1
|
Perez SJLP, Hsu ZF, Chang TT, Chen CL, Li WS. Harnessing the bishomolithocholic acid scaffold for selective sialyltransferase inhibition: A targeted approach to suppress breast cancer metastasis. Eur J Med Chem 2025; 292:117674. [PMID: 40324300 DOI: 10.1016/j.ejmech.2025.117674] [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: 12/26/2024] [Revised: 04/16/2025] [Accepted: 04/21/2025] [Indexed: 05/07/2025]
Abstract
ST6GAL1 plays a crucial role in the progression of triple-negative breast cancer (TNBC), highlighting its potential as a therapeutic target for this aggressive cancer subtype. Due to the high metastatic potential of TNBC and the limitations of current therapies, selective and potent ST6GAL1 inhibitors are urgently needed. In this study, a scaffold-hopping approach from lithocholic acid to bishomolithocholic acid successfully led to the discovery of novel ST6GAL1 inhibitors, SPP-037 and HZF01, with enhanced biological activity and selectivity. Both compounds significantly inhibited MDA-MB-231 cell migration, HUVEC tube formation, tumor growth, and metastasis in vitro and in vivo. Molecular docking studies revealed key interactions between the ST inhibitors and ST6GAL1, supporting their enhanced selectivity and binding affinity. Additionally, SPP-037 and HZF01 were found to block integrin α2,6-sialylation, disrupting integrin activation and downstream signaling pathways involving the phosphorylation of focal adhesion kinase (FAK) and paxillin, which are critical for cell migration. These results underscore the potential of targeting ST6GAL1 to suppress tumor progression and metastasis, offering a promising avenue for treating aggressive breast cancer.
Collapse
Affiliation(s)
- Ser John Lynon P Perez
- Institute of Chemistry, Academia Sinica, Taipei, 115, Taiwan; Department of Applied Chemistry, National Yang Ming Chiao Tung University, Hsinchu, 300, Taiwan; Sustainable Chemical Science and Technology, Taiwan International Graduate Program, Academia Sinica, Taipei, 115, Taiwan; Biomedical Translation Research Center, Academia Sinica, Taipei, 115, Taiwan
| | - Zih-Fan Hsu
- Institute of Chemistry, Academia Sinica, Taipei, 115, Taiwan; Department of Chemistry, National Taiwan Normal University, Taipei, 116, Taiwan
| | - Tzu-Ting Chang
- Biomedical Translation Research Center, Academia Sinica, Taipei, 115, Taiwan
| | - Chia-Ling Chen
- Institute of Chemistry, Academia Sinica, Taipei, 115, Taiwan
| | - Wen-Shan Li
- Institute of Chemistry, Academia Sinica, Taipei, 115, Taiwan; Sustainable Chemical Science and Technology, Taiwan International Graduate Program, Academia Sinica, Taipei, 115, Taiwan; Biomedical Translation Research Center, Academia Sinica, Taipei, 115, Taiwan; Department of Medicinal and Applied Chemistry, College of Life Science, Kaohsiung Medical University, Kaohsiung, 807, Taiwan; Ph.D. Program in Drug Discovery and Development Industry, College of Pharmacy, Taipei Medical University, Taipei, 110, Taiwan; Doctoral Degree Program in Marine Biotechnology, National Sun Yat-Sen University, Kaohsiung, 804, Taiwan.
| |
Collapse
|
2
|
Qusairy Z, Rada M. Glycosylation in cancer: mechanisms, diagnostic markers, and therapeutic applications. Mol Cell Biochem 2025:10.1007/s11010-025-05303-1. [PMID: 40389792 DOI: 10.1007/s11010-025-05303-1] [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: 03/04/2025] [Accepted: 05/04/2025] [Indexed: 05/21/2025]
Abstract
Glycosylation, a key post-translational modification, plays a pivotal role in cancer progression by influencing critical processes such as protein folding, immune modulation, and intercellular signaling. Altered glycosylation patterns are increasingly recognized as fundamental drivers of tumorigenesis, contributing to key cancer hallmarks like enhanced tumor migration, metastasis, and immune evasion. These aberrant glycosylation signatures not only offer insights into cancer biology but also serve as valuable diagnostic markers and potential therapeutic targets across a range of malignancies. This review explores the mechanisms underlying glycosylation alterations in cancer. We discuss the molecular basis of these changes, including genetic mutations, epigenetic regulation, and oncogene-driven shifts in glycosylation pathways. Additionally, we highlight recent advancements in glycomics research, with a focus on how these alterations influence tumor progression, angiogenesis, and the tumor microenvironment. Furthermore, the review considers the clinical implications of glycosylation changes, including their role in resistance to anti-cancer therapies and their potential as biomarkers for personalized treatment strategies. By bridging fundamental glycosylation research with clinical applications, this review underscores the promise of glycosylation as both a diagnostic tool and a therapeutic target in oncology, offering new avenues for improved patient stratification and precision medicine.
Collapse
Affiliation(s)
- Zahraa Qusairy
- McGill University Health Center Research Institute, Montreal, QC, H4A 3J1, Canada
| | - Miran Rada
- Medical Laboratory Science, Komar University of Science and Technology, Qularaisi, Sulaimani, Sulaymaniyah, Kurdistan Region, Iraq.
- Komar Cancer Research Program, Komar University of Science and Technology, Qularaisi, Sulaimani, Sulaymaniyah, Kurdistan Region, Iraq.
| |
Collapse
|
3
|
Slater AS, McDonald AG, Hickey RM, Davey GP. Glycosyltransferases: glycoengineers in human milk oligosaccharide synthesis and manufacturing. Front Mol Biosci 2025; 12:1587602. [PMID: 40370521 PMCID: PMC12074965 DOI: 10.3389/fmolb.2025.1587602] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2025] [Accepted: 04/11/2025] [Indexed: 05/16/2025] Open
Abstract
Human milk oligosaccharides (HMOs) are a diverse group of complex carbohydrates that play crucial roles in infant health, promoting a beneficial gut microbiota, modulating immune responses, and protecting against pathogens. Central to the synthesis of HMOs are glycosyltransferases, a specialized class of enzymes that catalyse the transfer of sugar moieties to form the complex glycan structures characteristic of HMOs. This review provides an in-depth analysis of glycosyltransferases, beginning with their classification based on structural and functional characteristics. The catalytic activity of these enzymes is explored, highlighting the mechanisms by which they facilitate the precise addition of monosaccharides in HMO biosynthesis. Structural insights into glycosyltransferases are also discussed, shedding light on how their conformational features enable specific glycosidic bond formations. This review maps out the key biosynthetic pathways involved in HMO production, including the synthesis of lactose, and subsequent fucosylation and sialylation processes, all of which are intricately regulated by glycosyltransferases. Industrial methods for HMO synthesis, including chemical, enzymatic, and microbial approaches, are examined, emphasizing the role of glycosyltransferases in these processes. Finally, the review discusses future directions in glycosyltransferase research, particularly in enhancing the efficiency of HMO synthesis and developing advanced analytical techniques to better understand the structural complexity and biological functions of HMOs.
Collapse
Affiliation(s)
- Alanna S. Slater
- Teagasc Food Research Centre, Moorepark, Fermoy, Ireland
- School of Biochemistry and Immunology, Trinity College Dublin, Dublin, Ireland
| | - Andrew G. McDonald
- School of Biochemistry and Immunology, Trinity College Dublin, Dublin, Ireland
| | - Rita M. Hickey
- Teagasc Food Research Centre, Moorepark, Fermoy, Ireland
| | - Gavin P. Davey
- School of Biochemistry and Immunology, Trinity College Dublin, Dublin, Ireland
| |
Collapse
|
4
|
Huang T, Xie W, Guo Y, Li Y, Yin J, Jin X, Ma Y, Zhang Y, Huang D, Chen C, Wang X, Zhu Z, Gan Y, Liesz A, Yu W, Yuan J, Li P. St3gal5-mediated sialylation of glyco-CD177 on neutrophils restricts neuroinflammation following CNS injury. Proc Natl Acad Sci U S A 2025; 122:e2426187122. [PMID: 40244680 PMCID: PMC12037025 DOI: 10.1073/pnas.2426187122] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2024] [Accepted: 03/04/2025] [Indexed: 04/18/2025] Open
Abstract
Neutrophils are the most abundant circulating leukocyte population that play critical roles in neuroinflammation following central nervous system (CNS) injury. CD177, a glycoprotein on neutrophils, is emerging as an important immune regulator which can fundamentally affect multiple human inflammatory diseases. However, the role and regulatory mechanism of CD177 glycobiology of neutrophils in neuroinflammation remain elusive. Here, we show that CD177+ neutrophils expand significantly and infiltrate the injured brain following CNS injury both in the human and mouse. Using single-cell RNA sequencing and genetic approaches, we find CD177+ neutrophils as an anti-inflammatory subset that is critical for modulating neuroinflammation after CNS injury. We further identify St3gal5, a sialyltransferase (ST), that can mediate the sialylation and cell surface presentation of glyco-CD177 on neutrophils. Glycoproteomics reveal downregulated sialylation levels in St3gal5-deficient neutrophils. Neutrophil-specific depletion of St3gal5 prevents the cell surface presentation of CD177 on brain-infiltrated neutrophils and exacerbates neuroinflammation. Administration of the FDA-approved anticonvulsant valproic acid (VPA), an St3gal5 upregulator, promotes the glycosylation of neutrophils and attenuates neuroinflammation following CNS injury. Our study reveals a glycoimmuno-regulatory effect of neutrophils and suggests VPA as a neutrophil glycobiology targeting approach to combat neuroinflammation following CNS injury.
Collapse
Affiliation(s)
- Tingting Huang
- Department of Anesthesiology, Clinical Research Center, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai200127, China
- Key Laboratory of Anesthesiology, Shanghai Jiao Tong University, Ministry of Education, Shanghai200127, China
| | - Wanqing Xie
- Department of Anesthesiology, Clinical Research Center, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai200127, China
- Key Laboratory of Anesthesiology, Shanghai Jiao Tong University, Ministry of Education, Shanghai200127, China
| | - Yunlu Guo
- Department of Anesthesiology, Clinical Research Center, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai200127, China
- Key Laboratory of Anesthesiology, Shanghai Jiao Tong University, Ministry of Education, Shanghai200127, China
| | - Yan Li
- Department of Anesthesiology, Clinical Research Center, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai200127, China
- Key Laboratory of Anesthesiology, Shanghai Jiao Tong University, Ministry of Education, Shanghai200127, China
| | - Jiemin Yin
- Department of Anesthesiology, Clinical Research Center, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai200127, China
- Key Laboratory of Anesthesiology, Shanghai Jiao Tong University, Ministry of Education, Shanghai200127, China
| | - Xia Jin
- Department of Anesthesiology, Clinical Research Center, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai200127, China
- Key Laboratory of Anesthesiology, Shanghai Jiao Tong University, Ministry of Education, Shanghai200127, China
| | - Yezhi Ma
- Department of Anesthesiology, Clinical Research Center, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai200127, China
- Key Laboratory of Anesthesiology, Shanghai Jiao Tong University, Ministry of Education, Shanghai200127, China
| | - Yueman Zhang
- Department of Anesthesiology, Clinical Research Center, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai200127, China
- Key Laboratory of Anesthesiology, Shanghai Jiao Tong University, Ministry of Education, Shanghai200127, China
| | - Dan Huang
- Department of Anesthesiology, Clinical Research Center, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai200127, China
- Key Laboratory of Anesthesiology, Shanghai Jiao Tong University, Ministry of Education, Shanghai200127, China
| | - Caiyang Chen
- Department of Anesthesiology, Clinical Research Center, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai200127, China
- Key Laboratory of Anesthesiology, Shanghai Jiao Tong University, Ministry of Education, Shanghai200127, China
| | - Xin Wang
- Department of Anesthesiology, Clinical Research Center, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai200127, China
- Key Laboratory of Anesthesiology, Shanghai Jiao Tong University, Ministry of Education, Shanghai200127, China
| | - Ziyu Zhu
- Department of Anesthesiology, Clinical Research Center, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai200127, China
- Key Laboratory of Anesthesiology, Shanghai Jiao Tong University, Ministry of Education, Shanghai200127, China
| | - Yu Gan
- Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai200127, China
| | - Arthur Liesz
- Institute for Stroke and Dementia Research, University Hospital, Ludwig-Maximilians-Universität München, Munich81377, Germany
- Munich Cluster for Systems Neurology (SyNergy), Munich81377, Germany
| | - Weifeng Yu
- Department of Anesthesiology, Clinical Research Center, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai200127, China
- Key Laboratory of Anesthesiology, Shanghai Jiao Tong University, Ministry of Education, Shanghai200127, China
| | - Junying Yuan
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Pudong, Shanghai201210, China
| | - Peiying Li
- Department of Anesthesiology, Clinical Research Center, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai200127, China
- Key Laboratory of Anesthesiology, Shanghai Jiao Tong University, Ministry of Education, Shanghai200127, China
| |
Collapse
|
5
|
Fan TC, Yeo HL, Hung TH, Chang NC, Tang YH, Yu J, Chen SH, Yu AL. ST3GAL1 regulates cancer cell migration through crosstalk between EGFR and neuropilin-1 signaling. J Biol Chem 2025; 301:108368. [PMID: 40024474 PMCID: PMC11984587 DOI: 10.1016/j.jbc.2025.108368] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2025] [Revised: 02/20/2025] [Accepted: 02/22/2025] [Indexed: 03/04/2025] Open
Abstract
Metastasis is a major cause of cancer-related morbidity and mortality. The overexpression of the sialyltransferase ST3GAL1 in breast cancer correlates with metastasis. However, the molecular mechanisms underlying the effect of ST3GAL1 on cell movement are poorly understood. We identified neuropilin-1/NRP1 as a substrate for ST3GAL1. Gene expression analysis revealed that recurrence-free survival (p = 0.0046) and distant metastasis-free survival (p = 0.0003) were significantly shorter in the ST3GAL1HighNRP1High cohort than in the both-low subgroup. We demonstrated that the ST3GAL1-mediated sialylation of NRP1 results in increased binding affinity toward EGFR at the molecular level. At the cellular level, ST3GAL1 silencing impaired cell migration and wound healing ability, which was linked to reduced activities of CAPN2 as a consequence of diminished EGF/EGFR signaling. These data establish a function for the ST3GAL1-mediated sialylation of NRP1, leading to increased EGF/EGFR downstream signaling and enhanced tumor cell motility. Furthermore, ST3GAL1 silencing augmented the sensitivity to cetuximab-mediated cell lysis. Our findings provide novel insight into the mechanisms underlying the function of ST3GAL1 in promoting tumor cell migration through the EGFR/NRP1 pathway. Our results suggest that ST3GAL1 may represent a valuable target for strategies aimed at inhibiting tumor migration.
Collapse
Affiliation(s)
- Tan-Chi Fan
- Institute of Stem Cell and Translational Cancer Research, Chang Gung Memorial Hospital, Linkou Branch, Taoyuan, Taiwan
| | - Hui Ling Yeo
- Institute of Stem Cell and Translational Cancer Research, Chang Gung Memorial Hospital, Linkou Branch, Taoyuan, Taiwan
| | - Tsai-Hsien Hung
- Institute of Stem Cell and Translational Cancer Research, Chang Gung Memorial Hospital, Linkou Branch, Taoyuan, Taiwan
| | - Nai-Chuan Chang
- Institute of Stem Cell and Translational Cancer Research, Chang Gung Memorial Hospital, Linkou Branch, Taoyuan, Taiwan
| | - Yun-Hsin Tang
- Institute of Stem Cell and Translational Cancer Research, Chang Gung Memorial Hospital, Linkou Branch, Taoyuan, Taiwan; Department of Obstetrics and Gynecology, Chang Gung Memorial Hospital, Linkou Branch, and Chang Gung University, College of Medicine, Taoyuan, Taiwan; Gynecologic Cancer Research Center, Chang Gung Memorial Hospital, Linkou Branch, Taoyuan, Taiwan
| | - John Yu
- Institute of Stem Cell and Translational Cancer Research, Chang Gung Memorial Hospital, Linkou Branch, Taoyuan, Taiwan
| | - Shih-Hsiang Chen
- Division of Hematology-Oncology, Department of Pediatrics, Chang Gung Memorial Hospital, Linkou Branch, and Chang Gung University, College of Medicine, Taoyuan, Taiwan.
| | - Alice L Yu
- Institute of Stem Cell and Translational Cancer Research, Chang Gung Memorial Hospital, Linkou Branch, Taoyuan, Taiwan; Genomics Research Center, Academia Sinica, Taipei, Taiwan; Department of Pediatrics/Hematology Oncology, University of California in San Diego, San Diego, California, USA.
| |
Collapse
|
6
|
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] [Download PDF] [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
|
7
|
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
|
8
|
Taguchi H, Fujishiro H, Sumi D. Arsenite increases sialic acid levels on the cellular surface through the inhibition of sialidase activity. Biochem Biophys Res Commun 2024; 739:150973. [PMID: 39541927 DOI: 10.1016/j.bbrc.2024.150973] [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: 10/03/2024] [Revised: 10/08/2024] [Accepted: 11/08/2024] [Indexed: 11/17/2024]
Abstract
Chronic exposure to arsenic has been shown to induce carcinogenesis in multiple organs, but the mechanisms underlying the multi-organ carcinogenicity of arsenic remain unknown. We here examined whether arsenic affects the amount of sialic acid on the cellular surface of immortalized HaCaT cells rather than cancerous cells to clarify the process of arsenic-induced carcinogenesis, since sialic acid is known to assist cancer cells in suppressing attacks by natural killer (NK) cells. Our results indicated that exposure to arsenite (As(III)) increases the amounts of sialic acid on the cell surface of HaCaT cells. To elucidate the mechanisms underlying the increase in the levels of sialic acid on the cell surface by As(III) exposure, we measured the activities of sialyltransferase and sialidase in HaCaT cells exposed to As(III). The results showed there was no significant change in the silalyltransferase activity, and the sialidase activity was significantly inhibited by As(III) exposure. When we examined the mRNA levels of NEU1-4, the four types of sialidases identified in mammals after exposure to As(III), no significant change was observed. Furthermore, sialidase activity was significantly reduced in NEU1 siRNA-transfected HaCaT cells, which showed the highest mRNA levels among NEU1-4 in HaCaT cells. These results suggest that inhibition of NEU1-derived sialidase activity by exposure to As(III) resulted in an increase in the amounts of sialic acid on the cell surface.
