1
|
Cebulla G, Hai L, Warnken U, Güngör C, Hoffmann DC, Korporal-Kuhnke M, Wildemann B, Wick W, Kessler T, Weiler M. Long-term CSF responses in adult patients with spinal muscular atrophy type 2 or 3 on treatment with nusinersen. J Neurol 2025; 272:270. [PMID: 40085221 PMCID: PMC11909034 DOI: 10.1007/s00415-025-12984-7] [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: 11/03/2024] [Revised: 01/21/2025] [Accepted: 02/10/2025] [Indexed: 03/16/2025]
Abstract
BACKGROUND 5q-associated spinal muscular atrophy (SMA) is a monogenic disease causing progressive alpha motor neuron degeneration, muscle atrophy, and weakness. Intrathecal therapy with the antisense oligonucleotide nusinersen modifies the disease course. However, biomarkers for understanding underlying molecular pathomechanisms and monitoring therapy are not yet known. METHODS A total of 130 cerebrospinal fluid (CSF) samples from 24 adult patients with SMA type 2 or 3 were collected over 3.5 years, and CSF proteome was analyzed using mass spectrometry (MS). By applying two complementary MS protein quantification methods, label-free quantification (LFQ) and tandem mass tag (TMT) isotopic labeling, specific protein patterns reflecting changes in the CSF in response to nusinersen therapy were identified. These results were combined with cellular and metabolic profiles. RESULTS Nusinersen therapy led to a median motor function improvement of 2.2 Hammersmith Functional Motor Scale-Expanded points after 10 months and 2.6 points after 34 months. CSF macrophages increased in number and showed an altered morphology. Albumin quotient (qAlb), glucose, and lactate concentrations were inversely correlated with clinical improvement. MS analysis of CSF identified 1,674 (TMT) and 441 (LFQ) proteins. Protein profiles reflected reduced inhibition of "nervous system development" and "axogenesis" pathways under therapy. In addition, clinical improvement was associated with upregulation of the interacting proteins α-dystroglycan and beta-1,4-glucuronyltransferase 1, reduction of complement factors, negative correlation in immunoglobulin- and B cell-related pathways, and reduction of cellular mediators such as lymphocytes. CONCLUSION The present multi-proteomic analysis contributes to the understanding of the molecular mechanisms underlying nusinersen's therapeutic effects and offers potential biomarkers for monitoring treatment response in SMA.
Collapse
Affiliation(s)
- Gina Cebulla
- Clinical Cooperation Unit Neurooncology, German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), Heidelberg, Germany
- Department of Neurology, Neurology and Neurooncology Program, National Center for Tumor Diseases, Heidelberg University Hospital, Im Neuenheimer Feld 400, 69120, Heidelberg, Germany
| | - Ling Hai
- Clinical Cooperation Unit Neurooncology, German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), Heidelberg, Germany
- Department of Neurology, Neurology and Neurooncology Program, National Center for Tumor Diseases, Heidelberg University Hospital, Im Neuenheimer Feld 400, 69120, Heidelberg, Germany
| | - Uwe Warnken
- Clinical Cooperation Unit Neurooncology, German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Cansu Güngör
- Department of Neurology, Heidelberg University Hospital, Im Neuenheimer Feld 400, 69120, Heidelberg, Germany
| | - Dirk C Hoffmann
- Clinical Cooperation Unit Neurooncology, German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), Heidelberg, Germany
- Department of Neurology, Neurology and Neurooncology Program, National Center for Tumor Diseases, Heidelberg University Hospital, Im Neuenheimer Feld 400, 69120, Heidelberg, Germany
| | - Mirjam Korporal-Kuhnke
- Department of Neurology, Heidelberg University Hospital, Im Neuenheimer Feld 400, 69120, Heidelberg, Germany
| | - Brigitte Wildemann
- Department of Neurology, Heidelberg University Hospital, Im Neuenheimer Feld 400, 69120, Heidelberg, Germany
| | - Wolfgang Wick
- Clinical Cooperation Unit Neurooncology, German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), Heidelberg, Germany
- Department of Neurology, Neurology and Neurooncology Program, National Center for Tumor Diseases, Heidelberg University Hospital, Im Neuenheimer Feld 400, 69120, Heidelberg, Germany
| | - Tobias Kessler
- Clinical Cooperation Unit Neurooncology, German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), Heidelberg, Germany.
- Department of Neurology, Neurology and Neurooncology Program, National Center for Tumor Diseases, Heidelberg University Hospital, Im Neuenheimer Feld 400, 69120, Heidelberg, Germany.
| | - Markus Weiler
- Department of Neurology, Heidelberg University Hospital, Im Neuenheimer Feld 400, 69120, Heidelberg, Germany.
| |
Collapse
|
2
|
Kotera K, Miyamoto R, Mochizuki G, Tamura T, Manabe N, Yamaguchi Y, Tamura JI. Chemical extension and glycodendrimer formation of the matriglycan decasaccharide, -(3Xylα1-3GlcAβ1) 5- and its affinity for laminin. Carbohydr Res 2025; 547:109328. [PMID: 39603177 DOI: 10.1016/j.carres.2024.109328] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2024] [Revised: 10/29/2024] [Accepted: 11/20/2024] [Indexed: 11/29/2024]
Abstract
Muscle tissue is stabilized by the strong interaction between laminin and matriglycan. Matriglycan is a polysaccharide composed of the repeating disaccharide, -3Xylα1-3GlcAβ1-, and is a pivotal part of the core M3 O-mannosyl glycan. Patients with muscular dystrophy cannot synthesize matriglycan or the core M3 O-mannosyl glycan due to a defect in or the lack of glycosyltransferases owing to glycan synthesis. Therefore, a supply of matriglycan may be a powerful tool for reconstructing muscle tissue in these patients. We herein report the synthesis of a matriglycan-repeating decasaccharide and a dendrimer comprising three branches of the decasaccharide. The glycan was regio- and stereoselectively synthesized by the stepwise addition of the corresponding disaccharide unit. The immobilized decasaccharide and glycodendrimer bound to laminin-G-like domains 4 and 5 of laminin-α2. The dissociation constants of the decasaccharide and dendrimer obtained from bio-layer interferometry were estimated to be 4.4 × 10-8 M and 6.8 × 10-8 M, respectively, showing higher affinity than those of a matriglycan-repeating hexasaccharide (1.6 × 10-7 M) and the dendrimer (1.8 × 10-7 M).
Collapse
Affiliation(s)
- Kota Kotera
- Graduate School of Sustainability Science, Department of Agricultural Science, Tottori University, Tottori, 680-8553, Japan
| | - Ren Miyamoto
- Division of Structural Glycobiology, Institute of Molecular Biomembrane and Glycobiology, Tohoku Medical and Pharmaceutical University, Sendai, 981-8558, Japan
| | - Gakuto Mochizuki
- Department of Agricultural, life and Environmental Sciences, Faculty of Agriculture, Tottori University, Tottori, 680-8553, Japan
| | - Takahiro Tamura
- The United Graduate School of Agricultural Sciences, Tottori University, Tottori, 680-8553, Japan
| | - Noriyoshi Manabe
- Division of Structural Glycobiology, Institute of Molecular Biomembrane and Glycobiology, Tohoku Medical and Pharmaceutical University, Sendai, 981-8558, Japan
| | - Yoshiki Yamaguchi
- Division of Structural Glycobiology, Institute of Molecular Biomembrane and Glycobiology, Tohoku Medical and Pharmaceutical University, Sendai, 981-8558, Japan.
| | - Jun-Ichi Tamura
- Graduate School of Sustainability Science, Department of Agricultural Science, Tottori University, Tottori, 680-8553, Japan; Department of Agricultural, life and Environmental Sciences, Faculty of Agriculture, Tottori University, Tottori, 680-8553, Japan; The United Graduate School of Agricultural Sciences, Tottori University, Tottori, 680-8553, Japan.
| |
Collapse
|
3
|
Lacin H, Zhu Y, DiPaola JT, Wilson BA, Zhu Y, Skeath JB. A genetic screen in Drosophila uncovers a role for senseless-2 in surface glia in the peripheral nervous system to regulate CNS morphology. G3 (BETHESDA, MD.) 2024; 14:jkae152. [PMID: 38996053 PMCID: PMC11373656 DOI: 10.1093/g3journal/jkae152] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/26/2024] [Revised: 07/01/2024] [Accepted: 07/02/2024] [Indexed: 07/14/2024]
Abstract
Despite increasing in mass approximately 100-fold during larval life, the Drosophila CNS maintains its characteristic form. Dynamic interactions between the overlying basement membrane and underlying surface glia are known to regulate CNS structure in Drosophila, but the genes and pathways that establish and maintain CNS morphology during development remain poorly characterized. To identify genes that regulate CNS shape in Drosophila, we conducted an EMS-based, forward genetic screen of the second chromosome, uncovered 50 mutations that disrupt CNS structure, and mapped these alleles to 17 genes. Analysis of whole genome sequencing data wedded to genetic studies uncovered the affected gene for all but 1 mutation. Identified genes include well-characterized regulators of tissue shape, like LanB1, viking, and Collagen type IV alpha1, and previously characterized genes, such as Toll-2 and Rme-8, with no known role in regulating CNS structure. We also uncovered that papilin and C1GalTA likely act in the same pathway to regulate CNS structure and found that the fly homolog of a glucuronosyltransferase, B4GAT1/LARGE1, that regulates Dystroglycan function in mammals is required to maintain CNS shape in Drosophila. Finally, we show that the senseless-2 transcription factor is expressed and functions specifically in surface glia found on peripheral nerves but not in the CNS to govern CNS structure, identifying a gene that functionally subdivides a glial subtype along the peripheral-central axis. Future work on these genes should clarify the genetic mechanisms that ensure the homeostasis of CNS form during development.
Collapse
Affiliation(s)
- Haluk Lacin
- Division of Biological and Biomedical Systems, University of Missouri-Kansas City, 5009 Rockhill Road, Kansas City, MO 64110, USA
| | - Yuqing Zhu
- Department of Genetics, Washington University School of Medicine, 4523 Clayton Avenue, St. Louis, MO 63110, USA
| | - Jose T DiPaola
- Department of Genetics, Washington University School of Medicine, 4523 Clayton Avenue, St. Louis, MO 63110, USA
| | - Beth A Wilson
- Department of Genetics, Washington University School of Medicine, 4523 Clayton Avenue, St. Louis, MO 63110, USA
| | - Yi Zhu
- Department of Genetics, Washington University School of Medicine, 4523 Clayton Avenue, St. Louis, MO 63110, USA
| | - James B Skeath
- Department of Genetics, Washington University School of Medicine, 4523 Clayton Avenue, St. Louis, MO 63110, USA
| |
Collapse
|
4
|
Melrose J. Dystroglycan-HSPG interactions provide synaptic plasticity and specificity. Glycobiology 2024; 34:cwae051. [PMID: 39223703 PMCID: PMC11368572 DOI: 10.1093/glycob/cwae051] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2024] [Accepted: 07/10/2024] [Indexed: 09/04/2024] Open
Abstract
AIM This study examined the roles of the laminin and proteoglycan receptor dystroglycan (DG) in extracellular matrix stabilization and cellular mechanosensory processes conveyed through communication between the extracellular matrix (ECM) and cytoskeleton facilitated by DG. Specific functional attributes of HS-proteoglycans (HSPGs) are conveyed through interactions with DG and provide synaptic specificity through diverse interactions with an extensive range of cell attachment and adaptor proteins which convey synaptic plasticity. HSPG-DG interactions are important in phototransduction and neurotransduction and facilitate retinal bipolar-photoreceptor neuronal signaling in vision. Besides synaptic stabilization, HSPG-DG interactions also stabilize basement membranes and the ECM and have specific roles in the assembly and function of the neuromuscular junction. This provides neuromuscular control of muscle systems that control conscious body movement as well as essential autonomic control of diaphragm, intercostal and abdominal muscles and muscle systems in the face, mouth and pharynx which assist in breathing processes. DG is thus a multifunctional cell regulatory glycoprotein receptor and regulates a diverse range of biological and physiological processes throughout the human body. The unique glycosylation of the αDG domain is responsible for its diverse interactions with ECM components in cell-ECM signaling. Cytoskeletal cell regulatory switches assembled by the βDG domain in its role as a nuclear scaffolding protein respond to such ECM cues to regulate cellular behavior and tissue homeostasis thus DG has fascinating and diverse roles in health and disease.
Collapse
Affiliation(s)
- James Melrose
- Raymond Purves Bone and Joint Research Laboratory, Kolling Institute, St. Leonards, NSW 2065, Australia
- School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney at Royal North Shore Hospital, St. Leonards, NSW 2065, Australia
- Graduate School of Biomedical Engineering, Faculty of Engineering, University of New South Wales, Sydney, NSW 2052, Australia
| |
Collapse
|
5
|
Mulim HA, Hernandez RO, Vanderhout R, Bai X, Willems O, Regmi P, Erasmus MA, Brito LF. Genetic background of walking ability and its relationship with leg defects, mortality, and performance traits in turkeys (Meleagris gallopavo). Poult Sci 2024; 103:103779. [PMID: 38788487 PMCID: PMC11145530 DOI: 10.1016/j.psj.2024.103779] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2024] [Revised: 03/28/2024] [Accepted: 04/17/2024] [Indexed: 05/26/2024] Open
Abstract
This study aimed to explore the genetic basis of walking ability and potentially related performance traits in turkey purebred populations. Phenotypic, pedigree, and genomic datasets from 2 turkey lines hatched between 2010 and 2023 were included in the study. Walking ability data, defined based on a scoring system ranging from 1 (worst) to 6 (best), were collected on 192,019 animals of a female line and 235,461 animals of a male line. Genomic information was obtained for 46,427 turkeys (22,302 from a female line and 24,125 from a male line) using a 65K single nucleotide polymorphism (SNP) panel. Variance components and heritability for walking ability were estimated. Furthermore, genetic and phenotypic correlations among walking ability, mortality and disorders, and performance traits were calculated. A genome-wide association study (GWAS) was also conducted to identify SNPs associated with walking ability. Walking ability is moderately heritable (0.23 ± 0.01) in both turkey lines. The genetic correlations between walking ability and the other evaluated traits ranged from -0.02 to -0.78, with leg defects exhibiting the strongest negative correlation with walking ability. In the female line, 31 SNPs were associated with walking ability and overlapped with 116 genes. These positional genes are linked to 6 gene ontology (GO) terms. Notably, genes such as CSRP2, DDX1, RHBDL1, SEZ6L, and CTSK are involved in growth, development, locomotion, and bone disorders. GO terms, including fibronectin binding (GO:0001968), peptide cross-linking (GO:0018149), and catabolic process (GO:0009057), are directly linked with mobility. In the male line, 66 markers associated with walking ability were identified and overlapped with 281 genes. These genes are linked to 12 GO terms. Genes such as RB1CC1, TNNI1, MSTN, FN1, SIK3, PADI2, ERBB4, B3GNT2, and BACE1 are associated with cell growth, myostatin development, and disorders. GO terms in the male line are predominantly related to lipid metabolism. In conclusion, walking ability is moderately heritable in both populations. Furthermore, walking ability can be enhanced through targeted genetic selection, emphasizing its relevance to both animal welfare and productivity.
