51
|
Dhara D, Mulard LA, Hollenstein M. Natural, modified and conjugated carbohydrates in nucleic acids. Chem Soc Rev 2025; 54:2948-2983. [PMID: 39936337 DOI: 10.1039/d4cs00799a] [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: 02/13/2025]
Abstract
Storage of genetic information in DNA occurs through a unique ordering of canonical base pairs. However, this would not be possible in the absence of the sugar-phosphate backbone which is essential for duplex formation. While over a hundred nucleobase modifications have been identified (mainly in RNA), Nature is rather conservative when it comes to alterations at the level of the (deoxy)ribose sugar moiety. This trend is not reflected in synthetic analogues of nucleic acids where modifications of the sugar entity is commonplace to improve the properties of DNA and RNA. In this review article, we describe the main incentives behind sugar modifications in nucleic acids and we highlight recent progress in this field with a particular emphasis on therapeutic applications, the development of xeno-nucleic acids (XNAs), and on interrogating nucleic acid etiology. We also describe recent strategies to conjugate carbohydrates and oligosaccharides to oligonucleotides since this represents a particularly powerful strategy to improve the therapeutic index of oligonucleotide drugs. The advent of glycoRNAs combined with progress in nucleic acid and carbohydrate chemistry, protein engineering, and delivery methods will undoubtedly yield more potent sugar-modified nucleic acids for therapeutic, biotechnological, and synthetic biology applications.
Collapse
Affiliation(s)
- Debashis Dhara
- Department of Structural Biology and Chemistry, Laboratory for Bioorganic Chemistry of Nucleic Acids, Institut Pasteur, Université Paris Cité, CNRS UMR 352328, rue du Docteur Roux, 75724 Paris Cedex 15, France.
- Department of Structural Biology and Chemistry, Laboratory for Chemistry of Biomolecules, Institut Pasteur, Université Paris Cité, CNRS UMR 3523, 28, rue du Docteur Roux, 75724 Paris Cedex 15, France
| | - Laurence A Mulard
- Department of Structural Biology and Chemistry, Laboratory for Chemistry of Biomolecules, Institut Pasteur, Université Paris Cité, CNRS UMR 3523, 28, rue du Docteur Roux, 75724 Paris Cedex 15, France
| | - Marcel Hollenstein
- Department of Structural Biology and Chemistry, Laboratory for Bioorganic Chemistry of Nucleic Acids, Institut Pasteur, Université Paris Cité, CNRS UMR 352328, rue du Docteur Roux, 75724 Paris Cedex 15, France.
| |
Collapse
|
52
|
Lee KW, Pham NT, Min HJ, Park HW, Lee JW, Lo HE, Kwon NY, Seo J, Shaginyan I, Cho H, Wei L, Manavalan B, Jeon YJ. DOGpred: A Novel Deep Learning Framework for Accurate Identification of Human O-linked Threonine Glycosylation Sites. J Mol Biol 2025; 437:168977. [PMID: 39900285 DOI: 10.1016/j.jmb.2025.168977] [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: 07/31/2024] [Revised: 01/06/2025] [Accepted: 01/28/2025] [Indexed: 02/05/2025]
Abstract
O-linked glycosylation is a crucial post-translational modification that regulates protein function and biological processes. Dysregulation of this process is associated with various diseases, underscoring the need to accurately identify O-linked glycosylation sites on proteins. Current experimental methods for identifying O-linked threonine glycosylation (OTG) sites are often complex and costly. Consequently, developing computational tools that predict these sites based on protein features is crucial. Such tools can complement experimental approaches, enhancing our understanding of the role of OTG dysregulation in diseases and uncovering potential therapeutic targets. In this study, we developed DOGpred, a deep learning-based predictor for precisely identifying human OTGs using high-latent feature representations. Initially, we extracted nine different conventional feature descriptors (CFDs) and nine pre-trained protein language model (PLM)-based embeddings. Notably, each feature was encoded as a 2D tensor, capturing both the sequential and inherent feature characteristics. Subsequently, we designed a stacked convolutional neural network (CNN) module to learn spatial feature representations from CFDs and a stacked recurrent neural network (RNN) module to learn temporal feature representations from PLM-based embeddings. These features were integrated using attention-based fusion mechanisms to generate high-level feature representations for final classification. Ablation analysis and independent tests demonstrated that the optimal model (DOGpred), employing a stacked 1D CNN and a stacked attention-based RNN modules with cross-attention feature fusion, achieved the best performance on the training dataset and significantly outperformed machine learning-based single-feature models and state-of-the-art methods on independent datasets. Furthermore, DOGpred is publicly available at https://github.com/JeonRPM/DOGpred/ for free access and usage.
Collapse
Affiliation(s)
- Ki Wook Lee
- Department of Integrative Biotechnology, College of Biotechnology and Bioengineering, Sungkyunkwan University, Suwon, Republic of Korea
| | - Nhat Truong Pham
- Department of Integrative Biotechnology, College of Biotechnology and Bioengineering, Sungkyunkwan University, Suwon, Republic of Korea
| | - Hye Jung Min
- Department of Integrative Biotechnology, College of Biotechnology and Bioengineering, Sungkyunkwan University, Suwon, Republic of Korea
| | - Hyun Woo Park
- Department of Integrative Biotechnology, College of Biotechnology and Bioengineering, Sungkyunkwan University, Suwon, Republic of Korea
| | - Ji Won Lee
- Department of Integrative Biotechnology, College of Biotechnology and Bioengineering, Sungkyunkwan University, Suwon, Republic of Korea
| | - Han-En Lo
- Department of Integrative Biotechnology, College of Biotechnology and Bioengineering, Sungkyunkwan University, Suwon, Republic of Korea
| | - Na Young Kwon
- Department of Integrative Biotechnology, College of Biotechnology and Bioengineering, Sungkyunkwan University, Suwon, Republic of Korea
| | - Jimin Seo
- Department of Integrative Biotechnology, College of Biotechnology and Bioengineering, Sungkyunkwan University, Suwon, Republic of Korea
| | - Illia Shaginyan
- Department of Integrative Biotechnology, College of Biotechnology and Bioengineering, Sungkyunkwan University, Suwon, Republic of Korea
| | - Heeje Cho
- Department of Integrative Biotechnology, College of Biotechnology and Bioengineering, Sungkyunkwan University, Suwon, Republic of Korea
| | - Leyi Wei
- Centre for Artificial Intelligence Driven Drug Discovery, Faculty of Applied Science, Macao Polytechnic University, Macau
| | - Balachandran Manavalan
- Department of Integrative Biotechnology, College of Biotechnology and Bioengineering, Sungkyunkwan University, Suwon, Republic of Korea
| | - Young-Jun Jeon
- Department of Integrative Biotechnology, College of Biotechnology and Bioengineering, Sungkyunkwan University, Suwon, Republic of Korea.
| |
Collapse
|
53
|
Chen L, Li Y, Guo Y, Wang G, Feng N, Sun J, Zhong Y, Yao Y, Ding L, Ju H. Two-Level Spatially Localized Proximity Labeling for Cross-Biological-Hierarchy Measurement and Manipulation. Angew Chem Int Ed Engl 2025; 64:e202421448. [PMID: 39805739 DOI: 10.1002/anie.202421448] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2024] [Revised: 12/31/2024] [Accepted: 01/13/2025] [Indexed: 01/16/2025]
Abstract
Proximity labeling (PL) has emerged as a powerful technique for the in situ elucidation of biomolecular interaction networks. However, PL methods generally rely on single-biological-hierarchy control of spatial localization at the labeling site, which limits their application in multi-tiered biological systems. Here, we introduced another enzymatic reaction upstream of an enzyme-based PL reaction and targeted the two enzymes to markers indicating different biological hierarchies, establishing a two-level spatially localized proximity labeling (P2L) platform for in situ molecular measurement and manipulation. Using the cellular- and glycan-level as the hierarchical models, we demonstrated the ability of P2L to efficiently execute a two-step logic operation and to discriminate target cells with different levels of glycosylation within mixed cell populations. By mounting clickable handles via P2L, we reprogrammed the robust covalent assembly of cells at designated sites. The combination of P2L with proteomics led to the profiling of the protein microenvironment of specific glycans on target cells, revealing changes in tumor-cell-surface interactions under immune pressure from a glycan perspective. P2L provides not only a solution for revealing the heterogeneity of biological systems, but also new insights in the fields of intelligent logic computation, enzyme engineering, tissue engineering, etc.
Collapse
Affiliation(s)
- Liusheng Chen
- State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing, 210023, China
| | - Yiran Li
- State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing, 210023, China
| | - Yuna Guo
- School of Clinical and Basic Medical Sciences, Shandong First Medical University, Jinan, 250117, China
| | - Guyu Wang
- State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing, 210023, China
| | - Nan Feng
- State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing, 210023, China
| | - Jiahui Sun
- State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing, 210023, China
| | - Yihong Zhong
- College of Chemistry and Materials, Jiangxi Normal University, Nanchang, 330022, China
| | - Yunyan Yao
- State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing, 210023, China
| | - Lin Ding
- State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing, 210023, China
- Chemistry and Biomedicine Innovation Center (ChemBIC), ChemBioMed Interdisciplinary Research Center, Nanjing University, Nanjing, 210023, China
| | - Huangxian Ju
- State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing, 210023, China
| |
Collapse
|
54
|
Puagsopa J, Tongviseskul N, Jaroentomeechai T, Meksiriporn B. Recent Progress in Developing Extracellular Vesicles as Nanovehicles to Deliver Carbohydrate-Based Therapeutics and Vaccines. Vaccines (Basel) 2025; 13:285. [PMID: 40266147 PMCID: PMC11946770 DOI: 10.3390/vaccines13030285] [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: 01/31/2025] [Revised: 02/22/2025] [Accepted: 03/04/2025] [Indexed: 04/24/2025] Open
Abstract
Cell-derived, nanoscale extracellular vesicles (EVs) have emerged as promising tools in diagnostic, therapeutic, and vaccine applications. Their unique properties including the capability to encapsulate diverse molecular cargo as well as the versatility in surface functionalization make them ideal candidates for safe and effective vehicles to deliver a range of biomolecules including gene editing cassettes, therapeutic proteins, glycans, and glycoconjugate vaccines. In this review, we discuss recent advances in the development of EVs derived from mammalian and bacterial cells for use in a delivery of carbohydrate-based protein therapeutics and vaccines. We highlight key innovations in EVs' molecular design, characterization, and deployment for treating diseases including Alzheimer's disease, infectious diseases, and cancers. We discuss challenges for their clinical translation and provide perspectives for future development of EVs within biopharmaceutical research and the clinical translation landscape.
Collapse
Affiliation(s)
- Japigorn Puagsopa
- Department of Physiology and Aging, College of Medicine, University of Florida, Gainesville, FL 32610, USA;
| | - Niksa Tongviseskul
- Department of Biology, School of Science, King Mongkut’s Institute of Technology Ladkrabang, Bangkok 10520, Thailand;
| | - Thapakorn Jaroentomeechai
- Copenhagen Center for Glycomics, Departments of Cellular and Molecular Medicine, Faculty of Health Sciences, University of Copenhagen, Blegdamsvej 3, 2200 Copenhagen, Denmark
| | - Bunyarit Meksiriporn
- Department of Biology, School of Science, King Mongkut’s Institute of Technology Ladkrabang, Bangkok 10520, Thailand;
| |
Collapse
|
55
|
Silsirivanit A, Alvarez MR, Grijaldo-Alvarez SJ, Gogte R, Kitkhuandee A, Piyawattanametha N, Seubwai W, Luang S, Panawan O, Mahalapbutr P, Vaeteewoottacharn K, Sawanyawisuth K, Let-itthiporn W, Saengboonmee C, Duangthongphon P, Jingjit K, Pankongsap A, Waraasawapati S, Aphivatanasiri C, Lebrilla CB. Serum N-Glycomics with Nano-LC-QToF LC-MS/MS Reveals N-Glycan Biomarkers for Glioblastoma, Meningioma, and High-Grade Meningioma. J Proteome Res 2025; 24:1402-1413. [PMID: 39905713 PMCID: PMC11894639 DOI: 10.1021/acs.jproteome.4c01090] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2024] [Revised: 01/21/2025] [Accepted: 01/28/2025] [Indexed: 02/06/2025]
Abstract
Alteration of glycosylation in cancer cells leads to the expression of tumor-associated glycans, which can be used as biomarkers for diagnosis and prognostic prediction of diseases. In this study, we used nano-LC-QToF to identify serum N-glycan biomarkers for the detection of brain tumors. We observed an increase in sialylated N-glycans and a decrease in fucosylated N-glycans in the serum of patients with glioblastoma (GBM) and meningioma (MG) compared to healthy individuals. In GBM, a combination of increased serum sialylated N-glycan (6_4_0_2 compound) and decreased fucosylated N-glycan (4_4_1_0 compound) was identified as the most appropriate panel, with an area under the curve (AUC) of 0.8660, 78.95% sensitivity, 84.21% specificity, and 82.89% accuracy. For MG, a combination of decreased 6_6_2_0 and 5_5_2_0 compounds and increased 4_4_1_1 compound achieved an AUC of 0.9260, 82.35% sensitivity, 78.57% specificity, and 80.26% accuracy for diagnosis of MG. Additionally, an increase in 5_5_1_0 and 4_3_0_0 compounds combined with a decrease in 7_7_4_3 was associated with high-grade MG (WHO grades II-III). In conclusion, we identified serum N-glycan profiles associated with brain tumors, highlighting their potential as biomarkers for the diagnosis and prognosis of these diseases.
Collapse
Affiliation(s)
- Atit Silsirivanit
- Department
of Biochemistry, Faculty of Medicine, Khon
Kaen University, Khon Kaen 40002, Thailand
| | | | | | - Riya Gogte
- Department
of Chemistry, University of California,
Davis, Davis, California 95616, United States
| | - Amnat Kitkhuandee
- Department
of Surgery, Faculty of Medicine, Khon Kaen
University, Khon Kaen 40002, Thailand
| | | | - Wunchana Seubwai
- Department
of Forensic Medicine, Faculty of Medicine, Khon Kaen University, Khon Kaen 40002, Thailand
| | - Sukanya Luang
- Department
of Biochemistry, Faculty of Medicine, Khon
Kaen University, Khon Kaen 40002, Thailand
| | - Orasa Panawan
- Department
of Biochemistry, Faculty of Medicine, Khon
Kaen University, Khon Kaen 40002, Thailand
| | - Panupong Mahalapbutr
- Department
of Biochemistry, Faculty of Medicine, Khon
Kaen University, Khon Kaen 40002, Thailand
| | | | - Kanlayanee Sawanyawisuth
- Department
of Biochemistry, Faculty of Medicine, Khon
Kaen University, Khon Kaen 40002, Thailand
| | - Worachart Let-itthiporn
- Department
of Biochemistry, Faculty of Medicine, Khon
Kaen University, Khon Kaen 40002, Thailand
| | - Charupong Saengboonmee
- Department
of Biochemistry, Faculty of Medicine, Khon
Kaen University, Khon Kaen 40002, Thailand
| | | | - Kritsakorn Jingjit
- Department
of Surgery, Faculty of Medicine, Khon Kaen
University, Khon Kaen 40002, Thailand
| | - Anuchit Pankongsap
- Department
of Surgery, Faculty of Medicine, Khon Kaen
University, Khon Kaen 40002, Thailand
| | - Sakda Waraasawapati
- Department
of Pathology, Faculty of Medicine, Khon
Kaen University, Khon Kaen 40002, Thailand
| | - Chaiwat Aphivatanasiri
- Department
of Pathology, Faculty of Medicine, Khon
Kaen University, Khon Kaen 40002, Thailand
| | - Carlito B. Lebrilla
- Department
of Chemistry, University of California,
Davis, Davis, California 95616, United States
| |
Collapse
|
56
|
Abdeen AH, Trist BG, Nikseresht S, Harwood R, Roudeau S, Rowlands BD, Kreilaus F, Cottam V, Mor D, Richardson M, Siciliano J, Forkgen J, Schaffer G, Genoud S, Li AA, Proschogo N, Antonio B, Falkenberg G, Brueckner D, Kysenius K, Liddell JR, Fat SCM, Wu S, Fifita J, Lockwood TE, Bishop DP, Blair I, Ortega R, Crouch PJ, Double KL. Parkinson-like wild-type superoxide dismutase 1 pathology induces nigral dopamine neuron degeneration in a novel murine model. Acta Neuropathol 2025; 149:22. [PMID: 40042537 PMCID: PMC11882636 DOI: 10.1007/s00401-025-02859-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2024] [Revised: 02/12/2025] [Accepted: 02/12/2025] [Indexed: 03/09/2025]
Abstract
Atypical wild-type superoxide dismutase 1 (SOD1) protein misfolding and deposition occurs specifically within the degenerating substantia nigra pars compacta (SNc) in Parkinson disease. Mechanisms driving the formation of this pathology and relationship with SNc dopamine neuron health are yet to be fully understood. We applied proteomic mass spectrometry and synchrotron-based biometal quantification to post-mortem brain tissues from the SNc of Parkinson disease patients and age-matched controls to uncover key factors underlying the formation of wild-type SOD1 pathology in this disorder. We also engineered two of these factors - brain copper deficiency and upregulated SOD1 protein levels - into a novel mouse strain, termed the SOCK mouse, to verify their involvement in the development of Parkinson-like wild-type SOD1 pathology and their impact on dopamine neuron health. Soluble SOD1 protein in the degenerating Parkinson disease SNc exhibited altered post-translational modifications, which may underlie changes to the enzymatic activity and aggregation of the protein in this region. These include decreased copper binding, dysregulation of physiological glycosylation, and atypical oxidation and glycation of key SOD1 amino acid residues. We demonstrated that the biochemical profile introduced in SOCK mice promotes the same post-translational modifications and the development of Parkinson-like wild-type SOD1 pathology in the midbrain and cortex. This pathology accumulates progressively with age and is accompanied by nigrostriatal degeneration and dysfunction, which occur in the absence of α-synuclein deposition. These mice do not exhibit weight loss nor spinal cord motor neuron degeneration, distinguishing them from transgenic mutant SOD1 mouse models. This study provides the first in vivo evidence that mismetallation and altered post-translational modifications precipitates wild-type SOD1 misfolding, dysfunction, and deposition in the Parkinson disease brain, which may contribute to SNc dopamine neuron degeneration. Our data position this pathology as a novel drug target for this disorder, with a particular focus on therapies capable of correcting alterations to SOD1 post-translational modifications.
