1
|
Cao J, Hu J, Zhang B, Zhang Y, Wen Z, Wu Y, Hu Z, Zhou Z, Liu X, Hou S. Polymorphisms of FUT9 and its relationship with susceptibility towards DHAV-3 in Pekin duck. Gene 2025; 955:149417. [PMID: 40090531 DOI: 10.1016/j.gene.2025.149417] [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: 11/18/2024] [Revised: 02/03/2025] [Accepted: 03/11/2025] [Indexed: 03/18/2025]
Abstract
Duck viral hepatitis severely threatens the development of the duck industry, leading to economic losses every year. Using selected Pekin duck populations exhibiting varying resistance towards Duck Hepatitis A Virus type 3 (DHAV-3), screening for genetic variations, such as single nucleotide polymorphisms (SNP), associated with disease susceptibility will facilitate the breeding of Pekin ducks with enhanced disease resistance. The biological role of fucosyltransferases, which are a type of glycosyltransferase enzymes, is to catalyze the transfer of fucose to molecules such as oligosaccharides, glycoproteins and glycolipids, which is crucial for maintaining immune function by promoting effective pathogen recognition and modulating immune responses through specific fucosylation patterns. Previous studies found that the expression level of the Fucosyltransferase 9 (FUT9) gene in the liver of resistant Pekin ducks was significantly higher than that in susceptible ducks, suggesting its potential association with disease resistance. However, the association between genetic variations in FUT9 and susceptibility to DHAV-3 in ducks remains unclear. This study aims to detect SNPs in the FUT9 gene and explore their relationships with disease mortality and susceptibility, the result will provide a scientific basis for developing effective control strategies in duck breeding. 242 Pekin ducks with varying resistance to DHAV-3 were used in this experiment. 12 SNPs were identified in the coding region of FUT9. And g.76953686 T > C and g.76954451C > T were significantly associated with susceptibility to DHAV-3 in Pekin ducks. The results indicate that variations in the FUT9 gene significantly influence the susceptibility of ducks towards DHAV-3, providing potential genetic markers for enhancing disease resistance breeding in Pekin ducks.
Collapse
Affiliation(s)
- Junting Cao
- Key Laboratory of Animal (Poultry) Genetics Breeding and Reproduction, Ministry of Agriculture and Rural Affairs, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing 100193, China; Institute of Feed Research, Chinese Academy of Agricultural Sciences, Beijing 100081, China; Shaanxi Key Laboratory of Molecular Biology for Agriculture, College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi 712100, China
| | - Jian Hu
- Key Laboratory of Animal (Poultry) Genetics Breeding and Reproduction, Ministry of Agriculture and Rural Affairs, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing 100193, China
| | - Bo Zhang
- Key Laboratory of Animal (Poultry) Genetics Breeding and Reproduction, Ministry of Agriculture and Rural Affairs, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing 100193, China; Institute of Animal Husbandry and Veterinary Medicine, Beijing Academy of Agriculture and Forestry Sciences, Beijing 100097, China
| | - Yunsheng Zhang
- Key Laboratory of Animal (Poultry) Genetics Breeding and Reproduction, Ministry of Agriculture and Rural Affairs, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing 100193, China
| | - Zhiguo Wen
- Institute of Feed Research, Chinese Academy of Agricultural Sciences, Beijing 100081, China
| | - Yongbao Wu
- Key Laboratory of Animal (Poultry) Genetics Breeding and Reproduction, Ministry of Agriculture and Rural Affairs, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing 100193, China; Institute of Feed Research, Chinese Academy of Agricultural Sciences, Beijing 100081, China
| | - Zhigang Hu
- Shaanxi Key Laboratory of Molecular Biology for Agriculture, College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi 712100, China
| | - Zhengkui Zhou
- Key Laboratory of Animal (Poultry) Genetics Breeding and Reproduction, Ministry of Agriculture and Rural Affairs, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing 100193, China
| | - Xiaolin Liu
- Shaanxi Key Laboratory of Molecular Biology for Agriculture, College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi 712100, China.
| | - Shuisheng Hou
- Key Laboratory of Animal (Poultry) Genetics Breeding and Reproduction, Ministry of Agriculture and Rural Affairs, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing 100193, China.
| |
Collapse
|
2
|
Li Z, Zhu Z, Wang P, Hou C, Ren L, Xu D, Wang X, Guo F, Meng Q, Liang W, Xue J, Zhi X. Diagnostic, prognostic, and immunological roles of FUT8 in lung adenocarcinoma and lung squamous cell carcinoma. PLoS One 2025; 20:e0321756. [PMID: 40373023 PMCID: PMC12080848 DOI: 10.1371/journal.pone.0321756] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2024] [Accepted: 03/11/2025] [Indexed: 05/17/2025] Open
Abstract
Lung cancer remains the leading cause of malignant tumors worldwide in terms of the incidence and mortality, posing a significant threat to human health. Given that distant metastases typically occur at the time of initial diagnosis, leading to a poor 5-year survival rate among patients, it is crucial to identify markers for diagnosis, prognosis, and therapeutic efficacy monitoring. Abnormal glycosylation is a hallmark of cancer cells, characterized by the disruption of core fucosylation, which is predominantly driven by the enzyme fucosyltransferase 8 (FUT8). Evidence indicates that FUT8 is a pivotal enzyme in cancer onset and progression, influencing cellular glycosylation pathways. Utilizing bioinformatics approaches, we have investigated FUT8 in lung cancer, resulting in a more systematic and comprehensive understanding of its role in the disease's pathogenesis. In this study, we employed bioinformatics to analyze the differential expression of FUT8 between LUAD and LUSC. We observed upregulation of FUT8 in both LUAD and LUSC, associated with unfavorable prognosis, and higher diagnostic utility in LUAD. GO/KEGG analysis revealed a primary association between LUAD and the spliceosome. Immunologically, FUT8 expression was significantly associated with immune cell infiltration and immune checkpoint activity, with a notable positive correlation with M2 macrophage infiltration. Our analysis of FUT8 indicates that it may serve as a potential biomarker for lung cancer diagnosis and prognosis, and could represent a therapeutic target for LUAD and LUSC immunotherapy.
Collapse
Affiliation(s)
- Zhijun Li
- Hebei North University, Zhangjiakou City, Hebei Province, China
| | - Zhenpeng Zhu
- Hebei North University, Zhangjiakou City, Hebei Province, China
| | - Peng Wang
- Hebei North University, Zhangjiakou City, Hebei Province, China
| | - Chenyang Hou
- Hebei North University, Zhangjiakou City, Hebei Province, China
| | - Lijuan Ren
- Hebei North University, Zhangjiakou City, Hebei Province, China
| | - Dandan Xu
- Hebei Key Laboratory of Systems Biology and Gene Regulation, Central Laboratory, The First Affiliated Hospital of Hebei North University, Zhangjiakou City, Hebei Province, China
| | - Xiran Wang
- Department of Bioinformatics, School of Health Care, Changchun Vocational College of Health, Changchun City, Jilin Province, China
| | - Fei Guo
- Department of Surgery, Hebei Key Laboratory of Systems Biology and Gene Regulation, The First Affiliated Hospital of Hebei North University, Zhangjiakou City, Hebei Province, China
| | - Qingxue Meng
- Technology Department, The First Affiliated Hospital of Hebei North University, Zhangjiakou City, Hebei Province, China
| | - Weizheng Liang
- Hebei Key Laboratory of Systems Biology and Gene Regulation, Central Laboratory, The First Affiliated Hospital of Hebei North University, Zhangjiakou City, Hebei Province, China
| | - Jun Xue
- Department of Surgery, Hebei Key Laboratory of Systems Biology and Gene Regulation, The First Affiliated Hospital of Hebei North University, Zhangjiakou City, Hebei Province, China
| | - Xuejun Zhi
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Hebei North University, Zhangjiakou City, Hebei Province, China
| |
Collapse
|
3
|
Klepper J. Glut1 Deficiency Syndrome: Novel Pathomechanisms, Current Concepts, and Challenges. J Inherit Metab Dis 2025; 48:e70044. [PMID: 40405536 PMCID: PMC12099281 DOI: 10.1002/jimd.70044] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/07/2025] [Revised: 05/02/2025] [Accepted: 05/05/2025] [Indexed: 05/24/2025]
Abstract
Glut1 Deficiency Syndrome (Glut1DS) has emerged as a treatable, but complex entity. Increasing data on pathogenic mechanisms, phenotype, genotype, and ketogenic dietary therapies (KDT) are available, as summarized in this review. Many challenges remain: novel symptoms emerge and vary with age. In Glut1DS, KDT in pregnancy and the clinical features in neonates and adults are poorly understood. KDT are ineffective in some patients for reasons yet unknown. Research reaches beyond the concept of brain energy depletion by impaired GLUT1-mediated glucose transfer across the blood-brain barrier. Novel concepts investigate alternative substrates, transport mechanisms, and metabolic interactions of different brain cell types. Future, yet currently unavailable prospects are neonatal screening for Glut1DS, reliable biomarkers, predictors for outcome, and alternative therapies, along with and beyond KDT.
Collapse
Affiliation(s)
- Joerg Klepper
- Department of Pediatrics and NeuropediatricsChildrens' Hospital AschaffenburgAschaffenburgGermany
| |
Collapse
|
4
|
Zhu B, Pitts MG, Buoncristiani MD, Bryant LT, Lopez-Nunez O, Gurria JP, Shedlock C, Ribas R, Keohane S, Liu J, Wang C, Gentry MS, Shelman NR, Allison DB, Evers BM, Sun RC, Rellinger EJ. GDP-mannose 4,6-dehydratase is a key driver of MYCN-amplified neuroblastoma core fucosylation and tumorigenesis. Oncogene 2025; 44:1272-1283. [PMID: 39956863 PMCID: PMC12048354 DOI: 10.1038/s41388-025-03297-0] [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: 07/10/2024] [Revised: 01/08/2025] [Accepted: 02/03/2025] [Indexed: 02/18/2025]
Abstract
MYCN-amplification is a genetic hallmark of ~40% of high-risk neuroblastomas (NBs). Altered glycosylation is a common feature of adult cancer progression, but little is known about how genetic signatures such as MYCN-amplification alter glycosylation profiles. Herein, matrix-assisted laser desorption/ionization mass spectrometry imaging (MALDI-MSI) revealed increased core fucosylated glycan abundance within neuroblast-rich regions of human MYCN-amplified NB tumors. GDP-mannose 4,6-dehydratase (GMDS) is responsible for the first-committed and rate-limiting step of de novo GDP-fucose synthesis. High GMDS expression was found to be associated with poor patient survival, advanced stage disease, and MYCN-amplification in human NB tumors. Chromatin immunoprecipitation and promoter reporter assays demonstrated that N-MYC directly binds and activates the GMDS promoter in NB cells. When GMDS was blocked through either genetic or pharmacological mechanisms, NBs were found to be dependent upon de novo GDP-fucose production to sustain cell surface and secreted core fucosylated glycan abundance, as well as adherence and motility. Moreover, genetic knockdown of GMDS inhibited tumor formation and progression in vivo. These critical findings identify de novo GDP-fucose production as a novel metabolic vulnerability that may be exploited in designing new treatment strategies for MYCN-amplified NBs.
Collapse
Affiliation(s)
- Beibei Zhu
- University of Kentucky, Department of Surgery, Lexington, KY, 40536, USA
| | - Michelle G Pitts
- University of Kentucky, Department of Surgery, Lexington, KY, 40536, USA
| | | | - Lindsay T Bryant
- University of Kentucky, Department of Surgery, Lexington, KY, 40536, USA
| | - Oscar Lopez-Nunez
- Cincinnati Children's Hospital Medical Center, Department of Pathology, Cincinnati, OH, 45229, USA
| | - Juan P Gurria
- Cincinnati Children's Hospital Medical Center, Department of Pediatric Surgery, Cincinnati, OH, 45229, USA
| | - Cameron Shedlock
- University of Florida, Department of Biochemistry & Molecular Biology, Gainesville, FL, 32610, USA
| | - Roberto Ribas
- University of Florida, Department of Biochemistry & Molecular Biology, Gainesville, FL, 32610, USA
| | - Shannon Keohane
- University of Florida, Department of Biochemistry & Molecular Biology, Gainesville, FL, 32610, USA
| | - Jinpeng Liu
- University of Kentucky, Department of Bioinformatics, Lexington, KY, 40536, USA
| | - Chi Wang
- University of Kentucky, Department of Bioinformatics, Lexington, KY, 40536, USA
| | - Matthew S Gentry
- University of Florida, Department of Biochemistry & Molecular Biology, Gainesville, FL, 32610, USA
| | - Nathan R Shelman
- University of Kentucky, Department of Pathology, Lexington, KY, 40536, USA
| | - Derek B Allison
- University of Kentucky, Department of Pathology, Lexington, KY, 40536, USA
- University of Kentucky, Markey Cancer Center, Lexington, KY, 40536, USA
| | - B Mark Evers
- University of Kentucky, Department of Surgery, Lexington, KY, 40536, USA
- University of Kentucky, Markey Cancer Center, Lexington, KY, 40536, USA
| | - Ramon C Sun
- University of Florida, Department of Biochemistry & Molecular Biology, Gainesville, FL, 32610, USA
| | - Eric J Rellinger
- University of Kentucky, Department of Surgery, Lexington, KY, 40536, USA.
- University of Kentucky, Markey Cancer Center, Lexington, KY, 40536, USA.
| |
Collapse
|
5
|
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
|
6
|
Bastian K, Orozco‐Moreno M, Thomas H, Hodgson K, Visser EA, Rossing E, Pijnenborg JFA, Eerden N, Wilson L, Saravannan H, Hanley O, Grimsley G, Frame F, Peng Z, Knight B, McCullagh P, McGrath J, Crundwell M, Harries L, Maitland NJ, Heer R, Wang N, Goddard‐Borger ED, Guerrero RH, Boltje TJ, Drake RR, Scott E, Elliott DJ, Munkley J. FUT8 Is a Critical Driver of Prostate Tumour Growth and Can Be Targeted Using Fucosylation Inhibitors. Cancer Med 2025; 14:e70959. [PMID: 40387385 PMCID: PMC12086987 DOI: 10.1002/cam4.70959] [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: 02/13/2025] [Revised: 04/20/2025] [Accepted: 04/29/2025] [Indexed: 05/20/2025] Open
Abstract
BACKGROUND An unmet clinical need requires the discovery of new treatments for men facing advanced prostate cancer. Aberrant glycosylation is a universal feature of cancer cells and plays a key role in tumour growth, immune evasion and metastasis. Alterations in tumour glycosylation are closely associated with prostate cancer progression, making glycans promising therapeutic targets. Fucosyltransferase 8 (FUT8) drives core fucosylation by adding α1,6-fucose to the innermost GlcNAc residue on N-glycans. While FUT8 is recognised as a crucial factor in cancer progression, its role in prostate cancer remains poorly understood. METHODS & RESULTS Here, we demonstrate using multiple independent clinical cohorts that FUT8 is upregulated in high grade and metastatic prostate tumours, and in the blood of prostate cancer patients with aggressive disease. Using novel tools, including PhosL lectin immunofluorescence and N-glycan MALDI mass spectrometry imaging (MALDI-MSI), we find FUT8 underpins the biosynthesis of malignant core fucosylated N-glycans in prostate cancer cells and using both in vitro and in vivo models, we find FUT8 promotes prostate tumour growth, cell motility and invasion. Mechanistically we show FUT8 regulates the expression of genes and signalling pathways linked to prostate cancer progression. Furthermore, we find that fucosylation inhibitors can inhibit the activity of FUT8 in prostate cancer to suppress the growth of prostate tumours. CONCLUSIONS Our study cements FUT8-mediated core fucosylation as an important driver of prostate cancer progression and suggests targeting FUT8 activity for prostate cancer therapy as an exciting area to explore.
