1
|
Scarcella M, Fecarotta S, Alagia M, Barretta F, Uomo F, De Pasquale V, Patel HS, Strisciuglio P, Parenti G, Frisso G, Pavone LM, Ruoppolo M. Digital microfluidic platform for dried blood spot newborn screening of lysosomal storage diseases in Campania region (Italy): Findings from the first year pilot project. Mol Genet Metab 2024:109008. [PMID: 39788860 DOI: 10.1016/j.ymgme.2024.109008] [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: 09/10/2024] [Revised: 12/25/2024] [Accepted: 12/26/2024] [Indexed: 01/12/2025]
Abstract
BACKGROUND Newborn screening (NBS) is a simple, non-invasive test that allows for the early identification of genetic diseases within the first days of a newborn's life. The aim of NBS is to detect potentially fatal or disabling conditions in newborns as early as possible, before the onset of disease symptoms. Early diagnosis enables timely treatments and improves the quality of life for affected patients. RESULTS A pilot project for dried blood spot (DBS) NBS of lysosomal storage diseases (LSDs), including Mucopolysaccharidosis I (MPSI, IDUA α-L-iduronidase deficiency), Pompe disease (GAA α-glucosidase acid deficiency), Gaucher disease (GBA β-glucosidase deficiency) and Fabry disease (GLA α-galactosidase deficiency), was conducted using the digital microfluidic (DMF) technique. DBS were analyzed in a multiplexed assays for the enzymatic activities of four lysosomal enzymes (IDUA, GAA, GBA, GLA), and subjects identified as deficient in any of these enzymes were referred to the clinical reference center for diagnosis confirmation. From June 6th, 2022, to May 12th, 2023, a total of 7650 newborns were analyzed and 1 subject affected by Pompe disease was identified together with two additional subjects, suspected of Pompe and Fabry disease respectively, for whom continued follow-up is mandatory to determine the phenotype. CONCLUSIONS The pilot project for DBS NBS of four LSDs in Campania Region validated the effectiveness of DMF method, established enzymatic activity cut-offs, and identified newborns referred to the clinical center for integrated diagnostics, including genetic analyses. The results suggest that this technique can effectively detect potentially affected newborns, who will require further diagnostic confirmation and clinical follow-up. This diagnostic flow chart provides the opportunity to initiate early treatments and improve LSD patients' life span.
Collapse
Affiliation(s)
- Melania Scarcella
- Department of Molecular Medicine and Medical Biotechnology, Medical School, University of Naples Federico II, 80131 Naples, Italy; CEINGE-Biotecnologie Avanzate Franco Salvatore s.c.ar.l., 80145 Naples, Italy
| | - Simona Fecarotta
- Department of Translational Medical Sciences, Medical School, University of Naples Federico II, 80131 Naples, Italy
| | - Marianna Alagia
- Department of Translational Medical Sciences, Medical School, University of Naples Federico II, 80131 Naples, Italy
| | - Ferdinando Barretta
- Department of Molecular Medicine and Medical Biotechnology, Medical School, University of Naples Federico II, 80131 Naples, Italy; CEINGE-Biotecnologie Avanzate Franco Salvatore s.c.ar.l., 80145 Naples, Italy; DAIMedLab AOU Federico II, 80131 Naples, Italy
| | - Fabiana Uomo
- Department of Molecular Medicine and Medical Biotechnology, Medical School, University of Naples Federico II, 80131 Naples, Italy; CEINGE-Biotecnologie Avanzate Franco Salvatore s.c.ar.l., 80145 Naples, Italy
| | - Valeria De Pasquale
- CEINGE-Biotecnologie Avanzate Franco Salvatore s.c.ar.l., 80145 Naples, Italy; Department of Veterinary Medicine and Animal Productions, University of Naples Federico II, 80137 Naples, Italy
| | | | - Pietro Strisciuglio
- Department of Translational Medical Sciences, Medical School, University of Naples Federico II, 80131 Naples, Italy
| | - Giancarlo Parenti
- Department of Translational Medical Sciences, Medical School, University of Naples Federico II, 80131 Naples, Italy; Telethon Institute of Genetics and Medicine (TIGEM), 80078 Pozzuoli, Italy
| | - Giulia Frisso
- Department of Molecular Medicine and Medical Biotechnology, Medical School, University of Naples Federico II, 80131 Naples, Italy; CEINGE-Biotecnologie Avanzate Franco Salvatore s.c.ar.l., 80145 Naples, Italy
| | - Luigi Michele Pavone
- Department of Molecular Medicine and Medical Biotechnology, Medical School, University of Naples Federico II, 80131 Naples, Italy; CEINGE-Biotecnologie Avanzate Franco Salvatore s.c.ar.l., 80145 Naples, Italy.
| | - Margherita Ruoppolo
- Department of Molecular Medicine and Medical Biotechnology, Medical School, University of Naples Federico II, 80131 Naples, Italy; CEINGE-Biotecnologie Avanzate Franco Salvatore s.c.ar.l., 80145 Naples, Italy.
| |
Collapse
|
2
|
Gragnaniello V, Cazzorla C, Gueraldi D, Loro C, Porcù E, Salviati L, Burlina AP, Burlina AB. Newborn Screening for Acid Sphingomyelinase Deficiency: Prevalence and Genotypic Findings in Italy. Int J Neonatal Screen 2024; 10:79. [PMID: 39728399 DOI: 10.3390/ijns10040079] [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: 11/09/2024] [Revised: 11/26/2024] [Accepted: 12/03/2024] [Indexed: 12/28/2024] Open
Abstract
Acid sphingomyelinase deficiency (ASMD) is a rare lysosomal storage disorder with a broad clinical spectrum. Early diagnosis and initiation of treatment are crucial for improving outcomes, yet the disease often goes undiagnosed due to its rarity and phenotypic heterogeneity. This study aims to evaluate the feasibility and disease incidence of newborn screening (NBS) for ASMD in Italy. Dried blood spot samples from 275,011 newborns were collected between 2015 and 2024 at the Regional Center for Expanded NBS in Padua. Acid sphingomyelinase activity was assayed using tandem mass spectrometry. Deidentified samples with reduced enzyme activity underwent second-tier testing with LysoSM quantification and SMPD1 gene analysis. Two samples were identified with reduced sphingomyelinase activity and elevated LysoSM levels. Both carried two SMPD1 variants, suggesting a diagnosis of ASMD. Molecular findings included novel and previously reported variants, some of uncertain significance. The overall incidence was 1 in 137,506 newborns and the PPV was 100%. This study demonstrates the feasibility of NBS for ASMD in Italy and provides evidence of a higher disease incidence than clinically reported, suggesting ASMD is an underdiagnosed condition. Optimized screening algorithms and second-tier biomarker testing can enhance the accuracy of NBS for ASMD. The long-term follow-up of identified cases is necessary for genotype-phenotype correlation and improving patient management.
Collapse
Affiliation(s)
- Vincenza Gragnaniello
- Division of Inherited Metabolic Diseases, Department of Women's and Children's Health, University Hospital of Padua, 35128 Padua, Italy
- Division of Inherited Metabolic Diseases, Department of Women's and Children's Health, University of Padua, 35128 Padua, Italy
| | - Chiara Cazzorla
- Division of Inherited Metabolic Diseases, Department of Women's and Children's Health, University Hospital of Padua, 35128 Padua, Italy
| | - Daniela Gueraldi
- Division of Inherited Metabolic Diseases, Department of Women's and Children's Health, University Hospital of Padua, 35128 Padua, Italy
| | - Christian Loro
- Division of Inherited Metabolic Diseases, Department of Women's and Children's Health, University Hospital of Padua, 35128 Padua, Italy
| | - Elena Porcù
- Division of Inherited Metabolic Diseases, Department of Women's and Children's Health, University Hospital of Padua, 35128 Padua, Italy
| | - Leonardo Salviati
- Clinical Genetics Unit, Department of Women's and Children's Health, University of Padua, 35128 Padua, Italy
| | | | - Alberto B Burlina
- Division of Inherited Metabolic Diseases, Department of Women's and Children's Health, University Hospital of Padua, 35128 Padua, Italy
- Division of Inherited Metabolic Diseases, Department of Women's and Children's Health, University of Padua, 35128 Padua, Italy
| |
Collapse
|
3
|
Bushnell C, Kernan WN, Sharrief AZ, Chaturvedi S, Cole JW, Cornwell WK, Cosby-Gaither C, Doyle S, Goldstein LB, Lennon O, Levine DA, Love M, Miller E, Nguyen-Huynh M, Rasmussen-Winkler J, Rexrode KM, Rosendale N, Sarma S, Shimbo D, Simpkins AN, Spatz ES, Sun LR, Tangpricha V, Turnage D, Velazquez G, Whelton PK. 2024 Guideline for the Primary Prevention of Stroke: A Guideline From the American Heart Association/American Stroke Association. Stroke 2024; 55:e344-e424. [PMID: 39429201 DOI: 10.1161/str.0000000000000475] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2024]
Abstract
AIM The "2024 Guideline for the Primary Prevention of Stroke" replaces the 2014 "Guidelines for the Primary Prevention of Stroke." This updated guideline is intended to be a resource for clinicians to use to guide various prevention strategies for individuals with no history of stroke. METHODS A comprehensive search for literature published since the 2014 guideline; derived from research involving human participants published in English; and indexed in MEDLINE, PubMed, Cochrane Library, and other selected and relevant databases was conducted between May and November 2023. Other documents on related subject matter previously published by the American Heart Association were also reviewed. STRUCTURE Ischemic and hemorrhagic strokes lead to significant disability but, most important, are preventable. The 2024 primary prevention of stroke guideline provides recommendations based on current evidence for strategies to prevent stroke throughout the life span. These recommendations align with the American Heart Association's Life's Essential 8 for optimizing cardiovascular and brain health, in addition to preventing incident stroke. We also have added sex-specific recommendations for screening and prevention of stroke, which are new compared with the 2014 guideline. Many recommendations for similar risk factor prevention were updated, new topics were reviewed, and recommendations were created when supported by sufficient-quality published data.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | | | - Eliza Miller
- American College of Obstetricians and Gynecologists liaison
| | | | | | | | | | | | | | - Alexis N Simpkins
- American Heart Association Stroke Council Scientific Statement Oversight Committee on Clinical Practice Guideline liaison
| | | | | | | | | | | | | |
Collapse
|
4
|
Kanack AJ, Prodoehl E, Ishihara-Aoki M, Aoki K, Dahms NM. Glycosphingolipids and their impact on platelet activity in a murine model of fabry disease. Sci Rep 2024; 14:29488. [PMID: 39604471 PMCID: PMC11603304 DOI: 10.1038/s41598-024-80633-6] [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: 05/13/2024] [Accepted: 11/21/2024] [Indexed: 11/29/2024] Open
Abstract
Fabry disease is an X-linked lysosomal storage disorder caused by deficiency of the lysosomal enzyme ⍺-galactosidase-A (⍺-Gal A), resulting in widespread accumulation of terminal galactose-containing glycosphingolipids (GSLs) and the impairment of multiple organ systems. Thrombotic events are common in Fabry patients, with strokes and heart attacks being significant contributors to a shortened lifespan in patients of both genders. Previously, we developed an ⍺-Gal A-knockout (KO) murine model that recapitulates most Fabry symptomologies and demonstrated that platelets from KO males become sensitized to agonist-mediated activation. In the current report, we used mass spectrometry, platelet-based assays and histology to define further the mechanisms linking GSL accumulation with thrombotic phenotypes in both sexes. Sera and platelets from ⍺-Gal A-KO females have elevated levels of Fabry-associated GSLs relative to wild-type females, but accumulated less of these GSLs than KO males. Correspondingly, KO females demonstrate a less severe thrombotic phenotypes than KO males. Notably, treatment of platelets from wild-type animals with globotriaosylceramide (Gb3) increased baseline platelet activation and aggregation. In contrast, several control GSLs did not stimulate platelet responses. These data suggest that chronically high concentrations of the Fabry-associated GSL, Gb3, contributes to the prothrombotic phenotypes experienced by Fabry patients by directly stimulating platelet activation.
Collapse
Affiliation(s)
- Adam J Kanack
- Department of Laboratory Medicine and Pathology, Mayo Clinic, 200 1st St. SW, Rochester, MN, 55905, USA.
| | - Eve Prodoehl
- Department of Biochemistry, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI, 53226, USA
| | - Mayumi Ishihara-Aoki
- Translational Metabolomics Shared Resource, Cancer Center, Medical College of Wisconsin, Milwaukee, WI, 53226, USA
| | - Kazuhiro Aoki
- Translational Metabolomics Shared Resource, Cancer Center, Medical College of Wisconsin, Milwaukee, WI, 53226, USA
- Department of Cell Biology, Neurobiology and Anatomy, Medical College of Wisconsin, Milwaukee, WI, 53226, USA
| | - Nancy M Dahms
- Department of Biochemistry, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI, 53226, USA.
| |
Collapse
|
5
|
Herbst ZM, Kubaski F, Pollard L, Basheeruddin K, Burton B, Orsini J, Henderson M, Chakraborty P, Gelb MH. High precision newborn screening for mucopolysaccharidosis type I by enzymatic activity followed by endogenous, non-reducing end glycosaminoglycan analysis. Mol Genet Metab 2024:108612. [PMID: 39645522 DOI: 10.1016/j.ymgme.2024.108612] [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: 07/11/2024] [Revised: 11/14/2024] [Accepted: 11/15/2024] [Indexed: 12/09/2024]
Abstract
Measurement of enzymatic activity in newborn dried blood spots (DBS) is the preferred first-tier method in newborn screening (NBS) for mucopolysaccharidosis (MPS) disorders. However, false positives are observed due mainly to the presence of pseudodeficiencies. Our previous publications on glycosaminoglycan (GAG) biomarker levels in dried blood spots (DBS) for mucopolysaccharidoses demonstrated that second-tier GAG biomarker analysis can dramatically reduce the false positive rate in NBS. In the present study, we extend this approach to the analysis of a large number of false positives for MPS-I obtained from the Illinois, New York, and Tennessee NBS programs and from Greenwood Genetics Center. Results show that GAG levels measured by the Endogenous-Non-Reducing End method (Endogenous-NRE) are in the normal reference range for all samples. In a second study, we analyzed 166 samples that showed below-cutoff MPS-I enzymatic activity level after testing 384,144 newborns in the Ontario, Canada NBS program. Both genotype and Endogenous-NRE GAG levels were determined for all 166 samples. Newborns at high risk for MPS-I based on genotype also showed elevated GAG levels and were clinically confirmed to be symptomatic for MPS-I. All newborns with pseudodeficiency or carrier status by genotyping all showed normal levels of the appropriate GAG biomarker. Samples found to be inconclusive based on one or more variants of unknown significance (VUS) all showed normal GAG biomarker levels and were found to be clinically normal during follow-up. These studies show that the Endogenous-NRE GAG second-tier NBS method is preferred over second-tier DNA analysis for the NBS of MPS-I with minimal false positives.
