1
|
Raynor A, Lebredonchel É, Foulquier F, Fenaille F, Bruneel A. Diagnostic and Therapeutic Approaches in Congenital Disorders of Glycosylation. Handb Exp Pharmacol 2025. [PMID: 40119203 DOI: 10.1007/164_2025_745] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/24/2025]
Abstract
Congenital disorders of glycosylation (CDG) constitute an increasing group of inborn metabolic disorders, with more than 170 described diseases to date. A disturbed glycosylation process characterizes them, with molecular defects localized in distinct cell compartments. In CDG, N-glycosylation, O-glycosylation, glycosylation of lipids (including phosphatidylinositol) as well as the glycosaminoglycan synthesis can be affected. Owing to the importance of glycosylation for the function of concerned proteins and lipids, glycosylation defects have diverse clinical consequences. CDG affected individuals often present with a non-specific multivisceral syndrome including neurological involvement, intellectual disability, dysmorphia, and hepatopathy. As CDG are rare diseases frequently lacking distinctive symptoms, biochemical and genetic testing bear important and complementary diagnostic roles.After an introduction on glycosylation and CDG, we review current biomarkers and analytical techniques in the field. Furthermore, we illustrate their interests in the follow-up of proven therapeutic approaches including D-mannose in MPI-CDG, D-galactose in PGM1-CDG, and manganese (MnSO4) in TMEM165-CDG.
Collapse
Affiliation(s)
- Alexandre Raynor
- AP-HP, Biochimie Métabolique et Cellulaire, Hôpital Bichat, Paris, France
| | | | - François Foulquier
- Université de Lille, CNRS, UMR 8576 - UGSF - Unité de Glycobiologie Structurale et Fonctionnelle, Lille, France
| | - François Fenaille
- Université Paris-Saclay, CEA, INRAE, Département Médicaments et Technologies pour la Santé (DMTS), MetaboHUB, Gif sur Yvette, France
| | - Arnaud Bruneel
- AP-HP, Biochimie Métabolique et Cellulaire, Hôpital Bichat, Paris, France.
- INSERM UMR1193, Faculté de Pharmacie, Université Paris-Saclay, Orsay, France.
| |
Collapse
|
2
|
Jankauskas SS, Varzideh F, Kansakar U, Al Tibi G, Densu Agyapong E, Gambardella J, Santulli G. Insights into molecular and cellular functions of the Golgi calcium/manganese-proton antiporter TMEM165. J Biol Chem 2024; 300:107567. [PMID: 39002685 PMCID: PMC11345563 DOI: 10.1016/j.jbc.2024.107567] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Revised: 06/19/2024] [Accepted: 07/08/2024] [Indexed: 07/15/2024] Open
Abstract
The Golgi compartment performs a number of crucial roles in the cell. However, the exact molecular mechanisms underlying these actions are not fully defined. Pathogenic mutations in genes encoding Golgi proteins may serve as an important source for expanding our knowledge. For instance, mutations in the gene encoding Transmembrane protein 165 (TMEM165) were discovered as a cause of a new type of congenital disorder of glycosylation (CDG). Comprehensive studies of TMEM165 in different model systems, including mammals, yeast, and fish uncovered the new realm of Mn2+ homeostasis regulation. TMEM165 was shown to act as a Ca2+/Mn2+:H+ antiporter in the medial- and trans-Golgi network, pumping the metal ions into the Golgi lumen and protons outside. Disruption of TMEM165 antiporter activity results in defects in N- and O-glycosylation of proteins and glycosylation of lipids. Impaired glycosylation of TMEM165-CDG arises from a lack of Mn2+ within the Golgi. Nevertheless, Mn2+ insufficiency in the Golgi is compensated by the activity of the ATPase SERCA2. TMEM165 turnover has also been found to be regulated by Mn2+ cytosolic concentration. Besides causing CDG, recent investigations have demonstrated the functional involvement of TMEM165 in several other pathologies including cancer and mental health disorders. This systematic review summarizes the available information on TMEM165 molecular structure, cellular function, and its roles in health and disease.
Collapse
Affiliation(s)
- Stanislovas S Jankauskas
- Department of Medicine, Wilf Family Cardiovascular Research Institute, Einstein-Mount Sinai Diabetes Research Center (ES-DRC), Albert Einstein College of Medicine, New York City, New York, USA
| | - Fahimeh Varzideh
- Department of Medicine, Wilf Family Cardiovascular Research Institute, Einstein-Mount Sinai Diabetes Research Center (ES-DRC), Albert Einstein College of Medicine, New York City, New York, USA
| | - Urna Kansakar
- Department of Medicine, Wilf Family Cardiovascular Research Institute, Einstein-Mount Sinai Diabetes Research Center (ES-DRC), Albert Einstein College of Medicine, New York City, New York, USA
| | - Ghaith Al Tibi
- Department of Medicine, Wilf Family Cardiovascular Research Institute, Einstein-Mount Sinai Diabetes Research Center (ES-DRC), Albert Einstein College of Medicine, New York City, New York, USA
| | - Esther Densu Agyapong
- Department of Medicine, Wilf Family Cardiovascular Research Institute, Einstein-Mount Sinai Diabetes Research Center (ES-DRC), Albert Einstein College of Medicine, New York City, New York, USA
| | - Jessica Gambardella
- Department of Medicine, Wilf Family Cardiovascular Research Institute, Einstein-Mount Sinai Diabetes Research Center (ES-DRC), Albert Einstein College of Medicine, New York City, New York, USA; Department of Advanced Biomedical Sciences, "Federico II" University, Naples, Italy
| | - Gaetano Santulli
- Department of Medicine, Wilf Family Cardiovascular Research Institute, Einstein-Mount Sinai Diabetes Research Center (ES-DRC), Albert Einstein College of Medicine, New York City, New York, USA; Department of Advanced Biomedical Sciences, "Federico II" University, Naples, Italy; International Translational Research and Medical Education (ITME) Consortium, Academic Research Unit, Naples, Italy; Department of Molecular Pharmacology, Einstein Institute for Aging Research, Fleischer Institute for Diabetes and Metabolism (FIDAM), Albert Einstein College of Medicine, New York City, New York, USA.
| |
Collapse
|
3
|
Durin Z, Raynor A, Fenaille F, Cholet S, Vuillaumier-Barrot S, Alili JM, Poupon J, Oussedik ND, Tuchmann-Durand C, Attali J, Touzé R, Dupré T, Lebredonchel E, Akaffou MA, Legrand D, de Lonlay P, Bruneel A, Foulquier F. Efficacy of oral manganese and D-galactose therapy in a patient bearing a novel TMEM165 variant. Transl Res 2024; 266:57-67. [PMID: 38013006 DOI: 10.1016/j.trsl.2023.11.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/17/2023] [Revised: 11/06/2023] [Accepted: 11/17/2023] [Indexed: 11/29/2023]
Abstract
TMEM165-CDG has first been reported in 2012 and manganese supplementation was shown highly efficient in rescuing glycosylation in isogenic KO cells. The unreported homozygous missense c.928G>C; p.Ala310Pro variant leading to a functional but unstable protein was identified. This patient was diagnosed at 2 months and displays a predominant bone phenotype and combined defects in N-, O- and GAG glycosylation. We administered for the first time a combined D-Gal and Mn2+ therapy to the patient. This fully suppressed the N-; O- and GAG hypoglycosylation. There was also striking improvement in biochemical parameters and in gastrointestinal symptoms. This study offers exciting therapeutic perspectives for TMEM165-CDG.
Collapse
Affiliation(s)
- Zoé Durin
- Univ. Lille, CNRS, UMR 8576 - UGSF - Unité de Glycobiologie Structurale et Fonctionnelle, F-59000 Lille, France
| | - Alexandre Raynor
- AP-HP, Biochimie Métabolique et Cellulaire, Hôpital Bichat-Claude Bernard, 46 rue Henri Huchard, 75018 Paris, France
| | - François Fenaille
- Université Paris-Saclay, CEA, INRAE, Département Médicaments et Technologies pour la Santé (DMTS), MetaboHUB, 91191 Gif sur Yvette, France
| | - Sophie Cholet
- Université Paris-Saclay, CEA, INRAE, Département Médicaments et Technologies pour la Santé (DMTS), MetaboHUB, 91191 Gif sur Yvette, France
| | - Sandrine Vuillaumier-Barrot
- AP-HP, Biochimie Métabolique et Cellulaire, Hôpital Bichat-Claude Bernard, 46 rue Henri Huchard, 75018 Paris, France; Laboratoire de biologie médicale multisites Seqoia - FMG2025, 75014 Paris, France
| | - Jean-Meidi Alili
- Filière G2m, Hôpital Universitaire Necker-Enfants-Malades, Assistance Publique-Hôpitaux de Paris (AP-HP), 75015 Paris, France
| | - Joël Poupon
- Laboratoire de Toxicologie biologique Groupe hospitalier Saint Louis - Lariboisière - Fernand Widal, 75475, Paris, France
| | - Nouzha Djebrani Oussedik
- Laboratoire de Toxicologie biologique Groupe hospitalier Saint Louis - Lariboisière - Fernand Widal, 75475, Paris, France
| | - Caroline Tuchmann-Durand
- Institut Imagine, Biothérapie, Hôpital Universitaire Necker-Enfants-Malades, Assistance Publique-Hôpitaux de Paris (AP-HP), Institut Imagine, 75015 Paris, France
| | - Jennifer Attali
- Service de Radiologie Pédiatrique, Hôpital Universitaire Necker-Enfants-Malades, Assistance Publique-Hôpitaux de Paris (AP-HP), Institut Imagine,75015 Paris, France
| | - Romain Touzé
- Service d'Ophtalmologie Pédiatrique, Hôpital Universitaire Necker-Enfants-Malades, Assistance Publique-Hôpitaux de Paris (AP-HP), 75015 Paris, France
| | - Thierry Dupré
- AP-HP, Biochimie Métabolique et Cellulaire, Hôpital Bichat-Claude Bernard, 46 rue Henri Huchard, 75018 Paris, France
| | - Elodie Lebredonchel
- AP-HP, Biochimie Métabolique et Cellulaire, Hôpital Bichat-Claude Bernard, 46 rue Henri Huchard, 75018 Paris, France
| | - Marlyse Angah Akaffou
- AP-HP, Biochimie Métabolique et Cellulaire, Hôpital Bichat-Claude Bernard, 46 rue Henri Huchard, 75018 Paris, France
| | - Dominique Legrand
- Univ. Lille, CNRS, UMR 8576 - UGSF - Unité de Glycobiologie Structurale et Fonctionnelle, F-59000 Lille, France
| | - Pascale de Lonlay
- Filière G2m, Hôpital Universitaire Necker-Enfants-Malades, Assistance Publique-Hôpitaux de Paris (AP-HP), 75015 Paris, France; Hôpital Universitaire Necker-Enfants-Malades, Institut Imagine, G2M, MetabERN, Université Paris Cité, 75015 Paris, France.
| | - Arnaud Bruneel
- AP-HP, Biochimie Métabolique et Cellulaire, Hôpital Bichat-Claude Bernard, 46 rue Henri Huchard, 75018 Paris, France; INSERM UMR1193, Faculté de Pharmacie, Université Paris-Saclay, bâtiment Henri Moissan, 92400 Orsay, France.
| | - François Foulquier
- Univ. Lille, CNRS, UMR 8576 - UGSF - Unité de Glycobiologie Structurale et Fonctionnelle, F-59000 Lille, France.
| |
Collapse
|
4
|
Zajac M, Mukherjee S, Anees P, Oettinger D, Henn K, Srikumar J, Zou J, Saminathan A, Krishnan Y. A mechanism of lysosomal calcium entry. SCIENCE ADVANCES 2024; 10:eadk2317. [PMID: 38354239 PMCID: PMC10866540 DOI: 10.1126/sciadv.adk2317] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/09/2023] [Accepted: 01/16/2024] [Indexed: 02/16/2024]
Abstract
Lysosomal calcium (Ca2+) release is critical to cell signaling and is mediated by well-known lysosomal Ca2+ channels. Yet, how lysosomes refill their Ca2+ remains hitherto undescribed. Here, from an RNA interference screen in Caenorhabditis elegans, we identify an evolutionarily conserved gene, lci-1, that facilitates lysosomal Ca2+ entry in C. elegans and mammalian cells. We found that its human homolog TMEM165, previously designated as a Ca2+/H+ exchanger, imports Ca2+ pH dependently into lysosomes. Using two-ion mapping and electrophysiology, we show that TMEM165, hereafter referred to as human LCI, acts as a proton-activated, lysosomal Ca2+ importer. Defects in lysosomal Ca2+ channels cause several neurodegenerative diseases, and knowledge of lysosomal Ca2+ importers may provide previously unidentified avenues to explore the physiology of Ca2+ channels.