Collapse
Affiliation(s)
- Henrique A Mulim
- Department of Animal Sciences, Purdue University, West Lafayette, IN, USA
| | - Rick O Hernandez
- Department of Animal Sciences, Purdue University, West Lafayette, IN, USA
| | | | - Xuechun Bai
- Hendrix Genetics Limited, Kitchener, ON, Canada
| | | | - Prafulla Regmi
- Department of Poultry Science, University of Georgia, Athens, GA, USA
| | - Marisa A Erasmus
- Department of Animal Sciences, Purdue University, West Lafayette, IN, USA
| | - Luiz F Brito
- Department of Animal Sciences, Purdue University, West Lafayette, IN, USA.
| |
Collapse
|
6
|
Naranjo Á, Álvarez-Soria MJ, Aranda-Villalobos P, Martínez-Rodríguez AM, Martínez-Lara E, Siles E. Hydroxytyrosol, a Promising Supplement in the Management of Human Stroke: An Exploratory Study. Int J Mol Sci 2024; 25:4799. [PMID: 38732018 PMCID: PMC11084205 DOI: 10.3390/ijms25094799] [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: 03/20/2024] [Revised: 04/24/2024] [Accepted: 04/24/2024] [Indexed: 05/13/2024] Open
Abstract
Hydroxytyrosol (HT) is a bioactive olive oil phenol with beneficial effects in a number of pathological situations. We have previously demonstrated that an HT-enriched diet could serve as a beneficial therapeutic approach to attenuate ischemic-stroke-associated damage in mice. Our exploratory pilot study examined this effect in humans. Particularly, a nutritional supplement containing 15 mg of HT/day was administered to patients 24 h after the onset of stroke, for 45 days. Biochemical and oxidative-stress-related parameters, blood pressure levels, serum proteome, and neurological and functional outcomes were evaluated at 45 and 90 days and compared to a control group. The main findings were that the daily administration of HT after stroke could: (i) favor the decrease in the percentage of glycated hemoglobin and diastolic blood pressure, (ii) control the increase in nitric oxide and exert a plausible protective effect in oxidative stress, (iii) modulate the evolution of the serum proteome and, particularly, the expression of apolipoproteins, and (iv) be beneficial for certain neurological and functional outcomes. Although a larger trial is necessary, this study suggests that HT could be a beneficial nutritional complement in the management of human stroke.
Collapse
Affiliation(s)
- Ángela Naranjo
- Departamento de Biología Experimental, Universidad de Jaén, 23071 Jaén, Spain;
| | | | | | | | | | - Eva Siles
- Departamento de Biología Experimental, Universidad de Jaén, 23071 Jaén, Spain;
| |
Collapse
|
7
|
Katz M, Diskin R. The underlying mechanisms of arenaviral entry through matriglycan. Front Mol Biosci 2024; 11:1371551. [PMID: 38516183 PMCID: PMC10955480 DOI: 10.3389/fmolb.2024.1371551] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2024] [Accepted: 02/15/2024] [Indexed: 03/23/2024] Open
Abstract
Matriglycan, a recently characterized linear polysaccharide, is composed of alternating xylose and glucuronic acid subunits bound to the ubiquitously expressed protein α-dystroglycan (α-DG). Pathogenic arenaviruses, like the Lassa virus (LASV), hijack this long linear polysaccharide to gain cellular entry. Until recently, it was unclear through what mechanisms LASV engages its matriglycan receptor to initiate infection. Additionally, how matriglycan is synthesized onto α-DG by the Golgi-resident glycosyltransferase LARGE1 remained enigmatic. Recent structural data for LARGE1 and for the LASV spike complex informs us about the synthesis of matriglycan as well as its usage as an entry receptor by arenaviruses. In this review, we discuss structural insights into the system of matriglycan generation and eventual recognition by pathogenic viruses. We also highlight the unique usage of matriglycan as a high-affinity host receptor compared with other polysaccharides that decorate cells.
Collapse
Affiliation(s)
| | - Ron Diskin
- Department of Chemical and Structural Biology, Weizmann Institute of Science, Rehovot, Israel
| |
Collapse
|
8
|
Yang T, Chandel I, Gonzales M, Okuma H, Prouty SJ, Zarei S, Joseph S, Garringer KW, Landa SO, Yonekawa T, Walimbe AS, Venzke DP, Anderson ME, Hord JM, Campbell KP. Identification of a short, single site matriglycan that maintains neuromuscular function in the mouse. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.12.20.572361. [PMID: 38187633 PMCID: PMC10769215 DOI: 10.1101/2023.12.20.572361] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/09/2024]
Abstract
Matriglycan (-1,3-β-glucuronic acid-1,3-α-xylose-) is a polysaccharide that is synthesized on α-dystroglycan, where it functions as a high-affinity glycan receptor for extracellular proteins, such as laminin, perlecan and agrin, thus anchoring the plasma membrane to the extracellular matrix. This biological activity is closely associated with the size of matriglycan. Using high-resolution mass spectrometry and site-specific mutant mice, we show for the first time that matriglycan on the T317/T319 and T379 sites of α-dystroglycan are not identical. T379-linked matriglycan is shorter than the previously characterized T317/T319-linked matriglycan, although it maintains its laminin binding capacity. Transgenic mice with only the shorter T379-linked matriglycan exhibited mild embryonic lethality, but those that survived were healthy. The shorter T379-linked matriglycan exists in multiple tissues and maintains neuromuscular function in adult mice. In addition, the genetic transfer of α-dystroglycan carrying just the short matriglycan restored grip strength and protected skeletal muscle from eccentric contraction-induced damage in muscle-specific dystroglycan knock-out mice. Due to the effects that matriglycan imparts on the extracellular proteome and its ability to modulate cell-matrix interactions, our work suggests that differential regulation of matriglycan length in various tissues optimizes the extracellular environment for unique cell types.
Collapse
Affiliation(s)
- Tiandi Yang
- Howard Hughes Medical Institute, Department of Molecular Physiology and Biophysics, Department of Neurology, University of Iowa Roy J. and Lucille A. Carver College of Medicine, Iowa City, IA 52242 USA
- Department of Immunology, Harvard Medical School, Boston, MA 02115, USA
| | - Ishita Chandel
- Howard Hughes Medical Institute, Department of Molecular Physiology and Biophysics, Department of Neurology, University of Iowa Roy J. and Lucille A. Carver College of Medicine, Iowa City, IA 52242 USA
| | - Miguel Gonzales
- Howard Hughes Medical Institute, Department of Molecular Physiology and Biophysics, Department of Neurology, University of Iowa Roy J. and Lucille A. Carver College of Medicine, Iowa City, IA 52242 USA
| | - Hidehiko Okuma
- Howard Hughes Medical Institute, Department of Molecular Physiology and Biophysics, Department of Neurology, University of Iowa Roy J. and Lucille A. Carver College of Medicine, Iowa City, IA 52242 USA
| | - Sally J Prouty
- Howard Hughes Medical Institute, Department of Molecular Physiology and Biophysics, Department of Neurology, University of Iowa Roy J. and Lucille A. Carver College of Medicine, Iowa City, IA 52242 USA
| | - Sanam Zarei
- Howard Hughes Medical Institute, Department of Molecular Physiology and Biophysics, Department of Neurology, University of Iowa Roy J. and Lucille A. Carver College of Medicine, Iowa City, IA 52242 USA
| | - Soumya Joseph
- Howard Hughes Medical Institute, Department of Molecular Physiology and Biophysics, Department of Neurology, University of Iowa Roy J. and Lucille A. Carver College of Medicine, Iowa City, IA 52242 USA
| | - Keith W Garringer
- Howard Hughes Medical Institute, Department of Molecular Physiology and Biophysics, Department of Neurology, University of Iowa Roy J. and Lucille A. Carver College of Medicine, Iowa City, IA 52242 USA
| | - Saul Ocampo Landa
- Howard Hughes Medical Institute, Department of Molecular Physiology and Biophysics, Department of Neurology, University of Iowa Roy J. and Lucille A. Carver College of Medicine, Iowa City, IA 52242 USA
| | - Takahiro Yonekawa
- Howard Hughes Medical Institute, Department of Molecular Physiology and Biophysics, Department of Neurology, University of Iowa Roy J. and Lucille A. Carver College of Medicine, Iowa City, IA 52242 USA
| | - Ameya S Walimbe
- Howard Hughes Medical Institute, Department of Molecular Physiology and Biophysics, Department of Neurology, University of Iowa Roy J. and Lucille A. Carver College of Medicine, Iowa City, IA 52242 USA
| | - David P Venzke
- Howard Hughes Medical Institute, Department of Molecular Physiology and Biophysics, Department of Neurology, University of Iowa Roy J. and Lucille A. Carver College of Medicine, Iowa City, IA 52242 USA
| | - Mary E Anderson
- Howard Hughes Medical Institute, Department of Molecular Physiology and Biophysics, Department of Neurology, University of Iowa Roy J. and Lucille A. Carver College of Medicine, Iowa City, IA 52242 USA
| | - Jeffery M Hord
- Howard Hughes Medical Institute, Department of Molecular Physiology and Biophysics, Department of Neurology, University of Iowa Roy J. and Lucille A. Carver College of Medicine, Iowa City, IA 52242 USA
| | - Kevin P Campbell
- Howard Hughes Medical Institute, Department of Molecular Physiology and Biophysics, Department of Neurology, University of Iowa Roy J. and Lucille A. Carver College of Medicine, Iowa City, IA 52242 USA
| |
Collapse
|
9
|
Structural basis for matriglycan synthesis by the LARGE1 dual glycosyltransferase. PLoS One 2022; 17:e0278713. [PMID: 36512577 PMCID: PMC9746966 DOI: 10.1371/journal.pone.0278713] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2022] [Accepted: 11/21/2022] [Indexed: 12/15/2022] Open
Abstract
LARGE1 is a bifunctional glycosyltransferase responsible for generating a long linear polysaccharide termed matriglycan that links the cytoskeleton and the extracellular matrix and is required for proper muscle function. This matriglycan polymer is made with an alternating pattern of xylose and glucuronic acid monomers. Mutations in the LARGE1 gene have been shown to cause life-threatening dystroglycanopathies through the inhibition of matriglycan synthesis. Despite its major role in muscle maintenance, the structure of the LARGE1 enzyme and how it assembles in the Golgi are unknown. Here we present the structure of LARGE1, obtained by a combination of X-ray crystallography and single-particle cryo-EM. We found that LARGE1 homo-dimerizes in a configuration that is dictated by its coiled-coil stem domain. The structure shows that this enzyme has two canonical GT-A folds within each of its catalytic domains. In the context of its dimeric structure, the two types of catalytic domains are brought into close proximity from opposing monomers to allow efficient shuttling of the substrates between the two domains. Together, with putative retention of matriglycan by electrostatic interactions, this dimeric organization offers a possible mechanism for the ability of LARGE1 to synthesize long matriglycan chains. The structural information further reveals the mechanisms in which disease-causing mutations disrupt the activity of LARGE1. Collectively, these data shed light on how matriglycan is synthesized alongside the functional significance of glycosyltransferase oligomerization.
Collapse
|
10
|
Lallar M, Kaur L, Preet M, Singh UP. B4GAT1 Gene Associated Congenital Muscular Dystrophy Presenting with Recurrent Severe Ventriculomegaly: Case Report and Review of Literature. Fetal Pediatr Pathol 2022; 41:837-842. [PMID: 34587870 DOI: 10.1080/15513815.2021.1982091] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
BACKGROUND Congenital muscular dystrophies (CMD) range in phenotype from an antenatal presentation with brain and eye anomalies to isolated muscular weakness. B4GAT1 gene has recently been associated with muscular dystrophy-dystroglycanopathy, type A, 13 and two families have been reported. CASE REPORT We report the third family with B4GAT1 associated CMD presenting as recurrent severe ventriculomegaly, cerebellar and vermian hypoplasia in fetal life, which was identified after the second affected pregnancy. The mutations identified were similar to those reported in a previously reported Indian family, homozygous, p.Asn390Asp, and p. Ala406Val, suggesting founder mutation. CONCLUSION B4GAT1 mutations are associated with CMD and may present in fetal life as severe ventriculomegaly. The homozygous B4GAT1 mutations, p.Asn390Asp, and p. Ala406Val, described in two Indian families (including this case) might represent a founder mutation.
Collapse
Affiliation(s)
- Meenakshi Lallar
- Prime Institute of Prenatal Imaging and Diagnostics, Chandigarh, India
| | - Ladbans Kaur
- Prime Institute of Prenatal Imaging and Diagnostics, Chandigarh, India
| | - Meetan Preet
- Prime Institute of Prenatal Imaging and Diagnostics, Chandigarh, India
| | - U P Singh
- Prime Institute of Prenatal Imaging and Diagnostics, Chandigarh, India
| |
Collapse
|
11
|
Concerted Regulation of Glycosylation Factors Sustains Tissue Identity and Function. Biomedicines 2022; 10:biomedicines10081805. [PMID: 36009354 PMCID: PMC9404854 DOI: 10.3390/biomedicines10081805] [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: 05/31/2022] [Revised: 06/27/2022] [Accepted: 07/21/2022] [Indexed: 11/17/2022] Open
Abstract
Glycosylation is a fundamental cellular process affecting human development and health. Complex machinery establishes the glycan structures whose heterogeneity provides greater structural diversity than other post-translational modifications. Although known to present spatial and temporal diversity, the evolution of glycosylation and its role at the tissue-specific level is poorly understood. In this study, we combined genome and transcriptome profiles of healthy and diseased tissues to uncover novel insights into the complex role of glycosylation in humans. We constructed a catalogue of human glycosylation factors, including transferases, hydrolases and other genes directly involved in glycosylation. These were categorized as involved in N-, O- and lipid-linked glycosylation, glypiation, and glycosaminoglycan synthesis. Our data showed that these glycosylation factors constitute an ancient family of genes, where evolutionary constraints suppressed large gene duplications, except for genes involved in O-linked and lipid glycosylation. The transcriptome profiles of 30 healthy human tissues revealed tissue-specific expression patterns preserved across mammals. In addition, clusters of tightly co-expressed genes suggest a glycosylation code underlying tissue identity. Interestingly, several glycosylation factors showed tissue-specific profiles varying with age, suggesting a role in ageing-related disorders. In cancer, our analysis revealed that glycosylation factors are highly perturbed, at the genome and transcriptome levels, with a strong predominance of copy number alterations. Moreover, glycosylation factor dysregulation was associated with distinct cellular compositions of the tumor microenvironment, reinforcing the impact of glycosylation in modulating the immune system. Overall, this work provides genome-wide evidence that the glycosylation machinery is tightly regulated in healthy tissues and impaired in ageing and tumorigenesis, unveiling novel potential roles as prognostic biomarkers or therapeutic targets.
Collapse
|
12
|
Sheikh MO, Capicciotti CJ, Liu L, Praissman J, Ding D, Mead DG, Brindley MA, Willer T, Campbell KP, Moremen KW, Wells L, Boons GJ. Cell surface glycan engineering reveals that matriglycan alone can recapitulate dystroglycan binding and function. Nat Commun 2022; 13:3617. [PMID: 35750689 PMCID: PMC9232514 DOI: 10.1038/s41467-022-31205-7] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2021] [Accepted: 06/07/2022] [Indexed: 12/29/2022] Open
Abstract
α-Dystroglycan (α-DG) is uniquely modified on O-mannose sites by a repeating disaccharide (-Xylα1,3-GlcAβ1,3-)n termed matriglycan, which is a receptor for laminin-G domain-containing proteins and employed by old-world arenaviruses for infection. Using chemoenzymatically synthesized matriglycans printed as a microarray, we demonstrate length-dependent binding to Laminin, Lassa virus GP1, and the clinically-important antibody IIH6. Utilizing an enzymatic engineering approach, an N-linked glycoprotein was converted into a IIH6-positive Laminin-binding glycoprotein. Engineering of the surface of cells deficient for either α-DG or O-mannosylation with matriglycans of sufficient length recovers infection with a Lassa-pseudovirus. Finally, free matriglycan in a dose and length dependent manner inhibits viral infection of wildtype cells. These results indicate that matriglycan alone is necessary and sufficient for IIH6 staining, Laminin and LASV GP1 binding, and Lassa-pseudovirus infection and support a model in which it is a tunable receptor for which increasing chain length enhances ligand-binding capacity.
