndrome, or mucopolysaccharidosis type III (MPSIII) is a lysosomal storage disease with predominant neurological manifestations in affected children. It is considered heterogeneous with respect to prevalence, clinical presentation, biochemistry (four biochemical forms of the disease referred to as MPSIIIA, B, C, and D are known), and causative mutations. The perspective of therapeutic options emphasizes the need for better knowledge of MPSIII incidence and natural history. We performed parallel retrospective epidemiological studies of patients diagnosed with MSPIII in France (n = 128), UK (n = 126), and Greece (n = 20) from 1990 to 2006. Incidences ranged from 0.68 per 100,000 live-births in France to 1.21 per 100,000 live-births in UK. MPSIIIA, which predominates in France and UK, was absent in Greece, where most patients have MPSIIIB. The study confirmed the large allelic heterogeneity of MPSIIIA and MPSIIIB and detected several yet undescribed mutations. Analysis of clinical manifestations at diagnosis and over a 6-7 years follow-up indicated that almost all patients, whatever the disease subtype, expressed neurological manifestations before the age of 5 years, including language acquisition delay, cognitive delay, and/or abnormal behavior. In contrast to relatively homogeneous early onset manifestations, disease progression showed significant variation depending on subtype and age at diagnosis. Different severities of disease progressions and different allele distribution between France and UK suggested that mutations are not equally deleterious, although genotype-phenotype correlation could not be established. Notwithstanding the rapidity of further clinical deterioration, all MPSIII patients suffer early onset devastating neurological manifestations that deserve early treatment when available.
Collapse
Affiliation(s)
- Bénédicte Héron
- Hôpital Trousseau, Centre de référence des maladies lysosomales, Paris, France
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
83
|
Langford-Smith KJ, Mercer J, Petty J, Tylee K, Church H, Roberts J, Moss G, Jones S, Wynn R, Wraith JE, Bigger BW. Heparin cofactor II-thrombin complex and dermatan sulphate:chondroitin sulphate ratio are biomarkers of short- and long-term treatment effects in mucopolysaccharide diseases. J Inherit Metab Dis 2011; 34:499-508. [PMID: 21170681 PMCID: PMC3063559 DOI: 10.1007/s10545-010-9254-8] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/27/2010] [Revised: 10/27/2010] [Accepted: 11/18/2010] [Indexed: 11/25/2022]
Abstract
Early detection of mucopolysaccharidosis (MPS) is an important factor in treatment success; therefore, good disease biomarkers are vital. We evaluate heparin cofactor II-thrombin complex (HCII-T) as a biomarker in serum and dried blood spots (DBS) of MPS patients. Serum HCII-T and urine dermatan sulphate:chondroitin sulphate (DS:CS) ratio are also compared longitudinally against clinical outcomes in MPSI, II and VI patients following treatment. Samples were collected from MPS patients at the Royal Manchester Children's Hospital. DS:CS ratio was obtained by measuring the area density of spots from 2D electrophoresis of urinary glycosaminoglycans. Serum and DBS HCII-T was measured by sandwich ELISA. Serum HCII-T is elevated approximately 25-fold in MPS diseases that store DS, clearly distinguishing untreated MPSI, II and VI patients from unaffected age-matched controls. Serum HCII-T is also elevated in MPSIII, which leads to storage of heparan sulphate, with an increase of approximately 4-fold over unaffected age-matched controls. Urine DS:CS ratio and serum HCII-T decrease in response to treatment of MPSI, II and VI patients. HCII-T appears to respond rapidly to perturbations in treatment, whilst DS:CS ratio responds more slowly. HCII-T is a suitable biomarker for MPSI, II and VI, and it may also be informative for MPS diseases storing HS alone, such as MPSIII, although the elevation observed is smaller. In treated MPS patients, HCII-T and DS:CS ratio appear to measure short-term and long-term treatment outcomes, respectively. The potential value of HCII-T measurement in DBS for newborn screening of MPS diseases warrants further investigation.