Collapse
Affiliation(s)
- Amr H Abdeen
- Brain and Mind Centre and School of Medical Sciences (Neuroscience), Faculty of Medicine and Health, The University of Sydney, 94-100 Mallett Street, Camperdown, Sydney, NSW, 2006, Australia
| | - Benjamin G Trist
- Brain and Mind Centre and School of Medical Sciences (Neuroscience), Faculty of Medicine and Health, The University of Sydney, 94-100 Mallett Street, Camperdown, Sydney, NSW, 2006, Australia
| | - Sara Nikseresht
- Department of Anatomy & Physiology, Faculty of Medicine, Dentistry and Health Sciences, The University of Melbourne, Parkville, VIC, 3052, Australia
| | - Richard Harwood
- Sydney Microscopy and Microanalysis, The University of Sydney, Sydney, NSW, 2006, Australia
| | - Stéphane Roudeau
- Univ. Bordeaux, CNRS, LP2I Bordeaux, UMR 5797, 33170, Gradignan, France
| | - Benjamin D Rowlands
- Brain and Mind Centre and School of Medical Sciences (Neuroscience), Faculty of Medicine and Health, The University of Sydney, 94-100 Mallett Street, Camperdown, Sydney, NSW, 2006, Australia
| | - Fabian Kreilaus
- Brain and Mind Centre and School of Medical Sciences (Neuroscience), Faculty of Medicine and Health, The University of Sydney, 94-100 Mallett Street, Camperdown, Sydney, NSW, 2006, Australia
| | - Veronica Cottam
- Brain and Mind Centre and School of Medical Sciences (Neuroscience), Faculty of Medicine and Health, The University of Sydney, 94-100 Mallett Street, Camperdown, Sydney, NSW, 2006, Australia
| | - David Mor
- Brain and Mind Centre and School of Medical Sciences (Neuroscience), Faculty of Medicine and Health, The University of Sydney, 94-100 Mallett Street, Camperdown, Sydney, NSW, 2006, Australia
| | - Miriam Richardson
- Brain and Mind Centre and School of Medical Sciences (Neuroscience), Faculty of Medicine and Health, The University of Sydney, 94-100 Mallett Street, Camperdown, Sydney, NSW, 2006, Australia
| | - Joel Siciliano
- Brain and Mind Centre and School of Medical Sciences (Neuroscience), Faculty of Medicine and Health, The University of Sydney, 94-100 Mallett Street, Camperdown, Sydney, NSW, 2006, Australia
| | - Julia Forkgen
- Brain and Mind Centre and School of Medical Sciences (Neuroscience), Faculty of Medicine and Health, The University of Sydney, 94-100 Mallett Street, Camperdown, Sydney, NSW, 2006, Australia
| | - Greta Schaffer
- Brain and Mind Centre and School of Medical Sciences (Neuroscience), Faculty of Medicine and Health, The University of Sydney, 94-100 Mallett Street, Camperdown, Sydney, NSW, 2006, Australia
| | - Sian Genoud
- Brain and Mind Centre and School of Medical Sciences (Neuroscience), Faculty of Medicine and Health, The University of Sydney, 94-100 Mallett Street, Camperdown, Sydney, NSW, 2006, Australia
| | - Anne A Li
- Brain and Mind Centre and School of Medical Sciences (Neuroscience), Faculty of Medicine and Health, The University of Sydney, 94-100 Mallett Street, Camperdown, Sydney, NSW, 2006, Australia
| | - Nicholas Proschogo
- Mass Spectrometry Facility, Faculty of Science, The University of Sydney, Sydney, NSW, 2006, Australia
| | - Bernadeth Antonio
- Mass Spectrometry Facility, Faculty of Science, The University of Sydney, Sydney, NSW, 2006, Australia
| | - Gerald Falkenberg
- Deutsches Elektronen-Synchrotron DESY, Notkestr. 85, 22607, Hamburg, Germany
| | - Dennis Brueckner
- Deutsches Elektronen-Synchrotron DESY, Notkestr. 85, 22607, Hamburg, Germany
| | - Kai Kysenius
- Department of Anatomy & Physiology, Faculty of Medicine, Dentistry and Health Sciences, The University of Melbourne, Parkville, VIC, 3052, Australia
| | - Jeffrey R Liddell
- Department of Anatomy & Physiology, Faculty of Medicine, Dentistry and Health Sciences, The University of Melbourne, Parkville, VIC, 3052, Australia
| | - Sandrine Chan Moi Fat
- Motor Neuron Disease Research Centre, Macquarie Medical School, Faculty of Medicine, Health and Human Sciences, Macquarie University, Sydney, Australia
| | - Sharlynn Wu
- Motor Neuron Disease Research Centre, Macquarie Medical School, Faculty of Medicine, Health and Human Sciences, Macquarie University, Sydney, Australia
| | - Jennifer Fifita
- Motor Neuron Disease Research Centre, Macquarie Medical School, Faculty of Medicine, Health and Human Sciences, Macquarie University, Sydney, Australia
| | - Thomas E Lockwood
- Hyphenated Mass Spectrometry Laboratory, Faculty of Science, University of Technology Sydney, Sydney, NSW, 2007, Australia
| | - David P Bishop
- Hyphenated Mass Spectrometry Laboratory, Faculty of Science, University of Technology Sydney, Sydney, NSW, 2007, Australia
| | - Ian Blair
- Motor Neuron Disease Research Centre, Macquarie Medical School, Faculty of Medicine, Health and Human Sciences, Macquarie University, Sydney, Australia
| | - Richard Ortega
- Univ. Bordeaux, CNRS, LP2I Bordeaux, UMR 5797, 33170, Gradignan, France
| | - Peter J Crouch
- Department of Anatomy & Physiology, Faculty of Medicine, Dentistry and Health Sciences, The University of Melbourne, Parkville, VIC, 3052, Australia
| | - Kay L Double
- Brain and Mind Centre and School of Medical Sciences (Neuroscience), Faculty of Medicine and Health, The University of Sydney, 94-100 Mallett Street, Camperdown, Sydney, NSW, 2006, Australia.
| |
Collapse
|
57
|
Cocchiararo I, Castets P. Recent advances in the clinical spectrum and pathomechanisms associated with X-linked myopathy with excessive autophagy and other VMA21-related disorders. J Neuromuscul Dis 2025:22143602251314767. [PMID: 40033998 DOI: 10.1177/22143602251314767] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/05/2025]
Abstract
X-linked myopathy with excessive autophagy (XMEA) is a rare neuromuscular disorder caused by mutations in the VMA21 gene, encoding a chaperone protein present in the endoplasmic reticulum (ER). In yeast and human, VMA21 has been shown to chaperone the assembly of the vacuolar (v)-ATPase proton pump required for the acidification of lysosomes and other organelles. In line with this, VMA21 deficiency in XMEA impairs autophagic degradation steps, which would be key in XMEA pathogenesis. Recent years have witnessed a surge of interest in VMA21, with the identification of novel mutations causing a congenital disorder of glycosylation (CDG) with liver affection, and its potent implication in cancer predisposition. With this, VMA21 deficiency has been further linked to defective glycosylation, lipid metabolism dysregulation and ER stress. Moreover, the identification of two VMA21 isoforms, namely VMA21-101 and VMA21-120, has opened novel avenues regarding the pathomechanisms leading to XMEA and VMA21-CDG. In this review, we discuss recent advances on the clinical spectrum associated with VMA21 deficiency and on the pathophysiological roles of VMA21.
Collapse
Affiliation(s)
- Ilaria Cocchiararo
- Department of Cell Physiology and Metabolism, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Perrine Castets
- Department of Cell Physiology and Metabolism, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| |
Collapse
|
58
|
Noel M, Suttapitugsakul S, Cummings RD, Mealer RG. O-GalNAc glycans are enriched in neuronal tracts and regulate nodes of Ranvier. Proc Natl Acad Sci U S A 2025; 122:e2418949122. [PMID: 39999163 DOI: 10.1073/pnas.2418949122] [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: 09/16/2024] [Accepted: 01/13/2025] [Indexed: 02/27/2025] Open
Abstract
Protein O-glycosylation is a critical modification in the brain, as genetic variants in the pathway are associated with common and severe neuropsychiatric phenotypes. However, little is known about the most abundant O-glycans in the mammalian brain, which are N-acetylgalactosamine (O-GalNAc) linked. Here, we determined the spatial localization, protein carriers, and cellular function of O-GalNAc glycans in the mouse brain. We observed striking spatial enrichment of O-GalNAc glycans in neuronal tracts, and specifically at nodes of Ranvier, specialized structures involved in signal propagation in the brain. Glycoproteomic analysis revealed that more than half of the identified O-GalNAc glycans were present on chondroitin sulfate proteoglycans termed lecticans, and display both domain enrichment and regional heterogeneity. Inhibition of O-GalNAc synthesis in neurons reduced binding of Siglec-4, a known regulator of neurite growth, and shortened the length of nodes of Ranvier. This work establishes a function of O-GalNAc glycans in the brain and will inform future studies on their role in development and disease.
Collapse
Affiliation(s)
- Maxence Noel
- Department of Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215
| | - Suttipong Suttapitugsakul
- Department of Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215
| | - Richard D Cummings
- Department of Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215
| | - Robert G Mealer
- Department of Psychiatry, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599
- Department of Genetics, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599
| |
Collapse
|
59
|
De la Fuente IM, Cortes JM, Malaina I, Pérez-Yarza G, Martinez L, López JI, Fedetz M, Carrasco-Pujante J. The main sources of molecular organization in the cell. Atlas of self-organized and self-regulated dynamic biostructures. PROGRESS IN BIOPHYSICS AND MOLECULAR BIOLOGY 2025; 195:167-191. [PMID: 39805422 DOI: 10.1016/j.pbiomolbio.2025.01.003] [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: 10/19/2024] [Accepted: 01/10/2025] [Indexed: 01/16/2025]
Abstract
One of the most important goals of contemporary biology is to understand the principles of the molecular order underlying the complex dynamic architecture of cells. Here, we present an overview of the main driving forces involved in the cellular molecular complexity and in the emergent functional dynamic structures, spanning from the most basic molecular organization levels to the complex emergent integrative systemic behaviors. First, we address the molecular information processing which is essential in many complex fundamental mechanisms such as the epigenetic memory, alternative splicing, regulation of transcriptional system, and the adequate self-regulatory adaptation to the extracellular environment. Next, we approach the biochemical self-organization, which is central to understand the emergency of metabolic rhythms, circadian oscillations, and spatial traveling waves. Such a complex behavior is also fundamental to understand the temporal compartmentalization of the cellular metabolism and the dynamic regulation of many physiological activities. Numerous examples of biochemical self-organization are considered here, which show that practically all the main physiological processes in the cell exhibit this type of dynamic molecular organization. Finally, we focus on the biochemical self-assembly which, at a primary level of organization, is a basic but important mechanism for the order in the cell allowing biomolecules in a disorganized state to form complex aggregates necessary for a plethora of essential structures and physiological functions. In total, more than 500 references have been compiled in this review. Due to these main sources of order, systemic functional structures emerge in the cell, driving the metabolic functionality towards the biological complexity.
Collapse
Affiliation(s)
- Ildefonso M De la Fuente
- Department of Mathematics, Faculty of Science and Technology, University of the Basque Country, UPV/EHU, Leioa, 48940, Spain.
| | - Jesus M Cortes
- Department of Cell Biology and Histology, Faculty of Medicine and Nursing, University of the Basque Country, UPV/EHU, Leioa, 48940, Spain; Biobizkaia Health Research Institute, Barakaldo, 48903, Spain; IKERBASQUE: The Basque Foundation for Science, Bilbao, Spain
| | - Iker Malaina
- Department of Mathematics, Faculty of Science and Technology, University of the Basque Country, UPV/EHU, Leioa, 48940, Spain
| | - Gorka Pérez-Yarza
- Department of Cell Biology and Histology, Faculty of Medicine and Nursing, University of the Basque Country, UPV/EHU, Leioa, 48940, Spain
| | - Luis Martinez
- Department of Mathematics, Faculty of Science and Technology, University of the Basque Country, UPV/EHU, Leioa, 48940, Spain
| | - José I López
- Biobizkaia Health Research Institute, Barakaldo, 48903, Spain
| | - Maria Fedetz
- Department of Cell Biology and Immunology, Institute of Parasitology and Biomedicine "López-Neyra", CSIC, Granada, 18016, Spain
| | - Jose Carrasco-Pujante
- Department of Cell Biology and Histology, Faculty of Medicine and Nursing, University of the Basque Country, UPV/EHU, Leioa, 48940, Spain
| |
Collapse
|
60
|
Pratt MR. Photocrosslinking and capture for the analysis of carbohydrate-dependent interactions. Bioorg Med Chem Lett 2025; 117:130077. [PMID: 39710139 PMCID: PMC11745908 DOI: 10.1016/j.bmcl.2024.130077] [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: 10/29/2024] [Revised: 12/10/2024] [Accepted: 12/13/2024] [Indexed: 12/24/2024]
Abstract
Carbohydrates play crucial roles in biological systems, including by mediating cell and protein interactions. The complexity and transient nature of carbohydrate-dependent interactions pose significant challenges for their characterization, as traditional techniques often fail to capture these low-affinity binding events. This review highlights the increasing utility of photocrosslinkers in studying carbohydrate-mediated interactions. Photocrosslinkers, such as aryl azides, benzophenones, and diazirines, allow for the capture of fleeting interactions by forming covalent bonds upon UV irradiation, enabling the downstream application of standard biochemical techniques. I discuss the three primary strategies for incorporating photocrosslinkers: synthetic small molecules, metabolic labeling, and exo-enzymatic labeling. I predict that the continued development and application of these methodologies will enhance our understanding of glycan-mediated interactions and their implications in health and disease.
Collapse
Affiliation(s)
- Matthew R Pratt
- Department of Chemistry, University of Southern California, Los Angeles, CA 90089, USA.
| |
Collapse
|
61
|
Lee TA, Tsai EY, Liu SH, Chou WC, Hsu Hung SD, Chang CY, Chao CH, Yamaguchi H, Lai YJ, Chen HL, Li CW. Regulation of PD-L1 glycosylation and advances in cancer immunotherapy. Cancer Lett 2025; 612:217498. [PMID: 39855377 DOI: 10.1016/j.canlet.2025.217498] [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: 09/18/2024] [Revised: 01/18/2025] [Accepted: 01/22/2025] [Indexed: 01/27/2025]
Abstract
Protein glycosylation plays a versatile role in regulating homeostasis, such as cell migration, protein sorting, and the immune response. Drugs aimed at targeting glycosylation have strong implications for immunity enhancement, diagnosis, and cancer regression. Programmed death-ligand 1 (PD-L1), expressed in cancer or antigen-presenting cells, binds to programmed cell death protein 1 (PD-1) and suppresses T cells. Glycosylation of PD-L1 at N35, N192, N200, and N219 stabilizes PD-L1 on the cancer cell surface, which contributes to immune evasion by inhibiting T cell activity. To date, at least six glycosyltransferases and four associate proteins are known to regulate PD-L1 glycosylation. Terminal modifications such as poly-N-acetyl-lactosamine (poly-LacNAC), sulfation, and sialylation are commonly found on PD-L1, acting as an immune recognition ligand and regulating certain immune responses. Studies have identified many mechanisms and potential therapeutic targets within the glycosylation pathways of PD-L1, revealing their involvement in cancer pathology, immune evasion, and resistance to immunotherapy. In this review, we covered the glycoforms, terminal moiety, binding lectin, glycosyltransferase, as well as sugar analogs focusing on glycosylated PD-L1. We present a mechanism that originates from the endoplasmic reticulum (ER)-Golgi apparatus (Golgi) and its subsequent translocation to the cell membrane. This pathway determines the immune suppression function of PD-L1 and therefore regulates the immune response such as T cells, monocytes, and macrophages. This collection of findings underscores the significance of glycosylation in the role of PD-L1 in cancer and highlights multiple potential targets and strategies for improving therapeutic intervention and diagnostic techniques.
Collapse
Affiliation(s)
- Te-An Lee
- Institute of Biomedical Sciences, Academia Sinica, Taipei, 115, Taiwan
| | - En-Yun Tsai
- Institute of Biomedical Sciences, Academia Sinica, Taipei, 115, Taiwan; School of Pharmacy, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Shou-Hou Liu
- Institute of Biomedical Sciences, Academia Sinica, Taipei, 115, Taiwan
| | - Wen-Cheng Chou
- Institute of Biomedical Sciences, Academia Sinica, Taipei, 115, Taiwan
| | - Shih-Duo Hsu Hung
- Institute of Biomedical Sciences, Academia Sinica, Taipei, 115, Taiwan
| | - Chen-Yu Chang
- Institute of Biomedical Sciences, Academia Sinica, Taipei, 115, Taiwan
| | - Chi-Hong Chao
- Center For Intelligent Drug Systems and Smart Bio-devices (IDS(2)B), National Yang Ming Chiao Tung University, Hsinchu, 30010, Taiwan; Department of Biological Science and Technology, National Yang Ming Chiao Tung University, Hsinchu, 30010, Taiwan
| | - Hirohito Yamaguchi
- Graduate Institute of Biomedical Sciences, Graduate Institute of Cell Biology, Research Center for Cancer Biology and Center for Molecular Medicine, China Medical University, Taichung, Taiwan
| | - Yun-Ju Lai
- Solomont School of Nursing, Zuckerberg College of Health Sciences, University of Massachusetts Lowell, 113 Wilder Street, Lowell, MA, 01854, USA
| | - Hung-Lin Chen
- Master Program in Clinical Genomics and Proteomics, Taipei Medical University, Taipei, 11031, Taiwan
| | - Chia-Wei Li
- Institute of Biomedical Sciences, Academia Sinica, Taipei, 115, Taiwan.
| |
Collapse
|
62
|
Li P, Xu X, Zhang C, Chang Q, Wang J, Wang W, Ren H. Glycosylation on extracellular vesicles and its detection strategy: Paving the way for clinical use. Int J Biol Macromol 2025; 295:139714. [PMID: 39798737 DOI: 10.1016/j.ijbiomac.2025.139714] [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: 06/25/2024] [Revised: 01/03/2025] [Accepted: 01/07/2025] [Indexed: 01/15/2025]
Abstract
Extracellular vesicles (EVs) contain various glycans during their life cycle, from biogenesis to cellular recognition and uptake by recipient cells. EV glycosylation has substantial diagnostic significance in multiple health conditions, highlighting the necessity of determining an accurate glycosylation pattern for EVs from diverse biological fluids. Reliable and accessible glycan detection techniques help to elaborate the glycosylation-related functional alterations of specific proteins or lipids. However, multiple obstacles exist, including the inconsistency in glycosylation patterns between an entire batch of EVs and a specific EV protein, and difficulty in distinguishing glycosylation types after tedious separation and purification procedures. This review outlines recent advances in EV glycan detection, either at the glycomic level for a collection of intact EVs or at the molecular level for a specific protein on EVs. Particular emphasis has been placed on the abundance of EVs in body fluids and their unique characteristics for drug delivery of EVs, indicating an opportunity for diagnostic and therapeutic purposes via EV glycans.
