1
|
Leung KK, Schaefer K, Lin Z, Yao Z, Wells JA. Engineered Proteins and Chemical Tools to Probe the Cell Surface Proteome. Chem Rev 2025; 125:4069-4110. [PMID: 40178992 DOI: 10.1021/acs.chemrev.4c00554] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/05/2025]
Abstract
The cell surface proteome, or surfaceome, is the hub for cells to interact and communicate with the outside world. Many disease-associated changes are hard-wired within the surfaceome, yet approved drugs target less than 50 cell surface proteins. In the past decade, the proteomics community has made significant strides in developing new technologies tailored for studying the surfaceome in all its complexity. In this review, we first dive into the unique characteristics and functions of the surfaceome, emphasizing the necessity for specialized labeling, enrichment, and proteomic approaches. An overview of surfaceomics methods is provided, detailing techniques to measure changes in protein expression and how this leads to novel target discovery. Next, we highlight advances in proximity labeling proteomics (PLP), showcasing how various enzymatic and photoaffinity proximity labeling techniques can map protein-protein interactions and membrane protein complexes on the cell surface. We then review the role of extracellular post-translational modifications, focusing on cell surface glycosylation, proteolytic remodeling, and the secretome. Finally, we discuss methods for identifying tumor-specific peptide MHC complexes and how they have shaped therapeutic development. This emerging field of neo-protein epitopes is constantly evolving, where targets are identified at the proteome level and encompass defined disease-associated PTMs, complexes, and dysregulated cellular and tissue locations. Given the functional importance of the surfaceome for biology and therapy, we view surfaceomics as a critical piece of this quest for neo-epitope target discovery.
Collapse
Affiliation(s)
- Kevin K Leung
- Department of Pharmaceutical Chemistry, University of California San Francisco, San Francisco, California 94158, United States
| | - Kaitlin Schaefer
- Department of Pharmaceutical Chemistry, University of California San Francisco, San Francisco, California 94158, United States
| | - Zhi Lin
- Department of Pharmaceutical Chemistry, University of California San Francisco, San Francisco, California 94158, United States
| | - Zi Yao
- Department of Pharmaceutical Chemistry, University of California San Francisco, San Francisco, California 94158, United States
| | - James A Wells
- Department of Pharmaceutical Chemistry, University of California San Francisco, San Francisco, California 94158, United States
- Department of Cellular and Molecular Pharmacology, University of California San Francisco, San Francisco, California 94158, United States
| |
Collapse
|
2
|
Ambati S, Choudhury QJ, Peter JA, Moremen KW, Chapla DG, Lewis ZA, Lin X, Meagher RB. Siglec-targeted liposomes to identify sialoglycans present on fungal pathogens. Antimicrob Agents Chemother 2025; 69:e0172024. [PMID: 40084878 PMCID: PMC11963605 DOI: 10.1128/aac.01720-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2024] [Accepted: 02/16/2025] [Indexed: 03/16/2025] Open
Abstract
The sialic acid Ig-like lectins Siglec-3 and Siglec-15 are pathogen receptors that bind sialic acid-modified glycoproteins, best characterized in metastatic cancers. Because fungi produce sialoglycans and sialo-glycoproteins, we wondered if Siglecs had the potential for targeted delivery of antifungal drugs. We purified the extracellular V-region Ig-like C2 ligand-binding domains and stalk regions of SIG3 and SIG15. We floated the two polypeptides on the surface of liposomes loaded with amphotericin B (AmB) and labeled with rhodamine B to prepare SIG3-Ls and SIG15-Ls. Using these two reagents, we explored the sialoglycans of two evolutionarily distant and deadly human fungal pathogens, the Mucormycete Rhizopus delemar and the Ascomycete Aspergillus fumigatus. We found that SIG3-Ls and SIG15-Ls localized in a continuous layer over the cell wall surface of germ tubes and hyphae of both fungal species and to the conidia of A. fumigatus. Binding was Neu5Ac-specific and appeared confined to N-linked sialoglycans on fungal proteins. SIG3 and SIG15 proteins bound to diverse sialo-glycoproteins extracted from the hyphae of both species. SIG3-Ls and SIG15-Ls delivering sub-micromolar concentrations of AmB were moderately more effective at inhibiting and/or killing both species relative to control liposomes. We discuss the roles that sialo-glycoproteins may play in fungal pathogens.
Collapse
Affiliation(s)
- Suresh Ambati
- Department of Genetics, University of Georgia, Athens, Georgia, USA
| | | | - Jesse Ann Peter
- Department of Genetics, University of Georgia, Athens, Georgia, USA
| | - Kelley W. Moremen
- Department of Biochemistry and Molecular Biology, University of Georgia, Athens, Georgia, USA
- Complex Carbohydrate Research Center, University of Georgia, Athens, Georgia, USA
| | - Digantkumar Gopaldas Chapla
- Department of Biochemistry and Molecular Biology, University of Georgia, Athens, Georgia, USA
- Complex Carbohydrate Research Center, University of Georgia, Athens, Georgia, USA
| | - Zachary A. Lewis
- Department of Microbiology, University of Georgia, Athens, Georgia, USA
| | - Xiaorong Lin
- Department of Microbiology, University of Georgia, Athens, Georgia, USA
| | | |
Collapse
|
3
|
Izadi S, Abrantes R, Gumpelmair S, Kunnummel V, Duarte HO, Steinberger P, Reis CA, Castilho A. An engineered PD1-Fc fusion produced in N. benthamiana plants efficiently blocks PD1/PDL1 interaction. PLANT CELL REPORTS 2025; 44:80. [PMID: 40119938 PMCID: PMC11929711 DOI: 10.1007/s00299-025-03475-0] [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: 12/16/2024] [Accepted: 03/04/2025] [Indexed: 03/25/2025]
Abstract
KEY MESSAGE Plant-made PD1-Fc fusions engineered for optimized glycosylation and Fc-receptor engagement are highly efficient in blocking PD1/PDL1 interactions and can be cost-effective alternatives to antibody-based immune checkpoint inhibitors. Immune checkpoint inhibitors (ICIs) are antibodies to receptors that have pivotal roles during T-cell activation processes. The programmed cell death 1 (PD1) can be regarded as the primary immune checkpoint and antibodies targeting PD1 or its ligand PDL1 have revolutionized immunotherapy of cancer. However, the majority of patients fail to respond, and treatment resistance as well as immune-related adverse events are commonly associated with this therapy. Alternatives to antibody-based ICIs targeting the PD1 pathway may bear the potential to overcome some of these shortcomings. Here, we have used a plant expression platform based on the tobacco relative Nicotiana benthamiana to generate immunoglobulin fusion proteins harboring the wild type or an affinity-enhanced PD1 ectodomain. We have exploited the versatility of our system to generate variants that differed regarding their glycosylation profile as well as their capability to engage Fc-receptors. Unlike its wild-type counterpart, the affinity-enhanced versions showed strongly augmented capabilities to engage PDL1 in both protein- and cell-based assays. Moreover, in contrast with clinical antibodies, their binding is not affected by the glycosylation status of PDL1. Importantly, we could demonstrate that the plant-made PD1 fusion proteins are highly efficient in blocking inhibitory PD1 signaling in a T cell reporter assay. Taken together, our study highlights the utility of our plant-based protein expression platform to generate biologics with therapeutic potential. Targeting PDL1 with plant derived affinity-enhanced PD1 immunoglobulin fusion proteins may reduce overstimulation associated with antibody-based therapies while retaining favorable features of ICIs such as long serum half-life.
Collapse
Affiliation(s)
- Shiva Izadi
- Department of Biotechnology and Food Science Institute of Plant Biotechnology and Cell Biology, BOKU University, Muthgasse 18, 1190, Vienna, Austria
| | - Rafaela Abrantes
- i3S Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
- Institute of Molecular Pathology and Immunology, University of Porto (IPATIMUP), Porto, Portugal
- Instituto de Ciências Biomédicas Abel Salazar (ICBAS), Universidade do Porto, Porto, Portugal
| | - Simon Gumpelmair
- Division of Immune Receptors and T Cell Activation, Institute of Immunology, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria
| | - Vinny Kunnummel
- Department of Biotechnology and Food Science Institute of Plant Biotechnology and Cell Biology, BOKU University, Muthgasse 18, 1190, Vienna, Austria
| | - Henrique O Duarte
- i3S Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
- Institute of Molecular Pathology and Immunology, University of Porto (IPATIMUP), Porto, Portugal
| | - Peter Steinberger
- Division of Immune Receptors and T Cell Activation, Institute of Immunology, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria
| | - Celso A Reis
- i3S Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
- Institute of Molecular Pathology and Immunology, University of Porto (IPATIMUP), Porto, Portugal
- Instituto de Ciências Biomédicas Abel Salazar (ICBAS), Universidade do Porto, Porto, Portugal
- Faculty of Medicine (FMUP), University of Porto, Porto, Portugal
| | - Alexandra Castilho
- Department of Biotechnology and Food Science Institute of Plant Biotechnology and Cell Biology, BOKU University, Muthgasse 18, 1190, Vienna, Austria.
| |
Collapse
|
4
|
Drummond-Guy O, Daly J, Wu A, Stewart N, Milne K, Duff C, Nelson BH, Williams KC, Wisnovsky S. Polysialic acid is upregulated on activated immune cells and negatively regulates anticancer immune activity. Front Oncol 2025; 15:1520948. [PMID: 40182033 PMCID: PMC11965634 DOI: 10.3389/fonc.2025.1520948] [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: 10/31/2024] [Accepted: 02/20/2025] [Indexed: 04/05/2025] Open
Abstract
Suppression of anticancer immune function is a key driver of tumorigenesis. Identifying molecular pathways that inhibit anticancer immunity is critical for developing novel immunotherapeutics. One such molecule that has recently been identified is the carbohydrate polysialic acid (polySia), whose expression is dramatically upregulated on both cancer cells and immune cells in breast cancer patient tissues. The role of polySia in the anticancer immune response, however, remains incompletely understood. In this study, we profile polySia expression on both healthy primary immune cells and on infiltrating immune cells in the tumour microenvironment (TME). These studies reveal polySia expression on multiple immune cell subsets in patient breast tumors. We find that stimulation of primary T-cells and macrophages in vitro induces a significant upregulation of polySia expression. We subsequently show that polySia is appended to a range of different carrier proteins within these immune cells. Finally, we find that selective removal of polySia can significantly potentiate killing of breast cancer cells by innate immune cells. These studies implicate polySia as a significant negative regulator of anticancer immunity.
