1
|
Zanetti A, D'Avanzo F, Tomanin R. Molecular basis of mucopolysaccharidosis type II (Hunter syndrome): first review and classification of published IDS gene variants. Hum Genomics 2024; 18:134. [PMID: 39617925 PMCID: PMC11610362 DOI: 10.1186/s40246-024-00701-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2024] [Accepted: 11/21/2024] [Indexed: 12/13/2024] Open
Abstract
PURPOSE Mucopolysaccharidosis type II (MPS II) is a rare X-linked lysosomal storage disorder caused by genetic alterations in the iduronate 2-sulfatase (IDS) gene. A wide range of variants has been reported for different countries and ethnic groups. We collected, analyzed and uniformly summarized all published IDS gene variants reported in literature up to June 2023, here providing the first worldwide review and classification. METHODS Data was obtained from a literature search, conducted in PubMed and Google. All data was analyzed to define the most common alleles, geographic distribution and genotype-phenotype correlation. Moreover, point variants were classified according to their pathogenicity, based on the ACMG guidelines. RESULTS Several types of variants have been described in the IDS gene, including intrachromosomal homologous recombination occurring between the homologous regions of IDS gene and its pseudogene IDSP1. Overall, we collected 2852 individuals from 2798 families, including 24 female patients. Most families carried missense variants, followed by large deletions-insertions and complex rearrangements, small frameshift deletions/insertions and nonsense variants. Based on ACMG guidelines, 62.9% of the 779 point variants were classified as "pathogenic", 35.4% as "likely pathogenic", and the remaining 13 variants as having "uncertain significance". CONCLUSION Data from this study confirmed that MPS II is a genetically very heterogeneous disorder, making genotype-phenotype correlation very challenging and in most cases merely unfeasible. Mutation updates are essential for the correct molecular diagnosis, genetic counseling, prenatal and preimplantation diagnosis, and disease management.
Collapse
Affiliation(s)
- Alessandra Zanetti
- Laboratory of Diagnosis and Therapy of Lysosomal Disorders, Department of Women's and Children's Health, University of Padova, Padova, Italy
- Fondazione Istituto di Ricerca Pediatrica Città della Speranza, Padova, Italy
| | - Francesca D'Avanzo
- Laboratory of Diagnosis and Therapy of Lysosomal Disorders, Department of Women's and Children's Health, University of Padova, Padova, Italy
- Fondazione Istituto di Ricerca Pediatrica Città della Speranza, Padova, Italy
| | - Rosella Tomanin
- Laboratory of Diagnosis and Therapy of Lysosomal Disorders, Department of Women's and Children's Health, University of Padova, Padova, Italy.
- Fondazione Istituto di Ricerca Pediatrica Città della Speranza, Padova, Italy.
| |
Collapse
|
2
|
Ayodele O, Fertek D, Evuarherhe O, Siffel C, Audi J, Yee KS, Burton BK. A Systematic Literature Review on the Global Status of Newborn Screening for Mucopolysaccharidosis II. Int J Neonatal Screen 2024; 10:71. [PMID: 39449359 PMCID: PMC11503380 DOI: 10.3390/ijns10040071] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/19/2024] [Revised: 09/16/2024] [Accepted: 09/19/2024] [Indexed: 10/26/2024] Open
Abstract
A systematic literature review was conducted to determine the global status of newborn screening (NBS) for mucopolysaccharidosis (MPS) II (Hunter syndrome; OMIM 309900). Electronic databases were searched in July 2023 for articles referencing NBS for lysosomal storage diseases: 53 featured MPS II. Until recently, only Taiwan and two US states (Illinois and Missouri) formally screened newborns for MPS II, although pilot programs have been conducted elsewhere (Japan, New York, and Washington). In 2022, MPS II was added to the US Recommended Uniform Screening Panel, with increased uptake of NBS anticipated across the USA. While the overall MPS II birth prevalence, determined from NBS initiatives, was higher than in previous reports, it was lower in the USA (approximately 1 in 73,000 according to recent studies in Illinois and Missouri) than in Asia (approximately 1 in 15,000 in Japan). NBS programs typically rely on tandem mass spectrometry quantification of iduronate-2-sulfatase activity for first-tier testing. Diagnosis is often confirmed via molecular genetic testing and/or biochemical testing but may be complicated by factors such as pseudodeficiency alleles and variants of unknown significance. Evidence relating to MPS II NBS is lacking outside Taiwan and the USA. Although broad benefits of NBS are recognized, few studies specifically explored the perspectives of families of children with MPS II.
Collapse
Affiliation(s)
- Olulade Ayodele
- Takeda Development Center Americas, Inc., Lexington, MA 02421, USA
| | - Daniel Fertek
- Takeda Pharmaceuticals International AG, 8152 Zurich, Switzerland
| | | | - Csaba Siffel
- Takeda Development Center Americas, Inc., Lexington, MA 02421, USA
- College of Allied Health Sciences, Augusta University, Augusta, GA 30912, USA
| | - Jennifer Audi
- Takeda Pharmaceuticals International AG, 8152 Zurich, Switzerland
| | - Karen S. Yee
- Takeda Development Center Americas, Inc., Cambridge, MA 02142, USA
| | - Barbara K. Burton
- Ann & Robert H. Lurie Children’s Hospital of Chicago, Northwestern University, Chicago, IL 60611, USA
| |
Collapse
|
3
|
Zanetti A, Tomanin R. Targeting Neurological Aspects of Mucopolysaccharidosis Type II: Enzyme Replacement Therapy and Beyond. BioDrugs 2024; 38:639-655. [PMID: 39177874 PMCID: PMC11358193 DOI: 10.1007/s40259-024-00675-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/30/2024] [Indexed: 08/24/2024]
Abstract
Mucopolysaccharidosis type II (MPS II) is a rare, pediatric, neurometabolic disorder due to the lack of activity of the lysosomal hydrolase iduronate 2-sulfatase (IDS), normally degrading heparan sulfate and dermatan sulfate within cell lysosomes. The deficit of activity is caused by mutations affecting the IDS gene, leading to the pathological accumulation of both glycosaminoglycans in the lysosomal compartment and in the extracellular matrix of most body districts. Although a continuum of clinical phenotypes is described, two main forms are commonly recognized-attenuated and severe-the latter being characterized by an earlier and faster clinical progression and by a progressive impairment of central nervous system (CNS) functions. However, attenuated forms have also been recently described as presenting some neurological involvement, although less deep, such as deficits of attention and hearing loss. The main treatment for the disease is represented by enzyme replacement therapy (ERT), applied in several countries since 2006, which, albeit showing partial efficacy on some peripheral organs, exhibited a very poor efficacy on bones and heart, and a total inefficacy on CNS impairment, due to the inability of the recombinant enzyme to cross the blood-brain barrier (BBB). Together with ERT, whose design enhancements, performed in the last few years, allowed a possible brain penetration of the drug through the BBB, other therapeutic approaches aimed at targeting CNS involvement in MPS II were proposed and evaluated in the last decades, such as intrathecal ERT, intracerebroventricular ERT, ex vivo gene therapy, or adeno-associated viral vector (AAV) gene therapy. The aim of this review is to summarize the main clinical aspects of MPS II in addition to current therapeutic options, with particular emphasis on the neurological ones and on the main CNS-targeted therapeutic approaches explored through the years.
Collapse
Affiliation(s)
- Alessandra Zanetti
- Laboratory of Diagnosis and Therapy of Lysosomal Disorders, Department of Women's and Children's Health SDB, University of Padova, Via Giustiniani, 3, 35128, Padua, Italy
- Istituto di Ricerca Pediatrica Città della Speranza, 35127, Padua, Italy
| | - Rosella Tomanin
- Laboratory of Diagnosis and Therapy of Lysosomal Disorders, Department of Women's and Children's Health SDB, University of Padova, Via Giustiniani, 3, 35128, Padua, Italy.
- Istituto di Ricerca Pediatrica Città della Speranza, 35127, Padua, Italy.
| |
Collapse
|
4
|
Darie-Ion L, Petre BA. An update on multiplexed mass spectrometry-based lysosomal storage disease diagnosis. MASS SPECTROMETRY REVIEWS 2024; 43:1135-1149. [PMID: 37584312 DOI: 10.1002/mas.21864] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/04/2023] [Revised: 08/01/2023] [Accepted: 08/04/2023] [Indexed: 08/17/2023]
Abstract
Lysosomal storage disorders (LSDs) are a type of inherited metabolic disorders in which biomolecules, accumulate as a specific substrate in lysosomes due to specific individual enzyme deficiencies. Despite the fact that LSDs are incurable, various approaches, including enzyme replacement therapy, hematopoietic stem cell transplantation, or gene therapy are now available. Therefore, a timely diagnosis is a critical initial step in patient treatment. The-state-of-the-art in LSD diagnostic uses, in the first stage, enzymatic activity determination by fluorimetry or by mass spectrometry (MS) with the aid of dry blood spots, based on different enzymatic substrate structures. Due to its sensitivity, high precision, and ability to screen for an unprecedented number of diseases in a single assay, multiplexed tandem MS-based enzyme activity assays for the screening of LSDs in newborns have recently received a lot of attention. Here, (i) we review the current approaches used for simultaneous enzymatic activity determination of LSDs in dried blood spots using multiplex-LC-MS/MS; (ii) we explore the need for designing novel enzymatic substrates that generate different enzymatic products with distinct molecular masses in multiplexed-MS studies; and (iii) we give examples of the relevance of affinity-MS technique as a basis for reversing undesirable immune-reactivity in enzyme replacement therapy.
Collapse
Affiliation(s)
- Laura Darie-Ion
- Group of Biochemistry, Faculty of Chemistry, Alexandru Ioan Cuza University of Iasi, Iaşi, Romania
| | - Brînduşa Alina Petre
- Group of Biochemistry, Faculty of Chemistry, Alexandru Ioan Cuza University of Iasi, Iaşi, Romania
- Laboratory of Proteomics, Center for Fundamental Research and Experimental Development in Translation Medicine-TRANSCEND, Regional Institute of Oncology, Iaşi, Romania
| |
Collapse
|
5
|
Grooms AJ, Burris BJ, Badu-Tawiah AK. Mass spectrometry for metabolomics analysis: Applications in neonatal and cancer screening. MASS SPECTROMETRY REVIEWS 2024; 43:683-712. [PMID: 36524560 PMCID: PMC10272294 DOI: 10.1002/mas.21826] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/07/2022] [Revised: 11/18/2022] [Accepted: 11/24/2022] [Indexed: 06/17/2023]
Abstract
Chemical analysis by analytical instrumentation has played a major role in disease diagnosis, which is a necessary step for disease treatment. While the treatment process often targets specific organs or compounds, the diagnostic step can occur through various means, including physical or chemical examination. Chemically, the genome may be evaluated to give information about potential genetic outcomes, the transcriptome to provide information about expression actively occurring, the proteome to offer insight on functions causing metabolite expression, or the metabolome to provide a picture of both past and ongoing physiological function in the body. Mass spectrometry (MS) has been elevated among other analytical instrumentation because it can be used to evaluate all four biological machineries of the body. In addition, MS provides enhanced sensitivity, selectivity, versatility, and speed for rapid turnaround time, qualities that are important for instance in clinical procedures involving the diagnosis of a pediatric patient in intensive care or a cancer patient undergoing surgery. In this review, we provide a summary of the use of MS to evaluate biomarkers for newborn screening and cancer diagnosis. As many reviews have recently appeared focusing on MS methods and instrumentation for metabolite analysis, we sought to describe the biological basis for many metabolomic and additional omics biomarkers used in newborn screening and how tandem MS methods have recently been applied, in comparison to traditional methods. Similar comparison is done for cancer screening, with emphasis on emerging MS approaches that allow biological fluids, tissues, and breath to be analyzed for the presence of diagnostic metabolites yielding insight for treatment options based on the understanding of prior and current physiological functions of the body.
Collapse
Affiliation(s)
- Alexander J Grooms
- Department of Chemistry and Biochemistry, The Ohio State University, Ohio, Columbus, USA
| | - Benjamin J Burris
- Department of Chemistry and Biochemistry, The Ohio State University, Ohio, Columbus, USA
| | - Abraham K Badu-Tawiah
- Department of Chemistry and Biochemistry, The Ohio State University, Ohio, Columbus, USA
| |
Collapse
|
6
|
Therrell BL, Padilla CD, Borrajo GJC, Khneisser I, Schielen PCJI, Knight-Madden J, Malherbe HL, Kase M. Current Status of Newborn Bloodspot Screening Worldwide 2024: A Comprehensive Review of Recent Activities (2020-2023). Int J Neonatal Screen 2024; 10:38. [PMID: 38920845 PMCID: PMC11203842 DOI: 10.3390/ijns10020038] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/06/2024] [Revised: 03/28/2024] [Accepted: 03/28/2024] [Indexed: 06/27/2024] Open
Abstract
Newborn bloodspot screening (NBS) began in the early 1960s based on the work of Dr. Robert "Bob" Guthrie in Buffalo, NY, USA. His development of a screening test for phenylketonuria on blood absorbed onto a special filter paper and transported to a remote testing laboratory began it all. Expansion of NBS to large numbers of asymptomatic congenital conditions flourishes in many settings while it has not yet been realized in others. The need for NBS as an efficient and effective public health prevention strategy that contributes to lowered morbidity and mortality wherever it is sustained is well known in the medical field but not necessarily by political policy makers. Acknowledging the value of national NBS reports published in 2007, the authors collaborated to create a worldwide NBS update in 2015. In a continuing attempt to review the progress of NBS globally, and to move towards a more harmonized and equitable screening system, we have updated our 2015 report with information available at the beginning of 2024. Reports on sub-Saharan Africa and the Caribbean, missing in 2015, have been included. Tables popular in the previous report have been updated with an eye towards harmonized comparisons. To emphasize areas needing attention globally, we have used regional tables containing similar listings of conditions screened, numbers of screening laboratories, and time at which specimen collection is recommended. Discussions are limited to bloodspot screening.
Collapse
Affiliation(s)
- Bradford L. Therrell
- Department of Pediatrics, University of Texas Health Science Center San Antonio, San Antonio, TX 78229, USA
- National Newborn Screening and Global Resource Center, Austin, TX 78759, USA
| | - Carmencita D. Padilla
- Department of Pediatrics, College of Medicine, University of the Philippines Manila, Manila 1000, Philippines;
| | - Gustavo J. C. Borrajo
- Detección de Errores Congénitos—Fundación Bioquímica Argentina, La Plata 1908, Argentina;
| | - Issam Khneisser
- Jacques LOISELET Genetic and Genomic Medical Center, Faculty of Medicine, Saint Joseph University, Beirut 1104 2020, Lebanon;
| | - Peter C. J. I. Schielen
- Office of the International Society for Neonatal Screening, Reigerskamp 273, 3607 HP Maarssen, The Netherlands;
| | - Jennifer Knight-Madden
- Caribbean Institute for Health Research—Sickle Cell Unit, The University of the West Indies, Mona, Kingston 7, Jamaica;
| | - Helen L. Malherbe
- Centre for Human Metabolomics, North-West University, Potchefstroom 2531, South Africa;
- Rare Diseases South Africa NPC, The Station Office, Bryanston, Sandton 2021, South Africa
| | - Marika Kase
- Strategic Initiatives Reproductive Health, Revvity, PL10, 10101 Turku, Finland;
| |
Collapse
|
7
|
Li JW, Mao SJ, Chao YQ, Hu CX, Qian YJ, Dai YL, Huang K, Shen Z, Zou CC. Application of tandem mass spectrometry in the screening and diagnosis of mucopolysaccharidoses. Orphanet J Rare Dis 2024; 19:179. [PMID: 38685110 PMCID: PMC11059687 DOI: 10.1186/s13023-024-03195-w] [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: 12/05/2023] [Accepted: 04/19/2024] [Indexed: 05/02/2024] Open
Abstract
Mucopolysaccharidoses (MPSs) are caused by a deficiency in the enzymes needed to degrade glycosaminoglycans (GAGs) in the lysosome. The storage of GAGs leads to the involvement of several systems and even to the death of the patient. In recent years, an increasing number of therapies have increased the treatment options available to patients. Early treatment is beneficial in improving the prognosis, but children with MPSs are often delayed in their diagnosis. Therefore, there is an urgent need to develop a method for early screening and diagnosis of the disease. Tandem mass spectrometry (MS/MS) is an analytical method that can detect multiple substrates or enzymes simultaneously. GAGs are reliable markers of MPSs. MS/MS can be used to screen children at an early stage of the disease, to improve prognosis by treating them before symptoms appear, to evaluate the effectiveness of treatment, and for metabolomic analysis or to find suitable biomarkers. In the future, MS/MS could be used to further identify suitable biomarkers for MPSs for early diagnosis and to detect efficacy.
Collapse
Affiliation(s)
- Jing-Wen Li
- Department of Endocrinology, the Children's Hospital of Zhejiang University School of Medicine, Hangzhou, 310052, China
| | - Shao-Jia Mao
- Department of Endocrinology, the Children's Hospital of Zhejiang University School of Medicine, Hangzhou, 310052, China
| | - Yun-Qi Chao
- Department of Endocrinology, the Children's Hospital of Zhejiang University School of Medicine, Hangzhou, 310052, China
| | - Chen-Xi Hu
- Department of Endocrinology, the Children's Hospital of Zhejiang University School of Medicine, Hangzhou, 310052, China
| | - Yan-Jie Qian
- Department of Endocrinology, the Children's Hospital of Zhejiang University School of Medicine, Hangzhou, 310052, China
| | - Yang-Li Dai
- Department of Endocrinology, the Children's Hospital of Zhejiang University School of Medicine, Hangzhou, 310052, China
| | - Ke Huang
- Department of Endocrinology, the Children's Hospital of Zhejiang University School of Medicine, Hangzhou, 310052, China
| | - Zheng Shen
- Lab Center, the Children's Hospital of Zhejiang University School of Medicine, Hangzhou, 310052, China
| | - Chao-Chun Zou
- Department of Endocrinology, the Children's Hospital of Zhejiang University School of Medicine, Hangzhou, 310052, China.
| |
Collapse
|
8
|
Seo JH, Kosuga M, Hamazaki T, Shintaku H, Okuyama T. Intracerebroventricular enzyme replacement therapy in patients with neuronopathic mucopolysaccharidosis type II: Final report of 5-year results from a Japanese open-label phase 1/2 study. Mol Genet Metab 2023; 140:107709. [PMID: 37922836 DOI: 10.1016/j.ymgme.2023.107709] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/01/2023] [Revised: 10/16/2023] [Accepted: 10/16/2023] [Indexed: 11/07/2023]
Abstract
Intravenous idursulfase is standard treatment for mucopolysaccharidosis II (MPS II) in Japan. In the interim analysis of this open-label, phase 1/2 study (Center for Clinical Trials, Japan Medical Association: JMA-IIA00350), intracerebroventricular (ICV) idursulfase beta was well tolerated, suppressed cerebrospinal fluid (CSF) heparan sulfate (HS) levels, and stabilized developmental decline over 100 weeks in Japanese children with MPS II. Here, we report the final study results, representing 5 years of ICV idursulfase beta treatment. Six male patients with MPS II and developmental delay were enrolled starting in June 2016 and followed until March 2021. Patients received up to 30 mg ICV idursulfase beta every 4 weeks. Outcomes included CSF HS levels, developmental age (DA) (assessed by the Kyoto Scale of Psychological Development), and safety (adverse events). Monitoring by laboratory biochemistry tests, urinary uronic tests, immunogenicity tests, and head computed tomography or magnetic resonance imaging were also conducted regularly. Following ICV idursulfase beta administration, mean CSF HS concentrations decreased from 7.75 μg/mL at baseline to 2.15 μg/mL at final injection (72.3% reduction). Mean DA increased from 23.2 months at screening to 36.0 months at final observation. In five patients with null mutations, mean DA at the final observation was higher than or did not regress compared with that of historical controls receiving intravenous idursulfase only, and the change in DA was greater in patients who started administration aged ≤3 years than in those aged >3 years (+28.7 vs -6.5 months). The difference in DA change versus historical controls in individual patients was +39.5, +40.8, +17.8, +10.5, +7.6 and - 4.5 (mean + 18.6). Common ICV idursulfase beta-related adverse events were vomiting, pyrexia, gastroenteritis, and upper respiratory tract infection (most mild/moderate). These results suggest that long-term ICV idursulfase beta treatment improved neurological symptoms in Japanese children with neuronopathic MPS II.