Collapse
Affiliation(s)
- Zackary M Herbst
- Departments of Chemistry, University of Washington, Seattle, WA 98195, USA; GelbChem, LLC, Seattle, WA 98195, USA
| | - Francyne Kubaski
- Greenwood Genetic Center, Biochemical Genetics Laboratory, Greenwood, SC 29646, USA
| | - Laura Pollard
- Greenwood Genetic Center, Biochemical Genetics Laboratory, Greenwood, SC 29646, USA
| | - Khaja Basheeruddin
- Newborn Screening Laboratory, Illinois Department of Public Health, Chicago, IL 60612, USA
| | - Barbara Burton
- Division of Genetics, Genomics & Metabolism, Ann & Robert H. Lurie Children's Hospital of Chicago and Feinberg School ofMedicine, Northwestern University, Chicago, IL 60611, USA
| | - Joseph Orsini
- David Axelrod Institute, Newborn Screening Program, Wadsworth Center, New York State Department of Health, Albany, NY 12201-2002, USA
| | - Matthew Henderson
- Children's Hospital of Eastern Ontario Research Institute, Ottawa, Canada
| | | | - Michael H Gelb
- Departments of Chemistry, University of Washington, Seattle, WA 98195, USA.
| |
Collapse
|
6
|
Hickey RE, Baker J. Newborn screening for acid sphingomyelinase deficiency in Illinois: A single center's experience. J Inherit Metab Dis 2024; 47:1363-1370. [PMID: 38992987 PMCID: PMC11586602 DOI: 10.1002/jimd.12780] [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: 04/25/2024] [Revised: 06/24/2024] [Accepted: 06/27/2024] [Indexed: 07/13/2024]
Abstract
Acid sphingomyelinase deficiency (ASMD) is a rare lysosomal storage disorder (LSD) caused by reduced activity of the acid sphingomyelinase (ASM) enzyme, which leads to progressive storage of sphingomyelin and related lipids in the body. ASMD is caused by biallelic variants in the SMPD1 gene, which encodes for the ASM enzyme. Current estimates of disease incidence range from 0.4 to 0.6 in 100 000 livebirths, although this is likely an underestimation of the true frequency of the disorder. While there is no cure for ASMD, comprehensive care guidelines and enzyme replacement therapy are available, making an early diagnosis crucial. Newborn screening (NBS) for ASMD is possible through measurement of ASM activity in dried blood spots and offers the opportunity for early diagnosis. In 2015, Illinois (IL) became the first to initiate statewide implementation of NBS for ASMD. This study describes the outcomes of screen-positive patients referred to Ann & Robert H. Lurie Children's Hospital (Lurie). Ten infants were referred for diagnostic evaluation at Lurie, and all 10 infants were classified as confirmed ASMD or at risk for ASMD through a combination of molecular and biochemical testing. Disease incidence was calculated using data from this statewide implementation program and was ~0.79 in 100 000 livebirths. This study demonstrates successful implementation of NBS for ASMD in IL, with high screen specificity and a notable absence of false positive screens.
Collapse
Affiliation(s)
- Rachel E. Hickey
- Ann & Robert H. Lurie Children's Hospital of ChicagoChicagoIllinoisUSA
| | - Joshua Baker
- Ann & Robert H. Lurie Children's Hospital of ChicagoChicagoIllinoisUSA
- Northwestern University Feinberg School of MedicineChicagoIllinoisUSA
| |
Collapse
|
7
|
Ruan J, Yu X, Xu H, Cui W, Zhang K, Liu C, Sun W, Huang X, An L, Zhang Y. Suppressor tRNA in gene therapy. SCIENCE CHINA. LIFE SCIENCES 2024; 67:2120-2131. [PMID: 38926247 DOI: 10.1007/s11427-024-2613-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/18/2024] [Accepted: 05/08/2024] [Indexed: 06/28/2024]
Abstract
Suppressor tRNAs are engineered or naturally occurring transfer RNA molecules that have shown promise in gene therapy for diseases caused by nonsense mutations, which result in premature termination codons (PTCs) in coding sequence, leading to truncated, often nonfunctional proteins. Suppressor tRNAs can recognize and pair with these PTCs, allowing the ribosome to continue translation and produce a full-length protein. This review introduces the mechanism and development of suppressor tRNAs, compares suppressor tRNAs with other readthrough therapies, discusses their potential for clinical therapy, limitations, and obstacles. We also summarize the applications of suppressor tRNAs in both in vitro and in vivo, offering new insights into the research and treatment of nonsense mutation diseases.
Collapse
Affiliation(s)
- Jingjing Ruan
- The Children's Hospital, National Clinical Research Center for Child Health, Zhejiang University School of Medicine, Liangzhu Laboratory, Hangzhou, 310000, China
- Liangzhu Laboratory, Zhejiang University School of Medicine, Hangzhou, 311121, China
| | - Xiaoxiao Yu
- Liangzhu Laboratory, Zhejiang University School of Medicine, Hangzhou, 311121, China
| | - Huixia Xu
- Department of Thoracic and Cardiovascular Surgery, Huaihe Hospital of Henan University, Henan University, Kaifeng, 475000, China
| | - Wenrui Cui
- Translational Medicine Center, Huaihe Hospital of Henan University, Henan University, Kaifeng, 475000, China
| | - Kaiye Zhang
- Eye Center, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310009, China
| | - Chenyang Liu
- Translational Medicine Center, Huaihe Hospital of Henan University, Henan University, Kaifeng, 475000, China
| | - Wenlong Sun
- Liangzhu Laboratory, Zhejiang University School of Medicine, Hangzhou, 311121, China
| | - Xiaodan Huang
- Eye Center, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310009, China
| | - Lei An
- Translational Medicine Center, Huaihe Hospital of Henan University, Henan University, Kaifeng, 475000, China.
| | - Yue Zhang
- The Children's Hospital, National Clinical Research Center for Child Health, Zhejiang University School of Medicine, Liangzhu Laboratory, Hangzhou, 310000, China.
- Liangzhu Laboratory, Zhejiang University School of Medicine, Hangzhou, 311121, China.
- Translational Medicine Center, Huaihe Hospital of Henan University, Henan University, Kaifeng, 475000, China.
| |
Collapse
|
8
|
Erdman ME, Ch S, Mohiuddin A, Al-Kirwi K, Rasper MR, Sokupa S, Low SWY, Skumatz CMB, De Stefano V, Kassem IS, Chaurasia SS. Fabry Disease Rat Model Develops Age- and Sex-Dependent Anterior Segment Ocular Abnormalities. Invest Ophthalmol Vis Sci 2024; 65:14. [PMID: 39110587 PMCID: PMC11314710 DOI: 10.1167/iovs.65.10.14] [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: 04/05/2024] [Accepted: 07/23/2024] [Indexed: 08/11/2024] Open
Abstract
Purpose Fabry disease is an X-linked lysosomal storage disorder that results in multi-systemic renal, cardiovascular, and neuropathological damage, including in the eyes. We evaluated anterior segment ocular abnormalities based on age, sex (male and female), and genotype (wild-type, knockout [KO] male, heterozygous [HET] female, and KO female) in a rat model of Fabry disease. Methods The α-Gal A KO and WT rats were divided into young (6-24 weeks), adult (25-60 weeks), and aged (61+ weeks) groups. Intraocular pressure (IOP) was measured. Eyes were clinically scored for corneal and lens opacity as well as evaluated for corneal epithelial integrity and tear break-up time (TBUT). Anterior chamber depth (ACD) and central corneal thickness (CCT) using anterior segment-optical coherence tomography (AS-OCT). Results The Fabry rats showed an age-dependent increase in IOP, predominantly in the male genotype. TBUT was decreased in both male and female groups with aging. Epithelial integrity was defective in KO males and HET females with age. However, it was highly compromised in KO females irrespective of age. Corneal and lens opacities were severely affected irrespective of sex or genotype in the aging Fabry rats. AS-OCT quantification of CCT and ACD also demonstrated age-dependent increases but were more pronounced in Fabry versus WT genotypes. Conclusions Epithelial integrity, corneal, and lens opacities worsened in Fabry rats, whereas IOP and TBUT changes were age-dependent. Similarly, CCT and ACD were age-related but more pronounced in Fabry rats, providing newer insights into the anterior segment ocular abnormalities with age, sex, and genotype in a rat model of Fabry disease.
Collapse
Affiliation(s)
- Madelyn E. Erdman
- Ocular Immunology & Angiogenesis Lab, Department of Ophthalmology & Visual Sciences, Medical College of Wisconsin, Milwaukee, Wisconsin, United States
| | - Sanjay Ch
- Ocular Immunology & Angiogenesis Lab, Department of Ophthalmology & Visual Sciences, Medical College of Wisconsin, Milwaukee, Wisconsin, United States
| | - Amer Mohiuddin
- Ocular Immunology & Angiogenesis Lab, Department of Ophthalmology & Visual Sciences, Medical College of Wisconsin, Milwaukee, Wisconsin, United States
| | - Khalid Al-Kirwi
- Ocular Immunology & Angiogenesis Lab, Department of Ophthalmology & Visual Sciences, Medical College of Wisconsin, Milwaukee, Wisconsin, United States
| | - Molly R. Rasper
- Ocular Immunology & Angiogenesis Lab, Department of Ophthalmology & Visual Sciences, Medical College of Wisconsin, Milwaukee, Wisconsin, United States
| | - Sibabalo Sokupa
- Ocular Immunology & Angiogenesis Lab, Department of Ophthalmology & Visual Sciences, Medical College of Wisconsin, Milwaukee, Wisconsin, United States
| | - Shermaine W. Y. Low
- Ocular Immunology & Angiogenesis Lab, Department of Ophthalmology & Visual Sciences, Medical College of Wisconsin, Milwaukee, Wisconsin, United States
| | - Christine M. B. Skumatz
- Ocular Immunology & Angiogenesis Lab, Department of Ophthalmology & Visual Sciences, Medical College of Wisconsin, Milwaukee, Wisconsin, United States
| | - Vinicius De Stefano
- Ocular Immunology & Angiogenesis Lab, Department of Ophthalmology & Visual Sciences, Medical College of Wisconsin, Milwaukee, Wisconsin, United States
- Cornea and Refractive Surgery, Froedtert and Medical College of Wisconsin Eye Institute, Milwaukee, Wisconsin, United States
| | - Iris S. Kassem
- Ocular Immunology & Angiogenesis Lab, Department of Ophthalmology & Visual Sciences, Medical College of Wisconsin, Milwaukee, Wisconsin, United States
- Department of Cell Biology, Neurobiology and Anatomy, Medical College of Wisconsin, Milwaukee, Wisconsin, United States
| | - Shyam S. Chaurasia
- Ocular Immunology & Angiogenesis Lab, Department of Ophthalmology & Visual Sciences, Medical College of Wisconsin, Milwaukee, Wisconsin, United States
- Department of Cell Biology, Neurobiology and Anatomy, Medical College of Wisconsin, Milwaukee, Wisconsin, United States
| |
Collapse
|
9
|
Duro G, Anania M, Zizzo C, Francofonte D, Giacalone I, D’Errico A, Marsana EM, Colomba P. Diagnosis of Fabry Disease Using Alpha-Galactosidase A Activity or LysoGb3 in Blood Fails to Identify Up to Two Thirds of Female Patients. Int J Mol Sci 2024; 25:5158. [PMID: 38791200 PMCID: PMC11121337 DOI: 10.3390/ijms25105158] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2024] [Accepted: 05/01/2024] [Indexed: 05/26/2024] Open
Abstract
Anderson-Fabry disease is a lysosomal storage disorder caused by mutations in the GLA gene, which encodes the enzyme α-galactosidase A. The GLA gene is located on the X-chromosome, causing an X-linked pathology: due to lyonization, female patients usually manifest a variable symptomatology, ranging from asymptomatic to severe phenotypes. The confirmation of the clinical diagnosis of Fabry disease, achieved by measuring α-galactosidase A activity, which is usually the first test used, shows differences between male and female patients. This assay is reliable in male patients with causative mutations in the GLA gene, in whom the enzymatic activity is lower than normal values; on the other hand, in female Fabry patients, the enzymatic activity is extremely variable between normal and pathological values. These fluctuations are also found in female patients' blood levels of globotriaosylsphingosine (LysoGb3) for the same reason. In this paper, we present a retrospective study conducted in our laboratories on 827 Fabry patients with causative mutations in the GLA gene. Our results show that 100% of male patients had α-galactosidase A activity below the reference value, while more than 70% of female patients had normal values. It can also be observed that almost half of the female patients with pathogenic mutations in the GLA gene showed normal values of LysoGb3 in blood. Furthermore, in women, blood LysoGb3 values can vary over time, as we show in a clinical case presented in this paper. Both these tests could lead to missed diagnoses of Fabry disease in female patients, so the analysis of the GLA gene represents the main diagnostic test for Fabry disease in women to date.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Paolo Colomba
- Institute for Biomedical Research and Innovation (IRIB), National Research Council (CNR), 90146 Palermo, Italy; (G.D.); (M.A.); (C.Z.); (D.F.); (I.G.); (A.D.); (E.M.M.)
| |
Collapse
|
10
|
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
|
11
|
Pieroni M, Namdar M, Olivotto I, Desnick RJ. Anderson-Fabry disease management: role of the cardiologist. Eur Heart J 2024; 45:1395-1409. [PMID: 38486361 DOI: 10.1093/eurheartj/ehae148] [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: 09/03/2023] [Revised: 02/04/2024] [Accepted: 02/27/2024] [Indexed: 04/22/2024] Open
Abstract
Anderson-Fabry disease (AFD) is a lysosomal storage disorder characterized by glycolipid accumulation in cardiac cells, associated with a peculiar form of hypertrophic cardiomyopathy (HCM). Up to 1% of patients with a diagnosis of HCM indeed have AFD. With the availability of targeted therapies for sarcomeric HCM and its genocopies, a timely differential diagnosis is essential. Specifically, the therapeutic landscape for AFD is rapidly evolving and offers increasingly effective, disease-modifying treatment options. However, diagnosing AFD may be difficult, particularly in the non-classic phenotype with prominent or isolated cardiac involvement and no systemic red flags. For many AFD patients, the clinical journey from initial clinical manifestations to diagnosis and appropriate treatment remains challenging, due to late recognition or utter neglect. Consequently, late initiation of treatment results in an exacerbation of cardiac involvement, representing the main cause of morbidity and mortality, irrespective of gender. Optimal management of AFD patients requires a dedicated multidisciplinary team, in which the cardiologist plays a decisive role, ranging from the differential diagnosis to the prevention of complications and the evaluation of timing for disease-specific therapies. The present review aims to redefine the role of cardiologists across the main decision nodes in contemporary AFD clinical care and drug discovery.
Collapse
Affiliation(s)
- Maurizio Pieroni
- Cardiovascular Department, San Donato Hospital, Via Pietro Nenni 22, 52100 Arezzo, Italy
| | - Mehdi Namdar
- Cardiology Division, Hôpitaux Universitaires de Genève, Geneva, Switzerland
| | - Iacopo Olivotto
- Cardiomyopathy Unit, Careggi Hospital and Meyer Children's Hospital IRCCS, Florence, Italy
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | - Robert J Desnick
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| |
Collapse
|
12
|
Coelho-Ribeiro B, Silva HG, Sampaio-Marques B, Fraga AG, Azevedo O, Pedrosa J, Ludovico P. Inflammation and Exosomes in Fabry Disease Pathogenesis. Cells 2024; 13:654. [PMID: 38667269 PMCID: PMC11049543 DOI: 10.3390/cells13080654] [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: 02/22/2024] [Revised: 04/01/2024] [Accepted: 04/07/2024] [Indexed: 04/28/2024] Open
Abstract
Fabry Disease (FD) is one of the most prevalent lysosomal storage disorders, resulting from mutations in the GLA gene located on the X chromosome. This genetic mutation triggers glo-botriaosylceramide (Gb-3) buildup within lysosomes, ultimately impairing cellular functions. Given the role of lysosomes in immune cell physiology, FD has been suggested to have a profound impact on immunological responses. During the past years, research has been focusing on this topic, and pooled evidence strengthens the hypothesis that Gb-3 accumulation potentiates the production of pro-inflammatory mediators, revealing the existence of an acute inflammatory process in FD that possibly develops to a chronic state due to stimulus persistency. In parallel, extracellular vesicles (EVs) have gained attention due to their function as intercellular communicators. Considering EVs' capacity to convey cargo from parent to distant cells, they emerge as potential inflammatory intermediaries capable of transporting cytokines and other immunomodulatory molecules. In this review, we revisit the evidence underlying the association between FD and altered immune responses and explore the potential of EVs to function as inflammatory vehicles.
Collapse
Affiliation(s)
- Bruna Coelho-Ribeiro
- Life and Health Sciences Research Institute (ICVS), 4710-057 Braga, Portugal; (B.C.-R.); (H.G.S.); (B.S.-M.); (A.G.F.); (J.P.)
- ICVS/3B’s-PT Government Associate Laboratory, 4710-057 Braga/4805-017 Guimarães, Portugal
| | - Helena G. Silva
- Life and Health Sciences Research Institute (ICVS), 4710-057 Braga, Portugal; (B.C.-R.); (H.G.S.); (B.S.-M.); (A.G.F.); (J.P.)
