1
|
Tu H, Yuan L, Ni B, Lin Y, Wang K. Siglecs-mediated immune regulation in neurological disorders. Pharmacol Res 2024; 210:107531. [PMID: 39615617 DOI: 10.1016/j.phrs.2024.107531] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/25/2024] [Revised: 11/18/2024] [Accepted: 11/27/2024] [Indexed: 12/06/2024]
Abstract
The surfaces of various immune cells are rich in glycan chains, including the sialic-acid-binding immunoglobulin-like lectins (Siglecs) family. As an emerging glyco-immune checkpoint, Siglecs have the ability to bind and interact with various glycoproteins, thereby eliciting a series of downstream reactions to modulate the immune response. The impact of Siglecs has been extensively studied in tumor immunotherapy. However, research in neurological disorders and neurological diseases is very limited, and therapeutic options involving Siglecs need further exploration. Siglecs play a crucial role in the development, homeostasis, and repair processes of the nervous system, especially in degenerative diseases. This review summarizes studies on the immunomodulatory role mediated by Siglecs expressed on different immune cells in various neurological disorders, elucidates how dysregulated sialic acid contributes to several psychiatric disorders, and discusses the progress and limitations of research on the treatment of neurological disorders.
Collapse
Affiliation(s)
- Huifang Tu
- Graduate School, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Limei Yuan
- Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, State Key Laboratory of Druggability Evaluation and Systematic Translational Medicine, Tianjin's Clinical Research Center for Cancer, Tianjin 300060, China
| | - Bo Ni
- Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, State Key Laboratory of Druggability Evaluation and Systematic Translational Medicine, Tianjin's Clinical Research Center for Cancer, Tianjin 300060, China
| | - Yufeng Lin
- Department of Neurology, Tianjin First Central Hospital, School of Medicine, Nankai University, Tianjin 300190, China.
| | - Kaiyuan Wang
- Graduate School, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China; Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, State Key Laboratory of Druggability Evaluation and Systematic Translational Medicine, Tianjin's Clinical Research Center for Cancer, Tianjin 300060, China.
| |
Collapse
|
2
|
Silva Z, Rabaça JA, Luz V, Lourenço RA, Salio M, Oliveira AC, Bule P, Springer S, Videira PA. New insights into the immunomodulatory potential of sialic acid on monocyte-derived dendritic cells. Cancer Immunol Immunother 2024; 74:9. [PMID: 39487861 PMCID: PMC11531459 DOI: 10.1007/s00262-024-03863-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2024] [Accepted: 10/11/2024] [Indexed: 11/04/2024]
Abstract
Sialic acids at the cell surface of dendritic cells (DCs) play an important immunomodulatory role, and their manipulation enhances DC maturation, leading to heightened T cell activation. Particularly, at the molecular level, the increased stability of surface MHC-I molecules in monocyte-derived DCs (MoDCs) underpins an improved DC: T cell interaction. In this study, we focused on the impact of sialic acid remodelling by treatment with Clostridium perfringens sialidase on MoDCs' phenotypic and functional characteristics. Our investigation juxtaposes this novel approach with the conventional cytokine-based maturation regimen commonly employed in clinical settings.Notably, C. perfringens sialidase remarkably increased MHC-I levels compared to other sialidases having different specificities, supporting the idea that higher MHC-I is due to the cleavage of specific sialoglycans on cell surface proteins. Sialidase treatment induced rapid elevated surface expression of MHC-I, MHC-II and CD40 within an hour, a response not fully replicated by 48 h cytokine cocktail treatment. These increases were also observable 48 h post sialidase treatment. While CD86 and PD-L1 showed significant increases after 48 h of cytokine maturation, 48 h post sialidase treatment showed a higher increase in CD86 and shorter increase in PD-L1. CCR-7 expression was significantly increased 48 h after sialidase treatment but not significantly affected by cytokine maturation. Both treatments promoted higher secretion of the IL-12 cytokine. However, the cytokine cocktail induced a more pronounced IL-12 production. SNA lectin staining analysis demonstrated that the sialic acid profile is significantly altered by sialidase treatment, but not by the cytokine cocktail, which causes only slight sialic acid upregulation. Notably, the lipid-presenting molecules CD1a, CD1b and CD1c remained unaffected by sialidase treatment in MoDCs, a finding also further supported by experiments performed on C1R cells. Inhibition of endogenous sialidases Neu1 and Neu3 during MoDC differentiation did not affect surface MHC-I expression and cytokine secretion. Yet, sialidase activity in MoDCs was minimal, suggesting that sialidase inhibition does not significantly alter MHC-I-related functions. Our study highlights the unique maturation profile induced by sialic acid manipulation in MoDCs. These findings provide insights into the potential of sialic acid manipulation as a rapid immunomodulatory strategy, offering promising avenues for targeted interventions in inflammatory contexts.
Collapse
Affiliation(s)
- Zélia Silva
- Associate Laboratory i4HB, NOVA School of Science and Technology, Institute for Health and Bioeconomy, Universidade NOVA de Lisboa, 2829-516, Caparica, Portugal
- Department of Life Sciences, Applied Molecular Biosciences Unit, UCIBIO, NOVA School of Science and Technology, Universidade NOVA de Lisboa, 2829-516, Caparica, Portugal
| | - João Amorim Rabaça
- Associate Laboratory i4HB, NOVA School of Science and Technology, Institute for Health and Bioeconomy, Universidade NOVA de Lisboa, 2829-516, Caparica, Portugal
- Department of Life Sciences, Applied Molecular Biosciences Unit, UCIBIO, NOVA School of Science and Technology, Universidade NOVA de Lisboa, 2829-516, Caparica, Portugal
| | - Vanessa Luz
- Associate Laboratory i4HB, NOVA School of Science and Technology, Institute for Health and Bioeconomy, Universidade NOVA de Lisboa, 2829-516, Caparica, Portugal
- Department of Life Sciences, Applied Molecular Biosciences Unit, UCIBIO, NOVA School of Science and Technology, Universidade NOVA de Lisboa, 2829-516, Caparica, Portugal
| | - Rita Adubeiro Lourenço
- Associate Laboratory i4HB, NOVA School of Science and Technology, Institute for Health and Bioeconomy, Universidade NOVA de Lisboa, 2829-516, Caparica, Portugal
- Department of Life Sciences, Applied Molecular Biosciences Unit, UCIBIO, NOVA School of Science and Technology, Universidade NOVA de Lisboa, 2829-516, Caparica, Portugal
| | - Mariolina Salio
- Medical Research Council Translational Immune Discovery Unit, Medical Research Council Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, OX39DS, UK
| | - Alexandra Couto Oliveira
- CIISA‑Centre for Interdisciplinary Research in Animal Health, Faculty of Veterinary Medicine, University of Lisbon, Avenida da Universidade Técnica, 1300‑477, Lisbon, Portugal
- Associate Laboratory for Animal and Veterinary Sciences (AL4AnimalS), 1300‑477, Lisbon, Portugal
| | - Pedro Bule
- CIISA‑Centre for Interdisciplinary Research in Animal Health, Faculty of Veterinary Medicine, University of Lisbon, Avenida da Universidade Técnica, 1300‑477, Lisbon, Portugal
- Associate Laboratory for Animal and Veterinary Sciences (AL4AnimalS), 1300‑477, Lisbon, Portugal
| | | | - Paula Alexandra Videira
- Associate Laboratory i4HB, NOVA School of Science and Technology, Institute for Health and Bioeconomy, Universidade NOVA de Lisboa, 2829-516, Caparica, Portugal.
- Department of Life Sciences, Applied Molecular Biosciences Unit, UCIBIO, NOVA School of Science and Technology, Universidade NOVA de Lisboa, 2829-516, Caparica, Portugal.
- Department of Life Sciences, CDG & Allies Professionals and Patient Associations International Network (CDG & Allies-PPAIN), NOVA School of Science and Technology, Universidade NOVA de Lisboa, 2829-516, Caparica, Portugal.
| |
Collapse
|
3
|
Ma X, Li M, Wang X, Qi G, Wei L, Zhang D. Sialylation in the gut: From mucosal protection to disease pathogenesis. Carbohydr Polym 2024; 343:122471. [PMID: 39174097 DOI: 10.1016/j.carbpol.2024.122471] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2024] [Revised: 06/19/2024] [Accepted: 07/07/2024] [Indexed: 08/24/2024]
Abstract
Sialylation, a crucial post-translational modification of glycoconjugates, entails the attachment of sialic acid (SA) to the terminal glycans of glycoproteins and glycolipids through a tightly regulated enzymatic process involving various enzymes. This review offers a comprehensive exploration of sialylation within the gut, encompassing its involvement in mucosal protection and its impact on disease progression. The sialylation of mucins and epithelial glycoproteins contributes to the integrity of the intestinal mucosal barrier. Furthermore, sialylation regulates immune responses in the gut, shaping interactions among immune cells, as well as their activation and tolerance. Additionally, the gut microbiota and gut-brain axis communication are involved in the role of sialylation in intestinal health. Altered sialylation patterns have been implicated in various intestinal diseases, including inflammatory bowel disease (IBD), colorectal cancer (CRC), and other intestinal disorders. Emerging research underscores sialylation as a promising avenue for diagnostic, prognostic, and therapeutic interventions in intestinal diseases. Potential strategies such as sialic acid supplementation, inhibition of sialidases, immunotherapy targeting sialylated antigens, and modulation of sialyltransferases have been utilized in the treatment of intestinal diseases. Future research directions will focus on elucidating the molecular mechanisms underlying sialylation alterations, identifying sialylation-based biomarkers, and developing targeted interventions for precision medicine approaches.
