1
|
Wei K, Zhang J, Qu W, Zhu J, Zhu Q, Yi W, Zou C, Ma D, Li X. FUT8 Regulates Cerebellar Neurogenesis and Development Through Maintaining the Level of Neural Cell Adhesion Molecule Cntn2. Mol Neurobiol 2025; 62:5679-5694. [PMID: 39604780 DOI: 10.1007/s12035-024-04620-8] [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: 11/30/2023] [Accepted: 11/07/2024] [Indexed: 11/29/2024]
Abstract
Core fucosylation at N-glycans, which is uniquely catalyzed by fucosyltransferase FUT8, plays essential roles in post-translational regulation of protein function. Aberrant core fucosylation leads to neurological disorders in individuals with congenital glycosylation disorders (CDG). However, the underlying mechanisms for these neurological defects remain largely unknown. In this study, we have showed that FUT8 and fucosylation are abundant in cerebellum. Specific deletion of Fut8 in cerebellar granule neuron progenitors (GNPs) results in the impaired proliferation and differentiation of GNPs, as well as the compromised neuronal development, synaptic physiology and motor coordination. Mechanistically, we have showed that Fut8 deficiency reduced Contactin 2 (Cntn2) expression, a member of neural cell adhesion molecules (NCAMs). Furthermore, ectopic Cntn2 can rescue the neuronal defects induced by Fut8 deficiency. Collectively, our study has revealed the important roles of FUT8 and core fucosylation in regulating cerebellar development and function through modulating Cntn2 expression.
Collapse
Affiliation(s)
- Kaiyan Wei
- The Children's Hospital, School of Medicine, National Clinical Research Center for Child Health, Zhejiang University, Hangzhou, 310052, China
- School of Medicine, The Institute of Translational Medicine, Zhejiang University, Hangzhou, 310029, China
| | - Jinyu Zhang
- The Children's Hospital, School of Medicine, National Clinical Research Center for Child Health, Zhejiang University, Hangzhou, 310052, China
- School of Medicine, The Institute of Translational Medicine, Zhejiang University, Hangzhou, 310029, China
- Binjiang Institute of Zhejiang University, Hangzhou, 310053, China
| | - Wenzheng Qu
- The Children's Hospital, School of Medicine, National Clinical Research Center for Child Health, Zhejiang University, Hangzhou, 310052, China
| | - Jinpiao Zhu
- The Children's Hospital, School of Medicine, National Clinical Research Center for Child Health, Zhejiang University, Hangzhou, 310052, China
| | - Qiang Zhu
- MOE Key Laboratory of Biosystems Homeostasis & Protection, College of Life Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Wen Yi
- MOE Key Laboratory of Biosystems Homeostasis & Protection, College of Life Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Chaochun Zou
- The Children's Hospital, School of Medicine, National Clinical Research Center for Child Health, Zhejiang University, Hangzhou, 310052, China.
| | - Daqing Ma
- The Children's Hospital, School of Medicine, National Clinical Research Center for Child Health, Zhejiang University, Hangzhou, 310052, China.
- School of Medicine, The Institute of Translational Medicine, Zhejiang University, Hangzhou, 310029, China.
- Division of Anaesthetics, Pain Medicine and Intensive Care, Department of Surgery and Cancer, Faculty of Medicine, Imperial College London, Chelsea and Westminster Hospital, London, UK.
| | - Xuekun Li
- The Children's Hospital, School of Medicine, National Clinical Research Center for Child Health, Zhejiang University, Hangzhou, 310052, China.
- School of Medicine, The Institute of Translational Medicine, Zhejiang University, Hangzhou, 310029, China.
- Binjiang Institute of Zhejiang University, Hangzhou, 310053, China.
| |
Collapse
|
2
|
Faustmann TJ, Hensel L, Bahic A, Wada Y, Grüneberg M, Marquardt T, Kamp D, Schilbach L. The long way to diagnosis: attention disorder, alcohol addiction or congenital disorder of glycosylation? A case report. BMC Psychiatry 2025; 25:435. [PMID: 40301838 PMCID: PMC12042650 DOI: 10.1186/s12888-025-06862-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/19/2024] [Accepted: 04/15/2025] [Indexed: 05/01/2025] Open
Abstract
Attention deficit hyperactivity disorder (ADHD) is a relatively common disorder in clinical psychiatry. Patients often suffer from symptoms long before the diagnosis due to an overlap with other psychiatric differential diagnosis. Importantly, alcohol addiction and other illicit drug dependence and withdrawal symptoms mimicking ADHD symptoms should be ruled out. Here we present a rare case of a young female patient with symptoms of ADHD and an extremely high carbohydrate-deficient transferrin (CDT) of 19,6% (< 1,3%) indicating the presence of a congenital disorder of glycosylation (CDG). A thorough diagnostic workup excluded alcohol addiction as a cause of the constantly high CDT levels. The CDT test was positive due a transferrin mutation affecting the glycosylation site. Nevertheless, psychiatric symptoms can be due to metabolic disorders which should be considered. Further, substance-use disorders (SUD) are a critical and potentially complicated differential diagnosis concerning diagnostic procedures and treatment in ADHD.
Collapse
Affiliation(s)
- Timo Jendrik Faustmann
- Department of Psychiatry and Psychotherapy, Medical Faculty, Heinrich Heine University Düsseldorf, Universitätsstraße 1, 40225, Düsseldorf, Germany.
- Department of General Psychiatry 2, LVR-Klinikum Düsseldorf, Bergische Landstraße 2, 40629, Düsseldorf, Germany.
| | - Lukas Hensel
- Department of Neurology, Medical Faculty, University Hospital Cologne, University of Cologne, Kerpenerstraße 62, 50937, Cologne, Germany
| | - Armin Bahic
- Department of Psychiatry and Psychotherapy, Medical Faculty, Heinrich Heine University Düsseldorf, Universitätsstraße 1, 40225, Düsseldorf, Germany
- Department of General Psychiatry 2, LVR-Klinikum Düsseldorf, Bergische Landstraße 2, 40629, Düsseldorf, Germany
| | - Yoshinao Wada
- Department of Molecular Medicine, Osaka Medical Center and Research Institute for Maternal and Child Health, 840 Murodo-cho, Izumi, Osaka, 594-1101, Japan
| | - Marianne Grüneberg
- Department of General Pediatrics, University Children's Hospital Münster, Albert-Schweitzer-Campus 1, 48149, Münster, Germany
| | - Thorsten Marquardt
- Department of General Pediatrics, University Children's Hospital Münster, Albert-Schweitzer-Campus 1, 48149, Münster, Germany
| | - Daniel Kamp
- Department of Psychiatry and Psychotherapy, Medical Faculty, Heinrich Heine University Düsseldorf, Universitätsstraße 1, 40225, Düsseldorf, Germany
- Department of General Psychiatry 2, LVR-Klinikum Düsseldorf, Bergische Landstraße 2, 40629, Düsseldorf, Germany
| | - Leonhard Schilbach
- Department of General Psychiatry 2, LVR-Klinikum Düsseldorf, Bergische Landstraße 2, 40629, Düsseldorf, Germany
- Department of Psychiatry and Psychotherapy, University Hospital, Ludwig Maximilians University Munich, Nußbaumstraße 7, 80336, Munich, Germany
| |
Collapse
|
3
|
Liu ZX, Zou TT, Liu HH, Jia HB, Zhang XQ. Knockout of the fcsk gene in zebrafish causes neurodevelopmental defects. Zool Res 2025; 46:313-324. [PMID: 40049660 PMCID: PMC12000135 DOI: 10.24272/j.issn.2095-8137.2024.229] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2024] [Accepted: 11/26/2024] [Indexed: 04/18/2025] Open
Abstract
Congenital disorders of glycosylation (CDG) are a cluster of monogenic disorders resulting from defects in glycosylation. FCSK encodes fucokinase, an enzyme that catalyzes the phosphorylation of L-fucose to generate fucose-1-phosphate, an important step in fucosylation. Mutations in FCSK lead to CDG with an autosomal recessive inheritance pattern, primarily manifesting as developmental delay, hypotonia, and brain abnormalities. However, no fcsk mutant animal models have yet been established. This study constructed the first fcsk knockout ( fcsk -/-) zebrafish model using CRISPR/Cas9 technology. Notably, fcsk -/- zebrafish exhibited impaired growth, characterized by delayed epiboly and DNA accumulation during early embryonic development, as well as brain atrophy in adulthood. Larval-stage fcsk -/- zebrafish displayed locomotor deficits and increased susceptibility to pentylenetetrazole-induced seizures. In adulthood, fcsk -/- zebrafish showed neurodevelopmental abnormalities, including increased anxiety, decreased aggression, reduced social preference, and impaired memory. Additionally, total protein fucosylation was markedly reduced in fcsk -/- zebrafish, accompanied by decreased expression of pofut2, which encodes protein O-fucosyltransferase 2, an enzyme involved in the fucosylation salvage pathway. Apoptotic activity was elevated in the midbrain-hindbrain boundary (MHB) of fcsk -/- zebrafish. Supplementation with GDP-L-fucose or the human FCSK gene restored developmental defects and total protein fucosylation in fcsk -/- zebrafish. RNA sequencing revealed dysregulated gene expression associated with glycosylation, apoptosis, and neurodegenerative diseases. These findings suggest that fcsk -/- zebrafish exhibit neurodevelopmental disorders, providing the first fcsk gene knockout animal model and offering a platform for investigating the molecular underpinnings of the disease and facilitating drug screening efforts.
Collapse
Affiliation(s)
- Zhen-Xing Liu
- Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and Technology and Center for Human Genome Research, Huazhong University of Science and Technology, Wuhan, Hubei 430074, China
| | - Ting-Ting Zou
- Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and Technology and Center for Human Genome Research, Huazhong University of Science and Technology, Wuhan, Hubei 430074, China
| | - Hui-Hui Liu
- Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and Technology and Center for Human Genome Research, Huazhong University of Science and Technology, Wuhan, Hubei 430074, China
| | - Hai-Bo Jia
- Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and Technology and Center for Human Genome Research, Huazhong University of Science and Technology, Wuhan, Hubei 430074, China. E-mail:
| | - Xian-Qin Zhang
- Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and Technology and Center for Human Genome Research, Huazhong University of Science and Technology, Wuhan, Hubei 430074, China. E-mail:
| |
Collapse
|
4
|
Nani JV, Muotri AR, Hayashi MAF. Peering into the mind: unraveling schizophrenia's secrets using models. Mol Psychiatry 2025; 30:659-678. [PMID: 39245692 DOI: 10.1038/s41380-024-02728-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/23/2023] [Revised: 08/21/2024] [Accepted: 08/27/2024] [Indexed: 09/10/2024]
Abstract
Schizophrenia (SCZ) is a complex mental disorder characterized by a range of symptoms, including positive and negative symptoms, as well as cognitive impairments. Despite the extensive research, the underlying neurobiology of SCZ remain elusive. To overcome this challenge, the use of diverse laboratory modeling techniques, encompassing cellular and animal models, and innovative approaches like induced pluripotent stem cell (iPSC)-derived neuronal cultures or brain organoids and genetically engineered animal models, has been crucial. Immortalized cellular models provide controlled environments for investigating the molecular and neurochemical pathways involved in neuronal function, while iPSCs and brain organoids, derived from patient-specific sources, offer significant advantage in translational research by facilitating direct comparisons of cellular phenotypes between patient-derived neurons and healthy-control neurons. Animal models can recapitulate the different psychopathological aspects that should be modeled, offering valuable insights into the neurobiology of SCZ. In addition, invertebrates' models are genetically tractable and offer a powerful approach to dissect the core genetic underpinnings of SCZ, while vertebrate models, especially mammals, with their more complex nervous systems and behavioral repertoire, provide a closer approximation of the human condition to study SCZ-related traits. This narrative review provides a comprehensive overview of the diverse modeling approaches, critically evaluating their strengths and limitations. By synthesizing knowledge from these models, this review offers a valuable source for researchers, clinicians, and stakeholders alike. Integrating findings across these different models may allow us to build a more holistic picture of SCZ pathophysiology, facilitating the exploration of new research avenues and informed decision-making for interventions.
Collapse
Affiliation(s)
- João V Nani
- Department of Pharmacology, Escola Paulista de Medicina (EPM), Universidade Federal de São Paulo (UNIFESP), São Paulo, SP, Brazil.
- National Institute for Translational Medicine (INCT-TM, CNPq/FAPESP/CAPES), Ribeirão Preto, Brazil.
| | - Alysson R Muotri
- Department of Pediatrics and Department of Molecular and Cellular Medicine, University of California, San Diego, La Jolla, CA, USA
| | - Mirian A F Hayashi
- Department of Pharmacology, Escola Paulista de Medicina (EPM), Universidade Federal de São Paulo (UNIFESP), São Paulo, SP, Brazil.
- National Institute for Translational Medicine (INCT-TM, CNPq/FAPESP/CAPES), Ribeirão Preto, Brazil.
| |
Collapse
|
5
|
Pan Q, Zhang XL. Roles of core fucosylation modification in immune system and diseases. CELL INSIGHT 2025; 4:100211. [PMID: 39624801 PMCID: PMC11609374 DOI: 10.1016/j.cellin.2024.100211] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/15/2024] [Revised: 10/01/2024] [Accepted: 10/02/2024] [Indexed: 01/04/2025]
Abstract
Core fucosylation, catalyzed by α1,6-fucosyltransferase (FUT8), is an important N-glycosylation modification process that attaches a fucose residue via an α1,6-linkage to the core N-acetylglucosamine of N-glycans in mammals. Research over the past three decades has revealed the critical role of FUT8-mediated core fucosylation modification in various physiological and pathological processes, including cell growth, adhesion, receptor activation, antibody-dependent cellular cytotoxicity (ADCC), tumor metastasis and infections. This review discusses the immune system function involving FUT8 and the mechanisms by which core fucosylation regulates immunity and contributes to disease. A deeper understanding of these mechanisms can provide insights into cellular biology and suggest new therapeutic approaches and targets for related diseases.
Collapse
Affiliation(s)
- Qiu Pan
- Hubei Province Key Laboratory of Allergy and Immunology, Department of Allergy Zhongnan Hospital of Wuhan University, Department of Immunology Wuhan University Taikang Medical School (School of Basic Medical Sciences), Wuhan, 430071, China
- State Key Laboratory of Virology, Frontier Science Center for Immunology and Metabolism, Wuhan University School of Medicine, Wuhan, 430071, China
| | - Xiao-Lian Zhang
- Hubei Province Key Laboratory of Allergy and Immunology, Department of Allergy Zhongnan Hospital of Wuhan University, Department of Immunology Wuhan University Taikang Medical School (School of Basic Medical Sciences), Wuhan, 430071, China
- State Key Laboratory of Virology, Frontier Science Center for Immunology and Metabolism, Wuhan University School of Medicine, Wuhan, 430071, China
| |
Collapse
|
6
|
Guo H, Sun Q, Huang X, Wang X, Zhang F, Qu W, Liu J, Cheng X, Zhu Q, Yi W, Shu Q, Li X. Fucosyltransferase 8 regulates adult neurogenesis and cognition of mice by modulating the Itga6-PI3K/Akt signaling pathway. SCIENCE CHINA. LIFE SCIENCES 2024; 67:1427-1440. [PMID: 38523237 DOI: 10.1007/s11427-023-2510-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/05/2023] [Accepted: 11/14/2023] [Indexed: 03/26/2024]
Abstract
Fucosyltransferase 8 (Fut8) and core fucosylation play critical roles in regulating various biological processes, including immune response, signal transduction, proteasomal degradation, and energy metabolism. However, the function and underlying mechanism of Fut8 and core fucosylation in regulating adult neurogenesis remains unknown. We have shown that Fut8 and core fucosylation display dynamic features during the differentiation of adult neural stem/progenitor cells (aNSPCs) and postnatal brain development. Fut8 depletion reduces the proliferation of aNSPCs and inhibits neuronal differentiation of aNSPCs in vitro and in vivo, respectively. Additionally, Fut8 deficiency impairs learning and memory in mice. Mechanistically, Fut8 directly interacts with integrin α6 (Itga6), an upstream regulator of the PI3k-Akt signaling pathway, and catalyzes core fucosylation of Itga6. Deletion of Fut8 enhances the ubiquitination of Itga6 by promoting the binding of ubiquitin ligase Trim21 to Itga6. Low levels of Itga6 inhibit the activity of the PI3K/Akt signaling pathway. Moreover, the Akt agonist SC79 can rescue neurogenic and behavioral deficits caused by Fut8 deficiency. In summary, our study uncovers an essential function of Fut8 and core fucosylation in regulating adult neurogenesis and sheds light on the underlying mechanisms.
