1
|
Li S, Li Z, Wu Q, Luo X, Shen Y. Psychiatric assessment and therapy in an adolescent with ALG6-CDG: a six-month follow-up case report. Eur Child Adolesc Psychiatry 2024:10.1007/s00787-024-02564-x. [PMID: 39141102 DOI: 10.1007/s00787-024-02564-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/07/2023] [Accepted: 08/07/2024] [Indexed: 08/15/2024]
Abstract
ALG6-congenital disorder of glycosylation (ALG6-CDG) is a complex of rare inherited disorders caused by mutations in the ALG6 gene, which encodes the α-1,3-glucosyltransferase enzyme required for N-glycosylation. ALG6-CDG affects multiple systems and exhibits clinical heterogeneity. Besides developmental delays and neurological signs and symptoms, behavioral and psychological symptoms are also an important group of clinical features of ALG6-CDG. Here, we present the case of a 17-year-old Chinese girl with ALG6-CDG who first visited the psychiatric department with apathy, language reduction, and substupor symptoms. The psychiatric assessments and treatment processes performed are described and discussed in this report. During diagnostic process, we found a novel mutation, c.849delT, in ALG6 by whole-exome sequencing. The patient's symptoms improved with escitalopram and risperidone treatment. However, above a certain dosage, she was sensitive to extrapyramidal side effects. This study accumulates clinical experience for diagnosing and treating ALG6-CDG and improves our understanding of this rare genetic disorder.
Collapse
Affiliation(s)
- Sihong Li
- Department of Psychiatry, and National Clinical Research Center for Mental Disorders, The Second Xiangya Hospital of Central South University, Changsha, 410011, Hunan, China
| | - Zexuan Li
- Department of Psychiatry, and National Clinical Research Center for Mental Disorders, The Second Xiangya Hospital of Central South University, Changsha, 410011, Hunan, China
| | - Qiuxia Wu
- Department of Psychiatry, and National Clinical Research Center for Mental Disorders, The Second Xiangya Hospital of Central South University, Changsha, 410011, Hunan, China
| | - Xuerong Luo
- Department of Psychiatry, and National Clinical Research Center for Mental Disorders, The Second Xiangya Hospital of Central South University, Changsha, 410011, Hunan, China.
| | - Yidong Shen
- Department of Psychiatry, and National Clinical Research Center for Mental Disorders, The Second Xiangya Hospital of Central South University, Changsha, 410011, Hunan, China.
| |
Collapse
|
2
|
Boulogne F, Claus LR, Wiersma H, Oelen R, Schukking F, de Klein N, Li S, Westra HJ, van der Zwaag B, van Reekum F, Sierks D, Schönauer R, Li Z, Bijlsma EK, Bos WJW, Halbritter J, Knoers NVAM, Besse W, Deelen P, Franke L, van Eerde AM. KidneyNetwork: using kidney-derived gene expression data to predict and prioritize novel genes involved in kidney disease. Eur J Hum Genet 2023; 31:1300-1308. [PMID: 36807342 PMCID: PMC10620423 DOI: 10.1038/s41431-023-01296-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2022] [Revised: 11/24/2022] [Accepted: 01/18/2023] [Indexed: 02/22/2023] Open
Abstract
Genetic testing in patients with suspected hereditary kidney disease may not reveal the genetic cause for the disorder as potentially pathogenic variants can reside in genes that are not yet known to be involved in kidney disease. We have developed KidneyNetwork, that utilizes tissue-specific expression to inform candidate gene prioritization specifically for kidney diseases. KidneyNetwork is a novel method constructed by integrating a kidney RNA-sequencing co-expression network of 878 samples with a multi-tissue network of 31,499 samples. It uses expression patterns and established gene-phenotype associations to predict which genes could be related to what (disease) phenotypes in an unbiased manner. We applied KidneyNetwork to rare variants in exome sequencing data from 13 kidney disease patients without a genetic diagnosis to prioritize candidate genes. KidneyNetwork can accurately predict kidney-specific gene functions and (kidney disease) phenotypes for disease-associated genes. The intersection of prioritized genes with genes carrying rare variants in a patient with kidney and liver cysts identified ALG6 as plausible candidate gene. We strengthen this plausibility by identifying ALG6 variants in several cystic kidney and liver disease cases without alternative genetic explanation. We present KidneyNetwork, a publicly available kidney-specific co-expression network with optimized gene-phenotype predictions for kidney disease phenotypes. We designed an easy-to-use online interface that allows clinicians and researchers to use gene expression and co-regulation data and gene-phenotype connections to accelerate advances in hereditary kidney disease diagnosis and research. TRANSLATIONAL STATEMENT: Genetic testing in patients with suspected hereditary kidney disease may not reveal the genetic cause for the patient's disorder. Potentially pathogenic variants can reside in genes not yet known to be involved in kidney disease, making it difficult to interpret the relevance of these variants. This reveals a clear need for methods to predict the phenotypic consequences of genetic variation in an unbiased manner. Here we describe KidneyNetwork, a tool that utilizes tissue-specific expression to predict kidney-specific gene functions. Applying KidneyNetwork to a group of undiagnosed cases identified ALG6 as a candidate gene in cystic kidney and liver disease. In summary, KidneyNetwork can aid the interpretation of genetic variants and can therefore be of value in translational nephrogenetics and help improve the diagnostic yield in kidney disease patients.
Collapse
Affiliation(s)
- Floranne Boulogne
- Department of Genetics, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
- Oncode Institute, Utrecht, The Netherlands
| | - Laura R Claus
- Department of Genetics, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Henry Wiersma
- Department of Genetics, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Roy Oelen
- Department of Genetics, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
- Oncode Institute, Utrecht, The Netherlands
| | - Floor Schukking
- Department of Genetics, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Niek de Klein
- Department of Genetics, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Shuang Li
- Department of Genetics, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
- Genomics Coordination Center, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Harm-Jan Westra
- Department of Genetics, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
- Oncode Institute, Utrecht, The Netherlands
| | - Bert van der Zwaag
- Department of Genetics, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Franka van Reekum
- Department of Nephrology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Dana Sierks
- Medical Department III - Endocrinology, Nephrology, Rheumatology Department of Internal Medicine, Division of Nephrology, University of Leipzig Medical Center, Leipzig, Germany
| | - Ria Schönauer
- Medical Department III - Endocrinology, Nephrology, Rheumatology Department of Internal Medicine, Division of Nephrology, University of Leipzig Medical Center, Leipzig, Germany
- Department of Nephrology and Medical Intensive Care, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Zhigui Li
- Department of Internal Medicine (Nephrology), Yale School of Medicine, New Haven, CT, USA
| | - Emilia K Bijlsma
- Department of Clinical Genetics, Leiden University Medical Center, Leiden, The Netherlands
| | - Willem Jan W Bos
- Department of Internal Medicine, St Antonius Hospital, Nieuwegein, The Netherlands
- Department of Internal Medicine, Leiden University Medical Center, Leiden, The Netherlands
| | - Jan Halbritter
- Medical Department III - Endocrinology, Nephrology, Rheumatology Department of Internal Medicine, Division of Nephrology, University of Leipzig Medical Center, Leipzig, Germany
- Department of Nephrology and Medical Intensive Care, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Nine V A M Knoers
- Department of Genetics, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Whitney Besse
- Department of Internal Medicine (Nephrology), Yale School of Medicine, New Haven, CT, USA
| | - Patrick Deelen
- Department of Genetics, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
- Oncode Institute, Utrecht, The Netherlands
- Department of Genetics, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Lude Franke
- Department of Genetics, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
- Oncode Institute, Utrecht, The Netherlands
| | - Albertien M van Eerde
- Department of Genetics, University Medical Center Utrecht, Utrecht, The Netherlands.