Collapse
Affiliation(s)
- Kayla Bastian
- Newcastle University Centre for CancerNewcastle University Institute of BiosciencesNewcastleUK
| | - Margarita Orozco‐Moreno
- Newcastle University Centre for CancerNewcastle University Institute of BiosciencesNewcastleUK
| | - Huw Thomas
- Newcastle University Centre for Cancer, Translational and Clinical Research Institute, Paul O'gorman BuildingNewcastle UniversityNewcastle upon TyneUK
| | - Kirsty Hodgson
- Newcastle University Centre for CancerNewcastle University Institute of BiosciencesNewcastleUK
| | - Eline A. Visser
- Synthetic Organic Chemistry, Institute for Molecules and MaterialsRadboud UniversityNijmegenthe Netherlands
| | - Emiel Rossing
- Synthetic Organic Chemistry, Institute for Molecules and MaterialsRadboud UniversityNijmegenthe Netherlands
| | | | | | - Laura Wilson
- Newcastle University Centre for Cancer, Translational and Clinical Research Institute, Paul O'gorman BuildingNewcastle UniversityNewcastle upon TyneUK
| | - Hasvini Saravannan
- Newcastle University Centre for CancerNewcastle University Institute of BiosciencesNewcastleUK
| | - Oliver Hanley
- Newcastle University Centre for CancerNewcastle University Institute of BiosciencesNewcastleUK
| | - Grace Grimsley
- Department of Cell and Molecular PharmacologyMedical University of South CarolinaCharlestonSouth CarolinaUSA
| | - Fiona Frame
- Cancer Research Unit, Department of BiologyUniversity of YorkNorth YorkshireUK
| | - Ziqian Peng
- Newcastle University Centre for CancerNewcastle University Institute of BiosciencesNewcastleUK
| | - Bridget Knight
- NIHR Exeter Clinical Research FacilityRoyal Devon and Exeter NHS Foundation TrustExeterUK
| | - Paul McCullagh
- Department of PathologyRoyal Devon and Exeter NHS Foundation TrustExeterUK
| | - John McGrath
- Exeter Surgical Health Services Research UnitRoyal Devon and Exeter NHS Foundation TrustExeterUK
| | - Malcolm Crundwell
- Institute of Biomedical and Clinical Sciences, Medical School, College of Medicine and HealthUniversity of ExeterExeterUK
| | - Lorna Harries
- Institute of Biomedical and Clinical Sciences, Medical School, College of Medicine and HealthUniversity of ExeterExeterUK
| | - Norman J. Maitland
- Cancer Research Unit, Department of BiologyUniversity of YorkNorth YorkshireUK
| | - Rakesh Heer
- Newcastle University Centre for Cancer, Translational and Clinical Research Institute, Paul O'gorman BuildingNewcastle UniversityNewcastle upon TyneUK
| | - Ning Wang
- The Mellanby Centre for Musculoskeletal Research, Division of Clinical MedicineThe University of SheffieldSheffieldUK
- Leicester Cancer Research Centre, Department of Genetics, Genomics, and Cancer SciencesUniversity of LeicesterLeicesterUK
| | - Ethan D. Goddard‐Borger
- The Walter and Eliza Hall Institute of Medical ResearchParkvilleVictoriaAustralia
- Department of Medical BiologyUniversity of MelbourneParkvilleVictoriaAustralia
| | - Ramon Hurtado Guerrero
- University of ZaragozaZaragozaSpain
- Copenhagen Center for Glycomics, Department of Cellular and Molecular MedicineUniversity of CopenhagenCopenhagenDenmark
| | - Thomas J. Boltje
- Synthetic Organic Chemistry, Institute for Molecules and MaterialsRadboud UniversityNijmegenthe Netherlands
| | - Richard R. Drake
- Department of Cell and Molecular PharmacologyMedical University of South CarolinaCharlestonSouth CarolinaUSA
| | - Emma Scott
- Newcastle University Centre for CancerNewcastle University Institute of BiosciencesNewcastleUK
| | - David J. Elliott
- Newcastle University Centre for CancerNewcastle University Institute of BiosciencesNewcastleUK
| | - Jennifer Munkley
- Newcastle University Centre for CancerNewcastle University Institute of BiosciencesNewcastleUK
| |
Collapse
|
7
|
Slater AS, McDonald AG, Hickey RM, Davey GP. Glycosyltransferases: glycoengineers in human milk oligosaccharide synthesis and manufacturing. Front Mol Biosci 2025; 12:1587602. [PMID: 40370521 PMCID: PMC12074965 DOI: 10.3389/fmolb.2025.1587602] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2025] [Accepted: 04/11/2025] [Indexed: 05/16/2025] Open
Abstract
Human milk oligosaccharides (HMOs) are a diverse group of complex carbohydrates that play crucial roles in infant health, promoting a beneficial gut microbiota, modulating immune responses, and protecting against pathogens. Central to the synthesis of HMOs are glycosyltransferases, a specialized class of enzymes that catalyse the transfer of sugar moieties to form the complex glycan structures characteristic of HMOs. This review provides an in-depth analysis of glycosyltransferases, beginning with their classification based on structural and functional characteristics. The catalytic activity of these enzymes is explored, highlighting the mechanisms by which they facilitate the precise addition of monosaccharides in HMO biosynthesis. Structural insights into glycosyltransferases are also discussed, shedding light on how their conformational features enable specific glycosidic bond formations. This review maps out the key biosynthetic pathways involved in HMO production, including the synthesis of lactose, and subsequent fucosylation and sialylation processes, all of which are intricately regulated by glycosyltransferases. Industrial methods for HMO synthesis, including chemical, enzymatic, and microbial approaches, are examined, emphasizing the role of glycosyltransferases in these processes. Finally, the review discusses future directions in glycosyltransferase research, particularly in enhancing the efficiency of HMO synthesis and developing advanced analytical techniques to better understand the structural complexity and biological functions of HMOs.
Collapse
Affiliation(s)
- Alanna S. Slater
- Teagasc Food Research Centre, Moorepark, Fermoy, Ireland
- School of Biochemistry and Immunology, Trinity College Dublin, Dublin, Ireland
| | - Andrew G. McDonald
- School of Biochemistry and Immunology, Trinity College Dublin, Dublin, Ireland
| | - Rita M. Hickey
- Teagasc Food Research Centre, Moorepark, Fermoy, Ireland
| | - Gavin P. Davey
- School of Biochemistry and Immunology, Trinity College Dublin, Dublin, Ireland
| |
Collapse
|
8
|
Kizuka Y. The last piece in fucosylation. Nat Chem Biol 2025; 21:470-471. [PMID: 40033095 DOI: 10.1038/s41589-025-01850-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/05/2025]
Affiliation(s)
- Yasuhiko Kizuka
- Institute for Glyco-core Research (iGCORE), Gifu University, Gifu, Japan.
| |
Collapse
|
9
|
Hao H, Yuan Y, Ito A, Eberand BM, Tjondro H, Cielesh M, Norris N, Moreno CL, Maxwell JWC, Neely GG, Payne RJ, Kebede MA, Urbauer RJB, Passam FH, Larance M, Haltiwanger RS. FUT10 and FUT11 are protein O-fucosyltransferases that modify protein EMI domains. Nat Chem Biol 2025; 21:598-610. [PMID: 39775168 PMCID: PMC11949838 DOI: 10.1038/s41589-024-01815-x] [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: 02/06/2024] [Accepted: 12/04/2024] [Indexed: 01/11/2025]
Abstract
O-Fucosylation plays crucial roles in various essential biological events. Alongside the well-established O-fucosylation of epidermal growth factor-like repeats by protein O-fucosyltransferase 1 (POFUT1) and thrombospondin type 1 repeats by POFUT2, we recently identified a type of O-fucosylation on the elastin microfibril interface (EMI) domain of Multimerin-1 (MMRN1). Here, using AlphaFold2 screens, co-immunoprecipitation, enzymatic assays combined with mass spectrometric analysis and CRISPR-Cas9 knockouts, we demonstrate that FUT10 and FUT11, originally annotated in UniProt as α1,3-fucosyltransferases, are actually POFUTs responsible for modifying EMI domains; thus, we renamed them as POFUT3 and POFUT4, respectively. Like POFUT1/2, POFUT3/4 function in the endoplasmic reticulum, require folded domain structures for modification and participate in a non-canonical endoplasmic reticulum quality control pathway for EMI domain-containing protein secretion. This finding expands the O-fucosylation repertoire and provides an entry point for further exploration in this emerging field of O-fucosylation.
Collapse
Affiliation(s)
- Huilin Hao
- Complex Carbohydrate Research Center, University of Georgia, Athens, GA, USA
| | - Youxi Yuan
- Complex Carbohydrate Research Center, University of Georgia, Athens, GA, USA
| | - Atsuko Ito
- Complex Carbohydrate Research Center, University of Georgia, Athens, GA, USA
- Regional Fish Institute, Ltd., Kyoto, Japan
| | - Benjamin M Eberand
- Charles Perkins Centre, School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales, Australia
| | - Harry Tjondro
- Central Clinical School, The University of Sydney, Sydney, New South Wales, Australia
| | - Michelle Cielesh
- Charles Perkins Centre, School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales, Australia
| | - Nicholas Norris
- Charles Perkins Centre, School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales, Australia
| | - Cesar L Moreno
- Charles Perkins Centre, School of Life and Environmental Sciences, Faculty of Science, The University of Sydney, Sydney, New South Wales, Australia
| | - Joshua W C Maxwell
- School of Chemistry, Faculty of Science, The University of Sydney, Sydney, New South Wales, Australia
- Australian Research Council Centre of Excellence for Innovations in Peptide and Protein Science, The University of Sydney, Sydney, New South Wales, Australia
| | - G Gregory Neely
- Charles Perkins Centre, School of Life and Environmental Sciences, Faculty of Science, The University of Sydney, Sydney, New South Wales, Australia
| | - Richard J Payne
- School of Chemistry, Faculty of Science, The University of Sydney, Sydney, New South Wales, Australia
- Australian Research Council Centre of Excellence for Innovations in Peptide and Protein Science, The University of Sydney, Sydney, New South Wales, Australia
| | - Melkam A Kebede
- Charles Perkins Centre, School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales, Australia
| | | | - Freda H Passam
- Central Clinical School, The University of Sydney, Sydney, New South Wales, Australia
| | - Mark Larance
- Charles Perkins Centre, School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales, Australia.
| | | |
Collapse
|
10
|
Hao H, Eberand BM, Larance M, Haltiwanger RS. Protein O-Fucosyltransferases: Biological Functions and Molecular Mechanisms in Mammals. Molecules 2025; 30:1470. [PMID: 40286076 PMCID: PMC11990869 DOI: 10.3390/molecules30071470] [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: 02/22/2025] [Revised: 03/20/2025] [Accepted: 03/21/2025] [Indexed: 04/29/2025] Open
Abstract
Domain-specific O-fucosylation is an unusual type of glycosylation, where the fucose is directly attached to the serine or threonine residues in specific protein domains via an O-linkage. O-fucosylated proteins play critical roles in a wide variety of biological events and hold important therapeutic values, with the most studied being the Notch receptors and ADAMTS proteins. O-fucose glycans modulate the function of the proteins they modify and are closely associated with various diseases including cancer. In mammals, alongside the well-documented protein O-fucosyltransferase (POFUT) 1-mediated O-fucosylation of epidermal growth factor-like (EGF) repeats and POFUT2-mediated O-fucosylation of thrombospondin type 1 repeats (TSRs), a new type of O-fucosylation was recently identified on elastin microfibril interface (EMI) domains, mediated by POFUT3 and POFUT4 (formerly FUT10 and FUT11). In this review, we present an overview of our current knowledge of O-fucosylation, integrating the latest findings and with a particular focus on its biological functions and molecular mechanisms.
Collapse
Affiliation(s)
- Huilin Hao
- Complex Carbohydrate Research Center, University of Georgia, Athens, GA 30605, USA;
| | - Benjamin M. Eberand
- Charles Perkins Centre, School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW 2006, Australia; (B.M.E.); (M.L.)
| | - Mark Larance
- Charles Perkins Centre, School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW 2006, Australia; (B.M.E.); (M.L.)
| | | |
Collapse
|
11
|
Renata S, Verma N, Peddinti RK. Surface-enhanced Raman spectroscopy as effective tool for detection of sialic acid as cancer biomarker. SPECTROCHIMICA ACTA. PART A, MOLECULAR AND BIOMOLECULAR SPECTROSCOPY 2025; 329:125631. [PMID: 39736186 DOI: 10.1016/j.saa.2024.125631] [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: 10/01/2024] [Revised: 12/07/2024] [Accepted: 12/17/2024] [Indexed: 01/01/2025]
Abstract
Sialic acid, a negatively charged nine-carbon monosaccharide, is mainly located at the terminal end of glycan chains on glycoproteins and glycolipids of cell surface and most secreted proteins. Elevated levels of sialylated glycans have been known as a hallmark in numerous cancers. As a result, sialic acid acts as a useful and accessible cancer biomarker for early cancer detection and monitoring the disease development during cancer treatment which is crucial in elevating the survival rate. The detection of sialic acid has been done by many tools including surface-enhanced Raman spectroscopy (SERS) which gained incredible attention due to its high selectivity and sensitivity. However, currently, comprehensive reviews of sialic acid detection and imaging as a cancer biomarker using SERS are still lacking. Here, we present the significant breakthroughs in SERS-based detection of sialic acid levels on cells, tissues, and body fluids due to the presence of cancer, different cancer metastasis stages, and in response to the external stimuli. This review covers the SERS substrate and novel SERS strategies, using lectin, boronic acid, metabolic glycan labelling and label-free methods, for sialic acid detection as cancer biomarker. The remaining challenges to detect sialic acid and prospect of future development of SERS for other carbohydrate-based cancer biomarker, for instance fucose, are also discussed.
Collapse
Affiliation(s)
- Septila Renata
- Department of Chemistry, Indian Institute of Technology Roorkee, Roorkee 247667, Uttarakhand, India.
| | - Nitish Verma
- Department of Chemistry, Indian Institute of Technology Roorkee, Roorkee 247667, Uttarakhand, India; Department of Chemistry, University of Natural Resources and Life Sciences Vienna, Muthgasse 18, 1190 Vienna, Austria
| | - Rama Krishna Peddinti
- Department of Chemistry, Indian Institute of Technology Roorkee, Roorkee 247667, Uttarakhand, India.
| |
Collapse
|
12
|
Li Y, Fu B, Wang M, Chen W, Fan J, Li Y, Liu X, Wang J, Zhang Z, Lu H, Zhang Y. Urinary extracellular vesicle N-glycomics identifies diagnostic glycosignatures for bladder cancer. Nat Commun 2025; 16:2292. [PMID: 40055327 PMCID: PMC11889218 DOI: 10.1038/s41467-025-57633-9] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2024] [Accepted: 02/27/2025] [Indexed: 05/13/2025] Open
Abstract
Bladder cancer (BC) is the most common urologic malignancy, facing enormous diagnostic challenges. Urinary extracellular vesicles (EVs) are promising source for developing diagnostic markers for bladder cancer because of the direct contact between urine and bladder. This study pioneers urinary EV N-glycomics for bladder cancer diagnosis. We have generated a comprehensive N-glycome landscape of urinary EVs through high-throughput N-glycome analysis, identifying a total of 252 N-glycans from 333 individuals. In bladder cancer patients, urinary EVs exhibit decreased fucosylation and increased sialylation level. An Eight N-glycan diagnostic model demonstrates strong performance in both validation cohorts, achieving ROC AUC values of 0.88 and 0.86, respectively. Furthermore, this model successfully differentiates both non-muscle invasive bladder cancer (NMIBC) and muscle-invasive bladder cancer (MIBC) from healthy individuals, underscoring the model's superiority. Moreover, urinary EVs N-glycoproteomic analysis reveals that the glycoproteins carrying cancer-associated N-glycan signatures are closely associated with immune activities. The N-glycome comparative analysis of EVs and their source cells indicate that the glycosylation profiles of EVs do not completely match the glycosylation backgrounds of their source cells. In summary, our study establishes urinary EV N-glycomics as a non-invasive BC screening tool and provide a framework for EV glycan biomarker discovery across cancers.
Collapse
Affiliation(s)
- Yang Li
- Department of Chemistry, Minhang hospital and Institutes of Biomedical Sciences, Fudan University, Shanghai, PR China
| | - Bin Fu
- Department of Chemistry, Minhang hospital and Institutes of Biomedical Sciences, Fudan University, Shanghai, PR China
| | - Maoyu Wang
- Department of Urology, First Affiliated Hospital, Naval Medical University, Shanghai, China
| | - Weiyu Chen
- Department of Chemistry, Minhang hospital and Institutes of Biomedical Sciences, Fudan University, Shanghai, PR China
| | - Jiawei Fan
- Department of Chemistry, Minhang hospital and Institutes of Biomedical Sciences, Fudan University, Shanghai, PR China
| | - Yueyue Li
- Department of Chemistry, Minhang hospital and Institutes of Biomedical Sciences, Fudan University, Shanghai, PR China
| | - Xuejiao Liu
- Department of Chemistry, Minhang hospital and Institutes of Biomedical Sciences, Fudan University, Shanghai, PR China
| | - Jun Wang
- Department of Chemistry, Minhang hospital and Institutes of Biomedical Sciences, Fudan University, Shanghai, PR China
| | - Zhensheng Zhang
- Department of Urology, First Affiliated Hospital, Naval Medical University, Shanghai, China.
| | - Haojie Lu
- Department of Chemistry, Minhang hospital and Institutes of Biomedical Sciences, Fudan University, Shanghai, PR China.
- NHC Key Laboratory of Glycoconjugates Research, Fudan University, Shanghai, PR China.
| | - Ying Zhang
- Department of Chemistry, Minhang hospital and Institutes of Biomedical Sciences, Fudan University, Shanghai, PR China.
- NHC Key Laboratory of Glycoconjugates Research, Fudan University, Shanghai, PR China.
| |
Collapse
|
13
|
Yin Y, Liao L, Xu Q, Xie S, Yuan L, Zhou R. Insight into the post-translational modifications in pregnancy and related complications†. Biol Reprod 2025; 112:204-224. [PMID: 39499652 DOI: 10.1093/biolre/ioae149] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2024] [Revised: 09/19/2024] [Indexed: 11/07/2024] Open
Abstract
Successful pregnancy is dependent on a number of essential events, including embryo implantation, decidualization, and placentation. Failure of the above process may lead to pregnancy-related complications, including preeclampsia, gestational diabetes mellitus, preterm birth, and fetal growth restriction, may affect 15% of pregnancies, and lead to increased mortality and morbidity of pregnant women and perinatal infants, as well as the occurrence of short-term and long-term diseases. These complications have distinct etiology and pathogenesis, and the present comprehension is still lacking. Post-translational modifications are important events in epigenetics, altering the properties of proteins through protein hydrolysis or the addition of modification groups to one or more amino acids, with different modification states regulating subcellular localization, protein degradation, protein-protein interaction, signal transduction, and gene transcription. In this review, we focus on the impact of various post-translational modifications on the progress of embryo and placenta development and pregnancy-related complications, which will provide important experimental bases for exploring new insights into the physiology of pregnancy and pathogenesis associated with pregnancy complications.