Collapse
Affiliation(s)
- Hongfeng Guo
- The Children's Hospital, National Clinical Research Center for Child Health, School of Medicine, Zhejiang University, Hangzhou, 310052, China
- The Institute of Translational Medicine, School of Medicine, Zhejiang University, Hangzhou, 310029, China
| | - Qihang Sun
- The Children's Hospital, National Clinical Research Center for Child Health, School of Medicine, Zhejiang University, Hangzhou, 310052, China
- The Institute of Translational Medicine, School of Medicine, Zhejiang University, Hangzhou, 310029, China
| | - Xiaoli Huang
- The Children's Hospital, National Clinical Research Center for Child Health, School of Medicine, Zhejiang University, Hangzhou, 310052, China
| | - Xiaohao Wang
- The Children's Hospital, National Clinical Research Center for Child Health, School of Medicine, Zhejiang University, Hangzhou, 310052, China
| | - Feng Zhang
- The Children's Hospital, National Clinical Research Center for Child Health, School of Medicine, Zhejiang University, Hangzhou, 310052, China
| | - Wenzheng Qu
- The Children's Hospital, National Clinical Research Center for Child Health, School of Medicine, Zhejiang University, Hangzhou, 310052, China
| | - Jinling Liu
- The Children's Hospital, National Clinical Research Center for Child Health, School of Medicine, Zhejiang University, Hangzhou, 310052, China
| | - Xuejun Cheng
- The Children's Hospital, National Clinical Research Center for Child Health, School of Medicine, Zhejiang University, Hangzhou, 310052, China
| | - Qiang Zhu
- College of Life Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Wen Yi
- College of Life Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Qiang Shu
- The Children's Hospital, National Clinical Research Center for Child Health, School of Medicine, Zhejiang University, Hangzhou, 310052, China.
| | - Xuekun Li
- The Children's Hospital, National Clinical Research Center for Child Health, School of Medicine, Zhejiang University, Hangzhou, 310052, China.
- The Institute of Translational Medicine, School of Medicine, Zhejiang University, Hangzhou, 310029, China.
- Binjiang Institute of Zhejiang University, Hangzhou, 310053, China.
- Zhejiang University Cancer Center, Zhejiang University, Hangzhou, 310029, China.
| |
Collapse
|
7
|
Kizuka Y. Regulation of intracellular activity of N-glycan branching enzymes in mammals. J Biol Chem 2024; 300:107471. [PMID: 38879010 PMCID: PMC11328876 DOI: 10.1016/j.jbc.2024.107471] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2024] [Revised: 06/01/2024] [Accepted: 06/06/2024] [Indexed: 07/07/2024] Open
Abstract
Most proteins in the secretory pathway are glycosylated, and N-glycans are estimated to be attached to over 7000 proteins in humans. As structural variation of N-glycans critically regulates the functions of a particular glycoprotein, it is pivotal to understand how structural diversity of N-glycans is generated in cells. One of the major factors conferring structural variation of N-glycans is the variable number of N-acetylglucosamine branches. These branch structures are biosynthesized by dedicated glycosyltransferases, including GnT-III (MGAT3), GnT-IVa (MGAT4A), GnT-IVb (MGAT4B), GnT-V (MGAT5), and GnT-IX (GnT-Vb, MGAT5B). In addition, the presence or absence of core modification of N-glycans, namely, core fucose (included as an N-glycan branch in this manuscript), synthesized by FUT8, also confers large structural variation on N-glycans, thereby crucially regulating many protein-protein interactions. Numerous biochemical and medical studies have revealed that these branch structures are involved in a wide range of physiological and pathological processes. However, the mechanisms regulating the activity of the biosynthetic glycosyltransferases are yet to be fully elucidated. In this review, we summarize the previous findings and recent updates regarding regulation of the activity of these N-glycan branching enzymes. We hope that such information will help readers to develop a comprehensive overview of the complex system regulating mammalian N-glycan maturation.
Collapse
Affiliation(s)
- Yasuhiko Kizuka
- Institute for Glyco-core Research (iGCORE), Gifu University, Gifu, Japan.
| |
Collapse
|
8
|
Fazelzadeh Haghighi M, Jafari Khamirani H, Fallahi J, Monfared AA, Ashrafi Dehkordi K, Tabei SMB. Novel insight into FCSK-congenital disorder of glycosylation through a CRISPR-generated cell model. Mol Genet Genomic Med 2024; 12:e2445. [PMID: 38722107 PMCID: PMC11080630 DOI: 10.1002/mgg3.2445] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2023] [Revised: 04/08/2024] [Accepted: 04/23/2024] [Indexed: 05/12/2024] Open
Abstract
BACKGROUND FCSK-congenital disorder of glycosylation (FCSK-CDG) is a recently discovered rare autosomal recessive genetic disorder with defective fucosylation due to mutations in the fucokinase encoding gene, FCSK. Despite the essential role of fucokinase in the fucose salvage pathway and severe multisystem manifestations of FCSK-CDG patients, it is not elucidated which cells or which types of fucosylation are affected by its deficiency. METHODS In this study, CRISPR/Cas9 was employed to construct an FCSK-CDG cell model and explore the molecular mechanisms of the disease by lectin flow cytometry and real-time PCR analyses. RESULTS Comparison of cellular fucosylation by lectin flow cytometry in the created CRISPR/Cas9 FCSK knockout and the same unedited cell lines showed no significant change in the amount of cell surface fucosylated glycans, which is consistent with the only documented previous study on different cell types. It suggests a probable effect of this disease on secretory glycoproteins. Investigating O-fucosylation by analysis of the NOTCH3 gene expression as a potential target revealed a significant decrease in the FCSK knockout cells compared with the same unedited ones, proving the effect of fucokinase deficiency on EGF-like repeats O-fucosylation. CONCLUSION This study expands insight into the FCSK-CDG molecular mechanism; to the best of our knowledge, it is the first research conducted to reveal a gene whose expression level alters due to this disease.
Collapse
Affiliation(s)
- Maryam Fazelzadeh Haghighi
- Department of Molecular Medicine, School of Advanced TechnologiesShahrekord University of Medical SciencesShahrekordIran
| | | | - Jafar Fallahi
- Molecular Medicine Department, School of Advanced Medical Sciences and TechnologiesShiraz University of Medical SciencesShirazIran
| | - Ali Arabi Monfared
- Central Research LaboratoryShiraz University of Medical SciencesShirazIran
| | - Korosh Ashrafi Dehkordi
- Department of Molecular Medicine, School of Advanced TechnologiesShahrekord University of Medical SciencesShahrekordIran
| | - Seyed Mohammad Bagher Tabei
- Department of Medical GeneticsShiraz University of Medical SciencesShirazIran
- Maternal‐Fetal Medicine Research CenterShiraz University of Medical SciencesShirazIran
| |
Collapse
|
9
|
Gefen AM, Zaritsky JJ. Review of childhood genetic nephrolithiasis and nephrocalcinosis. Front Genet 2024; 15:1381174. [PMID: 38606357 PMCID: PMC11007102 DOI: 10.3389/fgene.2024.1381174] [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: 02/03/2024] [Accepted: 03/04/2024] [Indexed: 04/13/2024] Open
Abstract
Nephrolithiasis (NL) is a common condition worldwide. The incidence of NL and nephrocalcinosis (NC) has been increasing, along with their associated morbidity and economic burden. The etiology of NL and NC is multifactorial and includes both environmental components and genetic components, with multiple studies showing high heritability. Causative gene variants have been detected in up to 32% of children with NL and NC. Children with NL and NC are genotypically heterogenous, but often phenotypically relatively homogenous, and there are subsequently little data on the predictors of genetic childhood NL and NC. Most genetic diseases associated with NL and NC are secondary to hypercalciuria, including those secondary to hypercalcemia, renal phosphate wasting, renal magnesium wasting, distal renal tubular acidosis (RTA), proximal tubulopathies, mixed or variable tubulopathies, Bartter syndrome, hyperaldosteronism and pseudohyperaldosteronism, and hyperparathyroidism and hypoparathyroidism. The remaining minority of genetic diseases associated with NL and NC are secondary to hyperoxaluria, cystinuria, hyperuricosuria, xanthinuria, other metabolic disorders, and multifactorial etiologies. Genome-wide association studies (GWAS) in adults have identified multiple polygenic traits associated with NL and NC, often involving genes that are involved in calcium, phosphorus, magnesium, and vitamin D homeostasis. Compared to adults, there is a relative paucity of studies in children with NL and NC. This review aims to focus on the genetic component of NL and NC in children.
Collapse
Affiliation(s)
- Ashley M. Gefen
- Phoenix Children’s Hospital, Department of Pediatrics, Division of Nephrology, Phoenix, AZ, United States
| | | |
Collapse
|
10
|
Pascoal C, Francisco R, Mexia P, Pereira BL, Granjo P, Coelho H, Barbosa M, dos Reis Ferreira V, Videira PA. Revisiting the immunopathology of congenital disorders of glycosylation: an updated review. Front Immunol 2024; 15:1350101. [PMID: 38550576 PMCID: PMC10972870 DOI: 10.3389/fimmu.2024.1350101] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2023] [Accepted: 02/26/2024] [Indexed: 04/02/2024] Open
Abstract
Glycosylation is a critical post-translational modification that plays a pivotal role in several biological processes, such as the immune response. Alterations in glycosylation can modulate the course of various pathologies, such as the case of congenital disorders of glycosylation (CDG), a group of more than 160 rare and complex genetic diseases. Although the link between glycosylation and immune dysfunction has already been recognized, the immune involvement in most CDG remains largely unexplored and poorly understood. In this study, we provide an update on the immune dysfunction and clinical manifestations of the 12 CDG with major immune involvement, organized into 6 categories of inborn errors of immunity according to the International Union of Immunological Societies (IUIS). The immune involvement in phosphomannomutase 2 (PMM2)-CDG - the most frequent CDG - was comprehensively reviewed, highlighting a higher prevalence of immune issues during infancy and childhood and in R141H-bearing genotypes. Finally, using PMM2-CDG as a model, we point to links between abnormal glycosylation patterns in host cells and possibly favored interactions with microorganisms that may explain the higher susceptibility to infection. Further characterizing immunopathology and unusual host-pathogen adhesion in CDG can not only improve immunological standards of care but also pave the way for innovative preventive measures and targeted glycan-based therapies that may improve quality of life for people living with CDG.
Collapse
Affiliation(s)
- Carlota Pascoal
- Associate Laboratory i4HB - Institute for Health and Bioeconomy, NOVA School of Science and Technology, Universidade NOVA de Lisboa, Caparica, Portugal
- UCIBIO– Applied Molecular Biosciences Unit, Department of Life Sciences, NOVA School of Science and Technology, Universidade NOVA de Lisboa, Caparica, Portugal
- CDG & Allies-Professionals and Patient Associations International Network, Caparica, Portugal
| | - Rita Francisco
- Associate Laboratory i4HB - Institute for Health and Bioeconomy, NOVA School of Science and Technology, Universidade NOVA de Lisboa, Caparica, Portugal
- UCIBIO– Applied Molecular Biosciences Unit, Department of Life Sciences, NOVA School of Science and Technology, Universidade NOVA de Lisboa, Caparica, Portugal
- CDG & Allies-Professionals and Patient Associations International Network, Caparica, Portugal
| | - Patrícia Mexia
- Associate Laboratory i4HB - Institute for Health and Bioeconomy, NOVA School of Science and Technology, Universidade NOVA de Lisboa, Caparica, Portugal
- UCIBIO– Applied Molecular Biosciences Unit, Department of Life Sciences, NOVA School of Science and Technology, Universidade NOVA de Lisboa, Caparica, Portugal
- CDG & Allies-Professionals and Patient Associations International Network, Caparica, Portugal
| | - Beatriz Luís Pereira
- Associate Laboratory i4HB - Institute for Health and Bioeconomy, NOVA School of Science and Technology, Universidade NOVA de Lisboa, Caparica, Portugal
- UCIBIO– Applied Molecular Biosciences Unit, Department of Life Sciences, NOVA School of Science and Technology, Universidade NOVA de Lisboa, Caparica, Portugal
- CDG & Allies-Professionals and Patient Associations International Network, Caparica, Portugal
| | - Pedro Granjo
- Associate Laboratory i4HB - Institute for Health and Bioeconomy, NOVA School of Science and Technology, Universidade NOVA de Lisboa, Caparica, Portugal
- UCIBIO– Applied Molecular Biosciences Unit, Department of Life Sciences, NOVA School of Science and Technology, Universidade NOVA de Lisboa, Caparica, Portugal
- CDG & Allies-Professionals and Patient Associations International Network, Caparica, Portugal
| | - Helena Coelho
- Associate Laboratory i4HB - Institute for Health and Bioeconomy, NOVA School of Science and Technology, Universidade NOVA de Lisboa, Caparica, Portugal
- UCIBIO – Applied Molecular Biosciences Unit, Department of Chemistry, NOVA School of Science and Technology, Universidade NOVA de Lisboa, Caparica, Portugal
| | - Mariana Barbosa
- Associate Laboratory i4HB - Institute for Health and Bioeconomy, NOVA School of Science and Technology, Universidade NOVA de Lisboa, Caparica, Portugal
- UCIBIO– Applied Molecular Biosciences Unit, Department of Life Sciences, NOVA School of Science and Technology, Universidade NOVA de Lisboa, Caparica, Portugal
- CDG & Allies-Professionals and Patient Associations International Network, Caparica, Portugal
| | - Vanessa dos Reis Ferreira
- Associate Laboratory i4HB - Institute for Health and Bioeconomy, NOVA School of Science and Technology, Universidade NOVA de Lisboa, Caparica, Portugal
- UCIBIO– Applied Molecular Biosciences Unit, Department of Life Sciences, NOVA School of Science and Technology, Universidade NOVA de Lisboa, Caparica, Portugal
- CDG & Allies-Professionals and Patient Associations International Network, Caparica, Portugal
| | - Paula Alexandra Videira
- Associate Laboratory i4HB - Institute for Health and Bioeconomy, NOVA School of Science and Technology, Universidade NOVA de Lisboa, Caparica, Portugal
- UCIBIO– Applied Molecular Biosciences Unit, Department of Life Sciences, NOVA School of Science and Technology, Universidade NOVA de Lisboa, Caparica, Portugal
- CDG & Allies-Professionals and Patient Associations International Network, Caparica, Portugal
| |
Collapse
|
11
|
Shi M, Nan XR, Liu BQ. The Multifaceted Role of FUT8 in Tumorigenesis: From Pathways to Potential Clinical Applications. Int J Mol Sci 2024; 25:1068. [PMID: 38256141 PMCID: PMC10815953 DOI: 10.3390/ijms25021068] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2023] [Revised: 01/07/2024] [Accepted: 01/13/2024] [Indexed: 01/24/2024] Open
Abstract
FUT8, the sole glycosyltransferase responsible for N-glycan core fucosylation, plays a crucial role in tumorigenesis and development. Aberrant FUT8 expression disrupts the function of critical cellular components and triggers the abnormality of tumor signaling pathways, leading to malignant transformations such as proliferation, invasion, metastasis, and immunosuppression. The association between FUT8 and unfavorable outcomes in various tumors underscores its potential as a valuable diagnostic marker. Given the remarkable variation in biological functions and regulatory mechanisms of FUT8 across different tumor types, gaining a comprehensive understanding of its complexity is imperative. Here, we review how FUT8 plays roles in tumorigenesis and development, and how this outcome could be utilized to develop potential clinical therapies for tumors.