Collapse
Affiliation(s)
- Xueni Ma
- Key Laboratory of Digestive Diseases, Lanzhou University Second Hospital, Lanzhou, China; The Second Clinical Medical College, Lanzhou University, Lanzhou, China
| | - Muyang Li
- Key Laboratory of Digestive Diseases, Lanzhou University Second Hospital, Lanzhou, China; The Second Clinical Medical College, Lanzhou University, Lanzhou, China
| | - Xiaochun Wang
- Department of Gastroenterology, Gansu Provincial Hospital, Lanzhou, China
| | - Guoqing Qi
- Department of Gastroenterology, Lanzhou University Second Hospital, Lanzhou, China
| | - Lina Wei
- Department of Gastroenterology, Lanzhou University Second Hospital, Lanzhou, China
| | - Dekui Zhang
- Key Laboratory of Digestive Diseases, Lanzhou University Second Hospital, Lanzhou, China; Department of Gastroenterology, Lanzhou University Second Hospital, Lanzhou, China.
| |
Collapse
|
4
|
Xu Y, Ren S, Wang H, Qin Y, Liu T, Sun C, Xiao Y, Shao B, Zhang J, Chen Q, Zhao P, Yang G, Liu X, Wang H. Endometrial regeneration cell-derived exosomes loaded with siSLAMF6 inhibit cardiac allograft rejection through the suppression of desialylation modification. Cell Mol Biol Lett 2024; 29:128. [PMID: 39354345 PMCID: PMC11443917 DOI: 10.1186/s11658-024-00645-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: 05/05/2024] [Accepted: 09/13/2024] [Indexed: 10/03/2024] Open
Abstract
BACKGROUNDS Acute transplant rejection is a major component of poor prognoses for organ transplantation. Owing to the multiple complex mechanisms involved, new treatments are still under exploration. Endometrial regenerative cells (ERCs) have been widely used in various refractory immune-related diseases, but the role of ERC-derived exosomes (ERC-Exos) in alleviating transplant rejection has not been extensively studied. Signaling lymphocyte activation molecule family 6 (SLAMF6) plays an important role in regulating immune responses. In this study, we explored the main mechanism by which ERC-Exos loaded with siSLAMF6 can alleviate allogeneic transplant rejection. METHODS C57BL/6 mouse recipients of BALB/c mouse kidney transplants were randomly divided into four groups and treated with exosomes. The graft pathology was evaluated by H&E staining. Splenic and transplanted heart immune cell populations were analyzed by flow cytometry. Recipient serum cytokine profiles were determined by enzyme-linked immunosorbent assay (ELISA). The proliferation and differentiation capacity of CD4+ T cell populations were evaluated in vitro. The α-2,6-sialylation levels in the CD4+ T cells were determined by SNA blotting. RESULTS In vivo, mice treated with ERC-siSLAMF6 Exo achieved significantly prolonged allograft survival. The serum cytokine profiles of the recipients were significantly altered in the ERC-siSLAMF6 Exo-treated recipients. In vitro, we found that ERC-siSLAMF6-Exo considerably downregulated α-2,6-sialyltransferase (ST6GAL1) expression in CD4+ T cells, and significantly reduced α-2,6-sialylation levels. Through desialylation, ERC-siSLAMF6 Exo therapy significantly decreased CD4+ T cell proliferation and inhibited CD4+ T cell differentiation into Th1 and Th17 cells while promoting regulatory T cell (Treg) differentiation. CONCLUSIONS Our study indicated that ERC-Exos loaded with siSLAMF6 reduce the amount of sialic acid connected to α-2,6 at the end of the N-glycan chain on the CD4+ T cell surface, increase the number of therapeutic exosomes endocytosed into CD4+ T cells, and inhibit the activation of T cell receptor signaling pathways, which prolongs allograft survival. This study confirms the feasibility of using ERC-Exos as natural carriers combined with gene therapy, which could be used as a potential therapeutic strategy to alleviate allograft rejection.
Collapse
Affiliation(s)
- Yini Xu
- Department of General Surgery, Tianjin Medical University General Hospital, 154 Anshan Road, Heping District, Tianjin, 300052, China
- Tianjin General Surgery Institute, 154 Anshan Road, Heping District, Tianjin, 300052, China
| | - Shaohua Ren
- Department of General Surgery, Tianjin Medical University General Hospital, 154 Anshan Road, Heping District, Tianjin, 300052, China
- Tianjin General Surgery Institute, 154 Anshan Road, Heping District, Tianjin, 300052, China
| | - Hongda Wang
- Department of General Surgery, Tianjin Medical University General Hospital, 154 Anshan Road, Heping District, Tianjin, 300052, China
- Tianjin General Surgery Institute, 154 Anshan Road, Heping District, Tianjin, 300052, China
| | - Yafei Qin
- Department of Vascular Surgery, Henan Provincial People's Hospital, The Affiliated People's Hospital of Zhengzhou University, Zhengzhou, 450003, Henan, China
| | - Tong Liu
- Department of General Surgery, Tianjin Medical University General Hospital, 154 Anshan Road, Heping District, Tianjin, 300052, China
- Tianjin General Surgery Institute, 154 Anshan Road, Heping District, Tianjin, 300052, China
| | - Chenglu Sun
- Department of General Surgery, Tianjin Medical University General Hospital, 154 Anshan Road, Heping District, Tianjin, 300052, China
- Tianjin General Surgery Institute, 154 Anshan Road, Heping District, Tianjin, 300052, China
| | - Yiyi Xiao
- Department of General Surgery, Tianjin Medical University General Hospital, 154 Anshan Road, Heping District, Tianjin, 300052, China
- Tianjin General Surgery Institute, 154 Anshan Road, Heping District, Tianjin, 300052, China
| | - Bo Shao
- Department of General Surgery, Tianjin Medical University General Hospital, 154 Anshan Road, Heping District, Tianjin, 300052, China
- Tianjin General Surgery Institute, 154 Anshan Road, Heping District, Tianjin, 300052, China
| | - Jingyi Zhang
- Department of General Surgery, Tianjin Medical University General Hospital, 154 Anshan Road, Heping District, Tianjin, 300052, China
- Tianjin General Surgery Institute, 154 Anshan Road, Heping District, Tianjin, 300052, China
| | - Qiang Chen
- Department of General Surgery, Tianjin Medical University General Hospital, 154 Anshan Road, Heping District, Tianjin, 300052, China
- Tianjin General Surgery Institute, 154 Anshan Road, Heping District, Tianjin, 300052, China
| | - Pengyu Zhao
- Department of General Surgery, Tianjin Medical University General Hospital, 154 Anshan Road, Heping District, Tianjin, 300052, China
- Tianjin General Surgery Institute, 154 Anshan Road, Heping District, Tianjin, 300052, China
| | - Guangmei Yang
- Department of General Surgery, Tianjin Medical University General Hospital, 154 Anshan Road, Heping District, Tianjin, 300052, China
- Tianjin General Surgery Institute, 154 Anshan Road, Heping District, Tianjin, 300052, China
| | - Xu Liu
- Department of General Surgery, Tianjin Medical University General Hospital, 154 Anshan Road, Heping District, Tianjin, 300052, China
- Tianjin General Surgery Institute, 154 Anshan Road, Heping District, Tianjin, 300052, China
| | - Hao Wang
- Department of General Surgery, Tianjin Medical University General Hospital, 154 Anshan Road, Heping District, Tianjin, 300052, China.
- Tianjin General Surgery Institute, 154 Anshan Road, Heping District, Tianjin, 300052, China.
