1
|
Barbero-Herranz R, Garriga-García M, Moreno-Blanco A, Palacios E, Ruiz-Sala P, Vicente-Santamaría S, Stanescu S, Belanger-Quintana A, Pintos-Morell G, Arconada B, del Campo R, Avendaño-Ortiz J. The Role of the Gut Microbiota in Sanfilippo Syndrome's Physiopathology: An Approach in Two Affected Siblings. Int J Mol Sci 2024; 25:8856. [PMID: 39201540 PMCID: PMC11354487 DOI: 10.3390/ijms25168856] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2024] [Revised: 08/09/2024] [Accepted: 08/12/2024] [Indexed: 09/02/2024] Open
Abstract
Sanfilippo syndrome, or mucopolysaccharidosis type III (MPS III), is a rare lysosomal disease caused by congenital enzymatic deficiencies in heparan sulfate (HS) degradation, leading to organ dysfunction. The most severe hallmark of MPS III comprises neurological alterations, although gastrointestinal symptoms (GISs) have also been shown to be relevant in many patients. Here, we explored the contribution of the gut microbiota to MPS III GISs. We analyzed the composition and functionality of the gut microbiota in two MPS III siblings with the same mutation (c.544C > T, c.1080delC, in the SGSH gene) and the same diet, but with differences in their GISs, including recurrent diarrhea in one of them. Using 16S sequencing, we observed that the MPS III patients exhibited decreased alpha diversity and a lower abundance of Lachnospiraceae and Bifidobacteriaceae accompanied by a higher abundance of the Ruminococcaceae and Rikenellaceae families than the healthy control subjects. Comparing siblings, we found an increased abundance of Bacteroidaceae and a lower abundance of Ruminococcaceae and Akkermansiaceae in the GIS-free patient. This patient also had a higher relative abundance of Sus genes (SusA, SusB, SusE, and SusG) involved in glycosaminoglycan metabolism. We found higher HS levels in the stool of the two MPS III patients than in healthy volunteers, particularly in the patient with GISs. Functionally, whole fecal metabolites from the patient with GISs induced oxidative stress in vitro in healthy monocytes. Finally, the Bacteroides thetaiotaomicron strain isolated from MPS III stool samples exhibited HS degradation ability. Overall, our results reveal different microbiota compositions and functionalities in MPS III siblings, who exhibited differential gastrointestinal symptomatology. Our study may serve as a gateway to explore the impact of the gut microbiota and its potential to enhance the quality of life in Sanfilippo syndrome patients.
Collapse
Affiliation(s)
- Raquel Barbero-Herranz
- Microbiology Department, Hospital Universitario Ramón y Cajal and Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), 28034 Madrid, Spain; (R.B.-H.); (A.M.-B.); (E.P.)
| | - María Garriga-García
- Endocrinology and Nutrition Service, Hospital Universitario Ramón y Cajal, 28034 Madrid, Spain; (M.G.-G.); (S.V.-S.)
| | - Ana Moreno-Blanco
- Microbiology Department, Hospital Universitario Ramón y Cajal and Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), 28034 Madrid, Spain; (R.B.-H.); (A.M.-B.); (E.P.)
- CIBER de Enfermedades Infecciosas, Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Esther Palacios
- Microbiology Department, Hospital Universitario Ramón y Cajal and Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), 28034 Madrid, Spain; (R.B.-H.); (A.M.-B.); (E.P.)
| | - Pedro Ruiz-Sala
- Centro de Diagnóstico de Enfermedades Moleculares (CEDEM), Autonomous University of Madrid (UAM), IdiPaz, 28049 Madrid, Spain;
- CIBER de Enfermedades Raras, Instituto de Salud Carlos III, 28049 Madrid, Spain
| | - Saioa Vicente-Santamaría
- Endocrinology and Nutrition Service, Hospital Universitario Ramón y Cajal, 28034 Madrid, Spain; (M.G.-G.); (S.V.-S.)
| | - Sinziana Stanescu
- Unidad de Enfermedades Metabólicas Hospital, CSUR, MetabERN, Pediatric Department, Hospital Universitario Ramón y Cajal, Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), 28034 Madrid, Spain; (S.S.); (A.B.-Q.)
| | - Amaya Belanger-Quintana
- Unidad de Enfermedades Metabólicas Hospital, CSUR, MetabERN, Pediatric Department, Hospital Universitario Ramón y Cajal, Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), 28034 Madrid, Spain; (S.S.); (A.B.-Q.)
| | - Guillem Pintos-Morell
- Vall d’Hebron Institut de Recerca (VHIR), Unidad de Enfermedades Raras, Hospital Vall d’Hebron Barcelona Hospital Campus, Comité Médico Consultivo MPS-Lisosomales, 08035 Barcelona, Spain;
| | - Beatriz Arconada
- Federación Española de Enfermedades Raras (FEDER), 28009 Madrid, Spain;
| | - Rosa del Campo
- Microbiology Department, Hospital Universitario Ramón y Cajal and Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), 28034 Madrid, Spain; (R.B.-H.); (A.M.-B.); (E.P.)
- CIBER de Enfermedades Infecciosas, Instituto de Salud Carlos III, 28029 Madrid, Spain
- Faculty of Health Sciences, Alfonso X El Sabio University, Villanueva de la Cañada, 28691 Madrid, Spain
| | - José Avendaño-Ortiz
- Microbiology Department, Hospital Universitario Ramón y Cajal and Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), 28034 Madrid, Spain; (R.B.-H.); (A.M.-B.); (E.P.)
- CIBER de Enfermedades Infecciosas, Instituto de Salud Carlos III, 28029 Madrid, Spain
| |
Collapse
|
2
|
Giugliano G, Schiavo M, Pirone D, Běhal J, Bianco V, Montefusco S, Memmolo P, Miccio L, Ferraro P, Medina DL. Investigation on lysosomal accumulation by a quantitative analysis of 2D phase-maps in digital holography microscopy. Cytometry A 2024; 105:323-331. [PMID: 38420869 DOI: 10.1002/cyto.a.24833] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2023] [Revised: 01/13/2024] [Accepted: 02/20/2024] [Indexed: 03/02/2024]
Abstract
Lysosomes are the terminal end of catabolic pathways in the cell, as well as signaling centers performing important functions such as the recycling of macromolecules, organelles, and nutrient adaptation. The importance of lysosomes in human health is supported by the fact that the deficiency of most lysosomal genes causes monogenic diseases called as a group Lysosomal Storage Diseases (LSDs). A common phenotypic hallmark of LSDs is the expansion of the lysosomal compartment that can be detected by using conventional imaging methods based on immunofluorescence protocols or overexpression of tagged lysosomal proteins. These methods require the alteration of the cellular architecture (i.e., due to fixation methods), can alter the behavior of cells (i.e., by the overexpression of proteins), and require sample preparation and the accurate selection of compatible fluorescent markers in relation to the type of analysis, therefore limiting the possibility of characterizing cellular status with simplicity. Therefore, a quantitative and label-free methodology, such as Quantitative Phase Imaging through Digital Holographic (QPI-DH), for the microscopic imaging of lysosomes in health and disease conditions may represent an important advance to study and effectively diagnose the presence of lysosomal storage in human disease. Here we proof the effectiveness of the QPI-DH method in accomplishing the detection of the lysosomal compartment using mouse embryonic fibroblasts (MEFs) derived from a Mucopolysaccharidosis type III-A (MSP-IIIA) mouse model, and comparing them with wild-type (WT) MEFs. We found that it is possible to identify label-free biomarkers able to supply a first pre-screening of the two populations, thus showing that QPI-DH can be a suitable candidate to surpass fluorescent drawbacks in the detection of lysosomes dysfunction. An appropriate numerical procedure was developed for detecting and evaluate such cellular substructures from in vitro cells cultures. Results reported in this study are encouraging about the further development of the proposed QPI-DH approach for such type of investigations about LSDs.
Collapse
Affiliation(s)
- Giusy Giugliano
- CNR-ISASI, Institute of Applied Sciences and Intelligent Systems "E. Caianiello", Pozzuoli, Napoli, Italy
| | - Michela Schiavo
- CNR-ISASI, Institute of Applied Sciences and Intelligent Systems "E. Caianiello", Pozzuoli, Napoli, Italy
| | - Daniele Pirone
- CNR-ISASI, Institute of Applied Sciences and Intelligent Systems "E. Caianiello", Pozzuoli, Napoli, Italy
| | - Jaromír Běhal
- CNR-ISASI, Institute of Applied Sciences and Intelligent Systems "E. Caianiello", Pozzuoli, Napoli, Italy
- Department of Optics, Palacký University, Olomouc, Czech Republic
| | - Vittorio Bianco
- CNR-ISASI, Institute of Applied Sciences and Intelligent Systems "E. Caianiello", Pozzuoli, Napoli, Italy
| | - Sandro Montefusco
- Telethon Institute of Genetics and Medicine (TIGEM), Pozzuoli, Naples, Italy
| | - Pasquale Memmolo
- CNR-ISASI, Institute of Applied Sciences and Intelligent Systems "E. Caianiello", Pozzuoli, Napoli, Italy
| | - Lisa Miccio
- CNR-ISASI, Institute of Applied Sciences and Intelligent Systems "E. Caianiello", Pozzuoli, Napoli, Italy
| | - Pietro Ferraro
- CNR-ISASI, Institute of Applied Sciences and Intelligent Systems "E. Caianiello", Pozzuoli, Napoli, Italy
| | - Diego L Medina
- Telethon Institute of Genetics and Medicine (TIGEM), Pozzuoli, Naples, Italy
- Department of Medical and Translational Science, Federico II University, Naples, Italy
| |
Collapse
|
3
|
Huang S, Beatty ZJ, Mckinney AM, Nascene DR. Increased pituitary volumes in patients with Sanfilippo syndrome (mucopolysaccharidosis type 3, MPS III). Neuroradiology 2023; 65:1381-1386. [PMID: 37127720 DOI: 10.1007/s00234-023-03157-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2023] [Accepted: 04/22/2023] [Indexed: 05/03/2023]
Abstract
PURPOSE To evaluate apparent pituitary gland enlargement in patients with Sanfilippo syndrome observed at our institution. METHODS Twelve patients with Sanfilippo syndrome with brain MRI were studied. Anterior, posterior, and whole pituitary volumes were estimated using the prolate ellipsoid volume calculation method (π/6 × L × W × H). Convexity along the upper pituitary margin (Elster's grade) was also measured. These values were compared to two age- and sex-matched groups (normal controls and patients with Hurler syndrome) using one-way ANOVA followed by Tukey's post hoc analysis for multiple comparisons. RESULTS In the Sanfilippo cohort, the mean whole pituitary volume was 529.9 mm, the mean anterior pituitary volume was 333.4 mm, and the mean posterior pituitary volume was 59.1 mm with Elster's grade of 4.2. In the control cohort, the mean whole pituitary volume was 217.4 mm, the mean anterior pituitary volume was 154.8 mm, and the mean posterior pituitary volume was 28.4 mm with Elster's grade of 2.5. In the Hurler syndrome cohort, the mean whole pituitary volume was 310.0 mm, the mean anterior pituitary volume was 178.2 mm, and the mean posterior pituitary volume was 35.4 mm with Elster's grade of 3.5. CONCLUSION In our cohort of patients with Sanfilippo syndrome, whole, anterior, and posterior pituitary volumes and degree of convexity along the upper pituitary border were all significantly greater than controls. The cause of these morphological changes is unclear, as is clinical correlation of the findings.
Collapse
Affiliation(s)
- Shiwei Huang
- Department of Neurosurgery, University of Minnesota, Room D-429 Mayo Building, Mayo Mail Code 96, 420 Delaware St. SE, Minneapolis, MN, 55455, USA.
| | - Zachary J Beatty
- Department of Radiology, University of Minnesota, 420 Delaware St. SE #MMC 292, Minneapolis, MN, 55455, USA
- Department of Neuroradiology, University of Minnesota, 420 Delaware St. SE #MMC 292, Minneapolis, MN, 55455, USA
| | - Alexander M Mckinney
- Department of Radiology, University of Minnesota, 420 Delaware St. SE #MMC 292, Minneapolis, MN, 55455, USA
- Department of Neuroradiology, University of Minnesota, 420 Delaware St. SE #MMC 292, Minneapolis, MN, 55455, USA
| | - David R Nascene
- Department of Radiology, University of Minnesota, 420 Delaware St. SE #MMC 292, Minneapolis, MN, 55455, USA
- Department of Neuroradiology, University of Minnesota, 420 Delaware St. SE #MMC 292, Minneapolis, MN, 55455, USA
| |
Collapse
|
4
|
Liang Y, Gao X, Lu D, Zhang H, Zhang. Mucopolysaccharidosis type IIIC in chinese mainland: clinical and molecular characteristics of ten patients and report of six novel variants in the HGSNAT gene. Metab Brain Dis 2023; 38:2013-2023. [PMID: 37014526 DOI: 10.1007/s11011-023-01204-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/13/2023] [Accepted: 03/21/2023] [Indexed: 04/05/2023]
Abstract
BACKGROUND Mucopolysaccharidosis type IIIC (MPS IIIC; Sanfilippo syndrome C) is a rare lysosomal storage disease caused by mutations in the heparan-α-glucosaminide N-acetyltransferase (HGSNAT) gene, resulting in the accumulation of heparan sulfate. MPS IIIC is characterized by severe neuropsychiatric symptoms and mild somatic symptoms. METHODS Our study analyzed the clinical presentation and biochemical characteristics of ten Chinese MPS IIIC patients from eight families. Whole exome sequencing was applied to identify the variants in HGSNAT gene. In one patient with only one mutant allele identified firstly, whole genome sequencing was applied. The pathogenic effect of novel variants was evaluated in silico. RESULTS The mean age at the onset of clinical symptoms was 4.2 ± 2.5 years old, and the mean age of diagnosis was 7.6 ± 4.5 years old, indicating a delay of diagnosis. The most common onset symptoms were speech deterioration, and the most frequent presenting symptoms are speech deterioration, mental deterioration, hyperactivity and hepatomegaly, sequentially. All mutant alleles of 10 patients have been identified. There were eleven different HGSNAT variants, and the most common one was a previously reported variant c.493 + 1G > A. There were six novel variants, p.R124T, p.G290A, p.G426E, c.743 + 101_743 + 102delTT, c.851 + 171T > A and p.V582Yfs*18 in our cohort. Extraordinarily, two deep intron variants were identified in our cohort, with the variant c.851 + 171T > A identified by whole genome sequencing. CONCLUSION This study analyzed the clinical, biochemical, and genetic characteristics of ten Chinese MPS IIIC patients, which would assist in the early diagnosis and genetic counselling of MPS IIIC.
Collapse
Affiliation(s)
- Yingjun Liang
- Pediatric Endocrinology and Genetic, Xinhua Hospital, Shanghai Institute for Pediatric Research, Shanghai Jiao Tong University School of Medicine, Kongjiang Road 1665 #, Shanghai, 200092, China
| | - Xiaolan Gao
- Pediatric Endocrinology and Genetic, Xinhua Hospital, Shanghai Institute for Pediatric Research, Shanghai Jiao Tong University School of Medicine, Kongjiang Road 1665 #, Shanghai, 200092, China
| | - Deyun Lu
- Pediatric Endocrinology and Genetic, Xinhua Hospital, Shanghai Institute for Pediatric Research, Shanghai Jiao Tong University School of Medicine, Kongjiang Road 1665 #, Shanghai, 200092, China
| | - Huiwen Zhang
- Pediatric Endocrinology and Genetic, Xinhua Hospital, Shanghai Institute for Pediatric Research, Shanghai Jiao Tong University School of Medicine, Kongjiang Road 1665 #, Shanghai, 200092, China.
| | - Zhang
- Pediatric Endocrinology and Genetic, Xinhua Hospital, Shanghai Institute for Pediatric Research, Shanghai Jiao Tong University School of Medicine, Kongjiang Road 1665 #, Shanghai, 200092, China
| |
Collapse
|
5
|
Muschol N, Giugliani R, Jones SA, Muenzer J, Smith NJC, Whitley CB, Donnell M, Drake E, Elvidge K, Melton L, O'Neill C. Sanfilippo syndrome: consensus guidelines for clinical care. Orphanet J Rare Dis 2022; 17:391. [PMID: 36303195 DOI: 10.1186/s13023-022-02484-6] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2022] [Accepted: 08/15/2022] [Indexed: 11/10/2022] Open
Abstract
Sanfilippo syndrome is a group of rare, complex, and progressive neurodegenerative lysosomal storage disorders that is characterized by childhood dementia. The clinical management of patients with progressive neurological decline and multisystem involvement requires a multidisciplinary team with experience in the management of neurodegenerative disorders. Best practice guidelines for the clinical management of patients with these types of rare disorders are critical to ensure prompt diagnosis and initiation of appropriate care. However, there are no published standard global clinical care guidelines for patients with Sanfilippo syndrome. To address this, a literature review was conducted to evaluate the current evidence base and to identify evidence gaps. The findings were reviewed by an international steering committee composed of clinical experts with extensive experience in managing patients with Sanfilippo syndrome. The goal was to create a consensus set of basic clinical guidelines that will be accessible to and informed by clinicians globally, as well as providing a practical resource for families to share with their local care team who may not have experience with this rare disease. This review distills 178 guideline statements into an easily digestible document that provides evidence-based, expert-led recommendations for how to approach common management challenges and appropriate monitoring schedules in the care of patients with Sanfilippo syndrome.