Collapse
Affiliation(s)
- Matthew Zajac
- Department of Chemistry, The University of Chicago, Chicago, IL 60637, USA
- Neuroscience Institute, The University of Chicago, Chicago, IL 60637, USA
| | - Sourajit Mukherjee
- Department of Chemistry, The University of Chicago, Chicago, IL 60637, USA
- Neuroscience Institute, The University of Chicago, Chicago, IL 60637, USA
| | - Palapuravan Anees
- Department of Chemistry, The University of Chicago, Chicago, IL 60637, USA
- Neuroscience Institute, The University of Chicago, Chicago, IL 60637, USA
| | - Daphne Oettinger
- Department of Chemistry, The University of Chicago, Chicago, IL 60637, USA
- Neuroscience Institute, The University of Chicago, Chicago, IL 60637, USA
| | - Katharine Henn
- Department of Chemistry, The University of Chicago, Chicago, IL 60637, USA
- Neuroscience Institute, The University of Chicago, Chicago, IL 60637, USA
| | - Jainaha Srikumar
- Department of Chemistry, The University of Chicago, Chicago, IL 60637, USA
- Neuroscience Institute, The University of Chicago, Chicago, IL 60637, USA
| | - Junyi Zou
- Department of Chemistry, The University of Chicago, Chicago, IL 60637, USA
- Neuroscience Institute, The University of Chicago, Chicago, IL 60637, USA
| | - Anand Saminathan
- Department of Chemistry, The University of Chicago, Chicago, IL 60637, USA
- Neuroscience Institute, The University of Chicago, Chicago, IL 60637, USA
| | - Yamuna Krishnan
- Department of Chemistry, The University of Chicago, Chicago, IL 60637, USA
- Neuroscience Institute, The University of Chicago, Chicago, IL 60637, USA
- Institute for Biophysical Dynamics, The University of Chicago, Chicago, IL 60637, USA
| |
Collapse
|
5
|
Legrand D, Herbaut M, Durin Z, Brysbaert G, Bardor M, Lensink MF, Foulquier F. New insights into the pathogenicity of TMEM165 variants using structural modeling based on AlphaFold 2 predictions. Comput Struct Biotechnol J 2023; 21:3424-3436. [PMID: 37416081 PMCID: PMC10319644 DOI: 10.1016/j.csbj.2023.06.015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2023] [Revised: 06/15/2023] [Accepted: 06/15/2023] [Indexed: 07/08/2023] Open
Abstract
TMEM165 is a Golgi protein playing a crucial role in Mn2+ transport, and whose mutations in patients are known to cause Congenital Disorders of Glycosylation. Some of those mutations affect the highly-conserved consensus motifs E-φ-G-D-[KR]-[TS] characterizing the CaCA2/UPF0016 family, presumably important for the transport of Mn2+ which is essential for the function of many Golgi glycosylation enzymes. Others, like the G>R304 mutation, are far away from these motifs in the sequence. Until recently, the classical membrane protein topology prediction methods were unable to provide a clear picture of the organization of TMEM165 inside the cell membrane, or to explain in a convincing manner the impact of patient and experimentally-generated mutations on the transporter function of TMEM165. In this study, AlphaFold 2 was used to build a TMEM165 model that was then refined by molecular dynamics simulation with membrane lipids and water. This model provides a realistic picture of the 3D protein scaffold formed from a two-fold repeat of three transmembrane helices/domains where the consensus motifs face each other to form a putative acidic cation-binding site at the cytosolic side of the protein. It sheds new light on the impact of mutations on the transporter function of TMEM165, found in patients and studied experimentally in vitro, formerly and within this study. More particularly and very interestingly, this model explains the impact of the G>R304 mutation on TMEM165's function. These findings provide great confidence in the predicted TMEM165 model whose structural features are discussed in the study and compared to other structural and functional TMEM165 homologs from the CaCA2/UPF0016 family and the LysE superfamily.
Collapse
Affiliation(s)
- Dominique Legrand
- Univ. Lille, CNRS, UMR 8576 – UGSF - Unité de Glycobiologie Structurale et Fonctionnelle, F-59000 Lille, France
| | - Mélissandre Herbaut
- Univ. Lille, CNRS, UMR 8576 – UGSF - Unité de Glycobiologie Structurale et Fonctionnelle, F-59000 Lille, France
| | - Zoé Durin
- Univ. Lille, CNRS, UMR 8576 – UGSF - Unité de Glycobiologie Structurale et Fonctionnelle, F-59000 Lille, France
| | - Guillaume Brysbaert
- Univ. Lille, CNRS, UMR 8576 – UGSF - Unité de Glycobiologie Structurale et Fonctionnelle, F-59000 Lille, France
| | - Muriel Bardor
- Univ. Lille, CNRS, UMR 8576 – UGSF - Unité de Glycobiologie Structurale et Fonctionnelle, F-59000 Lille, France
- Université de Rouen Normandie, Laboratoire GlycoMEV UR 4358, SFR Normandie Végétal FED 4277, Innovation Chimie Carnot, F-76000 Rouen, France
| | - Marc F. Lensink
- Univ. Lille, CNRS, UMR 8576 – UGSF - Unité de Glycobiologie Structurale et Fonctionnelle, F-59000 Lille, France
| | - François Foulquier
- Univ. Lille, CNRS, UMR 8576 – UGSF - Unité de Glycobiologie Structurale et Fonctionnelle, F-59000 Lille, France
| |
Collapse
|
6
|
Durin Z, Houdou M, Morelle W, Barré L, Layotte A, Legrand D, Ouzzine M, Foulquier F. Differential Effects of D-Galactose Supplementation on Golgi Glycosylation Defects in TMEM165 Deficiency. Front Cell Dev Biol 2022; 10:903953. [PMID: 35693943 PMCID: PMC9178294 DOI: 10.3389/fcell.2022.903953] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2022] [Accepted: 05/04/2022] [Indexed: 11/13/2022] Open
Abstract
Glycosylation is a ubiquitous and universal cellular process in all domains of life. In eukaryotes, many glycosylation pathways occur simultaneously onto proteins and lipids for generating a complex diversity of glycan structures. In humans, severe genetic diseases called Congenital Disorders of Glycosylation (CDG), resulting from glycosylation defects, demonstrate the functional relevance of these processes. No real cure exists so far, but oral administration of specific monosaccharides to bypass the metabolic defects has been used in few CDG, then constituting the simplest and safest treatments. Oral D-Galactose (Gal) therapy was seen as a promising tailored treatment for specific CDG and peculiarly for TMEM165-CDG patients. TMEM165 deficiency not only affects the N-glycosylation process but all the other Golgi-related glycosylation types, then contributing to the singularity of this defect. Our previous results established a link between TMEM165 deficiency and altered Golgi manganese (Mn2+) homeostasis. Besides the fascinating power of MnCl2 supplementation to rescue N-glycosylation in TMEM165-deficient cells, D-Gal supplementation has also been shown to be promising in suppressing the observed N-glycosylation defects. Its effect on the other Golgi glycosylation types, most especially O-glycosylation and glycosaminoglycan (GAG) synthesis, was however unknown. In the present study, we demonstrate the differential impact of D-Gal or MnCl2 supplementation effects on the Golgi glycosylation defects caused by TMEM165 deficiency. Whereas MnCl2 supplementation unambiguously fully rescues the N- and O-linked as well as GAG glycosylations in TMEM165-deficient cells, D-Gal supplementation only rescues the N-linked glycosylation, without any effects on the other Golgi-related glycosylation types. According to these results, we would recommend the use of MnCl2 for TMEM165-CDG therapy.
Collapse
Affiliation(s)
- Zoé Durin
- University of Lille, CNRS, UMR 8576—UGSF—Unité de Glycobiologie Structurale et Fonctionnelle, Lille, France
| | - Marine Houdou
- University of Lille, CNRS, UMR 8576—UGSF—Unité de Glycobiologie Structurale et Fonctionnelle, Lille, France
| | - Willy Morelle
- University of Lille, CNRS, UMR 8576—UGSF—Unité de Glycobiologie Structurale et Fonctionnelle, Lille, France
| | - Lydia Barré
- Faculty of Medicine, UMR7365 CNRS-University of Lorraine, Biopôle, Nancy, France
| | - Aurore Layotte
- University of Lille, CNRS, UMR 8576—UGSF—Unité de Glycobiologie Structurale et Fonctionnelle, Lille, France
| | - Dominique Legrand
- University of Lille, CNRS, UMR 8576—UGSF—Unité de Glycobiologie Structurale et Fonctionnelle, Lille, France
| | - Mohamed Ouzzine
- Faculty of Medicine, UMR7365 CNRS-University of Lorraine, Biopôle, Nancy, France
| | - François Foulquier
- University of Lille, CNRS, UMR 8576—UGSF—Unité de Glycobiologie Structurale et Fonctionnelle, Lille, France
- *Correspondence: François Foulquier,
| |
Collapse
|
7
|
Hellicar J, Stevenson NL, Stephens DJ, Lowe M. Supply chain logistics - the role of the Golgi complex in extracellular matrix production and maintenance. J Cell Sci 2022; 135:273996. [PMID: 35023559 PMCID: PMC8767278 DOI: 10.1242/jcs.258879] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
The biomechanical and biochemical properties of connective tissues are determined by the composition and quality of their extracellular matrix. This, in turn, is highly dependent on the function and organisation of the secretory pathway. The Golgi complex plays a vital role in directing matrix output by co-ordinating the post-translational modification and proteolytic processing of matrix components prior to their secretion. These modifications have broad impacts on the secretion and subsequent assembly of matrix components, as well as their function in the extracellular environment. In this Review, we highlight the role of the Golgi in the formation of an adaptable, healthy matrix, with a focus on proteoglycan and procollagen secretion as example cargoes. We then discuss the impact of Golgi dysfunction on connective tissue in the context of human disease and ageing.
Collapse
Affiliation(s)
- John Hellicar
- School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, The Michael Smith Building, Oxford Road, Manchester, M13 9PT, UK.,Institute of Molecular and Cell Biology (IMCB), Agency for Science, Technology and Research (A*STAR), 61 Biopolis Drive, Proteos, Singapore, 138673
| | - Nicola L Stevenson
- Cell Biology Laboratories, School of Biochemistry, Faculty of Life Sciences, University Walk, University of Bristol, Bristol, BS8 1TD, UK
| | - David J Stephens
- Cell Biology Laboratories, School of Biochemistry, Faculty of Life Sciences, University Walk, University of Bristol, Bristol, BS8 1TD, UK
| | - Martin Lowe
- School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, The Michael Smith Building, Oxford Road, Manchester, M13 9PT, UK
| |
Collapse
|
8
|
Lipiński P, Tylki-Szymańska A. Congenital Disorders of Glycosylation: What Clinicians Need to Know? Front Pediatr 2021; 9:715151. [PMID: 34540767 PMCID: PMC8446601 DOI: 10.3389/fped.2021.715151] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/26/2021] [Accepted: 08/10/2021] [Indexed: 12/27/2022] Open
Abstract
Congenital disorders of glycosylation (CDG) are a group of clinically heterogeneous disorders characterized by defects in the synthesis of glycans and their attachment to proteins and lipids. This manuscript aims to provide a classification of the clinical presentation, diagnostic methods, and treatment of CDG based on the literature review and our own experience (referral center in Poland). A diagnostic algorithm for CDG was also proposed. Isoelectric focusing (IEF) of serum transferrin (Tf) is still the method of choice for diagnosing N-glycosylation disorders associated with sialic acid deficiency. Nowadays, high-performance liquid chromatography, capillary zone electrophoresis, and mass spectrometry techniques are used, although they are not routinely available. Since next-generation sequencing became more widely available, an improvement in diagnostics has been observed, with more patients and novel CDG subtypes being reported. Early and accurate diagnosis of CDG is crucial for timely implementation of appropriate therapies and improving clinical outcomes. However, causative treatment is available only for few CDG types.