| |
Collapse
|
3
|
Uzunyayla-Inci G, Kiykim E, Zubarioglu T, Yesil G, Aktuglu Zeybek C. Autism Spectrum Disorder in Two Unrelated Patients with Homozygous Variants in Either ALG8 or ALG11. Mol Syndromol 2023; 14:428-432. [PMID: 37901858 PMCID: PMC10601802 DOI: 10.1159/000530118] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2023] [Accepted: 03/07/2023] [Indexed: 10/31/2023] Open
Abstract
Background Autism spectrum disorder (ASD) is used to describe individuals with a specific combination of disorders in social communication and repetitive behaviors, highly restricted interests, and/or sensory behavior that begin early in life. The prevalence of ASD has been increasing rapidly in recent years. Pathophysiology of ASDs remains still unclear; however, genetic defects and multifactorial causes have been reported to play an important role in genetic disorders. The prevalence of inborn errors of metabolism (IEM) reported among patients with ASD is 2-5%. The clinical presentation of congenital disorders of glycosylation (CDG) may be in the form of psychiatric disorder only. Case Study Case 1: a 5-year-old female patient was admitted for investigation of ASD. She had a dysmorphic facial appearance, inverted nipples, abnormal fat distribution, ataxic gait, and autistic features. Her transferrin isoelectric focusing test was compatible with a type 1 CDG pattern. A homozygous variant in ALG8 gene revealed the diagnosis of ALG8-CDG (CDG Type 1H). Case 2: a 2-year-old male patient was admitted with complaints of ASD for investigation of an underlying IEM due to speech delay. Physical examination revealed hypertelorism, small hands, and autistic behavior. Transferrin isoelectric focusing test was also found normal. As a result of the WES, a homozygous variant was detected in ALG11 confirming the diagnosis of CDG type 1p. Conclusion CDG should also be considered in the differential diagnosis of autistic patients with dysmorphic findings. The aim of our study was to emphasize that autism should be listed among the neurological findings of CDG.
Collapse
Affiliation(s)
- Gozde Uzunyayla-Inci
- Division of Nutrition and Metabolism, Department of Pediatrics, Cerrahpaşa Medical Faculty, Istanbul University-Cerrahpaşa, Istanbul, Turkey
| | - Ertugrul Kiykim
- Division of Nutrition and Metabolism, Department of Pediatrics, Cerrahpaşa Medical Faculty, Istanbul University-Cerrahpaşa, Istanbul, Turkey
| | - Tanyel Zubarioglu
- Division of Nutrition and Metabolism, Department of Pediatrics, Cerrahpaşa Medical Faculty, Istanbul University-Cerrahpaşa, Istanbul, Turkey
| | - Gozde Yesil
- Department of Medical Genetics, İstanbul Medical Faculty, Istanbul University, Istanbul, Turkey
| | - Cigdem Aktuglu Zeybek
- Division of Nutrition and Metabolism, Department of Pediatrics, Cerrahpaşa Medical Faculty, Istanbul University-Cerrahpaşa, Istanbul, Turkey
| |
Collapse
|
4
|
De Leon DD, Arnoux JB, Banerjee I, Bergada I, Bhatti T, Conwell LS, Fu J, Flanagan SE, Gillis D, Meissner T, Mohnike K, Pasquini TL, Shah P, Stanley CA, Vella A, Yorifuji T, Thornton PS. International Guidelines for the Diagnosis and Management of Hyperinsulinism. Horm Res Paediatr 2023; 97:279-298. [PMID: 37454648 PMCID: PMC11124746 DOI: 10.1159/000531766] [Citation(s) in RCA: 4] [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: 11/01/2022] [Accepted: 05/16/2023] [Indexed: 07/18/2023] Open
Abstract
BACKGROUND Hyperinsulinism (HI) due to dysregulation of pancreatic beta-cell insulin secretion is the most common and most severe cause of persistent hypoglycemia in infants and children. In the 65 years since HI in children was first described, there has been a dramatic advancement in the diagnostic tools available, including new genetic techniques and novel radiologic imaging for focal HI; however, there have been almost no new therapeutic modalities since the development of diazoxide. SUMMARY Recent advances in neonatal research and genetics have improved our understanding of the pathophysiology of both transient and persistent forms of neonatal hyperinsulinism. Rapid turnaround of genetic test results combined with advanced radiologic imaging can permit identification and localization of surgically-curable focal lesions in a large proportion of children with congenital forms of HI, but are only available in certain centers in "developed" countries. Diazoxide, the only drug currently approved for treating HI, was recently designated as an "essential medicine" by the World Health Organization but has been approved in only 16% of Latin American countries and remains unavailable in many under-developed areas of the world. Novel treatments for HI are emerging, but they await completion of safety and efficacy trials before being considered for clinical use. KEY MESSAGES This international consensus statement on diagnosis and management of HI was developed in order to assist specialists, general pediatricians, and neonatologists in early recognition and treatment of HI with the ultimate aim of reducing the prevalence of brain injury caused by hypoglycemia. A previous statement on diagnosis and management of HI in Japan was published in 2017. The current document provides an updated guideline for management of infants and children with HI and includes potential accommodations for less-developed regions of the world where resources may be limited.
Collapse
Affiliation(s)
- Diva D. De Leon
- Congenital Hyperinsulinism Center and Division of Endocrinology and Diabetes, Department of Pediatrics, Children’s Hospital of Philadelphia, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| | - Jean Baptiste Arnoux
- Reference Center for Inherited Metabolic Diseases, Necker-Enfants Malades Hospital, AP-HP, University of Paris-Cité, Paris, France
| | - Indraneel Banerjee
- Paediatric Endocrinology, Royal Manchester Children’s Hospital, University of Manchester, Manchester, UK
| | - Ignacio Bergada
- Centro de Investigaciones Endocrinológicas “Dr. César Bergadá” (CONICET – FEI), Division de Endrocrinología, Hospital de Niños Ricardo Gutiérrez, Buenos Aires, Argentina
| | - Tricia Bhatti
- Department of Clinical Pathology and Laboratory Medicine, Children’s Hospital of Philadelphia and Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| | - Louise S. Conwell
- Australia and Children’s Health Queensland Clinical Unit, Department of Endocrinology and Diabetes, Queensland Children’s Hospital, Children’s Health Queensland, Greater Brisbane Clinical School, Medical School, Faculty of Medicine, University of Queensland, Brisbane, QLD, Australia
| | - Junfen Fu
- National Clinical Research Center for Child Health, Department of Endocrinology, The Children’s Hospital of Zhejiang University School of Medicine, Hangzhou, China
| | - Sarah E. Flanagan
- Institute of Biomedical and Clinical Science, University of Exeter Medical School, Exeter, UK
| | - David Gillis
- Hadassah Medical Center, Department of Pediatrics, Ein-Kerem, Jerusalem and Faculty of Medicine, Hebrew-University, Jerusalem, Israel
| | - Thomas Meissner
- Department of General Pediatrics, Neonatology and Pediatric Cardiology, University Children’s Hospital, Medical Faculty, Heinrich Heine University, Duesseldorf, Germany
| | - Klaus Mohnike
- Department of General Pediatrics, Otto-von-Guericke University Magdeburg, Magdeburg, Germany
| | - Tai L.S. Pasquini
- Research and Policy Director, Congenital Hyperinsulinism International, Glen Ridge, NJ, USA
| | - Pratik Shah
- Pediatric Endocrinology, The Royal London Children’s Hospital, Queen Mary University of London, London, UK
| | - Charles A. Stanley
- Congenital Hyperinsulinism Center and Division of Endocrinology and Diabetes, Department of Pediatrics, Children’s Hospital of Philadelphia, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| | - Adrian Vella
- Division of Diabetes, Endocrinology and Metabolism, Mayo Clinic, Rochester, MN, USA
| | - Tohru Yorifuji
- Pediatric Endocrinology and Metabolism, Children’s Medical Center, Osaka City General Hospital, Osaka, Japan
| | - Paul S. Thornton
- Congenital Hyperinsulinism Center, Cook Children’s Medical Center and Texas Christian University Burnett School of Medicine, Fort Worth, TX, USA
| |
Collapse
|
5
|
Lee HF, Chi CS. Congenital disorders of glycosylation and infantile epilepsy. Epilepsy Behav 2023; 142:109214. [PMID: 37086590 DOI: 10.1016/j.yebeh.2023.109214] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/18/2022] [Revised: 04/01/2023] [Accepted: 04/04/2023] [Indexed: 04/24/2023]
Abstract
Congenital disorders of glycosylation (CDG) are a group of rare inherited metabolic disorders caused by defects in various defects of protein or lipid glycosylation pathways. The symptoms and signs of CDG usually develop in infancy. Epilepsy is commonly observed in CDG individuals and is often a presenting symptom. These epilepsies can present across the lifespan, share features of refractoriness to antiseizure medications, and are often associated with comorbid developmental delay, psychomotor regression, intellectual disability, and behavioral problems. In this review, we discuss CDG and infantile epilepsy, focusing on an overview of clinical manifestations and electroencephalographic features. Finally, we propose a tiered approach that will permit a clinician to systematically investigate and identify CDG earlier, and furthermore, to provide genetic counseling for the family.