- Tianjin Key Laboratory of Precise Vascular Reconstruction and Organ Function Repair, 154 Anshan Road, Heping District, Tianjin, 300052, China.
| |
Collapse
|
5
|
Zhu W, Zhou Y, Guo L, Feng S. Biological function of sialic acid and sialylation in human health and disease. Cell Death Discov 2024; 10:415. [PMID: 39349440 PMCID: PMC11442784 DOI: 10.1038/s41420-024-02180-3] [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: 06/28/2024] [Revised: 09/08/2024] [Accepted: 09/12/2024] [Indexed: 10/02/2024] Open
Abstract
Sialic acids are predominantly found at the terminal ends of glycoproteins and glycolipids and play key roles in cellular communication and function. The process of sialylation, a form of post-translational modification, involves the covalent attachment of sialic acid to the terminal residues of oligosaccharides and glycoproteins. This modification not only provides a layer of electrostatic repulsion to cells but also serves as a receptor for various biological signaling pathways. Sialylation is involved in several pathophysiological processes. Given its multifaceted involvement in cellular functions, sialylation presents a promising avenue for therapeutic intervention. Current studies are exploring agents that target sialic acid residues on sialoglycans or the sialylation process. These efforts are particularly focused on the fields of cancer therapy, stroke treatment, antiviral strategies, and therapies for central nervous system disorders. In this review, we aimed to summarize the biological functions of sialic acid and the process of sialylation, explore their roles in various pathophysiological contexts, and discuss their potential applications in the development of novel therapeutics.
Collapse
Affiliation(s)
- Wengen Zhu
- Department of Cardiology, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Yue Zhou
- Department of Ophthalmology, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Linjuan Guo
- Department of Cardiology, Jiangxi Provincial People's Hospital, The First Affiliated Hospital of Nanchang Medical College, Nanchang, China.
| | - Shenghui Feng
- Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.
| |
Collapse
|
6
|
Chen J, Wu T, Yang Y. Sialylation-associated long non-coding RNA signature predicts the prognosis, tumor microenvironment, and immunotherapy and chemotherapy options in uterine corpus endometrial carcinoma. Cancer Cell Int 2024; 24:314. [PMID: 39261877 PMCID: PMC11391619 DOI: 10.1186/s12935-024-03486-z] [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: 11/30/2023] [Accepted: 08/17/2024] [Indexed: 09/13/2024] Open
Abstract
BACKGROUND Sialylation in uterine corpus endometrial carcinoma (UCEC) differs significantly from apoptotic and ferroptosis pathways. It plays a crucial role in cancer progression and immune response modulation. Exploring how sialylation affects tumor behavior and its link with long non-coding RNAs (lncRNAs) may provide new insights into UCEC prognosis and treatment. METHODS We obtained RNA transcriptome, clinical, and mutation data of UCEC samples from the TCGA database. Our approach involved developing a risk model based on the co-expression patterns of sialylation genes and lncRNAs. Prognostic lncRNAs were identified through Cox regression and further refined using LASSO analysis. To understand the biological functions and pathways of model-associated differentially expressed genes (MADEGs), we conducted enrichment analyses. We also assessed the immune infiltration status of MADEGs using eight different algorithms, which helped in evaluating the potential for immunotherapy. Additionally, we validated the expression of these lncRNAs in UCEC using cell lines and clinical samples. RESULTS We developed a UCEC risk model using five sialylation-related lncRNAs (AC004884.2, AC026202.2, LINC01579, LINC00942, SLC16A1-AS1). This model, confirmed through Cox analysis and clinical evaluation, effectively predicted patient outcomes. Survival data analysis across entire cohort, as well as within training and test groups, indicated better survival in low-risk UCEC patients. Enrichment analyses linked MADEGs to sialylation functions and cancer pathways. High-risk patients showed increased responsiveness to immune checkpoint inhibitors (ICIs), as indicated by immunological assessments. Subgroup C2 patients showed superior outcomes and a robust response to immunotherapy and chemotherapy. Notably, LINC01579, LINC00942, and SLC16A1-AS1 were significantly overexpressed in UCEC clinical tumor samples as well as in Ishikawa and HEC-1-B cell lines, compared to the normal groups. CONCLUSIONS This lncRNA signature associated with sialylation could guide prognosis, enhance the understanding of molecular mechanisms, and inform treatment strategies in UCEC. It highlights the potential for the use of ICIs and chemotherapy.
Collapse
Affiliation(s)
- Jun Chen
- Department of Infectious Diseases, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center of Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Tingting Wu
- Department of Cardiovasology, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center of Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Yongwen Yang
- Department of Clinical Laboratory, Xiangya Hospital, Central South University, No. 87 Xiangya Road, Changsha, 410008, P. R. China.
- National Clinical Research Center of Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China.
| |
Collapse
|
7
|
Li Y, Chen H, Gao J, Wu P, Hong S. Glycoengineering in antigen-specific immunotherapies. Curr Opin Chem Biol 2024; 81:102503. [PMID: 39053235 PMCID: PMC11921784 DOI: 10.1016/j.cbpa.2024.102503] [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: 05/27/2024] [Revised: 06/27/2024] [Accepted: 06/28/2024] [Indexed: 07/27/2024]
Abstract
Advances in immunotherapy have revolutionized modern medical care paradigms. However, many patients respond poorly to the current FDA-approved treatment regimens that primarily target protein-based antigens or checkpoints. Current progress in developing therapeutic strategies that target disease-associated glycans has pinpointed a new class of glycoimmune checkpoints that function orthogonally to the established protein-immune checkpoints. Glycoengineering using chemical, enzymatic, and genetic methods is also increasingly recognized for its massive potential to improve biopharmaceuticals, such as tailoring therapies with antigen-targeting agents. Here, we review the recent development and applications of glycoengineering of antibodies and cells to suit therapeutic applications. We highlight living-cell glycoengineering strategies on cancer and immune cells for better therapeutic efficacy against specific antigens by leveraging the pre-existing immune machinery or instructing de novo creation of targeting agents. We also discuss glycoengineering strategies for studying basic immuno-oncology. Collectively, glycoengineering has a significant contribution to the design of antigen-specific immunotherapies.
Collapse
Affiliation(s)
- Yuxin Li
- State Key Laboratory of Natural and Biomimetic Drugs, Chemical Biology Center, School of Pharmaceutical Sciences, Peking University, Beijing, 100191, PR China
| | - Hongming Chen
- State Key Laboratory of Natural and Biomimetic Drugs, Chemical Biology Center, School of Pharmaceutical Sciences, Peking University, Beijing, 100191, PR China
| | - Jiuxiang Gao
- State Key Laboratory of Natural and Biomimetic Drugs, Chemical Biology Center, School of Pharmaceutical Sciences, Peking University, Beijing, 100191, PR China
| | - Peng Wu
- Department of Molecular and Cellular Biology, 10550 North Torrey Pines Road, The Scripps Research Institute, La Jolla, CA, 92037, USA
| | - Senlian Hong
- State Key Laboratory of Natural and Biomimetic Drugs, Chemical Biology Center, School of Pharmaceutical Sciences, Peking University, Beijing, 100191, PR China.
| |
Collapse
|
8
|
Carr A, Mateyka LM, Scheu SJC, Bici A, Paijmans J, Reijmers RM, Dieminger N, Dildebekova S, Hamed N, Wagner K, Busch DH, D'Ippolito E. Advances in preclinical TCR characterization: leveraging cell avidity to identify functional TCRs. Biol Chem 2024; 405:517-529. [PMID: 38666334 DOI: 10.1515/hsz-2023-0341] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2023] [Accepted: 03/28/2024] [Indexed: 07/14/2024]
Abstract
T-cell therapy has emerged as an effective approach for treating viral infections and cancers. However, a significant challenge is the selection of T-cell receptors (TCRs) that exhibit the desired functionality. Conventionally in vitro techniques, such as peptide sensitivity measurements and cytotoxicity assays, provide valuable insights into TCR potency but are labor-intensive. In contrast, measuring ligand binding properties (z-Movi technology) could provide an accelerated processing while showing robust correlations with T-cell functions. In this study, we assessed whether cell avidity can predict functionality also in the context of TCR-engineered T cells. To this end, we developed a flexible system for TCR re-expression by generating a Jurkat-derived T cell clone lacking TCR and CD3 expression through CRISPR-Cas9-mediated TRBC knockout. The knockin of a transgenic TCR into the TRAC locus restored TCR/CD3 expression, allowing for CD3-based purification of TCR-engineered T cells. Subsequently, we characterized these engineered cell lines by functional readouts, and assessment of binding properties through the z-Movi technology. Our findings revealed a strong correlation between the cell avidities and functional sensitivities of Jurkat TCR-T cells. Altogether, by integrating cell avidity measurements with our versatile T cell engineering platform, we established an accelerated system for enhancing the in vitro selection of clinically relevant TCRs.