Collapse
Affiliation(s)
- Nicole Muschol
- Department of Pediatrics, International Center for Lysosomal Disorders (ICLD), University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Roberto Giugliani
- DASA, Federal University of Rio Grande do Sul (UFRGS), Hospital de Clinicas de Porto Alegre (HCPA), Casa dos Raros, Porto Alegre, Brazil
| | | | - Joseph Muenzer
- University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Nicholas J C Smith
- Department of Neurology and Clinical Neurophysiology, Women's and Children's Health Network and the Discipline of Paediatrics, University of Adelaide, Adelaide, Australia
| | | | - Megan Donnell
- Sanfilippo Children's Foundation, Freshwater, NSW, Australia
| | - Elise Drake
- Cure Sanfilippo Foundation, Columbia, SC, USA
| | | | - Lisa Melton
- Sanfilippo Children's Foundation, Freshwater, NSW, Australia
| | | | | |
Collapse
|
6
|
Ballantyne M, Chiu B, Sergi CM. Sanfilippo Syndrome Type A: Early Cardiac Involvement of Two patients with Cardiac Manifestations. Cardiovasc Pathol 2022; 60:107430. [DOI: 10.1016/j.carpath.2022.107430] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Revised: 04/29/2022] [Accepted: 05/02/2022] [Indexed: 11/03/2022] Open
|
7
|
Porter KA, O'Neill C, Drake E, Andrews SM, Delaney K, Parker S, Escolar ML, Montgomery S, Moon W, Worrall C, Peay HL. Caregivers' assessment of meaningful and relevant clinical outcome assessments for Sanfilippo syndrome. J Patient Rep Outcomes 2022; 6:40. [PMID: 35467223 PMCID: PMC9038975 DOI: 10.1186/s41687-022-00447-w] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2021] [Accepted: 04/04/2022] [Indexed: 11/10/2022] Open
Abstract
OBJECTIVES Sanfilippo syndrome is a rare multisystem disease with no approved treatments. This study explores caregiver perspectives on the most impactful symptoms and patient-relevant clinical outcomes assessments. The pediatric onset and progressive neurodegenerative nature of Sanfilippo limits use of self-report in clinical research. This study obtains Sanfilippo caregiver data to support the selection of fit-for-purpose and patient-relevant clinical outcome assessments (COAs). METHODS We conducted an asynchronous online focus group (n = 11) followed by individual interviews with caregivers (n = 19) of children with Sanfilippo syndrome. All participants reported on the impact of disease symptoms and level of unmet treatment need across Sanfilippo symptom domains. Focus group participants reviewed existing assessments relating to 8 symptom domains (15 total assessments) and provided feedback on meaningfulness and relevance. Focus group data were used to reduce the number of assessments included in subsequent interviews to 8 COAs across 7 symptom domains: communication, eating, sleep, mobility, pain, behavior and adapting. Interview respondents provided data on meaningfulness and relevance of assessments. Data were coded using an item-tracking matrix. Data summaries were analyzed by caregivers' responses regarding meaningfulness; relevance to Sanfilippo syndrome; and based on caregiver indication of missing or problematic subdomains and items. RESULTS Participants' children were 2-24 years in age and varied in disease progression. Caregivers reported communication and mobility as highly impactful domains with unmet treatment needs, followed closely by pain and sleep. Domains such as eating, adaptive skills, and behaviors were identified as impactful but with relatively less priority, by comparison. Participants endorsed the relevance of clinical outcome assessments associated with communication, eating, sleep, and pain, and identified them as highly favorable for use in a clinical trial. Participants specified some refinements in existing assessments to best reflect Sanfilippo symptoms and disease course. DISCUSSION The identification of impactful symptoms to treat and relevant and meaningful clinical outcome assessments supports patient-focused drug development. Our results inform targets for drug development and the selection of primary and secondary outcome assessments with high meaningfulness and face validity to Sanfilippo syndrome caregivers. Assessments identified as less optimal might be refined, replaced, or remain if the clinical trial necessitates.
Collapse
Affiliation(s)
- Katherine Ackerman Porter
- Center for Genomics, Bioinformatics, and Translational Research, RTI International, Research Triangle Park, NC, USA
| | | | - Elise Drake
- Cure Sanfilippo Foundation, Columbia, SC, USA
| | - Sara M Andrews
- Center for Genomics, Bioinformatics, and Translational Research, RTI International, Research Triangle Park, NC, USA
| | - Kathleen Delaney
- Global Patient Advocacy and Engagement, BioMarin Pharmaceutical Inc., San Rafael, CA, USA
| | - Samantha Parker
- Patient and Policy Affairs, Lysogene, Neuilly sur Seine, France
| | - Maria L Escolar
- Department of Pediatrics, University of Pittsburgh Medical Center, Pittsburgh, PA, USA.,Children's Hospital of Pittsburgh of UPMC, Pittsburgh, PA, USA
| | | | - William Moon
- Cure Sanfilippo Foundation Parent Advocates, Columbia, SC, USA
| | - Carolyn Worrall
- Cure Sanfilippo Foundation Parent Advocates, Columbia, SC, USA
| | - Holly L Peay
- Center for Genomics, Bioinformatics, and Translational Research, RTI International, Research Triangle Park, NC, USA.
| |
Collapse
|
8
|
Kim MS, Yang A, Noh ES, Kim C, Bae GY, Lim HH, Park HD, Cho SY, Jin DK. Natural History and Molecular Characteristics of Korean Patients with Mucopolysaccharidosis Type III. J Pers Med 2022; 12:jpm12050665. [PMID: 35629088 PMCID: PMC9145712 DOI: 10.3390/jpm12050665] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2022] [Revised: 04/18/2022] [Accepted: 04/19/2022] [Indexed: 02/05/2023] Open
Abstract
Background: Mucopolysaccharidosis type III (MPS III) is an autosomal recessive lysosomal storage disorder characterised by progressive neurocognitive deterioration. MPS III subtypes are clinically indistinguishable, with a wide range of symptoms and variable severity. The natural history of this disorder within an Asian population has not yet been extensively studied. This study investigated the natural history of Korean patients with MPS III. Methods: Thirty-four patients from 31 families diagnosed with MPS III from January 1997 to May 2020 in Samsung Medical Centre were enrolled. Clinical, molecular, and biochemical characteristics were retrospectively collected from the patients’ medical records and via interviews. Results: 18 patients had MPS IIIA, 14 had IIIB, and two had IIIC. Twenty (58.9%) patients were male. Mean age at symptom onset was 2.8 ± 0.8 years and at diagnosis was 6.3 ± 2.2 years. All patients with MPS IIIA and IIIB were classified into the rapidly progressing (RP) phenotype. The most common symptom at diagnosis was language retardation (88.2%), followed by motor retardation (76.5%), general retardation (64.7%), and hyperactivity (41.2%). Language retardation was more predominant in IIIA, and motor retardation was more predominant in IIIB. The mean age of the 13 deceased patients at the time of the study was 14.4 ± 4.1 years. The age at diagnosis and lag time were significantly older and longer in the non-survivor group compared with the survivor group (p = 0.029 and 0.045, respectively). Genetic analysis was performed in 24 patients with MPS III and identified seven novel variants and three hot spots. Conclusion: This study is the first to analyse the genetic and clinical characteristics of MPS III patients in Korea. Better understanding of the natural history of MPS III might allow early diagnosis and timely management of the disease and evaluation of treatment outcomes in future clinical trials for MPS III.
Collapse
Affiliation(s)
- Min-Sun Kim
- Department of Pediatrics, Samsung Medical Centre, Sungkyunkwan University School of Medicine, Seoul 06351, Korea; (M.-S.K.); (E.-s.N.); (G.Y.B.); (D.-K.J.)
| | - Aram Yang
- Department of Pediatrics, Kangbuk Samsung Hospital, Sungkyunkwan University School of Medicine, Seoul 03181, Korea;
| | - Eu-seon Noh
- Department of Pediatrics, Samsung Medical Centre, Sungkyunkwan University School of Medicine, Seoul 06351, Korea; (M.-S.K.); (E.-s.N.); (G.Y.B.); (D.-K.J.)
| | - Chiwoo Kim
- Department of Pediatrics, Soonchunhyang University Bucheon Hospital, Soonchunhyang University College of Medicine, Bucheon 14584, Korea;
| | - Ga Young Bae
- Department of Pediatrics, Samsung Medical Centre, Sungkyunkwan University School of Medicine, Seoul 06351, Korea; (M.-S.K.); (E.-s.N.); (G.Y.B.); (D.-K.J.)
| | - Han Hyuk Lim
- Department of Pediatrics, Chungnam National University College of Medicine, Daejeon 35015, Korea;
| | - Hyung-Doo Park
- Department of Laboratory Medicine and Genetics, Samsung Medical Centre, Sungkyunkwan University School of Medicine, Seoul 06351, Korea;
| | - Sung Yoon Cho
- Department of Pediatrics, Samsung Medical Centre, Sungkyunkwan University School of Medicine, Seoul 06351, Korea; (M.-S.K.); (E.-s.N.); (G.Y.B.); (D.-K.J.)
- Correspondence: ; Tel.: +82-2-3410-3539; Fax: +82-2-3410-0043
| | - Dong-Kyu Jin
- Department of Pediatrics, Samsung Medical Centre, Sungkyunkwan University School of Medicine, Seoul 06351, Korea; (M.-S.K.); (E.-s.N.); (G.Y.B.); (D.-K.J.)
| |
Collapse
|
9
|
Wijburg FA, Aiach K, Chakrapani A, Eisengart JB, Giugliani R, Héron B, Muschol N, O'Neill C, Olivier S, Parker S. An observational, prospective, multicenter, natural history study of patients with mucopolysaccharidosis type IIIA. Mol Genet Metab 2022; 135:133-142. [PMID: 34991944 DOI: 10.1016/j.ymgme.2021.12.002] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/10/2021] [Revised: 12/04/2021] [Accepted: 12/05/2021] [Indexed: 10/19/2022]
Abstract
Mucopolysaccharidosis type IIIA (MPS IIIA, also known as Sanfilippo syndrome) is a rare genetic lysosomal storage disease characterized by early and progressive neurodegeneration resulting in a rapid decline in cognitive function affecting speech and language, adaptive behavior, and motor skills. We carried out a prospective observational study to assess the natural history of patients with MPS IIIA, using both standardized tests and patient-centric measures to determine the course of disease progression over a 2-year period. A cohort of 23 patients (7 girls, 16 boys; mean age 28-105 months at baseline) with a confirmed diagnosis of MPS IIIA were assessed and followed up at intervals of 3-6 months; cognitive function was measured using Bayley Scales of Infant and Toddler Development 3rd edition (BSID-III) to derive cognitive development quotients (DQ). Daily living, speech/language development and motor skills were measured using the Vineland Adaptive Behavior Scale (VABS-II). Sleep-wake patterns, behavior and quality-of-life questionnaires were also reported at each visit using parent/caregiver reported outcome tools. All patients had early onset severe MPS IIIA, were diagnosed before 74 months of age, and had cognitive scores below normal developmental levels at baseline. Patients less than 40 months of age at baseline were more likely to continue developing new skills over the first 6-12 months of follow-up. There was a high variability in cognitive developmental age (DA) in patients between 40 and 70 months of age; two-thirds of these patients already had profound cognitive decline, with a DA ≤10 months. The highest cognitive DA achieved in the full study cohort was 34 months. Post hoc, patients were divided into two groups based on baseline cognitive DQ (DQ ≥50 or <50). Cognitive DQ decreased linearly over time, with a decrease from baseline of 30.1 and 9.0 points in patients with cognitive DQ ≥50 at baseline and cognitive DQ <50 at baseline, respectively. Over the 2-year study, VABS-II language scores declined progressively. Motor skills, including walking, declined over time, although significantly later than cognitive decline. No clear pattern of sleep disturbance was observed, but night waking was common in younger patients. Pain scores, as measured on the quality-of-life questionnaire, increased over the study period. The findings of this study strengthen the natural history data on cognitive decline in MPS IIIA and importantly provide additional data on endpoints, validated by the patient community as important to treat, that may form the basis of a multidomain endpoint capturing the disease complexity.
Collapse
Affiliation(s)
- Frits A Wijburg
- Department of Pediatric Metabolic Diseases, Emma Children's Hospital, Amsterdam UMC, Amsterdam, Netherlands; Amsterdam Lysosome Center "Sphinx", University of Amsterdam, Amsterdam, Netherlands.
| | | | - Anupam Chakrapani
- Metabolic Medicine Department, Great Ormond Street Hospital for Children NHS Foundation Trust, Great Ormond Street, London, UK
| | - Julie B Eisengart
- Department of Pediatrics, University of Minnesota, Minneapolis, MN, USA
| | - Roberto Giugliani
- Department of Genetics, UFRGS, Medical Genetics Service and DR Brazil, HCPA, Porto Alegre, Brazil
| | - Bénédicte Héron
- Reference Center for Lysosomal Diseases, Pediatric Neurology Department, Armand Trousseau University Hospital, APHP, Paris, France
| | - Nicole Muschol
- Department of Pediatrics, International Center for Lysosomal Disorders (ICLD), University Medical Center Hamburg Eppendorf, Hamburg, Germany
| | | | | | | |
Collapse
|
10
|
MPS-IIIA or Autism Spectrum Disorder?: Discrimination and Treatment. REVIEW JOURNAL OF AUTISM AND DEVELOPMENTAL DISORDERS 2022. [DOI: 10.1007/s40489-021-00298-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
|
11
|
Seven-year follow-up of durability and safety of AAV CNS gene therapy for a lysosomal storage disorder in a large animal. MOLECULAR THERAPY-METHODS & CLINICAL DEVELOPMENT 2021; 23:370-389. [PMID: 34761052 PMCID: PMC8550992 DOI: 10.1016/j.omtm.2021.09.017] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/16/2021] [Revised: 08/25/2021] [Accepted: 09/29/2021] [Indexed: 12/15/2022]
Abstract
Delivery of adeno-associated viral vectors (AAVs) to cerebrospinal fluid (CSF) has emerged as a promising approach to achieve widespread transduction of the central nervous system (CNS) and peripheral nervous system (PNS), with direct applicability to the treatment of a wide range of neurological diseases, particularly lysosomal storage diseases. Although studies in small animal models have provided proof of concept and experiments in large animals demonstrated feasibility in bigger brains, there is not much information on long-term safety or durability of the effect. Here, we report a 7-year study in healthy beagle dogs after intra-CSF delivery of a single, clinically relevant dose (2 × 1013 vg/dog) of AAV9 vectors carrying the canine sulfamidase, the enzyme deficient in mucopolysaccharidosis type IIIA. Periodic monitoring of CSF and blood, clinical and neurological evaluations, and magnetic resonance and ultrasound imaging of target organs demonstrated no toxicity related to treatment. AAV9-mediated gene transfer resulted in detection of sulfamidase activity in CSF throughout the study. Analysis at tissue level showed widespread sulfamidase expression and activity in the absence of histological findings in any region of encephalon, spinal cord, or dorsal root ganglia. Altogether, these results provide proof of durability of expression and long-term safety for intra-CSF delivery of AAV-based gene transfer vectors encoding therapeutic proteins to the CNS.
Collapse
|
12
|
Gastrointestinal Manifestations in Mucopolysaccharidosis Type III: Review of Death Certificates and the Literature. J Clin Med 2021; 10:jcm10194445. [PMID: 34640463 PMCID: PMC8509825 DOI: 10.3390/jcm10194445] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2021] [Accepted: 09/23/2021] [Indexed: 12/31/2022] Open
Abstract
BACKGROUND Mucopolysaccharidosis type III (MPS III, Sanfilippo disease) is a life-limiting recessive lysosomal storage disorder caused by a deficiency in the enzymes involved in degrading glycosaminoglycan heparan sulfate. MPS III is characterized by progressive deterioration of the central nervous system. Respiratory tract infections have been reported as frequent and as the most common cause of death, but gastrointestinal (GI) manifestations have not been acknowledged as a cause of concern. The aim of this study was to determine the incidence of GI problems as a primary cause of death and to review GI symptoms reported in published studies. METHODS Causes of death from 221 UK death certificates (1957-2020) were reviewed and the literature was searched to ascertain reported GI symptoms. RESULTS GI manifestations were listed in 5.9% (n = 13) of death certificates. Median (IQR) age at death was 16.7 (5.3) years. Causes of death included GI failure, GI bleed, haemorrhagic pancreatitis, perforation due to gastrostomies, paralytic ileus and emaciation. Twenty-one GI conditions were reported in 30 studies, mostly related to functional GI disorders, including diarrhoea, dysphagia, constipation, faecal incontinence, abdominal pain/distension and cachexia. CONCLUSIONS GI manifestations may be an under-recognized but important clinical feature of MPS III. Early recognition of GI symptoms and timely interventions is an important part of the management of MPS III patients.
Collapse
|
13
|
Ammer LS, Dohrmann T, Muschol NM, Lang A, Breyer SR, Ozga AK, Petzoldt M. Disease Manifestations in Mucopolysaccharidoses and Their Impact on Anaesthesia-Related Complications-A Retrospective Analysis of 99 Patients. J Clin Med 2021; 10:jcm10163518. [PMID: 34441814 PMCID: PMC8397084 DOI: 10.3390/jcm10163518] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2021] [Revised: 08/03/2021] [Accepted: 08/05/2021] [Indexed: 01/22/2023] Open
Abstract
Patients with mucopolysaccharidoses (MPS) frequently require anaesthesia for diagnostic or surgical interventions and thereby experience high morbidity. This study aimed to develop a multivariable prediction model for anaesthesia-related complications in MPS. This two-centred study was performed by retrospective chart review of children and adults with MPS undergoing anaesthesia from 2002 until 2018. We retrieved the patients’ demographics, medical history, clinical manifestations, and indication by each anaesthesia. Multivariable mixed-effects logistic regression was calculated for a clinical model based on preoperative predictors preselected by lasso regression and another model based on disease subtypes only. Of the 484 anaesthesia cases in 99 patients, 22.7% experienced at least one adverse event. The clinical model resulted in a better forecast performance than the subtype-model (AICc 460.4 vs. 467.7). The most relevant predictors were hepatosplenomegaly (OR 3.10, CI 1.54–6.26), immobility (OR 3.80, CI 0.98–14.73), and planned major surgery (OR 6.64, CI 2.25–19.55), while disease-specific therapies, i.e., haematopoietic stem cell transplantation (OR 0.45, CI 0.20–1.03), produced a protective effect. Anaesthetic complications can best be predicted by surrogates for advanced disease stages and protective therapeutic factors. Further model validation in different cohorts is needed.