Collapse
Affiliation(s)
| | | | - Bao-Qin Liu
- Department of Biochemistry & Molecular Biology, School of Life Sciences, China Medical University, Shenyang 110122, China; (M.S.); (X.-R.N.)
| |
Collapse
|
12
|
Xu X, Fukuda T, Takai J, Morii S, Sun Y, Liu J, Ohno S, Isaji T, Yamaguchi Y, Nakano M, Moriguchi T, Gu J. Exogenous l-fucose attenuates neuroinflammation induced by lipopolysaccharide. J Biol Chem 2024; 300:105513. [PMID: 38042483 PMCID: PMC10772726 DOI: 10.1016/j.jbc.2023.105513] [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: 10/13/2023] [Revised: 11/16/2023] [Accepted: 11/21/2023] [Indexed: 12/04/2023] Open
Abstract
α1,6-Fucosyltransferase (Fut8) catalyzes the transfer of fucose to the innermost GlcNAc residue of N-glycan to form core fucosylation. Our previous studies showed that lipopolysaccharide (LPS) treatment highly induced neuroinflammation in Fut8 homozygous KO (Fut8-/-) or heterozygous KO (Fut8+/-) mice, compared with the WT (Fut8+/+) mice. To understand the underlying mechanism, we utilized a sensitive inflammation-monitoring mouse system that contains the human interleukin-6 (hIL6) bacterial artificial chromosome transgene modified with luciferase (Luc) reporter cassette. We successfully detected LPS-induced neuroinflammation in the central nervous system by exploiting this bacterial artificial chromosome transgenic monitoring system. Then we examined the effects of l-fucose on neuroinflammation in the Fut8+/- mice. The lectin blot and mass spectrometry analysis showed that l-fucose preadministration increased the core fucosylation levels in the Fut8+/- mice. Notably, exogenous l-fucose attenuated the LPS-induced IL-6 mRNA and Luc mRNA expression in the cerebral tissues, confirmed using the hIL6-Luc bioluminescence imaging system. The activation of microglial cells, which provoke neuroinflammatory responses upon LPS stimulation, was inhibited by l-fucose preadministration. l-Fucose also suppressed the downstream intracellular signaling of IL-6, such as the phosphorylation levels of JAK2 (Janus kinase 2), Akt (protein kinase B), and STAT3 (signal transducer and activator of transcription 3). l-Fucose administration increased gp130 core fucosylation levels and decreased the association of gp130 with the IL-6 receptor in Fut8+/- mice, which was further confirmed in BV-2 cells. These results indicate that l-fucose administration ameliorates the LPS-induced neuroinflammation in the Fut8+/- mice, suggesting that core fucosylation plays a vital role in anti-inflammation and that l-fucose is a potential prophylactic compound against neuroinflammation.
Collapse
Affiliation(s)
- Xing Xu
- Division of Regulatory Glycobiology, Institute of Molecular Biomembrane and Glycobiology, Tohoku Medical and Pharmaceutical University, Sendai, Miyagi, Japan
| | - Tomohiko Fukuda
- Division of Regulatory Glycobiology, Institute of Molecular Biomembrane and Glycobiology, Tohoku Medical and Pharmaceutical University, Sendai, Miyagi, Japan
| | - Jun Takai
- Division of Medical Biochemistry, Tohoku Medical and Pharmaceutical University, Sendai, Miyagi, Japan
| | - Sayaka Morii
- Graduate School of Integrated Sciences for Life, Hiroshima University, Higashi-Hiroshima, Japan
| | - Yuhan Sun
- Division of Regulatory Glycobiology, Institute of Molecular Biomembrane and Glycobiology, Tohoku Medical and Pharmaceutical University, Sendai, Miyagi, Japan
| | - Jianwei Liu
- Division of Regulatory Glycobiology, Institute of Molecular Biomembrane and Glycobiology, Tohoku Medical and Pharmaceutical University, Sendai, Miyagi, Japan
| | - Shiho Ohno
- Division of Structural Glycobiology, Institute of Molecular Biomembrane and Glycobiology, Tohoku Medical and Pharmaceutical University, Sendai, Miyagi, Japan
| | - Tomoya Isaji
- Division of Regulatory Glycobiology, Institute of Molecular Biomembrane and Glycobiology, Tohoku Medical and Pharmaceutical University, Sendai, Miyagi, Japan
| | - Yoshiki Yamaguchi
- Division of Structural Glycobiology, Institute of Molecular Biomembrane and Glycobiology, Tohoku Medical and Pharmaceutical University, Sendai, Miyagi, Japan
| | - Miyako Nakano
- Graduate School of Integrated Sciences for Life, Hiroshima University, Higashi-Hiroshima, Japan
| | - Takashi Moriguchi
- Division of Medical Biochemistry, Tohoku Medical and Pharmaceutical University, Sendai, Miyagi, Japan
| | - Jianguo Gu
- Division of Regulatory Glycobiology, Institute of Molecular Biomembrane and Glycobiology, Tohoku Medical and Pharmaceutical University, Sendai, Miyagi, Japan.
| |
Collapse
|
13
|
van Pul L, Maurer I, Boeser-Nunnink BD, Harskamp AM, van Dort KA, Kootstra NA. A genetic variation in fucosyltransferase 8 accelerates HIV-1 disease progression indicating a role for N-glycan fucosylation. AIDS 2023; 37:1959-1969. [PMID: 37598360 PMCID: PMC10552802 DOI: 10.1097/qad.0000000000003689] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2023] [Revised: 07/25/2023] [Accepted: 08/07/2023] [Indexed: 08/22/2023]
Abstract
OBJECTIVES Core fucosylation by fucosyltransferase 8 (FUT8) is an important posttranslational modification that impacts components of the immune system. Genetic variations in FUT8 can alter its function and could, therefore, play a role in the antiviral immune response and pathogenesis of HIV-1. This study analysed the effect of a single nucleotide polymorphism (SNP) in FUT8 on the clinical course of HIV-1 infection. DESIGN/METHODS The effect of SNPs in FUT8 on untreated HIV-1 disease outcome were analysed in a cohort of 304 people with HIV-1 (PWH) using survival analysis. Flow-cytometry was used to determine the effect of SNP on T-cell activation, differentiation and exhaustion/senescence. T-cell function was determined by proliferation assay and by measuring intracellular cytokine production. The effect of the SNP on HIV-1 replication was determined by in-vitro HIV-1 infections. Sensitivity of HIV-1 produced in PBMC with or without the SNP to broadly neutralizing antibodies was determined using a TZM-bl based neutralization assay. RESULTS Presence of the minor allele of SNP rs4131564 was associated with accelerated disease progression. The SNP had no effect on T-cell activation and T-cell differentiation in PWH. Additionally, no differences in T-cell functionality as determined by proliferation and cytokine production was observed. HIV-1 replication and neutralization sensitivity was also unaffected by the SNP in FUT8. CONCLUSION SNP rs4131564 in FUT8 showed a major impact on HIV-1 disease course underscoring a role for N-glycan fucosylation even though no clear effect on the immune system or HIV-1 could be determined in vitro .
Collapse
Affiliation(s)
- Lisa van Pul
- Amsterdam Institute for Infection and Immunity
- Department of Experimental Immunology, Amsterdam UMC, location University of Amsterdam, Amsterdam, The Netherlands
| | - Irma Maurer
- Amsterdam Institute for Infection and Immunity
- Department of Experimental Immunology, Amsterdam UMC, location University of Amsterdam, Amsterdam, The Netherlands
| | - Brigitte D.M. Boeser-Nunnink
- Amsterdam Institute for Infection and Immunity
- Department of Experimental Immunology, Amsterdam UMC, location University of Amsterdam, Amsterdam, The Netherlands
| | - Agnes M. Harskamp
- Amsterdam Institute for Infection and Immunity
- Department of Experimental Immunology, Amsterdam UMC, location University of Amsterdam, Amsterdam, The Netherlands
| | - Karel A. van Dort
- Amsterdam Institute for Infection and Immunity
- Department of Experimental Immunology, Amsterdam UMC, location University of Amsterdam, Amsterdam, The Netherlands
| | - Neeltje A. Kootstra
- Amsterdam Institute for Infection and Immunity
- Department of Experimental Immunology, Amsterdam UMC, location University of Amsterdam, Amsterdam, The Netherlands
| |
Collapse
|
14
|
Polenghi M, Taverna E. Intracellular traffic and polarity in brain development. Front Neurosci 2023; 17:1172016. [PMID: 37859764 PMCID: PMC10583573 DOI: 10.3389/fnins.2023.1172016] [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: 02/22/2023] [Accepted: 07/31/2023] [Indexed: 10/21/2023] Open
Abstract
Neurons forming the human brain are generated during embryonic development by neural stem and progenitor cells via a process called neurogenesis. A crucial feature contributing to neural stem cell morphological and functional heterogeneity is cell polarity, defined as asymmetric distribution of cellular components. Cell polarity is built and maintained thanks to the interplay between polarity proteins and polarity-generating organelles, such as the endoplasmic reticulum (ER) and the Golgi apparatus (GA). ER and GA affect the distribution of membrane components and work as a hub where glycans are added to nascent proteins and lipids. In the last decades our knowledge on the role of polarity in neural stem and progenitor cells have increased tremendously. However, the role of traffic and associated glycosylation in neural stem and progenitor cells is still relatively underexplored. In this review, we discuss the link between cell polarity, architecture, identity and intracellular traffic, and highlight how studies on neurons have shaped our knowledge and conceptual framework on traffic and polarity. We will then conclude by discussing how a group of rare diseases, called congenital disorders of glycosylation (CDG) offers the unique opportunity to study the contribution of traffic and glycosylation in the context of neurodevelopment.
Collapse
|
15
|
Abdelbary M, Nolz JC. N-linked glycans: an underappreciated key determinant of T cell development, activation, and function. IMMUNOMETABOLISM (COBHAM, SURREY) 2023; 5:e00035. [PMID: 38027254 PMCID: PMC10662610 DOI: 10.1097/in9.0000000000000035] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/29/2023] [Accepted: 10/31/2023] [Indexed: 12/01/2023]
Abstract
N-linked glycosylation is a post-translational modification that results in the decoration of newly synthesized proteins with diverse types of oligosaccharides that originate from the amide group of the amino acid asparagine. The sequential and collective action of multiple glycosidases and glycosyltransferases are responsible for determining the overall size, composition, and location of N-linked glycans that become covalently linked to an asparagine during and after protein translation. A growing body of evidence supports the critical role of N-linked glycan synthesis in regulating many features of T cell biology, including thymocyte development and tolerance, as well as T cell activation and differentiation. Here, we provide an overview of how specific glycosidases and glycosyltransferases contribute to the generation of different types of N-linked glycans and how these post-translational modifications ultimately regulate multiple facets of T cell biology.
Collapse
Affiliation(s)
- Mahmoud Abdelbary
- Department of Molecular Microbiology and Immunology, Oregon Health & Science University, Portland, OR, USA
| | - Jeffrey C. Nolz
- Department of Molecular Microbiology and Immunology, Oregon Health & Science University, Portland, OR, USA
- Department of Cell, Developmental and Cancer Biology, Oregon Health & Science University, Portland, OR, USA
- Department of Dermatology, Oregon Health & Science University, Portland, OR, USA
| |
Collapse
|
16
|
Kianičková K, Pažitná L, Kundalia PH, Pakanová Z, Nemčovič M, Baráth P, Katrlíková E, Šuba J, Trebatická J, Katrlík J. Alterations in the Glycan Composition of Serum Glycoproteins in Attention-Deficit Hyperactivity Disorder. Int J Mol Sci 2023; 24:ijms24108745. [PMID: 37240090 DOI: 10.3390/ijms24108745] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2023] [Revised: 05/04/2023] [Accepted: 05/11/2023] [Indexed: 05/28/2023] Open
Abstract
Changes in protein glycosylation are associated with most biological processes, and the importance of glycomic analysis in the research of disorders is constantly increasing, including in the neurodevelopmental field. We glycoprofiled sera in 10 children with attention-deficit hyperactivity disorder (ADHD) and 10 matching healthy controls for 3 types of samples: whole serum, sera after depletion of abundant proteins (albumin and IgG), and isolated IgG. The analytical methods used were a lectin-based glycoprotein microarray enabling high-throughput glycan analysis and matrix-assisted laser desorption/ionization time-of-flight (MALDI-TOF) mass spectrometry (MS) as a standard method for the identification of glycan structures. For microarray analysis, the samples printed on microarray slides were incubated with biotinylated lectins and detected using the fluorescent conjugate of streptavidin by a microarray scanner. In the ADHD patient samples, we found increased antennary fucosylation, decreased di-/triantennary N-glycans with bisecting N-acetylglucosamine (GlcNAc), and decreased α2-3 sialylation. The results obtained by both independent methods were consistent. The study's sample size and design do not allow far-reaching conclusions to be drawn. In any case, there is a strong demand for a better and more comprehensive diagnosis of ADHD, and the obtained results emphasize that the presented approach brings new horizons to studying functional associations of glycan alterations in ADHD.
Collapse
Affiliation(s)
- Kristína Kianičková
- Institute of Chemistry, Slovak Academy of Sciences, SK-84538 Bratislava, Slovakia
| | - Lucia Pažitná
- Institute of Chemistry, Slovak Academy of Sciences, SK-84538 Bratislava, Slovakia
| | - Paras H Kundalia
- Institute of Chemistry, Slovak Academy of Sciences, SK-84538 Bratislava, Slovakia
| | - Zuzana Pakanová
- Institute of Chemistry, Slovak Academy of Sciences, SK-84538 Bratislava, Slovakia
| | - Marek Nemčovič
- Institute of Chemistry, Slovak Academy of Sciences, SK-84538 Bratislava, Slovakia
| | - Peter Baráth
- Institute of Chemistry, Slovak Academy of Sciences, SK-84538 Bratislava, Slovakia
| | - Eva Katrlíková
- Department of Paediatric Psychiatry, Faculty of Medicine, Comenius University, The National Institute of Children's Diseases, SK-83340 Bratislava, Slovakia
| | - Ján Šuba
- Department of Paediatric Psychiatry, Faculty of Medicine, Comenius University, The National Institute of Children's Diseases, SK-83340 Bratislava, Slovakia
| | - Jana Trebatická
- Department of Paediatric Psychiatry, Faculty of Medicine, Comenius University, The National Institute of Children's Diseases, SK-83340 Bratislava, Slovakia
| | - Jaroslav Katrlík
- Institute of Chemistry, Slovak Academy of Sciences, SK-84538 Bratislava, Slovakia
| |
Collapse
|
17
|
Lee HF, Chi CS. Congenital disorders of glycosylation and infantile epilepsy. Epilepsy Behav 2023; 142:109214. [PMID: 37086590 DOI: 10.1016/j.yebeh.2023.109214] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/18/2022] [Revised: 04/01/2023] [Accepted: 04/04/2023] [Indexed: 04/24/2023]
Abstract
Congenital disorders of glycosylation (CDG) are a group of rare inherited metabolic disorders caused by defects in various defects of protein or lipid glycosylation pathways. The symptoms and signs of CDG usually develop in infancy. Epilepsy is commonly observed in CDG individuals and is often a presenting symptom. These epilepsies can present across the lifespan, share features of refractoriness to antiseizure medications, and are often associated with comorbid developmental delay, psychomotor regression, intellectual disability, and behavioral problems. In this review, we discuss CDG and infantile epilepsy, focusing on an overview of clinical manifestations and electroencephalographic features. Finally, we propose a tiered approach that will permit a clinician to systematically investigate and identify CDG earlier, and furthermore, to provide genetic counseling for the family.
Collapse
Affiliation(s)
- Hsiu-Fen Lee
- Department of Post-Baccalaureate Medicine, College of Medicine, National Chung Hsing University, 145, Xingda Rd., Taichung 402, Taiwan; Division of Pediatric Neurology, Children's Medical Center, Taichung Veterans General Hospital, 1650, Taiwan Boulevard Sec. 4, Taichung 407, Taiwan.
| | - Ching-Shiang Chi
- Division of Pediatric Neurology, Children's Medical Center, Taichung Veterans General Hospital, 1650, Taiwan Boulevard Sec. 4, Taichung 407, Taiwan.
| |
Collapse
|
18
|
Tudor L, Nedic Erjavec G, Nikolac Perkovic M, Konjevod M, Uzun S, Kozumplik O, Mimica N, Lauc G, Svob Strac D, Pivac N. The Association of the Polymorphisms in the FUT8-Related Locus with the Plasma Glycosylation in Post-Traumatic Stress Disorder. Int J Mol Sci 2023; 24:ijms24065706. [PMID: 36982780 PMCID: PMC10056189 DOI: 10.3390/ijms24065706] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2023] [Revised: 03/04/2023] [Accepted: 03/11/2023] [Indexed: 03/19/2023] Open
Abstract
The molecular underpinnings of post-traumatic stress disorder (PTSD) are still unclear due to the complex interactions of genetic, psychological, and environmental factors. Glycosylation is a common post-translational modification of proteins, and different pathophysiological states, such as inflammation, autoimmune diseases, and mental disorders including PTSD, show altered N-glycome. Fucosyltransferase 8 (FUT8) is the enzyme that catalyzes the addition of core fucose on glycoproteins, and mutations in the FUT8 gene are associated with defects in glycosylation and functional abnormalities. This is the first study that investigated the associations of plasma N-glycan levels with FUT8-related rs6573604, rs11621121, rs10483776, and rs4073416 polymorphisms and their haplotypes in 541 PTSD patients and control participants. The results demonstrated that the rs6573604 T allele was more frequent in the PTSD than in the control participants. Significant associations of plasma N-glycan levels with PTSD and FUT8-related polymorphisms were observed. We also detected associations of rs11621121 and rs10483776 polymorphisms and their haplotypes with plasma levels of specific N-glycan species in both the control and PTSD groups. In carriers of different rs6573604 and rs4073416 genotypes and alleles, differences in plasma N-glycan levels were only found in the control group. These molecular findings suggest a possible regulatory role of FUT8-related polymorphisms in glycosylation, the alternations of which could partially explain the development and clinical manifestation of PTSD.