- ICVS/3B’s-PT Government Associate Laboratory, 4710-057 Braga/4805-017 Guimarães, Portugal
| | - Belém Sampaio-Marques
- Life and Health Sciences Research Institute (ICVS), 4710-057 Braga, Portugal; (B.C.-R.); (H.G.S.); (B.S.-M.); (A.G.F.); (J.P.)
- ICVS/3B’s-PT Government Associate Laboratory, 4710-057 Braga/4805-017 Guimarães, Portugal
| | - Alexandra G. Fraga
- Life and Health Sciences Research Institute (ICVS), 4710-057 Braga, Portugal; (B.C.-R.); (H.G.S.); (B.S.-M.); (A.G.F.); (J.P.)
- ICVS/3B’s-PT Government Associate Laboratory, 4710-057 Braga/4805-017 Guimarães, Portugal
| | - Olga Azevedo
- Reference Center on Lysosomal Storage Disorders, Hospital Senhora da Oliveira, 4835-044 Guimarães, Portugal;
| | - Jorge Pedrosa
- Life and Health Sciences Research Institute (ICVS), 4710-057 Braga, Portugal; (B.C.-R.); (H.G.S.); (B.S.-M.); (A.G.F.); (J.P.)
- ICVS/3B’s-PT Government Associate Laboratory, 4710-057 Braga/4805-017 Guimarães, Portugal
| | - Paula Ludovico
- Life and Health Sciences Research Institute (ICVS), 4710-057 Braga, Portugal; (B.C.-R.); (H.G.S.); (B.S.-M.); (A.G.F.); (J.P.)
- ICVS/3B’s-PT Government Associate Laboratory, 4710-057 Braga/4805-017 Guimarães, Portugal
| |
Collapse
|
13
|
Majid H, Verma N, Bhandari S, Gupta S, Nidhi. A Systematic Review on Safety and Efficacy of Migalastat for the treatment of Fabry's Disease. Expert Opin Pharmacother 2024; 25:769-782. [PMID: 38753367 DOI: 10.1080/14656566.2024.2354466] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2024] [Accepted: 05/08/2024] [Indexed: 05/30/2024]
Abstract
INTRODUCTION Fabry's disease (FD) is a genetic lysosomal storage disorder characterized by α-galactosidase A (α-Gal A) lost/reduced activity. We aim to systematically assess the safety and efficacy of Migalastat, an oral pharmacological chaperone, that has been approved for the treatment of FD in patients with amenable mutations. METHODS We conducted literature search following the PRISMA guidelines in major databases up to 4 February 2024, for studies that assessed the clinical outcomes of migalastat in patients with FD. The New Castle Ottawa Scale was used to evaluate the quality of the included studies. RESULTS A total of 2141 records were identified through database searches and register searches, amongst which 26 records were screened, and 12 of these were excluded. The remaining 14 reports were sought for retrieval. The 12 retrieved articles were assessed for eligibility and their quality was assessed after their inclusion. Amongst the included studies, 5 were of high quality, 6 were of medium quality, and 1 was of low quality. CONCLUSION Migalastat showed varied effects on enzyme activity and substrate levels, with gender-specific differences noted in GL-3 substrate activity and eGFR. Overall, it improved cardiac and renal outcomes similarly to enzyme replacement therapy, with a comparable safety profile.
Collapse
Affiliation(s)
- Haya Majid
- Department of Translational and Clinical Research, School of Chemical and Life Sciences, Jamia Hamdard, New Delhi, India
| | - Neharika Verma
- Department of Translational and Clinical Research, School of Chemical and Life Sciences, Jamia Hamdard, New Delhi, India
| | - Shivani Bhandari
- Department of Translational and Clinical Research, School of Chemical and Life Sciences, Jamia Hamdard, New Delhi, India
| | - Sparsh Gupta
- Department of Pharmacology, Vardhman Mahavir Medical College, and Safdarjung Hospital, New Delhi, India
| | - Nidhi
- Department of Translational and Clinical Research, School of Chemical and Life Sciences, Jamia Hamdard, New Delhi, India
| |
Collapse
|
14
|
Millington DS. How mass spectrometry revolutionized newborn screening. J Mass Spectrom Adv Clin Lab 2024; 32:1-10. [PMID: 38333514 PMCID: PMC10847993 DOI: 10.1016/j.jmsacl.2024.01.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2023] [Revised: 01/24/2024] [Accepted: 01/26/2024] [Indexed: 02/10/2024] Open
Abstract
This article offers a personal account of a remarkable journey spanning over 30 years of applied mass spectrometry in a clinical setting. It begins with the author's inspiration from a clinician's story of rescuing a child from near death with a revolutionary therapeutic intervention. Motivated by this experience, the author delved into the field of chemistry and mass spectrometry to solve an analytical challenge. The breakthrough came with the development of the first front-line diagnostic test performed by MS/MS, which focused on analyzing acylcarnitines to detect and diagnose inherited disorders related to fatty acid and branched-chain amino acid catabolism. Building upon this success, the author expanded the application of the method to dried blood spots, incorporating additional analytical components such as essential amino acids. The result was a groundbreaking multiplex assay capable of screening newborns for more than 30 inherited metabolic conditions with just one test. This novel approach laid the foundation for a targeted metabolomics platform that facilitated the identification of new animal models of metabolic disease through screening the offspring of genetically modified adults. The development and utilization of MS/MS with UPLC has led to the creation of new assays for biomarkers of metabolic disease, benefiting both the diagnosis and therapeutic monitoring of these conditions. The article provides compelling examples from the author's laboratory, highlighting the value and vast applications of these methods in the field of metabolic disease research.
Collapse
Affiliation(s)
- David S Millington
- Duke University Medical Center, Department of Pediatrics, Durham, NC, USA
| |
Collapse
|
15
|
Hirachan R, Horman A, Burke D, Heales S. Evaluation, in a highly specialised enzyme laboratory, of a digital microfluidics platform for rapid assessment of lysosomal enzyme activity in dried blood spots. JIMD Rep 2024; 65:124-131. [PMID: 38444576 PMCID: PMC10910220 DOI: 10.1002/jmd2.12413] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/29/2023] [Revised: 01/24/2024] [Accepted: 01/29/2024] [Indexed: 03/07/2024] Open
Abstract
Lysosomal storage disorders (LSDs) are predominantly enzyme deficiencies leading to substrate accumulation, causing progressive damage to multiple organs. To date, a crucial part of diagnosing LSDs is measuring enzymatic activity in leucocytes, plasma, or dried blood spots (DBS). Here, we present results from a proof-of-principle study, evaluating an innovative digital microfluidics (DMF) platform, referred to as SEEKER®, that can measure the activity of the following four lysosomal enzymes from DBS: α-L-iduronidase (IDUA) for mucopolysaccharidosis I (MPS I), acid α-glucosidase (GAA) for Pompe disease, β-glucosidase (GBA) for Gaucher disease, and α-galactosidase A (GLA) for Fabry disease. Over 900 DBS were analysed from newborns, children, and adults. DMF successfully detected known patients with MPS I, Pompe disease, and Gaucher disease, and known males with Fabry disease. This is the first demonstration of this multiplexed DMF platform for identification of patients with LSDs in a specialised diagnostic enzyme laboratory environment. We conclude that this DMF platform is relatively simple, high-throughput, and could be readily accommodated into a specialised laboratory as a first-tier test for MPS I, Pompe disease, and Gaucher disease for all patients, and Fabry disease for male patients only.
Collapse
Affiliation(s)
- Rohit Hirachan
- Chemical PathologyCamelia Botnar Laboratories, Great Ormond Street Hospital for Children NHS Foundation TrustLondonUK
| | - Alistair Horman
- Chemical PathologyCamelia Botnar Laboratories, Great Ormond Street Hospital for Children NHS Foundation TrustLondonUK
| | - Derek Burke
- Chemical PathologyCamelia Botnar Laboratories, Great Ormond Street Hospital for Children NHS Foundation TrustLondonUK
| | - Simon Heales
- Chemical PathologyCamelia Botnar Laboratories, Great Ormond Street Hospital for Children NHS Foundation TrustLondonUK
- Neurometabolic UnitNational Hospital for Neurology and NeurosurgeryLondonUK
| |
Collapse
|
16
|
Gragnaniello V, Cazzorla C, Gueraldi D, Puma A, Loro C, Porcù E, Stornaiuolo M, Miglioranza P, Salviati L, Burlina AP, Burlina AB. Light and Shadows in Newborn Screening for Lysosomal Storage Disorders: Eight Years of Experience in Northeast Italy. Int J Neonatal Screen 2023; 10:3. [PMID: 38248631 PMCID: PMC10801488 DOI: 10.3390/ijns10010003] [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: 11/09/2023] [Revised: 12/13/2023] [Accepted: 12/21/2023] [Indexed: 01/23/2024] Open
Abstract
In the last two decades, the development of high-throughput diagnostic methods and the availability of effective treatments have increased the interest in newborn screening for lysosomal storage disorders. However, long-term follow-up experience is needed to clearly identify risks, benefits and challenges. We report our 8-year experience of screening and follow-up on about 250,000 neonates screened for four lysosomal storage diseases (Pompe disease, mucopolysaccharidosis type I, Fabry disease, Gaucher disease), using the enzyme activity assay by tandem mass spectrometry, and biomarker quantification as a second-tier test. Among the 126 positive newborns (0.051%), 51 infants were confirmed as affected (positive predictive value 40%), with an overall incidence of 1:4874. Of these, three patients with infantile-onset Pompe disease, two with neonatal-onset Gaucher disease and four with mucopolysaccharidosis type I were immediately treated. Furthermore, another four Gaucher disease patients needed treatment in the first years of life. Our study demonstrates the feasibility and effectiveness of newborn screening for lysosomal storage diseases. Early diagnosis and treatment allow the achievement of better patient outcomes. Challenges such as false-positive rates, the diagnosis of variants of uncertain significance or late-onset forms and the lack of treatment for neuronopathic forms, should be addressed.
Collapse
Affiliation(s)
- Vincenza Gragnaniello
- Division of Inherited Metabolic Diseases, Department of Diagnostic Services, University Hospital of Padua, 35128 Padua, Italy; (V.G.); (C.C.); (D.G.); (A.P.); (C.L.); (E.P.); (M.S.)
- Division of Inherited Metabolic Diseases, Department of Women’s and Children’s Health, University of Padua, 35128 Padua, Italy
| | - Chiara Cazzorla
- Division of Inherited Metabolic Diseases, Department of Diagnostic Services, University Hospital of Padua, 35128 Padua, Italy; (V.G.); (C.C.); (D.G.); (A.P.); (C.L.); (E.P.); (M.S.)
| | - Daniela Gueraldi
- Division of Inherited Metabolic Diseases, Department of Diagnostic Services, University Hospital of Padua, 35128 Padua, Italy; (V.G.); (C.C.); (D.G.); (A.P.); (C.L.); (E.P.); (M.S.)
| | - Andrea Puma
- Division of Inherited Metabolic Diseases, Department of Diagnostic Services, University Hospital of Padua, 35128 Padua, Italy; (V.G.); (C.C.); (D.G.); (A.P.); (C.L.); (E.P.); (M.S.)
| | - Christian Loro
- Division of Inherited Metabolic Diseases, Department of Diagnostic Services, University Hospital of Padua, 35128 Padua, Italy; (V.G.); (C.C.); (D.G.); (A.P.); (C.L.); (E.P.); (M.S.)
| | - Elena Porcù
- Division of Inherited Metabolic Diseases, Department of Diagnostic Services, University Hospital of Padua, 35128 Padua, Italy; (V.G.); (C.C.); (D.G.); (A.P.); (C.L.); (E.P.); (M.S.)
| | - Maria Stornaiuolo
- Division of Inherited Metabolic Diseases, Department of Diagnostic Services, University Hospital of Padua, 35128 Padua, Italy; (V.G.); (C.C.); (D.G.); (A.P.); (C.L.); (E.P.); (M.S.)
| | - Paolo Miglioranza
- Division of Inherited Metabolic Diseases, Department of Diagnostic Services, University Hospital of Padua, 35128 Padua, Italy; (V.G.); (C.C.); (D.G.); (A.P.); (C.L.); (E.P.); (M.S.)
| | - Leonardo Salviati
- Clinical Genetics Unit, Department of Women’s and Children’s Health, University of Padua, 35128 Padua, Italy;
| | | | - Alberto B. Burlina
- Division of Inherited Metabolic Diseases, Department of Diagnostic Services, University Hospital of Padua, 35128 Padua, Italy; (V.G.); (C.C.); (D.G.); (A.P.); (C.L.); (E.P.); (M.S.)
- Division of Inherited Metabolic Diseases, Department of Women’s and Children’s Health, University of Padua, 35128 Padua, Italy
| |
Collapse
|
17
|
Fillman T, Matteson J, Tang H, Mathur D, Zahedi R, Sen I, Bishop T, Neogi P, Feuchtbaum L, Olney RS, Sciortino S. First Three Years' Experience of Mucopolysaccharidosis Type-I Newborn Screening in California. J Pediatr 2023; 263:113644. [PMID: 37516270 DOI: 10.1016/j.jpeds.2023.113644] [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: 04/25/2023] [Revised: 06/23/2023] [Accepted: 07/25/2023] [Indexed: 07/31/2023]
Abstract
OBJECTIVE To report on the first 3 years of mucopolysaccharidosis type I (MPS I) newborn screening (NBS) in the large and diverse state of California. STUDY DESIGN The California Genetic Disease Screening Program began universal NBS for MPS I on August 29, 2018. The screening uses a 2-tiered approach: an α-L-iduronidase (IDUA) enzyme activity assay followed by DNA sequencing for variants in the IDUA gene. RESULTS As of August 29, 2021, 1 295 515 California newborns were screened for MPS I. In tier 1 of screening, 329 (0.025%) had an IDUA enzyme measurement below the cutoff and underwent tier-2 IDUA DNA sequencing. After tier 2, 146 (0.011%) newborns were screen positive, all of whom were referred to a metabolic Special Care Center for follow-up. After long-term follow-up, 7 cases were resolved as severe MPS I (Hurler syndrome) and 2 cases as attenuated MPS I for an MPS I birth prevalence of 1/143 946. DNA sequencing identified 107 unique IDUA variants among a total of 524 variants; 65% were known pseudodeficiency alleles, 25% were variants of uncertain significance, and 10% were pathogenic variants. CONCLUSIONS As a result of a 2-tiered NBS approach, 7 newborns diagnosed with Hurler syndrome had received early treatment for MPS I. Continuation of California's long-term follow-up program will be crucial for further understanding the complex genotype-phenotype relationships of MPS I.
Collapse
Affiliation(s)
- Toki Fillman
- Genetic Disease Screening Program, California Department of Public Health, Richmond, CA.
| | - Jamie Matteson
- Genetic Disease Screening Program, California Department of Public Health, Richmond, CA
| | - Hao Tang
- Genetic Disease Screening Program, California Department of Public Health, Richmond, CA
| | - Deepika Mathur
- Genetic Disease Screening Program, California Department of Public Health, Richmond, CA
| | - Rana Zahedi
- Genetic Disease Screening Program, California Department of Public Health, Richmond, CA
| | - Indranil Sen
- Genetic Disease Screening Program, California Department of Public Health, Richmond, CA
| | - Tracey Bishop
- Genetic Disease Screening Program, California Department of Public Health, Richmond, CA
| | - Partha Neogi
- Genetic Disease Screening Program, California Department of Public Health, Richmond, CA
| | - Lisa Feuchtbaum
- Genetic Disease Screening Program, California Department of Public Health, Richmond, CA
| | - Richard S Olney
- Genetic Disease Screening Program, California Department of Public Health, Richmond, CA
| | - Stanley Sciortino
- Genetic Disease Screening Program, California Department of Public Health, Richmond, CA
| |
Collapse
|
18
|
Erdal H, Turgut F. Thiol/disulfide homeostasis as a new oxidative stress marker in patients with Fabry disease. J Investig Med 2023; 71:865-870. [PMID: 37485970 DOI: 10.1177/10815589231191966] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/25/2023]
Abstract
This is the first study to show both dynamic thiol-disulfide balance and oxidative stress levels in patients with Fabry disease (FD). This prospective study consists of 30 FD patients and 30 healthy controls. Thiol and disulfide values of the study groups were evaluated using a new, cost-effective and fully automatic colorimetric method. A total of 60 subjects were included in the study. A statistically significant difference was found between the patient and control groups for native and total thiol levels (p < 0.001). In addition, disulfide levels were significantly higher in FD patients compared with the control group (p < 0.003). Native thiol levels showed significantly negative correlation with lysosomal globotriaosylceramide, total oxidant status (TOS), and oxidative stress index (OSI) levels. In addition, a positive correlation was found between disulfide/natural thiol and disulfide/total thiol ratios and TOS, OSI, and blood urea nitrogen. We found total antioxidant status levels were lower in the patient group compared to the control group, while TOS and OSI levels were higher and were statistically significant. This study highlights for the first time a novel, cost-effective and fully automated measurement of thiol-disulfide levels in patients with FD. Determination of thiol levels can make important contributions to understand the etiopathogenesis and follow-up of the disease in FD patients.