Collapse
Affiliation(s)
- Hsiu-Fen Lee
- Department of Post-Baccalaureate Medicine, College of Medicine, National Chung Hsing University, 145, Xingda Rd., Taichung 402, Taiwan; Division of Pediatric Neurology, Children's Medical Center, Taichung Veterans General Hospital, 1650, Taiwan Boulevard Sec. 4, Taichung 407, Taiwan.
| | - Ching-Shiang Chi
- Division of Pediatric Neurology, Children's Medical Center, Taichung Veterans General Hospital, 1650, Taiwan Boulevard Sec. 4, Taichung 407, Taiwan.
| |
Collapse
|
6
|
Clark DJ, Murray T, Drees M, Kulkarni N. A Case of ALG6-CDG with Explosive Onset of Intractable Epilepsy During Infancy. Child Neurol Open 2023; 10:2329048X231153781. [PMID: 36756224 PMCID: PMC9900650 DOI: 10.1177/2329048x231153781] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2022] [Revised: 12/15/2022] [Accepted: 01/09/2023] [Indexed: 02/05/2023] Open
Abstract
ALG6-CDG is a rare, but second most common, type 1 congenital disorder of glycosylation (CDG) caused by a defect in the α-1-3-glucosyltransferase (ALG6) enzyme in the N-glycan assembly pathway. Many mutations have been identified and inherited in an autosomal recessive pattern. There are less than 100 ALG6-CDG cases reported, all sharing the phenotype of hypotonia and developmental delay. The majority (perhaps >70%) have seizures, but a minority have intractable epilepsy or epileptic encephalopathy. We report the clinical course, EEG findings, and neuroimaging of a child found to have compound heterozygous alleles c.257 + 5G > A and c.680G > A (p.G227E) who developed explosive onset of intractable epilepsy and epileptic encephalopathy. Initially, CDG was not suspected due to its rarity and lack of multi-organ system involvement, but rapid whole exam sequence (8-day turnaround) revealed the specific diagnosis quickly.
Collapse
Affiliation(s)
- Daniel James Clark
- Division of Neurology, Nationwide Children's
Hospital, Columbus, OH, USA,Daniel James Clark, Division of Neurology,
Nationwide Children's Hospital, 700 Children's Drive, Columbus, OH 43205, USA.
| | - Thomas Murray
- Division of Neurology, Nationwide Children's
Hospital, Columbus, OH, USA
| | | | - Neil Kulkarni
- Division of Neurology, Nationwide Children's
Hospital, Columbus, OH, USA
| |
Collapse
|
7
|
Paprocka J. Neurological Consequences of Congenital Disorders of Glycosylation. ADVANCES IN NEUROBIOLOGY 2023; 29:219-253. [PMID: 36255677 DOI: 10.1007/978-3-031-12390-0_8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/16/2023]
Abstract
The chapter is devoted to neurological aspects of congenital disorders of glycosylation (CDG). At the beginning, the various types of CDG with neurological presentation of symptoms are summarized. Then, the occurrence of various neurological constellation of abnormalities (for example: epilepsy, brain anomalies on neuroimaging, ataxia, stroke-like episodes, autistic features) in different CDG types are discussed followed by data on possible biomarkers and limited treatment options.
Collapse
Affiliation(s)
- Justyna Paprocka
- Department of Pediatric Neurology, Faculty of Medical Sciences, Medical University of Silesia, Katowice, Poland.
| |
Collapse
|
8
|
Franzka P, Turecki G, Cubillos S, Kentache T, Steiner J, Walter M, Hübner CA, Engmann O. Altered mannose metabolism in chronic stress and depression is rapidly reversed by vitamin B12. Front Nutr 2022; 9:981511. [PMID: 36313076 PMCID: PMC9609420 DOI: 10.3389/fnut.2022.981511] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2022] [Accepted: 09/15/2022] [Indexed: 11/13/2022] Open
Abstract
GDP-Mannose Pyrophosphorylase B (GMPPB) is a key enzyme for glycosylation. Previous studies suggested a dysregulation of GMPBB and mannose in depression. Evidence, however, was sporadic and interventions to reverse these changes are unknown. Here, we show that GMPPB protein, but not RNA abundance is increased in the postmortem prefrontal cortex (PFC) of depressed patients and the chronic variable stress (CVS) mouse-model. This is accompanied by higher plasma mannose levels. Importantly, a single dose of intraperitoneally administered vitamin B12, which has previously been shown to rapidly reverse behavioral symptoms and molecular signatures of chronic stress in mice, normalized GMPPB plasma mannose levels and elevated GDP-mannose abundance. In summary, these data underline metabolic dysregulation in chronic stress and depression and provide further support for rapid effects of vitamin B12 on chronic stress.
Collapse
Affiliation(s)
- Patricia Franzka
- Institute of Human Genetics, University Hospital Jena, Friedrich Schiller University, Jena, Germany
| | - Gustavo Turecki
- Department of Psychiatry, Douglas Mental Health University Institute, McGill University, Montreal, QC, Canada
| | - Susana Cubillos
- Institute for Biochemistry and Biophysics, Friedrich-Schiller-University Jena, Jena, Germany
| | | | - Johann Steiner
- Clinic for Psychiatry and Psychotherapy, University Hospital of Otto-von-Guericke-Universität Magdeburg, Magdeburg, Germany
| | - Martin Walter
- Department of Psychiatry and Psychotherapy, Jena University Hospital, Jena, Germany
| | - Christian A. Hübner
- Institute of Human Genetics, University Hospital Jena, Friedrich Schiller University, Jena, Germany
| | - Olivia Engmann
- Institute of Human Genetics, University Hospital Jena, Friedrich Schiller University, Jena, Germany,Institute for Biochemistry and Biophysics, Friedrich-Schiller-University Jena, Jena, Germany,*Correspondence: Olivia Engmann,
| |
Collapse
|
9
|
Badshah N, Mattison KA, Ahmad S, Chopra P, Johnston HR, Ahmad S, Khan SH, Sarwar MT, Cutler DJ, Taylor M, Vadlamani G, Zwick ME, Escayg A. Novel Missense CNTNAP2 Variant Identified in Two Consanguineous Pakistani Families With Developmental Delay, Epilepsy, Intellectual Disability, and Aggressive Behavior. Front Neurol 2022; 13:918022. [PMID: 35911904 PMCID: PMC9329621 DOI: 10.3389/fneur.2022.918022] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2022] [Accepted: 06/13/2022] [Indexed: 11/13/2022] Open
Abstract
We report the genetic analysis of two consanguineous pedigrees of Pakistani ancestry in which two siblings in each family exhibited developmental delay, epilepsy, intellectual disability and aggressive behavior. Whole-genome sequencing was performed in Family 1, and we identified ~80,000 variants located in regions of homozygosity. Of these, 615 variants had a minor allele frequency ≤ 0.001, and 21 variants had CADD scores ≥ 15. Four homozygous exonic variants were identified in both affected siblings: PDZD7 (c.1348_1350delGAG, p.Glu450del), ALG6 (c.1033G>C, p.Glu345Gln), RBM20 (c.1587C>G, p.Ser529Arg), and CNTNAP2 (c.785G>A, p.Gly228Arg). Sanger sequencing revealed co-segregation of the PDZD7, RBM20, and CNTNAP2 variants with disease in Family 1. Pathogenic variants in PDZD7 and RBM20 are associated with autosomal recessive non-syndromic hearing loss and autosomal dominant dilated cardiomyopathy, respectively, suggesting that these variants are unlikely likely to contribute to the clinical presentation. Gene panel analysis was performed on the two affected siblings in Family 2, and they were found to also be homozygous for the p.Gly228Arg CNTNAP2 variant. Together these families provide a LOD score 2.9 toward p.Gly228Arg CNTNAP2 being a completely penetrant recessive cause of this disease. The clinical presentation of the affected siblings in both families is also consistent with previous reports from individuals with homozygous CNTNAP2 variants where at least one allele was a nonsense variant, frameshift or small deletion. Our data suggests that homozygous CNTNAP2 missense variants can also contribute to disease, thereby expanding the genetic landscape of CNTNAP2 dysfunction.