Collapse
Affiliation(s)
- Hiroki Taguchi
- Faculty of Pharmaceutical Sciences, Tokushima Bunri University, Yamashiro-cho, Tokushima, 770-8514, Japan
| | - Hitomi Fujishiro
- Faculty of Pharmaceutical Sciences, Tokushima Bunri University, Yamashiro-cho, Tokushima, 770-8514, Japan
| | - Daigo Sumi
- Faculty of Pharmaceutical Sciences, Tokushima Bunri University, Yamashiro-cho, Tokushima, 770-8514, Japan.
| |
Collapse
|
9
|
Xu L, Li X, Han S, Mu C, Zhu W. Galacto-oligosaccharides regulate intestinal mucosal sialylation to counteract antibiotic-induced mucin dysbiosis. Food Funct 2024; 15:12016-12032. [PMID: 39563647 DOI: 10.1039/d4fo04626a] [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: 11/21/2024]
Abstract
Intestinal mucin offers a physical barrier to maintain host-commensal homeostasis. Glycosylation is essential for the appropriate functioning of mucin. Galacto-oligosaccharides (GOS) have been used as a prebiotic with proven intestinal benefits, while their regulatory mechanism on mucin remains unclear. This study employed an antibiotic-treated rat model to mimic gut dysbiosis and attempted to restore gut dysbiosis using GOS. The gut microbiome and intestinal mucus O-glycosylations (O-glycans) in the small intestine were profiled by high-throughput sequencing and glycomics. The sialic acid phenotype at the end of O-glycans was further validated with lectin staining. Expressions of key enzymes in sialic acid metabolism and epithelial morphology were determined as well. Antibiotics significantly increased the relative abundance of Escherichia/Shigella and decreased the relative abundance of Lactobacillus. This was accompanied by decreased microbial sialidase activity and increased sialic acid in the digesta, as well as an increase in epithelial sialidase activity. Analysis of key sialylation enzymes showed the upregulation of α 2,6 sialylation (e.g. ST6GALNACs) and downregulation of α 2,3 sialylation (e.g. ST3GALs) after antibiotic treatment. The glycomics results revealed that antibiotics increased core 4 and α 2,6 sialylated O-glycans and decreased core 1, core 3 and α 2,3 sialylated O-glycans in the intestinal mucus of rats, which was further confirmed by lectin staining. Intestinal histology results demonstrated that antibiotic treatment led to the dysbiosis of intestinal mucus homeostasis. To further test the role of microbiota in regulating intestinal mucus sialylation, we supplemented GOS with antibiotics. The results showed that GOS reversed the effects of antibiotics on the gut microbiota and intestinal mucus O-glycans (especially sialylated O-glycans), characterized by an increase of Lactobacillus and α 2,3 sialylated O-glycans and a decrease of Escherichia/Shigella and α 2,6 sialylated O-glycans. What's more, GOS reduced the stimulation of the intestinal mucosa by lipopolysaccharide (LPS) by increasing α 2,3 sialylated intestinal alkaline phosphatase (IAP) to enhance IAP activity, thereby restoring intestinal mucus homeostasis. Overall, GOS counteracts antibiotic-induced mucin deficiency by remedying the gut ecology and changing the mucin sialylation pattern, as reflected by the increase of α 2,3 sialylated O-glycans and the decrease of α 2,6 sialylated O-glycans.
Collapse
Affiliation(s)
- Laipeng Xu
- Laboratory of Gastrointestinal Microbiology, Jiangsu Key Laboratory of Gastrointestinal Nutrition and Animal Health, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China.
- National Center for International Research on Animal Gut Nutrition, Nanjing Agricultural University, Nanjing 210095, China
| | - Xuan Li
- Laboratory of Gastrointestinal Microbiology, Jiangsu Key Laboratory of Gastrointestinal Nutrition and Animal Health, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China.
- National Center for International Research on Animal Gut Nutrition, Nanjing Agricultural University, Nanjing 210095, China
| | - Shuibing Han
- Laboratory of Gastrointestinal Microbiology, Jiangsu Key Laboratory of Gastrointestinal Nutrition and Animal Health, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China.
- National Center for International Research on Animal Gut Nutrition, Nanjing Agricultural University, Nanjing 210095, China
| | - Chunlong Mu
- Department of Biochemistry and Molecular Biology, Cumming School of Medicine, University of Calgary, Calgary, AB., Canada.
| | - Weiyun Zhu
- Laboratory of Gastrointestinal Microbiology, Jiangsu Key Laboratory of Gastrointestinal Nutrition and Animal Health, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China.
- National Center for International Research on Animal Gut Nutrition, Nanjing Agricultural University, Nanjing 210095, China
| |
Collapse
|
10
|
Liu D, Xue Y, Ding D, Zhu B, Shen J, Jin Z, Sun S. Distinct O-Acetylation Patterns of Serum Glycoproteins among Humans, Mice, and Rats. J Proteome Res 2024; 23:5511-5519. [PMID: 39533701 DOI: 10.1021/acs.jproteome.4c00653] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2024]
Abstract
O-Acetylation is a significant chemical modification of sialic acids on glycoproteins with diverse biological functions. As two important animal models, mice and rats have been widely used for various biomedical studies. In this study, we show that the sialic acid types and their O-acetylation patterns have large differences among serum glycoproteins of humans, rats, and mice. Based on intact N-glycopeptide analyses, all sialoglycopeptides in human sera were modified by Neu5Ac without any O-acetylation; 90% of sialoglycopeptides in rat sera were also modified by Neu5Ac, with more than 60% that were further O-acetylated. In contrast, 99% of sialoglycopeptides in mouse sera contained Neu5Gc including 12% in O-acetylated forms. Among all O-acetylated N-glycans, rat sera had hybrid glycans fivefold those of mouse sera, while mouse sera contained 5.5-fold core-fucosylated glycans and 4.6-31.5-fold mono-/penta-/hexa-antenna glycans compared to mice. The overall O-acetylation proportions of serum glycoproteins in rats were much higher than those in mice, and diverse O-acetylation proportions also commonly existed at different glycosites of the same glycoproteins. This study enhances our understanding of O-acetylated sialoglycan diversities and underscores the necessity of considering glycosylation profiles when selecting suitable animal models for various biomedical studies.
Collapse
Affiliation(s)
- Didi Liu
- Laboratory for Disease Glycoproteomics, College of Life Sciences, Northwest University, Xi'an 710069, P. R. China
| | - Yue Xue
- Laboratory for Disease Glycoproteomics, College of Life Sciences, Northwest University, Xi'an 710069, P. R. China
| | - Dan Ding
- Laboratory for Disease Glycoproteomics, College of Life Sciences, Northwest University, Xi'an 710069, P. R. China
| | - Bojing Zhu
- Laboratory for Disease Glycoproteomics, College of Life Sciences, Northwest University, Xi'an 710069, P. R. China
| | - Jiechen Shen
- Laboratory for Disease Glycoproteomics, College of Life Sciences, Northwest University, Xi'an 710069, P. R. China
| | - Zhehui Jin
- Laboratory for Disease Glycoproteomics, College of Life Sciences, Northwest University, Xi'an 710069, P. R. China
| | - Shisheng Sun
- Laboratory for Disease Glycoproteomics, College of Life Sciences, Northwest University, Xi'an 710069, P. R. China
| |
Collapse
|
11
|
Hunter CD, Cairo CW. Detection Strategies for Sialic Acid and Sialoglycoconjugates. Chembiochem 2024; 25:e202400402. [PMID: 39444251 DOI: 10.1002/cbic.202400402] [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: 05/01/2024] [Revised: 08/01/2024] [Indexed: 10/25/2024]
Abstract
Glycoconjugates are a vast class of biomolecules implicated in biological processes important for human health and disease. The structural complexity of glycoconjugates remains a challenge to deciphering their precise biological roles and for their development as biomarkers and therapeutics. Human glycoconjugates on the outside of the cell are modified with sialic (neuraminic) acid residues at their termini. The enzymes that install sialic acids are sialyltransferases (SiaTs), a family of 20 different isoenzymes. The removal and degradation of sialic acids is mediated by neuraminidase (NEU; sialidase) enzymes, of which there are four isoenzymes. In this review, we discuss chemical and biochemical approaches for the detection and analysis of sialoglycoconjugate (SGC) structures and their enzymatic products. The most common methods include affinity probes and synthetic substrates. Fluorogenic and radiolabelled substrates are also important tools for many applications, including screening for enzyme inhibitors. Strategies that give insight into the native substrate-specificity of enzymes that regulate SGCs (SiaT & NEU) are necessary to improve our understanding of the role of sialic acid metabolism in health and disease.
Collapse
Affiliation(s)
- Carmanah D Hunter
- Department of Chemistry, University of Alberta, Edmonton, Alberta, T6G 2G2, Canada
| | - Christopher W Cairo
- Department of Chemistry, University of Alberta, Edmonton, Alberta, T6G 2G2, Canada
| |
Collapse
|
12
|
Hashimoto N, Ito S, Harazono A, Tsuchida A, Mouri Y, Yamamoto A, Okajima T, Ohmi Y, Furukawa K, Kudo Y, Kawasaki N, Furukawa K. Bidirectional signals generated by Siglec-7 and its crucial ligand tri-sialylated T to escape of cancer cells from immune surveillance. iScience 2024; 27:111139. [PMID: 39507251 PMCID: PMC11539641 DOI: 10.1016/j.isci.2024.111139] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2024] [Revised: 08/05/2024] [Accepted: 10/07/2024] [Indexed: 11/08/2024] Open
Abstract
Siglec-7, an inhibitory receptor expressed on natural killer (NK) cells, recognizes sialic acid-containing glycans. However, the ligand glycan structures of Siglec-7 and its carrier proteins have not been comprehensively investigated. Here, we identified four sialyltransferases that are used for the synthesis of ligand glycans of Siglec-7 and two ligand O-glycan-carrier proteins, PODXL and MUC13, using a colon cancer line. Upon binding of these ligand glycans, Siglec-7-expressing immune cells showed reduced cytotoxic activity, whereas cancer cells expressing ligand glycans underwent signal activation, leading to enhanced invasion activity. To clarify the structure of the ligand glycan, podoplanin (PDPN) identified as a Siglec-7 ligand-carrier protein, was transfected into HEK293T cells using sialyltransferase cDNAs. Mass spectrometry of the products revealed a ligand glycan, tri-sialylated T antigen. These results indicate that Siglec-7 interaction with its ligand generates bidirectional signals in NK and cancer cells, leading to the efficient escape of cancers from host immune surveillance.
Collapse
Affiliation(s)
- Noboru Hashimoto
- Biochemistry II, Nagoya University Graduate School of Medicine, Nagoya 466-0065, Japan
- Tissue Regeneration, Tokushima University Graduate School of Biomedical Sciences, Tokushima 770-8504, Japan
| | - Shizuka Ito
- Biochemistry II, Nagoya University Graduate School of Medicine, Nagoya 466-0065, Japan
| | - Akira Harazono
- Biological Chemistry and Biologicals, National Institute of Health Sciences, Kanagawa 210-9501, Japan
| | - Akiko Tsuchida
- Laboratory of Glycobiology, The Noguchi Institute, Itabashi 173-0003, Japan
| | - Yasuhiro Mouri
- Oral Bioscience, Tokushima University Graduate School of Biomedical Sciences, Tokushima 770-8504, Japan
| | - Akihito Yamamoto
- Tissue Regeneration, Tokushima University Graduate School of Biomedical Sciences, Tokushima 770-8504, Japan
| | - Tetsuya Okajima
- Biochemistry II, Nagoya University Graduate School of Medicine, Nagoya 466-0065, Japan
| | - Yuhsuke Ohmi
- Clinical Engineering, Chubu University College of Life and Health Science, Aichi 487-8501, Japan
| | - Keiko Furukawa
- Biomedical Sciences, Chubu University College of Life and Health Sciences, Aichi 487-8501, Japan
| | - Yasusei Kudo
- Oral Bioscience, Tokushima University Graduate School of Biomedical Sciences, Tokushima 770-8504, Japan
| | - Nana Kawasaki
- Biopharmaceutical and Regenerative Sciences, Graduate School of Medical Life Science, Yokohama City University, Yokohama 230-0045, Japan
| | - Koichi Furukawa
- Biochemistry II, Nagoya University Graduate School of Medicine, Nagoya 466-0065, Japan
- Biomedical Sciences, Chubu University College of Life and Health Sciences, Aichi 487-8501, Japan
| |
Collapse
|
13
|
Zhong X, D’Antona AM, Rouse JC. Mechanistic and Therapeutic Implications of Protein and Lipid Sialylation in Human Diseases. Int J Mol Sci 2024; 25:11962. [PMID: 39596031 PMCID: PMC11594235 DOI: 10.3390/ijms252211962] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2024] [Revised: 10/28/2024] [Accepted: 11/05/2024] [Indexed: 11/28/2024] Open
Abstract
Glycan structures of glycoproteins and glycolipids on the surface glycocalyx and luminal sugar layers of intracellular membrane compartments in human cells constitute a key interface between intracellular biological processes and external environments. Sialic acids, a class of alpha-keto acid sugars with a nine-carbon backbone, are frequently found as the terminal residues of these glycoconjugates, forming the critical components of these sugar layers. Changes in the status and content of cellular sialic acids are closely linked to many human diseases such as cancer, cardiovascular, neurological, inflammatory, infectious, and lysosomal storage diseases. The molecular machineries responsible for the biosynthesis of the sialylated glycans, along with their biological interacting partners, are important therapeutic strategies and targets for drug development. The purpose of this article is to comprehensively review the recent literature and provide new scientific insights into the mechanisms and therapeutic implications of sialylation in glycoproteins and glycolipids across various human diseases. Recent advances in the clinical developments of sialic acid-related therapies are also summarized and discussed.
Collapse
Affiliation(s)
- Xiaotian Zhong
- BioMedicine Design, Discovery and Early Development, Pfizer Research and Development, 610 Main Street, Cambridge, MA 02139, USA;
| | - Aaron M. D’Antona
- BioMedicine Design, Discovery and Early Development, Pfizer Research and Development, 610 Main Street, Cambridge, MA 02139, USA;
| | - Jason C. Rouse
- Analytical Research and Development, Biotherapeutics Pharmaceutical Sciences, Pfizer Inc., Andover, MA 01810, USA;
| |
Collapse
|
14
|
Dahlmann F, Griesbach CE, Torres-Boy AY, von Helden G, Peczuh MW, Pagel K, Greis K. Direct Experimental Characterization of a Sialyl Cation. Chemistry 2024:e202403724. [PMID: 39499170 DOI: 10.1002/chem.202403724] [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] [Accepted: 11/01/2024] [Indexed: 11/07/2024]
Abstract
Sialic acids are monosaccharide residues involved in several biological processes. Controlling the stereoselectivity of sialylation reactions is challenging and mechanistic studies on the structure of its intermediate, the sialyl cation, are scarce. Here it is shown that a sialyl cation can be generated and isolated from an ionized sialic acid precursor. This short-lived species is structurally characterized for the first time using cryogenic infrared spectroscopy. In combination with quantum chemical calculations, the results reveal that the positive charge at the anomeric carbon of the sialyl cation is stabilized by remote participation of the C5-NHAc group leading to the formation of a bridged structure. In this structure, the β-side is shielded from nucleophilic attack, potentially explaining the α-selectivity of this building block in SN1-type sialylation reactions. Other modes of participation are energetically unfavored and cannot be observed experimentally.
Collapse
Affiliation(s)
- Franziska Dahlmann
- Department of Chemistry, Yale University, New Haven, 06520, Connecticut, USA
- Institut für Ionenphysik und Angewandte Physik, Universität Innsbruck, 6020, Innsbruck, Austria
- Department of Chemical Engineering, KTH Royal Institute of Technology, 114 28, Stockholm, Sweden
| | - Caleb E Griesbach
- Department of Chemistry, University of Connecticut, 06269, Connecticut, USA
- Department of Chemistry, University of Toronto, M5S 3H6, Toronto, Canada
| | | | - Gert von Helden
- Fritz Haber Institute of the Max Planck Society, 14195, Berlin, Germany
| | - Mark W Peczuh
- Department of Chemistry, University of Connecticut, 06269, Connecticut, USA
| | - Kevin Pagel
- Fritz Haber Institute of the Max Planck Society, 14195, Berlin, Germany
- Institute of Chemistry and Biochemistry, Freie Universität Berlin, 14195, Berlin, Germany
| | - Kim Greis
- Department of Chemistry, Yale University, New Haven, 06520, Connecticut, USA
- Fritz Haber Institute of the Max Planck Society, 14195, Berlin, Germany
- Institute of Chemistry and Biochemistry, Freie Universität Berlin, 14195, Berlin, Germany
- Department of Chemistry and Applied Biosciences, ETH Zürich, 8093, Zürich, Switzerland
| |
Collapse
|
15
|
Kumawat D, Gray TE, Garnier CR, Bui DT, Li Z, Jame-Chenarboo Z, Jerasi J, Wong WO, Klassen JS, Capicciotti CJ, Macauley MS. A Kinetic Trapping Approach for Facile Access to 3F axNeu5Ac and a Photo-Cross-Linkable Sialyltransferase Probe. J Am Chem Soc 2024; 146:28630-28634. [PMID: 39377645 DOI: 10.1021/jacs.4c10835] [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: 10/09/2024]
Abstract
Sialic acid (Neu5Ac) is installed onto glycoconjugates by sialyltransferases (STs) using cytidine monophosphate-Neu5Ac (CMP-β-d-Neu5Ac) as their donor. The only class of cell-active ST inhibitors are those based on a 3FaxNeu5Ac scaffold, which is metabolically converted into CMP-3FaxNeu5Ac within cells. It is essential for the fluorine to be axial, yet stereoselective installation of fluorine in this specific orientation is challenging. Sialic acid aldolase can convert 3-fluoropyruvate and 2-acetamido-2-deoxy-d-mannopyranose (ManNAc) to 3FNeu5Ac, but stereocontrol of the fluorine in the product has not been possible. We hypothesized that the 3Fax kinetic product of a sialic acid aldolase reaction could be trapped by coupling with CMP-sialic acid synthetase to yield CMP-3FaxNeu5Ac. Here, we report that highly active CMP-sialic acid synthetase and short reaction times produce exclusively CMP-3FaxNeu5Ac. Removal of CMP from CMP-3FaxNeu5Ac under acidic conditions unexpectedly led to 3-fluoro-β-d-Neu5Ac 2-phosphate (3FaxNeu5Ac-2P). Alkaline phosphatase successfully converted 3FaxNeu5Ac-2P to 3FaxNeu5Ac, enabling stereochemically controlled access to 3FaxNeu5Ac, which is effective in lowering the sialoglycan ligands for Siglecs on cells. Moreover, our kinetic trapping approach could be used to access CMP-3FaxNeu5Ac with modifications at the C5, C9, or both positions, which enabled the chemoenzymatic synthesis of a photo-cross-linkable version of CMP-3FaxNeu5Ac that selectively photo-cross-linked to ST6GAL1 over two other STs.