Collapse
Affiliation(s)
- Lucija Tudor
- Laboratory for Molecular Neuropsychiatry, Division of Molecular Medicine, Rudjer Boskovic Institute, 10000 Zagreb, Croatia; (L.T.); (G.N.E.); (M.N.P.); (M.K.)
| | - Gordana Nedic Erjavec
- Laboratory for Molecular Neuropsychiatry, Division of Molecular Medicine, Rudjer Boskovic Institute, 10000 Zagreb, Croatia; (L.T.); (G.N.E.); (M.N.P.); (M.K.)
| | - Matea Nikolac Perkovic
- Laboratory for Molecular Neuropsychiatry, Division of Molecular Medicine, Rudjer Boskovic Institute, 10000 Zagreb, Croatia; (L.T.); (G.N.E.); (M.N.P.); (M.K.)
| | - Marcela Konjevod
- Laboratory for Molecular Neuropsychiatry, Division of Molecular Medicine, Rudjer Boskovic Institute, 10000 Zagreb, Croatia; (L.T.); (G.N.E.); (M.N.P.); (M.K.)
| | - Suzana Uzun
- Department for Biological Psychiatry and Psychogeriatrics, University Hospital Vrapce, 10000 Zagreb, Croatia; (S.U.); (O.K.); (N.M.)
- School of Medicine, University of Zagreb, 10000 Zagreb, Croatia
- Faculty of Education and Rehabilitation Sciences, University of Zagreb, 10000 Zagreb, Croatia
| | - Oliver Kozumplik
- Department for Biological Psychiatry and Psychogeriatrics, University Hospital Vrapce, 10000 Zagreb, Croatia; (S.U.); (O.K.); (N.M.)
- Faculty of Education and Rehabilitation Sciences, University of Zagreb, 10000 Zagreb, Croatia
| | - Ninoslav Mimica
- Department for Biological Psychiatry and Psychogeriatrics, University Hospital Vrapce, 10000 Zagreb, Croatia; (S.U.); (O.K.); (N.M.)
- School of Medicine, University of Zagreb, 10000 Zagreb, Croatia
| | - Gordan Lauc
- Glycobiology Laboratory, Genos Ltd., 10000 Zagreb, Croatia;
| | - Dubravka Svob Strac
- Laboratory for Molecular Neuropsychiatry, Division of Molecular Medicine, Rudjer Boskovic Institute, 10000 Zagreb, Croatia; (L.T.); (G.N.E.); (M.N.P.); (M.K.)
- Correspondence: (D.S.S.); (N.P.)
| | - Nela Pivac
- Laboratory for Molecular Neuropsychiatry, Division of Molecular Medicine, Rudjer Boskovic Institute, 10000 Zagreb, Croatia; (L.T.); (G.N.E.); (M.N.P.); (M.K.)
- University of Applied Sciences Hrvatsko Zagorje Krapina, 49000 Krapina, Croatia
- Correspondence: (D.S.S.); (N.P.)
| |
Collapse
|
19
|
Huang Y, Zhang Z, Chen L. Diagnosis and prognosis of serum Fut8 for epilepsy and refractory epilepsy in children. PLoS One 2023; 18:e0284239. [PMID: 37053181 PMCID: PMC10101470 DOI: 10.1371/journal.pone.0284239] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2023] [Accepted: 03/27/2023] [Indexed: 04/14/2023] Open
Abstract
With adequate serum concentration of antiepileptic drugs, the epilepsy symptoms in many patients still cannot be controlled well. The alteration of glycosyltransferase has obvious influence on the pathogenesis of epilepsy. In this study, we focus on the diagnostic and prognostic value of fucosyltransferase 8 (Fut8) on epilepsy and refractory epilepsy. Serum samples of 199 patients with epilepsy, 59 patients with refractory epilepsy and 22 healthy controls who were diagnosed in Shenzhen Children's hospital from August 2018 to August 2019 were collected. The level of lectins was further analyzed by lectin chip and enzyme linked immunosorbent assay (ELISA). The diagnostic value of serum Fut8 for epilepsy and refractory epilepsy was evaluated by receiver operating characteristic curve. Finally, the difference in the recurrence rate of convulsion in patients with epilepsy or refractory epilepsy within 2 years were observed in different Fut8 expression patients. The concentration of valproic acid (VPA) were significant different between epilepsy and refractory epilepsy group. The expression of α1, 6-fucosylation and Fut8 was significantly increased in the refractory epilepsy group compared with healthy controls. The area under the curve of Fut8 as a biomarker for predicting epilepsy or refractory epilepsy was 0.620 and 0.856, respectively. There was a significant difference in the recurrence rate of convulsion within 2 years in the children with refractory epilepsy (p = 0.0493) not epilepsy (p = 0.1865) between the high and low Fut8 expression groups. Fut8 was one of the effective indicators for the diagnosis and prognosis of refractory epilepsy.
Collapse
Affiliation(s)
- Yunxiu Huang
- Department of Laboratory Medicine, Zhongshan People's Hospital, Zhongshan, Guangdong Province, China
| | - Zhou Zhang
- Department of Pharmacy, Shenzhen Children's Hospital, Shenzhen, Guangdong Province, China
| | - Linmu Chen
- Department of Pharmacy, Zhongshan People's Hospital, Zhongshan, Guangdong Province, China
| |
Collapse
|
20
|
α-1,6-Fucosyltransferase Is Essential for Myogenesis in Zebrafish. Cells 2022; 12:cells12010144. [PMID: 36611938 PMCID: PMC9818595 DOI: 10.3390/cells12010144] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2022] [Revised: 12/08/2022] [Accepted: 12/24/2022] [Indexed: 12/31/2022] Open
Abstract
Glycosylation is an important mechanism regulating various biological processes, including intercellular signaling and adhesion. α-1,6-fucosyltransferase (Fut8) belongs to a family of enzymes that determine the terminal structure of glycans. Fut8 is widely conserved from Caenorhabditis elegans to humans, and its mutants have been reported in humans, mice, and zebrafish. Although mutants show various symptoms, such as spinal deformity and growth retardation, its effects on skeletal muscles are unknown. We aimed to elucidate the function of Fut8 in skeletal muscle using zebrafish and C2C12 cells for evaluation. We observed that most fut8a morphants died at 2 days post-fertilization (dpf) or in earlier developmental stages even at low concentrations of morpholino oligonucleotides (MOs). Mutant juveniles also had small body sizes, and abnormal myocepta and sarcomere structures, suggesting that Fut8a plays important roles in myogenesis. Moreover, treatment of C2C12 cells with 2-fluorofucose (2FF), a fucosylation inhibitor, during cell differentiation dramatically reduced the expression of myogenic genes, such as Myomaker and other myogenic fusion genes, and inhibited myotube formation. These results indicate that Fut8 is an important factor in myogenesis, and myofusion in particular.
Collapse
|
21
|
Al Tuwaijri A, Alyafee Y, Umair M, Alsubait A, Alharbi M, AlEidi H, Ballow M, Aldrees M, Alam Q, Al Abdulrahman A, Alrifai MT, Alfadhel M. Congenital disorder of glycosylation with defective fucosylation 2 (
FCSK
gene defect): The third report in the literature with a mild phenotype. Mol Genet Genomic Med 2022; 11:e2117. [PMID: 36426412 PMCID: PMC10094070 DOI: 10.1002/mgg3.2117] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2022] [Revised: 10/29/2022] [Accepted: 11/01/2022] [Indexed: 11/27/2022] Open
Abstract
BACKGROUND Congenital disorders of glycosylation (CDG) are a group of heterogeneous disorders caused by abnormal lipid or protein glycosylation. Variants in the FCSK gene have been reported to cause CDG. Defective FCSK-induced CDG (FCSK-CDG) has only been reported previously in three unrelated children. METHODS In this study, we genetically and clinically examined a 3-year-old proband with resolved infantile spasms and normal development. Standard whole-exome sequencing (WES) and Sanger sequencing were performed to identify the functional impact of the variant. RESULTS WES revealed a rare biallelic missense variant (c.3013G>C; p.Val1005Leu) in FCSK. RT-qPCR showed a significant depletion in FCSK gene expression in the affected individual. Western blotting revealed reduced FCSK expression at the protein level compared to that in the control. Furthermore, 3D protein modeling suggested changes in the secondary structure, which might affect the overall FCSK protein function. CONCLUSION This study broadens the mutation and phenotypic spectrum of FCSK-associated developmental disorders.
Collapse
Affiliation(s)
- Abeer Al Tuwaijri
- Medical Genomics Research Department King Abdullah International Medical Research Center (KAIMRC) Riyadh Saudi Arabia
- King Saud Bin Abdulaziz University for Health Sciences, King Abdulaziz Medical City, Ministry of National Guard Health Affairs (MNG‐HA) Riyadh Saudi Arabia
- Clinical Laboratory Sciences Department College of Applied Medical Sciences, King Saud Bin Abdulaziz University for Health Sciences (KSAU‐HS) Riyadh Saudi Arabia
| | - Yusra Alyafee
- Medical Genomics Research Department King Abdullah International Medical Research Center (KAIMRC) Riyadh Saudi Arabia
| | - Muhammad Umair
- Medical Genomics Research Department King Abdullah International Medical Research Center (KAIMRC) Riyadh Saudi Arabia
- King Saud Bin Abdulaziz University for Health Sciences, King Abdulaziz Medical City, Ministry of National Guard Health Affairs (MNG‐HA) Riyadh Saudi Arabia
| | - Arwa Alsubait
- King Saud Bin Abdulaziz University for Health Sciences, King Abdulaziz Medical City, Ministry of National Guard Health Affairs (MNG‐HA) Riyadh Saudi Arabia
- Medical Research Core Facilities and Platforms King Abdullah International Medical Research Center (KAIMRC) Riyadh Saudi Arabia
| | - Mashael Alharbi
- Medical Genomics Research Department King Abdullah International Medical Research Center (KAIMRC) Riyadh Saudi Arabia
- King Saud Bin Abdulaziz University for Health Sciences, King Abdulaziz Medical City, Ministry of National Guard Health Affairs (MNG‐HA) Riyadh Saudi Arabia
| | - Hamad AlEidi
- Medical Genomics Research Department King Abdullah International Medical Research Center (KAIMRC) Riyadh Saudi Arabia
- King Saud Bin Abdulaziz University for Health Sciences, King Abdulaziz Medical City, Ministry of National Guard Health Affairs (MNG‐HA) Riyadh Saudi Arabia
| | - Mariam Ballow
- Medical Genomics Research Department King Abdullah International Medical Research Center (KAIMRC) Riyadh Saudi Arabia
- King Saud Bin Abdulaziz University for Health Sciences, King Abdulaziz Medical City, Ministry of National Guard Health Affairs (MNG‐HA) Riyadh Saudi Arabia
| | - Mohammed Aldrees
- Medical Genomics Research Department King Abdullah International Medical Research Center (KAIMRC) Riyadh Saudi Arabia
- King Saud Bin Abdulaziz University for Health Sciences, King Abdulaziz Medical City, Ministry of National Guard Health Affairs (MNG‐HA) Riyadh Saudi Arabia
| | - Qamre Alam
- Medical Genomics Research Department King Abdullah International Medical Research Center (KAIMRC) Riyadh Saudi Arabia
- King Saud Bin Abdulaziz University for Health Sciences, King Abdulaziz Medical City, Ministry of National Guard Health Affairs (MNG‐HA) Riyadh Saudi Arabia
| | - Abdulkareem Al Abdulrahman
- Medical Genomics Research Department King Abdullah International Medical Research Center (KAIMRC) Riyadh Saudi Arabia
- King Saud Bin Abdulaziz University for Health Sciences, King Abdulaziz Medical City, Ministry of National Guard Health Affairs (MNG‐HA) Riyadh Saudi Arabia
| | - Muhammad Talal Alrifai
- Neurology Division, Department of Pediatrics King Abdulaziz Medical City, Ministry of National Guard Health Affairs (NGHA) Riyadh Saudi Arabia
| | - Majid Alfadhel
- Medical Genomics Research Department King Abdullah International Medical Research Center (KAIMRC) Riyadh Saudi Arabia
- King Saud Bin Abdulaziz University for Health Sciences, King Abdulaziz Medical City, Ministry of National Guard Health Affairs (MNG‐HA) Riyadh Saudi Arabia
- Genetics and Precision Medicine Department (GPM) King Abdullah Specialized Children's Hospital Riyadh Saudi Arabia
| |
Collapse
|
22
|
Brasil S, Allocca M, Magrinho SCM, Santos I, Raposo M, Francisco R, Pascoal C, Martins T, Videira PA, Pereira F, Andreotti G, Jaeken J, Kantautas KA, Perlstein EO, Ferreira VDR. Systematic Review: Drug Repositioning for Congenital Disorders of Glycosylation (CDG). Int J Mol Sci 2022; 23:8725. [PMID: 35955863 PMCID: PMC9369176 DOI: 10.3390/ijms23158725] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2022] [Revised: 08/02/2022] [Accepted: 08/02/2022] [Indexed: 11/24/2022] Open
Abstract
Advances in research have boosted therapy development for congenital disorders of glycosylation (CDG), a group of rare genetic disorders affecting protein and lipid glycosylation and glycosylphosphatidylinositol anchor biosynthesis. The (re)use of known drugs for novel medical purposes, known as drug repositioning, is growing for both common and rare disorders. The latest innovation concerns the rational search for repositioned molecules which also benefits from artificial intelligence (AI). Compared to traditional methods, drug repositioning accelerates the overall drug discovery process while saving costs. This is particularly valuable for rare diseases. AI tools have proven their worth in diagnosis, in disease classification and characterization, and ultimately in therapy discovery in rare diseases. The availability of biomarkers and reliable disease models is critical for research and development of new drugs, especially for rare and heterogeneous diseases such as CDG. This work reviews the literature related to repositioned drugs for CDG, discovered by serendipity or through a systemic approach. Recent advances in biomarkers and disease models are also outlined as well as stakeholders' views on AI for therapy discovery in CDG.