Collapse
Affiliation(s)
- Noor Badshah
- Institute of Biotechnology and Genetic Engineering, University of Agriculture Peshawar, Peshawar, Pakistan
- Department of Human Genetics, Emory University, Atlanta, GA, United States
| | - Kari A. Mattison
- Department of Human Genetics, Emory University, Atlanta, GA, United States
- Genetics and Molecular Biology Graduate Program, Graduate Division of Biological and Biomedical Sciences, Laney Graduate School, Emory University, Atlanta, GA, United States
| | - Sohail Ahmad
- Institute of Biotechnology and Genetic Engineering, University of Agriculture Peshawar, Peshawar, Pakistan
| | - Pankaj Chopra
- Department of Human Genetics, Emory University, Atlanta, GA, United States
| | | | - Shakoor Ahmad
- Department of Animal Health, University of Agriculture Peshawar, Peshawar, Pakistan
| | - Sher Hayat Khan
- Institute of Biotechnology and Genetic Engineering, University of Agriculture Peshawar, Peshawar, Pakistan
| | - Muhammad Tahir Sarwar
- Department of Molecular Biology and Genetics, Institute of Basic Medical Sciences, Khyber Medical University, Peshawar, Pakistan
| | - David J. Cutler
- Department of Human Genetics, Emory University, Atlanta, GA, United States
| | - Micheal Taylor
- Department of Pediatric Neurology, Leeds Teaching Hospital NHS Trust, Leeds, United Kingdom
| | - Gayatri Vadlamani
- Department of Pediatric Neurology, Leeds Teaching Hospital NHS Trust, Leeds, United Kingdom
| | - Michael E. Zwick
- Department of Human Genetics, Emory University, Atlanta, GA, United States
| | - Andrew Escayg
- Department of Human Genetics, Emory University, Atlanta, GA, United States
- *Correspondence: Andrew Escayg
| |
Collapse
|
10
|
Boyer SW, Johnsen C, Morava E. Nutrition interventions in congenital disorders of glycosylation. Trends Mol Med 2022; 28:463-481. [PMID: 35562242 DOI: 10.1016/j.molmed.2022.04.003] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2021] [Revised: 03/30/2022] [Accepted: 04/04/2022] [Indexed: 12/13/2022]
Abstract
Congenital disorders of glycosylation (CDG) are a group of more than 160 inborn errors of metabolism affecting multiple pathways of protein and lipid glycosylation. Patients present with a wide range of symptoms and therapies are only available for very few subtypes. Specific nutritional treatment options for certain CDG types include oral supplementation of monosaccharide sugars, manganese, uridine, or pyridoxine. Additional management includes specific diets (i.e., complex carbohydrate or ketogenic diet), iron supplementation, and albumin infusions. We review the dietary management in CDG with a focus on two subgroups: N-linked glycosylation defects and GPI-anchor disorders.
Collapse
Affiliation(s)
- Suzanne W Boyer
- Department of Clinical Genomics, Mayo Clinic, Rochester, MN, USA
| | - Christin Johnsen
- Department of Clinical Genomics, Mayo Clinic, Rochester, MN, USA
| | - Eva Morava
- Department of Clinical Genomics, Mayo Clinic, Rochester, MN, USA.
| |
Collapse
|
11
|
Sena RM, Twiss JL, Gardiner AS, Dell’Orco M, Linsenbardt DN, Perrone-Bizzozero NI. The RNA-Binding Protein HuD Regulates Alternative Splicing and Alternative Polyadenylation in the Mouse Neocortex. Molecules 2021; 26:2836. [PMID: 34064652 PMCID: PMC8151252 DOI: 10.3390/molecules26102836] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2021] [Revised: 05/03/2021] [Accepted: 05/05/2021] [Indexed: 11/18/2022] Open
Abstract
The neuronal Hu/ELAV-like proteins HuB, HuC and HuD are a class of RNA-binding proteins that are crucial for proper development and maintenance of the nervous system. These proteins bind to AU-rich elements (AREs) in the untranslated regions (3'-UTRs) of target mRNAs regulating mRNA stability, transport and translation. In addition to these cytoplasmic functions, Hu proteins have been implicated in alternative splicing and alternative polyadenylation in the nucleus. The purpose of this study was to identify transcriptome-wide effects of HuD deletion on both of these nuclear events using RNA sequencing data obtained from the neocortex of Elavl4-/- (HuD KO) mice. HuD KO affected alternative splicing of 310 genes, including 17 validated HuD targets such as Cbx3, Cspp1, Snap25 and Gria2. In addition, deletion of HuD affected polyadenylation of 53 genes, with the majority of significantly altered mRNAs shifting towards usage of proximal polyadenylation signals (PAS), resulting in shorter 3'-UTRs. None of these genes overlapped with those showing alternative splicing events. Overall, HuD KO had a greater effect on alternative splicing than polyadenylation, with many of the affected genes implicated in several neuronal functions and neuropsychiatric disorders.
Collapse
Affiliation(s)
- Rebecca M. Sena
- Department Neurosciences, University of New Mexico School of Medicine, Albuquerque, NM 87131, USA; (R.M.S.); (A.S.G.); (M.D.)
| | - Jeffery L. Twiss
- Department Biological Sciences, University of South Carolina, Columbia, SC 29208, USA;
| | - Amy S. Gardiner
- Department Neurosciences, University of New Mexico School of Medicine, Albuquerque, NM 87131, USA; (R.M.S.); (A.S.G.); (M.D.)
- Department Cell Biology and Physiology, University of New Mexico School of Medicine, Albuquerque, NM 87131, USA
| | - Michela Dell’Orco
- Department Neurosciences, University of New Mexico School of Medicine, Albuquerque, NM 87131, USA; (R.M.S.); (A.S.G.); (M.D.)
| | - David N. Linsenbardt
- Department Neurosciences, University of New Mexico School of Medicine, Albuquerque, NM 87131, USA; (R.M.S.); (A.S.G.); (M.D.)
| | - Nora I. Perrone-Bizzozero
- Department Neurosciences, University of New Mexico School of Medicine, Albuquerque, NM 87131, USA; (R.M.S.); (A.S.G.); (M.D.)
| |
Collapse
|
12
|
Wong ML, Arcos-Burgos M, Liu S, Licinio AW, Yu C, Chin EWM, Yao WD, Lu XY, Bornstein SR, Licinio J. Rare Functional Variants Associated with Antidepressant Remission in Mexican-Americans: Short title: Antidepressant remission and pharmacogenetics in Mexican-Americans. J Affect Disord 2021; 279:491-500. [PMID: 33128939 PMCID: PMC7953425 DOI: 10.1016/j.jad.2020.10.027] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/10/2020] [Revised: 08/24/2020] [Accepted: 10/11/2020] [Indexed: 12/14/2022]
Abstract
INTRODUCTION Rare genetic functional variants can contribute to 30-40% of functional variability in genes relevant to drug action. Therefore, we investigated the role of rare functional variants in antidepressant response. METHOD Mexican-American individuals meeting the Diagnostic and Statistical Manual-IV criteria for major depressive disorder (MDD) participated in a prospective randomized, double-blind study with desipramine or fluoxetine. The rare variant analysis was performed using whole-exome genotyping data. Network and pathway analyses were carried out with the list of significant genes. RESULTS The Kernel-Based Adaptive Cluster method identified functional rare variants in 35 genes significantly associated with treatment remission (False discovery rate, FDR <0.01). Pathway analysis of these genes supports the involvement of the following gene ontology processes: olfactory/sensory transduction, regulation of response to cytokine stimulus, and meiotic cell cycleprocess. LIMITATIONS Our study did not have a placebo arm. We were not able to use antidepressant blood level as a covariate. Our study is based on a small sample size of only 65 Mexican-American individuals. Further studies using larger cohorts are warranted. CONCLUSION Our data identified several rare functional variants in antidepressant drug response in MDD patients. These have the potential to serve as genetic markers for predicting drug response. TRIAL REGISTRATION ClinicalTrials.gov NCT00265291.