Collapse
Affiliation(s)
- Kia Jane Langford-Smith
- MPS Stem Cell Research Group, Faculty of Medical and Human Sciences, University of Manchester, 3.721 Stopford Building, Manchester, M13 9PT UK
| | - Jean Mercer
- Genetic Medicine, St. Mary’s Hospital, Manchester, UK
| | - June Petty
- Genetic Medicine, St. Mary’s Hospital, Manchester, UK
| | - Karen Tylee
- Genetic Medicine, St. Mary’s Hospital, Manchester, UK
| | | | - Jane Roberts
- Genetic Medicine, St. Mary’s Hospital, Manchester, UK
| | - Gill Moss
- Genetic Medicine, St. Mary’s Hospital, Manchester, UK
| | - Simon Jones
- Genetic Medicine, St. Mary’s Hospital, Manchester, UK
| | - Rob Wynn
- Blood and Marrow Transplant Unit, Royal Manchester Children’s Hospital, Manchester, UK
| | - J. Ed Wraith
- Genetic Medicine, St. Mary’s Hospital, Manchester, UK
| | - Brian W. Bigger
- MPS Stem Cell Research Group, Faculty of Medical and Human Sciences, University of Manchester, 3.721 Stopford Building, Manchester, M13 9PT UK
| |
Collapse
|
84
|
Valstar MJ, Neijs S, Bruggenwirth HT, Olmer R, Ruijter GJG, Wevers RA, van Diggelen OP, Poorthuis BJ, Halley DJ, Wijburg FA. Mucopolysaccharidosis type IIIA: clinical spectrum and genotype-phenotype correlations. Ann Neurol 2011; 68:876-87. [PMID: 21061399 DOI: 10.1002/ana.22092] [Citation(s) in RCA: 134] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
OBJECTIVE Mucopolysaccharidosis (MPS) IIIA (Sanfilippo syndrome type A) is a lysosomal storage disorder caused by deficiency of the enzyme sulfamidase. Information on the natural course of MPS IIIA is scarce, but is much needed in view of emerging therapies. METHODS Clinical history and molecular defects of all 110 MPS IIIA patients identified by enzymatic studies in the Netherlands were collected and included in this study. RESULTS First clinical signs, mainly consisting of delayed speech development and behavioral problems, were noted between the ages of 1 and 6 years. Other symptoms included sleeping and hearing problems, recurrent upper airway infections, diarrhea, and epilepsy. The clinical course varied remarkably and could be correlated with the molecular defects. The frequent pathogenic mutations p.R245H, p.Q380R, p.S66W, and c.1080delC were associated with the classical severe phenotype. Patients compound heterozygous for the p.S298P mutation in combination with 1 of the mutations associated with the classical severe phenotype had a significantly longer preservation of psychomotor functions and a longer survival. Two patients homozygous for the p.S298P mutation, and 4 patients from 3 families heterozygous for 3 missense variants not reported previously (p.T421R, p.P180L, and p.L12Q), showed a remarkably attenuated phenotype. INTERPRETATION We report the natural history and mutational analysis in a large unbiased cohort of MPS IIIA patients. We demonstrate that the clinical spectrum of MPS IIIA is much broader than previously reported. A significant genotype-phenotype correlation was established in this cohort.