Collapse
Affiliation(s)
- Huseyin Erdal
- Department of Medical Genetics, Faculty of Medicine, Aksaray University, Aksaray, Turkey
| | - Faruk Turgut
- Department of Nephrology, Faculty of Medicine, Hatay Mustafa Kemal University, Hatay, Turkey
| |
Collapse
|
19
|
Bichet DG, Hopkin RJ, Aguiar P, Allam SR, Chien YH, Giugliani R, Kallish S, Kineen S, Lidove O, Niu DM, Olivotto I, Politei J, Rakoski P, Torra R, Tøndel C, Hughes DA. Consensus recommendations for the treatment and management of patients with Fabry disease on migalastat: a modified Delphi study. Front Med (Lausanne) 2023; 10:1220637. [PMID: 37727761 PMCID: PMC10505750 DOI: 10.3389/fmed.2023.1220637] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2023] [Accepted: 07/13/2023] [Indexed: 09/21/2023] Open
Abstract
Objective Fabry disease is a progressive disorder caused by deficiency of the α-galactosidase A enzyme (α-Gal A), leading to multisystemic organ damage with heterogenous clinical presentation. The addition of the oral chaperone therapy migalastat to the available treatment options for Fabry disease is not yet universally reflected in all treatment guidelines. These consensus recommendations are intended to provide guidance for the treatment and monitoring of patients with Fabry disease receiving migalastat. Methods A modified Delphi process was conducted to determine consensus on treatment decisions and monitoring of patients with Fabry disease receiving migalastat. The multidisciplinary panel comprised 14 expert physicians across nine specialties and two patients with Fabry disease. Two rounds of Delphi surveys were completed and recommendations on the use of biomarkers, multidisciplinary monitoring, and treatment decisions were generated based on statements that reached consensus. Results The expert panel reached consensus agreement on 49 of 54 statements, including 16 that reached consensus in round 1. Statements that reached consensus agreement are summarized in recommendations for migalastat treatment and monitoring, including baseline and follow-up assessments and frequency. All patients with Fabry disease and an amenable mutation may initiate migalastat treatment if they have evidence of Fabry-related symptoms and/or organ involvement. Treatment decisions should include holistic assessment of the patient, considering clinical symptoms and organ involvement as well as patient-reported outcomes and patient preference. The reliability of α-Gal A and globotriaosylsphingosine as pharmacodynamic response biomarkers remains unclear. Conclusion These recommendations build on previously published guidelines to highlight the importance of holistic, multidisciplinary monitoring for patients with Fabry disease receiving migalastat, in addition to shared decision-making regarding treatments and monitoring throughout the patient journey. GRAPHICAL ABSTRACT.
Collapse
Affiliation(s)
- Daniel G. Bichet
- Department of Medicine, Pharmacology and Physiology, Hôpital du Sacré-Coeur, University of Montréal, Montreal, QC, Canada
| | - Robert J. Hopkin
- Department of Pediatrics, Division of Human Genetics, University of Cincinnati College of Medicine, and Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, United States
| | - Patrício Aguiar
- Inborn Errors of Metabolism Reference Center, Centro Hospitalar Universitário Lisboa Norte, Lisbon, Portugal
- Faculty of Medicine, Lisbon University, Lisbon, Portugal
| | - Sridhar R. Allam
- Burnett School of Medicine, Texas Christian University, Fort Worth, TX, United States
- Tarrant Nephrology Associates/PPG Health, Fort Worth, TX, United States
| | - Yin-Hsiu Chien
- Department of Medical Genetics, National Taiwan University Hospital, Taipei, Taiwan
- Department of Pediatrics, National Taiwan University College of Medicine, Taipei, Taiwan
| | - Roberto Giugliani
- Postgraduate Program in Genetics and Molecular Biology (PPGBM) at Federal University of Rio Grande do Sul (UFRGS), Porto Alegre, Brazil
- BioDiscovery Laboratory at Hospital de Clinicas de Porto Alegre (HCPA), National Institute of Population Medical Genetics (INAGEMP), DASA, Casa dos Raros, Porto Alegre, Brazil
| | - Staci Kallish
- Division of Translational Medicine and Human Genetics, Department of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | | | - Olivier Lidove
- Department of Internal Medicine-Rheumatology, Croix Saint Simon Hospital, Paris, France
- French Network of Inherited Metabolic Disorders (G2m), France
| | - Dau-Ming Niu
- Department of Pediatrics, Taipei Veterans General Hospital, Taipei, Taiwan
- Institute of Clinical Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Iacopo Olivotto
- Department of Experimental and Clinical Medicine, Meyer University Children’s Hospital, Florence, Italy
| | - Juan Politei
- Department of Neurology, Fundacion Para el Estudio de Enfermedades Neurometabolicas (FESEN), Buenos Aires, Argentina
| | | | - Roser Torra
- Inherited Kidney Disorders, Department of Nephrology, Fundació Puigvert, Institut d’Investigació Biomèdica Sant Pau (IIB-SANT PAU), Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Camilla Tøndel
- Department of Clinical Science, University of Bergen, Bergen, Norway
- Department of Pediatrics, Haukeland University Hospital, Bergen, Norway
| | - Derralynn A. Hughes
- Lysosomal Storage Disorders Unit, Royal Free London NHS Foundation Trust and University College London, London, United Kingdom
| |
Collapse
|
20
|
Arslan N, Coker M, Gokcay GF, Kiykim E, Onenli Mungan HN, Ezgu F. Expert opinion on patient journey, diagnosis and clinical monitoring in acid sphingomyelinase deficiency in Turkey: a pediatric metabolic disease specialist's perspective. Front Pediatr 2023; 11:1113422. [PMID: 37435168 PMCID: PMC10330960 DOI: 10.3389/fped.2023.1113422] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/01/2022] [Accepted: 06/06/2023] [Indexed: 07/13/2023] Open
Abstract
This review by a panel of pediatric metabolic disease specialists aimed to provide a practical and implementable guidance document to assist clinicians in best clinical practice in terms of recognition, diagnosis and management of patients with acid sphingomyelinase deficiency (ASMD). The participating experts consider the clinical suspicion of ASMD by the physician to be of utmost importance in the prevention of diagnostic delay and strongly suggest the use of a diagnostic algorithm including/starting with dried blood spots assay in the timely diagnosis of ASMD in patients presenting with hepatosplenomegaly and a need for increased awareness among physicians in this regard to consider ASMD in the differential diagnosis. In anticipation of the introduction of enzyme replacement therapy, raising awareness of the disease among physicians to prevent diagnostic delay and further investigation addressing natural history of ASMD across the disease spectrum, potential presenting characteristics with a high index of suspicion, as well as biomarkers and genotype-phenotype correlations suggestive of poor prognosis seem important in terms of implementation of best practice patterns.
Collapse
Affiliation(s)
- Nur Arslan
- Division of Pediatric Metabolism, Department of Pediatrics, Dokuz Eylul University Faculty of Medicine, Izmir, Türkiye
| | - Mahmut Coker
- Division of Pediatric Metabolism, Department of Pediatrics, Ege University Faculty of Medicine, Izmir, Türkiye
| | - Gulden Fatma Gokcay
- Division of Pediatric Metabolism, Department of Pediatrics, Istanbul University Istanbul Faculty of Medicine, Istanbul, Türkiye
| | - Ertugrul Kiykim
- Division of Pediatric Metabolism, Department of Pediatrics, Istanbul University Cerrahpasa Faculty of Medicine, Istanbul, Türkiye
| | | | - Fatih Ezgu
- Division of Pediatric Metabolism and Pediatric Genetics, Department of Pediatrics, Gazi University Faculty of Medicine, Ankara, Türkiye
| |
Collapse
|
21
|
Courtney E, Pickens CA, Cuthbert C, Petritis K. Multiplexing Iduronate-2-Sulphatase (MPS-II) into a 7-Plex Lysosomal Storage Disorder MS/MS Assay Using Cold-Induced Phase Separation. Int J Neonatal Screen 2023; 9:32. [PMID: 37367213 DOI: 10.3390/ijns9020032] [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: 03/25/2023] [Revised: 05/31/2023] [Accepted: 06/01/2023] [Indexed: 06/28/2023] Open
Abstract
Mucopolysaccharidosis type II (MPS-II, Hunter syndrome, OMIM:30990) is a lysosomal storage disorder (LSD) that results in iduronate 2-sulphatase (I2S) enzyme deficiency. MPS-II was added to the Recommended Uniform Screening Panel (RUSP) in August 2022; thus, there is an increased demand for multiplexing I2S into existing LSD screening assays. After incubation with LSD synthetic substrates, extracts are cleaned using liquid-liquid extraction with ethyl acetate or protein precipitation using acetonitrile (ACN). We investigated cold-induced water ACN phase separation (CIPS) to improve the combination of 6-plex and I2S extracts to create a 7-plex assay, and compared it to room temperature ACN and ethyl acetate liquid-liquid extraction. The extracts were dried and resuspended in the mobile phase, and then analyzed using an optimized 1.9 min injection-to-injection liquid chromatography method coupled with tandem mass spectrometry (LC-MS/MS). The combination of ACN and CIPS improved the detection for I2S products without significant detriment to other analytes, which is attributable to a more complete coagulation and separation of heme, proteins, and extracted residual salts. Using CIPS for sample cleanup in dried blood spots (DBS) appears to represent a promising and straightforward way of achieving cleaner sample extracts in a new 7-plex LSD screening panel.
Collapse
Affiliation(s)
- Elya Courtney
- Centers for Disease Control and Prevention, Atlanta, GA 30341, USA
| | - C Austin Pickens
- Centers for Disease Control and Prevention, Atlanta, GA 30341, USA
| | - Carla Cuthbert
- Centers for Disease Control and Prevention, Atlanta, GA 30341, USA
| | | |
Collapse
|
22
|
Gragnaniello V, Burlina AP, Commone A, Gueraldi D, Puma A, Porcù E, Stornaiuolo M, Cazzorla C, Burlina AB. Newborn Screening for Fabry Disease: Current Status of Knowledge. Int J Neonatal Screen 2023; 9:31. [PMID: 37367212 DOI: 10.3390/ijns9020031] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/03/2023] [Revised: 05/24/2023] [Accepted: 06/01/2023] [Indexed: 06/28/2023] Open
Abstract
Fabry disease is an X-linked progressive lysosomal disorder, due to α-galactosidase A deficiency. Patients with a classic phenotype usually present in childhood as a multisystemic disease. Patients presenting with the later onset subtypes have cardiac, renal and neurological involvements in adulthood. Unfortunately, the diagnosis is often delayed until the organ damage is already irreversibly severe, making specific treatments less efficacious. For this reason, in the last two decades, newborn screening has been implemented to allow early diagnosis and treatment. This became possible with the application of the standard enzymology fluorometric method to dried blood spots. Then, high-throughput multiplexable assays, such as digital microfluidics and tandem mass spectrometry, were developed. Recently DNA-based methods have been applied to newborn screening in some countries. Using these methods, several newborn screening pilot studies and programs have been implemented worldwide. However, several concerns persist, and newborn screening for Fabry disease is still not universally accepted. In particular, enzyme-based methods miss a relevant number of affected females. Moreover, ethical issues are due to the large number of infants with later onset forms or variants of uncertain significance. Long term follow-up of individuals detected by newborn screening will improve our knowledge about the natural history of the disease, the phenotype prediction and the patients' management, allowing a better evaluation of risks and benefits of the newborn screening for Fabry disease.
Collapse
Affiliation(s)
- Vincenza Gragnaniello
- Division of Inherited Metabolic Diseases, Department of Diagnostic Services, University Hospital, 35128 Padua, Italy
| | | | - Anna Commone
- Division of Inherited Metabolic Diseases, Department of Diagnostic Services, University Hospital, 35128 Padua, Italy
| | - Daniela Gueraldi
- Division of Inherited Metabolic Diseases, Department of Diagnostic Services, University Hospital, 35128 Padua, Italy
| | - Andrea Puma
- Division of Inherited Metabolic Diseases, Department of Diagnostic Services, University Hospital, 35128 Padua, Italy
| | - Elena Porcù
- Division of Inherited Metabolic Diseases, Department of Diagnostic Services, University Hospital, 35128 Padua, Italy
| | - Maria Stornaiuolo
- Division of Inherited Metabolic Diseases, Department of Diagnostic Services, University Hospital, 35128 Padua, Italy
| | - Chiara Cazzorla
- Division of Inherited Metabolic Diseases, Department of Diagnostic Services, University Hospital, 35128 Padua, Italy
| | - Alberto B Burlina
- Division of Inherited Metabolic Diseases, Department of Diagnostic Services, University Hospital, 35128 Padua, Italy
| |
Collapse
|
23
|
Averbuch T, White JA, Fine NM. Anderson-Fabry disease cardiomyopathy: an update on epidemiology, diagnostic approach, management and monitoring strategies. Front Cardiovasc Med 2023; 10:1152568. [PMID: 37332587 PMCID: PMC10272370 DOI: 10.3389/fcvm.2023.1152568] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2023] [Accepted: 05/09/2023] [Indexed: 06/20/2023] Open
Abstract
Anderson-Fabry disease (AFD) is an X-linked lysosomal storage disorder caused by deficient activity of the enzyme alpha-galactosidase. While AFD is recognized as a progressive multi-system disorder, infiltrative cardiomyopathy causing a number of cardiovascular manifestations is recognized as an important complication of this disease. AFD affects both men and women, although the clinical presentation typically varies by sex, with men presenting at a younger age with more neurologic and renal phenotype and women developing a later onset variant with more cardiovascular manifestations. AFD is an important cause of increased myocardial wall thickness, and advances in imaging, in particular cardiac magnetic resonance imaging and T1 mapping techniques, have improved the ability to identify this disease non-invasively. Diagnosis is confirmed by the presence of low alpha-galactosidase activity and identification of a mutation in the GLA gene. Enzyme replacement therapy remains the mainstay of disease modifying therapy, with two formulations currently approved. In addition, newer treatments such as oral chaperone therapy are now available for select patients, with a number of other investigational therapies in development. The availability of these therapies has significantly improved outcomes for AFD patients. Improved survival and the availability of multiple agents has presented new clinical dilemmas regarding disease monitoring and surveillance using clinical, imaging and laboratory biomarkers, in addition to improved approaches to managing cardiovascular risk factors and AFD complications. This review will provide an update on clinical recognition and diagnostic approaches including differentiation from other causes of increased ventricular wall thickness, in addition to modern strategies for management and follow-up.