Collapse
Affiliation(s)
- Ma-Li Wong
- Department of Psychiatry and Behavioral Sciences, State University of New York, Upstate Medical University, Syracuse, NY, USA; Department of Neuroscience and Physiology, State University of New York, Upstate Medical University, Syracuse, NY, USA; Mind & Brain Theme, South Australian Health and Medical Research Institute Adelaide, South Australia, Australia; Department of Psychiatry, Flinders University College of Medicine and Public Health, Bedford Park, South Australia, Australia.
| | - Mauricio Arcos-Burgos
- Grupo de Investigación en Psiquiatría, Departamento de Psiquiatría, Instituto de Investigaciones Médicas, Facultad de Medicina, Universidad de Antioquia, Medellin, Antioquia, Colombia
| | - Sha Liu
- Mind & Brain Theme, South Australian Health and Medical Research Institute Adelaide, South Australia, Australia
| | - Alice W Licinio
- Mind & Brain Theme, South Australian Health and Medical Research Institute Adelaide, South Australia, Australia
| | - Chenglong Yu
- Mind & Brain Theme, South Australian Health and Medical Research Institute Adelaide, South Australia, Australia; Department of Psychiatry, Flinders University College of Medicine and Public Health, Bedford Park, South Australia, Australia
| | - Eunice W M Chin
- Department of Psychiatry and Behavioral Sciences, State University of New York, Upstate Medical University, Syracuse, NY, USA
| | - Wei-Dong Yao
- Department of Psychiatry and Behavioral Sciences, State University of New York, Upstate Medical University, Syracuse, NY, USA; Department of Neuroscience and Physiology, State University of New York, Upstate Medical University, Syracuse, NY, USA
| | - Xin-Yun Lu
- Department of Neuroscience & Regenerative Medicine, Medical College of Georgia at Augusta University, Augusta, GA, USA
| | - Stefan R Bornstein
- Medical Clinic III, Carl Gustav Carus University Hospital, Dresden University of Technology, Dresden, Germany
| | - Julio Licinio
- Department of Psychiatry and Behavioral Sciences, State University of New York, Upstate Medical University, Syracuse, NY, USA; Department of Neuroscience and Physiology, State University of New York, Upstate Medical University, Syracuse, NY, USA; Mind & Brain Theme, South Australian Health and Medical Research Institute Adelaide, South Australia, Australia; Department of Psychiatry, Flinders University College of Medicine and Public Health, Bedford Park, South Australia, Australia.
| |
Collapse
|
13
|
Paprocka J, Jezela-Stanek A, Tylki-Szymańska A, Grunewald S. Congenital Disorders of Glycosylation from a Neurological Perspective. Brain Sci 2021; 11:brainsci11010088. [PMID: 33440761 PMCID: PMC7827962 DOI: 10.3390/brainsci11010088] [Citation(s) in RCA: 54] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2020] [Revised: 01/01/2021] [Accepted: 01/04/2021] [Indexed: 12/11/2022] Open
Abstract
Most plasma proteins, cell membrane proteins and other proteins are glycoproteins with sugar chains attached to the polypeptide-glycans. Glycosylation is the main element of the post-translational transformation of most human proteins. Since glycosylation processes are necessary for many different biological processes, patients present a diverse spectrum of phenotypes and severity of symptoms. The most frequently observed neurological symptoms in congenital disorders of glycosylation (CDG) are: epilepsy, intellectual disability, myopathies, neuropathies and stroke-like episodes. Epilepsy is seen in many CDG subtypes and particularly present in the case of mutations in the following genes: ALG13, DOLK, DPAGT1, SLC35A2, ST3GAL3, PIGA, PIGW, ST3GAL5. On brain neuroimaging, atrophic changes of the cerebellum and cerebrum are frequently seen. Brain malformations particularly in the group of dystroglycanopathies are reported. Despite the growing number of CDG patients in the world and often neurological symptoms dominating in the clinical picture, the number of performed screening tests eg transferrin isoforms is systematically decreasing as broadened genetic testing is recently more favored. The aim of the review is the summary of selected neurological symptoms in CDG described in the literature in one paper. It is especially important for pediatric neurologists not experienced in the field of metabolic medicine. It may help to facilitate the diagnosis of this expanding group of disorders. Biochemically, this paper focuses on protein glycosylation abnormalities.
Collapse
Affiliation(s)
- Justyna Paprocka
- Department of Pediatric Neurology, Faculty of Medical Science in Katowice, Medical University of Silesia, 40-752 Katowice, Poland
- Correspondence: ; Tel.: +48-606-415-888
| | - Aleksandra Jezela-Stanek
- Department of Genetics and Clinical Immunology, National Institute of Tuberculosis and Lung Diseases, 01-138 Warsaw, Poland;
| | - Anna Tylki-Szymańska
- Department of Pediatrics, Nutrition and Metabolic Diseases, The Children’s Memorial Health Institute, W 04-730 Warsaw, Poland;
| | - Stephanie Grunewald
- NIHR Biomedical Research Center (BRC), Metabolic Unit, Great Ormond Street Hospital and Institute of Child Health, University College London, London SE1 9RT, UK;
| |
Collapse
|
14
|
Starosta RT, Boyer S, Tahata S, Raymond K, Lee HE, Wolfe LA, Lam C, Edmondson AC, Schwartz IVD, Morava E. Liver manifestations in a cohort of 39 patients with congenital disorders of glycosylation: pin-pointing the characteristics of liver injury and proposing recommendations for follow-up. Orphanet J Rare Dis 2021; 16:20. [PMID: 33413482 PMCID: PMC7788939 DOI: 10.1186/s13023-020-01630-2] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2020] [Accepted: 11/27/2020] [Indexed: 12/19/2022] Open
Abstract
Background The congenital disorders of glycosylation (CDG) are a heterogeneous group of rare metabolic diseases with multi-system involvement. The liver phenotype of CDG varies not only according to the specific disorder, but also from patient to patient. In this study, we sought to identify common patterns of liver injury among patients with a broad spectrum of CDG, and to provide recommendations for follow-up in clinical practice. Methods Patients were enrolled in the Frontiers in Congenital Disorders of Glycosylation natural history study. We analyzed clinical history, molecular genetics, serum markers of liver injury, liver ultrasonography and transient elastography, liver histopathology (when available), and clinical scores of 39 patients with 16 different CDG types (PMM2-CDG, n = 19), with a median age of 7 years (range: 10 months to 65 years). For patients with disorders which are treatable by specific interventions, we have added a description of liver parameters on treatment. Results Our principal findings are (1) there is a clear pattern in the evolution of the hepatocellular injury markers alanine aminotransferase and aspartate aminotransferase according to age, especially in PMM2-CDG patients but also in other CDG-I, and that the cholangiocellular injury marker gamma-glutamyltransferase is not elevated in most patients, pointing to an exclusive hepatocellular origin of injury; (2) there is a dissociation between liver ultrasound and transient elastography regarding signs of liver fibrosis; (3) histopathological findings in liver tissue of PMM2-CDG patients include cytoplasmic glycogen deposits; and (4) most CDG types show more than one type of liver injury.
Conclusions Based on these findings, we recommend that all CDG patients have regular systematic, comprehensive screening for liver disease, including physical examination (for hepatomegaly and signs of liver failure), laboratory tests (serum alanine aminotransferase and aspartate aminotransferase), liver ultrasound (for steatosis and liver tumors), and liver elastography (for fibrosis).
Collapse
Affiliation(s)
- Rodrigo Tzovenos Starosta
- Graduate Program in Genetics and Molecular Biology, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, RS, Brazil. .,Department of Clinical Genomics, Mayo Clinic, Rochester, MN, USA. .,Department of Pediatrics, Washington University in Saint Louis, St. Louis, MO, USA.
| | - Suzanne Boyer
- Department of Clinical Genomics, Mayo Clinic, Rochester, MN, USA
| | - Shawn Tahata
- Department of Internal Medicine, Mayo Clinic, Rochester, MN, USA
| | - Kimiyo Raymond
- Biochemical Genetics Laboratory, Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, USA
| | - Hee Eun Lee
- Biochemical Genetics Laboratory, Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, USA
| | - Lynne A Wolfe
- Undiagnosed Diseases Program, Common Fund, National Institutes of Health, Bethesda, MD, USA
| | - Christina Lam
- Division of Genetic Medicine, University of Washington, Seattle, WA, USA.,Center of Integrated Brain Research, Seattle Children's Research Institute, Seattle, WA, USA
| | - Andrew C Edmondson
- Section of Biochemical Genetics, Division of Human Genetics, Department of Pediatrics, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Ida Vanessa Doederlein Schwartz
- Graduate Program in Genetics and Molecular Biology, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, RS, Brazil.,Service of Medical Genetics, Hospital de Clínicas de Porto Alegre, UFRGS, Porto Alegre, RS, Brazil
| | - Eva Morava
- Department of Clinical Genomics, Mayo Clinic, Rochester, MN, USA.,Biochemical Genetics Laboratory, Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, USA.,Center for Individualized Medicine, Mayo Clinic, Rochester, MN, USA
| |
Collapse
|
15
|
Jaeken J. Congenital disorders of glycosylation: A multi-genetic disease family with multiple subcellular locations. JOURNAL OF MOTHER AND CHILD 2020; 24:14-20. [PMID: 33554500 PMCID: PMC8518092 DOI: 10.34763/jmotherandchild.20202402si.2005.000004] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
This review discusses a selection of congenital disorders of glycosylation that show peculiar features, such as an unusual presentation, different phenotypes, a novel biochemical/genetic mechanism, a relatively high frequency or a relatively efficient treatment.