Collapse
Affiliation(s)
- Olivia Drummond-Guy
- Faculty of Pharmaceutical Sciences, University of British Columbia, Vancouver, BC, Canada
| | - John Daly
- Faculty of Pharmaceutical Sciences, University of British Columbia, Vancouver, BC, Canada
| | - Angeline Wu
- Faculty of Pharmaceutical Sciences, University of British Columbia, Vancouver, BC, Canada
| | - Natalie Stewart
- Faculty of Pharmaceutical Sciences, University of British Columbia, Vancouver, BC, Canada
| | - Katy Milne
- Deeley Research Centre, British Columbia (BC) Cancer, Victoria, BC, Canada
| | - Chloe Duff
- Deeley Research Centre, British Columbia (BC) Cancer, Victoria, BC, Canada
| | - Brad H. Nelson
- Deeley Research Centre, British Columbia (BC) Cancer, Victoria, BC, Canada
- Department of Medical Genetics, University of British Columbia, Vancouver, BC, Canada
- Department of Biochemistry and Microbiology, University of Victoria, Victoria, BC, Canada
| | - Karla C. Williams
- Faculty of Pharmaceutical Sciences, University of British Columbia, Vancouver, BC, Canada
| | - Simon Wisnovsky
- Faculty of Pharmaceutical Sciences, University of British Columbia, Vancouver, BC, Canada
| |
Collapse
|
5
|
Radu KR, Baek KH. Insights on the Role of Sialic Acids in Acute Lymphoblastic Leukemia in Children. Int J Mol Sci 2025; 26:2233. [PMID: 40076855 PMCID: PMC11900591 DOI: 10.3390/ijms26052233] [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: 01/02/2025] [Revised: 02/11/2025] [Accepted: 02/21/2025] [Indexed: 03/14/2025] Open
Abstract
Sialic acids serve as crucial terminal sugars on glycoproteins or glycolipids present on cell surfaces. These sugars are involved in diverse physiological and pathological processes through their interactions with carbohydrate-binding proteins, facilitating cell-cell communication and influencing the outcomes of bacterial and viral infections. The role of hypersialylation in tumor growth and metastasis has been widely studied. Recent research has highlighted the significance of aberrant sialylation in enabling tumor cells to escape immune surveillance and sustain their malignant behavior. Acute lymphoblastic leukemia (ALL) is a heterogenous hematological malignancy that primarily affects children and is the second leading cause of mortality among individuals aged 1 to 14. ALL is characterized by the uncontrolled proliferation of immature lymphoid cells in the bone marrow, peripheral blood, and various organs. Sialic acid-binding immunoglobulin-like lectins (Siglecs) are cell surface proteins that can bind to sialic acids. Activation of Siglecs triggers downstream reactions, including induction of cell apoptosis. Siglec-7 and Siglec-9 have been reported to promote cancer progression by driving macrophage polarization, and their expressions on natural killer cells can inhibit tumor cell death. This comprehensive review aims to explore the sialylation mechanisms and their effects on ALL in children. Understanding the complex interplay between sialylation and ALL holds great potential for developing novel diagnostic tools and therapeutic interventions in managing this pediatric malignancy.
Collapse
Affiliation(s)
- Kimberley Rinai Radu
- Department of Life Science, Graduate School, CHA University, Seongnam-si 13488, Gyeonggi-do, Republic of Korea;
| | - Kwang-Hyun Baek
- Department of Life Science, Graduate School, CHA University, Seongnam-si 13488, Gyeonggi-do, Republic of Korea;
- Department of Bioconvergence, Graduate School, CHA University, Seongnam-si 13488, Gyeonggi-do, Republic of Korea
| |
Collapse
|
6
|
Lin SY, Schmidt EN, Takahashi-Yamashiro K, Macauley MS. Roles for Siglec-glycan interactions in regulating immune cells. Semin Immunol 2025; 77:101925. [PMID: 39706106 DOI: 10.1016/j.smim.2024.101925] [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: 10/04/2024] [Revised: 12/08/2024] [Accepted: 12/10/2024] [Indexed: 12/23/2024]
Abstract
Cell surface complex carbohydrates, known as glycans, are positioned to be the first point of contact between two cells. Indeed, interactions between glycans with glycan-binding can modulate cell-cell interactions. This concept is particularly relevant for immune cells, which use an array of glycan-binding proteins to help in the process of differentiating 'self' from 'non-self'. This is exemplified by the sialic acid-binding immunoglobulin-type lectins (Siglecs), which recognize sialic acid. Given that sialic acid is relatively unique to vertebrates, immune cells leverage Siglecs to recognize sialic acid as a marker of 'self'. Siglecs serve many biological roles, with most of these functions regulated through interactions with their sialoglycan ligands. In this review, we provide a comprehensive update on the ligands of Siglecs and how Siglec-sialoglycan interactions help regulate immune cells in the adaptive and innate immune system.
Collapse
Affiliation(s)
- Sung-Yao Lin
- Department of Medical Microbiology and Immunology, University of Alberta, Edmonton, Alberta, Canada
| | - Edward N Schmidt
- Department of Chemistry, University of Alberta, Edmonton, Alberta, Canada
| | | | - Matthew S Macauley
- Department of Medical Microbiology and Immunology, University of Alberta, Edmonton, Alberta, Canada; Department of Chemistry, University of Alberta, Edmonton, Alberta, Canada; Neuroscience and Mental Health Institute, University of Alberta, Edmonton, Alberta, Canada.
| |
Collapse
|
7
|
Feng Y, Chen H, Chen S, Zhang K, Yun D, Liu D, Zeng J, Yang C, Xie Q. Disulfiram-Loaded PLGA nanoparticles modified with a Phenyl borate chitosan Conjugate enhance hepatic carcinoma treatment. Int J Pharm 2025; 671:125293. [PMID: 39880140 DOI: 10.1016/j.ijpharm.2025.125293] [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: 10/01/2024] [Revised: 01/14/2025] [Accepted: 01/26/2025] [Indexed: 01/31/2025]
Abstract
Disulfiram (DSF), which has been traditionally used to treat alcoholism, has been shown to inhibit tumor growth, indicating its potential as an anticancer agent. However, its development and application are hindered by its poor water solubility, instability in physiological environments, and low bioavailability. In this study, phenylboronic acid-chitosan (PBA-CS) grafts were synthesized using the carbodiimide method. PBA-CS-modified DSF PLGA nanoparticles (DSF@PBA-CS-PLGA NPs) were constructed by coating the nanoparticle surfaces with PBA-CS to improve the stability of DSF in physiological environments and enhance its anti-tumor effects. The structures of PBA-CS and the DSF@PBA-CS-PLGA NPs were confirmed using FTIR UVs, DLS, ELS, TEM, 1HNMR, DSC. Our in vitro degradation experiments showed that PBA-CS-PLGA NPs significantly improved the stability of DSF in physiological environments. Cell experiments showed that PBA-CS-PLGA NPs improved drug uptake and strongly inhibited HepG2 cell migration. A mouse tumor model was established using Dutch H22 cells. DSF@PBA-CS-PLGA NPs showed better tumor-targeting ability than DSF@PLGA NPs, with a tumor inhibition rate of more than 60%, and they induced apoptosis and inhibited neovascularization in mouse tumor tissues. Both the in vitro and in vivo experiments indicated that the DSF@PBA-CS-PLGA NPs overcame the limitations of DSF, improving the dissolution rate and stability of the drug, ultimately offering low toxicity, sustained release, and targeted delivery. These findings demonstrated the potential of DSF@PBA-CS-PLGA NPs for hepatic carcinoma therapy.
Collapse
Affiliation(s)
- Yanyi Feng
- Center for New Drug Research and Development, Guangdong Pharmaceutical University, Guangzhou 510006 China
| | - Hongyu Chen
- Center for New Drug Research and Development, Guangdong Pharmaceutical University, Guangzhou 510006 China
| | - Simiao Chen
- Center for New Drug Research and Development, Guangdong Pharmaceutical University, Guangzhou 510006 China
| | - Kaijun Zhang
- Center for New Drug Research and Development, Guangdong Pharmaceutical University, Guangzhou 510006 China
| | - Dan Yun
- Center for New Drug Research and Development, Guangdong Pharmaceutical University, Guangzhou 510006 China
| | - Dengyuan Liu
- Center for New Drug Research and Development, Guangdong Pharmaceutical University, Guangzhou 510006 China
| | - Jinxin Zeng
- Center for New Drug Research and Development, Guangdong Pharmaceutical University, Guangzhou 510006 China
| | - Chutong Yang
- Center for New Drug Research and Development, Guangdong Pharmaceutical University, Guangzhou 510006 China
| | - Qingchun Xie
- Center for New Drug Research and Development, Guangdong Pharmaceutical University, Guangzhou 510006 China; Guangdong Provincial Key Laboratory for Research and Evaluation of Pharmaceutical Preparations, Guangdong Pharmaceutical University, Guangzhou 510006 China; Guangdong Provincial Engineering Center of Topical Precision Drug Delivery System, Guangdong Pharmaceutical University, Guangzhou 510006 China.
| |
Collapse
|
8
|
Jung J, Schmidt EN, Chang HC, Jame-Chenarboo Z, Enterina JR, McCord KA, Gray TE, Kageler L, St Laurent CD, Wang C, Flynn RA, Wu P, Khoo KH, Macauley MS. Understanding the Glycosylation Pathways Involved in the Biosynthesis of the Sulfated Glycan Ligands for Siglecs. ACS Chem Biol 2025; 20:386-400. [PMID: 39836965 DOI: 10.1021/acschembio.4c00677] [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: 01/23/2025]
Abstract
Carbohydrate sulfation plays a pivotal role in modulating the strength of Siglec-glycan interactions. Recently, new aspects of Siglec binding to sulfated cell surface carbohydrates have been discovered, but the class of glycan presenting these sulfated Siglec ligands has not been fully elucidated. In this study, the contribution of different classes of glycans to cis and trans Siglec ligands was investigated within cells expressing the carbohydrate sulfotransferase 1 (CHST1) or CHST2. For some Siglecs, the glycan class mediating binding was clear, such as O-glycans for Siglec-7 and N-glycans for Siglec-2 and Siglec-9. Both N-glycans and mucin-type O-glycans contributed to ligands for Siglec-3, -5, -8, and -15. However, significant levels of Siglec-3 and -8 ligands remained in CHST1-expressing cells lacking complex N-glycans and mucin-type O-glycans. A combination of genetic, pharmacological, and enzymatic treatment strategies ruled out heparan sulfates and glycoRNA as contributors, although Siglec-8 did exhibit some binding to glycolipids. Genetic disruption of O-mannose glycans within CHST1-expressing cells had a small but significant impact on Siglec-3 and -8 binding, demonstrating that this class of glycans can present sulfated Siglec ligands. We also investigated the ability of sulfated cis ligands to mask Siglec-3 and Siglec-7. For Siglec-7, cis ligands were again found to be mucin-type O-glycans. While N-glycans were the major sulfated trans ligands for Siglec-3, disruption of complex mucin-type O-glycans had the largest impact on Siglec-3 masking. Overall, this study enhances our knowledge of the types of sulfated glycans that can serve as Siglec ligands.