Collapse
Affiliation(s)
- Yangxue Yin
- Department of Obstetrics and Gynecology, West China Second University Hospital, Sichuan University, Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University) of Ministry of Education, Chengdu, China
- National Health Commission Key Laboratory of Chronobiology, Sichuan University, Chengdu, China
| | - Lingyun Liao
- Department of Obstetrics and Gynecology, West China Second University Hospital, Sichuan University, Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University) of Ministry of Education, Chengdu, China
- National Health Commission Key Laboratory of Chronobiology, Sichuan University, Chengdu, China
| | - Qin Xu
- Department of Obstetrics and Gynecology, West China Second University Hospital, Sichuan University, Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University) of Ministry of Education, Chengdu, China
- National Health Commission Key Laboratory of Chronobiology, Sichuan University, Chengdu, China
| | - Shuangshuang Xie
- Department of Obstetrics and Gynecology, West China Second University Hospital, Sichuan University, Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University) of Ministry of Education, Chengdu, China
- National Health Commission Key Laboratory of Chronobiology, Sichuan University, Chengdu, China
| | - Liming Yuan
- Department of Obstetrics and Gynecology, West China Second University Hospital, Sichuan University, Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University) of Ministry of Education, Chengdu, China
- National Health Commission Key Laboratory of Chronobiology, Sichuan University, Chengdu, China
| | - Rong Zhou
- Department of Obstetrics and Gynecology, West China Second University Hospital, Sichuan University, Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University) of Ministry of Education, Chengdu, China
- National Health Commission Key Laboratory of Chronobiology, Sichuan University, Chengdu, China
| |
Collapse
|
14
|
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
|
15
|
Yang Y, Holck J, Thorhallsson AT, Hunt CJ, Yang H, Morth JP, Meyer AS, Zeuner B. Structural elucidation and characterization of GH29A α-l-fucosidases and the effect of pH on their transglycosylation. FEBS J 2025; 292:653-680. [PMID: 39658312 PMCID: PMC11796335 DOI: 10.1111/febs.17347] [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: 06/19/2024] [Revised: 10/11/2024] [Accepted: 11/27/2024] [Indexed: 12/12/2024]
Abstract
GH29A α-l-fucosidases (EC 3.2.1.51) catalyze the release of α-l-fucosyl moieties from the nonreducing end of glycoconjugates by hydrolysis and some also catalyze transglycosylation. The latter is particularly interesting with regard to designing enzymatic synthesis of human milk oligosaccharides (HMOs). We combined the bioinformatics tool conserved unique peptide patterns (CUPP) and phylogenetic clustering to discover new microbial GH29A α-l-fucosidases of the underexplored CUPP group GH29:13.1. Three uncharacterized bacterial enzymes (EaGH29, SeGH29, and PmGH29) and two previously identified GH29A α-l-fucosidases (BF3242 and TfFuc1) were selected for reaction optimization, biochemical, and structural characterization. Kinetics, pH-temperature optima, and substrate preference for 2-chloro-4-nitrophenyl-α-l-fucopyranoside (CNP-α-l-Fuc) and 2'-fucosyllactose (2'FL) were determined. Transglycosylation was favored at high neutral to alkaline pH, especially for EaGH29, SeGH29, TfFuc1, and BF3242, mainly because hydrolysis was decreased. The α-l-fucosidases exhibited medium regioselectivity in transglycosylation, generally forming two out of five detected lacto-N-fucopentaose (LNFP) isomers from 2'FL and lacto-N-tetraose (LNT). Alkaline pH also affected the transglycosylation product regioselectivity of SeGH29, which was also affected by a Leu/Phe exchange in the acceptor binding site. New crystal structures of TfFuc1 and BF3242 showed congruence in active site topology between these two enzymes and contributed to understanding the function of GH29A α-l-fucosidases. Notably, the structural data provide new insight into the role of an Asn residue located between the two catalytic residues in the active site.
Collapse
Affiliation(s)
- Yaya Yang
- School of Food and Biological EngineeringJiangsu UniversityZhenjiangChina
- Section for Protein Chemistry and Enzyme Technology, Department of Biotechnology and Biomedicine, DTU BioengineeringTechnical University of DenmarkKgs. LyngbyDenmark
| | - Jesper Holck
- Section for Protein Chemistry and Enzyme Technology, Department of Biotechnology and Biomedicine, DTU BioengineeringTechnical University of DenmarkKgs. LyngbyDenmark
| | - Albert Thor Thorhallsson
- Section for Protein Chemistry and Enzyme Technology, Department of Biotechnology and Biomedicine, DTU BioengineeringTechnical University of DenmarkKgs. LyngbyDenmark
| | - Cameron J. Hunt
- Section for Protein Chemistry and Enzyme Technology, Department of Biotechnology and Biomedicine, DTU BioengineeringTechnical University of DenmarkKgs. LyngbyDenmark
| | - Huan Yang
- School of Food and Biological EngineeringJiangsu UniversityZhenjiangChina
| | - Jens Preben Morth
- Section for Protein Chemistry and Enzyme Technology, Department of Biotechnology and Biomedicine, DTU BioengineeringTechnical University of DenmarkKgs. LyngbyDenmark
| | - Anne S. Meyer
- Section for Protein Chemistry and Enzyme Technology, Department of Biotechnology and Biomedicine, DTU BioengineeringTechnical University of DenmarkKgs. LyngbyDenmark
| | - Birgitte Zeuner
- Section for Protein Chemistry and Enzyme Technology, Department of Biotechnology and Biomedicine, DTU BioengineeringTechnical University of DenmarkKgs. LyngbyDenmark
| |
Collapse
|
16
|
Xu H, Li Q, Zhang Y, He C, Zhang X, Wang Z, Zhao M, Chai Y, Zhuang W, Li B. Targeting fucosyltransferase FUT8 as a prospective therapeutic approach for DLBCL. Oncogenesis 2025; 14:1. [PMID: 39881135 PMCID: PMC11779920 DOI: 10.1038/s41389-025-00544-7] [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: 07/05/2024] [Revised: 12/19/2024] [Accepted: 01/17/2025] [Indexed: 01/31/2025] Open
Abstract
Diffuse large B-cell lymphoma (DLBCL) is characterized by its aggressive nature and resistance to standard chemotherapy, necessitating the development of new therapeutic approaches. The emergence of natural products and their derivatives has notably influenced cancer treatment, making morusinol, a medicine-derived monomer, a promising candidate. Here, we showed that morusinol exerted antitumor effects on DLBCL in vitro by inducing apoptosis and cell cycle arrest. Impressively, morusinol treatment exhibited potent tumor growth inhibition in vivo, proving both well-tolerated and safe in mouse models. Moreover, our investigation identified FUT8, a fucosyltransferase, as a potential target for morusinol. FUT8's role as an oncogene in DLBCL and its correlation with poor survival further underscored its significance. Furthermore, our screening efforts involving clinical and preclinical drugs unveiled a compelling synergistic effect between chidamide and morusinol. Additionally, morusinol's ability to hinder M2-like polarization of tumor-associated macrophages suggested its potential in immune response modulation within DLBCL. Collectively, morusinol showcased substantial promise as an anti-tumor agent for potential clinical application in DLBCL management, potentially augmenting established therapeutic strategies. Moreover, our findings offered promising prospects for natural products to effectively leverage its therapeutic advantages. Working model: The role of Morusinol in treating DLBCL.
Collapse
Affiliation(s)
- Hao Xu
- Department of Hematology, The Second Affiliated Hospital of Soochow University, Suzhou, China
- Department of Cell Biology, School of Biology & Basic Medical Sciences, Suzhou Medical College of Soochow University, Suzhou, China
- Department of Medical Oncology, The People's Hospital of Rugao, Nantong, China
| | - Qi Li
- Department of Hematology, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Yuchen Zhang
- Department of Hematology, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Chuan He
- Department of Hematology, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Xinyun Zhang
- Department of Hematology, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Zhiming Wang
- Department of Cell Biology, School of Biology & Basic Medical Sciences, Suzhou Medical College of Soochow University, Suzhou, China
| | - Meifang Zhao
- Department of Hematology, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Yali Chai
- Department of Cell Biology, School of Biology & Basic Medical Sciences, Suzhou Medical College of Soochow University, Suzhou, China
| | - Wenzhuo Zhuang
- Department of Cell Biology, School of Biology & Basic Medical Sciences, Suzhou Medical College of Soochow University, Suzhou, China.
| | - Bingzong Li
- Department of Hematology, The Second Affiliated Hospital of Soochow University, Suzhou, China.
| |
Collapse
|
17
|
Paredes Franco JC, Güther MLS, Ferguson MAJ. Use of Crithidia fasciculata extract for the facile enzymatic synthesis of GDP-L-[3H]Fucose. Glycobiology 2025; 35:cwae097. [PMID: 39706805 PMCID: PMC11738170 DOI: 10.1093/glycob/cwae097] [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: 04/22/2024] [Revised: 10/31/2024] [Accepted: 12/19/2024] [Indexed: 12/23/2024] Open
Abstract
For studies involving glycosyltransferases and nucleotide sugar transporters, radioactive nucleotide sugars are critical reagents. Of these, GDP-L-[3H]Fucose is currently commercially unavailable. Here, we present a facile approach for the preparation of GDP-[3H]-L-Fucose, using the enzymatic machinery present in the cytosol of the non-infectious and easily cultivated protozoan, Crithidia fasciculata, and its purification by solid phase extraction ion exchange chromatography.
Collapse
Affiliation(s)
- Jose Carlos Paredes Franco
- Wellcome Centre for Anti-Infectives Research, Biological Chemistry and Drug Discovery, School of Life Sciences, Dow Street, University of Dundee, Dundee DD1 5HN, United Kingdom
- Current address: Department of Biological Sciences, University of Notre Dame, Notre Dame, Indiana 46556, United States of America
| | - Maria Lucia Sampaio Güther
- Wellcome Centre for Anti-Infectives Research, Biological Chemistry and Drug Discovery, School of Life Sciences, Dow Street, University of Dundee, Dundee DD1 5HN, United Kingdom
| | - Michael A J Ferguson
- Wellcome Centre for Anti-Infectives Research, Biological Chemistry and Drug Discovery, School of Life Sciences, Dow Street, University of Dundee, Dundee DD1 5HN, United Kingdom
| |
Collapse
|
18
|
Könighofer E, Mirgorodskaya E, Nyström K, Stiasny K, Kärmander A, Bergström T, Nordén R. Identification of Three Novel O-Linked Glycans in the Envelope Protein of Tick-Borne Encephalitis Virus. Viruses 2024; 16:1891. [PMID: 39772199 PMCID: PMC11680210 DOI: 10.3390/v16121891] [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: 10/10/2024] [Revised: 12/05/2024] [Accepted: 12/06/2024] [Indexed: 01/11/2025] Open
Abstract
The tick-borne encephalitis virus is a pathogen endemic to northern Europe and Asia, transmitted through bites from infected ticks. It is a member of the Flaviviridae family and possesses a positive-sense, single-stranded RNA genome encoding a polypeptide that is processed into seven non-structural and three structural proteins, including the envelope (E) protein. The glycosylation of the E protein, involving a single N-linked glycan at position N154, plays a critical role in viral infectivity and pathogenesis. Here, we dissected the entire glycosylation profile of the E protein using liquid chromatography-tandem mass spectrometry and identified three novel O-linked glycans, which were found at relatively low frequency. One of the O-linked glycans was positioned close to the highly conserved N-linked glycan site, and structural analysis suggested that it may be relevant for the function of the E 150-loop. The N154 site was found to be glycosylated with a high frequency, containing oligomannose or complex-type structures, some of which were fucosylated. An unusually high portion of oligomannose N-linked glycan structures exhibited compositions that are normally observed on proteins when they are translocated from the endoplasmic reticulum to the trans-Golgi network, suggesting disruption of the glycan processing pathway in the infected cells from which the E protein was obtained.
Collapse
Affiliation(s)
- Ebba Könighofer
- Department of Infectious Diseases, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, 413 46 Gothenburg, Sweden
| | - Ekaterina Mirgorodskaya
- Proteomics Core Facility, Sahlgrenska Academy, University of Gothenburg, 405 30 Gothenburg, Sweden
| | - Kristina Nyström
- Department of Infectious Diseases, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, 413 46 Gothenburg, Sweden
| | - Karin Stiasny
- Center for Virology, Medical University of Vienna, 1090 Vienna, Austria
| | - Ambjörn Kärmander
- Department of Infectious Diseases, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, 413 46 Gothenburg, Sweden
| | - Tomas Bergström
- Department of Infectious Diseases, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, 413 46 Gothenburg, Sweden
| | - Rickard Nordén
- Department of Infectious Diseases, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, 413 46 Gothenburg, Sweden
- Department of Clinical Microbiology, Sahlgrenska University Hospital, 413 46 Gothenburg, Sweden
| |
Collapse
|
19
|
Yang S, Jeong CM, Park CS, Moon C, Jang L, Jang JY, Lee HS, Kim K, Byeon H, Eom D, Kim HH. Identification and quantification of unreported sialylated N-glycan isomers with α2-3 and α2-6 linkages in the egg yolk protein phosvitin. Food Res Int 2024; 197:115293. [PMID: 39577941 DOI: 10.1016/j.foodres.2024.115293] [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: 07/03/2024] [Revised: 10/02/2024] [Accepted: 10/31/2024] [Indexed: 11/24/2024]
Abstract
Phosvitin (PV), a highly phosphorylated protein found in chicken egg yolk, possesses multiple bioactivities (including anti-aging and anticancer) and functional properties (including emulsifier and metal-binding capacities). The carbohydrate moiety attached to PV has been reported, but its N-glycan structure is unknown. In this study, we performed structural and quantitative analyses of N-glycans from PV using liquid chromatography-tandem mass spectrometry (MS/MS). N-glycan structures were identified using observed precursor ion m/z and MS/MS fragment ions. Each quantity was obtained relative to the total N-glycans (100%). Thirty-seven N-glycans were identified, including 22 sialylations with a negative charge (a sum of the relative quantity of each, 96.4%) comprising 13 mono- (31.6%), 7 di- (57.5%), 2 tri- (7.3%) sialylations. The sialylated N-glycan isomers with α2-3 (flexible conformation) and α2-6 (rigid conformation) linkages were distinguished using α2-3- and α2-3,6 sialidase treatments and intensity ratios of the N-acetylglucosamine and sialic acid ions (Ln/Nn) with different fragmentation stabilities. The α2-6/α2-6 (53.8%), α2-6 (31.6%), α2-3/α2-6/α2-6 (6.5%), and α2-3/α2-6 (3.7%) linkages in mono-, di, or tri-antennary structures were identified. These negatively charged structures may affect the emulsification and metal-binding capacity of PV. This is the first study to identify and quantify N-glycans in PV, including predominantly 22 sialylated N-glycan isomers with more rigid α2-6 linkages than α2-3 linkages.
Collapse
Affiliation(s)
- Subin Yang
- Biotherapeutics and Glycomics Laboratory, College of Pharmacy, Chung-Ang University, 84 Heukseok-ro, Dongjak-gu, Seoul 06974, Republic of Korea; Department of Global Innovative Drugs, Graduate School of Chung-Ang University, 84 Heukseok-ro, Dongjak-gu, Seoul 06974, Republic of Korea
| | - Chang Myeong Jeong
- Biotherapeutics and Glycomics Laboratory, College of Pharmacy, Chung-Ang University, 84 Heukseok-ro, Dongjak-gu, Seoul 06974, Republic of Korea; Department of Global Innovative Drugs, Graduate School of Chung-Ang University, 84 Heukseok-ro, Dongjak-gu, Seoul 06974, Republic of Korea
| | - Chi Soo Park
- Biotherapeutics and Glycomics Laboratory, College of Pharmacy, Chung-Ang University, 84 Heukseok-ro, Dongjak-gu, Seoul 06974, Republic of Korea; Department of Global Innovative Drugs, Graduate School of Chung-Ang University, 84 Heukseok-ro, Dongjak-gu, Seoul 06974, Republic of Korea
| | - Chulmin Moon
- Biotherapeutics and Glycomics Laboratory, College of Pharmacy, Chung-Ang University, 84 Heukseok-ro, Dongjak-gu, Seoul 06974, Republic of Korea; Department of Global Innovative Drugs, Graduate School of Chung-Ang University, 84 Heukseok-ro, Dongjak-gu, Seoul 06974, Republic of Korea
| | - Leeseul Jang
- Biotherapeutics and Glycomics Laboratory, College of Pharmacy, Chung-Ang University, 84 Heukseok-ro, Dongjak-gu, Seoul 06974, Republic of Korea; Department of Global Innovative Drugs, Graduate School of Chung-Ang University, 84 Heukseok-ro, Dongjak-gu, Seoul 06974, Republic of Korea
| | - Ji Yeon Jang
- Biotherapeutics and Glycomics Laboratory, College of Pharmacy, Chung-Ang University, 84 Heukseok-ro, Dongjak-gu, Seoul 06974, Republic of Korea; Department of Global Innovative Drugs, Graduate School of Chung-Ang University, 84 Heukseok-ro, Dongjak-gu, Seoul 06974, Republic of Korea
| | - Han Seul Lee
- Biotherapeutics and Glycomics Laboratory, College of Pharmacy, Chung-Ang University, 84 Heukseok-ro, Dongjak-gu, Seoul 06974, Republic of Korea; Department of Global Innovative Drugs, Graduate School of Chung-Ang University, 84 Heukseok-ro, Dongjak-gu, Seoul 06974, Republic of Korea
| | - Kyuran Kim
- Biotherapeutics and Glycomics Laboratory, College of Pharmacy, Chung-Ang University, 84 Heukseok-ro, Dongjak-gu, Seoul 06974, Republic of Korea; Department of Global Innovative Drugs, Graduate School of Chung-Ang University, 84 Heukseok-ro, Dongjak-gu, Seoul 06974, Republic of Korea
| | - Haeun Byeon
- Biotherapeutics and Glycomics Laboratory, College of Pharmacy, Chung-Ang University, 84 Heukseok-ro, Dongjak-gu, Seoul 06974, Republic of Korea; Department of Global Innovative Drugs, Graduate School of Chung-Ang University, 84 Heukseok-ro, Dongjak-gu, Seoul 06974, Republic of Korea
| | - Daeun Eom
- Biotherapeutics and Glycomics Laboratory, College of Pharmacy, Chung-Ang University, 84 Heukseok-ro, Dongjak-gu, Seoul 06974, Republic of Korea; Department of Global Innovative Drugs, Graduate School of Chung-Ang University, 84 Heukseok-ro, Dongjak-gu, Seoul 06974, Republic of Korea
| | - Ha Hyung Kim
- Biotherapeutics and Glycomics Laboratory, College of Pharmacy, Chung-Ang University, 84 Heukseok-ro, Dongjak-gu, Seoul 06974, Republic of Korea; Department of Global Innovative Drugs, Graduate School of Chung-Ang University, 84 Heukseok-ro, Dongjak-gu, Seoul 06974, Republic of Korea.
| |
Collapse
|
20
|
Skurska E, Olczak M. Interplay between de novo and salvage pathways of GDP-fucose synthesis. PLoS One 2024; 19:e0309450. [PMID: 39446915 PMCID: PMC11501016 DOI: 10.1371/journal.pone.0309450] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2024] [Accepted: 08/12/2024] [Indexed: 10/26/2024] Open
Abstract
GDP-fucose is synthesised via two pathways: de novo and salvage. The first uses GDP-mannose as a substrate, and the second uses free fucose. To date, these pathways have been considered to work separately and not to have an influence on each other. We report the mutual response of the de novo and salvage pathways to the lack of enzymes from a particular route of GDP-fucose synthesis. We detected different efficiencies of GDP-fucose and fucosylated structure synthesis after a single inactivation of enzymes of the de novo pathway. Our study demonstrated the unequal influence of the salvage enzymes on the production of GDP-fucose by enzymes of the de novo biosynthesis pathway. Simultaneously, we detected an elevated level of one of the enzymes of the de novo pathway in the cell line lacking the enzyme of the salvage biosynthesis pathway. Additionally, we identified dissimilarities in fucose uptake between cells lacking TSTA3 and GMDS proteins.