Collapse
Affiliation(s)
- M Osman Sheikh
- Complex Carbohydrate Research Center, University of Georgia, Athens, GA, USA
| | - Chantelle J Capicciotti
- Complex Carbohydrate Research Center, University of Georgia, Athens, GA, USA
- Departments of Chemistry, Biomedical and Molecular Sciences, and Surgery, Queen's University, Kingston, ON, Canada
| | - Lin Liu
- Complex Carbohydrate Research Center, University of Georgia, Athens, GA, USA
| | - Jeremy Praissman
- Complex Carbohydrate Research Center, University of Georgia, Athens, GA, USA
| | - Dahai Ding
- Complex Carbohydrate Research Center, University of Georgia, Athens, GA, USA
- Department of Chemistry, University of Georgia, Athens, GA, USA
| | - Daniel G Mead
- College of Veterinary Medicine, University of Georgia, Athens, GA, USA
| | | | - Tobias Willer
- Howard Hughes Medical Institute, Senator Paul D. Wellstone Muscular Dystrophy Specialized Research Center, Department of Molecular Physiology and Biophysics, The University of Iowa, Iowa City, IA, USA
- Department of Neurology, Roy J. and Lucille A. Carver College of Medicine, The University of Iowa, Iowa City, IA, USA
| | - Kevin P Campbell
- Howard Hughes Medical Institute, Senator Paul D. Wellstone Muscular Dystrophy Specialized Research Center, Department of Molecular Physiology and Biophysics, The University of Iowa, Iowa City, IA, USA
- Department of Neurology, Roy J. and Lucille A. Carver College of Medicine, The University of Iowa, Iowa City, IA, USA
| | - Kelley W Moremen
- Complex Carbohydrate Research Center, University of Georgia, Athens, GA, USA
- Department of Biochemistry & Molecular Biology, University of Georgia, Athens, GA, USA
| | - Lance Wells
- Complex Carbohydrate Research Center, University of Georgia, Athens, GA, USA.
- Department of Biochemistry & Molecular Biology, University of Georgia, Athens, GA, USA.
| | - Geert-Jan Boons
- Complex Carbohydrate Research Center, University of Georgia, Athens, GA, USA.
- Department of Chemistry, University of Georgia, Athens, GA, USA.
- Department of Chemical Biology and Drug Discovery, Utrecht University, Utrecht, The Netherlands.
| |
Collapse
|
13
|
Cancer Malignancy Is Correlated with Upregulation of PCYT2-Mediated Glycerol Phosphate Modification of α-Dystroglycan. Int J Mol Sci 2022; 23:ijms23126662. [PMID: 35743105 PMCID: PMC9223686 DOI: 10.3390/ijms23126662] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2022] [Revised: 06/09/2022] [Accepted: 06/11/2022] [Indexed: 12/10/2022] Open
Abstract
The dystrophin–glycoprotein complex connects the cytoskeleton with base membrane components such as laminin through unique O-glycans displayed on α-dystroglycan (α-DG). Genetic impairment of elongation of these glycans causes congenital muscular dystrophies. We previously identified that glycerol phosphate (GroP) can cap the core part of the α-DG O-glycans and terminate their further elongation. This study examined the possible roles of the GroP modification in cancer malignancy, focusing on colorectal cancer. We found that the GroP modification critically depends on PCYT2, which serves as cytidine 5′-diphosphate-glycerol (CDP-Gro) synthase. Furthermore, we identified a significant positive correlation between cancer progression and GroP modification, which also correlated positively with PCYT2 expression. Moreover, we demonstrate that GroP modification promotes the migration of cancer cells. Based on these findings, we propose that the GroP modification by PCYT2 disrupts the glycan-mediated cell adhesion to the extracellular matrix and thereby enhances cancer metastasis. Thus, the present study suggests the possibility of novel approaches for cancer treatment by targeting the PCYT2-mediated GroP modification.
Collapse
|
14
|
Tamura JI, Tamura T, Hoshino S, Imae R, Kato R, Yokono M, Nagase M, Ohno S, Manabe N, Yamaguchi Y, Manya H, Endo T. Chemical and Chemo-Enzymatic Syntheses of Glycans Containing Ribitol Phosphate Scaffolding of Matriglycan. ACS Chem Biol 2022; 17:1513-1523. [PMID: 35670527 DOI: 10.1021/acschembio.2c00181] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Ribitol phosphate modifications to the core M3 O-mannosyl glycan are important for the functional maturation of α-dystroglycan. Three sequentially extended partial structures of the core M3 O-mannosyl glycan including a tandem ribitol phosphate were regio- and stereo-selectively synthesized: Rbo5P-3GalNAcβ, Rbo5P-1Rbo5P-3GalNAcβ, and Xylβ1-4Rbo5P-1Rbo5P-3GalNAcβ (Rbo5P, d-ribitol-5-phosphate; GalNAc, N-acetyl-d-galactosamine; Xyl, d-xylose). Rbo5P-3GalNAcβ with p-nitrophenyl at the aglycon part served as a substrate for ribitol phosphate transferase (FKRP, fukutin-related protein), and its product was glycosylated by the actions of a series of glycosyltransferases, namely, ribitol xylosyltransferase 1 (RXYLT1), β1,4-glucuronyltransferase 1 (B4GAT1), and like-acetyl-glucosaminyltransferase (LARGE). Rbo5P-3GalNAcβ equipped with an alkyne-type aglycon was also active for FKRP. The molecular information obtained on FKRP suggests that Rbo5P-3GalNAcβ derivatives are the minimal units required as the acceptor glycan for Rbo5P transfer and may serve as a precursor for the elongation of the core M3 O-mannosyl glycan.
Collapse
Affiliation(s)
- Jun-Ichi Tamura
- The United Graduate School of Agricultural Sciences, Tottori University, Tottori 680-8553, Japan
| | - Takahiro Tamura
- The United Graduate School of Agricultural Sciences, Tottori University, Tottori 680-8553, Japan
| | - Shunsuke Hoshino
- Molecular Glycobiology, Research Team for Mechanism of Aging, Tokyo Metropolitan Geriatric Hospital and Institute of Gerontology, Itabashi-ku, Tokyo 173-0015, Japan
| | - Rieko Imae
- Molecular Glycobiology, Research Team for Mechanism of Aging, Tokyo Metropolitan Geriatric Hospital and Institute of Gerontology, Itabashi-ku, Tokyo 173-0015, Japan
| | - Ryuichi Kato
- Structural Biology Research Center, Institute of Material Structure Science, High Energy Accelerator Research Organization (KEK), Tsukuba 305-0801, Japan
| | - Mizuki Yokono
- Technical Department, Tottori University, Tottori 680-8550, Japan
| | - Mao Nagase
- Division of Structural Glycobiology, Institute of Molecular Biomembrane and Glycobiology, Tohoku Medical and Pharmaceutical University, Sendai 981-8558, Japan
| | - Shiho Ohno
- Division of Structural Glycobiology, Institute of Molecular Biomembrane and Glycobiology, Tohoku Medical and Pharmaceutical University, Sendai 981-8558, Japan
| | - Noriyoshi Manabe
- Division of Structural Glycobiology, Institute of Molecular Biomembrane and Glycobiology, Tohoku Medical and Pharmaceutical University, Sendai 981-8558, Japan
| | - Yoshiki Yamaguchi
- Division of Structural Glycobiology, Institute of Molecular Biomembrane and Glycobiology, Tohoku Medical and Pharmaceutical University, Sendai 981-8558, Japan
| | - Hiroshi Manya
- Molecular Glycobiology, Research Team for Mechanism of Aging, Tokyo Metropolitan Geriatric Hospital and Institute of Gerontology, Itabashi-ku, Tokyo 173-0015, Japan
| | - Tamao Endo
- Molecular Glycobiology, Research Team for Mechanism of Aging, Tokyo Metropolitan Geriatric Hospital and Institute of Gerontology, Itabashi-ku, Tokyo 173-0015, Japan
| |
Collapse
|
15
|
Boyd A, Montandon M, Wood AJ, Currie PD. FKRP directed fibronectin glycosylation: A novel mechanism giving insights into muscular dystrophies? Bioessays 2022; 44:e2100270. [PMID: 35229908 DOI: 10.1002/bies.202100270] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2021] [Revised: 02/12/2022] [Accepted: 02/16/2022] [Indexed: 12/15/2022]
Abstract
The recently uncovered role of Fukutin-related protein (FKRP) in fibronectin glycosylation has challenged our understanding of the basis of disease pathogenesis in the muscular dystrophies. FKRP is a Golgi-resident glycosyltransferase implicated in a broad spectrum of muscular dystrophy (MD) pathologies that are not fully attributable to the well-described α-Dystroglycan hypoglycosylation. By revealing a new role for FKRP in the glycosylation of fibronectin, a modification critical for the development of the muscle basement membrane (MBM) and its associated muscle linkages, new possibilities for understanding clinical phenotype arise. This modification involves an interaction between FKRP and myosin-10, a protein involved in the Golgi organization and function. These observations suggest a FKRP nexus exists that controls two critical aspects to muscle fibre integrity, both fibre stability at the MBM and its elastic properties. This review explores the new potential disease axis in the context of our current knowledge of muscular dystrophies.
Collapse
Affiliation(s)
- Andrew Boyd
- Australian Regenerative Medicine Institute, Monash University, Clayton, Victoria, Australia
| | - Margo Montandon
- Australian Regenerative Medicine Institute, Monash University, Clayton, Victoria, Australia
| | - Alasdair J Wood
- Australian Regenerative Medicine Institute, Monash University, Clayton, Victoria, Australia
| | - Peter D Currie
- Australian Regenerative Medicine Institute, Monash University, Clayton, Victoria, Australia.,EMBL Australia, Monash University, Clayton, Victoria, Australia
| |
Collapse
|
16
|
Hang J, Wang J, Lu M, Xue Y, Qiao J, Tao L. Protein O-mannosylation across kingdoms and related diseases: From glycobiology to glycopathology. Biomed Pharmacother 2022; 148:112685. [PMID: 35149389 DOI: 10.1016/j.biopha.2022.112685] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2021] [Revised: 01/29/2022] [Accepted: 02/01/2022] [Indexed: 11/18/2022] Open
Abstract
The post-translational glycosylation of proteins by O-linked α-mannose is conserved from bacteria to humans. Due to advances in high-throughput mass spectrometry-based approaches, a variety of glycoproteins are identified to be O-mannosylated. Various proteins with O-mannosylation are involved in biological processes, providing essential necessity for proper growth and development. In this review, we summarize the process and regulation of O-mannosylation. The multi-step O-mannosylation procedures are quite dynamic and complex, especially when considering the structural and functional inspection of the involved enzymes. The widely studied O-mannosylated proteins in human include α-Dystroglycan (α-DG), cadherins, protocadherins, and plexin, and their aberrant O-mannosylation are associated with many diseases. In addition, O-mannosylation also contributes to diverse functions in lower eukaryotes and prokaryotes. Finally, we present the relationship between O-mannosylation and gut microbiota (GM), and elucidate that O-mannosylation in microbiome is of great importance in the dynamic balance of GM. Our study provides an overview of the processes of O-mannosylation in mammalian cells and other organisms, and also associated regulated enzymes and biological functions, which could contribute to the understanding of newly discovered O-mannosylated glycoproteins.
Collapse
Affiliation(s)
- Jing Hang
- Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing 100191, China; National Clinical Research Center for Obstetrics and Gynecology (Peking University Third Hospital), Beijing 100191, China; Key Laboratory of Assisted Reproduction (Peking University), Ministry of Education, Beijing 100191, China; Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology, Beijing 100191, China
| | - Jinpeng Wang
- Department of Orthopedics, First Hospital of China Medical University, Shenyang 110001, China
| | - Minzhen Lu
- Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing 100191, China; National Clinical Research Center for Obstetrics and Gynecology (Peking University Third Hospital), Beijing 100191, China; Key Laboratory of Assisted Reproduction (Peking University), Ministry of Education, Beijing 100191, China; Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology, Beijing 100191, China
| | - Yuchuan Xue
- The First Department of Clinical Medicine, China Medical University, Shenyang 110001, China
| | - Jie Qiao
- Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing 100191, China; National Clinical Research Center for Obstetrics and Gynecology (Peking University Third Hospital), Beijing 100191, China; Key Laboratory of Assisted Reproduction (Peking University), Ministry of Education, Beijing 100191, China; Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology, Beijing 100191, China.
| | - Lin Tao
- Department of Orthopedics, First Hospital of China Medical University, Shenyang 110001, China.
| |
Collapse
|
17
|
Tamura T, Omura Y, Kotera K, Ito R, Ohno S, Manabe N, Yamaguchi Y, Tamura JI. Synthesis of the matriglycan hexasaccharide, -3Xylα1-3GlcAβ1-trimer and its interaction with laminin. Org Biomol Chem 2022; 20:8489-8500. [DOI: 10.1039/d2ob01388f] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
Matriglycan hexasaccharide (-3Xylα1-3GlcAβ1)3-O(C2H4O)3CH2CCH and the biotin conjugate was synthesized. The hexasaccharide showed good interaction with laminin-G-like domains 4 and 5 of laminin-α2 using saturation transfer difference-NMR and bio-layer interferometry.
Collapse
Affiliation(s)
- Takahiro Tamura
- The United Graduate School of Agricultural Sciences, Tottori University, Tottori, 680-8553 Japan
| | - Yuka Omura
- Department of Agricultural, life and Environmental Sciences, Faculty of Agriculture, Tottori University, Tottori, 680-8553 Japan
| | - Kota Kotera
- Department of Agricultural, life and Environmental Sciences, Faculty of Agriculture, Tottori University, Tottori, 680-8553 Japan
| | - Ryota Ito
- Division of Structural Glycobiology, Institute of Molecular Biomembrane and Glycobiology, Tohoku Medical and Pharmaceutical University, Sendai, 981-8558 Japan
| | - Shiho Ohno
- Division of Structural Glycobiology, Institute of Molecular Biomembrane and Glycobiology, Tohoku Medical and Pharmaceutical University, Sendai, 981-8558 Japan
| | - Noriyoshi Manabe
- Division of Structural Glycobiology, Institute of Molecular Biomembrane and Glycobiology, Tohoku Medical and Pharmaceutical University, Sendai, 981-8558 Japan
| | - Yoshiki Yamaguchi
- Division of Structural Glycobiology, Institute of Molecular Biomembrane and Glycobiology, Tohoku Medical and Pharmaceutical University, Sendai, 981-8558 Japan
| | - Jun-ichi Tamura
- The United Graduate School of Agricultural Sciences, Tottori University, Tottori, 680-8553 Japan
- Department of Agricultural, life and Environmental Sciences, Faculty of Agriculture, Tottori University, Tottori, 680-8553 Japan
| |
Collapse
|
18
|
Ortiz-Cordero C, Bincoletto C, Dhoke NR, Selvaraj S, Magli A, Zhou H, Kim DH, Bang AG, Perlingeiro RCR. Defective autophagy and increased apoptosis contribute toward the pathogenesis of FKRP-associated muscular dystrophies. Stem Cell Reports 2021; 16:2752-2767. [PMID: 34653404 PMCID: PMC8581053 DOI: 10.1016/j.stemcr.2021.09.009] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2021] [Revised: 09/16/2021] [Accepted: 09/17/2021] [Indexed: 12/15/2022] Open
Abstract
Fukutin-related protein (FKRP) is a glycosyltransferase involved in glycosylation of alpha-dystroglycan (α-DG). Mutations in FKRP are associated with muscular dystrophies (MD) ranging from limb-girdle LGMDR9 to Walker-Warburg Syndrome (WWS), a severe type of congenital MD. Although hypoglycosylation of α-DG is the main hallmark of this group of diseases, a full understanding of the underlying pathophysiology is still missing. Here, we investigated molecular mechanisms impaired by FKRP mutations in pluripotent stem (PS) cell–derived myotubes. FKRP-deficient myotubes show transcriptome alterations in genes involved in extracellular matrix receptor interactions, calcium signaling, PI3K-Akt pathway, and lysosomal function. Accordingly, using a panel of patient-specific LGMDR9 and WWS induced PS cell–derived myotubes, we found a significant reduction in the autophagy-lysosome pathway for both disease phenotypes. In addition, we show that WWS myotubes display decreased ERK1/2 activity and increased apoptosis, which were restored in gene edited myotubes. Our results suggest the autophagy-lysosome pathway and apoptosis may contribute to the FKRP-associated MD pathogenesis. The lysosome pathway is deregulated in FKRP-deficient myotubes Autophagy is decreased in patient-specific LGMDR9 and WWS iPS cell–derived myotubes FKRP WWS and LGMDR9 iPS cell–derived myotubes have increased apoptosis FKRP correction in WWS myotubes rescues changes in autophagy and apoptosis
Collapse
Affiliation(s)
- Carolina Ortiz-Cordero
- Lillehei Heart Institute, Department of Medicine, University of Minnesota, 4-128 CCRB, 2231 6th St. SE, Minneapolis, MN 55455, USA; Department of Integrative Biology and Physiology, University of Minnesota, Minneapolis, MN, USA
| | - Claudia Bincoletto
- Lillehei Heart Institute, Department of Medicine, University of Minnesota, 4-128 CCRB, 2231 6th St. SE, Minneapolis, MN 55455, USA; Departamento de Farmacologia, Escola Paulista de Medicina (EPM), Universidade Federal de São Paulo (UNIFESP), São Paulo, Brazil
| | - Neha R Dhoke
- Lillehei Heart Institute, Department of Medicine, University of Minnesota, 4-128 CCRB, 2231 6th St. SE, Minneapolis, MN 55455, USA
| | - Sridhar Selvaraj
- Lillehei Heart Institute, Department of Medicine, University of Minnesota, 4-128 CCRB, 2231 6th St. SE, Minneapolis, MN 55455, USA
| | - Alessandro Magli
- Lillehei Heart Institute, Department of Medicine, University of Minnesota, 4-128 CCRB, 2231 6th St. SE, Minneapolis, MN 55455, USA
| | - Haowen Zhou
- Conrad Prebys Center for Chemical Genomics, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, USA
| | - Do-Hyung Kim
- Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota, Minneapolis, MN, USA
| | - Anne G Bang
- Conrad Prebys Center for Chemical Genomics, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, USA
| | - Rita C R Perlingeiro
- Lillehei Heart Institute, Department of Medicine, University of Minnesota, 4-128 CCRB, 2231 6th St. SE, Minneapolis, MN 55455, USA; Department of Integrative Biology and Physiology, University of Minnesota, Minneapolis, MN, USA.