Collapse
Affiliation(s)
- Ping Li
- The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao 266071, China; School of Rehabilitation Sciences and Engineering, University of Health and Rehabilitation Sciences, Qingdao 266024, China
| | - Xiao Xu
- The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao 266071, China
| | - Cong Zhang
- School of Food Science and Pharmaceutical Engineering, Nanjing Normal University, Nanjing 210042, China
| | - Qi Chang
- Department of Biomedical Engineering, School of Engineering, China Pharmaceutical University, Nanjing 211198, China
| | - Jie Wang
- The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao 266071, China.
| | - Weijie Wang
- School of Rehabilitation Sciences and Engineering, University of Health and Rehabilitation Sciences, Qingdao 266024, China.
| | - He Ren
- The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao 266071, China.
| |
Collapse
|
63
|
Xu J, Zhao Y, Tyler Mertens R, Ding Y, Xiao P. Sweet regulation - The emerging immunoregulatory roles of hexoses. J Adv Res 2025; 69:361-379. [PMID: 38631430 PMCID: PMC11954837 DOI: 10.1016/j.jare.2024.04.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2023] [Revised: 03/20/2024] [Accepted: 04/13/2024] [Indexed: 04/19/2024] Open
Abstract
BACKGROUND It is widely acknowledged that dietary habits have profound impacts on human health and diseases. As the most important sweeteners and energy sources in human diets, hexoses take part in a broad range of physiopathological processes. In recent years, emerging evidence has uncovered the crucial roles of hexoses, such as glucose, fructose, mannose, and galactose, in controlling the differentiation or function of immune cells. AIM OF REVIEW Herein, we reviewed the latest research progresses in the hexose-mediated modulation of immune responses, provided in-depth analyses of the underlying mechanisms, and discussed the unresolved issues in this field. KEY SCIENTIFIC CONCEPTS OF REVIEW Owing to their immunoregulatory effects, hexoses affect the onset and progression of various types of immune disorders, including inflammatory diseases, autoimmune diseases, and tumor immune evasion. Thus, targeting hexose metabolism is becoming a promising strategy for reversing immune abnormalities in diseases.
Collapse
Affiliation(s)
- Junjie Xu
- Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Yuening Zhao
- Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | | | - Yimin Ding
- Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Peng Xiao
- Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China; Institute of Immunology, Zhejiang University School of Medicine, Hangzhou, China; The Key Laboratory for Immunity and Inflammatory Diseases of Zhejiang Province, Hangzhou, China.
| |
Collapse
|
64
|
Raju C, Sankaranarayanan K. Insights on post-translational modifications in fatty liver and fibrosis progression. Biochim Biophys Acta Mol Basis Dis 2025; 1871:167659. [PMID: 39788217 DOI: 10.1016/j.bbadis.2025.167659] [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: 09/06/2024] [Revised: 12/20/2024] [Accepted: 01/02/2025] [Indexed: 01/12/2025]
Abstract
Metabolic dysfunction-associated steatotic liver disease [MASLD] is a pervasive multifactorial health burden. Post-translational modifications [PTMs] of amino acid residues in protein domains demonstrate pivotal roles for imparting dynamic alterations in the cellular micro milieu. The crux of identifying novel druggable targets relies on comprehensively studying the etiology of metabolic disorders. This review article presents how different chemical moieties of various PTMs like phosphorylation, methylation, ubiquitination, glutathionylation, neddylation, acetylation, SUMOylation, lactylation, crotonylation, hydroxylation, glycosylation, citrullination, S-sulfhydration and succinylation presents the cause-effect contribution towards the MASLD spectra. Additionally, the therapeutic prospects in the management of liver steatosis and hepatic fibrosis via targeting PTMs and regulatory enzymes are also encapsulated. This review seeks to understand the function of protein modifications in progression and promote the markers discovery of diagnostic, prognostic and drug targets towards MASLD management which could also halt the progression of a catalogue of related diseases.
Collapse
Affiliation(s)
- Chithra Raju
- Ion Channel Biology Laboratory, AU-KBC Research Centre, Madras Institute of Technology Campus, Anna University, Chrompet, Chennai 600 044, Tamil Nadu, India
| | - Kavitha Sankaranarayanan
- Ion Channel Biology Laboratory, AU-KBC Research Centre, Madras Institute of Technology Campus, Anna University, Chrompet, Chennai 600 044, Tamil Nadu, India.
| |
Collapse
|
65
|
Guo RR, Heijs B, Wang WJ, Wuhrer M, Liu L, Lageveen-Kammeijer GSM, Voglmeir J. Insight into distribution and composition of nonhuman N-Glycans in mammalian organs via MALDI-TOF and MALDI-MSI. Carbohydr Polym 2025; 351:123065. [PMID: 39778995 DOI: 10.1016/j.carbpol.2024.123065] [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: 09/04/2024] [Revised: 10/28/2024] [Accepted: 11/24/2024] [Indexed: 01/11/2025]
Abstract
The major hurdle of xenotransplantation is the immune response triggered by human natural antibodies interacting with carbohydrate antigens on the transplanted animal organ. Specifically, terminal glycoprotein motifs such as galactose-α1,3-galactose (α-Gal) and N-glycolylneuraminic acid (Neu5Gc) are significant obstacles. Little is known about the abundance and compositions of asparagine-linked complex carbohydrates (N-glycans) carrying these motifs in mammalian organs. By studying heart, kidney, and liver tissues from pig, cattle, and sheep, we aimed to gain insights into the abundance and spatial distribution of α-Gal- or Neu5Gc-containing N-glycans. N-glycomes were analyzed using matrix-assisted laser desorption/ionization time-of-flight mass spectrometry (MALDI-TOF-MS), MALDI-mass spectrometry imaging (MSI), and capillary electrophoresis-electrospray ionization (CE-ESI)-MS. Both α-Gal- and Neu5Gc-containing N-glycans were present in all samples, with α-Gal-modified N-glycans being the most abundant nonhuman carbohydrate motif. The abundance of N-glycans terminating with α-Gal or Neu5Gc was higher in heart and kidney samples than livers. MSI revealed kidneys had the highest glycosylation levels, and α-Gal-containing N-glycans were abundant in the kidney cortex but scarce in the medulla. This study enhances our understanding of α-Gal- and Neu5Gc-modified N-glycans in animal organs and may guide research on carbohydrate antigen-induced immune rejection in xenotransplantation.
Collapse
Affiliation(s)
- Rui-Rui Guo
- Glycomics and Glycan Bioengineering Research Center (GGBRC), College of Food Science and Technology, Nanjing Agricultural University, Nanjing 210095, China; School of Biological Engineering, Xinxiang University, Xinxiang, Henan 453000, China; Center for Proteomics and Metabolomics, Leiden University Medical Center, 2333, ZA, Leiden, the Netherlands
| | - Bram Heijs
- Center for Proteomics and Metabolomics, Leiden University Medical Center, 2333, ZA, Leiden, the Netherlands
| | - Wen-Jun Wang
- Center for Proteomics and Metabolomics, Leiden University Medical Center, 2333, ZA, Leiden, the Netherlands
| | - Manfred Wuhrer
- Center for Proteomics and Metabolomics, Leiden University Medical Center, 2333, ZA, Leiden, the Netherlands
| | - Li Liu
- Glycomics and Glycan Bioengineering Research Center (GGBRC), College of Food Science and Technology, Nanjing Agricultural University, Nanjing 210095, China.
| | - Guinevere S M Lageveen-Kammeijer
- Center for Proteomics and Metabolomics, Leiden University Medical Center, 2333, ZA, Leiden, the Netherlands; Analytical Biochemistry, Groningen Research Institute of Pharmacy, University of Groningen, 9713, AV, Groningen, the Netherlands.
| | - Josef Voglmeir
- Glycomics and Glycan Bioengineering Research Center (GGBRC), College of Food Science and Technology, Nanjing Agricultural University, Nanjing 210095, China.
| |
Collapse
|
66
|
Chernykh A, Sumer-Bayraktar Z, Lee JH, Meyer EJ, Torpy DJ, Thaysen-Andersen M. RCL glycosylation of serum corticosteroid-binding globulin: implications in cortisol delivery and septic shock. Glycobiology 2025; 35:cwaf013. [PMID: 40044123 PMCID: PMC11915215 DOI: 10.1093/glycob/cwaf013] [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: 06/03/2024] [Revised: 02/06/2025] [Accepted: 02/21/2025] [Indexed: 03/19/2025] Open
Abstract
Corticosteroid-binding globulin (CBG) is a serum glycoprotein that binds and delivers anti-inflammatory cortisol to inflammatory sites through neutrophil elastase-mediated proteolysis of an exposed reactive centre loop (RCL) on CBG. Timely and tissue-specific delivery of cortisol is critical to alleviate inflammation including in life-threatening septic shock conditions. Herein, we firstly summarise our recently published report of functional RCL O- and N-glycosylation events of serum CBG (Chernykh, J Biol Chem, 2023). A key finding of that published work was the LC-MS/MS-based discovery of RCL O-glycans at Thr342 and Thr345 of serum CBG and their inhibitory roles in neutrophil elastase-mediated RCL proteolysis. While these observations are of significance as they implicate RCL O-glycosylation as a potential regulator of cortisol delivery, the link to septic shock remains unexplored. To this end, we used a similar LC-MS/MS approach to profile the RCL O-glycosylation of CBG purified from serum of twelve septic shock patients. Serum CBG from all patients exhibited RCL O-glycosylation comprising (di)sialyl T (NeuAc1-2Gal1GalNAc1) core 1-type O-glycan structures decorating exclusively the Thr342 site. Importantly, relative to less severe cases, individuals presenting with the most severe illness displayed elevated RCL O-glycosylation upon ICU admission, suggesting a previously unknown link to septic shock severity. Overall, we have elucidated the coordinated RCL N- and O-glycosylation events of serum CBG, which improve our understanding of molecular mechanisms governing the timely and tissue-specific delivery of cortisol to inflammatory sites. This work provides clues to molecular aberrations and disease mechanisms underpinning septic shock.
Collapse
Affiliation(s)
- Anastasia Chernykh
- School of Natural Sciences, Macquarie University, 4 Wally's Walk, Macquarie Park, 2109, New South Wales, Australia
| | - Zeynep Sumer-Bayraktar
- School of Natural Sciences, Macquarie University, 4 Wally's Walk, Macquarie Park, 2109, New South Wales, Australia
| | - Jessica H Lee
- Department of Medicine, University of Adelaide, Corner of George Street and North Terrace, Adelaide, 5000, South Australia, Australia
- Endocrine and Metabolic Unit, Royal Adelaide Hospital, Port Road, Adelaide, 5000, South Australia, Australia
| | - Emily J Meyer
- Department of Medicine, University of Adelaide, Corner of George Street and North Terrace, Adelaide, 5000, South Australia, Australia
- Endocrine and Metabolic Unit, Royal Adelaide Hospital, Port Road, Adelaide, 5000, South Australia, Australia
| | - David J Torpy
- Department of Medicine, University of Adelaide, Corner of George Street and North Terrace, Adelaide, 5000, South Australia, Australia
- Endocrine and Metabolic Unit, Royal Adelaide Hospital, Port Road, Adelaide, 5000, South Australia, Australia
| | - Morten Thaysen-Andersen
- School of Natural Sciences, Macquarie University, 4 Wally's Walk, Macquarie Park, 2109, New South Wales, Australia
- Institute for Glyco-core Research (iGCORE), Nagoya University, Furocho, Chikusa Ward, Nagoya, 464-8601, Aichi, Japan
| |
Collapse
|
67
|
Kuo JCH, Colville MJ, Sorkin MR, Kuo JLK, Huang LT, Thornlow DN, Beacham GM, Hollopeter G, DeLisa MP, Alabi CA, Paszek MJ. Bio-orthogonal Glycan Imaging of Cultured Cells and Whole Animal C. elegans with Expansion Microscopy. ACS CENTRAL SCIENCE 2025; 11:193-207. [PMID: 40028367 PMCID: PMC11868961 DOI: 10.1021/acscentsci.4c01061] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 06/29/2024] [Revised: 10/22/2024] [Accepted: 10/23/2024] [Indexed: 03/05/2025]
Abstract
Complex carbohydrates called glycans play crucial roles in regulating cell and tissue physiology, but how they map to nanoscale anatomical features must still be resolved. Here, we present the first nanoscale map of mucin-type O-glycans throughout the entirety of the Caenorhabditis elegans model organism. We constructed a library of multifunctional linkers to probe and anchor metabolically labeled glycans in expansion microscopy (ExM). A flexible strategy was demonstrated for the chemical synthesis of linkers with a broad inventory of bio-orthogonal functional groups, fluorophores, anchorage chemistries, and linker arms. Employing C. elegans as a test bed, metabolically labeled O-glycans were resolved on the gut microvilli and other nanoscale anatomical features. Transmission electron microscopy images of C. elegans nanoanatomy validated the fidelity and isotropy of gel expansion. Whole organism maps of C. elegans O-glycosylation in the first larval stage revealed O-glycan "hotspots" in unexpected anatomical locations, including the body wall furrows. Beyond C. elegans, we validated ExM protocols for nanoscale imaging of metabolically labeled glycans on cultured mammalian cells. Together, our results suggest the broad applicability of the multifunctional reagents for imaging glycans and other metabolically labeled biomolecules at enhanced resolutions with ExM.
Collapse
Affiliation(s)
- Joe Chin-Hun Kuo
- Robert
Frederick Smith School of Chemical and Biomolecular Engineering, Cornell University, Ithaca, New York 14853, United States
| | - Marshall J. Colville
- Robert
Frederick Smith School of Chemical and Biomolecular Engineering, Cornell University, Ithaca, New York 14853, United States
| | - Michelle R. Sorkin
- Robert
Frederick Smith School of Chemical and Biomolecular Engineering, Cornell University, Ithaca, New York 14853, United States
| | - Jacky Lok Ka Kuo
- Robert
Frederick Smith School of Chemical and Biomolecular Engineering, Cornell University, Ithaca, New York 14853, United States
| | - Ling Ting Huang
- Robert
Frederick Smith School of Chemical and Biomolecular Engineering, Cornell University, Ithaca, New York 14853, United States
| | - Dana N. Thornlow
- Robert
Frederick Smith School of Chemical and Biomolecular Engineering, Cornell University, Ithaca, New York 14853, United States
| | - Gwendolyn M. Beacham
- Department
of Molecular Medicine, Cornell University, Ithaca, New York 14853, United States
| | - Gunther Hollopeter
- Department
of Molecular Medicine, Cornell University, Ithaca, New York 14853, United States
| | - Matthew P. DeLisa
- Robert
Frederick Smith School of Chemical and Biomolecular Engineering, Cornell University, Ithaca, New York 14853, United States
- Meinig
School of Biomedical Engineering, Cornell
University, Ithaca, New York 14853, United States
- Cornell
Institute of Biotechnology, Cornell University, Ithaca, New York 14853, United States
| | - Christopher A. Alabi
- Robert
Frederick Smith School of Chemical and Biomolecular Engineering, Cornell University, Ithaca, New York 14853, United States
| | - Matthew J. Paszek
- Robert
Frederick Smith School of Chemical and Biomolecular Engineering, Cornell University, Ithaca, New York 14853, United States
- Meinig
School of Biomedical Engineering, Cornell
University, Ithaca, New York 14853, United States
- Field
of Biophysics, Cornell University, Ithaca, New York 14853, United States
- Kavli
Institute
at Cornell for Nanoscale Science, Ithaca, New York 14853, United States
| |
Collapse
|
68
|
Sharaf-Eldin W. Malformations of Core M3 on α-Dystroglycan Are the Leading Cause of Dystroglycanopathies. J Mol Neurosci 2025; 75:28. [PMID: 39998573 PMCID: PMC11861012 DOI: 10.1007/s12031-025-02320-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2024] [Accepted: 02/07/2025] [Indexed: 02/27/2025]
Abstract
Dystroglycanopathies (DGPs) are a group of autosomal recessive neuromuscular diseases with significant clinical and genetic heterogeneity. They originate due to defects in the O-mannosyl glycosylation of α-dystroglycan (α-DG), a prominent linker between the intracellular cytoskeleton and the extracellular matrix (ECM). Fundamentally, such interactions are crucial for the integrity of muscle fibers and neuromuscular synapses, where their defects are mainly associated with muscle and brain dysfunction. To date, biallelic variants in 18 genes have been associated with DGPs, where the underlying cause is still undefined in a significant proportion of patients. Glycosylation of α-DG generates three core motifs where the core M3 is responsible for interaction with the basement membrane. Consistently, all gene defects that corrupt core M3 maturation have been identified as causes of DGPs. POMGNT1 which stimulates the generation of core M1 is also associated with DGPs, as it plays a central role in core M3 processing. Other genes involved in the glycosylation of α-DG seem unrelated to DPGs. The current review illustrates the O-mannosylation pathway of α-DG highlighting the functional properties of related genes and their contribution to the progression of DPGs. Different classes of DPGs are also elaborated characterizing the clinical features of each distinct type and phenotypes associated with each single gene. Finally, current therapeutic approaches with favorable outcomes are addressed. Potential achievements of preclinical and clinical studies would introduce effective curative therapies for this group of disorders in the near future.