Collapse
Affiliation(s)
- Sandra Brasil
- UCIBIO—Applied Molecular Biosciences Unit, School of Science and Technology, NOVA University of Lisbon, 2819-516 Caparica, Portugal
- Associate Laboratory i4HB—Institute for Health and Bioeconomy, School of Science and Technology, Nova University of Lisbon, 2829-516 Caparica, Portugal
- CDG & Allies PPAIN—Professionals and Patient Associations International Network, Department of Life Sciences, School of Science and Technology, NOVA University of Lisbon, 2819-516 Caparica, Portugal
| | - Mariateresa Allocca
- CDG & Allies PPAIN—Professionals and Patient Associations International Network, Department of Life Sciences, School of Science and Technology, NOVA University of Lisbon, 2819-516 Caparica, Portugal
- Institute of Biomolecular Chemistry, National Research Council of Italy, 80078 Pozzuoli, Italy
- Department of Environmental, Biological and Pharmaceutical Sciences and Technologies, University of Campania “Luigi Vanvitelli”, 81100 Caserta, Italy
| | - Salvador C. M. Magrinho
- UCIBIO—Applied Molecular Biosciences Unit, School of Science and Technology, NOVA University of Lisbon, 2819-516 Caparica, Portugal
- Associate Laboratory i4HB—Institute for Health and Bioeconomy, School of Science and Technology, Nova University of Lisbon, 2829-516 Caparica, Portugal
- CDG & Allies PPAIN—Professionals and Patient Associations International Network, Department of Life Sciences, School of Science and Technology, NOVA University of Lisbon, 2819-516 Caparica, Portugal
- LAQV-Requimte, Chemistry Department, School of Science and Technology, Nova University of Lisbon, 2819-516 Caparica, Portugal
| | - Inês Santos
- CDG & Allies PPAIN—Professionals and Patient Associations International Network, Department of Life Sciences, School of Science and Technology, NOVA University of Lisbon, 2819-516 Caparica, Portugal
- Sci and Volunteer Program from School of Science and Technology, NOVA University of Lisbon, 2819-516 Caparica, Portugal
| | - Madalena Raposo
- CDG & Allies PPAIN—Professionals and Patient Associations International Network, Department of Life Sciences, School of Science and Technology, NOVA University of Lisbon, 2819-516 Caparica, Portugal
- Sci and Volunteer Program from School of Science and Technology, NOVA University of Lisbon, 2819-516 Caparica, Portugal
| | - Rita Francisco
- UCIBIO—Applied Molecular Biosciences Unit, School of Science and Technology, NOVA University of Lisbon, 2819-516 Caparica, Portugal
- Associate Laboratory i4HB—Institute for Health and Bioeconomy, School of Science and Technology, Nova University of Lisbon, 2829-516 Caparica, Portugal
- CDG & Allies PPAIN—Professionals and Patient Associations International Network, Department of Life Sciences, School of Science and Technology, NOVA University of Lisbon, 2819-516 Caparica, Portugal
| | - Carlota Pascoal
- UCIBIO—Applied Molecular Biosciences Unit, School of Science and Technology, NOVA University of Lisbon, 2819-516 Caparica, Portugal
- Associate Laboratory i4HB—Institute for Health and Bioeconomy, School of Science and Technology, Nova University of Lisbon, 2829-516 Caparica, Portugal
- CDG & Allies PPAIN—Professionals and Patient Associations International Network, Department of Life Sciences, School of Science and Technology, NOVA University of Lisbon, 2819-516 Caparica, Portugal
| | - Tiago Martins
- CDG & Allies PPAIN—Professionals and Patient Associations International Network, Department of Life Sciences, School of Science and Technology, NOVA University of Lisbon, 2819-516 Caparica, Portugal
- Sci and Volunteer Program from School of Science and Technology, NOVA University of Lisbon, 2819-516 Caparica, Portugal
| | - Paula A. Videira
- UCIBIO—Applied Molecular Biosciences Unit, School of Science and Technology, NOVA University of Lisbon, 2819-516 Caparica, Portugal
- Associate Laboratory i4HB—Institute for Health and Bioeconomy, School of Science and Technology, Nova University of Lisbon, 2829-516 Caparica, Portugal
- CDG & Allies PPAIN—Professionals and Patient Associations International Network, Department of Life Sciences, School of Science and Technology, NOVA University of Lisbon, 2819-516 Caparica, Portugal
| | - Florbela Pereira
- CDG & Allies PPAIN—Professionals and Patient Associations International Network, Department of Life Sciences, School of Science and Technology, NOVA University of Lisbon, 2819-516 Caparica, Portugal
- LAQV-Requimte, Chemistry Department, School of Science and Technology, Nova University of Lisbon, 2819-516 Caparica, Portugal
| | - Giuseppina Andreotti
- Institute of Biomolecular Chemistry, National Research Council of Italy, 80078 Pozzuoli, Italy
| | - Jaak Jaeken
- CDG & Allies PPAIN—Professionals and Patient Associations International Network, Department of Life Sciences, School of Science and Technology, NOVA University of Lisbon, 2819-516 Caparica, Portugal
- Center for Metabolic Diseases, Department of Pediatrics, KU Leuven, 3000 Leuven, Belgium
| | | | | | - Vanessa dos Reis Ferreira
- UCIBIO—Applied Molecular Biosciences Unit, School of Science and Technology, NOVA University of Lisbon, 2819-516 Caparica, Portugal
- Associate Laboratory i4HB—Institute for Health and Bioeconomy, School of Science and Technology, Nova University of Lisbon, 2829-516 Caparica, Portugal
- CDG & Allies PPAIN—Professionals and Patient Associations International Network, Department of Life Sciences, School of Science and Technology, NOVA University of Lisbon, 2819-516 Caparica, Portugal
| |
Collapse
|
23
|
López-Cortés R, Muinelo-Romay L, Fernández-Briera A, Gil-Martín E. Inhibition of α(1,6)fucosyltransferase: Effects on Cell Proliferation, Migration, and Adhesion in an SW480/SW620 Syngeneic Colorectal Cancer Model. Int J Mol Sci 2022; 23:ijms23158463. [PMID: 35955598 PMCID: PMC9369121 DOI: 10.3390/ijms23158463] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2022] [Revised: 07/27/2022] [Accepted: 07/28/2022] [Indexed: 02/01/2023] Open
Abstract
The present study explored the impact of inhibiting α(1,6)fucosylation (core fucosylation) on the functional phenotype of a cellular model of colorectal cancer (CRC) malignization formed by the syngeneic SW480 and SW620 CRC lines. Expression of the FUT8 gene encoding α(1,6)fucosyltransferase was inhibited in tumor line SW480 by a combination of shRNA-based antisense knockdown and Lens culinaris agglutinin (LCA) selection. LCA-resistant clones were subsequently assayed in vitro for proliferation, migration, and adhesion. The α(1,6)FT-inhibited SW480 cells showed enhanced proliferation in adherent conditions, unlike their α(1,6)FT-depleted SW620 counterparts, which displayed reduced proliferation. Under non-adherent conditions, α(1,6)FT-inhibited SW480 cells also showed greater growth capacity than their respective non-targeted control (NTC) cells. However, cell migration decreased in SW480 after FUT8 knockdown, while adhesion to EA.hy926 cells was significantly enhanced. The reported results indicate that the FUT8 knockdown strategy with subsequent selection for LCA-resistant clones was effective in greatly reducing α(1,6)FT expression in SW480 and SW620 CRC lines. In addition, α(1,6)FT impairment affected the proliferation, migration, and adhesion of α(1,6)FT-deficient clones SW480 and SW620 in a tumor stage-dependent manner, suggesting that core fucosylation has a dynamic role in the evolution of CRC.
Collapse
Affiliation(s)
- Rubén López-Cortés
- Doctoral Program in Methods and Applications in Life Sciences, Faculty of Biology, Campus Lagoas-Marcosende, Universidade de Vigo, 36310 Vigo, Spain;
| | - Laura Muinelo-Romay
- Liquid Biopsy Analysis Unit, Translational Medical Oncology (Oncomet), Health Research Institute of Santiago de Compostela (IDIS), CIBERONC, Travesía da Choupana, 15706 Santiago de Compostela, Spain;
| | - Almudena Fernández-Briera
- Department of Biochemistry, Genetics and Immunology, Faculty of Biology, Campus Lagoas-Marcosende, Universidade de Vigo, 36310 Vigo, Spain;
| | - Emilio Gil-Martín
- Department of Biochemistry, Genetics and Immunology, Faculty of Biology, Campus Lagoas-Marcosende, Universidade de Vigo, 36310 Vigo, Spain;
- Correspondence: ; Tel.: +34-(986)-812-570
| |
Collapse
|
24
|
Loss of core-fucosylation of SPARC impairs collagen binding and contributes to COPD. Cell Mol Life Sci 2022; 79:348. [PMID: 35670884 PMCID: PMC9174126 DOI: 10.1007/s00018-022-04381-4] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2022] [Revised: 04/20/2022] [Accepted: 05/16/2022] [Indexed: 12/05/2022]
Abstract
Chronic obstructive pulmonary disease (COPD) is a progressive lung disease with high morbidity and mortality worldwide. Although several mechanisms to account for deleterious immune effects were proposed, molecular description for the underlying alveolar structural alterations for COPD is lacking. Here, silencing of α1,6-fucosyltransferase (Fut8), the enzyme for core-fucosylation and highly expressed in lung stem cells, resulted in alveolar structural changes in lung organoids, recapitulating COPD. Site-specific mass spectrometry analysis demonstrated that the secreted protein acidic and rich in cysteine (SPARC), which binds collagen, contains a core-fucosylation site in its VCSNDNcfK glycopeptide. Biacore assay showed markedly reduced collagen binding of SPARC lacking core fucosylation. Molecular dynamics analysis revealed that core fucosylation of SPARC-induced dynamic conformational changes in its N-glycan, allowing terminal galactose and N-acetylglucosamine to interact with K150, P261 and H264 residues, thereby promoting collagen binding. Site-specific mutagenesis of these residues also resulted in low affinity for collagen binding. Moreover, loss of collagen and decline of core fucosylation were observed in COPD lung tissues. These findings provide a new mechanistic insight into the role of core fucosylation of SPARC in cell–matrix communication and contribution to the abnormal alveolar structures in COPD.
Collapse
|
25
|
Boyer SW, Johnsen C, Morava E. Nutrition interventions in congenital disorders of glycosylation. Trends Mol Med 2022; 28:463-481. [PMID: 35562242 DOI: 10.1016/j.molmed.2022.04.003] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2021] [Revised: 03/30/2022] [Accepted: 04/04/2022] [Indexed: 12/13/2022]
Abstract
Congenital disorders of glycosylation (CDG) are a group of more than 160 inborn errors of metabolism affecting multiple pathways of protein and lipid glycosylation. Patients present with a wide range of symptoms and therapies are only available for very few subtypes. Specific nutritional treatment options for certain CDG types include oral supplementation of monosaccharide sugars, manganese, uridine, or pyridoxine. Additional management includes specific diets (i.e., complex carbohydrate or ketogenic diet), iron supplementation, and albumin infusions. We review the dietary management in CDG with a focus on two subgroups: N-linked glycosylation defects and GPI-anchor disorders.
Collapse
Affiliation(s)
- Suzanne W Boyer
- Department of Clinical Genomics, Mayo Clinic, Rochester, MN, USA
| | - Christin Johnsen
- Department of Clinical Genomics, Mayo Clinic, Rochester, MN, USA
| | - Eva Morava
- Department of Clinical Genomics, Mayo Clinic, Rochester, MN, USA.
| |
Collapse
|
26
|
Wu Z, Lin T, Kang P, Zhuang Z, Wang H, He W, Wei Q, Li Z. Overexpression of fucosyltransferase 8 reverses the inhibitory effect of high-dose dexamethasone on osteogenic response of MC3T3-E1 preosteoblasts. PeerJ 2021; 9:e12380. [PMID: 34966572 PMCID: PMC8667747 DOI: 10.7717/peerj.12380] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2021] [Accepted: 10/04/2021] [Indexed: 12/15/2022] Open
Abstract
Background Core fucosylation catalyzed by FUT8 is essential for TGF-β binding to TGF-β receptors. Methods Indirect TGF-β1 binding assay was used to evaluate the ability of TGF-β1 to bind to TGFBRs, Alizarin red and alkaline phosphatase staining were used to detect osteogenic differentiation and mineralization ability , western blot and quantitative RT-PCR were used to measure the differential expression of osteogenesis-related proteins and genes. Plasmid-mediated gain-of-function study. The scale of core fucosylation modification was detected by Lectin-blot and LCA laser confocal. Results Our results showed that compared with vehicle treatment, high-dose (10−6 and 10−5 M) dexamethasone significantly inhibited cell proliferation, osteogenic differentiation, and FUT8 mRNA expression while promoting mRNA expression of adipogenesis-related genes in MC3T3-E1 cells, suggesting that downregulation of FUT8 is involved in the inhibitory effect of high-dose dexamethasone on osteogenesis. Overexpression of FUT8 significantly promoted osteogenic differentiation and activated TGF-β/Smad signaling in MC3T3-E1 cells in the presence of high-dose dexamethasone, suggesting that FUT8 reverses the inhibitory effect of high-dose dexamethasone on osteogenesis. In addition, lectin fluorescent staining and blotting showed that overexpression of FUT8 significantly reversed the inhibitory effects of high-dose dexamethasone on core fucosylation of TGFBR1 and TGFBR2. Furthermore, indirect TGF-β1 binding assay showed that overexpression of FUT8 remarkably promoted TGF-β1 binding to TGFBRs in MC3T3-E1 cells in the presence of high-dose dexamethasone. Conclusions Taken together, these results suggest that overexpression of FUT8 facilitates counteracting the inhibitory effect of dexamethasone on TGF-β signaling and osteogenesis.
Collapse
Affiliation(s)
- Zhiming Wu
- Guangzhou University of Chinese Medicine, Guangzhou, China.,Department of Joint Orthopaedic, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China.,Lingnan Medical Research Center of Guangzhou University of Chinese Medicine, Guangzhou, China.,Shenzhen Hospital (Futian) of Guangzhou University of Chinese Medicine, Shenzhen City, Guangdong Province, China
| | - Tianye Lin
- Guangzhou University of Chinese Medicine, Guangzhou, China.,Department of Joint Orthopaedic, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China.,Lingnan Medical Research Center of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Pan Kang
- Guangzhou University of Chinese Medicine, Guangzhou, China.,Department of Joint Orthopaedic, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China.,Lingnan Medical Research Center of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Zhikun Zhuang
- Department of Joint Orthopaedic, Quanzhou Orthopedic-Traumatological Hospital of Fujian Traditional Chinese Medicine University, Quanzhou, China
| | - Haibin Wang
- Guangzhou University of Chinese Medicine, Guangzhou, China.,Department of Joint Orthopaedic, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China.,Lingnan Medical Research Center of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Wei He
- Guangzhou University of Chinese Medicine, Guangzhou, China.,Department of Joint Orthopaedic, The Third Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Qiushi Wei
- Guangzhou University of Chinese Medicine, Guangzhou, China.,Department of Joint Orthopaedic, The Third Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Ziqi Li
- Guangzhou University of Chinese Medicine, Guangzhou, China.,Department of Joint Orthopaedic, The Third Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou, China
| |
Collapse
|
27
|
Sanda M, Ahn J, Kozlik P, Goldman R. Analysis of site and structure specific core fucosylation in liver cirrhosis using exoglycosidase-assisted data-independent LC-MS/MS. Sci Rep 2021; 11:23273. [PMID: 34857845 PMCID: PMC8639754 DOI: 10.1038/s41598-021-02838-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2021] [Accepted: 11/03/2021] [Indexed: 12/16/2022] Open
Abstract
Carbohydrates form one of the major groups of biological macromolecules in living organisms. Many biological processes including protein folding, stability, immune response, and receptor activation are regulated by glycosylation. Fucosylation of proteins regulates such processes and is associated with various diseases including autoimmunity and cancer. Mass spectrometry efficiently identifies structures of fucosylated glycans or sites of core fucosylated N-glycopeptides but quantification of the glycopeptides remains less explored. We performed experiments that facilitate quantitative analysis of the core fucosylation of proteins with partial structural resolution of the glycans and we present results of the mass spectrometric SWATH-type DIA analysis of relative abundances of the core fucosylated glycoforms of 45 glycopeptides to their nonfucosylated glycoforms derived from 18 serum proteins in liver disease of different etiologies. Our results show that a combination of soft fragmentation with exoglycosidases is efficient at the assignment and quantification of the core fucosylated N-glycoforms at specific sites of protein attachment. In addition, our results show that disease-associated changes in core fucosylation are peptide-dependent and further differ by branching of the core fucosylated glycans. Further studies are needed to verify whether tri- and tetra-antennary core fucosylated glycopeptides could be used as markers of liver disease progression.
Collapse
Affiliation(s)
- Miloslav Sanda
- Department of Oncology, Lombardi Comprehensive Cancer Center, Georgetown University, Washington, DC, 20057, USA. .,Clinical and Translational Glycoscience Research Center, Georgetown University, Washington, DC, 20057, USA.
| | - Jaeil Ahn
- Department of Biostatistics, Bioinformatics and Biomathematics, Georgetown University Medical Center, Washington, DC, 20057, USA
| | - Petr Kozlik
- Department of Analytical Chemistry, Faculty of Science, Charles University, Prague, Czech Republic
| | - Radoslav Goldman
- Department of Oncology, Lombardi Comprehensive Cancer Center, Georgetown University, Washington, DC, 20057, USA.,Department of Biochemistry and Molecular and Cellular Biology, Georgetown University, Washington, DC, 20057, USA.,Clinical and Translational Glycoscience Research Center, Georgetown University, Washington, DC, 20057, USA
| |
Collapse
|
28
|
Hüllen A, Falkenstein K, Weigel C, Huidekoper H, Naumann-Bartsch N, Spenger J, Feichtinger RG, Schaefers J, Frenz S, Kotlarz D, Momen T, Khoshnevisan R, Riedhammer KM, Santer R, Herget T, Rennings A, Lefeber DJ, Mayr JA, Thiel C, Wortmann SB. Congenital disorders of glycosylation with defective fucosylation. J Inherit Metab Dis 2021; 44:1441-1452. [PMID: 34389986 DOI: 10.1002/jimd.12426] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/19/2021] [Revised: 08/02/2021] [Accepted: 08/10/2021] [Indexed: 11/05/2022]
Abstract
Fucosylation is essential for intercellular and intracellular recognition, cell-cell interaction, fertilization, and inflammatory processes. Only five types of congenital disorders of glycosylation (CDG) related to an impaired fucosylation have been described to date: FUT8-CDG, FCSK-CDG, POFUT1-CDG SLC35C1-CDG, and the only recently described GFUS-CDG. This review summarizes the clinical findings of all hitherto known 25 patients affected with those defects with regard to their pathophysiology and genotype. In addition, we describe five new patients with novel variants in the SLC35C1 gene. Furthermore, we discuss the efficacy of fucose therapy approaches within the different defects.