| |
Collapse
|
19
|
Biosynthetic Mechanisms and Biological Significance of Glycerol Phosphate-Containing Glycan in Mammals. Molecules 2021; 26:molecules26216675. [PMID: 34771084 PMCID: PMC8587909 DOI: 10.3390/molecules26216675] [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: 10/01/2021] [Revised: 11/01/2021] [Accepted: 11/02/2021] [Indexed: 11/25/2022] Open
Abstract
Bacteria contain glycerol phosphate (GroP)-containing glycans, which are important constituents of cell-surface glycopolymers such as the teichoic acids of Gram-positive bacterial cell walls. These glycopolymers comprising GroP play crucial roles in bacterial physiology and virulence. Recently, the first identification of a GroP-containing glycan in mammals was reported as a variant form of O-mannosyl glycan on α-dystroglycan (α-DG). However, the biological significance of such GroP modification remains largely unknown. In this review, we provide an overview of this new discovery of GroP-containing glycan in mammals and then outline the recent progress in elucidating the biosynthetic mechanisms of GroP-containing glycans on α-DG. In addition, we discuss the potential biological role of GroP modification along with the challenges and prospects for further research. The progress in this newly identified glycan modification will provide insights into the phylogenetic implications of glycan.
Collapse
|
20
|
Bigotti MG, Brancaccio A. High degree of conservation of the enzymes synthesizing the laminin-binding glycoepitope of α-dystroglycan. Open Biol 2021; 11:210104. [PMID: 34582712 PMCID: PMC8478517 DOI: 10.1098/rsob.210104] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
The dystroglycan (DG) complex plays a pivotal role for the stabilization of muscles in Metazoa. It is formed by two subunits, extracellular α-DG and transmembrane β-DG, originating from a unique precursor via a complex post-translational maturation process. The α-DG subunit is extensively glycosylated in sequential steps by several specific enzymes and employs such glycan scaffold to tightly bind basement membrane molecules. Mutations of several of these enzymes cause an alteration of the carbohydrate structure of α-DG, resulting in severe neuromuscular disorders collectively named dystroglycanopathies. Given the fundamental role played by DG in muscle stability, it is biochemically and clinically relevant to investigate these post-translational modifying enzymes from an evolutionary perspective. A first phylogenetic history of the thirteen enzymes involved in the fabrication of the so-called 'M3 core' laminin-binding epitope has been traced by an overall sequence comparison approach, and interesting details on the primordial enzyme set have emerged, as well as substantial conservation in Metazoa. The optimization along with the evolution of a well-conserved enzymatic set responsible for the glycosylation of α-DG indicate the importance of the glycosylation shell in modulating the connection between sarcolemma and surrounding basement membranes to increase skeletal muscle stability, and eventually support movement and locomotion.
Collapse
Affiliation(s)
- Maria Giulia Bigotti
- School of Translational Health Sciences, Research Floor Level 7, Bristol Royal Infirmary, Upper Maudlin Street, Bristol BS2 8HW, UK,School of Biochemistry, University Walk, University of Bristol, Bristol BS8 1TD, UK
| | - Andrea Brancaccio
- School of Biochemistry, University Walk, University of Bristol, Bristol BS8 1TD, UK,Institute of Chemical Sciences and Technologies ‘Giulio Natta’ (SCITEC) - CNR, Largo F.Vito 1, 00168, Rome, Italy
| |
Collapse
|
21
|
Imae R, Manya H, Tsumoto H, Miura Y, Endo T. PCYT2 synthesizes CDP-glycerol in mammals and reduced PCYT2 enhances the expression of functionally glycosylated α-dystroglycan. J Biochem 2021; 170:183-194. [PMID: 34255834 DOI: 10.1093/jb/mvab069] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2021] [Accepted: 05/31/2021] [Indexed: 11/14/2022] Open
Abstract
α-Dystroglycan (α-DG) is a highly glycosylated cell-surface protein. Defective O-mannosyl glycan on α-DG is associated with muscular dystrophies and cancer. In the biosynthetic pathway of the O-mannosyl glycan, fukutin (FKTN) and fukutin-related protein (FKRP) transfer ribitol phosphate (RboP). Previously, we reported that FKTN and FKRP can also transfer glycerol phosphate (GroP) from CDP-glycerol (CDP-Gro) and showed the inhibitory effects of CDP-Gro on functional glycan synthesis by preventing glycan elongation in vitro. However, whether mammalian cells have CDP-Gro or associated synthetic machinery has not been elucidated. Therefore, the function of CDP-Gro in mammals is largely unknown. Here, we reveal that cultured human cells and mouse tissues contain CDP-Gro using liquid chromatography tandem-mass spectrometry (LC-MS/MS). By performing the enzyme activity assay of candidate recombinant proteins, we found that ethanolamine-phosphate cytidylyltransferase (PCYT2), the key enzyme in de novo phosphatidylethanolamine biosynthesis, has CDP-Gro synthetic activity from glycerol-3-phosphate (Gro3P) and CTP. In addition, knockdown of PCYT2 dramatically reduced cellular CDP-Gro. These results indicate that PCYT2 is a CDP-Gro synthase in mammals. Furthermore, we found that the expression of functionally glycosylated α-DG is increased by reducing PCYT2 expression. Our results suggest an important role for CDP-Gro in the regulation of α-DG function in mammals.
Collapse
Affiliation(s)
| | | | - Hiroki Tsumoto
- Proteome Research, Research Team for Mechanism of Aging, Tokyo Metropolitan Geriatric Hospital and Institute of Gerontology, Tokyo, 173-0015, Japan
| | - Yuri Miura
- Proteome Research, Research Team for Mechanism of Aging, Tokyo Metropolitan Geriatric Hospital and Institute of Gerontology, Tokyo, 173-0015, Japan
| | | |
Collapse
|
22
|
Imae R, Kuwabara N, Manya H, Tanaka T, Tsuyuguchi M, Mizuno M, Endo T, Kato R. The structure of POMGNT2 provides new insights into the mechanism to determine the functional O-mannosylation site on α-dystroglycan. Genes Cells 2021; 26:485-494. [PMID: 33893702 PMCID: PMC8360118 DOI: 10.1111/gtc.12853] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2021] [Revised: 04/15/2021] [Accepted: 04/17/2021] [Indexed: 12/12/2022]
Abstract
Defects in the O‐mannosyl glycan of α‐dystroglycan (α‐DG) are associated with α‐dystroglycanopathy, a group of congenital muscular dystrophies. While α‐DG has many O‐mannosylation sites, only the specific positions can be modified with the functional O‐mannosyl glycan, namely, core M3‐type glycan. POMGNT2 is a glycosyltransferase which adds β1,4‐linked GlcNAc to the O‐mannose (Man) residue to acquire core M3‐type glycan. Although it is assumed that POMGNT2 extends the specific O‐Man residues around particular amino acid sequences, the details are not well understood. Here, we determined a series of crystal structures of POMGNT2 with and without the acceptor O‐mannosyl peptides and identified the critical interactions between POMGNT2 and the acceptor peptide. POMGNT2 has an N‐terminal catalytic domain and a C‐terminal fibronectin type III (FnIII) domain and forms a dimer. The acceptor peptide is sandwiched between the two protomers. The catalytic domain of one protomer recognizes the O‐mannosylation site (TPT motif), and the FnIII domain of the other protomer recognizes the C‐terminal region of the peptide. Structure‐based mutational studies confirmed that amino acid residues of the catalytic domain interacting with mannose or the TPT motif are essential for POMGNT2 enzymatic activity. In addition, the FnIII domain is also essential for the activity and it interacts with the peptide mainly by hydrophobic interaction. Our study provides the first atomic‐resolution insights into specific acceptor recognition by the FnIII domain of POMGNT2. The catalytic mechanism of POMGNT2 is proposed based on the structure.
Collapse
Affiliation(s)
- Rieko Imae
- Molecular Glycobiology, Research Team for Mechanism of Aging, Tokyo Metropolitan Geriatric Hospital and Institute of Gerontology, Itabashi-ku, Japan
| | - Naoyuki Kuwabara
- High Energy Accelerator Research Organization (KEK), Institute of Materials Structure Science, Structural Biology Research Center, Tsukuba, Japan
| | - Hiroshi Manya
- Molecular Glycobiology, Research Team for Mechanism of Aging, Tokyo Metropolitan Geriatric Hospital and Institute of Gerontology, Itabashi-ku, Japan
| | - Tomohiro Tanaka
- Laboratory of Glyco-organic Chemistry, The Noguchi Institute, Itabashi-ku, Japan
| | - Masato Tsuyuguchi
- High Energy Accelerator Research Organization (KEK), Institute of Materials Structure Science, Structural Biology Research Center, Tsukuba, Japan
| | - Mamoru Mizuno
- Laboratory of Glyco-organic Chemistry, The Noguchi Institute, Itabashi-ku, Japan
| | - Tamao Endo
- Molecular Glycobiology, Research Team for Mechanism of Aging, Tokyo Metropolitan Geriatric Hospital and Institute of Gerontology, Itabashi-ku, Japan
| | - Ryuichi Kato
- High Energy Accelerator Research Organization (KEK), Institute of Materials Structure Science, Structural Biology Research Center, Tsukuba, Japan
| |
Collapse
|
23
|
The promiscuous binding pocket of SLC35A1 ensures redundant transport of CDP-ribitol to the Golgi. J Biol Chem 2021; 296:100789. [PMID: 34015330 PMCID: PMC8192872 DOI: 10.1016/j.jbc.2021.100789] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2021] [Revised: 05/06/2021] [Accepted: 05/13/2021] [Indexed: 01/23/2023] Open
Abstract
The glycoprotein α-dystroglycan helps to link the intracellular cytoskeleton to the extracellular matrix. A unique glycan structure attached to this protein is required for its interaction with extracellular matrix proteins such as laminin. Up to now, this is the only mammalian glycan known to contain ribitol phosphate groups. Enzymes in the Golgi apparatus use CDP-ribitol to incorporate ribitol phosphate into the glycan chain of α-dystroglycan. Since CDP-ribitol is synthesized in the cytoplasm, we hypothesized that an unknown transporter must be required for its import into the Golgi apparatus. We discovered that CDP-ribitol transport relies on the CMP-sialic acid transporter SLC35A1 and the transporter SLC35A4 in a redundant manner. These two transporters are closely related, but bulky residues in the predicted binding pocket of SLC35A4 limit its size. We hypothesized that the large binding pocket SLC35A1 might accommodate the bulky CMP-sialic acid and the smaller CDP-ribitol, whereas SLC35A4 might only accept CDP-ribitol. To test this, we expressed SLC35A1 with mutations in its binding pocket in SLC35A1 KO cell lines. When we restricted the binding site of SLC35A1 by introducing the bulky residues present in SLC35A4, the mutant transporter was unable to support sialylation of proteins in cells but still supported ribitol phosphorylation. This demonstrates that the size of the binding pocket determines the substrate specificity of SLC35A1, allowing a variety of cytosine nucleotide conjugates to be transported. The redundancy with SLC35A4 also explains why patients with SLC35A1 mutations do not show symptoms of α-dystroglycan deficiency.
Collapse
|
24
|
Flynn RA, Pedram K, Malaker SA, Batista PJ, Smith BAH, Johnson AG, George BM, Majzoub K, Villalta PW, Carette JE, Bertozzi CR. Small RNAs are modified with N-glycans and displayed on the surface of living cells. Cell 2021; 184:3109-3124.e22. [PMID: 34004145 DOI: 10.1016/j.cell.2021.04.023] [Citation(s) in RCA: 294] [Impact Index Per Article: 73.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2020] [Revised: 12/18/2020] [Accepted: 04/14/2021] [Indexed: 12/20/2022]
Abstract
Glycans modify lipids and proteins to mediate inter- and intramolecular interactions across all domains of life. RNA is not thought to be a major target of glycosylation. Here, we challenge this view with evidence that mammals use RNA as a third scaffold for glycosylation. Using a battery of chemical and biochemical approaches, we found that conserved small noncoding RNAs bear sialylated glycans. These "glycoRNAs" were present in multiple cell types and mammalian species, in cultured cells, and in vivo. GlycoRNA assembly depends on canonical N-glycan biosynthetic machinery and results in structures enriched in sialic acid and fucose. Analysis of living cells revealed that the majority of glycoRNAs were present on the cell surface and can interact with anti-dsRNA antibodies and members of the Siglec receptor family. Collectively, these findings suggest the existence of a direct interface between RNA biology and glycobiology, and an expanded role for RNA in extracellular biology.