Collapse
Affiliation(s)
- Patryk Lipiński
- Department of Pediatrics, Nutrition and Metabolic Diseases, The Children's Memorial Health Institute, Warsaw, Poland
| | | |
Collapse
|
9
|
Lipiński P, Stępień KM, Ciara E, Tylki-Szymańska A, Jezela-Stanek A. Skeletal and Bone Mineral Density Features, Genetic Profile in Congenital Disorders of Glycosylation: Review. Diagnostics (Basel) 2021; 11:diagnostics11081438. [PMID: 34441372 PMCID: PMC8391432 DOI: 10.3390/diagnostics11081438] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2021] [Revised: 08/04/2021] [Accepted: 08/04/2021] [Indexed: 11/25/2022] Open
Abstract
Congenital disorders of glycosylation (CDGs) are a heterogeneous group of disorders with impaired glycosylation of proteins and lipids. These conditions have multisystemic clinical manifestations, resulting in gradually progressive complications including skeletal involvement and reduced bone mineral density. Contrary to PMM2-CDG, all remaining CDG, including ALG12-CDG, ALG3-CDG, ALG9-CDG, ALG6-CDG, PGM3-CDG, CSGALNACT1-CDG, SLC35D1-CDG and TMEM-165, are characterized by well-defined skeletal dysplasia. In some of them, prenatal-onset severe skeletal dysplasia is observed associated with early death. Osteoporosis or osteopenia are frequently observed in all CDG types and are more pronounced in adults. Hormonal dysfunction, limited mobility and inadequate diet are common risk factors for reduced bone mineral density. Skeletal involvement in CDGs is underestimated and, thus, should always be carefully investigated and managed to prevent fractures and chronic pain. With the advent of new therapeutic developments for CDGs, the severity of skeletal complications may be reduced. This review focuses on possible mechanisms of skeletal manifestations, risk factors for osteoporosis, and bone markers in reported paediatric and adult CDG patients.
Collapse
Affiliation(s)
- Patryk Lipiński
- Department of Pediatrics, Nutrition and Metabolic Diseases, The Children’s Memorial Health Institute, 04-730 Warsaw, Poland;
- Correspondence:
| | - Karolina M. Stępień
- Adult Inherited Metabolic Diseases, Salford Royal NHS Foundation Trust, Salford M6 8HD, UK;
| | - Elżbieta Ciara
- Department of Medical Genetics, The Children’s Memorial Health Institute, 04-730 Warsaw, Poland;
| | - Anna Tylki-Szymańska
- Department of Pediatrics, Nutrition and Metabolic Diseases, The Children’s Memorial Health Institute, 04-730 Warsaw, Poland;
| | - Aleksandra Jezela-Stanek
- Department of Genetics and Clinical Immunology, National Institute of Tuberculosis and Lung Diseases, 01-138 Warsaw, Poland;
| |
Collapse
|
10
|
Dubail J, Cormier-Daire V. Chondrodysplasias With Multiple Dislocations Caused by Defects in Glycosaminoglycan Synthesis. Front Genet 2021; 12:642097. [PMID: 34220933 PMCID: PMC8242584 DOI: 10.3389/fgene.2021.642097] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2020] [Accepted: 05/04/2021] [Indexed: 11/13/2022] Open
Abstract
Chondrodysplasias with multiple dislocations form a group of severe disorders characterized by joint laxity and multiple dislocations, severe short stature of pre- and post-natal onset, hand anomalies, and/or vertebral anomalies. The majority of chondrodysplasias with multiple dislocations have been associated with mutations in genes encoding glycosyltransferases, sulfotransferases, and transporters implicated in the synthesis or sulfation of glycosaminoglycans, long and unbranched polysaccharides composed of repeated disaccharide bond to protein core of proteoglycan. Glycosaminoglycan biosynthesis is a tightly regulated process that occurs mainly in the Golgi and that requires the coordinated action of numerous enzymes and transporters as well as an adequate Golgi environment. Any disturbances of this chain of reactions will lead to the incapacity of a cell to construct correct glycanic chains. This review focuses on genetic and glycobiological studies of chondrodysplasias with multiple dislocations associated with glycosaminoglycan biosynthesis defects and related animal models. Strong comprehension of the molecular mechanisms leading to those disorders, mostly through extensive phenotypic analyses of in vitro and/or in vivo models, is essential for the development of novel biomarkers for clinical screenings and innovative therapeutics for these diseases.
Collapse
Affiliation(s)
- Johanne Dubail
- Université de Paris, INSERM UMR 1163, Institut Imagine, Paris, France
| | - Valérie Cormier-Daire
- Université de Paris, INSERM UMR 1163, Institut Imagine, Paris, France.,Service de Génétique Clinique, Centre de Référence Pour Les Maladies Osseuses Constitutionnelles, AP-HP, Hôpital Necker-Enfants Malades, Paris, France
| |
Collapse
|
11
|
Wada Y, Okamoto N. Apolipoprotein C-III O-glycoform profiling of 500 serum samples by matrix-assisted laser desorption/ionization mass spectrometry for diagnosis of congenital disorders of glycosylation. JOURNAL OF MASS SPECTROMETRY : JMS 2021; 56:e4597. [PMID: 32677746 DOI: 10.1002/jms.4597] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/08/2020] [Revised: 06/06/2020] [Accepted: 06/16/2020] [Indexed: 06/11/2023]
Abstract
Congenital disorders of glycosylation (CDG) are caused by defects in various genes governing glycoconjugate biosynthesis. Several responsible genes have been identified in the protein N-glycosylation process. Analyses of mucin-type core-1 O-glycoform of apolipoprotein C-III (apoCIII) have recently revealed combined N- and O-glycosylation defects. We applied matrix-assisted laser desorption/ionization mass spectrometry profiling of apoCIII glycoforms to 500 serum samples for CDG screening, and reference values were determined. The content of unglycosylated apoCIII was low in early infancy, indicating that the O-glycan occupancy should be assessed based on age-matched reference values. The samples from patients with mutations in the ALG1, ATP6V0A2, B4GALT1, COG2, GCS1, PGM1, SLC35A2, and TRAPPC11 genes were analyzed. B4GALT1- and TRAPPC11-CDG were accompanied by under-sialylation of O-glycans and are now recognized as combined N- and O-glycosylation disorders.
Collapse
Affiliation(s)
- Yoshinao Wada
- Department of Molecular Medicine, Osaka Women's and Children's Hospital (OWCH), Osaka, Japan
| | - Nobuhiko Okamoto
- Department of Molecular Medicine, Osaka Women's and Children's Hospital (OWCH), Osaka, Japan
| |
Collapse
|
12
|
Hoecker N, Hennecke Y, Schrott S, Marino G, Schmidt SB, Leister D, Schneider A. Gene Replacement in Arabidopsis Reveals Manganese Transport as an Ancient Feature of Human, Plant and Cyanobacterial UPF0016 Proteins. FRONTIERS IN PLANT SCIENCE 2021; 12:697848. [PMID: 34194462 PMCID: PMC8236900 DOI: 10.3389/fpls.2021.697848] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/20/2021] [Accepted: 05/20/2021] [Indexed: 05/08/2023]
Abstract
The protein family 0016 (UPF0016) is conserved through evolution, and the few members characterized share a function in Mn2+ transport. So far, little is known about the history of these proteins in Eukaryotes. In Arabidopsis thaliana five such proteins, comprising four different subcellular localizations including chloroplasts, have been described, whereas non-photosynthetic Eukaryotes have only one. We used a phylogenetic approach to classify the eukaryotic proteins into two subgroups and performed gene-replacement studies to investigate UPF0016 genes of various origins. Replaceability can be scored readily in the Arabidopsis UPF0016 transporter mutant pam71, which exhibits a functional deficiency in photosystem II. The N-terminal region of the Arabidopsis PAM71 was used to direct selected proteins to chloroplast membranes. Transgenic pam71 lines overexpressing the closest plant homolog (CMT1), human TMEM165 or cyanobacterial MNX successfully restored photosystem II efficiency, manganese binding to photosystem II complexes and consequently plant growth rate and biomass production. Thus AtCMT1, HsTMEM165, and SynMNX can operate in the thylakoid membrane and substitute for PAM71 in a non-native environment, indicating that the manganese transport function of UPF0016 proteins is an ancient feature of the family. We propose that the two chloroplast-localized UPF0016 proteins, CMT1 and PAM71, in plants originated from the cyanobacterial endosymbiont that gave rise to the organelle.
Collapse
Affiliation(s)
- Natalie Hoecker
- Molekularbiologie der Pflanzen (Botanik), Fakultät für Biologie, Ludwig-Maximilians-Universität München, Martinsried, Germany
| | - Yvonne Hennecke
- Molekularbiologie der Pflanzen (Botanik), Fakultät für Biologie, Ludwig-Maximilians-Universität München, Martinsried, Germany
| | - Simon Schrott
- Molekularbiologie der Pflanzen (Botanik), Fakultät für Biologie, Ludwig-Maximilians-Universität München, Martinsried, Germany
| | - Giada Marino
- Molekularbiologie der Pflanzen (Botanik), Fakultät für Biologie, Ludwig-Maximilians-Universität München, Martinsried, Germany
- Massenspektrometrie von Biomolekülen an der LMU (MSBioLMU), Fakultät für Biologie, Ludwig-Maximilians-Universität München, Martinsried, Germany
| | - Sidsel Birkelund Schmidt
- Department of Plant and Environmental Sciences, Faculty of Science, University of Copenhagen, Frederiksberg, Denmark
| | - Dario Leister
- Molekularbiologie der Pflanzen (Botanik), Fakultät für Biologie, Ludwig-Maximilians-Universität München, Martinsried, Germany
| | - Anja Schneider
- Molekularbiologie der Pflanzen (Botanik), Fakultät für Biologie, Ludwig-Maximilians-Universität München, Martinsried, Germany
- *Correspondence: Anja Schneider,
| |
Collapse
|
13
|
Haouari W, Dubail J, Lounis-Ouaras S, Prada P, Bennani R, Roseau C, Huber C, Afenjar A, Colin E, Vuillaumier-Barrot S, Seta N, Foulquier F, Poüs C, Cormier-Daire V, Bruneel A. Serum bikunin isoforms in congenital disorders of glycosylation and linkeropathies. J Inherit Metab Dis 2020; 43:1349-1359. [PMID: 32700771 DOI: 10.1002/jimd.12291] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/05/2020] [Revised: 07/18/2020] [Accepted: 07/21/2020] [Indexed: 12/20/2022]
Abstract
Bikunin (Bkn) isoforms are serum chondroitin sulfate (CS) proteoglycans synthesized by the liver. They include two light forms, that is, the Bkn core protein and the Bkn linked to the CS chain (urinary trypsin inhibitor [UTI]), and two heavy forms, that is, pro-α-trypsin inhibitor and inter-α-trypsin inhibitor, corresponding to UTI esterified by one or two heavy chains glycoproteins, respectively. We previously showed that the Western-blot analysis of the light forms could allow the fast and easy detection of patients with linkeropathy, deficient in enzymes involved in the synthesis of the initial common tetrasaccharide linker of glycosaminoglycans. Here, we analyzed all serum Bkn isoforms in a context of congenital disorders of glycosylation (CDG) and showed very specific abnormal patterns suggesting potential interests for their screening and diagnosis. In particular, genetic deficiencies in V-ATPase (ATP6V0A2-CDG, CCDC115-CDG, ATP6AP1-CDG), in Golgi manganese homeostasis (TMEM165-CDG) and in the N-acetyl-glucosamine Golgi transport (SLC35A3-CDG) all share specific abnormal Bkn patterns. Furthermore, for each studied linkeropathy, we show that the light abnormal Bkn could be further in-depth characterized by two-dimensional electrophoresis. Moreover, besides being interesting as a specific biomarker of both CDG and linkeropathies, Bkn isoforms' analyses can provide new insights into the pathophysiology of the aforementioned diseases.