Collapse
Affiliation(s)
- Andreas Carr
- 9184 Institute for Medical Microbiology, Immunology and Hygiene, School of Medicine and Health, Technical University of Munich , Munich, Germany
| | - Laura M Mateyka
- 9184 Institute for Medical Microbiology, Immunology and Hygiene, School of Medicine and Health, Technical University of Munich , Munich, Germany
| | - Sebastian J C Scheu
- 9184 Institute for Medical Microbiology, Immunology and Hygiene, School of Medicine and Health, Technical University of Munich , Munich, Germany
| | - Ana Bici
- 9184 Institute for Medical Microbiology, Immunology and Hygiene, School of Medicine and Health, Technical University of Munich , Munich, Germany
| | - Joris Paijmans
- LUMICKS, Paalbergweg 3, NL-1105 AG, Amsterdam, The Netherlands
| | | | - Nina Dieminger
- 9184 Institute for Medical Microbiology, Immunology and Hygiene, School of Medicine and Health, Technical University of Munich , Munich, Germany
| | - Shirin Dildebekova
- 9184 Institute for Medical Microbiology, Immunology and Hygiene, School of Medicine and Health, Technical University of Munich , Munich, Germany
| | - Noomen Hamed
- 9184 Institute for Medical Microbiology, Immunology and Hygiene, School of Medicine and Health, Technical University of Munich , Munich, Germany
| | - Karolin Wagner
- 9184 Institute for Medical Microbiology, Immunology and Hygiene, School of Medicine and Health, Technical University of Munich , Munich, Germany
- CellPoint, a Galapagos Company, Oegstgeest, The Netherlands
| | - Dirk H Busch
- 9184 Institute for Medical Microbiology, Immunology and Hygiene, School of Medicine and Health, Technical University of Munich , Munich, Germany
- German Center for Infection Research (DZIF), Partner Site Munich, Munich, Germany
| | - Elvira D'Ippolito
- 9184 Institute for Medical Microbiology, Immunology and Hygiene, School of Medicine and Health, Technical University of Munich , Munich, Germany
- German Center for Infection Research (DZIF), Partner Site Munich, Munich, Germany
| |
Collapse
|
9
|
Silva Z, Soares CO, Barbosa M, Palma AS, Marcelo F, Videira PA. The role of sialoglycans in modulating dendritic cell function and tumour immunity. Semin Immunol 2024; 74-75:101900. [PMID: 39461124 DOI: 10.1016/j.smim.2024.101900] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/01/2024] [Revised: 10/15/2024] [Accepted: 10/15/2024] [Indexed: 10/29/2024]
Abstract
Dendritic cells (DCs) are crucial for initiating immune responses against tumours by presenting antigens to T cells. Glycosylation, particularly sialylation, plays a significant role in regulating cell functions, by modulating protein folding and signalling. This review aimed to provide a comprehensive overview of how sialic acids influence key aspects of DC biology, including maturation, migration, antigen presentation, and T cell interactions. Sialic acids influence DC endocytosis, affecting their ability to uptake and present antigens, while guiding their migration to lymph nodes and inflamed tissues. Removing sialic acids enhances DC-mediated antigen presentation to T cells, potentially boosting immune responses. Additionally, sialylated glycans on DCs modulate immune checkpoints, which can impact tumour immunity. Hypersialylation of tumour mucins further promotes immune evasion by interacting with DCs. Understanding the interplay between sialylation and DC functions offers promising avenues for enhancing cancer immunotherapy.
Collapse
Affiliation(s)
- Zélia Silva
- Institute for Health and Bioeconomy (i4HB), NOVA School of Science and Technology, Universidade NOVA de Lisboa, Caparica 2829-516, Portugal; UCIBIO - Applied Molecular Biosciences Unit, Department of Life Sciences, NOVA School of Science and Technology, Universidade NOVA de Lisboa, Caparica 2829-516, Portugal; CDG & Allies - Professionals and Patient Associations International Network (CDG & Allies - PPAIN), Department of Life Sciences, NOVA School of Science and Technology, Universidade NOVA de Lisboa, Caparica 2829-516, Portugal
| | - Cátia O Soares
- Institute for Health and Bioeconomy (i4HB), NOVA School of Science and Technology, Universidade NOVA de Lisboa, Caparica 2829-516, Portugal; UCIBIO - Applied Molecular Biosciences Unit, Department of Chemistry, NOVA School of Science and Technology, Universidade NOVA de Lisboa, Caparica 2829-516, Portugal
| | - Mariana Barbosa
- Institute for Health and Bioeconomy (i4HB), NOVA School of Science and Technology, Universidade NOVA de Lisboa, Caparica 2829-516, Portugal; UCIBIO - Applied Molecular Biosciences Unit, Department of Life Sciences, NOVA School of Science and Technology, Universidade NOVA de Lisboa, Caparica 2829-516, Portugal; CDG & Allies - Professionals and Patient Associations International Network (CDG & Allies - PPAIN), Department of Life Sciences, NOVA School of Science and Technology, Universidade NOVA de Lisboa, Caparica 2829-516, Portugal
| | - Angelina S Palma
- Institute for Health and Bioeconomy (i4HB), NOVA School of Science and Technology, Universidade NOVA de Lisboa, Caparica 2829-516, Portugal; UCIBIO - Applied Molecular Biosciences Unit, Department of Chemistry, NOVA School of Science and Technology, Universidade NOVA de Lisboa, Caparica 2829-516, Portugal
| | - Filipa Marcelo
- Institute for Health and Bioeconomy (i4HB), NOVA School of Science and Technology, Universidade NOVA de Lisboa, Caparica 2829-516, Portugal; UCIBIO - Applied Molecular Biosciences Unit, Department of Chemistry, NOVA School of Science and Technology, Universidade NOVA de Lisboa, Caparica 2829-516, Portugal
| | - Paula A Videira
- Institute for Health and Bioeconomy (i4HB), NOVA School of Science and Technology, Universidade NOVA de Lisboa, Caparica 2829-516, Portugal; UCIBIO - Applied Molecular Biosciences Unit, Department of Life Sciences, NOVA School of Science and Technology, Universidade NOVA de Lisboa, Caparica 2829-516, Portugal; CDG & Allies - Professionals and Patient Associations International Network (CDG & Allies - PPAIN), Department of Life Sciences, NOVA School of Science and Technology, Universidade NOVA de Lisboa, Caparica 2829-516, Portugal.
| |
Collapse
|
10
|
Tian W, Blomberg AL, Steinberg KE, Henriksen BL, Jørgensen JS, Skovgaard K, Skovbakke SL, Goletz S. Novel genetically glycoengineered human dendritic cell model reveals regulatory roles of α2,6-linked sialic acids in DC activation of CD4+ T cells and response to TNFα. Glycobiology 2024; 34:cwae042. [PMID: 38873803 DOI: 10.1093/glycob/cwae042] [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: 01/19/2024] [Revised: 05/08/2024] [Accepted: 05/31/2024] [Indexed: 06/15/2024] Open
Abstract
Dendritic cells (DCs) are central for the initiation and regulation of appropriate immune responses. While several studies suggest important regulatory roles of sialoglycans in DC biology, our understanding is still inadequate primarily due to a lack of appropriate models. Previous approaches based on enzymatic- or metabolic-glycoengineering and primary cell isolation from genetically modified mice have limitations related to specificity, stability, and species differences. This study addresses these challenges by introducing a workflow to genetically glycoengineer the human DC precursor cell line MUTZ-3, described to differentiate and maturate into fully functional dendritic cells, using CRISPR-Cas9, thereby providing and validating the first isogenic cell model for investigating glycan alteration on human DC differentiation, maturation, and activity. By knocking out (KO) the ST6GAL1 gene, we generated isogenic cells devoid of ST6GAL1-mediated α(2,6)-linked sialylation, allowing for a comprehensive investigation into its impact on DC function. Glycan profiling using lectin binding assay and functional studies revealed that ST6GAL1 KO increased the expression of important antigen presenting and co-stimulatory surface receptors and a specifically increased activation of allogenic human CD4 + T cells. Additionally, ST6GAL1 KO induces significant changes in surface marker expression and cytokine response to TNFα-induced maturation, and it affects migration and the endocytic capacity. These results indicate that genetic glycoengineering of the isogenic MUTZ-3 cellular model offers a valuable tool to study how specific glycan structures influence human DC biology, contributing to our understanding of glycoimmunology.