Collapse
Affiliation(s)
- Jaesoo Jung
- Department of Chemistry, University of Alberta, Edmonton T6G 2R3, Canada
| | - Edward N Schmidt
- Department of Chemistry, University of Alberta, Edmonton T6G 2R3, Canada
| | - Hua-Chien Chang
- Institute of Biological Chemistry, Academia Sinica, Taipei 115024, Taiwan
| | | | - Jhon R Enterina
- Department of Medical Microbiology and Immunology, University of Alberta, Edmonton T6G 2R3, Canada
| | - Kelli A McCord
- Department of Chemistry, University of Alberta, Edmonton T6G 2R3, Canada
| | - Taylor E Gray
- Department of Chemistry, University of Alberta, Edmonton T6G 2R3, Canada
| | - Lauren Kageler
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, Massachusetts 02138, United States
| | - Chris D St Laurent
- Department of Chemistry, University of Alberta, Edmonton T6G 2R3, Canada
| | - Chao Wang
- Department of Molecular and Cellular Biology, Scripps Research Institute, La Jolla, California 92037-1000, United States
| | - Ryan A Flynn
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, Massachusetts 02138, United States
- Stem Cell Program and Division of Hematology/Oncology, Boston Children's Hospital, Boston, Massachusetts 02115-5724, United States
| | - Peng Wu
- Department of Molecular and Cellular Biology, Scripps Research Institute, La Jolla, California 92037-1000, United States
| | - Kay-Hooi Khoo
- Institute of Biological Chemistry, Academia Sinica, Taipei 115024, Taiwan
| | - Matthew S Macauley
- Department of Chemistry, University of Alberta, Edmonton T6G 2R3, Canada
- Department of Medical Microbiology and Immunology, University of Alberta, Edmonton T6G 2R3, Canada
| |
Collapse
|
9
|
Lam YY, Tan A, Kempe K, Boyd BJ. Metabolic glycan labelling with bio-orthogonal targeting and its potential in drug delivery. J Control Release 2025; 378:880-898. [PMID: 39694071 DOI: 10.1016/j.jconrel.2024.12.018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2024] [Revised: 11/21/2024] [Accepted: 12/09/2024] [Indexed: 12/20/2024]
Abstract
New modes of targeted drug delivery are emerging with promise of enhancing therapeutic efficacy while reducing side effects. This review examines the landscape of metabolic glycan labelling-a technique gaining traction for its potential in specific drug targeting. By exploiting the natural glycan synthetic pathway of monosaccharides, unnatural sugar analogues are incorporated into glycoproteins, allowing for the presentation of unique functional groups on cells. This enables specific targeting using 'clickable' probes with complementary click chemistry functional groups. The selection of sugar analogues and chemical tags are crucial components explored in this review, alongside considerations for cell lines, tissues, and cargo selection. The review discusses non-therapeutic and therapeutic applications of metabolic glycan labelling, as well as its potential beyond labelling of cell surfaces. The review also highlights underexplored areas of metabolic glycan labelling by assessing the limited literature addressing labelling efficiency, turnover rates, the impact of sugar supplements in cell culture, and the critical cell to functionalised sugar ratio. Furthermore, this review delves into the future landscape and goals of metabolic glycan labelling, envisioning its potential in targeted drug delivery.
Collapse
Affiliation(s)
- Yuen Yi Lam
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University (Parkville Campus), 381 Royal Parade, Parkville, VIC 3052, Australia
| | - Angel Tan
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University (Parkville Campus), 381 Royal Parade, Parkville, VIC 3052, Australia
| | - Kristian Kempe
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University (Parkville Campus), 381 Royal Parade, Parkville, VIC 3052, Australia; Materials Science and Engineering, Monash University, Clayton, VIC 3800, Australia
| | - Ben J Boyd
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University (Parkville Campus), 381 Royal Parade, Parkville, VIC 3052, Australia; Materials Science and Engineering, Monash University, Clayton, VIC 3800, Australia; Department of Pharmacy, University of Copenhagen Universitetsparken 2, 2100 Copenhagen, Denmark.
| |
Collapse
|
10
|
Blanco R, Muñoz JP. The Role of Non-Human Sialic Acid Neu5Gc-Containing Glycoconjugates in Human Tumors: A Review of Clinical and Experimental Evidence. Biomolecules 2025; 15:253. [PMID: 40001556 PMCID: PMC11853303 DOI: 10.3390/biom15020253] [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: 12/03/2024] [Revised: 12/30/2024] [Accepted: 01/02/2025] [Indexed: 02/27/2025] Open
Abstract
N-Glycolylneuraminic acid (Neu5Gc) is a sialic acid variant commonly found in most mammals but not synthesized by humans due to an inactivating mutation in the CMP-Neu5Ac hydroxylase (CMAH) gene. Despite this, Neu5Gc-containing molecules are consistently detected in human tissues, particularly in malignant tumors. However, the mechanisms underlying Neu5Gc accumulation and its role in cancer development remain poorly understood. OBJECTIVES This review aims to analyze clinical and experimental evidence regarding the presence of Neu5Gc-containing glycoconjugates in both tumor and non-tumor human tissues, exploring potential mechanisms of the Neu5Gc expression and evaluating its contribution to tumor biology, with a particular focus on the Neu5Gc-GM3 ganglioside. METHODS A comprehensive review of the literature was conducted, integrating findings from immunohistochemistry, chromatography, and molecular studies to assess the expression and implications of Neu5Gc in cancer biology. RESULTS Neu5Gc-containing glycoconjugates were found to preferentially accumulate in various malignant tumors, while their presence in normal tissues was restricted to cells with high turnover rates. This accumulation is potentially mediated by dietary uptake, hypoxic conditions, and metabolic alterations in cancer cells. Additionally, Neu5Gc-containing molecules were associated with the activation of oncogenic pathways. CONCLUSION Neu5Gc-containing glycoconjugates play a multifaceted role in cancer progression and present potential as prognostic markers and therapeutic targets.
Collapse
Affiliation(s)
- Rancés Blanco
- Investigador Independiente, Av. Vicuña Mackenna Poniente 6315, La Florida 8240000, Chile;
| | - Juan P. Muñoz
- Laboratorio de Bioquímica, Departamento de Química, Facultad de Ciencias, Universidad de Tarapacá, Arica 1000007, Chile
| |
Collapse
|
11
|
Eggermont L, Lumen N, Van Praet C, Delanghe J, Rottey S, Vermassen T. A comprehensive view of N-glycosylation as clinical biomarker in prostate cancer. Biochim Biophys Acta Rev Cancer 2025; 1880:189239. [PMID: 39672278 DOI: 10.1016/j.bbcan.2024.189239] [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: 07/02/2024] [Revised: 10/25/2024] [Accepted: 12/06/2024] [Indexed: 12/15/2024]
Abstract
Alterations in the prostate cancer (PCa) N-glycome have gained attention as a potential biomarker. This comprehensive review explores the diversity of N-glycosylation patterns observed in PCa-related cell lines, tissue, serum and urine, focusing on prostate-specific antigen (PSA) and the total pool of glycoproteins. Within the context of PCa, altered N-glycosylation patterns are a mechanism of immune escape and a disruption in normal glycoprotein distribution and trafficking. Glycoproteins with PCa-induced N-glycosylation patterns tend to accumulate in prostate tissue and the bloodstream, thereby diminishing N-glycan proportions in urine. Based on literary observations, aberrations in N-glycan branching are probably a characteristic of metabolic reprogramming and (chronic) inflammation. Changes in (core) fucosylation, specific N-glycosylation structures (such as N,N'-diacetyllactosamine) and high-mannose glycans otherwise are more likely indicators of cancer development and progression. Further investigation into these PCa-specific alterations holds promise in the discovery of new diagnostic, prognostic and response prediction biomarkers in PCa.