Collapse
Affiliation(s)
- Luise Sophie Ammer
- Department of Paediatrics, International Centre for Lysosomal Disorders (ICLD), University Medical Centre Hamburg-Eppendorf, 20246 Hamburg, Germany; (N.M.M.); (A.L.); (S.R.B.)
- Correspondence: ; Tel.: +49-40-7410-53714
| | - Thorsten Dohrmann
- Department of Anaesthesiology, University Medical Centre Hamburg-Eppendorf, 20246 Hamburg, Germany; (T.D.); (M.P.)
| | - Nicole Maria Muschol
- Department of Paediatrics, International Centre for Lysosomal Disorders (ICLD), University Medical Centre Hamburg-Eppendorf, 20246 Hamburg, Germany; (N.M.M.); (A.L.); (S.R.B.)
| | - Annika Lang
- Department of Paediatrics, International Centre for Lysosomal Disorders (ICLD), University Medical Centre Hamburg-Eppendorf, 20246 Hamburg, Germany; (N.M.M.); (A.L.); (S.R.B.)
| | - Sandra Rafaela Breyer
- Department of Paediatrics, International Centre for Lysosomal Disorders (ICLD), University Medical Centre Hamburg-Eppendorf, 20246 Hamburg, Germany; (N.M.M.); (A.L.); (S.R.B.)
- Department of Paediatric Orthopaedics, Children’s Hospital Altona, 22763 Hamburg, Germany
- Department of Orthopaedics, University Medical Centre Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Ann-Kathrin Ozga
- Department of Medical Biometry and Epidemiology, University Medical Centre Hamburg-Eppendorf, 20246 Hamburg, Germany;
| | - Martin Petzoldt
- Department of Anaesthesiology, University Medical Centre Hamburg-Eppendorf, 20246 Hamburg, Germany; (T.D.); (M.P.)
| |
Collapse
|
14
|
Douek AM, Amiri Khabooshan M, Henry J, Stamatis SA, Kreuder F, Ramm G, Änkö ML, Wlodkowic D, Kaslin J. An Engineered sgsh Mutant Zebrafish Recapitulates Molecular and Behavioural Pathobiology of Sanfilippo Syndrome A/MPS IIIA. Int J Mol Sci 2021; 22:ijms22115948. [PMID: 34073041 PMCID: PMC8197930 DOI: 10.3390/ijms22115948] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2021] [Revised: 05/26/2021] [Accepted: 05/27/2021] [Indexed: 12/29/2022] Open
Abstract
Mucopolysaccharidosis IIIA (MPS IIIA, Sanfilippo syndrome type A), a paediatric neurological lysosomal storage disease, is caused by impaired function of the enzyme N-sulfoglucosamine sulfohydrolase (SGSH) resulting in impaired catabolism of heparan sulfate glycosaminoglycan (HS GAG) and its accumulation in tissues. MPS IIIA represents a significant proportion of childhood dementias. This condition generally leads to patient death in the teenage years, yet no effective therapy exists for MPS IIIA and a complete understanding of the mechanisms of MPS IIIA pathogenesis is lacking. Here, we employ targeted CRISPR/Cas9 mutagenesis to generate a model of MPS IIIA in the zebrafish, a model organism with strong genetic tractability and amenity for high-throughput screening. The sgshΔex5-6 zebrafish mutant exhibits a complete absence of Sgsh enzymatic activity, leading to progressive accumulation of HS degradation products with age. sgshΔex5-6 zebrafish faithfully recapitulate diverse CNS-specific features of MPS IIIA, including neuronal lysosomal overabundance, complex behavioural phenotypes, and profound, lifelong neuroinflammation. We further demonstrate that neuroinflammation in sgshΔex5-6 zebrafish is largely dependent on interleukin-1β and can be attenuated via the pharmacological inhibition of Caspase-1, which partially rescues behavioural abnormalities in sgshΔex5-6 mutant larvae in a context-dependent manner. We expect the sgshΔex5-6 zebrafish mutant to be a valuable resource in gaining a better understanding of MPS IIIA pathobiology towards the development of timely and effective therapeutic interventions.
Collapse
Affiliation(s)
- Alon M. Douek
- Australian Regenerative Medicine Institute, Monash University, Clayton, VIC 3800, Australia; (A.M.D.); (M.A.K.); (S.-A.S.); (F.K.)
| | - Mitra Amiri Khabooshan
- Australian Regenerative Medicine Institute, Monash University, Clayton, VIC 3800, Australia; (A.M.D.); (M.A.K.); (S.-A.S.); (F.K.)
| | - Jason Henry
- Neurotoxicology Lab, School of Science (Biosciences), RMIT University, Bundoora, VIC 3083, Australia; (J.H.); (D.W.)
| | - Sebastian-Alexander Stamatis
- Australian Regenerative Medicine Institute, Monash University, Clayton, VIC 3800, Australia; (A.M.D.); (M.A.K.); (S.-A.S.); (F.K.)
| | - Florian Kreuder
- Australian Regenerative Medicine Institute, Monash University, Clayton, VIC 3800, Australia; (A.M.D.); (M.A.K.); (S.-A.S.); (F.K.)
| | - Georg Ramm
- Ramaciotti Centre for Cryo-Electron Microscopy, Monash University, Clayton, VIC 3800, Australia;
- Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Clayton, VIC 3800, Australia
| | - Minna-Liisa Änkö
- Centre for Reproductive Health and Centre for Cancer Research, Hudson Institute of Medical Research, Clayton, VIC 3168, Australia;
- Department of Molecular and Translational Sciences, Monash University, Clayton, VIC 3800, Australia
| | - Donald Wlodkowic
- Neurotoxicology Lab, School of Science (Biosciences), RMIT University, Bundoora, VIC 3083, Australia; (J.H.); (D.W.)
| | - Jan Kaslin
- Australian Regenerative Medicine Institute, Monash University, Clayton, VIC 3800, Australia; (A.M.D.); (M.A.K.); (S.-A.S.); (F.K.)
- Correspondence: ; Tel.: +61-3-9902-9613; Fax: +61-3-9902-9729
| |
Collapse
|
15
|
Adetunji CO, Akram M, Michael OS, Shahzad K, Ayeni AE, Hasan S, Adetunji JB, Hasan SM, Inamuddin, Olaniyan M, Muhibi MA. Polysaccharides Derived From Natural Sources: A Panacea to Health and Nutritional Challenges. POLYSACCHARIDES 2021. [DOI: 10.1002/9781119711414.ch32] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
|
16
|
Shapiro EG, Eisengart JB. The natural history of neurocognition in MPS disorders: A review. Mol Genet Metab 2021; 133:8-34. [PMID: 33741271 DOI: 10.1016/j.ymgme.2021.03.002] [Citation(s) in RCA: 42] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/28/2021] [Revised: 03/02/2021] [Accepted: 03/04/2021] [Indexed: 01/22/2023]
Abstract
MPS disorders are associated with a wide spectrum of neurocognitive effects, from mild problems with attention and executive functions to progressive and degenerative neuronopathic disease. Studies of the natural history of neurocognition are necessary to determine the profile of abnormality and the rates of change, which are crucial to select endpoints for clinical trials of brain treatments and to make clinical recommendations for interventions to improve patients' quality of life. The goal of this paper is to review neurocognitive natural history studies to determine the current state of knowledge and assist in directing future research in all MPS disorders. There are seven different types of MPS diseases, each resulting from a specific enzyme deficiency and each having a separate natural history. MPS IX, will not be discussed as there are only 4 cases reported in the literature without cognitive abnormality. For MPS IH, hematopoietic cell transplant (HCT) is standard of care and many studies have documented the relationship between age at treatment and neurocognitive outcome, and to a lesser extent, neurocognitive status at baseline. However, the mortality and morbidity associated with the transplant process and residual long-term problems after transplant, have led to renewed efforts to find better treatments. Rather than natural history, new trials will likely need to use the developmental trajectories of the patients with HCT as a comparators. The literature has extensive data regarding developmental trajectories post-HCT. For attenuated MPS I, significant neurocognitive deficits have been documented, but more longitudinal data are needed in order to support a treatment directed at their attention and executive function abnormalities. The neuronopathic form of MPS II has been a challenge due to the variability of the trajectory of the disease with differences in timing of slowing of development and decline. Finding predictors of the course of the disease has only been partially successful, using mutation type and family history. Because of lack of systematic data and clinical trials that precede a thorough understanding of the disease, there is need for a major effort to gather natural history data on the entire spectrum of MPS II. Even in the attenuated disease, attention and executive function abnormalities need documentation. Lengthy detailed longitudinal studies are needed to encompass the wide variability in MPS II. In MPS IIIA, the existence of three good natural history studies allowed a quasi-meta-analysis. In patients with a rapid form of the disease, neurocognitive development slowed up until 42 to 47 months, halted up to about 54 months, then declined rapidly thereafter, with a leveling off at an extremely low age equivalent score below 22 months starting at about chronological age of 6. Those with slower or attenuated forms have been more variable and difficult to characterize. Because of the plethora of studies in IIIA, it has been recommended that data be combined from natural history studies to minimize the burden on parents and patients. Sufficient data exists to understand the natural history of cognition in MPS IIIA. MPS IIIB is quite similar to IIIA, but more attenuated patients in that phenotype have been reported. MPS IIIC and D, because they are so rare, have little documentation of natural history despite the prospects of treatments. MPS IV and VI are the least well documented of the MPS disorders with respect to their neurocognitive natural history. Because, like attenuated MPS I and II, they do not show progression of neurocognitive abnormality and most patients function in the range of normality, their behavioral, attentional, and executive function abnormalities have been ignored to the detriment of their quality of life. A peripheral treatment for MPS VII, extremely rare even among MPS types, has recently been approved with a post-approval monitoring system to provide neurocognitive natural history data in the future. More natural history studies in the MPS forms with milder cognitive deficits (MPS I, II, IV, and VI) are recommended with the goal of improving these patients' quality of life with and without new brain treatments, beyond the benefits of available peripheral enzyme replacement therapy. Recommendations are offered at-a-glance with respect to what areas most urgently need attention to clarify neurocognitive function in all MPS types.
Collapse
Affiliation(s)
- Elsa G Shapiro
- Department of Pediatrics, University of Minnesota, Minneapolis, MN, USA; Shapiro Neuropsychology Consulting LLC, Portland, OR, USA.
| | - Julie B Eisengart
- Department of Pediatrics, University of Minnesota, Minneapolis, MN, USA
| |
Collapse
|
17
|
Paget TL, Parkinson-Lawrence EJ, Trim PJ, Autilio C, Panchal MH, Koster G, Echaide M, Snel MF, Postle AD, Morrison JL, Pérez-Gil J, Orgeig S. Increased Alveolar Heparan Sulphate and Reduced Pulmonary Surfactant Amount and Function in the Mucopolysaccharidosis IIIA Mouse. Cells 2021; 10:849. [PMID: 33918094 PMCID: PMC8070179 DOI: 10.3390/cells10040849] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2021] [Revised: 03/18/2021] [Accepted: 03/24/2021] [Indexed: 02/07/2023] Open
Abstract
Mucopolysaccharidosis IIIA (MPS IIIA) is a lysosomal storage disease with significant neurological and skeletal pathologies. Respiratory dysfunction is a secondary pathology contributing to mortality in MPS IIIA patients. Pulmonary surfactant is crucial to optimal lung function and has not been investigated in MPS IIIA. We measured heparan sulphate (HS), lipids and surfactant proteins (SP) in pulmonary tissue and bronchoalveolar lavage fluid (BALF), and surfactant activity in healthy and diseased mice (20 weeks of age). Heparan sulphate, ganglioside GM3 and bis(monoacylglycero)phosphate (BMP) were increased in MPS IIIA lung tissue. There was an increase in HS and a decrease in BMP and cholesteryl esters (CE) in MPS IIIA BALF. Phospholipid composition remained unchanged, but BALF total phospholipids were reduced (49.70%) in MPS IIIA. There was a reduction in SP-A, -C and -D mRNA, SP-D protein in tissue and SP-A, -C and -D protein in BALF of MPS IIIA mice. Captive bubble surfactometry showed an increase in minimum and maximum surface tension and percent surface area compression, as well as a higher compressibility and hysteresis in MPS IIIA surfactant upon dynamic cycling. Collectively these biochemical and biophysical changes in alveolar surfactant are likely to be detrimental to lung function in MPS IIIA.
Collapse
Affiliation(s)
- Tamara L. Paget
- Mechanisms in Cell Biology and Disease Group, UniSA Clinical and Health Sciences, University of South Australia, Adelaide, SA 5000, Australia; (T.L.P.); (E.J.P.-L.)
| | - Emma J. Parkinson-Lawrence
- Mechanisms in Cell Biology and Disease Group, UniSA Clinical and Health Sciences, University of South Australia, Adelaide, SA 5000, Australia; (T.L.P.); (E.J.P.-L.)
| | - Paul J. Trim
- Proteomics, Metabolomics and MS-Imaging Core Facility, South Australian Health and Medical Research Institute, Adelaide, SA 5000, Australia; (P.J.T.); (M.F.S.)
| | - Chiara Autilio
- Department of Biochemistry, Faculty of Biology and Research Institute Hospital 12 de Octubre (Imas12), Complutense University, 28003 Madrid, Spain; (C.A.); (M.E.); (J.P.-G.)
| | - Madhuriben H. Panchal
- Faculty of Medicine, University of Southampton, Southampton SO16 6YD, UK; (M.H.P.); (G.K.); (A.D.P.)
| | - Grielof Koster
- Faculty of Medicine, University of Southampton, Southampton SO16 6YD, UK; (M.H.P.); (G.K.); (A.D.P.)
| | - Mercedes Echaide
- Department of Biochemistry, Faculty of Biology and Research Institute Hospital 12 de Octubre (Imas12), Complutense University, 28003 Madrid, Spain; (C.A.); (M.E.); (J.P.-G.)
| | - Marten F. Snel
- Proteomics, Metabolomics and MS-Imaging Core Facility, South Australian Health and Medical Research Institute, Adelaide, SA 5000, Australia; (P.J.T.); (M.F.S.)
| | - Anthony D. Postle
- Faculty of Medicine, University of Southampton, Southampton SO16 6YD, UK; (M.H.P.); (G.K.); (A.D.P.)
| | - Janna L. Morrison
- Early Origins Adult Health Research Group, Health and Biomedical Innovation, UniSA Clinical and Health Sciences, University of South Australia, Adelaide, SA 5000, Australia;
| | - Jésus Pérez-Gil
- Department of Biochemistry, Faculty of Biology and Research Institute Hospital 12 de Octubre (Imas12), Complutense University, 28003 Madrid, Spain; (C.A.); (M.E.); (J.P.-G.)
| | - Sandra Orgeig
- Mechanisms in Cell Biology and Disease Group, UniSA Clinical and Health Sciences, University of South Australia, Adelaide, SA 5000, Australia; (T.L.P.); (E.J.P.-L.)
| |
Collapse
|
18
|
Breyer SR, Vettorazzi E, Schmitz L, Gulati A, von Cossel KM, Spiro A, Rupprecht M, Stuecker R, Muschol NM. Hip pathologies in mucopolysaccharidosis type III. J Orthop Surg Res 2021; 16:201. [PMID: 33741007 PMCID: PMC7977579 DOI: 10.1186/s13018-021-02340-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/05/2020] [Accepted: 03/09/2021] [Indexed: 11/27/2022] Open
Abstract
Background Mucopolysaccharidosis type III (MPS III) comprises a group of rare lysosomal storage diseases. Although musculoskeletal symptoms are less pronounced than in other MPS subtypes, pathologies of hip and spine have been reported in MPS III patients. The purpose of this study was to describe hip pathologies and influencing parameters in MPS III patients. Methods A retrospective chart review was performed for 101 MPS III patients. Thirty-two patients met the inclusion criteria of enzymatically or genetically confirmed diagnosis and anteroposterior radiograph of the hips. Modified Ficat classification, Wiberg’s center-edge angle, and Reimer’s migration percentage were measured. Results The mean age at data assessment was 11.0 years (SD 5.7). Osteonecrosis of the femoral head was observed in 17/32 patients. No statistically significant association was found between these changes and age, sex, or MPS III subtype. Patients with a severe phenotype showed significantly higher rates of osteonecrosis (14/17) than patients with an intermediate phenotype. Hip dysplasia was present in 9/32 patients and was significantly associated with osteonecrosis of the femoral head (p = 0.04). Conclusions The present study demonstrates a high rate of hip pathologies in MPS III patients. Hip dysplasia and severe phenotype were significantly correlated with osteonecrosis of the femoral head. Therefore, radiographs of the hips are highly recommended in baseline and follow-up assessments of MPS III patients. Trial registration Retrospectively registered.