Collapse
Affiliation(s)
- Dhanraj Kumawat
- Department of Chemistry, University of Alberta, Edmonton T6G 2G2, Canada
| | - Taylor E Gray
- Department of Chemistry, University of Alberta, Edmonton T6G 2G2, Canada
| | - Cole R Garnier
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston K7L 3N6, Canada
| | - Duong T Bui
- Department of Chemistry, University of Alberta, Edmonton T6G 2G2, Canada
| | - Zhixiong Li
- Department of Chemistry, University of Alberta, Edmonton T6G 2G2, Canada
| | | | - Jeremy Jerasi
- Department of Chemistry, University of Alberta, Edmonton T6G 2G2, Canada
| | - Warren O Wong
- Department of Chemistry, University of Alberta, Edmonton T6G 2G2, Canada
| | - John S Klassen
- Department of Chemistry, University of Alberta, Edmonton T6G 2G2, Canada
| | - Chantelle J Capicciotti
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston K7L 3N6, Canada
- Department of Chemistry, Queen's University, Kingston K7L 2S8, Canada
- Department of Surgery, Queen's University, Kingston K7L 2V7, Canada
| | - Matthew S Macauley
- Department of Chemistry, University of Alberta, Edmonton T6G 2G2, Canada
- Department of Medical Microbiology and Immunology, University of Alberta, Edmonton T6G 2E1, Canada
| |
Collapse
|
16
|
Mohamed KA, Kruf S, Büll C. Putting a cap on the glycome: Dissecting human sialyltransferase functions. Carbohydr Res 2024; 544:109242. [PMID: 39167930 DOI: 10.1016/j.carres.2024.109242] [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: 07/24/2024] [Accepted: 08/13/2024] [Indexed: 08/23/2024]
Abstract
Human glycans are capped with sialic acids and these nine-carbon sugars mediate many of the biological functions and interactions of glycans. Structurally diverse sialic acid caps mark human cells as self and they form the ligands for the Siglec immune receptors and other glycan-binding proteins. Sialic acids enable host interactions with the human microbiome and many human pathogens utilize sialic acids to infect host cells. Alterations in sialic acid-carrying glycans, sialoglycans, can be found in every major human disease including inflammatory conditions and cancer. Twenty sialyltransferase family members in the Golgi apparatus of human cells transfer sialic acids to distinct glycans and glycoconjugates. Sialyltransferases catalyze specific reactions to form unique sialoglycans or they have shared functions where multiple family members generate the same sialoglycan product. Moreover, some sialyltransferases compete for the same glycan substrate, but create different sialic acid caps. The redundant and competing functions make it difficult to understand the individual roles of the human sialyltransferases in biology and to reveal the specific contributions to pathobiological processes. Recent insights hint towards the existence of biosynthetic rules formed by the individual functions of sialyltransferases, their interactions, and cues from the local Golgi environment that coordinate sialoglycan biosynthesis. In this review, we discuss the current structural and functional understanding of the human sialyltransferase family and we review recent technological advances that enable the dissection of individual sialyltransferase activities.
Collapse
Affiliation(s)
- Khadra A Mohamed
- Department of Biomolecular Chemistry, Institute for Molecules and Materials, Faculty of Science, Radboud University Nijmegen, Heyendaalseweg 135, 6525AJ, Nijmegen, the Netherlands
| | - Stijn Kruf
- Department of Biomolecular Chemistry, Institute for Molecules and Materials, Faculty of Science, Radboud University Nijmegen, Heyendaalseweg 135, 6525AJ, Nijmegen, the Netherlands
| | - Christian Büll
- Department of Biomolecular Chemistry, Institute for Molecules and Materials, Faculty of Science, Radboud University Nijmegen, Heyendaalseweg 135, 6525AJ, Nijmegen, the Netherlands.
| |
Collapse
|
17
|
Yao Q, Hu X, Bian T, Zhang Q, Xue Z, Lv Y, Ren S, Chen Y, Zhang D, Chen L. Role of KLF4 and SIAT7A interaction accelerates myocardial hypertrophy induced by Ang II. J Cell Mol Med 2024; 28:e70144. [PMID: 39431583 PMCID: PMC11492152 DOI: 10.1111/jcmm.70144] [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: 01/02/2024] [Revised: 09/05/2024] [Accepted: 10/03/2024] [Indexed: 10/22/2024] Open
Abstract
Sialylation catalysed by sialyltransferase 7A (SIAT7A) plays a role in the development of cardiac hypertrophy. However, the regulatory mechanisms upstream of SIAT7A in this context remain poorly elucidated. Previous study demonstrated that KLF4 activates the SIAT7A gene in ischemic myocardium by binding to its promoter region. Nevertheless, the potential involvement of KLF4 in regulating SIAT7A expression in Ang II-induced hypertrophic cardiomyocytes remains uncertain. This study seeks to deepen the underlying mechanisms of the KLF4 and SIAT7A interaction in the progression of Ang II-induced cardiac hypertrophy. The results showed a concurrent increase in SIAT7A and KLF4 levels in hypertrophic myocardium of essential hypertension patients and in hypertrophic cardiomyocytes stimulated by Ang II. In vitro experiments revealed that reducing KLF4 levels led to a decrease in both SIAT7A synthesis and Sialyl-Tn antigen expression, consequently inhibiting Ang II-induced cardiomyocyte hypertrophy. Intriguingly, reducing SIAT7A levels also resulted in decreased KLF4 expression and suppression cardiomyocyte hypertrophy. Consistent with this, elevating SIAT7A levels increased KLF4 expression and exacerbated cardiomyocyte hypertrophy in both in vivo and in vitro experiments. Additionally, a time-course analysis indicated that KLF4 expression preceded that of SIAT7A. Luciferase reporter assays further confirmed that modulating SIAT7A levels directly influenced the transcriptional activity of KLF4 in cardiomyocytes. In summary, KLF4 expression is upregulated in cardiomyocytes treated with Ang II, which subsequently induces the expression of SIAT7A. The elevated levels of SIAT7A, in turn, enhance the transcription of KLF4. These findings suggest a positive feedback loop between KLF4 and SIAT7A-Sialyl-Tn, ultimately promoting Ang II-induced cardiac hypertrophy.
Collapse
Affiliation(s)
- Qiying Yao
- Department of PhysiologyDalian Medical UniversityDalianLiaoningChina
| | - Xinrui Hu
- Department of PhysiologyDalian Medical UniversityDalianLiaoningChina
| | - Tiantian Bian
- Department of PhysiologyDalian Medical UniversityDalianLiaoningChina
| | - Qing Zhang
- Department of PhysiologyDalian Medical UniversityDalianLiaoningChina
| | - Zhao Xue
- Department of CardiologyThe Second Affiliated Hospital of Dalian Medical UniversityDalianChina
| | - Yuesheng Lv
- Institute of Cancer Stem CellDalian Medical UniversityDalianChina
| | - Shupeng Ren
- Department of CardiologyThe Second Affiliated Hospital of Dalian Medical UniversityDalianChina
| | - Yue Chen
- Department of PhysiologyDalian Medical UniversityDalianLiaoningChina
| | - Dongmei Zhang
- Department of PhysiologyDalian Medical UniversityDalianLiaoningChina
| | - Liang Chen
- Department of CardiologyThe Second Affiliated Hospital of Dalian Medical UniversityDalianChina
| |
Collapse
|
18
|
Wang T, Ran B, Luo Y, Ma J, Li J, Li P, Li M, Li D. Functional study of the ST6GAL2 gene regulating skeletal muscle growth and development. Heliyon 2024; 10:e37311. [PMID: 39296044 PMCID: PMC11407927 DOI: 10.1016/j.heliyon.2024.e37311] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2024] [Revised: 08/30/2024] [Accepted: 08/30/2024] [Indexed: 09/21/2024] Open
Abstract
ST6GAL2, a member of the sialoglycosyltransferase family, primarily localizes within the cellular Golgi apparatus. However, the role of the ST6GAL2 gene in skeletal muscle growth and development remains elusive. In this study, the impact of the ST6GAL2 gene on the proliferation, differentiation, and apoptosis of primary chicken myoblasts at the cellular level was investigated. Quantitative fluorescent PCR was used to measure the expression levels of genes. Subsequently, using gene knockout mice, we assessed its effects on skeletal muscle growth and development in vivo. Our findings reveal that the ST6GAL2 gene promotes the expression of cell cycle and proliferation-related genes, including CCNB2 and PCNA, and apoptosis-related genes, such as Fas and Caspase-9. At the individual level, double knockout of ST6GAL2 inhibited the formation of both fast and slow muscle fibers in the quadriceps, extensor digitorum longus, and tibial anterior muscle, while promoting their formation in the gastrocnemius and soleus. These results collectively demonstrate that the ST6GAL2 gene facilitates the proliferation, apoptosis, and fusion processes of primary chicken myoblasts. Additionally, it promotes the enlargement of cross-sectional muscle fiber areas and regulates the formation of fast and slow muscle fibers at the individual level, albeit inhibiting muscle fusion. This study provides valuable insights into the role of the ST6GAL2 gene in promoting proliferation of skeletal muscle.
Collapse
Affiliation(s)
- Tao Wang
- School of Pharmacy, Chengdu University, Chengdu, 610106, China
| | - Bo Ran
- Sichuan Animal Science Academy, Chengdu, 610066, China
| | - Yingyu Luo
- State Key Laboratory of Swine and Poultry Breeding Industry, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, 611130, China
| | - Jideng Ma
- State Key Laboratory of Swine and Poultry Breeding Industry, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, 611130, China
| | - Jing Li
- State Key Laboratory of Swine and Poultry Breeding Industry, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, 611130, China
| | - Penghao Li
- Jinxin Research Institute for Reproductive Medicine and Genetics, Chengdu Xi Nan Gynecological Hospital Co., Ltd., 66 Bisheng Road, Chengdu, 610000, China
| | - Mingzhou Li
- State Key Laboratory of Swine and Poultry Breeding Industry, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, 611130, China
| | - Diyan Li
- School of Pharmacy, Chengdu University, Chengdu, 610106, China
| |
Collapse
|
19
|
Bonab MKF, Guo Z, Li Q. Glycosphingolipids: from metabolism to chemoenzymatic total synthesis. Org Biomol Chem 2024; 22:6665-6683. [PMID: 39120686 PMCID: PMC11341264 DOI: 10.1039/d4ob00695j] [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] [Indexed: 08/10/2024]
Abstract
GSLs are the major glycolipids in vertebrates and mediate many key biological processes from intercellular recognition to cis regulation of signal transduction. The fast-expanding field of glycobiology has led to a growing demand for diverse and structurally defined GSLs, and enzymatic GSL synthesis is developing rapidly in accordance. This article provides an overview of natural GSL biosynthetic pathways and surveys the bacterial enzymes applied to GSL synthesis and recent progress in synthesis strategies. By correlating these three areas, this article aims to define the gaps between GSL biosynthesis and chemoenzymatic synthesis and evaluate the opportunities for harnessing natural forces to access GSLs efficiently.
Collapse
Affiliation(s)
- Mitra K F Bonab
- Department of Chemistry, University of Massachusetts Boston, Boston, Massachusetts 02125, USA.
| | - Zhongwu Guo
- Department of Chemistry, University of Florida, Gainesville, Florida 32611, USA.
| | - Qingjiang Li
- Department of Chemistry, University of Massachusetts Boston, Boston, Massachusetts 02125, USA.
| |
Collapse
|
20
|
Szabo R, Dobie C, Montgomery AP, Steele H, Yu H, Skropeta D. Synthesis of α-Hydroxy-1,2,3-Triazole-linked Sialyltransferase Inhibitors and Evaluation of Selectivity Towards ST3GAL1, ST6GAL1 and ST8SIA2. ChemMedChem 2024; 19:e202400088. [PMID: 38758134 DOI: 10.1002/cmdc.202400088] [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: 01/30/2024] [Revised: 05/15/2024] [Accepted: 05/15/2024] [Indexed: 05/18/2024]
Abstract
Tumour-derived sialoglycans, bearing the charged nonulosonic sugar sialic acid at their termini, play a critical role in tumour cell adhesion and invasion, as well as evading cell death and immune surveillance. Sialyltransferases (ST), the enzymes responsible for the biosynthesis of sialylated glycans, are highly upregulated in cancer, with tumour hypersialylation strongly correlated with tumour growth, metastasis and drug resistance. As a result, desialylation of the tumour cell surface using either targeted delivery of a pan-ST inhibitor (or sialidase) or systemic delivery of a non-toxic selective ST inhibitors are being pursued as potential new anti-metastatic strategies against multiple cancers including pancreatic, ovarian, breast, melanoma and lung cancer. Herein, we have employed molecular modelling to give insights into the selectivity observed in a series of selective ST inhibitors that incorporate a uridyl ring in place of the cytidine of the natural donor (CMP-Neu5Ac) and replace the charged phosphodiester linker of classical ST inhibitors with a neutral α-hydroxy-1,2,3-triazole linker. The inhibitory activities of the nascent compounds were determined against recombinant human ST enzymes (ST3GAL1, ST6GAL1, ST8SIA2) showing promising activity and selectivity towards specific ST sub-types. Our ST inhibitors are non-toxic and show improved synthetic accessibility and drug-likeness compared to earlier nucleoside-based ST inhibitors.
Collapse
Affiliation(s)
- Rémi Szabo
- School of Chemistry & Molecular Bioscience, University of Wollongong, Wollongong, NSW, Australia
| | - Chris Dobie
- School of Chemistry & Molecular Bioscience, University of Wollongong, Wollongong, NSW, Australia
- Molecular Horizons, University of Wollongong, Wollongong, NSW, Australia
| | - Andrew P Montgomery
- School of Chemistry & Molecular Bioscience, University of Wollongong, Wollongong, NSW, Australia
| | - Harrison Steele
- School of Chemistry & Molecular Bioscience, University of Wollongong, Wollongong, NSW, Australia
| | - Haibo Yu
- School of Chemistry & Molecular Bioscience, University of Wollongong, Wollongong, NSW, Australia
- Molecular Horizons, University of Wollongong, Wollongong, NSW, Australia
- ARC Centre of Excellence in Quantum Biotechnology, University of Wollongong, Wollongong, NSW, Australia
| | - Danielle Skropeta
- School of Chemistry & Molecular Bioscience, University of Wollongong, Wollongong, NSW, Australia
- Molecular Horizons, University of Wollongong, Wollongong, NSW, Australia
| |
Collapse
|
21
|
Irons EE, Sajina GC, Lau JT. Sialic acid in the regulation of blood cell production, differentiation and turnover. Immunology 2024; 172:517-532. [PMID: 38503445 PMCID: PMC11223974 DOI: 10.1111/imm.13780] [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: 11/27/2023] [Accepted: 03/04/2024] [Indexed: 03/21/2024] Open
Abstract
Sialic acid is a unique sugar moiety that resides in the distal and most accessible position of the glycans on mammalian cell surface and extracellular glycoproteins and glycolipids. The potential for sialic acid to obscure underlying structures has long been postulated, but the means by which such structural changes directly affect biological processes continues to be elucidated. Here, we appraise the growing body of literature detailing the importance of sialic acid for the generation, differentiation, function and death of haematopoietic cells. We conclude that sialylation is a critical post-translational modification utilized in haematopoiesis to meet the dynamic needs of the organism by enforcing rapid changes in availability of lineage-specific cell types. Though long thought to be generated only cell-autonomously within the intracellular ER-Golgi secretory apparatus, emerging data also demonstrate previously unexpected diversity in the mechanisms of sialylation. Emphasis is afforded to the mechanism of extrinsic sialylation, whereby extracellular enzymes remodel cell surface and extracellular glycans, supported by charged sugar donor molecules from activated platelets.
Collapse
Affiliation(s)
| | | | - Joseph T.Y. Lau
- Department of Molecular and Cellular Biology, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14203 USA
| |
Collapse
|
22
|
Fox A, Leonard GD, Adzibolosu N, Wong T, Tedja R, Sharma S, Gogoi R, Morris R, Mor G, Fehl C, Alvero AB. Adipose microenvironment promotes hypersialylation of ovarian cancer cells. Front Oncol 2024; 14:1432333. [PMID: 39104719 PMCID: PMC11299042 DOI: 10.3389/fonc.2024.1432333] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2024] [Accepted: 06/28/2024] [Indexed: 08/07/2024] Open
Abstract
Introduction Ovarian and other peritoneal cancers have a strong tendency to metastasize into the surrounding adipose tissue. This study describes an effect of the adipose microenvironment on upregulation of sialic acid-containing glycans in ovarian cancer (OC). Heterogeneous populations of glycosylated OC tumors converged to a highly sialylated cell state that regulates tumorigenesis in an immune-dependent manner. Methods We modeled the adipose microenvironment by conditioning growth media with human patient-derived adipose tissue. OC cell lines grown in the presence vs. absence of adipose conditioned media (ACM) were characterized by transcriptomics, western blotting, and chemical biology glycan labeling methods. Fluorescence-activated cell sorting was used to separate adipose-driven upregulation of hypersialylated ("SNA-high") vs. hyposialylated ("SNA-low") OC subpopulations. The two subpopulations were characterized by further transcriptomic and quantitative polymerase chain reaction analyses, then injected into a syngeneic mouse model. Immune system involvement was implicated using wild type and athymic nude mice with a primary endpoint of overall survival. Results Adipose conditioning resulted in upregulation of sialyltransferases ST3GAL1, ST6GAL1, ST6GALNAC3, and ST8Sia1. In culture, OC cells displayed two distinct sialylated subpopulations that were stable for up to 9 passages, suggesting inherent heterogeneity in sialylation that is maintained throughout cell division and media changes. OC tumors that implanted in the omental adipose tissue exclusively reprogrammed to the highly sialylated subpopulation. In wild type C57BL/6 mice, only the hypersialylated SNA-high subpopulation implanted in the adipose, whereas the hyposialylated SNA-low subpopulation failed to be tumorigenic (p=0.023, n=5). In the single case where SNA-low established a tumor, post-mortem analysis revealed reprogramming of the tumor to the SNA-high state in vivo. In athymic nude mice, both subpopulations rapidly formed tumors, implicating a role of the adaptive immune system. Conclusions These findings suggest a model of glycan-dependent tumor evolution wherein the adipose microenvironment reprograms OC to a tumorigenic state that resists the adaptive immune system. Mechanistically, adipose factors upregulate sialyltransferases. To our knowledge, this is the first demonstration of the effect of adipose microenvironment on OC tumor sialylation. Our results set the stage for translational applications targeting sialic acid pathways in OC and other peritoneal cancer tumorigenesis and metastasis.