Collapse
Affiliation(s)
- Ryan A Flynn
- Department of Chemistry, Stanford University, Stanford, CA, USA.
| | - Kayvon Pedram
- Department of Chemistry, Stanford University, Stanford, CA, USA
| | - Stacy A Malaker
- Department of Chemistry, Stanford University, Stanford, CA, USA
| | - Pedro J Batista
- Laboratory of Cell Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Benjamin A H Smith
- Department of Chemical and Systems Biology and ChEM-H, Stanford University, Stanford, CA, USA
| | - Alex G Johnson
- Department of Chemical and Systems Biology, Stanford University, Stanford, CA, USA
| | - Benson M George
- Department of Cancer Biology, Stanford University, Stanford, CA, USA
| | - Karim Majzoub
- Department of Microbiology and Immunology, Stanford University, Stanford, CA, USA; IGMM, CNRS, University of Montpellier, Montpellier, France
| | - Peter W Villalta
- Masonic Cancer Center and Department of Medicinal Chemistry, University of Minnesota, Minneapolis, MN, USA
| | - Jan E Carette
- Department of Microbiology and Immunology, Stanford University, Stanford, CA, USA
| | - Carolyn R Bertozzi
- Department of Chemistry, Stanford University, Stanford, CA, USA; Howard Hughes Medical Institute, Stanford University, Stanford, CA, USA.
| |
Collapse
|
25
|
Yang JY, Halmo SM, Praissman J, Chapla D, Singh D, Wells L, Moremen KW, Lanzilotta WN. Crystal structures of β-1,4-N-acetylglucosaminyltransferase 2: structural basis for inherited muscular dystrophies. Acta Crystallogr D Struct Biol 2021; 77:486-495. [PMID: 33825709 PMCID: PMC8025878 DOI: 10.1107/s2059798321001261] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2020] [Accepted: 02/02/2021] [Indexed: 11/11/2022] Open
Abstract
The canonical O-mannosylation pathway in humans is essential for the functional glycosylation of α-dystroglycan. Disruption of this post-translational modification pathway leads to congenital muscular dystrophies. The first committed step in the construction of a functional matriglycan structure involves the post-translational modification of α-dystroglycan. This is essential for binding extracellular matrix proteins and arenaviruses, and is catalyzed by β-1,4-N-acetylglucosaminyltransferase 2 (POMGNT2). While another glycosyl transferase, β-1,4-N-acetylglucosaminyltransferase 1 (POMGNT1), has been shown to be promiscuous in extending O-mannosylated sites, POMGNT2 has been shown to display significant primary amino-acid selectivity near the site of O-mannosylation. Moreover, several single point mutations in POMGNT2 have been identified in patients with assorted dystroglycanopathies such as Walker-Warburg syndrome and limb girdle muscular dystrophy. To gain insight into POMGNT2 function in humans, the enzyme was expressed as a soluble, secreted fusion protein by transient infection of HEK293 suspension cultures. Here, crystal structures of POMGNT2 (amino-acid residues 25-580) with and without UDP bound are reported. Consistent with a novel fold and a unique domain organization, no molecular-replacement model was available and phases were obtained through crystallization of a selenomethionine variant of the enzyme in the same space group. Tetragonal (space group P4212; unit-cell parameters a = b = 129.8, c = 81.6 Å, α = γ = β = 90°) crystals with UDP bound diffracted to 1.98 Å resolution and contained a single monomer in the asymmetric unit. Orthorhombic (space group P212121; unit-cell parameters a = 142.3, b = 153.9, c = 187.4 Å, α = γ = β = 90°) crystals were also obtained; they diffracted to 2.57 Å resolution and contained four monomers with differential glycosylation patterns and conformations. These structures provide the first rational basis for an explanation of the loss-of-function mutations and offer significant insights into the mechanics of this important human enzyme.
Collapse
Affiliation(s)
- Jeong Yeh Yang
- The Complex Carbohydrate Research Center, University of Georgia, Athens, GA 30602, USA
| | - Stephanie M. Halmo
- The Complex Carbohydrate Research Center, University of Georgia, Athens, GA 30602, USA
| | - Jeremy Praissman
- The Complex Carbohydrate Research Center, University of Georgia, Athens, GA 30602, USA
| | - Digantkumar Chapla
- The Complex Carbohydrate Research Center, University of Georgia, Athens, GA 30602, USA
| | - Danish Singh
- The Complex Carbohydrate Research Center, University of Georgia, Athens, GA 30602, USA
| | - Lance Wells
- The Complex Carbohydrate Research Center, University of Georgia, Athens, GA 30602, USA
| | - Kelley W. Moremen
- The Complex Carbohydrate Research Center, University of Georgia, Athens, GA 30602, USA
| | - William N. Lanzilotta
- Department of Biochemistry and Molecular Biology, University of Georgia, Athens, GA 30602, USA
| |
Collapse
|
26
|
Ortiz-Cordero C, Magli A, Dhoke NR, Kuebler T, Selvaraj S, Oliveira NA, Zhou H, Sham YY, Bang AG, Perlingeiro RC. NAD+ enhances ribitol and ribose rescue of α-dystroglycan functional glycosylation in human FKRP-mutant myotubes. eLife 2021; 10:65443. [PMID: 33513091 PMCID: PMC7924940 DOI: 10.7554/elife.65443] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2020] [Accepted: 01/28/2021] [Indexed: 12/22/2022] Open
Abstract
Mutations in the fukutin-related protein (FKRP) cause Walker-Warburg syndrome (WWS), a severe form of congenital muscular dystrophy. Here, we established a WWS human induced pluripotent stem cell-derived myogenic model that recapitulates hallmarks of WWS pathology. We used this model to investigate the therapeutic effect of metabolites of the pentose phosphate pathway in human WWS. We show that functional recovery of WWS myotubes is promoted not only by ribitol but also by its precursor ribose. Moreover, we found that the combination of each of these metabolites with NAD+ results in a synergistic effect, as demonstrated by rescue of α-dystroglycan glycosylation and laminin binding capacity. Mechanistically, we found that FKRP residual enzymatic capacity, characteristic of many recessive FKRP mutations, is required for rescue as supported by functional and structural mutational analyses. These findings provide the rationale for testing ribose/ribitol in combination with NAD+ to treat WWS and other diseases associated with FKRP mutations.
Collapse
Affiliation(s)
- Carolina Ortiz-Cordero
- Department of Integrative Biology and Physiology, University of Minnesota, Minneapolis, United States.,Lillehei Heart Institute, Department of Medicine, University of Minnesota, Minneapolis, United States.,Stem Cell Institute, University of Minnesota, Minneapolis, United States
| | - Alessandro Magli
- Lillehei Heart Institute, Department of Medicine, University of Minnesota, Minneapolis, United States.,Stem Cell Institute, University of Minnesota, Minneapolis, United States
| | - Neha R Dhoke
- Lillehei Heart Institute, Department of Medicine, University of Minnesota, Minneapolis, United States
| | - Taylor Kuebler
- Bioinformatics and Computational Biology Program, University of Minnesota, Minneapolis, United States
| | - Sridhar Selvaraj
- Lillehei Heart Institute, Department of Medicine, University of Minnesota, Minneapolis, United States
| | - Nelio Aj Oliveira
- Lillehei Heart Institute, Department of Medicine, University of Minnesota, Minneapolis, United States
| | - Haowen Zhou
- Conrad Prebys Center for Chemical Genomics, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, United States
| | - Yuk Y Sham
- Department of Integrative Biology and Physiology, University of Minnesota, Minneapolis, United States.,Bioinformatics and Computational Biology Program, University of Minnesota, Minneapolis, United States
| | - Anne G Bang
- Conrad Prebys Center for Chemical Genomics, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, United States
| | - Rita Cr Perlingeiro
- Department of Integrative Biology and Physiology, University of Minnesota, Minneapolis, United States.,Lillehei Heart Institute, Department of Medicine, University of Minnesota, Minneapolis, United States.,Stem Cell Institute, University of Minnesota, Minneapolis, United States
| |
Collapse
|
27
|
Salinas-Marín R, Villanueva-Cabello TM, Martínez-Duncker I. Biology of Proteoglycans and Associated Glycosaminoglycans. COMPREHENSIVE GLYCOSCIENCE 2021:63-102. [DOI: 10.1016/b978-0-12-819475-1.00065-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2025]
|
28
|
Kadirvelraj R, Yang JY, Kim HW, Sanders JH, Moremen KW, Wood ZA. Comparison of human poly-N-acetyl-lactosamine synthase structure with GT-A fold glycosyltransferases supports a modular assembly of catalytic subsites. J Biol Chem 2021; 296:100110. [PMID: 33229435 PMCID: PMC7948508 DOI: 10.1074/jbc.ra120.015305] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2020] [Revised: 11/19/2020] [Accepted: 11/23/2020] [Indexed: 01/05/2023] Open
Abstract
Poly-N-acetyl-lactosamine (poly-LacNAc) structures are composed of repeating [-Galβ(1,4)-GlcNAcβ(1,3)-]n glycan extensions. They are found on both N- and O-glycoproteins and glycolipids and play an important role in development, immune function, and human disease. The majority of mammalian poly-LacNAc is synthesized by the alternating iterative action of β1,3-N-acetylglucosaminyltransferase 2 (B3GNT2) and β1,4-galactosyltransferases. B3GNT2 is in the largest mammalian glycosyltransferase family, GT31, but little is known about the structure, substrate recognition, or catalysis by family members. Here we report the structures of human B3GNT2 in complex with UDP:Mg2+ and in complex with both UDP:Mg2+ and a glycan acceptor, lacto-N-neotetraose. The B3GNT2 structure conserves the GT-A fold and the DxD motif that coordinates a Mg2+ ion for binding the UDP-GlcNAc sugar donor. The acceptor complex shows interactions with only the terminal Galβ(1,4)-GlcNAcβ(1,3)- disaccharide unit, which likely explains the specificity for both N- and O-glycan acceptors. Modeling of the UDP-GlcNAc donor supports a direct displacement inverting catalytic mechanism. Comparative structural analysis indicates that nucleotide sugar donors for GT-A fold glycosyltransferases bind in similar positions and conformations without conserving interacting residues, even for enzymes that use the same donor substrate. In contrast, the B3GNT2 acceptor binding site is consistent with prior models suggesting that the evolution of acceptor specificity involves loops inserted into the stable GT-A fold. These observations support the hypothesis that GT-A fold glycosyltransferases employ coevolving donor, acceptor, and catalytic subsite modules as templates to achieve the complex diversity of glycan linkages in biological systems.
Collapse
Affiliation(s)
- Renuka Kadirvelraj
- Department of Biochemistry & Molecular Biology, University of Georgia, Athens, Georgia, USA
| | - Jeong-Yeh Yang
- Complex Carbohydrate Research Center, University of Georgia, Athens, Georgia, USA
| | - Hyun W Kim
- Department of Biochemistry & Molecular Biology, University of Georgia, Athens, Georgia, USA
| | - Justin H Sanders
- Department of Biochemistry & Molecular Biology, University of Georgia, Athens, Georgia, USA
| | - Kelley W Moremen
- Department of Biochemistry & Molecular Biology, University of Georgia, Athens, Georgia, USA; Complex Carbohydrate Research Center, University of Georgia, Athens, Georgia, USA.
| | - Zachary A Wood
- Department of Biochemistry & Molecular Biology, University of Georgia, Athens, Georgia, USA.
| |
Collapse
|
29
|
Ortiz-Cordero C, Azzag K, Perlingeiro RCR. Fukutin-Related Protein: From Pathology to Treatments. Trends Cell Biol 2020; 31:197-210. [PMID: 33272829 DOI: 10.1016/j.tcb.2020.11.003] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2020] [Revised: 11/02/2020] [Accepted: 11/04/2020] [Indexed: 12/27/2022]
Abstract
Fukutin-related protein (FKRP) is a glycosyltransferase involved in the functional glycosylation of α-dystroglycan (DG), a key component in the link between the cytoskeleton and the extracellular matrix (ECM). Mutations in FKRP lead to dystroglycanopathies with broad severity, including limb-girdle and congenital muscular dystrophy. Studies over the past 5 years have elucidated the function of FKRP, which has expanded the number of therapeutic opportunities for patients carrying FKRP mutations. These include small molecules, gene delivery, and cell therapy. Here we summarize recent findings on the function of FKRP and describe available models for studying diseases and testing therapeutics. Lastly, we highlight preclinical studies that hold potential for the treatment of FKRP-associated dystroglycanopathies.
Collapse
Affiliation(s)
- Carolina Ortiz-Cordero
- Lillehei Heart Institute, Department of Medicine, University of Minnesota, Minneapolis, MN, USA; Department of Integrative Biology and Physiology, University of Minnesota, Minneapolis, MN, USA
| | - Karim Azzag
- Lillehei Heart Institute, Department of Medicine, University of Minnesota, Minneapolis, MN, USA
| | - Rita C R Perlingeiro
- Lillehei Heart Institute, Department of Medicine, University of Minnesota, Minneapolis, MN, USA; Department of Integrative Biology and Physiology, University of Minnesota, Minneapolis, MN, USA; Stem Cell Institute, University of Minnesota, Minneapolis, MN, USA.
| |
Collapse
|
30
|
Imae R, Kuwabara N, Manya H, Kato R, Endo T. Biosynthetic Mechanisms of a Unique Ribitol Phosphate-containing Glycan by FKRP, a Ribitol Phosphate Transferase. TRENDS GLYCOSCI GLYC 2020. [DOI: 10.4052/tigg.2008.1j] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Affiliation(s)
- Rieko Imae
- Molecular Glycobiology, Research Team for Mechanism of Aging, Tokyo Metropolitan Geriatric Hospital and Institute of Gerontology
| | - Naoyuki Kuwabara
- Structural Biology Research Center, Institute of Materials Structure Science, High Energy Accelerator Research Organization (KEK)
| | - Hiroshi Manya
- Molecular Glycobiology, Research Team for Mechanism of Aging, Tokyo Metropolitan Geriatric Hospital and Institute of Gerontology
| | - Ryuichi Kato
- Structural Biology Research Center, Institute of Materials Structure Science, High Energy Accelerator Research Organization (KEK)
| | - Tamao Endo
- Molecular Glycobiology, Research Team for Mechanism of Aging, Tokyo Metropolitan Geriatric Hospital and Institute of Gerontology
| |
Collapse
|
31
|
Imae R, Kuwabara N, Manya H, Kato R, Endo T. Biosynthetic Mechanisms of a Unique Ribitol Phosphate-containing Glycan by FKRP, a Ribitol Phosphate Transferase. TRENDS GLYCOSCI GLYC 2020. [DOI: 10.4052/tigg.2008.1e] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Affiliation(s)
- Rieko Imae
- Molecular Glycobiology, Research Team for Mechanism of Aging, Tokyo Metropolitan Geriatric Hospital and Institute of Gerontology
| | - Naoyuki Kuwabara
- Structural Biology Research Center, Institute of Materials Structure Science, High Energy Accelerator Research Organization (KEK)
| | - Hiroshi Manya
- Molecular Glycobiology, Research Team for Mechanism of Aging, Tokyo Metropolitan Geriatric Hospital and Institute of Gerontology
| | - Ryuichi Kato
- Structural Biology Research Center, Institute of Materials Structure Science, High Energy Accelerator Research Organization (KEK)
| | - Tamao Endo
- Molecular Glycobiology, Research Team for Mechanism of Aging, Tokyo Metropolitan Geriatric Hospital and Institute of Gerontology
| |
Collapse
|
32
|
Tamura T, Omura Y, Tamura JI. Stereo- and Regioselective Synthesis of O-Mannosyl Glycan Containing Matriglycan and a Part of Tandem Ribitol Phosphate. J Org Chem 2020; 85:12935-12946. [PMID: 32930586 DOI: 10.1021/acs.joc.0c01569] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
We herein successfully synthesized two pivotal structures of O-mannosyl glycan: (1) the matriglycan-repeating tetrasaccharide Xylα1-3GlcAβ1-3Xylα1-3GlcAβ and (2) the link between matriglycan and a part of tandem ribitol phosphate, Xylα1-3GlcAβ1-4Xylβ1-4Rbo, in a regio- and stereocontrolled manner. The disaccharide unit with the α-linkage of xylose was obtained by adopting the conformational fixation of the xylopyranoside ring and a specific solvation system of diastereoselective solubility.