Collapse
Affiliation(s)
- Joo-Hyun Seo
- Department of Clinical Genomics, Saitama Medical University, 1397-1 Yamane, Hidaka, Saitama 350-1241, Japan
| | - Motomichi Kosuga
- Division of Medical Genetics, National Center for Child Health and Development, 2-10-1 Okura, Setagaya-ku, Tokyo 157-8535, Japan
| | - Takashi Hamazaki
- Department of Pediatrics, Osaka Metropolitan University Hospital, 1-4-3 Asahi-machi, Abeno-ku, Osaka 545-8585, Japan
| | - Haruo Shintaku
- Department of Pediatrics, Osaka Metropolitan University Hospital, 1-4-3 Asahi-machi, Abeno-ku, Osaka 545-8585, Japan
| | - Torayuki Okuyama
- Department of Clinical Genomics, Saitama Medical University, 1397-1 Yamane, Hidaka, Saitama 350-1241, Japan; Division of Medical Genetics, National Center for Child Health and Development, 2-10-1 Okura, Setagaya-ku, Tokyo 157-8535, Japan.
| |
Collapse
|
9
|
Yee KS, Alexanderian D, Merberg D, Natarajan M, Wang S, Wu Y, Whiteman DAH. Cognitive and adaptive behaviors associated with disease severity and genotype in patients with mucopolysaccharidosis II. Mol Genet Metab 2023; 140:107652. [PMID: 37506513 DOI: 10.1016/j.ymgme.2023.107652] [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: 04/21/2023] [Revised: 07/07/2023] [Accepted: 07/08/2023] [Indexed: 07/30/2023]
Abstract
BACKGROUND Mucopolysaccharidosis II (MPS II) is a rare, X-linked lysosomal storage disease caused by pathogenic variants of the iduronate-2-sulfatase gene (IDS) and is characterized by a highly variable disease spectrum. MPS II severity is difficult to predict based on IDS variants alone; while some genotypes are associated with specific phenotypes, the disease course of most genotypes remains unknown. This study aims to refine the genotype-phenotype categorization by combining information from the scientific literature with data from two clinical studies in MPS II. METHODS Genotype, cognitive, and behavioral data from 88 patients in two clinical studies (NCT01822184, NCT02055118) in MPS II were analyzed post hoc in combination with published information on IDS variants from the biomedical literature through a semi-automated multi-stage review process. The Differential Ability Scales, second edition (DAS-II) and the Vineland Adaptive Behavior Scales™, second edition (VABS-II) were used to measure cognitive function and adaptive behavior. RESULTS The most common category of IDS variant was missense (47/88, 53.4% of total variants). The mean (standard deviation [SD]) baseline DAS-II General Conceptual Ability (GCA) and VABS-II Adaptive Behavior Composite (ABC) scores were 74.0 (16.4) and 82.6 (14.7), respectively. All identified IDS complete deletions/large rearrangements (n = 7) and large deletions (n = 1) were associated with a published 'severe' or 'predicted severe' progressive neuronopathic phenotype, characterized by central nervous system involvement. In categories comprising more than one participant, mean baseline DAS-II GCA scores (SD) were lowest among individuals with complete deletions/large rearrangements 64.0 (9.1, n = 4) and highest among those with splice site variants 83.8 (14.2, n = 4). Mean baseline VABS-II ABC scores (SD) were lowest among patients with unclassifiable variants 79.3 (4.9, n = 3) and highest among those with a splice site variant 87.2 (16.1, n = 5), in variant categories with more than one participant. CONCLUSIONS Most patients in the studies had an MPS II phenotype categorized as 'severe' or 'predicted severe' according to classifications, as reported in the literature. Patients with IDS complete deletion/large rearrangement variants had lower mean DAS-II GCA scores than those with other variants, as well as low VABS-II ABC, confirming an association with the early progressive 'severe' (neuronopathic) disease. These data provide a starting point to improve the classification of MPS II phenotypes and the characterization of the genotype-phenotype relationship.
Collapse
Affiliation(s)
- Karen S Yee
- Takeda Development Center Americas, Inc., Cambridge, MA, USA
| | | | - David Merberg
- Takeda Development Center Americas, Inc., Cambridge, MA, USA
| | | | - Scarlett Wang
- Takeda Development Center Americas, Inc., Cambridge, MA, USA
| | - Yuna Wu
- Takeda Development Center Americas, Inc., Lexington, MA, USA
| | | |
Collapse
|
10
|
Tsai MJM, Hung MZ, Lin YL, Lee NC, Chien YH, Hwu WL. Curated incidence of lysosomal storage diseases from the Taiwan Biobank. NPJ Genom Med 2023; 8:27. [PMID: 37741878 PMCID: PMC10517920 DOI: 10.1038/s41525-023-00372-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2023] [Accepted: 09/04/2023] [Indexed: 09/25/2023] Open
Abstract
Lysosomal storage diseases (LSDs) are a group of metabolic disorders resulting from a deficiency in one of the lysosomal hydrolases. Most LSDs are inherited in an autosomal or X-linked recessive manner. As LSDs are rare, their true incidence in Taiwan remains unknown. In this study, we used high-coverage whole-genome sequencing data from 1,495 Taiwanese individuals obtained from the Taiwan Biobank. We found 3826 variants in 71 genes responsible for autosomal recessive LSDs. We first excluded benign variants by allele frequency and other criteria. As a result, 270 variants were considered disease-causing. We curated these variants using published guidelines from the American College of Medical Genetics and Genomics (ACMG). Our results revealed a combined incidence rate of 13 per 100,000 (conservative estimation by pathologic and likely pathogenic variants; 95% CI 6.92-22.23) to 94 per 100,000 (extended estimation by the inclusion of variants of unknown significance; 95% CI 75.96-115.03) among 71 autosomal recessive disease-associated genes. The conservative estimations were similar to those in published clinical data. No disease-causing mutations were found for 18 other diseases; thus, these diseases are likely extremely rare in Taiwan. The study results are important for designing screening and treatment methods for LSDs in Taiwan and demonstrate the importance of mutation curation to avoid overestimating disease incidences from genomic data.
Collapse
Affiliation(s)
- Meng-Ju Melody Tsai
- Department of Pediatrics, National Taiwan University Hospital and College of Medicine, National Taiwan University, Taipei, Taiwan
- Department of Pediatrics, National Taiwan University Hospital Yunlin Branch, Yunlin, Taiwan
| | - Miao-Zi Hung
- Department of Medical Genetics, National Taiwan University Hospital, Taipei, Taiwan
| | - Yi-Lin Lin
- Department of Medical Genetics, National Taiwan University Hospital, Taipei, Taiwan
| | - Ni-Chung Lee
- Department of Pediatrics, National Taiwan University Hospital and College of Medicine, National Taiwan University, Taipei, Taiwan
- Department of Medical Genetics, National Taiwan University Hospital, Taipei, Taiwan
| | - Yin-Hsiu Chien
- Department of Pediatrics, National Taiwan University Hospital and College of Medicine, National Taiwan University, Taipei, Taiwan
- Department of Medical Genetics, National Taiwan University Hospital, Taipei, Taiwan
| | - Wuh-Liang Hwu
- Department of Pediatrics, National Taiwan University Hospital and College of Medicine, National Taiwan University, Taipei, Taiwan.
- Department of Medical Genetics, National Taiwan University Hospital, Taipei, Taiwan.
- Center for Precision Medicine, China Medical University Hospital, Taichung, Taiwan.
| |
Collapse
|
11
|
Kubaski F, Sousa I, Amorim T, Pereira D, Silva C, Chaves V, Brusius-Facchin AC, Netto ABO, Soares J, Vairo F, Poletto E, Trometer J, Souza A, Ranieri E, Polo G, Hong X, Herbst ZM, Burlina A, Gelb MH, Giugliani R. Pilot study of newborn screening for six lysosomal diseases in Brazil. Mol Genet Metab 2023; 140:107654. [PMID: 37507255 DOI: 10.1016/j.ymgme.2023.107654] [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: 03/07/2023] [Revised: 07/10/2023] [Accepted: 07/11/2023] [Indexed: 07/30/2023]
Abstract
BACKGROUND Lysosomal diseases (LDs) are progressive life-threatening disorders that are usually asymptomatic at birth. Specific treatments are available for several LDs, and early intervention improves patient's outcomes. Thus, these diseases benefit from newborn screening (NBS). We have performed a pilot study for six LDs in Brazil by tandem mass spectrometry. METHODS Dried blood spot (DBS) samples of unselected newborns were analyzed by the Neo-LSD™ kit (Perkin-Elmer) by MS/MS. Samples with low enzyme activity were submitted to the evaluation of specific biomarkers by ultra-performance liquid chromatography tandem-mass spectrometry as the second-tier, and were analyzed by a next-generation sequencing (NGS) multi-gene panel as the third-tier. All tests were performed in the same DBS sample. RESULTS In 20,066 newborns analyzed, 15 samples showed activity of one enzyme below the cutoff. Two newborns had biochemical and molecular results compatible with Fabry disease, and five newborns had biochemical results and pathogenic variants or variants of unknown significance (VUS) in GAA. CONCLUSIONS This study indicates that the use of enzyme assay as the first-tier test gives an acceptably low number of positive results that requires second/third tier testing. The possibility to run all tests in a DBS sample makes this protocol applicable to large-scale NBS programs.
Collapse
Affiliation(s)
- Francyne Kubaski
- Greenwood Genetic Center, Biochemical Genetics Laboratory, Greenwood, USA; BioDiscovery Laboratory, HCPA, Porto Alegre, Brazil; Medical Genetics Service, HCPA, Porto Alegre, Brazil; PPGBM, UFRGS, Porto Alegre, Brazil.
| | | | - Tatiana Amorim
- APAE Salvador, Salvador, Brazil; Colegiado de Medicina, Universidade do Estado da Bahia, Salvador, Brazil.
| | | | - Camilo Silva
- Waters technologies do Brazil, Sao Paulo, Brazil.
| | - Vitor Chaves
- Waters technologies do Brazil, Sao Paulo, Brazil.
| | | | - Alice B O Netto
- BioDiscovery Laboratory, HCPA, Porto Alegre, Brazil; PPGBM, UFRGS, Porto Alegre, Brazil.
| | | | - Filippo Vairo
- Department of Clinical Genomics, Center for Individualized Medicine, Mayo Clinic, Rochester, USA.
| | - Edina Poletto
- Medical Genetics Service, HCPA, Porto Alegre, Brazil; PPGBM, UFRGS, Porto Alegre, Brazil
| | | | | | - Enzo Ranieri
- Women's and Children Hospital, Adelaide, Australia.
| | - Giulia Polo
- Division of Inherited Metabolic Diseases, Regional Center for Expanded Neonatal Screening, Department of Women and Children's Health, University Hospital of Padova, Padova, Italy
| | - Xinying Hong
- Department of Chemistry, University of Washington, Seattle, USA; Department of Pathology and Laboratory of Medicine, Children's Hospital of Philadelphia, Philadelphia, USA.
| | - Zackary M Herbst
- Department of Chemistry, University of Washington, Seattle, USA.
| | - Alberto Burlina
- Division of Inherited Metabolic Diseases, Regional Center for Expanded Neonatal Screening, Department of Women and Children's Health, University Hospital of Padova, Padova, Italy.
| | - Michael H Gelb
- Department of Chemistry, University of Washington, Seattle, USA.
| | - Roberto Giugliani
- BioDiscovery Laboratory, HCPA, Porto Alegre, Brazil; Medical Genetics Service, HCPA, Porto Alegre, Brazil; PPGBM, UFRGS, Porto Alegre, Brazil; DASA, Sao Paulo, Brazil; Casa dos Raros, Porto Alegre, Brazil.
| |
Collapse
|
12
|
Burton BK, Shively V, Quadri A, Warn L, Burton J, Grange DK, Christensen K, Groepper D, Ashbaugh L, Ehrhardt J, Basheeruddin K. Newborn screening for mucopolysaccharidosis type II: Lessons learned. Mol Genet Metab 2023; 140:107557. [PMID: 36907694 DOI: 10.1016/j.ymgme.2023.107557] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/03/2022] [Revised: 03/01/2023] [Accepted: 03/02/2023] [Indexed: 03/08/2023]
Abstract
We describe our experience with population-based newborn screening for mucopolysaccharidosis type II (MPS II) in 586,323 infants by measurement of iduronate-2-sulfatase activity in dried blood spots between December 12, 2017 and April 30, 2022. A total of 76 infants were referred for diagnostic testing, 0.01% of the screened population. Of these, eight cases of MPS II were diagnosed for an incidence of 1 in 73,290. At least four of the eight cases detected had an attenuated phenotype. In addition, cascade testing revealed a diagnosis in four extended family members. Fifty-three cases of pseudodeficiency were also identified, for an incidence of 1 in 11,062. Our data suggest that MPS II may be more common than previously recognized with a higher prevalence of attenuated cases.
Collapse
Affiliation(s)
- Barbara K Burton
- Ann & Robert H. Lurie Children's Hospital of Chicago, Chicago, IL, USA; Northwestern University Feinberg School of Medicine, Chicago, IL, USA.
| | - Vera Shively
- Ann & Robert H. Lurie Children's Hospital of Chicago, Chicago, IL, USA
| | - Allegra Quadri
- Ann & Robert H. Lurie Children's Hospital of Chicago, Chicago, IL, USA
| | - Lauren Warn
- University of Illinois College of Medicine at Peoria, Peoria, IL, USA
| | - Jennifer Burton
- University of Illinois College of Medicine at Peoria, Peoria, IL, USA; OSF St. Francis Hospital and Children's Hospital of Illinois, Peoria, IL, USA
| | | | | | - Daniel Groepper
- Southern Illinois University, School of Medicine, Springfield, IL, USA
| | - Laura Ashbaugh
- Illinois Department of Public Health, Newborn Screening Follow-up Program, Springfield, IL, USA
| | - Joan Ehrhardt
- Illinois Department of Public Health, Newborn Screening Follow-up Program, Springfield, IL, USA
| | - Khaja Basheeruddin
- Illinois Department of Public Health, Newborn Screening Laboratory, Chicago, IL, USA
| |
Collapse
|
13
|
Chien YH, Hwu WL. The modern face of newborn screening. Pediatr Neonatol 2023; 64 Suppl 1:S22-S29. [PMID: 36481189 DOI: 10.1016/j.pedneo.2022.11.001] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/17/2022] [Accepted: 11/07/2022] [Indexed: 11/16/2022] Open
Abstract
Newborn screening (NBS) has been developed for years to identify newborns with severe but treatable conditions. Taiwan's NBS system, after the initial setup for a total coverage of newborns in 1990s, was later optimized to ensure the timely return of results in infants with abnormal results. Advancements in techniques such as Tandem mass spectrometry enable the screening into a multiplex format and increase the conditions to be screened. Furthermore, advances in therapies, such as enzyme replacement therapy, stem cell transplantation, and gene therapy, significantly expand the needs for newborn screening. Advances in genomics and biomarkers discovery improve the test accuracy with the assistance of second-tier tests, and have the potential to be the first-tier test in the future. Therefore, challenge of NBS now is the knowledge gap, including the evidence of the long-term clinical benefits in large cohorts especially in conditions with new therapies, phenotypic variations and the corresponding management of some screened diseases, and cost-effectiveness of extended NBS programs. A short-term and a long-term follow-up program should be implemented to gather those outcomes better especially in the genomic era. Ethical and psychosocial issues are also potentially encountered frequently. Essential education and better informed consent should be considered fundamental to parallel those new tests into future NBS.
Collapse
Affiliation(s)
- Yin-Hsiu Chien
- Department of Medical Genetics, National Taiwan University Hospital, Taipei, Taiwan; Department of Pediatrics, National Taiwan University Hospital, Taipei, Taiwan; Department of Pediatrics, National Taiwan University College of Medicine, Taipei, Taiwan.
| | - Wuh-Liang Hwu
- Department of Medical Genetics, National Taiwan University Hospital, Taipei, Taiwan; Department of Pediatrics, National Taiwan University Hospital, Taipei, Taiwan; Department of Pediatrics, National Taiwan University College of Medicine, Taipei, Taiwan
| |
Collapse
|
14
|
Updated Confirmatory Diagnosis for Mucopolysaccharidoses in Taiwanese Infants and the Application of Gene Variants. Int J Mol Sci 2022; 23:ijms23179979. [PMID: 36077388 PMCID: PMC9456254 DOI: 10.3390/ijms23179979] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2022] [Revised: 08/26/2022] [Accepted: 08/30/2022] [Indexed: 12/03/2022] Open
Abstract
Mucopolysaccharidosis (MPS) is a lysosomal storage disease caused by genetic defects that result in deficiency of one specific enzyme activity, consequently impairing the stepwise degradation of glycosaminoglycans (GAGs). Except for MPS II, the other types of MPS have autosomal recessive inheritance in which two copies of an abnormal allele must be present in order for the disease to develop. In this study, we present the status of variant alleles and biochemistry results found in infants suspected of having MPS I, II, IVA, and VI. A total of 324 suspected infants, including 12 for MPS I, 223 for MPS II, 72 for MPS IVA, and 17 for MPS VI, who were referred for MPS confirmation from newborn screening centers in Taiwan, were enrolled. In all of these infants, one specific enzyme activity in dried blood spot filter paper was lower than the cut-off value in the first blood sample, as well asin a second follow-up sample. The confirmatory methods used in this study included Sanger sequencing, next-generation sequencing, leukocyte enzyme fluorometric assay, and GAG-derived disaccharides in urine using tandem mass spectrometry assays. The results showed that five, nine, and six infants had MPS I, II, and IVA, respectively, and all of them were asymptomatic. Thus, a laboratory diagnosis is extremely important to confirm the diagnosis of MPS. The other infants with identified nucleotide variations and reductions in leukocyte enzyme activities were categorized as being highly suspected cases requiring long-term and intensive follow-up examinations. In summary, the final confirmation of MPS depends on the most powerful biomarkers found in urine, i.e., the quantification of GAG-derived disaccharides including dermatan sulfate, heparan sulfate, and keratan sulfate, and analysis of genetic variants can help predict outcomes and guide treatment.
Collapse
|
15
|
Lin HY, Chang YH, Lee CL, Tu YR, Lo YT, Hung PW, Niu DM, Liu MY, Liu HY, Chen HJ, Kao SM, Wang LY, Ho HJ, Chuang CK, Lin SP. Newborn Screening Program for Mucopolysaccharidosis Type II and Long-Term Follow-Up of the Screen-Positive Subjects in Taiwan. J Pers Med 2022; 12:jpm12071023. [PMID: 35887520 PMCID: PMC9320252 DOI: 10.3390/jpm12071023] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2022] [Revised: 06/18/2022] [Accepted: 06/20/2022] [Indexed: 11/16/2022] Open
Abstract
Background: Mucopolysaccharidosis II (MPS II) is an X-linked disorder resulting from a deficiency in lysosomal enzyme iduronate-2-sulfatase (IDS), which causes the accumulation of glycosaminoglycans (GAGs) in the lysosomes of many tissues and organs, leading to progressive cellular dysfunction. An MPS II newborn screening program has been available in Taiwan since 2015. The aim of the current study was to collect and analyze the long-term follow-up data of the screen-positive subjects in this program. Methods: From August 2015 to April 2022, 548,624 newborns were screened for MPS II by dried blood spots using tandem mass spectrometry, of which 202 suspected infants were referred to our hospital for confirmation. The diagnosis of MPS II was confirmed by IDS enzyme activity assay in leukocytes, quantitative determination of urinary GAGs by mass spectrometry, and identification of the IDS gene variant. Results: Among the 202 referred infants, 10 (5%) with seven IDS gene variants were diagnosed with confirmed MPS II (Group 1), 151 (75%) with nine IDS gene variants were classified as having suspected MPS II or pseudodeficiency (Group 2), and 41 (20%) with five IDS gene variants were classified as not having MPS II (Group 3). Long-term follow-up every 6 months was arranged for the infants in Group 1 and Group 2. Intravenous enzyme replacement therapy (ERT) was started in four patients at 1, 0.5, 0.4, and 0.5 years of age, respectively. Three patients also received hematopoietic stem cell transplantation (HSCT) at 1.5, 0.9, and 0.6 years of age, respectively. After ERT and/or HSCT, IDS enzyme activity and the quantity of urinary GAGs significantly improved in all of these patients compared with the baseline data. Conclusions: Because of the progressive nature of MPS II, early diagnosis via a newborn screening program and timely initiation of ERT and/or HSCT before the occurrence of irreversible organ damage may lead to better clinical outcomes. The findings of the current study could serve as baseline data for the analysis of the long-term effects of ERT and HSCT in these patients.
Collapse
Affiliation(s)
- Hsiang-Yu Lin
- Department of Pediatrics, MacKay Memorial Hospital, Taipei 10449, Taiwan; (H.-Y.L.); (Y.-H.C.); (C.-L.L.)