Collapse
Affiliation(s)
- Alexandra Fox
- Department of Obstetrics and Gynecology, Wayne State University, Detroit, MI, United States
| | - Garry D. Leonard
- Department of Chemistry, Wayne State University, Detroit, MI, United States
| | - Nicholas Adzibolosu
- Department of Obstetrics and Gynecology, Wayne State University, Detroit, MI, United States
| | - Terrence Wong
- Department of Obstetrics and Gynecology, Wayne State University, Detroit, MI, United States
- Karmanos Cancer Institute, Detroit, MI, United States
| | - Roslyn Tedja
- Department of Obstetrics and Gynecology, Wayne State University, Detroit, MI, United States
- Karmanos Cancer Institute, Detroit, MI, United States
| | - Sapna Sharma
- Department of Obstetrics and Gynecology, Wayne State University, Detroit, MI, United States
| | - Radhika Gogoi
- Department of Obstetrics and Gynecology, Wayne State University, Detroit, MI, United States
- Karmanos Cancer Institute, Detroit, MI, United States
| | - Robert Morris
- Karmanos Cancer Institute, Detroit, MI, United States
| | - Gil Mor
- Department of Obstetrics and Gynecology, Wayne State University, Detroit, MI, United States
- Karmanos Cancer Institute, Detroit, MI, United States
| | - Charlie Fehl
- Department of Chemistry, Wayne State University, Detroit, MI, United States
- Karmanos Cancer Institute, Detroit, MI, United States
| | - Ayesha B. Alvero
- Department of Obstetrics and Gynecology, Wayne State University, Detroit, MI, United States
- Karmanos Cancer Institute, Detroit, MI, United States
| |
Collapse
|
23
|
Kasahara T, Chang TC, Yoshioka H, Urano S, Egawa Y, Inoue M, Tahara T, Morimoto K, Pradipta AR, Tanaka K. Anticancer approach by targeted activation of a global inhibitor of sialyltransferases with acrolein. Chem Sci 2024; 15:9566-9573. [PMID: 38939146 PMCID: PMC11206204 DOI: 10.1039/d4sc00969j] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2024] [Accepted: 04/28/2024] [Indexed: 06/29/2024] Open
Abstract
Cells are covered with a thick layer of sugar molecules known as glycans. Abnormal glycosylation is a hallmark of cancer, and hypersialylation increases tumor metastasis by promoting immune evasion and inducing tumor cell invasion and migration. Inhibiting sialylation is thus a potential anticancer treatment strategy. However, targeting sialic acids is difficult because of the lack of selective delivery tools. Here, we present a prodrug strategy for selectively releasing the global inhibitor of sialylation peracetylated 3Fax-Neu5Ac (PFN) in cancer cells using the reaction between phenyl azide and endogenous acrolein, which is overproduced in most cancer cells. The prodrug significantly suppressed tumor growth in mice as effectively as PFN without causing kidney dysfunction, which is associated with PFN. The use of sialylated glycans as immune checkpoints is gaining increasing attention, and the proposed method for precisely targeting aberrant sialylation provides a novel avenue for expanding current cancer treatments.
Collapse
Affiliation(s)
- Takatsugu Kasahara
- Department of Chemical Science and Engineering, School of Materials and Chemical Technology, Tokyo Institute of Technology 2-12-1 Ookayama, Meguro-ku Tokyo 152-8552 Japan
| | - Tsung-Che Chang
- Department of Chemical Science and Engineering, School of Materials and Chemical Technology, Tokyo Institute of Technology 2-12-1 Ookayama, Meguro-ku Tokyo 152-8552 Japan
- Biofunctional Synthetic Chemistry Laboratory, RIKEN Cluster for Pioneering Research 2-1 Hirosawa Wako-shi Saitama 351-0198 Japan
| | - Hiromasa Yoshioka
- Biofunctional Synthetic Chemistry Laboratory, RIKEN Cluster for Pioneering Research 2-1 Hirosawa Wako-shi Saitama 351-0198 Japan
| | - Sayaka Urano
- Biofunctional Synthetic Chemistry Laboratory, RIKEN Cluster for Pioneering Research 2-1 Hirosawa Wako-shi Saitama 351-0198 Japan
| | - Yasuko Egawa
- Biofunctional Synthetic Chemistry Laboratory, RIKEN Cluster for Pioneering Research 2-1 Hirosawa Wako-shi Saitama 351-0198 Japan
| | - Michiko Inoue
- Laboratory for Biofunction Dynamics Imaging, RIKEN Center for Biosystems Dynamics Research 6-7-3 Minatojima-minamimachi, Chuo-ku Kobe 650-0047 Japan
| | - Tsuyoshi Tahara
- Department of In vivo Imaging, Advanced Research Promoting Center, Tokushima University 3-18-15 Kuramto-cho Tokushima Tokushima 770-8503 Japan
| | - Koji Morimoto
- Biofunctional Synthetic Chemistry Laboratory, RIKEN Cluster for Pioneering Research 2-1 Hirosawa Wako-shi Saitama 351-0198 Japan
| | - Ambara R Pradipta
- Department of Chemical Science and Engineering, School of Materials and Chemical Technology, Tokyo Institute of Technology 2-12-1 Ookayama, Meguro-ku Tokyo 152-8552 Japan
| | - Katsunori Tanaka
- Department of Chemical Science and Engineering, School of Materials and Chemical Technology, Tokyo Institute of Technology 2-12-1 Ookayama, Meguro-ku Tokyo 152-8552 Japan
- Biofunctional Synthetic Chemistry Laboratory, RIKEN Cluster for Pioneering Research 2-1 Hirosawa Wako-shi Saitama 351-0198 Japan
| |
Collapse
|
24
|
Perez SJLP, Chen CL, Chang TT, Li WS. Biological evaluation of sulfonate and sulfate analogues of lithocholic acid: A bioisosterism-guided approach towards the discovery of potential sialyltransferase inhibitors for antimetastatic study. Bioorg Med Chem Lett 2024; 105:129760. [PMID: 38641151 DOI: 10.1016/j.bmcl.2024.129760] [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: 08/27/2023] [Revised: 02/12/2024] [Accepted: 04/16/2024] [Indexed: 04/21/2024]
Abstract
The naturally occurring bile acid lithocholic acid (LCA) has been a crucial core structure for many non-sugar-containing sialyltranferase (ST) inhibitors documented in literature. With the aim of elucidating the impact of the terminal carboxyl acid substituent of LCA on its ST inhibition, in this present study, we report the (bio)isosteric replacement-based design and synthesis of sulfonate and sulfate analogues of LCA. Among these compounds, the sulfate analogue SPP-002 was found to selectively inhibit N-glycan sialylation by at least an order of magnitude, indicating a substantial improvement in both potency and selectivity when compared to the unmodified parent bile acid. Molecular docking analysis supported the stronger binding of the synthetic analogue in the enzyme active site. Treatment with SPP-002 also hampered the migration, adhesion, and invasion of MDA-MB-231 cells in vitro by suppressing the expression of signaling proteins involved in the cancer metastasis-associated integrin/FAK/paxillin pathway. In totality, these findings offer not only a novel structural scaffold but also valuable insights for the future development of more potent and selective ST inhibitors with potential therapeutic effects against tumor cancer metastasis.
Collapse
Affiliation(s)
- Ser John Lynon P Perez
- Institute of Chemistry, Academia Sinica, Taipei 115, Taiwan; Department of Applied Chemistry, National Yang Ming Chiao Tung University, Hsinchu 300, Taiwan; Sustainable Chemical Science and Technology, Taiwan International Graduate Program, Academia Sinica, Taipei 115, Taiwan; Biomedical Translation Research Center, Academia Sinica, Taipei 115, Taiwan
| | - Chia-Ling Chen
- Institute of Chemistry, Academia Sinica, Taipei 115, Taiwan
| | - Tzu-Ting Chang
- Biomedical Translation Research Center, Academia Sinica, Taipei 115, Taiwan
| | - Wen-Shan Li
- Institute of Chemistry, Academia Sinica, Taipei 115, Taiwan; Sustainable Chemical Science and Technology, Taiwan International Graduate Program, Academia Sinica, Taipei 115, Taiwan; Biomedical Translation Research Center, Academia Sinica, Taipei 115, Taiwan; Department of Medicinal and Applied Chemistry, College of Life Science, Kaohsiung Medical University, Kaohsiung 807, Taiwan; Ph.D. Program in Drug Discovery and Development Industry, College of Pharmacy, Taipei Medical University; Doctoral Degree Program in Marine Biotechnology, National Sun Yat-Sen University, Kaohsiung 804, Taiwan; Department of Chemistry, College of Science, Tamkang University, New Taipei City 251, Taiwan.
| |
Collapse
|
25
|
Pei C, Peng X, Wu Y, Jiao R, Li T, Jiao S, Zhou L, Li J, Du Y, Qian EW. Characterization and application of active human α2,6-sialyltransferases ST6GalNAc V and ST6GalNAc VI recombined in Escherichia coli. Enzyme Microb Technol 2024; 177:110426. [PMID: 38503081 DOI: 10.1016/j.enzmictec.2024.110426] [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: 01/09/2024] [Revised: 02/25/2024] [Accepted: 03/11/2024] [Indexed: 03/21/2024]
Abstract
Eukaryotic sialyltransferases play key roles in many physiological and pathological events. The expression of active human recombinant sialyltransferases in bacteria is still challenging. In the current study, the genes encoding human N-acetylgalactosaminide α2,6-sialyltransferase V (hST6GalNAc V) and N-acetylgalactosaminide α2,6-sialyltransferase VI (hST6GalNAc VI) lacking the N-terminal transmembrane domains were cloned into the expression vectors, pET-32a and pET-22b, respectively. Soluble and active forms of recombinant hST6GalNAc V and hST6GalNAc VI when coexpressed with the chaperone plasmid pGro7 were successfully achieved in Escherichia coli. Further, lactose (Lac), Lacto-N-triose II (LNT II), lacto-N-tetraose (LNT), and sialyllacto-N-tetraose a (LSTa) were used as acceptor substrates to investigate their activities and substrate specificities. Unexpectedly, both can transfer sialic acid onto all those substrates. Compared with hST6GalNAc V expressed in the mammalian cells, the recombinant two α2,6-sialyltransferases in bacteria displayed flexible substrate specificities and lower enzymatic efficiency. In addition, an important human milk oligosaccharide disialyllacto-N-tetraose (DSLNT) can be synthesized by both human α2,6-sialyltransferases expressed in E. coli using LSTa as an acceptor substrate. To the best of our knowledge, these two active human α2,6-sialyltransferases enzymes were expressed in bacteria for the first time. They showed a high potential to be applied in biotechnology and investigating the molecular mechanisms of biological and pathological interactions related to sialylated glycoconjugates.
Collapse
Affiliation(s)
- Caixia Pei
- Graduate School of Bio-Applications and Systems Engineering, Tokyo University of Agriculture and Technology, Nakacho 2-24-16, Koganei, Tokyo 184-8588, Japan; State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing 100190, China; Key Laboratory of Biopharmaceutical Preparation and Delivery, Chinese Academy of Sciences, Beijing 100190, China
| | - Xinlv Peng
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing 100190, China; Key Laboratory of Biopharmaceutical Preparation and Delivery, Chinese Academy of Sciences, Beijing 100190, China; School of Chemical Engineering, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Yiran Wu
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing 100190, China; Key Laboratory of Biopharmaceutical Preparation and Delivery, Chinese Academy of Sciences, Beijing 100190, China; School of Chemical Engineering, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Runmiao Jiao
- Graduate School of Bio-Applications and Systems Engineering, Tokyo University of Agriculture and Technology, Nakacho 2-24-16, Koganei, Tokyo 184-8588, Japan; State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing 100190, China; Key Laboratory of Biopharmaceutical Preparation and Delivery, Chinese Academy of Sciences, Beijing 100190, China
| | - Tiehai Li
- Carbohydrate-Based Drug Research Center, CAS Key Laboratory of Receptor Research, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
| | - Siming Jiao
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing 100190, China; Key Laboratory of Biopharmaceutical Preparation and Delivery, Chinese Academy of Sciences, Beijing 100190, China; Innovation Academy for Green Manufacture Institute, Chinese Academy of Sciences, Beijing 100190, China
| | - Lei Zhou
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing 100190, China; Key Laboratory of Biopharmaceutical Preparation and Delivery, Chinese Academy of Sciences, Beijing 100190, China; Innovation Academy for Green Manufacture Institute, Chinese Academy of Sciences, Beijing 100190, China
| | - Jianjun Li
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing 100190, China; Key Laboratory of Biopharmaceutical Preparation and Delivery, Chinese Academy of Sciences, Beijing 100190, China; Innovation Academy for Green Manufacture Institute, Chinese Academy of Sciences, Beijing 100190, China.
| | - Yuguang Du
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing 100190, China; Key Laboratory of Biopharmaceutical Preparation and Delivery, Chinese Academy of Sciences, Beijing 100190, China; Innovation Academy for Green Manufacture Institute, Chinese Academy of Sciences, Beijing 100190, China.
| | - Eika W Qian
- Graduate School of Bio-Applications and Systems Engineering, Tokyo University of Agriculture and Technology, Nakacho 2-24-16, Koganei, Tokyo 184-8588, Japan.
| |
Collapse
|
26
|
Mitry MMA, Dallas ML, Boateng SY, Greco F, Osborn HMI. Selective activation of prodrugs in breast cancer using metabolic glycoengineering and the tetrazine ligation bioorthogonal reaction. Bioorg Chem 2024; 147:107304. [PMID: 38643563 DOI: 10.1016/j.bioorg.2024.107304] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2024] [Revised: 03/14/2024] [Accepted: 03/19/2024] [Indexed: 04/23/2024]
Abstract
Increasing the selectivity of chemotherapies by converting them into prodrugs that can be activated at the tumour site decreases their side effects and allows discrimination between cancerous and non-cancerous cells. Herein, the use of metabolic glycoengineering (MGE) to selectively label MCF-7 breast cancer cells with tetrazine (Tz) activators for subsequent activation of prodrugs containing the trans-cyclooctene (TCO) moiety by a bioorthogonal reaction is demonstrated. Three novel Tz-modified monosaccharides, Ac4ManNTz 7, Ac4GalNTz 8, and Ac4SiaTz 16, were used for expression of the Tz activator within sialic-acid rich breast cancer cells' surface glycans through MGE. Tz expression on breast cancer cells (MCF-7) was evaluated versus the non-cancerous L929 fibroblasts showing a concentration-dependant effect and excellent selectivity with ≥35-fold Tz expression on the MCF-7 cells versus the non-cancerous L929 fibroblasts. Next, a novel TCO-N-mustard prodrug and a TCO-doxorubicin prodrug were analyzed in vitro on the Tz-bioengineered cells to probe our hypothesis that these could be activated via a bioorthogonal reaction. Selective prodrug activation and restoration of cytotoxicity were demonstrated for the MCF-7 breast cancer cells versus the non-cancerous L929 cells. Restoration of the parent drug's cytotoxicity was shown to be dependent on the level of Tz expression where the Ac4ManNTz 7 and Ac4GalNTz 8 derivatives (20 µM) lead to the highest Tz expression and full restoration of the parent drug's cytotoxicity. This work suggests the feasibility of combining MGE and tetrazine ligation for selective prodrug activation in breast cancer.
Collapse
Affiliation(s)
- Madonna M A Mitry
- Reading School of Pharmacy, University of Reading, Whiteknights, Reading RG6 6AD. UK; Dept. of Pharmaceutical Chemistry, Faculty of Pharmacy, Ain Shams University, Cairo 11566, Egypt.
| | - Mark L Dallas
- Reading School of Pharmacy, University of Reading, Whiteknights, Reading RG6 6AD. UK.
| | - Samuel Y Boateng
- School of Biological Sciences, University of Reading, Whiteknights, Reading RG6 6UB, UK.
| | - Francesca Greco
- Reading School of Pharmacy, University of Reading, Whiteknights, Reading RG6 6AD. UK.
| | - Helen M I Osborn
- Reading School of Pharmacy, University of Reading, Whiteknights, Reading RG6 6AD. UK.
| |
Collapse
|
27
|
Isaji T, Gu J. Novel regulatory mechanisms of N-glycan sialylation: Implication of integrin and focal adhesion kinase in the regulation. Biochim Biophys Acta Gen Subj 2024; 1868:130617. [PMID: 38614280 DOI: 10.1016/j.bbagen.2024.130617] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2024] [Revised: 04/05/2024] [Accepted: 04/09/2024] [Indexed: 04/15/2024]
Abstract
BACKGROUND Sialylation of glycoproteins, including integrins, is crucial in various cancers and diseases such as immune disorders. These modifications significantly impact cellular functions and are associated with cancer progression. Sialylation, catalyzed by specific sialyltransferases (STs), has traditionally been considered to be regulated at the mRNA level. SCOPE OF REVIEW Recent research has expanded our understanding of sialylation, revealing ST activity changes beyond mRNA level variations. This includes insights into COPI vesicle formation and Golgi apparatus maintenance and identifying specific target proteins of STs that are not predictable through recombinant enzyme assays. MAJOR CONCLUSIONS This review summarizes that Golgi-associated pathways largely influence the regulation of STs. GOLPH3, GORAB, PI4K, and FAK have become critical elements in sialylation regulation. Some STs have been revealed to possess specificity for specific target proteins, suggesting the presence of additional, enzyme-specific regulatory mechanisms. GENERAL SIGNIFICANCE This study enhances our understanding of the molecular interplay in sialylation regulation, mainly focusing on the role of integrin and FAK. It proposes a bidirectional system where sialylations might influence integrins and vice versa. The diversity of STs and their specific linkages offer new perspectives in cancer research, potentially broadening our understanding of cellular mechanisms and opening avenues for new therapeutic approaches in targeting sialylation pathways.
Collapse
Affiliation(s)
- Tomoya Isaji
- Division of Regulatory Glycobiology, Institute of Molecular Biomembrane and Glycobiology, Tohoku Medical and Pharmaceutical University, 4-4-1 Komatsushima, Aoba-ku, Sendai, Miyagi 981-8558, Japan.
| | - Jianguo Gu
- Division of Regulatory Glycobiology, Institute of Molecular Biomembrane and Glycobiology, Tohoku Medical and Pharmaceutical University, 4-4-1 Komatsushima, Aoba-ku, Sendai, Miyagi 981-8558, Japan.
| |
Collapse
|
28
|
Fox A, Leonard GD, Adzibolosu N, Wong T, Tedja R, Sharma S, Gogoi R, Morris R, Mor G, Fehl C, Alvero AB. Adipose microenvironment promotes hypersialylation of ovarian cancer cells. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.13.593990. [PMID: 38798490 PMCID: PMC11118282 DOI: 10.1101/2024.05.13.593990] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/29/2024]
Abstract
Sialylation, the addition of negatively charged sialic acid sugars to terminal ends of glycans, is upregulated in most cancers. Hypersialylation supports multiple pro-tumor mechanisms such as enhanced migration and invasion, resistance to apoptosis and immune evasion. A current gap in knowledge is the lack of understanding on how the tumor microenvironment regulates cancer cell sialylation. The adipose niche is a main component of most peritoneal cancers' microenvironment. This includes ovarian cancer (OC), which causes most deaths from all gynecologic cancers. In this report, we demonstrate that the adipose microenvironment is a critical regulator of OC cell sialylation. In vitro adipose conditioning led to an increase in both ⍺2,3- and ⍺2,6-linked cell surface sialic acids in both human and mouse models of OC. Adipose-induced sialylation reprogramming was also observed in vivo from intra-peritoneal OC tumors seeded in the adipose-rich omentum. Mechanistically, we observed upregulation of at least three sialyltransferases, ST3GAL1, ST6GAL1 and ST3GALNAC3. Hypersialylated OC cells consistently formed intra-peritoneal tumors in both immune-competent mice and immune-compromised athymic nude mice. In contrast, hyposiaylated OC cells persistently formed tumors only in athymic nude mice demonstrating that sialylation impacts OC tumor formation in an immune dependent manner. To our knowledge, this is the first demonstration of the effect of adipose microenvironment on OC tumor sialylation. Our results set the stage for translational applications targeting sialic acid pathways in OC and other peritoneal cancers.