Collapse
Affiliation(s)
- Takahiro Tamura
- The United Graduate School of Agricultural Sciences, Tottori University, Tottori 680-8553, Japan
| | - Yuka Omura
- Graduate School of Sustainability Science, Department of Agricultural Science, Tottori University, Tottori 680-8553, Japan
| | - Jun-Ichi Tamura
- The United Graduate School of Agricultural Sciences, Tottori University, Tottori 680-8553, Japan.,Graduate School of Sustainability Science, Department of Agricultural Science, Tottori University, Tottori 680-8553, Japan
| |
Collapse
|
33
|
Righino B, Bozzi M, Pirolli D, Sciandra F, Bigotti MG, Brancaccio A, De Rosa MC. Identification and Modeling of a GT-A Fold in the α-Dystroglycan Glycosylating Enzyme LARGE1. J Chem Inf Model 2020; 60:3145-3156. [PMID: 32356985 PMCID: PMC7340341 DOI: 10.1021/acs.jcim.0c00281] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
![]()
The
acetylglucosaminyltransferase-like protein LARGE1 is an enzyme
that is responsible for the final steps of the post-translational
modifications of dystroglycan (DG), a membrane receptor that links
the cytoskeleton with the extracellular matrix in the skeletal muscle
and in a variety of other tissues. LARGE1 acts by adding the repeating
disaccharide unit [-3Xyl-α1,3GlcAβ1-] to the extracellular
portion of the DG complex (α-DG); defects in the LARGE1 gene result in an aberrant glycosylation of α-DG and consequent
impairment of its binding to laminin, eventually affecting the connection
between the cell and the extracellular environment. In the skeletal
muscle, this leads to degeneration of the muscular tissue and muscular
dystrophy. So far, a few missense mutations have been identified within
the LARGE1 protein and linked to congenital muscular dystrophy, and
because no structural information is available on this enzyme, our
understanding of the molecular mechanisms underlying these pathologies
is still very limited. Here, we generated a 3D model structure of
the two catalytic domains of LARGE1, combining different molecular
modeling approaches. Furthermore, by using molecular dynamics simulations,
we analyzed the effect on the structure and stability of the first
catalytic domain of the pathological missense mutation S331F that
gives rise to a severe form of muscle–eye–brain disease.
Collapse
Affiliation(s)
- Benedetta Righino
- Dipartimento di Scienze Biotecnologiche di Base, Cliniche Intensivologiche e Perioperatorie, Università Cattolica del Sacro Cuore, L.go F. Vito 1, 00168 Rome, Italy
| | - Manuela Bozzi
- Dipartimento di Scienze Biotecnologiche di Base, Cliniche Intensivologiche e Perioperatorie, Università Cattolica del Sacro Cuore, L.go F. Vito 1, 00168 Rome, Italy.,Institute of Chemical Sciences and Technologies "Giulio Natta" (SCITEC)-CNR, L.go F. Vito 1, 00168 Rome, Italy
| | - Davide Pirolli
- Institute of Chemical Sciences and Technologies "Giulio Natta" (SCITEC)-CNR, L.go F. Vito 1, 00168 Rome, Italy
| | - Francesca Sciandra
- Institute of Chemical Sciences and Technologies "Giulio Natta" (SCITEC)-CNR, L.go F. Vito 1, 00168 Rome, Italy
| | - Maria Giulia Bigotti
- School of Translational Health Sciences, Research Floor Level 7, Bristol Royal Infirmary, Upper Maudlin Street, BS2 8HW Bristol, U.K.,School of Biochemistry, University Walk, University of Bristol, BS8 1TD Bristol, U.K
| | - Andrea Brancaccio
- Institute of Chemical Sciences and Technologies "Giulio Natta" (SCITEC)-CNR, L.go F. Vito 1, 00168 Rome, Italy.,School of Biochemistry, University Walk, University of Bristol, BS8 1TD Bristol, U.K
| | - Maria Cristina De Rosa
- Institute of Chemical Sciences and Technologies "Giulio Natta" (SCITEC)-CNR, L.go F. Vito 1, 00168 Rome, Italy
| |
Collapse
|
34
|
Ribitol enhances matriglycan of α-dystroglycan in breast cancer cells without affecting cell growth. Sci Rep 2020; 10:4935. [PMID: 32188898 PMCID: PMC7080755 DOI: 10.1038/s41598-020-61747-z] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2019] [Accepted: 02/25/2020] [Indexed: 11/24/2022] Open
Abstract
The laminin-binding glycan (matriglycan) on α-dystroglycan (α-DG) enables diverse roles, from neuronal development to muscle integrity. Reduction or loss of matriglycan has also been implicated in cancer development and metastasis, and specifically associated with high-grade tumors and poor prognoses in breast cancers. Hyperglycosylation of α-DG with LARGE overexpression is shown to inhibit cancer cell growth and tumorigenicity. We recently demonstrated that ribitol, considered to be a metabolic end-product, enhances matriglycan expression in dystrophic muscles in vivo. In the current study, we tested the hypothesis that ribitol could also enhance matriglycan expression in cancer cells. Our results showed for the first time that ribitol is able to significantly enhance the expression of matriglycan on α-DG in breast cancer cells. The ribitol effect is associated with an increase in levels of CDP-ribitol, the substrate for the ribitol-5-phosphate transferases FKRP and FKTN. Direct use of CDP-ribitol is also effective for matriglycan expression. Ribitol treatment does not alter the expression of FKRP, FKTN as well as LARGEs and ISPD which are critical for the synthesis of matriglycan. The results suggest that alteration in substrates could also be involved in regulation of matriglycan expression. Interestingly, expression of matriglycan is related to cell cycle progression with highest levels in S and G2 phases and ribitol treatment does not alter the pattern. Although matriglycan up-regulation does not affect cell cycle progression and proliferation of the cancer cells tested, the novel substrate-mediated treatment opens a new approach easily applicable to experimental systems in vivo for further exploitation of matriglycan expression in cancer progression and for therapeutic potential.
Collapse
|
35
|
Sheikh MO, Venzke D, Anderson ME, Yoshida-Moriguchi T, Glushka JN, Nairn AV, Galizzi M, Moremen KW, Campbell KP, Wells L. HNK-1 sulfotransferase modulates α-dystroglycan glycosylation by 3-O-sulfation of glucuronic acid on matriglycan. Glycobiology 2020; 30:817-829. [PMID: 32149355 DOI: 10.1093/glycob/cwaa024] [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: 07/14/2019] [Revised: 02/26/2020] [Accepted: 02/26/2020] [Indexed: 12/18/2022] Open
Abstract
Mutations in multiple genes required for proper O-mannosylation of α-dystroglycan are causal for congenital/limb-girdle muscular dystrophies and abnormal brain development in mammals. Previously, we and others further elucidated the functional O-mannose glycan structure that is terminated by matriglycan, [(-GlcA-β3-Xyl-α3-)n]. This repeating disaccharide serves as a receptor for proteins in the extracellular matrix. Here, we demonstrate in vitro that HNK-1 sulfotransferase (HNK-1ST/carbohydrate sulfotransferase) sulfates terminal glucuronyl residues of matriglycan at the 3-hydroxyl and prevents further matriglycan polymerization by the LARGE1 glycosyltransferase. While α-dystroglycan isolated from mouse heart and kidney is susceptible to exoglycosidase digestion of matriglycan, the functional, lower molecular weight α-dystroglycan detected in brain, where HNK-1ST expression is elevated, is resistant. Removal of the sulfate cap by a sulfatase facilitated dual-glycosidase digestion. Our data strongly support a tissue specific mechanism in which HNK-1ST regulates polymer length by competing with LARGE for the 3-position on the nonreducing GlcA of matriglycan.
Collapse
Affiliation(s)
- M Osman Sheikh
- Complex Carbohydrate Research Center, University of Georgia, Athens, GA 30602, USA
| | - David Venzke
- Department of Molecular Physiology and Biophysics, Department of Neurology, Howard Hughes Medical Institute, University of Iowa Roy J. and Lucille A. Carver College of Medicine, Iowa City, IA 52242, USA
| | - Mary E Anderson
- Department of Molecular Physiology and Biophysics, Department of Neurology, Howard Hughes Medical Institute, University of Iowa Roy J. and Lucille A. Carver College of Medicine, Iowa City, IA 52242, USA
| | - Takako Yoshida-Moriguchi
- Department of Molecular Physiology and Biophysics, Department of Neurology, Howard Hughes Medical Institute, University of Iowa Roy J. and Lucille A. Carver College of Medicine, Iowa City, IA 52242, USA
| | - John N Glushka
- Complex Carbohydrate Research Center, University of Georgia, Athens, GA 30602, USA
| | - Alison V Nairn
- Complex Carbohydrate Research Center, University of Georgia, Athens, GA 30602, USA
| | - Melina Galizzi
- Complex Carbohydrate Research Center, University of Georgia, Athens, GA 30602, USA
| | - Kelley W Moremen
- Complex Carbohydrate Research Center, University of Georgia, Athens, GA 30602, USA.,Department of Biochemistry and Molecular Biology, University of Georgia, Athens, GA 30602, USA
| | - Kevin P Campbell
- Department of Molecular Physiology and Biophysics, Department of Neurology, Howard Hughes Medical Institute, University of Iowa Roy J. and Lucille A. Carver College of Medicine, Iowa City, IA 52242, USA
| | - Lance Wells
- Complex Carbohydrate Research Center, University of Georgia, Athens, GA 30602, USA.,Department of Biochemistry and Molecular Biology, University of Georgia, Athens, GA 30602, USA
| |
Collapse
|
36
|
A Markov model of glycosylation elucidates isozyme specificity and glycosyltransferase interactions for glycoengineering. CURRENT RESEARCH IN BIOTECHNOLOGY 2020; 2:22-36. [PMID: 32285041 DOI: 10.1016/j.crbiot.2020.01.001] [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] [Indexed: 12/12/2022] Open
Abstract
Glycosylated biopharmaceuticals are important in the global pharmaceutical market. Despite the importance of their glycan structures, our limited knowledge of the glycosylation machinery still hinders controllability of this critical quality attribute. To facilitate discovery of glycosyltransferase specificity and predict glycoengineering efforts, here we extend the approach to model N-linked protein glycosylation as a Markov process. Our model leverages putative glycosyltransferase (GT) specificity to define the biosynthetic pathways for all measured glycans, and the Markov chain modelling is used to learn glycosyltransferase isoform activities and predict glycosylation following glycosyltransferase knock-in/knockout. We apply our methodology to four different glycoengineered therapeutics (i.e., Rituximab, erythropoietin, Enbrel, and alpha-1 antitrypsin) produced in CHO cells. Our model accurately predicted N-linked glycosylation following glycoengineering and further quantified the impact of glycosyltransferase mutations on reactions catalyzed by other glycosyltransferases. By applying these learned GT-GT interaction rules identified from single glycosyltransferase mutants, our model further predicts the outcome of multi-gene glycosyltransferase mutations on the diverse biotherapeutics. Thus, this modeling approach enables rational glycoengineering and the elucidation of relationships between glycosyltransferases, thereby facilitating biopharmaceutical research and aiding the broader study of glycosylation to elucidate the genetic basis of complex changes in glycosylation.
Collapse
|
37
|
Kuwabara N, Imae R, Manya H, Tanaka T, Mizuno M, Tsumoto H, Kanagawa M, Kobayashi K, Toda T, Senda T, Endo T, Kato R. Crystal structures of fukutin-related protein (FKRP), a ribitol-phosphate transferase related to muscular dystrophy. Nat Commun 2020; 11:303. [PMID: 31949166 PMCID: PMC6965139 DOI: 10.1038/s41467-019-14220-z] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2019] [Accepted: 12/10/2019] [Indexed: 12/16/2022] Open
Abstract
α-Dystroglycan (α-DG) is a highly-glycosylated surface membrane protein. Defects in the O-mannosyl glycan of α-DG cause dystroglycanopathy, a group of congenital muscular dystrophies. The core M3 O-mannosyl glycan contains tandem ribitol-phosphate (RboP), a characteristic feature first found in mammals. Fukutin and fukutin-related protein (FKRP), whose mutated genes underlie dystroglycanopathy, sequentially transfer RboP from cytidine diphosphate-ribitol (CDP-Rbo) to form a tandem RboP unit in the core M3 glycan. Here, we report a series of crystal structures of FKRP with and without donor (CDP-Rbo) and/or acceptor [RboP-(phospho-)core M3 peptide] substrates. FKRP has N-terminal stem and C-terminal catalytic domains, and forms a tetramer both in crystal and in solution. In the acceptor complex, the phosphate group of RboP is recognized by the catalytic domain of one subunit, and a phosphate group on O-mannose is recognized by the stem domain of another subunit. Structure-based functional studies confirmed that the dimeric structure is essential for FKRP enzymatic activity. Fukutin-related protein (FKRP) catalyses the addition of ribitol-phosphate (RboP) to the O-mannosyl glycan of α-dystroglycan and mutations in FKRP cause dystroglycanopathy. Here the authors provide insights into its oligomerization and recognition of the substrates, CDP-Rbo and the RboP-(phospho-)core M3 glycan, by determining the crystal structures of human FKRP.
Collapse
Affiliation(s)
- Naoyuki Kuwabara
- Structural Biology Research Center, Institute of Materials Structure Science, High Energy Accelerator Research Organization, Tsukuba, Ibaraki, 305-0801, Japan
| | - Rieko Imae
- Molecular Glycobiology, Research Team for Mechanism of Aging, Tokyo Metropolitan Geriatric Hospital and Institute of Gerontology, Itabashi-ku, Tokyo, 173-0015, Japan
| | - Hiroshi Manya
- Molecular Glycobiology, Research Team for Mechanism of Aging, Tokyo Metropolitan Geriatric Hospital and Institute of Gerontology, Itabashi-ku, Tokyo, 173-0015, Japan
| | - Tomohiro Tanaka
- Laboratory of Glyco-organic Chemistry, The Noguchi Institute, Itabashi-ku, Tokyo, 173-0003, Japan
| | - Mamoru Mizuno
- Laboratory of Glyco-organic Chemistry, The Noguchi Institute, Itabashi-ku, Tokyo, 173-0003, Japan
| | - Hiroki Tsumoto
- Proteome Research, Research Team for Mechanism of Aging, Tokyo Metropolitan Geriatric Hospital and Institute of Gerontology, Itabashi-ku, Tokyo, 173-0015, Japan
| | - Motoi Kanagawa
- Division of Molecular Brain Science, Kobe University Graduate School of Medicine, Kobe, Hyogo, 650-0017, Japan
| | - Kazuhiro Kobayashi
- Division of Molecular Brain Science, Kobe University Graduate School of Medicine, Kobe, Hyogo, 650-0017, Japan
| | - Tatsushi Toda
- Division of Molecular Brain Science, Kobe University Graduate School of Medicine, Kobe, Hyogo, 650-0017, Japan.,Department of Neurology, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8655, Japan
| | - Toshiya Senda
- Structural Biology Research Center, Institute of Materials Structure Science, High Energy Accelerator Research Organization, Tsukuba, Ibaraki, 305-0801, Japan.,School of High Energy Accelerator Science, SOKENDAI, Tsukuba, Ibaraki, 305-0801, Japan
| | - Tamao Endo
- Molecular Glycobiology, Research Team for Mechanism of Aging, Tokyo Metropolitan Geriatric Hospital and Institute of Gerontology, Itabashi-ku, Tokyo, 173-0015, Japan.
| | - Ryuichi Kato
- Structural Biology Research Center, Institute of Materials Structure Science, High Energy Accelerator Research Organization, Tsukuba, Ibaraki, 305-0801, Japan. .,School of High Energy Accelerator Science, SOKENDAI, Tsukuba, Ibaraki, 305-0801, Japan.
| |
Collapse
|
38
|
Kim J, Lana B, Torelli S, Ryan D, Catapano F, Ala P, Luft C, Stevens E, Konstantinidis E, Louzada S, Fu B, Paredes‐Redondo A, Chan AWE, Yang F, Stemple DL, Liu P, Ketteler R, Selwood DL, Muntoni F, Lin Y. A new patient-derived iPSC model for dystroglycanopathies validates a compound that increases glycosylation of α-dystroglycan. EMBO Rep 2019; 20:e47967. [PMID: 31566294 PMCID: PMC6832011 DOI: 10.15252/embr.201947967] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2019] [Revised: 08/24/2019] [Accepted: 08/29/2019] [Indexed: 12/24/2022] Open
Abstract
Dystroglycan, an extracellular matrix receptor, has essential functions in various tissues. Loss of α-dystroglycan-laminin interaction due to defective glycosylation of α-dystroglycan underlies a group of congenital muscular dystrophies often associated with brain malformations, referred to as dystroglycanopathies. The lack of isogenic human dystroglycanopathy cell models has limited our ability to test potential drugs in a human- and neural-specific context. Here, we generated induced pluripotent stem cells (iPSCs) from a severe dystroglycanopathy patient with homozygous FKRP (fukutin-related protein gene) mutation. We showed that CRISPR/Cas9-mediated gene correction of FKRP restored glycosylation of α-dystroglycan in iPSC-derived cortical neurons, whereas targeted gene mutation of FKRP in wild-type cells disrupted this glycosylation. In parallel, we screened 31,954 small molecule compounds using a mouse myoblast line for increased glycosylation of α-dystroglycan. Using human FKRP-iPSC-derived neural cells for hit validation, we demonstrated that compound 4-(4-bromophenyl)-6-ethylsulfanyl-2-oxo-3,4-dihydro-1H-pyridine-5-carbonitrile (4BPPNit) significantly augmented glycosylation of α-dystroglycan, in part through upregulation of LARGE1 glycosyltransferase gene expression. Together, isogenic human iPSC-derived cells represent a valuable platform for facilitating dystroglycanopathy drug discovery and therapeutic development.
