51
|
The diagnostic utility of genome sequencing in a pediatric cohort with suspected mitochondrial disease. Genet Med 2020; 22:1254-1261. [DOI: 10.1038/s41436-020-0793-6] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2020] [Revised: 03/23/2020] [Accepted: 03/24/2020] [Indexed: 12/15/2022] Open
|
52
|
Fujishiro H, Hamao S, Isawa M, Himeno S. Segment-specific and direction-dependent transport of cadmium and manganese in immortalized S1, S2, and S3 cells derived from mouse kidney proximal tubules. J Toxicol Sci 2020; 44:611-619. [PMID: 31474742 DOI: 10.2131/jts.44.611] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Abstract
The kidney proximal tubule is a target of many renal toxicants, including cadmium (Cd), and also a place of reabsorption of essential metals in glomerular filtrate to systemic circulation. Although the mechanisms of metal transport in the convoluted proximal tubule (S1 and S2 segments) and the straight proximal tubule (S3 segment) may differ, little is known about the segment-specific modes of metal transport. Here, we utilized immortalized cell lines derived from the S1, S2, and S3 segments of mouse kidney proximal tubules, and examined the segment-specific and direction-dependent transport of Cd and manganese (Mn) using a trans-well culture system. The results showed that the uptakes of Cd2+ and Mn2+ from apical sides were the highest in S3 cells, and Cd2+, Mn2+, and Zn2+ mutually inhibited the apical uptake of each metal. As the expression of ZIP8, a zinc transporter having affinities for Cd2+ and Mn2+, was the highest in S3 cells, ZIP8 may contribute largely to the apical uptakes of these metals. The efficient uptake of Mn2+ from apical side of S3 cells may suggest an important role of ZIP8 in proximal tubule in reabsorption of Mn, an essential metal. Our study demonstrated that S1, S2, and S3 cells provide a useful tool for studying the segment-specific and direction-dependent transport of both toxic and essential metals in the kidney's proximal tubules.
Collapse
Affiliation(s)
- Hitomi Fujishiro
- Laboratory of Molecular Nutrition and Toxicology, Faculty of Pharmaceutical Sciences, Tokushima Bunri University
| | - Satoko Hamao
- Laboratory of Molecular Nutrition and Toxicology, Faculty of Pharmaceutical Sciences, Tokushima Bunri University
| | - Misaki Isawa
- Laboratory of Molecular Nutrition and Toxicology, Faculty of Pharmaceutical Sciences, Tokushima Bunri University
| | - Seiichiro Himeno
- Laboratory of Molecular Nutrition and Toxicology, Faculty of Pharmaceutical Sciences, Tokushima Bunri University
| |
Collapse
|
53
|
Pascoal C, Francisco R, Ferro T, Dos Reis Ferreira V, Jaeken J, Videira PA. CDG and immune response: From bedside to bench and back. J Inherit Metab Dis 2020; 43:90-124. [PMID: 31095764 DOI: 10.1002/jimd.12126] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/18/2019] [Revised: 05/13/2019] [Accepted: 05/15/2019] [Indexed: 12/20/2022]
Abstract
Glycosylation is an essential biological process that adds structural and functional diversity to cells and molecules, participating in physiological processes such as immunity. The immune response is driven and modulated by protein-attached glycans that mediate cell-cell interactions, pathogen recognition and cell activation. Therefore, abnormal glycosylation can be associated with deranged immune responses. Within human diseases presenting immunological defects are congenital disorders of glycosylation (CDG), a family of around 130 rare and complex genetic diseases. In this review, we have identified 23 CDG with immunological involvement, characterized by an increased propensity to-often life-threatening-infection. Inflammatory and autoimmune complications were found in 7 CDG types. CDG natural history(ies) and the mechanisms behind the immunological anomalies are still poorly understood. However, in some cases, alterations in pathogen recognition and intracellular signaling (eg, TGF-β1, NFAT, and NF-κB) have been suggested. Targeted therapies to restore immune defects are only available for PGM3-CDG and SLC35C1-CDG. Fostering research on glycoimmunology may elucidate the involved pathophysiological mechanisms and open new therapeutic avenues, thus improving CDG patients' quality of life.
Collapse
Affiliation(s)
- Carlota Pascoal
- Portuguese Association for CDG, Lisbon, Portugal
- CDG & Allies - Professionals and Patient Associations International Network (CDG & Allies - PPAIN), Caparica, Portugal
- UCIBIO, Departamento Ciências da Vida, Faculdade de Ciências e Tecnologia, Universidade NOVA de Lisboa, Caparica, Portugal
| | - Rita Francisco
- Portuguese Association for CDG, Lisbon, Portugal
- CDG & Allies - Professionals and Patient Associations International Network (CDG & Allies - PPAIN), Caparica, Portugal
- UCIBIO, Departamento Ciências da Vida, Faculdade de Ciências e Tecnologia, Universidade NOVA de Lisboa, Caparica, Portugal
| | - Tiago Ferro
- CDG & Allies - Professionals and Patient Associations International Network (CDG & Allies - PPAIN), Caparica, Portugal
- UCIBIO, Departamento Ciências da Vida, Faculdade de Ciências e Tecnologia, Universidade NOVA de Lisboa, Caparica, Portugal
| | - Vanessa Dos Reis Ferreira
- Portuguese Association for CDG, Lisbon, Portugal
- CDG & Allies - Professionals and Patient Associations International Network (CDG & Allies - PPAIN), Caparica, Portugal
| | - Jaak Jaeken
- CDG & Allies - Professionals and Patient Associations International Network (CDG & Allies - PPAIN), Caparica, Portugal
- Center for Metabolic Diseases, Department of Development and Regeneration, UZ and KU Leuven, Leuven, Belgium
| | - Paula A Videira
- Portuguese Association for CDG, Lisbon, Portugal
- CDG & Allies - Professionals and Patient Associations International Network (CDG & Allies - PPAIN), Caparica, Portugal
- UCIBIO, Departamento Ciências da Vida, Faculdade de Ciências e Tecnologia, Universidade NOVA de Lisboa, Caparica, Portugal
| |
Collapse
|
54
|
Bakulski KM, Seo YA, Hickman RC, Brandt D, Vadari HS, Hu H, KyunPark S. Heavy Metals Exposure and Alzheimer's Disease and Related Dementias. J Alzheimers Dis 2020; 76:1215-1242. [PMID: 32651318 PMCID: PMC7454042 DOI: 10.3233/jad-200282] [Citation(s) in RCA: 173] [Impact Index Per Article: 34.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Alzheimer's disease and related dementias lack effective treatment or cures and are major public health challenges. Risk for Alzheimer's disease and related dementias is partially attributable to environmental factors. The heavy metals lead, cadmium, and manganese are widespread and persistent in our environments. Once persons are exposed to these metals, they are adept at entering cells and reaching the brain. Lead and cadmium are associated with numerous health outcomes even at low levels of exposure. Although manganese is an essential metal, deficiency or environmental exposure or high levels of the metal can be toxic. In cell and animal model systems, lead, cadmium, and manganese are well documented neurotoxicants that contribute to canonical Alzheimer's disease pathologies. Adult human epidemiologic studies have consistently shown lead, cadmium, and manganese are associated with impaired cognitive function and cognitive decline. No longitudinal human epidemiology study has assessed lead or manganese exposure on Alzheimer's disease specifically though two studies have reported a link between cadmium and Alzheimer's disease mortality. More longitudinal epidemiologic studies with high-quality time course exposure data and incident cases of Alzheimer's disease and related dementias are warranted to confirm and estimate the proportion of risk attributable to these exposures. Given the widespread and global exposure to lead, cadmium, and manganese, even small increases in the risks of Alzheimer's disease and related dementias would have a major population impact on the burden on disease. This article reviews the experimental and epidemiologic literature of the associations between lead, cadmium, and manganese on Alzheimer's disease and related dementias and makes recommendations of critical areas of future investment.
Collapse
Affiliation(s)
- Kelly M. Bakulski
- Department of Epidemiology, School of Public Health, University of Michigan, Ann Arbor, MI, USA
| | - Young Ah Seo
- Department of Nutritional Sciences, School of Public Health, University of Michigan, Ann Arbor, MI, USA
| | - Ruby C. Hickman
- Department of Epidemiology, School of Public Health, University of Michigan, Ann Arbor, MI, USA
| | - Daniel Brandt
- Department of Epidemiology, School of Public Health, University of Michigan, Ann Arbor, MI, USA
| | - Harita S. Vadari
- Department of Epidemiology, School of Public Health, University of Michigan, Ann Arbor, MI, USA
| | - Howard Hu
- School of Public Health, University of Washington, Seattle, WA, USA
| | - Sung KyunPark
- Department of Epidemiology, School of Public Health, University of Michigan, Ann Arbor, MI, USA
- Department of Environmental Health Sciences, School of Public Health, University of Michigan, Ann Arbor, MI, USA
| |
Collapse
|
55
|
Fujishiro H, Himeno S. New Insights into the Roles of ZIP8, a Cadmium and Manganese Transporter, and Its Relation to Human Diseases. Biol Pharm Bull 2019; 42:1076-1082. [PMID: 31257283 DOI: 10.1248/bpb.b18-00637] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
ZIP8, a Zrt-/Irt-related protein encoded by Slc39A8, was originally discovered as a zinc transporter, but since then its roles as a transporter for cadmium (Cd) and manganese (Mn) have also been well characterized. ZIP8 is highly expressed in the S3 segment of the proximal tubules of the mouse kidney and may play a significant role in reabsorption of both toxic Cd and essential Mn from the lumen to the epithelial cells of the proximal tubule. In recent years, associations between various human diseases and genetic variations of ZIP8 have been reported. Missense mutations in the human SLC39A8 gene are associated with serious disorders of Mn metabolism, showing symptoms similar to congenital glycosylation deficiency. Enhanced excretion of Mn via bile or urine might be the cause of extremely low blood Mn levels in ZIP8-mutated patients, leading to the defects in Mn-dependent glycosylation. Several genome-wide association studies have demonstrated the associations of multiple diseases and ZIP8 SNPs constituting missense mutations. These findings suggest that ZIP8 plays more important roles than previously expected as a modulator of Mn homeostasis in the body. Elucidation of biochemical mechanisms regarding the metal-transporting ability of ZIP8 and its alteration by mutation is required for better understanding of the role of ZIP8 in human diseases.
Collapse
Affiliation(s)
- Hitomi Fujishiro
- Laboratory of Molecular Nutrition and Toxicology, Faculty of Pharmaceutical Sciences, Tokushima Bunri University
| | - Seiichiro Himeno
- Laboratory of Molecular Nutrition and Toxicology, Faculty of Pharmaceutical Sciences, Tokushima Bunri University
| |
Collapse
|
56
|
Thompson KJ, Wessling-Resnick M. ZIP14 is degraded in response to manganese exposure. Biometals 2019; 32:829-843. [PMID: 31541377 PMCID: PMC7755095 DOI: 10.1007/s10534-019-00216-1] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2019] [Accepted: 09/10/2019] [Indexed: 12/16/2022]
Abstract
Manganese (Mn) is an essential element necessary for proper development and brain function. Circulating Mn levels are regulated by hepatobiliary clearance to limit toxic levels and prevent tissue deposition. To characterize mechanisms involved in hepatocyte Mn uptake, polarized human HepaRG cells were used for this study. Western blot analysis and immunofluorescence microscopy showed the Mn transporter ZIP14 was expressed and localized to the basolateral surface of polarized HepaRG cells. HepaRG cells took up 54Mn in a time- and temperature-dependent manner but uptake was reduced after exposure to Mn. This loss in transport activity was associated with decreased ZIP14 protein levels in response to Mn exposure. Mn-induced degradation of ZIP14 was blocked by bafilomycin A1, which increased localization of the transporter in Lamp1-positive vesicles. Mn exposure also down-regulated the Golgi proteins TMEM165 and GPP130 while the ER stress marker BiP was induced. These results indicate that Mn exposure decreases ZIP14 protein levels to limit subsequent uptake of Mn as a cytoprotective response. Thus, high levels of Mn may compromise first-pass-hepatic clearance mechanisms.