Collapse
Affiliation(s)
- Edyta Skurska
- Department of Biochemistry, Faculty of Biotechnology, University of Wroclaw, Wroclaw, Poland
| | - Mariusz Olczak
- Department of Biochemistry, Faculty of Biotechnology, University of Wroclaw, Wroclaw, Poland
| |
Collapse
|
21
|
Lembo A, Molinaro A, De Castro C, Berti F, Biagini M. Impact of glycosylation on viral vaccines. Carbohydr Polym 2024; 342:122402. [PMID: 39048237 DOI: 10.1016/j.carbpol.2024.122402] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2024] [Revised: 05/24/2024] [Accepted: 06/11/2024] [Indexed: 07/27/2024]
Abstract
Glycosylation is the most prominent modification important for vaccines and its specific pattern depends on several factors that need to be considered when developing a new biopharmaceutical. Tailor-made glycosylation can be exploited to develop more effective and safer vaccines; for this reason, a deep understanding of both glycoengineering strategies and glycans structures and functions is required. In this review we discuss the recent advances concerning glycoprotein expression systems and the explanation of glycans immunomodulation mechanisms. Furthermore, we highlight how glycans tune the immunological properties among different vaccines platforms (whole virus, recombinant protein, nucleic acid), also comparing commercially available formulations and describing the state-of-the-art analytical technologies for glycosylation analysis. The whole review stresses the aspect of glycoprotein glycans as a potential tool to overcome nowadays medical needs in vaccine field.
Collapse
Affiliation(s)
- Antonio Lembo
- Department of Chemical Sciences, University of Naples Federico II, Naples, Italy; GSK, Siena, Italy
| | - Antonio Molinaro
- Department of Chemical Sciences, University of Naples Federico II, Naples, Italy
| | - Cristina De Castro
- Department of Chemical Sciences, University of Naples Federico II, Naples, Italy.
| | | | | |
Collapse
|
22
|
Chen J, Wen P, Tang YH, Li H, Wang Z, Wang X, Zhou X, Gao XD, Fujita M, Yang G. Proteome and Glycoproteome Analyses Reveal Regulation of Protein Glycosylation Site-Specific Occupancy and Lysosomal Hydrolase Maturation by N-Glycan-Dependent ER-Quality Control. J Proteome Res 2024; 23:4409-4421. [PMID: 39235835 DOI: 10.1021/acs.jproteome.4c00378] [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] [Indexed: 09/06/2024]
Abstract
N-Glycan-dependent endoplasmic reticulum quality control (ERQC) primarily mediates protein folding, which determines the fate of the polypeptide. Monoglucose residues on N-glycans determine whether the nascent N-glycosylated proteins enter into and escape from the calnexin (CANX)/calreticulin (CALR) cycle, which is a central system of the ERQC. To reveal the impact of ERQC on glycosylation and protein fate, we performed comprehensive quantitative proteomic and glycoproteomic analyses using cells defective in N-glycan-dependent ERQC. Deficiency of MOGS encoding the ER α-glucosidase I, CANX, or/and CALR broadly affected protein expression and glycosylation. Among the altered glycoproteins, the occupancy of oligomannosidic N-glycans was significantly affected. Besides the expected ER stress, proteins and glycoproteins involved in pathways for lysosome and viral infection are differentially changed in those deficient cells. We demonstrated that lysosomal hydrolases were not correctly modified with mannose-6-phosphates on the N-glycans and were directly secreted to the culture medium in N-glycan-dependent ERQC mutant cells. Overall, the CANX/CALR cycle promotes the correct folding of glycosylated peptides and influences the transport of lysosomal hydrolases.
Collapse
Affiliation(s)
- Jingru Chen
- Key Laboratory of Carbohydrate Chemistry and Biotechnology, Ministry of Education, School of Biotechnology, Jiangnan University, Wuxi, Jiangsu 214122, China
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing 100190, China
- Key Laboratory of Biopharmaceutical Preparation and Delivery, Chinese Academy of Sciences, Beijing 100190, China
| | - Piaopiao Wen
- Key Laboratory of Carbohydrate Chemistry and Biotechnology, Ministry of Education, School of Biotechnology, Jiangnan University, Wuxi, Jiangsu 214122, China
| | - Yu-He Tang
- Key Laboratory of Carbohydrate Chemistry and Biotechnology, Ministry of Education, School of Biotechnology, Jiangnan University, Wuxi, Jiangsu 214122, China
| | - Hanjie Li
- Key Laboratory of Carbohydrate Chemistry and Biotechnology, Ministry of Education, School of Biotechnology, Jiangnan University, Wuxi, Jiangsu 214122, China
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing 100190, China
- Key Laboratory of Biopharmaceutical Preparation and Delivery, Chinese Academy of Sciences, Beijing 100190, China
| | - Zibo Wang
- Key Laboratory of Carbohydrate Chemistry and Biotechnology, Ministry of Education, School of Biotechnology, Jiangnan University, Wuxi, Jiangsu 214122, China
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing 100190, China
- Key Laboratory of Biopharmaceutical Preparation and Delivery, Chinese Academy of Sciences, Beijing 100190, China
| | - Xiuyuan Wang
- Key Laboratory of Carbohydrate Chemistry and Biotechnology, Ministry of Education, School of Biotechnology, Jiangnan University, Wuxi, Jiangsu 214122, China
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing 100190, China
- Key Laboratory of Biopharmaceutical Preparation and Delivery, Chinese Academy of Sciences, Beijing 100190, China
| | - Xiaoman Zhou
- Key Laboratory of Carbohydrate Chemistry and Biotechnology, Ministry of Education, School of Biotechnology, Jiangnan University, Wuxi, Jiangsu 214122, China
| | - Xiao-Dong Gao
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing 100190, China
- Key Laboratory of Biopharmaceutical Preparation and Delivery, Chinese Academy of Sciences, Beijing 100190, China
| | - Morihisa Fujita
- Key Laboratory of Carbohydrate Chemistry and Biotechnology, Ministry of Education, School of Biotechnology, Jiangnan University, Wuxi, Jiangsu 214122, China
- Institute for Glyco-core Research (iGCORE), Gifu University, Gifu 501-1193, Japan
| | - Ganglong Yang
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing 100190, China
- Key Laboratory of Biopharmaceutical Preparation and Delivery, Chinese Academy of Sciences, Beijing 100190, China
| |
Collapse
|
23
|
Stanley P. Genetics of glycosylation in mammalian development and disease. Nat Rev Genet 2024; 25:715-729. [PMID: 38724711 DOI: 10.1038/s41576-024-00725-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/12/2024] [Indexed: 09/19/2024]
Abstract
Glycosylation of proteins and lipids in mammals is essential for embryogenesis and the development of all tissues. Analyses of glycosylation mutants in cultured mammalian cells and model organisms have been key to defining glycosylation pathways and the biological functions of glycans. More recently, applications of genome sequencing have revealed the breadth of rare congenital disorders of glycosylation in humans and the influence of genetics on the synthesis of glycans relevant to infectious diseases, cancer progression and diseases of the immune system. This improved understanding of glycan synthesis and functions is paving the way for advances in the diagnosis and treatment of glycosylation-related diseases, including the development of glycoprotein therapeutics through glycosylation engineering.
Collapse
Affiliation(s)
- Pamela Stanley
- Department of Cell Biology, Albert Einstein College of Medicine, New York, NY, USA.
| |
Collapse
|
24
|
Gough EK, Edens TJ, Carr L, Robertson RC, Mutasa K, Ntozini R, Chasekwa B, Geum HM, Baharmand I, Gill SK, Mutasa B, Mbuya MNN, Majo FD, Tavengwa N, Francis F, Tome J, Evans C, Kosek M, Prendergast AJ, Manges AR. Bifidobacterium longum and microbiome maturation modify a nutrient intervention for stunting in Zimbabwean infants. EBioMedicine 2024; 108:105362. [PMID: 39341154 PMCID: PMC11467582 DOI: 10.1016/j.ebiom.2024.105362] [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: 04/13/2024] [Revised: 09/06/2024] [Accepted: 09/10/2024] [Indexed: 09/30/2024] Open
Abstract
BACKGROUND Small-quantity lipid-based nutrient supplements (SQ-LNS), which has been widely tested to reduce child stunting, has largely modest effects to date, but the mechanisms underlying these modest effects are unclear. Child stunting is a longstanding indicator of chronic undernutrition and it remains a prevalent public health problem. The infant gut microbiome may be a key contributor to stunting; and mother and infant fucosyltransferase (FUT) phenotypes are important determinants of infant microbiome composition. METHODS We investigated whether mother-infant FUT status (n = 792) and infant gut microbiome composition (n = 354 fecal specimens from 172 infants) modified the impact of an infant and young child feeding (IYCF) intervention, that included SQ-LNS, on stunting at age 18 months in secondary analysis of a randomized trial in rural Zimbabwe. FINDINGS We found that the impact of the IYCF intervention on stunting was modified by: (i) mother-infant FUT2+/FUT3- phenotype (difference-in-differences -32.6% [95% CI: -55.3%, -9.9%]); (ii) changes in species composition that reflected microbiome maturation (difference-in-differences -68.1% [95% CI: -99.0%, -28.5%); and (iii) greater relative abundance of B. longum (differences-in-differences 49.1% [95% CI: 26.6%, 73.6%]). The dominant strains of B. longum when the intervention started were most similar to the proficient milk oligosaccharide utilizer subspecies infantis, which decreased with infant age and differed by mother-infant FUT2+/FUT3- phenotypes. INTERPRETATION These findings indicate that a persistently "younger" microbiome at initiation of the intervention reduced its benefits on stunting in areas with a high prevalence of growth restriction. FUNDING Bill and Melinda Gates Foundation, UK DFID/Aid, Wellcome Trust, Swiss Agency for Development and Cooperation, US National Institutes of Health, UNICEF, and Nutricia Research Foundation.
Collapse
Affiliation(s)
- Ethan K Gough
- Department of International Health, Johns Hopkins Bloomberg School of Public Health; Baltimore, MD, USA.
| | | | - Lynnea Carr
- Department of Microbiology and Immunology, University of British Columbia; Vancouver, BC, Canada
| | | | - Kuda Mutasa
- Zvitambo Institute for Maternal and Child Health Research, Harare, Zimbabwe
| | - Robert Ntozini
- Zvitambo Institute for Maternal and Child Health Research, Harare, Zimbabwe
| | - Bernard Chasekwa
- Zvitambo Institute for Maternal and Child Health Research, Harare, Zimbabwe
| | - Hyun Min Geum
- School of Population and Public Health, University of British Columbia, Vancouver, BC, Canada
| | - Iman Baharmand
- School of Population and Public Health, University of British Columbia, Vancouver, BC, Canada
| | - Sandeep K Gill
- School of Population and Public Health, University of British Columbia, Vancouver, BC, Canada
| | - Batsirai Mutasa
- Zvitambo Institute for Maternal and Child Health Research, Harare, Zimbabwe
| | - Mduduzi N N Mbuya
- Zvitambo Institute for Maternal and Child Health Research, Harare, Zimbabwe; Global Alliance for Improved Nutrition, Washington, DC, 20036, USA
| | - Florence D Majo
- Zvitambo Institute for Maternal and Child Health Research, Harare, Zimbabwe
| | - Naume Tavengwa
- Zvitambo Institute for Maternal and Child Health Research, Harare, Zimbabwe
| | - Freddy Francis
- Department of Experimental Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Joice Tome
- Zvitambo Institute for Maternal and Child Health Research, Harare, Zimbabwe
| | - Ceri Evans
- Blizard Institute, Queen Mary University of London, London, UK; Zvitambo Institute for Maternal and Child Health Research, Harare, Zimbabwe; Department of Clinical Infection, Microbiology and Immunology, University of Liverpool, Liverpool, UK
| | - Margaret Kosek
- University of Virginia School of Medicine, Charlottesville, VA, USA
| | - Andrew J Prendergast
- Blizard Institute, Queen Mary University of London, London, UK; Zvitambo Institute for Maternal and Child Health Research, Harare, Zimbabwe
| | - Amee R Manges
- School of Population and Public Health, University of British Columbia, Vancouver, BC, Canada; British Columbia Centre for Disease Control (BCCDC), Vancouver, BC, Canada
| |
Collapse
|
25
|
Zemkollari M, Ruprecht C, Blaukopf M, Grabherr R, Staudacher E. Cloning, expression and characterisation of a novel mollusc α-1,2-Fucosyltransferase from Crassostrea gigas (CgFUT2). Glycoconj J 2024; 41:255-265. [PMID: 39162891 PMCID: PMC11522050 DOI: 10.1007/s10719-024-10162-x] [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: 05/14/2024] [Revised: 07/01/2024] [Accepted: 07/29/2024] [Indexed: 08/21/2024]
Abstract
Glycans containing fucose play crucial roles in cell biology, particularly in recognition processes. In humans, fucose found in H-blood group antigens is recognized by various pathogens, thereby influencing host-pathogen interactions. However, in invertebrate biology the specific functions of these modifications and the corresponding glycosyltransferases are not fully elucidated. Therefore, cloning these glycosyltransferases from different model systems will provide valuable insights into this process. Little is known about fucosyltransferases in molluscs. For this study, a sequence of the Pacific oyster, Crassostrea gigas, based on amino acid sequence homologies with rabbit and human α-1,2-fucosyltransferases, was chosen. The recombinant enzyme (350 amino acids) was able to transfer fucose from GDP-fucose to the galactose residue of type II disaccharides, terminal galactoses in complex N-glycan structures and several linear and branched galactans which were tested using a glycan microarray. The α-1,2-linkage formed was confirmed by NMR analysis. The enzyme was active in a broad pH-range, it was relatively stable upon storage conditions and its activity was not dependent on the presence of divalent cations. In this study, we were able to clone, express and characterise a novel α-1,2-fucosyltrasferase from Crassostrea gigas (CgFUT2).
Collapse
Affiliation(s)
- Marilica Zemkollari
- Department of Chemistry, University of Natural Resources and Life Sciences, Vienna, Austria
| | - Colin Ruprecht
- Department of Chemistry, University of Natural Resources and Life Sciences, Vienna, Austria
| | - Markus Blaukopf
- Department of Chemistry, University of Natural Resources and Life Sciences, Vienna, Austria
| | - Reingard Grabherr
- Department of Biotechnology, University of Natural Resources and Life Sciences, Vienna, Austria
| | - Erika Staudacher
- Department of Chemistry, University of Natural Resources and Life Sciences, Vienna, Austria.
| |
Collapse
|
26
|
Zhu Q, Chaubard JL, Geng D, Shen J, Ban L, Cheung ST, Wei F, Liu Y, Sun H, Calderon A, Dong W, Qin W, Li T, Wen L, Wang PG, Sun S, Yi W, Hsieh-Wilson LC. Chemoenzymatic Labeling, Detection and Profiling of Core Fucosylation in Live Cells. J Am Chem Soc 2024; 146:26408-26415. [PMID: 39279393 DOI: 10.1021/jacs.4c09303] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/18/2024]
Abstract
Core fucosylation, the attachment of an α-1,6-linked-fucose to the N-glycan core pentasaccharide, is an abundant protein modification that plays critical roles in various biological processes such as cell signaling, B cell development, antibody-dependent cellular cytotoxicity, and oncogenesis. However, the tools currently used to detect core fucosylation suffer from poor specificity, exhibiting cross-reactivity against all types of fucosylation. Herein we report the development of a new chemoenzymatic strategy for the rapid and selective detection of core fucosylated glycans. This approach employs a galactosyltransferase enzyme identified fromCaenorhabditis elegansthat specifically transfers an azido-appended galactose residue onto core fucose via a β-1,4 glycosidic linkage. We demonstrate that the approach exhibits superior specificity toward core fucose on a variety of complex N-glycans. The method enables detection of core fucosylated glycoproteins from complex cell lysates, as well as on live cell surfaces, and it can be integrated into a diagnostic platform to profile protein-specific core fucosylation levels. This chemoenzymatic labeling approach offers a new strategy for the identification of disease biomarkers and will allow researchers to further characterize the fundamental role of this important glycan in normal and disease physiology.