Collapse
Affiliation(s)
- Jihee Kim
- Centre for Genomics and Child HealthBlizard Institute, Barts and the London School of Medicine and DentistryQueen Mary University of LondonLondonUK
- Stem Cell LaboratoryNational Bowel Research CentreBlizard Institute, Barts and the London School of Medicine and DentistryQueen Mary University of LondonLondonUK
| | - Beatrice Lana
- Centre for Genomics and Child HealthBlizard Institute, Barts and the London School of Medicine and DentistryQueen Mary University of LondonLondonUK
- Stem Cell LaboratoryNational Bowel Research CentreBlizard Institute, Barts and the London School of Medicine and DentistryQueen Mary University of LondonLondonUK
| | - Silvia Torelli
- UCL Great Ormond Street Institute of Child HealthLondonUK
| | - David Ryan
- Wellcome Sanger InstituteHinxtonCambridgeUK
| | | | - Pierpaolo Ala
- UCL Great Ormond Street Institute of Child HealthLondonUK
| | - Christin Luft
- MRC Laboratory for Molecular Cell BiologyUniversity College LondonLondonUK
| | | | - Evangelos Konstantinidis
- Centre for Genomics and Child HealthBlizard Institute, Barts and the London School of Medicine and DentistryQueen Mary University of LondonLondonUK
- Stem Cell LaboratoryNational Bowel Research CentreBlizard Institute, Barts and the London School of Medicine and DentistryQueen Mary University of LondonLondonUK
| | | | - Beiyuan Fu
- Wellcome Sanger InstituteHinxtonCambridgeUK
| | - Amaia Paredes‐Redondo
- Centre for Genomics and Child HealthBlizard Institute, Barts and the London School of Medicine and DentistryQueen Mary University of LondonLondonUK
- Stem Cell LaboratoryNational Bowel Research CentreBlizard Institute, Barts and the London School of Medicine and DentistryQueen Mary University of LondonLondonUK
| | - AW Edith Chan
- The Wolfson Institute for Biomedical ResearchUniversity College LondonLondonUK
| | | | | | - Pentao Liu
- Wellcome Sanger InstituteHinxtonCambridgeUK
| | - Robin Ketteler
- MRC Laboratory for Molecular Cell BiologyUniversity College LondonLondonUK
| | - David L Selwood
- The Wolfson Institute for Biomedical ResearchUniversity College LondonLondonUK
| | - Francesco Muntoni
- UCL Great Ormond Street Institute of Child HealthLondonUK
- NIHR Biomedical Research Centre at Great Ormond Street HospitalLondonUK
| | - Yung‐Yao Lin
- Centre for Genomics and Child HealthBlizard Institute, Barts and the London School of Medicine and DentistryQueen Mary University of LondonLondonUK
- Stem Cell LaboratoryNational Bowel Research CentreBlizard Institute, Barts and the London School of Medicine and DentistryQueen Mary University of LondonLondonUK
| |
Collapse
|
39
|
Kanagawa M, Toda T. Muscular Dystrophy with Ribitol-Phosphate Deficiency: A Novel Post-Translational Mechanism in Dystroglycanopathy. J Neuromuscul Dis 2019; 4:259-267. [PMID: 29081423 PMCID: PMC5701763 DOI: 10.3233/jnd-170255] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
Muscular dystrophy is a group of genetic disorders characterized by progressive muscle weakness. In the early 2000s, a new classification of muscular dystrophy, dystroglycanopathy, was established. Dystroglycanopathy often associates with abnormalities in the central nervous system. Currently, at least eighteen genes have been identified that are responsible for dystroglycanopathy, and despite its genetic heterogeneity, its common biochemical feature is abnormal glycosylation of alpha-dystroglycan. Abnormal glycosylation of alpha-dystroglycan reduces its binding activities to ligand proteins, including laminins. In just the last few years, remarkable progress has been made in determining the sugar chain structures and gene functions associated with dystroglycanopathy. The normal sugar chain contains tandem structures of ribitol-phosphate, a pentose alcohol that was previously unknown in humans. The dystroglycanopathy genes fukutin, fukutin-related protein (FKRP), and isoprenoid synthase domain-containing protein (ISPD) encode essential enzymes for the synthesis of this structure: fukutin and FKRP transfer ribitol-phosphate onto sugar chains of alpha-dystroglycan, and ISPD synthesizes CDP-ribitol, a donor substrate for fukutin and FKRP. These findings resolved long-standing questions and established a disease subgroup that is ribitol-phosphate deficient, which describes a large population of dystroglycanopathy patients. Here, we review the history of dystroglycanopathy, the properties of the sugar chain structure of alpha-dystroglycan, dystroglycanopathy gene functions, and therapeutic strategies.
Collapse
Affiliation(s)
- Motoi Kanagawa
- Division of Neurology/Molecular Brain Science, Kobe University Graduate School of Medicine, Chuo-ku, Kobe, Japan
| | - Tatsushi Toda
- Division of Neurology/Molecular Brain Science, Kobe University Graduate School of Medicine, Chuo-ku, Kobe, Japan.,Department of Neurology, Division of Neuroscience, Graduate School of Medicine, The University of Tokyo, Bunkyo-ku, Tokyo, Japan
| |
Collapse
|
40
|
Bardell D, Milner PI, Goljanek-Whysall K, Peffers MJ. Differences in plasma and peritoneal fluid proteomes identifies potential biomarkers associated with survival following strangulating small intestinal disease. Equine Vet J 2019; 51:727-732. [PMID: 30854696 DOI: 10.1111/evj.13094] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2018] [Accepted: 03/02/2019] [Indexed: 02/06/2023]
Abstract
BACKGROUND Strangulating small intestinal disease (SSID) carries a poor prognosis for survival in comparison to other types of colic, particularly if resection is required. Identification of markers which aid early diagnosis may prevent the need for resection, assist with more accurate prognostication and/or support the decision on whether surgical intervention is likely to be successful, would be of significant welfare benefit. OBJECTIVES To apply an unbiased methodology to investigate the plasma and peritoneal fluid proteomes in horses diagnosed with SSID requiring resection, to identify novel biomarkers which may be of diagnostic or prognostic value. STUDY DESIGN Prospective clinical study. METHODS Plasma and peritoneal fluid from horses presented with acute abdominal signs consistent with SSID was collected at initial clinical examination. Samples from eight horses diagnosed with SSID at surgery in which resection of affected bowel was performed and four control horses subjected to euthanasia for orthopaedic conditions were submitted for liquid chromatography tandem mass spectrometry. Protein expression profiles were determined using label-free quantification. Data were analysed using analysis of variance to identify differentially expressed proteins between control and all SSID horses and SSID horses which survived to hospital discharge and those which did not. Significance was assumed at P≤0.05. RESULTS A greater number of proteins were identified in peritoneal fluid than plasma of both SSID cases and controls, with 123 peritoneal fluid and 13 plasma proteins significantly differentially expressed (DE) between cases and controls (P<0.05, ≥2 fold change). Twelve peritoneal fluid proteins (P<0.036) and four plasma proteins (P<0.05) were significantly DE between SSID horses which survived and those which did not. MAIN LIMITATIONS A low number of samples were analysed, there was variation in duration and severity of SSID and only short-term outcome was considered. CONCLUSIONS Changes in peritoneal fluid proteome may provide a sensitive indicator of small intestinal strangulation and provide biomarkers relevant to prognosis.
Collapse
Affiliation(s)
- D Bardell
- Institute of Ageing and Chronic Disease, Department of Musculoskeletal Biology, University of Liverpool, Liverpool, UK.,Institute of Veterinary Science, University of Liverpool, Neston, Wirral, UK
| | - P I Milner
- Institute of Ageing and Chronic Disease, Department of Musculoskeletal Biology, University of Liverpool, Liverpool, UK.,Institute of Veterinary Science, University of Liverpool, Neston, Wirral, UK
| | - K Goljanek-Whysall
- Institute of Ageing and Chronic Disease, Department of Musculoskeletal Biology, University of Liverpool, Liverpool, UK
| | - M J Peffers
- Institute of Ageing and Chronic Disease, Department of Musculoskeletal Biology, University of Liverpool, Liverpool, UK
| |
Collapse
|
41
|
ENDO T. Mammalian O-mannosyl glycans: Biochemistry and glycopathology. PROCEEDINGS OF THE JAPAN ACADEMY. SERIES B, PHYSICAL AND BIOLOGICAL SCIENCES 2019; 95:39-51. [PMID: 30643095 PMCID: PMC6395781 DOI: 10.2183/pjab.95.004] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/05/2018] [Accepted: 11/05/2018] [Indexed: 05/20/2023]
Abstract
Glycosylation is an important posttranslational modification in mammals. The glycans of glycoproteins are classified into two groups, namely, N-glycans and O-glycans, according to their glycan-peptide linkage regions. Recently, O-mannosyl glycan, an O-glycan, has been shown to be important in muscle and brain development. A clear relationship between O-mannosyl glycans and the pathomechanisms of some congenital muscular dystrophies has been established in humans. Ribitol-5-phosphate is a newly identified glycan component in mammals, and its biosynthetic pathway has been elucidated. The discovery of new glycan structures and the identification of highly regulated mechanisms of glycan processing will help researchers to understand glycan functions and develop therapeutic strategies.
Collapse
Affiliation(s)
- Tamao ENDO
- Tokyo Metropolitan Institute of Gerontology, Tokyo, Japan
- Correspondence should be addressed: T. Endo, Tokyo Metropolitan Institute of Gerontology, 35-2 Sakaecho, Itabashi-ku, Tokyo 173-0015, Japan (e-mail: )
| |
Collapse
|
42
|
Vannoy CH, Blaeser A, Lu QL. Dystroglycanopathy Gene Therapy: Unlocking the Potential of Genetic Engineering. MUSCLE GENE THERAPY 2019:469-490. [DOI: 10.1007/978-3-030-03095-7_27] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2025]
|
43
|
Hohenester E. Laminin G-like domains: dystroglycan-specific lectins. Curr Opin Struct Biol 2018; 56:56-63. [PMID: 30530204 DOI: 10.1016/j.sbi.2018.11.007] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2018] [Revised: 11/23/2018] [Accepted: 11/23/2018] [Indexed: 01/31/2023]
Abstract
A unique O-mannose-linked glycan on the transmembrane protein dystroglycan binds a number of extracellular matrix proteins containing laminin G-like (LG) domains. The dystroglycan-matrix interaction is essential for muscle function: disrupted biosynthesis of the matrix-binding modification causes several forms of muscular dystrophy. The complete chemical structure of this modification has been deciphered in the past few years. We now know that LG domains bind to a glycosaminoglycan-like polysaccharide of [-3GlcAβ1,3Xylα1-] units, termed matriglycan, that is attached to a highly unusual heptasaccharide linker. X-ray crystallography has revealed the principles of Ca2+-dependent matriglycan binding by LG domains. In this review, the new structural insights are applied to the growing number of LG domain-containing proteins that bind dystroglycan. It is proposed that LG domains be recognised as 'D-type' lectins to indicate their conserved function in dystroglycan binding.
Collapse
Affiliation(s)
- Erhard Hohenester
- Department of Life Sciences, Imperial College London, London SW7 2AZ, United Kingdom.
| |
Collapse
|
44
|
Amos RA, Pattathil S, Yang JY, Atmodjo MA, Urbanowicz BR, Moremen KW, Mohnen D. A two-phase model for the non-processive biosynthesis of homogalacturonan polysaccharides by the GAUT1:GAUT7 complex. J Biol Chem 2018; 293:19047-19063. [PMID: 30327429 DOI: 10.1074/jbc.ra118.004463] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2018] [Revised: 10/08/2018] [Indexed: 11/06/2022] Open
Abstract
Homogalacturonan (HG) is a pectic glycan in the plant cell wall that contributes to plant growth and development and cell wall structure and function, and interacts with other glycans and proteoglycans in the wall. HG is synthesized by the galacturonosyltransferase (GAUT) gene family. Two members of this family, GAUT1 and GAUT7, form a heteromeric enzyme complex in Arabidopsis thaliana Here, we established a heterologous GAUT expression system in HEK293 cells and show that co-expression of recombinant GAUT1 with GAUT7 results in the production of a soluble GAUT1:GAUT7 complex that catalyzes elongation of HG products in vitro The reaction rates, progress curves, and product distributions exhibited major differences dependent upon small changes in the degree of polymerization (DP) of the oligosaccharide acceptor. GAUT1:GAUT7 displayed >45-fold increased catalytic efficiency with DP11 acceptors relative to DP7 acceptors. Although GAUT1:GAUT7 synthesized high-molecular-weight polymeric HG (>100 kDa) in a substrate concentration-dependent manner typical of distributive (nonprocessive) glycosyltransferases with DP11 acceptors, reactions primed with short-chain acceptors resulted in a bimodal product distribution of glycan products that has previously been reported as evidence for a processive model of GT elongation. As an alternative to the processive glycosyltransfer model, a two-phase distributive elongation model is proposed in which a slow phase, which includes the de novo initiation of HG and elongation of short-chain acceptors, is distinguished from a phase of rapid elongation of intermediate- and long-chain acceptors. Upon reaching a critical chain length of DP11, GAUT1:GAUT7 elongates HG to high-molecular-weight products.
Collapse
Affiliation(s)
- Robert A Amos
- From the Complex Carbohydrate Research Center and.,the Department of Biochemistry and Molecular Biology, University of Georgia, Athens, Georgia 30602
| | | | | | - Melani A Atmodjo
- From the Complex Carbohydrate Research Center and.,the Department of Biochemistry and Molecular Biology, University of Georgia, Athens, Georgia 30602
| | | | - Kelley W Moremen
- From the Complex Carbohydrate Research Center and.,the Department of Biochemistry and Molecular Biology, University of Georgia, Athens, Georgia 30602
| | - Debra Mohnen
- From the Complex Carbohydrate Research Center and .,the Department of Biochemistry and Molecular Biology, University of Georgia, Athens, Georgia 30602
| |
Collapse
|
45
|
Sheikh MO, Halmo SM, Patel S, Middleton D, Takeuchi H, Schafer CM, West CM, Haltiwanger RS, Avci FY, Moremen KW, Wells L. Rapid screening of sugar-nucleotide donor specificities of putative glycosyltransferases. Glycobiology 2018; 27:206-212. [PMID: 28177478 DOI: 10.1093/glycob/cww114] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2016] [Revised: 10/20/2016] [Accepted: 11/17/2016] [Indexed: 01/09/2023] Open
Abstract
Determining the correct enzymatic activity of putative glycosyltransferases (GTs) can be challenging as these enzymes can utilize multiple donor and acceptor substrates. Upon initial determination of the donor-sugar nucleotide(s), a GT utilizes various acceptor molecules that can then be tested. Here, we describe a quick method to screen sugar-nucleotide donor specificities of GTs utilizing a sensitive, nonradioactive, commercially available bioluminescent uridine diphosphate detection kit. This in vitro method allowed us to validate the sugar-nucleotide donor-substrate specificities of recombinantly expressed human, bovine, bacterial and protozoan GTs. Our approach, which is less time consuming than many traditional assays that utilize radiolabeled sugars and chromatographic separations, should facilitate discovery of novel GTs that participate in diverse biological processes.