Collapse
Affiliation(s)
- Tauben Averbuch
- Division of Cardiology, Department of Cardiac Sciences, University of Calgary, Calgary, AB, Canada
| | - James A. White
- Division of Cardiology, Department of Cardiac Sciences, University of Calgary, Calgary, AB, Canada
- Stephenson Cardiac Imaging Center, Alberta Health Services, Libin Cardiovascular Institute, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Nowell M. Fine
- Division of Cardiology, Department of Cardiac Sciences, University of Calgary, Calgary, AB, Canada
| |
Collapse
|
24
|
Gilchrist M, Casanova F, Tyrrell JS, Cannon S, Wood AR, Fife N, Young K, Oram RA, Weedon MN. Prevalence of Fabry disease-causing variants in the UK Biobank. J Med Genet 2023; 60:391-396. [PMID: 35977816 PMCID: PMC10086508 DOI: 10.1136/jmg-2022-108523] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2022] [Accepted: 07/15/2022] [Indexed: 11/03/2022]
Abstract
BACKGROUND Fabry disease is an X-linked lysosomal storage disorder resulting from deficiency of the alpha-galactosidase A enzyme leading to accumulation of globotriaosylceramide in multiple organ sites with prominent cardiovascular and renal involvement. Global prevalence estimates of Fabry disease based on clinical ascertainment range from 1 in 40 000 to 1 in 170 000. We aimed to determine the prevalence of Fabry disease-causing variants in UK Biobank. METHODS We sought GLA gene variants in exome sequencing data from 200 643 individuals from UK Biobank. We used ACMG/AMP guidelines (American College of Medical Genetics/Association for Molecular Pathology) to classify pathogenicity and compared baseline biomarker data, hospital ICD-10 (International Classification of Diseases version-10) codes, general practitioner records and self-reported health data with those without pathogenic variants. RESULTS We identified 81 GLA coding variants. We identified eight likely pathogenic variants on the basis of being rare (<1/10 000 individuals) and either previously reported to cause Fabry disease, or being protein-truncating variants. Thirty-six individuals carried one of these variants. In the UK Biobank, the prevalence of likely pathogenic Fabry disease-causing variants is 1/5732 for late-onset disease-causing variants and 1/200 643 for variants causing classic Fabry disease. CONCLUSION Fabry disease-causing GLA variants are more prevalent in an unselected population sample than the reported prevalence of Fabry disease. These are overwhelmingly variants associated with later onset. It is possible the prevalence of later-onset Fabry disease exceeds current estimates.
Collapse
Affiliation(s)
- Mark Gilchrist
- College of Medicine and Health, University of Exeter Medical School, Exeter, UK
| | | | - Jess S Tyrrell
- College of Medicine and Health, University of Exeter, Exeter, UK
| | - Stuart Cannon
- College of Medicine and Health, University of Exeter Medical School, Exeter, UK
| | - Andrew R Wood
- Genetics of Complex Traits, University of Exeter Medical School, Exeter, UK
| | - Nicole Fife
- College of Medicine and Health, University of Exeter Medical School, Exeter, UK
| | - Katherine Young
- College of Medicine and Health, University of Exeter Medical School, Exeter, UK
| | - Richard A Oram
- Institute of Biomedical and Clinical Science, University of Exeter Medical School, Exeter, UK
| | - Michael N Weedon
- Institute of Biomedical and Clinical Science, University of Exeter Medical School, Exeter, UK
| |
Collapse
|
25
|
Oliva P, Schwarz M, Mechtler TP, Sansen S, Keutzer J, Prusa AR, Streubel B, Kasper DC. Importance to include differential diagnostics for acid sphingomyelinase deficiency (ASMD) in patients suspected to have to Gaucher disease. Mol Genet Metab 2023; 139:107563. [PMID: 37086570 DOI: 10.1016/j.ymgme.2023.107563] [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: 12/19/2022] [Revised: 02/12/2023] [Accepted: 03/26/2023] [Indexed: 04/24/2023]
Abstract
The clinical manifestation of sphingolipidosis leads often to misclassification between acid sphingomyelinase deficiency (ASMD) and Gaucher disease. In this multicenter, prospective study, we investigated a cohort of 31,838 individuals suspected to have Gaucher disease, due to clinical presentation, from 61 countries between 2017 and 2022. For all samples, both Acid-β-glucocerebrosidase and acid sphingomyelinase enzyme activities were measured in dried blood spot specimens by tandem mass spectrometry followed by genetic confirmatory testing in potential positive cases. In total, 5933 symptomatic cases showed decreased enzyme activities and were submitted for genetic confirmatory testing. 1411/5933 (24%) cases were finally identified with Gaucher disease and 550/5933 (9%) with ASMD. Most of the confirmed ASMD cases were newborns and children below 2 years of age (63%). This study reveals that one in four cases suspected for Gaucher disease is diagnosed with ASMD. An early appropriate diagnostic work-up is essential because of the availability of a recently approved enzyme replacement therapy for ASMD. In conclusion, a diagnostic strategy using differential biochemical testing including genetic confirmatory testing in clinically suspected cases for sphingolipidosis is highly recommended.
Collapse
Affiliation(s)
- Petra Oliva
- ARCHIMED Life Science GmbH (ARCHIMEDlife), Vienna, Austria.
| | - Markus Schwarz
- ARCHIMED Life Science GmbH (ARCHIMEDlife), Vienna, Austria.
| | | | | | - Joan Keutzer
- Sanofi Genzyme, Amsterdam, Netherlands; Independent consultant, Littleton MA 01460, USA
| | - Andrea-Romana Prusa
- Deptartment of Children and Adolescent Medicine, Medical University of Vienna, Vienna, Austria.
| | - Berthold Streubel
- Clinical Institute of Pathology, Medical University of Vienna, Vienna, Austria.
| | - David C Kasper
- ARCHIMED Life Science GmbH (ARCHIMEDlife), Vienna, Austria.
| |
Collapse
|
26
|
Pfrieger FW. The Niemann-Pick type diseases – A synopsis of inborn errors in sphingolipid and cholesterol metabolism. Prog Lipid Res 2023; 90:101225. [PMID: 37003582 DOI: 10.1016/j.plipres.2023.101225] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2023] [Revised: 03/27/2023] [Accepted: 03/27/2023] [Indexed: 04/03/2023]
Abstract
Disturbances of lipid homeostasis in cells provoke human diseases. The elucidation of the underlying mechanisms and the development of efficient therapies represent formidable challenges for biomedical research. Exemplary cases are two rare, autosomal recessive, and ultimately fatal lysosomal diseases historically named "Niemann-Pick" honoring the physicians, whose pioneering observations led to their discovery. Acid sphingomyelinase deficiency (ASMD) and Niemann-Pick type C disease (NPCD) are caused by specific variants of the sphingomyelin phosphodiesterase 1 (SMPD1) and NPC intracellular cholesterol transporter 1 (NPC1) or NPC intracellular cholesterol transporter 2 (NPC2) genes that perturb homeostasis of two key membrane components, sphingomyelin and cholesterol, respectively. Patients with severe forms of these diseases present visceral and neurologic symptoms and succumb to premature death. This synopsis traces the tortuous discovery of the Niemann-Pick diseases, highlights important advances with respect to genetic culprits and cellular mechanisms, and exposes efforts to improve diagnosis and to explore new therapeutic approaches.
Collapse
|
27
|
McGarry ME, Ren CL, Wu R, Farrell PM, McColley SA. Detection of disease-causing CFTR variants in state newborn screening programs. Pediatr Pulmonol 2023; 58:465-474. [PMID: 36237137 PMCID: PMC9870974 DOI: 10.1002/ppul.26209] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/23/2022] [Revised: 09/29/2022] [Accepted: 10/12/2022] [Indexed: 01/26/2023]
Abstract
BACKGROUND Newborn screening (NBS) algorithms for cystic fibrosis (CF) vary in the United State of America and include different cystic fibrosis transmembrane conductance regulator (CFTR) variants. CFTR variant distribution varies among racial and ethnic groups. OBJECTIVE Our objectives were to identify differences in detection rate by race and ethnicity for CFTR variant panels, identify each US state detection rate for CFTR variant panels, and describe the rate of false-negative NBS and delayed diagnoses by race and ethnicity. METHODS This is a cross-sectional analysis of the detection rate of at least 1 CFTR variant for seven panels by race and ethnicity in genotyped people with CF (PwCF) or CFTR-related metabolic syndrome (CRMS)/CFTR-related disorders in CF Foundation Patient Registry (CFFPR) in 2020. We estimated the case detection rate of CFTR variant panels by applying the detection rate to Census data. Using data from CFFPR, we compared the rate of delayed diagnosis or false-negative NBS by race and ethnicity. RESULTS For all panels, detection of at least 1 CFTR variant was highest in non-Hispanic White PwCF (87.5%-97.0%), and lowest in Black, Asian, and Hispanic PwCF (41.9%-93.1%). Detection of at least 1 CFTR variant was lowest in Black and Asian people with CRMS/CFTR-related disorders (48.4%-64.8%). States with increased racial and ethnic diversity have lower detection rates for all panels. Overall, 3.8% PwCF had a false-negative NBS and 11.8% had a delayed diagnosis; Black, Hispanic, and mixed-race PwCF were overrepresented. CONCLUSION CFTR variant panels have lower detection rates in minoritized racial and ethnic groups leading to false-negative NBS, delayed diagnosis, and likely health disparities.
Collapse
Affiliation(s)
- Meghan E McGarry
- Department of Pediatrics, University of California, San Francisco, California, USA
| | - Clement L Ren
- Department of Pediatrics, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
| | - Runyu Wu
- Cystic Fibrosis Foundation, Bethesda, Maryland, USA
| | - Philip M Farrell
- Department of Pediatrics, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin, USA
| | - Susanna A McColley
- Department of Pediatrics, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| |
Collapse
|
28
|
The Spanish Fabry women study: a retrospective observational study describing the phenotype of females with GLA variants. Orphanet J Rare Dis 2023; 18:8. [PMID: 36624527 PMCID: PMC9830917 DOI: 10.1186/s13023-022-02599-w] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2022] [Accepted: 12/19/2022] [Indexed: 01/11/2023] Open
Abstract
BACKGROUND Fabry disease (FD) is an X-linked condition caused by variants in the GLA gene. Since females have two X chromosomes, they were historically thought to be carriers. Although increased knowledge has shown that females often develop the disease, data from Spain and other countries reported that females were undertreated. The aim of this study was to provide a wider and more recent description of the disease characteristics and associated management of females with a GLA variant in a Spanish cohort. RESULTS Ninety-seven females from 12 hospitals were included in this retrospective study. Mean age was 50.1 ± 17.2 years. Median follow-up time from GLA variant identification was 36.1 months, and most (70.1%) were identified through family screening. Variants associated with classic/non-classic phenotypes were similarly distributed (40.2%/53.6%). Missense variants were the most prevalent (n = 84, 86.6%). In the overall group, 70.4% had major organ involvement (i.e., cardiac, renal, cerebrovascular, peripheral nervous system or gastrointestinal), and 47.3% also had typical Fabry signs (angiokeratoma, cornea verticillata or increased plasma lyso-Gb3). Cardiac involvement was the most prevalent (49.5%) and the main reason for treatment initiation. A total of 33 (34%) patients received disease-specific therapy, 55% of whom were diagnosed by family screening. Females carrying variants associated with a classic phenotype had higher frequencies of clinical manifestations (92.3%) and were predominant in the treated subgroup (69.7%). Despite this, there were 34 untreated females (56.7% of total untreated), with both phenotypes represented, who had major organ involvement, with 27 of cardiac, renal or cerebrovascular nature. Age or comorbidities in this subgroup were comparable to the treated subgroup (P = 0.8 and P = 0.8, respectively). CONCLUSIONS Efforts have been made in recent years to diagnose and treat timely Fabry females in Spain. A high percentage of females with pathogenic variants, regardless of their associated phenotype, will likely develop disease. A proportion of females with severe disease in this cohort received specific treatment. Still a significant number of females, even with same profile as the treated ones, who may be eligible for treatment according to European recommendations, remained untreated. Reasons for this merit further investigation.
Collapse
|
29
|
Castillon G, Chang SC, Moride Y. Global Incidence and Prevalence of Gaucher Disease: A Targeted Literature Review. J Clin Med 2022; 12:jcm12010085. [PMID: 36614898 PMCID: PMC9821068 DOI: 10.3390/jcm12010085] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2022] [Revised: 12/16/2022] [Accepted: 12/19/2022] [Indexed: 12/24/2022] Open
Abstract
Incidence and prevalence estimates for Gaucher disease (GD) are scarce for this rare disease and can be variable within the same region. This review provides a qualitative synthesis of global GD incidence and prevalence estimates, GD1-3 type-specific and overall, published in the last 10 years. A targeted literature search was conducted across multiple databases from January 2011 to September 2020, including web-based sources and congress proceedings to May 2021. Searches yielded 490 publications, with 31 analyzed: 20 cohort studies (15 prospective, 5 retrospective), 6 cross-sectional studies, 5 online reports (most from Europe (n = 11) or North America (n = 11); one multiregional). Across all GD types, incidence estimates ranged 0.45-25.0/100,000 live births (16 studies), lowest for Asia-Pacific. Incidence of GD1: 0.45-22.9/100,000 live births (Europe and North America) and GD3: 1.36/100,000 live births (Asia-Pacific only). GD type-specific prevalence estimates per 100,000 population were GD1: 0.26-0.63; GD2 and GD3: 0.02-0.08 (Europe only); estimates for GD type unspecified or overall ranged 0.11-139.0/100,000 inhabitants (17 studies), highest for North America. Generalizability was assessed as "adequate"or "intermediate" for all regions with data. GD incidence and prevalence estimates for the last 10 years varied considerably between regions and were poorly documented outside Europe and North America. Data for GD2 and GD3 were limited.
Collapse
Affiliation(s)
| | - Shun-Chiao Chang
- Takeda Development Center Americas, Inc., Cambridge, MA 02142, USA
- Correspondence:
| | - Yola Moride
- YolaRX Consultants Inc., Montreal, QC H3H 1V4, Canada
- Center for Pharmacoepidemiology and Treatment Science, Rutgers, The State University of New Jersey, New Brunswick, NJ 08901, USA
| |
Collapse
|
30
|
Liquid Chromatography-Tandem Mass Spectrometry in Newborn Screening Laboratories. Int J Neonatal Screen 2022; 8:ijns8040062. [PMID: 36547379 PMCID: PMC9781967 DOI: 10.3390/ijns8040062] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/15/2022] [Revised: 11/11/2022] [Accepted: 11/16/2022] [Indexed: 11/29/2022] Open
Abstract
Tandem mass spectrometry (MS/MS) is the most universal platform currently available for the analysis of enzymatic activities and biomarkers in dried blood spots (DBS) for applications in newborn screening (NBS). Among the MS/MS applications in NBS, the most common is flow-injection analysis (FIA-) MS/MS, where the sample is introduced as a bolus injection into the mass spectrometer without the prior fractionation of analytes. Liquid chromatography combined with MS/MS (LC-MS/MS) has been employed for second-tier tests to reduce the false-positive rate associated with several nonspecific screening markers, beginning two decades ago. More recently, LC-MS/MS has been applied to primary screening for new conditions for which FIA-MS/MS or other methods, including genomic screening, are not yet adequate. In addition to providing a list of the currently used LC-MS/MS-based assays for NBS, the authors share their experience regarding the maintenance requirements of LC-MS/MS vs. FIA-MS/MS systems. The consensus is that the maintenance of LC-MS/MS and FIA-MS/MS instrumentation is similar, and LC-MS/MS has the advantage of allowing for a larger number of diseases to be screened for in a multiplex, cost-effective fashion with a high throughput and an adequate turnaround time.