Collapse
Affiliation(s)
- Walid Haouari
- INSERM UMR1193, Université Paris-Saclay, Châtenay-Malabry, France
| | - Johanne Dubail
- Department of Clinical Genetics and Reference Centre for Constitutional Bone Diseases, INSERM U1163, Université de Paris, Imagine Institute, Necker-Enfants Malades Hospital, AP-HP, Paris, France
| | - Samra Lounis-Ouaras
- INSERM UMR1193, Université Paris-Saclay, Châtenay-Malabry, France
- AP-HP, Biochimie-Hormonologie, Hôpital Antoine Béclère, Clamart, France
| | - Pierre Prada
- AP-HP, Biochimie Métabolique et Cellulaire, Hôpital Bichat-Claude Bernard, Paris, France
| | - Rizk Bennani
- AP-HP, Biochimie Métabolique et Cellulaire, Hôpital Bichat-Claude Bernard, Paris, France
| | - Charles Roseau
- INSERM UMR1193, Université Paris-Saclay, Châtenay-Malabry, France
| | - Céline Huber
- Department of Clinical Genetics and Reference Centre for Constitutional Bone Diseases, INSERM U1163, Université de Paris, Imagine Institute, Necker-Enfants Malades Hospital, AP-HP, Paris, France
| | - Alexandra Afenjar
- Département de Génétique et Embryologie Médicale, Sorbonne Universités, Centre de Référence Malformations et Maladies Congénitales du Cervelet et Déficiences Intellectuelles de Causes Rares, Hôpital Trousseau, AP-HP, Paris, France
| | - Estelle Colin
- Department of Biochemistry and Genetics, University Hospital, Angers, France
- MitoLab Team, Institut MitoVasc, UMR CNRS6015, INSERM U1083, Angers, France
| | | | - Nathalie Seta
- AP-HP, Biochimie Métabolique et Cellulaire, Hôpital Bichat-Claude Bernard, Paris, France
- Université de Paris, Paris, France
| | - François Foulquier
- Université de Lille, CNRS, UMR 8576 - UGSF - Unité de Glycobiologie Structurale et Fonctionnelle, Lille, France
| | - Christian Poüs
- INSERM UMR1193, Université Paris-Saclay, Châtenay-Malabry, France
- AP-HP, Biochimie-Hormonologie, Hôpital Antoine Béclère, Clamart, France
| | - Valérie Cormier-Daire
- Department of Clinical Genetics and Reference Centre for Constitutional Bone Diseases, INSERM U1163, Université de Paris, Imagine Institute, Necker-Enfants Malades Hospital, AP-HP, Paris, France
| | - Arnaud Bruneel
- INSERM UMR1193, Université Paris-Saclay, Châtenay-Malabry, France
- AP-HP, Biochimie Métabolique et Cellulaire, Hôpital Bichat-Claude Bernard, Paris, France
| |
Collapse
|
14
|
Ondruskova N, Cechova A, Hansikova H, Honzik T, Jaeken J. Congenital disorders of glycosylation: Still "hot" in 2020. Biochim Biophys Acta Gen Subj 2020; 1865:129751. [PMID: 32991969 DOI: 10.1016/j.bbagen.2020.129751] [Citation(s) in RCA: 91] [Impact Index Per Article: 18.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2020] [Revised: 08/12/2020] [Accepted: 08/27/2020] [Indexed: 12/11/2022]
Abstract
BACKGROUND Congenital disorders of glycosylation (CDG) are inherited metabolic diseases caused by defects in the genes important for the process of protein and lipid glycosylation. With the ever growing number of the known subtypes and discoveries regarding the disease mechanisms and therapy development, it remains a very active field of study. SCOPE OF REVIEW This review brings an update on the CDG-related research since 2017, describing the novel gene defects, pathobiomechanisms, biomarkers and the patients' phenotypes. We also summarize the clinical guidelines for the most prevalent disorders and the current therapeutical options for the treatable CDG. MAJOR CONCLUSIONS In the majority of the 23 new CDG, neurological involvement is associated with other organ disease. Increasingly, different aspects of cellular metabolism (e.g., autophagy) are found to be perturbed in multiple CDG. GENERAL SIGNIFICANCE This work highlights the recent trends in the CDG field and comprehensively overviews the up-to-date clinical recommendations.
Collapse
Affiliation(s)
- Nina Ondruskova
- Department of Paediatrics and Inherited Metabolic Disorders, First Faculty of Medicine, Charles University and General University Hospital in Prague, Prague, Czech Republic
| | - Anna Cechova
- Department of Paediatrics and Inherited Metabolic Disorders, First Faculty of Medicine, Charles University and General University Hospital in Prague, Prague, Czech Republic
| | - Hana Hansikova
- Department of Paediatrics and Inherited Metabolic Disorders, First Faculty of Medicine, Charles University and General University Hospital in Prague, Prague, Czech Republic
| | - Tomas Honzik
- Department of Paediatrics and Inherited Metabolic Disorders, First Faculty of Medicine, Charles University and General University Hospital in Prague, Prague, Czech Republic.
| | - Jaak Jaeken
- Department of Paediatrics and Centre for Metabolic Diseases, KU Leuven and University Hospital Leuven, Leuven, Belgium.
| |
Collapse
|
15
|
A Great Catch for Investigating Inborn Errors of Metabolism-Insights Obtained from Zebrafish. Biomolecules 2020; 10:biom10091352. [PMID: 32971894 PMCID: PMC7564250 DOI: 10.3390/biom10091352] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2020] [Revised: 09/18/2020] [Accepted: 09/19/2020] [Indexed: 12/14/2022] Open
Abstract
Inborn errors of metabolism cause abnormal synthesis, recycling, or breakdown of amino acids, neurotransmitters, and other various metabolites. This aberrant homeostasis commonly causes the accumulation of toxic compounds or depletion of vital metabolites, which has detrimental consequences for the patients. Efficient and rapid intervention is often key to survival. Therefore, it requires useful animal models to understand the pathomechanisms and identify promising therapeutic drug targets. Zebrafish are an effective tool to investigate developmental mechanisms and understanding the pathophysiology of disorders. In the past decades, zebrafish have proven their efficiency for studying genetic disorders owing to the high degree of conservation between human and zebrafish genes. Subsequently, several rare inherited metabolic disorders have been successfully investigated in zebrafish revealing underlying mechanisms and identifying novel therapeutic targets, including methylmalonic acidemia, Gaucher’s disease, maple urine disorder, hyperammonemia, TRAPPC11-CDGs, and others. This review summarizes the recent impact zebrafish have made in the field of inborn errors of metabolism.
Collapse
|
16
|
Abstract
La glycosylation est un processus cellulaire complexe conduisant à des transferts successifs de monosaccharides sur une molécule acceptrice, le plus souvent une protéine ou un lipide. Ce processus est universel chez tous les organismes vivants et est très conservé au cours de l’évolution. Chez l’homme, des perturbations survenant au cours d’une ou plusieurs réactions de glycosylation sont à l’origine de glycopathologies génétiques rares, appelées anomalies congénitales de la glycosylation ou congenital disorders of glycosylation (CDG). Cette revue propose de revisiter ces CDG, de 1980 à aujourd’hui, en présentant leurs découvertes, leurs diagnostics, leurs causes biochimiques et les traitements actuellement disponibles.
Collapse
|
17
|
Murali P, Johnson BP, Lu Z, Climer L, Scott DA, Foulquier F, Oprea-Ilies G, Lupashin V, Drake RR, Abbott KL. Novel role for the Golgi membrane protein TMEM165 in control of migration and invasion for breast carcinoma. Oncotarget 2020; 11:2747-2762. [PMID: 32733646 PMCID: PMC7367651 DOI: 10.18632/oncotarget.27668] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2019] [Accepted: 06/20/2020] [Indexed: 12/21/2022] Open
Abstract
The TMEM165 gene encodes for a multiple pass membrane protein localized in the Golgi that has been linked to congenital disorders of glycosylation. The TMEM165 protein is a putative ion transporter that regulates H+/Ca++/Mn++ homeostasis and pH in the Golgi. Previously, we identified TMEM165 as a potential biomarker for breast carcinoma in a glycoproteomic study using late stage invasive ductal carcinoma tissues with patient- matched adjacent normal tissues. The TMEM165 protein was not detected in non-malignant matched breast tissues and was detected in invasive ductal breast carcinoma tissues by mass spectrometry. Our hypothesis is that the TMEM165 protein confers a growth advantage to breast cancer. In this preliminary study we have investigated the expression of TMEM165 in earlier stage invasive ductal carcinoma and ductal carcinoma in situ cases. We created a CRISPR/Cas9 knockout of TMEM165 in the human invasive breast cancer cell line MDAMB231. Our results indicate that removal of TMEM165 in these cells results in a significant reduction of cell migration, tumor growth, and tumor vascularization in vivo. Furthermore, we find that TMEM165 expression alters the glycosylation of breast cancer cells and these changes promote the invasion and growth of breast cancer by altering the expression levels of key glycoproteins involved in regulation of the epithelial to mesenchymal transition such as E-cadherin. These studies illustrate new potential functions for this Golgi membrane protein in the control of breast cancer growth and invasion.
Collapse
Affiliation(s)
- Pavitra Murali
- University of Oklahoma Health Sciences Center, Department of Biochemistry and Molecular Biology, Oklahoma City, OK, United States
| | - Blake P Johnson
- Ouchita Baptist University, Department of Biology, Arkadelphia, AR, United States
| | - Zhongpeng Lu
- University of Oklahoma Health Sciences Center, Department of Biochemistry and Molecular Biology, Oklahoma City, OK, United States
| | - Leslie Climer
- University of Arkansas for Medical Sciences, Department of Biophysics and Physiology, Little Rock, AR, United States
| | - Danielle A Scott
- Department of Cell and Molecular Pharmacology, Medical University of South Carolina, Charleston, SC, United States
| | - Francois Foulquier
- Centre National de la Recherche Scientifique, Unité Mixte de Recherche, Unité de Glycobiologie Structurale et Fonctionnelle, University of Lille, Lille, France
| | | | - Vladimir Lupashin
- University of Arkansas for Medical Sciences, Department of Biophysics and Physiology, Little Rock, AR, United States
| | - Richard R Drake
- Department of Cell and Molecular Pharmacology, Medical University of South Carolina, Charleston, SC, United States
| | - Karen L Abbott
- University of Oklahoma Health Sciences Center, Department of Biochemistry and Molecular Biology, Oklahoma City, OK, United States
| |
Collapse
|
18
|
Investigating the functional link between TMEM165 and SPCA1. Biochem J 2020; 476:3281-3293. [PMID: 31652305 DOI: 10.1042/bcj20190488] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2019] [Revised: 10/07/2019] [Accepted: 10/17/2019] [Indexed: 01/09/2023]
Abstract
TMEM165 was highlighted in 2012 as the first member of the Uncharacterized Protein Family 0016 (UPF0016) related to human glycosylation diseases. Defects in TMEM165 are associated with strong Golgi glycosylation abnormalities. Our previous work has shown that TMEM165 rapidly degrades with supraphysiological manganese supplementation. In this paper, we establish a functional link between TMEM165 and SPCA1, the Golgi Ca2+/Mn2+ P-type ATPase pump. A nearly complete loss of TMEM165 was observed in SPCA1-deficient Hap1 cells. We demonstrate that TMEM165 was constitutively degraded in lysosomes in the absence of SPCA1. Complementation studies showed that TMEM165 abundance was directly dependent on SPCA1's function and more specifically its capacity to pump Mn2+ from the cytosol into the Golgi lumen. Among SPCA1 mutants that differentially impair Mn2+ and Ca2+ transport, only the Q747A mutant that favors Mn2+ pumping rescues the abundance and Golgi subcellular localization of TMEM165. Interestingly, the overexpression of SERCA2b also rescues the expression of TMEM165. Finally, this paper highlights that TMEM165 expression is linked to the function of SPCA1.