Collapse
Affiliation(s)
- Jaak Jaeken
- Department of Development and Regeneration, Center for Metabolic Diseases, University Hospital Gasthuisberg, KU Leuven, Leuven, Belgium
| |
Collapse
|
16
|
Rossi M, van der Veen S, Merello M, Tijssen MAJ, van de Warrenburg B. Myoclonus-Ataxia Syndromes: A Diagnostic Approach. Mov Disord Clin Pract 2020; 8:9-24. [PMID: 33426154 DOI: 10.1002/mdc3.13106] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2020] [Revised: 09/30/2020] [Accepted: 10/14/2020] [Indexed: 12/30/2022] Open
Abstract
Background A myriad of disorders combine myoclonus and ataxia. Most causes are genetic and an increasing number of genes are being associated with myoclonus-ataxia syndromes (MAS), due to recent advances in genetic techniques. A proper etiologic diagnosis of MAS is clinically relevant, given the consequences for genetic counseling, treatment, and prognosis. Objectives To review the causes of MAS and to propose a diagnostic algorithm. Methods A comprehensive and structured literature search following PRISMA criteria was conducted to identify those disorders that may combine myoclonus with ataxia. Results A total of 135 causes of combined myoclonus and ataxia were identified, of which 30 were charted as the main causes of MAS. These include four acquired entities: opsoclonus-myoclonus-ataxia syndrome, celiac disease, multiple system atrophy, and sporadic prion diseases. The distinction between progressive myoclonus epilepsy and progressive myoclonus ataxia poses one of the main diagnostic dilemmas. Conclusions Diagnostic algorithms for pediatric and adult patients, based on clinical manifestations including epilepsy, are proposed to guide the differential diagnosis and corresponding work-up of the most important and frequent causes of MAS. A list of genes associated with MAS to guide genetic testing strategies is provided. Priority should be given to diagnose or exclude acquired or treatable disorders.
Collapse
Affiliation(s)
- Malco Rossi
- Movement Disorders Section Neuroscience Department Buenos Aires Argentina.,Argentine National Scientific and Technological Research Council (CONICET) Buenos Aires Argentina
| | - Sterre van der Veen
- Pontificia Universidad Católica Argentina (UCA) Buenos Aires Argentina.,Department of Neurology University of Groningen, University Medical Center Groningen Groningen The Netherlands
| | - Marcelo Merello
- Movement Disorders Section Neuroscience Department Buenos Aires Argentina.,Argentine National Scientific and Technological Research Council (CONICET) Buenos Aires Argentina.,Pontificia Universidad Católica Argentina (UCA) Buenos Aires Argentina
| | - Marina A J Tijssen
- Department of Neurology University of Groningen, University Medical Center Groningen Groningen The Netherlands.,Expertise Center Movement Disorders Groningen University Medical Center Groningen (UMCG) Groningen The Netherlands
| | - Bart van de Warrenburg
- Department of Neurology, Donders Institute for Brain, Cognition & Behaviour Radboud University Medical Center Nijmegen The Netherlands
| |
Collapse
|
17
|
Abstract
Based on the analysis of literature, the authors describe the neuropathophysiological mechanism of the formation of synapses, synaptic transmission and plasticity, which may underlie the pathogenesis of autism. The results of some studies confirm the involvement of aberrant expression of genes and proteins of synaptic contacts, cell adhesion molecules p120ctn, CNTN5, CNTN6, activation of NMDA glutamate, TrkB, p75 receptors, Ca2+-input, BDNF, serotonin and testosterone. This leads to an imbalance in the exciting, inhibitory synaptic transmission and forms of synaptic plasticity, including long-term potentiation (LTP) and long-term depression (LTD) at the level of individual neurons and their chains due to suppression of GABA synthesis, expression of its ionotropic and metabotropic receptors, G proteins, NGF, TrkA receptors, a reduction in the number of GABAergic neurons, their contacts and disruption of differentiation. The pathology of the nuclei of the thalamus, especially the reticular nucleus (RN), is associated with a disturbance of the expression of the subunits of metabotropic GABAβ receptors, Ca2+ channels, GABA excretion and the work of chlorine transmitters. These failures do not ensure the inhibitory effect of OC on the exciting associative and ventral nuclei of the thalamus, nor modify the incoming information to the cerebral cortex (CC) from these thalamus nuclei, the dentate gyrus of the hippocampus and the nuclei of the reticular formation. Information propagating into the somatosensory and associative regions of CC is not modified by mirror neurons (MN) when performing arbitrary actions, which prevents the formation of an adequate image in the neural networks of the associative cortex and promotes the development of hyperexcitability, irritability, increased visual and auditory sensitivity, anxiety, and the ability to form a holistic image based on the actions of other people.
Collapse
Affiliation(s)
- A N Chernov
- Almazov National Medical Research Center of the Ministry of Health of Russia, St. Petersburg, Russia
| |
Collapse
|
18
|
Platelets and Defective N-Glycosylation. Int J Mol Sci 2020; 21:ijms21165630. [PMID: 32781578 PMCID: PMC7460655 DOI: 10.3390/ijms21165630] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2020] [Revised: 08/04/2020] [Accepted: 08/04/2020] [Indexed: 12/13/2022] Open
Abstract
N-glycans are covalently linked to an asparagine residue in a simple acceptor sequence of proteins, called a sequon. This modification is important for protein folding, enhancing thermodynamic stability, and decreasing abnormal protein aggregation within the endoplasmic reticulum (ER), for the lifetime and for the subcellular localization of proteins besides other functions. Hypoglycosylation is the hallmark of a group of rare genetic diseases called congenital disorders of glycosylation (CDG). These diseases are due to defects in glycan synthesis, processing, and attachment to proteins and lipids, thereby modifying signaling functions and metabolic pathways. Defects in N-glycosylation and O-glycosylation constitute the largest CDG groups. Clotting and anticlotting factor defects as well as a tendency to thrombosis or bleeding have been described in CDG patients. However, N-glycosylation of platelet proteins has been poorly investigated in CDG. In this review, we highlight normal and deficient N-glycosylation of platelet-derived molecules and discuss the involvement of platelets in the congenital disorders of N-glycosylation.
Collapse
|
19
|
Bosch-Boonstra-Schaaf optic atrophy syndrome (BBSOAS) initially diagnosed as ALG6-CDG: Functional evidence for benignity of the ALG6 c.391T>C (p.Tyr131His) variant and further expanding the BBSOAS phenotype. Eur J Med Genet 2020; 63:103941. [PMID: 32407885 DOI: 10.1016/j.ejmg.2020.103941] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2020] [Revised: 04/29/2020] [Accepted: 05/02/2020] [Indexed: 11/23/2022]
Abstract
Bosch-Boonstra-Schaaf optic atrophy syndrome (BBSOAS) is a recently described autosomal dominant syndrome of developmental delay, cortical vision loss with optic nerve atrophy, epilepsy, and autism spectrum disorder. Due to its many overlapping features with congenital disorders of glycosylation (CDG), the differential diagnosis between these disorders may be difficult and relies on molecular genetic testing. We report on a 31-year-old female initially diagnosed with ALG6-CDG based on glycosylation abnormalities on transferrin isoelectrofocusing and targeted genetic testing, and later diagnosed with BBSOAS by whole-exome sequencing (WES). Functional studies on cultured fibroblasts including Western blotting and RT-qPCR, as well as mass spectrometry of glycosylated transferrin and MALDI-TOF glycan analysis in serum, demonstrated normal glycosylation in this patient. In this report, we extend the phenotype of BBSOAS with ataxia and protein-losing enteropathy. This case is illustrative of the utility of whole exome sequencing in the diagnostic odyssey, and the potential pitfalls of relying on focused genetic testing results for diagnosis of conditions with complex overlapping phenotypes.