Collapse
Affiliation(s)
- Qiang Zhu
- College of Life Sciences, Zhejiang University, Hangzhou 310012, China
| | - Jean-Luc Chaubard
- Division of Chemistry and Chemical Engineering, California Institute of Technology, 1200 E. California Blvd, Pasadena, California 91125, United States
| | - Didi Geng
- College of Life Sciences, Zhejiang University, Hangzhou 310012, China
| | - Jiechen Shen
- College of Life Sciences, Northwest University, Xi'an 710069, China
| | - Lan Ban
- Division of Chemistry and Chemical Engineering, California Institute of Technology, 1200 E. California Blvd, Pasadena, California 91125, United States
| | - Sheldon T Cheung
- Division of Chemistry and Chemical Engineering, California Institute of Technology, 1200 E. California Blvd, Pasadena, California 91125, United States
| | - Fangyu Wei
- Carbohydrate-Based Drug Research Center, Shanghai Institute of Materia Medica, The Chinese Academy of Sciences, Shanghai 201203, China
| | - Yating Liu
- Carbohydrate-Based Drug Research Center, Shanghai Institute of Materia Medica, The Chinese Academy of Sciences, Shanghai 201203, China
| | - Haofan Sun
- State Key Laboratory of Proteomics, Beijing Institute of Lifeomics, National Center for Protein Sciences Beijing, Beijing 102206, China
| | - Angie Calderon
- Department of Pharmacology, School of Medicine, Southern University of Science and Technology Institution, Shenzhen, Guangdong 518055, China
| | - Wenbo Dong
- College of Life Sciences, Northwest University, Xi'an 710069, China
| | - Weijie Qin
- State Key Laboratory of Proteomics, Beijing Institute of Lifeomics, National Center for Protein Sciences Beijing, Beijing 102206, China
| | - Tiehai Li
- Carbohydrate-Based Drug Research Center, Shanghai Institute of Materia Medica, The Chinese Academy of Sciences, Shanghai 201203, China
| | - Liuqing Wen
- Carbohydrate-Based Drug Research Center, Shanghai Institute of Materia Medica, The Chinese Academy of Sciences, Shanghai 201203, China
| | - Peng George Wang
- Department of Pharmacology, School of Medicine, Southern University of Science and Technology Institution, Shenzhen, Guangdong 518055, China
| | - Shisheng Sun
- College of Life Sciences, Northwest University, Xi'an 710069, China
| | - Wen Yi
- College of Life Sciences, Zhejiang University, Hangzhou 310012, China
| | - Linda C Hsieh-Wilson
- Division of Chemistry and Chemical Engineering, California Institute of Technology, 1200 E. California Blvd, Pasadena, California 91125, United States
| |
Collapse
|
27
|
Feng Y, Sun L, Dang X, Liu D, Liao Z, Yao J, Zhang Y, Deng Z, Li J, Zhao M, Liu F. Aberrant glycosylation in schizophrenia: insights into pathophysiological mechanisms and therapeutic potentials. Front Pharmacol 2024; 15:1457811. [PMID: 39286629 PMCID: PMC11402814 DOI: 10.3389/fphar.2024.1457811] [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: 07/01/2024] [Accepted: 08/22/2024] [Indexed: 09/19/2024] Open
Abstract
Schizophrenia (SCZ) is a severe neuropsychiatric disorder characterized by cognitive, affective, and social dysfunction, resulting in hallucinations, delusions, emotional blunting, and disordered thinking. In recent years, proteomics has been increasingly influential in SCZ research. Glycosylation, a key post-translational modification, can alter neuronal stability and normal signaling in the nervous system by affecting protein folding, stability, and cellular signaling. Recent research evidence suggests that abnormal glycosylation patterns exist in different brain regions in autopsy samples from SCZ patients, and that there are significant differences in various glycosylation modification types and glycosylation modifying enzymes. Therefore, this review explores the mechanisms of aberrant modifications of N-glycosylation, O-glycosylation, glycosyltransferases, and polysialic acid in the brains of SCZ patients, emphasizing their roles in neurotransmitter receptor function, synaptic plasticity, and neural adhesion. Additionally, the effects of antipsychotic drugs on glycosylation processes and the potential for glycosylation-targeted therapies are discussed. By integrating these findings, this review aims to provide a comprehensive perspective to further understand the role of aberrant glycosylation modifications in the pathophysiology of SCZ.
Collapse
Affiliation(s)
- Yanchen Feng
- The First Clinical Medical School, Henan University of Chinese Medicine, Zhengzhou, China
- Traditional Chinese Medicine (Zhong Jing) School, Henan University of Chinese Medicine, Zhengzhou, China
| | - Lu Sun
- The First Clinical Medical School, Henan University of Chinese Medicine, Zhengzhou, China
| | - Xue Dang
- Traditional Chinese Medicine (Zhong Jing) School, Henan University of Chinese Medicine, Zhengzhou, China
| | - Diyan Liu
- Traditional Chinese Medicine (Zhong Jing) School, Henan University of Chinese Medicine, Zhengzhou, China
| | - Ziyun Liao
- College of Acupuncture, Moxibustion and Tuina, Henan University of Chinese Medicine, Zhengzhou, China
| | - Jianping Yao
- Traditional Chinese Medicine (Zhong Jing) School, Henan University of Chinese Medicine, Zhengzhou, China
| | - Yunke Zhang
- School of Rehabilitation Medicine, Henan University of Chinese Medicine, Zhengzhou, China
| | - Ziqi Deng
- School of Pharmacy, Henan University of Chinese Medicine, Zhengzhou, China
| | - Jinyao Li
- Traditional Chinese Medicine (Zhong Jing) School, Henan University of Chinese Medicine, Zhengzhou, China
| | - Min Zhao
- The First Clinical Medical School, Henan University of Chinese Medicine, Zhengzhou, China
- Hospital of Encephalopathy, The First Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou, China
| | - Feixiang Liu
- The First Clinical Medical School, Henan University of Chinese Medicine, Zhengzhou, China
- Hospital of Encephalopathy, The First Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou, China
| |
Collapse
|
28
|
Ma Q, Zhang YH, Guo W, Feng K, Huang T, Cai YD. Machine Learning in Identifying Marker Genes for Congenital Heart Diseases of Different Cardiac Cell Types. Life (Basel) 2024; 14:1032. [PMID: 39202774 PMCID: PMC11355424 DOI: 10.3390/life14081032] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2024] [Revised: 07/31/2024] [Accepted: 08/14/2024] [Indexed: 09/03/2024] Open
Abstract
Congenital heart disease (CHD) represents a spectrum of inborn heart defects influenced by genetic and environmental factors. This study advances the field by analyzing gene expression profiles in 21,034 cardiac fibroblasts, 73,296 cardiomyocytes, and 35,673 endothelial cells, utilizing single-cell level analysis and machine learning techniques. Six CHD conditions: dilated cardiomyopathy (DCM), donor hearts (used as healthy controls), hypertrophic cardiomyopathy (HCM), heart failure with hypoplastic left heart syndrome (HF_HLHS), Neonatal Hypoplastic Left Heart Syndrome (Neo_HLHS), and Tetralogy of Fallot (TOF), were investigated for each cardiac cell type. Each cell sample was represented by 29,266 gene features. These features were first analyzed by six feature-ranking algorithms, resulting in several feature lists. Then, these lists were fed into incremental feature selection, containing two classification algorithms, to extract essential gene features and classification rules and build efficient classifiers. The identified essential genes can be potential CHD markers in different cardiac cell types. For instance, the LASSO identified key genes specific to various heart cell types in CHD subtypes. FOXO3 was found to be up-regulated in cardiac fibroblasts for both Dilated and hypertrophic cardiomyopathy. In cardiomyocytes, distinct genes such as TMTC1, ART3, ARHGAP24, SHROOM3, and XIST were linked to dilated cardiomyopathy, Neo-Hypoplastic Left Heart Syndrome, hypertrophic cardiomyopathy, HF-Hypoplastic Left Heart Syndrome, and Tetralogy of Fallot, respectively. Endothelial cell analysis further revealed COL25A1, NFIB, and KLF7 as significant genes for dilated cardiomyopathy, hypertrophic cardiomyopathy, and Tetralogy of Fallot. LightGBM, Catboost, MCFS, RF, and XGBoost further delineated key genes for specific CHD subtypes, demonstrating the efficacy of machine learning in identifying CHD-specific genes. Additionally, this study developed quantitative rules for representing the gene expression patterns related to CHDs. This research underscores the potential of machine learning in unraveling the molecular complexities of CHD and establishes a foundation for future mechanism-based studies.
Collapse
Affiliation(s)
- Qinglan Ma
- School of Life Sciences, Shanghai University, Shanghai 200444, China;
| | - Yu-Hang Zhang
- Channing Division of Network Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA;
| | - Wei Guo
- Key Laboratory of Stem Cell Biology, Shanghai Jiao Tong University School of Medicine (SJTUSM) & Shanghai Institutes for Biological Sciences (SIBS), Chinese Academy of Sciences (CAS), Shanghai 200030, China;
| | - Kaiyan Feng
- Department of Computer Science, Guangdong AIB Polytechnic College, Guangzhou 510507, China;
| | - Tao Huang
- Bio-Med Big Data Center, CAS Key Laboratory of Computational Biology, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Yu-Dong Cai
- School of Life Sciences, Shanghai University, Shanghai 200444, China;
| |
Collapse
|
29
|
Jang L, Kim A, Park CS, Moon C, Kim M, Kim J, Yang S, Jang JY, Jeong CM, Lee HS, Park J, Kim K, Byeon H, Kim HH. Fucosylation and galactosylation in N-glycans of bovine intestinal alkaline phosphatase and their role in its enzymatic activity. Arch Biochem Biophys 2024; 758:110069. [PMID: 38914216 DOI: 10.1016/j.abb.2024.110069] [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: 01/03/2024] [Revised: 06/20/2024] [Accepted: 06/20/2024] [Indexed: 06/26/2024]
Abstract
Bovine intestinal alkaline phosphatase (biALP), a membrane-bound plasma metalloenzyme, maintains intestinal homeostasis, regulates duodenal surface pH, and protects against infections caused by pathogenic bacteria. The N-glycans of biALP regulate its enzymatic activity, protein folding, and thermostability, but their structures are not fully reported. In this study, the structures and quantities of the N-glycans of biALP were analyzed by liquid chromatography-electrospray ionization-high energy collision dissociation-tandem mass spectrometry. In total, 48 N-glycans were identified and quantified, comprising high-mannose [6 N-glycans, 33.1 % (sum of relative quantities of each N-glycan)], hybrid (6, 11.9 %), and complex (36, 55.0 %) structures [bi- (13, 26.1 %), tri- (16, 21.5 %), and tetra-antennary (7, 7.4 %)]. These included bisecting N-acetylglucosamine (33, 56.6 %), mono-to tri-fucosylation (32, 53.3 %), mono-to tri-α-galactosylation (16, 20.7 %), and mono-to tetra-β-galactosylation (36, 58.5 %). No sialylation was identified. N-glycans with non-bisecting GlcNAc (9, 10.3 %), non-fucosylation (10, 13.6 %), non-α-galactosylation (26, 46.2 %), and non-β-galactosylation (6, 8.4 %) were also identified. The activity (100 %) of biALP was reduced to 37.3 ± 0.2 % (by de-fucosylation), 32.7 ± 2.9 % (by de-α-galactosylation), and 0.2 ± 0.2 % (by de-β-galactosylation), comparable to inhibition by 10-4 to 101 mM EDTA, a biALP inhibitor. These results indicate that fucosylated and galactosylated N-glycans, especially β-galactosylation, affected the activity of biALP. This study is the first to identify 48 diverse N-glycan structures and quantities of bovine as well as human intestinal ALP and to demonstrate the importance of the role of fucosylation and galactosylation for maintaining the activity of biALP.
Collapse
Affiliation(s)
- Leeseul Jang
- Department of Global Innovative Drugs, Graduate School of Chung-Ang University, 84 Heukseok-ro, Dongjak-gu, Seoul, 06974, Republic of Korea
| | - Ahyeon Kim
- Department of Global Innovative Drugs, Graduate School of Chung-Ang University, 84 Heukseok-ro, Dongjak-gu, Seoul, 06974, Republic of Korea
| | - Chi Soo Park
- Department of Global Innovative Drugs, Graduate School of Chung-Ang University, 84 Heukseok-ro, Dongjak-gu, Seoul, 06974, Republic of Korea
| | - Chulmin Moon
- Department of Global Innovative Drugs, Graduate School of Chung-Ang University, 84 Heukseok-ro, Dongjak-gu, Seoul, 06974, Republic of Korea
| | - Mirae Kim
- Department of Global Innovative Drugs, Graduate School of Chung-Ang University, 84 Heukseok-ro, Dongjak-gu, Seoul, 06974, Republic of Korea
| | - Jieun Kim
- Department of Global Innovative Drugs, Graduate School of Chung-Ang University, 84 Heukseok-ro, Dongjak-gu, Seoul, 06974, Republic of Korea
| | - Subin Yang
- Department of Global Innovative Drugs, Graduate School of Chung-Ang University, 84 Heukseok-ro, Dongjak-gu, Seoul, 06974, Republic of Korea
| | - Ji Yeon Jang
- Department of Global Innovative Drugs, Graduate School of Chung-Ang University, 84 Heukseok-ro, Dongjak-gu, Seoul, 06974, Republic of Korea
| | - Chang Myeong Jeong
- Department of Global Innovative Drugs, Graduate School of Chung-Ang University, 84 Heukseok-ro, Dongjak-gu, Seoul, 06974, Republic of Korea
| | - Han Seul Lee
- Department of Global Innovative Drugs, Graduate School of Chung-Ang University, 84 Heukseok-ro, Dongjak-gu, Seoul, 06974, Republic of Korea
| | - Juhee Park
- Department of Pharmaceutical Regulatory Sciences, Graduate School of Chung-Ang University, 84 Heukseok-ro, Dongjak-gu, Seoul, 06974, Republic of Korea
| | - Kyuran Kim
- Department of Global Innovative Drugs, Graduate School of Chung-Ang University, 84 Heukseok-ro, Dongjak-gu, Seoul, 06974, Republic of Korea
| | - Haeun Byeon
- Department of Global Innovative Drugs, Graduate School of Chung-Ang University, 84 Heukseok-ro, Dongjak-gu, Seoul, 06974, Republic of Korea
| | - Ha Hyung Kim
- Department of Global Innovative Drugs, Graduate School of Chung-Ang University, 84 Heukseok-ro, Dongjak-gu, Seoul, 06974, Republic of Korea; Department of Pharmaceutical Regulatory Sciences, Graduate School of Chung-Ang University, 84 Heukseok-ro, Dongjak-gu, Seoul, 06974, Republic of Korea.
| |
Collapse
|
30
|
Witecka A, Kazak V, Kwiatkowski S, Kiersztan A, Jagielski AK, Kozminski W, Augustyniak R, Drozak J. Hydroxysteroid 17-β dehydrogenase 14 (HSD17B14) is an L-fucose dehydrogenase, the initial enzyme of the L-fucose degradation pathway. J Biol Chem 2024; 300:107501. [PMID: 38944119 PMCID: PMC11293516 DOI: 10.1016/j.jbc.2024.107501] [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: 03/29/2024] [Revised: 06/03/2024] [Accepted: 06/15/2024] [Indexed: 07/01/2024] Open
Abstract
L-Fucose (6-deoxy-L-galactose), a monosaccharide abundant in glycolipids and glycoproteins produced by mammalian cells, has been extensively studied for its role in intracellular biosynthesis and recycling of GDP-L-fucose for fucosylation. However, in certain mammalian species, L-fucose is efficiently broken down to pyruvate and lactate in a poorly understood metabolic pathway. In the 1970s, L-fucose dehydrogenase, an enzyme responsible for the initial step of this pathway, was partially purified from pig and rabbit livers and characterized biochemically. However, its molecular identity remained elusive until recently. This study reports the purification, identification, and biochemical characterization of the mammalian L-fucose dehydrogenase. The enzyme was purified from rabbit liver approximately 340-fold. Mass spectrometry analysis of the purified protein preparation identified mammalian hydroxysteroid 17-β dehydrogenase 14 (HSD17B14) as the sole candidate enzyme. Rabbit and human HSD17B14 were expressed in HEK293T and Escherichia coli, respectively, purified, and demonstrated to catalyze the oxidation of L-fucose to L-fucono-1,5-lactone, as confirmed by mass spectrometry and NMR analysis. Substrate specificity studies revealed that L-fucose is the preferred substrate for both enzymes. The human enzyme exhibited a catalytic efficiency for L-fucose that was 359-fold higher than its efficiency for estradiol. Additionally, recombinant rat HSD17B14 exhibited negligible activity towards L-fucose, consistent with the absence of L-fucose metabolism in this species. The identification of the gene-encoding mammalian L-fucose dehydrogenase provides novel insights into the substrate specificity of enzymes belonging to the 17-β-hydroxysteroid dehydrogenase family. This discovery also paves the way for unraveling the physiological functions of the L-fucose degradation pathway, which remains enigmatic.
Collapse
Affiliation(s)
- Apolonia Witecka
- Department of Metabolic Regulation, Institute of Biochemistry, Faculty of Biology, University of Warsaw, Warsaw, Poland
| | - Varvara Kazak
- Department of Metabolic Regulation, Institute of Biochemistry, Faculty of Biology, University of Warsaw, Warsaw, Poland
| | - Sebastian Kwiatkowski
- Department of Metabolic Regulation, Institute of Biochemistry, Faculty of Biology, University of Warsaw, Warsaw, Poland; Biotechnology Division, Research & Development Centre, Celon Pharma S.A., Kazun Nowy, Poland
| | - Anna Kiersztan
- Department of Metabolic Regulation, Institute of Biochemistry, Faculty of Biology, University of Warsaw, Warsaw, Poland
| | - Adam K Jagielski
- Department of Metabolic Regulation, Institute of Biochemistry, Faculty of Biology, University of Warsaw, Warsaw, Poland
| | - Wiktor Kozminski
- Biological and Chemical Research Centre, Faculty of Chemistry, University of Warsaw, Warsaw, Poland
| | - Rafal Augustyniak
- Biological and Chemical Research Centre, Faculty of Chemistry, University of Warsaw, Warsaw, Poland.
| | - Jakub Drozak
- Department of Metabolic Regulation, Institute of Biochemistry, Faculty of Biology, University of Warsaw, Warsaw, Poland.
| |
Collapse
|
31
|
Skurska E, Szulc B, Kreczko K, Olczak M. Mutations in the SLC35C1 gene, contributing to significant differences in fucosylation patterns, may underlie the diverse phenotypic manifestations observed in leukocyte adhesion deficiency type II patients. Int J Biochem Cell Biol 2024; 173:106602. [PMID: 38843991 DOI: 10.1016/j.biocel.2024.106602] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2024] [Revised: 05/31/2024] [Accepted: 06/03/2024] [Indexed: 06/10/2024]
Abstract
Congenital disorders of glycosylation (CDG) are a large family of genetic diseases resulting from defects in the synthesis of glycans and the attachment of glycans to macromolecules. The CDG known as leukocyte adhesion deficiency II (LAD II) is an autosomal, recessive disorder caused by mutations in the SLC35C1 gene, encoding a transmembrane protein of the Golgi apparatus, involved in GDP-fucose transport from the cytosol to the Golgi lumen. In this study, a cell-based model was used as a tool to characterize the molecular background of a therapy based on a fucose-supplemented diet. Such therapies have been successfully introduced in some (but not all) known cases of LAD II. In this study, the effect of external fucose was analyzed in SLC35C1 KO cell lines, expressing 11 mutated SLC35C1 proteins, previously discovered in patients with an LAD II diagnosis. For many of them, the cis-Golgi subcellular localization was affected; however, some proteins were localized properly. Additionally, although mutated SLC35C1 caused different α-1-6 core fucosylation of N-glycans, which explains previously described, more or less severe disorder symptoms, the differences practically disappeared after external fucose supplementation, with fucosylation restored to the level observed in healthy cells. This indicates that additional fucose in the diet should improve the condition of all patients. Thus, for patients diagnosed with LAD II we advocate careful analysis of particular mutations using the SLC35C1-KO cell line-based model, to predict changes in localization and fucosylation rate. We also recommend searching for additional mutations in the human genome of LAD II patients, when fucose supplementation does not influence patients' state.