Collapse
Affiliation(s)
- Khristy J Thompson
- Department of Genetics and Complex Diseases, Harvard T.H. Chan School of Public Health, 665 Huntington Avenue, Boston, MA, 02115, USA.
| | - Marianne Wessling-Resnick
- Department of Genetics and Complex Diseases, Harvard T.H. Chan School of Public Health, 665 Huntington Avenue, Boston, MA, 02115, USA
| |
Collapse
|
57
|
Rius R, Van Bergen NJ, Compton AG, Riley LG, Kava MP, Balasubramaniam S, Amor DJ, Fanjul-Fernandez M, Cowley MJ, Fahey MC, Koenig MK, Enns GM, Sadedin S, Wilson MJ, Tan TY, Thorburn DR, Christodoulou J. Clinical Spectrum and Functional Consequences Associated with Bi-Allelic Pathogenic PNPT1 Variants. J Clin Med 2019; 8:jcm8112020. [PMID: 31752325 PMCID: PMC6912252 DOI: 10.3390/jcm8112020] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2019] [Revised: 11/11/2019] [Accepted: 11/14/2019] [Indexed: 02/01/2023] Open
Abstract
PNPT1 (PNPase—polynucleotide phosphorylase) is involved in multiple RNA processing functions in the mitochondria. Bi-allelic pathogenic PNPT1 variants cause heterogeneous clinical phenotypes affecting multiple organs without any established genotype–phenotype correlations. Defects in PNPase can cause variable combined respiratory chain complex defects. Recently, it has been suggested that PNPase can lead to activation of an innate immune response. To better understand the clinical and molecular spectrum of patients with bi-allelic PNPT1 variants, we captured detailed clinical and molecular phenotypes of all 17 patients reported in the literature, plus seven new patients, including a 78-year-old male with the longest reported survival. A functional follow-up of genomic sequencing by cDNA studies confirmed a splicing defect in a novel, apparently synonymous, variant. Patient fibroblasts showed an accumulation of mitochondrial unprocessed PNPT1 transcripts, while blood showed an increased interferon response. Our findings suggest that functional analyses of the RNA processing function of PNPase are more sensitive than testing downstream defects in oxidative phosphorylation (OXPHPOS) enzyme activities. This research extends our knowledge of the clinical and functional consequences of bi-allelic pathogenic PNPT1 variants that may guide management and further efforts into understanding the pathophysiological mechanisms for therapeutic development.
Collapse
Affiliation(s)
- Rocio Rius
- Murdoch Children’s Research Institute, Melbourne, VIC 3052, Australia
- Department of Paediatrics, University of Melbourne, Melbourne, VIC 3052, Australia
| | - Nicole J. Van Bergen
- Murdoch Children’s Research Institute, Melbourne, VIC 3052, Australia
- Department of Paediatrics, University of Melbourne, Melbourne, VIC 3052, Australia
| | - Alison G. Compton
- Murdoch Children’s Research Institute, Melbourne, VIC 3052, Australia
- Department of Paediatrics, University of Melbourne, Melbourne, VIC 3052, Australia
| | - Lisa G. Riley
- Kids Research, The Children’s Hospital at Westmead, Sydney, NSW 2145, Australia
- Discipline of Child & Adolescent Health, Sydney Medical School, University of Sydney, Sydney, NSW 2050, Australia
| | - Maina P. Kava
- Department of Neurology, Perth Children’s Hospital, Perth, WA 6009, Australia
- Department of Metabolic Medicine and Rheumatology, Perth Children’s Hospital, Perth, WA 6009, Australia
| | - Shanti Balasubramaniam
- Department of Metabolic Medicine and Rheumatology, Perth Children’s Hospital, Perth, WA 6009, Australia
- Genetic Metabolic Disorders Service, Western Sydney Genetics Program, The Children’s Hospital at Westmead, Sydney, NSW 2145, Australia
- Discipline of Genetic Medicine, Sydney Medical School, University of Sydney, Sydney, NSW 2145, Australia
| | - David J. Amor
- Murdoch Children’s Research Institute, Melbourne, VIC 3052, Australia
- Department of Paediatrics, University of Melbourne, Melbourne, VIC 3052, Australia
- Victorian Clinical Genetic Services, Melbourne, VIC 3052, Australia
| | - Miriam Fanjul-Fernandez
- Department of Paediatrics, University of Melbourne, Melbourne, VIC 3052, Australia
- Victorian Clinical Genetic Services, Melbourne, VIC 3052, Australia
| | - Mark J. Cowley
- Precision Medicine Theme, Children’s Cancer Institute, Kensington, NSW 2750, Australia
- Kinghorn Centre for Clinical Genomics, Garvan Institute, University of New South Wales, Randwick, NSW 2010, Australia
- School of Women’s and Children’s Health, University of New South Wales, Randwick, NSW 2031, Australia
| | - Michael C. Fahey
- Department of Paediatrics, Monash University, Melbourne, VIC 3168, Australia
| | - Mary K. Koenig
- The University of Texas McGovern Medical School, Houston, TX 77030, USA
| | - Gregory M. Enns
- Department of Pediatrics, Division of Medical Genetics, Stanford University, Stanford, CA 94305, USA
| | - Simon Sadedin
- Murdoch Children’s Research Institute, Melbourne, VIC 3052, Australia
- Victorian Clinical Genetic Services, Melbourne, VIC 3052, Australia
| | - Meredith J. Wilson
- Department of Clinical Genetics, The Children’s Hospital at Westmead, Sydney, NSW 2145, Australia
- Discipline of Genomic Medicine, Faculty of Medicine and Health, University of Sydney, Sydney, NSW 2006, Australia
| | - Tiong Y. Tan
- Murdoch Children’s Research Institute, Melbourne, VIC 3052, Australia
- Department of Paediatrics, University of Melbourne, Melbourne, VIC 3052, Australia
- Victorian Clinical Genetic Services, Melbourne, VIC 3052, Australia
| | - David R. Thorburn
- Murdoch Children’s Research Institute, Melbourne, VIC 3052, Australia
- Department of Paediatrics, University of Melbourne, Melbourne, VIC 3052, Australia
- Victorian Clinical Genetic Services, Melbourne, VIC 3052, Australia
| | - John Christodoulou
- Murdoch Children’s Research Institute, Melbourne, VIC 3052, Australia
- Department of Paediatrics, University of Melbourne, Melbourne, VIC 3052, Australia
- Discipline of Child & Adolescent Health, Sydney Medical School, University of Sydney, Sydney, NSW 2050, Australia
- Victorian Clinical Genetic Services, Melbourne, VIC 3052, Australia
- Correspondence: ; Tel.: +613-9936-6353
| |
Collapse
|
58
|
Steimle BL, Smith FM, Kosman DJ. The solute carriers ZIP8 and ZIP14 regulate manganese accumulation in brain microvascular endothelial cells and control brain manganese levels. J Biol Chem 2019; 294:19197-19208. [PMID: 31699897 DOI: 10.1074/jbc.ra119.009371] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2019] [Revised: 10/29/2019] [Indexed: 12/29/2022] Open
Abstract
Manganese supports numerous neuronal functions but in excess is neurotoxic. Consequently, regulation of manganese flux at the blood-brain barrier (BBB) is critical to brain homeostasis. However, the molecular pathways supporting the transcellular trafficking of divalent manganese ions within the microvascular capillary endothelial cells (BMVECs) that constitute the BBB have not been examined. In this study, we have determined that ZIP8 and ZIP14 (Zrt- and Irt-like proteins 8 and 14) support Mn2+ uptake by BMVECs and that neither DMT1 nor an endocytosis-dependent pathway play any significant role in Mn2+ uptake. Specifically, siRNA-mediated knockdown of ZIP8 and ZIP14 coincided with a decrease in manganese uptake, and kinetic analyses revealed that manganese uptake depends on pH and bicarbonate and is up-regulated by lipopolysaccharide, all biochemical markers of ZIP8 or ZIP14 activity. Mn2+ uptake also was associated with cell-surface membrane presentation of ZIP8 and ZIP14, as indicated by membrane protein biotinylation. Importantly, surface ZIP8 and ZIP14 biotinylation and Mn2+-uptake experiments together revealed that these transporters support manganese uptake at both the apical, blood and basal, brain sides of BMVECs. This indicated that in the BMVECs of the BBB, these two transporters support a bidirectional Mn2+ flux. We conclude that BMVECs play a critical role in controlling manganese homeostasis in the brain.
Collapse
Affiliation(s)
- Brittany L Steimle
- Department of Biochemistry, State University of New York at Buffalo, Jacobs School of Medicine and Biomedical Sciences, Buffalo, New York 14203
| | - Frances M Smith
- Department of Biochemistry, State University of New York at Buffalo, Jacobs School of Medicine and Biomedical Sciences, Buffalo, New York 14203
| | - Daniel J Kosman
- Department of Biochemistry, State University of New York at Buffalo, Jacobs School of Medicine and Biomedical Sciences, Buffalo, New York 14203
| |
Collapse
|
59
|
Gayevskiy V, Roscioli T, Dinger ME, Cowley MJ. Seave: a comprehensive web platform for storing and interrogating human genomic variation. Bioinformatics 2019; 35:122-125. [PMID: 30561546 PMCID: PMC6298057 DOI: 10.1093/bioinformatics/bty540] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2018] [Accepted: 06/28/2018] [Indexed: 01/06/2023] Open
Abstract
Motivation Genome sequencing has had a remarkable impact on our ability to study the effects of human genetic variation, however, variant interpretation remains the major bottleneck. Understanding the potential impact of variants, including structural variants, requires extensive annotation from disparate sources of knowledge, and in silico prediction algorithms. Results We introduce Seave, an intuitive web platform that enables all types of variants to be securely stored, annotated and filtered. Variants are annotated with allele frequencies and pathogenicity assessments from many popular databases and in silico pathogenicity prediction scores. Seave enables filtering of variants with specific inheritance patterns, including somatic variants, by quality, allele frequencies and gene lists which can be curated and saved. Seave was made for whole genome data and is capable of storing and querying copy number and structural variants. Availability and implementation To demo Seave with public data, see https://www.seave.bio. Source code is available at http://code.seave.bio and extensive documentation is available at http://documentation.seave.bio. Seave can be locally installed on an Apache server with PHP and MySQL, or we provide an Amazon Machine Image for quick deployment. For commercial and clinical diagnostic licensing, contact the corresponding author. Supplementary information Supplementary data are available at Bioinformatics online.