Collapse
|
19
|
Haijes HA, Jaeken J, van Hasselt PM. Hypothesis: determining phenotypic specificity facilitates understanding of pathophysiology in rare genetic disorders. J Inherit Metab Dis 2020; 43:701-711. [PMID: 31804708 PMCID: PMC7383723 DOI: 10.1002/jimd.12201] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/23/2019] [Revised: 11/28/2019] [Accepted: 12/03/2019] [Indexed: 12/17/2022]
Abstract
In the rapidly growing group of rare genetic disorders, data scarcity demands an intelligible use of available data, in order to improve understanding of underlying pathophysiology. We hypothesize, based on the principle that clinical similarities may be indicative of shared pathophysiology, that determining phenotypic specificity could provide unsuspected insights in pathophysiology of rare genetic disorders. We explored our hypothesis by studying subunit deficiencies of the conserved oligomeric Golgi (COG) complex, a subgroup of congenital disorders of glycosylation (CDG). In this systematic data assessment, all 45 reported patients with COG-CDG were included. The vocabulary of the Human Phenotype Ontology was used to annotate all phenotypic features and to assess occurrence in other genetic disorders. Gene occurrence ratios were calculated by dividing the frequency in the patient cohort over the number of associated genes, according to the Human Phenotype Ontology. Prioritisation based on phenotypic specificity was highly informative and captured phenotypic features commonly associated with glycosylation disorders. Moreover, it captured features not seen in any other glycosylation disorder, among which episodic fever, likely reflecting underappreciated other cellular functions of the COG complex. Interestingly, the COG complex was recently implicated in the autophagy pathway, as are more than half of the genes underlying disorders that present with episodic fever. This suggests that whereas many phenotypic features in these patients are caused by disrupted glycosylation, episodic fever might be caused by disrupted autophagy. Thus, we here demonstrate support for our hypothesis that determining phenotypic specificity could facilitate understanding of pathophysiology in rare genetic disorders.
Collapse
Affiliation(s)
- Hanneke A. Haijes
- Department of Biomedical Genetics, Section Metabolic DiagnosticsWilhelmina Children's Hospital, University Medical Centre Utrecht, Utrecht UniversityUtrechtThe Netherlands
- Department of Pediatrics, Subdivision Metabolic DiseasesWilhelmina Children's Hospital, University Medical Centre Utrecht, Utrecht UniversityUtrechtThe Netherlands
| | - Jaak Jaeken
- Department of PediatricsCentre for Metabolic Diseases, University Hospital GasthuisbergLeuvenBelgium
| | - Peter M. van Hasselt
- Department of Pediatrics, Subdivision Metabolic DiseasesWilhelmina Children's Hospital, University Medical Centre Utrecht, Utrecht UniversityUtrechtThe Netherlands
| |
Collapse
|
20
|
Foulquier F, Legrand D. Biometals and glycosylation in humans: Congenital disorders of glycosylation shed lights into the crucial role of Golgi manganese homeostasis. Biochim Biophys Acta Gen Subj 2020; 1864:129674. [PMID: 32599014 DOI: 10.1016/j.bbagen.2020.129674] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2020] [Revised: 06/18/2020] [Accepted: 06/24/2020] [Indexed: 12/24/2022]
Abstract
About half of the eukaryotic proteins bind biometals that participate in their structure and functions in virtually all physiological processes, including glycosylation. After reviewing the biological roles and transport mechanisms of calcium, magnesium, manganese, zinc and cobalt acting as cofactors of the metalloproteins involved in sugar metabolism and/or glycosylation, the paper will outline the pathologies resulting from a dysregulation of these metals homeostasis and more particularly Congenital Disorders of Glycosylation (CDGs) caused by ion transporter defects. Highlighting of CDGs due to defects in SLC39A8 (ZIP8) and TMEM165, two proteins transporting manganese from the extracellular space to cytosol and from cytosol to the Golgi lumen, respectively, has emphasized the importance of manganese homeostasis for glycosylation. Based on our current knowledge of TMEM165 structure and functions, this review will draw a picture of known and putative mechanisms regulating manganese homeostasis in the secretory pathway.
Collapse
Affiliation(s)
- François Foulquier
- Univ. Lille, CNRS, UMR 8576 - UGSF - Unité de Glycobiologie Structurale et Fonctionnelle, Lille F-59000, France
| | - Dominique Legrand
- Univ. Lille, CNRS, UMR 8576 - UGSF - Unité de Glycobiologie Structurale et Fonctionnelle, Lille F-59000, France.
| |
Collapse
|
21
|
Thines L, Stribny J, Morsomme P. From the Uncharacterized Protein Family 0016 to the GDT1 family: Molecular insights into a newly-characterized family of cation secondary transporters. MICROBIAL CELL 2020; 7:202-214. [PMID: 32743000 PMCID: PMC7380456 DOI: 10.15698/mic2020.08.725] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
The Uncharacterized Protein Family 0016 (UPF0016) gathers poorly studied membrane proteins well conserved through evolution that possess one or two copies of the consensus motif Glu-x-Gly-Asp-(Arg/Lys)-(Ser/Thr). Members are found in many eukaryotes, bacteria and archaea. The interest for this protein family arose in 2012 when its human member TMEM165 was linked to the occurrence of Congenital Disorders of Glycosylation (CDGs) when harbouring specific mutations. Study of the UPF0016 family is undergone through the characterization of the bacterium Vibrio cholerae (MneA), cyanobacterium Synechocystis (SynPAM71), yeast Saccharomyces cerevisiae (Gdt1p), plant Arabidopsis thaliana (PAM71 and CMT1), and human (TMEM165) members. These proteins have all been identified as transporters of cations, more precisely of Mn2+, with an extra reported function in Ca2+ and/or H+ transport for some of them. Apart from glycosylation in humans, the UPF0016 members are required for lactation in humans, photosynthesis in plants and cyanobacteria, Ca2+ signaling in yeast, and Mn2+ homeostasis in the five aforementioned species. The requirement of the UPF0016 members for key physiological processes most likely derives from their transport activity at the Golgi membrane in human and yeast, the chloroplasts membranes in plants, the thylakoid and plasma membranes in cyanobacteria, and the cell membrane in bacteria. In the light of these studies on various UPF0016 members, this family is not considered as uncharacterized anymore and has been renamed the Gdt1 family according to the name of its S. cerevisiae member. This review aims at assembling and confronting the current knowledge in order to identify shared and distinct features in terms of transported molecules, mode of action, structure, etc., as well as to better understand their corresponding physiological roles.
Collapse
Affiliation(s)
- Louise Thines
- Louvain Institute of Biomolecular Science and Technology, UCLouvain, Louvain-la-Neuve, Belgium
| | - Jiri Stribny
- Louvain Institute of Biomolecular Science and Technology, UCLouvain, Louvain-la-Neuve, Belgium
| | - Pierre Morsomme
- Louvain Institute of Biomolecular Science and Technology, UCLouvain, Louvain-la-Neuve, Belgium
| |
Collapse
|
22
|
Vicogne D, Houdou M, Garat A, Climer L, Lupashin V, Morelle W, Foulquier F. Fetal bovine serum impacts the observed N-glycosylation defects in TMEM165 KO HEK cells. J Inherit Metab Dis 2020; 43:357-366. [PMID: 31415112 PMCID: PMC7021577 DOI: 10.1002/jimd.12161] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/19/2019] [Revised: 07/28/2019] [Accepted: 07/29/2019] [Indexed: 01/10/2023]
Abstract
TMEM165 is involved in a rare genetic human disease named TMEM165-CDG (congenital disorders of glycosylation). It is Golgi localized, highly conserved through evolution and belongs to the uncharacterized protein family 0016 (UPF0016). The use of isogenic TMEM165 KO HEK cells was crucial in deciphering the function of TMEM165 in Golgi manganese homeostasis. Manganese is a major cofactor of many glycosylation enzymes. Severe Golgi glycosylation defects are observed in TMEM165 Knock Out Human Embryonic Kidney (KO HEK) cells and are rescued by exogenous manganese supplementation. Intriguingly, we demonstrate in this study that the observed Golgi glycosylation defect mainly depends on fetal bovine serum, particularly its manganese level. Our results also demonstrate that iron and/or galactose can modulate the observed glycosylation defects in TMEM165 KO HEK cells. While isogenic cultured cells are widely used to study the impact of gene defects on proteins' glycosylation patterns, these results emphasize the importance of the use of validated fetal bovine serum in glycomics studies.
Collapse
Affiliation(s)
- Dorothée Vicogne
- Univ. Lille, CNRS, UMR 8576 – UGSF - Unité de Glycobiologie Structurale et Fonctionnelle, F- 59000 Lille, France
| | - Marine Houdou
- Univ. Lille, CNRS, UMR 8576 – UGSF - Unité de Glycobiologie Structurale et Fonctionnelle, F- 59000 Lille, France
| | - Anne Garat
- Univ. Lille, EA 4483 - IMPECS - IMPact de l’Environnement Chimique sur la Santé humaine, F-59000 Lille, France
- CHU Lille, Unité Fonctionnelle de Toxicologie, F- 59000 Lille, France
| | | | - Vladimir Lupashin
- Department of Physiology and Biophysics, College of Medicine, University of Arkansas for Medical Sciences, Biomed 261-2, Slot 505, 200 South Cedar St., Little Rock, AR 72205, USA
| | - Willy Morelle
- Univ. Lille, CNRS, UMR 8576 – UGSF - Unité de Glycobiologie Structurale et Fonctionnelle, F- 59000 Lille, France
| | - François Foulquier
- Univ. Lille, CNRS, UMR 8576 – UGSF - Unité de Glycobiologie Structurale et Fonctionnelle, F- 59000 Lille, France
- Address correspondence should be sent to: François Foulquier ()
| |
Collapse
|
23
|
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
|
24
|
Houdou M, Lebredonchel E, Garat A, Duvet S, Legrand D, Decool V, Klein A, Ouzzine M, Gasnier B, Potelle S, Foulquier F. Involvement of thapsigargin- and cyclopiazonic acid-sensitive pumps in the rescue of TMEM165-associated glycosylation defects by Mn 2. FASEB J 2018; 33:2669-2679. [PMID: 30307768 DOI: 10.1096/fj.201800387r] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Congenital disorders of glycosylation are severe inherited diseases in which aberrant protein glycosylation is a hallmark. Transmembrane protein 165 (TMEM165) is a novel Golgi transmembrane protein involved in type II congenital disorders of glycosylation. Although its biologic function is still a controversial issue, we have demonstrated that the Golgi glycosylation defect due to TMEM165 deficiency resulted from a Golgi Mn2+ homeostasis defect. The goal of this study was to delineate the cellular pathway by which extracellular Mn2+ rescues N-glycosylation in TMEM165 knockout (KO) cells. We first demonstrated that after extracellular exposure, Mn2+ uptake by HEK293 cells at the plasma membrane did not rely on endocytosis but was likely done by plasma membrane transporters. Second, we showed that the secretory pathway Ca2+-ATPase 1, also known to mediate the influx of cytosolic Mn2+ into the lumen of the Golgi apparatus, is not crucial for the Mn2+-induced rescue glycosylation of lysosomal-associated membrane protein 2 (LAMP2). In contrast, our results demonstrate the involvement of cyclopiazonic acid- and thapsigargin (Tg)-sensitive pumps in the rescue of TMEM165-associated glycosylation defects by Mn2+. Interestingly, overexpression of sarco/endoplasmic reticulum Ca2+-ATPase (SERCA) 2b isoform in TMEM165 KO cells partially rescues the observed LAMP2 glycosylation defect. Overall, this study indicates that the rescue of Golgi N-glycosylation defects in TMEM165 KO cells by extracellular Mn2+ involves the activity of Tg and cyclopiazonic acid-sensitive pumps, probably the SERCA pumps.-Houdou, M., Lebredonchel, E., Garat, A., Duvet, S., Legrand, D., Decool, V., Klein, A., Ouzzine, M., Gasnier, B., Potelle, S., Foulquier, F. Involvement of thapsigargin- and cyclopiazonic acid-sensitive pumps in the rescue of TMEM165-associated glycosylation defects by Mn2+.