- Department of Medical Research, MacKay Memorial Hospital, Taipei 10449, Taiwan;
- The Rare Disease Center, MacKay Memorial Hospital, Taipei 10449, Taiwan; (Y.-T.L.); (P.-W.H.)
- Department of Medicine, MacKay Medical College, New Taipei City 25245, Taiwan
- MacKay Junior College of Medicine, Nursing and Management, Taipei 11260, Taiwan
- Department of Medical Research, China Medical University Hospital, China Medical University, Taichung 40402, Taiwan
| | - Ya-Hui Chang
- Department of Pediatrics, MacKay Memorial Hospital, Taipei 10449, Taiwan; (H.-Y.L.); (Y.-H.C.); (C.-L.L.)
- The Rare Disease Center, MacKay Memorial Hospital, Taipei 10449, Taiwan; (Y.-T.L.); (P.-W.H.)
| | - Chung-Lin Lee
- Department of Pediatrics, MacKay Memorial Hospital, Taipei 10449, Taiwan; (H.-Y.L.); (Y.-H.C.); (C.-L.L.)
- Department of Medical Research, MacKay Memorial Hospital, Taipei 10449, Taiwan;
- The Rare Disease Center, MacKay Memorial Hospital, Taipei 10449, Taiwan; (Y.-T.L.); (P.-W.H.)
- Department of Medicine, MacKay Medical College, New Taipei City 25245, Taiwan
- MacKay Junior College of Medicine, Nursing and Management, Taipei 11260, Taiwan
| | - Yuan-Rong Tu
- Department of Medical Research, MacKay Memorial Hospital, Taipei 10449, Taiwan;
| | - Yun-Ting Lo
- The Rare Disease Center, MacKay Memorial Hospital, Taipei 10449, Taiwan; (Y.-T.L.); (P.-W.H.)
| | - Pei-Wen Hung
- The Rare Disease Center, MacKay Memorial Hospital, Taipei 10449, Taiwan; (Y.-T.L.); (P.-W.H.)
| | - Dau-Ming Niu
- Department of Pediatrics, Taipei Veterans General Hospital, Taipei 11217, Taiwan;
| | - Mei-Ying Liu
- The Chinese Foundation of Health, Neonatal Screening Center, Taipei 10699, Taiwan; (M.-Y.L.); (H.-Y.L.); (H.-J.C.); (S.-M.K.)
| | - Hsin-Yun Liu
- The Chinese Foundation of Health, Neonatal Screening Center, Taipei 10699, Taiwan; (M.-Y.L.); (H.-Y.L.); (H.-J.C.); (S.-M.K.)
| | - Hsiao-Jan Chen
- The Chinese Foundation of Health, Neonatal Screening Center, Taipei 10699, Taiwan; (M.-Y.L.); (H.-Y.L.); (H.-J.C.); (S.-M.K.)
| | - Shu-Min Kao
- The Chinese Foundation of Health, Neonatal Screening Center, Taipei 10699, Taiwan; (M.-Y.L.); (H.-Y.L.); (H.-J.C.); (S.-M.K.)
| | - Li-Yun Wang
- Taipei Institute of Pathology, Neonatal Screening Center, Taipei 10374, Taiwan; (L.-Y.W.); (H.-J.H.)
| | - Huey-Jane Ho
- Taipei Institute of Pathology, Neonatal Screening Center, Taipei 10374, Taiwan; (L.-Y.W.); (H.-J.H.)
| | - Chih-Kuang Chuang
- Department of Medical Research, MacKay Memorial Hospital, Taipei 10449, Taiwan;
- College of Medicine, Fu-Jen Catholic University, Taipei 24205, Taiwan
- Correspondence: (C.-K.C.); (S.-P.L.); Tel.: +886-2-2809-4661 (ext. 2348) (C.-K.C.); +886-2-2543-3535 (ext. 3090) (S.-P.L.); Fax: +886-2-2808-5952 (C.-K.C.); +886-2-2543-3642 (S.-P.L.)
| | - Shuan-Pei Lin
- Department of Pediatrics, MacKay Memorial Hospital, Taipei 10449, Taiwan; (H.-Y.L.); (Y.-H.C.); (C.-L.L.)
- Department of Medical Research, MacKay Memorial Hospital, Taipei 10449, Taiwan;
- The Rare Disease Center, MacKay Memorial Hospital, Taipei 10449, Taiwan; (Y.-T.L.); (P.-W.H.)
- Department of Medicine, MacKay Medical College, New Taipei City 25245, Taiwan
- Department of Infant and Child Care, National Taipei University of Nursing and Health Sciences, Taipei 11219, Taiwan
- Correspondence: (C.-K.C.); (S.-P.L.); Tel.: +886-2-2809-4661 (ext. 2348) (C.-K.C.); +886-2-2543-3535 (ext. 3090) (S.-P.L.); Fax: +886-2-2808-5952 (C.-K.C.); +886-2-2543-3642 (S.-P.L.)
| |
Collapse
|
16
|
Abstract
Mucopolysaccharidosis type I (MPS I), a lysosomal storage disease caused by a deficiency of α-L-iduronidase, leads to storage of the glycosaminoglycans, dermatan sulfate and heparan sulfate. Available therapies include enzyme replacement and hematopoietic stem cell transplantation. In the last two decades, newborn screening (NBS) has focused on early identification of the disorder, allowing early intervention and avoiding irreversible manifestations. Techniques developed and optimized for MPS I NBS include tandem mass-spectrometry, digital microfluidics, and glycosaminoglycan quantification. Several pilot studies have been conducted and screening programs have been implemented worldwide. NBS for MPS I has been established in Taiwan, the United States, Brazil, Mexico, and several European countries. All these programs measure α-L-iduronidase enzyme activity in dried blood spots, although there are differences in the analytical strategies employed. Screening algorithms based on published studies are discussed. However, some limitations remain: one is the high rate of false-positive results due to frequent pseudodeficiency alleles, which has been partially solved using post-analytical tools and second-tier tests; another involves the management of infants with late-onset forms or variants of uncertain significance. Nonetheless, the risk-benefit ratio is favorable. Furthermore, long-term follow-up of patients detected by neonatal screening will improve our knowledge of the natural history of the disease and inform better management.
Collapse
Affiliation(s)
- Alberto B Burlina
- Division of Inherited Metabolic Diseases, Department of Diagnostic Services, University Hospital, Padua, Italy
| | - Vincenza Gragnaniello
- Division of Inherited Metabolic Diseases, Department of Diagnostic Services, University Hospital, Padua, Italy
| |
Collapse
|
17
|
D’Avanzo F, Zanetti A, De Filippis C, Tomanin R. Mucopolysaccharidosis Type VI, an Updated Overview of the Disease. Int J Mol Sci 2021; 22:ijms222413456. [PMID: 34948256 PMCID: PMC8707598 DOI: 10.3390/ijms222413456] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Revised: 11/29/2021] [Accepted: 12/09/2021] [Indexed: 12/17/2022] Open
Abstract
Mucopolysaccharidosis type VI, or Maroteaux-Lamy syndrome, is a rare, autosomal recessive genetic disease, mainly affecting the pediatric age group. The disease is due to pathogenic variants of the ARSB gene, coding for the lysosomal hydrolase N-acetylgalactosamine 4-sulfatase (arylsulfatase B, ASB). The enzyme deficit causes a pathological accumulation of the undegraded glycosaminoglycans dermatan-sulphate and chondroitin-sulphate, natural substrates of ASB activity. Intracellular and extracellular deposits progressively take to a pathological scenario, often severe, involving most organ-systems and generally starting from the osteoarticular apparatus. Neurocognitive and behavioral abilities, commonly described as maintained, have been actually investigated by few studies. The disease, first described in 1963, has a reported prevalence between 0.36 and 1.3 per 100,000 live births across the continents. With this paper, we wish to contribute an updated overview of the disease from the clinical, diagnostic, and therapeutic sides. The numerous in vitro and in vivo preclinical studies conducted in the last 10-15 years to dissect the disease pathogenesis, the efficacy of the available therapeutic treatment (enzyme replacement therapy), as well as new therapies under study are here described. This review also highlights the need to identify new disease biomarkers, potentially speeding up the diagnostic process and the monitoring of therapeutic efficacy.
Collapse
Affiliation(s)
- Francesca D’Avanzo
- Laboratory of Diagnosis and Therapy of Lysosomal Disorders, Department of Women’s and Children’s Health, University of Padova, 35128 Padova, Italy; (F.D.); (A.Z.); (C.D.F.)
- Fondazione Istituto di Ricerca Pediatrica Città della Speranza, Corso Stati Uniti 4, 35127 Padova, Italy
| | - Alessandra Zanetti
- Laboratory of Diagnosis and Therapy of Lysosomal Disorders, Department of Women’s and Children’s Health, University of Padova, 35128 Padova, Italy; (F.D.); (A.Z.); (C.D.F.)
- Fondazione Istituto di Ricerca Pediatrica Città della Speranza, Corso Stati Uniti 4, 35127 Padova, Italy
| | - Concetta De Filippis
- Laboratory of Diagnosis and Therapy of Lysosomal Disorders, Department of Women’s and Children’s Health, University of Padova, 35128 Padova, Italy; (F.D.); (A.Z.); (C.D.F.)
- Fondazione Istituto di Ricerca Pediatrica Città della Speranza, Corso Stati Uniti 4, 35127 Padova, Italy
| | - Rosella Tomanin
- Laboratory of Diagnosis and Therapy of Lysosomal Disorders, Department of Women’s and Children’s Health, University of Padova, 35128 Padova, Italy; (F.D.); (A.Z.); (C.D.F.)
- Fondazione Istituto di Ricerca Pediatrica Città della Speranza, Corso Stati Uniti 4, 35127 Padova, Italy
- Correspondence: ; Tel.: +39-049-821-1264
| |
Collapse
|
18
|
Nationwide Newborn Screening Program for Mucopolysaccharidoses in Taiwan and an Update of the "Gold Standard" Criteria Required to Make a Confirmatory Diagnosis. Diagnostics (Basel) 2021; 11:diagnostics11091583. [PMID: 34573925 PMCID: PMC8465393 DOI: 10.3390/diagnostics11091583] [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: 08/02/2021] [Revised: 08/29/2021] [Accepted: 08/29/2021] [Indexed: 11/17/2022] Open
Abstract
Mucopolysaccharidoses (MPSs) are a group of lysosomal storage diseases (LSDs) caused by an inherited gene defect. MPS patients can remain undetected unless the initial signs or symptoms have been identified. Newborn screening (NBS) programs for MPSs have been implemented in Taiwan since 2015, and more than 48.5% of confirmed cases of MPS have since been referred from these NBS programs. The purpose of this study was to report the current status of NBS for MPSs in Taiwan and update the gold standard criteria required to make a confirmative diagnosis of MPS, which requires the presence of the following three laboratory findings: (1) elevation of individual urinary glycosaminoglycan (GAG)-derived disaccharides detected by MS/MS-based assay; (2) deficient activity of a particular leukocyte enzyme by fluorometric assay; and (3) verification of heterogeneous or homogeneous variants by Sanger sequencing or next generation sequencing. Up to 30 April 2021, 599,962 newborn babies have been screened through the NBS programs for MPS type I, II, VI, and IVA, and a total of 255 infants have been referred to MacKay Memorial Hospital for a confirmatory diagnosis. Of these infants, four cases were confirmed to have MPS I, nine cases MPS II, and three cases MPS IVA, with prevalence rates of 0.67, 2.92, and 4.13 per 100,000 live births, respectively. Intensive long-term regular physical and laboratory examinations for asymptomatic infants with confirmed MPS or with highly suspected MPS can enhance the ability to administer ERT in a timely fashion.
Collapse
|
19
|
Zebrafish, an In Vivo Platform to Screen Drugs and Proteins for Biomedical Use. Pharmaceuticals (Basel) 2021; 14:ph14060500. [PMID: 34073947 PMCID: PMC8225009 DOI: 10.3390/ph14060500] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Revised: 05/14/2021] [Accepted: 05/20/2021] [Indexed: 12/28/2022] Open
Abstract
The nearly simultaneous convergence of human genetics and advanced molecular technologies has led to an improved understanding of human diseases. At the same time, the demand for drug screening and gene function identification has also increased, albeit time- and labor-intensive. However, bridging the gap between in vitro evidence from cell lines and in vivo evidence, the lower vertebrate zebrafish possesses many advantages over higher vertebrates, such as low maintenance, high fecundity, light-induced spawning, transparent embryos, short generation interval, rapid embryonic development, fully sequenced genome, and some phenotypes similar to human diseases. Such merits have popularized the zebrafish as a model system for biomedical and pharmaceutical studies, including drug screening. Here, we reviewed the various ways in which zebrafish serve as an in vivo platform to perform drug and protein screening in the fields of rare human diseases, social behavior and cancer studies. Since zebrafish mutations faithfully phenocopy many human disorders, many compounds identified from zebrafish screening systems have advanced to early clinical trials, such as those for Adenoid cystic carcinoma, Dravet syndrome and Diamond-Blackfan anemia. We also reviewed and described how zebrafish are used to carry out environmental pollutant detection and assessment of nanoparticle biosafety and QT prolongation.
Collapse
|
20
|
Dangouloff T, Vrščaj E, Servais L, Osredkar D. Newborn screening programs for spinal muscular atrophy worldwide: Where we stand and where to go. Neuromuscul Disord 2021; 31:574-582. [PMID: 33985857 DOI: 10.1016/j.nmd.2021.03.007] [Citation(s) in RCA: 110] [Impact Index Per Article: 27.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2021] [Revised: 03/10/2021] [Accepted: 03/16/2021] [Indexed: 02/05/2023]
Abstract
Spinal muscular atrophy (SMA) is a rare and devastating disease. New disease-modifying treatments have recently been approved and early treatment has been related to a better outcome. In this context, several newborn screening (NBS) programs have been implemented. The aim of the study was to obtain a global overview on the current situation and perspectives on SMA NBS. We conducted a survey and contacted experts from 152 countries, from which we gathered 87 responses. We identified 9 SMA NBS programs that have so far detected 288 newborns with SMA out of 3,674,277 newborns screened. Funding, screening methods, organisation, and consent process were variable between SMA NBS programs. Many respondents pointed the lack of cost/benefit data as a major obstacle to SMA NBS implementation. In the next four years, our data suggest a 24% coverage of newborns from countries where a disease-modifying drug is available and 8,5% coverage in countries with no diseases-modifying drugs. The annual proportion of newborns to be screened in the coming years is expected to increase steadily. The experts expressed a strong need for the implementation of SMA NBS as means to improve care for patients with SMA.
Collapse
Affiliation(s)
- Tamara Dangouloff
- Division of Child Neurology, Reference Center for Neuromuscular Diseases, Department of Pediatrics, University Hospital Liège & University of Liège, CRMN Liège, CHR de la Citadelle, Boulevard du 12ème de Ligne, 4000 Liège, Belgium
| | - Eva Vrščaj
- Department of Pediatric Neurology, University Children's Hospital, University Medical Centre Ljubljana, Bohoričeva 20, 1525 Ljubljana, Slovenia
| | - Laurent Servais
- Division of Child Neurology, Reference Center for Neuromuscular Diseases, Department of Pediatrics, University Hospital Liège & University of Liège, CRMN Liège, CHR de la Citadelle, Boulevard du 12ème de Ligne, 4000 Liège, Belgium; MDUK Neuromuscular Centre, Department of Paediatrics, University of Oxford, United Kingdom.
| | - Damjan Osredkar
- Department of Pediatric Neurology, University Children's Hospital, University Medical Centre Ljubljana, Bohoričeva 20, 1525 Ljubljana, Slovenia; Medical Faculty, University of Ljubljana, Slovenia.
| |
Collapse
|
21
|
Lin HY, Chen MR, Lee CL, Lin SM, Hung CL, Niu DM, Chang TM, Chuang CK, Lin SP. Natural progression of cardiac features and long-term effects of enzyme replacement therapy in Taiwanese patients with mucopolysaccharidosis II. Orphanet J Rare Dis 2021; 16:99. [PMID: 33622387 PMCID: PMC7903682 DOI: 10.1186/s13023-021-01743-2] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2020] [Accepted: 02/11/2021] [Indexed: 11/29/2022] Open
Abstract
Background Cardiac abnormalities have been observed in patients with mucopolysaccharidosis type II (MPS II). The aim of this study was to investigate the cardiac features and natural progression of Taiwanese patients with MPS II, and evaluate the impact of enzyme replacement therapy (ERT) on cardiac structure and function. Methods The medical records and echocardiograms of 48 Taiwanese patients with MPS II (median age, 6.9 years; age range, 0.1–27.9 years) were reviewed. The relationships between age and each echocardiographic parameter were analyzed. Results The mean z-scores of left ventricular mass index (LVMI), interventricular septum diameter in diastole (IVSd), left ventricular posterior wall diameter in diastole (LVPWd), and aortic diameter were 1.10, 2.70, 0.95 and 1.91, respectively. Z scores > 2 were identified in 33%, 54%, 13%, and 46% for LVMI, IVSd, LVPWd, and aortic diameter, respectively. The most prevalent cardiac valve abnormality was mitral regurgitation (MR) (56%), followed by aortic regurgitation (AR) (33%). The severity of mitral stenosis (MS), MR, aortic stenosis (AS), AR, and the existence of valvular heart disease were all positively correlated with increasing age (p < 0.01). We also compared the echocardiographic parameters between two groups: (1) 12 patients who had up to 17 years of follow-up echocardiographic data without ERT, and (2) nine patients who had up to 12 years of follow-up data with ERT. The results showed that z-score changes of LVMI significantly improved in the patients who received ERT compared to those who did not receive ERT (0.05 versus 1.52, p < 0.05). However, the severity score changes of MS, MR, AS, and AR all showed gradual progression in both groups (p > 0.05). Conclusions High prevalence rates of valvular heart disease and cardiac hypertrophy were observed in the MPS II patients in this study. The existence and severity of cardiac hypertrophy and valvular heart disease in these patients worsened with increasing age, reinforcing the concept of the progressive nature of this disease. ERT for MPS II appeared to be effective in stabilizing or reducing the progression of cardiac hypertrophy, but it only had a limited effect on valvulopathy.