Collapse
Affiliation(s)
- Velimir Gayevskiy
- Kinghorn Centre for Clinical Genomics, Garvan Institute of Medical Research, Darlinghurst, NSW, Australia
| | - Tony Roscioli
- Centre for Clinical Genetics, Sydney Children's Hospital, Randwick, NSW, Australia.,Prince of Wales Clinical School, University of New South Wales, UNSW Sydney, NSW, Australia.,Neuroscience Research Australia, University of New South Wales, UNSW Sydney, NSW, Australia
| | - Marcel E Dinger
- Kinghorn Centre for Clinical Genomics, Garvan Institute of Medical Research, Darlinghurst, NSW, Australia.,St Vincent's Clinical School, University of New South Wales, UNSW Sydney, NSW, Australia.,Genome.One, Darlinghurst, NSW, Australia
| | - Mark J Cowley
- Kinghorn Centre for Clinical Genomics, Garvan Institute of Medical Research, Darlinghurst, NSW, Australia.,St Vincent's Clinical School, University of New South Wales, UNSW Sydney, NSW, Australia.,Children's Cancer Institute, UNSW Sydney, NSW, Australia
| |
Collapse
|
60
|
Nebert DW, Liu Z. SLC39A8 gene encoding a metal ion transporter: discovery and bench to bedside. Hum Genomics 2019; 13:51. [PMID: 31521203 PMCID: PMC6744627 DOI: 10.1186/s40246-019-0233-3] [Citation(s) in RCA: 72] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2019] [Accepted: 08/14/2019] [Indexed: 01/08/2023] Open
Abstract
SLC39A8 is an evolutionarily highly conserved gene that encodes the ZIP8 metal cation transporter in all vertebrates. SLC39A8 is ubiquitously expressed, including pluripotent embryonic stem cells; SLC39A8 expression occurs in every cell type examined. Uptake of ZIP8-mediated Mn2+, Zn2+, Fe2+, Se4+, and Co2+ represents endogenous functions-moving these cations into the cell. By way of mouse genetic differences, the phenotype of "subcutaneous cadmium-induced testicular necrosis" was assigned to the Cdm locus in the 1970s. This led to identification of the mouse Slc39a8 gene, its most closely related Slc39a14 gene, and creation of Slc39a8-overexpressing, Slc39a8(neo/neo) knockdown, and cell type-specific conditional knockout mouse lines; the Slc39a8(-/-) global knockout mouse is early-embryolethal. Slc39a8(neo/neo) hypomorphs die between gestational day 16.5 and postnatal day 1-exhibiting severe anemia, dysregulated hematopoiesis, hypoplastic spleen, dysorganogenesis, stunted growth, and hypomorphic limbs. Not surprisingly, genome-wide association studies subsequently revealed human SLC39A8-deficiency variants exhibiting striking pleiotropy-defects correlated with clinical disorders in virtually every organ, tissue, and cell-type: numerous developmental and congenital disorders, the immune system, cardiovascular system, kidney, lung, liver, coagulation system, central nervous system, musculoskeletal system, eye, and gastrointestinal tract. Traits with which SLC39A8-deficiency variants are currently associated include Mn2+-deficient hypoglycosylation; numerous birth defects; Leigh syndrome-like mitochondrial redox deficiency; decreased serum high-density lipoprotein-cholesterol levels; increased body mass index; greater risk of coronary artery disease, hypotension, cardiovascular death, allergy, ischemic stroke, schizophrenia, Parkinson disease, inflammatory bowel disease, Crohn disease, myopia, and adolescent idiopathic scoliosis; systemic lupus erythematosus with primary Sjögren syndrome; decreased height; and inadvertent participation in the inflammatory progression of osteoarthritis.
Collapse
Affiliation(s)
- Daniel W Nebert
- Department of Environmental Health and Center for Environmental Genetics, University of Cincinnati Medical Center, Cincinnati, OH, 45267-0056, USA.
- Division of Human Genetics, Department of Pediatrics & Molecular Developmental Biology, Cincinnati Children's Hospital, Cincinnati, OH, 45229-2899, USA.
| | - Zijuan Liu
- Department of Biological Sciences, Oakland University, Rochester, MI, 48309, USA
| |
Collapse
|
61
|
Jinnah H, Sun YV. Dystonia genes and their biological pathways. Neurobiol Dis 2019; 129:159-168. [DOI: 10.1016/j.nbd.2019.05.014] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2019] [Revised: 05/05/2019] [Accepted: 05/17/2019] [Indexed: 12/27/2022] Open
|
62
|
Sachse B, Kolbaum AE, Ziegenhagen R, Andres S, Berg K, Dusemund B, Hirsch-Ernst KI, Kappenstein O, Müller F, Röhl C, Lindtner O, Lampen A, Schäfer B. Dietary Manganese Exposure in the Adult Population in Germany-What Does it Mean in Relation to Health Risks? Mol Nutr Food Res 2019; 63:e1900065. [PMID: 31216097 DOI: 10.1002/mnfr.201900065] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2019] [Revised: 04/29/2019] [Indexed: 11/10/2022]
Abstract
Manganese is both an essential nutrient and a potential neurotoxicant. Therefore, the question arises whether the dietary manganese intake in the German population is on the low or high side. Results from a pilot total diet study in Germany presented here reveal that the average dietary manganese intake in the general population in Germany aged 14-80 years is about 2.8 mg day-1 for a person of 70 kg body weight. This exposure level is within the intake range of 2-5 mg per person and day as recommended by the societies for nutrition in Germany, Austria, and Switzerland. No information on the dietary exposure of children in Germany can be provided so far. Although reliable information on health effects related to oral manganese exposure is limited, there is no indication from the literature that these dietary intake levels are associated with adverse health effects either by manganese deficiency or excess. However, there is limited evidence that manganese taken up as a highly bioavailable bolus, for example, uptake via drinking water or food supplements, could pose a potential risk to human health-particularly in certain subpopulations-when certain intake amounts, which are currently not well defined, are exceeded.
Collapse
Affiliation(s)
- Benjamin Sachse
- German Federal Institute for Risk Assessment (BfR), Department of Food Safety, 10589, Berlin, Germany
| | - Anna Elena Kolbaum
- German Federal Institute for Risk Assessment (BfR), Department of Exposure, 12277, Berlin, Germany
| | - Rainer Ziegenhagen
- German Federal Institute for Risk Assessment (BfR), Department of Food Safety, 10589, Berlin, Germany
| | - Susanne Andres
- German Federal Institute for Risk Assessment (BfR), Department of Food Safety, 10589, Berlin, Germany
| | - Katharina Berg
- German Federal Institute for Risk Assessment (BfR), Department of Exposure, 12277, Berlin, Germany
| | - Birgit Dusemund
- German Federal Institute for Risk Assessment (BfR), Department of Food Safety, 10589, Berlin, Germany
| | - Karen Ildico Hirsch-Ernst
- German Federal Institute for Risk Assessment (BfR), Department of Food Safety, 10589, Berlin, Germany
| | - Oliver Kappenstein
- German Federal Institute for Risk Assessment (BfR), Department of Chemicals and Product Safety, 10589, Berlin, Germany
| | - Frederic Müller
- German Federal Institute for Risk Assessment (BfR), Department of Chemicals and Product Safety, 10589, Berlin, Germany
| | - Claudia Röhl
- German Federal Institute for Risk Assessment (BfR), Department of Food Safety, 10589, Berlin, Germany.,State Agency for Social Services Schleswig-Holstein (LAsD), Department of Environmental Health Protection, 24105, Kiel, Germany
| | - Oliver Lindtner
- German Federal Institute for Risk Assessment (BfR), Department of Exposure, 12277, Berlin, Germany
| | - Alfonso Lampen
- German Federal Institute for Risk Assessment (BfR), Department of Food Safety, 10589, Berlin, Germany
| | - Bernd Schäfer
- German Federal Institute for Risk Assessment (BfR), Department of Food Safety, 10589, Berlin, Germany
| |
Collapse
|
63
|
Scheiber IF, Alarcon NO, Zhao N. Manganese Uptake by A549 Cells is Mediated by Both ZIP8 and ZIP14. Nutrients 2019; 11:nu11071473. [PMID: 31261654 PMCID: PMC6682971 DOI: 10.3390/nu11071473] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2019] [Revised: 06/20/2019] [Accepted: 06/26/2019] [Indexed: 12/16/2022] Open
Abstract
The alveolar epithelia of the lungs require manganese (Mn) as an essential nutrient, but also provide an entry route for airborne Mn that can cause neurotoxicity. Transporters involved in Mn uptake by alveolar epithelial cells are unknown. Recently, two members of the Zrt- and Irt-like protein (ZIP) family of metal transporters, ZIP8 and ZIP14, have been identified as crucial Mn importers in vivo. ZIP8 is by far most abundantly expressed in the lungs, whereas ZIP14 expression in the lungs is low compared to other tissues. We hypothesized that Mn uptake by alveolar epithelial cells is primarily mediated by ZIP8. To test our hypothesis, we used A549 cells, a type II alveolar cell line. Mirroring the in vivo situation, A549 cells expressed higher levels of ZIP8 than cell models for the liver, intestines, and kidney. Quantification of ZIP8 and ZIP14 revealed a strong enrichment of ZIP8 over ZIP14 in A549 cells. Using siRNA technology, we identified ZIP8 and ZIP14 as the major transporters mediating Mn uptake by A549 cells. To our surprise, knockdown of either ZIP8 or ZIP14 impaired Mn accumulation to a similar extent, which we traced back to similar amounts of ZIP8 and ZIP14 at the plasma membrane. Our study highlights the importance of both ZIP8 and ZIP14 in Mn metabolism of alveolar epithelial cells.
Collapse
Affiliation(s)
- Ivo F Scheiber
- Department of Nutritional Sciences, The University of Arizona, Tucson, AZ 85721, USA
| | | | - Ningning Zhao
- Department of Nutritional Sciences, The University of Arizona, Tucson, AZ 85721, USA.
| |
Collapse
|
64
|
Abstract
Purpose of Review This article provides an overview of the pathogenesis, clinical presentation and treatment of inherited manganese transporter defects. Recent Findings Identification of a new group of manganese transportopathies has greatly advanced our understanding of how manganese homeostasis is regulated in vivo. While the manganese efflux transporter SLC30A10 and the uptake transporter SLC39A14 work synergistically to reduce the manganese load, SLC39A8 has an opposing function facilitating manganese uptake into the organism. Bi-allelic mutations in any of these transporter proteins disrupt the manganese equilibrium and lead to neurological disease: Hypermanganesaemia with dystonia 1 (SLC30A10 deficiency) and hypermanganesaemia with dystonia 2 (SLC39A14 deficiency) are characterised by manganese neurotoxicity while SLC39A8 mutations cause a congenital disorder of glycosylation type IIn due to Mn deficiency. Summary Inherited manganese transporter defects are an important differential diagnosis of paediatric movement disorders. Manganese blood levels and MRI brain are diagnostic and allow early diagnosis to avoid treatment delay.
Collapse
Affiliation(s)
- S Anagianni
- Department of Cell and Developmental Biology, University College London, Gower Street, WC1E 6BT, London, UK
| | - K Tuschl
- Department of Cell and Developmental Biology, University College London, Gower Street, WC1E 6BT, London, UK. .,Department of Developmental Neurobiology, King's College London, New Hunt's House, Guy's Campus, London, SE1 1UL, UK. .,UCL GOS Institute of Child Health, 30 Guilford Street, London,, WC1N 1EH, UK.
| |
Collapse
|
65
|
Affiliation(s)
- Luke Tillman
- Genetics and Complex Diseases, Harvard T.H. Chan School of Public Health, Harvard University, Boston, MA, USA
| |
Collapse
|
66
|
Abstract
Congenital disorders of glycosylation (CDG) is a genetically heterogeneous and clinically polymorphic group of diseases caused by defects in various enzymes, the synthesis and processing of N-linked glycans or oligosaccharides into glycoproteins. Approximately half of all proteins expressed in cells are glycosylated to achieve their full functionality. Basically there are 2 variants of glycosylation: N-glycosylation and O-glycosylation. N-glycans are bound to the amide group of aspartine, whereas O-glycans are bonded to the hydroxyl group of serine or threonine. Synthesis of N-glycans occurs in 3 stages: the formation of nucleotide-linked sugars, assembly (in the cytosol and endoplasmic reticulum) and treatment (in the Golgi apparatus). Synthesis of O-glycans occurs mainly in the Golgi apparatus. The most frequently identified types of CDG are associated with a defect in the N-glycosylation pathway. CDGs are typically multisystem disorders with varying clinical manifestations such as hepatomegaly, cholestasis, liver failure, developmental delay, hypotonia, convulsions, facial dysmorphism and gastrointestinal disorders. Also histological findings showed liver fibrosis, malformation of the ducts, cirrhosis, and steatosis. CDGs typically present in the first months of life, and about 20% of patients do not survive to 5 years. The first line of CDG screening is based on the analysis of N-glycosylation of transf ferin. Exome sequencing or targeted gene panel is used for diagnosis. Several CDG subtypes are amenable to teraphy with mannose and galactose.