Collapse
Affiliation(s)
- Marine Houdou
- Centre National de la Recherche Scientifique (CNRS), Unité Mixte de Recherche (UMR) 8576, Unité de Glycobiologie Structurale et Fonctionnelle (UGSF), University of Lille, Lille, France
| | - Elodie Lebredonchel
- Centre National de la Recherche Scientifique (CNRS), Unité Mixte de Recherche (UMR) 8576, Unité de Glycobiologie Structurale et Fonctionnelle (UGSF), University of Lille, Lille, France
| | - Anne Garat
- Centre Hospitalier Universitaire (CHU) Lille, Institut Pasteur de Lille, Equipe d'Accueil (EA) 4483, Impact de l'Environnement Chimique sur la Santé Humaine (IMPECS), University of Lille, Lille, France
| | - Sandrine Duvet
- Centre National de la Recherche Scientifique (CNRS), Unité Mixte de Recherche (UMR) 8576, Unité de Glycobiologie Structurale et Fonctionnelle (UGSF), University of Lille, Lille, France
| | - Dominique Legrand
- Centre National de la Recherche Scientifique (CNRS), Unité Mixte de Recherche (UMR) 8576, Unité de Glycobiologie Structurale et Fonctionnelle (UGSF), University of Lille, Lille, France
| | - Valérie Decool
- Centre Hospitalier Universitaire (CHU) Lille, Institut Pasteur de Lille, Equipe d'Accueil (EA) 4483, Impact de l'Environnement Chimique sur la Santé Humaine (IMPECS), University of Lille, Lille, France
| | - André Klein
- Centre National de la Recherche Scientifique (CNRS), Unité Mixte de Recherche (UMR) 8576, Unité de Glycobiologie Structurale et Fonctionnelle (UGSF), University of Lille, Lille, France
| | - Mohamed Ouzzine
- Unité Mixte de Recherche (UMR) 7365, Centre National de la Recherche Scientifique (CNRS)-Université de Lorraine, Biopôle-Faculté de Médecine, Vandoeuvre-lès-Nancy, France
| | - Bruno Gasnier
- Neurophotonics Laboratory, Centre National de la Recherche Scientifique (CNRS) Unité Mixte de Recherche (UMR) 8250, Paris Descartes University, Sorbonne Paris Cité, Paris, France
| | - Sven Potelle
- Centre National de la Recherche Scientifique (CNRS), Unité Mixte de Recherche (UMR) 8576, Unité de Glycobiologie Structurale et Fonctionnelle (UGSF), University of Lille, Lille, France
| | - François Foulquier
- Centre National de la Recherche Scientifique (CNRS), Unité Mixte de Recherche (UMR) 8576, Unité de Glycobiologie Structurale et Fonctionnelle (UGSF), University of Lille, Lille, France
| |
Collapse
|
25
|
Harvey DJ. Analysis of carbohydrates and glycoconjugates by matrix-assisted laser desorption/ionization mass spectrometry: An update for 2013-2014. MASS SPECTROMETRY REVIEWS 2018; 37:353-491. [PMID: 29687922 DOI: 10.1002/mas.21530] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/16/2016] [Accepted: 11/29/2016] [Indexed: 06/08/2023]
Abstract
This review is the eighth update of the original article published in 1999 on the application of Matrix-assisted laser desorption/ionization mass spectrometry (MALDI) mass spectrometry to the analysis of carbohydrates and glycoconjugates and brings coverage of the literature to the end of 2014. Topics covered in the first part of the review include general aspects such as theory of the MALDI process, matrices, derivatization, MALDI imaging, fragmentation, and arrays. The second part of the review is devoted to applications to various structural types such as oligo- and poly- saccharides, glycoproteins, glycolipids, glycosides, and biopharmaceuticals. Much of this material is presented in tabular form. The third part of the review covers medical and industrial applications of the technique, studies of enzyme reactions, and applications to chemical synthesis. © 2018 Wiley Periodicals, Inc. Mass Spec Rev 37:353-491, 2018.
Collapse
Affiliation(s)
- David J Harvey
- Target Discovery Institute, Nuffield Department of Medicine, University of Oxford, Roosevelt Drive, Oxford, OX3 7FZ, United Kingdom
| |
Collapse
|
26
|
Bai X, Moraes TF, Reithmeier RAF. Structural biology of solute carrier (SLC) membrane transport proteins. Mol Membr Biol 2018; 34:1-32. [PMID: 29651895 DOI: 10.1080/09687688.2018.1448123] [Citation(s) in RCA: 102] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
The human solute carriers (SLCs) comprise over 400 different transporters, organized into 65 families ( http://slc.bioparadigms.org/ ) based on their sequence homology and transport function. SLCs are responsible for transporting extraordinarily diverse solutes across biological membranes, including inorganic ions, amino acids, lipids, sugars, neurotransmitters and drugs. Most of these membrane proteins function as coupled symporters (co-transporters) utilizing downhill ion (H+ or Na+) gradients as the driving force for the transport of substrate against its concentration gradient into cells. Other members work as antiporters (exchangers) that typically contain a single substrate-binding site with an alternating access mode of transport, while a few members exhibit channel-like properties. Dysfunction of SLCs is correlated with numerous human diseases and therefore they are potential therapeutic drug targets. In this review, we identified all of the SLC crystal structures that have been determined, most of which are from prokaryotic species. We further sorted all the SLC structures into four main groups with different protein folds and further discuss the well-characterized MFS (major facilitator superfamily) and LeuT (leucine transporter) folds. This review provides a systematic analysis of the structure, molecular basis of substrate recognition and mechanism of action in different SLC family members.
Collapse
Affiliation(s)
- Xiaoyun Bai
- a Department of Biochemistry , University of Toronto , Toronto , Canada
| | - Trevor F Moraes
- a Department of Biochemistry , University of Toronto , Toronto , Canada
| | | |
Collapse
|
27
|
H + and Pi Byproducts of Glycosylation Affect Ca 2+ Homeostasis and Are Retrieved from the Golgi Complex by Homologs of TMEM165 and XPR1. G3-GENES GENOMES GENETICS 2017; 7:3913-3924. [PMID: 29042410 PMCID: PMC5714488 DOI: 10.1534/g3.117.300339] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Glycosylation reactions in the Golgi complex and the endoplasmic reticulum utilize nucleotide sugars as donors and produce inorganic phosphate (Pi) and acid (H+) as byproducts. Here we show that homologs of mammalian XPR1 and TMEM165 (termed Erd1 and Gdt1) recycle luminal Pi and exchange luminal H+ for cytoplasmic Ca2+, respectively, thereby promoting growth of yeast cells in low Pi and low Ca2+ environments. As expected for reversible H+/Ca2+ exchangers, Gdt1 also promoted growth in high Ca2+ environments when the Golgi-localized V-ATPase was operational but had the opposite effect when the V-ATPase was eliminated. Gdt1 activities were negatively regulated by calcineurin signaling and by Erd1, which recycled the Pi byproduct of glycosylation reactions and prevented the loss of this nutrient to the environment via exocytosis. Thus, Erd1 transports Pi in the opposite direction from XPR1 and other EXS family proteins and facilitates byproduct removal from the Golgi complex together with Gdt1.
Collapse
|
28
|
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
|
29
|
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
|
30
|
Morelle W, Potelle S, Witters P, Wong S, Climer L, Lupashin V, Matthijs G, Gadomski T, Jaeken J, Cassiman D, Morava E, Foulquier F. Galactose Supplementation in Patients With TMEM165-CDG Rescues the Glycosylation Defects. J Clin Endocrinol Metab 2017; 102:1375-1386. [PMID: 28323990 PMCID: PMC6283449 DOI: 10.1210/jc.2016-3443] [Citation(s) in RCA: 64] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/13/2016] [Accepted: 12/22/2016] [Indexed: 01/18/2023]
Abstract
CONTEXT TMEM165 deficiency is a severe multisystem disease that manifests with metabolic, endocrine, and skeletal involvement. It leads to one type of congenital disorders of glycosylation (CDG), a rapidly growing group of inherited diseases in which the glycosylation process is altered. Patients have decreased galactosylation by serum glycan analysis. There are >100 CDGs, but only specific types are treatable. OBJECTIVE Galactose has been shown to be beneficial in other CDG types with abnormal galactosylation. The aim of this study was to characterize the effects of galactose supplementation on Golgi glycosylation in TMEM165-depleted HEK293 cells, as well as in 2 patients with TMEM165-CDG and in their cultured skin fibroblast cells. DESIGN AND SETTING Glycosylation was assessed by mass spectrometry, western blot analysis, and transferrin isoelectrofocusing. PATIENTS AND INTERVENTIONS Both unrelated patients with TMEM165-CDG with the same deep intronic homozygous mutation (c.792+182G>A) were allocated to receive d-galactose in a daily dose of 1 g/kg. RESULTS We analyzed N-linked glycans and glycolipids in knockout TMEM165 HEK293 cells, revealing severe hypogalactosylation and GalNAc transfer defects. Although these defects were completely corrected by the addition of Mn2+, we demonstrated that the observed N-glycosylation defect could also be overcome by galactose supplementation. We then demonstrated that oral galactose supplementation in patients with TMEM165-deficient CDG improved biochemical and clinical parameters, including a substantial increase in the negatively charged transferrin isoforms, and a decrease in hypogalactosylated total N-glycan structures, endocrine function, and coagulation parameters. CONCLUSION To our knowledge, this is the first description of abnormal glycosylation of lipids in the TMEM165 defect and the first report of successful dietary treatment in TMEM165 deficiency. We recommend the use of oral d-galactose therapy in TMEM165-CDG.
Collapse
Affiliation(s)
- Willy Morelle
- Université Lille, Centre National de la Recherche Française, UMR 8576-Unité de Glycobiologie Structurale et Fonctionnelle-Unité de Glycobiologie Structurale et Fonctionnelle, F-59000 Lille, France
| | - Sven Potelle
- Université Lille, Centre National de la Recherche Française, UMR 8576-Unité de Glycobiologie Structurale et Fonctionnelle-Unité de Glycobiologie Structurale et Fonctionnelle, F-59000 Lille, France
| | - Peter Witters
- Metabolic Center, Department of Pediatrics, University Hospitals Leuven, Leuven B-3000, Belgium
| | - Sunnie Wong
- Hayward Genetics Center, Tulane University School of Medicine, New Orleans, Louisiana 70112
| | - Leslie Climer
- Department of Physiology and Biophysics, College of Medicine, University of Arkansas for Medical Sciences, Little Rock, Arkansas 72205
| | - Vladimir Lupashin
- Department of Physiology and Biophysics, College of Medicine, University of Arkansas for Medical Sciences, Little Rock, Arkansas 72205
| | - Gert Matthijs
- Center for Human Genetics, KU Leuven, Leuven B-3000, Belgium
| | - Therese Gadomski
- Hayward Genetics Center, Tulane University School of Medicine, New Orleans, Louisiana 70112
| | - Jaak Jaeken
- Metabolic Center, Department of Pediatrics, University Hospitals Leuven, Leuven B-3000, Belgium
| | - David Cassiman
- Metabolic Center, Department of Pediatrics, University Hospitals Leuven, Leuven B-3000, Belgium
| | - Eva Morava
- Metabolic Center, Department of Pediatrics, University Hospitals Leuven, Leuven B-3000, Belgium
- Hayward Genetics Center, Tulane University School of Medicine, New Orleans, Louisiana 70112
| | - François Foulquier
- Université Lille, Centre National de la Recherche Française, UMR 8576-Unité de Glycobiologie Structurale et Fonctionnelle-Unité de Glycobiologie Structurale et Fonctionnelle, F-59000 Lille, France
| |
Collapse
|
31
|
Evidence for splice transcript variants of TMEM165, a gene involved in CDG. Biochim Biophys Acta Gen Subj 2017; 1861:737-748. [DOI: 10.1016/j.bbagen.2017.01.011] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2016] [Revised: 01/06/2017] [Accepted: 01/10/2017] [Indexed: 02/06/2023]
|
32
|
Hoecker N, Leister D, Schneider A. Plants contain small families of UPF0016 proteins including the PHOTOSYNTHESIS AFFECTED MUTANT71 transporter. PLANT SIGNALING & BEHAVIOR 2017; 12:e1278101. [PMID: 28075225 PMCID: PMC5351731 DOI: 10.1080/15592324.2016.1278101] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/05/2023]
Abstract
PHOTOSYNTHESIS AFFECTED MUTANT71 (PAM71) is an integral thylakoid membrane protein that functions in manganese uptake into the lumen. Manganese is needed in the thylakoid lumen to build up the inorganic Mn4CaO5 cluster, the catalytic center for water oxidation, and is hence indispensable for oxygen evolution. A recent study revealed that PAM71 is well conserved in plants and shares homology to GCR1 DEPENDENT TRANSLATION FACTOR1 (GDT1) and TRANSMEMBRANE PROTEIN 165 (TMEM165) in Saccharomyces cerevisiae and Homo sapiens, respectively. In most eukaryotes only single members of this family, designated "Uncharacterized Protein Family 0016" (UPF0016), are present; however, plant genomes contain genes for several UPF0016 proteins. In Arabidopsis thaliana, this protein family comprises 5 members, which mainly differ in their N-terminal regions. PAM71 and its closest homolog PAM71-HL possess chloroplast transit peptides at their N-terminus. Two of the remaining 3 members are derived from a segmental chromosomal duplication event and lack an N-terminal extension. Thus, plants have evolved UPF0016 members residing in various compartments of the cell, whereas in non-plant eukaryotes just a Golgi localization occurs. The identification of PAM71 as a candidate Mn2+ transporter opens the question on the function of the remaining plant members. Here we resume briefly our current knowledge of UPF0016 members in Arabidopsis in comparison to their yeast and human UPF0016 members.