Collapse
Affiliation(s)
- Wessam Sharaf-Eldin
- Medical Molecular Genetics Department, National Research Centre, Cairo, Egypt.
| |
Collapse
|
69
|
Huang HW, Zeng YF, Shivatare VS, Tseng TH, Wong CH. Cell-based glycoengineering for production of homogeneous and specific glycoform-enriched antibodies with improved effector functions. Proc Natl Acad Sci U S A 2025; 122:e2423853122. [PMID: 39969996 PMCID: PMC11874607 DOI: 10.1073/pnas.2423853122] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2024] [Accepted: 01/18/2025] [Indexed: 02/21/2025] Open
Abstract
Glycosylation of humanized antibody at Fc-Asn297 significantly affects the Fc-mediated killing of target cells through effector functions, especially antibody-dependent cellular cytotoxicity (ADCC), antibody-dependent cell-mediated phagocytosis (ADCP), and antibody-dependent vaccinal effect (ADVE). Previous studies showed that therapeutic immunoglobulin G (IgG) antibodies with α2,6-sialyl complex type (SCT) glycan attached to Fc-Asn297 exhibited optimal binding to the Fc receptors on effector cells associated with ADCC, ADCP, and ADVE. However, the production of antibodies with homogeneous Fc-SCT glycan requires multiple in vitro enzymatic and purification steps. In this study, we report two cell-based methods to produce Fc-GlcNAc antibody and Fc-SCT-enriched antibodies with improved effector functions. First, we expressed endoglycosidase S2 in Expi293F GnT1- cells to trim all N-glycans to Fc-GlcNAc antibody for in vitro transglycosylation to generate homogeneous antibodies with well-defined Fc glycan. Second, we engineered the glycosylation pathway of HEK293T cells through knock-out of undesired glycosyltransferases and knock-in of desired glycosyltransferases to produce Fc-SCT-enriched antibodies and evaluated their binding to Fc receptors, and we found that the Fc-SCT-enriched antibody is like or better than the homogeneous Fc-SCT antibody in binding to the Fc receptors associated with ADCC, ADCP, and ADVE.
Collapse
Affiliation(s)
- Han-Wen Huang
- Department of Chemistry, The Scripps Research Institute, La Jolla, CA92037
| | - Yi-Fang Zeng
- Department of Chemistry, The Scripps Research Institute, La Jolla, CA92037
| | - Vidya S. Shivatare
- Department of Chemistry, The Scripps Research Institute, La Jolla, CA92037
| | - Tzu-Hao Tseng
- Department of Chemistry, The Scripps Research Institute, La Jolla, CA92037
| | - Chi-Huey Wong
- Department of Chemistry, The Scripps Research Institute, La Jolla, CA92037
| |
Collapse
|
70
|
Gao F, Wang JH, Ma H, Xia B, Wen L, Long YT, Ying YL. Identification of Oligosaccharide Isomers Using Electrostatically Asymmetric OmpF Nanopore. Angew Chem Int Ed Engl 2025; 64:e202422118. [PMID: 39856493 DOI: 10.1002/anie.202422118] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2024] [Revised: 01/09/2025] [Accepted: 01/22/2025] [Indexed: 01/27/2025]
Abstract
Glycans, unlike uniformly charged DNA and compositionally diverse peptides, are typically uncharged and possess rich stereoisomeric diversity in the glycosidic bonds between two monosaccharide units. These unique features, including charge heterogeneity and structural complexity, pose significant challenges for accurate analysis. Herein, we developed a novel single-molecule oligosaccharide sensor, OmpF nanopore. The natural electroosmotic flow within OmpF generates a robust driving force for unlabeled neutral oligosaccharides, enabling detection at a concentration as low as 6.4 μM. Furthermore, the asymmetric constriction zone of OmpF was employed to construct a stereoselective recognition site, enabling sensitive identification of glycosidic bond differences in cell lysate samples. With the assistance of machine learning algorithms, the OmpF nanopore achieved a recognition accuracy of 99.9 % for tetrasaccharides differing in only one glycosidic bond was achieved. This nanopore sensor provides a highly sensitive analytical tool with a broad dynamic range. It enables chiral recognition of oligosaccharides at low concentrations and is suitable for analysing both low-abundance and practical samples.
Collapse
Affiliation(s)
- Fan Gao
- Molecular Sensing and Imaging Center, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing, 210023, P. R. China
| | - Jia-Hong Wang
- Molecular Sensing and Imaging Center, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing, 210023, P. R. China
| | - Hui Ma
- Molecular Sensing and Imaging Center, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing, 210023, P. R. China
| | - Bingqing Xia
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, P. R. China
- University of Chinese Academy of Sciences, Beijing, 100049, P. R. China
| | - Liuqing Wen
- Carbohydrate-Based Drug Research Center, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, P. R. China
- University of Chinese Academy of Sciences, Beijing, 100049, P. R. China
| | - Yi-Tao Long
- Molecular Sensing and Imaging Center, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing, 210023, P. R. China
| | - Yi-Lun Ying
- Molecular Sensing and Imaging Center, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing, 210023, P. R. China
- Chemistry and Biomedicine Innovation Center, Nanjing University, Nanjing, 210023, P. R. China
| |
Collapse
|
71
|
Gao J, Chen H, Yin H, Chen X, Yang Z, Wang Y, Wu J, Tian Y, Shao H, Wen L, Zhou H. Decoding Protein Glycosylation by an Integrative Mass Spectrometry-Based De Novo Sequencing Strategy. JACS AU 2025; 5:702-713. [PMID: 40017757 PMCID: PMC11863158 DOI: 10.1021/jacsau.4c00960] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/14/2024] [Revised: 12/31/2024] [Accepted: 01/03/2025] [Indexed: 03/01/2025]
Abstract
Glycoproteins, representing more than 50% of human proteins and most biopharmaceuticals, are crucial for regulating various biological processes. The complexity of multiple glycosylation sites often leads to incomplete sequence coverage and ambiguous glycan modification profiles. Here, we developed an integrative mass spectrometry-based approach for decoding unknown glycoproteins, which is featured with the combination of deglycosylation-mediated de novo sequencing with glycosylation site characterization. We utilized the enzymatic deglycosylation of N-/ O-glycans to achieve comprehensive sequence coverage. Additionally, EThcD fragmentation enables the identification of high-quality long peptides, facilitating precise protein assembly. We subsequently applied this method to de novo sequencing of the highly glycosylated therapeutic fusion protein Etanercept (Enbrel). We also sequenced three new tumor necrosis factor receptor:Fc-fusion biologics with largely unknown sequences, unveiling subtle distinctions in the primary sequences. Furthermore, we characterized N- and O-glycosylation modifications of these proteins at subunit, glycopeptide, and glycan levels. This strategy bridges the gap between the de novo sequencing and glycosylation modification, providing comprehensive information on the primary structure and glycosylation modifications for glycoproteins. Notably, our method could be a robust solution for accurate sequencing of the glycoproteins and has practical value not only in basic research but also in the biopharmaceutical industry.
Collapse
Affiliation(s)
- Jing Gao
- Analytical
Research Center for Organic and Biological Molecules, State Key Laboratory
of Drug Research, Carbohydrate-Based Drug Research Center, Shanghai
Institute of Materia Medica, Chinese Academy
of Sciences, 555 Zuchongzhi
Road, Shanghai 201203, China
| | - Hongxu Chen
- School of
Chinese Materia Medica, Nanjing University
of Chinese Medicine, 138 Xianlin Avenue, Nanjing, Jiangsu 210023, China
| | - Hongrui Yin
- NMPA Key
Laboratory for Quality Control of Therapeutic Monoclonal Antibodies, Shanghai Institute for Food and Drug Control, 1500 Zhangheng Road, Shanghai 201203, China
| | - Xin Chen
- School of
Chinese Materia Medica, Nanjing University
of Chinese Medicine, 138 Xianlin Avenue, Nanjing, Jiangsu 210023, China
| | - Zhicheng Yang
- Analytical
Research Center for Organic and Biological Molecules, State Key Laboratory
of Drug Research, Carbohydrate-Based Drug Research Center, Shanghai
Institute of Materia Medica, Chinese Academy
of Sciences, 555 Zuchongzhi
Road, Shanghai 201203, China
- University
of the Chinese Academy of Sciences, 19A Yuquan Road, Beijing 100049, China
| | - Yuqiu Wang
- Analytical
Research Center for Organic and Biological Molecules, State Key Laboratory
of Drug Research, Carbohydrate-Based Drug Research Center, Shanghai
Institute of Materia Medica, Chinese Academy
of Sciences, 555 Zuchongzhi
Road, Shanghai 201203, China
- Department
of Otolaryngology, Eye & ENT Hospital, Fudan University, Shanghai 200031, China
| | - Jianhong Wu
- Thermo
Fisher
Scientific, 2517 Jinke
Road, Shanghai 201206, China
| | - Yinping Tian
- Analytical
Research Center for Organic and Biological Molecules, State Key Laboratory
of Drug Research, Carbohydrate-Based Drug Research Center, Shanghai
Institute of Materia Medica, Chinese Academy
of Sciences, 555 Zuchongzhi
Road, Shanghai 201203, China
| | - Hong Shao
- NMPA Key
Laboratory for Quality Control of Therapeutic Monoclonal Antibodies, Shanghai Institute for Food and Drug Control, 1500 Zhangheng Road, Shanghai 201203, China
| | - Liuqing Wen
- Analytical
Research Center for Organic and Biological Molecules, State Key Laboratory
of Drug Research, Carbohydrate-Based Drug Research Center, Shanghai
Institute of Materia Medica, Chinese Academy
of Sciences, 555 Zuchongzhi
Road, Shanghai 201203, China
- School of
Chinese Materia Medica, Nanjing University
of Chinese Medicine, 138 Xianlin Avenue, Nanjing, Jiangsu 210023, China
| | - Hu Zhou
- Analytical
Research Center for Organic and Biological Molecules, State Key Laboratory
of Drug Research, Carbohydrate-Based Drug Research Center, Shanghai
Institute of Materia Medica, Chinese Academy
of Sciences, 555 Zuchongzhi
Road, Shanghai 201203, China
- School of
Chinese Materia Medica, Nanjing University
of Chinese Medicine, 138 Xianlin Avenue, Nanjing, Jiangsu 210023, China
- University
of the Chinese Academy of Sciences, 19A Yuquan Road, Beijing 100049, China
- School of
Pharmaceutical Science and Technology, Hangzhou Institute for Advanced
Study, University of Chinese Academy of
Sciences, Hangzhou 310024, China
| |
Collapse
|
72
|
Svecla M, Li-Gao R, Falck D, Bonacina F. N-glycosylation signature and its relevance in cardiovascular immunometabolism. Vascul Pharmacol 2025; 159:107474. [PMID: 39988310 DOI: 10.1016/j.vph.2025.107474] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2024] [Revised: 01/21/2025] [Accepted: 02/20/2025] [Indexed: 02/25/2025]
Abstract
Glycosylation is a post-translational modification in which complex, branched carbohydrates (glycans) are covalently attached to proteins or lipids. Asparagine-link protein (N-) glycosylation is among the most common types of glycosylation. This process is essential for many biological and cellular functions, and impaired N-glycosylation has been widely implicated in inflammation and cardiovascular diseases. Different technical approaches have been used to increase the coverage of the N-glycome, revealing a high level of complexity of glycans, regarding their structure and attachment site on a protein. In this context, new insights from genomic studies have revealed a genetic regulation of glycosylation, linking genetic variants to total plasma N-glycosylation and N-glycosylation of immunoglobulin G (IgG). In addition, RNAseq approaches have revealed a degree of transcriptional regulation for the glycoenzymes involved in glycan structure. However, our understanding of the association between cardiovascular risk and glycosylation, determined by a complex overlay of genetic and environmental factors, remains limited. Mostly, plasma N-glycosylation profiling in different human cohorts or experimental investigations of specific enzyme functions in models of atherosclerosis have been reported. Most of the uncovered glycosylation associations with pathological mechanisms revolve around the recruitment of inflammatory cells to the vessel wall and lipoprotein metabolism. This review aims to summarise insights from omics studies into the immune and metabolic regulation of N-glycosylation and its association with cardiovascular and metabolic disease risk and to provide mechanistic insights from experimental models. The combination of emerging techniques for glycomics and glycoproteomics with already achieved omics approaches to map the transcriptomic, epigenomic, and metabolomic profile at single-cell resolution will deepen our understanding of the molecular regulation of glycosylation as well as identify novel biomarkers and targets for cardiovascular disease prevention and treatment.
Collapse
Affiliation(s)
- Monika Svecla
- Department of Neurosurgery, Charité Universitätsmedizin Berlin, Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Ruifang Li-Gao
- Department of Clinical Epidemiology, Leiden University Medical Center, Leiden, the Netherlands
| | - David Falck
- Leiden University Medical Center, Center for Proteomics and Metabolomics, Glycomics Group, Leiden, the Netherlands
| | - Fabrizia Bonacina
- Department of Excellence of Pharmacological and Biomolecular Sciences "Rodolfo Paoletti", University of Milan, Milan, Italy.
| |
Collapse
|
73
|
Gill A, Kinghorn K, Bautch VL, Mac Gabhann F. Mechanistic computational modeling of sFLT1 secretion dynamics. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.02.12.637983. [PMID: 40027776 PMCID: PMC11870409 DOI: 10.1101/2025.02.12.637983] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/05/2025]
Abstract
Constitutively secreted by endothelial cells, soluble FLT1 (sFLT1 or sVEGFR1) binds and sequesters extracellular vascular endothelial growth factors (VEGF), thereby reducing VEGF binding to VEGF receptor tyrosine kinases and their downstream signaling. In doing so, sFLT1 plays an important role in vascular development and in the patterning of new blood vessels in angiogenesis. Here, we develop multiple mechanistic models of sFLT1 secretion and identify a minimal mechanistic model that recapitulates key qualitative and quantitative features of temporal experimental datasets of sFLT1 secretion from multiple studies. We show that the experimental data on sFLT1 secretion is best represented by a delay differential equation (DDE) system including a maturation term, reflecting the time required between synthesis and secretion. Using optimization to identify appropriate values for the key mechanistic parameters in the model, we show that two model parameters (extracellular degradation rate constant and maturation time) are very strongly constrained by the experimental data, and that the remaining parameters are related by two strongly constrained constants. Thus, only one degree of freedom remains, and measurements of the intracellular levels of sFLT1 would fix the remaining parameters. Comparison between simulation predictions and additional experimental data of the outcomes of chemical inhibitors and genetic perturbations suggest that intermediate values of the secretion rate constant best match the simulation with experiments, which would completely constrain the model. However, some of the inhibitors tested produce results that cannot be reproduced by the model simulations, suggesting that additional mechanisms not included here are required to explain those inhibitors. Overall, the model reproduces most available experimental data and suggests targets for further quantitative investigation of the sFLT1 system.
Collapse
Affiliation(s)
- Amy Gill
- Institute for Computational Medicine, Johns Hopkins University, Baltimore, MD, USA
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, USA
| | - Karina Kinghorn
- Department of Biology, University of North Carolina, Chapel Hill, NC, USA
- Curriculum in Cell Biology and Physiology, University of North Carolina, Chapel Hill, NC, USA
| | - Victoria L Bautch
- Department of Biology, University of North Carolina, Chapel Hill, NC, USA
- Curriculum in Cell Biology and Physiology, University of North Carolina, Chapel Hill, NC, USA
- McAllister Heart Institute, University of North Carolina, Chapel Hill, NC, USA
| | - Feilim Mac Gabhann
- Institute for Computational Medicine, Johns Hopkins University, Baltimore, MD, USA
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, USA
| |
Collapse
|
74
|
Das R, Mukhopadhyay B. The effect of neighbouring group participation and possible long range remote group participation in O-glycosylation. Beilstein J Org Chem 2025; 21:369-406. [PMID: 39996165 PMCID: PMC11849559 DOI: 10.3762/bjoc.21.27] [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: 07/07/2024] [Accepted: 01/27/2025] [Indexed: 02/26/2025] Open
Abstract
Stereoselective glycosylations are one of the most challenging tasks of synthetic glycochemists. The protecting building blocks on the glycosides contribute significantly in attaining the required stereochemistry of the resulting glycosides. Strategic installation of suitable protecting groups in the C-2 position, vicinal to the anomeric carbon, renders neighbouring group participation, whereas protecting groups in the distal C-3, C-4, and C-6 positions are often claimed to exhibit remote group participation with the anomeric carbon. Neighbouring group participation and remote group participation are being widely studied to help the glycochemists design the synthetic protocols for multistep synthesis of complex oligosaccharides and in turn, standardise the process of the glycosylation towards a particular stereochemical output. While neighbouring group participation has been quite effective in achieving the required stereochemistry of the produced glycosides, remote participation exhibits comparatively less efficacy in achieving complete stereoselectivity in the glycosylation reactions. Remote participation is a still highly debated topic in the scientific community. However, implementing the participating role of the remote groups in glycosylation reactions is widely practised to achieve better stereocontrol and to facilitate the formation of synthetically challenging glycosidic linkages.