Collapse
|
67
|
Thompson KJ, Hein J, Baez A, Sosa JC, Wessling-Resnick M. Manganese transport and toxicity in polarized WIF-B hepatocytes. Am J Physiol Gastrointest Liver Physiol 2018; 315:G351-G363. [PMID: 29792530 PMCID: PMC6335010 DOI: 10.1152/ajpgi.00103.2018] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Manganese (Mn) toxicity arises from nutritional problems, community and occupational exposures, and genetic risks. Mn blood levels are controlled by hepatobiliary clearance. The goals of this study were to determine the cellular distribution of Mn transporters in polarized hepatocytes, to establish an in vitro assay for hepatocyte Mn efflux, and to examine possible roles the Mn transporters would play in metal import and export. For these experiments, hepatocytoma WIF-B cells were grown for 12-14 days to achieve maximal polarity. Immunoblots showed that Mn transporters ZIP8, ZnT10, ferroportin (Fpn), and ZIP14 were present. Indirect immunofluorescence microscopy localized Fpn and ZIP14 to WIF-B cell basolateral domains whereas ZnT10 and ZIP8 associated with intracellular vesicular compartments. ZIP8-positive structures were distributed uniformly throughout the cytoplasm, but ZnT10-positive vesicles were adjacent to apical bile compartments. WIF-B cells were sensitive to Mn toxicity, showing decreased viability after 16 h exposure to >250 μM MnCl2. However, the hepatocytes were resistant to 4-h exposures of up to 500 μM MnCl2 despite 50-fold increased Mn content. Washout experiments showed time-dependent efflux with 80% Mn released after a 4 h chase period. Hepcidin reduced levels of Fpn in WIF-B cells, clearing Fpn from the cell surface, but Mn efflux was unaffected. The secretory inhibitor, brefeldin A, did block release of Mn from WIF-B cells, suggesting vesicle fusion may be involved in export. These results point to a possible role of ZnT10 to import Mn into vesicles that subsequently fuse with the apical membrane and empty their contents into bile. NEW & NOTEWORTHY Polarized WIF-B hepatocytes express manganese (Mn) transporters ZIP8, ZnT10, ferroportin (Fpn), and ZIP14. Fpn and ZIP14 localize to basolateral domains. ZnT10-positive vesicles were adjacent to apical bile compartments, and ZIP8-positive vesicles were distributed uniformly throughout the cytoplasm. WIF-B hepatocyte Mn export was resistant to hepcidin but inhibited by brefeldin A, pointing to an efflux mechanism involving ZnT10-mediated uptake of Mn into vesicles that subsequently fuse with and empty their contents across the apical bile canalicular membrane.
Collapse
Affiliation(s)
- Khristy J. Thompson
- Department of Genetics and Complex Diseases, Harvard T. H. Chan School of Public Health, Harvard University, Boston, Massachusetts
| | - Jennifer Hein
- Department of Genetics and Complex Diseases, Harvard T. H. Chan School of Public Health, Harvard University, Boston, Massachusetts
| | - Andrew Baez
- Department of Genetics and Complex Diseases, Harvard T. H. Chan School of Public Health, Harvard University, Boston, Massachusetts
| | - Jose Carlo Sosa
- Department of Genetics and Complex Diseases, Harvard T. H. Chan School of Public Health, Harvard University, Boston, Massachusetts
| | - Marianne Wessling-Resnick
- Department of Genetics and Complex Diseases, Harvard T. H. Chan School of Public Health, Harvard University, Boston, Massachusetts
| |
Collapse
|
68
|
Chen J, Gálvez-Peralta M, Zhang X, Deng J, Liu Z, Nebert DW. In utero gene expression in the Slc39a8(neo/neo) knockdown mouse. Sci Rep 2018; 8:10703. [PMID: 30013175 PMCID: PMC6048144 DOI: 10.1038/s41598-018-29109-y] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2018] [Accepted: 07/05/2018] [Indexed: 12/13/2022] Open
Abstract
Slc39a8 encodes ZIP8, a divalent cation/bicarbonate symporter expressed in pluripotent mouse embryonic stem cells, and therefore ubiquitous in adult tissues; ZIP8 influxes Zn2+, Mn2+ and Fe2+. Slc39a8(neo/neo) knockdown mice exhibit 10-15% of wild-type ZIP8 mRNA and protein levels, and show pleiotropic phenotype of stunted growth, neonatal lethality, multi-organ dysmorphogenesis, and dysregulated hematopoiesis manifested as severe anemia. Herein we performed RNA-seq analysis of gestational day (GD)13.5 yolk sac and placenta, and GD16.5 liver, kidney, lung, heart and cerebellum, comparing Slc39a8(neo/neo) with Slc39a8(+/+) wild-type. Meta-data analysis of differentially-expressed genes revealed 29 unique genes from all tissues - having enriched GO categories associated with hematopoiesis and hypoxia and KEGG categories of complement, response to infection, and coagulation cascade - consistent with dysregulated hematopoietic stem cell fate. Based on transcription factor (TF) profiles in the JASPAR database, and searching for TF-binding sites enriched by Pscan, we identified numerous genes encoding zinc-finger and other TFs associated with hematopoietic stem cell functions. We conclude that, in this mouse model, deficient ZIP8-mediated divalent cation transport affects zinc-finger (e.g. GATA proteins) and other TFs interacting with GATA proteins (e.g. TAL1), predominantly in yolk sac. These data strongly support the phenotype of dysmorphogenesis and anemia seen in Slc39a8(neo/neo) mice in utero.
Collapse
Affiliation(s)
- Jing Chen
- Division of Biomedical Informatics, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, 45229, USA
| | - Marina Gálvez-Peralta
- Department of Environmental Health, University of Cincinnati College of Medicine, Cincinnati, Ohio, 45267, USA.,Department of Pharmaceutical Sciences, West Virginia University Medical Center, Morgantown, WV, 26506, USA
| | - Xiang Zhang
- Department of Environmental Health, University of Cincinnati College of Medicine, Cincinnati, Ohio, 45267, USA
| | - Jingyuan Deng
- Department of Environmental Health, University of Cincinnati College of Medicine, Cincinnati, Ohio, 45267, USA.,Amazon.com, Inc., Seattle, WA, 98101, USA
| | - Zijuan Liu
- Department of Biological Sciences, Oakland University, Rochester, MI, 48309, USA
| | - Daniel W Nebert
- Department of Environmental Health, University of Cincinnati College of Medicine, Cincinnati, Ohio, 45267, USA.
| |
Collapse
|
69
|
Rodan LH, Hauptman M, D'Gama AM, Qualls AE, Cao S, Tuschl K, Al-Jasmi F, Hertecant J, Hayflick SJ, Wessling-Resnick M, Yang ET, Berry GT, Gropman A, Woolf AD, Agrawal PB. Novel founder intronic variant in SLC39A14 in two families causing Manganism and potential treatment strategies. Mol Genet Metab 2018; 124:161-167. [PMID: 29685658 PMCID: PMC5976541 DOI: 10.1016/j.ymgme.2018.04.002] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/22/2018] [Revised: 04/04/2018] [Accepted: 04/04/2018] [Indexed: 01/06/2023]
Abstract
Congenital disorders of manganese metabolism are rare occurrences in children, and medical management of these disorders is complex and challenging. Homozygous exonic mutations in the manganese transporter SLC39A14 have recently been associated with a pediatric-onset neurodegenerative disorder characterized by brain manganese accumulation and clinical signs of manganese neurotoxicity, including parkinsonism-dystonia. We performed whole exome sequencing on DNA samples from two unrelated female children from the United Arab Emirates with progressive movement disorder and brain mineralization, identified a novel homozygous intronic mutation in SLC39A14 in both children, and demonstrated that the mutation leads to aberrant splicing. Both children had consistently elevated serum manganese levels and were diagnosed with SLC39A14-associated manganism. Over a four-year period, we utilized a multidisciplinary management approach for Patient 1 combining decreased manganese dietary intake and chelation with symptomatic management of dystonia. Our treatment strategy appeared to slow disease progression, but did not lead to a cure or reversal of already established deficits. Clinicians should consider testing for noncoding mutations in the diagnosis of congenital disorders of manganese metabolism and utilizing multidisciplinary approaches in the management of these disorders.
Collapse
Affiliation(s)
- Lance H Rodan
- Department of Neurology, Boston Children's Hospital, Boston, MA, United States; Harvard Medical School, Boston, MA, United States; Division of Genetics and Genomics, Boston Children's Hospital, Boston, MA, United States.
| | - Marissa Hauptman
- Harvard Medical School, Boston, MA, United States; Pediatric Environmental Health Center, Division of General Pediatrics, Boston Children's Hospital, Boston, MA, United States; Region 1 New, England, Pediatric Environmental Health Specialty Unit (PEHSU), Boston, MA, United States
| | - Alissa M D'Gama
- Harvard Medical School, Boston, MA, United States; Division of Genetics and Genomics, Boston Children's Hospital, Boston, MA, United States
| | - Anita E Qualls
- Division of Newborn Medicine, Boston Children's Hospital, Boston, MA, United States; The Manton Center for Orphan Disease Research, Boston Children's Hospital, Boston, MA, United States
| | - Siqi Cao
- Division of Genetics and Genomics, Boston Children's Hospital, Boston, MA, United States; Division of Newborn Medicine, Boston Children's Hospital, Boston, MA, United States; The Manton Center for Orphan Disease Research, Boston Children's Hospital, Boston, MA, United States
| | - Karin Tuschl
- Department of Cell and Developmental Biology, University College London Great Ormond Street Institute of Child Health, London, UK
| | - Fatma Al-Jasmi
- Department of Pediatrics, United Arab Emirates University, Al Ain, United Arab Emirates
| | - Jozef Hertecant
- Department of Pediatrics, United Arab Emirates University, Al Ain, United Arab Emirates
| | - Susan J Hayflick
- Department of Molecular and Medical Genetics, Oregon Health & Science University, Portland, OR, United States; Department of Pathology, Oregon Health & Science University, Portland, OR, United States
| | - Marianne Wessling-Resnick
- Department of Genetics and Complex Diseases, Harvard T.H. Chan School of Public Health, Boston, MA, United States
| | - Edward T Yang
- Department of Radiology, Boston Children's Hospital, Boston, MA, United States
| | - Gerard T Berry
- Harvard Medical School, Boston, MA, United States; Division of Genetics and Genomics, Boston Children's Hospital, Boston, MA, United States
| | - Andrea Gropman
- Division of Neurodevelopmental Disabilities and Neurogenetics, Children's National Health System, Washington, DC, United States
| | - Alan D Woolf
- Harvard Medical School, Boston, MA, United States; Pediatric Environmental Health Center, Division of General Pediatrics, Boston Children's Hospital, Boston, MA, United States; Region 1 New, England, Pediatric Environmental Health Specialty Unit (PEHSU), Boston, MA, United States
| | - Pankaj B Agrawal
- Division of Genetics and Genomics, Boston Children's Hospital, Boston, MA, United States; Division of Newborn Medicine, Boston Children's Hospital, Boston, MA, United States; The Manton Center for Orphan Disease Research, Boston Children's Hospital, Boston, MA, United States
| |
Collapse
|
70
|
Ferreira CR, Altassan R, Marques-Da-Silva D, Francisco R, Jaeken J, Morava E. Recognizable phenotypes in CDG. J Inherit Metab Dis 2018; 41:541-553. [PMID: 29654385 PMCID: PMC5960425 DOI: 10.1007/s10545-018-0156-5] [Citation(s) in RCA: 73] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/15/2017] [Revised: 12/23/2017] [Accepted: 02/06/2018] [Indexed: 01/06/2023]
Abstract
Pattern recognition, using a group of characteristic, or discriminating features, is a powerful tool in metabolic diagnostic. A classic example of this approach is used in biochemical analysis of urine organic acid analysis, where the reporting depends more on the correlation of pertinent positive and negative findings, rather than on the absolute values of specific markers. Similar uses of pattern recognition in the field of biochemical genetics include the interpretation of data obtained by metabolomics, like glycomics, where a recognizable pattern or the presence of a specific glycan sub-fraction can lead to the direct diagnosis of certain types of congenital disorders of glycosylation. Another indispensable tool is the use of clinical pattern recognition-or syndromology-relying on careful phenotyping. While genomics might uncover variants not essential in the final clinical expression of disease, and metabolomics could point to a mixture of primary but also secondary changes in biochemical pathways, phenomics describes the clinically relevant manifestations and the full expression of the disease. In the current review we apply phenomics to the field of congenital disorders of glycosylation, focusing on recognizable differentiating findings in glycosylation disorders, characteristic dysmorphic features and malformations in PMM2-CDG, and overlapping patterns among the currently known glycosylation disorders based on their pathophysiological basis.