Collapse
Affiliation(s)
- Lissa Eggermont
- Dept. Medical Oncology, Ghent University Hospital, Ghent, Belgium; Biomarkers in Cancer research group, Dept. Basic and Applied Medicine, Ghent University, Ghent, Belgium; Cancer Research Institute Ghent, Ghent, Belgium
| | - Nicolaas Lumen
- Cancer Research Institute Ghent, Ghent, Belgium; Dept. Urology, Ghent University Hospital, Ghent, Belgium; Uro-Oncology research group, Dept. Human Structure and Repair, Ghent University, Ghent, Belgium
| | - Charles Van Praet
- Cancer Research Institute Ghent, Ghent, Belgium; Dept. Urology, Ghent University Hospital, Ghent, Belgium; Uro-Oncology research group, Dept. Human Structure and Repair, Ghent University, Ghent, Belgium
| | - Joris Delanghe
- Cancer Research Institute Ghent, Ghent, Belgium; Dept. Diagnostic Sciences, Faculty of Medicine and Health Sciences, Ghent University, Ghent, Belgium
| | - Sylvie Rottey
- Dept. Medical Oncology, Ghent University Hospital, Ghent, Belgium; Biomarkers in Cancer research group, Dept. Basic and Applied Medicine, Ghent University, Ghent, Belgium; Cancer Research Institute Ghent, Ghent, Belgium; Drug Research Unit Ghent, Ghent University Hospital, Ghent, Belgium
| | - Tijl Vermassen
- Dept. Medical Oncology, Ghent University Hospital, Ghent, Belgium; Biomarkers in Cancer research group, Dept. Basic and Applied Medicine, Ghent University, Ghent, Belgium; Cancer Research Institute Ghent, Ghent, Belgium.
| |
Collapse
|
12
|
Yue S, Chen Y, Cui W, Lu X, Shen Y, Zhou F, Guan J, Chen J, Wen Q, Chen Y. Multi-center study on the application potential of Siaα-2,6Gal in early and differential diagnosis of lung cancer. Clin Chim Acta 2025; 566:120031. [PMID: 39547554 DOI: 10.1016/j.cca.2024.120031] [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: 08/28/2024] [Revised: 11/04/2024] [Accepted: 11/05/2024] [Indexed: 11/17/2024]
Abstract
OBJECTIVE This study aimed to investigate the application potential of the abnormal glycan structure Siaα-2,6Gal in the early and differential diagnosis of lung cancer. METHODS Clinical data and serum samples from 730 patients and 120 healthy individuals participating in clinical trials on Siaα-2,6Gal were collected at three medical centers between January 2022 and June 2023. The levels of Siaα-2,6Gal, carcinoembryonic antigen (CEA), cytokeratin 19 fragment antigen (CYFRA21-1), squamous cell carcinoma antigen (SCC), neuron-specific enolase (NSE), and pro-gastrin-releasing peptide (ProGRP) in serum were measured. The application potentials of these markers in the early and differential diagnosis, classification, and staging of lung cancer were explored. RESULTS (1) Serum Siaα-2,6Gal levels in the lung cancer group were 2,606 (1,970-3,458) U/mL, significantly higher than those in the benign lung disease, miscellaneous malignant tumor, miscellaneous benign disease, and healthy individual groups at 1,359 (950-1,528), 1,252 (903-1,532), 1,196 (850-1,490), and 1,210 (1,100-1,287) U/mL (P < 0.0001). (2) Serum Siaα-2,6Gal levels in the early-stage lung cancer (stages 0-II) group were 2,576 (1,929-3,338) U/mL, significantly higher than those in the benign pulmonary nodule group at 1,419 (1,105-1,820) U/mL (P < 0.0001). (3) Receiver operating characteristic curves showed that Siaα-2,6Gal had a high diagnostic efficiency for lung cancer (area under the curve (AUC) = 0.9217), significantly superior to CEA, CYFRA21-1, SCC, NSE, and ProGRP (AUCs of 0.6618, 0.6605, 0.5783, 0.5985, and 0.6381). CONCLUSION Siaα-2,6Gal is a promising biomarker for lung cancer diagnosis and may offer superior differential diagnosis of early-stage lung cancer from benign pulmonary nodules compared to traditional tumor markers.
Collapse
Affiliation(s)
- Si Yue
- Wenzhou Medical University, China
| | | | - Wenhao Cui
- Department of Clinical Laboratory Medicine, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong, 510000, China
| | - Xiuwei Lu
- Zhejiang Zijing Biotechnology Co., Ltd, China
| | - Yuhuan Shen
- Laboratory Medicine Center, Department of Clinical Laboratory, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Feifei Zhou
- Laboratory Medicine Center, Department of Clinical Laboratory, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Jinju Guan
- Laboratory Medicine Center, Department of Clinical Laboratory, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Jierong Chen
- Department of Clinical Laboratory Medicine, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong, 510000, China
| | - Qiuyuan Wen
- Department of Pathology, The Second Xiangya Hospital, Central South University, Changsha 410011, Hunan, China
| | - Yongjian Chen
- Laboratory Medicine Center, Department of Clinical Laboratory, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, China.
| |
Collapse
|
13
|
Wang S, Ling L, Ding CF. Integrating Chemoselective Labeling and Laser-Cleavable Mass Tagging for Determination of Sialic Acids in Glycoconjugates. Anal Chem 2025; 97:38-42. [PMID: 39749466 DOI: 10.1021/acs.analchem.4c03591] [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: 01/04/2025]
Abstract
Sialic acids are the terminal units of glycans in glycoproteins or glycolipids. The determination of sialic acids in glycoconjugates is crucial since they regulate essential biological functions and have a significant nutritional value. To achieve a specific and high-throughput in situ determination of sialic acids in glycoconjugates, a laser-desorption/ionization mass spectrometry (LDI-MS)-based strategy is reported by integrating chemoselective labeling and laser-cleavable mass tagging. 1-Pyrenebutyric hydrazide (PBH), a commercially available reagent that contains a pyrene moiety and a hydrazide group, has been developed as a novel laser-cleavable mass tag. For chemoselective labeling, an aldehyde group is introduced to the polyhydroxy side chain of sialic acids through mild periodate oxidation and then reacted with PBH, achieving the in situ determination of sialic acids. The quantitative determination of sialic acids in the range of 5-1000 μM (R2 = 0.99984) was achieved using an internal standard method. Thus, a specific, quantitative, and high-throughput method was developed for the in situ determination of sialic acids in glycoconjugates. Finally, this method has been successfully used to quantify the sialic acid content in EBN proteins and glycoprotein biopharmaceuticals, showing its practical application in the quality control of nutritional and therapeutic glycoprotein products. Additionally, the pyrene moiety, when linked to other reactive groups, can also be utilized to analyze other biomolecules, offering a new route for the rational design of mass tags.
Collapse
Affiliation(s)
- Shuyi Wang
- Key Laboratory of Advanced Mass Spectrometry and Molecular Analysis of Zhejiang Province, Institute of Mass Spectrometry, School of Material Science and Chemical Engineering, Ningbo University, Ningbo 315211, China
| | - Ling Ling
- Key Laboratory of Advanced Mass Spectrometry and Molecular Analysis of Zhejiang Province, Institute of Mass Spectrometry, School of Material Science and Chemical Engineering, Ningbo University, Ningbo 315211, China
| | - Chuan-Fan Ding
- Key Laboratory of Advanced Mass Spectrometry and Molecular Analysis of Zhejiang Province, Institute of Mass Spectrometry, School of Material Science and Chemical Engineering, Ningbo University, Ningbo 315211, China
| |
Collapse
|
14
|
He J, Zhang F, Wu B, Yu W. ST8SIA6 Sialylates CD24 to Enhance Its Membrane Localization in BRCA. Cells 2024; 14:9. [PMID: 39791710 PMCID: PMC11719756 DOI: 10.3390/cells14010009] [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: 11/14/2024] [Revised: 12/23/2024] [Accepted: 12/23/2024] [Indexed: 01/12/2025] Open
Abstract
CD24, a highly sialylated glycosyl-phosphatidyl-inositol (GPI) cell surface protein that interacts with sialic acid-binding immunoglobulin-like lectins (Siglecs), serves as an innate immune checkpoint and plays a crucial role in inflammatory diseases and tumor progression. Recently, cytoplasmic CD24 has been observed in samples from patients with cancer. However, whether sialylation governs the subcellular localization of CD24 in cancer remains unclear, and the impact of CD24 expression and localization on the clinical prognosis of cancer remains controversial. Here, we performed a systematic pan-cancer analysis of the gene expression levels and clinical correlation of CD24. Our analysis revealed that CD24 was highly expressed in breast tumor tissues and tumor cells, significantly shortening patient survival time. However, this correlation was not evident in other types of cancer. Additionally, a correlation analysis of CD24 levels with sialyltransferases (STs) revealed that ST8SIA6 is the key ST affecting CD24 sialylation. Further investigation demonstrated that ST8SIA6 directly modified CD24, promoting its localization to the cell membrane. Taken together, these findings elucidate, for the first time, the mechanisms by which ST8SIA6 regulates CD24 subcellular localization, providing new insights into the biological functions and applications of CD24.
Collapse
Affiliation(s)
- Jinxia He
- Key Laboratory of Marine Drugs (Ministry of Education), Shandong Provincial Key Laboratory of Glycoscience and Glycoengineering, School of Medicine and Pharmacy, Ocean University of China, Qingdao 266003, China; (J.H.); (F.Z.); (B.W.)
- Laboratory for Marine Drugs and Bioproducts, Qingdao Marine Science and Technology Center, Qingdao 266237, China
| | - Fengchao Zhang
- Key Laboratory of Marine Drugs (Ministry of Education), Shandong Provincial Key Laboratory of Glycoscience and Glycoengineering, School of Medicine and Pharmacy, Ocean University of China, Qingdao 266003, China; (J.H.); (F.Z.); (B.W.)
- Laboratory for Marine Drugs and Bioproducts, Qingdao Marine Science and Technology Center, Qingdao 266237, China
| | - Baihai Wu
- Key Laboratory of Marine Drugs (Ministry of Education), Shandong Provincial Key Laboratory of Glycoscience and Glycoengineering, School of Medicine and Pharmacy, Ocean University of China, Qingdao 266003, China; (J.H.); (F.Z.); (B.W.)
- Laboratory for Marine Drugs and Bioproducts, Qingdao Marine Science and Technology Center, Qingdao 266237, China
| | - Wengong Yu
- Key Laboratory of Marine Drugs (Ministry of Education), Shandong Provincial Key Laboratory of Glycoscience and Glycoengineering, School of Medicine and Pharmacy, Ocean University of China, Qingdao 266003, China; (J.H.); (F.Z.); (B.W.)