Collapse
|
29
|
He Y, Miao C, Yang S, Xu C, Liu Y, Zhu X, Wen Y, Wu R, Zhao Q, Huang X, Yan Q, Lang Y, Zhao S, Wang Y, Han X, Cao S, Hu Y, Du S. Sialic acids as attachment factors in mosquitoes mediating Japanese encephalitis virus infection. J Virol 2024; 98:e0195923. [PMID: 38634598 PMCID: PMC11092328 DOI: 10.1128/jvi.01959-23] [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: 12/14/2023] [Accepted: 03/26/2024] [Indexed: 04/19/2024] Open
Abstract
The role of Culex mosquitoes in the transmission of Japanese encephalitis virus (JEV) is crucial, yet the mechanisms of JEV infection in these vectors remain unclear. Previous research has indicated that various host factors participate in JEV infection. Herein, we present evidence that mosquito sialic acids enhance JEV infection both in vivo and in vitro. By treating mosquitoes and C6/36 cells with neuraminidase or lectin, the function of sialic acids is effectively blocked, resulting in significant inhibition of JEV infection. Furthermore, knockdown of the sialic acid biosynthesis genes in Culex mosquitoes also leads to a reduction in JEV infection. Moreover, our research revealed that sialic acids play a role in the attachment of JEV to mosquito cells, but not in its internalization. To further explore the mechanisms underlying the promotion of JEV attachment by sialic acids, we conducted immunoprecipitation experiments to confirm the direct binding of sialic acids to the last α-helix in JEV envelope protein domain III. Overall, our study contributes to a molecular comprehension of the interaction between mosquitoes and JEV and offers potential strategies for preventing the dissemination of flavivirus in natural environments.IMPORTANCEIn this study, we aimed to investigate the impact of glycoconjugate sialic acids on mosquito infection with Japanese encephalitis virus (JEV). Our findings demonstrate that sialic acids play a crucial role in enhancing JEV infection by facilitating the attachment of the virus to the cell membrane. Furthermore, our investigation revealed that sialic acids directly bind to the final α-helix in the JEV envelope protein domain III, thereby accelerating virus adsorption. Collectively, our results highlight the significance of mosquito sialic acids in JEV infection within vectors, contributing to a better understanding of the interaction between mosquitoes and JEV.
Collapse
Affiliation(s)
- Yi He
- Research Center for Swine Diseases, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| | - Chang Miao
- Research Center for Swine Diseases, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| | - Shiping Yang
- Research Center for Swine Diseases, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| | - Changhao Xu
- Research Center for Swine Diseases, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| | - Yuwei Liu
- Research Center for Swine Diseases, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| | - Xi Zhu
- Research Center for Swine Diseases, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| | - Yiping Wen
- Research Center for Swine Diseases, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
- Sichuan Science-Observation Experimental Station for Veterinary Drugs and Veterinary Diagnostic Technology, Ministry of Agriculture, Chengdu, China
- Engineering Research Center of Southwest Animal Disease Prevention and Control Technology, Ministry of Education of the People’s Republic of China, Chengdu, China
| | - Rui Wu
- Research Center for Swine Diseases, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
- Sichuan Science-Observation Experimental Station for Veterinary Drugs and Veterinary Diagnostic Technology, Ministry of Agriculture, Chengdu, China
- Engineering Research Center of Southwest Animal Disease Prevention and Control Technology, Ministry of Education of the People’s Republic of China, Chengdu, China
| | - Qin Zhao
- Research Center for Swine Diseases, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
- Sichuan Science-Observation Experimental Station for Veterinary Drugs and Veterinary Diagnostic Technology, Ministry of Agriculture, Chengdu, China
- Engineering Research Center of Southwest Animal Disease Prevention and Control Technology, Ministry of Education of the People’s Republic of China, Chengdu, China
| | - Xiaobo Huang
- Research Center for Swine Diseases, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
- Sichuan Science-Observation Experimental Station for Veterinary Drugs and Veterinary Diagnostic Technology, Ministry of Agriculture, Chengdu, China
- Engineering Research Center of Southwest Animal Disease Prevention and Control Technology, Ministry of Education of the People’s Republic of China, Chengdu, China
| | - Qigui Yan
- Research Center for Swine Diseases, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
- Sichuan Science-Observation Experimental Station for Veterinary Drugs and Veterinary Diagnostic Technology, Ministry of Agriculture, Chengdu, China
- Engineering Research Center of Southwest Animal Disease Prevention and Control Technology, Ministry of Education of the People’s Republic of China, Chengdu, China
| | - Yifei Lang
- Research Center for Swine Diseases, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
- Sichuan Science-Observation Experimental Station for Veterinary Drugs and Veterinary Diagnostic Technology, Ministry of Agriculture, Chengdu, China
- Engineering Research Center of Southwest Animal Disease Prevention and Control Technology, Ministry of Education of the People’s Republic of China, Chengdu, China
| | - Shan Zhao
- Research Center for Swine Diseases, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
- Sichuan Science-Observation Experimental Station for Veterinary Drugs and Veterinary Diagnostic Technology, Ministry of Agriculture, Chengdu, China
- Engineering Research Center of Southwest Animal Disease Prevention and Control Technology, Ministry of Education of the People’s Republic of China, Chengdu, China
| | - Yiping Wang
- Research Center for Swine Diseases, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
- Sichuan Science-Observation Experimental Station for Veterinary Drugs and Veterinary Diagnostic Technology, Ministry of Agriculture, Chengdu, China
- Engineering Research Center of Southwest Animal Disease Prevention and Control Technology, Ministry of Education of the People’s Republic of China, Chengdu, China
| | - Xinfeng Han
- Sichuan Science-Observation Experimental Station for Veterinary Drugs and Veterinary Diagnostic Technology, Ministry of Agriculture, Chengdu, China
- Engineering Research Center of Southwest Animal Disease Prevention and Control Technology, Ministry of Education of the People’s Republic of China, Chengdu, China
| | - Sanjie Cao
- Research Center for Swine Diseases, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
- Sichuan Science-Observation Experimental Station for Veterinary Drugs and Veterinary Diagnostic Technology, Ministry of Agriculture, Chengdu, China
- Engineering Research Center of Southwest Animal Disease Prevention and Control Technology, Ministry of Education of the People’s Republic of China, Chengdu, China
| | - Yajie Hu
- Sichuan Center for Disease Control and Prevention, Chengdu, China
| | - Senyan Du
- Research Center for Swine Diseases, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
- Sichuan Science-Observation Experimental Station for Veterinary Drugs and Veterinary Diagnostic Technology, Ministry of Agriculture, Chengdu, China
- Engineering Research Center of Southwest Animal Disease Prevention and Control Technology, Ministry of Education of the People’s Republic of China, Chengdu, China
| |
Collapse
|
30
|
Wang B, Hou C, Yu X, Liu J, Wang J. The prognostic value of sialylation-related long non-coding RNAs in lung adenocarcinoma. Sci Rep 2024; 14:8879. [PMID: 38632255 PMCID: PMC11024174 DOI: 10.1038/s41598-024-59130-3] [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: 11/13/2023] [Accepted: 04/08/2024] [Indexed: 04/19/2024] Open
Abstract
There has been increasing interest in the role of epigenetic modification in cancers recently. Among the various modifications, sialylation has emerged as a dominant subtype implicated in tumor progression, metastasis, immune evasion, and chemoresistance. The prognostic significance of sialylation-related molecules has been demonstrated in colorectal cancer. However, the potential roles and regulatory mechanisms of sialylation in lung adenocarcinoma (LUAD) have not been thoroughly investigated. Through Pearson correlation, univariate Cox hazards proportional regression, and random survival forest model analyses, we identified several prognostic long non-coding RNAs (lncRNAs) associated with aberrant sialylation and tumor progression, including LINC00857, LINC00968, LINC00663, and ITGA9-AS1. Based on the signatures of four lncRNAs, we classified patients into two clusters with different landscapes using a non-negative matrix factorization approach. Collectively, patients in Cluster 1 (C1) exhibited worse prognoses than those in Cluster 2 (C2), as well as heavier tumor mutation burden. Functional enrichment analysis showed the enrichment of several pro-tumor pathways in C1, differing from the upregulated Longevity and programmed cell death pathways in C2. Moreover, we profiled immune infiltration levels of important immune cell lineages in two subgroups using MCPcounter scores and single sample gene set enrichment analysis scores, revealing a relatively immunosuppressive microenvironment in C1. Risk analysis indicated that LINC00857 may serve as a pro-tumor regulator, while the other three lncRNAs may be protective contributors. Consistently, we observed upregulated LINC00857 in C1, whereas increased expressive levels of LINC00968, LINC00663, and ITGA9-AS1 were observed in C2. Finally, drug sensitivity analysis suggested that patients in the two groups may benefit from different therapeutic strategies, contributing to precise treatment in LUAD. By integrating multi-omics data, we identified four core sialylation-related lncRNAs and successfully established a prognostic model to distinguish patients with different characterizations. These findings may provide some insights into the underlying mechanism of sialylation, and offer a new stratification way as well as clinical guidance in LUAD.
Collapse
Grants
- 2022ZD08 National Traditional Chinese Medicine Inheritance and Innovation Center, the First Clinical Medical College of Guangzhou University of Traditional Chinese Medicine, China
- 2022ZD08 National Traditional Chinese Medicine Inheritance and Innovation Center, the First Clinical Medical College of Guangzhou University of Traditional Chinese Medicine, China
- 2022ZD08 National Traditional Chinese Medicine Inheritance and Innovation Center, the First Clinical Medical College of Guangzhou University of Traditional Chinese Medicine, China
- 20241105 Administration of Traditional Chinese Medicine of Guangdong Province, China
- 20241105 Administration of Traditional Chinese Medicine of Guangdong Province, China
- 20221402 Science and Technology Planning Project of Guangdong Province, China
- 20221402 Science and Technology Planning Project of Guangdong Province, China
- 20221402 Science and Technology Planning Project of Guangdong Province, China
Collapse
Affiliation(s)
- Beiru Wang
- First Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou, 510405, Guangdong, China
| | - Chengyu Hou
- First Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou, 510405, Guangdong, China
| | - Xiang Yu
- First Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou, 510405, Guangdong, China
| | - Jiaxin Liu
- First Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou, 510405, Guangdong, China
| | - Jiyong Wang
- Department of Thoracic Surgery, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, 510405, Guangdong, China.
| |
Collapse
|
31
|
Tsai HE, Chen CL, Chang TT, Fu CW, Chen WC, Perez SJLP, Hsiao PW, Tai MH, Li WS. Development of a Novel, Potent, and Selective Sialyltransferase Inhibitor for Suppressing Cancer Metastasis. Int J Mol Sci 2024; 25:4283. [PMID: 38673867 PMCID: PMC11050067 DOI: 10.3390/ijms25084283] [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: 03/03/2024] [Revised: 04/01/2024] [Accepted: 04/08/2024] [Indexed: 04/28/2024] Open
Abstract
Sialyltransferase-catalyzed membrane protein and lipid glycosylation plays a vital role as one of the most abundant post-translational modifications and diversification reactions in eukaryotes. However, aberrant sialylation has been associated with cancer malignancy and metastasis. Sialyltransferases thus represent emerging targets for the development of small molecule cancer drugs. Herein, we report the inhibitory effects of a recently discovered lithocholic acid derivative FCW393 on sialyltransferase catalytic activity, integrin sialyation, cancer-associated signal transduction, MDA-MB-231 and B16F10 cell migration and invasion, and in in vivo studies, on tumor growth, metastasis, and angiogenesis. FCW393 showed effective and selective inhibition of the sialyltransferases ST6GAL1 (IC50 = 7.8 μM) and ST3GAL3 (IC50 = 9.45 μM) relative to ST3GAL1 (IC50 > 400 μM) and ST8SIA4 (IC50 > 100 μM). FCW393 reduced integrin sialylation in breast cancer and melanoma cells dose-dependently and downregulated proteins associated with the integrin-regulated FAK/paxillin and GEF/Rho/ROCK pathways, and with the VEGF-regulated Akt/NFκB/HIF-1α pathway. FCW393 inhibited cell migration (IC50 = 2.6 μM) and invasion in in vitro experiments, and in in vivo studies of tumor-bearing mice, FCW393 reduced tumor size, angiogenesis, and metastatic potential. Based on its demonstrated selectivity, cell permeability, relatively low cytotoxicity (IC50 = 55 μM), and high efficacy, FCW393 shows promising potential as a small molecule experimental tool compound and a lead for further development of a novel cancer therapeutic.
Collapse
Grants
- AS-KPQ-110-EIMD, AS-KPQ-109-BioMed, AS-KPQ-110-BioMed and AS-KPQ-111-KNT Academia Sinica
- MOST, Taiwan, MOST 110-0210-01-22-02, MOST-108-3114-Y-001-002, MOST 108-3111-Y-001-056, MOST 106-2113-M-001-011, MOST 103-2325-B-001-001 and MOST108-2314-B-110-003-MY2 Ministry of Science and Technology, TAIWAN
- 108-36 Kaohsiung Armed Forces General Hospital, TAIWAN
Collapse
Affiliation(s)
- Han-En Tsai
- Institute of Chemistry, Academia Sinica, Taipei 115, Taiwan (C.-W.F.); (S.J.L.P.P.)
| | - Chia-Ling Chen
- Institute of Chemistry, Academia Sinica, Taipei 115, Taiwan (C.-W.F.); (S.J.L.P.P.)
| | - Tzu-Ting Chang
- Biomedical Translation Research Center, Academia Sinica, National Biotechnology Research Park, Taipei 115, Taiwan
| | - Chih-Wei Fu
- Institute of Chemistry, Academia Sinica, Taipei 115, Taiwan (C.-W.F.); (S.J.L.P.P.)
- Department of Chemistry, National Central University, Taoyuan 320, Taiwan
| | - Wei-Chia Chen
- Institute of Chemistry, Academia Sinica, Taipei 115, Taiwan (C.-W.F.); (S.J.L.P.P.)
- Department of Chemistry, National Taiwan Normal University, Taipei 106, Taiwan
| | - Ser John Lynon P. Perez
- Institute of Chemistry, Academia Sinica, Taipei 115, Taiwan (C.-W.F.); (S.J.L.P.P.)
- Biomedical Translation Research Center, Academia Sinica, National Biotechnology Research Park, Taipei 115, Taiwan
- Department of Applied Chemistry, National Yang Ming Chiao Tung University, Hsinchu 300, Taiwan
- Sustainable Chemical Science and Technology, Taiwan International Graduate Program, National Yang Ming Chiao Tung University and Academia Sinica, Taipei 115, Taiwan
| | - Pei-Wen Hsiao
- Agricultural Biotechnology Research Center, Academia Sinica, Taipei 115, Taiwan
| | - Ming-Hong Tai
- Doctoral Degree Program in Marine Biotechnology, National Sun Yat-Sen University, Kaohsiung 804, Taiwan
- Center for Neuroscience, National Sun Yat-Sen University, Kaohsiung 804, Taiwan
- Institute of Biomedical Sciences, National Sun Yat-Sen University, Kaohsiung 804, Taiwan
| | - Wen-Shan Li
- Institute of Chemistry, Academia Sinica, Taipei 115, Taiwan (C.-W.F.); (S.J.L.P.P.)
- Biomedical Translation Research Center, Academia Sinica, National Biotechnology Research Park, Taipei 115, Taiwan
- Sustainable Chemical Science and Technology, Taiwan International Graduate Program, National Yang Ming Chiao Tung University and Academia Sinica, Taipei 115, Taiwan
- Doctoral Degree Program in Marine Biotechnology, National Sun Yat-Sen University, Kaohsiung 804, Taiwan
- Ph.D. Program in Drug Discovery and Development Industry, College of Pharmacy, Taipei Medical University, Taipei 110, Taiwan
- Department of Medicinal and Applied Chemistry, College of Life Science, Kaohsiung Medical University, Kaohsiung 807, Taiwan
| |
Collapse
|
32
|
Liu H, Li X, Ren Y, Yang Y, Chen Y, Ju H. In Situ Visualization of RNA-Specific Sialylation on Living Cell Membranes to Explore N-Glycosylation Sites. J Am Chem Soc 2024; 146:8780-8786. [PMID: 38497732 DOI: 10.1021/jacs.4c01826] [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: 03/19/2024]
Abstract
The small RNAs on living cell membranes were recently found to be N-glycosylated and terminated with sialic acids, although the glycosylation sites and potential functions remain unclear. Herein, we designed a second-generation hierarchical coding strategy (HieCo 2) for in situ visualization of cell surface RNA-specific sialylation. After covalently binding DNA codes to sialic acids and then binding a DNA code to a target RNA via sequence specificity, cascade decoding processes were performed with subsequent signal amplification that enabled sensitive in situ visualization of low-abundance Y5 RNA-specific sialic acids on living cell membranes. The proposed strategy unveils the number of glycosylation sites on a single RNA and reveals the binding preference of glycosylated RNAs to different sialic acid binding-immunoglobulin lectin-type receptors, demonstrating a new route for exploration of the glycosylated RNA-related biological and pathological processes.
Collapse
Affiliation(s)
- Huipu Liu
- State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing 210023, P. R. China
| | - Xuemei Li
- Shandong Province Key Laboratory of Detection Technology for Tumor Markers, School of Chemistry and Chemical Engineering, Linyi University, Linyi 276005, P. R. China
| | - Yi Ren
- State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing 210023, P. R. China
| | - Yuanjiao Yang
- State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing 210023, P. R. China
| | - Yunlong Chen
- State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing 210023, P. R. China
| | - Huangxian Ju
- State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing 210023, P. R. China
| |
Collapse
|
33
|
Agrawal P, Chen S, de Pablos A, Jame-Chenarboo F, Miera Saenz de Vega E, Darvishian F, Osman I, Lujambio A, Mahal LK, Hernando E. Integrated in vivo functional screens and multi-omics analyses identify α-2,3-sialylation as essential for melanoma maintenance. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.03.08.584072. [PMID: 38559078 PMCID: PMC10979837 DOI: 10.1101/2024.03.08.584072] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/04/2024]
Abstract
Glycosylation is a hallmark of cancer biology, and altered glycosylation influences multiple facets of melanoma growth and progression. To identify glycosyltransferases, glycans, and glycoproteins essential for melanoma maintenance, we conducted an in vivo growth screen with a pooled shRNA library of glycosyltransferases, lectin microarray profiling of benign nevi and melanoma patient samples, and mass spectrometry-based glycoproteomics. We found that α-2,3 sialyltransferases ST3GAL1 and ST3GAL2 and corresponding α-2,3-linked sialosides are upregulated in melanoma compared to nevi and are essential for melanoma growth in vivo and in vitro. Glycoproteomics revealed that glycoprotein targets of ST3GAL1 and ST3GAL2 are enriched in transmembrane proteins involved in growth signaling, including the amino acid transporter Solute Carrier Family 3 Member 2 (SLC3A2/CD98hc). CD98hc suppression mimicked the effect of ST3GAL1 and ST3GAL2 silencing, inhibiting melanoma cell proliferation. We found that both CD98hc protein stability and its pro-survival effect in melanoma are dependent upon α-2,3 sialylation mediated by ST3GAL1 and ST3GAL2. In summary, our studies reveal that α-2,3-sialosides functionally contribute to melanoma maintenance, supporting ST3GAL1 and ST3GAL2 as novel therapeutic targets in these tumors.