Collapse
Affiliation(s)
- Marlies J Valstar
- Department of Pediatrics and Amsterdam Lysosome Center Sphinx, Academic Medical Center, University of Amsterdam, Amsterdam, the Netherlands
| | | | | | | | | | | | | | | | | | | |
Collapse
|
85
|
McIntyre C, Byers S, Anson DS. Correction of mucopolysaccharidosis type IIIA somatic and central nervous system pathology by lentiviral-mediated gene transfer. J Gene Med 2010; 12:717-28. [PMID: 20683858 DOI: 10.1002/jgm.1489] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022] Open
Abstract
BACKGROUND The hallmark of lysosomal storage disorders (LSDs) is microscopically demonstrable lysosomal distension. In mucopolysaccharidosis type IIIA (MPS IIIA), this occurs as a result of an inherited deficiency of the lysosomal hydrolase sulphamidase. Consequently, heparan sulphate, a highly sulphated glycosaminoglycan, accumulates primarily within the cells of the reticulo-endothelial and monocyte-macrophage systems and, most importantly, neurones. Children affected by MPS IIIA experience a severe, progressive neuropathology that ultimately leads to death at around 15 years of age. METHODS MPS IIIA pathology was addressed in a mouse model using two separate methods of therapeutic gene delivery. A lentiviral vector expressing murine sulphamidase was delivered to 6-week-old MPS IIIA affected mice either by intravenous injection, or by intraventricular infusion. Therapeutic outcomes were assessed 7 months after gene transfer. RESULTS After intravenous gene delivery, liver sulphamidase was restored to approximately 30% of wild-type levels. The resultant widespread delivery of enzyme secreted from transduced cells to somatic tissues via the peripheral circulation corrected most somatic pathology. However, unlike an earlier study, central nervous system (CNS) pathology remained unchanged. Conversely, intraventricular gene delivery resulted in widespread sulphamidase gene delivery in (and reduced lysosomal storage throughout) the brain. Improvements in behaviour were observed in these mice, and interestingly, pathological urinary retention was prevented. CONCLUSIONS The CNS remains the last major barrier to effective therapy for children affected by LSDs. The blood-brain barrier (BBB) limits the uptake of lysosomal enzymes from the peripheral circulation into the CNS, making direct gene delivery to the brain a reasonable, albeit more challenging, therapeutic option. Future work will further assess the relative advantages of directly targeting the brain with somatic gene delivery with sulphamidase modified to increase the efficiency of transport across the BBB.
Collapse
Affiliation(s)
- Chantelle McIntyre
- Genetics and Molecular Pathology, SA Pathology, North Adelaide, Australia.
| | | | | |
Collapse
|
86
|
Jardim LB, Villanueva MM, de Souza CFM, Netto CBO. Clinical aspects of neuropathic lysosomal storage disorders. J Inherit Metab Dis 2010; 33:315-29. [PMID: 20490930 DOI: 10.1007/s10545-010-9079-5] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/08/2009] [Revised: 03/06/2010] [Accepted: 03/16/2010] [Indexed: 10/19/2022]
Abstract
The purpose of this review is to describe neurological phenotypes associated with lysosomal storage diseases (LSDs), focusing on features arising from primary neuronal involvement. Clinical presentation, progression and genetic data, are discussed in detail in Part 2, the electronic material. Main features are summarized in Part 1. Insights gained from several observational studies are discussed. Prospective studies of the natural history of most neuronopathic LSDs have been hampered by the rarity of these conditions and the short survival of affected patients. Increasingly, longitudinal observations relating to neurological manifestations are being reported. Better clinical studies are necessary, including repeated measurements of disease progression to facilitate the development of sensitive scoring systems and appropriate counseling of affected individuals and their families. Ideally, clinical studies should involve a large cohort. As treatment becomes available, knowledge of disease expression and factors that influence the phenotype may enable critical assessment of therapeutic outcomes. It is hoped that increased familiarity with the clinical expression of individual LSDs will allow early diagnosis, so families at risk are given options to consider during future pregnancies. Early diagnosis also permits the introduction of timely intervention, to favoring improved outcome in cases that are potentially treatable.
Collapse
Affiliation(s)
- Laura Bannach Jardim
- Department of Internal Medicine, Universidade Federal do Rio Grande do Sul, Rua Ramiro Barcelos 2350, 90035-903, Porto Alegre, Brazil.
| | | | | | | |
Collapse
|
87
|
Malm G, Månsson JE. Mucopolysaccharidosis type III (Sanfilippo disease) in Sweden: clinical presentation of 22 children diagnosed during a 30-year period. Acta Paediatr 2010; 99:1253-7. [PMID: 20337777 DOI: 10.1111/j.1651-2227.2010.01800.x] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
AIM The aim of this study was to present the natural clinical course in children and adolescents with MPS III diagnosed during a 30-year period in Sweden. METHODS The patients were identified from metabolic laboratory records between 1975 and 2004. Patient data were assessed from interviews of parents and by clinical examination and records from the patients. RESULTS A total of 15 children, 68%, with MPS IIIA were diagnosed at a median age of 6.8 years (range 1.2-18.9 years). One boy had MPS IIIB and five children MPS IIIC, diagnosed at ages between 1.9 and 11.6 years. In one child the type was not determined. The median age of children with type IIIA who had deceased was 16.2 years (range 10.4-31.2 years). Ten individuals with MPS III are alive at ages between 5 and 29 years. In four families, two children were affected. CONCLUSION In 22 Swedish children with Sanfilippo disease an early normal development followed by a delay in speech and an appearance of behaviour problems was found in most children during the early preschool period. Mental retardation was diagnosed in almost all individuals before starting school. Early diagnosis is important in this devastating, progressive disorder, not only for genetic counselling but also for participation in future treatments.