- Laboratory for Marine Drugs and Bioproducts, Qingdao Marine Science and Technology Center, Qingdao 266237, China
| |
Collapse
|
15
|
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
|
16
|
Lima GM, Jame-Chenarboo Z, Sojitra M, Sarkar S, Carpenter EJ, Yang CY, Schmidt E, Lai J, Atrazhev A, Yazdan D, Peng C, Volker EA, Ho R, Monteiro G, Lai R, Mahal LK, Macauley MS, Derda R. The liquid lectin array detects compositional glycocalyx differences using multivalent DNA-encoded lectins on phage. Cell Chem Biol 2024; 31:1986-2001.e9. [PMID: 39454580 DOI: 10.1016/j.chembiol.2024.09.010] [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: 01/22/2024] [Revised: 07/05/2024] [Accepted: 09/26/2024] [Indexed: 10/28/2024]
Abstract
Selective detection of disease-associated changes in the glycocalyx is an emerging field in modern targeted therapies. Detecting minor glycan changes on the cell surface is a challenge exacerbated by the lack of correspondence between cellular DNA/RNA and glycan structures. We demonstrate that multivalent displays of lectins on DNA-barcoded phages-liquid lectin array (LiLA)-detect subtle differences in density of glycans on cells. LiLA constructs displaying 73 copies of diCBM40 (CBM) lectin per virion (φ-CBM73) exhibit non-linear ON/OFF-like recognition of sialoglycans on the surface of normal and cancer cells. A high-valency φ-CBM290 display, or soluble CBM protein, cannot amplify the subtle differences detected by φ-CBM73. Similarly, multivalent displays of CBM and Siglec-7 detect differences in the glycocalyx between stem-like and non-stem populations in cancer. Multivalent display of lectins offer in situ detection of minor differences in glycocalyx in cells both in vitro and in vivo not feasible to currently available technologies.
Collapse
Affiliation(s)
- Guilherme M Lima
- Department of Chemistry, University of Alberta, Edmonton, AB T6G 2G2, Canada
| | | | - Mirat Sojitra
- Department of Chemistry, Stanford University, Stanford, CA 94305, USA
| | - Susmita Sarkar
- Department of Chemistry, University of Alberta, Edmonton, AB T6G 2G2, Canada
| | - Eric J Carpenter
- Department of Chemistry, University of Alberta, Edmonton, AB T6G 2G2, Canada
| | - Claire Y Yang
- Department of Chemistry, University of Alberta, Edmonton, AB T6G 2G2, Canada
| | - Edward Schmidt
- Department of Chemistry, University of Alberta, Edmonton, AB T6G 2G2, Canada
| | - Justine Lai
- Department of Medicine, University of Alberta, Edmonton, AB T6G 1C9, Canada
| | - Alexey Atrazhev
- Department of Chemistry, University of Alberta, Edmonton, AB T6G 2G2, Canada
| | - Danial Yazdan
- Department of Chemistry, University of Alberta, Edmonton, AB T6G 2G2, Canada
| | - Chuanhao Peng
- Department of Chemistry, University of Alberta, Edmonton, AB T6G 2G2, Canada
| | - Elizabeth A Volker
- Department of Chemistry, University of Alberta, Edmonton, AB T6G 2G2, Canada
| | - Ray Ho
- Department of Chemistry, University of Alberta, Edmonton, AB T6G 2G2, Canada
| | - Gisele Monteiro
- Departamento de Tecnologia Bioquímico-Farmacêutica, Faculdade de Ciências Farmacêuticas, Universidade de São Paulo, São Paulo, SP 05508 000, Brazil
| | - Raymond Lai
- Department of Laboratory Medicine and Pathology, University of Alberta, Edmonton, AB T6G 1C9, Canada
| | - Lara K Mahal
- Department of Chemistry, University of Alberta, Edmonton, AB T6G 2G2, Canada
| | - Matthew S Macauley
- Department of Chemistry, University of Alberta, Edmonton, AB T6G 2G2, Canada; Department of Medical Microbiology and Immunology, University of Alberta, Edmonton, AB T6G 2J7, Canada
| | - Ratmir Derda
- Department of Chemistry, University of Alberta, Edmonton, AB T6G 2G2, Canada.
| |
Collapse
|
17
|
Habeeb IF, Alao TE, Delgado D, Buffone A. When a negative (charge) is not a positive: sialylation and its role in cancer mechanics and progression. Front Oncol 2024; 14:1487306. [PMID: 39628991 PMCID: PMC11611868 DOI: 10.3389/fonc.2024.1487306] [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: 08/27/2024] [Accepted: 10/10/2024] [Indexed: 12/06/2024] Open
Abstract
Sialic acids and sialoglycans are critical actors in cancer progression and metastasis. These terminal sugar residues on glycoproteins and glycolipids modulate key cellular processes such as immune evasion, cell adhesion, and migration. Aberrant sialylation is driven by overexpression of sialyltransferases, resulting in hypersialylation on cancer cell surfaces as well as enhancing tumor aggressiveness. Sialylated glycans alter the structure of the glycocalyx, a protective barrier that fosters cancer cell detachment, migration, and invasion. This bulky glycocalyx also increases membrane tension, promoting integrin clustering and downstream signaling pathways that drive cell proliferation and metastasis. They play a critical role in immune evasion by binding to Siglecs, inhibitory receptors on immune cells, which transmit signals that protect cancer cells from immune-mediated destruction. Targeting sialylation pathways presents a promising therapeutic opportunity to understand the complex roles of sialic acids and sialoglycans in cancer mechanics and progression, which is crucial for developing novel diagnostic and therapeutic strategies that can disrupt these processes and improve cancer treatment outcomes.
Collapse
Affiliation(s)
- Issa Funsho Habeeb
- Department of Biomedical Engineering, New Jersey Institute of Technlogy, Newark, NJ, United States
| | - Toheeb Eniola Alao
- Department of Biomedical Engineering, New Jersey Institute of Technlogy, Newark, NJ, United States
| | - Daniella Delgado
- Department of Biomedical Engineering, New Jersey Institute of Technlogy, Newark, NJ, United States
| | - Alexander Buffone
- Department of Biomedical Engineering, New Jersey Institute of Technlogy, Newark, NJ, United States
- Chemical and Materials Engineering, New Jersey Institute of Technlogy, Newark, NJ, United States
| |
Collapse
|
18
|
Roy N, Dasgupta T, Ghosh S, Jayaprakash M, Pal M, Shanavas S, Pal SK, Muthukumar V, Senthil Kumar A, Tamizhselvi R, Roy M, Bose B, Panda D, Chakrabarty R, Paira P. Sialic Acid-Targeted Ru(II)/Ir(III)/Re(I) Complexes for Ferroptosis Induction in Triple-Negative Breast Cancer. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2024. [PMID: 39556719 DOI: 10.1021/acs.langmuir.4c02043] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/20/2024]
Abstract
Ferroptosis has been recognized as an iron-based nonapoptotic-regulated cell death process. In the quest of resisting the unyielding vehemence of triple-negative breast cancer (TNBC), herein we have showcased the ferroptosis-inducing heteroleptic [LIrcRu], [LIrcIrh], and [LIrcRe] complexes, enabling them to selectively target "sialic acid", an overexpressed cancer cell-surface marker. The open-circuit potential (OCP) measurements in live cancer cells revealed the specific interaction between TNBC and the complexes, whereas control experiments with normal cells did not exhibit such interactions. GSH depletion, GPx4 inhibition, NADH/NADPH oxidation, lipid peroxidation, COX-2 activation, and Nrf2 inactivation were meticulously investigated upon treatment with these complexes to establish a strong basis for ferroptosis. Among all complexes, the complex [LIrcIrh] (IC50 = 25 ± 2.17 μM) has been well-documented as a potent ferroptosis inducer, which unveils the sturdy interaction with sialic acid possessing the highest binding constant (Kb = 0.71 × 105 M-1, ΔG = -279345.8026 kcal/mol) along with the highest serum albumin binding affinity (KHSA = 0.67 × 106 M-1) and significant DNA intercalation (Kb = 0.56 × 105 M-1, Kapp = 1.06 × 106 M-1, and C50 of intercalation is 76.56 μM), displaying the decreased current intensity in differential pulse voltammetry (DPV). Moreover, the complex [LIrcIrh] exhibited mitochondrial dysfunction and membrane damage (diminished MMP, ΔΨm) through the production of copious reactive oxygen species (ROS) in MDA-MB-231 cells upon considerable accumulation in mitochondria (Pearson's coefficient = 0.842). The analysis of the field emission scanning electron microscopy (FE-SEM) image has marked the vivid membrane damage induced by the complex [LIrcIrh], exhibiting ablaze evidence for the destruction of TNBC cells through ferroptosis.