Collapse
Affiliation(s)
- Andreas Hüllen
- Centre for Child and Adolescent Medicine, Department 1, University of Heidelberg, Heidelberg, Germany
| | - Kristina Falkenstein
- Centre for Child and Adolescent Medicine, Department 1, University of Heidelberg, Heidelberg, Germany
| | - Corina Weigel
- Department of Pediatrics and Adolescent Medicine, Friedrich-Alexander-University Erlangen-Nuremberg, Erlangen, Germany
| | - Hidde Huidekoper
- Department of Pediatrics, Center for Lysosomal and Metabolic Diseases, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Nora Naumann-Bartsch
- Department of Pediatrics and Adolescent Medicine, Friedrich-Alexander-University Erlangen-Nuremberg, Erlangen, Germany
| | - Johannes Spenger
- University Children's Hospital, Salzburger Landeskliniken (SALK) and Paracelsus Medical University (PMU), Salzburg, Austria
| | - René G Feichtinger
- University Children's Hospital, Salzburger Landeskliniken (SALK) and Paracelsus Medical University (PMU), Salzburg, Austria
| | - Jacqueline Schaefers
- Department of Pediatrics, Center for Lysosomal and Metabolic Diseases, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Stephanie Frenz
- Department of Pediatrics, Dr von Hauner Children's Hospital, University Hospital, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Daniel Kotlarz
- Department of Pediatrics, Dr von Hauner Children's Hospital, University Hospital, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Tooba Momen
- Department of Asthma, Allergy and Clinical Immunology, Child Growth and Development Research Center, Research Institute for Primordial Prevention of Non-Communicable Disease, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Razieh Khoshnevisan
- Department of Immunology, Medical Faculty, Isfahan University of Medical Sciences, Isfahan, Iran
- Acquired Immunodeficiency Research Center, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Korbinian M Riedhammer
- Institute of Human Genetics, Klinikum Rechts der Isar, School of Medicine, Technical University of Munich, Munich, Germany
- Department of Nephrology, Klinikum Rechts der Isar, School of Medicine, Technical University of Munich, Munich, Germany
| | - René Santer
- Department of Pediatrics, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Theresia Herget
- Institute of Human Genetics, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Alexander Rennings
- Department of Pediatrics, Radboud Center for Mitochondrial Medicine, Amalia Children's Hospital, Nijmegen, The Netherlands
| | - Dirk J Lefeber
- Department of Neurology, Translational Metabolic Laboratory, Donders Center for Brain, Cognition and Behavior, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Johannes A Mayr
- University Children's Hospital, Salzburger Landeskliniken (SALK) and Paracelsus Medical University (PMU), Salzburg, Austria
| | - Christian Thiel
- Centre for Child and Adolescent Medicine, Department 1, University of Heidelberg, Heidelberg, Germany
| | - Saskia B Wortmann
- University Children's Hospital, Salzburger Landeskliniken (SALK) and Paracelsus Medical University (PMU), Salzburg, Austria
- Department of Pediatrics, Radboud Center for Mitochondrial Medicine, Amalia Children's Hospital, Nijmegen, The Netherlands
| |
Collapse
|
29
|
Park JH, Marquardt T. Treatment Options in Congenital Disorders of Glycosylation. Front Genet 2021; 12:735348. [PMID: 34567084 PMCID: PMC8461064 DOI: 10.3389/fgene.2021.735348] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2021] [Accepted: 08/23/2021] [Indexed: 12/15/2022] Open
Abstract
Despite advances in the identification and diagnosis of congenital disorders of glycosylation (CDG), treatment options remain limited and are often constrained to symptomatic management of disease manifestations. However, recent years have seen significant advances in treatment and novel therapies aimed both at the causative defect and secondary disease manifestations have been transferred from bench to bedside. In this review, we aim to give a detailed overview of the available therapies and rising concepts to treat these ultra-rare diseases.
Collapse
Affiliation(s)
- Julien H Park
- Department of General Pediatrics, Metabolic Diseases, University Children's Hospital Münster, Münster, Germany
| | - Thorsten Marquardt
- Department of General Pediatrics, Metabolic Diseases, University Children's Hospital Münster, Münster, Germany
| |
Collapse
|
30
|
Feichtinger RG, Hüllen A, Koller A, Kotzot D, Grote V, Rapp E, Hofbauer P, Brugger K, Thiel C, Mayr JA, Wortmann SB. A spoonful of L-fucose-an efficient therapy for GFUS-CDG, a new glycosylation disorder. EMBO Mol Med 2021; 13:e14332. [PMID: 34468083 PMCID: PMC8422078 DOI: 10.15252/emmm.202114332] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Revised: 07/26/2021] [Accepted: 07/28/2021] [Indexed: 12/29/2022] Open
Abstract
Congenital disorders of glycosylation are a genetically and phenotypically heterogeneous family of diseases affecting the co- and posttranslational modification of proteins. Using exome sequencing, we detected biallelic variants in GFUS (NM_003313.4) c.[632G>A];[659C>T] (p.[Gly211Glu];[Ser220Leu]) in a patient presenting with global developmental delay, mild coarse facial features and faltering growth. GFUS encodes GDP-L-fucose synthase, the terminal enzyme in de novo synthesis of GDP-L-fucose, required for fucosylation of N- and O-glycans. We found reduced GFUS protein and decreased GDP-L-fucose levels leading to a general hypofucosylation determined in patient's glycoproteins in serum, leukocytes, thrombocytes and fibroblasts. Complementation of patient fibroblasts with wild-type GFUS cDNA restored fucosylation. Making use of the GDP-L-fucose salvage pathway, oral fucose supplementation normalized fucosylation of proteins within 4 weeks as measured in serum and leukocytes. During the follow-up of 19 months, a moderate improvement of growth was seen, as well as a clear improvement of cognitive skills as measured by the Kaufmann ABC and the Nijmegen Pediatric CDG Rating Scale. In conclusion, GFUS-CDG is a new glycosylation disorder for which oral L-fucose supplementation is promising.
Collapse
Affiliation(s)
- René G Feichtinger
- University Children’s HospitalSalzburger Landeskliniken (SALK) and Paracelsus Medical University (PMU)SalzburgAustria
| | - Andreas Hüllen
- Department PediatricsCentre for Child and Adolescent MedicineUniversity of HeidelbergHeidelbergGermany
| | - Andreas Koller
- Research Program for Experimental OphthalmologyDepartment of Ophthalmology and OptometrySalzburger Landeskliniken (SALK) and Paracelsus Medical University (PMU)SalzburgAustria
| | - Dieter Kotzot
- Clinical Genetics UnitSalzburger Landeskliniken (SALK) and Paracelsus Medical University (PMU)SalzburgAustria
| | - Valerian Grote
- Max Planck Institute for Dynamics of Complex Technical Systems, Bioprocess EngineeringMagdeburgGermany
| | - Erdmann Rapp
- Max Planck Institute for Dynamics of Complex Technical Systems, Bioprocess EngineeringMagdeburgGermany
- glyXera GmbHMagdeburgGermany
| | - Peter Hofbauer
- Department of ProductionLandesapotheke SalzburgHospital PharmacySalzburgAustria
| | - Karin Brugger
- University Children’s HospitalSalzburger Landeskliniken (SALK) and Paracelsus Medical University (PMU)SalzburgAustria
| | - Christian Thiel
- Department PediatricsCentre for Child and Adolescent MedicineUniversity of HeidelbergHeidelbergGermany
| | - Johannes A Mayr
- University Children’s HospitalSalzburger Landeskliniken (SALK) and Paracelsus Medical University (PMU)SalzburgAustria
| | - Saskia B Wortmann
- University Children’s HospitalSalzburger Landeskliniken (SALK) and Paracelsus Medical University (PMU)SalzburgAustria
- Department of PediatricsAmalia Children’s HospitalRadboud Center for Mitochondrial MedicineRadboudumcNijmegenThe Netherlands
| |
Collapse
|
31
|
Zhou Q, Xie Y, Lam M, Lebrilla CB. N-Glycomic Analysis of the Cell Shows Specific Effects of Glycosyl Transferase Inhibitors. Cells 2021; 10:cells10092318. [PMID: 34571967 PMCID: PMC8465854 DOI: 10.3390/cells10092318] [Citation(s) in RCA: 9] [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: 07/21/2021] [Revised: 08/31/2021] [Accepted: 09/02/2021] [Indexed: 11/16/2022] Open
Abstract
Glycomic profiling methods were used to determine the effect of metabolic inhibitors on glycan production. These inhibitors are commonly used to alter the cell surface glycosylation. However, structural analysis of the released glycans has been limited. In this research, the cell membranes were enriched and the glycans were released to obtain the N-glycans of the glycocalyx. Glycomic analysis using liquid chromatography–mass spectrometry (LC–MS) with a PGC chip column was used to profile the structures in the cell membrane. Glycans of untreated cells were compared to glycans of cells treated with inhibitors, including kifunensine, which inhibits the formation of complex- and hybrid-type structures, 2,4,7,8,9-Penta-O-acetyl-N-acetyl-3-fluoro-b-d-neuraminic acid methyl ester for sialylated glycans, 2-deoxy-2-fluorofucose, and 6-alkynyl fucose for fucosylated glycans. Kifunensine was the most effective, converting nearly 95% of glycans to high mannose types. The compound 6-alkynyl fucose inhibited some fucosylation but also incorporated into the glycan structure. Proteomic analysis of the enriched membrane for the four inhibitors showed only small changes in the proteome accompanied by large changes in the N-glycome for Caco-2. Future works may use these inhibitors to study the cellular behavior associated with the alteration of glycosylation in various biological systems, e.g., viral and bacterial infection, drug binding, and cell–cell interactions.
Collapse
Affiliation(s)
- Qingwen Zhou
- Department of Chemistry, University of California, Davis, CA 95616, USA; (Q.Z.); (Y.X.); (M.L.)
| | - Yixuan Xie
- Department of Chemistry, University of California, Davis, CA 95616, USA; (Q.Z.); (Y.X.); (M.L.)
| | - Matthew Lam
- Department of Chemistry, University of California, Davis, CA 95616, USA; (Q.Z.); (Y.X.); (M.L.)
| | - Carlito B. Lebrilla
- Department of Chemistry, University of California, Davis, CA 95616, USA; (Q.Z.); (Y.X.); (M.L.)
- Department of Biochemistry, University of California, Davis, CA 95616, USA
- Correspondence:
| |
Collapse
|
32
|
Jia L, Li J, Li P, Liu D, Li J, Shen J, Zhu B, Ma C, Zhao T, Lan R, Dang L, Li W, Sun S. Site-specific glycoproteomic analysis revealing increased core-fucosylation on FOLR1 enhances folate uptake capacity of HCC cells to promote EMT. Am J Cancer Res 2021; 11:6905-6921. [PMID: 34093861 PMCID: PMC8171077 DOI: 10.7150/thno.56882] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2020] [Accepted: 04/14/2021] [Indexed: 12/24/2022] Open
Abstract
Rationale: Epithelial-mesenchymal transition (EMT) has been recognized as an important step toward high invasion and metastasis of many cancers including hepatocellular carcinoma (HCC), while the mechanism for EMT promotion is still ambiguous. Methods: The dynamic alterations of site-specific glycosylation during HGF/TGF-β1-induced EMT process of three HCC cell lines were systematically investigated using precision glycoproteomic methods. The possible roles of EMT-related glycoproteins and site-specific glycans were further confirmed by various molecular biological approaches. Results: Using mass spectrometry-based glycoproteomic methods, we totally identified 2306 unique intact glycopeptides from SMMC-7721 and HepG2 cell lines, and found that core-fucosylated glycans were accounted for the largest proportion of complex N-glycans. Through quantification analysis of intact glycopeptides, we found that the majority of core-fucosylated intact glycopeptides from folate receptor α (FOLR1) were up-regulated in the three HGF-treated cell lines. Similarly, core-fucosylation of FOLR1 were up-regulated in SMMC-7721 and Hep3B cells with TGF-β1 treatment. Using molecular approaches, we further demonstrated that FUT8 was a driver for HGF/TGF-β1-induced EMT. The silencing of FUT8 reduced core-fucosylation and partially blocked the progress of HGF-induced EMT. Finally, we confirmed that the level of core-fucosylation on FOLR1 especially at the glycosite Asn-201 positively regulated the cellular uptake capacity of folates, and enhanced uptake of folates could promote the EMT of HCC cells. Conclusions: Based on the results, we proposed a potential pathway for HGF or TGF-β1-induced EMT of HCC cells: HGF or TGF-β1 treatment of HCC cells can increase the expression of glycosyltransferase FUT8 to up-regulate the core-fucosylation of N-glycans on glycoproteins including the FOLR1; core-fucosylation on FOLR1 can then enhance the folate uptake capacity to finally promote the EMT progress of HCC cells.
Collapse
|
33
|
Ohkawa Y, Harada Y, Taniguchi N. Keratan sulfate-based glycomimetics using Langerin as a target for COPD: lessons from studies on Fut8 and core fucose. Biochem Soc Trans 2021; 49:441-453. [PMID: 33616615 PMCID: PMC7924997 DOI: 10.1042/bst20200780] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2020] [Revised: 01/14/2021] [Accepted: 01/29/2021] [Indexed: 12/19/2022]
Abstract
Glycosylation represents one of the most abundant posttranslational modification of proteins. Glycosylation products are diverse and are regulated by the cooperative action of various glycosyltransferases, glycosidases, substrates thereof: nucleoside sugars and their transporters, and chaperons. In this article, we focus on a glycosyltransferase, α1,6-fucosyltransferase (Fut8) and its product, the core fucose structure on N-glycans, and summarize the potential protective functions of this structure against emphysema and chronic obstructive pulmonary disease (COPD). Studies of FUT8 and its enzymatic product, core fucose, are becoming an emerging area of interest in various fields of research including inflammation, cancer and therapeutics. This article discusses what we can learn from studies of Fut8 and core fucose by using knockout mice or in vitro studies that were conducted by our group as well as other groups. We also include a discussion of the potential protective functions of the keratan sulfate (KS) disaccharide, namely L4, against emphysema and COPD as a glycomimetic. Glycomimetics using glycan analogs is one of the more promising therapeutics that compensate for the usual therapeutic strategy that involves targeting the genome and the proteome. These typical glycans using KS derivatives as glycomimetics, will likely become a clue to the development of novel and effective therapeutic strategies.