Collapse
Affiliation(s)
- Carlos R Ferreira
- Medical Genetics Branch National Human Genome Research Institute, National Institutes of Health, Bethesda, MD, USA
- Division of Genetics and Metabolism, Children's National Medical Center, Washington, DC, USA
| | - Ruqaia Altassan
- Metabolic Center, Department of Pediatrics, University Hospitals Leuven, Herestraat 49, B-3000, Leuven, Belgium
- Department of Development and Regeneration, Faculty of Medicine, KU Leuven, Leuven, Belgium
| | - Dorinda Marques-Da-Silva
- UCIBIO, Departamento Ciências da Vida, Faculdade de Ciências e Tecnologia, Universidade NOVA de Lisboa, Lisboa, Portugal
- Portuguese Association for CDG, Lisboa, Portugal
| | - Rita Francisco
- UCIBIO, Departamento Ciências da Vida, Faculdade de Ciências e Tecnologia, Universidade NOVA de Lisboa, Lisboa, Portugal
- Portuguese Association for CDG, Lisboa, Portugal
| | - Jaak Jaeken
- Metabolic Center, Department of Pediatrics, University Hospitals Leuven, Herestraat 49, B-3000, Leuven, Belgium
- Department of Development and Regeneration, Faculty of Medicine, KU Leuven, Leuven, Belgium
| | - Eva Morava
- Metabolic Center, Department of Pediatrics, University Hospitals Leuven, Herestraat 49, B-3000, Leuven, Belgium.
- Department of Development and Regeneration, Faculty of Medicine, KU Leuven, Leuven, Belgium.
- Department of Clinical Genomics, Center for Individualized Medicine, Mayo Clinic, Rochester, MN, USA.
| |
Collapse
|
71
|
Brasil S, Pascoal C, Francisco R, Marques-da-Silva D, Andreotti G, Videira PA, Morava E, Jaeken J, Dos Reis Ferreira V. CDG Therapies: From Bench to Bedside. Int J Mol Sci 2018; 19:ijms19051304. [PMID: 29702557 PMCID: PMC5983582 DOI: 10.3390/ijms19051304] [Citation(s) in RCA: 60] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2018] [Revised: 04/14/2018] [Accepted: 04/21/2018] [Indexed: 12/20/2022] Open
Abstract
Congenital disorders of glycosylation (CDG) are a group of genetic disorders that affect protein and lipid glycosylation and glycosylphosphatidylinositol synthesis. More than 100 different disorders have been reported and the number is rapidly increasing. Since glycosylation is an essential post-translational process, patients present a large range of symptoms and variable phenotypes, from very mild to extremely severe. Only for few CDG, potentially curative therapies are being used, including dietary supplementation (e.g., galactose for PGM1-CDG, fucose for SLC35C1-CDG, Mn2+ for TMEM165-CDG or mannose for MPI-CDG) and organ transplantation (e.g., liver for MPI-CDG and heart for DOLK-CDG). However, for the majority of patients, only symptomatic and preventive treatments are in use. This constitutes a burden for patients, care-givers and ultimately the healthcare system. Innovative diagnostic approaches, in vitro and in vivo models and novel biomarkers have been developed that can lead to novel therapeutic avenues aiming to ameliorate the patients’ symptoms and lives. This review summarizes the advances in therapeutic approaches for CDG.
Collapse
Affiliation(s)
- Sandra Brasil
- Portuguese Association for Congenital Disorders of Glycosylation (CDG), Departamento Ciências da Vida, Faculdade de Ciências e Tecnologia, Universidade NOVA de Lisboa, 2820-287 Lisboa, Portugal.
- Professionals and Patient Associations International Network (CDG & Allies-PPAIN), Departamento Ciências da Vida, Faculdade de Ciências e Tecnologia, Universidade NOVA de Lisboa, 2820-287 Lisboa, Portugal.
| | - Carlota Pascoal
- Portuguese Association for Congenital Disorders of Glycosylation (CDG), Departamento Ciências da Vida, Faculdade de Ciências e Tecnologia, Universidade NOVA de Lisboa, 2820-287 Lisboa, Portugal.
- Professionals and Patient Associations International Network (CDG & Allies-PPAIN), Departamento Ciências da Vida, Faculdade de Ciências e Tecnologia, Universidade NOVA de Lisboa, 2820-287 Lisboa, Portugal.
- Research Unit on Applied Molecular Biosciences (UCIBIO), Departamento Ciências da Vida, Faculdade de Ciências e Tecnologia, Universidade NOVA de Lisboa, 2829-516 Lisboa, Portugal.
| | - Rita Francisco
- Portuguese Association for Congenital Disorders of Glycosylation (CDG), Departamento Ciências da Vida, Faculdade de Ciências e Tecnologia, Universidade NOVA de Lisboa, 2820-287 Lisboa, Portugal.
- Professionals and Patient Associations International Network (CDG & Allies-PPAIN), Departamento Ciências da Vida, Faculdade de Ciências e Tecnologia, Universidade NOVA de Lisboa, 2820-287 Lisboa, Portugal.
- Research Unit on Applied Molecular Biosciences (UCIBIO), Departamento Ciências da Vida, Faculdade de Ciências e Tecnologia, Universidade NOVA de Lisboa, 2829-516 Lisboa, Portugal.
| | - Dorinda Marques-da-Silva
- Portuguese Association for Congenital Disorders of Glycosylation (CDG), Departamento Ciências da Vida, Faculdade de Ciências e Tecnologia, Universidade NOVA de Lisboa, 2820-287 Lisboa, Portugal.
- Professionals and Patient Associations International Network (CDG & Allies-PPAIN), Departamento Ciências da Vida, Faculdade de Ciências e Tecnologia, Universidade NOVA de Lisboa, 2820-287 Lisboa, Portugal.
- Research Unit on Applied Molecular Biosciences (UCIBIO), Departamento Ciências da Vida, Faculdade de Ciências e Tecnologia, Universidade NOVA de Lisboa, 2829-516 Lisboa, Portugal.
| | - Giuseppina Andreotti
- Istituto di Chimica Biomolecolare-Consiglio Nazionale delle Ricerche (CNR), 80078 Pozzuoli, Italy.
| | - Paula A Videira
- Portuguese Association for Congenital Disorders of Glycosylation (CDG), Departamento Ciências da Vida, Faculdade de Ciências e Tecnologia, Universidade NOVA de Lisboa, 2820-287 Lisboa, Portugal.
- Professionals and Patient Associations International Network (CDG & Allies-PPAIN), Departamento Ciências da Vida, Faculdade de Ciências e Tecnologia, Universidade NOVA de Lisboa, 2820-287 Lisboa, Portugal.
- Research Unit on Applied Molecular Biosciences (UCIBIO), Departamento Ciências da Vida, Faculdade de Ciências e Tecnologia, Universidade NOVA de Lisboa, 2829-516 Lisboa, Portugal.
| | - Eva Morava
- Professionals and Patient Associations International Network (CDG & Allies-PPAIN), Departamento Ciências da Vida, Faculdade de Ciências e Tecnologia, Universidade NOVA de Lisboa, 2820-287 Lisboa, Portugal.
- Department of Clinical Genomics, Mayo Clinic, Rochester, MN 55905, USA.
| | - Jaak Jaeken
- Professionals and Patient Associations International Network (CDG & Allies-PPAIN), Departamento Ciências da Vida, Faculdade de Ciências e Tecnologia, Universidade NOVA de Lisboa, 2820-287 Lisboa, Portugal.
- Center for Metabolic Diseases, Universitaire Ziekenhuizen (UZ) and Katholieke Universiteit (KU) Leuven, 3000 Leuven, Belgium.
| | - Vanessa Dos Reis Ferreira
- Portuguese Association for Congenital Disorders of Glycosylation (CDG), Departamento Ciências da Vida, Faculdade de Ciências e Tecnologia, Universidade NOVA de Lisboa, 2820-287 Lisboa, Portugal.
- Professionals and Patient Associations International Network (CDG & Allies-PPAIN), Departamento Ciências da Vida, Faculdade de Ciências e Tecnologia, Universidade NOVA de Lisboa, 2820-287 Lisboa, Portugal.
| |
Collapse
|
72
|
Gardeitchik T, Wyckmans J, Morava E. Complex Phenotypes in Inborn Errors of Metabolism: Overlapping Presentations in Congenital Disorders of Glycosylation and Mitochondrial Disorders. Pediatr Clin North Am 2018; 65:375-388. [PMID: 29502919 DOI: 10.1016/j.pcl.2017.11.012] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Congenital disorders of glycosylation (CDG) and mitochondrial disorders have overlapping clinical features, including central nervous system, cardiac, gastrointestinal, hepatic, muscular, endocrine, and psychiatric disease. Specific abnormalities orienting the clinician toward the right diagnostic approach include abnormal fat distribution, coagulation abnormalities, together with anticoagulation abnormalities, hyperinsulinism, and congenital malformations in CDG. Diabetes, sensorineural deafness, and depression are very rare in CDG but common in mitochondrial disease. Chronic lactic acidosis is highly suggestive of mitochondrial dysfunction. Serum transferrin isoform analysis is specific for glycosylation abnormalities but not abnormal in all types of CDG.
Collapse
Affiliation(s)
- Thatjana Gardeitchik
- Department of Human Genetics, Radboudumc Medical Center, Geert Grooteplein, 6500 HB, Nijmegen, The Netherlands
| | - Jeroen Wyckmans
- Department of Pediatrics, University Hospitals Leuven, Leuven, Belgium
| | - Eva Morava
- Department of Pediatrics, University Hospitals Leuven, Leuven, Belgium; Hayward Genetics Center, Tulane University Medical School, New Orleans, LA, USA.
| |
Collapse
|
73
|
Ng BG, Freeze HH. Perspectives on Glycosylation and Its Congenital Disorders. Trends Genet 2018; 34:466-476. [PMID: 29606283 DOI: 10.1016/j.tig.2018.03.002] [Citation(s) in RCA: 171] [Impact Index Per Article: 24.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2018] [Revised: 03/04/2018] [Accepted: 03/05/2018] [Indexed: 12/12/2022]
Abstract
Congenital disorders of glycosylation (CDG) are a rapidly expanding group of metabolic disorders that result from abnormal protein or lipid glycosylation. They are often difficult to clinically diagnose because they broadly affect many organs and functions and lack clinical uniformity. However, recent technological advances in next-generation sequencing have revealed a treasure trove of new genetic disorders, expanded the knowledge of known disorders, and showed a critical role in infectious diseases. More comprehensive genetic tools specifically tailored for mammalian cell-based models have revealed a critical role for glycosylation in pathogen-host interactions, while also identifying new CDG susceptibility genes. We highlight recent advancements that have resulted in a better understanding of human glycosylation disorders, perspectives for potential future therapies, and mysteries for which we continue to seek new insights and solutions.
Collapse
Affiliation(s)
- Bobby G Ng
- Human Genetics Program, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA 92037, USA
| | - Hudson H Freeze
- Human Genetics Program, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA 92037, USA.
| |
Collapse
|
74
|
Choi EK, Nguyen TT, Gupta N, Iwase S, Seo YA. Functional analysis of SLC39A8 mutations and their implications for manganese deficiency and mitochondrial disorders. Sci Rep 2018; 8:3163. [PMID: 29453449 PMCID: PMC5816659 DOI: 10.1038/s41598-018-21464-0] [Citation(s) in RCA: 62] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2017] [Accepted: 02/05/2018] [Indexed: 11/19/2022] Open
Abstract
SLC39A8 encodes ZIP8, a divalent metal ion transporter. Mutations in the SLC39A8 gene are associated with congenital disorder of glycosylation type II and Leigh syndrome. Notably, affected patients with both disorders exhibited severe manganese (Mn) deficiency. The cellular function of human SLC39A8 (hSLC39A8) and the mechanisms by which mutations in this protein lead to human diseases are unclear. Herein, we show that hSLC39A8 mediates 54Mn uptake by the cells, and its expression is regulated by Mn. While expression of wild-type hSLC39A8 increased 54Mn uptake activity, disease-associated mutations abrogated the ability of the transporter to mediate Mn uptake into the cells, thereby providing a causal link to severe Mn deficiency. All mutants failed to localize on the cell surface and were retained within the endoplasmic reticulum. Interestingly, expression of hSLC39A8 mutants of both CDG type II and Leigh syndrome reduced mitochondrial 54Mn levels and activity of Mn-dependent mitochondrial superoxide dismutase MnSOD, and in turn increased oxidative stress. The expression of wild-type hSLC39A8, but not the disease-associated mutants, promoted mitochondrial functions. Moreover, loss of function analyses further corroborate hSLC39A8's critical role in mediating Mn uptake and mitochondrial function. Our results provide a potential pathogenic mechanism of diseases that are associated with hSLC39A8 mutations.