Collapse
|
20
|
Durrant C, Fuehring JI, Willemetz A, Chrétien D, Sala G, Ghidoni R, Katz A, Rötig A, Thelestam M, Ermonval M, Moore SEH. Defects in Galactose Metabolism and Glycoconjugate Biosynthesis in a UDP-Glucose Pyrophosphorylase-Deficient Cell Line Are Reversed by Adding Galactose to the Growth Medium. Int J Mol Sci 2020; 21:ijms21062028. [PMID: 32188137 PMCID: PMC7139386 DOI: 10.3390/ijms21062028] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2020] [Revised: 03/05/2020] [Accepted: 03/10/2020] [Indexed: 12/28/2022] Open
Abstract
UDP-glucose (UDP-Glc) is synthesized by UGP2-encoded UDP-Glc pyrophosphorylase (UGP) and is required for glycoconjugate biosynthesis and galactose metabolism because it is a uridyl donor for galactose-1-P (Gal1P) uridyltransferase. Chinese hamster lung fibroblasts harboring a hypomrphic UGP(G116D) variant display reduced UDP-Glc levels and cannot grow if galactose is the sole carbon source. Here, these cells were cultivated with glucose in either the absence or presence of galactose in order to investigate glycoconjugate biosynthesis and galactose metabolism. The UGP-deficient cells display < 5% control levels of UDP-Glc/UDP-Gal and > 100-fold reduction of [6-3H]galactose incorporation into UDP-[6-3H]galactose, as well as multiple deficits in glycoconjugate biosynthesis. Cultivation of these cells in the presence of galactose leads to partial restoration of UDP-Glc levels, galactose metabolism and glycoconjugate biosynthesis. The Vmax for recombinant human UGP(G116D) with Glc1P is 2000-fold less than that of the wild-type protein, and UGP(G116D) displayed a mildly elevated Km for Glc1P, but no activity of the mutant enzyme towards Gal1P was detectable. To conclude, although the mechanism behind UDP-Glc/Gal production in the UGP-deficient cells remains to be determined, the capacity of this cell line to change its glycosylation status as a function of extracellular galactose makes it a useful, reversible model with which to study different aspects of galactose metabolism and glycoconjugate biosynthesis.
Collapse
Affiliation(s)
- Christelle Durrant
- INSERM U1149, Université de Paris, 16 rue Henri Huchard, 75018 Paris, France; (C.D.); (A.W.)
| | - Jana I. Fuehring
- Institute for Clinical Biochemistry, Hannover Medical School, Carl-Neuberg-Str. 1, 30625 Hannover, Germany;
| | - Alexandra Willemetz
- INSERM U1149, Université de Paris, 16 rue Henri Huchard, 75018 Paris, France; (C.D.); (A.W.)
| | - Dominique Chrétien
- UMR1163, Université Paris Decartes, Sorbonnes Paris Cité, Institut Imagine, 24 Boulevard du Montparnasse, 75015 Paris, France; (D.C.); (A.R.)
| | - Giusy Sala
- “Aldo Ravelli” Research Center and Department of Health Sciences, University of Milan, 20146 Milan, Italy; (G.S.); (R.G.)
| | - Riccardo Ghidoni
- “Aldo Ravelli” Research Center and Department of Health Sciences, University of Milan, 20146 Milan, Italy; (G.S.); (R.G.)
| | - Abram Katz
- Department of Physiology and Pharmacology, Karolinska Institutet, 17177 Stockholm, Sweden;
| | - Agnès Rötig
- UMR1163, Université Paris Decartes, Sorbonnes Paris Cité, Institut Imagine, 24 Boulevard du Montparnasse, 75015 Paris, France; (D.C.); (A.R.)
| | - Monica Thelestam
- Department of Cell and Molecular Biology, Karolinska Institutet, 17177 Stockholm, Sweden;
| | - Myriam Ermonval
- Institut Pasteur, Department of Virology, 25 rue du Dr. Roux, 75015 Paris, France;
| | - Stuart E. H. Moore
- INSERM U1149, Université de Paris, 16 rue Henri Huchard, 75018 Paris, France; (C.D.); (A.W.)
- Correspondence:
| |
Collapse
|
21
|
CRISPR/Cas9-mediated mutation of asparagine-linked glycosylation 13 transcript variant 1 causes epilepsy in mice. JOURNAL OF BIO-X RESEARCH 2020. [DOI: 10.1097/jbr.0000000000000059] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
|
22
|
Structure and mechanism of the ER-based glucosyltransferase ALG6. Nature 2020; 579:443-447. [PMID: 32103179 DOI: 10.1038/s41586-020-2044-z] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2019] [Accepted: 01/06/2020] [Indexed: 01/03/2023]
Abstract
In eukaryotic protein N-glycosylation, a series of glycosyltransferases catalyse the biosynthesis of a dolichylpyrophosphate-linked oligosaccharide before its transfer onto acceptor proteins1. The final seven steps occur in the lumen of the endoplasmic reticulum (ER) and require dolichylphosphate-activated mannose and glucose as donor substrates2. The responsible enzymes-ALG3, ALG9, ALG12, ALG6, ALG8 and ALG10-are glycosyltransferases of the C-superfamily (GT-Cs), which are loosely defined as containing membrane-spanning helices and processing an isoprenoid-linked carbohydrate donor substrate3,4. Here we present the cryo-electron microscopy structure of yeast ALG6 at 3.0 Å resolution, which reveals a previously undescribed transmembrane protein fold. Comparison with reported GT-C structures suggests that GT-C enzymes contain a modular architecture with a conserved module and a variable module, each with distinct functional roles. We used synthetic analogues of dolichylphosphate-linked and dolichylpyrophosphate-linked sugars and enzymatic glycan extension to generate donor and acceptor substrates using purified enzymes of the ALG pathway to recapitulate the activity of ALG6 in vitro. A second cryo-electron microscopy structure of ALG6 bound to an analogue of dolichylphosphate-glucose at 3.9 Å resolution revealed the active site of the enzyme. Functional analysis of ALG6 variants identified a catalytic aspartate residue that probably acts as a general base. This residue is conserved in the GT-C superfamily. Our results define the architecture of ER-luminal GT-C enzymes and provide a structural basis for understanding their catalytic mechanisms.
Collapse
|
23
|
Therapeutic approaches in Congenital Disorders of Glycosylation (CDG) involving N-linked glycosylation: an update. Genet Med 2020; 22:268-279. [PMID: 31534212 PMCID: PMC8720509 DOI: 10.1038/s41436-019-0647-2] [Citation(s) in RCA: 48] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2019] [Accepted: 08/20/2019] [Indexed: 02/07/2023] Open
Abstract
Congenital disorders of glycosylation (CDG) are a group of clinically and genetically heterogeneous metabolic disorders. Over 150 CDG types have been described. Most CDG types are ultrarare disorders. CDG types affecting N-glycosylation are the most common type of CDG with emerging therapeutic possibilities. This review is an update on the available therapies for disorders affecting the N-linked glycosylation pathway. In the first part of the review, we highlight the clinical presentation, general principles of management, and disease-specific therapies for N-linked glycosylation CDG types, organized by organ system. The second part of the review focuses on the therapeutic strategies currently available and under development. We summarize the successful (pre-) clinical application of nutritional therapies, transplantation, activated sugars, gene therapy, and pharmacological chaperones and outline the anticipated expansion of the therapeutic possibilities in CDG. We aim to provide a comprehensive update on the treatable aspects of CDG types involving N-linked glycosylation, with particular emphasis on disease-specific treatment options for the involved organ systems; call for natural history studies; and present current and future therapeutic strategies for CDG.