Collapse
MESH Headings
- Animals
- Antigens, CD/genetics
- Antigens, CD/metabolism
- Antigens, Surface/genetics
- Antigens, Surface/metabolism
- Antigens, Surface/physiology
- Biomimetic Materials/chemistry
- Biomimetic Materials/therapeutic use
- Fucose/metabolism
- Fucosyltransferases/physiology
- Glycosylation
- Humans
- Keratan Sulfate/chemistry
- Lectins, C-Type/antagonists & inhibitors
- Lectins, C-Type/genetics
- Lectins, C-Type/metabolism
- Lectins, C-Type/physiology
- Mannose-Binding Lectins/antagonists & inhibitors
- Mannose-Binding Lectins/genetics
- Mannose-Binding Lectins/metabolism
- Mannose-Binding Lectins/physiology
- Mice
- Mice, Knockout
- Molecular Targeted Therapy/methods
- Polysaccharides/chemistry
- Polysaccharides/metabolism
- Pulmonary Disease, Chronic Obstructive/drug therapy
- Pulmonary Disease, Chronic Obstructive/genetics
- Pulmonary Disease, Chronic Obstructive/metabolism
Collapse
Affiliation(s)
- Yuki Ohkawa
- Department of Glyco-Oncology and Medical Biochemistry, Osaka International Cancer Institute, 3-1-69 Otemae, Chuo-ku, Osaka 541-8567, Japan
| | - Yoichiro Harada
- Department of Glyco-Oncology and Medical Biochemistry, Osaka International Cancer Institute, 3-1-69 Otemae, Chuo-ku, Osaka 541-8567, Japan
| | - Naoyuki Taniguchi
- Department of Glyco-Oncology and Medical Biochemistry, Osaka International Cancer Institute, 3-1-69 Otemae, Chuo-ku, Osaka 541-8567, Japan
| |
Collapse
|
34
|
Abstract
N-glycosylation is a highly conserved glycan modification, and more than 7000 proteins are N-glycosylated in humans. N-glycosylation has many biological functions such as protein folding, trafficking, and signal transduction. Thus, glycan modification to proteins is profoundly involved in numerous physiological and pathological processes. The N-glycan precursor is biosynthesized in the endoplasmic reticulum (ER) from dolichol phosphate by sequential enzymatic reactions to generate the dolichol-linked oligosaccharide composed of 14 sugar residues, Glc3Man9GlcNAc2. The oligosaccharide is then en bloc transferred to the consensus sequence N-X-S/T (X represents any amino acid except proline) of nascent proteins. Subsequently, the N-glycosylated nascent proteins enter the folding step, in which N-glycans contribute largely to attaining the correct protein fold by recruiting the lectin-like chaperones, calnexin, and calreticulin. Despite the N-glycan-dependent folding process, some glycoproteins do not fold correctly, and these misfolded glycoproteins are destined to degradation by proteasomes in the cytosol. Properly folded proteins are transported to the Golgi, and N-glycans undergo maturation by the sequential reactions of glycosidases and glycosyltransferases, generating complex-type N-glycans. N-Acetylglucosaminyltransferases (GnT-III, GnT-IV, and GnT-V) produce branched N-glycan structures, affording a higher complexity to N-glycans. In this chapter, we provide an overview of the biosynthetic pathway of N-glycans in the ER and Golgi.
Collapse
|
35
|
Wongtrakul-Kish K, Herbert BR, Packer NH. Bisecting GlcNAc Protein N-Glycosylation Is Characteristic of Human Adipogenesis. J Proteome Res 2020; 20:1313-1327. [PMID: 33383989 DOI: 10.1021/acs.jproteome.0c00702] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Human adipose tissue contains a major source of adipose-derived stem cells (ADSCs) that have the ability to differentiate into various cell types: in vitro, ADSCs can differentiate into mesenchymal lineages including adipocytes, while in vivo, ADSCs become mature adipocytes. Protein glycosylation has been shown to change in stem cell differentiation, and while ADSCs have been acknowledged for their therapeutic potential, little is known about protein glycosylation during human ADSC adipogenic differentiation. In the present study, the global membrane protein glycosylation of native adipocytes was compared to ADSCs from the same individuals as a model of in vivo adipogenesis. For in vitro adipogenesis, ADSCs were adipogenically differentiated in cell culture using an optimized, large-scale differentiation procedure. The membrane glycome of the differentiated ADSCs (dADSCs) was compared with mature adipocytes and the progenitor ADSCs. A total of 137 glycan structures were characterized across the three cell types using PGC-LC coupled with negative-ion electrospray ionization mass spectrometry (ESI-MS)/MS. Significantly higher levels of bisecting GlcNAc-type N-glycans were detected in mature adipocytes (32.1% of total glycans) and in in vitro dADSC progeny (1.9% of total glycans) compared to ADSCs. This was further correlated by the mRNA expression of the MGAT3 gene responsible for the enzymatic synthesis of this structural type. The bisecting GlcNAc structures were found on the majority of human native adipocyte membrane proteins, suggesting an important role in human adipocyte biology. Core fucosylation was also significantly increased during in vivo adipogenesis but did not correlate with an increase in Fut8 gene transcript. Unexpectedly, low abundance structures carrying rare β-linked Gal-Gal termini were also detected. Overall, the N-glycan profiles of the in vitro differentiated progeny did not reflect native adipocytes, and the results show that bisecting GlcNAc structures are a characteristic feature of human adipocyte membrane protein N-glycosylation. Raw MS files are available on GlycoPOST (ID: GPST000153 https://glycopost.glycosmos.org/).
Collapse
Affiliation(s)
- Katherine Wongtrakul-Kish
- Biomolecular Discovery Research Centre, Department of Molecular Sciences, Macquarie University, Sydney, NSW 2109, Australia.,ARC Centre for Nanoscale BioPhotonics, Department of Molecular Sciences, Macquarie University, Sydney, NSW 2109, Australia
| | - Benjamin R Herbert
- Biomolecular Discovery Research Centre, Department of Molecular Sciences, Macquarie University, Sydney, NSW 2109, Australia
| | - Nicolle H Packer
- Biomolecular Discovery Research Centre, Department of Molecular Sciences, Macquarie University, Sydney, NSW 2109, Australia.,ARC Centre for Nanoscale BioPhotonics, Department of Molecular Sciences, Macquarie University, Sydney, NSW 2109, Australia
| |
Collapse
|
36
|
Boruah BM, Kadirvelraj R, Liu L, Ramiah A, Li C, Zong G, Bosman GP, Yang JY, Wang LX, Boons GJ, Wood ZA, Moremen KW. Characterizing human α-1,6-fucosyltransferase (FUT8) substrate specificity and structural similarities with related fucosyltransferases. J Biol Chem 2020; 295:17027-17045. [PMID: 33004438 PMCID: PMC7863877 DOI: 10.1074/jbc.ra120.014625] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2020] [Revised: 09/28/2020] [Indexed: 12/18/2022] Open
Abstract
Mammalian Asn-linked glycans are extensively processed as they transit the secretory pathway to generate diverse glycans on cell surface and secreted glycoproteins. Additional modification of the glycan core by α-1,6-fucose addition to the innermost GlcNAc residue (core fucosylation) is catalyzed by an α-1,6-fucosyltransferase (FUT8). The importance of core fucosylation can be seen in the complex pathological phenotypes of FUT8 null mice, which display defects in cellular signaling, development, and subsequent neonatal lethality. Elevated core fucosylation has also been identified in several human cancers. However, the structural basis for FUT8 substrate specificity remains unknown.Here, using various crystal structures of FUT8 in complex with a donor substrate analog, and with four distinct glycan acceptors, we identify the molecular basis for FUT8 specificity and activity. The ordering of three active site loops corresponds to an increased occupancy for bound GDP, suggesting an induced-fit folding of the donor-binding subsite. Structures of the various acceptor complexes were compared with kinetic data on FUT8 active site mutants and with specificity data from a library of glycan acceptors to reveal how binding site complementarity and steric hindrance can tune substrate affinity. The FUT8 structure was also compared with other known fucosyltransferases to identify conserved and divergent structural features for donor and acceptor recognition and catalysis. These data provide insights into the evolution of modular templates for donor and acceptor recognition among GT-B fold glycosyltransferases in the synthesis of diverse glycan structures in biological systems.
Collapse
Affiliation(s)
- Bhargavi M Boruah
- Complex Carbohydrate Research Center, University of Georgia, Athens, Georgia, USA
| | - Renuka Kadirvelraj
- Department of Biochemistry and Molecular Biology, University of Georgia, Athens, Georgia, USA
| | - Lin Liu
- Complex Carbohydrate Research Center, University of Georgia, Athens, Georgia, USA
| | - Annapoorani Ramiah
- Complex Carbohydrate Research Center, University of Georgia, Athens, Georgia, USA
| | - Chao Li
- Department of Chemistry and Biochemistry, University of Maryland, College Park, Maryland, USA
| | - Guanghui Zong
- Department of Chemistry and Biochemistry, University of Maryland, College Park, Maryland, USA
| | - Gerlof P Bosman
- Department of Chemical Biology and Drug Discovery, Utrecht Institute for Pharmaceutical Sciences, and Utrecht Institute for Pharmaceutical Sciences, Utrecht, The Netherlands
| | - Jeong-Yeh Yang
- Complex Carbohydrate Research Center, University of Georgia, Athens, Georgia, USA
| | - Lai-Xi Wang
- Department of Chemistry and Biochemistry, University of Maryland, College Park, Maryland, USA
| | - Geert-Jan Boons
- Complex Carbohydrate Research Center, University of Georgia, Athens, Georgia, USA; Department of Chemical Biology and Drug Discovery, Utrecht Institute for Pharmaceutical Sciences, and Utrecht Institute for Pharmaceutical Sciences, Utrecht, The Netherlands
| | - Zachary A Wood
- Department of Biochemistry and Molecular Biology, University of Georgia, Athens, Georgia, USA.
| | - Kelley W Moremen
- Complex Carbohydrate Research Center, University of Georgia, Athens, Georgia, USA; Department of Biochemistry and Molecular Biology, University of Georgia, Athens, Georgia, USA.
| |
Collapse
|
37
|
L-Fucose treatment of FUT8-CDG. Mol Genet Metab Rep 2020; 25:100680. [PMID: 33312876 PMCID: PMC7719959 DOI: 10.1016/j.ymgmr.2020.100680] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2020] [Revised: 11/12/2020] [Accepted: 11/12/2020] [Indexed: 11/22/2022] Open
Abstract
FUT8-CDG is a severe multisystem disorder caused by mutations in FUT8, encoding the α-1,6-fucosyltransferase. We report on dizygotic twins with FUT8-CDG presenting with dysmorphisms, failure to thrive, and respiratory abnormalities. Due to the severe phenotype, oral L-fucose supplementation was started. Glycosylation analysis using mass spectrometry indicated a limited response to fucose therapy while the clinical presentation stabilized. Further research is needed to assess the concept of substrate supplementation in FUT8-CDG.
Collapse
|
38
|
Taniguchi N, Ohkawa Y, Maeda K, Harada Y, Nagae M, Kizuka Y, Ihara H, Ikeda Y. True significance of N-acetylglucosaminyltransferases GnT-III, V and α1,6 fucosyltransferase in epithelial-mesenchymal transition and cancer. Mol Aspects Med 2020; 79:100905. [PMID: 33010941 DOI: 10.1016/j.mam.2020.100905] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2020] [Accepted: 09/02/2020] [Indexed: 12/13/2022]
Abstract
It is well known that numerous cancer-related changes occur in glycans that are attached to glycoproteins, glycolipids and proteoglycans on the cell surface and these changes in structure and the expression of the glycans are largely regulated by glycosyl-transferases, glycosidases, nucleotide sugars and their related genes. Such structural changes in glycans on cell surface proteins may accelerate the progression, invasion and metastasis of cancer cells. Among the over 200 known glycosyltransferases and related genes, β 1,6 N-acetylglucosaminyltransferase V (GnT-V) (the MGAT5 gene) and α 1,6 fucosyltransferase (FUT8) (the FUT8 gene) are representative enzymes in this respect because changes in glycans caused by these genes appear to be related to cancer metastasis and invasion in vitro as well as in vivo, and a number of reports on these genes in related to epithelial-mesenchymal transition (EMT) have also appeared. Another enzyme, one of the N-glycan branching enzymes, β1,4 N-acetylglucosaminyltransferase III (GnT-III) (the MGAT3 gene) has been reported to suppress EMT. However, there are intermediate states between EMT and mesenchymal-epithelial transition (MET) and some of these genes have been implicated in both EMT and MET and are also probably in an intermediate state. Therefore, it would be difficult to clearly define which specific glycosyltransferase is involved in EMT or MET or an intermediate state. The significance of EMT and N-glycan branching glycosyltransferases needs to be reconsidered and the inhibition of their corresponding genes would also be desirable in therapeutics. This review mainly focuses on GnT-III, GnT-V and FUT8, major players as N-glycan branching enzymes in cancer in relation to EMT programs, and also discusses the catalytic mechanisms of GnT-V and FUT8 whose crystal structures have now been obtained.
Collapse
Affiliation(s)
- Naoyuki Taniguchi
- Department of Glyco-Oncology and Medical Biochemistry, Osaka International Cancer Institute, Osaka, Japan.
| | - Yuki Ohkawa
- Department of Glyco-Oncology and Medical Biochemistry, Osaka International Cancer Institute, Osaka, Japan.
| | - Kento Maeda
- Department of Glyco-Oncology and Medical Biochemistry, Osaka International Cancer Institute, Osaka, Japan.
| | - Yoichiro Harada
- Department of Glyco-Oncology and Medical Biochemistry, Osaka International Cancer Institute, Osaka, Japan.
| | - Masamichi Nagae
- Department of Molecular Immunology, RIMD, Osaka University, Osaka, Japan.
| | - Yasuhiko Kizuka
- Glyco-biochemistry Laboratory, G-Chain, Gifu University, Gifu, Japan.
| | - Hideyuki Ihara
- Division of Molecular Cell Biology, Department of Biomolecular Sciences, Saga University Faculty of Medicine, Saga, Japan.
| | - Yoshitaka Ikeda
- Division of Molecular Cell Biology, Department of Biomolecular Sciences, Saga University Faculty of Medicine, Saga, Japan.
| |
Collapse
|
39
|
Abstract
In this review, we focus on the metabolism of mammalian glycan-associated monosaccharides, where the vast majority of our current knowledge comes from research done during the 1960s and 1970s. Most monosaccharides enter the cell using distinct, often tissue specific transporters from the SLC2A family. If not catabolized, these monosaccharides can be activated to donor nucleotide sugars and used for glycan synthesis. Apart from exogenous and dietary sources, all monosaccharides and their associated nucleotide sugars can be synthesized de novo, using mostly glucose to produce all nine nucleotide sugars present in human cells. Today, monosaccharides are used as treatment options for a small number of rare genetic disorders and even some common conditions. Here, we cover therapeutic applications of these sugars and highlight biochemical gaps that must be revisited as we go forward.
Collapse
Affiliation(s)
- Paulina Sosicka
- Human Genetics Program, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, California 92037, United States
| | - Bobby G. Ng
- Human Genetics Program, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, California 92037, United States
| | - Hudson H. Freeze
- Human Genetics Program, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, California 92037, United States
| |
Collapse
|
40
|
Ng BG, Dastsooz H, Silawi M, Habibzadeh P, Jahan SB, Fard MAF, Halliday BJ, Raymond K, Ruzhnikov MRZ, Tabatabaei Z, Taghipour-Sheshdeh A, Brimble E, Robertson SP, Faghihi MA, Freeze HH. Expanding the molecular and clinical phenotypes of FUT8-CDG. J Inherit Metab Dis 2020; 43:871-879. [PMID: 32049367 PMCID: PMC7359201 DOI: 10.1002/jimd.12221] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/27/2019] [Revised: 02/06/2020] [Accepted: 02/10/2020] [Indexed: 12/11/2022]
Abstract
Pathogenic variants in the Golgi localised alpha 1,6 fucosyltransferase, FUT8, cause a rare inherited metabolic disorder known as FUT8-CDG. To date, only three affected individuals have been reported presenting with a constellation of symptoms including intrauterine growth restriction, severe delays in growth and development, other neurological impairments, significantly shortened limbs, respiratory complications, and shortened lifespan. Here, we report an additional four unrelated affected individuals homozygous for novel pathogenic variants in FUT8. Analysis of serum N-glycans revealed a complete lack of core fucosylation, an important diagnostic biomarker of FUT8-CDG. Our data expands both the molecular and clinical phenotypes of FUT8-CDG and highlights the importance of identifying a reliable biomarker for confirming potentially pathogenic variants.