Collapse
Affiliation(s)
- Weihua Tian
- Biotherapeutic Glycoengineering and Immunology, Section for Medical Biotechnology, Department of Biotechnology and Biomedicine, Technical University of Denmark, Søltofts Plads, Kgs Lyngby 2800, Denmark
| | - Anne Louise Blomberg
- Biotherapeutic Glycoengineering and Immunology, Section for Medical Biotechnology, Department of Biotechnology and Biomedicine, Technical University of Denmark, Søltofts Plads, Kgs Lyngby 2800, Denmark
| | - Kaylin Elisabeth Steinberg
- Biotherapeutic Glycoengineering and Immunology, Section for Medical Biotechnology, Department of Biotechnology and Biomedicine, Technical University of Denmark, Søltofts Plads, Kgs Lyngby 2800, Denmark
| | - Betina Lyngfeldt Henriksen
- Biotherapeutic Glycoengineering and Immunology, Section for Medical Biotechnology, Department of Biotechnology and Biomedicine, Technical University of Denmark, Søltofts Plads, Kgs Lyngby 2800, Denmark
| | - Josefine Søborg Jørgensen
- Biotherapeutic Glycoengineering and Immunology, Section for Medical Biotechnology, Department of Biotechnology and Biomedicine, Technical University of Denmark, Søltofts Plads, Kgs Lyngby 2800, Denmark
| | - Kerstin Skovgaard
- Biotherapeutic Glycoengineering and Immunology, Section for Medical Biotechnology, Department of Biotechnology and Biomedicine, Technical University of Denmark, Søltofts Plads, Kgs Lyngby 2800, Denmark
| | - Sarah Line Skovbakke
- Biotherapeutic Glycoengineering and Immunology, Section for Medical Biotechnology, Department of Biotechnology and Biomedicine, Technical University of Denmark, Søltofts Plads, Kgs Lyngby 2800, Denmark
| | - Steffen Goletz
- Biotherapeutic Glycoengineering and Immunology, Section for Medical Biotechnology, Department of Biotechnology and Biomedicine, Technical University of Denmark, Søltofts Plads, Kgs Lyngby 2800, Denmark
| |
Collapse
|
11
|
Filipsky F, Läubli H. Regulation of sialic acid metabolism in cancer. Carbohydr Res 2024; 539:109123. [PMID: 38669826 DOI: 10.1016/j.carres.2024.109123] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2024] [Revised: 04/17/2024] [Accepted: 04/17/2024] [Indexed: 04/28/2024]
Abstract
Sialic acid, the terminal structure of cell surface glycans, has essential functions in regulating immune response, cell-to-cell communication, and cell adhesion. More importantly, an increased level of sialic acid, termed hypersialylation, has emerged as a commonly observed phenotype in cancer. Therefore, targeting sialic acid ligands (sialoglycans) and their receptors (Siglecs) may provide a new therapeutic approach for cancer immunotherapy. We highlight the complexity of the sialic acid metabolism and its involvement in malignant transformation within individual cancer subtypes. In this review, we focus on the dysregulation of sialylation, the intricate nature of sialic acid synthesis, and clinical perspective. We aim to provide a brief insight into the mechanism of hypersialylation and how our understanding of these processes can be leveraged for the development of novel therapeutics.
Collapse
Affiliation(s)
- Filip Filipsky
- Department of Biomedicine, University Hospital and University of Basel, Switzerland
| | - Heinz Läubli
- Department of Biomedicine, University Hospital and University of Basel, Switzerland; Division of Oncology, University Hospital Basel, Switzerland.
| |
Collapse
|
12
|
Ayyalasomayajula R, Cudic M. Targeting Siglec-Sialylated MUC1 Immune Axis in Cancer. Cancers (Basel) 2024; 16:1334. [PMID: 38611013 PMCID: PMC11011055 DOI: 10.3390/cancers16071334] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2023] [Revised: 03/12/2024] [Accepted: 03/27/2024] [Indexed: 04/14/2024] Open
Abstract
Siglecs play a key role in mediating cell-cell interactions via the recognition of different sialylated glycoconjugates, including tumor-associated MUC1, which can lead to the activation or inhibition of the immune response. The activation occurs through the signaling of Siglecs with the cytoplasmic immunoreceptor tyrosine-based activation motif (ITAM)-containing proteins, while the inhibition signal is a result of the interaction of intracellular immunoreceptor tyrosine-based inhibition motif (ITIM)-bearing receptors. The interaction of tumor-associated MUC1 sialylated glycans with Siglecs via ITIM motifs decreases antitumor immunity. Consequently, these interactions are expected to play a key role in tumor evasion. Efforts to modulate the response of immune cells by blocking the immune-suppressive effects of inhibitory Siglecs, driving immune-activating Siglecs, and/or altering the synthesis and expression of the sialic acid glycocalyx are new therapeutic strategies deserving further investigation. We will highlight the role of Siglec's family receptors in immune evasion through interactions with glycan ligands in their natural context, presented on the protein such as MUC1, factors affecting their fine binding specificities, such as the role of multivalency either at the ligand or receptor side, their spatial organization, and finally the current and future therapeutic interventions targeting the Siglec-sialylated MUC1 immune axis in cancer.
Collapse
Affiliation(s)
| | - Mare Cudic
- Department of Chemistry and Biochemistry, Florida Atlantic University, 777 Glades Rd., Boca Raton, FL 33431, USA;
| |
Collapse
|
13
|
Boelaars K, van Kooyk Y. Targeting myeloid cells for cancer immunotherapy: Siglec-7/9/10/15 and their ligands. Trends Cancer 2024; 10:230-241. [PMID: 38160071 DOI: 10.1016/j.trecan.2023.11.009] [Citation(s) in RCA: 14] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2023] [Revised: 11/17/2023] [Accepted: 11/28/2023] [Indexed: 01/03/2024]
Abstract
Advances in immunotherapy have revolutionized cancer treatment, yet many patients do not show clinical responses. While most immunotherapies target T cells, myeloid cells are the most abundant cell type in solid tumors and are key orchestrators of the immunosuppressive tumor microenvironment (TME), hampering effective T cell responses. Therefore, unraveling the immune suppressive pathways within myeloid cells could unveil new avenues for cancer immunotherapy. Over the past decade, Siglec receptors and their ligand, sialic acids, have emerged as a novel immune checkpoint on myeloid cells. In this review, we highlight key findings on how sialic acids modify immunity in the TME through engagement of Siglec-7/9/10/15 expressed on myeloid cells, and how the sialic acid-Siglec axis can be targeted for future cancer immunotherapies.
Collapse
Affiliation(s)
- Kelly Boelaars
- Amsterdam UMC location Vrije Universiteit Amsterdam, Molecular Cell Biology and Immunology, Cancer Center Amsterdam, Amsterdam Institute for Infection and Immunity, De Boelelaan, 1117, Amsterdam, The Netherlands
| | - Yvette van Kooyk
- Amsterdam UMC location Vrije Universiteit Amsterdam, Molecular Cell Biology and Immunology, Cancer Center Amsterdam, Amsterdam Institute for Infection and Immunity, De Boelelaan, 1117, Amsterdam, The Netherlands.
| |
Collapse
|
14
|
Feng H, Feng J, Han X, Ying Y, Lou W, Liu L, Zhang L. The Potential of Siglecs and Sialic Acids as Biomarkers and Therapeutic Targets in Tumor Immunotherapy. Cancers (Basel) 2024; 16:289. [PMID: 38254780 PMCID: PMC10813689 DOI: 10.3390/cancers16020289] [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: 11/17/2023] [Revised: 12/24/2023] [Accepted: 01/02/2024] [Indexed: 01/24/2024] Open
Abstract
The dysregulation of sialic acid is closely associated with oncogenesis and tumor progression. Most tumor cells exhibit sialic acid upregulation. Sialic acid-binding immunoglobulin-like lectins (Siglecs) are receptors that recognize sialic acid and are expressed in various immune cells. The activity of Siglecs in the tumor microenvironment promotes immune escape, mirroring the mechanisms of the well-characterized PD-1/PD-L1 pathway in cancer. Cancer cells utilize sialic acid-linked glycans to evade immune surveillance. As Siglecs exhibit similar mechanisms as the established immune checkpoint inhibitors (ICIs), they are potential therapeutic targets for different forms of cancer, especially ICI-resistant malignancies. Additionally, the upregulation of sialic acid serves as a potential tumor biomarker. This review examines the feasibility of using sialic acid and Siglecs for early malignant tumor detection and discusses the potential of targeting Siglec-sialic acid interaction as a novel cancer therapeutic strategy.
Collapse
Affiliation(s)
- Haokang Feng
- Department of Pancreatic Surgery, Zhongshan Hospital, Fudan University, Shanghai 200032, China; (H.F.); (J.F.); (X.H.); (Y.Y.); (W.L.)
- Cancer Center, Zhongshan Hospital, Fudan University, Shanghai 200032, China
- Department of General Surgery, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Jiale Feng
- Department of Pancreatic Surgery, Zhongshan Hospital, Fudan University, Shanghai 200032, China; (H.F.); (J.F.); (X.H.); (Y.Y.); (W.L.)
- Cancer Center, Zhongshan Hospital, Fudan University, Shanghai 200032, China
- Department of General Surgery, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Xu Han
- Department of Pancreatic Surgery, Zhongshan Hospital, Fudan University, Shanghai 200032, China; (H.F.); (J.F.); (X.H.); (Y.Y.); (W.L.)
- Cancer Center, Zhongshan Hospital, Fudan University, Shanghai 200032, China
- Department of General Surgery, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Ying Ying
- Department of Pancreatic Surgery, Zhongshan Hospital, Fudan University, Shanghai 200032, China; (H.F.); (J.F.); (X.H.); (Y.Y.); (W.L.)
- Cancer Center, Zhongshan Hospital, Fudan University, Shanghai 200032, China
- Department of General Surgery, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Wenhui Lou
- Department of Pancreatic Surgery, Zhongshan Hospital, Fudan University, Shanghai 200032, China; (H.F.); (J.F.); (X.H.); (Y.Y.); (W.L.)