Collapse
Affiliation(s)
- Praveen Agrawal
- Department of Pathology, NYU Grossman School of Medicine, New York
- Interdisciplinary Melanoma Cooperative Group, Perlmutter Cancer Center, NYU Langone Health
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, New York
| | - Shuhui Chen
- Department of Chemistry, New York University
| | - Ana de Pablos
- Department of Pathology, NYU Grossman School of Medicine, New York
- Interdisciplinary Melanoma Cooperative Group, Perlmutter Cancer Center, NYU Langone Health
- Centro Nacional de Investigaciones Oncologicas (CNIO), Madrid, Spain
| | | | | | | | - Iman Osman
- Interdisciplinary Melanoma Cooperative Group, Perlmutter Cancer Center, NYU Langone Health
- Department of Dermatology, NYU Grossman School of Medicine, New York
| | | | - Lara K. Mahal
- Department of Chemistry, New York University
- Department of Chemistry, University of Alberta, Edmonton, Canada
| | - Eva Hernando
- Department of Pathology, NYU Grossman School of Medicine, New York
- Interdisciplinary Melanoma Cooperative Group, Perlmutter Cancer Center, NYU Langone Health
| |
Collapse
|
34
|
Teng D, Wang W, Jia W, Song J, Gong L, Zhong L, Yang J. The effects of glycosylation modifications on monocyte recruitment and foam cell formation in atherosclerosis. Biochim Biophys Acta Mol Basis Dis 2024; 1870:167027. [PMID: 38237743 DOI: 10.1016/j.bbadis.2024.167027] [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: 09/16/2023] [Revised: 01/10/2024] [Accepted: 01/10/2024] [Indexed: 01/23/2024]
Abstract
The monocyte recruitment and foam cell formation have been intensively investigated in atherosclerosis. Nevertheless, as the study progressed, it was obvious that crucial molecules participated in the monocyte recruitment and the membrane proteins in macrophages exhibited substantial glycosylation modifications. These modifications can exert a significant influence on protein functions and may even impact the overall progression of diseases. This article provides a review of the effects of glycosylation modifications on monocyte recruitment and foam cell formation. By elaborating on these effects, we aim to understand the underlying mechanisms of atherogenesis further and to provide new insights into the future treatment of atherosclerosis.
Collapse
Affiliation(s)
- Da Teng
- Yantai Yuhuangding Hospital affiliated to Qingdao University, Yantai, Shandong, People's Republic of China; Qingdao University, Qingdao, Shandong, People's Republic of China
| | - Wenlong Wang
- Yantai Yuhuangding Hospital affiliated to Qingdao University, Yantai, Shandong, People's Republic of China; Qingdao University, Qingdao, Shandong, People's Republic of China
| | - Wenjuan Jia
- Yantai Yuhuangding Hospital affiliated to Qingdao University, Yantai, Shandong, People's Republic of China; Qingdao University, Qingdao, Shandong, People's Republic of China
| | - Jikai Song
- Yantai Yuhuangding Hospital affiliated to Qingdao University, Yantai, Shandong, People's Republic of China; Qingdao University, Qingdao, Shandong, People's Republic of China
| | - Lei Gong
- Yantai Yuhuangding Hospital affiliated to Qingdao University, Yantai, Shandong, People's Republic of China
| | - Lin Zhong
- Yantai Yuhuangding Hospital affiliated to Qingdao University, Yantai, Shandong, People's Republic of China.
| | - Jun Yang
- Yantai Yuhuangding Hospital affiliated to Qingdao University, Yantai, Shandong, People's Republic of China; Qingdao University, Qingdao, Shandong, People's Republic of China.
| |
Collapse
|
35
|
Choi H, Song KH, Kim HD, Park JY, Lee YC, Choi HJ, Kim CH. Human ST3Gal II and ST6GalNAc IV genes increase human serum-mediated cytotoxicity to xenogeneic cells. Xenotransplantation 2024; 31:exen12855. [PMID: 38602029 DOI: 10.1111/xen.12855] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Revised: 01/24/2024] [Accepted: 02/10/2024] [Indexed: 04/12/2024]
Abstract
Carbohydrate-antigens widely existed on glycoproteins and glycosphingolipids of all mammalian cells play a crucial role in self-defense and immunity. Xeno-reactive antibodies included in natural human sera play a protecting role in an acute phase-rejection of xenotransplantation. In this study, we investigated the effect of an alteration of glycosylation-pattern, caused by human sialyltransferases such as hST3Gal II or hST6GalNAc IV, on human serum mediated cytotoxicity in pig kidney PK15 cells. From LDH cytotoxicity assay, cytotoxicity to human serum was significantly increased in hST3Gal II and hST6GalNAc IV-transfected PK15 cells, as compared to the control. In the hST6Gal I-carrying cells, the cytotoxicity to human serum was rather decreased. Moreover, flow cytometry analysis revealed that an alteration of pig glycosylation-pattern by hST3Gal II or hST6GalNAc IV influences on a binding of human IgM or IgG, respectively, in pig kidney cells, regardless of Gal antigen alteration. Finally, we found that hST6GalNAc IV contributed to increase of terminal disialylated tetrasaccharide structure, disialyl T antigen, as evidenced by increase of the MAL II lectin binding capacity in the hST6GalNAc IV-transfected PK15 cells, compared with control. Therefore, our results suggest that carbohydrate antigens, such as disialyl T antigen, newly synthesized by the ST3Gal II- and ST6GalNAc IV are potentially believed to be new xeno-reactive elements.
Collapse
Affiliation(s)
- Hyunju Choi
- Molecular and Cellular Glycobiology Unit, Department of Biological Sciences, SungKyunKwan University, Suwon, South Korea
| | - Kwon-Ho Song
- Molecular and Cellular Glycobiology Unit, Department of Biological Sciences, SungKyunKwan University, Suwon, South Korea
- Department of Cell Biology, Daegu Catholic University School of Medicine, Daegu, South Korea
| | - Hee-Do Kim
- Molecular and Cellular Glycobiology Unit, Department of Biological Sciences, SungKyunKwan University, Suwon, South Korea
| | - Jun-Young Park
- Molecular and Cellular Glycobiology Unit, Department of Biological Sciences, SungKyunKwan University, Suwon, South Korea
| | - Young-Choon Lee
- Department of Medicinal Biotechnology, College of Health Sciences, Dong-A University, Saha-Gu, Busan, South Korea
| | - Hee-Jung Choi
- Jin BioCell Co., Ltd. R&D Center, #101-103, National Clinical Research Center for Korean Medicine, Pusan National University Korean Medicine Hospital, Yangsan, Gyeongsangnam-do, Republic of Korea
| | - Cheorl-Ho Kim
- Molecular and Cellular Glycobiology Unit, Department of Biological Sciences, SungKyunKwan University, Suwon, South Korea
| |
Collapse
|
36
|
Baboo S, Diedrich JK, Torres JL, Copps J, Singh B, Garrett PT, Ward AB, Paulson JC, Yates JR. Evolving spike-protein N-glycosylation in SARS-CoV-2 variants. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.05.08.539897. [PMID: 37214937 PMCID: PMC10197516 DOI: 10.1101/2023.05.08.539897] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/24/2023]
Abstract
Since >3 years, SARS-CoV-2 has plunged humans into a colossal pandemic. Henceforth, multiple waves of infection have swept through the human population, led by variants that were able to partially evade acquired immunity. The co-evolution of SARS-CoV-2 variants with human immunity provides an excellent opportunity to study the interaction between viral pathogens and their human hosts. The heavily N-glycosylated spike-protein of SARS-CoV-2 plays a pivotal role in initiating infection and is the target for host immune-response, both of which are impacted by host-installed N-glycans. Using highly-sensitive DeGlyPHER approach, we compared the N-glycan landscape on spikes of the SARS-CoV-2 Wuhan-Hu-1 strain to seven WHO-defined variants of concern/interest, using recombinantly expressed, soluble spike-protein trimers, sharing same stabilizing-mutations. We found that N-glycan processing is conserved at most sites. However, in multiple variants, processing of N-glycans from high mannose- to complex-type is reduced at sites N165, N343 and N616, implicated in spike-protein function.
Collapse
Affiliation(s)
- Sabyasachi Baboo
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, California 92037, United States
| | - Jolene K. Diedrich
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, California 92037, United States
| | - Jonathan L. Torres
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, California 92037, United States
| | - Jeffrey Copps
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, California 92037, United States
| | - Bhavya Singh
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, California 92037, United States
| | - Patrick T. Garrett
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, California 92037, United States
| | - Andrew B. Ward
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, California 92037, United States
| | - James C. Paulson
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, California 92037, United States
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, California 92037, United States
| | - John R. Yates
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, California 92037, United States
| |
Collapse
|
37
|
An SY, Lee JW, Kim HD, Kim KS, Cho JH, Kim CH, Lee YC. Regulatory mechanism for the human glioblastoma cell-specific expression of the human GD1c/GT1a/GQ1b synthase (hST8Sia V) gene. Glycoconj J 2023; 40:621-630. [PMID: 37921922 DOI: 10.1007/s10719-023-10136-5] [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: 07/28/2023] [Revised: 10/05/2023] [Accepted: 10/19/2023] [Indexed: 11/05/2023]
Abstract
In this study we observed that human GD1c/GT1a/GQ1b synthase (hST8Sia V) is particularly expressed in human glioblastoma cells. To address the mechanism regulating human glioblastoma-specific gene expression of the hST8Sia V, after the transcription start site (TSS) was identified by the 5'-rapid amplification of cDNA end with total RNA from human glioblastoma U87MG cells, the 5'-flanking region (2.5 kb) of the hST8Sia V gene was isolated and its promoter activity was examined. By luciferase reporter assay, this 5'-flanking region revealed strong promoter activity in only U-87MG cells, but not in other tissue-derived cancer cells. 5'-deletion mutant analysis showed that the region from -1140 to -494 is crucial for transcription of the hST8Sia V gene in U87MG cells. This region contains the activator protein-1 (AP-1) binding site, the main target of the c-Jun N-terminal kinase (JNK) downstream. The AP-1 binding site at -1043/-1037 was proved to be indispensable for the hST8Sia V gene-specific expression in U87MG cells by site-directed mutagenesis. Moreover, the transcriptional activation of hST8Sia V gene in U87MG cells was strongly inhibited by a specific JNK inhibitor, SP600125. These results suggest that the hST8Sia V gene-specific expression in U87MG cells is controlled by JNK/AP-1 signaling pathway.
Collapse
Affiliation(s)
- So-Young An
- Department of Medicinal Biotechnology, College of Health Sciences, Dong-A University, Busan, 49315, South Korea
| | - Ji-Won Lee
- Department of Medicinal Biotechnology, College of Health Sciences, Dong-A University, Busan, 49315, South Korea
| | - Hee-Do Kim
- Molecular and Cellular Glycobiology Unit, Department of Biological Sciences, SungKyunKwan University, Kyunggi-Do, 16419, South Korea
| | - Kyoung-Sook Kim
- Department of Medicinal Biotechnology, College of Health Sciences, Dong-A University, Busan, 49315, South Korea
| | - Jong-Hyun Cho
- Department of Medicinal Biotechnology, College of Health Sciences, Dong-A University, Busan, 49315, South Korea
| | - Cheorl-Ho Kim
- Molecular and Cellular Glycobiology Unit, Department of Biological Sciences, SungKyunKwan University, Kyunggi-Do, 16419, South Korea.
| | - Young-Choon Lee
- Department of Medicinal Biotechnology, College of Health Sciences, Dong-A University, Busan, 49315, South Korea.
| |
Collapse
|
38
|
Kuliesiute U, Joseph K, Straehle J, Madapusi Ravi V, Kueckelhaus J, Kada Benotmane J, Zhang J, Vlachos A, Beck J, Schnell O, Neniskyte U, Heiland DH. Sialic acid metabolism orchestrates transcellular connectivity and signaling in glioblastoma. Neuro Oncol 2023; 25:1963-1975. [PMID: 37288604 PMCID: PMC10628944 DOI: 10.1093/neuonc/noad101] [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: 11/02/2022] [Indexed: 06/09/2023] Open
Abstract
BACKGROUND In glioblastoma (GBM), the effects of altered glycocalyx are largely unexplored. The terminal moiety of cell coating glycans, sialic acid, is of paramount importance for cell-cell contacts. However, sialic acid turnover in gliomas and its impact on tumor networks remain unknown. METHODS We streamlined an experimental setup using organotypic human brain slice cultures as a framework for exploring brain glycobiology, including metabolic labeling of sialic acid moieties and quantification of glycocalyx changes. By live, 2-photon and high-resolution microscopy we have examined morphological and functional effects of altered sialic acid metabolism in GBM. By calcium imaging we investigated the effects of the altered glycocalyx on a functional level of GBM networks. RESULTS The visualization and quantitative analysis of newly synthesized sialic acids revealed a high rate of de novo sialylation in GBM cells. Sialyltrasferases and sialidases were highly expressed in GBM, indicating that significant turnover of sialic acids is involved in GBM pathology. Inhibition of either sialic acid biosynthesis or desialylation affected the pattern of tumor growth and lead to the alterations in the connectivity of glioblastoma cells network. CONCLUSIONS Our results indicate that sialic acid is essential for the establishment of GBM tumor and its cellular network. They highlight the importance of sialic acid for glioblastoma pathology and suggest that dynamics of sialylation have the potential to be targeted therapeutically.
Collapse
Affiliation(s)
- Ugne Kuliesiute
- Microenvironment and Immunology Research Laboratory, Medical Center, University of Freiburg, Freiburg, Germany
- Department of Neurosurgery, Medical Center, University of Freiburg, Freiburg, Germany
- Faculty of Medicine, Freiburg University, Freiburg, Germany
- Institute of Biosciences, Life Sciences Center, Vilnius University, Vilnius, Lithuania
- VU LSC-EMBL Partnership for Genome Editing Technologies, Life Sciences Center, Vilnius University, Vilnius, Lithuania
| | - Kevin Joseph
- Microenvironment and Immunology Research Laboratory, Medical Center, University of Freiburg, Freiburg, Germany
- Department of Neurosurgery, Medical Center, University of Freiburg, Freiburg, Germany
- Faculty of Medicine, Freiburg University, Freiburg, Germany
- Department of Neuroanatomy, Institute of Anatomy and Cell Biology, Faculty of Medicine, University of Freiburg, Freiburg, Germany
- Center Brain Links Brain Tools, University of Freiburg, Freiburg, Germany
| | - Jakob Straehle
- Department of Neurosurgery, Medical Center, University of Freiburg, Freiburg, Germany
- Faculty of Medicine, Freiburg University, Freiburg, Germany
- Department of Neuroanatomy, Institute of Anatomy and Cell Biology, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Vidhya Madapusi Ravi
- Microenvironment and Immunology Research Laboratory, Medical Center, University of Freiburg, Freiburg, Germany
- Department of Neurosurgery, Medical Center, University of Freiburg, Freiburg, Germany
- Faculty of Medicine, Freiburg University, Freiburg, Germany
- Department of Neuroanatomy, Institute of Anatomy and Cell Biology, Faculty of Medicine, University of Freiburg, Freiburg, Germany
- Center Brain Links Brain Tools, University of Freiburg, Freiburg, Germany
| | - Jan Kueckelhaus
- Microenvironment and Immunology Research Laboratory, Medical Center, University of Freiburg, Freiburg, Germany
- Department of Neurosurgery, Medical Center, University of Freiburg, Freiburg, Germany
- Faculty of Medicine, Freiburg University, Freiburg, Germany
- Department of Neuroanatomy, Institute of Anatomy and Cell Biology, Faculty of Medicine, University of Freiburg, Freiburg, Germany
- Center Brain Links Brain Tools, University of Freiburg, Freiburg, Germany
| | - Jasim Kada Benotmane
- Microenvironment and Immunology Research Laboratory, Medical Center, University of Freiburg, Freiburg, Germany
- Department of Neurosurgery, Medical Center, University of Freiburg, Freiburg, Germany
- Faculty of Medicine, Freiburg University, Freiburg, Germany
- Department of Neuroanatomy, Institute of Anatomy and Cell Biology, Faculty of Medicine, University of Freiburg, Freiburg, Germany
- Center Brain Links Brain Tools, University of Freiburg, Freiburg, Germany
| | - Junyi Zhang
- Microenvironment and Immunology Research Laboratory, Medical Center, University of Freiburg, Freiburg, Germany
- Department of Neurosurgery, Medical Center, University of Freiburg, Freiburg, Germany
- Faculty of Medicine, Freiburg University, Freiburg, Germany
- Department of Neuroanatomy, Institute of Anatomy and Cell Biology, Faculty of Medicine, University of Freiburg, Freiburg, Germany
- Center Brain Links Brain Tools, University of Freiburg, Freiburg, Germany
| | - Andreas Vlachos
- Department of Neuroanatomy, Institute of Anatomy and Cell Biology, Faculty of Medicine, University of Freiburg, Freiburg, Germany
- Center Brain Links Brain Tools, University of Freiburg, Freiburg, Germany
- Center for Basics in Neuromodulation, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Juergen Beck
- Department of Neurosurgery, Medical Center, University of Freiburg, Freiburg, Germany
- Faculty of Medicine, Freiburg University, Freiburg, Germany
- Department of Neuroanatomy, Institute of Anatomy and Cell Biology, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Oliver Schnell
- Microenvironment and Immunology Research Laboratory, Medical Center, University of Freiburg, Freiburg, Germany
- Department of Neurosurgery, Medical Center, University of Freiburg, Freiburg, Germany
- Faculty of Medicine, Freiburg University, Freiburg, Germany
| | - Urte Neniskyte
- Institute of Biosciences, Life Sciences Center, Vilnius University, Vilnius, Lithuania
- VU LSC-EMBL Partnership for Genome Editing Technologies, Life Sciences Center, Vilnius University, Vilnius, Lithuania
| | - Dieter Henrik Heiland
- Microenvironment and Immunology Research Laboratory, Medical Center, University of Freiburg, Freiburg, Germany
- Department of Neurosurgery, Medical Center, University of Freiburg, Freiburg, Germany
- Faculty of Medicine, Freiburg University, Freiburg, Germany
- Department of Neurological Surgery, Lou and Jean Malnati Brain Tumor Institute, Robert H. Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, Illinois
- Comprehensive Cancer Center Freiburg (CCCF), Faculty of Medicine and Medical Center—University of Freiburg, Freiburg, Germany
- German Cancer Consortium (DKTK), partner siteFreiburg
| |
Collapse
|
39
|
Naber A, Demus D, Slieker R, Nicolardi S, Beulens JWJ, Elders PJM, Lieverse AG, Sijbrands EJG, 't Hart LM, Wuhrer M, van Hoek M. Apolipoprotein-CIII O-Glycosylation, a Link between GALNT2 and Plasma Lipids. Int J Mol Sci 2023; 24:14844. [PMID: 37834292 PMCID: PMC10573541 DOI: 10.3390/ijms241914844] [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: 08/15/2023] [Revised: 09/29/2023] [Accepted: 09/30/2023] [Indexed: 10/15/2023] Open
Abstract
Apolipoprotein-CIII (apo-CIII) is involved in triglyceride-rich lipoprotein metabolism and linked to beta-cell damage, insulin resistance, and cardiovascular disease. Apo-CIII exists in four main proteoforms: non-glycosylated (apo-CIII0a), and glycosylated apo-CIII with zero, one, or two sialic acids (apo-CIII0c, apo-CIII1 and apo-CIII2). Our objective is to determine how apo-CIII glycosylation affects lipid traits and type 2 diabetes prevalence, and to investigate the genetic basis of these relations with a genome-wide association study (GWAS) on apo-CIII glycosylation. We conducted GWAS on the four apo-CIII proteoforms in the DiaGene study in people with and without type 2 diabetes (n = 2318). We investigated the relations of the identified genetic loci and apo-CIII glycosylation with lipids and type 2 diabetes. The associations of the genetic variants with lipids were replicated in the Diabetes Care System (n = 5409). Rs4846913-A, in the GALNT2-gene, was associated with decreased apo-CIII0a. This variant was associated with increased high-density lipoprotein cholesterol and decreased triglycerides, while high apo-CIII0a was associated with raised high-density lipoprotein-cholesterol and triglycerides. Rs67086575-G, located in the IFT172-gene, was associated with decreased apo-CIII2 and with hypertriglyceridemia. In line, apo-CIII2 was associated with low triglycerides. On a genome-wide scale, we confirmed that the GALNT2-gene plays a major role i O-glycosylation of apolipoprotein-CIII, with subsequent associations with lipid parameters. We newly identified the IFT172/NRBP1 region, in the literature previously associated with hypertriglyceridemia, as involved in apolipoprotein-CIII sialylation and hypertriglyceridemia. These results link genomics, glycosylation, and lipid metabolism, and represent a key step towards unravelling the importance of O-glycosylation in health and disease.