Collapse
Affiliation(s)
- Sandra Rafaela Breyer
- Department of Pediatric Orthopedics, Children's Hospital Altona, Bleickenallee 38, 22763, Hamburg, Germany. .,Department of Orthopedics, University Medical Center Hamburg-Eppendorf, 20246, Hamburg, Germany. .,International Center for Lysosomal Disorders, University Medical Center Hamburg-Eppendorf, 20246, Hamburg, Germany.
| | - Eik Vettorazzi
- Department of Medical Biometry and Epidemiology, University Medical Center Hamburg-Eppendorf, 20246, Hamburg, Germany
| | - Leonie Schmitz
- Department of Medical Biometry and Epidemiology, University Medical Center Hamburg-Eppendorf, 20246, Hamburg, Germany
| | - Amit Gulati
- Department of Medical Biometry and Epidemiology, University Medical Center Hamburg-Eppendorf, 20246, Hamburg, Germany
| | - Katharina Maria von Cossel
- International Center for Lysosomal Disorders, University Medical Center Hamburg-Eppendorf, 20246, Hamburg, Germany.,Department of Pediatrics, University Medical Center Hamburg-Eppendorf, 20246, Hamburg, Germany
| | - Alexander Spiro
- Department of Pediatric Orthopedics, Children's Hospital Altona, Bleickenallee 38, 22763, Hamburg, Germany.,Department of Orthopedics, University Medical Center Hamburg-Eppendorf, 20246, Hamburg, Germany.,International Center for Lysosomal Disorders, University Medical Center Hamburg-Eppendorf, 20246, Hamburg, Germany
| | - Martin Rupprecht
- Department of Pediatric Orthopedics, Children's Hospital Altona, Bleickenallee 38, 22763, Hamburg, Germany.,Department of Orthopedics, University Medical Center Hamburg-Eppendorf, 20246, Hamburg, Germany.,International Center for Lysosomal Disorders, University Medical Center Hamburg-Eppendorf, 20246, Hamburg, Germany
| | - Ralf Stuecker
- Department of Pediatric Orthopedics, Children's Hospital Altona, Bleickenallee 38, 22763, Hamburg, Germany.,Department of Orthopedics, University Medical Center Hamburg-Eppendorf, 20246, Hamburg, Germany.,International Center for Lysosomal Disorders, University Medical Center Hamburg-Eppendorf, 20246, Hamburg, Germany
| | - Nicole Maria Muschol
- International Center for Lysosomal Disorders, University Medical Center Hamburg-Eppendorf, 20246, Hamburg, Germany.,Department of Pediatrics, University Medical Center Hamburg-Eppendorf, 20246, Hamburg, Germany
| |
Collapse
|
19
|
Porter KA, O'Neill C, Drake E, Parker S, Escolar ML, Montgomery S, Moon W, Worrall C, Peay HL. Parent Experiences of Sanfilippo Syndrome Impact and Unmet Treatment Needs: A Qualitative Assessment. Neurol Ther 2020; 10:197-212. [PMID: 33263924 PMCID: PMC8139994 DOI: 10.1007/s40120-020-00226-z] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/14/2020] [Accepted: 11/19/2020] [Indexed: 12/04/2022] Open
Abstract
Introduction Sanfilippo syndrome (MPS III) is a rare, degenerative condition characterized by symptoms impacting cognitive ability, mobility, behavior, and quality of life. Currently there are no approved therapies for this severe life-limiting disease. Integrating patient and caregiver experience data into drug development and regulatory decision-making has become a priority of the Food and Drug Administration and rare disease patient communities. Methods This study assesses parents’ perceptions of their child’s Sanfilippo syndrome disease-related symptoms using a research approach that is consistent with the Center for Drug Evaluation and Research (CDER) guidance. This study was initiated by the Cure Sanfilippo Foundation, and all steps in the research process were informed by a multidisciplinary advisory committee, with an objective of informing biopharmaceutical companies and regulatory agencies. We explored caregiver burden, symptoms with greatest impact, and meaningful but unmet treatment needs. Data were collected from 25 parents through three focus groups and a questionnaire. Transcripts were coded and analyzed using inductive thematic analysis, and descriptive analysis of quantitative data was conducted. Results Participating parents’ children ranged in age from 4 to 36 years. Participants endorsed high caregiving burden across all stages of the disease. Analysis revealed multiple domains of unmet need that impact child and family quality of life, including cognitive-behavioral challenges in communication, relationships, behavior, anxiety, and child safety; and physical health symptoms including sleep, pain, and mobility. Participants reported placing high value on incremental benefits targeting those symptoms, and on a treatment that would slow or stop symptom progression. Conclusion Even modest treatment benefits for Sanfilippo syndrome were shown to be highly valued. Despite high caregiver burden, most parents expressed a willingness to “try anything,” including treatments with potentially high risk profiles, to maintain their child’s current state. Electronic supplementary material The online version of this article (10.1007/s40120-020-00226-z) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Katherine Ackerman Porter
- Center for Genomics, Bioinformatics, and Translational Research, RTI International, Research Triangle Park, NC, USA.
| | | | - Elise Drake
- Cure Sanfilippo Foundation, Columbia, SC, USA
| | - Samantha Parker
- Patient and Policy Affairs, Lysogene, Neuilly sur Seine, France
| | - Maria L Escolar
- University of Pittsburgh Medical Center, Pittsburgh, PA, USA.,Children's Hospital of Pittsburgh of UPMC, Pittsburgh, PA, USA
| | | | - William Moon
- Cure Sanfilippo Foundation Parent Advocates, Columbia, SC, USA
| | - Carolyn Worrall
- Cure Sanfilippo Foundation Parent Advocates, Columbia, SC, USA
| | - Holly L Peay
- Center for Genomics, Bioinformatics, and Translational Research, RTI International, Research Triangle Park, NC, USA
| |
Collapse
|
20
|
Pierzynowska K, Mański A, Limanówka M, Wierzba J, Gaffke L, Anikiej P, Węgrzyn G. Untypically mild phenotype of a patient suffering from Sanfilippo syndrome B with the c.638C>T/c.889C>T (p.Pro213Leu/p.Arg297Ter) mutations in the NAGLU gene. Mol Genet Genomic Med 2020; 8:e1356. [PMID: 32578945 PMCID: PMC7507323 DOI: 10.1002/mgg3.1356] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2020] [Revised: 05/19/2020] [Accepted: 05/21/2020] [Indexed: 12/19/2022] Open
Abstract
Background Sanfilippo syndrome B (or mucopolysaccharidosis type IIIB [MPS IIIB]) is a severe inherited metabolic disorder caused by mutations in the NAGLU gene, encoding α‐N‐acetylglucosaminidase. Dysfunction of this enzyme results in impaired degradation of heparan sulfate, one of glycosaminoglycans, and accumulation of this complex carbohydrate in lysosomes. Severe symptoms occurring in this disease are related to progressive neurodegeneration and include extreme hyperactivity, sleeping problems, aggressive‐like behavior, reduced fear, and progressive mental and cognitive deterioration. No cure is currently available for Sanfilippo disease. Methods Clinical characterization of the patient's symptoms has been performed. Biochemical analyses included glycosaminoglycan level determination and measurement of α‐N‐acetylglucosaminidase activity. Molecular analyses included exome sequencing and detailed analysis of the NAGLU gene. Psychological tests included assessment of attention, communication and behavior. Results We describe a patient with an untypically mild phenotype, who was diagnosed at the age of 13 years. Many cognitive, communication, and motoric functions were preserved in this patient, contrary to vast majority of those suffering from MPS IIIB. The patient is a compound heterozygote (c.638C>T/c.889C>T) in the NAGLU gene, and relatively high residual activity (about 25%) of α‐N‐acetylglucosaminidase was measured in serum (while no activity of this enzyme could be detected in dry blood spot). Conclusions We suggest that the mild phenotype might arise from the partially preserved function of the mutant enzyme (p.Pro213Leu), suggesting the genotype‐phenotype correlation in this case.
Collapse
Affiliation(s)
- Karolina Pierzynowska
- Department of Molecular Biology, Faculty of Biology, University of Gdańsk, Gdansk, Poland
| | - Arkadiusz Mański
- Psychological Counselling Centre of Rare Genetic Diseases, University of Gdańsk, Gdansk, Poland
| | - Monika Limanówka
- Departement of Pediatrics, Hematology and Oncology, Medical University of Gdańsk, Gdansk, Poland
| | - Jolanta Wierzba
- Department of Internal and Pediatric Nursing, Medical University of Gdańsk, Gdansk, Poland
| | - Lidia Gaffke
- Department of Molecular Biology, Faculty of Biology, University of Gdańsk, Gdansk, Poland
| | - Paulina Anikiej
- Psychological Counselling Centre of Rare Genetic Diseases, University of Gdańsk, Gdansk, Poland
| | - Grzegorz Węgrzyn
- Department of Molecular Biology, Faculty of Biology, University of Gdańsk, Gdansk, Poland
| |
Collapse
|
21
|
Köhn AF, Grigull L, du Moulin M, Kabisch S, Ammer L, Rudolph C, Muschol NM. Hematopoietic stem cell transplantation in mucopolysaccharidosis type IIIA: A case description and comparison with a genotype-matched control group. Mol Genet Metab Rep 2020; 23:100578. [PMID: 32226768 PMCID: PMC7093801 DOI: 10.1016/j.ymgmr.2020.100578] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2019] [Revised: 02/24/2020] [Accepted: 03/07/2020] [Indexed: 11/01/2022] Open
Abstract
BACKGROUND Mucopolysaccharidosis type IIIA (MPS IIIA, Sanfilippo A syndrome) is a chronic progressive neurodegenerative storage disorder caused by a deficiency of lysosomal sulfamidase. The clinical hallmarks are sleep disturbances, behavioral abnormalities and loss of cognitive, speech and motor abilities. Affected children show developmental slowing from the second year of life, dementia occurs by the age of 5 years followed by death in the second decade of life. Only a few studies concerning HSCT in MPS IIIA have been published and do not document a clear benefit of treatment. METHODS The present study summarizes the clinical outcome of a girl with MPS IIIA who received HSCT at the age of 2.5 years. Her clinical course was compared with the natural history of six untreated MPS IIIA patients carrying the same mutations (p.R74C and p. R245H) in the SGSH-gene. RESULTS Eight years after successful HSCT, the patient showed a global developmental delay. However, cognitive abilities continued to develop, albeit very slowly. There was no sign of regression. She could talk in short sentences, had good motor abilities and performed basic daily living activities by herself. She did not present with sleeping problems, but behavioral abnormalities were profound. In contrast, the six untreated patients with identical mutations in the SGSH-gene showed the typical progressive course of disease with early and continuous loss of abilities. CONCLUSIONS The present data suggest a beneficial effect of HSCT performed at an early stage of MPS IIIA on cognitive skills, motor function and quality of life.
Collapse
Key Words
- AEq, age-equivalent score
- ATG, antithymocyte globulin
- Avg., Average
- DQ, developmental quotient
- FPSS, four point scoring system
- GAG, Glykosaminoglycans
- HSCT
- HSCT, hematopoietic stem cell transplantation
- ICLD, International Center for Lysosomal Disorders
- MPS IH, mucopolysaccharidosis type I (Hurler syndrome)
- MPS IIIA
- MPS IIIA, mucopolysaccharidosis type IIIA
- MPS IIIB, mucopolysaccharidosis type IIIB
- Mucopolysaccharidosis type III
- Natural history
- SGSH, N-sulfoglucosamine sulfohydrolase
- Sanfilippo syndrome
- Stem cell transplantation
- TDS, total disability score
- UCBT, umbilical cord blood-derived hematopoietic stem cell transplantation
- VABS-II, Vineland Adaptive Behavior Scales
- y, years
Collapse
Affiliation(s)
- Anja F. Köhn
- Department of Pediatrics, University Medical Center Hamburg Eppendorf, Hamburg, Germany
| | - Lorenz Grigull
- Department of Pediatric Hematology and Oncology, Hannover Medical School, Hannover, Germany
| | - Marcel du Moulin
- Department of Pediatrics, University Medical Center Hamburg Eppendorf, Hamburg, Germany
| | - Sarah Kabisch
- Department of Pediatrics, University Medical Center Hamburg Eppendorf, Hamburg, Germany
| | - Luise Ammer
- Department of Pediatrics, University Medical Center Hamburg Eppendorf, Hamburg, Germany
| | - Cornelia Rudolph
- Department of Pediatrics, University Medical Center Hamburg Eppendorf, Hamburg, Germany
| | - Nicole M. Muschol
- Department of Pediatrics, University Medical Center Hamburg Eppendorf, Hamburg, Germany
| |
Collapse
|
22
|
Kong W, Meng Y, Zou L, Yang G, Wang J, Shi X. Mucopolysaccharidosis III in Mainland China: natural history, clinical and molecular characteristics of 34 patients. J Pediatr Endocrinol Metab 2020; 33:793-802. [PMID: 32447333 DOI: 10.1515/jpem-2019-0505] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/27/2019] [Accepted: 03/23/2020] [Indexed: 11/15/2022]
Abstract
Objectives Sanfilippo syndrome (Mucopolysaccharidosis III, MPS III) is a rare autosomal recessive hereditary disease, which is caused by lysosomal enzyme deficiency. This study was operated to investigate clinical and molecular characteristics of patients with MPS III, which will improve the diagnosis and treatment of MPS III. Method Thirty four patients with MPS III were assessed using clinical evaluation, questionnaire, and scoring system. Results Among the 34 patients, 14 had MPS IIIA, 19 had MPS III B, and one had MPS III C. Speech delay (100%) and intellectual disability (100%) were the most prevalent clinical manifestations in this cohort, followed by hyperactivity (94.12%), hirsutism (91.18%), enlarged head circumference (73.52%), repeated diarrhea (67.64%), sparse teeth (67.64%), and Mongolian spots (64.71%). There were two clinical manifestations that were significantly different between IIIA and IIIB: Hepatosplenomegaly and serrated teeth. The most common initial symptoms at diagnosis were speech delay (52.94%), hyperactivity (35.29%), and mental retardation (29.41%). Genetic analysis of 25 patients was conducted, which identified 12 novel mutations. Conclusion When language retardation, mental retardation, and rough facial features occurred, MPS III should be considered. At same time, more examination should be operated, such as examination of changes in cranial magnetic resonance imaging of cerebral cortex atrophy. Hepatosplenomegaly and serrated teeth could be used clinically to preliminarily distinguish IIIA from IIIB.
Collapse
Affiliation(s)
- Weijing Kong
- Department of Pediatrics, Chinese PLA General Hospital, Beijing, 100853, China.,Department of Pediatrics, Beijing Friendship Hospital, Capital Medical University, Beijing, 100050, China
| | - Yan Meng
- Department of Pediatrics, Chinese PLA General Hospital, Beijing, 100853, China
| | - Liping Zou
- Department of Pediatrics, Chinese PLA General Hospital, Beijing, 100853, China
| | - Guang Yang
- Department of Pediatrics, Chinese PLA General Hospital, Beijing, 100853, China
| | - Jing Wang
- Department of Pediatrics, Chinese PLA General Hospital, Beijing, 100853, China
| | - Xiuyu Shi
- Department of Pediatrics, Chinese PLA General Hospital, Beijing, 100853, China
| |
Collapse
|
23
|
Marcó S, Haurigot V, Bosch F. In Vivo Gene Therapy for Mucopolysaccharidosis Type III (Sanfilippo Syndrome): A New Treatment Horizon. Hum Gene Ther 2020; 30:1211-1221. [PMID: 31482754 DOI: 10.1089/hum.2019.217] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
For most lysosomal storage diseases (LSDs), there is no cure. Gene therapy is an attractive tool for treatment of LSDs caused by deficiencies in secretable lysosomal enzymes, in which neither full restoration of normal enzymatic activity nor transduction of all cells of the affected organ is necessary. However, some LSDs, such as mucopolysaccharidosis type III (MPSIII) diseases or Sanfilippo syndrome, represent a difficult challenge because patients suffer severe neurodegeneration with mild somatic alterations. The disease's main target is the central nervous system (CNS) and enzymes do not efficiently cross the blood-brain barrier (BBB) even if present at very high concentration in circulation. No specific treatment has been approved for MPSIII. In this study, we discuss the adeno-associated virus (AAV) vector-mediated gene transfer strategies currently being developed for MPSIII disease. These strategies rely on local delivery of AAV vectors to the CNS either through direct intraparenchymal injection at several sites or through delivery to the cerebrospinal fluid (CSF), which bathes the whole CNS, or exploit the properties of certain AAV serotypes capable of crossing the BBB upon systemic administration. Although studies in small and large animal models of MPSIII diseases have provided evidence supporting the efficacy and safety of all these strategies, there are considerable differences between the different routes of administration in terms of procedure-associated risks, vector dose requirements, sensitivity to the effect of circulating neutralizing antibodies that block AAV transduction, and potential toxicity. Ongoing clinical studies should shed light on which gene transfer strategy leads to highest clinical benefits while minimizing risks. The development of all these strategies opens a new horizon for treatment of not only MPSIII and other LSDs but also of a wide range of neurological diseases.
Collapse
Affiliation(s)
- Sara Marcó
- Center of Animal Biotechnology and Gene Therapy and Department of Biochemistry and Molecular Biology, Universitat Autònoma de Barcelona (UAB), Bellaterra, Spain
| | - Virginia Haurigot
- Center of Animal Biotechnology and Gene Therapy and Department of Biochemistry and Molecular Biology, Universitat Autònoma de Barcelona (UAB), Bellaterra, Spain
| | - Fatima Bosch
- Center of Animal Biotechnology and Gene Therapy and Department of Biochemistry and Molecular Biology, Universitat Autònoma de Barcelona (UAB), Bellaterra, Spain.,CIBER de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Madrid, Spain
| |
Collapse
|
24
|
Story BD, Miller ME, Bradbury AM, Million ED, Duan D, Taghian T, Faissler D, Fernau D, Beecy SJ, Gray-Edwards HL. Canine Models of Inherited Musculoskeletal and Neurodegenerative Diseases. Front Vet Sci 2020; 7:80. [PMID: 32219101 PMCID: PMC7078110 DOI: 10.3389/fvets.2020.00080] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2019] [Accepted: 01/31/2020] [Indexed: 12/11/2022] Open
Abstract
Mouse models of human disease remain the bread and butter of modern biology and therapeutic discovery. Nonetheless, more often than not mouse models do not reproduce the pathophysiology of the human conditions they are designed to mimic. Naturally occurring large animal models have predominantly been found in companion animals or livestock because of their emotional or economic value to modern society and, unlike mice, often recapitulate the human disease state. In particular, numerous models have been discovered in dogs and have a fundamental role in bridging proof of concept studies in mice to human clinical trials. The present article is a review that highlights current canine models of human diseases, including Alzheimer's disease, degenerative myelopathy, neuronal ceroid lipofuscinosis, globoid cell leukodystrophy, Duchenne muscular dystrophy, mucopolysaccharidosis, and fucosidosis. The goal of the review is to discuss canine and human neurodegenerative pathophysiologic similarities, introduce the animal models, and shed light on the ability of canine models to facilitate current and future treatment trials.