Collapse
Affiliation(s)
- Hsiang-Yu Lin
- Department of Medicine, MacKay Medical College, New Taipei City, Taiwan.,Department of Pediatrics, MacKay Memorial Hospital, No.92, Sec. 2, Chung-Shan North Road, Taipei, 10449, Taiwan.,Department of Medical Research, MacKay Memorial Hospital, 92 Chung-Shan N. Rd., Sec. 2, Taipei, 10449, Taiwan.,Department of Childhood Care and Education, MacKay Junior College of Medicine, Nursing and Management, Taipei, Taiwan.,Department of Medical Research, China Medical University Hospital, China Medical University, Taichung, Taiwan.,Department of Rare Disease Center, MacKay Memorial Hospital, Taipei, Taiwan
| | - Ming-Ren Chen
- Department of Medicine, MacKay Medical College, New Taipei City, Taiwan.,Department of Pediatrics, MacKay Memorial Hospital, No.92, Sec. 2, Chung-Shan North Road, Taipei, 10449, Taiwan.,Department of Childhood Care and Education, MacKay Junior College of Medicine, Nursing and Management, Taipei, Taiwan
| | - Chung-Lin Lee
- Department of Childhood Care and Education, MacKay Junior College of Medicine, Nursing and Management, Taipei, Taiwan.,Department of Pediatrics, MacKay Memorial Hospital, Hsinchu, Taiwan.,Institute of Clinical Medicine, National Yang-Ming University, Taipei, Taiwan
| | - Shan-Miao Lin
- Department of Medicine, MacKay Medical College, New Taipei City, Taiwan.,Department of Pediatrics, MacKay Memorial Hospital, No.92, Sec. 2, Chung-Shan North Road, Taipei, 10449, Taiwan.,Department of Childhood Care and Education, MacKay Junior College of Medicine, Nursing and Management, Taipei, Taiwan
| | - Chung-Lieh Hung
- Department of Medicine, MacKay Medical College, New Taipei City, Taiwan.,Division of Cardiology, Department of Internal Medicine, MacKay Memorial Hospital, Taipei, Taiwan
| | - Dau-Ming Niu
- Institute of Clinical Medicine, National Yang-Ming University, Taipei, Taiwan.,Department of Pediatrics, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Tung-Ming Chang
- Department of Pediatric Neurology, Changhua Christian Children's Hospital, Changhua, Taiwan.,School of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Chih-Kuang Chuang
- Department of Medical Research, MacKay Memorial Hospital, 92 Chung-Shan N. Rd., Sec. 2, Taipei, 10449, Taiwan. .,College of Medicine, Fu-Jen Catholic University, Taipei, Taiwan.
| | - Shuan-Pei Lin
- Department of Medicine, MacKay Medical College, New Taipei City, Taiwan. .,Department of Pediatrics, MacKay Memorial Hospital, No.92, Sec. 2, Chung-Shan North Road, Taipei, 10449, Taiwan. .,Department of Medical Research, MacKay Memorial Hospital, 92 Chung-Shan N. Rd., Sec. 2, Taipei, 10449, Taiwan. .,Department of Rare Disease Center, MacKay Memorial Hospital, Taipei, Taiwan. .,Department of Infant and Child Care, National Taipei University of Nursing and Health Sciences, Taipei, Taiwan.
| |
Collapse
|
22
|
Epidemiology of Mucopolysaccharidoses Update. Diagnostics (Basel) 2021; 11:diagnostics11020273. [PMID: 33578874 PMCID: PMC7916572 DOI: 10.3390/diagnostics11020273] [Citation(s) in RCA: 55] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2021] [Revised: 02/03/2021] [Accepted: 02/05/2021] [Indexed: 12/26/2022] Open
Abstract
Mucopolysaccharidoses (MPS) are a group of lysosomal storage disorders caused by a lysosomal enzyme deficiency or malfunction, which leads to the accumulation of glycosaminoglycans in tissues and organs. If not treated at an early stage, patients have various health problems, affecting their quality of life and life-span. Two therapeutic options for MPS are widely used in practice: enzyme replacement therapy and hematopoietic stem cell transplantation. However, early diagnosis of MPS is crucial, as treatment may be too late to reverse or ameliorate the disease progress. It has been noted that the prevalence of MPS and each subtype varies based on geographic regions and/or ethnic background. Each type of MPS is caused by a wide range of the mutational spectrum, mainly missense mutations. Some mutations were derived from the common founder effect. In the previous study, Khan et al. 2018 have reported the epidemiology of MPS from 22 countries and 16 regions. In this study, we aimed to update the prevalence of MPS across the world. We have collected and investigated 189 publications related to the prevalence of MPS via PubMed as of December 2020. In total, data from 33 countries and 23 regions were compiled and analyzed. Saudi Arabia provided the highest frequency of overall MPS because of regional or consanguineous marriages (or founder effect), followed by Portugal, Brazil, the Netherlands, and Australia. The newborn screening is an efficient and early diagnosis for MPS. MPS I has been approved for newborn screening in the United States. After the newborn screening of MPS I, the frequency of MPS I increased, compared with the past incidence rates. Overall, we conclude that the current identification methods are not enough to recognize all MPS patients, leading to an inaccurate incidence and status. Differences in ethnic background and/or founder effects impact on the frequency of MPS, which affects the prevalence of MPS. Two-tier newborn screening has accelerated early recognition of MPS I, providing an accurate incidence of patients.
Collapse
|
23
|
Lin HY, Chen MR, Lee CL, Lin SM, Hung CL, Niu DM, Chang TM, Chuang CK, Lin SP. Aortic Root Dilatation in Taiwanese Patients with Mucopolysaccharidoses and the Long-Term Effects of Enzyme Replacement Therapy. Diagnostics (Basel) 2020; 11:diagnostics11010016. [PMID: 33374885 PMCID: PMC7823494 DOI: 10.3390/diagnostics11010016] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2020] [Revised: 12/04/2020] [Accepted: 12/18/2020] [Indexed: 12/02/2022] Open
Abstract
Background: Cardiovascular abnormalities have been observed in patients with mucopolysaccharidosis (MPS) of any type, with the most documented abnormalities being valvular regurgitation and stenosis and cardiac hypertrophy. Only a few studies have focused on aortic root dilatation and the long-term effects of enzyme replacement therapy (ERT) in these patients. Methods: We reviewed echocardiograms of 125 Taiwanese MPS patients (age range, 0.1 to 19.1 years; 11 with MPS I, 49 with MPS II, 25 with MPS III, 29 with MPS IVA, and 11 with MPS VI). The aortic root diameter was measured at the sinus of Valsalva. Results: Aortic root dilatation (z score >2) was observed in 47% of the MPS patients, including 66% of MPS IV, 51% of MPS II, 45% of MPS VI, 28% of MPS III, and 27% of MPS I patients. The mean aortic root diameter z score was 2.14 (n = 125). The patients with MPS IV had the most severe aortic root dilatation with a mean aortic root diameter z score of 3.03, followed by MPS II (2.12), MPS VI (2.06), MPS III (1.68), and MPS I (1.03). The aortic root diameter z score was positively correlated with increasing age (n = 125, p < 0.01). For the patients with MPS II, III, and IV, aortic root diameter z score was also positively correlated with increasing age (p < 0.01). For 16 patients who had received ERT and had follow-up echocardiographic data (range 2.0–16.2 years), the mean aortic root diameter z score change was −0.46 compared to baseline (baseline 2.49 versus follow-up 2.03, p = 0.490). Conclusions: Aortic root dilatation was common in the patients with all types of MPS, with the most severe aortic root dilatation observed in those with MPS IV. The severity of aortic root dilatation worsened with increasing age, reinforcing the concept of the progressive nature of this disease. ERT for MPS appears to stabilize the progression of aortic root dilatation.
Collapse
Affiliation(s)
- Hsiang-Yu Lin
- Department of Medicine, MacKay Medical College, New Taipei City 25245, Taiwan; (H.-Y.L.); (M.-R.C.); (S.-M.L.); (C.-L.H.)
- Department of Pediatrics, MacKay Memorial Hospital, Taipei 10449, Taiwan
- Department of Medical Research, MacKay Memorial Hospital, New Taipei City 25160, Taiwan
- MacKay Junior College of Medicine, Nursing and Management, Taipei 11260, Taiwan;
- Department of Medical Research, China Medical University Hospital, China Medical University, Taichung 40402, Taiwan
- Department of Rare Disease Center, MacKay Memorial Hospital, Taipei 10449, Taiwan
| | - Ming-Ren Chen
- Department of Medicine, MacKay Medical College, New Taipei City 25245, Taiwan; (H.-Y.L.); (M.-R.C.); (S.-M.L.); (C.-L.H.)
- Department of Pediatrics, MacKay Memorial Hospital, Taipei 10449, Taiwan
- MacKay Junior College of Medicine, Nursing and Management, Taipei 11260, Taiwan;
| | - Chung-Lin Lee
- MacKay Junior College of Medicine, Nursing and Management, Taipei 11260, Taiwan;
- Department of Rare Disease Center, MacKay Memorial Hospital, Taipei 10449, Taiwan
- Department of Pediatrics, MacKay Memorial Hospital, Hsinchu 30071, Taiwan
- Institute of Clinical Medicine, National Yang-Ming University, Taipei 11221, Taiwan;
| | - Shan-Miao Lin
- Department of Medicine, MacKay Medical College, New Taipei City 25245, Taiwan; (H.-Y.L.); (M.-R.C.); (S.-M.L.); (C.-L.H.)
- Department of Pediatrics, MacKay Memorial Hospital, Taipei 10449, Taiwan
- MacKay Junior College of Medicine, Nursing and Management, Taipei 11260, Taiwan;
| | - Chung-Lieh Hung
- Department of Medicine, MacKay Medical College, New Taipei City 25245, Taiwan; (H.-Y.L.); (M.-R.C.); (S.-M.L.); (C.-L.H.)
- Division of Cardiology, Department of Internal Medicine, MacKay Memorial Hospital, Taipei 10449, Taiwan
| | - Dau-Ming Niu
- Institute of Clinical Medicine, National Yang-Ming University, Taipei 11221, Taiwan;
- Department of Pediatrics, Taipei Veterans General Hospital, Taipei 11217, Taiwan
| | - Tung-Ming Chang
- Department of Pediatric Neurology, Changhua Christian Children’s Hospital, Changhua 500, Taiwan;
- School of Medicine, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
| | - Chih-Kuang Chuang
- Department of Medical Research, MacKay Memorial Hospital, New Taipei City 25160, Taiwan
- Department of Rare Disease Center, MacKay Memorial Hospital, Taipei 10449, Taiwan
- College of Medicine, Fu-Jen Catholic University, Taipei 24205, Taiwan
- Correspondence: (C.-K.C.); (S.-P.L.); Tel.: +886-2-2809-4661 (ext. 2348) (C.-K.C.); +886-2-2543-3535 (ext. 3090) (S.-P.L.); Fax: +886-2-2808-5952 (C.-K.C.); +886-2-2543-3642 (S.-P.L.)
| | - Shuan-Pei Lin
- Department of Medicine, MacKay Medical College, New Taipei City 25245, Taiwan; (H.-Y.L.); (M.-R.C.); (S.-M.L.); (C.-L.H.)
- Department of Pediatrics, MacKay Memorial Hospital, Taipei 10449, Taiwan
- Department of Medical Research, MacKay Memorial Hospital, New Taipei City 25160, Taiwan
- Department of Rare Disease Center, MacKay Memorial Hospital, Taipei 10449, Taiwan
- Department of Infant and Child Care, National Taipei University of Nursing and Health Sciences, Taipei 11219, Taiwan
- Correspondence: (C.-K.C.); (S.-P.L.); Tel.: +886-2-2809-4661 (ext. 2348) (C.-K.C.); +886-2-2543-3535 (ext. 3090) (S.-P.L.); Fax: +886-2-2808-5952 (C.-K.C.); +886-2-2543-3642 (S.-P.L.)
| |
Collapse
|
24
|
Kubaski F, Sousa I, Amorim T, Pereira D, Trometer J, Souza A, Ranieri E, Polo G, Burlina A, Brusius-Facchin AC, Netto ABO, Tomatsu S, Giugliani R. Neonatal Screening for MPS Disorders in Latin America: A Survey of Pilot Initiatives. Int J Neonatal Screen 2020; 6:ijns6040090. [PMID: 33203019 PMCID: PMC7711587 DOI: 10.3390/ijns6040090] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/09/2020] [Revised: 11/07/2020] [Accepted: 11/10/2020] [Indexed: 11/30/2022] Open
Abstract
Newborn screening enables the diagnosis of treatable disorders at the early stages, and because of its countless benefits, conditions have been continuously added to screening panels, allowing early intervention, aiming for the prevention of irreversible manifestations and even premature death. Mucopolysaccharidoses (MPS) are lysosomal storage disorders than can benefit from an early diagnosis, and thus are being recommended for newborn screening. They are multisystemic progressive disorders, with treatment options already available for several MPS types. MPS I was the first MPS disorder enrolled in the newborn screening (NBS) panel in the USA and a few other countries, and other MPS types are expected to be added. Very few studies about NBS for MPS in Latin America have been published so far. In this review, we report the results of pilot studies performed in Mexico and Brazil using different methodologies: tandem mass spectrometry, molecular analysis, digital microfluidics, and fluorimetry. These experiences are important to report and discuss, as we expect to have several MPS types added to NBS panels shortly. This addition will enable timely diagnosis of MPS, avoiding the long diagnostic odyssey that is part of the current natural history of this group of diseases, and leading to a better outcome for the affected patients.
Collapse
Affiliation(s)
- Francyne Kubaski
- Postgraduate Program in Genetics and Molecular Biology, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre 90040-060, Brazil;
- Medical Genetics Service, Hospital de Clínicas de Porto Alegre (HCPA), Porto Alegre 90035-903, Brazil; (A.C.B.-F.); (A.B.O.N.)
- Instituto Nacional de Genética Médica Populacional (iNaGeMP), Porto Alegre 90035-003, Brazil
- Correspondence: ; Tel.: +55-51-3359-8011
| | - Inês Sousa
- Associação de Pais e Amigos dos Excepcionais (APAE) Salvador, Salvador 41830-141, Brazil; (I.S.); (T.A.)
| | - Tatiana Amorim
- Associação de Pais e Amigos dos Excepcionais (APAE) Salvador, Salvador 41830-141, Brazil; (I.S.); (T.A.)
| | - Danilo Pereira
- Department of Research and Innovation, Innovatox, São Paulo 06455-020, Brazil;
| | | | | | - Enzo Ranieri
- SA Pathology, Women’s and Children’s Hospital, Adelaide, SA 5006, Australia;
| | - Giulia Polo
- Division of Inherited Metabolic Diseases, Department of Diagnostic Services, University Hospital of Padua, 35129 Padua, Italy; (G.P.); (A.B.)
| | - Alberto Burlina
- Division of Inherited Metabolic Diseases, Department of Diagnostic Services, University Hospital of Padua, 35129 Padua, Italy; (G.P.); (A.B.)
| | - Ana Carolina Brusius-Facchin
- Medical Genetics Service, Hospital de Clínicas de Porto Alegre (HCPA), Porto Alegre 90035-903, Brazil; (A.C.B.-F.); (A.B.O.N.)
| | - Alice Brinckmann Oliveira Netto
- Medical Genetics Service, Hospital de Clínicas de Porto Alegre (HCPA), Porto Alegre 90035-903, Brazil; (A.C.B.-F.); (A.B.O.N.)
- Instituto Nacional de Genética Médica Populacional (iNaGeMP), Porto Alegre 90035-003, Brazil
- Department of Biological Sciences, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre 90040-060, Brazil
| | - Shunji Tomatsu
- Department of Orthopedics and Biomedical, Nemours/Alfred I. duPont Hospital for Children, Wilmington, DE 19803, USA;
| | - Roberto Giugliani
- Postgraduate Program in Genetics and Molecular Biology, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre 90040-060, Brazil;
- Medical Genetics Service, Hospital de Clínicas de Porto Alegre (HCPA), Porto Alegre 90035-903, Brazil; (A.C.B.-F.); (A.B.O.N.)
- Instituto Nacional de Genética Médica Populacional (iNaGeMP), Porto Alegre 90035-003, Brazil
| |
Collapse
|
25
|
Lin HY, Lee CL, Chang CY, Chiu PC, Chien YH, Niu DM, Tsai FJ, Hwu WL, Lin SJ, Lin JL, Chao MC, Chang TM, Tsai WH, Wang TJ, Chuang CK, Lin SP. Survival and diagnostic age of 175 Taiwanese patients with mucopolysaccharidoses (1985-2019). Orphanet J Rare Dis 2020; 15:314. [PMID: 33160388 PMCID: PMC7648385 DOI: 10.1186/s13023-020-01598-z] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2020] [Accepted: 10/26/2020] [Indexed: 01/03/2023] Open
Abstract
Background Mucopolysaccharidoses (MPSs) are a group of inherited metabolic diseases, which are characterized by the accumulation of glycosaminoglycans, and eventually lead to the progressive damage of various tissues and organs. Methods An epidemiological study of MPS in Taiwan was performed using multiple sources. The survival and diagnostic age for different types of MPS between 1985 and 2019 were evaluated.
Results Between 1985 and 2019, there were 175 patients diagnosed with MPS disorders in the Taiwanese population, with a median diagnostic age of 3.9 years. There were 21 (12%), 78 (45%), 33 (19%), 32 (18%) and 11 (6%) patients diagnosed with MPS I, II, III, IV and VI, respectively, with median diagnostic ages of 1.5, 3.8, 4.7, 4.5 and 3.7 years, respectively. Diagnosis of MPS patients was significantly earlier in recent decades (p < 0.01). Pilot newborn screening programs for MPS I, II, VI, IVA, and IIIB were progressively introduced in Taiwan from 2016, and 48% (16/33) of MPS patients diagnosed between 2016 and 2019 were diagnosed by one of these screening programs, with a median diagnostic age at 0.2 years. For patients born between 2016 and 2019, up to 94% (16/17) were diagnosed with MPS via the newborn screening programs. At the time of this study, 81 patients had passed away with a median age at death of 15.6 years. Age at diagnosis was positively correlated with life expectancy (p < 0.01). Life expectancy also significantly increased between 1985 and 2019, however this increase was gradual (p < 0.01). Conclusions The life expectancy of Taiwanese patients with MPS has improved in recent decades and patients are being diagnosed earlier. Because of the progressive nature of the disease, early diagnosis by newborn screening programs and timely implementation of early therapeutic interventions may lead to better clinical outcomes.
Collapse
Affiliation(s)
- Hsiang-Yu Lin
- Department of Medicine, MacKay Medical College, New Taipei City, Taiwan.,Department of Pediatrics, MacKay Memorial Hospital, No.92, Sec. 2 Chung-Shan North Road, Taipei, 10449, Taiwan.,Department of Medical Research, MacKay Memorial Hospital, 92 Chung-Shan N. Rd., Sec. 2, Taipei, 10449, Taiwan.,Nursing and Management, MacKay Junior College of Medicine, Taipei, Taiwan.,Department of Medical Research, China Medical University Hospital, China Medical University, Taichung, Taiwan.,Institute of Biomedical Sciences, MacKay Medical College, New Taipei City, Taiwan
| | - Chung-Lin Lee
- Nursing and Management, MacKay Junior College of Medicine, Taipei, Taiwan.,Department of Pediatrics, MacKay Memorial Hospital, Hsinchu, Taiwan.,Institute of Clinical Medicine, National Yang-Ming University, Taipei, Taiwan
| | - Chia-Ying Chang
- Department of Pediatrics, MacKay Memorial Hospital, Hsinchu, Taiwan
| | - Pao Chin Chiu
- Department of Pediatrics, Kaohsiung Veterans General Hospital, Kaohsiung, Taiwan
| | - Yin-Hsiu Chien
- Department of Pediatrics, National Taiwan University Hospital, Taipei, Taiwan
| | - Dau-Ming Niu
- Institute of Clinical Medicine, National Yang-Ming University, Taipei, Taiwan.,Department of Pediatrics, Taipei Veterans General 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
| | - Ju-Li Lin
- Department of Pediatrics, Chang Gung Memorial Hospital at Linkou, Taoyuan, Taiwan
| | - Mei-Chyn Chao
- Department of Pediatrics, Changhua Christian Children's Hospital, Changhua, Taiwan
| | - Tung-Ming Chang
- Department of Pediatric Neurology, Changhua Christian Children's Hospital, Changhua, Taiwan.,School of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Wen-Hui Tsai
- Department of Pediatrics, Chi Mei Medical Center, Tainan, Taiwan
| | - Tzu-Jou Wang
- Department of Pediatrics, Chang Gung Memorial Hospital, Kaohsiung, Taiwan
| | - Chih-Kuang Chuang
- Department of Medical Research, MacKay Memorial Hospital, 92 Chung-Shan N. Rd., Sec. 2, Taipei, 10449, Taiwan. .,College of Medicine, Fu-Jen Catholic University, Taipei, Taiwan.
| | - Shuan-Pei Lin
- Department of Medicine, MacKay Medical College, New Taipei City, Taiwan. .,Department of Pediatrics, MacKay Memorial Hospital, No.92, Sec. 2 Chung-Shan North Road, Taipei, 10449, Taiwan. .,Department of Medical Research, MacKay Memorial Hospital, 92 Chung-Shan N. Rd., Sec. 2, Taipei, 10449, Taiwan. .,Department of Infant and Child Care, National Taipei University of Nursing and Health Sciences, Taipei, Taiwan.
| |
Collapse
|
26
|
Lin CY, Lin HY, Chuang CK, Zhang PH, Tu RY, Lin SP, Tsai HJ. Effect of Mutated ids Overexpression on IDS Enzyme Activity and Developmental Phenotypes in Zebrafish Embryos: A Valuable Index for Assessing Critical Point-Mutations Associated with Mucopolysaccharidosis Type II Occurrence in Humans. Diagnostics (Basel) 2020; 10:diagnostics10100854. [PMID: 33096603 PMCID: PMC7589091 DOI: 10.3390/diagnostics10100854] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2020] [Revised: 10/09/2020] [Accepted: 10/19/2020] [Indexed: 01/08/2023] Open
Abstract
Mucopolysaccharidosis type II (MPS II) is an X-linked disorder resulting from a deficiency in iduronate 2-sulfatase (IDS), which is reported to be caused by gene mutations in the iduronate 2-sulfatase (IDS) gene. Many IDS mutation sites have not yet had their causal relationship with MPS II characterized. We employed a gain-of-function strategy whereby we microinjected different mutated zebrafish ids (z-ids) mRNAs corresponded to human IDS gene into zebrafish embryos, and then measured their total IDS enzymatic activity and observed the occurrence of defective phenotypes during embryonic development. We examined three known mutation sites for human IDS genes (h-IDS) associated with MPS II symptoms, including h-IDS-P86L, -S333L and -R468W, which corresponded to z-ids-P80L, -S327L and -R454W. When these three mutated z-ids mRNAs were overexpressed in zebrafish embryos, the IDS enzymatic activity of the total proteins extracted from the injected embryos was not increased compared with the endogenous IDS of the untreated embryos, which suggests that the IDS enzymatic activity of these three mutated z-ids was totally lost, as expected. Additionally, we observed defective phenotypes in these injected embryos, resulting from the failed IDS enzyme breakdown, which, in turn, has a dominant negative effect on the endogenous wild-type IDS function. These phenotypes were similar to the clinical symptoms observed in MPS II pathogenesis. We further studied six uncharacterized IDS mutation sites as identified by the Taiwanese MPS newborn screening programs. We propose a novel IDS enzyme activity assay combined with phenotypic observation in zebrafish embryos, as an alternative platform for quickly providing a valuable index for preliminarily assessment of any identified IDS point mutation gene that has not yet been characterized, in the context of its role in MPS II development.