- Cancer Center, Zhongshan Hospital, Fudan University, Shanghai 200032, China
- Department of General Surgery, Zhongshan Hospital, Fudan University, Shanghai 200032, China
- The Shanghai Geriatrics Medical Center, Zhongshan Hospital MinHang MeiLong Branch, Fudan University, Shanghai 200032, China
| | - Liang Liu
- Department of Pancreatic Surgery, Zhongshan Hospital, Fudan University, Shanghai 200032, China; (H.F.); (J.F.); (X.H.); (Y.Y.); (W.L.)
- Cancer Center, Zhongshan Hospital, Fudan University, Shanghai 200032, China
- Department of General Surgery, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Lei Zhang
- Department of Pancreatic Surgery, Zhongshan Hospital, Fudan University, Shanghai 200032, China; (H.F.); (J.F.); (X.H.); (Y.Y.); (W.L.)
- Cancer Center, Zhongshan Hospital, Fudan University, Shanghai 200032, China
- Department of General Surgery, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| |
Collapse
|
15
|
Yang Y, Wang Y, Chao Z, Yang Y, Fang Y, Liu Y, Ding L, Chen Y, Ju H. Triply Enhanced Immunotherapy via Dual Glycan Reforming Integrated with Perforation. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2304971. [PMID: 37870206 PMCID: PMC10787084 DOI: 10.1002/advs.202304971] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/20/2023] [Revised: 09/11/2023] [Indexed: 10/24/2023]
Abstract
The enhancement of immunotherapy is an emerging direction to develop highly effective and practical cancer therapeutic methods. Here a triply enhanced immunotherapy drug (TEID) is designed for ingeniously integrating in situ dual glycan reforming with perforation on cell membrane. The TEID is composed of galactose and neuraminidase conjugated streptolysin O (SLO-Gal and SLO-NEU), which are encapsulated in a hyaluronic acid (HA) shell for targeted recognition to tumor tissue via cell surface CD44. After targeted delivery and HAase-mediated degradation in the tumor region, the TEID releases SLO-Gal and SLO-NEU, which can easily anchor Gal and NEU on the tumor cell membrane via the perforation of SLO to perform dual glycan reforming for the introduction of Gal and the cleavage of sialic acid. The former can activate immune cells to secret cytokines for immune-killing, and the latter can weaken the immune inhibition to improve the immunotherapeutic efficacy. Meanwhile, the perforation of SLO can promote the delivery of cytokines into the tumor cells to further enhance the efficacy. The designed triply enhanced immunotherapy strategy opens a significant and promising route to promote clinical immunotherapy of cancer.
Collapse
Affiliation(s)
- Yuanjiao Yang
- State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical EngineeringNanjing UniversityNanjing210023China
| | - Yuru Wang
- State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical EngineeringNanjing UniversityNanjing210023China
| | - Zhicong Chao
- State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical EngineeringNanjing UniversityNanjing210023China
| | - Yuhui Yang
- State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical EngineeringNanjing UniversityNanjing210023China
| | - Yanyun Fang
- State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical EngineeringNanjing UniversityNanjing210023China
| | - Ying Liu
- State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical EngineeringNanjing UniversityNanjing210023China
| | - Lin Ding
- State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical EngineeringNanjing UniversityNanjing210023China
| | - Yunlong Chen
- State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical EngineeringNanjing UniversityNanjing210023China
| | - Huangxian Ju
- State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical EngineeringNanjing UniversityNanjing210023China
| |
Collapse
|
16
|
Moons SJ, Hornikx DLAH, Aasted MKM, Pijnenborg JFA, Calzari M, White PB, Narimatsu Y, Clausen H, Wandall HH, Boltje TJ, Büll C. UV light-induced spatial loss of sialic acid capping using a photoactivatable sialyltransferase inhibitor. RSC Chem Biol 2023; 4:506-511. [PMID: 37415865 PMCID: PMC10320844 DOI: 10.1039/d3cb00006k] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2023] [Accepted: 05/06/2023] [Indexed: 07/08/2023] Open
Abstract
Sialic acids cap glycans displayed on mammalian glycoproteins and glycolipids and mediate many glycan-receptor interactions. Sialoglycans play a role in diseases such as cancer and infections where they facilitate immune evasion and metastasis or serve as cellular receptors for viruses, respectively. Strategies that specifically interfere with cellular sialoglycan biosynthesis, such as sialic acid mimetics that act as metabolic sialyltransferase inhibitors, enable research into the diverse biological functions of sialoglycans. Sialylation inhibitors are also emerging as potential therapeutics for cancer, infection, and other diseases. However, sialoglycans serve many important biological functions and systemic inhibition of sialoglycan biosynthesis can have adverse effects. To enable local and inducible inhibition of sialylation, we have synthesized and characterized a caged sialyltransferase inhibitor that can be selectively activated with UV-light. A photolabile protecting group was conjugated to a known sialyltransferase inhibitor (P-SiaFNEtoc). This yielded a photoactivatable inhibitor, UV-SiaFNEtoc, that remained inactive in human cell cultures and was readily activated through radiation with 365 nm UV light. Direct and short radiation of a human embryonic kidney (HEK293) cell monolayer was well-tolerated and resulted in photoactivation of the inhibitor and subsequent spatial restricted synthesis of asialoglycans. The developed photocaged sialic acid mimetic holds the potential to locally hinder the synthesis of sialoglycans through focused treatment with UV light and may be applied to bypass the adverse effects related to systemic loss of sialylation.
Collapse
Affiliation(s)
- Sam J Moons
- Cluster for Molecular Chemistry, Institute for Molecules and Materials, Radboud University Nijmegen Nijmegen The Netherlands
| | - Daniël L A H Hornikx
- Department of Biomolecular Chemistry, Institute for Molecules and Materials, Radboud University Nijmegen Nijmegen The Netherlands
| | - Mikkel K M Aasted
- Copenhagen Center for Glycomics, Department of Cellular and Molecular Medicine, Faculty of Health Sciences, University of Copenhagen Copenhagen Denmark
| | - Johan F A Pijnenborg
- Cluster for Molecular Chemistry, Institute for Molecules and Materials, Radboud University Nijmegen Nijmegen The Netherlands
| | - Matteo Calzari
- Cluster for Molecular Chemistry, Institute for Molecules and Materials, Radboud University Nijmegen Nijmegen The Netherlands
| | - Paul B White
- Cluster for Molecular Chemistry, Institute for Molecules and Materials, Radboud University Nijmegen Nijmegen The Netherlands
| | - Yoshiki Narimatsu
- Copenhagen Center for Glycomics, Department of Cellular and Molecular Medicine, Faculty of Health Sciences, University of Copenhagen Copenhagen Denmark
| | - Henrik Clausen
- Copenhagen Center for Glycomics, Department of Cellular and Molecular Medicine, Faculty of Health Sciences, University of Copenhagen Copenhagen Denmark
| | - Hans H Wandall
- Copenhagen Center for Glycomics, Department of Cellular and Molecular Medicine, Faculty of Health Sciences, University of Copenhagen Copenhagen Denmark
| | - Thomas J Boltje
- Cluster for Molecular Chemistry, Institute for Molecules and Materials, Radboud University Nijmegen Nijmegen The Netherlands
| | - Christian Büll
- Department of Biomolecular Chemistry, Institute for Molecules and Materials, Radboud University Nijmegen Nijmegen The Netherlands
- Copenhagen Center for Glycomics, Department of Cellular and Molecular Medicine, Faculty of Health Sciences, University of Copenhagen Copenhagen Denmark
| |
Collapse
|
17
|
Stanczak MA, Läubli H. Siglec receptors as new immune checkpoints in cancer. Mol Aspects Med 2023; 90:101112. [PMID: 35948467 DOI: 10.1016/j.mam.2022.101112] [Citation(s) in RCA: 57] [Impact Index Per Article: 28.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2022] [Revised: 07/26/2022] [Accepted: 07/28/2022] [Indexed: 02/08/2023]
Abstract
Cancer immunotherapy in the form of immune checkpoint inhibitors and cellular therapies has improved the treatment and prognosis of many patients. Nevertheless, most cancers are still resistant to currently approved cancer immunotherapies. New approaches and rational combinations are needed to overcome these resistances. There is emerging evidence that Siglec receptors could be regarded as new immune checkpoints and targets for cancer immunotherapy. In this review, we summarize the experimental evidence supporting Siglec receptors as new immune checkpoints in cancer and discuss their mechanisms of action, as well as current efforts to target Siglec receptors and their interactions with sialoglycan Siglec-ligands.