Collapse
Affiliation(s)
- Annemieke Naber
- Department of Internal Medicine, Erasmus MC University Medical Center Rotterdam, P.O. Box 2040, 3000 CA Rotterdam, The Netherlands
| | - Daniel Demus
- Center for Proteomics and Metabolomics, Leiden University Medical Center, P.O. Box 9600, 2300 RC Leiden, The Netherlands
| | - Roderick Slieker
- Department of Cell and Chemical Biology, Leiden University Medical Center, P.O. Box 9600, 2300 RC Leiden, The Netherlands
- Department of Epidemiology and Data Science, Amsterdam UMC, Location Vrije Universiteit Amsterdam, P.O. Box 7057, 1007 MB Amsterdam, The Netherlands
| | - Simone Nicolardi
- Center for Proteomics and Metabolomics, Leiden University Medical Center, P.O. Box 9600, 2300 RC Leiden, The Netherlands
| | - Joline W J Beulens
- Department of Epidemiology and Data Science, Amsterdam UMC, Location Vrije Universiteit Amsterdam, P.O. Box 7057, 1007 MB Amsterdam, The Netherlands
- Amsterdam Public Health, Amsterdam Cardiovascular Sciences, Meibergdreef 9, 1105 AZ Amsterdam, The Netherlands
- Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, P.O. Box 85500, 3508 GA Utrecht, The Netherlands
| | - Petra J M Elders
- Department of General Practice, Amsterdam Public Health Institute, Amsterdam UMC, Location VUmc, P.O. Box 7057, 1007 MB Amsterdam, The Netherlands
| | - Aloysius G Lieverse
- Department of Internal Medicine, Maxima Medical Center, P.O. Box 90052, 5600 PD Eindhoven, The Netherlands
| | - Eric J G Sijbrands
- Department of Internal Medicine, Erasmus MC University Medical Center Rotterdam, P.O. Box 2040, 3000 CA Rotterdam, The Netherlands
| | - Leen M 't Hart
- Department of Cell and Chemical Biology, Leiden University Medical Center, P.O. Box 9600, 2300 RC Leiden, The Netherlands
- Department of Epidemiology and Data Science, Amsterdam UMC, Location Vrije Universiteit Amsterdam, P.O. Box 7057, 1007 MB Amsterdam, The Netherlands
- Amsterdam Public Health, Amsterdam Cardiovascular Sciences, Meibergdreef 9, 1105 AZ Amsterdam, The Netherlands
- Department of Biomedical Data Science, Section Molecular Epidemiology, Leiden University Medical Center, Postal Zone S5-P, P.O. Box 9600, 2300 RC Leiden, The Netherlands
| | - Manfred Wuhrer
- Center for Proteomics and Metabolomics, Leiden University Medical Center, P.O. Box 9600, 2300 RC Leiden, The Netherlands
| | - Mandy van Hoek
- Department of Internal Medicine, Erasmus MC University Medical Center Rotterdam, P.O. Box 2040, 3000 CA Rotterdam, The Netherlands
| |
Collapse
|
40
|
Decloquement M, Venuto MT, Cogez V, Steinmetz A, Schulz C, Lion C, Noel M, Rigolot V, Teppa RE, Biot C, Rebl A, Galuska SP, Harduin-Lepers A. Salmonid polysialyltransferases to generate a variety of sialic acid polymers. Sci Rep 2023; 13:15610. [PMID: 37730806 PMCID: PMC10511417 DOI: 10.1038/s41598-023-42095-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2023] [Accepted: 09/05/2023] [Indexed: 09/22/2023] Open
Abstract
The human polysialyltransferases ST8Sia II and ST8Sia IV catalyze the transfer of several Neu5Ac residues onto glycoproteins forming homopolymers with essential roles during different physiological processes. In salmonids, heterogeneous set of sialic acids polymers have been described in ovary and on eggs cell surface and three genes st8sia4, st8sia2-r1 and st8sia2-r2 were identified that could be implicated in these heteropolymers. The three polysialyltransferases from the salmonid Coregonus maraena were cloned, recombinantly expressed in HEK293 cells and the ST8Sia IV was biochemically characterized. The MicroPlate Sialyltransferase Assay and the non-natural donor substrate CMP-SiaNAl were used to demonstrate enzyme activity and optimize polysialylation reactions. Polysialylation was also carried out with natural donor substrates CMP-Neu5Ac, CMP-Neu5Gc and CMP-Kdn in cell-free and cell-based assays and structural analyses of polysialylated products using the anti-polySia monoclonal antibody 735 and endoneuraminidase N and HPLC approaches. Our data highlighted distinct specificities of human and salmonid polysialyltransferases with notable differences in donor substrates use and the capacity of fish enzymes to generate heteropolymers. This study further suggested an evolution of the biological functions of polySia. C. maraena ST8Sia IV of particular interest to modify glycoproteins with a variety of polySia chains.
Collapse
Affiliation(s)
- Mathieu Decloquement
- Univ. Lille, CNRS, UMR 8576 - UGSF - Unité de Glycobiologie Structurale et Fonctionnelle, 59000, Lille, France
| | - Marzia Tindara Venuto
- Institute of Reproductive Biology, Research Institute for Farm Animal Biology (FBN), Wilhelm-Stahl-Allee 2, 18196, Dummerstorf, Germany
| | - Virginie Cogez
- Univ. Lille, CNRS, UMR 8576 - UGSF - Unité de Glycobiologie Structurale et Fonctionnelle, 59000, Lille, France
| | - Anna Steinmetz
- Institute of Reproductive Biology, Research Institute for Farm Animal Biology (FBN), Wilhelm-Stahl-Allee 2, 18196, Dummerstorf, Germany
| | - Céline Schulz
- Univ. Lille, CNRS, UMR 8576 - UGSF - Unité de Glycobiologie Structurale et Fonctionnelle, 59000, Lille, France
| | - Cédric Lion
- Univ. Lille, CNRS, UMR 8576 - UGSF - Unité de Glycobiologie Structurale et Fonctionnelle, 59000, Lille, France
| | - Maxence Noel
- Univ. Lille, CNRS, UMR 8576 - UGSF - Unité de Glycobiologie Structurale et Fonctionnelle, 59000, Lille, France
| | - Vincent Rigolot
- Univ. Lille, CNRS, UMR 8576 - UGSF - Unité de Glycobiologie Structurale et Fonctionnelle, 59000, Lille, France
| | - Roxana Elin Teppa
- Univ. Lille, CNRS, UMR 8576 - UGSF - Unité de Glycobiologie Structurale et Fonctionnelle, 59000, Lille, France
| | - Christophe Biot
- Univ. Lille, CNRS, UMR 8576 - UGSF - Unité de Glycobiologie Structurale et Fonctionnelle, 59000, Lille, France
| | - Alexander Rebl
- Institute of Genome Biology, Research Institute for Farm Animal Biology FBN, Wilhelm-Stahl-Allee 2, 18196, Dummerstorf, Germany
| | - Sebastian Peter Galuska
- Institute of Reproductive Biology, Research Institute for Farm Animal Biology (FBN), Wilhelm-Stahl-Allee 2, 18196, Dummerstorf, Germany.
| | - Anne Harduin-Lepers
- Univ. Lille, CNRS, UMR 8576 - UGSF - Unité de Glycobiologie Structurale et Fonctionnelle, 59000, Lille, France.
- Unité de Glycobiologie Structurale et Fonctionnelle, UMR CNRS 8576, Faculté des sciences et Technologies, Univ. Lille, 59655, Villeneuve d'Ascq, France.
| |
Collapse
|
41
|
Al Saoud R, Hamrouni A, Idris A, Mousa WK, Abu Izneid T. Recent advances in the development of sialyltransferase inhibitors to control cancer metastasis: A comprehensive review. Biomed Pharmacother 2023; 165:115091. [PMID: 37421784 DOI: 10.1016/j.biopha.2023.115091] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2023] [Revised: 06/23/2023] [Accepted: 06/26/2023] [Indexed: 07/10/2023] Open
Abstract
Metastasis accounts for the majority of cancer-associated mortalities, representing a huge health and economic burden. One of the mechanisms that enables metastasis is hypersialylation, characterized by an overabundance of sialylated glycans on the tumor surface, which leads to repulsion and detachment of cells from the original tumor. Once the tumor cells are mobilized, sialylated glycans hijack the natural killer T-cells through self-molecular mimicry and activatea downstream cascade of molecular events that result in inhibition of cytotoxicity and inflammatory responses against cancer cells, ultimately leading to immune evasion. Sialylation is mediated by a family of enzymes known as sialyltransferases (STs), which catalyse the transfer of sialic acid residue from the donor, CMP-sialic acid, onto the terminal end of an acceptor such as N-acetylgalactosamine on the cell-surface. Upregulation of STs increases tumor hypersialylation by up to 60% which is considered a distinctive hallmark of several types of cancers such as pancreatic, breast, and ovarian cancer. Therefore, inhibiting STs has emerged as a potential strategy to prevent metastasis. In this comprehensive review, we discuss the recent advances in designing novel sialyltransferase inhibitors using ligand-based drug design and high-throughput screening of natural and synthetic entities, emphasizing the most successful approaches. We analyse the limitations and challenges of designing selective, potent, and cell-permeable ST inhibitors that hindered further development of ST inhibitors into clinical trials. We conclude by analysing emerging opportunities, including advanced delivery methods which further increase the potential of these inhibitors to enrich the clinics with novel therapeutics to combat metastasis.
Collapse
Affiliation(s)
- Ranim Al Saoud
- Pharmaceutical Sciences Program, College of Pharmacy, Al Ain University, P.O. Box 112612, Al Ain, Abu Dhabi, United Arab Emirates; AAU Health and Biomedical Research Center, Al Ain University, P.O. Box 112612, Abu Dhabi, United Arab Emirates
| | - Amar Hamrouni
- Pharmaceutical Sciences Program, College of Pharmacy, Al Ain University, P.O. Box 112612, Al Ain, Abu Dhabi, United Arab Emirates; AAU Health and Biomedical Research Center, Al Ain University, P.O. Box 112612, Abu Dhabi, United Arab Emirates
| | - Adi Idris
- School of Biomedical Sciences, Queensland University of Technology, Gardens Point, QLD, Australia; School of Pharmacy and Medical Science, Menzies Health Institute Queensland, Griffith University, Gold Coast, QLD, Australia
| | - Walaa K Mousa
- Pharmaceutical Sciences Program, College of Pharmacy, Al Ain University, P.O. Box 112612, Al Ain, Abu Dhabi, United Arab Emirates; AAU Health and Biomedical Research Center, Al Ain University, P.O. Box 112612, Abu Dhabi, United Arab Emirates
| | - Tareq Abu Izneid
- Pharmaceutical Sciences Program, College of Pharmacy, Al Ain University, P.O. Box 112612, Al Ain, Abu Dhabi, United Arab Emirates; AAU Health and Biomedical Research Center, Al Ain University, P.O. Box 112612, Abu Dhabi, United Arab Emirates.
| |
Collapse
|
42
|
Liou LB, Tsai PH, Fang YF, Chen YF, Chen CC, Lai JH. Sialic-Acid-Related Enzymes of B Cells and Monocytes as Novel Markers to Discriminate Improvement Categories and to Fulfill Two Remission Definitions in Rheumatoid Arthritis. Int J Mol Sci 2023; 24:12998. [PMID: 37629178 PMCID: PMC10455111 DOI: 10.3390/ijms241612998] [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: 06/16/2023] [Revised: 08/10/2023] [Accepted: 08/17/2023] [Indexed: 08/27/2023] Open
Abstract
The enzymes α-2,6-sialyltransferase 1 (ST6Gal1), neuraminidase 1 (Neu1), α-2,3-sialyltransferase 1 (ST3Gal1), and neuraminidase 3 (Neu3) are known to affect immune cell function. However, it is not known whether the levels of these enzymes relate to remission definitions or differentiate American College of Rheumatology (ACR), European League Against Rheumatism (EULAR), and Simplified Disease Activity Index (SDAI) responses in patients with rheumatoid arthritis (RA). We measured the ST6Gal1, Neu1, ST3Gal1, and Neu3 levels of B cells and monocytes in RA patients and correlated the cells' enzyme levels/ratios with the improvement in the ACR, EULAR and SDAI responses and with the two remission definitions. The difference in the B-cell Neu1 levels differed between the ACR 70% improvement and non-improvement groups (p = 0.043), between the EULAR good major response (improvement) and non-good response groups (p = 0.014), and also between the SDAI 50% or 70% improvement and non-improvement groups (p = 0.001 and 0.018, respectively). The same held true when the RA patients were classified by positive rheumatoid factor or the use of biologics. The B-cell Neu1 levels significantly indicated 2005 modified American Rheumatism Association and 2011 ACR/EULAR remission definitions (area under the curve (AUC) = 0.674 with p = 0.001, and AUC = 0.682 with p < 0.001, respectively) in contrast to the CRP and ESR (all AUCs < 0.420). We suggest that B-cell Neu1 is superior for discriminating ACR, EULAR, and SDAI improvement and is good for predicting two kinds of remission definitions.
Collapse
Affiliation(s)
- Lieh-Bang Liou
- Division of Rheumatology, Allergy and Immunology, New Taipei Municipal Tucheng Hospital, New Taipei City 236, Taiwan; (P.-H.T.); (C.-C.C.)
- Division of Rheumatology, Allergy and Immunology, Chang Gung Memorial Hospital at Linkou, Taoyuan 333, Taiwan; (Y.-F.F.); (Y.-F.C.); (J.-H.L.)
- School of Medicine, Chang Gung University College of Medicine, Taoyuan 333, Taiwan
| | - Ping-Han Tsai
- Division of Rheumatology, Allergy and Immunology, New Taipei Municipal Tucheng Hospital, New Taipei City 236, Taiwan; (P.-H.T.); (C.-C.C.)
- Division of Rheumatology, Allergy and Immunology, Chang Gung Memorial Hospital at Linkou, Taoyuan 333, Taiwan; (Y.-F.F.); (Y.-F.C.); (J.-H.L.)
| | - Yao-Fan Fang
- Division of Rheumatology, Allergy and Immunology, Chang Gung Memorial Hospital at Linkou, Taoyuan 333, Taiwan; (Y.-F.F.); (Y.-F.C.); (J.-H.L.)
| | - Yen-Fu Chen
- Division of Rheumatology, Allergy and Immunology, Chang Gung Memorial Hospital at Linkou, Taoyuan 333, Taiwan; (Y.-F.F.); (Y.-F.C.); (J.-H.L.)
| | - Chih-Chieh Chen
- Division of Rheumatology, Allergy and Immunology, New Taipei Municipal Tucheng Hospital, New Taipei City 236, Taiwan; (P.-H.T.); (C.-C.C.)
- Division of Rheumatology, Allergy and Immunology, Chang Gung Memorial Hospital at Linkou, Taoyuan 333, Taiwan; (Y.-F.F.); (Y.-F.C.); (J.-H.L.)
| | - Jenn-Haung Lai
- Division of Rheumatology, Allergy and Immunology, Chang Gung Memorial Hospital at Linkou, Taoyuan 333, Taiwan; (Y.-F.F.); (Y.-F.C.); (J.-H.L.)
| |
Collapse
|
43
|
Mitry MMA, Boateng SY, Greco F, Osborn HMI. Bioorthogonal activation of prodrugs, for the potential treatment of breast cancer, using the Staudinger reaction. RSC Med Chem 2023; 14:1537-1548. [PMID: 37593579 PMCID: PMC10429771 DOI: 10.1039/d3md00137g] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2023] [Accepted: 06/03/2023] [Indexed: 08/19/2023] Open
Abstract
Selective prodrug activation at a tumor site is crucial to maximise the efficiency of chemotherapy approaches and minimise side effects due to off-site activation. In this paper, a new prodrug activation strategy is reported based on the bioorthogonal Staudinger reaction. The feasibility of this prodrug activation strategy was initially demonstrated using 9-azido sialic acid 4 as a trigger and two novel triphenylphosphine-modified N-mustard-PRO 10 and doxorubicin-PRO 12 prodrugs in an HPLC-monitored release study. Then, the azide reporter group was introduced on cancer cells' surfaces through metabolic glycoengineering of sialic acid-rich surface glycans using azide-modified monosaccharides (9-azido sialic acid 4, tetra-O-acetylated-9-azido sialic acid 5 and tetra-O-acetyl azidomannosamine). Next, the N-mustard-PRO 10 and doxorubicin-PRO 12 prodrugs were employed in vitro with the bioengineered cells, and activation of the prodrugs, which allowed selective release of the cytotoxic moiety at the tumour cell, was assessed. Release of the parent drugs from the prodrugs was shown to be dependent on the level of metabolic labelling, where tetra-O-acetyl azidomannosamine allowed the highest level of azide reporter generation in tumor cells and led to full recovery of the parent cytotoxic drug's potency. The selectivity of azide expression on breast cancer MCF-7 cells versus normal fibroblast L929 cells was also probed, with the 9-azido sialic acid and tetra-O-acetylated-9-azido sialic acid showing ∼17-fold higher azide expression on the former. Taken together, these data demonstrate the feasibility of the Staudinger reaction for selective activation of prodrugs targeted to the MCF-7 breast cancer cells.