Collapse
Affiliation(s)
- E Skurska
- Laboratory of Biochemistry, Faculty of Biotechnology, University of Wrocław, Wrocław, Poland
| | - B Szulc
- Laboratory of Biochemistry, Faculty of Biotechnology, University of Wrocław, Wrocław, Poland
| | - K Kreczko
- Laboratory of Biochemistry, Faculty of Biotechnology, University of Wrocław, Wrocław, Poland
| | - M Olczak
- Laboratory of Biochemistry, Faculty of Biotechnology, University of Wrocław, Wrocław, Poland.
| |
Collapse
|
32
|
Sowa K, Okuda-Shimazaki J, Fukawa E, Sode K. Direct Electron Transfer-Type Oxidoreductases for Biomedical Applications. Annu Rev Biomed Eng 2024; 26:357-382. [PMID: 38424090 DOI: 10.1146/annurev-bioeng-110222-101926] [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] [Indexed: 03/02/2024]
Abstract
Among the various types of enzyme-based biosensors, sensors utilizing enzymes capable of direct electron transfer (DET) are recognized as the most ideal. However, only a limited number of redox enzymes are capable of DET with electrodes, that is, dehydrogenases harboring a subunit or domain that functions specifically to accept electrons from the redox cofactor of the catalytic site and transfer the electrons to the external electron acceptor. Such subunits or domains act as built-in mediators for electron transfer between enzymes and electrodes; consequently, such enzymes enable direct electron transfer to electrodes and are designated as DET-type enzymes. DET-type enzymes fall into several categories, including redox cofactors of catalytic reactions, built-in mediators for DET with electrodes and by their protein hierarchic structures, DET-type oxidoreductases with oligomeric structures harboring electron transfer subunits, and monomeric DET-type oxidoreductases harboring electron transfer domains. In this review, we cover the science of DET-type oxidoreductases and their biomedical applications. First, we introduce the structural biology and current understanding of DET-type enzyme reactions. Next, we describe recent technological developments based on DET-type enzymes for biomedical applications, such as biosensors and biochemical energy harvesting for self-powered medical devices. Finally, after discussing how to further engineer and create DET-type enzymes, we address the future prospects for DET-type enzymes in biomedical engineering.
Collapse
Affiliation(s)
- Keisei Sowa
- Division of Applied Life Sciences, Graduate School of Agriculture, Kyoto University, Sakyo, Kyoto, Japan
| | - Junko Okuda-Shimazaki
- Department of Biotechnology and Life Science, Graduate School of Engineering, Tokyo University of Agriculture and Technology, Kogane, Tokyo, Japan
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University, Chapel Hill, North Carolina, USA;
| | - Eole Fukawa
- Division of Applied Life Sciences, Graduate School of Agriculture, Kyoto University, Sakyo, Kyoto, Japan
| | - Koji Sode
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University, Chapel Hill, North Carolina, USA;
| |
Collapse
|
33
|
Liang Y, Liu Z, Zuo D, Chen S, Chen J, Yan X, Liu P, Wang Q. Single cell glycan-linkages profiling for hepatocellular carcinoma early diagnosis using lanthanide encoded bacteriophage MS2 based ICP-MS. Talanta 2024; 274:126056. [PMID: 38599123 DOI: 10.1016/j.talanta.2024.126056] [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: 01/11/2024] [Revised: 04/01/2024] [Accepted: 04/04/2024] [Indexed: 04/12/2024]
Abstract
Early diagnosis is paramount for enhancing survival rates and prognosis in the context of malignant diseases. Hepatocellular carcinoma (HCC), the second leading cause of cancer-related deaths worldwide, poses significant challenges for its early detection. In this study, we present an innovative approach which contributed to the early diagnosis of HCC. By lanthanide encoding signal amplification to map glycan-linkages at the single-cell level, the minute quantities of "soft" glycan-linkages on single cell surface were converted into "hard" elemental tags through the use of an MS2 signal amplifier. Harnessing the power of lanthanides encoded within MS2, we achieve nearly three orders of magnitude signal amplification. These encoded tags are subsequently quantified using single-cell inductively coupled plasma mass spectrometry (SC-ICP-MS). Linear discriminant analysis (LDA) identifies seven specific glycan-linkages (α-2,3-Sia, α-Gal, α-1,2-Fuc, α-1,6-Fuc, α-2,6-Sia, α-GalNAc, and Gal-β-1,3-GalNAc) as biomarkers. Our methodology is initially validated at the cellular level with 100% accuracy in discriminating between hepatic carcinoma HepG2 cells and their normal HL7702 cells. We apply this approach to quantify and classify glycan-linkages on the surfaces of 55 clinical surgical HCC specimens. Leveraging these seven glycan-linkages as biomarkers, we achieve precise differentiation between 8 normal hepatic specimens, 40 early HCC specimens, and 7 colorectal metastasis HCC specimens. This pioneering work represents the first instance of employing single-cell glycan-linkages as biomarkers promising for the early diagnosis of HCC with a remarkable 100% predictive accuracy rate, which holds immense potential for enhancing the feasibility and precision of HCC diagnosis in clinical practice.
Collapse
Affiliation(s)
- Yong Liang
- Department of Chemistry & the MOE Key Laboratory of Spectrochemical Analysis and Instrumentation, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen, 361005, China; Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, 510405, China.
| | - Zhen Liu
- Department of Chemistry & the MOE Key Laboratory of Spectrochemical Analysis and Instrumentation, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen, 361005, China
| | - Dongliang Zuo
- Department of Hepatobiliary Surgery, Zhongshan Hospital, Xiamen University, Fujian Provincial Key Laboratory for Chronic Liver Disease and Hepatocellular Carcinoma, Xiamen, 361004, China; The Central Hospital of Enshi Tujia and Miao Autonomous Prefecture, Enshi, 445000, China
| | - Shi Chen
- Department of Chemistry & the MOE Key Laboratory of Spectrochemical Analysis and Instrumentation, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen, 361005, China
| | - Jianbin Chen
- Department of Chemistry & the MOE Key Laboratory of Spectrochemical Analysis and Instrumentation, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen, 361005, China
| | - Xiaowen Yan
- Department of Chemistry & the MOE Key Laboratory of Spectrochemical Analysis and Instrumentation, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen, 361005, China
| | - Pingguo Liu
- Department of Hepatobiliary Surgery, Zhongshan Hospital, Xiamen University, Fujian Provincial Key Laboratory for Chronic Liver Disease and Hepatocellular Carcinoma, Xiamen, 361004, China.
| | - Qiuquan Wang
- Department of Chemistry & the MOE Key Laboratory of Spectrochemical Analysis and Instrumentation, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen, 361005, China.
| |
Collapse
|
34
|
He S, Luo Y, Ma W, Wang X, Yan C, Hao W, Fang Y, Su H, Lai B, Liu J, Xiong Y, Bai T, Ren X, Liu E, Han H, Wu Y, Yuan Z, Wang Y. Endothelial POFUT1 controls injury-induced liver fibrosis by repressing fibrinogen synthesis. J Hepatol 2024; 81:135-148. [PMID: 38460791 DOI: 10.1016/j.jhep.2024.02.032] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/20/2023] [Revised: 02/19/2024] [Accepted: 02/27/2024] [Indexed: 03/11/2024]
Abstract
BACKGROUND & AIMS NOTCH signaling in liver sinusoidal endothelial cells (LSECs) regulates liver fibrosis, a pathological feature of chronic liver diseases. POFUT1 is an essential regulator of NOTCH signaling. Here, we investigated the role of LSEC-expressed POFUT1 in liver fibrosis. METHODS Endothelial-specific Pofut1 knockout mice were generated and experimental liver fibrosis was induced by chronic carbon tetrachloride exposure or common bile duct ligation. Liver samples were assessed by ELISA, histology, electron microscopy, immunostaining and RNA in situ hybridization. LSECs and hepatic stellate cells (HSCs) were isolated for gene expression analysis by RNA sequencing, qPCR, and western blotting. Signaling crosstalk between LSECs and HSCs was investigated by treating HSCs with supernatant from LSEC cultures. Liver single-cell RNA sequencing datasets from patients with cirrhosis and healthy individuals were analyzed to evaluate the clinical relevance of gene expression changes observed in mouse studies. RESULTS POFUT1 loss promoted injury-induced LSEC capillarization and HSC activation, leading to aggravated liver fibrosis. RNA sequencing analysis revealed that POFUT1 deficiency upregulated fibrinogen expression in LSECs. Consistently, fibrinogen was elevated in LSECs of patients with cirrhosis. HSCs treated with supernatant from LSECs of Pofut1 null mice showed exacerbated activation compared to those treated with supernatant from control LSECs, and this effect was attenuated by knockdown of fibrinogen or by pharmacological inhibition of fibrinogen receptor signaling, altogether suggesting that LSEC-derived fibrinogen induced the activation of HSCs. Mechanistically, POFUT1 loss augmented fibrinogen expression by enhancing NOTCH/HES1/STAT3 signaling. CONCLUSIONS Endothelial POFUT1 prevents injury-induced liver fibrosis by repressing the expression of fibrinogen, which functions as a profibrotic paracrine signal to activate HSCs. Therapies targeting the POFUT1/fibrinogen axis offer a promising strategy for the prevention and treatment of fibrotic liver diseases. IMPACT AND IMPLICATIONS Paracrine signals produced by liver vasculature play a major role in the development of liver fibrosis, which is a pathological hallmark of most liver diseases. Identifying those paracrine signals is clinically relevant in that they may serve as therapeutic targets. In this study, we discovered that genetic deletion of Pofut1 aggravated experimental liver fibrosis in mouse models. Moreover, fibrinogen was identified as a downstream target repressed by Pofut1 in liver endothelial cells and functioned as a novel paracrine signal that drove liver fibrosis. In addition, fibrinogen was found to be relevant to cirrhosis and may serve as a potential therapeutic target for this devastating human disease.
Collapse
Affiliation(s)
- Shan He
- The Institute of Cardiovascular Sciences, School of Basic Medical Sciences, Xi'an Jiaotong University, Xi'an, Shaanxi, China; Department of Stomatology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Yuru Luo
- The Institute of Cardiovascular Sciences, School of Basic Medical Sciences, Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Wangge Ma
- Cardiovascular Department, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Xiaoke Wang
- Cardiovascular Department, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Chengrong Yan
- The Institute of Cardiovascular Sciences, School of Basic Medical Sciences, Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Wenyang Hao
- The Institute of Cardiovascular Sciences, School of Basic Medical Sciences, Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Yuan Fang
- The Institute of Cardiovascular Sciences, School of Basic Medical Sciences, Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Hongyu Su
- The Institute of Cardiovascular Sciences, School of Basic Medical Sciences, Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Baochang Lai
- The Institute of Cardiovascular Sciences, School of Basic Medical Sciences, Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Junhui Liu
- Clinical Laboratory, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Ying Xiong
- Cardiovascular Department, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Ting Bai
- Cardiovascular Department, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Xiaoyong Ren
- Department of Stomatology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Enqi Liu
- Department of Laboratory Animal Science, School of Basic Medical Sciences, Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Hua Han
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancer and Department of Biochemistry and Molecular Biology, Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Yue Wu
- Cardiovascular Department, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China; Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education, Xi'an Jiaotong University, Xi'an, Shaanxi, China; Cardiometabolic Innovation Center, Ministry of Education, Xi'an Jiaotong University, Xi'an, Shaanxi, China.
| | - Zuyi Yuan
- Cardiovascular Department, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China; Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education, Xi'an Jiaotong University, Xi'an, Shaanxi, China; Cardiometabolic Innovation Center, Ministry of Education, Xi'an Jiaotong University, Xi'an, Shaanxi, China.
| | - Yidong Wang
- The Institute of Cardiovascular Sciences, School of Basic Medical Sciences, Xi'an Jiaotong University, Xi'an, Shaanxi, China; Cardiovascular Department, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China; Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education, Xi'an Jiaotong University, Xi'an, Shaanxi, China; Cardiometabolic Innovation Center, Ministry of Education, Xi'an Jiaotong University, Xi'an, Shaanxi, China; Department of Cardiology, Wenling First People's Hospital, The Affiliated Hospital of Wenzhou Medical University, Wenling, Zhejiang, China.
| |
Collapse
|
35
|
Tokoro Y, Nagae M, Nakano M, Harduin-Lepers A, Kizuka Y. LacdiNAc synthase B4GALNT3 has a unique PA14 domain and suppresses N-glycan capping. J Biol Chem 2024; 300:107450. [PMID: 38844136 PMCID: PMC11254600 DOI: 10.1016/j.jbc.2024.107450] [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: 04/05/2024] [Revised: 05/17/2024] [Accepted: 05/30/2024] [Indexed: 06/28/2024] Open
Abstract
Structural variation of N-glycans is essential for the regulation of glycoprotein functions. GalNAcβ1-4GlcNAc (LacdiNAc or LDN), a unique subterminal glycan structure synthesized by B4GALNT3 or B4GALNT4, is involved in the clearance of N-glycoproteins from the blood and maintenance of cell stemness. Such regulation of glycoprotein functions by LDN is largely different from that by the dominant subterminal structure, N-acetyllactosamine (Galβ1-4GlcNAc, LacNAc). However, the mechanisms by which B4GALNT activity is regulated and how LDN plays different roles from LacNAc remain unclear. Here, we found that B4GALNT3 and four have unique domain organization containing a noncatalytic PA14 domain, which is a putative glycan-binding module. A mutant lacking this domain dramatically decreases the activity toward various substrates, such as N-glycan, O-GalNAc glycan, and glycoproteins, indicating that this domain is essential for enzyme activity and forms part of the catalytic region. In addition, to clarify the mechanism underlying the functional differences between LDN and LacNAc, we examined the effects of LDN on the maturation of N-glycans, focusing on the related glycosyltransferases upstream and downstream of B4GALNT. We revealed that, unlike LacNAc synthesis, prior formation of bisecting GlcNAc in N-glycan almost completely inhibits LDN synthesis by B4GALNT3. Moreover, the presence of LDN negatively impacted the actions of many glycosyltransferases for terminal modifications, including sialylation, fucosylation, and human natural killer-1 synthesis. These findings demonstrate that LDN has significant impacts on N-glycan maturation in a completely different way from LacNAc, which could contribute to obtaining a comprehensive overview of the system regulating complex N-glycan biosynthesis.
Collapse
Affiliation(s)
- Yuko Tokoro
- Institute for Glyco-core Research (iGCORE), Gifu University, Gifu, Japan
| | - Masamichi Nagae
- Department of Molecular Immunology, Research Institute for Microbial Diseases, Osaka University, Suita, Japan; Laboratory of Molecular Immunology, Immunology Frontier Research Center (IFReC), Osaka University, Suita, Japan
| | - Miyako Nakano
- Graduate School of Integrated Sciences for Life, Hiroshima University, Higashihiroshima, Japan
| | - Anne Harduin-Lepers
- University of Lille, CNRS, UMR 8576 -UGSF- Unité de Glycobiologie Structurale et Fonctionnelle, Lille, France
| | - Yasuhiko Kizuka
- Institute for Glyco-core Research (iGCORE), Gifu University, Gifu, Japan.
| |
Collapse
|
36
|
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
|
37
|
Zhang L, Feng Y, Zhang Y, Sun X, Ma Q, Ma F. The Sweet Relationship between the Endometrium and Protein Glycosylation. Biomolecules 2024; 14:770. [PMID: 39062484 PMCID: PMC11274983 DOI: 10.3390/biom14070770] [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: 05/27/2024] [Revised: 06/17/2024] [Accepted: 06/25/2024] [Indexed: 07/28/2024] Open
Abstract
The endometrium is an important part of women's bodies for menstruation and pregnancy. Various proteins are widely expressed on the surface of endometrial cells, and glycosylation is an important post-translational modification of proteins. Glycosylation modification is closely related not only to endometrial receptivity but also to common diseases related to endometrial receptivity. Glycosylation can improve endometrial receptivity, promote embryo localization and trophoblast cell adhesion and invasion, and contribute to successful implantation. Two diseases related to endometrial receptivity include endometriosis and endometrial cancer. As a common benign disease in women, endometriosis is often accompanied by an increased menstrual volume, prolonged menstrual periods, progressive and aggravated dysmenorrhea, and may be accompanied by infertility. Protein glycosylation modification of the endometrial surface indicates the severity of the disease and may be an important pathogenesis of endometriosis. In cancer, glycosylation modifications on the surface of tumor cells can be a marker to distinguish the type and severity of endometrial cancer. This review highlights the role of protein glycosylation in embryo-maternal endometrial dialogue and explores its potential mechanisms in diseases related to endometrial receptivity, which could provide a new clinical approach for their diagnosis and treatment.