Collapse
Affiliation(s)
- Michal A Stanczak
- The Bloomberg-Kimmel Institute for Cancer Immunotherapy at Johns Hopkins, Baltimore, MD, 21287, USA
| | - Heinz Läubli
- Laboratory for Cancer Immunotherapy, Department of Biomedicine, University of Basel, Division of Oncology, University Hospital Basel, Switzerland.
| |
Collapse
|
18
|
Hu M, Zhang R, Yang J, Zhao C, Liu W, Huang Y, Lyu H, Xiao S, Guo D, Zhou C, Tang J. The role of N-glycosylation modification in the pathogenesis of liver cancer. Cell Death Dis 2023; 14:222. [PMID: 36990999 PMCID: PMC10060418 DOI: 10.1038/s41419-023-05733-z] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2022] [Revised: 03/02/2023] [Accepted: 03/13/2023] [Indexed: 03/31/2023]
Abstract
N-glycosylation is one of the most common types of protein modifications and it plays a vital role in normal physiological processes. However, aberrant N-glycan modifications are closely associated with the pathogenesis of diverse diseases, including processes such as malignant transformation and tumor progression. It is known that the N-glycan conformation of the associated glycoproteins is altered during different stages of hepatocarcinogenesis. Characterizing the heterogeneity and biological functions of glycans in liver cancer patients will facilitate a deeper understanding of the molecular mechanisms of liver injury and hepatocarcinogenesis. In this article, we review the role of N-glycosylation in hepatocarcinogenesis, focusing on epithelial-mesenchymal transition, extracellular matrix changes, and tumor microenvironment formation. We highlight the role of N-glycosylation in the pathogenesis of liver cancer and its potential applications in the treatment or diagnosis of liver cancer.
Collapse
Affiliation(s)
- Mengyu Hu
- National "111" Center for Cellular Regulation and Molecular Pharmaceutics, Key Laboratory of Fermentation Engineering (Ministry of Education), Cooperative Innovation Center of Industrial Fermentation (Ministry of Education & Hubei Province), Hubei Key Laboratory of Industrial Microbiology, Hubei University of Technology, Wuhan, China
| | - Rui Zhang
- National "111" Center for Cellular Regulation and Molecular Pharmaceutics, Key Laboratory of Fermentation Engineering (Ministry of Education), Cooperative Innovation Center of Industrial Fermentation (Ministry of Education & Hubei Province), Hubei Key Laboratory of Industrial Microbiology, Hubei University of Technology, Wuhan, China
| | - Jiaren Yang
- National "111" Center for Cellular Regulation and Molecular Pharmaceutics, Key Laboratory of Fermentation Engineering (Ministry of Education), Cooperative Innovation Center of Industrial Fermentation (Ministry of Education & Hubei Province), Hubei Key Laboratory of Industrial Microbiology, Hubei University of Technology, Wuhan, China
| | - Chenshu Zhao
- National "111" Center for Cellular Regulation and Molecular Pharmaceutics, Key Laboratory of Fermentation Engineering (Ministry of Education), Cooperative Innovation Center of Industrial Fermentation (Ministry of Education & Hubei Province), Hubei Key Laboratory of Industrial Microbiology, Hubei University of Technology, Wuhan, China
| | - Wei Liu
- National "111" Center for Cellular Regulation and Molecular Pharmaceutics, Key Laboratory of Fermentation Engineering (Ministry of Education), Cooperative Innovation Center of Industrial Fermentation (Ministry of Education & Hubei Province), Hubei Key Laboratory of Industrial Microbiology, Hubei University of Technology, Wuhan, China
| | - Yuan Huang
- National "111" Center for Cellular Regulation and Molecular Pharmaceutics, Key Laboratory of Fermentation Engineering (Ministry of Education), Cooperative Innovation Center of Industrial Fermentation (Ministry of Education & Hubei Province), Hubei Key Laboratory of Industrial Microbiology, Hubei University of Technology, Wuhan, China
| | - Hao Lyu
- National "111" Center for Cellular Regulation and Molecular Pharmaceutics, Key Laboratory of Fermentation Engineering (Ministry of Education), Cooperative Innovation Center of Industrial Fermentation (Ministry of Education & Hubei Province), Hubei Key Laboratory of Industrial Microbiology, Hubei University of Technology, Wuhan, China
| | - Shuai Xiao
- National "111" Center for Cellular Regulation and Molecular Pharmaceutics, Key Laboratory of Fermentation Engineering (Ministry of Education), Cooperative Innovation Center of Industrial Fermentation (Ministry of Education & Hubei Province), Hubei Key Laboratory of Industrial Microbiology, Hubei University of Technology, Wuhan, China
| | - Dong Guo
- National "111" Center for Cellular Regulation and Molecular Pharmaceutics, Key Laboratory of Fermentation Engineering (Ministry of Education), Cooperative Innovation Center of Industrial Fermentation (Ministry of Education & Hubei Province), Hubei Key Laboratory of Industrial Microbiology, Hubei University of Technology, Wuhan, China
| | - Cefan Zhou
- National "111" Center for Cellular Regulation and Molecular Pharmaceutics, Key Laboratory of Fermentation Engineering (Ministry of Education), Cooperative Innovation Center of Industrial Fermentation (Ministry of Education & Hubei Province), Hubei Key Laboratory of Industrial Microbiology, Hubei University of Technology, Wuhan, China.
| | - Jingfeng Tang
- National "111" Center for Cellular Regulation and Molecular Pharmaceutics, Key Laboratory of Fermentation Engineering (Ministry of Education), Cooperative Innovation Center of Industrial Fermentation (Ministry of Education & Hubei Province), Hubei Key Laboratory of Industrial Microbiology, Hubei University of Technology, Wuhan, China.
| |
Collapse
|
19
|
Hu M, Zhang R, Yang J, Zhao C, Liu W, Huang Y, Lyu H, Xiao S, Guo D, Zhou C, Tang J. The role of N-glycosylation modification in the pathogenesis of liver cancer. Cell Death Dis 2023; 14:222. [PMID: 36990999 DOI: 10.1038/s41419-023-05733-z.pmid:] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2022] [Revised: 03/02/2023] [Accepted: 03/13/2023] [Indexed: 10/14/2024]
Abstract
N-glycosylation is one of the most common types of protein modifications and it plays a vital role in normal physiological processes. However, aberrant N-glycan modifications are closely associated with the pathogenesis of diverse diseases, including processes such as malignant transformation and tumor progression. It is known that the N-glycan conformation of the associated glycoproteins is altered during different stages of hepatocarcinogenesis. Characterizing the heterogeneity and biological functions of glycans in liver cancer patients will facilitate a deeper understanding of the molecular mechanisms of liver injury and hepatocarcinogenesis. In this article, we review the role of N-glycosylation in hepatocarcinogenesis, focusing on epithelial-mesenchymal transition, extracellular matrix changes, and tumor microenvironment formation. We highlight the role of N-glycosylation in the pathogenesis of liver cancer and its potential applications in the treatment or diagnosis of liver cancer.
Collapse
Affiliation(s)
- Mengyu Hu
- National "111" Center for Cellular Regulation and Molecular Pharmaceutics, Key Laboratory of Fermentation Engineering (Ministry of Education), Cooperative Innovation Center of Industrial Fermentation (Ministry of Education & Hubei Province), Hubei Key Laboratory of Industrial Microbiology, Hubei University of Technology, Wuhan, China
| | - Rui Zhang
- National "111" Center for Cellular Regulation and Molecular Pharmaceutics, Key Laboratory of Fermentation Engineering (Ministry of Education), Cooperative Innovation Center of Industrial Fermentation (Ministry of Education & Hubei Province), Hubei Key Laboratory of Industrial Microbiology, Hubei University of Technology, Wuhan, China
| | - Jiaren Yang
- National "111" Center for Cellular Regulation and Molecular Pharmaceutics, Key Laboratory of Fermentation Engineering (Ministry of Education), Cooperative Innovation Center of Industrial Fermentation (Ministry of Education & Hubei Province), Hubei Key Laboratory of Industrial Microbiology, Hubei University of Technology, Wuhan, China
| | - Chenshu Zhao
- National "111" Center for Cellular Regulation and Molecular Pharmaceutics, Key Laboratory of Fermentation Engineering (Ministry of Education), Cooperative Innovation Center of Industrial Fermentation (Ministry of Education & Hubei Province), Hubei Key Laboratory of Industrial Microbiology, Hubei University of Technology, Wuhan, China
| | - Wei Liu
- National "111" Center for Cellular Regulation and Molecular Pharmaceutics, Key Laboratory of Fermentation Engineering (Ministry of Education), Cooperative Innovation Center of Industrial Fermentation (Ministry of Education & Hubei Province), Hubei Key Laboratory of Industrial Microbiology, Hubei University of Technology, Wuhan, China
| | - Yuan Huang
- National "111" Center for Cellular Regulation and Molecular Pharmaceutics, Key Laboratory of Fermentation Engineering (Ministry of Education), Cooperative Innovation Center of Industrial Fermentation (Ministry of Education & Hubei Province), Hubei Key Laboratory of Industrial Microbiology, Hubei University of Technology, Wuhan, China
| | - Hao Lyu
- National "111" Center for Cellular Regulation and Molecular Pharmaceutics, Key Laboratory of Fermentation Engineering (Ministry of Education), Cooperative Innovation Center of Industrial Fermentation (Ministry of Education & Hubei Province), Hubei Key Laboratory of Industrial Microbiology, Hubei University of Technology, Wuhan, China
| | - Shuai Xiao
- National "111" Center for Cellular Regulation and Molecular Pharmaceutics, Key Laboratory of Fermentation Engineering (Ministry of Education), Cooperative Innovation Center of Industrial Fermentation (Ministry of Education & Hubei Province), Hubei Key Laboratory of Industrial Microbiology, Hubei University of Technology, Wuhan, China
| | - Dong Guo
- National "111" Center for Cellular Regulation and Molecular Pharmaceutics, Key Laboratory of Fermentation Engineering (Ministry of Education), Cooperative Innovation Center of Industrial Fermentation (Ministry of Education & Hubei Province), Hubei Key Laboratory of Industrial Microbiology, Hubei University of Technology, Wuhan, China
| | - Cefan Zhou
- National "111" Center for Cellular Regulation and Molecular Pharmaceutics, Key Laboratory of Fermentation Engineering (Ministry of Education), Cooperative Innovation Center of Industrial Fermentation (Ministry of Education & Hubei Province), Hubei Key Laboratory of Industrial Microbiology, Hubei University of Technology, Wuhan, China.