Collapse
Affiliation(s)
- M Osman Sheikh
- Complex Carbohydrate Research Center, University of Georgia, Athens, GA, USA
| | - Stephanie M Halmo
- Complex Carbohydrate Research Center, University of Georgia, Athens, GA, USA.,Department of Biochemistry and Molecular Biology, University of Georgia, Athens, GA, USA
| | - Sneha Patel
- Complex Carbohydrate Research Center, University of Georgia, Athens, GA, USA.,Department of Biochemistry and Molecular Biology, University of Georgia, Athens, GA, USA
| | - Dustin Middleton
- Complex Carbohydrate Research Center, University of Georgia, Athens, GA, USA.,Department of Biochemistry and Molecular Biology, University of Georgia, Athens, GA, USA.,Center for Molecular Medicine, University of Georgia, Athens, GA, USA
| | - Hideyuki Takeuchi
- Complex Carbohydrate Research Center, University of Georgia, Athens, GA, USA.,Department of Biochemistry and Molecular Biology, University of Georgia, Athens, GA, USA
| | | | - Christopher M West
- Department of Biochemistry and Molecular Biology, University of Georgia, Athens, GA, USA
| | - Robert S Haltiwanger
- Complex Carbohydrate Research Center, University of Georgia, Athens, GA, USA.,Department of Biochemistry and Molecular Biology, University of Georgia, Athens, GA, USA
| | - Fikri Y Avci
- Complex Carbohydrate Research Center, University of Georgia, Athens, GA, USA.,Department of Biochemistry and Molecular Biology, University of Georgia, Athens, GA, USA.,Center for Molecular Medicine, University of Georgia, Athens, GA, USA
| | - Kelley W Moremen
- Complex Carbohydrate Research Center, University of Georgia, Athens, GA, USA.,Department of Biochemistry and Molecular Biology, University of Georgia, Athens, GA, USA
| | - Lance Wells
- Complex Carbohydrate Research Center, University of Georgia, Athens, GA, USA.,Department of Biochemistry and Molecular Biology, University of Georgia, Athens, GA, USA
| |
Collapse
|
46
|
Imae R, Manya H, Tsumoto H, Osumi K, Tanaka T, Mizuno M, Kanagawa M, Kobayashi K, Toda T, Endo T. CDP-glycerol inhibits the synthesis of the functional O-mannosyl glycan of α-dystroglycan. J Biol Chem 2018; 293:12186-12198. [PMID: 29884773 DOI: 10.1074/jbc.ra118.003197] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2018] [Revised: 06/06/2018] [Indexed: 12/24/2022] Open
Abstract
α-Dystroglycan (α-DG) is a highly glycosylated cell-surface laminin receptor. Defects in the O-mannosyl glycan of an α-DG with laminin-binding activity can cause α-dystroglycanopathy, a group of congenital muscular dystrophies. In the biosynthetic pathway of functional O-mannosyl glycan, fukutin (FKTN) and fukutin-related protein (FKRP), whose mutated genes underlie α-dystroglycanopathy, sequentially transfer ribitol phosphate (RboP) from CDP-Rbo to form a tandem RboP unit (RboP-RboP) required for the synthesis of the laminin-binding epitope on O-mannosyl glycan. Both RboP- and glycerol phosphate (GroP)-substituted glycoforms have recently been detected in recombinant α-DG. However, it is unclear how GroP is transferred to the O-mannosyl glycan or whether GroP substitution affects the synthesis of the O-mannosyl glycan. Here, we report that, in addition to having RboP transfer activity, FKTN and FKRP can transfer GroP to O-mannosyl glycans by using CDP-glycerol (CDP-Gro) as a donor substrate. Kinetic experiments indicated that CDP-Gro is a less efficient donor substrate for FKTN than is CDP-Rbo. We also show that the GroP-substituted glycoform synthesized by FKTN does not serve as an acceptor substrate for FKRP and that therefore further elongation of the outer glycan chain cannot occur with this glycoform. Finally, CDP-Gro inhibited the RboP transfer activities of both FKTN and FKRP. These results suggest that CDP-Gro inhibits the synthesis of the functional O-mannosyl glycan of α-DG by preventing further elongation of the glycan chain. This is the first report of GroP transferases in mammals.
Collapse
Affiliation(s)
- Rieko Imae
- Molecular Glycobiology, Tokyo Metropolitan Geriatric Hospital and Institute of Gerontology, Tokyo 173-0015, Japan
| | - Hiroshi Manya
- Molecular Glycobiology, Tokyo Metropolitan Geriatric Hospital and Institute of Gerontology, Tokyo 173-0015, Japan.
| | - Hiroki Tsumoto
- Proteome Research, Research Team for Mechanism of Aging, Tokyo Metropolitan Geriatric Hospital and Institute of Gerontology, Tokyo 173-0015, Japan
| | - Kenji Osumi
- Laboratory of Glyco-organic Chemistry, The Noguchi Institute, Tokyo 173-0003, Japan
| | - Tomohiro Tanaka
- Laboratory of Glyco-organic Chemistry, The Noguchi Institute, Tokyo 173-0003, Japan
| | - Mamoru Mizuno
- Laboratory of Glyco-organic Chemistry, The Noguchi Institute, Tokyo 173-0003, Japan
| | - Motoi Kanagawa
- Division of Neurology/Molecular Brain Science, Kobe University Graduate School of Medicine, Hyogo 650-0017, Japan
| | - Kazuhiro Kobayashi
- Division of Neurology/Molecular Brain Science, Kobe University Graduate School of Medicine, Hyogo 650-0017, Japan
| | - Tatsushi Toda
- Division of Neurology/Molecular Brain Science, Kobe University Graduate School of Medicine, Hyogo 650-0017, Japan; Department of Neurology, Division of Neuroscience, Graduate School of Medicine, The University of Tokyo, Tokyo 113-8655, Japan
| | - Tamao Endo
- Molecular Glycobiology, Tokyo Metropolitan Geriatric Hospital and Institute of Gerontology, Tokyo 173-0015, Japan
| |
Collapse
|
47
|
Tucker JD, Lu PJ, Xiao X, Lu QL. Overexpression of Mutant FKRP Restores Functional Glycosylation and Improves Dystrophic Phenotype in FKRP Mutant Mice. MOLECULAR THERAPY-NUCLEIC ACIDS 2018; 11:216-227. [PMID: 29858056 PMCID: PMC5992437 DOI: 10.1016/j.omtn.2018.02.008] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/17/2017] [Revised: 02/20/2018] [Accepted: 02/23/2018] [Indexed: 11/17/2022]
Abstract
Autosomal recessive homozygous or compound heterozygous mutations in FKRP result in forms of muscular dystrophy-dystroglycanopathy varying in age of onset, clinical presentation, and disease progression, ranging from the severe Walker-Warburg, type A,5 (MDDGA5), muscle-eye-brain (MDDGB5) with or without cognitive deficit, to limb-girdle type 2I (MDDGC5). Phenotypic variation indicates degrees of functionality of individual FKRP mutation, which has been supported by the presence of residual expression of functionally glycosylated α-dystroglycan (DG) in muscles of both animal models and patients. However, direct evidence showing enhancement in glycosylation of α-DG by mutant FKRP is lacking. Using AAV9-mediated overexpression of mutant human FKRP bearing the P448L mutation (mhFKRP-P448L) associated with severe congenital muscular dystrophy (CMD), we demonstrate the restoration of functional glycosylation of α-DG and reduction in markers of disease progression. Expression of mhFKRP-P448L also corrects dystrophic phenotypes in the models of L276I mutation with mild disease phenotype and causes no obvious histological or biomarker alteration in C57BL/6 normal mice. Our results confirm the existing function of mutant FKRP. The results also suggest that mutant FKRP could be an alternative approach for potential gene therapy should normal FKRP gene products be immunogenic.
Collapse
Affiliation(s)
- Jason D Tucker
- McColl-Lockwood Laboratory for Muscular Dystrophy Research, James G. Cannon Research Center, Carolinas Medical Center, Charlotte, NC 28203, USA
| | - Pei J Lu
- McColl-Lockwood Laboratory for Muscular Dystrophy Research, James G. Cannon Research Center, Carolinas Medical Center, Charlotte, NC 28203, USA
| | - Xiao Xiao
- Division of Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Qi L Lu
- McColl-Lockwood Laboratory for Muscular Dystrophy Research, James G. Cannon Research Center, Carolinas Medical Center, Charlotte, NC 28203, USA.
| |
Collapse
|
48
|
Moremen KW, Ramiah A, Stuart M, Steel J, Meng L, Forouhar F, Moniz HA, Gahlay G, Gao Z, Chapla D, Wang S, Yang JY, Prabahkar PK, Johnson R, dela Rosa M, Geisler C, Nairn AV, Wu SC, Tong L, Gilbert HJ, LaBaer J, Jarvis DL. Expression system for structural and functional studies of human glycosylation enzymes. Nat Chem Biol 2018; 14:156-162. [PMID: 29251719 PMCID: PMC5774587 DOI: 10.1038/nchembio.2539] [Citation(s) in RCA: 185] [Impact Index Per Article: 26.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2017] [Accepted: 11/08/2017] [Indexed: 12/14/2022]
Abstract
Vertebrate glycoproteins and glycolipids are synthesized in complex biosynthetic pathways localized predominantly within membrane compartments of the secretory pathway. The enzymes that catalyze these reactions are exquisitely specific, yet few have been extensively characterized because of challenges associated with their recombinant expression as functional products. We used a modular approach to create an expression vector library encoding all known human glycosyltransferases, glycoside hydrolases, and sulfotransferases, as well as other glycan-modifying enzymes. We then expressed the enzymes as secreted catalytic domain fusion proteins in mammalian and insect cell hosts, purified and characterized a subset of the enzymes, and determined the structure of one enzyme, the sialyltransferase ST6GalNAcII. Many enzymes were produced at high yields and at similar levels in both hosts, but individual protein expression levels varied widely. This expression vector library will be a transformative resource for recombinant enzyme production, broadly enabling structure-function studies and expanding applications of these enzymes in glycochemistry and glycobiology.
Collapse
Affiliation(s)
- Kelley W. Moremen
- Complex Carbohydrate Research Center, University of Georgia, Athens, GA
| | | | - Melissa Stuart
- Department of Molecular Biology, University of Wyoming, Laramie, WY 82071
| | - Jason Steel
- Biodesign Institute, Arizona State University, Tempe, AZ 85287
| | - Lu Meng
- Complex Carbohydrate Research Center, University of Georgia, Athens, GA
| | - Farhad Forouhar
- Department of Biological Sciences, Northeast Structural Genomics Consortium, Columbia University, New York, New York 10027
| | - Heather A. Moniz
- Complex Carbohydrate Research Center, University of Georgia, Athens, GA
| | - Gagandeep Gahlay
- Department of Molecular Biology, University of Wyoming, Laramie, WY 82071
| | - Zhongwei Gao
- Complex Carbohydrate Research Center, University of Georgia, Athens, GA
| | | | - Shuo Wang
- Complex Carbohydrate Research Center, University of Georgia, Athens, GA
| | - Jeong-Yeh Yang
- Complex Carbohydrate Research Center, University of Georgia, Athens, GA
| | | | - Roy Johnson
- Complex Carbohydrate Research Center, University of Georgia, Athens, GA
| | - Mitche dela Rosa
- Complex Carbohydrate Research Center, University of Georgia, Athens, GA
| | - Christoph Geisler
- Department of Molecular Biology, University of Wyoming, Laramie, WY 82071
| | - Alison V. Nairn
- Complex Carbohydrate Research Center, University of Georgia, Athens, GA
| | - Sheng-Cheng Wu
- Complex Carbohydrate Research Center, University of Georgia, Athens, GA
| | - Liang Tong
- Department of Biological Sciences, Northeast Structural Genomics Consortium, Columbia University, New York, New York 10027
| | - Harry J. Gilbert
- Complex Carbohydrate Research Center, University of Georgia, Athens, GA
| | - Joshua LaBaer
- Biodesign Institute, Arizona State University, Tempe, AZ 85287
| | - Donald L. Jarvis
- Department of Molecular Biology, University of Wyoming, Laramie, WY 82071
| |
Collapse
|
49
|
Kanagawa M, Toda T. Ribitol-phosphate—a newly identified posttranslational glycosylation unit in mammals: structure, modification enzymes and relationship to human diseases. J Biochem 2018; 163:359-369. [DOI: 10.1093/jb/mvy020] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2017] [Accepted: 01/23/2018] [Indexed: 01/05/2023] Open
Affiliation(s)
- Motoi Kanagawa
- Division of Molecular Brain Science, Kobe University Graduate School of Medicine, 7-5-1 Kusunoki-cho, Chuo-ku, Kobe 650-0017, Japan
| | - Tatsushi Toda
- Division of Molecular Brain Science, Kobe University Graduate School of Medicine, 7-5-1 Kusunoki-cho, Chuo-ku, Kobe 650-0017, Japan
- Department of Neurology, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8655, Japan
| |
Collapse
|
50
|
Sheikh MO, Halmo SM, Wells L. Recent advancements in understanding mammalian O-mannosylation. Glycobiology 2017; 27:806-819. [PMID: 28810660 PMCID: PMC6082599 DOI: 10.1093/glycob/cwx062] [Citation(s) in RCA: 78] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2017] [Revised: 06/25/2017] [Accepted: 06/28/2017] [Indexed: 02/07/2023] Open
Abstract
The post-translational glycosylation of select proteins by O-linked mannose (O-mannose or O-man) is a conserved modification from yeast to humans and has been shown to be necessary for proper development and growth. The most well studied O-mannosylated mammalian protein is α-dystroglycan (α-DG). Hypoglycosylation of α-DG results in varying severities of congenital muscular dystrophies, cancer progression and metastasis, and inhibited entry and infection of certain arenaviruses. Defects in the gene products responsible for post-translational modification of α-DG, primarily glycosyltransferases, are the basis for these diseases. The multitude of clinical phenotypes resulting from defective O-mannosylation highlights the biomedical significance of this unique modification. Elucidation of the various O-mannose biosynthetic pathways is imperative to understanding a broad range of human diseases and for the development of novel therapeutics. In this review, we will focus on recent discoveries delineating the various enzymes, structures and functions associated with O-mannose-initiated glycoproteins. Additionally, we discuss current gaps in our knowledge of mammalian O-mannosylation, discuss the evolution of this pathway, and illustrate the utility and limitations of model systems to study functions of O-mannosylation.
Collapse
Affiliation(s)
- M Osman Sheikh
- Complex Carbohydrate Research Center, University of Georgia, 315 Riverbend Road, Athens, GA 30602, USA
| | - Stephanie M Halmo
- Complex Carbohydrate Research Center, University of Georgia, 315 Riverbend Road, Athens, GA 30602, USA
- Department of Biochemistry and Molecular Biology, University of Georgia, 315 Riverbend Road, Athens, GA 30602, USA
| | - Lance Wells
- Complex Carbohydrate Research Center, University of Georgia, 315 Riverbend Road, Athens, GA 30602, USA
- Department of Biochemistry and Molecular Biology, University of Georgia, 315 Riverbend Road, Athens, GA 30602, USA
| |
Collapse
|