Collapse
Affiliation(s)
- Madonna M A Mitry
- Reading School of Pharmacy, University of Reading Whiteknights Reading RG6 6AD UK
- Dept. of Pharmaceutical Chemistry, Faculty of Pharmacy, Ain Shams University Cairo 11566 Egypt
| | - Samuel Y Boateng
- School of Biological Sciences, University of Reading Whiteknights Reading RG6 6ES UK
| | - Francesca Greco
- Reading School of Pharmacy, University of Reading Whiteknights Reading RG6 6AD UK
| | - Helen M I Osborn
- Reading School of Pharmacy, University of Reading Whiteknights Reading RG6 6AD UK
| |
Collapse
|
44
|
Costa AF, Senra E, Faria-Ramos I, Teixeira A, Morais J, Pacheco M, Reis CA, Gomes C. ST3GalIV drives SLeX biosynthesis in gastrointestinal cancer cells and associates with cancer cell motility. Glycoconj J 2023; 40:421-433. [PMID: 37074623 PMCID: PMC10335957 DOI: 10.1007/s10719-023-10113-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Revised: 01/31/2023] [Accepted: 03/15/2023] [Indexed: 04/20/2023]
Abstract
Expression of sialyl Lewis X (SLeX) is a well-documented event during malignant transformation of cancer cells, and largely associates with their invasive and metastatic properties. Glycoproteins and glycolipids are the main carriers of SLeX, whose biosynthesis is known to be performed by different glycosyltransferases, namely by the family of β-galactoside-α2,3-sialyltransferases (ST3Gals). In this study, we sought to elucidate the role of ST3GalIV in the biosynthesis of SLeX and in malignant properties of gastrointestinal (GI) cancer cells. By immunofluorescent screening, we selected SLeX-positive GI cancer cell lines and silenced ST3GalIV expression via CRISPR/Cas9. Flow cytometry, immunofluorescence and western blot analysis showed that ST3GalIV KO efficiently impaired SLeX expression in most cancer cell lines, with the exception of the colon cancer cell line LS174T. The impact of ST3GalIV KO in the biosynthesis of SLeX isomer SLeA and non sialylated Lewis X and A were also evaluated and overall, ST3GalIV KO led to a decreased expression of SLeA and an increased expression in both LeX and LeA. In addition, the abrogation of SLeX on GI cancer cells led to a reduction in cell motility. Furthermore, ST3GalVI KO was performed in LS174T ST3GalIV KO cells, resulting in the complete abolishment of SLeX expression and consequent reduced motility capacity of those cells. Overall, these findings portray ST3GalIV as the main, but not the only, enzyme driving the biosynthesis of SLeX in GI cancer cells, with a functional impact on cancer cell motility.
Collapse
Affiliation(s)
- Ana F Costa
- I3S-Instituto de Investigação e Inovação em Saúde, University of Porto, Porto, Portugal
- IPATIMUP-Institute of Molecular Pathology and Immunology, University of Porto, Porto, Portugal
- ICBAS-School of Medicine and Biomedical Sciences, University of Porto, Porto, Portugal
| | - Emanuel Senra
- I3S-Instituto de Investigação e Inovação em Saúde, University of Porto, Porto, Portugal
- IPATIMUP-Institute of Molecular Pathology and Immunology, University of Porto, Porto, Portugal
| | - Isabel Faria-Ramos
- I3S-Instituto de Investigação e Inovação em Saúde, University of Porto, Porto, Portugal
- IPATIMUP-Institute of Molecular Pathology and Immunology, University of Porto, Porto, Portugal
| | - Andreia Teixeira
- I3S-Instituto de Investigação e Inovação em Saúde, University of Porto, Porto, Portugal
- IPATIMUP-Institute of Molecular Pathology and Immunology, University of Porto, Porto, Portugal
- Faculty of Science, University of Porto, Porto, Portugal
| | - João Morais
- I3S-Instituto de Investigação e Inovação em Saúde, University of Porto, Porto, Portugal
- IPATIMUP-Institute of Molecular Pathology and Immunology, University of Porto, Porto, Portugal
| | - Mariana Pacheco
- I3S-Instituto de Investigação e Inovação em Saúde, University of 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, University of Porto, Porto, Portugal.
- IPATIMUP-Institute of Molecular Pathology and Immunology, University of Porto, Porto, Portugal.
- Faculty of Medicine, University of Porto, Porto, Portugal.
| | - Catarina Gomes
- I3S-Instituto de Investigação e Inovação em Saúde, University of Porto, Porto, Portugal.
- IPATIMUP-Institute of Molecular Pathology and Immunology, University of Porto, Porto, Portugal.
| |
Collapse
|
45
|
Li M, Ma Z, Zhang Y, Feng H, Li Y, Sang W, Zhu R, Huang R, Yan J. Integrative analysis of the ST6GALNAC family identifies GATA2-upregulated ST6GALNAC5 as an adverse prognostic biomarker promoting prostate cancer cell invasion. Cancer Cell Int 2023; 23:141. [PMID: 37468844 DOI: 10.1186/s12935-023-02983-x] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2023] [Accepted: 06/29/2023] [Indexed: 07/21/2023] Open
Abstract
BACKGROUND ST6GALNAC family members function as sialyltransferases and have been implicated in cancer progression. However, their aberrant expression levels, prognostic values and specific roles in metastatic prostate cancer (PCa) remain largely unclear. METHODS Two independent public datasets (TCGA-PRAD and GSE21032), containing 648 PCa samples in total, were employed to comprehensively examine the mRNA expression changes of ST6GALNAC family members in PCa, as well as their associations with clinicopathological parameters and prognosis. The dysregulation of ST6GALNAC5 was further validated in a mouse PCa model and human PCa samples from our cohort (n = 64) by immunohistochemistry (IHC). Gene Set Enrichment Analysis, Gene Ontology, Kyoto Encyclopedia of Genes and Genomes and drug sensitivity analyses were performed to enrich the biological processes most related to ST6GALNAC5. Sulforhodamine B, transwell, luciferase reporter and chromatin immunoprecipitation (ChIP) assays were used to examine the PCa cell proliferation, invasion and transcriptional regulation, respectively. RESULTS Systematical investigation of six ST6GALNAC family members in public datasets revealed that ST6GALNAC5 was the only gene consistently and significantly upregulated in metastatic PCa, and ST6GALNAC5 overexpression was also positively associated with Gleason score and predicted poor prognosis in PCa patients. IHC results showed that (1) ST6GALNAC5 protein expression was increased in prostatic intraepithelial neoplasia and further elevated in PCa from a PbCre;PtenF/F mouse model; (2) overexpressed ST6GALNAC5 protein was confirmed in human PCa samples comparing with benign prostatic hyperplasia samples from our cohort (p < 0.001); (3) ST6GALNAC5 overexpression was significantly correlated with perineural invasion of PCa. Moreover, we first found transcription factor GATA2 positively and directly regulated ST6GALNAC5 expression at transcriptional level. ST6GALNAC5 overexpression could partially reverse GATA2-depletion-induced inhibition of PCa cell invasion. The GATA2-ST6GALNAC5 signature exhibited better prediction on the poor prognosis in PCa patients than GATA2 or ST6GALNAC5 alone. CONCLUSIONS Our results indicated that GATA2-upregulated ST6GALNAC5 might serve as an adverse prognostic biomarker promoting prostate cancer cell invasion.
Collapse
Affiliation(s)
- Meiqian Li
- Model Animal Research Center, Medical School of Nanjing University, Nanjing University, Nanjing, China
| | - Zhihui Ma
- Center for Drug Safety Evaluation and Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Yuqing Zhang
- Department of Urology, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai, China
| | - Hanyi Feng
- School of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing, China
| | - Yang Li
- Department of Urology, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai, China
| | - Weicong Sang
- Department of Urology, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai, China
| | - Rujian Zhu
- Department of Urology, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai, China.
| | - Ruimin Huang
- Center for Drug Safety Evaluation and Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China.
- University of Chinese Academy of Sciences, Beijing, China.
- School of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing, China.
| | - Jun Yan
- Department of Laboratory Animal Science, Fudan University, Shanghai, China.
- Department of Urology, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai, China.
- Model Animal Research Center, Nanjing University, Nanjing, China.
| |
Collapse
|
46
|
Natoni A, Cerreto M, De Propris MS, Del Giudice I, Soscia R, Peragine N, Intoppa S, Milani ML, Guarini A, Foà R. Sialylation regulates migration in chronic lymphocytic leukemia. Haematologica 2023; 108:1851-1860. [PMID: 36779594 PMCID: PMC10316253 DOI: 10.3324/haematol.2022.281999] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2020] [Accepted: 01/27/2023] [Indexed: 02/11/2023] Open
Abstract
Sialylation is the terminal addition of sialic acid to underlying glycans. It plays a prominent role in cell adhesion and immune regulation. Sialylated structures found on adhesion molecules, such as CD49d, mediate the interactions between cancer cells and the microenvironment, facilitating metastatic seeding in target organs. Chronic lymphocytic leukemia (CLL) is a clonal B-cell malignancy characterized by the accumulation of CD5-positive B cells in the peripheral blood, bone marrow and lymph nodes. CLL cells proliferate mainly in the lymph node "proliferation centers", where the microenvironment provides pro-survival signals. Thus, migration and homing into these protective niches play a crucial role in CLL biology. In recent years, therapeutic strategies aimed at inducing the egress of CLL cells from the lymph nodes and bone marrow into the circulation have been highly successful. In this study, the sialylation status of 79 untreated and 24 ibrutinib-treated CLL patients was characterized by flow cytometry. Moreover, the effect of sialic acid removal on migration was tested by a transwell assay. Finally, we examined the sialylation status of CD49d by Western blot analysis. We found that CLL cells are highly sialylated, particularly those characterized by an "activated" immune phenotype. Notably, sialylation regulates CLL migration through the post-translational modification of CD49d. Finally, we showed that therapeutic agents that induce CLL mobilization from their protective niches, such as ibrutinib, modulate sialic acid levels. We propose that sialylation is an important regulator of CLL trafficking and may represent a novel target to further improve CLL therapy.
Collapse
Affiliation(s)
- Alessandro Natoni
- Hematology, Department of Translational and Precision Medicine, Sapienza University, Rome.
| | - Marina Cerreto
- Hematology, Department of Translational and Precision Medicine, Sapienza University, Rome
| | | | - Ilaria Del Giudice
- Hematology, Department of Translational and Precision Medicine, Sapienza University, Rome
| | - Roberta Soscia
- Hematology, Department of Translational and Precision Medicine, Sapienza University, Rome
| | - Nadia Peragine
- Hematology, Department of Translational and Precision Medicine, Sapienza University, Rome
| | - Stefania Intoppa
- Hematology, Department of Translational and Precision Medicine, Sapienza University, Rome
| | - Maria Laura Milani
- Hematology, Department of Translational and Precision Medicine, Sapienza University, Rome
| | - Anna Guarini
- Department of Molecular Medicine, Sapienza University, Rome
| | - Robin Foà
- Hematology, Department of Translational and Precision Medicine, Sapienza University, Rome
| |
Collapse
|
47
|
Zhou X, Chi K, Zhang C, Liu Q, Yang G. Sialylation: A Cloak for Tumors to Trick the Immune System in the Microenvironment. BIOLOGY 2023; 12:832. [PMID: 37372117 DOI: 10.3390/biology12060832] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/10/2023] [Revised: 06/03/2023] [Accepted: 06/06/2023] [Indexed: 06/29/2023]
Abstract
The tumor microenvironment (TME), where the tumor cells incite the surrounding normal cells to create an immune suppressive environment, reduces the effectiveness of immune responses during cancer development. Sialylation, a type of glycosylation that occurs on cell surface proteins, lipids, and glycoRNAs, is known to accumulate in tumors and acts as a "cloak" to help tumor cells evade immunological surveillance. In the last few years, the role of sialylation in tumor proliferation and metastasis has become increasingly evident. With the advent of single-cell and spatial sequencing technologies, more research is being conducted to understand the effects of sialylation on immunity regulation. This review provides updated insights into recent research on the function of sialylation in tumor biology and summarizes the latest developments in sialylation-targeted tumor therapeutics, including antibody-mediated and metabolic-based sialylation inhibition, as well as interference with sialic acid-Siglec interaction.
Collapse
Affiliation(s)
- Xiaoman Zhou
- The Key Laboratory of Carbohydrate Chemistry and Biotechnology, Ministry of Education, School of Biotechnology, Jiangnan University, Wuxi 214122, China
| | - Kaijun Chi
- The Key Laboratory of Carbohydrate Chemistry and Biotechnology, Ministry of Education, School of Biotechnology, Jiangnan University, Wuxi 214122, China
| | - Chairui Zhang
- The Key Laboratory of Carbohydrate Chemistry and Biotechnology, Ministry of Education, School of Biotechnology, Jiangnan University, Wuxi 214122, China
| | - Quan Liu
- Department of Medical Oncology, Affiliated Hospital of Jiangnan University, Wuxi 214122, China
| | - Ganglong Yang
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing 100190, China
| |
Collapse
|
48
|
Wang Y, Chen H. Protein glycosylation alterations in hepatocellular carcinoma: function and clinical implications. Oncogene 2023:10.1038/s41388-023-02702-w. [PMID: 37193819 DOI: 10.1038/s41388-023-02702-w] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2023] [Revised: 04/17/2023] [Accepted: 04/19/2023] [Indexed: 05/18/2023]
Abstract
Hepatocellular carcinoma (HCC) is the third leading cause of cancer death worldwide. Understanding the cancer mechanisms provides novel diagnostic, prognostic, and therapeutic markers for the management of HCC disease. In addition to genomic and epigenomic regulation, post-translational modification exerts a profound influence on protein functions and plays a critical role in regulating various biological processes. Protein glycosylation is one of the most common and complex post-translational modifications of newly synthesized proteins and acts as an important regulatory mechanism that is implicated in fundamental molecular and cell biology processes. Recent studies in glycobiology suggest that aberrant protein glycosylation in hepatocytes contributes to the malignant transformation to HCC by modulating a wide range of pro-tumorigenic signaling pathways. The dysregulated protein glycosylation regulates cancer growth, metastasis, stemness, immune evasion, and therapy resistance, and is regarded as a hallmark of HCC. Changes in protein glycosylation could serve as potential diagnostic, prognostic, and therapeutic factors in HCC. In this review, we summarize the functional importance, molecular mechanism, and clinical application of protein glycosylation alterations in HCC.
Collapse
Affiliation(s)
- Yifei Wang
- School of Biomedical Sciences, The Chinese University of Hong Kong, Hong Kong, China
| | - Huarong Chen
- Department of Anaesthesia and Intensive Care and Peter Hung Pain Research Institute, The Chinese University of Hong Kong, Hong Kong, China.
- Institute of Digestive Disease and Department of Medicine and Therapeutics, State Key Laboratory of Digestive Disease, Li Ka Shing Institute of Health Sciences, CUHK Shenzhen Research Institute, The Chinese University of Hong Kong, Hong Kong, China.
| |
Collapse
|
49
|
Xie X, Kong S, Cao W. Targeting protein glycosylation to regulate inflammation in the respiratory tract: novel diagnostic and therapeutic candidates for chronic respiratory diseases. Front Immunol 2023; 14:1168023. [PMID: 37256139 PMCID: PMC10225578 DOI: 10.3389/fimmu.2023.1168023] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2023] [Accepted: 05/02/2023] [Indexed: 06/01/2023] Open
Abstract
Protein glycosylation is a widespread posttranslational modification that can impact the function of proteins. Dysregulated protein glycosylation has been linked to several diseases, including chronic respiratory diseases (CRDs). CRDs pose a significant public health threat globally, affecting the airways and other lung structures. Emerging researches suggest that glycosylation plays a significant role in regulating inflammation associated with CRDs. This review offers an overview of the abnormal glycoenzyme activity and corresponding glycosylation changes involved in various CRDs, including chronic obstructive pulmonary disease, asthma, cystic fibrosis, idiopathic pulmonary fibrosis, pulmonary arterial hypertension, non-cystic fibrosis bronchiectasis, and lung cancer. Additionally, this review summarizes recent advances in glycomics and glycoproteomics-based protein glycosylation analysis of CRDs. The potential of glycoenzymes and glycoproteins for clinical use in the diagnosis and treatment of CRDs is also discussed.
Collapse
Affiliation(s)
- Xiaofeng Xie
- Shanghai Fifth People’s Hospital and Institutes of Biomedical Sciences, Fudan University, Shanghai, China
| | - Siyuan Kong
- Shanghai Fifth People’s Hospital and Institutes of Biomedical Sciences, Fudan University, Shanghai, China
| | - Weiqian Cao
- Shanghai Fifth People’s Hospital and Institutes of Biomedical Sciences, Fudan University, Shanghai, China
- NHC Key Laboratory of Glycoconjugates Research, Fudan University, Shanghai, China
| |
Collapse
|
50
|
Makarava N, Baskakov IV. Role of sialylation of N-linked glycans in prion pathogenesis. Cell Tissue Res 2023; 392:201-214. [PMID: 35088180 PMCID: PMC9329487 DOI: 10.1007/s00441-022-03584-2] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2021] [Accepted: 01/12/2022] [Indexed: 01/10/2023]
Abstract
Mammalian prion or PrPSc is a proteinaceous infectious agent that consists of a misfolded, self-replicating state of the prion protein or PrPC. PrPC and PrPSc are posttranslationally modified with N-linked glycans, which are sialylated at the terminal positions. More than 30 years have passed since the first characterization of the composition and structural diversity of N-linked glycans associated with the prion protein, yet the role of carbohydrate groups that constitute N-glycans and, in particular, their terminal sialic acid residues in prion disease pathogenesis remains poorly understood. A number of recent studies shed a light on the role of sialylation in the biology of prion diseases. This review article discusses several mechanisms by which terminal sialylation dictates the spread of PrPSc across brain regions and the outcomes of prion infection in an organism. In particular, relationships between the sialylation status of PrPSc and important strain-specific features including lymphotropism, neurotropism, and neuroinflammation are discussed. Moreover, emerging evidence pointing out the roles of sialic acid residues in prion replication, cross-species transmission, strain competition, and strain adaptation are reviewed. A hypothesis according to which selective, strain-specified recruitment of PrPC sialoglycoforms dictates unique strain-specific disease phenotypes is examined. Finally, the current article proposes that prion strains evolve as a result of a delicate balance between recruiting highly sialylated glycoforms to avoid an "eat-me" response by glia and limiting heavily sialylated glycoforms for enabling rapid prion replication.
Collapse
Affiliation(s)
- Natallia Makarava
- Center for Biomedical Engineering and Technology and Department of Anatomy and Neurobiology, University of Maryland School of Medicine, Baltimore, MD, 21201, USA
| | - Ilia V Baskakov
- Center for Biomedical Engineering and Technology and Department of Anatomy and Neurobiology, University of Maryland School of Medicine, Baltimore, MD, 21201, USA.
| |
Collapse
|