Collapse
Affiliation(s)
- Eun-Kyung Choi
- Department of Nutritional Sciences, University of Michigan School of Public Health, Ann Arbor, MI, 48109, USA
| | - Trang-Tiffany Nguyen
- Department of Nutritional Sciences, University of Michigan School of Public Health, Ann Arbor, MI, 48109, USA
| | - Neil Gupta
- Department of Nutritional Sciences, University of Michigan School of Public Health, Ann Arbor, MI, 48109, USA
| | - Shigeki Iwase
- Department of Human Genetics, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Young Ah Seo
- Department of Nutritional Sciences, University of Michigan School of Public Health, Ann Arbor, MI, 48109, USA.
| |
Collapse
|
75
|
Marti-Sanchez L, Ortigoza-Escobar JD, Darling A, Villaronga M, Baide H, Molero-Luis M, Batllori M, Vanegas MI, Muchart J, Aquino L, Artuch R, Macaya A, Kurian MA, Dueñas P. Hypermanganesemia due to mutations in SLC39A14: further insights into Mn deposition in the central nervous system. Orphanet J Rare Dis 2018; 13:28. [PMID: 29382362 PMCID: PMC5791243 DOI: 10.1186/s13023-018-0758-x] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2017] [Accepted: 01/03/2018] [Indexed: 11/13/2022] Open
Abstract
Background The SLC39A14, SLC30A10 and SLC39A8 are considered to be key genes involved in manganese (Mn) homeostasis in humans. Mn levels in plasma and urine are useful tools for early recognition of these disorders. We aimed to explore further biomarkers of Mn deposition in the central nervous system in two siblings presenting with acute dystonia and hypermanganesemia due to mutations in SLC39A14. These biomarkers may help clinicians to establish faster and accurate diagnosis and to monitor disease progression after chelation therapy is administered. Results A customized gene panel for movement disorders revealed a novel missense variant (c.311G > T; p.Ser104Ile) in SLC39A14 gene in two siblings presenting at the age of 10 months with acute dystonia and motor regression. Mn concentrations were analyzed using inductively coupled mass spectrometry in plasma and cerebrospinal fluid, disclosing elevated Mn levels in the index case compared to control patients. Surprisingly, Mn values were 3-fold higher in CSF than in plasma. We quantified the pallidal index, defined as the ratio between the signal intensity in the globus pallidus and the subcortical frontal white matter in axial T1-weighted MRI, and found significantly higher values in the SLC39A14 patient than in controls. These values increased over a period of 10 years, suggesting the relentless pallidal accumulation of Mn. Following genetic confirmation, a trial with the Mn chelator Na2CaEDTA led to a reduction in plasma Mn, zinc and selenium levels. However, parents reported worsening of cervical dystonia, irritability and sleep difficulties and chelation therapy was discontinued. Conclusions Our study expands the very few descriptions of patients with SLC39A14 mutations. We report for the first time the elevation of Mn in CSF of SLC39A14 mutated patients, supporting the hypothesis that brain is an important organ of Mn deposition in SLC39A14-related disease. The pallidal index is an indirect and non-invasive method that can be used to rate disease progression on follow-up MRIs. Finally, we propose that patients with inherited defects of manganese transport should be initially treated with low doses of Na2CaEDTA followed by gradual dose escalation, together with a close monitoring of blood trace elements in order to avoid side effects.
Collapse
Affiliation(s)
- L Marti-Sanchez
- Department of Biochemistry, Institut de Recerca - Hospital Sant Joan de Déu, University of Barcelona, Barcelona, Spain
| | - J D Ortigoza-Escobar
- Department of Child Neurology, Institut de Recerca - Hospital Sant Joan de Déu, University of Barcelona, Barcelona, Spain
| | - A Darling
- Department of Child Neurology, Institut de Recerca - Hospital Sant Joan de Déu, University of Barcelona, Barcelona, Spain
| | - M Villaronga
- Department of Pharmacy, Institut de Recerca - Hospital Sant Joan de Déu, University of Barcelona, Barcelona, Spain
| | - H Baide
- Department of Child Neurology, Institut de Recerca - Hospital Sant Joan de Déu, University of Barcelona, Barcelona, Spain
| | - M Molero-Luis
- Department of Biochemistry, Institut de Recerca - Hospital Sant Joan de Déu, University of Barcelona, Barcelona, Spain
| | - M Batllori
- Department of Biochemistry, Institut de Recerca - Hospital Sant Joan de Déu, University of Barcelona, Barcelona, Spain
| | - M I Vanegas
- Department of Child Neurology, Institut de Recerca - Hospital Sant Joan de Déu, University of Barcelona, Barcelona, Spain
| | - J Muchart
- Department of Radiology, Institut de Recerca - Hospital Sant Joan de Déu, University of Barcelona, Barcelona, Spain
| | - L Aquino
- Deparment of Pediatrics, Hospital de Mataró, Barcelona, Spain
| | - R Artuch
- Department of Biochemistry, Institut de Recerca - Hospital Sant Joan de Déu, University of Barcelona, Barcelona, Spain
| | - A Macaya
- Pediatric Neurology Research Group, Vall d'Hebron Research Institute (VHIR), Universitat Autònoma de Barcelona, Passeig de la Vall d'Hebron, 119-129, 08035, Barcelona, Catalonia, Spain
| | - M A Kurian
- Molecular Neurosciences, Developmental Neurosciences Programme, UCL-Great Ormond Street Institute of Child Health, London, UK
| | - Pérez Dueñas
- Department of Child Neurology, Institut de Recerca - Hospital Sant Joan de Déu, University of Barcelona, Barcelona, Spain. .,Pediatric Neurology Research Group, Vall d'Hebron Research Institute (VHIR), Universitat Autònoma de Barcelona, Passeig de la Vall d'Hebron, 119-129, 08035, Barcelona, Catalonia, Spain.
| |
Collapse
|
76
|
Abstract
Trace elements are chemical elements needed in minute amounts for normal physiology. Some of the physiologically relevant trace elements include iodine, copper, iron, manganese, zinc, selenium, cobalt and molybdenum. Of these, some are metals, and in particular, transition metals. The different electron shells of an atom carry different energy levels, with those closest to the nucleus being lowest in energy. The number of electrons in the outermost shell determines the reactivity of such an atom. The electron shells are divided in sub-shells, and in particular the third shell has s, p and d sub-shells. Transition metals are strictly defined as elements whose atom has an incomplete d sub-shell. This incomplete d sub-shell makes them prone to chemical reactions, particularly redox reactions. Transition metals of biologic importance include copper, iron, manganese, cobalt and molybdenum. Zinc is not a transition metal, since it has a complete d sub-shell. Selenium, on the other hand, is strictly speaking a nonmetal, although given its chemical properties between those of metals and nonmetals, it is sometimes considered a metalloid. In this review, we summarize the current knowledge on the inborn errors of metal and metalloid metabolism.
Collapse
Affiliation(s)
- Carlos R. Ferreira
- Division of Genetics and Metabolism, Children’s National Health System, Washington, DC, USA
- Department of Pediatrics, George Washington University School of Medicine and Health Sciences, Washington, DC, USA
- Section on Human Biochemical Genetics, Medical Genetics Branch, National Human Genome Research Institute, NIH, Bethesda, MD, USA
| | - William A. Gahl
- Section on Human Biochemical Genetics, Medical Genetics Branch, National Human Genome Research Institute, NIH, Bethesda, MD, USA
| |
Collapse
|
77
|
Maas RR, Iwanicka‐Pronicka K, Kalkan Ucar S, Alhaddad B, AlSayed M, Al‐Owain MA, Al‐Zaidan HI, Balasubramaniam S, Barić I, Bubshait DK, Burlina A, Christodoulou J, Chung WK, Colombo R, Darin N, Freisinger P, Garcia Silva MT, Grunewald S, Haack TB, van Hasselt PM, Hikmat O, Hörster F, Isohanni P, Ramzan K, Kovacs‐Nagy R, Krumina Z, Martin‐Hernandez E, Mayr JA, McClean P, De Meirleir L, Naess K, Ngu LH, Pajdowska M, Rahman S, Riordan G, Riley L, Roeben B, Rutsch F, Santer R, Schiff M, Seders M, Sequeira S, Sperl W, Staufner C, Synofzik M, Taylor RW, Trubicka J, Tsiakas K, Unal O, Wassmer E, Wedatilake Y, Wolff T, Prokisch H, Morava E, Pronicka E, Wevers RA, de Brouwer AP, Wortmann SB. Progressive deafness-dystonia due to SERAC1 mutations: A study of 67 cases. Ann Neurol 2017; 82:1004-1015. [PMID: 29205472 PMCID: PMC5847115 DOI: 10.1002/ana.25110] [Citation(s) in RCA: 49] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2017] [Revised: 11/13/2017] [Accepted: 11/26/2017] [Indexed: 11/12/2022]
Abstract
OBJECTIVE 3-Methylglutaconic aciduria, dystonia-deafness, hepatopathy, encephalopathy, Leigh-like syndrome (MEGDHEL) syndrome is caused by biallelic variants in SERAC1. METHODS This multicenter study addressed the course of disease for each organ system. Metabolic, neuroradiological, and genetic findings are reported. RESULTS Sixty-seven individuals (39 previously unreported) from 59 families were included (age range = 5 days-33.4 years, median age = 9 years). A total of 41 different SERAC1 variants were identified, including 20 that have not been reported before. With the exception of 2 families with a milder phenotype, all affected individuals showed a strikingly homogeneous phenotype and time course. Severe, reversible neonatal liver dysfunction and hypoglycemia were seen in >40% of all cases. Starting at a median age of 6 months, muscular hypotonia (91%) was seen, followed by progressive spasticity (82%, median onset = 15 months) and dystonia (82%, 18 months). The majority of affected individuals never learned to walk (68%). Seventy-nine percent suffered hearing loss, 58% never learned to speak, and nearly all had significant intellectual disability (88%). Magnetic resonance imaging features were accordingly homogenous, with bilateral basal ganglia involvement (98%); the characteristic "putaminal eye" was seen in 53%. The urinary marker 3-methylglutaconic aciduria was present in virtually all patients (98%). Supportive treatment focused on spasticity and drooling, and was effective in the individuals treated; hearing aids or cochlear implants did not improve communication skills. INTERPRETATION MEGDHEL syndrome is a progressive deafness-dystonia syndrome with frequent and reversible neonatal liver involvement and a strikingly homogenous course of disease. Ann Neurol 2017;82:1004-1015.