Collapse
Affiliation(s)
- Nilmadhab Roy
- Department of Chemistry, School of Advanced Sciences (SAS), Vellore Institute of Technology (VIT), Vellore, Tamilnadu 632014, India
| | - Tiasha Dasgupta
- Department of Bioscience, School of Bio-Sciences and Technology (SBST), Vellore Institute of Technology (VIT), Vellore, Tamilnadu 632014, India
| | - Sreejani Ghosh
- Department of Chemistry, School of Advanced Sciences (SAS), Vellore Institute of Technology (VIT), Vellore, Tamilnadu 632014, India
| | - Meena Jayaprakash
- Department of Chemistry, School of Advanced Sciences (SAS), Vellore Institute of Technology (VIT), Vellore, Tamilnadu 632014, India
- Nano and Bioelectrochemistry Research Laboratory, Department of Chemistry, School of Advanced Sciences (SAS), Vellore Institute of Technology (VIT), Vellore, Tamilnadu 632014, India
| | - Maynak Pal
- Department of Chemistry, National Institute of Technology (NIT) Agartala, Agartala, Tripura 799046, India
- Department of Chemistry, National Institute of Technology (NIT) Manipur, Imphal, Manipur 795004, India
| | - Shanooja Shanavas
- Department of Stem Cells and Regenerative Medicine, Yenepoya Research Centre, Yenepoya (Deemed to be University), Mangalore, Karnataka 575018, India
| | - Surja Kanta Pal
- Department of Chemistry, Nano-Bio Spectroscopy Lab, Sciences and Humanities, Rajiv Gandhi Institute of Petroleum Technology (RGIPT), Jais, Rae Bareli, Uttar Pradesh 229304, India
| | - Venkatesan Muthukumar
- Department of Chemistry, School of Advanced Sciences (SAS), Vellore Institute of Technology (VIT), Vellore, Tamilnadu 632014, India
| | - Annamalai Senthil Kumar
- Department of Chemistry, School of Advanced Sciences (SAS), Vellore Institute of Technology (VIT), Vellore, Tamilnadu 632014, India
- Nano and Bioelectrochemistry Research Laboratory, Department of Chemistry, School of Advanced Sciences (SAS), Vellore Institute of Technology (VIT), Vellore, Tamilnadu 632014, India
| | - Ramasamy Tamizhselvi
- Department of Bioscience, School of Bio-Sciences and Technology (SBST), Vellore Institute of Technology (VIT), Vellore, Tamilnadu 632014, India
| | - Mithun Roy
- Department of Chemistry, National Institute of Technology (NIT) Agartala, Agartala, Tripura 799046, India
- Department of Chemistry, National Institute of Technology (NIT) Manipur, Imphal, Manipur 795004, India
| | - Bipasha Bose
- Department of Stem Cells and Regenerative Medicine, Yenepoya Research Centre, Yenepoya (Deemed to be University), Mangalore, Karnataka 575018, India
| | - Debashis Panda
- Department of Chemistry, Nano-Bio Spectroscopy Lab, Sciences and Humanities, Rajiv Gandhi Institute of Petroleum Technology (RGIPT), Jais, Rae Bareli, Uttar Pradesh 229304, India
| | - Rinku Chakrabarty
- Department of Chemistry, Alipurduar University, Alipurduar, West Bengal 736122, India
| | - Priyankar Paira
- Department of Chemistry, School of Advanced Sciences (SAS), Vellore Institute of Technology (VIT), Vellore, Tamilnadu 632014, India
| |
Collapse
|
19
|
Fan N, Yang K, Bian X, Chen Y, Zhang L, Ai Z, Li X, Ding S, Li S, Cheng W. GlycoSS: A DNA Glycosignal Sieve for Deciphering Spatially Resolved EpCAM-Specific Glycoforms. ACS NANO 2024; 18:29106-29120. [PMID: 39374425 DOI: 10.1021/acsnano.4c10565] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/09/2024]
Abstract
Malignant transformation of cancer is often accompanied by aberrant glycopatterns. Epithelial-mesenchymal transition (EMT) is a crucial biological process in cancer migration and invasion, accelerating cancer deterioration. High-precision analysis of protein-glycan spatial profiling in the EMT process is essential for elucidating glycosylation functions and cancer progression. However, the diversity of glycans in composition and conformation complicates their spatial analysis. Here, we develop a DNA glycosignal sieve (GlycoSS) visualization platform for screening glycoform expression with a protein spatial dimension. GlycoSS utilizes protein-anchored DNA nanoscanners of distinct lengths to control glycosignal readout, enabling protein-glycan distance modulations, and simultaneously orthogonally amplify glycoform output through signal amplification by an exchange reaction. Using GlycoSS, we screened EpCAM-specific hypoglycosylated glycoform signals in different breast cancer cell subtypes, especially characterizing the spatial distribution of glycans on the MCF-7 cell surface. Considering that the EpCAM-specific N-glycan dysregulation in EMT is pivotal, GlycoSS revealed dynamic glycan fluctuations during IGF-1-induced EMT, revealing that the N-glycans were positively associated with tumor malignancy and metastasis. GlycoSS is anticipated to accelerate the identification of aberrant N-glycosylation in tumor progression, advancing systemic glycobiology insights. Notably, GlycoSS is capable of analyzing diverse glycoprotein profiles, offering additional dimensions into the role of glycoprotein nanoenvironments in regulating membrane protein function.
Collapse
Affiliation(s)
- Ningke Fan
- Key Laboratory of Clinical Laboratory Diagnostics (Ministry of Education), College of Laboratory Medicine, Chongqing Medical University, Chongqing 400016, China
| | - Ketong Yang
- Key Laboratory of Clinical Laboratory Diagnostics (Ministry of Education), College of Laboratory Medicine, Chongqing Medical University, Chongqing 400016, China
| | - Xintong Bian
- Department of Laboratory Medicine, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Yirong Chen
- Key Laboratory of Clinical Laboratory Diagnostics (Ministry of Education), College of Laboratory Medicine, Chongqing Medical University, Chongqing 400016, China
| | - Lu Zhang
- Key Laboratory of Clinical Laboratory Diagnostics (Ministry of Education), College of Laboratory Medicine, Chongqing Medical University, Chongqing 400016, China
| | - Zhujun Ai
- Key Laboratory of Clinical Laboratory Diagnostics (Ministry of Education), College of Laboratory Medicine, Chongqing Medical University, Chongqing 400016, China
| | - Xinyu Li
- Key Laboratory of Clinical Laboratory Diagnostics (Ministry of Education), College of Laboratory Medicine, Chongqing Medical University, Chongqing 400016, China
| | - Shijia Ding
- Key Laboratory of Clinical Laboratory Diagnostics (Ministry of Education), College of Laboratory Medicine, Chongqing Medical University, Chongqing 400016, China
| | - Siqiao Li
- School of Basic Medical Sciences, Chongqing Medical University, Chongqing 400016, China
| | - Wei Cheng
- The Center for Clinical Molecular Medical Detection, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| |
Collapse
|
20
|
Yu Y, Shang Y, He Y, Shi S, Wang Q, Ma J, Wang M, Shi W, Chen H. Identification of a potential sialylation-related pattern for the prediction of prognosis and immunotherapy response in small cell lung cancer. Medicine (Baltimore) 2024; 103:e40060. [PMID: 39465788 PMCID: PMC11479454 DOI: 10.1097/md.0000000000040060] [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: 01/23/2024] [Accepted: 09/24/2024] [Indexed: 10/29/2024] Open
Abstract
Our study aimed to establish a novel system for quantifying sialylation patterns and comprehensively analyze their relationship with immune cell infiltration (ICI) characterization, prognosis, and therapeutic sensitivity in small cell lung cancer (SCLC). We conducted a thorough assessment of the sialylation patterns in 100 patients diagnosed with SCLC. Our primary focus was on analyzing the expression levels of 7 prognostic sialylation-related genes. To evaluate and quantify these sialylation patterns, we devised a sialylation score (SS) using principal component analysis algorithms. Prognostic value and therapeutic sensitivities were then evaluated using multiple methods. The GSE176307 was used to verify the predictive ability of SS for immunotherapy. Our study identified 2 distinct clusters based on sialylation patterns. Sialylation cluster B exhibited a lower level of induced ICI therapy and immune-related signaling enrichment, which was associated with a poorer prognosis. Furthermore, there were significant differences in prognosis, response to targeted inhibitors, and immunotherapy between the high and low SS groups. Patients with high SS were characterized by decreased immune cell infiltration, chemokine and immune checkpoint expression, and poorer response to immunotherapy, while the low SS group was more likely to benefit from immunotherapy. This work showed that the evaluation of sialylation subtypes will help to gain insight into the heterogeneity of SCLC. The quantification of sialylation patterns played a non-negligible role in the prediction of ICI characterization, prognosis and individualized therapy strategies.
Collapse
Affiliation(s)
- Yao Yu
- Harbin Medical University, Harbin, Heilongjiang Province, China
- Department of Pulmonary and Critical Care Medicine, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province, China
| | - Yu Shang
- Department of Respiration, The First Hospital of Harbin, Harbin, Heilongjiang Province, China
| | - Yaowu He
- Harbin Medical University, Harbin, Heilongjiang Province, China
- Department of Pulmonary and Critical Care Medicine, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province, China
| | - Si Shi
- Harbin Medical University, Harbin, Heilongjiang Province, China
- Department of Pulmonary and Critical Care Medicine, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province, China
| | - Qi Wang
- Harbin Medical University, Harbin, Heilongjiang Province, China
- Department of Pulmonary and Critical Care Medicine, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province, China
| | - Jing Ma
- Harbin Medical University, Harbin, Heilongjiang Province, China
- Department of Pulmonary and Critical Care Medicine, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province, China
| | - Menghan Wang
- Harbin Medical University, Harbin, Heilongjiang Province, China
- Department of Pulmonary and Critical Care Medicine, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province, China
| | - Wenchao Shi
- Harbin Medical University, Harbin, Heilongjiang Province, China
- Department of Pulmonary and Critical Care Medicine, The Fourth Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province, China
| | - Hong Chen
- Harbin Medical University, Harbin, Heilongjiang Province, China
- Department of Pulmonary and Critical Care Medicine, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province, China
| |
Collapse
|
21
|
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
|
22
|
Zhao J, Zhang K, Sui D, Wang S, Li Y, Tang X, Liu X, Song Y, Deng Y. Recent advances in sialic acid-based active targeting chemoimmunotherapy promoting tumor shedding: a systematic review. NANOSCALE 2024; 16:14621-14639. [PMID: 39023195 DOI: 10.1039/d4nr01740d] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/20/2024]
Abstract
Tumors have always been a major public health concern worldwide, and attempts to look for effective treatments have never ceased. Sialic acid is known to be a crucial element for tumor development and its receptors are highly expressed on tumor-associated immune cells, which perform significant roles in establishing the immunosuppressive tumor microenvironment and further boosting tumorigenesis, progression, and metastasis. Obviously, it is essential to consider sophisticated crosstalk between tumors, the immune system, and preparations, and understand the links between pharmaceutics and immunology. Sialic acid-based chemoimmunotherapy enables active targeting drug delivery via mediating the recognition between the sialic acid-modified nano-drug delivery system represented by liposomes and sialic acid-binding receptors on tumor-associated immune cells, which inhibit their activity and utilize their homing ability to deliver drugs. Such a "Trojan horse" strategy has remarkably improved the shortcomings of traditional passive targeting treatments, unexpectedly promoted tumor shedding, and persistently induced robust immunological memory, thus highlighting its prospective application potential for targeting various tumors. Herein, we review recent advances in sialic acid-based active targeting chemoimmunotherapy to promote tumor shedding, summarize the current viewpoints on the tumor shedding mechanism, especially the formation of durable immunological memory, and analyze the challenges and opportunities of this attractive approach.