Collapse
Affiliation(s)
- Linyu Zhang
- Center for Translational Medicine, Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu 610041, China
- Department of Obstetrics and Gynecology, West China Second Hospital, Sichuan University, Chengdu 610041, China
| | - Ying Feng
- West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu 610041, China
| | - Yue Zhang
- Center for Translational Medicine, Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu 610041, China
- Department of Obstetrics and Gynecology, West China Second Hospital, Sichuan University, Chengdu 610041, China
| | - Xinrui Sun
- Center for Translational Medicine, Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu 610041, China
- Department of Obstetrics and Gynecology, West China Second Hospital, Sichuan University, Chengdu 610041, China
| | - Qianhong Ma
- Department of Obstetrics and Gynecology, West China Second Hospital, Sichuan University, Chengdu 610041, China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu 610041, China
| | - Fang Ma
- Center for Translational Medicine, Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu 610041, China
- Department of Obstetrics and Gynecology, West China Second Hospital, Sichuan University, Chengdu 610041, China
| |
Collapse
|
38
|
Li Q, Guo W, Qian Y, Li S, Li L, Zhu Z, Wang F, Tong Y, Xia Q, Liu Y. Protein O-fucosyltransferase 1 promotes PD-L1 stability to drive immune evasion and directs liver cancer to immunotherapy. J Immunother Cancer 2024; 12:e008917. [PMID: 38908854 PMCID: PMC11328658 DOI: 10.1136/jitc-2024-008917] [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] [Accepted: 05/30/2024] [Indexed: 06/24/2024] Open
Abstract
BACKGROUND AND AIMS The immunosuppressive tumor microenvironment (TME) plays an essential role in cancer progression and immunotherapy response. Despite the considerable advancements in cancer immunotherapy, the limited response to immune checkpoint blockade (ICB) therapies in patients with hepatocellular carcinoma (HCC) remains a major challenge for its clinical implications. Here, we investigated the molecular basis of the protein O-fucosyltransferase 1 (POFUT1) that drives HCC immune evasion and explored a potential therapeutic strategy for enhancing ICB efficacy. METHODS De novo MYC/Trp53-/- liver tumor and the xenograft tumor models were used to evaluate the function of POFUT1 in immune evasion. Biochemical assays were performed to elucidate the underlying mechanism of POFUT1-mediated immune evasion. RESULTS We identified POFUT1 as a crucial promoter of immune evasion in liver cancer. Notably, POFUT1 promoted HCC progression and inhibited T-cell infiltration in the xenograft tumor and de novo MYC/Trp53-/- mouse liver tumor models. Mechanistically, we demonstrated that POFUT1 stabilized programmed death ligand 1 (PD-L1) protein by preventing tripartite motif containing 21-mediated PD-L1 ubiquitination and degradation independently of its protein-O-fucosyltransferase activity. In addition, we further demonstrated that PD-L1 was required for the tumor-promoting and immune evasion effects of POFUT1 in HCC. Importantly, inhibition of POFUT1 could synergize with anti-programmed death receptor 1 therapy by remodeling TME in the xenograft tumor mouse model. Clinically, POFUT1 high expression displayed a lower response rate and worse clinical outcome to ICB therapies. CONCLUSIONS Our findings demonstrate that POFUT1 functions as a novel regulator of tumor immune evasion and inhibition of POFUT1 may be a potential therapeutic strategy to enhance the efficacy of immune therapy in HCC.
Collapse
Affiliation(s)
- Qianyu Li
- Department of Liver Surgery, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Renji-Med-X Clinical Stem Cell Research Center, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Wenyun Guo
- Department of Liver Surgery, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Renji-Med-X Clinical Stem Cell Research Center, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yifei Qian
- Department of Liver Surgery, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Renji-Med-X Clinical Stem Cell Research Center, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Songling Li
- School of Biomedical Engineering & Med-X Research Institute, Shanghai Jiao Tong University, Shanghai, China
| | - Linfeng Li
- Renji-Med-X Clinical Stem Cell Research Center, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zijun Zhu
- Department of Liver Surgery, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Renji-Med-X Clinical Stem Cell Research Center, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Fan Wang
- Renji-Med-X Clinical Stem Cell Research Center, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yu Tong
- Renji-Med-X Clinical Stem Cell Research Center, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Qiang Xia
- Department of Liver Surgery, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Engineering Research Center of Transplantation and Immunology, Shanghai Institute of Transplantation, Shanghai, China
| | - Yanfeng Liu
- Department of Liver Surgery, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Engineering Research Center of Transplantation and Immunology, Shanghai Institute of Transplantation, Shanghai, China
| |
Collapse
|
39
|
He X, Zhao L, Tian Y, Li R, Chu Q, Gu Z, Zheng M, Wang Y, Li S, Jiang H, Jiang Y, Wen L, Wang D, Cheng X. Highly accurate carbohydrate-binding site prediction with DeepGlycanSite. Nat Commun 2024; 15:5163. [PMID: 38886381 PMCID: PMC11183243 DOI: 10.1038/s41467-024-49516-2] [Citation(s) in RCA: 14] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2023] [Accepted: 06/10/2024] [Indexed: 06/20/2024] Open
Abstract
As the most abundant organic substances in nature, carbohydrates are essential for life. Understanding how carbohydrates regulate proteins in the physiological and pathological processes presents opportunities to address crucial biological problems and develop new therapeutics. However, the diversity and complexity of carbohydrates pose a challenge in experimentally identifying the sites where carbohydrates bind to and act on proteins. Here, we introduce a deep learning model, DeepGlycanSite, capable of accurately predicting carbohydrate-binding sites on a given protein structure. Incorporating geometric and evolutionary features of proteins into a deep equivariant graph neural network with the transformer architecture, DeepGlycanSite remarkably outperforms previous state-of-the-art methods and effectively predicts binding sites for diverse carbohydrates. Integrating with a mutagenesis study, DeepGlycanSite reveals the guanosine-5'-diphosphate-sugar-recognition site of an important G-protein coupled receptor. These findings demonstrate DeepGlycanSite is invaluable for carbohydrate-binding site prediction and could provide insights into molecular mechanisms underlying carbohydrate-regulation of therapeutically important proteins.
Collapse
Affiliation(s)
- Xinheng He
- State Key Laboratory of Drug Research and State Key Laboratory of Chemical Biology, Carbohydrate-Based Drug Research Center, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Lifen Zhao
- State Key Laboratory of Drug Research and State Key Laboratory of Chemical Biology, Carbohydrate-Based Drug Research Center, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
| | - Yinping Tian
- State Key Laboratory of Drug Research and State Key Laboratory of Chemical Biology, Carbohydrate-Based Drug Research Center, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
| | - Rui Li
- State Key Laboratory of Drug Research and State Key Laboratory of Chemical Biology, Carbohydrate-Based Drug Research Center, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
- School of Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Qinyu Chu
- School of Pharmaceutical Science and Technology, Hangzhou Institute of Advanced Study, Hangzhou, China
| | - Zhiyong Gu
- School of Pharmaceutical Science and Technology, Hangzhou Institute of Advanced Study, Hangzhou, China
| | - Mingyue Zheng
- State Key Laboratory of Drug Research and State Key Laboratory of Chemical Biology, Carbohydrate-Based Drug Research Center, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
- University of Chinese Academy of Sciences, Beijing, China
- School of Pharmaceutical Science and Technology, Hangzhou Institute of Advanced Study, Hangzhou, China
| | - Yusong Wang
- National Key Laboratory of Human-Machine Hybrid Augmented Intelligence, National Engineering Research Center for Visual Information and Applications, and Institute of Artificial Intelligence and Robotics, Xi'an Jiaotong University, Xi'an, China
| | - Shaoning Li
- Department of Computer Science and Engineering, The Chinese University of Hong Kong, Hong Kong, China
| | - Hualiang Jiang
- State Key Laboratory of Drug Research and State Key Laboratory of Chemical Biology, Carbohydrate-Based Drug Research Center, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
- University of Chinese Academy of Sciences, Beijing, China
- School of Pharmaceutical Science and Technology, Hangzhou Institute of Advanced Study, Hangzhou, China
- Lingang Laboratory, Shanghai, China
| | - Yi Jiang
- Lingang Laboratory, Shanghai, China
| | - Liuqing Wen
- State Key Laboratory of Drug Research and State Key Laboratory of Chemical Biology, Carbohydrate-Based Drug Research Center, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China.
- University of Chinese Academy of Sciences, Beijing, China.
| | | | - Xi Cheng
- State Key Laboratory of Drug Research and State Key Laboratory of Chemical Biology, Carbohydrate-Based Drug Research Center, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China.
- University of Chinese Academy of Sciences, Beijing, China.
- School of Pharmaceutical Science and Technology, Hangzhou Institute of Advanced Study, Hangzhou, China.
| |
Collapse
|
40
|
Yang X, Zhang J, Zhu J, Yang R, Tong Y. Molecular insights into FucR transcription factor to control the metabolism of L-fucose in Bifidobacterium longum subsp. infantis. Microbiol Res 2024; 283:127709. [PMID: 38593579 DOI: 10.1016/j.micres.2024.127709] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2023] [Revised: 03/27/2024] [Accepted: 03/30/2024] [Indexed: 04/11/2024]
Abstract
Bifidobacterium longum subsp. infantis commonly colonizes the human gut and is capable of metabolizing L-fucose, which is abundant in the gut. Multiple studies have focused on the mechanisms of L-fucose utilization by B. longum subsp. infantis, but the regulatory pathways governing the expression of these catabolic processes are still unclear. In this study, we have conducted a structural and functional analysis of L-fucose metabolism transcription factor FucR derived from B. longum subsp. infantis Bi-26. Our results indicated that FucR is a L-fucose-sensitive repressor with more α-helices, fewer β-sheets, and β-turns. Transcriptional analysis revealed that FucR displays weak negative self-regulation, which is counteracted in the presence of L-fucose. Isothermal titration calorimetry indicated that FucR has a 2:1 stoichiometry with L-fucose. The key amino acid residues for FucR binding L-fucose are Asp280 and Arg331, with mutation of Asp280 to Ala resulting in a decrease in the affinity between FucR and L-fucose with the Kd value from 2.58 to 11.68 μM, and mutation of Arg331 to Ala abolishes the binding ability of FucR towards L-fucose. FucR specifically recognized and bound to a 20-bp incomplete palindrome sequence (5'-ACCCCAATTACGAAAATTTTT-3'), and the affinity of the L-fucose-loaded FucR for the DNA fragment was lower than apo-FucR. The results provided new insights into the regulating L-fucose metabolism by B. longum subsp. infantis.
Collapse
Affiliation(s)
- Xiaojun Yang
- School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu 214122, China
| | - Jing Zhang
- School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu 214122, China
| | - Jing Zhu
- School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu 214122, China
| | - Ruijin Yang
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi, Jiangsu 214122, China; School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu 214122, China
| | - Yanjun Tong
- School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu 214122, China.
| |
Collapse
|
41
|
Nguan HS, Chen JL, Ni CK. Collision-Induced Dissociation of Fucose and Identification of Anomericity. J Phys Chem A 2024; 128:3812-3820. [PMID: 38690855 PMCID: PMC11103703 DOI: 10.1021/acs.jpca.4c00640] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2024] [Revised: 04/15/2024] [Accepted: 04/24/2024] [Indexed: 05/03/2024]
Abstract
Structural determination of carbohydrates using mass spectrometry remains challenging, particularly, the differentiation of anomeric configurations. In this work, we studied the collision-induced dissociation (CID) mechanisms of sodiated α- and β-l-fucose using an experimental method and quantum chemistry calculations. The calculations show that α-l-fucose is more likely to undergo dehydration due to the fact that O1 and O2 are on the same side of the sugar ring. In contrast, β-l-fucose is more prone to the ring-opening reaction because more OH groups are on the same side of the sugar ring as O1. These differences suggest a higher preference for the dehydration reaction in sodiated α-l-fucose but a lower preference for ring-opening compared to that of β-l-fucose. The calculation results, which are used to assign the CID mass spectra of α- and β-l-fucose separated by high-performance liquid chromatography, are supported by the fucose produced from the CID of disaccharides Fuc-β-(1 → 3)-GlcNAc and Fuc-α-(1 → 4)-GlcNAc. This study demonstrates that the correlation of cis- and trans-configurations of O1 and O2 to the relative branching ratios of dehydration and cross-ring dissociation in CID, observed in aldohexose and ketohexose in the pyranose form, can be extended to deoxyhexoses for anomericity determination.
Collapse
Affiliation(s)
- Hock-Seng Nguan
- Institute
of Atomic and Molecular Sciences, Academia
Sinica, P.O. Box 23-166, Taipei 10617, Taiwan
| | - Jien-Lian Chen
- Institute
of Atomic and Molecular Sciences, Academia
Sinica, P.O. Box 23-166, Taipei 10617, Taiwan
| | - Chi-Kung Ni
- Institute
of Atomic and Molecular Sciences, Academia
Sinica, P.O. Box 23-166, Taipei 10617, Taiwan
- Department
of Chemistry, National Tsing Hua University, Hsinchu 30013, Taiwan
| |
Collapse
|
42
|
Rokhsartalab Azar P, Maleki Aghdam M, Karimi S, Haghtalab A, Sadeghpour S, Mellatyar H, Taheri-Anganeh M, Ghasemnejad-Berenji H. Uterine fluid microRNAs in repeated implantation failure. Clin Chim Acta 2024; 558:119678. [PMID: 38641194 DOI: 10.1016/j.cca.2024.119678] [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: 01/25/2024] [Revised: 04/13/2024] [Accepted: 04/15/2024] [Indexed: 04/21/2024]
Abstract
Recurrent implantation failure (RIF) is a significant obstacle in assisted reproductive procedures, primarily because of compromised receptivity. As such, there is a need for a dependable and accurate clinical test to evaluate endometrial receptiveness, particularly during embryo transfer. MicroRNAs (miRNAs) have diverse functions in the processes of implantation and pregnancy. Dysregulation of miRNAs results in reproductive diseases such as recurrent implantation failure (RIF). The endometrium secretes several microRNAs (miRNAs) during the implantation period, which could potentially indicate whether the endometrium is suitable for in vitro fertilization (IVF). The goal of this review is to examine endometrial miRNAs as noninvasive biomarkers that successfully predict endometrium receptivity in RIF.
Collapse
Affiliation(s)
| | - Mahdi Maleki Aghdam
- Student Research Committee, Urmia University of Medical Sciences, Urmia, Iran
| | - Sarmad Karimi
- Student Research Committee, Urmia University of Medical Sciences, Urmia, Iran
| | - Arian Haghtalab
- School of Medicine, Urmia University of Medical Sciences, Urmia, Iran
| | - Sonia Sadeghpour
- Department of Obstetrics and Gynecology, School of Medicine, Urmia University of Medical Sciences, Urmia, Iran; Reproductive Health Research Center, Clinical Research Institute, Urmia University of Medical Sciences, Urmia, Iran
| | | | - Mortaza Taheri-Anganeh
- Cellular and Molecular Research Center, Cellular and Molecular Medicine Research Institute, Urmia University of Medical Sciences, Urmia, Iran.
| | - Hojat Ghasemnejad-Berenji
- Reproductive Health Research Center, Clinical Research Institute, Urmia University of Medical Sciences, Urmia, Iran.
| |
Collapse
|
43
|
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
|
44
|
Okada T, Teramoto T, Ihara H, Ikeda Y, Kakuta Y. Crystal structure of mango α1,3/α1,4-fucosyltransferase elucidates unique elements that regulate Lewis A-dominant oligosaccharide assembly. Glycobiology 2024; 34:cwae015. [PMID: 38376259 DOI: 10.1093/glycob/cwae015] [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: 10/19/2023] [Revised: 01/28/2024] [Accepted: 02/14/2024] [Indexed: 02/21/2024] Open
Abstract
In various organisms, α1,3/α1,4-fucosyltransferases (CAZy GT10 family enzymes) mediate the assembly of type I (Galβ1,3GlcNAc) and/or type II (Galβ1,4GlcNAc)-based Lewis structures that are widely distributed in glycoconjugates. Unlike enzymes of other species, plant orthologues show little fucosyltransferase activity for type II-based glycans and predominantly catalyze the assembly of the Lewis A structure [Galβ1,3(Fucα1,4)GlcNAc] on the type I disaccharide unit of their substrates. However, the structural basis underlying this unique substrate selectivity remains elusive. In this study, we investigated the structure-function relationship of MiFUT13A, a mango α1,3/α1,4-fucosyltransferase. The prepared MiFUT13A displayed distinct α1,4-fucosyltransferase activity. Consistent with the enzymatic properties of this molecule, X-ray crystallography revealed that this enzyme has a typical GT-B fold-type structure containing a set of residues that are responsible for its SN2-like catalysis. Site-directed mutagenesis and molecular docking analyses proposed a rational binding mechanism for type I oligosaccharides. Within the catalytic cleft, the pocket surrounding Trp121 serves as a binding site, anchoring the non-reducing terminal β1,3-galactose that belongs to the type I disaccharide unit. Furthermore, Glu177 was postulated to function as a general base catalyst through its interaction with the 4-hydroxy group of the acceptor N-acetylglucosamine residue. Adjacent residues, specifically Thr120, Thr157 and Asp175 were speculated to assist in binding of the reducing terminal residues. Intriguingly, these structural elements were not fully conserved in mammalian orthologue which also shows predominant α1,4-fucosyltransferase activity. In conclusion, we have proposed that MiFUT13A generates the Lewis A structure on type I glycans through a distinct mechanism, divergent from that of mammalian enzymes.