Collapse
Affiliation(s)
- Brett D. Story
- Auburn University College of Veterinary Medicine, Auburn, AL, United States
- University of Florida College of Veterinary Medicine, Gainesville, FL, United States
| | - Matthew E. Miller
- Auburn University College of Veterinary Medicine, Auburn, AL, United States
| | - Allison M. Bradbury
- Department of Clinical Sciences and Advanced Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Emily D. Million
- Department of Molecular Microbiology and Immunology, School of Medicine, University of Missouri, Columbia, MO, United States
| | - Dongsheng Duan
- Department of Molecular Microbiology and Immunology, School of Medicine, University of Missouri, Columbia, MO, United States
- Department of Biomedical, Biological and Chemical Engineering, College of Engineering, University of Missouri, Columbia, MO, United States
- Department of Biomedical Sciences, College of Veterinary Medicine, University of Missouri, Columbia, MO, United States
- Department of Neurology, School of Medicine, University of Missouri, Columbia, MO, United States
| | - Toloo Taghian
- Horae Gene Therapy Center, University of Massachusetts Medical School, Worcester, MA, United States
| | - Dominik Faissler
- Cummings School of Veterinary Medicine at Tufts University, North Grafton, MA, United States
| | - Deborah Fernau
- Horae Gene Therapy Center, University of Massachusetts Medical School, Worcester, MA, United States
| | - Sidney J. Beecy
- Horae Gene Therapy Center, University of Massachusetts Medical School, Worcester, MA, United States
- Cummings School of Veterinary Medicine at Tufts University, North Grafton, MA, United States
| | - Heather L. Gray-Edwards
- Horae Gene Therapy Center, University of Massachusetts Medical School, Worcester, MA, United States
- Department of Radiology, University of Massachusetts Medical School, Worcester, MA, United States
| |
Collapse
|
25
|
Saville JT, Fuller M. Sphingolipid dyshomeostasis in the brain of the mouse model of mucopolysaccharidosis type IIIA. Mol Genet Metab 2020; 129:111-116. [PMID: 31494022 DOI: 10.1016/j.ymgme.2019.08.008] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/29/2019] [Revised: 08/28/2019] [Accepted: 08/28/2019] [Indexed: 10/26/2022]
Abstract
Gangliosides are complex glycosphingolipids that are vital for proper brain development and function. Alterations in ganglioside metabolism are evident in neurological disorders including the inherited metabolic disease mucopolysaccharidosis type IIIA (MPS IIIA/Sanfilippo A syndrome). Here we sought to comprehensively analyse alterations in ganglioside metabolism within the brain of a naturally occurring MPS IIIA mouse model at early (one month) and late (six months of age) stages of disease progression, as well as the impact on related sphingolipids in the ganglioside metabolic pathway. The simple gangliosides GM2 and GM3 were elevated in the brain stem, cerebellum and sub-cortex of the MPS IIIA mouse at one month of age, but not in the cortex. By six months accumulation was significant throughout the brain, with GD2 gangliosides also elevated. Elevations in other sphingolipids were limited to the upstream synthetic precursors, ceramide and dihexosylceramide (DHC) species containing 18:0 and 20:0 acyl chains, likely due to the abundance of these fatty acids in the elevated gangliosides. In contrast, sphingomyelin, sulphatide and DHC containing a 24:1 fatty acid, were all decreased in the brain stem of the MPS IIIA mice, suggestive of alterations in myelination. These perturbations in sphingolipid metabolism could provide an avenue for therapeutic intervention by manipulation with specific drugs that target the production of these lipids.
Collapse
Affiliation(s)
- Jennifer T Saville
- Genetics and Molecular Pathology, SA Pathology at Women's and Children's Hospital, 72 King William Road, North Adelaide 5006, Australia
| | - Maria Fuller
- Genetics and Molecular Pathology, SA Pathology at Women's and Children's Hospital, 72 King William Road, North Adelaide 5006, Australia; School of Medicine, University of Adelaide, Adelaide 5005, Australia.
| |
Collapse
|
26
|
Heon-Roberts R, Nguyen ALA, Pshezhetsky AV. Molecular Bases of Neurodegeneration and Cognitive Decline, the Major Burden of Sanfilippo Disease. J Clin Med 2020; 9:jcm9020344. [PMID: 32012694 PMCID: PMC7074161 DOI: 10.3390/jcm9020344] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2019] [Revised: 01/20/2020] [Accepted: 01/22/2020] [Indexed: 12/13/2022] Open
Abstract
The mucopolysaccharidoses (MPS) are a group of diseases caused by the lysosomal accumulation of glycosaminoglycans, due to genetic deficiencies of enzymes involved in their degradation. MPS III or Sanfilippo disease, in particular, is characterized by early-onset severe, progressive neurodegeneration but mild somatic involvement, with patients losing milestones and previously acquired skills as the disease progresses. Despite being the focus of extensive research over the past years, the links between accumulation of the primary molecule, the glycosaminoglycan heparan sulfate, and the neurodegeneration seen in patients have yet to be fully elucidated. This review summarizes the current knowledge on the molecular bases of neurological decline in Sanfilippo disease. It emerges that this deterioration results from the dysregulation of multiple cellular pathways, leading to neuroinflammation, oxidative stress, impaired autophagy and defects in cellular signaling. However, many important questions about the neuropathological mechanisms of the disease remain unanswered, highlighting the need for further research in this area.
Collapse
Affiliation(s)
- Rachel Heon-Roberts
- Division of Medical Genetics, CHU Ste-Justine Research Centre, Montreal, QC H3T 1C5, Canada; (R.H.-R.); (A.L.A.N.)
- Department of Anatomy and Cell Biology, McGill University, Montreal, QC H3A 0C7, Canada
| | - Annie L. A. Nguyen
- Division of Medical Genetics, CHU Ste-Justine Research Centre, Montreal, QC H3T 1C5, Canada; (R.H.-R.); (A.L.A.N.)
- Department of Medicine, University of Montreal, Montreal, QC H3T 1J4, Canada
| | - Alexey V. Pshezhetsky
- Division of Medical Genetics, CHU Ste-Justine Research Centre, Montreal, QC H3T 1C5, Canada; (R.H.-R.); (A.L.A.N.)
- Department of Anatomy and Cell Biology, McGill University, Montreal, QC H3A 0C7, Canada
- Department of Paediatrics, University of Montreal, Montreal, QC H3T 1C5, Canada
- Correspondence: ; Tel.: +1-(514)-345-4931 (ext. 2736)
| |
Collapse
|
27
|
Escolar M, Bradshaw J, Byers VT, Giugliani R, Golightly L, Lourenço CM, McDonald K, Muschol N, Newsom-Davis I, O’Neill C, Peay HL, Siedman J, Solano ML, Wirt T, Wood T, Zwaigenbaum L. Development of a Clinical Algorithm for the Early Diagnosis of Mucopolysaccharidosis III. JOURNAL OF INBORN ERRORS OF METABOLISM AND SCREENING 2020. [DOI: 10.1590/2326-4594-jiems-2020-0002] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Affiliation(s)
| | | | | | - Roberto Giugliani
- Universidade Federal do Rio Grande do Sul, Brasil; Instituto Nacional de Ciência e Tecnologia de Genética Médica Populacional, Brasil
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
28
|
Nijmeijer SCM, van den Born LI, Kievit AJA, Stepien KM, Langendonk J, Marchal JP, Roosing S, Wijburg FA, Wagenmakers MAEM. The attenuated end of the phenotypic spectrum in MPS III: from late-onset stable cognitive impairment to a non-neuronopathic phenotype. Orphanet J Rare Dis 2019; 14:249. [PMID: 31718697 PMCID: PMC6852993 DOI: 10.1186/s13023-019-1232-0] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2019] [Accepted: 10/22/2019] [Indexed: 01/17/2023] Open
Abstract
BACKGROUND The phenotypic spectrum of many rare disorders is much wider than previously considered. Mucopolysaccharidosis type III (Sanfilippo syndrome, MPS III), is a lysosomal storage disorder traditionally considered to be characterized by childhood onset, progressive neurocognitive deterioration with a rapidly or slowly progressing phenotype. The presented MPS III case series demonstrates adult onset phenotypes with mild cognitive impairment or non-neuronopathic phenotypes. METHODS In this case series all adult MPS III patients with a mild- or non-neuronopathic phenotype, who attend the outpatient clinic of 3 expert centers for lysosomal storage disorders were included. A mild- or non-neuronopathic phenotype was defined as having completed regular secondary education and attaining a level of independency during adulthood, involving either independent living or a paid job. RESULTS Twelve patients from six families, with a median age at diagnosis of 43 years (range 3-68) were included (11 MPS IIIA, 1 MPS IIIB). In the four index patients symptoms which led to diagnostic studies (whole exome sequencing and metabolomics) resulting in the diagnosis of MPS III; two patients presented with retinal dystrophy, one with hypertrophic cardiomyopathy and one with neurocognitive decline. The other eight patients were diagnosed by family screening. At a median age of 47 years (range 19-74) 9 out of the 12 patients had normal cognitive functions. Nine patients had retinal dystrophy and 8 patients hypertrophic cardiomyopathy. CONCLUSION We show the very mild end of the phenotypic spectrum of MPS III, ranging from late-onset stable neurocognitive impairment to a fully non-neuronopathic phenotype. Awareness of this phenotype could lead to timely diagnosis and genetic counseling.
Collapse
Affiliation(s)
- Stephanie C M Nijmeijer
- Amsterdam UMC, Pediatric Metabolic Diseases, Amsterdam Lysosome Center "Sphinx", University of Amsterdam, H8-264, Meibergdreef 9, Amsterdam, The Netherlands
| | | | - Anneke J A Kievit
- Erasmus MC, Department of Clinical Genetics, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Karolina M Stepien
- Salford Royal NHS Foundation Trust, Adult Inherited Metabolic Disorders, Mark Holland Metabolic Unit, Salford, UK
| | - Janneke Langendonk
- Erasmus MC, Center for Lysosomal and Metabolic disease, Department of Internal Medicine, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Jan Pieter Marchal
- Amsterdam UMC, Psychosocial Department, Amsterdam Public Health Research Institute, University of Amsterdam, Amsterdam, The Netherlands
| | - Susanne Roosing
- Radboud University Medical Center, Donders Institute for Brain, Cognition and Behaviour, Department of Human Genetics, Nijmegen, The Netherlands
| | - Frits A Wijburg
- Amsterdam UMC, Pediatric Metabolic Diseases, Amsterdam Lysosome Center "Sphinx", University of Amsterdam, H8-264, Meibergdreef 9, Amsterdam, The Netherlands.
| | - Margreet A E M Wagenmakers
- Erasmus MC, Center for Lysosomal and Metabolic disease, Department of Internal Medicine, University Medical Center Rotterdam, Rotterdam, The Netherlands
| |
Collapse
|
29
|
Saville JT, Flanigan KM, Truxal KV, McBride KL, Fuller M. Evaluation of biomarkers for Sanfilippo syndrome. Mol Genet Metab 2019; 128:68-74. [PMID: 31104888 DOI: 10.1016/j.ymgme.2019.05.005] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/24/2019] [Accepted: 05/07/2019] [Indexed: 11/16/2022]
Abstract
Sanfilippo syndrome or mucopolysaccharidosis type III (MPS III) is a childhood metabolic disorder marked by neuropathology arising due to impaired heparan sulphate (HS) catabolism. Consequently, partially degraded HS accumulates in the lysosomes of affected cells and is excreted in the urine. The measurement of HS in urine has long been considered a biomarker of Sanfilippo syndrome although it is largely non-specific. Using blood, urine and CSF collected from a cohort of Sanfilippo patients we investigated the utility of primary and secondary biomarkers to inform on disease activity. These included enzyme activity, specific oligosaccharides with non-reducing end residues reflective of the enzyme deficiency, and gangliosides. The diagnostic oligosaccharides - a HS disaccharide and tetrasaccharide - were elevated in the urine, plasma and CSF of all MPS IIIA and IIIB patients, respectively. There was no correlation between the concentrations in any of the matrices suggesting they reflect specific tissues and not overall disease burden. Enzyme activity did not inform on disease severity, with no measurable activity in CSF and activity approaching normal in MPS IIIA plasma. The concentration of gangliosides, GM2 and GM3, were significantly higher in the CSF of all MPS III subjects when compared to controls and correlated with the age of onset of first symptoms. Given that these gangliosides reflect delayed brain development they may be useful measures of disease burden, within the limitations of the clinical surrogates. Observation of these biochemical measurements in MPS III patients enrolled in clinical trials may determine whether they represent true pharmacodynamics biomarkers.
Collapse
Affiliation(s)
- Jennifer T Saville
- Genetics and Molecular Pathology, SA Pathology at Women's and Children's Hospital, 72 King William Road, North Adelaide 5006, Australia
| | - Kevin M Flanigan
- Center for Gene Therapy, The Research Institute at Nationwide Children's Hospital, Columbus, OH, USA
| | - Kristen V Truxal
- The Division of Genetics and Genomic Medicine, Nationwide Children's Hospital, Columbus, OH, USA
| | - Kim L McBride
- The Division of Genetics and Genomic Medicine, Nationwide Children's Hospital, Columbus, OH, USA
| | - Maria Fuller
- Genetics and Molecular Pathology, SA Pathology at Women's and Children's Hospital, 72 King William Road, North Adelaide 5006, Australia; School of Medicine, University of Adelaide, Adelaide 5005, Australia.
| |
Collapse
|
30
|
Lee CL, Lin HY, Chuang CK, Chiu HC, Tu RY, Huang YH, Hwu WL, Tsai FJ, Chiu PC, Niu DM, Chen YJ, Chao MC, Chang TM, Lin JL, Chang CY, Kao YC, Lin SP. Functional independence of Taiwanese patients with mucopolysaccharidoses. Mol Genet Genomic Med 2019; 7:e790. [PMID: 31215158 PMCID: PMC6687640 DOI: 10.1002/mgg3.790] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2019] [Revised: 05/05/2019] [Accepted: 05/17/2019] [Indexed: 11/12/2022] Open
Abstract
BACKGROUND Information on functional strengths and weaknesses of mucopolysaccharidosis (MPS) patients is important for early intervention programs and enzyme replacement therapy (ERT). METHODS We used the Functional Independence Measure for Children (WeeFIM) questionnaire to assess the functional skills of 63 Taiwanese MPS patients (median age, 13 years 3 months; range, 3-20 years) from January 2012 to December 2018. RESULTS Mean total WeeFIM score was 75.4 of a potential score of 126. Mean total WeeFIM scores of each type (MPS I, MPS II, MPS IIIB, MPS IVA, and MPS VI) were 103.8, 76.2, 41.6, 92.2, and 113.6, respectively. Mean scores for self-care, mobility, and cognition domains were 30 (maximum 56), 23 (maximum 35), and 22 (maximum 35), respectively. MPS type IIIB patients had the lowest scores in self-care, mobility, cognition, and total domains compared to other types of MPS. All patients with ERT in MPS I, II, and IVA had higher scores in self-care and mobility domains than patients without ERT. Most patients required assistance for self-care skills, especially in grooming and bathing. CONCLUSION MPS patients require support and supervision in self-care tasks. For cognition tasks, MPS IIIB patients also require help. This questionnaire is useful to identify the strengths and limitations of MPS patients.