Collapse
|
24
|
Beaudin M, Matilla-Dueñas A, Soong BW, Pedroso JL, Barsottini OG, Mitoma H, Tsuji S, Schmahmann JD, Manto M, Rouleau GA, Klein C, Dupre N. The Classification of Autosomal Recessive Cerebellar Ataxias: a Consensus Statement from the Society for Research on the Cerebellum and Ataxias Task Force. CEREBELLUM (LONDON, ENGLAND) 2019; 18:1098-1125. [PMID: 31267374 PMCID: PMC6867988 DOI: 10.1007/s12311-019-01052-2] [Citation(s) in RCA: 67] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
There is currently no accepted classification of autosomal recessive cerebellar ataxias, a group of disorders characterized by important genetic heterogeneity and complex phenotypes. The objective of this task force was to build a consensus on the classification of autosomal recessive ataxias in order to develop a general approach to a patient presenting with ataxia, organize disorders according to clinical presentation, and define this field of research by identifying common pathogenic molecular mechanisms in these disorders. The work of this task force was based on a previously published systematic scoping review of the literature that identified autosomal recessive disorders characterized primarily by cerebellar motor dysfunction and cerebellar degeneration. The task force regrouped 12 international ataxia experts who decided on general orientation and specific issues. We identified 59 disorders that are classified as primary autosomal recessive cerebellar ataxias. For each of these disorders, we present geographical and ethnical specificities along with distinctive clinical and imagery features. These primary recessive ataxias were organized in a clinical and a pathophysiological classification, and we present a general clinical approach to the patient presenting with ataxia. We also identified a list of 48 complex multisystem disorders that are associated with ataxia and should be included in the differential diagnosis of autosomal recessive ataxias. This classification is the result of a consensus among a panel of international experts, and it promotes a unified understanding of autosomal recessive cerebellar disorders for clinicians and researchers.
Collapse
Affiliation(s)
- Marie Beaudin
- Axe Neurosciences, CHU de Québec-Université Laval, Québec, QC, Canada
- Department of Medicine, Faculty of Medicine, Université Laval, Quebec City, QC, Canada
| | - Antoni Matilla-Dueñas
- Department of Neuroscience, Health Sciences Research Institute Germans Trias i Pujol (IGTP), Universitat Autònoma de Barcelona, Badalona, Barcelona, Spain
| | - Bing-Weng Soong
- Department of Neurology, Shuang Ho Hospital and Taipei Neuroscience Institute, Taipei Medical University, Taipei, Taiwan, Republic of China
- National Yang-Ming University School of Medicine, Taipei Veterans General Hospital, Taipei, Taiwan, Republic of China
| | - Jose Luiz Pedroso
- Ataxia Unit, Department of Neurology, Universidade Federal de São Paulo, São Paulo, SP, Brazil
| | - Orlando G Barsottini
- Ataxia Unit, Department of Neurology, Universidade Federal de São Paulo, São Paulo, SP, Brazil
| | - Hiroshi Mitoma
- Medical Education Promotion Center, Tokyo Medical University, Tokyo, Japan
| | - Shoji Tsuji
- The University of Tokyo, Tokyo, Japan
- International University of Health and Welfare, Chiba, Japan
| | - Jeremy D Schmahmann
- Department of Neurology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Mario Manto
- Service de Neurologie, Médiathèque Jean Jacquy, CHU-Charleroi, 6000, Charleroi, Belgium
- Service des Neurosciences, UMons, Mons, Belgium
| | | | | | - Nicolas Dupre
- Axe Neurosciences, CHU de Québec-Université Laval, Québec, QC, Canada.
- Department of Medicine, Faculty of Medicine, Université Laval, Quebec City, QC, Canada.
| |
Collapse
|
25
|
Tobler M, Caslavska J, Burda P, Thormann W. High-resolution capillary zone electrophoresis for transferrin glycoform analysis associated with congenital disorders of glycosylation. J Sep Sci 2018; 41:2808-2818. [PMID: 29701302 DOI: 10.1002/jssc.201800082] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2018] [Revised: 04/18/2018] [Accepted: 04/19/2018] [Indexed: 11/10/2022]
Abstract
High-resolution capillary zone electrophoresis is used to assess the transferrin profile in serum of patients with eight different congenital disorders of glycosylation that represent type I, type II, and mixed type I/II disorders. Capillary zone electrophoresis data are compared to patterns obtained by gel isoelectric focusing. The high-resolution capillary zone electrophoresis method is shown to represent an effective tool to assess the diversity of transferrin patterns. Hypoglycosylated disialo-, monosialo-, and asialo-transferrin in type I cases can be distinguished from the corresponding underdesialylated transferrin glycoforms present in type II disorders. The latter can be separated from and detected ahead of their corresponding hypoglycosylated forms of type I patients. Both types of glycoforms are detected in sera of mixed type I/II patients. The assay has the potential to be used as screening method for congenital disorders of glycosylation. It can be run with a few microliters of serum when microvials are used.
Collapse
Affiliation(s)
- Micha Tobler
- Division of Metabolism, University Children's Hospital Zürich, Zürich, Switzerland
| | - Jitka Caslavska
- Clinical Pharmacology Laboratory, Institute for Infectious Diseases, University of Bern, Bern, Switzerland
| | - Patricie Burda
- Division of Metabolism, University Children's Hospital Zürich, Zürich, Switzerland
| | - Wolfgang Thormann
- Clinical Pharmacology Laboratory, Institute for Infectious Diseases, University of Bern, Bern, Switzerland
| |
Collapse
|
26
|
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: 68] [Impact Index Per Article: 11.3] [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
|
27
|
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: 1.8] [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
|
28
|
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: 2.9] [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
|
29
|
Marques-da-Silva D, Dos Reis Ferreira V, Monticelli M, Janeiro P, Videira PA, Witters P, Jaeken J, Cassiman D. Liver involvement in congenital disorders of glycosylation (CDG). A systematic review of the literature. J Inherit Metab Dis 2017; 40:195-207. [PMID: 28108845 DOI: 10.1007/s10545-016-0012-4] [Citation(s) in RCA: 70] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/10/2016] [Revised: 12/20/2016] [Accepted: 12/22/2016] [Indexed: 12/15/2022]
Abstract
Congenital disorders of glycosylation (CDG) are a rapidly growing family of genetic diseases caused by defects in glycosylation. Nearly 100 CDG types are known so far. Patients present a great phenotypic diversity ranging from poly- to mono-organ/system involvement and from very mild to extremely severe presentation. In this literature review, we summarize the liver involvement reported in CDG patients. Although liver involvement is present in only a minority of the reported CDG types (22 %), it can be debilitating or even life-threatening. Sixteen of the patients we collated here developed cirrhosis, 10 had liver failure. We distinguish two main groups: on the one hand, the CDG types with predominant or isolated liver involvement including MPI-CDG, TMEM199-CDG, CCDC115-CDG, and ATP6AP1-CDG, and on the other hand, the CDG types associated with liver disease but not as a striking, unique or predominant feature, including PMM2-CDG, ALG1-CDG, ALG3-CDG, ALG6-CDG, ALG8-CDG, ALG9-CDG, PGM1-CDG, and COG-CDG. This review aims to facilitate CDG patient identification and to understand CDG liver involvement, hopefully leading to earlier diagnosis, and better management and treatment.
Collapse
Affiliation(s)
- D 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
- CDG & Allies - Professionals and Patient Associations International Network (CDG & Allies - PPAIN), Caparica, Portugal
| | - V Dos Reis Ferreira
- Portuguese Association for CDG, Lisboa, Portugal
- CDG & Allies - Professionals and Patient Associations International Network (CDG & Allies - PPAIN), Caparica, Portugal
| | - M Monticelli
- UCIBIO, Departamento Ciências da Vida, Faculdade de Ciências e Tecnologia, Universidade NOVA de Lisboa, Lisboa, Portugal
- Dipartimento di Biologia, Università degli Studi di Napoli "Federico II", Napoli, Italy
| | - P Janeiro
- Departamento de Pediatria, Unidade de Doenças Metabólicas, CHLN, Hospital de Sta. Maria, Lisboa, Portugal
| | - P A Videira
- UCIBIO, Departamento Ciências da Vida, Faculdade de Ciências e Tecnologia, Universidade NOVA de Lisboa, Lisboa, Portugal
- Portuguese Association for CDG, Lisboa, Portugal
- CDG & Allies - Professionals and Patient Associations International Network (CDG & Allies - PPAIN), Caparica, Portugal
| | - P Witters
- CDG & Allies - Professionals and Patient Associations International Network (CDG & Allies - PPAIN), Caparica, Portugal
- Center for Metabolic Diseases, UZ and KU Leuven, Leuven, Belgium
| | - J Jaeken
- CDG & Allies - Professionals and Patient Associations International Network (CDG & Allies - PPAIN), Caparica, Portugal.
- Center for Metabolic Diseases, UZ and KU Leuven, Leuven, Belgium.
| | - D Cassiman
- CDG & Allies - Professionals and Patient Associations International Network (CDG & Allies - PPAIN), Caparica, Portugal.
- Center for Metabolic Diseases, UZ and KU Leuven, Leuven, Belgium.
| |
Collapse
|