Collapse
Affiliation(s)
- Natalie Hoecker
- Department of Biology I, Plant Sciences, Ludwig-Maximilians Universität München, Martinsried, Germany
| | - Dario Leister
- Department of Biology I, Plant Sciences, Ludwig-Maximilians Universität München, Martinsried, Germany
| | - Anja Schneider
- Department of Biology I, Plant Sciences, Ludwig-Maximilians Universität München, Martinsried, Germany
- CONTACT Anja Schneider
| |
Collapse
|
33
|
Dulary E, Potelle S, Legrand D, Foulquier F. TMEM165 deficiencies in Congenital Disorders of Glycosylation type II (CDG-II): Clues and evidences for roles of the protein in Golgi functions and ion homeostasis. Tissue Cell 2016; 49:150-156. [PMID: 27401145 DOI: 10.1016/j.tice.2016.06.006] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2016] [Revised: 06/10/2016] [Accepted: 06/14/2016] [Indexed: 12/28/2022]
Abstract
Congenital Disorders of Glycosylation (CDG) are rare inherited diseases causing glycosylation defects responsible for severe growth and psychomotor retardations in patients. Whereas most genetic defects affect enzymes directly involved in the glycosylation process, like glycosyltransferases or sugar transporters, recent findings revealed the impact of gene mutations on proteins implicated in both Golgi vesicular trafficking and ion homeostasis. TMEM165 is one of these deficient Golgi proteins found in CDG patients whose function in the secretory pathway has been deduced from several recent studies using TMEM165 deficient mammalian cells or yeast cells deficient in Gtd1p, the yeast TMEM165 ortholog. These studies actually confirm previous observations based on both sequence and predicted topology of this transmembrane protein and the phenotypes of human and yeast cells, namely that TMEM165 is very probably a transporter involved in ion homeostasis. Whereas the exact function of TMEM165 remains to be fully characterized, several studies hypothesize that TMEM165 could be a Golgi localized Ca2+/H+ antiporter. However, recent data also support the role of TMEM165 in Golgi Mn2+ homeostasis then arguing for a putative role of Mn2+ transporter for TMEM165 essential to achieve the correct N-glycosylation process of proteins in the secretory pathway. This manuscript is a review of the current state of knowledge on TMEM165 deficiencies in Congenital Disorders of Glycosylation as well as new data on function of TMEM165 and some speculative models on TMEM165/Golgi functions are discussed.
Collapse
Affiliation(s)
- Eudoxie Dulary
- Univ. Lille, CNRS, UMR 8576 - UGSF - Unité de Glycobiologie Structurale et Fonctionnelle, F- 59000 Lille, France
| | - Sven Potelle
- Univ. Lille, CNRS, UMR 8576 - UGSF - Unité de Glycobiologie Structurale et Fonctionnelle, F- 59000 Lille, France
| | - Dominique Legrand
- Univ. Lille, CNRS, UMR 8576 - UGSF - Unité de Glycobiologie Structurale et Fonctionnelle, F- 59000 Lille, France.
| | - François Foulquier
- Univ. Lille, CNRS, UMR 8576 - UGSF - Unité de Glycobiologie Structurale et Fonctionnelle, F- 59000 Lille, France.
| |
Collapse
|
34
|
Palagano E, Blair HC, Pangrazio A, Tourkova I, Strina D, Angius A, Cuccuru G, Oppo M, Uva P, Van Hul W, Boudin E, Superti-Furga A, Faletra F, Nocerino A, Ferrari MC, Grappiolo G, Monari M, Montanelli A, Vezzoni P, Villa A, Sobacchi C. Buried in the Middle but Guilty: Intronic Mutations in the TCIRG1 Gene Cause Human Autosomal Recessive Osteopetrosis. J Bone Miner Res 2015; 30:1814-21. [PMID: 25829125 DOI: 10.1002/jbmr.2517] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/11/2014] [Revised: 03/16/2015] [Accepted: 03/22/2015] [Indexed: 11/10/2022]
Abstract
Autosomal recessive osteopetrosis (ARO) is a rare genetic bone disease with genotypic and phenotypic heterogeneity, sometimes translating into delayed diagnosis and treatment. In particular, cases of intermediate severity often constitute a diagnostic challenge and represent good candidates for exome sequencing. Here, we describe the tortuous path to identification of the molecular defect in two siblings, in which osteopetrosis diagnosed in early childhood followed a milder course, allowing them to reach the adult age in relatively good conditions with no specific therapy. No clearly pathogenic mutation was identified either with standard amplification and resequencing protocols or with exome sequencing analysis. While evaluating the possible impact of a 3'UTR variant on the TCIRG1 expression, we found a novel single nucleotide change buried in the middle of intron 15 of the TCIRG1 gene, about 150 nucleotides away from the closest canonical splice site. By sequencing a number of independent cDNA clones covering exons 14 to 17, we demonstrated that this mutation reduced splicing efficiency but did not completely abrogate the production of the normal transcript. Prompted by this finding, we sequenced the same genomic region in 33 patients from our unresolved ARO cohort and found three additional novel single nucleotide changes in a similar location and with a predicted disruptive effect on splicing, further confirmed in one of them at the transcript level. Overall, we identified an intronic region in TCIRG1 that seems to be particularly prone to splicing mutations, allowing the production of a small amount of protein sufficient to reduce the severity of the phenotype usually associated with TCIRG1 defects. On this basis, we would recommend including TCIRG1 not only in the molecular work-up of severe infantile osteopetrosis but also in intermediate cases and carefully evaluating the possible effects of intronic changes.
Collapse
Affiliation(s)
- Eleonora Palagano
- UOS/IRGB, Milan Unit, National Research Council (CNR), Milan, Italy.,Humanitas Clinical and Research Center, Rozzano, Italy
| | - Harry C Blair
- Veteran's Affairs Medical Center and Department of Pathology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Alessandra Pangrazio
- UOS/IRGB, Milan Unit, National Research Council (CNR), Milan, Italy.,Humanitas Clinical and Research Center, Rozzano, Italy
| | - Irina Tourkova
- Veteran's Affairs Medical Center and Department of Pathology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Dario Strina
- UOS/IRGB, Milan Unit, National Research Council (CNR), Milan, Italy.,Humanitas Clinical and Research Center, Rozzano, Italy
| | - Andrea Angius
- CRS4, Science and Technology Park Polaris, Piscina Manna, Pula, Italy.,Institute of Genetic and Biomedical Research (IRGB), National Research Council (CNR), Monserrato, Italy
| | - Gianmauro Cuccuru
- CRS4, Science and Technology Park Polaris, Piscina Manna, Pula, Italy
| | - Manuela Oppo
- CRS4, Science and Technology Park Polaris, Piscina Manna, Pula, Italy
| | - Paolo Uva
- CRS4, Science and Technology Park Polaris, Piscina Manna, Pula, Italy
| | - Wim Van Hul
- Department of Medical Genetics, University of Antwerp, Antwerp, Belgium
| | - Eveline Boudin
- Department of Medical Genetics, University of Antwerp, Antwerp, Belgium
| | - Andrea Superti-Furga
- Department of Pediatrics, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - Flavio Faletra
- Institute for Maternal and Child Health-IRCCS "Burlo Garofolo", Trieste, Italy
| | - Agostino Nocerino
- Clinica Pediatrica, Azienda Ospedaliero-Universitaria "S Maria della Misericordia", Udine, Italy
| | - Matteo C Ferrari
- Hip and Prosthetic Replacement Unit, Humanitas Clinical and Research Center, Rozzano, Italy
| | - Guido Grappiolo
- Hip and Prosthetic Replacement Unit, Humanitas Clinical and Research Center, Rozzano, Italy
| | - Marta Monari
- Clinical Investigation Laboratory, Humanitas Clinical and Research Center, Rozzano, Italy
| | - Alessandro Montanelli
- Clinical Investigation Laboratory, Humanitas Clinical and Research Center, Rozzano, Italy
| | - Paolo Vezzoni
- UOS/IRGB, Milan Unit, National Research Council (CNR), Milan, Italy.,Humanitas Clinical and Research Center, Rozzano, Italy
| | - Anna Villa
- UOS/IRGB, Milan Unit, National Research Council (CNR), Milan, Italy.,Humanitas Clinical and Research Center, Rozzano, Italy
| | - Cristina Sobacchi
- UOS/IRGB, Milan Unit, National Research Council (CNR), Milan, Italy.,Humanitas Clinical and Research Center, Rozzano, Italy
| |
Collapse
|
35
|
Ondrušková N, Honzík T, Kytnarová J, Matoulek M, Zeman J, Hansíková H. Isoelectric Focusing of Serum Apolipoprotein C-III as a Sensitive Screening Method for the Detection of O-glycosylation Disturbances. Prague Med Rep 2015; 116:73-86. [PMID: 26093664 DOI: 10.14712/23362936.2015.48] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022] Open
Abstract
Apolipoprotein C-III (ApoC-III) is a glycoprotein carrying the most common O-linked glycan structure and is abundantly present in serum, what renders it a suitable marker for analysis of O-glycosylation abnormalities. Isoelectric focusing followed by a Western blot of ApoC-III, using PhastSystem™ Electrophoresis System (GE Healthcare), was introduced as a rather simple and rapid method for screening of certain subtypes of inherited glycosylation disorders. The study's aim was to establish this method in our laboratory, what included performing the analysis in a group of 170 healthy individuals to set the reference range of detected relative amounts of sialylated ApoC-III isoforms and to evaluate the gender- and age-dependent differences. A significant relative increase of asialo-ApoC-III with growing age was found. Secondly, we examined serum from patients with selected metabolic disorders and detected minor O-glycosylation changes in diseases such as Prader-Willi syndrome, PGM1 (phosphoglucomutase 1) or MAN1B (class 1B alpha-1,2-mannosidase) deficiency. Our results show that this method allows for a sensitive detection of ApoC-III O-glycosylation status, however this might be modulated by several factors (i.e. nutrition, medication) whose exact role remains to be determined.