Collapse
Affiliation(s)
- Chung-Lin Lee
- Department of Pediatrics, Mackay Memorial Hospital, Hsinchu, Taiwan
| | - Hsiang-Yu Lin
- Department of Pediatrics, Mackay Memorial Hospital, Taipei, Taiwan.,Department of Medicine, Mackay Medical College, New Taipei City, Taiwan.,Division of Genetics and Metabolism, Department of Medical Research, MacKay Memorial Hospital, Taipei, Taiwan.,Mackay Junior College of Medicine, Nursing and Management, Taipei, Taiwan.,Department of Medical Research, China Medical University Hospital, China Medical University, Taichung, Taiwan
| | - Chih-Kuang Chuang
- Division of Genetics and Metabolism, Department of Medical Research, MacKay Memorial Hospital, Taipei, Taiwan.,College of Medicine, Fu-Jen Catholic University, Taipei, Taiwan
| | - Huei-Ching Chiu
- Department of Pediatrics, Mackay Memorial Hospital, Taipei, Taiwan
| | - Ru-Yi Tu
- Division of Genetics and Metabolism, Department of Medical Research, MacKay Memorial Hospital, Taipei, Taiwan
| | - You-Hsin Huang
- Department of Pediatrics, Mackay Memorial Hospital, Taipei, Taiwan
| | - Wuh-Liang Hwu
- Department of Pediatrics, National Taiwan University Hospital, Taipei, Taiwan
| | - Fuu-Jen Tsai
- Department of Medical Research, Genetics Center, China Medical University Hospital, Taichung, Taiwan
| | - Pao-Chin Chiu
- Department of Pediatrics, Kaohsiung Veterans General Hospital, Kaohsiung, Taiwan
| | - Dau-Ming Niu
- Institute of Clinical Medicine, National Yang-Ming University, Taipei, Taiwan.,Department of Pediatrics, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Yann-Jang Chen
- Institute of Clinical Medicine, National Yang-Ming University, Taipei, Taiwan.,Department of Pediatrics, Renai Branch, Taipei City Hospital, Taipei, Taiwan
| | - Mei-Chyn Chao
- Department of Pediatrics, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan.,Department of Pediatric Neurology, Changhua Christian Children's Hospital, Changhua, Taiwan
| | - Tung-Ming Chang
- Department of Pediatric Neurology, Changhua Christian Children's Hospital, Changhua, Taiwan.,Department of Biological Science and Technology, College of Biological Science and Technology, National Chiao Tung University, Hsinchu, Taiwan
| | - Ju-Li Lin
- Department of Pediatrics, Chang-Gung Memorial Hospital, Taoyuan, Taiwan
| | - Chia-Ying Chang
- Department of Pediatrics, Mackay Memorial Hospital, Hsinchu, Taiwan
| | - Yu-Chia Kao
- Department of Pediatrics, E-DA Hospital, Kaohsiung, Taiwan
| | - Shuan-Pei Lin
- Department of Pediatrics, Mackay Memorial Hospital, Taipei, Taiwan.,Department of Medicine, Mackay Medical College, New Taipei City, Taiwan.,Division of Genetics and Metabolism, Department of Medical Research, MacKay Memorial Hospital, Taipei, Taiwan.,Department of Infant and Child Care, National Taipei University of Nursing and Health Sciences, Taipei, Taiwan
| |
Collapse
|
31
|
McIntyre C, Saville J, Fuller M. Collection of cerebrospinal fluid from murine lateral ventricles for biomarker determination in mucopolysaccharidosis type IIIA. J Neurosci Methods 2019; 324:108314. [PMID: 31202732 DOI: 10.1016/j.jneumeth.2019.108314] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2019] [Revised: 06/12/2019] [Accepted: 06/12/2019] [Indexed: 10/26/2022]
Abstract
BACKGROUND Cerebrospinal fluid (CSF) collection is currently the only feasible means to obtain biological fluid for the quantitative determination of biomarkers that may reflect disease activity within the brain. Studies in mouse models of human neurological disease benefit from ascertainment and subsequent analysis of brain tissue that is not afforded in human patients. The CSF provides a translational forum, however, due to the practical constraints presented by the mouse's small size, CSF is often ignored in experimental mouse models. NEW METHOD Here we report a method for the controlled, precise and predictable collection of 10 μL of CSF from the lateral ventricles of adult mice using stereotaxic equipment and a micro-syringe pump. RESULTS Collected CSF was clear and manifested the consistency of water and moreover, quantification of a disease-specific biomarker for the neurodegenerative disorder, mucopolysaccharidosis type IIIA (MPS IIIA) was possible in this small volume of CSF. In the naturally occurring mouse model of MPS IIIA, that faithfully recapitulates the human form of the disease, this biomarker was present at concentrations of >100 pmol/mL and undetectable in wild-type mice. COMPARISON WITH EXISTING METHOD Advantages of this method over the most commonly used cisterna magna collection technique include increased CSF sample volume (10 μL) and reduced blood contamination. CONCLUSION The ability to collect CSF from mouse models of neurological disease enables a forum for translating research outcomes in experimental models to the human equivalent in which CSF collection is also possible.
Collapse
Affiliation(s)
- Chantelle McIntyre
- Genetics and Molecular Pathology, SA Pathology at Women's and Children's Hospital, 72 King William Rd, North Adelaide, South Australia, 5006, Australia; School of Medicine, University of Adelaide, Adelaide Health and Medical Sciences building, 4 North Terrace, Adelaide, South Australia, 5000, Australia
| | - Jennifer Saville
- Genetics and Molecular Pathology, SA Pathology at Women's and Children's Hospital, 72 King William Rd, North Adelaide, South Australia, 5006, Australia
| | - Maria Fuller
- Genetics and Molecular Pathology, SA Pathology at Women's and Children's Hospital, 72 King William Rd, North Adelaide, South Australia, 5006, Australia; School of Medicine, University of Adelaide, Adelaide Health and Medical Sciences building, 4 North Terrace, Adelaide, South Australia, 5000, Australia.
| |
Collapse
|
32
|
Muschol NM, Pape D, Kossow K, Ullrich K, Arash-Kaps L, Hennermann JB, Stücker R, Breyer SR. Growth charts for patients with Sanfilippo syndrome (Mucopolysaccharidosis type III). Orphanet J Rare Dis 2019; 14:93. [PMID: 31046785 PMCID: PMC6498678 DOI: 10.1186/s13023-019-1065-x] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2019] [Accepted: 04/16/2019] [Indexed: 01/17/2023] Open
Abstract
Background Mucopolysaccharidosis (MPS) type III (Sanfilippo syndrome) comprises a group of rare, lysosomal storage diseases caused by the deficiency of one of four enzymes involved in the degradation of heparan sulfate. The clinical hallmark of the disease is severe neurological deterioration leading to dementia and death in the second decade of life. Adult MPS patients are generally of short stature. To date there is no clear description of the physical development of MPS III patients. The aim of this study was to document growth reference data for MPS III patients. We collected growth data of 182 German MPS III patients and were able to develop growth charts for this cohort. Growth curves for height, weight, head circumference, and body mass index were calculated and compared to German reference charts. Results Birth height, weight and head circumference were within the physiological ranges. Both genders were significantly taller than healthy children at 2 years of age, while only male patients were taller at the age of four. Growth velocity decelerated after the ages of 4.5 and 5 years for female and male patients, respectively. Both genders were significantly shorter than the reference group at the age of 17.5 years. Head circumference was larger compared to healthy matched controls within the first 2 years of life and remained enlarged until physical maturity. Conclusion MPS III is a not yet treatable severe neuro-degenerative disease, developing new therapeutic strategies might change the course of the disease significantly. The present charts contribute to the understanding of the natural history of MPS III. Specific growth charts represent an important tool for families and physicians as the expected height at physical maturity can be estimated and therapeutic effects can be monitored. Electronic supplementary material The online version of this article (10.1186/s13023-019-1065-x) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Nicole M Muschol
- Department of Pediatrics, University Medical Center Hamburg-Eppendorf, Martinistr. 52, 20246, Hamburg, Germany
| | - Daniel Pape
- Department of Pediatrics, University Medical Center Hamburg-Eppendorf, Martinistr. 52, 20246, Hamburg, Germany
| | - Kai Kossow
- Department of Medical Psychology, University Medical Center Hamburg-Eppendorf, Martinistr. 52, 20246, Hamburg, Germany
| | - Kurt Ullrich
- Martin Zeitz Center for Rare Diseases, University Medical Center Hamburg-Eppendorf, Martinistr.52, 20246, Hamburg, Germany
| | - Laila Arash-Kaps
- Villa Metabolica, Department of Pediatric and Adolescent Medicine, University Medical Center Mainz, Langenbeckstr. 1, 55131, Mainz, Germany
| | - Julia B Hennermann
- Villa Metabolica, Department of Pediatric and Adolescent Medicine, University Medical Center Mainz, Langenbeckstr. 1, 55131, Mainz, Germany
| | - Ralf Stücker
- Department of Pediatric Orthopedics, Altonaer Children's Hospital, Bleickenallee 38, 22763, Hamburg, Germany.,Department of Orthopedics, University Medical Center Hamburg-Eppendorf, Martinistr. 52, 20246, Hamburg, Germany
| | - Sandra R Breyer
- Department of Pediatric Orthopedics, Altonaer Children's Hospital, Bleickenallee 38, 22763, Hamburg, Germany. .,Department of Orthopedics, University Medical Center Hamburg-Eppendorf, Martinistr. 52, 20246, Hamburg, Germany.
| |
Collapse
|
33
|
Abstract
The mucopolysaccharidoses (MPS) are clinically similar but also heterogeneous in terms of major or minor involvement of different organs/systems, burden of disease, and rate of progression. The attenuated forms of MPS, due to their less severe presentations, are more difficult to diagnose and often receive a significantly delayed diagnosis. On the other hand, the diagnosis is very important since the attenuated forms may benefit from earlier treatments. The aim of this paper is to describe the natural history and the clinical signs useful to arise a suspicion of an attenuated form of MPS. MPS patients usually show a cluster of signs and symptoms, one of which may be the trigger for an evaluation by a specialist. Individuals with attenuated MPS are mostly cognitively normal, and dysmorphisms of the facies may be mild or absent. The most frequently involved organs/systems are the osteoarticular system, heart, and eyes. These patients may also have hepatosplenomegaly, hearing loss, and respiratory problems. When they are referred to a specialist (rheumatologist, cardiologist, ophthalmologist, surgeon, orthopedist, etc.) for their main complaint, the other signs and symptoms are likely to be missed in the medical history. To avoid missing data and to save time, we propose a semistructured medical history form to be filled in by the patients or their caregivers while waiting for evaluation by a specialist.
Collapse
Affiliation(s)
- Miriam Rigoldi
- Centro Malattie Rare, ASST-Monza, Ospedale San Gerardo, Via Pergolesi, 33 20900, Monza, MB, Italy.
| | - Elena Verrecchia
- Centro delle febbri periodiche e malattie rare, Policlinico Gemelli, Università Cattolica Roma, Rome, Italy
| | - Raffaele Manna
- Centro delle febbri periodiche e malattie rare, Policlinico Gemelli, Università Cattolica Roma, Rome, Italy
| | - Maria Teresa Mascia
- Patologie dell'apparato locomotore a genesi immunologica, Università di Modena e Reggio Emilia, Azienda Ospedaliero-Universitaria, Policlinico di Modena, Modena, Italy
| |
Collapse
|
34
|
Abstract
Mucopolysaccharidoses (MPS) are a group of lysosomal multisystemic, chronic, and progressive diseases characterized by the storage of glycosaminoglycans (GAGs) that may affect the central nervous system. Neuronopathic MPS such as MPS IH, MPS II, MPS IIIA–D, and MPS VII are characterized by neurocognitive regression. In severe MPS I (MPS IH, or Hurler syndrome) initial developmental trajectory is usually unremarkable but cognitive development shows a plateau by 2 to 4 years of age and then progressively regresses with aging. Patients with neuronopathic MPS II have a plateau of cognitive and adaptive development on average by 4 to 4.5 years of age, although there is significant variability, followed by progressive neurocognitive decline. In patients with classic MPS III, developmental trajectory reaches a plateau around 3 years of age, followed by regression. Sleep disturbances and behavioral problems occur early in MPS II and III with features of externalizing disorders. Acquired autism-like behavior is often observed in children with MPS III after 4–6 years of age. Impaired social and communication abilities do occur, but MPS III children do not have restricted and repetitive interests such as in autism spectrum disorder. MPS type VII is an ultra-rare neuronopathic MPS with a wide clinical spectrum from very severe with early mortality to milder phenotypes with longer survival into adolescence and adulthood. Most patients with MPS VII have intellectual disability and severely delayed speech development, usually associated with hearing impairment. Cognitive regression in neuronopathic MPS runs parallel to a significant decrease in brain tissue volume. Assessment of the developmental profile is challenging because of low cognitive abilities, physical impairment, and behavioral disturbances. Early diagnosis is crucial as different promising treatment approaches have been extensively studied in animal MPS models and are currently being applied in clinical trials.
Collapse
Affiliation(s)
- Rita Barone
- Neuropsichiatria Infantile, Dipartimento di Medicina Clinica e Sperimentale, Università di Catania, Catania, Italy. .,Neuropsichiatria Infantile, Policlinico, Università di Catania, Via S. Sofia 78, 95123, Catania, Italy.
| | - Alessandra Pellico
- Neuropsichiatria Infantile, Dipartimento di Medicina Clinica e Sperimentale, Università di Catania, Catania, Italy
| | - Annarita Pittalà
- Centro di Riferimento Regionale per le malattie metaboliche congenite, Policlinico, Università di Catania, Catania, Italy
| | - Serena Gasperini
- UOS Malattie Metaboliche Rare, Clinica Pediatrica, Fondazione MBBM, ATS Monza, Monza, Italy
| |
Collapse
|
35
|
Tebani A, Abily-Donval L, Schmitz-Afonso I, Héron B, Piraud M, Ausseil J, Zerimech F, Gonzalez B, Marret S, Afonso C, Bekri S. Unveiling metabolic remodeling in mucopolysaccharidosis type III through integrative metabolomics and pathway analysis. J Transl Med 2018; 16:248. [PMID: 30180851 PMCID: PMC6122730 DOI: 10.1186/s12967-018-1625-1] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2018] [Accepted: 08/30/2018] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND Metabolomics represent a valuable tool to recover biological information using body fluids and may help to characterize pathophysiological mechanisms of the studied disease. This approach has not been widely used to explore inherited metabolic diseases. This study investigates mucopolysaccharidosis type III (MPS III). A thorough and holistic understanding of metabolic remodeling in MPS III may allow the development, improvement and personalization of patient care. METHODS We applied both targeted and untargeted metabolomics to urine samples obtained from a French cohort of 49 patients, consisting of 13 MPS IIIA, 16 MPS IIIB, 13 MPS IIIC, and 7 MPS IIID, along with 66 controls. The analytical strategy is based on ultra-high-performance liquid chromatography combined with ion mobility and high-resolution mass spectrometry. Twenty-four amino acids have been assessed using tandem mass spectrometry combined with liquid chromatography. Multivariate data modeling has been used for discriminant metabolite selection. Pathway analysis has been performed to retrieve metabolic pathways impairments. RESULTS Data analysis revealed distinct biochemical profiles. These metabolic patterns, particularly those related to the amino acid metabolisms, allowed the different studied groups to be distinguished. Pathway analysis unveiled major amino acid pathways impairments in MPS III mainly arginine-proline metabolism and urea cycle metabolism. CONCLUSION This represents one of the first metabolomics-based investigations of MPS III. These results may shed light on MPS III pathophysiology and could help to set more targeted studies to infer the biomarkers of the affected pathways, which is crucial for rare conditions such as MPS III.
Collapse
Affiliation(s)
- Abdellah Tebani
- Department of Metabolic Biochemistry, Rouen University Hospital, 76000, Rouen Cedex, France.,Normandie Univ, UNIROUEN, CHU Rouen, INSERM U1245, 76000, Rouen, France.,Normandie Univ, UNIROUEN, INSA Rouen, CNRS, COBRA, 76000, Rouen, France
| | - Lenaig Abily-Donval
- Normandie Univ, UNIROUEN, CHU Rouen, INSERM U1245, 76000, Rouen, France.,Department of Neonatal Pediatrics, Intensive Care and Neuropediatrics, Rouen University Hospital, 76031, Rouen, France
| | | | - Bénédicte Héron
- Department of Pediatric Neurology, Reference Center of Lysosomal Diseases, Trousseau Hospital, APHP and Sorbonne Université, GRC No 19, Pathologies Congénitales du Cervelet-LeucoDystrophies, AP-HP, Hôpital Armand Trousseau, 75012, Paris, France
| | - Monique Piraud
- Service de Biochimie et Biologie Moléculaire Grand Est, Unité des Maladies Héréditaires du Métabolisme et Dépistage Néonatal, Centre de Biologie et de Pathologie Est, CHU de Lyon, Lyon, France
| | - Jérôme Ausseil
- INSERM U1088, Laboratoire de Biochimie Métabolique, Centre de Biologie Humaine, CHU Sud, 80054, Amiens Cedex, France
| | - Farid Zerimech
- Laboratoire de Biochimie et Biologie Moléculaire, Université de Lille et Pôle de Biologie Pathologie Génétique du CHRU de Lille, 59000, Lille, France
| | - Bruno Gonzalez
- Normandie Univ, UNIROUEN, CHU Rouen, INSERM U1245, 76000, Rouen, France
| | - Stéphane Marret
- Normandie Univ, UNIROUEN, CHU Rouen, INSERM U1245, 76000, Rouen, France.,Department of Neonatal Pediatrics, Intensive Care and Neuropediatrics, Rouen University Hospital, 76031, Rouen, France
| | - Carlos Afonso
- Normandie Univ, UNIROUEN, INSA Rouen, CNRS, COBRA, 76000, Rouen, France
| | - Soumeya Bekri
- Department of Metabolic Biochemistry, Rouen University Hospital, 76000, Rouen Cedex, France. .,Normandie Univ, UNIROUEN, CHU Rouen, INSERM U1245, 76000, Rouen, France.
| |
Collapse
|
36
|
Lin HY, Chuang CK, Lee CL, Tu RY, Lo YT, Chiu PC, Niu DM, Fang YY, Chen TL, Tsai FJ, Hwu WL, Lin SJ, Chang TM, Lin SP. Mucopolysaccharidosis III in Taiwan: Natural history, clinical and molecular characteristics of 28 patients diagnosed during a 21-year period. Am J Med Genet A 2018; 176:1799-1809. [PMID: 30070758 DOI: 10.1002/ajmg.a.40351] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2018] [Revised: 05/04/2018] [Accepted: 05/07/2018] [Indexed: 01/01/2023]
Abstract
Mucopolysaccharidosis type III (MPS III, Sanfilippo syndrome) has a variable age of onset and variable rate of progression. However, information regarding the natural history of this disorder in Asian populations is limited. A retrospective analysis was carried out for 28 patients with MPS III (types IIIA [n = 3], IIIB [n = 23], and IIIC [n = 2]; 15 males and 13 females; median age, 8.2 years; age range, 2.7-26.5 years) seen in six medical centers in Taiwan from January 1996 through October 2017. The median age at confirmed diagnosis was 4.6 years. The most common initial symptom was speech delay (75%), followed by hirsutism (64%) and hyperactivity (54%). Both z scores for height and weight were negatively correlated with age (r = -.693 and -0.718, respectively; p < .01). The most prevalent clinical manifestations were speech delay (100%) and intellectual disability (100%), followed by hirsutism (93%), hyperactivity (79%), coarse facial features (68%), sleep disorders (61%), and hepatosplenomegaly (61%). Ten patients (36%) had epilepsy, and the median age at the first seizure was 11 years. Thirteen patients (46%) experienced at least one surgical procedure. At the time of the present study, 7 of the 28 patients had passed away at the median age of 13.0 years. Molecular studies showed an allelic heterogeneity without clear genotype and phenotype correlations. MPS IIIB is the most frequent subtype among MPS III in the Taiwanese population. An understanding of the natural history of MPS III may allow early diagnosis and timely management of the disease facilitating better treatment outcomes.