Collapse
Affiliation(s)
- Cheng-Yung Lin
- Institute of Biomedical Sciences, MacKay Medical College, New Taipei City 25245, Taiwan; (C.-Y.L.); (H.-Y.L.); (P.-H.Z.)
| | - Hsiang-Yu Lin
- Institute of Biomedical Sciences, MacKay Medical College, New Taipei City 25245, Taiwan; (C.-Y.L.); (H.-Y.L.); (P.-H.Z.)
- Department of Medicine, MacKay Medical College, New Taipei City 25245, Taiwan
- Department of Pediatrics, MacKay Memorial Hospital, Taipei 10449, Taiwan
- Department of Medical Research, MacKay Memorial Hospital, New Taipei City 25160, Taiwan; (C.-K.C.); (R.-Y.T.)
- MacKay Junior College of Medicine, Nursing and Management, Taipei 11260, Taiwan
- Department of Medical Research, China Medical University Hospital, China Medical University, Taichung 40402, Taiwan
| | - Chih-Kuang Chuang
- Department of Medical Research, MacKay Memorial Hospital, New Taipei City 25160, Taiwan; (C.-K.C.); (R.-Y.T.)
- College of Medicine, Fu-Jen Catholic University, Taipei 24205, Taiwan
| | - Po-Hsiang Zhang
- Institute of Biomedical Sciences, MacKay Medical College, New Taipei City 25245, Taiwan; (C.-Y.L.); (H.-Y.L.); (P.-H.Z.)
- Department of Medical Research, MacKay Memorial Hospital, New Taipei City 25160, Taiwan; (C.-K.C.); (R.-Y.T.)
| | - Ru-Yi Tu
- Department of Medical Research, MacKay Memorial Hospital, New Taipei City 25160, Taiwan; (C.-K.C.); (R.-Y.T.)
| | - Shuan-Pei Lin
- Department of Medicine, MacKay Medical College, New Taipei City 25245, Taiwan
- Department of Pediatrics, MacKay Memorial Hospital, Taipei 10449, Taiwan
- Department of Medical Research, MacKay Memorial Hospital, New Taipei City 25160, Taiwan; (C.-K.C.); (R.-Y.T.)
- Department of Infant and Child Care, National Taipei University of Nursing and Health Sciences, Taipei 11219, Taiwan
- Correspondence: (S.-P.L.); (H.-J.T.)
| | - Huai-Jen Tsai
- Institute of Biomedical Sciences, MacKay Medical College, New Taipei City 25245, Taiwan; (C.-Y.L.); (H.-Y.L.); (P.-H.Z.)
- Correspondence: (S.-P.L.); (H.-J.T.)
| |
Collapse
|
27
|
Washburn J, Millington DS. Digital Microfluidics in Newborn Screening for Mucopolysaccharidoses: A Progress Report. Int J Neonatal Screen 2020; 6:ijns6040078. [PMID: 33124616 PMCID: PMC7711648 DOI: 10.3390/ijns6040078] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/11/2020] [Revised: 09/29/2020] [Accepted: 10/04/2020] [Indexed: 11/16/2022] Open
Abstract
Newborn screening (NBS) for mucopolysaccharidosis type I (MPS I, Hurler syndrome) is currently conducted in about two-fifths of the NBS programs in the United States and in a few other countries. Screening is performed by measurement of residual activity of the enzyme alpha-l-iduronidase in dried blood spots using either tandem mass spectrometry or digital microfluidic fluorometry (DMF). In this article, we focus on the development and practical experience of using DMF to screen for MPS I in the USA. By means of their responses to a questionnaire, we determined for each responding program that is screening for MPS I using DMF the screen positive rate, follow-up methods, and classification of confirmed cases as either severe or attenuated. Overall, the results show that at the time of reporting, over 1.3 million newborns in the US were screened for MPS I using DMF, 2094 (0.173%) of whom were screen positive. Of these, severe MPS I was confirmed in five cases, attenuated MPS I was confirmed in two cases, and undetermined phenotype was reported in one case. We conclude that DMF is an effective and economical method to screen for MPS I and recommend second-tier testing owing to high screen positive rates. Preliminary results of NBS for MPS II and MPS III using DMF are discussed.
Collapse
Affiliation(s)
| | - David S. Millington
- Duke University Hospital Biochemical Genetics Lab, Durham, NC 27709, USA
- Correspondence: ; Tel.: +1-919-448-8221
| |
Collapse
|
28
|
Burton BK, Hickey R, Hitchins L. Newborn Screening for Mucopolysaccharidosis Type II in Illinois: An Update. Int J Neonatal Screen 2020; 6:73. [PMID: 33117908 PMCID: PMC7569764 DOI: 10.3390/ijns6030073] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/12/2020] [Accepted: 09/01/2020] [Indexed: 11/16/2022] Open
Abstract
Mucopolysaccharidosis type II (MPS II, Hunter syndrome) is a rare, progressive multisystemic lysosomal storage disorder with significant morbidity and premature mortality. Infants with MPS II develop signs and symptoms of the disorder in the early years of life, yet diagnostic delays are very common. Enzyme replacement therapy is an effective treatment option. It has been shown to prolong survival and improve or stabilize many somatic manifestations of the disorder. Our initial experience with newborn screening in 162,000 infants was previously reported. Here, we update that experience with the findings in 339,269 infants. Measurement of iduronate-2-sulfatase (I2S) activity was performed on dried blood spot samples submitted for other newborn screening disorders. A positive screen was defined as I2S activity less than or equal to 10% of the daily median. In this series, 28 infants had a positive screening test result, and four other infants had a borderline result. Three positive diagnoses of MPS II were established, and 25 were diagnosed as having I2S pseudodeficiency. The natural history and the clinical features of MPS II make it an ideal target for newborn screening. Newborn screening was effective in identifying affected infants in our population with an acceptable rate of false positive results.
Collapse
Affiliation(s)
- Barbara K Burton
- Department of Pediatrics, Ann & Robert H. Lurie Children's Hospital of Chicago, Chicago, IL 60611, USA; (R.H.); (L.H.)
- Department of Pediatrics, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Rachel Hickey
- Department of Pediatrics, Ann & Robert H. Lurie Children's Hospital of Chicago, Chicago, IL 60611, USA; (R.H.); (L.H.)
| | - Lauren Hitchins
- Department of Pediatrics, Ann & Robert H. Lurie Children's Hospital of Chicago, Chicago, IL 60611, USA; (R.H.); (L.H.)
| |
Collapse
|
29
|
Sabir ES, Lafhal K, Ezoubeiri A, Harkati I, Sbyea S, Aldámiz-Echevarría L, Andrade F, Ait Babram M, Maoulainine FMR, Draiss G, Rada N, Bouskraoui M, Karim A, Fdil N. Usefulness of urinary glycosaminoglycans assay for a mucopolysaccharidosis-specific screening. Pediatr Int 2020; 62:1077-1085. [PMID: 32357280 DOI: 10.1111/ped.14278] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/28/2020] [Revised: 03/17/2020] [Accepted: 04/22/2020] [Indexed: 01/26/2023]
Abstract
BACKGROUND Mucopolysaccharidoses (MPS), a group of inherited metabolic disorders characterized by the accumulation of glycosaminoglycans, can be diagnosed early through newborn screening programs. Establishing newborn screening in Morocco is a challenging task for multiple economic and social reasons. Screening in a Moroccan population using 1,9-dimethylmethylene blue urinary glycosaminoglycan (GAG) assays may allow for an earlier diagnosis of MPS. We studied the feasibility of implementing screening in Moroccan children as an alternative to national newborn screening. We determined the reference ranges for GAGs in the Moroccan population, their stability during transport, the effectiveness of this test as a screening procedure for MPS in patients, and its use as a screening test for MPS in the Imssouane region, where the rate of consanguineous marriage is 38%. METHODS Using dimethylmethylene blue assays, urine samples of 47 MPS patients were analyzed, together with urine samples from healthy controls (n = 368, age ranging from 1 month to 25 years), and from Imssouane region children (n = 350, age ranging from 6 months to 24 month). Precision, linearity, recovery, limits, and stability were tested. RESULTS Urinary GAGs reference values are age and ethnicity dependent. The validation parameters established displayed great precision and accuracy leading to recoveries according to internationally accepted values for bioanalytical methods. Urinary GAGs were stable for a maximum of 7 weeks at 40 °C. Screening of Imssouane children resulted in the detection of a 6-month-old child, diagnosed with MPS I. CONCLUSIONS Our results demonstrate the usefulness of quantifying glycosaminoglycans for early screening of MPS.
Collapse
Affiliation(s)
- Es-Said Sabir
- Metabolics Platform, Biochemistry Laboratory, Faculty of Medicine, Cadi Ayad University, Marrakech, Morocco
| | - Karima Lafhal
- Metabolics Platform, Biochemistry Laboratory, Faculty of Medicine, Cadi Ayad University, Marrakech, Morocco
| | - Aicha Ezoubeiri
- Medical Biology Laboratory - Biochemistry Unit, Ibn Tofail Hospital, CHU Mohamed VI of Marrakech, Marrakech, Morocco
| | - Imane Harkati
- Medical Biology Laboratory - Biochemistry Unit, Ibn Tofail Hospital, CHU Mohamed VI of Marrakech, Marrakech, Morocco
| | - Safia Sbyea
- Metabolics Platform, Biochemistry Laboratory, Faculty of Medicine, Cadi Ayad University, Marrakech, Morocco
| | - Luis Aldámiz-Echevarría
- Metabolomics Platform, Biocruces Bizkaia Health Research Institute, Clinical Linked Group to CIBER of Rare Diseases (CIBERER), Barakaldo, Spain
| | - Fernando Andrade
- Metabolomics Platform, Biocruces Bizkaia Health Research Institute, Clinical Linked Group to CIBER of Rare Diseases (CIBERER), Barakaldo, Spain
| | - Mohammed Ait Babram
- Department of Mathematics, Faculty of Science and Technology, Cadi Ayad University, Marrakech, Morocco
| | - Fadl Mrabih Rabou Maoulainine
- Neonatal Intensive Care Department, Team for Childhood, Health and Development, Marrakesh Faculty of Medicine, Mohammed VI University Hospital and Research, Cadi Ayad University, Marrakech, Morocco
| | - Ghizlane Draiss
- Department of Pediatrics A, Faculty of Medicine and Pharmacy, Mohammed VI University Hospital, Cadi Ayad University, Marrakech, Morocco
| | - Noureddine Rada
- Department of Pediatrics A, Faculty of Medicine and Pharmacy, Mohammed VI University Hospital, Cadi Ayad University, Marrakech, Morocco
| | - Mohammed Bouskraoui
- Department of Pediatrics A, Faculty of Medicine and Pharmacy, Mohammed VI University Hospital, Cadi Ayad University, Marrakech, Morocco
| | - Abdallah Karim
- Laboratory of Coordination Chemistry, Faculty of Sciences Semlalia, Cadi Ayad University, Marrakech, Morocco
| | - Naima Fdil
- Metabolics Platform, Biochemistry Laboratory, Faculty of Medicine, Cadi Ayad University, Marrakech, Morocco
| |
Collapse
|
30
|
Arunkumar N, Langan TJ, Stapleton M, Kubaski F, Mason RW, Singh R, Kobayashi H, Yamaguchi S, Suzuki Y, Orii K, Orii T, Fukao T, Tomatsu S. Newborn screening of mucopolysaccharidoses: past, present, and future. J Hum Genet 2020; 65:557-567. [PMID: 32277174 DOI: 10.1038/s10038-020-0744-8] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2020] [Accepted: 03/03/2020] [Indexed: 11/09/2022]
Abstract
Mucopolysaccharidoses (MPS) are a subtype of lysosomal storage disorders (LSDs) characterized by the deficiency of the enzyme involved in the breakdown of glycosaminoglycans (GAGs). Mucopolysaccharidosis type I (MPS I, Hurler Syndrome) was endorsed by the U.S. Secretary of the Department of Health and Human Services for universal newborn screening (NBS) in February 2016. Its endorsement exemplifies the need to enhance the accuracy of diagnostic testing for disorders that are considered for NBS. The progression of MPS disorders typically incudes irreversible CNS involvement, severe bone dysplasia, and cardiac and respiratory issues. Patients with MPS have a significantly decreased quality of life if untreated and require timely diagnosis and management for optimal outcomes. NBS provides the opportunity to diagnose and initiate treatment plans for MPS patients as early as possible. Most newborns with MPS are asymptomatic at birth; therefore, it is crucial to have biomarkers that can be identified in the newborn. At present, there are tiered methods and different instrumentation available for this purpose. The screening of quick, cost-effective, sensitive, and specific biomarkers in patients with MPS at birth is important. Rapid newborn diagnosis enables treatments to maximize therapeutic efficacy and to introduce immune tolerance during the neonatal period. Currently, newborn screening for MPS I and II has been implemented and/or in pilot testing in several countries. In this review article, historical aspects of NBS for MPS and the prospect of newborn screening for MPS are described, including the potential tiers of screening.
Collapse
Affiliation(s)
- Nivethitha Arunkumar
- Nemours/Alfred I. duPont Hospital for Children, Wilmington, DE, USA.,Department of Health Sciences, University of Delaware, Newark, DE, USA
| | - Thomas J Langan
- Departments of Neurology Pediatrics, and Physiology and Biophysics, State University of New York at Buffalo, Buffalo, NY, USA
| | - Molly Stapleton
- Nemours/Alfred I. duPont Hospital for Children, Wilmington, DE, USA.,Department of Biological Sciences, University of Delaware, Newark, DE, USA
| | - Francyne Kubaski
- Medical Genetics Service, HCPA, Porto Alegre, Brazil.,Department of Genetics and Molecular Biology-PPGBM, UFRGS, Porto Alegre, Brazil.,INAGEMP, Porto Alegre, Brazil
| | - Robert W Mason
- Nemours/Alfred I. duPont Hospital for Children, Wilmington, DE, USA.,Department of Biological Sciences, University of Delaware, Newark, DE, USA
| | | | - Hironori Kobayashi
- Department of Pediatrics, Shimane University Faculty of Medicine, Shimane, Japan
| | - Seiji Yamaguchi
- Department of Pediatrics, Shimane University Faculty of Medicine, Shimane, Japan
| | - Yasuyuki Suzuki
- Medical Education Development Center, Gifu University, Gifu, Japan
| | - Kenji Orii
- Department of Pediatrics, Graduate School of Medicine, Gifu University, Gifu, Japan
| | - Tadao Orii
- Department of Pediatrics, Graduate School of Medicine, Gifu University, Gifu, Japan
| | - Toshiyuki Fukao
- Department of Pediatrics, Graduate School of Medicine, Gifu University, Gifu, Japan
| | - Shunji Tomatsu
- Nemours/Alfred I. duPont Hospital for Children, Wilmington, DE, USA. .,Department of Biological Sciences, University of Delaware, Newark, DE, USA. .,Department of Pediatrics, Shimane University Faculty of Medicine, Shimane, Japan. .,Department of Pediatrics, Graduate School of Medicine, Gifu University, Gifu, Japan. .,Department of Pediatrics, Thomas Jefferson University, Philadelphia, PA, USA.
| |
Collapse
|
31
|
Kiem Hao T, Diem Chi NT, Hong Duc NT, Kim Hoa NT. A case study of three patients with mucopolysaccharidoses in Hue Central Hospital. SAGE Open Med Case Rep 2020; 8:2050313X20938245. [PMID: 32647582 PMCID: PMC7325546 DOI: 10.1177/2050313x20938245] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2020] [Accepted: 06/03/2020] [Indexed: 11/16/2022] Open
Abstract
Mucopolysaccharidosis is a group of rare metabolic disorders characterized by a deficiency of enzymes in the degradation of glycosaminoglycans. The incomplete degradation process leads to the accumulation of glycosaminoglycans in lysosomes of various tissues, which interferes with cell function. We report three cases that were classified as Hurler-Mucopolysaccharidosis I, Morquio-Mucopolysaccharidosis IV A, and Maroteaux-Lamy-Mucopolysaccharidosis VI. Clinical presentations of these cases vary, depending on each type of enzyme defect. All the patients appeared healthy at birth, and symptoms appear at around 1 or 2 years. Clinical features, radiological findings, and especially enzyme assays have allowed us to establish a definitive diagnosis in these cases. These cases highlight that abnormal clinical symptoms, such as growth failure, coarse facial features, and joint problems, are key points for further investigation relating to mucopolysaccharidosis disease. However, in low- and middle-income countries, it is difficult to have a definitive diagnosis of one of the mucopolysaccharidoses due to lacking enzyme assays.
Collapse
Affiliation(s)
- Tran Kiem Hao
- Pediatric Center, Hue Central Hospital, Hue City, Vietnam
| | | | | | | |
Collapse
|
32
|
Sanders KA, Gavrilov DK, Oglesbee D, Raymond KM, Tortorelli S, Hopwood JJ, Lorey F, Majumdar R, Kroll CA, McDonald AM, Lacey JM, Turgeon CT, Tucker JN, Tang H, Currier R, Isaya G, Rinaldo P, Matern D. A Comparative Effectiveness Study of Newborn Screening Methods for Four Lysosomal Storage Disorders. Int J Neonatal Screen 2020; 6:44. [PMID: 32802993 PMCID: PMC7423013 DOI: 10.3390/ijns6020044] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/04/2020] [Accepted: 05/27/2020] [Indexed: 01/13/2023] Open
Abstract
Newborn screening for one or more lysosomal disorders has been implemented in several US states, Japan and Taiwan by multiplexed enzyme assays using either tandem mass spectrometry or digital microfluidics. Another multiplex assay making use of immunocapture technology has also been proposed. To investigate the potential variability in performance of these analytical approaches, we implemented three high-throughput screening assays for the simultaneous screening for four lysosomal disorders: Fabry disease, Gaucher disease, mucopolysaccharidosis type I, and Pompe disease. These assays were tested in a prospective comparative effectiveness study using nearly 100,000 residual newborn dried blood spot specimens. In addition, 2nd tier enzyme assays and confirmatory molecular genetic testing were employed. Post-analytical interpretive tools were created using the software Collaborative Laboratory Integrated Reports (CLIR) to determine its ability to improve the performance of each assay vs. the traditional result interpretation based on analyte-specific reference ranges and cutoffs. This study showed that all three platforms have high sensitivity, and the application of CLIR tools markedly improves the performance of each platform while reducing the need for 2nd tier testing by 66% to 95%. Moreover, the addition of disease-specific biochemical 2nd tier tests ensures the lowest false positive rates and the highest positive predictive values for any platform.
Collapse
Affiliation(s)
- Karen A. Sanders
- Biochemical Genetics Laboratory, Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN 55905, USA; (K.A.S.); (D.K.G.); (D.O.); (K.M.R.); (S.T.); (R.M.); (C.A.K.); (A.M.M.); (J.M.L.); (C.T.T.); (P.R.)
| | - Dimitar K. Gavrilov
- Biochemical Genetics Laboratory, Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN 55905, USA; (K.A.S.); (D.K.G.); (D.O.); (K.M.R.); (S.T.); (R.M.); (C.A.K.); (A.M.M.); (J.M.L.); (C.T.T.); (P.R.)
- Department of Clinical Genomics, Mayo Clinic, Rochester, MN 55905, USA
| | - Devin Oglesbee
- Biochemical Genetics Laboratory, Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN 55905, USA; (K.A.S.); (D.K.G.); (D.O.); (K.M.R.); (S.T.); (R.M.); (C.A.K.); (A.M.M.); (J.M.L.); (C.T.T.); (P.R.)
- Department of Clinical Genomics, Mayo Clinic, Rochester, MN 55905, USA
| | - Kimiyo M. Raymond
- Biochemical Genetics Laboratory, Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN 55905, USA; (K.A.S.); (D.K.G.); (D.O.); (K.M.R.); (S.T.); (R.M.); (C.A.K.); (A.M.M.); (J.M.L.); (C.T.T.); (P.R.)
- Department of Clinical Genomics, Mayo Clinic, Rochester, MN 55905, USA
| | - Silvia Tortorelli
- Biochemical Genetics Laboratory, Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN 55905, USA; (K.A.S.); (D.K.G.); (D.O.); (K.M.R.); (S.T.); (R.M.); (C.A.K.); (A.M.M.); (J.M.L.); (C.T.T.); (P.R.)