Collapse
Affiliation(s)
- Bobby G. Ng
- Human Genetics Program, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, California
| | - Hassan Dastsooz
- Department of Life Sciences and Systems Biology, Italian Institute for Genomic Medicine (IIGM), University of Turin, Turin, Italy
- Persian BayanGene Research and Training Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Mohammad Silawi
- Persian BayanGene Research and Training Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Parham Habibzadeh
- Persian BayanGene Research and Training Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Shima B. Jahan
- Persian BayanGene Research and Training Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Mohammad A. F. Fard
- Persian BayanGene Research and Training Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Benjamin J. Halliday
- Department of Women’s and Children’s Health, Dunedin School of Medicine, University of Otago, Dunedin, New Zealand
| | - Kimiyo Raymond
- Biochemical Genetics Laboratory, Mayo Clinic College of Medicine, Rochester, Minnesota
| | - Maura R. Z. Ruzhnikov
- Department of Neurology and Neurological Sciences, Stanford Medicine, Stanford, California
- Division of Medical Genetics, Department of Pediatrics, Stanford Medicine, Stanford, California
| | - Zahra Tabatabaei
- Persian BayanGene Research and Training Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | | | - Elise Brimble
- Department of Neurology and Neurological Sciences, Stanford Medicine, Stanford, California
| | - Stephen P. Robertson
- Department of Women’s and Children’s Health, Dunedin School of Medicine, University of Otago, Dunedin, New Zealand
| | - Mohammad A. Faghihi
- Persian BayanGene Research and Training Center, Shiraz University of Medical Sciences, Shiraz, Iran
- Center for Therapeutic Innovation, Department of Psychiatry and Behavioral Sciences, University of Miami Miller School of Medicine, Miami, Florida
| | - Hudson H. Freeze
- Human Genetics Program, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, California
| |
Collapse
|
41
|
A lectin-based glycomic approach identifies FUT8 as a driver of radioresistance in oesophageal squamous cell carcinoma. Cell Oncol (Dordr) 2020; 43:695-707. [PMID: 32474852 DOI: 10.1007/s13402-020-00517-5] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2019] [Revised: 03/30/2020] [Accepted: 04/03/2020] [Indexed: 02/06/2023] Open
Abstract
PURPOSE Radio-resistance is recognized as a main factor in the failure of radiotherapy in oesophageal squamous cell carcinoma (ESCC). Aberrant cell surface glycosylation has been reported to correlate with radio-resistance in different kinds of tumours. However, glycomic alterations and the corresponding enzymes associated with ESCC radio-resistance have not yet been defined. METHODS Two radioresistant cell lines, EC109R and TE-1R, were established from parental ESCC cell lines EC109 and TE-1 by fractionated irradiation. A lectin microarray was used to screen for altered glycan patterns. RNA-sequencing (RNA-seq) was employed to identify differentially expressed glycosyltransferases. Cell Counting Kit-8, colony formation and flow cytometry assays were used to measure cell viability and radiosensitivity. Expression of glycosyltransferase in ESCC tissues was assessed by immunohistochemistry. In vivo radiosensitivity was analysed using a nude mouse xenograft model. Downstream effectors of the enzyme were verified using a lectin-based pull-down assay combined with mass spectrometry. RESULTS We found that EC109R and TE-1R cells were more resistant to irradiation than the parental EC109 and TE-1 cells. Using lectin microarrays combined with RNA sequencing, we found that α1, 6-fucosyltransferase (FUT8) was overexpressed in the radioresistant ESCC cell lines. Both gain- and loss-of-function studies confirmed that FUT8 regulates the sensitivity of ESCC cells to irradiation. Importantly, we found that high FUT8 expression was positively linked to radio-resistance and a poor prognosis in ESCC patients who received radiation therapy. Moreover, FUT8 inhibition suppressed the growth and formation of xenograft tumours in nude mice after irradiation. Using a lectin-based pull-down assay and mass spectrometry, we found that CD147 could be glycosylated by FUT8. As expected, inhibition of CD147 partly reversed FUT8-induced radio-resistance in ESCC cells. CONCLUSIONS Our results indicate that FUT8 functions as a driver of radio-resistance in ESCC by targeting CD147. Therefore, FUT8 may serve as a marker for predicting the response to radiation therapy in patients with ESCC.
Collapse
|
42
|
Järvå MA, Dramicanin M, Lingford JP, Mao R, John A, Jarman KE, Grinter R, Goddard-Borger ED. Structural basis of substrate recognition and catalysis by fucosyltransferase 8. J Biol Chem 2020; 295:6677-6688. [PMID: 32220931 DOI: 10.1074/jbc.ra120.013291] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2020] [Revised: 03/26/2020] [Indexed: 12/11/2022] Open
Abstract
Fucosylation of the innermost GlcNAc of N-glycans by fucosyltransferase 8 (FUT8) is an important step in the maturation of complex and hybrid N-glycans. This simple modification can dramatically affect the activities and half-lives of glycoproteins, effects that are relevant to understanding the invasiveness of some cancers, development of mAb therapeutics, and the etiology of a congenital glycosylation disorder. The acceptor substrate preferences of FUT8 are well-characterized and provide a framework for understanding N-glycan maturation in the Golgi; however, the structural basis of these substrate preferences and the mechanism through which catalysis is achieved remain unknown. Here we describe several structures of mouse and human FUT8 in the apo state and in complex with GDP, a mimic of the donor substrate, and with a glycopeptide acceptor substrate at 1.80-2.50 Å resolution. These structures provide insights into a unique conformational change associated with donor substrate binding, common strategies employed by fucosyltransferases to coordinate GDP, features that define acceptor substrate preferences, and a likely mechanism for enzyme catalysis. Together with molecular dynamics simulations, the structures also revealed how FUT8 dimerization plays an important role in defining the acceptor substrate-binding site. Collectively, this information significantly builds on our understanding of the core fucosylation process.
Collapse
Affiliation(s)
- Michael A Järvå
- The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria 3052, Australia.,Department of Medical Biology, University of Melbourne, Parkville, Victoria 3010, Australia
| | - Marija Dramicanin
- The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria 3052, Australia.,Department of Medical Biology, University of Melbourne, Parkville, Victoria 3010, Australia
| | - James P Lingford
- The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria 3052, Australia.,Department of Medical Biology, University of Melbourne, Parkville, Victoria 3010, Australia
| | - Runyu Mao
- The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria 3052, Australia.,Department of Medical Biology, University of Melbourne, Parkville, Victoria 3010, Australia
| | - Alan John
- The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria 3052, Australia.,Department of Medical Biology, University of Melbourne, Parkville, Victoria 3010, Australia
| | - Kate E Jarman
- The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria 3052, Australia.,Department of Medical Biology, University of Melbourne, Parkville, Victoria 3010, Australia
| | - Rhys Grinter
- Department of Microbiology, Monash Biomedicine Discovery Institute, Monash University, Clayton, Victoria 3800, Australia
| | - Ethan D Goddard-Borger
- The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria 3052, Australia .,Department of Medical Biology, University of Melbourne, Parkville, Victoria 3010, Australia
| |
Collapse
|
43
|
Involvement of the α-helical and Src homology 3 domains in the molecular assembly and enzymatic activity of human α1,6-fucosyltransferase, FUT8. Biochim Biophys Acta Gen Subj 2020; 1864:129596. [PMID: 32147455 DOI: 10.1016/j.bbagen.2020.129596] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2019] [Revised: 02/20/2020] [Accepted: 03/03/2020] [Indexed: 01/05/2023]
Abstract
BACKGROUND Previous structural analyses showed that human α1,6-fucosyltransferase, FUT8 contains a catalytic domain along with two additional domains, N-terminal α-helical domain and C-terminal Src homology 3 domain, but these domains are unique to FUT8 among glycosyltransferases. The role that these domains play in formation of the active form of FUT8 has not been investigated. This study reports on attempts to determine the involvement of these domains in the functions of FUT8. METHODS Based on molecular modeling, the domain mutants were constructed by truncation and site-directed mutagenesis, and were heterologously expressed in Sf21 or COS-1 cells. The mutants were analyzed by SDS-PAGE and assayed for enzymatic activity. In vivo cross-linking experiments by introducing disulfide bonds were also carried out to examine the orientation of the domains in the molecular assembly. RESULTS Mutagenesis and molecular modeling findings suggest that human FUT8 potentially forms homodimer in vivo via intermolecular hydrophobic interactions involving α-helical domains. Truncation or site-directed mutagenesis findings indicated that α-helical and SH3 domains are all required for enzymatic activity. In addition, in vivo cross-linking experiments clearly indicated that the SH3 domain located in close proximity to the α-helical domain in an intermolecular manner. CONCLUSIONS α-Helical and SH3 domains are required for a fully active enzyme, and are also involved in homophilic dimerization, which probably results in the formation of the active form of human FUT8. GENERAL SIGNIFICANCE α-Helical and SH3 domains, which are not commonly found in glycosyltransferases, play roles in the formation of the functional quaternary structure of human FUT8.
Collapse
|
44
|
Duan C, Fukuda T, Isaji T, Qi F, Yang J, Wang Y, Takahashi S, Gu J. Deficiency of core fucosylation activates cellular signaling dependent on FLT3 expression in a Ba/F3 cell system. FASEB J 2020; 34:3239-3252. [DOI: 10.1096/fj.201902313rr] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2019] [Revised: 12/16/2019] [Accepted: 12/23/2019] [Indexed: 12/29/2022]
Affiliation(s)
- Chengwei Duan
- Division of Regulatory Glycobiology Institute of Molecular Biomembrane and Glycobiology Tohoku Medical and Pharmaceutical University Sendai Japan
| | - Tomohiko Fukuda
- Division of Regulatory Glycobiology Institute of Molecular Biomembrane and Glycobiology Tohoku Medical and Pharmaceutical University Sendai Japan
| | - Tomoya Isaji
- Division of Regulatory Glycobiology Institute of Molecular Biomembrane and Glycobiology Tohoku Medical and Pharmaceutical University Sendai Japan
| | - Feng Qi
- Division of Regulatory Glycobiology Institute of Molecular Biomembrane and Glycobiology Tohoku Medical and Pharmaceutical University Sendai Japan
| | - Jie Yang
- Division of Regulatory Glycobiology Institute of Molecular Biomembrane and Glycobiology Tohoku Medical and Pharmaceutical University Sendai Japan
| | - Yuqin Wang
- Department of Pharmacology Pharmacy College Nantong University Nantong China
| | - Shinichiro Takahashi
- Division of Laboratory Medicine Faculty of Medicine Tohoku Medical and Pharmaceutical University Sendai Japan
| | - Jianguo Gu
- Division of Regulatory Glycobiology Institute of Molecular Biomembrane and Glycobiology Tohoku Medical and Pharmaceutical University Sendai Japan
| |
Collapse
|
45
|
Mishra N, Spearman M, Donald L, Perreault H, Butler M. Comparison of two glycoengineering strategies to control the fucosylation of a monoclonal antibody. J Biotechnol 2020; 324S:100015. [DOI: 10.1016/j.btecx.2020.100015] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2019] [Revised: 01/16/2020] [Accepted: 02/14/2020] [Indexed: 12/21/2022]
|
46
|
Pascoal C, Francisco R, Ferro T, Dos Reis Ferreira V, Jaeken J, Videira PA. CDG and immune response: From bedside to bench and back. J Inherit Metab Dis 2020; 43:90-124. [PMID: 31095764 DOI: 10.1002/jimd.12126] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/18/2019] [Revised: 05/13/2019] [Accepted: 05/15/2019] [Indexed: 12/20/2022]
Abstract
Glycosylation is an essential biological process that adds structural and functional diversity to cells and molecules, participating in physiological processes such as immunity. The immune response is driven and modulated by protein-attached glycans that mediate cell-cell interactions, pathogen recognition and cell activation. Therefore, abnormal glycosylation can be associated with deranged immune responses. Within human diseases presenting immunological defects are congenital disorders of glycosylation (CDG), a family of around 130 rare and complex genetic diseases. In this review, we have identified 23 CDG with immunological involvement, characterized by an increased propensity to-often life-threatening-infection. Inflammatory and autoimmune complications were found in 7 CDG types. CDG natural history(ies) and the mechanisms behind the immunological anomalies are still poorly understood. However, in some cases, alterations in pathogen recognition and intracellular signaling (eg, TGF-β1, NFAT, and NF-κB) have been suggested. Targeted therapies to restore immune defects are only available for PGM3-CDG and SLC35C1-CDG. Fostering research on glycoimmunology may elucidate the involved pathophysiological mechanisms and open new therapeutic avenues, thus improving CDG patients' quality of life.
Collapse
Affiliation(s)
- Carlota Pascoal
- Portuguese Association for CDG, Lisbon, Portugal
- CDG & Allies - Professionals and Patient Associations International Network (CDG & Allies - PPAIN), Caparica, Portugal
- UCIBIO, Departamento Ciências da Vida, Faculdade de Ciências e Tecnologia, Universidade NOVA de Lisboa, Caparica, Portugal
| | - Rita Francisco
- Portuguese Association for CDG, Lisbon, Portugal
- CDG & Allies - Professionals and Patient Associations International Network (CDG & Allies - PPAIN), Caparica, Portugal
- UCIBIO, Departamento Ciências da Vida, Faculdade de Ciências e Tecnologia, Universidade NOVA de Lisboa, Caparica, Portugal
| | - Tiago Ferro
- CDG & Allies - Professionals and Patient Associations International Network (CDG & Allies - PPAIN), Caparica, Portugal
- UCIBIO, Departamento Ciências da Vida, Faculdade de Ciências e Tecnologia, Universidade NOVA de Lisboa, Caparica, Portugal
| | - Vanessa Dos Reis Ferreira
- Portuguese Association for CDG, Lisbon, Portugal
- CDG & Allies - Professionals and Patient Associations International Network (CDG & Allies - PPAIN), Caparica, Portugal
| | - Jaak Jaeken
- CDG & Allies - Professionals and Patient Associations International Network (CDG & Allies - PPAIN), Caparica, Portugal
- Center for Metabolic Diseases, Department of Development and Regeneration, UZ and KU Leuven, Leuven, Belgium
| | - Paula A Videira
- Portuguese Association for CDG, Lisbon, Portugal
- CDG & Allies - Professionals and Patient Associations International Network (CDG & Allies - PPAIN), Caparica, Portugal
- UCIBIO, Departamento Ciências da Vida, Faculdade de Ciências e Tecnologia, Universidade NOVA de Lisboa, Caparica, Portugal
| |
Collapse
|
47
|
Schweigert A, Areaux RG. Childhood glaucoma in association with congenital disorder of glycosylation caused by mutations in fucosyltransferase 8. J AAPOS 2019; 23:351-352. [PMID: 31580894 DOI: 10.1016/j.jaapos.2019.08.272] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/12/2019] [Revised: 08/09/2019] [Accepted: 08/14/2019] [Indexed: 10/25/2022]
Abstract
A rare form of congenital disorder of glycosylation (CDG) was recently discovered in individuals with biallelic mutations in fucosyltransferase 8 (FUT8). The clinical characteristics of patients with FUT8-CDG include intrauterine growth retardation, feeding difficulties, hypotonia, microcephaly, seizures, short stature, developmental delay, and respiratory abnormalities. We report the first case of glaucoma in an infant with FUT8-CGD and hypothesize a pathogenesis for glaucoma.
Collapse
Affiliation(s)
- Anna Schweigert
- Department of Ophthalmology and Visual Neurosciences, University of Minnesota, Minneapolis.
| | - Raymond G Areaux
- Department of Ophthalmology and Visual Neurosciences, University of Minnesota, Minneapolis
| |
Collapse
|
48
|
Lu X, Zhang D, Shoji H, Duan C, Zhang G, Isaji T, Wang Y, Fukuda T, Gu J. Deficiency of α1,6-fucosyltransferase promotes neuroinflammation by increasing the sensitivity of glial cells to inflammatory mediators. Biochim Biophys Acta Gen Subj 2019; 1863:598-608. [DOI: 10.1016/j.bbagen.2018.12.008] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2018] [Revised: 12/05/2018] [Accepted: 12/13/2018] [Indexed: 10/27/2022]
|
49
|
Ng BG, Rosenfeld JA, Emrick L, Jain M, Burrage LC, Lee B, Craigen WJ, Bearden DR, Graham BH, Freeze HH, Freeze HH. Pathogenic Variants in Fucokinase Cause a Congenital Disorder of Glycosylation. Am J Hum Genet 2018; 103:1030-1037. [PMID: 30503518 DOI: 10.1016/j.ajhg.2018.10.021] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2018] [Accepted: 10/25/2018] [Indexed: 01/20/2023] Open
Abstract
FUK encodes fucokinase, the only enzyme capable of converting L-fucose to fucose-1-phosphate, which will ultimately be used for synthesizing GDP-fucose, the donor substrate for all fucosyltransferases. Although it is essential for fucose salvage, this pathway is thought to make only a minor contribution to the total amount of GDP-fucose. A second pathway, the major de novo pathway, involves conversion of GDP-mannose to GDP-fucose. Here we describe two unrelated individuals who have pathogenic variants in FUK and who presented with severe developmental delays, encephalopathy, intractable seizures, and hypotonia. The first individual was compound heterozygous for c.667T>C (p.Ser223Pro) and c.2047C>T (p.Arg683Cys), and the second individual was homozygous for c.2980A>C (p.Lys994Gln). Skin fibroblasts from the first individual confirmed the variants as loss of function and showed significant decreases in total GDP-[3H] fucose and [3H] fucose-1-phosphate. There was also a decrease in the incorporation of [5,6-3H]-fucose into fucosylated glycoproteins. Lys994 has previously been shown to be an important site for ubiquitin conjugation. Here, we show that loss-of-function variants in FUK cause a congenital glycosylation disorder characterized by a defective fucose-salvage pathway.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | - Hudson H Freeze
- Human Genetics Program, Sanford-Burnham-Prebys Medical Discovery Institute, La Jolla, CA 92037, USA.
| |
Collapse
|