Collapse
Affiliation(s)
- Roeltje R. Maas
- Translational Metabolic Laboratory, Department of Laboratory MedicineRadboud University Medical CenterNijmegenthe Netherlands
| | | | - Sema Kalkan Ucar
- Division of Metabolic Disease, Ege University Medical Faculty, Department of PediatricsIzmirTurkey
| | - Bader Alhaddad
- Institute of Human GeneticsTechnische UniversitätMünchenMunichGermany
| | - Moeenaldeen AlSayed
- Department of GeneticsKing Faisal Specialist Hospital and Research CenterRiyadhSaudi Arabia
- Department of Anatomy and Cell BiologyCollege of Medicine, Alfaisal UniversityRiyadhSaudi Arabia
| | - Mohammed A. Al‐Owain
- Department of GeneticsKing Faisal Specialist Hospital and Research CenterRiyadhSaudi Arabia
- Department of Anatomy and Cell BiologyCollege of Medicine, Alfaisal UniversityRiyadhSaudi Arabia
| | - Hamad I. Al‐Zaidan
- Department of GeneticsKing Faisal Specialist Hospital and Research CenterRiyadhSaudi Arabia
- Department of Anatomy and Cell BiologyCollege of Medicine, Alfaisal UniversityRiyadhSaudi Arabia
| | - Shanti Balasubramaniam
- Western Sydney Genetics Program, Children's Hospital at Westmead, SydneyNew South WalesAustralia
- Discipline of Genetic Medicine & Paediatrics and Child Health, University of SydneySydneyNew South WalesAustralia
| | - Ivo Barić
- Department of PediatricsUniversity Hospital CenterZagrebCroatia
- School of Medicine, University of ZagrebZagrebCroatia
| | - Dalal K. Bubshait
- Department of Pediatrics, College of MedicineImam Abdulrahman Bin Faisal UniversityDammamSaudi Arabia
| | - Alberto Burlina
- Division of Inherited Metabolic Diseases, Department of PediatricsUniversity Hospital of PaduaPaduaItaly
| | - John Christodoulou
- Neurodevelopmental Genomics Research Group, Murdoch Children's Research Institute, and Department of PaediatricsMelbourne Medical School, University of MelbourneMelbourneVictoriaAustralia
- Genetic Metabolic Disorders Research Unit and Western Sydney Genetics Program, Children's Hospital at WestmeadSydneyNew South WalesAustralia
- Discipline of Child and Adolescent Health and Genetic Medicine, Sydney Medical School, University of SydneySydneyNew South WalesAustralia
| | - Wendy K. Chung
- Departments of Pediatrics and MedicineColumbia UniversityNew YorkNY
| | - Roberto Colombo
- Institute of Clinical Biochemistry, Faculty of Medicine, Catholic University of the Sacred HeartRomeItaly
- Center for the Study of Rare Hereditary Diseases, Niguarda Ca' Granda Metropolitan HospitalMilanItaly
| | - Niklas Darin
- Department of PediatricsInstitute of Clinical Sciences, University of Gothenburg, Queen Silvia's Children's HospitalGothenburgSweden
| | | | - Maria Teresa Garcia Silva
- Inborn Errors of Metabolism and Mitochondrial Disease Unit“12 de Octubre” University Hospital, Avenida de Cordoba sn, 28041 Madrid, Spain. Rare Diseases Biomedical Research Centre (CIBERER)MadridSpain
- Complutense UniversityMadridSpain
| | - Stephanie Grunewald
- Metabolic Medicine DepartmentGreat Ormond Street Hospital for Children National Health Service Foundation Trust, University College London Institute of Child HealthLondonUnited Kingdom
| | - Tobias B. Haack
- Institute of Human GeneticsTechnische UniversitätMünchenMunichGermany
- Institute of Medical Genetics and Applied GenomicsTübingenGermany
| | - Peter M. van Hasselt
- Wilhelmina Children's Hospital Utrecht, University Medical Center UtrechtUtrechtthe Netherlands
| | - Omar Hikmat
- Department of PediatricsHaukeland University HospitalBergenNorway
- Department of Clinical Medicine (K1)University of BergenBergenNorway
| | - Friederike Hörster
- Department of General Pediatrics, Division of Neuropediatrics and Pediatric Metabolic MedicineUniversity Hospital HeidelbergHeidelbergGermany
| | - Pirjo Isohanni
- Children's Hospital, University of Helsinki and Helsinki University HospitalHelsinkiFinland
- Research Programs Unit, Molecular Neurology, Biomedicum Helsinki, University of HelsinkiHelsinkiFinland
| | - Khushnooda Ramzan
- Department of GeneticsKing Faisal Specialist Hospital and Research CenterRiyadhSaudi Arabia
- Department of Anatomy and Cell BiologyCollege of Medicine, Alfaisal UniversityRiyadhSaudi Arabia
| | - Reka Kovacs‐Nagy
- Institute of Human GeneticsTechnische UniversitätMünchenMunichGermany
| | - Zita Krumina
- Department of Biology and MicrobiologyRiga Stradin's UniversityRigaLatvia
| | - Elena Martin‐Hernandez
- Inborn Errors of Metabolism and Mitochondrial Disease Unit“12 de Octubre” University Hospital, Avenida de Cordoba sn, 28041 Madrid, Spain. Rare Diseases Biomedical Research Centre (CIBERER)MadridSpain
- Complutense UniversityMadridSpain
| | - Johannes A. Mayr
- Department of PediatricsSalzburg State Hospitals and Paracelsus Medical UniversitySalzburgAustria
| | - Patricia McClean
- Leeds Teaching Hospitals National Health Service TrustLeedsUnited Kingdom
| | | | - Karin Naess
- Department of Pediatric NeurologyKarolinska University HospitalStockholmSweden
| | - Lock H. Ngu
- Division of Clinical Genetics, Department of GeneticsKuala Lumpur HospitalKuala LumpurMalaysia
| | - Magdalena Pajdowska
- Department of Clinical Biochemistry, Radioimmunology, and Experimental MedicineChildren's Memorial Health InstituteWarsawPoland
| | - Shamima Rahman
- University College London Great Ormond Street Institute of Child HealthLondonUnited Kingdom
| | - Gillian Riordan
- Department of Pediatric NeurologyRed Cross War Memorial Children's HospitalCape TownSouth Africa
| | - Lisa Riley
- Genetic Metabolic Disorders Research Unit and Western Sydney Genetics Program, Children's Hospital at WestmeadSydneyNew South WalesAustralia
- Discipline of Child and Adolescent Health and Genetic Medicine, Sydney Medical School, University of SydneySydneyNew South WalesAustralia
| | - Benjamin Roeben
- Department of NeurodegenerationHertie Institute for Clinical Brain Research, University of TübingenTübingenGermany
- German Center for Neurodegenerative Diseases (DZNE)TübingenGermany
| | - Frank Rutsch
- Department of General PediatricsMünster University Children's HospitalMünsterGermany
| | - Rene Santer
- Department of PediatricsUniversity Medical Center EppendorfHamburgGermany
| | - Manuel Schiff
- Reference Center for Inherited Metabolic Diseases, AP‐HP, Robert Debré Hospital, University Paris Diderot‐Sorbonne Paris Cité, Paris, France AND INSERM U1141ParisFrance
| | - Martine Seders
- Department of Human GeneticsRadboud University Medical CenterNijmegenthe Netherlands
| | | | - Wolfgang Sperl
- Department of PediatricsSalzburg State Hospitals and Paracelsus Medical UniversitySalzburgAustria
| | - Christian Staufner
- Department of General Pediatrics, Division of Neuropediatrics and Pediatric Metabolic MedicineUniversity Hospital HeidelbergHeidelbergGermany
| | - Matthis Synofzik
- Department of NeurodegenerationHertie Institute for Clinical Brain Research, University of TübingenTübingenGermany
- German Center for Neurodegenerative Diseases (DZNE)TübingenGermany
| | - Robert W. Taylor
- Wellcome Centre for Mitochondrial ResearchInstitute of Neuroscience, The Medical School, Newcastle UniversityNewcastle upon TyneUnited Kingdom
| | - Joanna Trubicka
- Department of Medical GeneticsChildren's Memorial Health InstituteWarsawPoland
| | | | - Ozlem Unal
- Division of Metabolic DiseasesHacettepe University Children's HospitalAnkaraTurkey
| | | | - Yehani Wedatilake
- University College London Great Ormond Street Institute of Child HealthLondonUnited Kingdom
| | - Toni Wolff
- Nottingham University Hospitals National Health Service Trust, Nottingham Children's HospitalNottinghamUnited Kingdom
| | - Holger Prokisch
- Institute of Human GeneticsTechnische UniversitätMünchenMunichGermany
- Institute of Human Genetics, Helmholtz Center MunichNeuherbergGermany
| | - Eva Morava
- Hayward Genetics Center and Department of PediatricsTulane University Medical SchoolNew OrleansLA
| | - Ewa Pronicka
- Department of Pediatrics, Nutrition and Metabolic DiseasesChildren's Memorial Health InstituteWarsawPoland
| | - Ron A. Wevers
- Translational Metabolic Laboratory, Department of Laboratory MedicineRadboud University Medical CenterNijmegenthe Netherlands
| | - Arjan P. de Brouwer
- Department of Human GeneticsRadboud University Medical CenterNijmegenthe Netherlands
- Donders Institute for Brain, Cognition, and Behavior, Radboud University Medical CenterNijmegenthe Netherlands
| | - Saskia B. Wortmann
- Institute of Human GeneticsTechnische UniversitätMünchenMunichGermany
- Department of PediatricsSalzburg State Hospitals and Paracelsus Medical UniversitySalzburgAustria
- Institute of Human Genetics, Helmholtz Center MunichNeuherbergGermany
| |
Collapse
|
78
|
EPG5-Related Vici Syndrome: A Primary Defect of Autophagic Regulation with an Emerging Phenotype Overlapping with Mitochondrial Disorders. JIMD Rep 2017; 42:19-29. [PMID: 29159459 PMCID: PMC6226401 DOI: 10.1007/8904_2017_71] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/06/2017] [Revised: 10/24/2017] [Accepted: 10/25/2017] [Indexed: 02/01/2023] Open
Abstract
Vici syndrome is a rare, under-recognised, relentlessly progressive congenital multisystem disorder characterised by five principal features of callosal agenesis, cataracts, cardiomyopathy, combined immunodeficiency and oculocutaneous hypopigmentation. In addition, three equally consistent features (profound developmental delay, progressive failure to thrive and acquired microcephaly) are highly supportive of the diagnosis. Since its recognition as a distinct entity in 1988, an extended phenotype with sensorineural hearing loss, skeletal myopathy and variable involvement of virtually any organ system, including the lungs, thyroid, liver and kidneys, have been described.Autosomal recessive mutations in EPG5 encoding ectopic P-granules autophagy protein 5 (EPG5), a key autophagy regulator implicated in the formation of autolysosomes, were identified as the genetic cause of Vici syndrome. The eight key features outlined above are highly predictive of EPG5 involvement, with pathogenic EPG5 mutations identified in >90% of cases where six or more of these features are present. The manifestation of all eight features has a specificity of 97% and sensitivity of 89% for EPG5-related Vici syndrome. Nevertheless, substantial clinical overlap exists with other multisystem disorders, in particular congenital disorders of glycosylation and mitochondrial disorders. Clinical and pathological findings suggest Vici syndrome as a paradigm of congenital disorders of autophagy, a novel group of inherited neurometabolic conditions linking neurodevelopment and neurodegeneration due to primary autophagy defects.Here we describe the diagnostic odyssey in a 4-year-old boy whose clinical presentation with multisystem manifestations including skeletal myopathy mimicked a mitochondrial disorder. A genetic diagnosis of Vici syndrome was made through whole genome sequencing which identified compound heterozygous variants in EPG5. We also review the myopathic presentation and morphological characterisation of previously reported cases.
Collapse
|
79
|
Witters P, Cassiman D, Morava E. Nutritional Therapies in Congenital Disorders of Glycosylation (CDG). Nutrients 2017; 9:nu9111222. [PMID: 29112118 PMCID: PMC5707694 DOI: 10.3390/nu9111222] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2017] [Revised: 11/01/2017] [Accepted: 11/01/2017] [Indexed: 12/22/2022] Open
Abstract
Congenital disorders of glycosylation (CDG) are a group of more than 130 inborn errors of metabolism affecting N-linked, O-linked protein and lipid-linked glycosylation. The phenotype in CDG patients includes frequent liver involvement, especially the disorders belonging to the N-linked protein glycosylation group. There are only a few treatable CDG. Mannose-Phosphate Isomerase (MPI)-CDG was the first treatable CDG by high dose mannose supplements. Recently, with the successful use of d-galactose in Phosphoglucomutase 1 (PGM1)-CDG, other CDG types have been trialed on galactose and with an increasing number of potential nutritional therapies. Current mini review focuses on therapies in glycosylation disorders affecting liver function and dietary intervention in general in N-linked glycosylation disorders. We also emphasize now the importance of early screening for CDG in patients with mild hepatopathy but also in cholestasis.