Collapse
Affiliation(s)
- Rituparna Das
- SWEET Lab, Department of Chemical Sciences, Indian Institute of Science Education and Research (IISER) Kolkata, Mohanpur, Nadia 741246, India
| | - Balaram Mukhopadhyay
- SWEET Lab, Department of Chemical Sciences, Indian Institute of Science Education and Research (IISER) Kolkata, Mohanpur, Nadia 741246, India
| |
Collapse
|
75
|
Zhang RY, He X, Chen JJ, Wu CM, Lin YL, Wang YB, Wang Q, Zheng L, Hu XM. UGP2, a novel target gene of TP53, inhibits endothelial cells apoptosis and atherosclerosis. Life Sci 2025; 363:123393. [PMID: 39818283 DOI: 10.1016/j.lfs.2025.123393] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2024] [Revised: 12/31/2024] [Accepted: 01/10/2025] [Indexed: 01/18/2025]
Abstract
The dysfunction of the endothelial lining in lesion-prone areas of the arterial vasculature significantly contributes to the pathobiology of atherosclerotic cardiovascular disease. Recent studies suggested that UDP-glucose pyrophosphorylase 2 (UGP2) plays a role in cell proliferation and survival. This study investigates the anti-apoptotic and anti-atherogenic effects of UGP2 both in vitro and in vivo. We explored the effects and mechanisms of UGP2 on apoptosis in endothelial cells using flow cytometry and Western blot analysis. Additionally, we evaluate apoptosis levels in atherosclerotic lesions with ldlr-/- ugp2+/- mice. Microarray analysis revealed reduced UGP2 expression in human atherosclerotic plaques. In vitro experiments demonstrated that TP53 interacts with the promoter region of the UGP2 gene, upregulating UGP2 expression. Enhanced UGP2 expression led to decreased reactive oxygen species (ROS) levels, reduced Cleaved caspase-3 expression, and lower apoptosis levels in endothelial cells. The anti-apoptotic effects of UGP2 were significantly diminished by H2O2. In vivo, UGP2 deficiency in ldlr-/- mice fed a Western high-fat diet promoted atherosclerosis, increased ROS levels, and elevated Cleaved caspase-3 expression and apoptosis in atherosclerotic lesions. Our findings identify UGP2 as a novel TP53 target gene that contributes to anti-apoptotic effects by regulating ROS homeostasis via a non-canonical pathway. UGP2 represents a potential therapeutic target for ameliorating atherosclerosis-related diseases.
Collapse
Affiliation(s)
- Ru-Yi Zhang
- Department of Laboratory Medicine, Guangdong Provincial Key Laboratory of Precision Medical Diagnostics, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Xin He
- Division of Nephrology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Juan-Jiang Chen
- Laboratory Medicine Center, Zhujiang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Chang-Meng Wu
- Department of Laboratory Medicine, Peking University Shenzhen Hospital, Shenzhen, 518036, China
| | - Yu-Ling Lin
- Laboratory Medicine Center, Zhujiang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Yu-Bing Wang
- Laboratory Medicine Center, Zhujiang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Qian Wang
- Department of Laboratory Medicine, Guangdong Provincial Key Laboratory of Precision Medical Diagnostics, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China; Laboratory Medicine Center, Zhujiang Hospital, Southern Medical University, Guangzhou, 510515, China.
| | - Lei Zheng
- Department of Laboratory Medicine, Guangdong Provincial Key Laboratory of Precision Medical Diagnostics, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China.
| | - Xiu-Mei Hu
- Department of Laboratory Medicine, Guangdong Provincial Key Laboratory of Precision Medical Diagnostics, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China.
| |
Collapse
|
76
|
Zhang P, Wang L, Liu H, Lin S, Guo D. Unveiling the crucial role of glycosylation modification in lung adenocarcinoma metastasis through artificial neural network-based spatial multi-omics single-cell analysis and Mendelian randomization. BMC Cancer 2025; 25:249. [PMID: 39948531 PMCID: PMC11823056 DOI: 10.1186/s12885-025-13650-x] [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: 04/10/2024] [Accepted: 02/05/2025] [Indexed: 02/17/2025] Open
Abstract
BACKGROUND Investigations into the intricacies of glycosylation modifications, a prevalent post-translational alteration observed in neoplasms, especially remain elusive in the context of lung adenocarcinoma. Through the integration of multiple omics approaches, the investigation aimed to delineate the significance of glycosylation in lung adenocarcinoma, with an objective to pinpoint viable biological targets. METHODS Initial steps involved the identification of genes differentially expressed in relation to glycosylation at the aggregate transcriptome level within lung adenocarcinoma tissues. This was followed by analyses of localization and function employing both single-cell and spatial transcriptomics to provide a more nuanced understanding. In pursuit of elucidating functional disparities in glycosylation patterns, a predictive framework employing artificial neural networks was constructed. To ascertain causal relationships between specific genes and lung adenocarcinoma, Mendelian randomization was applied, culminating in the experimental validation of these genes' roles. RESULTS Analysis at the single-cell level uncovered marked glycosylation modification expressions in metastatic tissues of lung adenocarcinoma. Moreover, tissues of lung adenocarcinoma with elevated expression of genes associated with glycosylation displayed enhanced differentiation and activation across signaling pathways including TGF-β, oxidative stress, and WNT. Through spatial transcriptomics, zones of intense glycosylation modification were pinpointed within tumor nests and proximate to tumor-associated blood vessels. An artificial neural network-derived prognostic model demonstrated outstanding predictive capability, with AUC scores achieving 0.84, 0.83, and 0.89 for 1, 3, and 5-year forecasts, respectively. The group identified as high-risk was characterized by pronounced immunosuppression and diminished responsiveness to immunotherapy. Mendelian randomization analysis pinpointed GLANT2 (OR = 1.3654, p < 0.05) and GYS1 (OR = 1.2668, p < 0.05) as genes contributing to the pathogenesis of lung adenocarcinoma. Cell assays have reaffirmed that the inhibition of GYS1 significantly reduces proliferation and invasion in lung adenocarcinoma cell lines, while also decreasing glycogen storage and the formation of glycosylation end products, indicating suppression of glycosylation processes. These findings identify GYS1 as a prospective glycosylation-linked biological target for lung adenocarcinoma therapy.
Collapse
Affiliation(s)
- Penngcheng Zhang
- Department of General Surgery, The Third Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
- The First Affiliated Hospital of Zhejiang Chinese Medical University (Zheiiang Provincial Hospital of Chinese Medicine), Hangzhou, Zhejiang, China
| | - Lexin Wang
- General Hospital of Ningxia Medical University, Yinchuan, Ningxia, China
- Western Institute of Digital-Intelligent Medicine, Chongqing, China
| | - Hanwen Liu
- Department of General Surgery, The Third Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
| | - Shengyou Lin
- The First Affiliated Hospital of Zhejiang Chinese Medical University (Zheiiang Provincial Hospital of Chinese Medicine), Hangzhou, Zhejiang, China.
| | - Dechao Guo
- Department of General Surgery, The Third Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China.
| |
Collapse
|
77
|
Bourganou MV, Chondrogianni ME, Kyrou I, Flessa CM, Chatzigeorgiou A, Oikonomou E, Lambadiari V, Randeva HS, Kassi E. Unraveling Metabolic Dysfunction-Associated Steatotic Liver Disease Through the Use of Omics Technologies. Int J Mol Sci 2025; 26:1589. [PMID: 40004054 PMCID: PMC11855544 DOI: 10.3390/ijms26041589] [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: 01/05/2025] [Revised: 02/07/2025] [Accepted: 02/10/2025] [Indexed: 02/27/2025] Open
Abstract
Non-alcoholic fatty liver disease (NAFLD), now referred to as metabolic dysfunction-associated steatotic liver disease (MASLD), is the most prevalent liver disorder globally, linked to obesity, type 2 diabetes, and cardiovascular risk. Understanding its potential progression from simple steatosis to cirrhosis and hepatocellular carcinoma (HCC) is crucial for patient management and treatment strategies. The disease's complexity requires innovative approaches for early detection and personalized care. Omics technologies-such as genomics, transcriptomics, proteomics, metabolomics, and exposomics-are revolutionizing the study of MASLD. These high-throughput techniques allow for a deeper exploration of the molecular mechanisms driving disease progression. Genomics can identify genetic predispositions, whilst transcriptomics and proteomics reveal changes in gene expression and protein profiles during disease evolution. Metabolomics offers insights into the metabolic alterations associated with MASLD, while exposomics links environmental exposures to MASLD progression and pathology. By integrating data from various omics platforms, researchers can map out the intricate biochemical pathways involved in liver disease progression. This review discusses the roles of omics technologies in enhancing the understanding of disease progression and highlights potential diagnostic and therapeutic targets within the MASLD spectrum, emphasizing the need for non-invasive tools in disease staging and treatment development.
Collapse
Affiliation(s)
- Maria V. Bourganou
- Department of Biological Chemistry, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece; (M.V.B.); (M.E.C.); (C.-M.F.)
| | - Maria Eleni Chondrogianni
- Department of Biological Chemistry, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece; (M.V.B.); (M.E.C.); (C.-M.F.)
- Endocrine Unit, 1st Department of Propaedeutic Internal Medicine, Laiko Hospital, National and Kapodistrian University of Athens, 11527 Athens, Greece
| | - Ioannis Kyrou
- Laboratory of Dietetics and Quality of Life, Department of Food Science and Human Nutrition, School of Food and Nutritional Sciences, Agricultural University of Athens, 11855 Athens, Greece
- Warwickshire Institute for the Study of Diabetes, Endocrinology and Metabolism (WISDEM), University Hospitals Coventry and Warwickshire NHS Trust, Coventry CV2 2DX, UK
- Institute for Cardiometabolic Medicine, University Hospitals Coventry and Warwickshire NHS Trust, Coventry CV2 2DX, UK
- Warwick Medical School, University of Warwick, Coventry CV4 7AL, UK
- Centre for Health & Life Sciences, Coventry University, Coventry CV1 5FB, UK
- Aston Medical School, College of Health and Life Sciences, Aston University, Birmingham B4 7ET, UK
- College of Health, Psychology and Social Care, University of Derby, Derby DE22 IGB, UK
| | - Christina-Maria Flessa
- Department of Biological Chemistry, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece; (M.V.B.); (M.E.C.); (C.-M.F.)
| | - Antonios Chatzigeorgiou
- Department of Physiology, Medical School, National and Kapodistrian University of Athens, 75 Mikras Asias Str., 11527 Athens, Greece;
| | - Evangelos Oikonomou
- 3rd Department of Cardiology, “Sotiria” Thoracic Diseases Hospital of Athens, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece;
| | - Vaia Lambadiari
- 2nd Department of Internal-Medicine, Diabetes Centre, Attikon University Hospital, Medical School, National and Kapodistrian University of Athens, 12462 Athens, Greece;
| | - Harpal S. Randeva
- Warwickshire Institute for the Study of Diabetes, Endocrinology and Metabolism (WISDEM), University Hospitals Coventry and Warwickshire NHS Trust, Coventry CV2 2DX, UK
- Institute for Cardiometabolic Medicine, University Hospitals Coventry and Warwickshire NHS Trust, Coventry CV2 2DX, UK
- Warwick Medical School, University of Warwick, Coventry CV4 7AL, UK
- Centre for Health & Life Sciences, Coventry University, Coventry CV1 5FB, UK
| | - Eva Kassi
- Department of Biological Chemistry, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece; (M.V.B.); (M.E.C.); (C.-M.F.)
- Endocrine Unit, 1st Department of Propaedeutic Internal Medicine, Laiko Hospital, National and Kapodistrian University of Athens, 11527 Athens, Greece
| |
Collapse
|
78
|
Felixberger PT, Andrieux G, Maul-Pavicic A, Goldacker S, Harder I, Gutenberger S, Landry JJM, Benes V, Jakob TF, Boerries M, Nitschke L, Voll RE, Warnatz K, Keller B. CD21 low B cells reveal a unique glycosylation pattern with hypersialylation and hyperfucosylation. Front Immunol 2025; 16:1512279. [PMID: 40013136 PMCID: PMC11861550 DOI: 10.3389/fimmu.2025.1512279] [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: 10/16/2024] [Accepted: 01/10/2025] [Indexed: 02/28/2025] Open
Abstract
Background The posttranslational modification of cellular macromolecules by glycosylation is considered to contribute to disease pathogenesis in autoimmune and inflammatory conditions. In a subgroup of patients with common variable immunodeficiency (CVID), the occurrence of such complications is associated with an expansion of naïve-like CD21low B cells during a chronic type 1 immune activation. The glycosylation pattern of B cells in CVID patients has not been addressed to date. Objective The objective of this study was to examine the surface glycome of B cells in patients with CVID and associated immune dysregulation. Methods We performed surface lectin staining on B cells from peripheral blood and tonsils, both ex vivo and after in vitro stimulation. Additionally, we examined the expression of glycosylation-related genes by RNAseq in naïve-like CD21low B cells ex vivo, as well as in naïve CD21pos B cells from healthy controls after in vitro stimulation. Results Unlike CD21pos B cells, naïve-like CD21low B cells from CVID patients and CD21low B cells from healthy controls exhibited a unique glycosylation pattern with high levels of α2,6 sialic acids and fucose. This hypersialylation and hyperfucosylation were particularly induced by activation with anti-IgM and interferon-γ (IFN-γ). Transcriptome analysis suggested that naïve-like CD21low B cells possess a comprehensively reorganised glycosylation machinery, with anti-IgM/IFN-γ having the potential to initiate these changes in vitro. Conclusion CD21low B cells are hypersialylated and hyperfucosylated. This may implicate altered lectin-ligand interactions on the cell surface potentially affecting the CD21low B-cell function. These glycome changes appear to be driven by the prominent type I immune response in complicated CVID patients. A better understanding of how altered glycosylation influences immune cell function could lead to new therapeutic strategies.
Collapse
Affiliation(s)
- Peter Tobias Felixberger
- Department of Rheumatology and Clinical Immunology, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
- Center for Chronic Immunodeficiency (CCI), Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Geoffroy Andrieux
- Institute of Medical Bioinformatics and Systems Medicine, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Andrea Maul-Pavicic
- Department of Rheumatology and Clinical Immunology, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
- Center for Chronic Immunodeficiency (CCI), Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Sigune Goldacker
- Department of Rheumatology and Clinical Immunology, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
- Center for Chronic Immunodeficiency (CCI), Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Ina Harder
- Department of Rheumatology and Clinical Immunology, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
- Center for Chronic Immunodeficiency (CCI), Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Sylvia Gutenberger
- Department of Rheumatology and Clinical Immunology, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
- Center for Chronic Immunodeficiency (CCI), Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | | | - Vladimir Benes
- Genomics Core Facility, European Molecular Biology Laboratory, Heidelberg, Germany
| | - Till Fabian Jakob
- Department of Oto-Rhino-Laryngology, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Melanie Boerries
- Institute of Medical Bioinformatics and Systems Medicine, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
- German Cancer Consortium (DKTK), Partner site Freiburg, a partnership between DKFZ and Medical Center - University of Freiburg, Freiburg, Germany
| | - Lars Nitschke
- Division of Genetics, Department of Biology, University of Erlangen, Erlangen, Germany
| | - Reinhard Edmund Voll
- Department of Rheumatology and Clinical Immunology, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
- Center for Chronic Immunodeficiency (CCI), Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Klaus Warnatz
- Department of Rheumatology and Clinical Immunology, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
- Center for Chronic Immunodeficiency (CCI), Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Baerbel Keller
- Department of Rheumatology and Clinical Immunology, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
- Center for Chronic Immunodeficiency (CCI), Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| |
Collapse
|
79
|
Lam YY, Tan A, Kempe K, Boyd BJ. Metabolic glycan labelling with bio-orthogonal targeting and its potential in drug delivery. J Control Release 2025; 378:880-898. [PMID: 39694071 DOI: 10.1016/j.jconrel.2024.12.018] [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: 07/21/2024] [Revised: 11/21/2024] [Accepted: 12/09/2024] [Indexed: 12/20/2024]
Abstract
New modes of targeted drug delivery are emerging with promise of enhancing therapeutic efficacy while reducing side effects. This review examines the landscape of metabolic glycan labelling-a technique gaining traction for its potential in specific drug targeting. By exploiting the natural glycan synthetic pathway of monosaccharides, unnatural sugar analogues are incorporated into glycoproteins, allowing for the presentation of unique functional groups on cells. This enables specific targeting using 'clickable' probes with complementary click chemistry functional groups. The selection of sugar analogues and chemical tags are crucial components explored in this review, alongside considerations for cell lines, tissues, and cargo selection. The review discusses non-therapeutic and therapeutic applications of metabolic glycan labelling, as well as its potential beyond labelling of cell surfaces. The review also highlights underexplored areas of metabolic glycan labelling by assessing the limited literature addressing labelling efficiency, turnover rates, the impact of sugar supplements in cell culture, and the critical cell to functionalised sugar ratio. Furthermore, this review delves into the future landscape and goals of metabolic glycan labelling, envisioning its potential in targeted drug delivery.
Collapse
Affiliation(s)
- Yuen Yi Lam
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University (Parkville Campus), 381 Royal Parade, Parkville, VIC 3052, Australia
| | - Angel Tan
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University (Parkville Campus), 381 Royal Parade, Parkville, VIC 3052, Australia
| | - Kristian Kempe
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University (Parkville Campus), 381 Royal Parade, Parkville, VIC 3052, Australia; Materials Science and Engineering, Monash University, Clayton, VIC 3800, Australia
| | - Ben J Boyd
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University (Parkville Campus), 381 Royal Parade, Parkville, VIC 3052, Australia; Materials Science and Engineering, Monash University, Clayton, VIC 3800, Australia; Department of Pharmacy, University of Copenhagen Universitetsparken 2, 2100 Copenhagen, Denmark.
| |
Collapse
|
80
|
Liang S, Tang Q, Guo X, Li Z, Guo Y, Chang J, Cheng B, Song Q, Sun J, Dai P, Chen X. Mutant glycosidases for labeling sialoglycans with high specificity and affinity. Nat Commun 2025; 16:1427. [PMID: 39915445 PMCID: PMC11802738 DOI: 10.1038/s41467-025-56629-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2024] [Accepted: 01/22/2025] [Indexed: 02/09/2025] Open
Abstract
Affinity labeling of biomacromolecules is vital for bioimaging and functional studies. However, affinity probes recognizing glycans with high specificity remain scarce. Here we report the development of glycan recombinant affinity binders (GRABs) based on mutant bacterial sialidases, which are enzymatically inactive but preserve stringent specificity for sialoglycan substrates. By mutating a key catalytic residue of Streptococcus pneumoniae neuraminidase A (SpNanA) and Ruminococcus gnavus neuraminidase H (RgNanH), we develop GRAB-Sia and GRAB-Sia3 recognizing total sialoglycans and α2,3-sialosides, respectively. The GRABs exhibit strict substrate and linkage specificity, and tetramerization with streptavidin substantially increases their avidity. The GRABs and tetrameric GRABs (tetra-GRABs) are effective tools for probing sialoglycans in immunoblotting, flow cytometry, immunoprecipitation, and fluorescence imaging. Furthermore, multiplex analysis with tetra-GRABs uncovers spatially distinct sialoglycans in the various mouse organs. This work provides a versatile toolkit for labeling and analyzing sialoglycans with high specificity, sensitivity, and convenience.