Collapse
Affiliation(s)
- Nina Ondrušková
- Department of Pediatrics and Adolescent Medicine, First Faculty of Medicine, Charles University in Prague and General University Hospital in Prague, Prague, Czech Republic
| | - Tomáš Honzík
- Department of Pediatrics and Adolescent Medicine, First Faculty of Medicine, Charles University in Prague and General University Hospital in Prague, Prague, Czech Republic
| | - Jitka Kytnarová
- Department of Pediatrics and Adolescent Medicine, First Faculty of Medicine, Charles University in Prague and General University Hospital in Prague, Prague, Czech Republic
| | - Martin Matoulek
- 3rd Department of Medicine - Department of Endocrinology and Metabolism, First Faculty of Medicine, Charles University in Prague and General University Hospital in Prague, Prague, Czech Republic
| | - Jiří Zeman
- Department of Pediatrics and Adolescent Medicine, First Faculty of Medicine, Charles University in Prague and General University Hospital in Prague, Prague, Czech Republic
| | - Hana Hansíková
- Department of Pediatrics and Adolescent Medicine, First Faculty of Medicine, Charles University in Prague and General University Hospital in Prague, Prague, Czech Republic.
| |
Collapse
|
36
|
Bammens R, Mehta N, Race V, Foulquier F, Jaeken J, Tiemeyer M, Steet R, Matthijs G, Flanagan-Steet H. Abnormal cartilage development and altered N-glycosylation in Tmem165-deficient zebrafish mirrors the phenotypes associated with TMEM165-CDG. Glycobiology 2015; 25:669-82. [PMID: 25609749 DOI: 10.1093/glycob/cwv009] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2014] [Accepted: 01/13/2015] [Indexed: 11/13/2022] Open
Abstract
The congenital disorders of glycosylation (CDG), a group of inherited diseases characterized by aberrant glycosylation, encompass a wide range of defects, including glycosyltransferases, glycosidases, nucleotide-sugar transporters as well as proteins involved in maintaining Golgi architecture, pH and vesicular trafficking. Mutations in a previously undescribed protein, TMEM165, were recently shown to cause a new form of CDG, termed TMEM165-CDG. TMEM165-CDG patients exhibit cartilage and bone dysplasia and altered glycosylation of serum glycoproteins. We utilized a morpholino knockdown strategy in zebrafish to investigate the physiologic and pathogenic functions of TMEM165. Inhibition of tmem165 expression in developing zebrafish embryos caused craniofacial abnormalities, largely attributable to fewer chondrocytes. Decreased expression of several markers of cartilage and bone development suggests that Tmem165 deficiency alters both chondrocyte and osteoblast differentiation. Glycomic analysis of tmem165 morphants also revealed altered initiation, processing and extension of N-glycans, paralleling some of the glycosylation changes noted in human patients. Collectively, these findings highlight the utility of zebrafish to elucidate pathogenic mechanisms associated with glycosylation disorders and suggest that the cartilage and bone dysplasia manifested in TMEM165-CDG patients may stem from abnormal development of chondrocytes and osteoblasts.
Collapse
Affiliation(s)
- Riet Bammens
- Center for Human Genetics, University of Leuven, 3000 Leuven, Belgium
| | - Nickita Mehta
- Complex Carbohydrate Research Center, Department of Biochemistry and Molecular Biology, University of Georgia, Athens, GA 30602, USA
| | - Valérie Race
- Center for Human Genetics, University of Leuven, 3000 Leuven, Belgium
| | - François Foulquier
- CNRS-UMR 8576, Structural and Functional Glycobiology Unit, IFR 147, University of Lille 1, 59655 Villeneuve d'Ascq, France
| | - Jaak Jaeken
- Center for Metabolic Disease, University Hospital Gasthuisberg, 3000 Leuven, Belgium
| | - Michael Tiemeyer
- Complex Carbohydrate Research Center, Department of Biochemistry and Molecular Biology, University of Georgia, Athens, GA 30602, USA
| | - Richard Steet
- Complex Carbohydrate Research Center, Department of Biochemistry and Molecular Biology, University of Georgia, Athens, GA 30602, USA
| | - Gert Matthijs
- Center for Human Genetics, University of Leuven, 3000 Leuven, Belgium
| | - Heather Flanagan-Steet
- Complex Carbohydrate Research Center, Department of Biochemistry and Molecular Biology, University of Georgia, Athens, GA 30602, USA
| |
Collapse
|
37
|
Rymen D, Jaeken J. Skin manifestations in CDG. J Inherit Metab Dis 2014; 37:699-708. [PMID: 24554337 DOI: 10.1007/s10545-014-9678-7] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/30/2013] [Revised: 12/01/2013] [Accepted: 01/20/2014] [Indexed: 10/25/2022]
Abstract
The group of congenital disorders of glycosylation (CDG) has expanded tremendously since its first description in 1980, with around 70 distinct disorders described to date. A great phenotypic variability exists, ranging from multisystem disease to single organ involvement. Skin manifestations, although inconsistently present, are part of this broad clinical spectrum. Indeed, the presence of inverted nipples, fat pads and orange peel skin in a patient with developmental delay are considered as a hallmark of CDG, particularly seen in PMM2 deficiency. However, over the years many more dermatological findings have been observed (e.g., ichthyosis, cutis laxa, tumoral calcinosis…). In this review we will discuss the variety of skin manifestations reported in CDG. Moreover, we will explore the possible mechanisms that link a certain glycosylation deficiency to its skin phenotype.
Collapse
Affiliation(s)
- D Rymen
- Center for Human Genetics, University of Leuven, Leuven, Belgium,
| | | |
Collapse
|
38
|
Scott K, Gadomski T, Kozicz T, Morava E. Congenital disorders of glycosylation: new defects and still counting. J Inherit Metab Dis 2014; 37:609-17. [PMID: 24831587 PMCID: PMC4141334 DOI: 10.1007/s10545-014-9720-9] [Citation(s) in RCA: 93] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/14/2014] [Revised: 04/16/2014] [Accepted: 04/22/2014] [Indexed: 12/11/2022]
Abstract
Almost 50 inborn errors of metabolism have been described due to congenital defects in N-linked glycosylation. These phenotypically diverse disorders typically present as clinical syndromes, affecting multiple systems including the central nervous system, muscle function, transport, regulation, immunity, endocrine system, and coagulation. An increasing number of disorders have been discovered using novel techniques that combine glycobiology with next-generation sequencing or use tandem mass spectrometry in combination with molecular gene-hunting techniques. The number of "classic" congenital disorders of glycosylation (CDGs) due to N-linked glycosylation defects is still rising. Eight novel CDGs affecting N-linked glycans were discovered in 2013 alone. Newly discovered genes teach us about the significance of glycosylation in cell-cell interaction, signaling, organ development, cell survival, and mosaicism, in addition to the consequences of abnormal glycosylation for muscle function. We have learned how important glycosylation is in posttranslational modification and how glycosylation defects can imitate recognizable, previously described phenotypes. In many CDG subtypes, patients unexpectedly presented with long-term survival, whereas some others presented with nonsyndromic intellectual disability. In this review, recently discovered N-linked CDGs are described, with a focus on clinical presentations and therapeutic ideas. A diagnostic approach in unsolved N-linked CDG cases with abnormal transferrin screening results is also suggested.
Collapse
Affiliation(s)
- Kyle Scott
- Hayward Genetics Center, Tulane University School of Medicine, 1430 Tulane Ave, New Orleans, LA, 70112, USA
| | | | | | | |
Collapse
|
39
|
Yuste-Checa P, Medrano C, Gámez A, Desviat LR, Matthijs G, Ugarte M, Pérez-Cerdá C, Pérez B. Antisense-mediated therapeutic pseudoexon skipping in TMEM165-CDG. Clin Genet 2014; 87:42-8. [PMID: 24720419 DOI: 10.1111/cge.12402] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2014] [Revised: 04/08/2014] [Accepted: 04/08/2014] [Indexed: 01/18/2023]
Abstract
Deficiencies in glycosyltransferases, glycosidases or nucleotide-sugar transporters involved in protein glycosylation lead to congenital disorders of glycosylation (CDG), a group of genetic diseases mostly showing multisystem phenotype. Despite recent advances in the biochemical and molecular knowledge of these diseases, no effective therapy exists for most. Efforts are now being directed toward therapies based on identifying new targets, which would allow to treat specific patients in a personalized way. This work presents proof-of concept for the antisense RNA rescue of the Golgi-resident protein TMEM165, a gene involved in a new type of CDG with a characteristic skeletal phenotype. Using a functional in vitro splicing assay based on minigenes, it was found that the deep intronic change c.792+182G>A is responsible for the insertion of an aberrant exon, corresponding to an intronic sequence. Antisense morpholino oligonucleotide therapy targeted toward TMEM165 mRNA recovered normal protein levels in the Golgi apparatus of patient-derived fibroblasts. This work expands the application of antisense oligonucleotide-mediated pseudoexon skipping to the treatment of a Golgi-resident protein, and opens up a promising treatment option for this specific TMEM165-CDG.
Collapse
Affiliation(s)
- P Yuste-Checa
- Centro de Diagnóstico de Enfermedades Moleculares, Centro de Biología Molecular-SO UAM-CSIC, Campus de Cantoblanco, Universidad Autónoma de Madrid, Madrid, Spain; Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), IDIPaz, Madrid, Spain; Instituto de Investigación Biomedica, IDIPaz, Madrid
| | | | | | | | | | | | | | | |
Collapse
|
40
|
Ong YS, Tran THT, Gounko NV, Hong W. TMEM115 is an integral membrane protein of the Golgi complex involved in retrograde transport. J Cell Sci 2014; 127:2825-39. [PMID: 24806965 PMCID: PMC4077589 DOI: 10.1242/jcs.136754] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Searching and evaluating the Human Protein Atlas for transmembrane proteins enabled us to identify an integral membrane protein, TMEM115, that is enriched in the Golgi complex. Biochemical and cell biological analysis suggested that TMEM115 has four candidate transmembrane domains located in the N-terminal region. Both the N- and C-terminal domains are oriented towards the cytoplasm. Immunofluorescence analysis supports that TMEM115 is enriched in the Golgi cisternae. Functionally, TMEM115 knockdown or overexpression delays Brefeldin-A-induced Golgi-to-ER retrograde transport, phenocopying cells with mutations or silencing of the conserved oligomeric Golgi (COG) complex. Co-immunoprecipitation and in vitro binding experiments reveals that TMEM115 interacts with the COG complex, and might self-interact to form dimers or oligomers. A short region (residues 206–229) immediately to the C-terminal side of the fourth transmembrane domain is both necessary and sufficient for Golgi targeting. Knockdown of TMEM115 also reduces the binding of the lectins peanut agglutinin (PNA) and Helix pomatia agglutinin (HPA), suggesting an altered O-linked glycosylation profile. These results establish that TMEM115 is an integral membrane protein of the Golgi stack regulating Golgi-to-ER retrograde transport and is likely to be part of the machinery of the COG complex.
Collapse
Affiliation(s)
- Yan Shan Ong
- Institute of Molecular and Cell Biology, 61 Biopolis Drive, Singapore 138673, Singapore
| | - Ton Hoai Thi Tran
- Institute of Molecular and Cell Biology, 61 Biopolis Drive, Singapore 138673, Singapore
| | - Natalia V Gounko
- Institute of Molecular and Cell Biology, 61 Biopolis Drive, Singapore 138673, Singapore IMB-IMCB Joint Electron Microscopy Suite, 20 Biopolis Street, Singapore 138671, Singapore
| | - Wanjin Hong
- Institute of Molecular and Cell Biology, 61 Biopolis Drive, Singapore 138673, Singapore Department of Biochemistry, National University of Singapore, Singapore 117599, Singapore
| |
Collapse
|
41
|
Bexiga MG, Simpson JC. Human diseases associated with form and function of the Golgi complex. Int J Mol Sci 2013; 14:18670-81. [PMID: 24025425 PMCID: PMC3794802 DOI: 10.3390/ijms140918670] [Citation(s) in RCA: 62] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2013] [Revised: 08/09/2013] [Accepted: 09/03/2013] [Indexed: 11/16/2022] Open
Abstract
The Golgi complex lies at the heart of the secretory pathway and is responsible for modifying proteins and lipids, as well as sorting newly synthesized molecules to their correct destination. As a consequence of these important roles, any changes in its proteome can negatively affect its function and in turn lead to disease. Recently, a number of proteins have been identified, which when either depleted or mutated, result in diseases that affect various organ systems. Here we describe how these proteins have been linked to the Golgi complex, and specifically how they affect either the morphology, membrane traffic or glycosylation ability of this organelle.
Collapse
Affiliation(s)
| | - Jeremy C. Simpson
- Author to whom correspondence should be addressed; E-Mail: ; Tel.: +353-1-716-2345; Fax: +353-1-716-1153
| |
Collapse
|