- Department of Clinical Genomics, Mayo Clinic, Rochester, MN 55905, USA
| | - John J. Hopwood
- Lysosomal Diseases Research Unit, South Australian Health and Medical Research Institute, Adelaide 5000, Australia; (J.J.H.); (J.N.T.)
| | - Fred Lorey
- Genetic Disease Screening Program, California Department of Public Health, Richmond, CA 94804, USA; (F.L.); (H.T.); (R.C.)
| | - Ramanath Majumdar
- Biochemical Genetics Laboratory, Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN 55905, USA; (K.A.S.); (D.K.G.); (D.O.); (K.M.R.); (S.T.); (R.M.); (C.A.K.); (A.M.M.); (J.M.L.); (C.T.T.); (P.R.)
| | - Charles A. Kroll
- Biochemical Genetics Laboratory, Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN 55905, USA; (K.A.S.); (D.K.G.); (D.O.); (K.M.R.); (S.T.); (R.M.); (C.A.K.); (A.M.M.); (J.M.L.); (C.T.T.); (P.R.)
| | - Amber M. McDonald
- Biochemical Genetics Laboratory, Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN 55905, USA; (K.A.S.); (D.K.G.); (D.O.); (K.M.R.); (S.T.); (R.M.); (C.A.K.); (A.M.M.); (J.M.L.); (C.T.T.); (P.R.)
| | - Jean M. Lacey
- Biochemical Genetics Laboratory, Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN 55905, USA; (K.A.S.); (D.K.G.); (D.O.); (K.M.R.); (S.T.); (R.M.); (C.A.K.); (A.M.M.); (J.M.L.); (C.T.T.); (P.R.)
| | - Coleman T. Turgeon
- Biochemical Genetics Laboratory, Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN 55905, USA; (K.A.S.); (D.K.G.); (D.O.); (K.M.R.); (S.T.); (R.M.); (C.A.K.); (A.M.M.); (J.M.L.); (C.T.T.); (P.R.)
| | - Justin N. Tucker
- Lysosomal Diseases Research Unit, South Australian Health and Medical Research Institute, Adelaide 5000, Australia; (J.J.H.); (J.N.T.)
| | - Hao Tang
- Genetic Disease Screening Program, California Department of Public Health, Richmond, CA 94804, USA; (F.L.); (H.T.); (R.C.)
| | - Robert Currier
- Genetic Disease Screening Program, California Department of Public Health, Richmond, CA 94804, USA; (F.L.); (H.T.); (R.C.)
- Department of Pediatrics, University of California, San Francisco, CA 94143, USA
| | - Grazia Isaya
- Department of Pediatric and Adolescent Medicine, Mayo Clinic, Rochester, MN 55905, USA;
| | - Piero Rinaldo
- Biochemical Genetics Laboratory, Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN 55905, USA; (K.A.S.); (D.K.G.); (D.O.); (K.M.R.); (S.T.); (R.M.); (C.A.K.); (A.M.M.); (J.M.L.); (C.T.T.); (P.R.)
- Department of Clinical Genomics, Mayo Clinic, Rochester, MN 55905, USA
- Department of Pediatric and Adolescent Medicine, Mayo Clinic, Rochester, MN 55905, USA;
| | - Dietrich Matern
- Biochemical Genetics Laboratory, Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN 55905, USA; (K.A.S.); (D.K.G.); (D.O.); (K.M.R.); (S.T.); (R.M.); (C.A.K.); (A.M.M.); (J.M.L.); (C.T.T.); (P.R.)
- Department of Clinical Genomics, Mayo Clinic, Rochester, MN 55905, USA
- Department of Pediatric and Adolescent Medicine, Mayo Clinic, Rochester, MN 55905, USA;
| |
Collapse
|
33
|
Singh R, Chopra S, Graham C, Langer M, Ng R, Ullal AJ, Pamula VK. Emerging Approaches for Fluorescence-Based Newborn Screening of Mucopolysaccharidoses. Diagnostics (Basel) 2020; 10:E294. [PMID: 32403245 PMCID: PMC7277946 DOI: 10.3390/diagnostics10050294] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2020] [Revised: 05/06/2020] [Accepted: 05/07/2020] [Indexed: 11/23/2022] Open
Abstract
Interest in newborn screening for mucopolysaccharidoses (MPS) is growing, due in part to ongoing efforts to develop new therapies for these disorders and new screening assays to identify increased risk for the individual MPSs on the basis of deficiency in the cognate enzyme. Existing tests for MPSs utilize either fluorescence or mass spectrometry detection methods to measure biomarkers of disease (e.g., enzyme function or glycosaminoglycans) using either urine or dried blood spot (DBS) samples. There are currently two approaches to fluorescence-based enzyme function assays from DBS: (1) manual reaction mixing, incubation, and termination followed by detection on a microtiter plate reader; and (2) miniaturized automation of these same assay steps using digital microfluidics technology. This article describes the origins of laboratory assays for enzyme activity measurement, the maturation and clinical application of fluorescent enzyme assays for MPS newborn screening, and considerations for future expansion of the technology.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Vamsee K. Pamula
- Baebies, Inc., P.O. Box 14403, Durham, NC 27709, USA; (R.S.); (S.C.); (C.G.); (M.L.); (R.N.); (A.J.U.)
| |
Collapse
|
34
|
A highly multiplexed biochemical assay for analytes in dried blood spots: application to newborn screening and diagnosis of lysosomal storage disorders and other inborn errors of metabolism. Genet Med 2020; 22:1262-1268. [DOI: 10.1038/s41436-020-0790-9] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2020] [Revised: 03/17/2020] [Accepted: 03/19/2020] [Indexed: 11/09/2022] Open
|
35
|
Mashima R, Okuyama T, Ohira M. Biomarkers for Lysosomal Storage Disorders with an Emphasis on Mass Spectrometry. Int J Mol Sci 2020; 21:ijms21082704. [PMID: 32295281 PMCID: PMC7215887 DOI: 10.3390/ijms21082704] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2020] [Revised: 04/02/2020] [Accepted: 04/04/2020] [Indexed: 01/01/2023] Open
Abstract
Lysosomal storage disorders (LSDs) are characterized by an accumulation of various substances, such as sphingolipids, mucopolysaccharides, and oligosaccharides. The LSD enzymes responsible for the catabolism are active at acidic pH in the lysosomal compartment. In addition to the classically established lysosomal degradation biochemistry, recent data have suggested that lysosome plays a key role in the autophagy where the fusion of autophagosome and lysosome facilitates the degradation of amino acids. A failure in the lysosomal function leads to a variety of manifestations, including neurovisceral disorders. While affected individuals appear to be normal at birth, they gradually become symptomatic in childhood. Biomarkers for each condition have been well-documented and their proper selection helps to perform accurate clinical diagnoses. Based on the natural history of disorders, it is now evident that the existing treatment becomes most effective when initiated during presymptomatic period. Neonatal screening provides such a platform for inborn error of metabolism in general and is now expanding to LSDs as well. These are implemented in some areas and countries, including Taiwan and the U.S. In this short review, we will discuss several issues on some selected biomarkers for LSDs involving Fabry, Niemann–Pick disease type C, mucopolysaccharidosis, and oligosaccharidosis, with a focus on mass spectrometry application to biomarker discovery and detection.
Collapse
|
36
|
Kubaski F, de Oliveira Poswar F, Michelin-Tirelli K, Burin MG, Rojas-Málaga D, Brusius-Facchin AC, Leistner-Segal S, Giugliani R. Diagnosis of Mucopolysaccharidoses. Diagnostics (Basel) 2020; 10:E172. [PMID: 32235807 PMCID: PMC7151013 DOI: 10.3390/diagnostics10030172] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2020] [Revised: 03/16/2020] [Accepted: 03/18/2020] [Indexed: 12/13/2022] Open
Abstract
The mucopolysaccharidoses (MPSs) include 11 different conditions caused by specific enzyme deficiencies in the degradation pathway of glycosaminoglycans (GAGs). Although most MPS types present increased levels of GAGs in tissues, including blood and urine, diagnosis is challenging as specific enzyme assays are needed for the correct diagnosis. Enzyme assays are usually performed in blood, with some samples (as leukocytes) providing a final diagnosis, while others (such as dried blood spots) still being considered as screening methods. The identification of variants in the specific genes that encode each MPS-related enzyme is helpful for diagnosis confirmation (when needed), carrier detection, genetic counseling, prenatal diagnosis (preferably in combination with enzyme assays) and phenotype prediction. Although the usual diagnostic flow in high-risk patients starts with the measurement of urinary GAGs, it continues with specific enzyme assays and is completed with mutation identification; there is a growing trend to have genotype-based investigations performed at the beginning of the investigation. In such cases, confirmation of pathogenicity of the variants identified should be confirmed by measurement of enzyme activity and/or identification and/or quantification of GAG species. As there is a growing number of countries performing newborn screening for MPS diseases, the investigation of a low enzyme activity by the measurement of GAG species concentration and identification of gene mutations in the same DBS sample is recommended before the suspicion of MPS is taken to the family. With specific therapies already available for most MPS patients, and with clinical trials in progress for many conditions, the specific diagnosis of MPS as early as possible is becoming increasingly necessary. In this review, we describe traditional and the most up to date diagnostic methods for mucopolysaccharidoses.
Collapse
Affiliation(s)
- Francyne Kubaski
- Postgraduate Program in Genetics and Molecular Biology, UFRGS, Porto Alegre 91501-970, Brazil; (F.K.); (F.d.O.P.); (D.R.-M.)
- Medical Genetics Service, HCPA, Porto Alegre 90035-903, Brazil; (K.M.-T.); (M.G.B.); (A.C.B.-F.); (S.L.-S.)
- INAGEMP, Porto Alegre 90035-903, Brazil
- Biodiscovery Research Group, Experimental Research Center, HCPA, Porto Alegre 90035-903, Brazil
| | - Fabiano de Oliveira Poswar
- Postgraduate Program in Genetics and Molecular Biology, UFRGS, Porto Alegre 91501-970, Brazil; (F.K.); (F.d.O.P.); (D.R.-M.)
- Medical Genetics Service, HCPA, Porto Alegre 90035-903, Brazil; (K.M.-T.); (M.G.B.); (A.C.B.-F.); (S.L.-S.)
| | - Kristiane Michelin-Tirelli
- Medical Genetics Service, HCPA, Porto Alegre 90035-903, Brazil; (K.M.-T.); (M.G.B.); (A.C.B.-F.); (S.L.-S.)
- Biodiscovery Research Group, Experimental Research Center, HCPA, Porto Alegre 90035-903, Brazil
| | - Maira Graeff Burin
- Medical Genetics Service, HCPA, Porto Alegre 90035-903, Brazil; (K.M.-T.); (M.G.B.); (A.C.B.-F.); (S.L.-S.)
- Biodiscovery Research Group, Experimental Research Center, HCPA, Porto Alegre 90035-903, Brazil
| | - Diana Rojas-Málaga
- Postgraduate Program in Genetics and Molecular Biology, UFRGS, Porto Alegre 91501-970, Brazil; (F.K.); (F.d.O.P.); (D.R.-M.)
- Medical Genetics Service, HCPA, Porto Alegre 90035-903, Brazil; (K.M.-T.); (M.G.B.); (A.C.B.-F.); (S.L.-S.)
| | - Ana Carolina Brusius-Facchin
- Medical Genetics Service, HCPA, Porto Alegre 90035-903, Brazil; (K.M.-T.); (M.G.B.); (A.C.B.-F.); (S.L.-S.)
- INAGEMP, Porto Alegre 90035-903, Brazil
- Biodiscovery Research Group, Experimental Research Center, HCPA, Porto Alegre 90035-903, Brazil
- Postgraduate Program in Medicine, Clinical Sciences, UFRGS, Porto Alegre 90035-003, Brazil
| | - Sandra Leistner-Segal
- Medical Genetics Service, HCPA, Porto Alegre 90035-903, Brazil; (K.M.-T.); (M.G.B.); (A.C.B.-F.); (S.L.-S.)
- INAGEMP, Porto Alegre 90035-903, Brazil
- Biodiscovery Research Group, Experimental Research Center, HCPA, Porto Alegre 90035-903, Brazil
- Postgraduate Program in Medicine, Clinical Sciences, UFRGS, Porto Alegre 90035-003, Brazil
| | - Roberto Giugliani
- Postgraduate Program in Genetics and Molecular Biology, UFRGS, Porto Alegre 91501-970, Brazil; (F.K.); (F.d.O.P.); (D.R.-M.)
- Medical Genetics Service, HCPA, Porto Alegre 90035-903, Brazil; (K.M.-T.); (M.G.B.); (A.C.B.-F.); (S.L.-S.)
- INAGEMP, Porto Alegre 90035-903, Brazil
- Biodiscovery Research Group, Experimental Research Center, HCPA, Porto Alegre 90035-903, Brazil
- Postgraduate Program in Medicine, Clinical Sciences, UFRGS, Porto Alegre 90035-003, Brazil
| |
Collapse
|
37
|
Kubaski F, de Oliveira Poswar F, Michelin-Tirelli K, Matte UDS, Horovitz DD, Barth AL, Baldo G, Vairo F, Giugliani R. Mucopolysaccharidosis Type I. Diagnostics (Basel) 2020; 10:E161. [PMID: 32188113 PMCID: PMC7151028 DOI: 10.3390/diagnostics10030161] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2020] [Revised: 03/09/2020] [Accepted: 03/10/2020] [Indexed: 12/31/2022] Open
Abstract
Mucopolysaccharidosis type I (MPS I) is caused by the deficiency of α-l-iduronidase, leading to the storage of dermatan and heparan sulfate. There is a broad phenotypical spectrum with the presence or absence of neurological impairment. The classical form is known as Hurler syndrome, the intermediate form as Hurler-Scheie, and the most attenuated form is known as Scheie syndrome. Phenotype seems to be largely influenced by genotype. Patients usually develop several somatic symptoms such as abdominal hernias, extensive dermal melanocytosis, thoracolumbar kyphosis odontoid dysplasia, arthropathy, coxa valga and genu valgum, coarse facial features, respiratory and cardiac impairment. The diagnosis is based on the quantification of α-l-iduronidase coupled with glycosaminoglycan analysis and gene sequencing. Guidelines for treatment recommend hematopoietic stem cell transplantation for young Hurler patients (usually at less than 30 months of age). Intravenous enzyme replacement is approved and is the standard of care for attenuated-Hurler-Scheie and Scheie-forms (without cognitive impairment) and for the late-diagnosed severe-Hurler-cases. Intrathecal enzyme replacement therapy is under evaluation, but it seems to be safe and effective. Other therapeutic approaches such as gene therapy, gene editing, stop codon read through, and therapy with small molecules are under development. Newborn screening is now allowing the early identification of MPS I patients, who can then be treated within their first days of life, potentially leading to a dramatic change in the disease's progression. Supportive care is very important to improve quality of life and might include several surgeries throughout the life course.
Collapse
Affiliation(s)
- Francyne Kubaski
- Postgraduate Program in Genetics and Molecular Biology, UFRGS, Porto Alegre 91501970, Brazil; (F.K.); (F.d.O.P.); (U.d.S.M.); (G.B.)
- Medical Genetics Service, HCPA, Porto Alegre 90035903, Brazil;
- INAGEMP, Porto Alegre 90035903, Brazil
- Biodiscovery Research Group, Experimental Research Center, HCPA, Porto Alegre 90035903, Brazil
| | - Fabiano de Oliveira Poswar
- Postgraduate Program in Genetics and Molecular Biology, UFRGS, Porto Alegre 91501970, Brazil; (F.K.); (F.d.O.P.); (U.d.S.M.); (G.B.)
- Medical Genetics Service, HCPA, Porto Alegre 90035903, Brazil;
| | - Kristiane Michelin-Tirelli
- Medical Genetics Service, HCPA, Porto Alegre 90035903, Brazil;
- Biodiscovery Research Group, Experimental Research Center, HCPA, Porto Alegre 90035903, Brazil
| | - Ursula da Silveira Matte
- Postgraduate Program in Genetics and Molecular Biology, UFRGS, Porto Alegre 91501970, Brazil; (F.K.); (F.d.O.P.); (U.d.S.M.); (G.B.)
- INAGEMP, Porto Alegre 90035903, Brazil
- Biodiscovery Research Group, Experimental Research Center, HCPA, Porto Alegre 90035903, Brazil
- Gene Therapy Center, HCPA, Porto Alegre 90035903, Brazil
- Department of Genetics, UFRGS, Porto Alegre 91501970, Brazil
| | - Dafne D. Horovitz
- Medical Genetics Department, National Institute of Women, Children, and Adolescent Health, Oswaldo Cruz Foundation, Rio de Janeiro 21040900, Brazil; (D.D.H.); (A.L.B.)
| | - Anneliese Lopes Barth
- Medical Genetics Department, National Institute of Women, Children, and Adolescent Health, Oswaldo Cruz Foundation, Rio de Janeiro 21040900, Brazil; (D.D.H.); (A.L.B.)
| | - Guilherme Baldo
- Postgraduate Program in Genetics and Molecular Biology, UFRGS, Porto Alegre 91501970, Brazil; (F.K.); (F.d.O.P.); (U.d.S.M.); (G.B.)
- INAGEMP, Porto Alegre 90035903, Brazil
- Biodiscovery Research Group, Experimental Research Center, HCPA, Porto Alegre 90035903, Brazil
- Gene Therapy Center, HCPA, Porto Alegre 90035903, Brazil
- Department of Physiology, UFRGS, Porto Alegre 90050170, Brazil
| | - Filippo Vairo
- Center for Individualized Medicine, Mayo Clinic, Rochester, MN 55905, USA;
- Department of Clinical Genomics, Mayo Clinic, Rochester, MN 55905, USA
| | - Roberto Giugliani
- Postgraduate Program in Genetics and Molecular Biology, UFRGS, Porto Alegre 91501970, Brazil; (F.K.); (F.d.O.P.); (U.d.S.M.); (G.B.)
- Medical Genetics Service, HCPA, Porto Alegre 90035903, Brazil;
- INAGEMP, Porto Alegre 90035903, Brazil
- Biodiscovery Research Group, Experimental Research Center, HCPA, Porto Alegre 90035903, Brazil
- Gene Therapy Center, HCPA, Porto Alegre 90035903, Brazil
- Department of Genetics, UFRGS, Porto Alegre 91501970, Brazil
- Postgraduation Program in Medicine, Clinical Sciences, UFRGS, Porto Alegre 90035003, Brazil
| |
Collapse
|
38
|
Peck DS, Lacey JM, White AL, Pino G, Studinski AL, Fisher R, Ahmad A, Spencer L, Viall S, Shallow N, Siemon A, Hamm JA, Murray BK, Jones KL, Gavrilov D, Oglesbee D, Raymond K, Matern D, Rinaldo P, Tortorelli S. Incorporation of Second-Tier Biomarker Testing Improves the Specificity of Newborn Screening for Mucopolysaccharidosis Type I. Int J Neonatal Screen 2020; 6:10. [PMID: 33073008 PMCID: PMC7422968 DOI: 10.3390/ijns6010010] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/07/2020] [Accepted: 02/05/2020] [Indexed: 11/17/2022] Open
Abstract
Enzyme-based newborn screening for Mucopolysaccharidosis type I (MPS I) has a high false-positive rate due to the prevalence of pseudodeficiency alleles, often resulting in unnecessary and costly follow up. The glycosaminoglycans (GAGs), dermatan sulfate (DS) and heparan sulfate (HS) are both substrates for α-l-iduronidase (IDUA). These GAGs are elevated in patients with MPS I and have been shown to be promising biomarkers for both primary and second-tier testing. Since February 2016, we have measured DS and HS in 1213 specimens submitted on infants at risk for MPS I based on newborn screening. Molecular correlation was available for 157 of the tested cases. Samples from infants with MPS I confirmed by IDUA molecular analysis all had significantly elevated levels of DS and HS compared to those with confirmed pseudodeficiency and/or heterozygosity. Analysis of our testing population and correlation with molecular results identified few discrepant outcomes and uncovered no evidence of false-negative cases. We have demonstrated that blood spot GAGs analysis accurately discriminates between patients with confirmed MPS I and false-positive cases due to pseudodeficiency or heterozygosity and increases the specificity of newborn screening for MPS I.