Collapse
Affiliation(s)
- Jingyi Zhao
- College of Pharmacy, Shenyang Pharmaceutical University, Wenhua Road, No. 103, Shenyang 110016, China.
| | - Kunfeng Zhang
- College of Pharmacy, Shenyang Pharmaceutical University, Wenhua Road, No. 103, Shenyang 110016, China.
| | - Dezhi Sui
- College of Pharmacy, Shenyang Pharmaceutical University, Wenhua Road, No. 103, Shenyang 110016, China.
| | - Shuo Wang
- College of Pharmacy, Shenyang Pharmaceutical University, Wenhua Road, No. 103, Shenyang 110016, China.
| | - Yantong Li
- College of Pharmacy, Shenyang Pharmaceutical University, Wenhua Road, No. 103, Shenyang 110016, China.
| | - Xueying Tang
- College of Pharmacy, Shenyang Pharmaceutical University, Wenhua Road, No. 103, Shenyang 110016, China.
| | - Xinrong Liu
- College of Pharmacy, Shenyang Pharmaceutical University, Wenhua Road, No. 103, Shenyang 110016, China.
| | - Yanzhi Song
- College of Pharmacy, Shenyang Pharmaceutical University, Wenhua Road, No. 103, Shenyang 110016, China.
| | - Yihui Deng
- College of Pharmacy, Shenyang Pharmaceutical University, Wenhua Road, No. 103, Shenyang 110016, China.
| |
Collapse
|
23
|
Li X, Liu D, Wu Z, Xu Y. Diffuse tumors: Molecular determinants shared by different cancer types. Comput Biol Med 2024; 178:108703. [PMID: 38850961 DOI: 10.1016/j.compbiomed.2024.108703] [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: 12/05/2023] [Revised: 05/02/2024] [Accepted: 06/01/2024] [Indexed: 06/10/2024]
Abstract
Most cancer types have both diffuse and non-diffuse subtypes, which have rather distinct morphologies, namely scattered tiny tumors vs. one solid tumor, and different levels of aggressiveness. However, the causes for forming such distinct subtypes remain largely unknown. Using the diffuse and non-diffuse gastric cancers (GCs) as the illustrative example, we present a computational study based on the transcriptomic data from the TCGA and GEO databases, to address the following questions: (i) What are the key molecular determinants that give rise to the distinct morphologies between diffuse and non-diffuse cancers? (ii) What are the main reasons for diffuse cancers to be generally more aggressive than non-diffuse ones of the same cancer type? (iii) What are the reasons for their distinct immunoactivities? And (iv) why do diffuse cancers on average tend to take place in younger patients? The study is conducted using the framework we have previously developed for elucidation of general drivers cancer formation and development. Our main discoveries are: (a) the level of (poly-) sialic acids deployed on the surface of cancer cells is a significant factor contributing to questions (i) and (ii); (b) poly-sialic acids synthesized by ST8SIA4 are the key to question (iii); and (c) the circulating growth factors specifically needed by the diffuse subtype dictate the answer to question (iv). All these predictions are substantiated by published experimental studies. Our further analyses on breast, prostate, lung, liver, and thyroid cancers reveal that these discoveries generally apply to the diffuse subtypes of these cancer types, hence indicating the generality of our discoveries.
Collapse
Affiliation(s)
- Xuan Li
- Key Laboratory of Symbolic Computation and Knowledge Engineering of Ministry of Education, College of Computer Science and Technology, Jilin University, Changchun, 130012, China; School of Medicine, Southern University of Science and Technology, Shenzhen, China
| | - Dingyun Liu
- Key Laboratory of Symbolic Computation and Knowledge Engineering of Ministry of Education, College of Computer Science and Technology, Jilin University, Changchun, 130012, China
| | - Zhipeng Wu
- Key Laboratory of Symbolic Computation and Knowledge Engineering of Ministry of Education, College of Computer Science and Technology, Jilin University, Changchun, 130012, China
| | - Ying Xu
- School of Medicine, Southern University of Science and Technology, Shenzhen, China.
| |
Collapse
|
24
|
Godbole S, Voß H, Gocke A, Schlumbohm S, Schumann Y, Peng B, Mynarek M, Rutkowski S, Dottermusch M, Dorostkar MM, Korshunov A, Mair T, Pfister SM, Kwiatkowski M, Hotze M, Neumann P, Hartmann C, Weis J, Liesche-Starnecker F, Guan Y, Moritz M, Siebels B, Struve N, Schlüter H, Schüller U, Krisp C, Neumann JE. Multiomic profiling of medulloblastoma reveals subtype-specific targetable alterations at the proteome and N-glycan level. Nat Commun 2024; 15:6237. [PMID: 39043693 PMCID: PMC11266559 DOI: 10.1038/s41467-024-50554-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2023] [Accepted: 07/11/2024] [Indexed: 07/25/2024] Open
Abstract
Medulloblastomas (MBs) are malignant pediatric brain tumors that are molecularly and clinically heterogenous. The application of omics technologies-mainly studying nucleic acids-has significantly improved MB classification and stratification, but treatment options are still unsatisfactory. The proteome and their N-glycans hold the potential to discover clinically relevant phenotypes and targetable pathways. We compile a harmonized proteome dataset of 167 MBs and integrate findings with DNA methylome, transcriptome and N-glycome data. We show six proteome MB subtypes, that can be assigned to two main molecular programs: transcription/translation (pSHHt, pWNT and pG3myc), and synapses/immunological processes (pSHHs, pG3 and pG4). Multiomic analysis reveals different conservation levels of proteome features across MB subtypes at the DNA methylome level. Aggressive pGroup3myc MBs and favorable pWNT MBs are most similar in cluster hierarchies concerning overall proteome patterns but show different protein abundances of the vincristine resistance-associated multiprotein complex TriC/CCT and of N-glycan turnover-associated factors. The N-glycome reflects proteome subtypes and complex-bisecting N-glycans characterize pGroup3myc tumors. Our results shed light on targetable alterations in MB and set a foundation for potential immunotherapies targeting glycan structures.
Collapse
Affiliation(s)
- Shweta Godbole
- Center for Molecular Neurobiology (ZMNH), University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Institute of Neuropathology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Hannah Voß
- Section of Mass Spectrometry and Proteomics, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Antonia Gocke
- Center for Molecular Neurobiology (ZMNH), University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Section of Mass Spectrometry and Proteomics, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Simon Schlumbohm
- Chair for High Performance Computing, Helmut Schmidt University, Hamburg, Germany
| | - Yannis Schumann
- Chair for High Performance Computing, Helmut Schmidt University, Hamburg, Germany
| | - Bojia Peng
- Section of Mass Spectrometry and Proteomics, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Martin Mynarek
- Department of Pediatric Hematology and Oncology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Mildred Scheel Cancer Career Center HaTriCS4, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Stefan Rutkowski
- Department of Pediatric Hematology and Oncology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Matthias Dottermusch
- Center for Molecular Neurobiology (ZMNH), University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Institute of Neuropathology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Mario M Dorostkar
- Center for Neuropathology, Ludwig-Maximilians-University, Munich, Germany
- German Center for Neurodegenerative Diseases, Munich, Germany
| | - Andrey Korshunov
- Department of Neuropathology, University Hospital Heidelberg, Heidelberg, Germany
- Clinical Cooperation Unit Neuropathology, German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Thomas Mair
- Section of Mass Spectrometry and Proteomics, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Stefan M Pfister
- Hopp Children's Cancer Center at the NCT Heidelberg (KiTZ), Heidelberg, Germany
- Division of Pediatric Neurooncology, German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), Heidelberg, Germany
- Department of Pediatric Hematology and Oncology, Heidelberg University Hospital, Heidelberg, Germany
| | | | - Madlen Hotze
- Institute of Biochemistry, University of Innsbruck, Innsbruck, Austria
| | - Philipp Neumann
- Chair for High Performance Computing, Helmut Schmidt University, Hamburg, Germany
| | - Christian Hartmann
- Department of Neuropathology, Hannover Medical School (MHH), Hannover, Germany
| | - Joachim Weis
- Institute of Neuropathology, RWTH Aachen University Hospital, Aachen, Germany
| | | | - Yudong Guan
- Section of Mass Spectrometry and Proteomics, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Manuela Moritz
- Section of Mass Spectrometry and Proteomics, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Bente Siebels
- Section of Mass Spectrometry and Proteomics, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Nina Struve
- Mildred Scheel Cancer Career Center HaTriCS4, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Department of Radiotherapy & Radiation Oncology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Hartmut Schlüter
- Section of Mass Spectrometry and Proteomics, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Ulrich Schüller
- Institute of Neuropathology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Department of Pediatric Hematology and Oncology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Research Institute Children's Cancer Center Hamburg, Hamburg, Germany
| | - Christoph Krisp
- Section of Mass Spectrometry and Proteomics, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Julia E Neumann
- Center for Molecular Neurobiology (ZMNH), University Medical Center Hamburg-Eppendorf, Hamburg, Germany.
- Institute of Neuropathology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.
| |
Collapse
|
25
|
Hodgson K, Orozco-Moreno M, Goode EA, Fisher M, Garnham R, Beatson R, Turner H, Livermore K, Zhou Y, Wilson L, Visser EA, Pijnenborg JF, Eerden N, Moons SJ, Rossing E, Hysenaj G, Krishna R, Peng Z, Nangkana KP, Schmidt EN, Duxfield A, Dennis EP, Heer R, Lawson MA, Macauley M, Elliott DJ, Büll C, Scott E, Boltje TJ, Drake RR, Wang N, Munkley J. Sialic acid blockade inhibits the metastatic spread of prostate cancer to bone. EBioMedicine 2024; 104:105163. [PMID: 38772281 PMCID: PMC11134892 DOI: 10.1016/j.ebiom.2024.105163] [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: 07/07/2023] [Revised: 05/02/2024] [Accepted: 05/06/2024] [Indexed: 05/23/2024] Open
Abstract
BACKGROUND Bone metastasis is a common consequence of advanced prostate cancer. Bisphosphonates can be used to manage symptoms, but there are currently no curative treatments available. Altered tumour cell glycosylation is a hallmark of cancer and is an important driver of a malignant phenotype. In prostate cancer, the sialyltransferase ST6GAL1 is upregulated, and studies show ST6GAL1-mediated aberrant sialylation of N-glycans promotes prostate tumour growth and disease progression. METHODS Here, we monitor ST6GAL1 in tumour and serum samples from men with aggressive prostate cancer and using in vitro and in vivo models we investigate the role of ST6GAL1 in prostate cancer bone metastasis. FINDINGS ST6GAL1 is upregulated in patients with prostate cancer with tumours that have spread to the bone and can promote prostate cancer bone metastasis in vivo. The mechanisms involved are multi-faceted and involve modification of the pre-metastatic niche towards bone resorption to promote the vicious cycle, promoting the development of M2 like macrophages, and the regulation of immunosuppressive sialoglycans. Furthermore, using syngeneic mouse models, we show that inhibiting sialylation can block the spread of prostate tumours to bone. INTERPRETATION Our study identifies an important role for ST6GAL1 and α2-6 sialylated N-glycans in prostate cancer bone metastasis, provides proof-of-concept data to show that inhibiting sialylation can suppress the spread of prostate tumours to bone, and highlights sialic acid blockade as an exciting new strategy to develop new therapies for patients with advanced prostate cancer. FUNDING Prostate Cancer Research and the Mark Foundation For Cancer Research, the Medical Research Council and Prostate Cancer UK.