Collapse
Affiliation(s)
- G Malm
- Division of Paediatrics, Department of Clinical Science, Intervention and Technology, Karolinska Institutet, Karolinska University Hospital, Huddinge, Sweden.
| | | |
Collapse
|
88
|
Kalkan Ucar S, Ozbaran B, Demiral N, Yuncu Z, Erermis S, Coker M. Clinical overview of children with mucopolysaccharidosis type III A and effect of Risperidone treatment on children and their mothers psychological status. Brain Dev 2010; 32:156-61. [PMID: 19217229 DOI: 10.1016/j.braindev.2008.12.010] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/08/2008] [Revised: 12/12/2008] [Accepted: 12/18/2008] [Indexed: 11/30/2022]
Abstract
Mucopolysaccharidosis IIIA (MPS IIIA) is a lysosomal storage disorder characterized by progressive mental deterioration and severe behavioral problems. We conducted an open-label, crossover study of the efficacy and safety of Risperidone on behavioral disorder in children with MPS IIIA. A total of 12 patients (5.5+/-2.2 years) with enzymatic diagnosis of MPS IIIA were randomly assigned to receive Risperidone (0.125-2mg/d) for 6 months. The hyperactivity and disruptive behavior level of children before and after treatment was evaluated regarding the scores from Turgay DSM IV Based Child and Adolescent Behavior Disorders Screening and Rating Scale (T-DSM-IV-S). Clinic Global Impression Scale - Severity (CGIS-S) was used for all cases for determining the psychiatric disorder severity. The anxiety and depression levels of mothers before and after treatment were evaluated using Hamilton Anxiety Scale (HAM-A) and Beck Depression Inventory (BDI). The adverse effects were evaluated by monitoring weight, serum prolactin, glucose and lipid levels. The response to the treatment was measured by decrease in values of CGI-S (from 6+/-1.12 to 2.91+/-0.66, p=0.001). According to T-DSM-IV-S scores the best improvement was observed in hyperactivity scores (16.25+/-8.57/11.58+/-7.26, p=0.001), followed by opposition/defiance (6.66+/-5.92/5.08+/-4.88, p=0.032), and conduct disorder scores (1.00+/-1.85/0.41+/-.99, p=0.67). No clinically relevant elevations in weight and serum prolactin, glucose or lipid levels have been documented (p>0.05). There was a significant decrease in anxiety and depression scores of mothers (HAM-A: 20.33+/-8.28/17.91+/-6.89, BDI: 23.58+/-7.14/20.5+/-5.93, p<0.001). To our knowledge, research on the pharmacological treatment of MPS IIIA with Risperidone has not been reported. According to our data, Risperidone appeared to be safe and effective in MPS IIIA patients.
Collapse
Affiliation(s)
- Sema Kalkan Ucar
- Ege University Medical Faculty, Department of Pediatric Endocrinology and Metabolism, Bornova, Izmir, Turkey.
| | | | | | | | | | | |
Collapse
|
89
|
Tchan MC, Sillence D. Extrapyramidal symptoms and medication use in Mucopolysaccharidosis type III. JOURNAL OF INTELLECTUAL & DEVELOPMENTAL DISABILITY 2009; 34:275-279. [PMID: 19681008 DOI: 10.1080/13668250903070891] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/28/2023]
Abstract
BACKGROUND We report the case of a 16-year-old male with Mucopolysaccharidosis III type A (Sanfilippo syndrome) who was commenced on risperidone for behaviour management. He rapidly developed extrapyramidal symptoms that have not resolved. METHOD The medication histories of 20 patients with Mucopolysaccharidosis III seen at a Lysosomal Storage Diseases Clinic were reviewed to determine the incidence of extrapyramidal side effects. RESULTS Six patients had been treated with risperidone, olanzapine, or lamotrigine. Five of these patients developed extrapyramidal side effects. CONCLUSIONS The incidence of extrapyramidal side effects was considerably higher than expected. We suggest that these medications be used with considerable caution in these patients.