Collapse
Affiliation(s)
- Shuyu Liang
- College of Chemistry and Molecular Engineering, Peking University, Beijing, China
- Beijing National Laboratory for Molecular Sciences, Peking University, Beijing, China
| | - Qi Tang
- College of Chemistry and Molecular Engineering, Peking University, Beijing, China
- Beijing National Laboratory for Molecular Sciences, Peking University, Beijing, China
| | - Xunzi Guo
- College of Chemistry and Molecular Engineering, Peking University, Beijing, China
- Beijing National Laboratory for Molecular Sciences, Peking University, Beijing, China
| | - Zi'an Li
- College of Chemistry and Molecular Engineering, Peking University, Beijing, China
- Beijing National Laboratory for Molecular Sciences, Peking University, Beijing, China
| | - Yilan Guo
- College of Chemistry and Molecular Engineering, Peking University, Beijing, China
- Beijing National Laboratory for Molecular Sciences, Peking University, Beijing, China
| | - Jinghan Chang
- College of Chemistry and Molecular Engineering, Peking University, Beijing, China
- Beijing National Laboratory for Molecular Sciences, Peking University, Beijing, China
| | - Bo Cheng
- College of Chemistry and Molecular Engineering, Peking University, Beijing, China
- Beijing National Laboratory for Molecular Sciences, Peking University, Beijing, China
| | - Qitao Song
- College of Chemistry and Molecular Engineering, Peking University, Beijing, China
- Peking-Tsinghua Center for Life Sciences, Peking University, Beijing, China
| | - Jiayu Sun
- College of Chemistry and Molecular Engineering, Peking University, Beijing, China
- Beijing National Laboratory for Molecular Sciences, Peking University, Beijing, China
| | - Peng Dai
- College of Chemistry and Molecular Engineering, Peking University, Beijing, China.
- Beijing National Laboratory for Molecular Sciences, Peking University, Beijing, China.
- Synthetic and Functional Biomolecules Center, Peking University, Beijing, China.
- Key Laboratory of Bioorganic Chemistry and Molecular Engineering of Ministry of Education, Peking University, Beijing, China.
| | - Xing Chen
- College of Chemistry and Molecular Engineering, Peking University, Beijing, China.
- Beijing National Laboratory for Molecular Sciences, Peking University, Beijing, China.
- Peking-Tsinghua Center for Life Sciences, Peking University, Beijing, China.
- Synthetic and Functional Biomolecules Center, Peking University, Beijing, China.
- Key Laboratory of Bioorganic Chemistry and Molecular Engineering of Ministry of Education, Peking University, Beijing, China.
| |
Collapse
|
81
|
Ferreira BH, Silva IS, Mendes A, Leite-Pinheiro F, Paton AW, Paton JC, Duarte IF, Pierre P, Almeida CR. Promoting ER stress in a plasmacytoid dendritic cell line drives fibroblast activation. Cell Commun Signal 2025; 23:66. [PMID: 39920698 PMCID: PMC11804055 DOI: 10.1186/s12964-025-02057-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2024] [Accepted: 01/18/2025] [Indexed: 02/09/2025] Open
Abstract
BACKGROUND Fibrosis remains a major complication in several chronic diseases, including systemic sclerosis (SSc). Plasmacytoid dendritic cells (pDCs) are innate immune cells that play a key role in the development of fibrosis in SSc patients, through still poorly defined mechanisms. Interestingly, endoplasmic reticulum (ER) stress signaling pathways are dysregulated in pDCs from patients with SSc, but their contribution to fibrosis remains unclear. Thus, this study aimed to unravel the mechanisms behind the involvement of pDCs and ER stress in fibrosis. METHODS To address this question, we established an in vitro model designed to study the interactions between pDCs and fibroblasts. More specifically, IMR-90 fibroblasts were co-cultured with CAL-1, a pDC cell line. ER stress was then induced by the bacterial toxin SubAB. Extracellular matrix (ECM) production was assessed using immunoblotting, qPCR and confocal microscopy. The importance of cell-to-cell contact was investigated using conditioned media (CM) and transwell assays. RESULTS Direct contact of CAL-1 and IMR-90 cells under ER stress conditions led to increased expression of fibronectin and alpha-smooth muscle actin (α-SMA). This effect required expression of the ER stress signaling sensor protein kinase R-like ER kinase (PERK) in pDCs and was observed only upon direct contact between both cell types. CONCLUSIONS Overall, our data suggest that ER stress induction in pDCs promotes fibroblast activation, which may contribute to the development of fibrosis in SSc.
Collapse
Affiliation(s)
- Beatriz H Ferreira
- Institute of Biomedicine (iBiMED), Department of Medical Sciences, University of Aveiro, Aveiro, Portugal
- CICECO - Aveiro Institute of Materials, Department of Chemistry, University of Aveiro, Aveiro, Portugal
| | - Inês S Silva
- Institute of Biomedicine (iBiMED), Department of Medical Sciences, University of Aveiro, Aveiro, Portugal
| | - Andreia Mendes
- Institute of Biomedicine (iBiMED), Department of Medical Sciences, University of Aveiro, Aveiro, Portugal
| | - Fátima Leite-Pinheiro
- Institute of Biomedicine (iBiMED), Department of Medical Sciences, University of Aveiro, Aveiro, Portugal
| | - Adrienne W Paton
- Department of Molecular and Biomedical Science, Research Centre for Infectious Diseases, University of Adelaide, Adelaide, Australia
| | - James C Paton
- Department of Molecular and Biomedical Science, Research Centre for Infectious Diseases, University of Adelaide, Adelaide, Australia
| | - Iola F Duarte
- CICECO - Aveiro Institute of Materials, Department of Chemistry, University of Aveiro, Aveiro, Portugal
- LAQV-REQUIMTE, Department of Chemistry, University of Aveiro, Aveiro, Portugal
| | - Philippe Pierre
- Institute of Biomedicine (iBiMED), Department of Medical Sciences, University of Aveiro, Aveiro, Portugal
- Centre d'Immunologie de Marseille-Luminy, Aix Marseille Université, CNRS, INSERM, Marseille Cedex 9, 13288, France
- Shanghai Institute of Immunology, Department of Microbiology and Immunology, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Catarina R Almeida
- Institute of Biomedicine (iBiMED), Department of Medical Sciences, University of Aveiro, Aveiro, Portugal.
| |
Collapse
|
82
|
Videcrantz Faurschou N, Sauer SPA, Pedersen CM. Unraveling the Mechanism of Stereospecific Self-Promoted N-Glycosylations. Chemistry 2025; 31:e202403909. [PMID: 39601674 DOI: 10.1002/chem.202403909] [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: 10/22/2024] [Revised: 11/15/2024] [Accepted: 11/27/2024] [Indexed: 11/29/2024]
Abstract
In this study, the mechanism of self-promoted N-glycosylations is extensively investigated through kinetic experiments, computational studies, and nucleophilic competition experiments. Based on the findings, the mechanism is proposed to be initiated by proton transfer from the acidic sulfonyl carbamate to the trichloroacetimidate, upon formation of an associated contact ion pair. This ion pair then collapses in an SN1-like fashion, with formation of an oxocarbeniumion-like intermediate. According to the proposed mechanism, stereospecificity arises from the associated nature of all intermediates formed throughout the reaction. During the mechanistic study, it was also found that the sulfonyl carbamates have catalytic properties if a competing nucleophile is present.
Collapse
Affiliation(s)
| | - Stephan P A Sauer
- Department of Chemistry, University of Copenhagen, Universitetsparken 5, 2100, Copenhagen, Denmark
| | | |
Collapse
|
83
|
Suzuki S, Itoh M. Synergistic effects of mutation and glycosylation on disease progression. Front Mol Biosci 2025; 12:1550815. [PMID: 39967653 PMCID: PMC11832388 DOI: 10.3389/fmolb.2025.1550815] [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: 12/24/2024] [Accepted: 01/20/2025] [Indexed: 02/20/2025] Open
Abstract
Glycosylation, a post-translational modification, plays a crucial role in proper localization and function of proteins. It is regulated by multiple glycosyltransferases and can be influenced by various factors. Inherited missense mutations in glycosylated proteins such as NOTCH3, Low-density lipoprotein receptor (LDLR), and Amyloid precursor protein (APP) could affect their glycosylation states, leading to cerebral small vessel disease, hypercholesterolemia, and Alzheimer's disease, respectively. Additionally, physiological states and aging-related conditions can affect the expression levels of glycosyltransferases. However, the interplay between mutations in glycosylated proteins and changes in their glycosylation levels remains poorly understood. This mini-review summarizes the effects of glycosylation on transmembrane proteins with pathogenic mutations, including NOTCH3, LDLR, and APP. We highlight the synergistic contributions of missense amino acids in the mutant proteins and alterations in their glycosylation states to their molecular pathogenesis.
Collapse
Affiliation(s)
- Shodai Suzuki
- Department of Biochemistry, Graduate School of Pharmaceutical Sciences, Chiba University, Chiba, Japan
| | - Motoyuki Itoh
- Department of Biochemistry, Graduate School of Pharmaceutical Sciences, Chiba University, Chiba, Japan
- Research Institute of Disaster Medicine, Chiba University, Chiba, Japan
- Health and Disease Omics Center, Chiba University, Chiba, Japan
| |
Collapse
|
84
|
Dangerfield EM, Foster AJ, Lynch AT, Stocker BL, Timmer MSM. The Synthesis of Glycosyl Amides from Unprotected Carbohydrates and Acylsilanes. Org Lett 2025. [PMID: 39902732 DOI: 10.1021/acs.orglett.4c04666] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2025]
Abstract
We report on a new methodology for the stereoselective synthesis of β-anomeric N-glycosyl amides from unprotected carbohydrates via the reaction of in situ generated carbohydrate oximes with acylsilanes. By variation of the acylsilane, N-glycosyl amides with azide and chloride conjugation handles are prepared in excellent yield and β-selectivity from unprotected sugars.
Collapse
Affiliation(s)
- Emma M Dangerfield
- School of Chemical and Physical Sciences, Victoria University of Wellington, PO Box 600, 6140, Wellington, New Zealand
- Centre for Biodiscovery, Victoria University of Wellington, PO Box 600, 6140, Wellington, New Zealand
| | - Amy J Foster
- School of Chemical and Physical Sciences, Victoria University of Wellington, PO Box 600, 6140, Wellington, New Zealand
- Centre for Biodiscovery, Victoria University of Wellington, PO Box 600, 6140, Wellington, New Zealand
| | - Amy T Lynch
- School of Chemical and Physical Sciences, Victoria University of Wellington, PO Box 600, 6140, Wellington, New Zealand
- Centre for Biodiscovery, Victoria University of Wellington, PO Box 600, 6140, Wellington, New Zealand
| | - Bridget L Stocker
- School of Chemical and Physical Sciences, Victoria University of Wellington, PO Box 600, 6140, Wellington, New Zealand
- Centre for Biodiscovery, Victoria University of Wellington, PO Box 600, 6140, Wellington, New Zealand
| | - Mattie S M Timmer
- School of Chemical and Physical Sciences, Victoria University of Wellington, PO Box 600, 6140, Wellington, New Zealand
- Centre for Biodiscovery, Victoria University of Wellington, PO Box 600, 6140, Wellington, New Zealand
| |
Collapse
|
85
|
Clairene Filipe K, Dangudubiyyam S, Lion C, Decloquement M, Elin Teppa R, Biot C, Harduin-Lepers A. A Rapid and Sensitive MicroPlate Assay (MPSA) Using an Alkyne-Modified CMP-Sialic Acid Donor to Evaluate Human Sialyltransferase Specificity. Chembiochem 2025; 26:e202400539. [PMID: 39470683 DOI: 10.1002/cbic.202400539] [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: 06/21/2024] [Revised: 10/18/2024] [Accepted: 10/29/2024] [Indexed: 10/30/2024]
Abstract
Human sialyltransferases primarily utilize CMP-Sias, especially transferring Neu5Ac from CMP-Neu5Ac to various acceptors. Advances in chemical biology have led to the synthesis of novel CMP-Sia donors suitable for bioorthogonal reactions in cell-based assays. However, the compatibility of these donors with all human enzymes remains uncertain. We synthesized a non-natural CMP-Sia donor with an alkyne modification on the N-acyl group of Neu5Ac, which was effectively used by human ST6Gal I and ST3Gal I. A sensitive MicroPlate Sialyltransferase Assay (MPSA) was developed and expanded to a panel of 13 human STs acting on glycoproteins. All assayed enzymes tolerated CMP-SiaNAl, allowing for the determination of kinetic parameters and turnover numbers. This study enhances the biochemical characterization of human sialyltransferases and opens new avenues for developing sialyltransferase inhibitors.
Collapse
Affiliation(s)
- Kiamungongo Clairene Filipe
- Univ. Lille, CNRS, UMR 8576, UGSF, Unité de Glycobiologie Structurale et Fonctionnelle, Faculté des sciences et Technologies, F-59000, Lille, France
| | - Sushmaa Dangudubiyyam
- Univ. Lille, CNRS, UMR 8576, UGSF, Unité de Glycobiologie Structurale et Fonctionnelle, Faculté des sciences et Technologies, F-59000, Lille, France
| | - Cédric Lion
- Univ. Lille, CNRS, UMR 8576, UGSF, Unité de Glycobiologie Structurale et Fonctionnelle, Faculté des sciences et Technologies, F-59000, Lille, France
| | - Mathieu Decloquement
- Univ. Lille, CNRS, UMR 8576, UGSF, Unité de Glycobiologie Structurale et Fonctionnelle, Faculté des sciences et Technologies, F-59000, Lille, France
| | - Roxana Elin Teppa
- Univ. Lille, CNRS, UMR 8576, UGSF, Unité de Glycobiologie Structurale et Fonctionnelle, Faculté des sciences et Technologies, F-59000, Lille, France
| | - Christophe Biot
- Univ. Lille, CNRS, UMR 8576, UGSF, Unité de Glycobiologie Structurale et Fonctionnelle, Faculté des sciences et Technologies, F-59000, Lille, France
| | - Anne Harduin-Lepers
- Univ. Lille, CNRS, UMR 8576, UGSF, Unité de Glycobiologie Structurale et Fonctionnelle, Faculté des sciences et Technologies, F-59000, Lille, France
| |
Collapse
|
86
|
Chen J, Martin R. Ni-Catalyzed Stereodivergent Synthesis of N-Glycosides. Chemistry 2025; 31:e202403822. [PMID: 39612346 DOI: 10.1002/chem.202403822] [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: 10/15/2024] [Revised: 11/25/2024] [Accepted: 11/27/2024] [Indexed: 12/01/2024]
Abstract
Herein, we describe a stereoselective Ni-catalyzed N-glycosylation of glycals. The reaction is enabled by addition of an in situ generated nickel hydride across an olefin prior to C-N bond-formation. Stereodivergence can be accomplished on kinetic or thermodynamic grounds, thus giving access to either α- or β-N-glycosides with equal ease. The protocol is distinguished by its operational simplicity, generality and exquisite selectivity, thus offering a new gateway to expedite the synthesis of N-glycosides.
Collapse
Affiliation(s)
- Jinhong Chen
- Institute of Chemical Research of Catalonia (ICIQ), The Barcelona Institute of Science and Technology, Av. Països Catalans 16, 43007, Tarragona, Spain
- Universitat Rovira i Virgili, Departament de Química Analítica i Química Orgànica, c/Marcel⋅lí Domingo, 1, 43007, Tarragona, Spain
| | - Ruben Martin
- Institute of Chemical Research of Catalonia (ICIQ), The Barcelona Institute of Science and Technology, Av. Països Catalans 16, 43007, Tarragona, Spain
- Catalan Institution for Research and Advanced Studies (ICREA), Passeig Lluïs Companys, 23, 08010, Barcelona, Spain
| |
Collapse
|
87
|
Yang Y, Zhou X, Wang W, Dai H. Glycobiology of psoriasis: A review. J Autoimmun 2025; 151:103361. [PMID: 39808852 DOI: 10.1016/j.jaut.2025.103361] [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: 12/19/2023] [Accepted: 01/07/2025] [Indexed: 01/16/2025]
Abstract
Psoriasis is a chronic inflammatory skin disease with etiologies related to genetics, immunity, and the environment. It is characterized by excessive proliferation of keratinocytes and infiltration of inflammatory immune cells. Glycosylation is a post-translational modification of proteins that plays important roles in cell adhesion, signal transduction, and immune cell activation. Abnormal glycosylation is associated with inflammation, tumors, autoimmunity, and several diseases. Glycan profiles and glycosylation-related enzymes are altered in patients with psoriasis. Specific glycan structures, such as glycosaminoglycans and gangliosides, inhibit the development of psoriasis through various pathways. Lectins are glycan-binding proteins that are widely involved in the pathogenesis of psoriasis. The differential serum, epidermal, and dermal expression of galectins in patients with psoriasis distinguishes psoriasis from other nonspecific psoriasis-like dermatitis. This article summarizes relevant literature on psoriasis-related glycans to help clarify the potential molecular mechanisms of psoriasis and identify novel biomarkers and targets for the treatment of psoriasis.