Collapse
|
31
|
A Roadmap for Potential Improvement of Newborn Screening for Inherited Metabolic Diseases Following Recent Developments and Successful Applications of Bivariate Normal Limits for Pre-Symptomatic Detection of MPS I, Pompe Disease, and Krabbe Disease. Int J Neonatal Screen 2022; 8:ijns8040061. [PMID: 36412587 PMCID: PMC9680456 DOI: 10.3390/ijns8040061] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/06/2022] [Revised: 11/08/2022] [Accepted: 11/10/2022] [Indexed: 11/16/2022] Open
Abstract
The mucopolysaccharidoses (MPS), Pompe Disease (PD), and Krabbe disease (KD) are inherited conditions known as lysosomal storage disorders (LSDs) The resulting enzyme deficiencies give rise to progressive symptoms. The United States Department of Health and Human Services' Recommended Uniform Screening Panel (RUSP) suggests LSDs for inclusion in state universal newborn screening (NBS) programs and has identified screening deficiencies in MPS I, KD, and PD NBS programs. MPS I NBS programs utilize newborn dried blood spots and assay alpha L-iduronidase (IDUA) enzyme to screen for potential cases. Glycosaminoglycans (GAGs) offer potential as a confirmatory test. KD NBS programs utilize galactocerebrosidase (GaLC) as an initial test, with psychosine (PSY) activity increasingly used as a confirmatory test for predicting onset of Krabbe disease, though with an excessive false positive rate. PD is marked by a deficiency in acid α-glucosidase (GAA), causing increased glycogen, creatine (CRE), and other biomarkers. Bivariate normal limit (BVNL) methods have been applied to GaLC and PSY activity to produce a NBS tool for KD, and more recently, to IDUA and GAG activity to develop a NBS tool for MPS I. A BVNL tool based on GAA and CRE is in development for infantile PD diagnosis. Early infantile KD, MPS I, and PD cases were pre-symptomatically identified by BVNL-based NBS tools. This article reviews these developments, discusses how they address screening deficiencies identified by the RUSP and may improve NBS more generally.
Collapse
|
32
|
Gragnaniello V, Pijnappel PW, Burlina AP, In 't Groen SL, Gueraldi D, Cazzorla C, Maines E, Polo G, Salviati L, Di Salvo G, Burlina AB. Newborn screening for Pompe disease in Italy: Long-term results and future challenges. Mol Genet Metab Rep 2022; 33:100929. [PMID: 36310651 PMCID: PMC9597184 DOI: 10.1016/j.ymgmr.2022.100929] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2022] [Accepted: 10/14/2022] [Indexed: 11/06/2022] Open
Abstract
Pompe disease (PD) is a progressive neuromuscular disorder caused by a lysosomal acid α-glucosidase (GAA) deficiency. Enzymatic replacement therapy is available, but early diagnosis by newborn screening (NBS) is essential for early treatment and better outcomes, especially with more severe forms. We present results from 7 years of NBS for PD and the management of infantile-onset (IOPD) and late-onset (LOPD) patients, during which we sought candidate predictive parameters of phenotype severity at baseline and during follow-up. We used a tandem mass spectrometry assay for α-glucosidase activity to screen 206,741 newborns and identified 39 positive neonates (0.019%). Eleven had two pathogenic variants of the GAA gene (3 IOPD, 8 LOPD); six carried variants of uncertain significance (VUS). IOPD patients were treated promptly and had good outcomes. LOPD and infants with VUS were followed; all were asymptomatic at the last visit (mean age 3.4 years, range 0.5–5.5). Urinary glucose tetrasaccharide was a useful and biomarker for rapidly differentiating IOPD from LOPD and monitoring response to therapy during follow-up. Our study, the largest reported to date in Europe, presents data from longstanding NBS for PD, revealing an incidence in North East Italy of 1/18,795 (IOPD 1/68,914; LOPD 1/25,843), and the absence of mortality in IOPD treated from birth. In LOPD, rigorous long-term follow-up is needed to evaluate the best time to start therapy. The high pseudodeficiency frequency, ethical issues with early LOPD diagnosis, and difficulty predicting phenotypes based on biochemical parameters and genotypes, especially in LOPD, need further study.
Collapse
Key Words
- Acid α-glucosidase
- CLIR, Collaborative Laboratory Integrated Reports
- CRIM, cross-reactive immunological material
- DBS, dried blood spot
- DMF, digital microfluidics
- ECG, electrocardiogram
- EF, ejection fraction
- EMG, electromyography
- ERT, enzyme replacement therapy
- Enzyme replacement therapy
- GAA, acid α-glucosidase
- GMFM-88, Gross Motor Function Measure
- Glc4, glucose tetrasaccharide
- IOPD, infantile-onset Pompe disease
- ITI, immunotolerance induction
- LOPD, late-onset Pompe disease
- LVMI, left ventricular max index
- MFM-20, motor function measurement
- MRC, Medical Research Council Scale
- MRI, magnetic resonance imaging
- MS/MS, tandem mass spectrometry
- NBS, newborn screening
- Newborn screening
- PBMC, peripheral blood mononuclear cells
- PD, Pompe disease
- PPV, positive predictive value
- Pompe disease
- RUSP, Recommended Uniform Screening Panel
- Tandem mass-spectrometry
- Urinary tetrasaccharide
- VUS, variants of uncertain significance.
- nv, normal values
- rhGAA, recombinant human GAA
Collapse
Affiliation(s)
- Vincenza Gragnaniello
- Division of Inherited Metabolic Diseases, Department of Diagnostic Services, University Hospital, Padua, Italy
| | - Pim W.W.M. Pijnappel
- Department of Pediatrics, Erasmus University Medical Center, Rotterdam, the Netherlands
- Department of Clinical Genetics, Erasmus University Medical Center, Rotterdam, the Netherlands
- Center for Lysosomal and Metabolic Diseases, Erasmus University Medical Center, Rotterdam, the Netherlands
| | | | - Stijn L.M. In 't Groen
- Department of Pediatrics, Erasmus University Medical Center, Rotterdam, the Netherlands
- Department of Clinical Genetics, Erasmus University Medical Center, Rotterdam, the Netherlands
- Center for Lysosomal and Metabolic Diseases, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Daniela Gueraldi
- Division of Inherited Metabolic Diseases, Department of Diagnostic Services, University Hospital, Padua, Italy
| | - Chiara Cazzorla
- Division of Inherited Metabolic Diseases, Department of Diagnostic Services, University Hospital, Padua, Italy
| | - Evelina Maines
- Division of Pediatrics, S. Chiara General Hospital, Trento, Italy
| | - Giulia Polo
- Division of Inherited Metabolic Diseases, Department of Diagnostic Services, University Hospital, Padua, Italy
| | - Leonardo Salviati
- Clinical Genetics Unit, Department of Women's and Children's Health, and Myology Center, University of Padova, Padova, Italy
| | - Giovanni Di Salvo
- Division of Paediatric Cardiology, Department of Women's and Children's Health, University Hospital Padua, Padua, Italy
| | - Alberto B. Burlina
- Division of Inherited Metabolic Diseases, Department of Diagnostic Services, University Hospital, Padua, Italy
- Corresponding author at: Division of Inherited Metabolic Diseases, Department of Diagnostic Services, University Hospital, via Orus 2/c, 35129 Padua, Italy.
| |
Collapse
|
33
|
Goomber S, Huggins E, Rehder CW, Cohen JL, Bali DS, Kishnani PS. Development of a clinically validated in vitro functional assay to assess pathogenicity of novel GAA variants in patients with Pompe disease identified via newborn screening. Front Genet 2022; 13:1001154. [PMID: 36246652 PMCID: PMC9562992 DOI: 10.3389/fgene.2022.1001154] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2022] [Accepted: 09/12/2022] [Indexed: 11/13/2022] Open
Abstract
Purpose: The addition of Pompe disease (Glycogen Storage Disease Type II) to the Recommended Uniform Screening Panel in the United States has led to an increase in the number of variants of uncertain significance (VUS) and novel variants identified in the GAA gene. This presents a diagnostic challenge, especially in the setting of late-onset Pompe disease when symptoms are rarely apparent at birth. There is an unmet need for validated functional studies to aid in classification of GAA variants. Methods: We developed an in vitro mammalian cell expression and functional analysis system based on guidelines established by the Clinical Genome Resource (ClinGen) Sequence Variant Interpretation Working Group for PS3/BS3. We validated the assay with 12 control variants and subsequently analyzed eight VUS or novel variants in GAA identified in patients with a positive newborn screen for Pompe disease without phenotypic evidence of infantile-onset disease. Results: The control variants were analyzed in our expression system and an activity range was established. The pathogenic controls had GAA activity between 0% and 11% of normal. The benign or likely benign controls had an activity range of 54%–100%. The pseudodeficiency variant had activity of 17%. These ranges were then applied to the variants selected for functional studies. Using the threshold of <11%, we were able to apply PS3_ supporting to classify two variants as likely pathogenic (c.316C > T and c.1103G > A) and provide further evidence to support the classification of likely pathogenic for two variants (c.1721T > C and c.1048G > A). One variant (c.1123C > T) was able to be reclassified based on other supporting evidence. We were unable to reclassify three variants (c.664G > A, c.2450A > G, and c.1378G > A) due to insufficient or conflicting evidence. Conclusion: We investigated eight GAA variants as proof of concept using our validated and reproducible in vitro expression and functional analysis system. While additional work is needed to further refine our system with additional controls and different variant types in order to apply the PS3/BS3 criteria at a higher level, this tool can be utilized for variant classification to meet the growing need for novel GAA variant classification in the era of newborn screening for Pompe disease.
Collapse
Affiliation(s)
- Shelly Goomber
- Division of Medical Genetics, Department of Pediatrics, Duke University School of Medicine, Durham, NC, United States
| | - Erin Huggins
- Division of Medical Genetics, Department of Pediatrics, Duke University School of Medicine, Durham, NC, United States
| | - Catherine W. Rehder
- Department of Pathology, Duke University Medical Center, Durham, NC, United States
| | - Jennifer L. Cohen
- Division of Medical Genetics, Department of Pediatrics, Duke University School of Medicine, Durham, NC, United States
| | - Deeksha S. Bali
- Division of Medical Genetics, Department of Pediatrics, Duke University School of Medicine, Durham, NC, United States
- Biochemical Genetics Laboratory, Duke University Medical Center, Durham, NC, United States
| | - Priya S. Kishnani
- Division of Medical Genetics, Department of Pediatrics, Duke University School of Medicine, Durham, NC, United States
- *Correspondence: Priya S. Kishnani,
| |
Collapse
|
34
|
Do YR, Choi Y, Heo MH, Kim JS, Yoon JH, Lee JH, Park JS, Sohn SK, Kim SH, Lim S, Chung JS, Jo DY, Eom HS, Kim H, Jeon SY, Won JH, Lee HJ, Shin JW, Jang JH, Yoon SS. Early diagnosis of Gaucher disease in Korean patients with unexplained splenomegaly: a multicenter observational study. Blood Res 2022; 57:207-215. [PMID: 35880496 PMCID: PMC9492528 DOI: 10.5045/br.2022.2022089] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2022] [Revised: 06/23/2022] [Accepted: 06/27/2022] [Indexed: 11/17/2022] Open
Abstract
Background Gaucher disease (GD) is an autosomal recessive disorder characterized by excessive accumulation of glucosylceramide in multiple organs. This study was performed to determine the detection rate of GD in a selected patient population with unexplained splenomegaly in Korea. Methods This was a multicenter, observational study conducted at 18 sites in Korea between December 2016 and February 2020. Adult patients with unexplained splenomegaly were enrolled and tested for β-glucosidase enzyme activity on dried blood spots (DBS) and in peripheral blood leukocytes. Mutation analysis was performed if the test was positive or indeterminate for the enzyme assay. The primary endpoint was the percentage of patients with GD in patients with unexplained splenomegaly. Results A total of 352 patients were enrolled in this study (male patients, 199; mean age, 48.42 yr). Amongst them, 14.77% of patients had concomitant hepatomegaly. The most common sign related to GD was splenomegaly (100%), followed by thrombocytopenia (44.32%) and, anemia (40.91%). The β-glucosidase activity assay on DBS and peripheral leukocytes showed abnormal results in sixteen and six patients, respectively. Eight patients were tested for the mutation, seven of whom were negative and one patient showed a positive mutation analysis result. One female patient who presented with splenomegaly and thrombocytopenia was diagnosed with type 1 GD. The detection rate of GD was 0.2841% (exact 95% CI, 0.0072‒1.5726). Conclusion The detection rate of GD in probable high-risk patients in Korea was lower than expected. However, the role of hemato-oncologists is still important in the diagnosis of GD.
Collapse
Affiliation(s)
- Young Rok Do
- Department of Internal Medicine, Keimyung University Dongsan Hospital, Keimyung University School of Medicine, Daegu, Korea
| | - Yunsuk Choi
- Department of Hematology and Oncology, Ulsan University Hospital, University of Ulsan College of Medicine, Ulsan, Korea
| | - Mi Hwa Heo
- Department of Hematology and Oncology, Ulsan University Hospital, University of Ulsan College of Medicine, Ulsan, Korea
| | - Jin Seok Kim
- Division of Hematology, Department of Internal Medicine, Severance Hospital, Yonsei University College of Medicine, Seoul, Korea
| | - Jae-Ho Yoon
- Division of Hematology, Department of Internal Medicine, Catholic Hematology Hospital and Leukemia Research Institute, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Je-Hwan Lee
- Department of Hematology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Joon Seong Park
- Department of Hematology-Oncology, Ajou University School of Medicine, Suwon, Korea
| | - Sang Kyun Sohn
- Departments of Oncology/Hematology, Kyungpook National University Hospital, Kyungpook National University School of Medicine, Daegu, Korea
| | - Sung Hyun Kim
- Division of Hematology and Oncology, Department of Internal Medicine, Dong-A University College of Medicine, Busan, Korea
| | - Sungnam Lim
- Department of Internal Medicine, Inje University College of Medicine, Haeundae Paik Hospital, Busan, Korea
| | - Joo Seop Chung
- Division of Hematology-Oncology, Department of Internal Medicine, Pusan National University School of Medicine and Biomedical Research Institute, Pusan National University Hospital, Busan, Korea
| | - Deog-Yeon Jo
- Division of Hematology/Oncology, Department of Internal Medicine, Chungnam National University Hospital, Chungnam National University College of Medicine, Daejeon, Korea
| | - Hyeon Seok Eom
- Department of Hematology-Oncology, Center for Hematologic Malignancy, National Cancer Center, Goyang, Korea
| | - Hawk Kim
- Division of Hematology, Gachon University Gil Medical Center, Gachon University College of Medicine, Incheon, Korea
| | - So Yeon Jeon
- Division of Hematology/Oncology, Department of Internal Medicine, Jeonbuk National University Hospital, Jeonbuk National University Medical School, Jeonju, Korea
| | - Jong-Ho Won
- Division of Hematology and Oncology, Department of Internal Medicine, Soonchunhyang University Hospital, Seoul, Korea
| | - Hee Jeong Lee
- Department of Internal Medicine, Hemato-Oncology, Chosun University Hospital, Gwangju, Korea
| | - Jung Won Shin
- Sanofi, Division of Hematology/Oncology, Department of Medicine, Seoul National University College of Medicine, Seoul, Korea
| | - Jun-Ho Jang
- Samsung Medical Center, Seoul National University College of Medicine, Seoul, Korea
| | - Sung-Soo Yoon
- Seoul National University Hospital, Seoul National University College of Medicine, Seoul, Korea
| |
Collapse
|
35
|
Global Epidemiology of Gaucher Disease: an Updated Systematic Review and Meta-analysis. J Pediatr Hematol Oncol 2022; 45:181-188. [PMID: 35867706 PMCID: PMC10115488 DOI: 10.1097/mph.0000000000002506] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/27/2022] [Accepted: 05/01/2022] [Indexed: 11/27/2022]
Abstract
BACKGROUND Gaucher disease [GD], an autosomal recessive lysosomal storage disorder, is characterized by progressive lysosomal storage of glucocerebroside in macrophages predominantly in bone, bone marrow, liver, and spleen. Meta-analysis of global GD epidemiology was not available before this study. METHODS To provide a systematic review and meta-analysis of birth prevalence and prevalence of GD in multiple countries. MEDLINE and EMBASE databases were searched for original research articles on the epidemiology of GD from inception until July 21, 2021. Meta-analysis, adopting a random-effects logistic model, was performed to estimate the birth prevalence and prevalence of GD. RESULTS Eighteen studies that were screened out of 1874 records were included for data extraction. The studies that fulfilled the criteria for inclusion involved 15 areas/countries. The global birth prevalence of GD was 1.5 cases [95% confidence interval: 1.0 to 2.0] per 100,000 live births. The global prevalence of GD was 0.9 cases [95% confidence interval: 0.7 to 1.1] per 100,000 inhabitants. CONCLUSIONS This is the first comprehensive systematic review that presented quantitative data of GD global epidemiology. Quantitative data on global epidemiology of GD could be the fundamental to evaluate the global efforts on building a better world for GD patients.