| | - Jingfeng Tang
- National "111" Center for Cellular Regulation and Molecular Pharmaceutics, Key Laboratory of Fermentation Engineering (Ministry of Education), Cooperative Innovation Center of Industrial Fermentation (Ministry of Education & Hubei Province), Hubei Key Laboratory of Industrial Microbiology, Hubei University of Technology, Wuhan, China.
| |
Collapse
|
20
|
Wei L, Zhao D, Sun W, Lin L, Sui D, Li W, Gui Y, Wang J, Deng Y, Song Y. Targeting of TAMs with freeze-dried monosialotetrahexosylganglioside and sialic acid-octadecylamine co-modified liposomes remodels the tumor microenvironment and enhances anti-tumor activity. Eur J Pharm Biopharm 2023; 184:50-61. [PMID: 36682511 DOI: 10.1016/j.ejpb.2023.01.011] [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: 06/18/2022] [Revised: 12/25/2022] [Accepted: 01/16/2023] [Indexed: 01/22/2023]
Abstract
Although anti-tumor strategies targeting tumor-associated immune cells were being rapidly developed, the preparations were usually limited in targeting efficiency. To overcome this barrier, this study reported a novel sialic acid-octadecylamine (SA-ODA) and monosialotetrahexosylganglioside (GM1) co-modified epirubicin liposomes (5-5-SAGL-EPI), which improved tumor-targeting ability through the active targeting of tumor-associated macrophages (TAMs) by SA-ODA and the long circulation of GM1. Thus, we evaluated 5-5-SAGL-EPI in vitro and in vivo. Analysis of cellular uptake by RAW264.7 cells using flow cytometry and confocal microscopy showed a higher rate of cellular uptake for 5-5-SAGL-EPI than for the common liposomes (CL-EPI). In pharmacokinetic studies using Wistar rats, compared to CL-EPI, 5-5-SAGL-EPI showed a higher circulation time in vivo. Tissue distribution studies in Kunming mice bearing S180 tumors revealed increased distribution of 5-5-SAGL-EPI in tumor tissues compared with liposomes modified with single ligands (SA-ODA [5-SAL-EPI] or GM1 [5-GL-EPI]). In vivo anti-tumor experiments using the S180 tumor-bearing mice revealed a high tumor inhibition rate and low toxicity for 5-5-SAGL-EPI. Moreover, freeze-dried 5-5-SAGL-EPI had good storage stability, and the anti-tumor effect was comparable to that before freeze-drying. Overall, 5-5-SAGL-EPI exhibited excellent anti-tumor effects before and after lyophilization.
Collapse
Affiliation(s)
- Lu Wei
- College of Pharmacy, Shenyang Pharmaceutical University, Shenyang 110016, China.
| | - Dan Zhao
- Sinovac Life Sciences Co., Ltd., Beijing 100085, China.
| | - Wenliang Sun
- College of Pharmacy, Shenyang Pharmaceutical University, Shenyang 110016, China.
| | - Lin Lin
- College of Pharmacy, Shenyang Pharmaceutical University, Shenyang 110016, China.
| | - Dezhi Sui
- College of Pharmacy, Shenyang Pharmaceutical University, Shenyang 110016, China.
| | - Wen Li
- College of Pharmacy, Shenyang Pharmaceutical University, Shenyang 110016, China.
| | - Yangxu Gui
- College of Pharmacy, Shenyang Pharmaceutical University, Shenyang 110016, China.
| | - Jia Wang
- College of Pharmacy, Shenyang Pharmaceutical University, Shenyang 110016, China.
| | - Yihui Deng
- College of Pharmacy, Shenyang Pharmaceutical University, Shenyang 110016, China.
| | - Yanzhi Song
- College of Pharmacy, Shenyang Pharmaceutical University, Shenyang 110016, China.
| |
Collapse
|
21
|
Wang Y, Jin J, Wang HJ, Ju LA. Acoustic Force-Based Cell-Matrix Avidity Measurement in High Throughput. BIOSENSORS 2023; 13:95. [PMID: 36671930 PMCID: PMC9855465 DOI: 10.3390/bios13010095] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/17/2022] [Revised: 12/09/2022] [Accepted: 12/21/2022] [Indexed: 06/17/2023]
Abstract
Cancer cells interacting with the extracellular matrix (ECM) in the tumor microenvironment is pivotal for tumorigenesis, invasion, and metastasis. Cell-ECM adhesion has been intensively studied in cancer biology in the past decades to understand the molecular mechanisms underlying the adhesion events and extracellular mechanosensing, as well as develop therapeutic strategies targeting the cell adhesion molecules. Many methods have been established to measure the cell-ECM adhesion strength and correlate it with the metastatic potential of certain cancer types. However, those approaches are either low throughput, not quantitative, or with poor sensitivity and reproducibility. Herein, we developed a novel acoustic force spectroscopy based method to quantify the cell-ECM adhesion strength during adhesion maturation process using the emerging z-Movi® technology. This can be served as a fast, simple, and high-throughput platform for functional assessment of cell adhesion molecules in a highly predictive and reproducible manner.
Collapse
Affiliation(s)
- Yao Wang
- School of Biomedical Engineering, The University of Sydney, Darlington, NSW 2008, Australia
| | - Jasmine Jin
- School of Biomedical Engineering, The University of Sydney, Darlington, NSW 2008, Australia
| | - Haoqing Jerry Wang
- School of Biomedical Engineering, The University of Sydney, Darlington, NSW 2008, Australia
- Charles Perkins Centre, The University of Sydney, Camperdown, NSW 2006, Australia
| | - Lining Arnold Ju
- School of Biomedical Engineering, The University of Sydney, Darlington, NSW 2008, Australia
- Charles Perkins Centre, The University of Sydney, Camperdown, NSW 2006, Australia
- The University of Sydney Nano Institute (Sydney Nano), The University of Sydney, Camperdown, NSW 2006, Australia
| |
Collapse
|
22
|
Rossing E, Pijnenborg JFA, Boltje TJ. Chemical tools to track and perturb the expression of sialic acid and fucose monosaccharides. Chem Commun (Camb) 2022; 58:12139-12150. [PMID: 36222364 PMCID: PMC9623448 DOI: 10.1039/d2cc04275d] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2022] [Accepted: 10/05/2022] [Indexed: 11/24/2022]
Abstract
The biosynthesis of glycans is a highly conserved biological process and found in all domains of life. The expression of cell surface glycans is increasingly recognized as a target for therapeutic intervention given the role of glycans in major pathologies such as cancer and microbial infection. Herein, we summarize our contributions to the development of unnatural monosaccharide derivatives to infiltrate and alter the expression of both mammalian and bacterial glycans and their therapeutic application.
Collapse
Affiliation(s)
- Emiel Rossing
- Department of Synthetic Organic Chemistry, Institute for Molecules and Materials, Radboud University, Heyendaalseweg 135, 6525AJ, Nijmegen, The Netherlands.
| | - Johan F A Pijnenborg
- Department of Synthetic Organic Chemistry, Institute for Molecules and Materials, Radboud University, Heyendaalseweg 135, 6525AJ, Nijmegen, The Netherlands.
| | - Thomas J Boltje
- Department of Synthetic Organic Chemistry, Institute for Molecules and Materials, Radboud University, Heyendaalseweg 135, 6525AJ, Nijmegen, The Netherlands.
| |
Collapse
|