Collapse
Affiliation(s)
- Yinuo Yang
- Department of Dermatology, Peking University Third Hospital, No.49, Huayuan North Road, Haidian, Beijing, 100191, China; Department of Immunology, School of Basic Medical Sciences, NHC Key Laboratory of Medical Immunology, Peking University, No.38, Xueyuan Road, Haidian, Beijing, 100191, China
| | - Xin Zhou
- Department of Dermatology, Peking University Third Hospital, No.49, Huayuan North Road, Haidian, Beijing, 100191, China; Department of Immunology, School of Basic Medical Sciences, NHC Key Laboratory of Medical Immunology, Peking University, No.38, Xueyuan Road, Haidian, Beijing, 100191, China
| | - Wenhui Wang
- Department of Dermatology, Peking University Third Hospital, No.49, Huayuan North Road, Haidian, Beijing, 100191, China.
| | - Hui Dai
- Department of Immunology, School of Basic Medical Sciences, NHC Key Laboratory of Medical Immunology, Peking University, No.38, Xueyuan Road, Haidian, Beijing, 100191, China.
| |
Collapse
|
88
|
Lee S, Ono T, Masaaki S, Fujita A, Matsubara M, Zappa A, Yamada I, Aoki-Kinoshita KF. Updates implemented in version 4 of the GlyCosmos Glycoscience Portal. Anal Bioanal Chem 2025; 417:907-919. [PMID: 39690313 PMCID: PMC11782317 DOI: 10.1007/s00216-024-05692-0] [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: 09/17/2024] [Revised: 11/25/2024] [Accepted: 11/28/2024] [Indexed: 12/19/2024]
Abstract
Glycosylation, characterized by its complexity and diversity, is a common system across all domains of life. The glycosylation of proteins or lipids imparts them with structural and functional roles, ranging from development to infectious or Mendelian disease. The high-throughput-based omics data has revealed that glycans are involved in important cellular processes. Comprehensive knowledge of glycosylation has contributed not only to the fundamental concepts in glycoscience but also to its applications, including the development of molecular markers for diagnosis and therapeutic tools for treating diseases. The GlyCosmos Glycoscience Portal (GlyCosmos) has undergone significant updates to better support the scientific community in studying glycosylation-related phenomena. Key enhancements include the integration of expanded datasets linking glycans to other omics fields, improved tools for glycan structure prediction and analysis, and upgraded visualization capabilities to streamline data interpretation. A strengthened focus on data standardization has also been introduced, fostering interoperability between glycoscience resources and external databases. Since its release in 2019, the portal has seen a fivefold increase in user engagement, reflecting its growing relevance. These recent advancements aim to provide researchers with a more comprehensive and user-friendly platform, enabling deeper insights into glycan roles in cellular processes and disease mechanisms. GlyCosmos will continue to evolve, prioritizing community needs and advancing the integration of glycoscience with broader biological and biomedical research.
Collapse
Affiliation(s)
- Sunmyoung Lee
- Glycan and Life Systems Integration Center (GaLSIC), Soka University, Hachioji, Tokyo, Japan
| | - Tamiko Ono
- Glycan and Life Systems Integration Center (GaLSIC), Soka University, Hachioji, Tokyo, Japan
| | - Shiota Masaaki
- Glycan and Life Systems Integration Center (GaLSIC), Soka University, Hachioji, Tokyo, Japan
| | - Akihiro Fujita
- Institute for Glyco-Core Research, Nagoya University, Nagoya, Japan
| | | | - Achille Zappa
- Glycan and Life Systems Integration Center (GaLSIC), Soka University, Hachioji, Tokyo, Japan
| | | | - Kiyoko F Aoki-Kinoshita
- Glycan and Life Systems Integration Center (GaLSIC), Soka University, Hachioji, Tokyo, Japan.
- Graduate School of Science and Engineering, Soka University, Hachioji, Tokyo, Japan.
- Institute for Glyco-Core Research, Nagoya University, Nagoya, Japan.
| |
Collapse
|
89
|
Chalkley RJ, Baker PR. Improving the Depth and Reliability of Glycopeptide Identification Using Protein Prospector. Mol Cell Proteomics 2025; 24:100903. [PMID: 39788319 PMCID: PMC11851224 DOI: 10.1016/j.mcpro.2025.100903] [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: 09/06/2024] [Revised: 11/27/2024] [Accepted: 12/26/2024] [Indexed: 01/12/2025] Open
Abstract
Glycosylation is the most common and diverse modification of proteins. It can affect protein function and stability and is associated with many diseases. While proteomic methods to study most post-translational modifications are now quite mature, glycopeptide analysis is still a challenge, particularly from the aspect of data analysis. Several software packages have been developed in the last few years that aim to support omic-level N-linked glycopeptide analysis. This study presents developments of Protein Prospector for the analysis of N-glycopeptide data. Results are compared to other software, showing that Protein Prospector reports many more glycoforms of glycopeptides than competing software. The advantages and disadvantages of considering glycan adducts are also evaluated, showing how considering them can correct previously wrong assignments.
Collapse
Affiliation(s)
- Robert J Chalkley
- Department of Pharmaceutical Chemistry, University of California, San Francisco, USA.
| | - Peter R Baker
- Department of Pharmaceutical Chemistry, University of California, San Francisco, USA
| |
Collapse
|
90
|
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
|
91
|
Lei C, Li X, Li W, Chen Z, Liu S, Cheng B, Hu Y, Song Q, Qiu Y, Zhou Y, Meng X, Yu H, Zhou W, Chen X, Li J. Chemical Glycoproteomic Profiling in Rice Seedlings Reveals N-glycosylation in the ERAD-L Machinery. Mol Cell Proteomics 2025; 24:100883. [PMID: 39577566 PMCID: PMC11869521 DOI: 10.1016/j.mcpro.2024.100883] [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: 03/22/2024] [Revised: 11/14/2024] [Accepted: 11/15/2024] [Indexed: 11/24/2024] Open
Abstract
As a ubiquitous and essential posttranslational modification occurring in both plants and animals, protein N-linked glycosylation regulates various important biological processes. Unlike the well-studied animal N-glycoproteomes, the landscape of rice N-glycoproteome remains largely unexplored. Here, by developing a chemical glycoproteomic strategy based on metabolic glycan labeling, we report a comprehensive profiling of the N-glycoproteome in rice seedlings. The rice seedlings are incubated with N-azidoacetylgalactosamine-a monosaccharide analog containing a bioorthogonal functional group-to metabolically label N-glycans, followed by conjugation with an affinity probe via click chemistry for the enrichment of the N-glycoproteins. Subsequent mass spectrometry analyses identify a total of 403 N-glycosylation sites and 673 N-glycosylated proteins, which are involved in various important biological processes. In particular, the core components of the endoplasmic reticulum-associated protein degradation machinery are N-glycosylated, and the N-glycosylation is important for the endoplasmic reticulum-associated protein degradation-L function. This work not only provides an invaluable resource for studying rice N-glycosylation but also demonstrates the applicability of metabolic glycan labeling in glycoproteomic profiling for crop species.
Collapse
Affiliation(s)
- Cong Lei
- College of Chemistry and Molecular Engineering, Peking University, Beijing, China; Beijing National Laboratory for Molecular Sciences, Peking University, Beijing, China; Yazhouwan National Laboratory, Sanya, China
| | - Xilong Li
- Yazhouwan National Laboratory, Sanya, China.
| | - Wenjia Li
- Yazhouwan National Laboratory, Sanya, China; School of Tropical Agriculture and Forestry, Hainan University, Haikou, China
| | - Zihan Chen
- College of Chemistry and Molecular Engineering, Peking University, Beijing, China; Beijing National Laboratory for Molecular Sciences, Peking University, Beijing, China
| | - Simiao Liu
- State Key Laboratory of Plant Genomics and National Center for Plant Gene Research, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, China
| | - Bo Cheng
- College of Chemistry and Molecular Engineering, Peking University, Beijing, China; Beijing National Laboratory for Molecular Sciences, Peking University, Beijing, China
| | - Yili Hu
- Yazhouwan National Laboratory, Sanya, China; School of Tropical Agriculture and Forestry, Hainan University, Haikou, China
| | - Qitao Song
- College of Chemistry and Molecular Engineering, Peking University, Beijing, China; Peking-Tsinghua Center for Life Sciences, Peking University, Beijing, China
| | - Yahong Qiu
- Yazhouwan National Laboratory, Sanya, China
| | - Yilan Zhou
- Yazhouwan National Laboratory, Sanya, China
| | - Xiangbing Meng
- State Key Laboratory of Plant Genomics and National Center for Plant Gene Research, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, China
| | - Hong Yu
- Yazhouwan National Laboratory, Sanya, China
| | - Wen Zhou
- College of Chemistry and Molecular Engineering, Peking University, Beijing, China; Beijing National Laboratory for Molecular Sciences, Peking University, Beijing, China
| | - Xing Chen
- College of Chemistry and Molecular Engineering, Peking University, Beijing, China; Beijing National Laboratory for Molecular Sciences, Peking University, Beijing, China; Peking-Tsinghua Center for Life Sciences, Peking University, Beijing, China; Synthetic and Functional Biomolecules Center, Peking University, Beijing, China; Key Laboratory of Bioorganic Chemistry and Molecular Engineering of Ministry of Education, Peking University, Beijing, China.
| | - Jiayang Li
- Yazhouwan National Laboratory, Sanya, China; State Key Laboratory of Plant Genomics and National Center for Plant Gene Research, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, China; University of Chinese Academy of Sciences, Beijing, China.
| |
Collapse
|
92
|
Li MY, Jiang J, Li JG, Niu H, Ying YL, Tian R, Long YT. Nanopore approaches for single-molecule temporal omics: promises and challenges. Nat Methods 2025; 22:241-253. [PMID: 39558099 DOI: 10.1038/s41592-024-02492-3] [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: 10/31/2023] [Accepted: 09/18/2024] [Indexed: 11/20/2024]
Abstract
The great molecular heterogeneity within single cells demands omics analysis from a single-molecule perspective. Moreover, considering the perpetual metabolism and communication within cells, it is essential to determine the time-series changes of the molecular library, rather than obtaining data at only one time point. Thus, there is an urgent need to develop a single-molecule strategy for this omics analysis to elucidate the biosystem heterogeneity and temporal dynamics. In this Perspective, we explore the potential application of nanopores for single-molecule temporal omics to characterize individual molecules beyond mass, in both a single-molecule and high-throughput manner. Accordingly, recent advances in nanopores available for single-molecule temporal omics are reviewed from the view of single-molecule mass identification, revealing single-molecule heterogeneity and illustrating temporal evolution. Furthermore, we discuss the primary challenges associated with using nanopores for single-molecule temporal omics in complex biological samples, and present the potential strategies and notes to respond to these challenges.
Collapse
Affiliation(s)
- Meng-Yin Li
- State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing, China.
- Chemistry and Biomedicine Innovation Center, Nanjing University, Nanjing, China.
| | - Jie Jiang
- State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing, China
| | - Jun-Ge Li
- State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing, China
| | - Hongyan Niu
- State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing, China
- Chemistry and Biomedicine Innovation Center, Nanjing University, Nanjing, China
| | - Yi-Lun Ying
- State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing, China
- Chemistry and Biomedicine Innovation Center, Nanjing University, Nanjing, China
| | - Ruijun Tian
- Department of Chemistry, School of Science, Southern University of Science and Technology, Shenzhen, China
| | - Yi-Tao Long
- State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing, China.
| |
Collapse
|
93
|
Gadi MR, Han J, Shen T, Fan S, Xiao Z, Li L. Divergent synthesis of amino acid-linked O-GalNAc glycan core structures. Nat Protoc 2025; 20:480-517. [PMID: 39327537 DOI: 10.1038/s41596-024-01051-6] [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: 07/27/2023] [Accepted: 07/19/2024] [Indexed: 09/28/2024]
Abstract
O-GalNAc glycans, also known as mucin-type O-glycans, are primary constituents of mucins on various mucosal sites of the body and also ubiquitously expressed on cell surface and secreted proteins. They have crucial roles in a wide range of physiological and pathological processes, including tumor growth and progression. In addition, altered expression of O-GalNAc glycans is frequently observed during different disease states. Research dedicated to unraveling the structure-function relationships of O-GalNAc glycans has led to the discovery of disease biomarkers and diagnostic tools and the development of O-glycopeptide-based cancer vaccines. Many of these efforts require amino acid-linked O-GalNAc core structures as building blocks to assemble complex O-glycans and glycopeptides. There are eight core structures (cores one to eight), from which all mucin-type O-glycans are derived. In this protocol, we describe the first divergent synthesis of all eight cores from a versatile precursor in practical scales. The protocol involves (i) chemical synthesis of the orthogonally protected precursor (3 days) from commercially available materials, (ii) chemical synthesis of five unique glycosyl donors (1-2 days for each donor) and (iii) selective deprotection of the precursor and assembly of the eight cores (2-4 days for each core). The procedure can be adopted to prepare O-GalNAc cores linked to serine, threonine and tyrosine, which can then be utilized directly for solid-phase glycopeptide synthesis or chemoenzymatic synthesis of complex O-glycans. The procedure empowers researchers with fundamental organic chemistry skills to prepare gram scales of any desired O-GalNAc core(s) or all eight cores concurrently.
Collapse
Affiliation(s)
- Madhusudhan Reddy Gadi
- Department of Chemistry and Center for Diagnostics and Therapeutics, Georgia State University, Atlanta, GA, USA
| | - Jinghua Han
- Department of Chemistry and Center for Diagnostics and Therapeutics, Georgia State University, Atlanta, GA, USA
| | - Tangliang Shen
- Department of Chemistry and Center for Diagnostics and Therapeutics, Georgia State University, Atlanta, GA, USA
| | - Shuquan Fan
- Department of Chemistry and Center for Diagnostics and Therapeutics, Georgia State University, Atlanta, GA, USA
| | - Zhongying Xiao
- Department of Chemistry and Center for Diagnostics and Therapeutics, Georgia State University, Atlanta, GA, USA
| | - Lei Li
- Department of Chemistry and Center for Diagnostics and Therapeutics, Georgia State University, Atlanta, GA, USA.
| |
Collapse
|
94
|
Chadwick SR, Stack-Couture S, Berg MD, Di Gregorio S, Lung B, Genereaux J, Moir RD, Brandl CJ, Willis IM, Snapp EL, Lajoie P. TUDCA modulates drug bioavailability to regulate resistance to acute ER stress in Saccharomyces cerevisiae. Mol Biol Cell 2025; 36:ar13. [PMID: 39661468 PMCID: PMC11809307 DOI: 10.1091/mbc.e24-04-0147] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2024] [Revised: 12/02/2024] [Accepted: 12/06/2024] [Indexed: 12/13/2024] Open
Abstract
Cells counter accumulation of misfolded secretory proteins in the endoplasmic reticulum (ER) through activation of the Unfolded Protein Response (UPR). Small molecules termed chemical chaperones can promote protein folding to alleviate ER stress. The bile acid tauroursodeoxycholic acid (TUDCA) has been described as a chemical chaperone. While promising in models of protein folding diseases, TUDCA's mechanism of action remains unclear. Here, we found TUDCA can rescue growth of yeast treated with the ER stressor tunicamycin (Tm), even in the absence of a functional UPR. In contrast, TUDCA failed to rescue growth on other ER stressors. Nor could TUDCA attenuate chronic UPR associated with specific gene deletions or overexpression of a misfolded mutant secretory protein. Neither pretreatment with nor delayed addition of TUDCA conferred protection against Tm. Importantly, attenuation of Tm-induced toxicity required TUDCA's critical micelle forming concentration, suggesting a mechanism where TUDCA directly sequesters drugs. Indeed, in several assays, TUDCA-treated cells closely resembled cells treated with lower doses of Tm. In addition, we found TUDCA can inhibit dyes from labeling intracellular compartments. Thus, our study challenges the model of TUDCA as a chemical chaperone and suggests that TUDCA decreases drug bioavailability, allowing cells to adapt to ER stress.
Collapse
Affiliation(s)
- Sarah R. Chadwick
- Department of Anatomy and Cell Biology, The University of Western Ontario, London, Ontario N6A 5C1, Canada
| | - Samuel Stack-Couture
- Department of Anatomy and Cell Biology, The University of Western Ontario, London, Ontario N6A 5C1, Canada
| | - Matthew D. Berg
- Department of Biochemistry, The University of Western Ontario, London, Ontario N6A 5C1, Canada
| | - Sonja Di Gregorio
- Department of Anatomy and Cell Biology, The University of Western Ontario, London, Ontario N6A 5C1, Canada
| | - Bryan Lung
- Department of Anatomy and Cell Biology, The University of Western Ontario, London, Ontario N6A 5C1, Canada
| | - Julie Genereaux
- Department of Anatomy and Cell Biology, The University of Western Ontario, London, Ontario N6A 5C1, Canada
- Department of Biochemistry, The University of Western Ontario, London, Ontario N6A 5C1, Canada
| | - Robyn D. Moir
- Department of Biochemistry, Albert Einstein College of Medicine, Bronx, NY 10461
| | - Christopher J. Brandl
- Department of Biochemistry, The University of Western Ontario, London, Ontario N6A 5C1, Canada
| | - Ian M. Willis
- Department of Biochemistry, Albert Einstein College of Medicine, Bronx, NY 10461
| | - Erik L. Snapp
- Janelia Research Campus of the Howard Hughes Medical Institute, Ashburn, VA 20147
| | - Patrick Lajoie
- Department of Anatomy and Cell Biology, The University of Western Ontario, London, Ontario N6A 5C1, Canada
- Children's Health Research Institute, Lawson Health Research Institute, London, Ontario N6C 2V5, Canada
| |
Collapse
|
95
|
Costa AF, Teixeira A, Reis CA, Gomes C. Novel anticancer drug discovery efforts targeting glycosylation: the emergence of fluorinated monosaccharides analogs. Expert Opin Drug Discov 2025; 20:193-203. [PMID: 39749684 DOI: 10.1080/17460441.2024.2444375] [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: 09/05/2024] [Revised: 12/11/2024] [Accepted: 12/16/2024] [Indexed: 01/04/2025]
Abstract
INTRODUCTION Glycosylation is an essential enzymatic process of building glycan structures that occur mainly within the cell and gives rise to a diversity of cell surface and secreted glycoconjugates. These glycoconjugates play vital roles, for instance in cellcell adhesion, interaction and communication, activation of cell surface receptors, inflammatory response and immune recognition. This controlled and wellcoordinated enzymatic process is altered in cancer, leading to the biosynthesis of cancerassociated glycans, which impact glycandependent biological roles. AREAS COVERED In this review, the authors discuss the importance of targeting cancerassociated glycans through potent glycan biosynthesis inhibitors. It focuses on the use of analogs, providing an overview of findings involving these in cancer. The highly explored fluorinated monosaccharide analogs targeting aberrant glycosylation are described, aiming to inspire advances in the field. EXPERT OPINION Altered glycosylation, such as increased sialylation and fucosylation, is a feature in cancer and has been shown to play key roles in several malignant properties of cancer cells. Strategies aiming at remodeling cancer cells´ glycome are emerging and present a huge potential for cancer therapy. Fluorinated monosaccharides have been gathering promising preclinical results as novel cancer drugs. Nevertheless, cancer specific targeting strategies must be considered to avoid significant sideeffects.