Collapse
|
36
|
Tremblay J, Kim S, Philbin E, Beers K, Lightle A, Belov D. Late-onset Fabry disease: the cardiac sequela. BMJ Case Rep 2022; 15:e247917. [PMID: 35680278 PMCID: PMC9185384 DOI: 10.1136/bcr-2021-247917] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/20/2022] [Indexed: 11/04/2022] Open
Abstract
We describe a patient with Fabry disease (FD) who initially presented with atrial fibrillation without left ventricular hypertrophy (LVH) 14 years before being correctly diagnosed with FD. In the interim, he survived a myocardial infarction complicated by ventricular fibrillation, and his severe LVH was misdiagnosed as sarcomeric hypertrophic cardiomyopathy. In the following 4 years, he developed proteinuric kidney disease, neuropathy, sensorineural hearing loss and gastrointestinal symptoms. The patient was eventually readmitted for an overt heart failure (HF) exacerbation and was seen by an HF cardiologist. The constellation of systemic findings led to further diagnostic testing, including an endomyocardial biopsy, tests to determine alpha-galactosidase A enzyme activity and α-galactosidase A gene (GLA) analysis. The results of the patient's tests were consistent with FD and he was started on enzyme replacement therapy. To our knowledge, this is the first detailed description of a late-onset phenotype of FD with c.146 G>C GLA variant. In addition, this case serves as a potent reminder to pay meticulous attention to 'red flags' accompanying LVH.
Collapse
Affiliation(s)
- John Tremblay
- Internal Medicine, Albany Medical Center, Albany, New York, USA
| | - Samuel Kim
- Department of Cardiology, Albany Medical Center, Albany, New York, USA
| | - Edward Philbin
- Department of Cardiology, Albany Medical Center, Albany, New York, USA
| | - Kelly Beers
- Department of Nephrology, Albany Medical Center, Albany, New York, USA
| | - Andrea Lightle
- Department of Pathology, Albany Medical Center, Albany, New York, USA
| | - Dmitri Belov
- Department of Cardiology, Albany Medical Center, Albany, New York, USA
| |
Collapse
|
37
|
Ellinwood NM. Newborn screening and the recommended uniform screening panel: Optimal submissions and suggested improvements based on an advocacy organization's decade-long experience. AMERICAN JOURNAL OF MEDICAL GENETICS. PART C, SEMINARS IN MEDICAL GENETICS 2022; 190:156-161. [PMID: 36135708 DOI: 10.1002/ajmg.c.32001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/19/2022] [Revised: 07/26/2022] [Accepted: 08/27/2022] [Indexed: 06/16/2023]
Abstract
The National MPS Society, Inc., founded in 1974, is a rare disease advocacy non-profit with a tripartite mission addressing the needs of the mucopolysaccharidosis and mucolipidosis communities through advocacy, research, and family and patient support. The Recommended Uniform Screening Panel (RUSP) of conditions for newborn screening (NBS), legislatively mandated in 2008, was implemented in 2010 by the Secretary of Health and Human Services (HSS), through the adoption of 29 core conditions. Since its inception the RUSP has grown to 35 core conditions. Each addition followed a defined nomination process that has itself undergone further definition over time. Since the adoption of the RUSP, the Society has nominated two conditions that have been approved by the Advisory Committee on Heritable Disorders in Children and Newborns (ACHDNC) and forwarded to the Secretary of HSS for inclusion on the RUSP. This history places the Society in a position to reflect on the process of successfully nominating conditions. Additionally, the Society is well placed by this experience to provide observations on the RUSP process. We will highlight best practices for pending and future nominations and reflect on potential improvements to the process and infrastructure of NBS, the RUSP, and the ACHDNC.
Collapse
Affiliation(s)
- N Matthew Ellinwood
- The National MPS Society, Inc., 1007 Slater Rd. Suite 220, Durham, North Carolina, 27703, USA
| |
Collapse
|
38
|
LI G, TIAN L, GUO Y, LI Y, SUN M, ZOU H. Cut-off values of neonatal lysosomal storage disease-related enzymes detected by tandem mass spectrometry. Zhejiang Da Xue Xue Bao Yi Xue Ban 2022; 51:321-325. [PMID: 35462462 PMCID: PMC9511489 DOI: 10.3724/zdxbyxb-2022-0095] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
OBJECTIVE To establish cut-off values of lysosomal storage disease (LSD)-related enzymes by tandem mass spectrometry. METHODS A total of 26 689 newborns and 7 clinically confirmed LSD children underwent screening for LSDs (glycogen storage disease typeⅡ, Fabry disease, mucopolysaccharidosis type Ⅰ, Krabbe disease, Niemann-Pick disease A/B and Gaucher disease). The activities of LSD-related enzymes were detected by tandem mass spectrometry. The 20% of the median enzyme activity of each batch of acid β-glucocerebrosidase, acid sphingomyelinase, β-galactocerebroside, α- L-iduronidase and acid α-glucosidase, and the 30% of the median enzyme activity of α-galactosidase were taken as cut-off values of corresponding enzymes. The genetic diagnosis was performed in neonates whose enzyme activity was lower than 70% of the cut-off value. RESULTS The enzyme activities of 7 clinically confirmed cases were all lower than the cut-off values. Among 26 689 newborns, 142 cases (0.53%) were suspected positive for LSDs, including 25 cases of β-galactocerebroside deficiency, 1 case of α- L-iduronidase deficiency, 19 cases of α-galactosidase deficiency, and 97 cases of acid α-glucosidase deficiency. Eight infants were genetically diagnosed with LSDs, including 3 cases of glycogen storage disease type Ⅱ, 3 cases of Krabbe disease, and 2 cases of Fabry disease, with a positive predictive value of about 5.6%. Cut-off values of the 6 LSD enzyme activities all showed a downward trend from March to August, and an upward trend from September to December. There was a statistically significant difference in LSD enzyme activity among different months ( P<0.05). CONCLUSION The established cut-off values of LSD-related enzyme activities detected by tandem mass spectrometry can be used for screening LSDs in neonates, and the enzyme activity would be affected by temperature and humidity.
Collapse
|
39
|
Viall S, Dennis A, Yang A. Newborn screening for Fabry disease in Oregon: Approaching the iceberg of A143T and variants of uncertain significance. AMERICAN JOURNAL OF MEDICAL GENETICS. PART C, SEMINARS IN MEDICAL GENETICS 2022; 190:206-214. [PMID: 36156392 DOI: 10.1002/ajmg.c.31998] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/15/2022] [Revised: 08/01/2022] [Accepted: 08/27/2022] [Indexed: 06/16/2023]
Abstract
Fabry disease newborn screening (NBS) has been ongoing in Oregon for over 41 months by first-tier enzyme quantitation and second-tier DNA testing. During that period the majority of abnormal referrals received (34/60) were for the presence of the controversial c.427G > A (p.Ala143Thr) aka A143T and the majority of non-A143T referrals were for other variants of uncertain significance (17/60) resulting in at least 32 infants with an inconclusive case outcome even after clinical evaluation and/or diagnostic testing. To date there has been no significant family history or onset of symptoms in individuals with an inconclusive outcome. Based on our experience, we have developed a framework for approaching A143T and other variants of uncertain clinical significance in an attempt to balance sensitivity with the unnecessary medicalization of healthy infants.
Collapse
Affiliation(s)
- Sarah Viall
- Oregon Health & Science University, Portland, Oregon, USA
| | - Anna Dennis
- Oregon Health & Science University, Portland, Oregon, USA
| | - Amy Yang
- Oregon Health & Science University, Portland, Oregon, USA
| |
Collapse
|
40
|
Sawada T, Kido J, Sugawara K, Yoshida S, Matsumoto S, Shimazu T, Matsushita Y, Inoue T, Hirose S, Endo F, Nakamura K. Newborn screening for Gaucher disease in Japan. Mol Genet Metab Rep 2022; 31:100850. [PMID: 35242582 PMCID: PMC8866142 DOI: 10.1016/j.ymgmr.2022.100850] [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: 12/29/2021] [Revised: 02/13/2022] [Accepted: 02/13/2022] [Indexed: 12/04/2022] Open
Abstract
Gaucher disease (GD) is an autosomal recessive inborn metabolic disorder caused by a glucocerebrosidase (GCase) defect. GD is classified into three main types depending on accompanying neurological symptoms. Enzyme replacement therapy and substrate reduction therapy are limited in the treatment of neurological symptoms, and using genotype and GCase activity to discriminate between non-neuronopathic and neuronopathic GD may be challenging as the two sometimes phenotypically overlap. The number of patients exhibiting neurological symptoms in Japan is significantly higher than that in Europe and the United States, and newborn screening (NBS) is still not actively performed in Japan. Definitive determination of the actual frequency and proportion of the type of GD from the results of NBS remains inconclusive. We performed NBS for Fabry disease, Pompe disease, and GD, mainly in the Kyushu area in Japan. Herein, we discuss the results of NBS for GD, as well as, the insights gained from following the clinical course of patients diagnosed through NBS. A total of 155,442 newborns were screened using an enzyme activity assay using dried blood spots. We found four newborns showing lower GCase activity and were definitively diagnosed with GD by GBA gene analysis. The frequency of GD diagnosis through NBS was 1 in 77,720 when limited to the probands. This frequency is higher than that previously estimated in Japan. In the future, NBS for GD is expected to be performed in many regions of Japan and contribute to detecting more patients with GD. Early screening and diagnosis may have a very significant impact on the quality of life and potentially longevity in infants with GD. Newborn screening (NBS) identified 4 cases of Gaucher disease (GD) with few false positives in Japan. The frequency of GD diagnosis through NBS was 1 in 77,720, being higher than the previously estimated. Early diagnosis may have a very significant impact on the quality of life and potentially longevity in infants with GD.
Collapse
|
41
|
Fabry Disease in Slovakia: How the Situation Has Changed over 20 Years of Treatment. J Pers Med 2022; 12:jpm12060922. [PMID: 35743707 PMCID: PMC9224707 DOI: 10.3390/jpm12060922] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2022] [Revised: 05/19/2022] [Accepted: 05/27/2022] [Indexed: 11/17/2022] Open
Abstract
Fabry disease (FD, OMIM#301500) is a rare inborn error of the lysosomal enzyme α-galactosidase (α-Gal A, EC 3.2.1.22) and results in progressive substrate accumulation in tissues with a wide range of clinical presentations. Despite the X-linked inheritance, heterozygous females may also be affected. Hemizygous males are usually affected more severely, with an earlier manifestation of the symptoms. Rising awareness among health care professionals and more accessible diagnostics have positioned FD among the most-common inherited metabolic diseases in adults. An early and correct diagnosis of FD is crucial with a focus on personalised therapy. Preventing irreversible destruction of vital organs is the main goal of modern medicine. The aim of this study was to offer a complex report mapping the situation surrounding FD patients in Slovakia. A total of 48 patients (21 males, 27 females) with FD are registered in the Centre for Inborn Errors of Metabolism in Bratislava, Slovakia. In our cohort, we have identified three novel pathogenic variants in five patients. Three patients presented with the frameshift mutation c.736delA, and two others presented with the missense mutations c.203T>C, c.157A>C. Moreover, we present a new clinical picture of the pathogenic variant c.801+1G>A, which was previously described and associated with the renal phenotype.
Collapse
|
42
|
Weinreb NJ, Goker-Alpan O, Kishnani PS, Longo N, Burrow TA, Bernat JA, Gupta P, Henderson N, Pedro H, Prada CE, Vats D, Pathak RR, Wright E, Ficicioglu C. The diagnosis and management of Gaucher disease in pediatric patients: Where do we go from here? Mol Genet Metab 2022; 136:4-21. [PMID: 35367141 DOI: 10.1016/j.ymgme.2022.03.001] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Revised: 03/04/2022] [Accepted: 03/04/2022] [Indexed: 02/07/2023]
Abstract
Gaucher disease (GD) is an autosomal recessive inherited lysosomal storage disease that often presents in early childhood and is associated with damage to multiple organ systems. Many challenges associated with GD diagnosis and management arise from the considerable heterogeneity of disease presentations and natural history. Phenotypic classification has traditionally been based on the absence (in type 1 GD) or presence (in types 2 and 3 GD) of neurological involvement of varying severity. However, patient management and prediction of prognosis may be best served by a dynamic, evolving definition of individual phenotype rather than by a rigid system of classification. Patients may experience considerable delays in diagnosis, which can potentially be reduced by effective screening programs; however, program implementation can involve ethical and practical challenges. Variation in the clinical course of GD and an uncertain prognosis also complicate decisions concerning treatment initiation, with differing stakeholder perspectives around efficacy and acceptable cost/benefit ratio. We review the challenges faced by physicians in the diagnosis and management of GD in pediatric patients. We also consider future directions and goals, including acceleration of accurate diagnosis, improvements in the understanding of disease heterogeneity (natural history, response to treatment, and prognosis), the need for new treatments to address unmet needs for all forms of GD, and refinement of the tools for monitoring disease progression and treatment efficacy, such as specific biomarkers.
Collapse
Affiliation(s)
- Neal J Weinreb
- Department of Human Genetics, University of Miami Miller School of Medicine, Miami, FL, USA.
| | - Ozlem Goker-Alpan
- Lysosomal and Rare Disorders Research and Treatment Center, Fairfax, VA, USA.
| | - Priya S Kishnani
- Division of Medical Genetics, Department of Pediatrics, Duke University Medical Center, Durham, NC, USA.
| | - Nicola Longo
- Division of Medical Genetics, University of Utah, Salt Lake City, UT, USA.
| | - T Andrew Burrow
- Department of Pediatrics, University of Arkansas for Medical Sciences and Arkansas Children's Hospital, Little Rock, AR, USA.
| | - John A Bernat
- Division of Medical Genetics and Genomics, Stead Family Department of Pediatrics, University of Iowa, Iowa City, IA, USA.
| | - Punita Gupta
- St Joseph's University Hospital, Paterson, NJ, USA.
| | - Nadene Henderson
- Division of Genetic and Genomic Medicine, UPMC Children's Hospital of Pittsburgh, Pittsburgh, PA, USA.
| | - Helio Pedro
- Center for Genetic and Genomic Medicine, Hackensack University Medical Center, Hackensack, NJ, USA.
| | - Carlos E Prada
- Division of Genetics, Birth Defects & Metabolism, Ann & Robert H. Lurie Children's Hospital and Department of Pediatrics, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA.
| | - Divya Vats
- Kaiser Permanente Southern California, Los Angeles, CA, USA.
| | - Ravi R Pathak
- Takeda Pharmaceuticals USA, Inc., Lexington, MA, USA.
| | | | - Can Ficicioglu
- Division of Human Genetics and Metabolism, The Children's Hospital of Philadelphia, Philadelphia and the Perelman School of Medicine at the University of Pennsylvania, PA, USA.
| |
Collapse
|
43
|
Huggins E, Holland M, Case LE, Blount J, Landstrom AP, Jones HN, Kishnani PS. Early clinical phenotype of late onset Pompe disease: Lessons learned from newborn screening. Mol Genet Metab 2022; 135:179-185. [PMID: 35123877 DOI: 10.1016/j.ymgme.2022.01.003] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/14/2022] [Accepted: 01/14/2022] [Indexed: 01/14/2023]
Abstract
PURPOSE Thoroughly phenotype children with late-onset Pompe disease (LOPD) diagnosed via newborn screening (NBS) to provide guidance for long-term follow up. METHODS Twenty infants ages 6-21 months with LOPD diagnosed by NBS underwent systematic clinical evaluation at Duke University including cardiac imaging, biomarker testing, physical therapy evaluation, and speech-language pathology evaluation. RESULTS Of the 20 infants, four were homozygous for the "late-onset" IVS1 splice site variant c.-32-13 T > G, fourteen were compound heterozygous, and two did not have any copies of this variant. None of the patients had evidence of cardiomyopathy or cardiac rhythm disturbances. Biomarker testing showed an increase in CK, AST, and ALT in 8 patients (40%) and increase in Glc4 in two patients (10%). All patients demonstrated postural and kinematic concerns. Three patients (17%) scored below the 10%ile on the Alberta Infant Motor Scale (AIMS) and 15 patients (83%) scored above the 10%ile. Speech-language pathology assessments were normal in all patients and mild feeding/swallowing abnormalities were noted in nine patients (45%). CONCLUSION Our data show high variability among children with LOPD diagnosed via NBS. Careful physical therapy evaluation is necessary to monitor for subtle musculoskeletal signs that may reflect early muscle involvement. Patients should be monitored closely for symptom progression.