Collapse
Affiliation(s)
- Hsiang-Yu Lin
- Department of Medicine, Mackay Medical College, New Taipei City, Taiwan.,Department of Pediatrics, Mackay Memorial Hospital, Taipei, Taiwan.,Department of Medical Research, Mackay Memorial Hospital, Taipei, Taiwan.,Mackay Junior College of Medicine, Nursing and Management, Taipei, Taiwan.,Department of Medical Research, China Medical University Hospital, China Medical University, Taichung, Taiwan
| | - Chih-Kuang Chuang
- Department of Medical Research, Mackay Memorial Hospital, Taipei, Taiwan.,Medical College, Fu-Jen Catholic University, Taipei, Taiwan
| | - Chung-Lin Lee
- Department of Pediatrics, Mackay Memorial Hospital, Taipei, Taiwan
| | - Ru-Yi Tu
- Department of Medical Research, Mackay Memorial Hospital, Taipei, Taiwan
| | - Yun-Ting Lo
- Department of Laboratory Medicine, Mackay Memorial Hospital, Taipei, Taiwan
| | - Pao Chin Chiu
- Department of Pediatrics, Kaohsiung Veterans General Hospital, Kaohsiung, Taiwan
| | - Dau-Ming Niu
- Department of Pediatrics, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Yi-Ya Fang
- Department of Medical Research, Mackay Memorial Hospital, Taipei, Taiwan
| | - Tzu-Lin Chen
- Department of Laboratory Medicine, Mackay Memorial Hospital, Taipei, Taiwan
| | - Fuu-Jen Tsai
- Department of Pediatrics, China Medical University Hospital, Taichung, Taiwan
| | - Wuh-Liang Hwu
- Department of Pediatrics, National Taiwan University Hospital, Taipei, Taiwan
| | - Shio Jean Lin
- Department of Pediatrics, Chi Mei Medical Center, Tainan, Taiwan
| | - Tung-Ming Chang
- Department of Pediatric Neurology, Changhua Christian Children's Hospital, Changhua, Taiwan.,Department of Biological Science and Technology, College of Biological Science and Technology, National Chiao Tung University, Hsinchu, Taiwan
| | - Shuan-Pei Lin
- Department of Medicine, Mackay Medical College, New Taipei City, Taiwan.,Department of Pediatrics, Mackay Memorial Hospital, Taipei, Taiwan.,Department of Medical Research, Mackay Memorial Hospital, Taipei, Taiwan.,Department of Infant and Child Care, National Taipei University of Nursing and Health Sciences, Taipei, Taiwan
| |
Collapse
|
37
|
Lotfi P, Tse DY, Di Ronza A, Seymour ML, Martano G, Cooper JD, Pereira FA, Passafaro M, Wu SM, Sardiello M. Trehalose reduces retinal degeneration, neuroinflammation and storage burden caused by a lysosomal hydrolase deficiency. Autophagy 2018; 14:1419-1434. [PMID: 29916295 PMCID: PMC6103706 DOI: 10.1080/15548627.2018.1474313] [Citation(s) in RCA: 74] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
The accumulation of undegraded molecular material leads to progressive neurodegeneration in a number of lysosomal storage disorders (LSDs) that are caused by functional deficiencies of lysosomal hydrolases. To determine whether inducing macroautophagy/autophagy via small-molecule therapy would be effective for neuropathic LSDs due to enzyme deficiency, we treated a mouse model of mucopolysaccharidosis IIIB (MPS IIIB), a storage disorder caused by deficiency of the enzyme NAGLU (alpha-N-acetylglucosaminidase [Sanfilippo disease IIIB]), with the autophagy-inducing compound trehalose. Treated naglu–/ – mice lived longer, displayed less hyperactivity and anxiety, retained their vision (and retinal photoreceptors), and showed reduced inflammation in the brain and retina. Treated mice also showed improved clearance of autophagic vacuoles in neuronal and glial cells, accompanied by activation of the TFEB transcriptional network that controls lysosomal biogenesis and autophagic flux. Therefore, small-molecule-induced autophagy enhancement can improve the neurological symptoms associated with a lysosomal enzyme deficiency and could provide a viable therapeutic approach to neuropathic LSDs. Abbreviations: ANOVA: analysis of variance; Atg7: autophagy related 7; AV: autophagic vacuoles; CD68: cd68 antigen; ERG: electroretinogram; ERT: enzyme replacement therapy; GAPDH: glyceraldehyde-3-phosphate dehydrogenase; GFAP: glial fibrillary acidic protein; GNAT2: guanine nucleotide binding protein, alpha transducing 2; HSCT: hematopoietic stem cell transplantation; INL: inner nuclear layer; LC3: microtubule-associated protein 1 light chain 3 alpha; MPS: mucopolysaccharidoses; NAGLU: alpha-N-acetylglucosaminidase (Sanfilippo disease IIIB); ONL: outer nuclear layer; PBS: phosphate-buffered saline; PRKCA/PKCα: protein kinase C, alpha; S1BF: somatosensory cortex; SQSTM1: sequestosome 1; TEM: transmission electron microscopy; TFEB: transcription factor EB; VMP/VPL: ventral posterior nuclei of the thalamus
Collapse
Affiliation(s)
- Parisa Lotfi
- a Department of Molecular and Human Genetics , Baylor College of Medicine, Jan and Dan Duncan Neurological Research Institute at Texas Children's Hospital , Houston , TX , USA
| | - Dennis Y Tse
- b Department of Ophthalmology , Cullen Eye Institute, Baylor College of Medicine , Houston , TX , USA.,c School of Optometry , The Hong Kong Polytechnic University , Kowloon , Hong Kong
| | - Alberto Di Ronza
- a Department of Molecular and Human Genetics , Baylor College of Medicine, Jan and Dan Duncan Neurological Research Institute at Texas Children's Hospital , Houston , TX , USA
| | - Michelle L Seymour
- d Huffington Center on Aging, Department of Molecular and Cellular Biology , Baylor College of Medicine , Houston , TX , USA.,e Department of Otolaryngology-Head & Neck Surgery , Baylor College of Medicine , Houston , TX , USA
| | | | - Jonathan D Cooper
- g Department of Basic and Clinical Neuroscience , Maurice Wohl Clinical Neuroscience Institute, Institute of Psychiatry, Psychology and Neuroscience , Kings College London , London , UK.,h Department of Pediatrics, Los Angeles Biomedical Research Institute at Harbor-UCLA Medical Center , David Geffen School of Medicine, UCLA , Torrance , CA , USA
| | - Fred A Pereira
- d Huffington Center on Aging, Department of Molecular and Cellular Biology , Baylor College of Medicine , Houston , TX , USA.,e Department of Otolaryngology-Head & Neck Surgery , Baylor College of Medicine , Houston , TX , USA
| | | | - Samuel M Wu
- b Department of Ophthalmology , Cullen Eye Institute, Baylor College of Medicine , Houston , TX , USA
| | - Marco Sardiello
- a Department of Molecular and Human Genetics , Baylor College of Medicine, Jan and Dan Duncan Neurological Research Institute at Texas Children's Hospital , Houston , TX , USA
| |
Collapse
|
38
|
Neuronal-specific impairment of heparan sulfate degradation in Drosophila reveals pathogenic mechanisms for Mucopolysaccharidosis type IIIA. Exp Neurol 2018; 303:38-47. [PMID: 29408731 DOI: 10.1016/j.expneurol.2018.01.020] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2016] [Revised: 12/13/2017] [Accepted: 01/31/2018] [Indexed: 01/17/2023]
Abstract
Mucopolysaccharidosis type IIIA (MPS IIIA) is a lysosomal storage disorder resulting from the deficit of the N-sulfoglucosamine sulfohydrolase (SGSH) enzyme that leads to accumulation of partially-degraded heparan sulfate. MPS IIIA is characterized by severe neurological symptoms, clinically presenting as Sanfilippo syndrome, for which no effective therapy is available. The lysosomal SGSH enzyme is conserved in Drosophila and we have identified increased levels of heparan sulfate in flies with ubiquitous knockdown of SGSH/CG14291. Using neuronal specific knockdown of SGSH/CG14291 we have also observed a higher abundance of Lysotracker-positive puncta as well as increased expression of GFP tagged Ref(2)P supporting disruption to lysosomal function. We have also observed a progressive defect in climbing ability, a hallmark of neurological dysfunction. Genetic screens indicate proteins and pathways that can functionally modify the climbing phenotype, including autophagy-related proteins (Atg1 and Atg18), superoxide dismutase enzymes (Sod1 and Sod2) and heat shock protein (HSPA1). In addition, reducing heparan sulfate biosynthesis by knocking down sulfateless or slalom expression significantly worsens the phenotype; an important observation given that substrate inhibition is being evaluated clinically as a treatment for MPS IIIA. Identifying the cellular pathways that can modify MPS IIIA neuropathology is an essential step in the development of novel therapeutic approaches to prevent and/or ameliorate symptoms in children with Sanfilippo syndrome.
Collapse
|
39
|
Kuiper GA, Meijer OLM, Langereis EJ, Wijburg FA. Failure to shorten the diagnostic delay in two ultra-orphan diseases (mucopolysaccharidosis types I and III): potential causes and implications. Orphanet J Rare Dis 2018; 13:2. [PMID: 29310675 PMCID: PMC5759238 DOI: 10.1186/s13023-017-0733-y] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2017] [Accepted: 12/04/2017] [Indexed: 12/04/2022] Open
Abstract
Background Rare diseases are often un- or misdiagnosed for extended periods, resulting in a long diagnostic delay that may significantly add to the burden of the disease. An early diagnosis is particularly essential if a disease-modifying treatment is available. The purpose of this study was to assess the extent of the diagnostic delay in the two ultra-rare diseases, i.e., mucopolysaccharidosis I (MPS I) and III (MPS III), both of which are lysosomal storage disorders with different phenotypic severities (MPS 1 is characterized by the severe Hurler and the more attenuated non-Hurler phenotypes, MPS III is characterized by the severe rapidly progressing (RP) phenotype and more attenuated slowly progressing (SP) phenotype). We investigated whether the diagnostic delay changed over the previous decades. Results The diagnostic delay, which is defined as the time between the first visit to a medical doctor for disease-related symptoms and the final diagnosis, was assessed using telephone interviews with patients diagnosed between 1988 and 2017 and/or their parents or legal guardian(s). In addition, the medical charts were reviewed. For MPS I (n = 29), the median diagnostic delay was 8 months (range 1-24 months) for Hurler patients and 28 months (range 2-147 months) for non-Hurler patients. For MPS III (n = 46), the median diagnostic delay was 33 months (range 1-365 months). No difference was observed between the RP and SP phenotypic groups. Comparing the diagnostic delay over time using 5-year time intervals, no reduction in the diagnostic delay was observed for MPS I or MPS III. Conclusions In the Netherlands, the time to diagnosis for patients with MPS I and MPS III has not changed between 1988 and 2017, and an extensive delay still exists between the first visit to a medical doctor for disease-related symptoms and the final diagnosis. The numerous campaigns launched to increase awareness, leading to earlier diagnosis of these rare disorders, particularly of MPS I, have failed to achieve their goal. Robust selected screening protocols embedded in national guidelines and newborn screening for disorders that meet the criteria for population screening may be the only effective approaches for reducing the diagnostic delay.
Collapse
Affiliation(s)
- Gé-Ann Kuiper
- Department of Pediatric Metabolic Diseases, Emma Children's Hospital and Amsterdam Lysosome Center "Sphinx", Academic Medical Center, Meibergdreef 9, 1105, AZ, Amsterdam, The Netherlands
| | - Olga L M Meijer
- Department of Pediatric Metabolic Diseases, Emma Children's Hospital and Amsterdam Lysosome Center "Sphinx", Academic Medical Center, Meibergdreef 9, 1105, AZ, Amsterdam, The Netherlands
| | - Eveline J Langereis
- Department of Pediatric Metabolic Diseases, Emma Children's Hospital and Amsterdam Lysosome Center "Sphinx", Academic Medical Center, Meibergdreef 9, 1105, AZ, Amsterdam, The Netherlands
| | - Frits A Wijburg
- Department of Pediatric Metabolic Diseases, Emma Children's Hospital and Amsterdam Lysosome Center "Sphinx", Academic Medical Center, Meibergdreef 9, 1105, AZ, Amsterdam, The Netherlands.
| |
Collapse
|
40
|
Nijmeijer SCM, Wijburg FA. Mucopolysaccharidosis type III: current clinical trials, challenges and recommendations. Expert Opin Orphan Drugs 2017. [DOI: 10.1080/21678707.2018.1411797] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Affiliation(s)
- Stephanie CM Nijmeijer
- Department of Pediatric Metabolic Diseases, Emma Children’s Hospital and Amsterdam Lysosome Center ‘Sphinx,’ Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | - Frits A Wijburg
- Department of Pediatric Metabolic Diseases, Emma Children’s Hospital and Amsterdam Lysosome Center ‘Sphinx,’ Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| |
Collapse
|
41
|
Shapiro EG, Escolar ML, Delaney KA, Mitchell JJ. Assessments of neurocognitive and behavioral function in the mucopolysaccharidoses. Mol Genet Metab 2017; 122S:8-16. [PMID: 29128371 DOI: 10.1016/j.ymgme.2017.09.007] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/26/2017] [Revised: 09/12/2017] [Accepted: 09/13/2017] [Indexed: 12/25/2022]
Abstract
The mucopolysaccharidoses (MPS) are a group of rare, inherited lysosomal storage disorders in which accumulation of glycosaminoglycans (GAGs) leads to progressive tissue and organ dysfunction. In addition to a variety of somatic signs and symptoms, patients with rapidly progressing MPS I (Hurler), II, III, and VII can present with significant neurological manifestations, including impaired cognitive abilities, difficulties in language and speech, behavioral abnormalities, sleep problems, and/or seizures. Neurological symptoms have a substantial impact on the quality of life of MPS patients and their families. Due to the progressive nature of cognitive impairment in these MPS patients, neurocognitive function is a sensitive indicator of disease progression, and a relevant outcome when testing efficacy of therapies for these disorders. In order to effectively manage and develop therapies that address neurological manifestations of MPS, it is important to use appropriate neurocognitive assessment tools that are sensitive to changes in neurocognitive function in MPS patients. This review discusses expert opinions on key issues and considerations for effective neurocognitive testing in MPS patients. In addition, it describes the neurocognitive assessment tools that have been used in clinical practice for these patients. The content of this review is based on existing literature and information from a meeting of international experts with extensive experience in managing and treating MPS disorders.
Collapse
Affiliation(s)
- Elsa G Shapiro
- Shapiro Neuropsychology Consultants, LLC, Portland, OR, USA; Department of Pediatrics and Neurology, University of Minnesota, Minneapolis, MN, USA.