Collapse
Affiliation(s)
- Dawn S Peck
- Biochemical Genetics Laboratory, Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN 55905, USA; (J.M.L.); (A.L.W.); (G.P.); (A.L.S.); (D.G.); (D.O.); (K.R.); (D.M.); (P.R.)
| | - Jean M Lacey
- Biochemical Genetics Laboratory, Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN 55905, USA; (J.M.L.); (A.L.W.); (G.P.); (A.L.S.); (D.G.); (D.O.); (K.R.); (D.M.); (P.R.)
| | - Amy L White
- Biochemical Genetics Laboratory, Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN 55905, USA; (J.M.L.); (A.L.W.); (G.P.); (A.L.S.); (D.G.); (D.O.); (K.R.); (D.M.); (P.R.)
| | - Gisele Pino
- Biochemical Genetics Laboratory, Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN 55905, USA; (J.M.L.); (A.L.W.); (G.P.); (A.L.S.); (D.G.); (D.O.); (K.R.); (D.M.); (P.R.)
| | - April L Studinski
- Biochemical Genetics Laboratory, Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN 55905, USA; (J.M.L.); (A.L.W.); (G.P.); (A.L.S.); (D.G.); (D.O.); (K.R.); (D.M.); (P.R.)
| | - Rachel Fisher
- Division of Pediatric Genetics, Metabolism and Genomic Medicine, Department of Pediatrics, University of Michigan, Ann Arbor, MI 48109, USA; (R.F.); (A.A.)
| | - Ayesha Ahmad
- Division of Pediatric Genetics, Metabolism and Genomic Medicine, Department of Pediatrics, University of Michigan, Ann Arbor, MI 48109, USA; (R.F.); (A.A.)
| | - Linda Spencer
- Division of Genetic, Genomic and Metabolic Disorders, Children's Hospital of Michigan, Detroit, MI 48201, USA;
| | - Sarah Viall
- Rare Disease Institute, Children's National Health System, Washington, DC 20010, USA;
| | - Natalie Shallow
- Division of Medical Genetics and Genomic Medicine, Monroe Carell Jr. Children's Hospital at Vanderbilt, Nashville, TN 37232, USA;
| | - Amy Siemon
- Division of Genetic and Genomic Medicine, Nationwide Children's Hospital, Columbus, OH 43205, USA;
| | - J Austin Hamm
- Pediatric Genetics, East Tennessee Children's Hospital, Knoxville, TN 37916, USA;
| | - Brianna K Murray
- Division of Medical Genetics and Metabolism, Children's Hospital of the King's Daughters, Norfolk, VA 23507, USA; (B.K.M.); (K.L.J.)
| | - Kelly L Jones
- Division of Medical Genetics and Metabolism, Children's Hospital of the King's Daughters, Norfolk, VA 23507, USA; (B.K.M.); (K.L.J.)
| | - Dimitar Gavrilov
- Biochemical Genetics Laboratory, Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN 55905, USA; (J.M.L.); (A.L.W.); (G.P.); (A.L.S.); (D.G.); (D.O.); (K.R.); (D.M.); (P.R.)
| | - Devin Oglesbee
- Biochemical Genetics Laboratory, Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN 55905, USA; (J.M.L.); (A.L.W.); (G.P.); (A.L.S.); (D.G.); (D.O.); (K.R.); (D.M.); (P.R.)
| | - Kimiyo Raymond
- Biochemical Genetics Laboratory, Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN 55905, USA; (J.M.L.); (A.L.W.); (G.P.); (A.L.S.); (D.G.); (D.O.); (K.R.); (D.M.); (P.R.)
| | - Dietrich Matern
- Biochemical Genetics Laboratory, Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN 55905, USA; (J.M.L.); (A.L.W.); (G.P.); (A.L.S.); (D.G.); (D.O.); (K.R.); (D.M.); (P.R.)
| | - Piero Rinaldo
- Biochemical Genetics Laboratory, Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN 55905, USA; (J.M.L.); (A.L.W.); (G.P.); (A.L.S.); (D.G.); (D.O.); (K.R.); (D.M.); (P.R.)
| | - Silvia Tortorelli
- Biochemical Genetics Laboratory, Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN 55905, USA; (J.M.L.); (A.L.W.); (G.P.); (A.L.S.); (D.G.); (D.O.); (K.R.); (D.M.); (P.R.)
| |
Collapse
|
39
|
D’Avanzo F, Rigon L, Zanetti A, Tomanin R. Mucopolysaccharidosis Type II: One Hundred Years of Research, Diagnosis, and Treatment. Int J Mol Sci 2020; 21:E1258. [PMID: 32070051 PMCID: PMC7072947 DOI: 10.3390/ijms21041258] [Citation(s) in RCA: 79] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2020] [Revised: 02/10/2020] [Accepted: 02/11/2020] [Indexed: 12/11/2022] Open
Abstract
Mucopolysaccharidosis type II (MPS II, Hunter syndrome) was first described by Dr. Charles Hunter in 1917. Since then, about one hundred years have passed and Hunter syndrome, although at first neglected for a few decades and afterwards mistaken for a long time for the similar disorder Hurler syndrome, has been clearly distinguished as a specific disease since 1978, when the distinct genetic causes of the two disorders were finally identified. MPS II is a rare genetic disorder, recently described as presenting an incidence rate ranging from 0.38 to 1.09 per 100,000 live male births, and it is the only X-linked-inherited mucopolysaccharidosis. The complex disease is due to a deficit of the lysosomal hydrolase iduronate 2-sulphatase, which is a crucial enzyme in the stepwise degradation of heparan and dermatan sulphate. This contributes to a heavy clinical phenotype involving most organ-systems, including the brain, in at least two-thirds of cases. In this review, we will summarize the history of the disease during this century through clinical and laboratory evaluations that allowed its definition, its correct diagnosis, a partial comprehension of its pathogenesis, and the proposition of therapeutic protocols. We will also highlight the main open issues related to the possible inclusion of MPS II in newborn screenings, the comprehension of brain pathogenesis, and treatment of the neurological compartment.
Collapse
Affiliation(s)
- Francesca D’Avanzo
- Laboratory of Diagnosis and Therapy of Lysosomal Disorders, Department of Women’s and Children ‘s Health, University of Padova, Via Giustiniani 3, 35128 Padova, Italy; (F.D.); (A.Z.)
- Fondazione Istituto di Ricerca Pediatrica “Città della Speranza”, Corso Stati Uniti 4, 35127 Padova, Italy;
| | - Laura Rigon
- Fondazione Istituto di Ricerca Pediatrica “Città della Speranza”, Corso Stati Uniti 4, 35127 Padova, Italy;
- Molecular Developmental Biology, Life & Medical Science Institute (LIMES), University of Bonn, 53115 Bonn, Germany
| | - Alessandra Zanetti
- Laboratory of Diagnosis and Therapy of Lysosomal Disorders, Department of Women’s and Children ‘s Health, University of Padova, Via Giustiniani 3, 35128 Padova, Italy; (F.D.); (A.Z.)
- Fondazione Istituto di Ricerca Pediatrica “Città della Speranza”, Corso Stati Uniti 4, 35127 Padova, Italy;
| | - Rosella Tomanin
- Laboratory of Diagnosis and Therapy of Lysosomal Disorders, Department of Women’s and Children ‘s Health, University of Padova, Via Giustiniani 3, 35128 Padova, Italy; (F.D.); (A.Z.)
- Fondazione Istituto di Ricerca Pediatrica “Città della Speranza”, Corso Stati Uniti 4, 35127 Padova, Italy;
| |
Collapse
|
40
|
Lin HY, Chuang CK, Lee CL, Chen MR, Sung KT, Lin SM, Hou CJY, Niu DM, Chang TM, Hung CL, Lin SP. Cardiac Evaluation using Two-Dimensional Speckle-Tracking Echocardiography and Conventional Echocardiography in Taiwanese Patients with Mucopolysaccharidoses. Diagnostics (Basel) 2020; 10:diagnostics10020062. [PMID: 31979324 PMCID: PMC7168914 DOI: 10.3390/diagnostics10020062] [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: 11/29/2019] [Revised: 01/12/2020] [Accepted: 01/21/2020] [Indexed: 12/19/2022] Open
Abstract
Background: Mucopolysaccharidoses (MPSs) are a group of rare inherited metabolic disorders that can damage various organs, including the heart. Cardiac abnormalities have been observed in patients with all MPS types, with the most documented abnormalities being cardiac valvular regurgitation and stenosis, valvular thickening, and hypertrophic cardiomyopathy. Methods: Cardiac features of 53 Taiwanese patients with MPS (31 men and 22 women; age range 1.1–34.9 years; seven with MPS I, 16 with MPS II, nine with MPS III, 14 with MPS IVA, and seven with MPS VI) were evaluated using two-dimensional speckle-tracking echocardiography and conventional echocardiography. Results: The mean z scores of the global longitudinal strain (GLS), left ventricular mass index (LVMI), interventricular septum diameter in diastole (IVSd), left ventricular posterior wall diameter in diastole (LVPWd), and aortic diameter of the 53 patients with MPS were 1.71, 0.35, 1.66, 1.03, and 3.15, respectively. Furthermore, z scores >2 were identified in 45%, 13%, 40%, 13%, and 70% of the GLS, LVMI, IVSd, LVPWd, and aortic diameter, respectively. The most severe GLS was observed in those with MPS VI, followed by in those with MPS II and MPS I. The GLS z score was positively correlated with the LVMI z score (p < 0.01). Moreover, diastolic dysfunction (reversed ratio between early and late (atrial) ventricular filling velocity (E/A ratio < 1)) was identified in 12 patients (23%). Ejection and shortening fractions were abnormal in four (8%) and seven (13%) patients, respectively. Mitral regurgitation (MR) (92%) was the most common valvular heart disease, followed by aortic regurgitation (AR) (57%), mitral stenosis (MS) (21%), and aortic stenosis (AS) (15%). The z scores of the GLS and LVMI and severity scores of the MS, MR, AS, and AR were all positively correlated with increasing age (p < 0.05). Twenty patients (38%) had a left ventricular remodeling pattern. Conclusions: The most significant left ventricular myocardial deformation, hypertrophy and valvular heart disease were observed in the patients with MPS VI, II, and I, followed by those with MPS IV; in contrast, patients with MPS III had the mildest manifestations. Cardiac abnormalities in patients with MPS worsened with increasing age in accordance with the progressive nature of this disease.
Collapse
Affiliation(s)
- Hsiang-Yu Lin
- Department of Medicine, MacKay Medical College, New Taipei City 25245, Taiwan; (H.-Y.L.); (M.-R.C.); (K.-T.S.); (S.-M.L.)
- Department of Pediatrics, MacKay Memorial Hospital, Taipei 10449, Taiwan
- Department of Medical Research, MacKay Memorial Hospital, New Taipei City 25160, Taiwan;
- MacKay Junior College of Medicine, Nursing and Management, Taipei 11260, Taiwan
- Department of Medical Research, China Medical University Hospital, China Medical University, Taichung 40402, Taiwan
- Institute of Biomedical Sciences, MacKay Medical College, New Taipei City 25245, Taiwan
| | - Chih-Kuang Chuang
- Department of Medical Research, MacKay Memorial Hospital, New Taipei City 25160, Taiwan;
- College of Medicine, Fu-Jen Catholic University, Taipei 24205, Taiwan
| | - Chung-Lin Lee
- Department of Pediatrics, MacKay Memorial Hospital, Hsinchu 30071, Taiwan;
- Institute of Clinical Medicine, National Yang-Ming University, Taipei 11221, Taiwan;
| | - Ming-Ren Chen
- Department of Medicine, MacKay Medical College, New Taipei City 25245, Taiwan; (H.-Y.L.); (M.-R.C.); (K.-T.S.); (S.-M.L.)
- Department of Pediatrics, MacKay Memorial Hospital, Taipei 10449, Taiwan
- MacKay Junior College of Medicine, Nursing and Management, Taipei 11260, Taiwan
| | - Kuo-Tzu Sung
- Department of Medicine, MacKay Medical College, New Taipei City 25245, Taiwan; (H.-Y.L.); (M.-R.C.); (K.-T.S.); (S.-M.L.)
- Division of Cardiology, Department of Internal Medicine, MacKay Memorial Hospital, Taipei 10449, Taiwan;
| | - Shan-Miao Lin
- Department of Medicine, MacKay Medical College, New Taipei City 25245, Taiwan; (H.-Y.L.); (M.-R.C.); (K.-T.S.); (S.-M.L.)
- Department of Pediatrics, MacKay Memorial Hospital, Taipei 10449, Taiwan
- MacKay Junior College of Medicine, Nursing and Management, Taipei 11260, Taiwan
| | - Charles Jia-Yin Hou
- Division of Cardiology, Department of Internal Medicine, MacKay Memorial Hospital, Taipei 10449, Taiwan;
| | - Dau-Ming Niu
- Institute of Clinical Medicine, National Yang-Ming University, Taipei 11221, Taiwan;
- Department of Pediatrics, Taipei Veterans General Hospital, Taipei 11217, Taiwan
| | - Tung-Ming Chang
- Department of Pediatric Neurology, Changhua Christian Children’s Hospital, Changhua 500, Taiwan;
- School of Medicine, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
| | - Chung-Lieh Hung
- Department of Medicine, MacKay Medical College, New Taipei City 25245, Taiwan; (H.-Y.L.); (M.-R.C.); (K.-T.S.); (S.-M.L.)
- Department of Medical Research, MacKay Memorial Hospital, New Taipei City 25160, Taiwan;
- Division of Cardiology, Department of Internal Medicine, MacKay Memorial Hospital, Taipei 10449, Taiwan;
- Correspondence: (C.-L.H.); (S.-P.L.); Tel.: +886-2-2543-3535 (ext. 3090) (S.-P.L.); Fax: +886-2-2543-3642 (S.-P.L.)
| | - Shuan-Pei Lin
- Department of Medicine, MacKay Medical College, New Taipei City 25245, Taiwan; (H.-Y.L.); (M.-R.C.); (K.-T.S.); (S.-M.L.)
- Department of Pediatrics, MacKay Memorial Hospital, Taipei 10449, Taiwan
- Department of Medical Research, MacKay Memorial Hospital, New Taipei City 25160, Taiwan;
- Department of Infant and Child Care, National Taipei University of Nursing and Health Sciences, Taipei 11219, Taiwan
- Correspondence: (C.-L.H.); (S.-P.L.); Tel.: +886-2-2543-3535 (ext. 3090) (S.-P.L.); Fax: +886-2-2543-3642 (S.-P.L.)
| |
Collapse
|
41
|
Scott CR, Elliott S, Hong X, Huang JY, Kumar AB, Yi F, Pendem N, Chennamaneni NK, Gelb MH. Newborn Screening for Mucopolysaccharidoses: Results of a Pilot Study with 100 000 Dried Blood Spots. J Pediatr 2020; 216:204-207. [PMID: 31732130 PMCID: PMC7159818 DOI: 10.1016/j.jpeds.2019.09.036] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/30/2019] [Revised: 08/06/2019] [Accepted: 09/13/2019] [Indexed: 12/20/2022]
Abstract
OBJECTIVE To test, in a newborn screening (NBS) laboratory, the performance of liquid chromatography-tandem mass spectrometry (LC-MS/MS) to assay 5 enzymatic activities in dried blood spots (DBS) for NBS of 5 lysosomal storage diseases (mucopolysaccharidosis [MPS]-II, MPS-IIIB, MPS-IVA, MPS-VI, and MPS-VII). STUDY DESIGN Three mm punches from de-identified DBS were obtained from the Washington NBS laboratory and submitted to the 5-plex LC-MS/MS assay. Screen cut-offs were established by analyzing the enzymatic activity in patients confirmed to have the MPS disorder. DNA sequencing of the relevant gene was performed on a second DBS punch for all samples with enzyme activity below 10% of the mean daily activity. RESULTS (1) For MPS-II, 18 below cut-off samples, 1 pathogenic genotype, and 2 "high risk" genotypes; (2) For MPS-IIIB, no below cut-off samples; (3) For MPS-IVA, 8 below cut-off samples, 4 non-pathogenic genotypes, 4 genotypes unobtainable; (4) For MPS-VI, 4 below cut-off samples and no high-risk genotypes; (5) For MPS-VII, 1 below cut-off sample confirmed by genotype and clinical report to be affected. CONCLUSIONS These results establish that the number of initial screen positive samples is low and manageable. Thus, population newborn screening for these conditions is feasible in a state newborn screening laboratory.
Collapse
Affiliation(s)
| | - Susan Elliott
- Department of Pediatrics, University of Washington, Seattle. WA
| | - Xinying Hong
- Department of Chemistry, University of Washington, Seattle, WA
| | - Jie-Yu Huang
- Department of Pediatrics, University of Washington, Seattle. WA
| | - Arun Babu Kumar
- Department of Chemistry, University of Washington, Seattle, WA
| | - Fan Yi
- Department of Chemistry, University of Washington, Seattle, WA
| | - Nagendar Pendem
- Department of Chemistry, University of Washington, Seattle, WA
| | | | - Michael H. Gelb
- Department of Chemistry, University of Washington, Seattle, WA
| |
Collapse
|
42
|
Muñoz G, García-Seisdedos D, Ciubotariu C, Piris-Villaespesa M, Gandía M, Martín-Moro F, Gutiérrez-Solana LG, Morado M, López-Jiménez J, Sánchez-Herranz A, Villarrubia J, Del Castillo FJ. Early detection of lysosomal diseases by screening of cases of idiopathic splenomegaly and/or thrombocytopenia with a next-generation sequencing gene panel. JIMD Rep 2019; 51:53-61. [PMID: 32071839 PMCID: PMC7012743 DOI: 10.1002/jmd2.12078] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/01/2019] [Revised: 09/06/2019] [Accepted: 09/12/2019] [Indexed: 02/02/2023] Open
Abstract
Lysosomal diseases (LD) are a group of about 70 rare hereditary disorders (combined incidence 1:5000) in which diverse lysosomal functions are impaired, impacting multiple organs and systems. The first clinical signs and symptoms are usually unspecific and shared by hundreds of other disorders. Diagnosis of LD traditionally relies on performing specific enzymatic assays, if available, upon clinical suspicion of the disorder. However, the combination of the insidious onset of LD and the lack of awareness on these rare diseases among medical personnel results in undesirable diagnostic delays, with unchecked disease progression, appearance of complications and a worsened prognosis. We tested the usefulness of a next‐generation sequencing‐based gene panel for quick, early detection of LD among cases of idiopathic splenomegaly and/or thrombocytopenia, two of the earliest clinical signs observed in most LD. Our 73‐gene panel interrogated 28 genes for LD, 1 biomarker and 44 genes underlying non‐LD differential diagnoses. Among 38 unrelated patients, we elucidated eight cases (21%), five with LD (GM1 gangliosidosis, Sanfilippo disease A and B, Niemann‐Pick disease B, Gaucher disease) and three with non‐LD conditions. Interestingly, we identified three LD patients harboring pathogenic mutations in two LD genes each, which may result in unusual disease presentations and impact treatment. Turnaround time for panel screening and genetic validation was 1 month. Our results underline the usefulness of resequencing gene panels for quick and cost‐effective screening of LDs and disorders sharing with them early clinical signs.
Collapse
Affiliation(s)
- Gloria Muñoz
- UCA de Genómica Traslacional Hospital Universitario Ramón y Cajal, IRYCIS Madrid Spain
| | | | - Crina Ciubotariu
- UCA de Genómica Traslacional Hospital Universitario Ramón y Cajal, IRYCIS Madrid Spain
| | | | - Marta Gandía
- UCA de Genómica Traslacional Hospital Universitario Ramón y Cajal, IRYCIS Madrid Spain
| | - Fernando Martín-Moro
- Servicio de Hematología Hospital Universitario Ramón y Cajal, IRYCIS Madrid Spain
| | - Luis G Gutiérrez-Solana
- Consulta de Neurodegenerativas, Servicio de Neurología Pediátrica Hospital Infantil Universitario Niño Jesús Madrid Spain.,Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER) Madrid Spain
| | - Marta Morado
- Servicio de Hematología Hospital Universitario La Paz Madrid Spain
| | - Javier López-Jiménez
- Servicio de Hematología Hospital Universitario Ramón y Cajal, IRYCIS Madrid Spain
| | | | - Jesús Villarrubia
- UCA de Genómica Traslacional Hospital Universitario Ramón y Cajal, IRYCIS Madrid Spain.,Servicio de Hematología Hospital Universitario Ramón y Cajal, IRYCIS Madrid Spain
| | - Francisco J Del Castillo
- UCA de Genómica Traslacional Hospital Universitario Ramón y Cajal, IRYCIS Madrid Spain.,Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER) Madrid Spain.,Servicio de Genética Hospital Universitario Ramón y Cajal, IRYCIS Madrid Spain
| |
Collapse
|
43
|
Menkovic I, Marchand AS, Boutin M, Auray-Blais C. Neonatal Mass Urine Screening Approach for Early Detection of Mucopolysaccharidoses by UPLC-MS/MS. Diagnostics (Basel) 2019; 9:diagnostics9040195. [PMID: 31752121 PMCID: PMC6963508 DOI: 10.3390/diagnostics9040195] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2019] [Revised: 11/13/2019] [Accepted: 11/14/2019] [Indexed: 11/16/2022] Open
Abstract
Mucopolysaccharidoses (MPSs) are lysosomal storage disorders caused by deficiencies of enzymes involved in the catabolism of glycosaminoglycans (GAGs). Various treatments such as enzyme replacement therapy and/or hematopoietic stem cell transplant are available for MPSs. Early initiation of treatment improves the outcome and delays the onset of symptoms, highlighting the need for newborn screening for MPSs. The main objective of this project was to devise and validate a multiplex urine filter paper method for GAG analysis using a tandem mass spectrometry (MS/MS) approach to screen newborns for MPSs. Eluted urine samples from 21-day-old newborns were evaporated and a methanolysis reaction was performed. Samples were resuspended and analyzed using a UPLC-MS/MS system. A one-minute chromatographic method allowed the absolute quantification of heparan sulfate (HS), dermatan sulfate (DS), and creatinine. Method validation revealed high precision (< 9% relative standard deviation) and accuracy (< 7% bias) for all analytes. The reference values normalized to creatinine obtained by the analysis of five hundred 21-day-old newborn urine samples were 34.6 +/-6.2 mg/mmol of creatinine and 17.3 +/-3.9 mg/mmol of creatinine for HS and DS, respectively. We present a rapid and efficient method for populational newborn urine screening using MS/MS, which could also be applied to high-risk screening.