Collapse
Affiliation(s)
- Erin Huggins
- Division of Medical Genetics, Department of Pediatrics, Duke University School of Medicine, Durham, NC, USA
| | - Maggie Holland
- Department of Physical and Occupational Therapy, Duke University Health System, Durham, NC, USA
| | - Laura E Case
- Doctor of Physical Therapy Division, Department of Orthopedic Surgery, Duke University School of Medicine, Durham, NC, USA
| | - Janet Blount
- Division of Medical Genetics, Department of Pediatrics, Duke University School of Medicine, Durham, NC, USA
| | - Andrew P Landstrom
- Department of Pediatrics, Division of Cardiology and Department of Cell Biology, Duke University School of Medicine, Durham, NC, USA
| | - Harrison N Jones
- Department of Head and Neck Surgery & Communication Sciences, Duke University School of Medicine, Durham, NC, USA
| | - Priya S Kishnani
- Division of Medical Genetics, Department of Pediatrics, Duke University School of Medicine, Durham, NC, USA.
| |
Collapse
|
44
|
Kishnani PS, Al-Hertani W, Balwani M, Göker-Alpan Ö, Lau HA, Wasserstein M, Weinreb NJ, Grabowski G. Screening, patient identification, evaluation, and treatment in patients with Gaucher disease: Results from a Delphi consensus. Mol Genet Metab 2022; 135:154-162. [PMID: 34972655 DOI: 10.1016/j.ymgme.2021.12.009] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/27/2021] [Revised: 12/15/2021] [Accepted: 12/16/2021] [Indexed: 02/06/2023]
Abstract
Several guidelines are available for identification and management of patients with Gaucher disease, but the most recent guideline was published in 2013. Since then, there have been significant advances in newborn screening, phenotypic characterization, identification of biomarkers and their integration into clinical practice, and the development and approval of new treatment options. Accordingly, the goal of this Delphi consensus exercise was to extend prior initiatives of this type by addressing issues related to newborn screening, diagnostic evaluations, and treatment (both disease directed and adjunctive). The iterative Delphi process involved creation of an initial slate of statements, review by a steering committee, and three rounds of consensus development by an independent panel. A preliminary set of statements was developed by the supporting agency based on literature searches covering the period from 1965 to 2020. The Delphi process reduced an initial set of 185 statements to 65 for which there was unanimous support from the panel. The statements supported may ultimately provide a framework for more detailed treatment guidelines. In addition, the statements for which unanimous support could not be achieved help to identify evidence gaps that are targets for future research.
Collapse
Affiliation(s)
- Priya S Kishnani
- Duke University Medical Center, Department of Molecular Genetics and Microbiology, 905 Lasalle Street, GSRB1, 4th Floor, Durham, NC 27710, USA.
| | - Walla Al-Hertani
- Harvard Medical School, Boston Children's Hospital, Department of Pediatrics, Division of Genetics and Genomics, 300 Longwood Avenue, Boston, MA 02115, USA
| | - Manisha Balwani
- Icahn School of Medicine at Mount Sinai, Division of Medical Genetics and Genomics, Department of Genetics and Genomic Sciences, 1428 Madison Avenue, 1st Floor, New York, NY 10029, USA
| | - Özlem Göker-Alpan
- Lysosomal & Rare Disorders Research & Treatment Center, 3702 Pender Drive, Suite 170, Fairfax, VA 22030, USA
| | - Heather A Lau
- Ultragenyx Pharmaceutical Inc., Global Clinical Development, 840 Memorial Drive, Cambridge, MA 02139, USA
| | - Melissa Wasserstein
- The Children's Hospital at Montefiore and the Albert Einstein College of Medicine, Division of Pediatric Genetic Medicine, Departments of Pediatrics and Genetics, 3411 Wayne Ave, 9th Floor, Bronx, NY 10467, USA
| | - Neal J Weinreb
- University of Miami Miller School of Medicine, Departments of Human Genetics and Medicine, Hematology Division, 7367 Wexford Terrace, Boca Raton, FL 33433, USA
| | - Gregory Grabowski
- University of Cincinnati College of Medicine, Department of Pediatrics, and Department of Molecular Genetics, Biochemistry and Microbiology, Division of Human Genetics, Cincinnati Children's Hospital Research Foundation, Cincinnati, OH, USA
| |
Collapse
|
45
|
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
|
46
|
Prevalence of lysosomal storage disorders in Australia from 2009 to 2020. THE LANCET REGIONAL HEALTH. WESTERN PACIFIC 2022; 19:100344. [PMID: 35024668 PMCID: PMC8671750 DOI: 10.1016/j.lanwpc.2021.100344] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Abstract
Background Lysosomal storage disorders (LSD) are a family of genetic diseases that have a devastating impact on the patient and family with a concomitant health burden. Although considered rare disorders, improved diagnostic capabilities, newborn screening programs and public awareness has witnessed the frequency of many LSD increase considerably over recent years. To quantify their footprint, the number of LSD diagnosed in the multicultural Australian population in a 12-year period was determined. The principle objective was to yield contemporary prevalence figures to inform public health policies. Methods From the national referral laboratory for LSD diagnoses in Australia, retrospective data from patient referrals and prenatal testing for the period January 1 2009 to December 31 2020 were collated. Diagnosis was established biochemically by enzyme activity and/or metabolite determinations, as well as molecular genetic testing. The incidence of each disorder was determined by dividing the number of postnatal diagnoses by the number of births with prevalence including prenatal diagnoses. Findings During this 12-year period 766 diagnosis of LSD were confirmed inclusive of 32 prenatal outcomes representing 38 individual disorders. Total diagnosis per 100,000 live births averaged 21 per year (range 16 – 26) with Fabry disease the most prevalent representing 34% of all diagnoses in the current (up to 2020) report. Interpretation The combined prevalence of LSD for this study period at 1 per 4,800 live births is considerably higher than 1 per 7,700 reported for a 17-year period up to 1996. Additionally, more adults were diagnosed than children, implying that LSD are more common in adulthood than childhood. These data highlight the requirements for physicians to consider LSD in symptomatic adults and should refigure public health policies steering newborn screening programs in the direction of adult-onset conditions. Funding No funding was received for this study.
Collapse
|
47
|
Giraldo P, Andrade-Campos M. Novel Management and Screening Approaches for Haematological Complications of Gaucher's Disease. J Blood Med 2021; 12:1045-1056. [PMID: 34908889 PMCID: PMC8665828 DOI: 10.2147/jbm.s279756] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Accepted: 11/23/2021] [Indexed: 12/17/2022] Open
Abstract
Purpose Gaucher disease (GD) is the most common lysosomal storage disorder. The principal manifestations for its diagnosis and further monitoring include haematological manifestations such as anaemia, thrombocytopaenia, spleen enlargement, and bleeding disorders, among others. This review aims to summarise and update the role of haematological complications in GD diagnosis and follow-up, describe their management strategies, and to use these indicators as part of the diagnostic approach. Materials and Methods A systematic review following the recommendations of PRISMA-P 2020 was carried out. Publications indexed in the databases PubMed, Embase, Science Open, Mendeley, and Web of Science were electronically searched by three independent reviewers, and publications up to June 2021 were accessed. A total of 246 publications were initially listed, of which 129 were included for further review and analysis. Case reports were considered if they were representative of a relevant hematologic complication. Results From the first review dated in 1974 to the latest publication in 2021, including different populations confirmed that the haematological manifestations such as thrombocytopaenia and splenomegaly at diagnosis of GD type 1 are the most frequent features of the disease. The incorporation of haematological parameters to diagnosis strategies increases their cost-effectiveness. Hematologic parameters are part of the scoring system for disease assessment and the evaluation of therapeutic outcomes, providing reliable and accessible data to improve the management of GD. However, cytopaenia, underlying coagulation disorders, and platelet dysfunction need to be addressed, especially during pregnancy or surgery. Long-term haematological complications include the risk of neoplasia and immune impairment, an area of unmet need that is currently under research. Conclusion Haematological features are key for GD suspicion, diagnosis, and management. Normalization of hematological parameters is achieved with the treatment; however, there are unmet needs such as the underlying inflammatory status and the long-term risk of hematologic neoplasia.
Collapse
Affiliation(s)
- Pilar Giraldo
- Haematology, Hospital Quironsalud, Zaragoza, Spain.,Foundation FEETEG, Zaragoza, Spain
| | - Marcio Andrade-Campos
- Foundation FEETEG, Zaragoza, Spain.,Haematology Department, Hospital del Mar, Barcelona, Spain
| |
Collapse
|
48
|
Kingma SDK, Jonckheere AI. MPS I: Early diagnosis, bone disease and treatment, where are we now? J Inherit Metab Dis 2021; 44:1289-1310. [PMID: 34480380 DOI: 10.1002/jimd.12431] [Citation(s) in RCA: 2] [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: 07/01/2021] [Revised: 08/12/2021] [Accepted: 09/01/2021] [Indexed: 12/22/2022]
Abstract
Mucopolysaccharidosis type I (MPS I) is a lysosomal storage disorder characterized by α-L-iduronidase deficiency. Patients present with a broad spectrum of disease severity ranging from the most severe phenotype (Hurler) with devastating neurocognitive decline, bone disease and early death to intermediate (Hurler-Scheie) and more attenuated (Scheie) phenotypes, with a normal life expectancy. The most severely affected patients are preferably treated with hematopoietic stem cell transplantation, which halts the neurocognitive decline. Patients with more attenuated phenotypes are treated with enzyme replacement therapy. There are several challenges to be met in the treatment of MPS I patients. First, to optimize outcome, early recognition of the disease and clinical phenotype is needed to guide decisions on therapeutic strategies. Second, there is thus far no effective treatment available for MPS I bone disease. The pathophysiological mechanisms behind bone disease are largely unknown, limiting the development of effective therapeutic strategies. This article is a state of the art that comprehensively discusses three of the most urgent open issues in MPS I: early diagnosis of MPS I patients, pathophysiology of MPS I bone disease, and emerging therapeutic strategies for MPS I bone disease.
Collapse
Affiliation(s)
- Sandra D K Kingma
- Centre for Metabolic Diseases, University Hospital Antwerp, University of Antwerp, Edegem, Antwerp, Belgium
| | - An I Jonckheere
- Centre for Metabolic Diseases, University Hospital Antwerp, University of Antwerp, Edegem, Antwerp, Belgium
| |
Collapse
|
49
|
Sawada T, Kido J, Sugawara K, Nakamura K. High-Risk Screening for Fabry Disease: A Nationwide Study in Japan and Literature Review. Diagnostics (Basel) 2021; 11:diagnostics11101779. [PMID: 34679477 PMCID: PMC8534369 DOI: 10.3390/diagnostics11101779] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Revised: 09/23/2021] [Accepted: 09/24/2021] [Indexed: 12/26/2022] Open
Abstract
Fabry disease (FD) is an X-linked inherited disorder caused by mutations in the GLA gene, which encodes the lysosomal enzyme α-galactosidase A (α-Gal A). FD detection in patients at an early stage is essential to achieve sufficient treatment effects, and high-risk screening may be effective. Here, we performed high-risk screening for FD in Japan and showed that peripheral neurological manifestations are important in young patients with FD. Moreover, we reviewed the literature on high-risk screening in patients with renal, cardiac, and central neurological manifestations. Based on the results of this study and review of research abroad, we believe that FD can be detected more effectively by targeting individuals based on age. In recent years, the methods for high-risk screening have been ameliorated, and high-risk screening studies using GLA next-generation sequencing have been conducted. Considering the cost-effectiveness of screening, GLA sequencing should be performed in individuals with reduced α-Gal A activity and females with certain FD manifestations and/or a family history of FD. The findings suggest that family analysis would likely detect FD patients, although GLA sequencing of asymptomatic family members requires adequate genetic counseling.
Collapse
Affiliation(s)
- Takaaki Sawada
- Department of Pediatrics, Faculty of Life Sciences, Kumamoto University, 1-1-1 Honjo, Kumamoto City 860-8556, Japan; (T.S.); (K.S.); (K.N.)
- Department of Pediatrics, Graduate School of Medical Sciences, Kumamoto University, Kumamoto City 860-8556, Japan
| | - Jun Kido
- Department of Pediatrics, Faculty of Life Sciences, Kumamoto University, 1-1-1 Honjo, Kumamoto City 860-8556, Japan; (T.S.); (K.S.); (K.N.)
- Correspondence: ; Tel.: +81-096-373-5191; Fax: +81-096-373-5335
| | - Keishin Sugawara
- Department of Pediatrics, Faculty of Life Sciences, Kumamoto University, 1-1-1 Honjo, Kumamoto City 860-8556, Japan; (T.S.); (K.S.); (K.N.)
| | - Kimitoshi Nakamura
- Department of Pediatrics, Faculty of Life Sciences, Kumamoto University, 1-1-1 Honjo, Kumamoto City 860-8556, Japan; (T.S.); (K.S.); (K.N.)
| |
Collapse
|
50
|
Mak J, Cowan TM. Detecting lysosomal storage disorders by glycomic profiling using liquid chromatography mass spectrometry. Mol Genet Metab 2021; 134:43-52. [PMID: 34474962 PMCID: PMC9069563 DOI: 10.1016/j.ymgme.2021.08.006] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/03/2021] [Revised: 08/14/2021] [Accepted: 08/15/2021] [Indexed: 01/15/2023]
Abstract
BACKGROUND Urine and plasma biomarker testing for lysosomal storage disorders by liquid chromatography mass spectrometry (LC-MS) currently requires multiple analytical methods to detect the abnormal accumulation of oligosaccharides, mucopolysaccharides, and glycolipids. To improve clinical testing efficiency, we developed a single LC-MS method to simultaneously identify disorders of oligosaccharide, mucopolysaccharide, and glycolipid metabolism with minimal sample preparation. METHODS We created a single chromatographic method for separating free glycans and glycolipids in their native form, using an amide column and high pH conditions. We used this glycomic profiling method both in untargeted analyses of patient and control urines using LC ion-mobility high-resolution MS (biomarker discovery), and targeted analyses of urine, serum, and dried blood spot samples by LC-MS/MS (clinical validation). RESULTS Untargeted glycomic profiling revealed twenty biomarkers that could identify and subtype mucopolysaccharidoses. We incorporated these with known oligosaccharide and glycolipid biomarkers into a rapid test that identifies at least 27 lysosomal storage disorders, including oligosaccharidoses, mucopolysaccharidoses, sphingolipidoses, glycogen storage disorders, and congenital disorders of glycosylation and de-glycosylation. In a validation set containing 115 urine samples from patients with lysosomal storage disorders, all were unambiguously distinguished from normal controls, with correct disease subtyping for 88% (101/115) of cases. Glucosylsphingosine was reliably elevated in dried blood spots from Gaucher disease patients with baseline resolution from galactosylsphingosine. CONCLUSION Glycomic profiling by liquid chromatography mass spectrometry identifies a range of lysosomal storage disorders. This test can be used in clinical evaluations to rapidly focus a diagnosis, as well as to clarify or support additional gene sequencing and enzyme studies.
Collapse
Affiliation(s)
- Justin Mak
- Clinical Biochemical Genetics Laboratory, Stanford Health Care, United States of America.
| | - Tina M Cowan
- Clinical Biochemical Genetics Laboratory, Stanford Health Care, United States of America; Department of Pathology, Stanford University Medical Center, United States of America
| |
Collapse
|