| | - Maria L Escolar
- Department of Pediatric Neurodevelopment, Children's Hospital of Pittsburgh, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | | | - John J Mitchell
- Departments of Endocrinology and Metabolism & Medical Genetics, Montreal Children's Hospital, McGill University Health Centre, Montreal, QC, Canada
| |
Collapse
|
42
|
Piguet F, Alves S, Cartier N. Clinical Gene Therapy for Neurodegenerative Diseases: Past, Present, and Future. Hum Gene Ther 2017; 28:988-1003. [DOI: 10.1089/hum.2017.160] [Citation(s) in RCA: 71] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Affiliation(s)
- Françoise Piguet
- Translational Medicine and Neurogenetics Department, Institut de Genetique et de Biologie Moleculaire et Cellulaire, Strasbourg, France
- Inserm U596, Illkirch, France; CNRS, UMR7104, Illkirch, France
- Faculte des Sciences de la Vie, Universite de Strasbourg, Strasbourg, France
| | | | - Nathalie Cartier
- INSERM/CEA UMR1169, MIRCen Fontenay aux Roses, France
- Universite Paris-Sud, Orsay, France
| |
Collapse
|
43
|
Knottnerus SJG, Nijmeijer SCM, IJlst L, Te Brinke H, van Vlies N, Wijburg FA. Prediction of phenotypic severity in mucopolysaccharidosis type IIIA. Ann Neurol 2017; 82:686-696. [PMID: 29023963 PMCID: PMC5725696 DOI: 10.1002/ana.25069] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2017] [Revised: 08/11/2017] [Accepted: 10/02/2017] [Indexed: 01/03/2023]
Abstract
Objective Mucopolysaccharidosis IIIA or Sanfilippo disease type A is a progressive neurodegenerative disorder presenting in early childhood, caused by an inherited deficiency of the lysosomal hydrolase sulfamidase. New missense mutations, for which genotype–phenotype correlations are currently unknown, are frequently reported, hampering early prediction of phenotypic severity and efficacy assessment of new disease‐modifying treatments. We aimed to design a method to determine phenotypic severity early in the disease course. Methods Fifty‐three patients were included for whom skin fibroblasts and data on disease course and mutation analysis were available. Patients were phenotypically characterized on clinical data as rapidly progressing or slowly progressing. Sulfamidase activity was measured in fibroblasts cultured at 37 °C and at 30 °C. Results Sulfamidase activity in fibroblasts from patients homozygous or compound heterozygous for a combination of known severe mutations remained below the limit of quantification under both culture conditions. In contrast, sulfamidase activity in fibroblasts from patients homozygous or compound heterozygous for a known mild mutation increased above the limit of quantification when cultured at 30 °C. With division on the basis of the patients' phenotype, fibroblasts from slowly progressing patients could be separated from rapidly progressing patients by increase in sulfamidase activity when cultured at 30 °C (p < 0.001, sensitivity = 96%, specificity = 93%). Interpretation Phenotypic severity strongly correlates with the potential to increase sulfamidase activity in fibroblasts cultured at 30 °C, allowing reliable distinction between patients with rapidly progressing or slowly progressing phenotypes. This method may provide an essential tool for assessment of treatment effects and for health care and life planning decisions. Ann Neurol 2017;82:686–696
Collapse
Affiliation(s)
- Suzan J G Knottnerus
- Department of Pediatric Metabolic Diseases, Emma Children's Hospital and Amsterdam Lysosome Center "Sphinx," Academic Medical Center, University of Amsterdam.,Laboratory Genetic Metabolic Diseases, Academic Medical Center, University of Amsterdam, Amsterdam, the Netherlands
| | - Stephanie C M Nijmeijer
- Department of Pediatric Metabolic Diseases, Emma Children's Hospital and Amsterdam Lysosome Center "Sphinx," Academic Medical Center, University of Amsterdam
| | - Lodewijk IJlst
- Laboratory Genetic Metabolic Diseases, Academic Medical Center, University of Amsterdam, Amsterdam, the Netherlands
| | - Heleen Te Brinke
- Laboratory Genetic Metabolic Diseases, Academic Medical Center, University of Amsterdam, Amsterdam, the Netherlands
| | - Naomi van Vlies
- Laboratory Genetic Metabolic Diseases, Academic Medical Center, University of Amsterdam, Amsterdam, the Netherlands
| | - Frits A Wijburg
- Department of Pediatric Metabolic Diseases, Emma Children's Hospital and Amsterdam Lysosome Center "Sphinx," Academic Medical Center, University of Amsterdam.,Laboratory Genetic Metabolic Diseases, Academic Medical Center, University of Amsterdam, Amsterdam, the Netherlands
| |
Collapse
|
44
|
Ghosh A, Shapiro E, Rust S, Delaney K, Parker S, Shaywitz AJ, Morte A, Bubb G, Cleary M, Bo T, Lavery C, Bigger BW, Jones SA. Recommendations on clinical trial design for treatment of Mucopolysaccharidosis Type III. Orphanet J Rare Dis 2017. [PMID: 28651568 PMCID: PMC5485703 DOI: 10.1186/s13023-017-0675-4] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/02/2022] Open
Abstract
Background Mucopolysaccharidosis type III is a progressive, neurodegenerative lysosomal storage disorder for which there is currently no effective therapy. Though numerous potential therapies are in development, there are several challenges to conducting clinical research in this area. We seek to make recommendations on the approach to clinical research in MPS III, including the selection of outcome measures and trial endpoints, in order to improve the quality and impact of research in this area. Results An international workshop involving academic researchers, clinical experts and industry groups was held in June 2015, with presentations and discussions on disease pathophysiology, biomarkers, potential therapies and clinical outcome measures. A set of recommendations was subsequently prepared by a working group and reviewed by all delegates. We present a series of 11 recommendations regarding the conduct of clinical research, outcome measures and management of natural history data in Mucopolysaccharidosis type III. Conclusions Improving the quality of clinical research in Mucopolysaccharidosis type III will require an open, collaborative and systematic approach between academic researchers, clinicians and industry. Natural history data should be published as soon as possible and ideally collated in a central repository. There should be agreement on outcome measures and instruments for evaluation of clinical outcomes to maximise the effectiveness of current and future clinical research.
Collapse
Affiliation(s)
- Arunabha Ghosh
- Willink Biochemical Genetics Unit, Manchester Centre For Genomic Medicine, Manchester Academic Health Science Centre, Central Manchester University Hospitals NHS Foundation Trust, Manchester, UK.,School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| | - Elsa Shapiro
- Shapiro & Delaney LLC, Mendota Heights, MN, USA.,Paediatrics and Neurology, University of Minnesota, Minneapolis, MN, USA
| | - Stewart Rust
- Paediatric Psychosocial Service, Royal Manchester Children's Hospital, Central Manchester University Hospitals NHS Foundation Trust, Manchester, UK
| | | | | | | | | | | | | | | | | | - Brian W Bigger
- Stem Cell & Neurotherapies, Division of Cell Matrix Biology and Regenerative Medicine, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK.
| | - Simon A Jones
- Willink Biochemical Genetics Unit, Manchester Centre For Genomic Medicine, Manchester Academic Health Science Centre, Central Manchester University Hospitals NHS Foundation Trust, Manchester, UK
| |
Collapse
|
45
|
Kartal A. Delayed speech, hyperactivity, and coarse facies: Does Sanfilippo syndrome come to mind? J Pediatr Neurosci 2016; 11:282-284. [PMID: 27857809 PMCID: PMC5108143 DOI: 10.4103/1817-1745.193378] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Mucopolysaccharidosis Type IIIA (MPS IIIA) or Sanfilippo-A syndrome is caused by a deficiency in lysosomal a-heparan N-sulfatase. Its clinical manifestations include progressive dementia, hyperactivity, and aggressive behavior. Unlike other mucopolysaccharide disorders, the diagnosis of MPS IIIA is challenging in both adults and children. This diagnostic challenge has been associated with the high incidence of false negative results encountered on urinary screening tests. We herein describe Sanfilippo-A syndrome in a pediatric patient who presented with progressive hyperactivity, delayed language, and developmental delay and a negative urine screening test. We emphasized that these findings may serve as possible initial presentations of MPS IIIA; therefore, screening for MPS should be done in all patients with unexplained psychomotor retardation and progressive hyperactivity.
Collapse
Affiliation(s)
- Ayşe Kartal
- Department of Pediatric Neurology, School of Medicine, Selçuk University, Konya, Turkey
| |
Collapse
|
46
|
Marcó S, Pujol A, Roca C, Motas S, Ribera A, Garcia M, Molas M, Villacampa P, Melia CS, Sánchez V, Sánchez X, Bertolin J, Ruberte J, Haurigot V, Bosch F. Progressive neurologic and somatic disease in a novel mouse model of human mucopolysaccharidosis type IIIC. Dis Model Mech 2016; 9:999-1013. [PMID: 27491071 PMCID: PMC5047683 DOI: 10.1242/dmm.025171] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2016] [Accepted: 07/26/2016] [Indexed: 02/02/2023] Open
Abstract
Mucopolysaccharidosis type IIIC (MPSIIIC) is a severe lysosomal storage disease caused by deficiency in activity of the transmembrane enzyme heparan-α-glucosaminide N-acetyltransferase (HGSNAT) that catalyses the N-acetylation of α-glucosamine residues of heparan sulfate. Enzyme deficiency causes abnormal substrate accumulation in lysosomes, leading to progressive and severe neurodegeneration, somatic pathology and early death. There is no cure for MPSIIIC, and development of new therapies is challenging because of the unfeasibility of cross-correction. In this study, we generated a new mouse model of MPSIIIC by targeted disruption of the Hgsnat gene. Successful targeting left LacZ expression under control of the Hgsnat promoter, allowing investigation into sites of endogenous expression, which was particularly prominent in the CNS, but was also detectable in peripheral organs. Signs of CNS storage pathology, including glycosaminoglycan accumulation, lysosomal distension, lysosomal dysfunction and neuroinflammation were detected in 2-month-old animals and progressed with age. Glycosaminoglycan accumulation and ultrastructural changes were also observed in most somatic organs, but lysosomal pathology seemed most severe in liver. Furthermore, HGSNAT-deficient mice had altered locomotor and exploratory activity and shortened lifespan. Hence, this animal model recapitulates human MPSIIIC and provides a useful tool for the study of disease physiopathology and the development of new therapeutic approaches.
Collapse
Affiliation(s)
- Sara Marcó
- Center of Animal Biotechnology and Gene Therapy, Universitat Autònoma de Barcelona, Bellaterra 08193, Spain Department of Biochemistry and Molecular Biology, Universitat Autònoma de Barcelona, Bellaterra 08193, Spain
| | - Anna Pujol
- Center of Animal Biotechnology and Gene Therapy, Universitat Autònoma de Barcelona, Bellaterra 08193, Spain Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas, Barcelona 08036, Spain
| | - Carles Roca
- Center of Animal Biotechnology and Gene Therapy, Universitat Autònoma de Barcelona, Bellaterra 08193, Spain Department of Biochemistry and Molecular Biology, Universitat Autònoma de Barcelona, Bellaterra 08193, Spain Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas, Barcelona 08036, Spain
| | - Sandra Motas
- Center of Animal Biotechnology and Gene Therapy, Universitat Autònoma de Barcelona, Bellaterra 08193, Spain Department of Biochemistry and Molecular Biology, Universitat Autònoma de Barcelona, Bellaterra 08193, Spain
| | - Albert Ribera
- Center of Animal Biotechnology and Gene Therapy, Universitat Autònoma de Barcelona, Bellaterra 08193, Spain Department of Biochemistry and Molecular Biology, Universitat Autònoma de Barcelona, Bellaterra 08193, Spain Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas, Barcelona 08036, Spain
| | - Miguel Garcia
- Center of Animal Biotechnology and Gene Therapy, Universitat Autònoma de Barcelona, Bellaterra 08193, Spain Department of Biochemistry and Molecular Biology, Universitat Autònoma de Barcelona, Bellaterra 08193, Spain Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas, Barcelona 08036, Spain
| | - Maria Molas
- Center of Animal Biotechnology and Gene Therapy, Universitat Autònoma de Barcelona, Bellaterra 08193, Spain Department of Biochemistry and Molecular Biology, Universitat Autònoma de Barcelona, Bellaterra 08193, Spain Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas, Barcelona 08036, Spain
| | - Pilar Villacampa
- Center of Animal Biotechnology and Gene Therapy, Universitat Autònoma de Barcelona, Bellaterra 08193, Spain Department of Biochemistry and Molecular Biology, Universitat Autònoma de Barcelona, Bellaterra 08193, Spain Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas, Barcelona 08036, Spain
| | - Cristian S Melia
- Center of Animal Biotechnology and Gene Therapy, Universitat Autònoma de Barcelona, Bellaterra 08193, Spain Department of Biochemistry and Molecular Biology, Universitat Autònoma de Barcelona, Bellaterra 08193, Spain
| | - Víctor Sánchez
- Center of Animal Biotechnology and Gene Therapy, Universitat Autònoma de Barcelona, Bellaterra 08193, Spain Department of Biochemistry and Molecular Biology, Universitat Autònoma de Barcelona, Bellaterra 08193, Spain
| | - Xavier Sánchez
- Center of Animal Biotechnology and Gene Therapy, Universitat Autònoma de Barcelona, Bellaterra 08193, Spain Department of Biochemistry and Molecular Biology, Universitat Autònoma de Barcelona, Bellaterra 08193, Spain
| | - Joan Bertolin
- Center of Animal Biotechnology and Gene Therapy, Universitat Autònoma de Barcelona, Bellaterra 08193, Spain Department of Biochemistry and Molecular Biology, Universitat Autònoma de Barcelona, Bellaterra 08193, Spain
| | - Jesús Ruberte
- Center of Animal Biotechnology and Gene Therapy, Universitat Autònoma de Barcelona, Bellaterra 08193, Spain Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas, Barcelona 08036, Spain Department of Animal Health and Anatomy, School of Veterinary Medicine, Universitat Autònoma de Barcelona, Bellaterra 08193, Spain
| | - Virginia Haurigot
- Center of Animal Biotechnology and Gene Therapy, Universitat Autònoma de Barcelona, Bellaterra 08193, Spain Department of Biochemistry and Molecular Biology, Universitat Autònoma de Barcelona, Bellaterra 08193, Spain Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas, Barcelona 08036, Spain
| | - Fatima Bosch
- Center of Animal Biotechnology and Gene Therapy, Universitat Autònoma de Barcelona, Bellaterra 08193, Spain Department of Biochemistry and Molecular Biology, Universitat Autònoma de Barcelona, Bellaterra 08193, Spain Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas, Barcelona 08036, Spain
| |
Collapse
|
47
|
Shapiro EG, Nestrasil I, Delaney KA, Rudser K, Kovac V, Nair N, Richard CW, Haslett P, Whitley CB. A Prospective Natural History Study of Mucopolysaccharidosis Type IIIA. J Pediatr 2016; 170:278-87.e1-4. [PMID: 26787381 PMCID: PMC4769976 DOI: 10.1016/j.jpeds.2015.11.079] [Citation(s) in RCA: 78] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/14/2015] [Revised: 10/09/2015] [Accepted: 11/30/2015] [Indexed: 11/17/2022]
Abstract
OBJECTIVES To characterize the clinical course of mucopolysaccharidosis type IIIA (MPS IIIA), and identify potential endpoints for future treatment trials. STUDY DESIGN Children with a confirmed diagnosis of MPS IIIA, functioning above a developmental age of 1 year, were followed for up to 2 years. Cognitive status and brain atrophy were assessed by standardized tests and volumetric magnetic resonance imaging, respectively. Liver and spleen volumes and cerebrospinal fluid and urine biomarker levels were measured. RESULTS Twenty-five children, from 1.1 to 18.4 years old, were enrolled, and 24 followed for at least 12 months. 19 exhibited a rapidly progressing (RP) form of MPS IIIA, and 5, a more slowly progressing form. Children with RP plateaued in development by 30 months, followed by rapid regression after 40-50 months. In patients with RP, cognitive developmental quotients showed consistent steep declines associated with progressive cortical gray matter atrophy. Children with slowly progressing had a similar but more prolonged course. Liver and spleen volumes were approximately double normal size, and cerebrospinal fluid and urine heparin sulfate levels were elevated and relatively constant over time. CONCLUSION Developmental quotient and cortical gray matter volume are sensitive markers of disease progression in MPS IIIA, and may have utility as clinical endpoints in treatment trials. For optimal outcomes, treatment may need to be instituted in children before the onset of steep cognitive decline and brain atrophy. TRIAL REGISTRATION ClinicalTrials.gov: NCT01047306.
Collapse
Affiliation(s)
- Elsa G Shapiro
- Department of Pediatrics, University of Minnesota, Minneapolis, MN.
| | - Igor Nestrasil
- Department of Pediatrics, University of Minnesota, Minneapolis, MN
| | | | - Kyle Rudser
- Division of Biostatistics, University of Minnesota, Minneapolis, MN
| | - Victor Kovac
- Department of Pediatrics, University of Minnesota, Minneapolis, MN
| | | | | | | | | |
Collapse
|
48
|
A novel LC-MS/MS assay for heparan sulfate screening in the cerebrospinal fluid of mucopolysaccharidosis IIIA patients. Bioanalysis 2016; 8:285-95. [PMID: 26847798 DOI: 10.4155/bio.15.243] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
AIMS Heparan sulfate (HS) accumulates in the central nervous system in mucopolysaccharidosis III type A (MPS IIIA). A validated LC-MS/MS assay was developed to measure HS in human cerebrospinal fluid (CSF). METHODS & RESULTS HS was extracted and digested and the resultant disaccharides were derivatized with a novel label, 4-butylaniline, enabling isoform separation and isotope-tagged analog introduction as an internal standard for LC-MS/MS. The assay has a LLOQ for disaccharides of 0.1 μM, ±20% accuracy and ≤20% precision. CSF samples from patients with MPS IIIA showed elevated HS levels (mean 4.9 μM) compared with negative controls (0.37 μM). CONCLUSION This assay detected elevated HS levels in the CSF of patients with MPS IIIA and provides a method to assess experimental therapies.
Collapse
|
49
|
Tse DY, Lotfi P, Simons DL, Sardiello M, Wu SM. Electrophysiological and Histological Characterization of Rod-Cone Retinal Degeneration and Microglia Activation in a Mouse Model of Mucopolysaccharidosis Type IIIB. Sci Rep 2015; 5:17143. [PMID: 26607664 PMCID: PMC4660851 DOI: 10.1038/srep17143] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2015] [Accepted: 10/26/2015] [Indexed: 02/07/2023] Open
Abstract
Sanfilippo syndrome Type B or Mucopolysaccharidosis IIIB (MPS IIIB) is a neurodegenerative autosomal recessive lysosomal storage disorder in which patients suffer severe vision loss from associated retinopathy. Here we sought to study the underlying retinal functional and morphological changes associated with MPS IIIB disease progression using the established model of MPS IIIB, the B6.129S6-Naglu(tm1Efn)/J mouse line. Electroretinogram (ERG) was recorded from MPS IIIB and wild-type (WT) mice at the age of 28 and 46 weeks, and retinal tissues were subsequently collected for immunohistochemistry analysis. At the 28th week, rod a- and b-wave amplitudes were significantly diminished in MPS IIIB compared to WT mice. The cone a- and b-waves of MPS IIIB mice were not significantly different from those of the control at the 28th week but were significantly diminished at the 46 th week, when MPS IIIB mice showed a major loss of rods and rod bipolar cells in both central and peripheral regions and a minor loss of cones in the periphery. Activation of microglia and neovascularization were also detected in the MPS IIIB retina. The new findings that cones and rod bipolar cells also undergo degeneration, and that retinal microglia are activated, will inform future development of therapeutic strategies.
Collapse
Affiliation(s)
- Dennis Y Tse
- Department of Ophthalmology, Cullen Eye Institute, Baylor College of Medicine, Houston TX, USA.,School of Optometry, The Hong Kong Polytechnic University, Hong Kong
| | - Parisa Lotfi
- Department of Human and Molecular Genetics, Baylor College of Medicine, Jan and Dun Duncan Neurological Research Institute, Texas Children's Hospital, Houston TX, USA
| | - David L Simons
- Department of Ophthalmology, Cullen Eye Institute, Baylor College of Medicine, Houston TX, USA
| | - Marco Sardiello
- Department of Human and Molecular Genetics, Baylor College of Medicine, Jan and Dun Duncan Neurological Research Institute, Texas Children's Hospital, Houston TX, USA
| | - Samuel M Wu
- Department of Ophthalmology, Cullen Eye Institute, Baylor College of Medicine, Houston TX, USA
| |
Collapse
|
50
|
Ethische Aspekte der Ernährungstherapie bei Kindern mit Demenzerkrankungen. Monatsschr Kinderheilkd 2015. [DOI: 10.1007/s00112-015-3432-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|