Collapse
|
44
|
Burton BK, Hoganson GE, Fleischer J, Grange DK, Braddock SR, Hickey R, Hitchins L, Groepper D, Christensen KM, Kirby A, Moody C, Shryock H, Ashbaugh L, Shao R, Basheeruddin K. Population-Based Newborn Screening for Mucopolysaccharidosis Type II in Illinois: The First Year Experience. J Pediatr 2019; 214:165-167.e1. [PMID: 31477379 DOI: 10.1016/j.jpeds.2019.07.053] [Citation(s) in RCA: 13] [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: 05/24/2019] [Revised: 06/28/2019] [Accepted: 07/23/2019] [Indexed: 01/24/2023]
Abstract
OBJECTIVES To assess the outcome of population-based newborn screening for mucopolysaccharidosis type II (MPS II) during the first year of screening in Illinois. STUDY DESIGN Tandem mass spectrometry was used to measure iduronate-2-sulfatase (I2S) activity in dried blood spot specimens obtained from 162 000 infant samples sent to the Newborn Screening Laboratory of the Illinois Department of Public Health in Chicago. RESULTS One case of MPS II and 14 infants with pseudodeficiency for I2S were identified. CONCLUSIONS Newborn screening for MPS II by measurement of I2S enzyme activity was successfully integrated into the statewide newborn screening program in Illinois.
Collapse
Affiliation(s)
- Barbara K Burton
- Ann and Robert H. Lurie Children's Hospital of Chicago, Chicago, IL; Northwestern University Feinberg School of Medicine, Chicago, IL.
| | | | | | | | - Stephen R Braddock
- Saint Louis University, St. Louis, MO; SSM Health Cardinal Glennon Children's Medical Center, St. Louis, MO
| | - Rachel Hickey
- Ann and Robert H. Lurie Children's Hospital of Chicago, Chicago, IL
| | - Lauren Hitchins
- Ann and Robert H. Lurie Children's Hospital of Chicago, Chicago, IL; Northwestern University Feinberg School of Medicine, Chicago, IL
| | | | - Katherine M Christensen
- Saint Louis University, St. Louis, MO; SSM Health Cardinal Glennon Children's Medical Center, St. Louis, MO
| | - Amelia Kirby
- Saint Louis University, St. Louis, MO; SSM Health Cardinal Glennon Children's Medical Center, St. Louis, MO
| | - Conny Moody
- Office of Health Promotion, Illinois Department of Public Health, Springfield, IL
| | - Heather Shryock
- Office of Health Promotion, Illinois Department of Public Health, Springfield, IL
| | - Laura Ashbaugh
- Office of Health Promotion, Illinois Department of Public Health, Springfield, IL
| | - Rong Shao
- Newborn Screening Laboratory, Illinois Department of Public Health, Chicago, IL
| | - Khaja Basheeruddin
- Newborn Screening Laboratory, Illinois Department of Public Health, Chicago, IL
| |
Collapse
|
45
|
Abstract
Mucopolysaccharidoses (MPS) are inborn errors of metabolism produced by a deficiency of one of the enzymes involved in the degradation of glycosaminoglycans (GAGs). Although taken separately, each type is rare. As a group, MPS are relatively frequent, with an overall estimated incidence of around 1 in 20,000-25,000 births. Development of therapeutic options for MPS, including hematopoietic stem cell transplantation (HSCT) and enzyme replacement therapy (ERT), has modified the natural history of many MPS types. In spite of the improvement in some tissues and organs, significant challenges remain unsolved, including blood-brain barrier (BBB) penetration and treatment of lesions in avascular cartilage, heart valves, and corneas. Newer approaches, such as intrathecal ERT, ERT with fusion proteins to cross the BBB, gene therapy, substrate reduction therapy (SRT), chaperone therapy, and some combination of these strategies may provide better outcomes for MPS patients in the near future. As early diagnosis and early treatment are imperative to improve therapeutic efficacy, the inclusion of MPS in newborn screening programs should enhance the potential impact of treatment in reducing the morbidity associated with MPS diseases. In this review, we evaluate available treatments, including ERT and HSCT, and future treatments, such as gene therapy, SRT, and chaperone therapy, and describe the advantages and disadvantages. We also assess the current clinical endpoints and biomarkers used in clinical trials.
Collapse
|
46
|
Lin HY, Lee CL, Lo YT, Tu RY, Chang YH, Chang CY, Chiu PC, Chang TM, Tsai WH, Niu DM, Chuang CK, Lin SP. An At-Risk Population Screening Program for Mucopolysaccharidoses by Measuring Urinary Glycosaminoglycans in Taiwan. Diagnostics (Basel) 2019; 9:diagnostics9040140. [PMID: 31590383 PMCID: PMC6963841 DOI: 10.3390/diagnostics9040140] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2019] [Revised: 10/02/2019] [Accepted: 10/02/2019] [Indexed: 01/13/2023] Open
Abstract
Background: The mucopolysaccharidoses (MPSs) are a group of rare lysosomal storage disorders characterized by the accumulation of glycosaminoglycans (GAGs) and which eventually cause progressive damage to various tissues and organs. We developed a feasible MPS screening algorithm and established a cross-specialty collaboration platform between medical geneticists and other medical specialists based on at-risk criteria to allow for an earlier confirmative diagnosis of MPS. Methods: Children (<19 years of age) with clinical signs and symptoms compatible with MPS were prospectively enrolled from pediatric clinics between July 2013 and June 2018. Urine samples were collected for a non-specific total GAG analysis using the dimethylmethylene blue (DMB) spectrophotometric method, and the quantitation of three urinary GAGs (dermatan sulfate (DS), heparan sulfate (HS), and keratan sulfate (KS)) was performed by liquid chromatography/tandem mass spectrometry (LC-MS/MS). The subjects with elevated urinary GAG levels were recalled for leukocyte enzyme activity assay and genetic testing for confirmation. Results: Among 153 subjects enrolled in this study, 13 had a confirmative diagnosis of MPS (age range, 0.6 to 10.9 years—three with MPS I, four with MPS II, five with MPS IIIB, and one with MPS IVA). The major signs and symptoms with regards to different systems recorded by pediatricians at the time of the decision to test for MPS were the musculoskeletal system (55%), followed by the neurological system (45%) and coarse facial features (39%). For these 13 patients, the median age at the diagnosis of MPS was 2.9 years. The false negative rate of urinary DMB ratio using the dye-based method for these 13 patients was 31%, including one MPS I, two MPS IIIB, and one MPS IVA. However, there were no false negative results with urinary DS, HS and KS using the MS/MS-based method. Conclusions: We established an at-risk population screening program for MPS by measuring urinary GAG fractionation biomarkers using the LC-MS/MS method. The program included medical geneticists and other medical specialists to increase awareness and enable an early diagnosis by detecting MPS at the initial onset of clinical symptoms.
Collapse
Affiliation(s)
- Hsiang-Yu Lin
- Department of Medicine, MacKay Medical College, New Taipei City 252, Taiwan.
- Department of Pediatrics, MacKay Memorial Hospital, Taipei 100, Taiwan.
- Department of Medical Research, MacKay Memorial Hospital, Taipei 100, Taiwan.
- MacKay Junior College of Medicine, Nursing and Management, Taipei 100, Taiwan.
- Department of Medical Research, China Medical University Hospital, China Medical University, Taichung 400, Taiwan.
- Institute of Biomedical Sciences, MacKay Medical College, New Taipei City 252, Taiwan.
| | - Chung-Lin Lee
- Department of Pediatrics, MacKay Memorial Hospital, Hsinchu 300, Taiwan.
- Institute of Clinical Medicine, National Yang-Ming University, Taipei 100, Taiwan.
| | - Yun-Ting Lo
- Department of Laboratory Medicine, MacKay Memorial Hospital, Taipei 100, Taiwan.
| | - Ru-Yi Tu
- Department of Medical Research, MacKay Memorial Hospital, Taipei 100, Taiwan.
| | - Ya-Hui Chang
- Department of Pediatrics, MacKay Memorial Hospital, Taipei 100, Taiwan.
| | - Chia-Ying Chang
- Department of Pediatrics, MacKay Memorial Hospital, Hsinchu 300, Taiwan.
| | - Pao Chin Chiu
- Department of Pediatrics, Kaohsiung Veterans General Hospital, Kaohsiung 800, Taiwan.
| | - Tung-Ming Chang
- Department of Pediatric Neurology, Changhua Christian Children's Hospital, Changhua 500, Taiwan.
- Department of Biological Science and Technology, College of Biological Science and Technology, National Chiao Tung University, Hsinchu 300, Taiwan.
| | - Wen-Hui Tsai
- Department of Pediatrics, Chi Mei Medical Center, Tainan 700, Taiwan.
| | - Dau-Ming Niu
- Institute of Clinical Medicine, National Yang-Ming University, Taipei 100, Taiwan.
- Department of Pediatrics, Taipei Veterans General Hospital, Taipei 100, Taiwan.
| | - Chih-Kuang Chuang
- Department of Medical Research, MacKay Memorial Hospital, Taipei 100, Taiwan.
- College of Medicine, Fu-Jen Catholic University, Taipei 100, Taiwan.
| | - Shuan-Pei Lin
- Department of Medicine, MacKay Medical College, New Taipei City 252, Taiwan.
- Department of Pediatrics, MacKay Memorial Hospital, Taipei 100, Taiwan.
- Department of Medical Research, MacKay Memorial Hospital, Taipei 100, Taiwan.
- Department of Infant and Child Care, National Taipei University of Nursing and Health Sciences, Taipei 100, Taiwan.
| |
Collapse
|
47
|
Clarke LA, Giugliani R, Guffon N, Jones SA, Keenan HA, Munoz-Rojas MV, Okuyama T, Viskochil D, Whitley CB, Wijburg FA, Muenzer J. Genotype-phenotype relationships in mucopolysaccharidosis type I (MPS I): Insights from the International MPS I Registry. Clin Genet 2019; 96:281-289. [PMID: 31194252 PMCID: PMC6852151 DOI: 10.1111/cge.13583] [Citation(s) in RCA: 47] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2019] [Revised: 06/06/2019] [Accepted: 06/12/2019] [Indexed: 01/22/2023]
Abstract
Mucopolysaccharidosis type I (MPS I) is a rare autosomal recessive disorder resulting from pathogenic variants in the α-L-iduronidase (IDUA) gene. Clinical phenotypes range from severe (Hurler syndrome) to attenuated (Hurler-Scheie and Scheie syndromes) and vary in age of onset, severity, and rate of progression. Defining the phenotype at diagnosis is essential for disease management. To date, no systematic analysis of genotype-phenotype correlation in large MPS I cohorts have been performed. Understanding genotype-phenotype is critical now that newborn screening for MPS I is being implemented. Data from 538 patients from the MPS I Registry (380 severe, 158 attenuated) who had 2 IDUA alleles identified were examined. In the 1076 alleles identified, 148 pathogenic variants were reported; of those, 75 were unique. Of the 538 genotypes, 147 (27%) were unique; 40% of patients with attenuated and 22% of patients with severe MPS I had unique genotypes. About 67.6% of severe patients had genotypes where both variants identified are predicted to severely disrupt protein/gene function and 96.1% of attenuated patients had at least one missense or intronic variant. This dataset illustrates a close genotype/phenotype correlation in MPS I but the presence of unique IDUA missense variants remains a challenge for disease prediction.
Collapse
Affiliation(s)
- Lorne A Clarke
- Department of Medical Genetics, B.C. Children's Hospital Research Institute, University of British Columbia, Vancouver, British Columbia, Canada
| | - Roberto Giugliani
- Department of Genetics, Federal University of Rio Grande do Sul and Medical Genetics Service, Hospital de Clínicas de Porto Alegre, Porto Alegre, Brazil
| | - Nathalie Guffon
- Centre de Référence des Maladies Héréditaires du Métabolisme, Hôpital Femme Mère Enfant, Bron Cedex, France
| | - Simon A Jones
- Manchester Centre for Genomic Medicine, Manchester University NHS Trust, Manchester, UK
| | | | | | - Torayuki Okuyama
- Department of Clinical Laboratory Medicine, National Center for Child Health and Development, Tokyo, Japan
| | | | - Chester B Whitley
- Department of Pediatrics, University of Minnesota, Minneapolis, Minnesota.,Experimental and Clinical Pharmacology, University of Minnesota, Minneapolis, Minnesota
| | - Frits A Wijburg
- Department of Pediatrics, Academic Medical Center, Amsterdam, The Netherlands
| | - Joseph Muenzer
- University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| |
Collapse
|
48
|
Lin HY, Chen MR, Lin SM, Hung CL, Niu DM, Chang TM, Chuang CK, Lin SP. Cardiac characteristics and natural progression in Taiwanese patients with mucopolysaccharidosis III. Orphanet J Rare Dis 2019; 14:140. [PMID: 31196149 PMCID: PMC6567572 DOI: 10.1186/s13023-019-1112-7] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2019] [Accepted: 06/04/2019] [Indexed: 11/22/2022] Open
Abstract
BACKGROUND Mucopolysaccharidosis type III (MPS III), or Sanfilippo syndrome, is caused by a deficiency in one of the four enzymes involved in the lysosomal degradation of heparan sulfate. Cardiac abnormalities have been observed in patients with all types of MPS except MPS IX, however few studies have focused on cardiac alterations in patients with MPS III. METHODS We reviewed medical records, echocardiograms, and electrocardiograms of 26 Taiwanese patients with MPS III (five with IIIA, 20 with IIIB, and one with IIIC; 14 males and 12 females; median age, 7.4 years; age range, 1.8-26.5 years). The relationships between age and each echocardiographic parameter were analyzed. RESULTS Echocardiographic examinations (n = 26) revealed that 10 patients (38%) had valvular heart disease. Four (15%) and eight (31%) patients had valvular stenosis or regurgitation, respectively. The most prevalent cardiac valve abnormality was mitral regurgitation (31%), followed by aortic regurgitation (19%). However, most of the cases of valvular heart disease were mild. Three (12%), five (19%) and five (19%) patients had mitral valve prolapse, a thickened interventricular septum, and asymmetric septal hypertrophy, respectively. The severity of aortic regurgitation and the existence of valvular heart disease, aortic valve abnormalities and valvular stenosis were all positively correlated with increasing age (p < 0.05). Z scores > 2 were identified in 0, 38, 8, and 27% of left ventricular mass index, interventricular septal end-diastolic dimension, left ventricular posterior wall end-diastolic dimension, and aortic diameter, respectively. Electrocardiograms in 11 patients revealed the presence of sinus arrhythmia (n = 3), sinus bradycardia (n = 2), and sinus tachycardia (n = 1). Six patients with MPS IIIB had follow-up echocardiographic data at 1.9-18.1 years to compare with the baseline data, which showed some patients had increased thickness of the interventricular septum, as well as more patients had valvular abnormalities at follow-up. CONCLUSIONS Cardiac involvement in MPS III is less common and milder compared with other types of MPS. The existence of valvular heart disease, aortic valve abnormalities and valvular stenosis in the patients worsened with increasing age, reinforcing the concept of the progressive nature of this disease.
Collapse
Affiliation(s)
- Hsiang-Yu Lin
- Department of Medicine, Mackay Medical College, New Taipei City, Taiwan
- Department of Pediatrics, Mackay Memorial Hospital, No.92, Sec. 2, Chung-Shan North Road, Taipei, 10449, Taiwan
- Department of Medical Research, Mackay Memorial Hospital, 92 Chung-Shan N. Rd., Sec. 2, Taipei, 10449, Taiwan
- Mackay Junior College of Medicine, Nursing and Management, Taipei, Taiwan
- Department of Medical Research, China Medical University Hospital, China Medical University, Taichung, Taiwan
- Institute of Biomedical Sciences, Mackay Medical College, New Taipei City, Taiwan
| | - Ming-Ren Chen
- Department of Medicine, Mackay Medical College, New Taipei City, Taiwan
- Department of Pediatrics, Mackay Memorial Hospital, No.92, Sec. 2, Chung-Shan North Road, Taipei, 10449, Taiwan
- Mackay Junior College of Medicine, Nursing and Management, Taipei, Taiwan
| | - Shan-Miao Lin
- Department of Medicine, Mackay Medical College, New Taipei City, Taiwan
- Department of Pediatrics, Mackay Memorial Hospital, No.92, Sec. 2, Chung-Shan North Road, Taipei, 10449, Taiwan
- Mackay Junior College of Medicine, Nursing and Management, Taipei, Taiwan
| | - Chung-Lieh Hung
- Department of Medicine, Mackay Medical College, New Taipei City, Taiwan
- Division of Cardiology, Department of Internal Medicine, Mackay Memorial Hospital, Taipei, Taiwan
| | - Dau-Ming Niu
- Department of Pediatrics, Taipei Veterans General Hospital, Taipei, 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
| | - Chih-Kuang Chuang
- Department of Medical Research, Mackay Memorial Hospital, 92 Chung-Shan N. Rd., Sec. 2, Taipei, 10449, Taiwan.
- College of Medicine, Fu-Jen Catholic University, Taipei, Taiwan.
| | - Shuan-Pei Lin
- Department of Medicine, Mackay Medical College, New Taipei City, Taiwan.
- Department of Pediatrics, Mackay Memorial Hospital, No.92, Sec. 2, Chung-Shan North Road, Taipei, 10449, Taiwan.
- Department of Medical Research, Mackay Memorial Hospital, 92 Chung-Shan N. Rd., Sec. 2, Taipei, 10449, Taiwan.
- Department of Infant and Child Care, National Taipei University of Nursing and Health Sciences, Taipei, Taiwan.
| |
Collapse
|
49
|
Newborn Screening for Lysosomal Storage Disorders: Methodologies for Measurement of Enzymatic Activities in Dried Blood Spots. Int J Neonatal Screen 2019; 5:1. [PMID: 30957052 PMCID: PMC6448570 DOI: 10.3390/ijns5010001] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
All worldwide newborn screening (NBS) for lysosomal storage diseases (LSDs) is performed as a first-tier test by measurement of lysosomal enzymatic activities in dried blood spots (DBS). The currently two available methodologies used for measurement of enzymatic activities are tandem mass spectrometry (MS/MS) and digital microfluidics fluorimetry (DMF-F). In this chapter we summarize the workflows for the two platforms. Neither platform is fully automated, but the relative ease of workflow will be dependent upon the specific operation of each newborn screening laboratory on a case-by-case basis. We provide the screen positive rate (the number of below cutoff newborns per 100,000 newborns) from all NBS laboratories worldwide carrying out MS/MS-based NBS of one or more LSDs. The analytical precision of the MS/MS method is higher than that for DMF-F as shown by analysis of a common set of quality control DBS by the Centers for Disease Control and Prevention (CDC). Both the MS/MS and DMF-F platforms enable multiplexing of the LSD enzymes. An advantage of MS/MS over DMF-F is the ability to include assays of enzymatic activities and biomarkers for which no fluorimetric methods exist. Advantages of DMF-F over MS/MS are: 1) Simple to use technology with same-day turn-around time for the lysosomal enzymes with the fastest rates compared to MS/MS requiring overnight analytical runs.; 2) The DMF-F instrumentation, because of its simplicity, requires less maintenance than the MS/MS platform.
Collapse
|