Collapse
Affiliation(s)
- Peter Witters
- Metabolic Center, University Hospitals Leuven, B-3000 Leuven, Belgium.
- Department of Development and Regeneration, Faculty of Medicine, KU Leuven, B-3000 Leuven, Belgium.
| | - David Cassiman
- Department of Gastroenterology-Hepatology and Metabolic Center, University Hospitals Leuven, B-3000 Leuven, Belgium.
| | - Eva Morava
- Metabolic Center, University Hospitals Leuven, B-3000 Leuven, Belgium.
- Department of Development and Regeneration, Faculty of Medicine, KU Leuven, B-3000 Leuven, Belgium.
- Hayward Genetics Center, Tulane University School of Medicine, New Orleans, LA 70112, USA.
| |
Collapse
|
80
|
Bastaki F, Bizzari S, Hamici S, Nair P, Mohamed M, Saif F, Malik EM, Al-Ali MT, Hamzeh AR. Single-center experience of N-linked Congenital Disorders of Glycosylation with a Summary of Molecularly Characterized Cases in Arabs. Ann Hum Genet 2017; 82:35-47. [PMID: 28940310 DOI: 10.1111/ahg.12220] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2017] [Revised: 08/14/2017] [Accepted: 08/16/2017] [Indexed: 12/18/2022]
Abstract
Congenital disorders of glycosylation (CDG) represent an expanding group of conditions that result from defects in protein and lipid glycosylation. Different subgroups of CDG display considerable clinical and genetic heterogeneity due to the highly complex nature of cellular glycosylation. This is further complicated by ethno-geographic differences in the mutational landscape of each of these subgroups. Ten Arab CDG patients from Latifa Hospital in Dubai, United Arab Emirates, were assessed using biochemical (glycosylation status of transferrin) and molecular approaches (next-generation sequencing [NGS] and Sanger sequencing). In silico tools including CADD and PolyPhen-2 were used to predict the functional consequences of uncovered mutations. In our sample of patients, five novel mutations were uncovered in the genes: MPDU1, PMM2, MAN1B1, and RFT1. In total, 9 mutations were harbored by the 10 patients in 7 genes. These are missense and nonsense mutations with deleterious functional consequences. This article integrates a single-center experience within a list of reported CDG mutations in the Arab world, accompanied by full molecular and clinical details pertaining to the studied cases. It also sheds light on potential ethnic differences that were not noted before in regards to CDG in the Arab world.
Collapse
Affiliation(s)
- Fatma Bastaki
- Pediatric Department, Latifa Hospital, Dubai Health Authority, Dubai, UAE
| | | | - Sana Hamici
- Pediatric Department, Latifa Hospital, Dubai Health Authority, Dubai, UAE
| | | | - Madiha Mohamed
- Pediatric Department, Latifa Hospital, Dubai Health Authority, Dubai, UAE
| | - Fatima Saif
- Pediatric Department, Latifa Hospital, Dubai Health Authority, Dubai, UAE
| | | | | | | |
Collapse
|
81
|
SLC39A8 deficiency: biochemical correction and major clinical improvement by manganese therapy. Genet Med 2017; 20:259-268. [PMID: 28749473 DOI: 10.1038/gim.2017.106] [Citation(s) in RCA: 52] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2017] [Accepted: 06/01/2017] [Indexed: 02/08/2023] Open
Abstract
PurposeSLC39A8 deficiency is a severe inborn error of metabolism that is caused by impaired function of manganese metabolism in humans. Mutations in SLC39A8 lead to impaired function of the manganese transporter ZIP8 and thus manganese deficiency. Due to the important role of Mn2+ as a cofactor for a variety of enzymes, the resulting phenotype is complex and severe. The manganese-dependence of β-1,4-galactosyltransferases leads to secondary hypoglycosylation, making SLC39A8 deficiency both a disorder of trace element metabolism and a congenital disorder of glycosylation. Some hypoglycosylation disorders have previously been treated with galactose administration. The development of an effective treatment of the disorder by high-dose manganese substitution aims at correcting biochemical, and hopefully, clinical abnormalities.MethodsTwo SCL39A8 deficient patients were treated with 15 and 20 mg MnSO4/kg bodyweight per day. Glycosylation and blood manganese were monitored closely. In addition, magnetic resonance imaging was performed to detect potential toxic effects of manganese.ResultsAll measured enzyme dysfunctions resolved completely and considerable clinical improvement regarding motor abilities, hearing, and other neurological manifestations was observed.ConclusionHigh-dose manganese substitution was effective in two patients with SLC39A8 deficiency. Close therapy monitoring by glycosylation assays and blood manganese measurements is necessary to prevent manganese toxicity.
Collapse
|
82
|
Clayton PT. Inherited disorders of transition metal metabolism: an update. J Inherit Metab Dis 2017; 40:519-529. [PMID: 28303424 DOI: 10.1007/s10545-017-0030-x] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/24/2017] [Revised: 02/16/2017] [Accepted: 02/20/2017] [Indexed: 12/30/2022]
Abstract
Elements with a biological role include six trace transition metals: manganese, iron, cobalt, copper, zinc and molybdenum. Transition metals participate in group transfer reactions such as glycosylation and phosphorylation and those that can transfer an electron by alternating between two redox states such as iron (3+/2+) and copper (2+/1+) are also very important in biological redox reactions including the reduction of molecular oxygen and the transport of oxygen. However, these trace metals are also potentially toxic, generating reactive oxygen species through Fenton chemistry. Recently, a role of trace metals in host defence ("nutritional immunity") has been recognized. The host can deprive the pathogen of a trace metal or poison it with a toxic concentration. Disorders leading to low concentrations of a trace metal can often be treated by supplementing that metal; disorders leading to excessively high concentrations can often be treated with chelating agents such as penicillamine and disodium calcium edetate. This update will address: i) the manganese/zinc transporters (because two new treatable disorders were described in 2016 - SLC39A8 deficiency and SLC39A14 deficiency); ii) copper transporter disorders because we need to improve the treatment of patients with neurological symptoms due to Wilson's disease; and iii) iron homeostasis because recent progress in research into the metabolism of iron and its regulation helps us better understand several inborn errors affecting these pathways.
Collapse
Affiliation(s)
- Peter T Clayton
- Centre for Translational Omics, Genetics and Genomic Medicine, UCL Great Ormond Street Institute of Child Health, 30 Guilford Street, London, WC1N 1EH, UK.
| |
Collapse
|
83
|
Thomason RT, Pettiglio MA, Herrera C, Kao C, Gitlin JD, Bartnikas TB. Characterization of trace metal content in the developing zebrafish embryo. PLoS One 2017; 12:e0179318. [PMID: 28617866 PMCID: PMC5472288 DOI: 10.1371/journal.pone.0179318] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2017] [Accepted: 05/26/2017] [Indexed: 11/18/2022] Open
Abstract
Trace metals are essential for health but toxic when present in excess. The maintenance of trace metals at physiologic levels reflects both import and export by cells and absorption and excretion by organs. The mechanism by which this maintenance is achieved in vertebrate organisms is incompletely understood. To explore this, we chose zebrafish as our model organism, as they are amenable to both pharmacologic and genetic manipulation and comprise an ideal system for genetic screens and toxicological studies. To characterize trace metal content in developing zebrafish, we measured levels of three trace elements, copper, zinc, and manganese, from the oocyte stage to 30 days post-fertilization using inductively coupled plasma mass spectrometry. Our results indicate that metal levels are stable until zebrafish can acquire metals from the environment and imply that the early embryo relies on maternal contribution of metals to the oocyte. We also measured metal levels in bodies and yolks of embryos reared in presence and absence of the copper chelator neocuproine. All three metals exhibited different relative abundances between yolks and bodies of embryos. While neocuproine treatment led to an expected phenotype of copper deficiency, total copper levels were unaffected, indicating that measurement of total metal levels does not equate with measurement of biologically active metal levels. Overall, our data not only can be used in the design and execution of genetic, physiologic, and toxicologic studies but also has implications for the understanding of vertebrate metal homeostasis.
Collapse
Affiliation(s)
| | - Michael A. Pettiglio
- Department of Pathology and Laboratory Medicine, Brown University, Providence, Rhode Island, United States of America
| | - Carolina Herrera
- Department of Pathology and Laboratory Medicine, Brown University, Providence, Rhode Island, United States of America
| | - Clara Kao
- Eugene Bell Center, Marine Biological Laboratory, Woods Hole, Massachusetts, United States of America
| | - Jonathan D. Gitlin
- Eugene Bell Center, Marine Biological Laboratory, Woods Hole, Massachusetts, United States of America
| | - Thomas B. Bartnikas
- Department of Pathology and Laboratory Medicine, Brown University, Providence, Rhode Island, United States of America
| |
Collapse
|
84
|
Lin W, Vann DR, Doulias PT, Wang T, Landesberg G, Li X, Ricciotti E, Scalia R, He M, Hand NJ, Rader DJ. Hepatic metal ion transporter ZIP8 regulates manganese homeostasis and manganese-dependent enzyme activity. J Clin Invest 2017; 127:2407-2417. [PMID: 28481222 DOI: 10.1172/jci90896] [Citation(s) in RCA: 123] [Impact Index Per Article: 15.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2016] [Accepted: 03/07/2017] [Indexed: 01/08/2023] Open
Abstract
Genetic variants at the solute carrier family 39 member 8 (SLC39A8) gene locus are associated with the regulation of whole-blood manganese (Mn) and multiple physiological traits. SLC39A8 encodes ZIP8, a divalent metal ion transporter best known for zinc transport. Here, we hypothesized that ZIP8 regulates Mn homeostasis and Mn-dependent enzymes to influence metabolism. We generated Slc39a8-inducible global-knockout (ZIP8-iKO) and liver-specific-knockout (ZIP8-LSKO) mice and observed markedly decreased Mn levels in multiple organs and whole blood of both mouse models. By contrast, liver-specific overexpression of human ZIP8 (adeno-associated virus-ZIP8 [AAV-ZIP8]) resulted in increased tissue and whole blood Mn levels. ZIP8 expression was localized to the hepatocyte canalicular membrane, and bile Mn levels were increased in ZIP8-LSKO and decreased in AAV-ZIP8 mice. ZIP8-LSKO mice also displayed decreased liver and kidney activity of the Mn-dependent enzyme arginase. Both ZIP8-iKO and ZIP8-LSKO mice had defective protein N-glycosylation, and humans homozygous for the minor allele at the lead SLC39A8 variant showed hypogalactosylation, consistent with decreased activity of another Mn-dependent enzyme, β-1,4-galactosyltransferase. In summary, hepatic ZIP8 reclaims Mn from bile and regulates whole-body Mn homeostasis, thereby modulating the activity of Mn-dependent enzymes. This work provides a mechanistic basis for the association of SLC39A8 with whole-blood Mn, potentially linking SLC39A8 variants with other physiological traits.
Collapse
Affiliation(s)
- Wen Lin
- Department of Medicine, Perelman School of Medicine, and
| | - David R Vann
- Department of Earth and Environmental Science, School of Arts and Sciences, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Paschalis-Thomas Doulias
- Children's Hospital of Philadelphia Research Institute and Department of Pharmacology, Children's Hospital of Philadelphia and University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Tao Wang
- Department of Medicine, Perelman School of Medicine, and
| | - Gavin Landesberg
- Department of Physiology, Temple University, Philadelphia, Pennsylvania, USA
| | - Xueli Li
- Palmieri Metabolic Disease Laboratory, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
| | | | - Rosario Scalia
- Department of Physiology, Temple University, Philadelphia, Pennsylvania, USA
| | - Miao He
- Palmieri Metabolic Disease Laboratory, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA.,Department of Pathology and Laboratory Medicine, Perelman School of Medicine, and
| | - Nicholas J Hand
- Department of Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Daniel J Rader
- Department of Medicine, Perelman School of Medicine, and.,Institute for Translational Medicine and Therapeutics.,Department of Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| |
Collapse
|