Collapse
|
90
|
Piotrowska E, Jakóbkiewicz-Banecka J, Tylki-Szymańska A, Czartoryska B, Wegrzyn A, Wegrzyn G. Correlation between severity of mucopolysaccharidoses and combination of the residual enzyme activity and efficiency of glycosaminoglycan synthesis. Acta Paediatr 2009; 98:743-9. [PMID: 19046346 DOI: 10.1111/j.1651-2227.2008.01153.x] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
AIM To develop a method for prediction of severity and clinical course of mucopolysaccharidoses (MPS), a group of inherited metabolic diseases. METHODS Various biochemical and clinical parameters (including estimation of the level of clinical severity, presence of specific mutations, residual enzyme activity, urinary glycosaminoglycan (GAG) excretion, storage of GAG in fibroblasts and efficiency of GAG synthesis) of patients suffering from MPS types II, IIIA and IIIB were determined. Correlations between genetic, biochemical and clinical parameters were tested. RESULTS We found that efficiency of GAG synthesis may contribute to the level of severity of MPS. It appears that (i) combination of low or average efficiency of GAG synthesis and the presence of residual activity of the enzyme is responsible for an attenuated phenotype, (ii) a lack of detectable residual enzyme activity causes a severe phenotype, irrespective of the efficiency of GAG synthesis and (iii) high efficiency of GAG synthesis leads to a severe phenotype, even if residual enzyme activity is detected. This correlation was found to be valid in 15 out of 17 patients tested. CONCLUSION Analysis of efficiency of GAG synthesis and residual activity of the enzyme may be considered for prediction of severity of MPS patients' clinical phenotypes.
Collapse
Affiliation(s)
- Ewa Piotrowska
- Department of Molecular Biology, University of Gdańsk, Gdańsk, Poland
| | | | | | | | | | | |
Collapse
|
91
|
Valstar MJ, Ruijter GJG, van Diggelen OP, Poorthuis BJ, Wijburg FA. Sanfilippo syndrome: a mini-review. J Inherit Metab Dis 2008; 31:240-52. [PMID: 18392742 DOI: 10.1007/s10545-008-0838-5] [Citation(s) in RCA: 260] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/10/2007] [Revised: 02/04/2008] [Accepted: 02/05/2008] [Indexed: 12/18/2022]
Abstract
Mucopolysaccharidosis type III (MPS III, Sanfilippo syndrome) is an autosomal recessive disorder, caused by a deficiency in one of the four enzymes involved in the lysosomal degradation of the glycosaminoglycan heparan sulfate. Based on the enzyme deficiency, four different subtypes, MPS IIIA, B, C, and D, are recognized. The genes encoding these four enzymes have been characterized and various mutations have been reported. The probable diagnosis of all MPS III subtypes is based on increased concentration of heparan sulfate in the urine. Enzymatic assays in leukocytes and/or fibroblasts confirm the diagnosis and allow for discrimination between the different subtypes of the disease. The clinical course of MPS III can be divided into three phases. In the first phase, which usually starts between 1 and 4 years of age, a developmental delay becomes apparent after an initial normal development during the first 1-2 years of life. The second phase generally starts around 3-4 years and is characterized by severe behavioural problems and progressive mental deterioration ultimately leading to severe dementia. In the third and final stage, behavioural problems slowly disappear, but motor retardation with swallowing difficulties and spasticity emerge. Patients usually die at the end of the second or beginning of the third decade of life, although survival into the fourth decade has been reported. Although currently no effective therapy is yet available for MPS III, several promising developments raise hope that therapeutic interventions, halting the devastating mental and behavioural deterioration, might be feasible in the near future.
Collapse
Affiliation(s)
- M J Valstar
- Department of Pediatrics, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | | | | | | | | |
Collapse
|