Collapse
Affiliation(s)
- Ana F Costa
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
- IPATIMUP - Institute of Molecular Pathology and Immunology, University of Porto, Porto, Portugal
- Institute of Biomedical Sciences of Abel Salazar - ICBAS, University of Porto, Porto, Portugal
| | - Andreia Teixeira
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
- IPATIMUP - Institute of Molecular Pathology and Immunology, University of Porto, Porto, Portugal
| | - Celso A Reis
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
- IPATIMUP - Institute of Molecular Pathology and Immunology, University of Porto, Porto, Portugal
- Institute of Biomedical Sciences of Abel Salazar - ICBAS, University of Porto, Porto, Portugal
- Medical Faculty, University of Porto, Porto, Portugal
| | - Catarina Gomes
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
- IPATIMUP - Institute of Molecular Pathology and Immunology, University of Porto, Porto, Portugal
| |
Collapse
|
96
|
Qin S, Tian Z. Deep structure-level N-glycan identification using feature-induced structure diagnosis integrated with a deep learning model. Anal Bioanal Chem 2025; 417:1001-1014. [PMID: 39212697 DOI: 10.1007/s00216-024-05505-4] [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: 06/10/2024] [Revised: 07/30/2024] [Accepted: 08/14/2024] [Indexed: 09/04/2024]
Abstract
Being a widely occurring protein post-translational modification, N-glycosylation features unique multi-dimensional structures including sequence and linkage isomers. There have been successful bioinformatics efforts in N-glycan structure identification using N-glycoproteomics data; however, symmetric "mirror" branch isomers and linkage isomers are largely unresolved. Here, we report deep structure-level N-glycan identification using feature-induced structure diagnosis (FISD) integrated with a deep learning model. A neural network model is integrated to conduct the identification of featured N-glycan motifs and boosts the process of structure diagnosis and distinction for linkage isomers. By adopting publicly available N-glycoproteomics datasets of five mouse tissues (17,136 intact N-glycopeptide spectrum matches) and a consideration of 23 motif features, a deep learning model integrated with a convolutional autoencoder and a multilayer perceptron was trained to be capable of predicting N-glycan featured motifs in the MS/MS spectra with previously identified compositions. In the test of the trained model, a prediction accuracy of 0.8 and AUC value of 0.95 were achieved; 5701 previously unresolved N-glycan structures were assigned by matched structure-diagnostic ions; and by using an explainable learning algorithm, two new fragmentation features of m/z = 674.25 and m/z = 835.28 were found to be significant to three N-glycan structure motifs with fucose, NeuAc, and NeuGc, proving the capability of FISD to discover new features in the MS/MS spectra.
Collapse
Affiliation(s)
- Suideng Qin
- School of Chemical Science & Engineering, Shanghai Key Laboratory of Chemical Assessment and Sustainability, Tongji University, Shanghai, 200092, China
| | - Zhixin Tian
- School of Chemical Science & Engineering, Shanghai Key Laboratory of Chemical Assessment and Sustainability, Tongji University, Shanghai, 200092, China.
| |
Collapse
|
97
|
Sun Q, Hong S. Glycoscience in Advancing PD-1/PD-L1-Axis-Targeted Tumor Immunotherapy. Int J Mol Sci 2025; 26:1238. [PMID: 39941004 PMCID: PMC11818636 DOI: 10.3390/ijms26031238] [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: 12/11/2024] [Revised: 01/23/2025] [Accepted: 01/28/2025] [Indexed: 02/16/2025] Open
Abstract
Immune checkpoint blockade therapy, represented by anti-PD-1/PD-L1 monoclonal antibodies, has significantly changed the immunotherapy landscape. However, the treatment is still limited by unsatisfactory response rates, immune-related adverse effects, and drug resistance. Current studies have established that glycosylation, a common post-translational modification, is crucial in promoting cancer progression and immune invasion. Targeting aberrant glycosylation in cancers presents precision medicine regimens for monitoring cancer progression and developing personalized medicine. Notably, the immune checkpoints PD-1 and PD-L1 are highly glycosylated, which affects PD-1/PD-L1 interaction and the binding of anti-PD-1/PD-L1 monoclonal antibodies. Recent achievements in glycoscience to enhance patient outcomes, referred to as glycotherapy, have underscored their high potency in advancing PD-1/PD-L1 blockade therapies, i.e., glycoengineered antibodies with improved binding toward PD-1/PD-L1, pharmaceutic inhibitors for core fucosylation and sialylation, and synergistic treatment with the antibody-sialidase conjugate. This review briefly introduces the PD-1/PD-L1 axis and glycosylation and highlights the fundamental and applied advances in glycoscience that improve PD-1/PD-L1 immunoblockade therapies.
Collapse
Affiliation(s)
| | - Senlian Hong
- State Key Laboratory of Natural and Biomimetic Drugs, Chemical Biology Center, Department of Chemical Biology, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China;
| |
Collapse
|
98
|
Jochim BE, Topalidou I, Lehrbach NJ. Protein sequence editing defines distinct and overlapping functions of SKN-1A/Nrf1 and SKN-1C/Nrf2. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.01.29.635299. [PMID: 39975340 PMCID: PMC11838306 DOI: 10.1101/2025.01.29.635299] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 02/21/2025]
Abstract
The Nrf/NFE2L family of transcription factors regulates redox balance, xenobiotic detoxification, metabolism, proteostasis, and aging. Nrf1/NFE2L1 is primarily responsible for stress-responsive upregulation of proteasome subunit genes and is essential for adaptation to proteotoxic stress. Nrf2/NFE2L2 is mainly involved in activating oxidative stress responses and promoting xenobiotic detoxification. Nrf1 and Nrf2 contain very similar DNA binding domains and can drive similar transcriptional responses. In C. elegans, a single gene, skn-1, encodes distinct protein isoforms, SKN-1A and SKN-1C, that function analogously to mammalian Nrf1 and Nrf2, respectively, and share an identical DNA binding domain. Thus, the extent to which SKN-1A/Nrf1 and SKN-1C/Nrf2 functions are distinct or overlapping has been unclear. Regulation of the proteasome by SKN-1A/Nrf1 requires post-translational conversion of N-glycosylated asparagine residues to aspartate by the PNG-1/NGLY1 peptide:N-glycanase, a process we term 'sequence editing'. Here, we reveal the consequences of sequence editing for the transcriptomic output of activated SKN-1A. We confirm that activation of proteasome subunit genes is strictly dependent on sequence editing. In addition, we find that sequence edited SKN-1A can also activate genes linked to redox homeostasis and xenobiotic detoxification that are also regulated by SKN-1C, but the extent of these genes' activation is antagonized by sequence editing. Using mutant alleles that selectively inactivate either SKN-1A or SKN-1C, we show that both isoforms promote optimal oxidative stress resistance, acting as effectors for distinct signaling pathways. These findings suggest that sequence editing governs SKN-1/Nrf functions by tuning the SKN-1A/Nrf1 regulated transcriptome.
Collapse
|
99
|
Chen S, Xie Y, Alvarez MR, Sheng Y, Bouchibti Y, Chang V, Lebrilla CB. Quantitative Glycan-Protein Cross-Linking Mass Spectrometry Using Enrichable Linkers Reveals Extensive Glycan-Mediated Protein Interaction Networks. Anal Chem 2025; 97:1584-1593. [PMID: 39805041 PMCID: PMC11780575 DOI: 10.1021/acs.analchem.4c04134] [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: 08/05/2024] [Revised: 12/19/2024] [Accepted: 01/03/2025] [Indexed: 01/16/2025]
Abstract
Protein-protein interactions in the cell membrane are typically mediated by glycans, with terminal sialic acid often involved in these interactions. To probe the nature of the interactions, we developed quantitative cross-linking methods involving the glycans of the glycoproteins and the polypeptide moieties of proteins. We designed and synthesized biotinylated enrichable cross-linkers that were click-tagged to metabolically incorporate azido-sialic acid on cell surface glycans to allow cross-linking of the azido-glycans with lysine residues on proximal polypeptides. The glycopeptide-peptide cross-links (GPx) were enriched using biotin groups through affinity purification with streptavidin resin beads. Workflows using two linkers, one photocleavable and the other disulfide, were developed and applied to reveal the sialic acid-mediated cell-surface protein networks of PNT2 (prostate) cells. Glycopeptide-peptide pairs were identified, with 12000 GPx linked by sialylated glycoforms revealing interactions between source glycoproteins and nearly 700 target proteins. Protein-protein interactions were characterized by as many as 40 peptide pairs, and the extent of the interactions between proteins was prioritized by the number of GPx. Quantitation was performed by counting the number of GPx that identify the protein pairs. Abundant membrane proteins such as ITGB1 yielded an interactome consisting of around 400 other proteins, which were ranked from the most extensive interaction, having the largest number of GPx, to at least one. The interactome was further confirmed separately by published databases. This tool will enhance our understanding of glycosylation on protein-protein interactions and provide new potential targets for therapeutics.
Collapse
Affiliation(s)
- Siyu Chen
- Department
of Chemistry, University of California, Davis, California 95616, United States
| | - Yixuan Xie
- Department
of Chemistry, University of California, Davis, California 95616, United States
- State
Key Laboratory of Genetic Engineering, Greater Bay Area Institute
of Precision Medicine (Guangzhou), School of Life Sciences and Institutes
of Biomedical Sciences, Fudan University, Shanghai, 200438, China
| | | | - Ying Sheng
- Department
of Chemistry, University of California, Davis, California 95616, United States
| | - Yasmine Bouchibti
- Department
of Chemistry, University of California, Davis, California 95616, United States
| | - Vincent Chang
- Department
of Chemistry, University of California, Davis, California 95616, United States
| | - Carlito B. Lebrilla
- Department
of Chemistry, University of California, Davis, California 95616, United States
- Department
of Biochemistry, University of California, Davis, California 95616, United States
| |
Collapse
|
100
|
Soares C, Laugieri ME, Grosso AS, Natale M, Coelho H, Behren S, Yu J, Cai H, Franconetti A, Oyenarte I, Magnasco M, Gimeno A, Ramos N, Chai W, Corzana F, Westerlind U, Jiménez-Barbero J, Palma AS, Videira PA, Ereño-Orbea J, Marcelo F. Decoding the Molecular Basis of the Specificity of an Anti-sTn Antibody. JACS AU 2025; 5:225-236. [PMID: 39886580 PMCID: PMC11775696 DOI: 10.1021/jacsau.4c00921] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/30/2024] [Revised: 11/11/2024] [Accepted: 11/13/2024] [Indexed: 02/01/2025]
Abstract
The mucin O-glycan sialyl Tn antigen (sTn, Neu5Acα2-6GalNAcα1-O-Ser/Thr) is an antigen associated with different types of cancers, often linked with a higher risk of metastasis and poor prognosis. Despite efforts to develop anti-sTn antibodies with high specificity for diagnostics and immunotherapy, challenges in eliciting high-affinity antibodies for glycan structures have limited their effectiveness, leading to low titers and short protection durations. Experimental structural insights into anti-sTn antibody specificity are lacking, hindering their optimization for cancer cell recognition. In this study, we used a comprehensive structural approach, combining X-ray crystallography, NMR spectroscopy, computational methods, glycan/glycopeptide microarrays, and biophysical techniques, to thoroughly investigate the molecular basis of sTn recognition by L2A5, a novel preclinical anti-sTn monoclonal antibody (mAb). Our data unequivocally show that the L2A5 fragment antigen-binding (Fab) specifically binds to core sTn moieties. NMR and X-ray structural data suggest a similar binding mode for the complexes formed by the sTn moiety linked to Ser or Thr and the L2A5 Fab. The sugar moieties are similarly oriented in the paratope of mAb, with the Neu5Ac moiety establishing key interactions with the receptor and the GalNAc moiety providing additional contacts. Furthermore, L2A5 exhibits fine specificity toward cancer-related MUC1 and MUC4 mucin-derived sTn glycopeptides, which might contribute to its selective targeting against tumor cells. This newfound knowledge holds promise for the rational improvement and potential application of this anti-sTn antibody in diagnosis and targeted therapy against sTn expressing cancers such as breast, colorectal, and bladder cancer, improving patient care.
Collapse
Affiliation(s)
- Cátia
O. Soares
- UCIBIO—Applied
Molecular Biosciences Unit, Department of Chemistry, NOVA School of
Science and Technology, NOVA University
Lisbon, 2829-516 Caparica, Portugal
- Associate
Laboratory i4HB, Institute for Health and Bioeconomy, NOVA School
of Science and Technology, NOVA University
Lisbon, 2829-516 Caparica, Portugal
| | - Maria Elena Laugieri
- CIC
bioGUNE, Basque Research and Technology
Alliance, Bizkaia Technology
Park, Ed. 800, E-48160 Derio, Spain
| | - Ana Sofia Grosso
- UCIBIO—Applied
Molecular Biosciences Unit, Department of Chemistry, NOVA School of
Science and Technology, NOVA University
Lisbon, 2829-516 Caparica, Portugal
- Associate
Laboratory i4HB, Institute for Health and Bioeconomy, NOVA School
of Science and Technology, NOVA University
Lisbon, 2829-516 Caparica, Portugal
| | - Mariangela Natale
- UCIBIO—Applied
Molecular Biosciences Unit, Department of Chemistry, NOVA School of
Science and Technology, NOVA University
Lisbon, 2829-516 Caparica, Portugal
- Associate
Laboratory i4HB, Institute for Health and Bioeconomy, NOVA School
of Science and Technology, NOVA University
Lisbon, 2829-516 Caparica, Portugal
| | - Helena Coelho
- UCIBIO—Applied
Molecular Biosciences Unit, Department of Chemistry, NOVA School of
Science and Technology, NOVA University
Lisbon, 2829-516 Caparica, Portugal
- Associate
Laboratory i4HB, Institute for Health and Bioeconomy, NOVA School
of Science and Technology, NOVA University
Lisbon, 2829-516 Caparica, Portugal
| | - Sandra Behren
- Department
of Chemistry, Umeå University, 90187 Umeå, Sweden
| | - Jin Yu
- Department
of Chemistry, Umeå University, 90187 Umeå, Sweden
- Glycosciences
Laboratory, Faculty of Medicine, Imperial
College London, Du Cane Road, London W12
0NN, U.K.
| | - Hui Cai
- Department
of Chemistry, Umeå University, 90187 Umeå, Sweden
- School of
Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou, Guangdong 510275, China
| | - Antonio Franconetti
- CIC
bioGUNE, Basque Research and Technology
Alliance, Bizkaia Technology
Park, Ed. 800, E-48160 Derio, Spain
| | - Iker Oyenarte
- CIC
bioGUNE, Basque Research and Technology
Alliance, Bizkaia Technology
Park, Ed. 800, E-48160 Derio, Spain
| | - Maria Magnasco
- CIC
bioGUNE, Basque Research and Technology
Alliance, Bizkaia Technology
Park, Ed. 800, E-48160 Derio, Spain
| | - Ana Gimeno
- CIC
bioGUNE, Basque Research and Technology
Alliance, Bizkaia Technology
Park, Ed. 800, E-48160 Derio, Spain
| | - Nuno Ramos
- UCIBIO—Applied
Molecular Biosciences Unit, Department of Chemistry, NOVA School of
Science and Technology, NOVA University
Lisbon, 2829-516 Caparica, Portugal
- Associate
Laboratory i4HB, Institute for Health and Bioeconomy, NOVA School
of Science and Technology, NOVA University
Lisbon, 2829-516 Caparica, Portugal
| | - Wengang Chai
- Glycosciences
Laboratory, Department of Metabolism, Digestion and Reproduction, Imperial College London, London SW7 2AZ, U.K.
| | - Francisco Corzana
- Departamento
de Química, Instituto de Investigación en Química
de la Universidad de La Rioja (IQUR), Universidad
de La Rioja, 26006 Logroño, La Rioja, Spain
| | | | - Jesús Jiménez-Barbero
- CIC
bioGUNE, Basque Research and Technology
Alliance, Bizkaia Technology
Park, Ed. 800, E-48160 Derio, Spain
- Ikerbasque
Basque Foundation for Science, Euskadi Plaza 5, 48009 Bilbao, Bizkaia, Spain
- Department
of Organic and Inorganic Chemistry, Faculty of Science and Technology, University of the Basque Country, EHU-UPV, 48940 Leioa, Bizkaia, Spain
- Centro de Investigacion Biomedica En Red de Enfermedades
Respiratorias, 28029 Madrid, Spain
| | - Angelina S. Palma
- UCIBIO—Applied
Molecular Biosciences Unit, Department of Chemistry, NOVA School of
Science and Technology, NOVA University
Lisbon, 2829-516 Caparica, Portugal
- Associate
Laboratory i4HB, Institute for Health and Bioeconomy, NOVA School
of Science and Technology, NOVA University
Lisbon, 2829-516 Caparica, Portugal
| | - Paula A. Videira
- UCIBIO—Applied
Molecular Biosciences Unit, Department of Chemistry, NOVA School of
Science and Technology, NOVA University
Lisbon, 2829-516 Caparica, Portugal
- Associate
Laboratory i4HB, Institute for Health and Bioeconomy, NOVA School
of Science and Technology, NOVA University
Lisbon, 2829-516 Caparica, Portugal
| | - June Ereño-Orbea
- CIC
bioGUNE, Basque Research and Technology
Alliance, Bizkaia Technology
Park, Ed. 800, E-48160 Derio, Spain
- Ikerbasque
Basque Foundation for Science, Euskadi Plaza 5, 48009 Bilbao, Bizkaia, Spain
| | - Filipa Marcelo
- UCIBIO—Applied
Molecular Biosciences Unit, Department of Chemistry, NOVA School of
Science and Technology, NOVA University
Lisbon, 2829-516 Caparica, Portugal
- Associate
Laboratory i4HB, Institute for Health and Bioeconomy, NOVA School
of Science and Technology, NOVA University
Lisbon, 2829-516 Caparica, Portugal
| |
Collapse
|