Collapse
Affiliation(s)
- Takahiro Okada
- Division of Molecular Cell Biology, Department of Biomolecular Sciences, Faculty of Medicine, Saga University, 5-1-1 Nabeshima, Saga 849-8501, Japan
| | - Takamasa Teramoto
- Laboratory of Biophysical Chemistry, Department of Bioscience and Biotechnology, Faculty of Agriculture, Kyushu University, 744 Motooka, Nishi-ku, Fukuoka 819-0395, Japan
| | - Hideyuki Ihara
- Division of Molecular Cell Biology, Department of Biomolecular Sciences, Faculty of Medicine, Saga University, 5-1-1 Nabeshima, Saga 849-8501, Japan
| | - Yoshitaka Ikeda
- Division of Molecular Cell Biology, Department of Biomolecular Sciences, Faculty of Medicine, Saga University, 5-1-1 Nabeshima, Saga 849-8501, Japan
| | - Yoshimitsu Kakuta
- Laboratory of Biophysical Chemistry, Department of Bioscience and Biotechnology, Faculty of Agriculture, Kyushu University, 744 Motooka, Nishi-ku, Fukuoka 819-0395, Japan
| |
Collapse
|
45
|
Li J, Liu D, Zhang Y, Shen J, Dan W, Chen Z, Sun S. Site-Specific Analysis of Core and Antenna Fucosylation on Serum Glycoproteins. Anal Chem 2024; 96:5741-5745. [PMID: 38573003 DOI: 10.1021/acs.analchem.4c00445] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/05/2024]
Abstract
Fucosylation is an important structural feature of glycans and plays an essential role in the regulation of glycoprotein functions. Fucosylation can be classified into core- (CF) and antenna-fucosylation (AF, also known as (sialyl-) Lewis) based on the location on N-glycans, and they perform distinct biological functions. In this study, core- and antenna-fucosylated N-glycans on human serum glycoproteins that hold great clinical application values were systematically characterized at the site-specific level using StrucGP combined with the recently developed fucosylation assignment method. The results showed that fucosylation was widely distributed on serum glycoproteins, with 50% of fucosylated glycopeptides modified by AF N-glycans, 37% by CF N-glycans, and 13% by dual-fucosylated N-glycans. Interestingly, CF and AF N-glycans preferred to modify different groups of serum glycoproteins with different tissue origins and were involved in distinctive biological processes. Specifically, AF N-glycoproteins are mainly from the liver and participated in complement activation, blood coagulation, and endopeptidase activities, while CF N-glycoproteins originate from diverse tissues and are mainly involved in cell adhesion and signaling transduction. These data further enhanced our understanding of fucosylation on circulation glycoproteins.
Collapse
Affiliation(s)
- Jun Li
- College of Life Sciences, Northwest University, Xi'an 710069, China P.R
| | - Didi Liu
- College of Life Sciences, Northwest University, Xi'an 710069, China P.R
| | - Yingjie Zhang
- College of Life Sciences, Northwest University, Xi'an 710069, China P.R
| | - Jiechen Shen
- College of Life Sciences, Northwest University, Xi'an 710069, China P.R
| | - Wei Dan
- College of Life Sciences, Northwest University, Xi'an 710069, China P.R
| | - Zexuan Chen
- College of Life Sciences, Northwest University, Xi'an 710069, China P.R
| | - Shisheng Sun
- College of Life Sciences, Northwest University, Xi'an 710069, China P.R
| |
Collapse
|
46
|
Burton C, Bitaraf A, Snyder K, Zhang C, Yoder SJ, Avram D, Du D, Yu X, Lau EK. The functional role of L-fucose on dendritic cell function and polarization. Front Immunol 2024; 15:1353570. [PMID: 38646527 PMCID: PMC11026564 DOI: 10.3389/fimmu.2024.1353570] [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: 12/10/2023] [Accepted: 02/21/2024] [Indexed: 04/23/2024] Open
Abstract
Despite significant advances in the development and refinement of immunotherapies administered to combat cancer over the past decades, a number of barriers continue to limit their efficacy. One significant clinical barrier is the inability to mount initial immune responses towards the tumor. As dendritic cells are central initiators of immune responses in the body, the elucidation of mechanisms that can be therapeutically leveraged to enhance their functions to drive anti-tumor immune responses is urgently needed. Here, we report that the dietary sugar L-fucose can be used to enhance the immunostimulatory activity of dendritic cells (DCs). L-fucose polarizes immature myeloid cells towards specific DC subsets, specifically cDC1 and moDC subsets. In vitro, L-fucose treatment enhances antigen uptake and processing of DCs. Furthermore, our data suggests that L-fucose-treated DCs increase stimulation of T cell populations. Consistent with our functional assays, single-cell RNA sequencing of intratumoral DCs from melanoma- and breast tumor-bearing mice confirmed transcriptional regulation and antigen processing as pathways that are significantly altered by dietary L-fucose. Together, this study provides the first evidence of the ability of L-fucose to bolster DC functionality and provides rational to further investigate how L-fucose can be used to leverage DC function in order to enhance current immunotherapy.
Collapse
Affiliation(s)
- Chase Burton
- Department of Immunology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL, United States
- Cancer Biology Ph.D. Program, University of South Florida, Tampa, FL, United States
- Immunology Program, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL, United States
| | - Amirreza Bitaraf
- Cancer Biology Ph.D. Program, University of South Florida, Tampa, FL, United States
- Molecular Medicine Program, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL, United States
- Department of Tumor Microenvironment and Metastasis, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL, United States
| | - Kara Snyder
- Molecular Medicine Program, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL, United States
- Department of Tumor Microenvironment and Metastasis, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL, United States
- Department of Molecular Medicine, University of South Florida, Tampa, FL, United States
| | - Chaomei Zhang
- Molecular Genomics Core, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL, United States
| | - Sean J. Yoder
- Molecular Genomics Core, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL, United States
| | - Dorina Avram
- Department of Immunology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL, United States
- Immunology Program, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL, United States
| | - Dongliang Du
- Department of Biostatistics and Bioinformatics, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL, United States
| | - Xiaoqing Yu
- Department of Biostatistics and Bioinformatics, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL, United States
| | - Eric K. Lau
- Molecular Medicine Program, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL, United States
- Department of Tumor Microenvironment and Metastasis, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL, United States
| |
Collapse
|
47
|
Zhang Q, Ma C, Chin LS, Pan S, Li L. Human brain glycoform coregulation network and glycan modification alterations in Alzheimer's disease. SCIENCE ADVANCES 2024; 10:eadk6911. [PMID: 38579000 PMCID: PMC10997212 DOI: 10.1126/sciadv.adk6911] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/05/2023] [Accepted: 03/05/2024] [Indexed: 04/07/2024]
Abstract
Despite the importance of protein glycosylation to brain health, current knowledge of glycosylated proteoforms or glycoforms in human brain and their alterations in Alzheimer's disease (AD) is limited. Here, we report a proteome-wide glycoform profiling study of human AD and control brains using intact glycopeptide-based quantitative glycoproteomics coupled with systems biology. Our study identified more than 10,000 human brain N-glycoforms from nearly 1200 glycoproteins and uncovered disease signatures of altered glycoforms and glycan modifications, including reduced sialylation and N-glycan branching and elongation as well as elevated mannosylation and N-glycan truncation in AD. Network analyses revealed a higher-order organization of brain glycoproteome into networks of coregulated glycoforms and glycans and discovered glycoform and glycan modules associated with AD clinical phenotype, amyloid-β accumulation, and tau pathology. Our findings provide valuable insights into disease pathogenesis and a rich resource of glycoform and glycan changes in AD and pave the way forward for developing glycosylation-based therapies and biomarkers for AD.
Collapse
Affiliation(s)
- Qi Zhang
- Department of Pharmacology and Chemical Biology, Center for Neurodegenerative Disease, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Cheng Ma
- The Brown Foundation Institute of Molecular Medicine, University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Lih-Shen Chin
- Department of Pharmacology and Chemical Biology, Center for Neurodegenerative Disease, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Sheng Pan
- The Brown Foundation Institute of Molecular Medicine, University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Lian Li
- Department of Pharmacology and Chemical Biology, Center for Neurodegenerative Disease, Emory University School of Medicine, Atlanta, GA 30322, USA
| |
Collapse
|
48
|
Biricioiu MR, Sarbu M, Ica R, Vukelić Ž, Clemmer DE, Zamfir AD. Human Cerebellum Gangliosides: A Comprehensive Analysis by Ion Mobility Tandem Mass Spectrometry. JOURNAL OF THE AMERICAN SOCIETY FOR MASS SPECTROMETRY 2024; 35:683-695. [PMID: 38518248 DOI: 10.1021/jasms.3c00360] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/24/2024]
Abstract
The human cerebellum is an ultraspecialized region of the brain responsible for cognitive functions and movement coordination. The fine mechanisms through which the process of aging impacts such functions are not well understood; therefore, a rigorous exploration of this brain region at the molecular level is deemed necessary. Gangliosides, sialylated glycosphingolipids, highly and specifically expressed in the human central nervous system, represent possible molecular markers of cerebellum development and aging. In this context, for a comprehensive determination of development- and age-specific components, we have conducted here a comparative profiling and structural determination of the gangliosides expressed in fetal cerebellum in two intrauterine developmental stages and aged cerebellum by ion mobility separation (IMS) mass spectrometry (MS) and tandem MS (MS/MS). Due to the high sensitivity and efficiency of separation provided by IMS MS, no less than 551 chemically distinct species were identified, which represents 4.5 times more gangliosides than ever discovered in this brain region. The detailed assessment of fetal vs aged cerebellum gangliosidome showed marked discrepancies not only in the general number of the species expressed, but also in their sialylation patterns, the modifications of the glycan core, and the composition of the ceramides. All of these characteristics are potential markers of cerebellum development and aging. The structural analysis by collision-induced dissociation (CID) documented the occurrence of GD1b (d18:1/18:0) isomer in the fetal cerebellum in the second gestational trimester, with all probability of GQ1b (t18:1/18:0) in the near-term fetus and of GQ1b (d18:1/18:0) in aged cerebellum.
Collapse
Affiliation(s)
- Maria Roxana Biricioiu
- Department of Condensed Matter, National Institute for Research and Development in Electrochemistry and Condensed Matter, Timisoara, 300224, Romania
- Department of Physics, West University of Timisoara, Timisoara 300223, Romania
| | - Mirela Sarbu
- Department of Condensed Matter, National Institute for Research and Development in Electrochemistry and Condensed Matter, Timisoara, 300224, Romania
| | - Raluca Ica
- Department of Condensed Matter, National Institute for Research and Development in Electrochemistry and Condensed Matter, Timisoara, 300224, Romania
| | - Željka Vukelić
- Department of Chemistry and Biochemistry, School of Medicine, University of Zagreb, Zagreb 10000, Croatia
| | - David E Clemmer
- Department of Chemistry, Indiana University, Bloomington, Indiana 47405, United States
| | - Alina D Zamfir
- Department of Condensed Matter, National Institute for Research and Development in Electrochemistry and Condensed Matter, Timisoara, 300224, Romania
- Institute for Research, Development and Innovation in Natural and Technical Sciences, Aurel Vlaicu University of Arad, Arad 310330, Romania
| |
Collapse
|
49
|
Xie Y, Chen S, Alvarez MR, Sheng Y, Li Q, Maverakis E, Lebrilla CB. Protein oxidation of fucose environments (POFE) reveals fucose-protein interactions. Chem Sci 2024; 15:5256-5267. [PMID: 38577366 PMCID: PMC10988611 DOI: 10.1039/d3sc06432h] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2023] [Accepted: 03/03/2024] [Indexed: 04/06/2024] Open
Abstract
Cell membrane glycoproteins are generally highly fucosylated and sialylated, and post-translational modifications play important roles in the proteins' functions of signaling, binding and cellular processing. For these reasons, methods for measuring sialic acid-mediated protein-protein interactions have been developed. However, determining the role of fucose in these interactions has been limited by technological barriers that have thus far hindered the ability to characterize and observe fucose-mediated protein-protein interactions. Herein, we describe a method to metabolically label mammalian cells with modified fucose, which incorporates a bioorthogonal group into cell membrane glycoproteins thereby enabling the characterization of cell-surface fucose interactome. Copper-catalyzed click chemistry was used to conjugate a proximity labeling probe, azido-FeBABE. Following the addition of hydrogen peroxide (H2O2), the fucose-azido-FeBABE catalyzed the formation of hydroxyl radicals, which in turn oxidized the amino acids in the proximity of the labeled fucose residue. The oxidized peptides were identified using liquid chromatography coupled with tandem mass spectrometry (LC-MS/MS). Variations in degree of protein oxidation were obtained with different H2O2 reaction times yielding the acquisition of spatial information of the fucose-interacting proteins. In addition, specific glycoprotein-protein interactions were constructed for Galectin-3 (LEG3) and Galectin-3-binding protein (LG3BP) illustrating the further utility of the method. This method identifies new fucose binding partners thereby enhancing our understanding of the cell glycocalyx.
Collapse
Affiliation(s)
- Yixuan Xie
- Department of Chemistry, University of California, Davis Davis California USA
- Department of Biochemistry and Molecular Biophysics, Washington University School of Medicine St. Louis Missouri 63110 USA
| | - Siyu Chen
- Department of Chemistry, University of California, Davis Davis California USA
| | | | - Ying Sheng
- Department of Chemistry, University of California, Davis Davis California USA
| | - Qiongyu Li
- Department of Chemistry, University of California, Davis Davis California USA
| | - Emanual Maverakis
- Department of Dermatology, University of California, Davis Sacramento California USA
| | - Carlito B Lebrilla
- Department of Chemistry, University of California, Davis Davis California USA
- Department of Biochemistry, University of California, Davis Davis California USA
| |
Collapse
|
50
|
Nie H, Saini P, Miyamoto T, Liao L, Zielinski RJ, Liu H, Zhou W, Wang C, Murphy B, Towers M, Yang T, Qi Y, Kannan T, Kossenkov A, Tateno H, Claiborne DT, Zhang N, Abdel-Mohsen M, Zhang R. Targeting branched N-glycans and fucosylation sensitizes ovarian tumors to immune checkpoint blockade. Nat Commun 2024; 15:2853. [PMID: 38565883 PMCID: PMC10987604 DOI: 10.1038/s41467-024-47069-y] [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: 09/21/2023] [Accepted: 03/19/2024] [Indexed: 04/04/2024] Open
Abstract
Aberrant glycosylation is a crucial strategy employed by cancer cells to evade cellular immunity. However, it's unclear whether homologous recombination (HR) status-dependent glycosylation can be therapeutically explored. Here, we show that the inhibition of branched N-glycans sensitizes HR-proficient, but not HR-deficient, epithelial ovarian cancers (EOCs) to immune checkpoint blockade (ICB). In contrast to fucosylation whose inhibition sensitizes EOCs to anti-PD-L1 immunotherapy regardless of HR-status, we observe an enrichment of branched N-glycans on HR-proficient compared to HR-deficient EOCs. Mechanistically, BRCA1/2 transcriptionally promotes the expression of MGAT5, the enzyme responsible for catalyzing branched N-glycans. The branched N-glycans on HR-proficient tumors augment their resistance to anti-PD-L1 by enhancing its binding with PD-1 on CD8+ T cells. In orthotopic, syngeneic EOC models in female mice, inhibiting branched N-glycans using 2-Deoxy-D-glucose sensitizes HR-proficient, but not HR-deficient EOCs, to anti-PD-L1. These findings indicate branched N-glycans as promising therapeutic targets whose inhibition sensitizes HR-proficient EOCs to ICB by overcoming immune evasion.
Collapse
Affiliation(s)
- Hao Nie
- Department of Experimental Therapeutics, University of Texas MD Anderson Cancer Center, Houston, TX, 77054, USA
| | - Pratima Saini
- Vaccine and Immunotherapy Center, The Wistar Institute, Philadelphia, PA, 19104, USA
| | - Taito Miyamoto
- Immunology, Microenvironment and Metastasis Program, The Wistar Institute, Philadelphia, PA, 19104, USA
| | - Liping Liao
- Department of Experimental Therapeutics, University of Texas MD Anderson Cancer Center, Houston, TX, 77054, USA
| | - Rafal J Zielinski
- Department of Experimental Therapeutics, University of Texas MD Anderson Cancer Center, Houston, TX, 77054, USA
| | - Heng Liu
- Immunology, Microenvironment and Metastasis Program, The Wistar Institute, Philadelphia, PA, 19104, USA
| | - Wei Zhou
- Immunology, Microenvironment and Metastasis Program, The Wistar Institute, Philadelphia, PA, 19104, USA
| | - Chen Wang
- Department of Experimental Therapeutics, University of Texas MD Anderson Cancer Center, Houston, TX, 77054, USA
| | - Brennah Murphy
- Immunology, Microenvironment and Metastasis Program, The Wistar Institute, Philadelphia, PA, 19104, USA
| | - Martina Towers
- Department of Experimental Therapeutics, University of Texas MD Anderson Cancer Center, Houston, TX, 77054, USA
| | - Tyler Yang
- Immunology, Microenvironment and Metastasis Program, The Wistar Institute, Philadelphia, PA, 19104, USA
| | - Yuan Qi
- Department of Bioinformatics & Computational Biology, University of Texas MD Anderson Cancer Center, Houston, TX, 77054, USA
| | - Toshitha Kannan
- Bioinformatics Facility, The Wistar Institute, Philadelphia, PA, 19104, USA
| | - Andrew Kossenkov
- Gene Expression and Regulation Program, The Wistar Institute, Philadelphia, PA, 19104, USA
| | - Hiroaki Tateno
- Cellular and Molecular Biotechnology Research Institute, National Institute of Advanced Industrial Science and Technology (AIST), Tsukuba, Ibaraki, 305-8566, Japan
| | - Daniel T Claiborne
- Immunology, Microenvironment and Metastasis Program, The Wistar Institute, Philadelphia, PA, 19104, USA
| | - Nan Zhang
- Immunology, Microenvironment and Metastasis Program, The Wistar Institute, Philadelphia, PA, 19104, USA
| | - Mohamed Abdel-Mohsen
- Vaccine and Immunotherapy Center, The Wistar Institute, Philadelphia, PA, 19104, USA.
| | - Rugang Zhang
- Department of Experimental Therapeutics, University of Texas MD Anderson Cancer Center, Houston, TX, 77054, USA.
- Immunology, Microenvironment and Metastasis Program, The Wistar Institute, Philadelphia, PA, 19104, USA.
| |
Collapse
|