Collapse
Affiliation(s)
- Kirsty Hodgson
- Newcastle University Centre for Cancer, Newcastle University Institute of Biosciences, Newcastle upon Tyne NE1 3BZ, UK
| | - Margarita Orozco-Moreno
- Newcastle University Centre for Cancer, Newcastle University Institute of Biosciences, Newcastle upon Tyne NE1 3BZ, UK
| | - Emily Archer Goode
- Newcastle University Centre for Cancer, Newcastle University Institute of Biosciences, Newcastle upon Tyne NE1 3BZ, UK
| | - Matthew Fisher
- The Mellanby Centre for Musculoskeletal Research, Division of Clinical Medicine, The University of Sheffield, Sheffield, UK
| | - Rebecca Garnham
- Newcastle University Centre for Cancer, Newcastle University Institute of Biosciences, Newcastle upon Tyne NE1 3BZ, UK
| | - Richard Beatson
- Centre for Inflammation and Tissue Repair, UCL Respiratory, Division of Medicine, University College London (UCL), Rayne 9 Building, London WC1E 6JF, UK
| | - Helen Turner
- Cellular Pathology, The Royal Victoria Infirmary, Queen Victoria Road, Newcastle upon Tyne NE1 4LP, UK
| | - Karen Livermore
- Newcastle University Centre for Cancer, Newcastle University Institute of Biosciences, Newcastle upon Tyne NE1 3BZ, UK
| | - Yuhan Zhou
- The Mellanby Centre for Musculoskeletal Research, Division of Clinical Medicine, The University of Sheffield, Sheffield, UK
| | - Laura Wilson
- Newcastle University Centre for Cancer, Translational and Clinical Research Institute, Newcastle University, Paul O'Gorman Building, Newcastle upon Tyne NE2 4HH, UK
| | - Eline A Visser
- Synthetic Organic Chemistry, Institute for Molecules and Materials, Radboud University, Nijmegen, the Netherlands
| | | | - Nienke Eerden
- Synthetic Organic Chemistry, Institute for Molecules and Materials, Radboud University, Nijmegen, the Netherlands; GlycoTherapeutics B.V., Nijmegen, the Netherlands
| | | | - Emiel Rossing
- Synthetic Organic Chemistry, Institute for Molecules and Materials, Radboud University, Nijmegen, the Netherlands
| | - Gerald Hysenaj
- Newcastle University Centre for Cancer, Newcastle University Institute of Biosciences, Newcastle upon Tyne NE1 3BZ, UK
| | - Rashi Krishna
- Newcastle University Centre for Cancer, Newcastle University Institute of Biosciences, Newcastle upon Tyne NE1 3BZ, UK
| | - Ziqian Peng
- Newcastle University Centre for Cancer, Newcastle University Institute of Biosciences, Newcastle upon Tyne NE1 3BZ, UK
| | - Kyla Putri Nangkana
- Newcastle University Centre for Cancer, Newcastle University Institute of Biosciences, Newcastle upon Tyne NE1 3BZ, UK
| | - Edward N Schmidt
- Department of Chemistry, University of Alberta, Edmonton, AB T6G 2G2, Canada; Department of Medical Microbiology and Immunology, University of Alberta, Edmonton, AB T6G 2E1, Canada
| | - Adam Duxfield
- Newcastle University Centre for Cancer, Newcastle University Institute of Biosciences, Newcastle upon Tyne NE1 3BZ, UK; International Centre for Life, Biosciences Institute, Newcastle University, Newcastle Upon Tyne NE1 3BZ, UK
| | - Ella P Dennis
- International Centre for Life, Biosciences Institute, Newcastle University, Newcastle Upon Tyne NE1 3BZ, UK
| | - Rakesh Heer
- Newcastle University Centre for Cancer, Translational and Clinical Research Institute, Newcastle University, Paul O'Gorman Building, Newcastle upon Tyne NE2 4HH, UK; Department of Urology, Freeman Hospital, The Newcastle upon Tyne Hospitals NHS Foundation Trust, Newcastle upon Tyne NE7 7DN, UK
| | - Michelle A Lawson
- The Mellanby Centre for Musculoskeletal Research, Division of Clinical Medicine, The University of Sheffield, Sheffield, UK
| | - Matthew Macauley
- Department of Chemistry, University of Alberta, Edmonton, AB T6G 2G2, Canada; Department of Medical Microbiology and Immunology, University of Alberta, Edmonton, AB T6G 2E1, Canada
| | - David J Elliott
- Newcastle University Centre for Cancer, Newcastle University Institute of Biosciences, Newcastle upon Tyne NE1 3BZ, UK
| | - Christian Büll
- Biomolecular Chemistry, Institute for Molecules and Materials, Radboud University Nijmegen, the Netherlands
| | - Emma Scott
- Newcastle University Centre for Cancer, Newcastle University Institute of Biosciences, Newcastle upon Tyne NE1 3BZ, UK
| | - Thomas J Boltje
- Synthetic Organic Chemistry, Institute for Molecules and Materials, Radboud University, Nijmegen, the Netherlands
| | - Richard R Drake
- Department of Cell and Molecular Pharmacology, Medical University of South Carolina, Charleston, SC, USA
| | - Ning Wang
- The Mellanby Centre for Musculoskeletal Research, Division of Clinical Medicine, The University of Sheffield, Sheffield, UK; Leicester Cancer Research Centre, Department of Genetics and Genome Biology, University of Leicester, LE2 7LX, UK.
| | - Jennifer Munkley
- Newcastle University Centre for Cancer, Newcastle University Institute of Biosciences, Newcastle upon Tyne NE1 3BZ, UK.
| |
Collapse
|
26
|
Aguilar Díaz de león JS, Thirumurty M, Ly N. Surface plasmon resonance microscopy identifies glycan heterogeneity in pancreatic cancer cells that influences mucin-4 binding interactions. PLoS One 2024; 19:e0304154. [PMID: 38776309 PMCID: PMC11111020 DOI: 10.1371/journal.pone.0304154] [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: 12/21/2023] [Accepted: 05/07/2024] [Indexed: 05/24/2024] Open
Abstract
Membrane proteins are the main targets of therapeutic drugs and most of them are glycosylated. Glycans play pivotal roles in several biological processes, and glycosylation changes are a well-established hallmark of several types of cancer, including pancreatic cancer, that contribute to tumor growth. Mucin-4 (MUC-4) is a membrane glycoprotein which is associated with pancreatic cancer and metastasis, and it has been targeted as a promising vaccine candidate. In this study, Surface Plasmon Resonance Microscopy (SPRM) was implemented to study complex influences of the native N-glycan cellular environment on binding interactions to the MUC-4 receptor as this is currently the only commercially available label-free technique with high enough sensitivity and resolution to measure binding kinetics and heterogeneity on single cells. Such unique capability enables for a more accurate understanding of the "true" binding interactions on human cancer cells without disrupting the native environment of the target MUC-4 receptor. Removal of N-linked glycans in pancreatic cancer cells using PNGase F exposed heterogeneity in Concanavalin (Con A) binding by revealing three new binding populations with higher affinities than the glycosylated control cells. Anti-MUC-4 binding interactions of enzymatically N-linked deglycosylated pancreatic cancer cells produced a 25x faster association and 37x higher affinity relative to the glycosylated control cells. Lastly, four interaction modes were observed for Helix Pomatia Agglutinin (HPA) binding to the glycosylated control cells, but shifted and increased in activity upon removal of N-linked glycans. These results identified predominant interaction modes of glycan and MUC-4 in pancreatic cancer cells, the kinetics of their binding interactions were quantified, and the influence of N-linked glycans in MUC-4 binding interactions was revealed.
Collapse
Affiliation(s)
| | - Miyuki Thirumurty
- Biosensing Instrument Inc., Tempe, Arizona, United States of America
| | - Nguyen Ly
- Biosensing Instrument Inc., Tempe, Arizona, United States of America
| |
Collapse
|
27
|
Yang Z, Hou Y, Grande G, Cho JH, Wang C, Shi Y, Zak J, Wan Y, Qin K, Liu D, Teijaro JR, Lerner RA, Wu P. Targeted desialylation and cytolysis of tumour cells by fusing a sialidase to a bispecific T-cell engager. Nat Biomed Eng 2024; 8:499-512. [PMID: 38693431 PMCID: PMC11577304 DOI: 10.1038/s41551-024-01202-w] [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: 04/21/2022] [Accepted: 03/26/2024] [Indexed: 05/03/2024]
Abstract
Bispecific T-cell engagers (BiTEs) bring together tumour cells and cytotoxic T cells by binding to specific cell-surface tumour antigens and T-cell receptors, and have been clinically successful for the treatment of B-cell malignancies. Here we show that a BiTE-sialidase fusion protein enhances the susceptibility of solid tumours to BiTE-mediated cytolysis of tumour cells via targeted desialylation-that is, the removal of terminal sialic acid residues on glycans-at the BiTE-induced T-cell-tumour-cell interface. In xenograft and syngeneic mouse models of leukaemia and of melanoma and breast cancer, and compared with the parental BiTE molecules, targeted desialylation via the BiTE-sialidase fusion proteins enhanced the formation of immunological synapses, T-cell activation and T-cell-mediated tumour-cell cytolysis in the presence of the target antigen. The targeted desialylation of tumour cells may enhance the potency of therapies relying on T-cell engagers.
Collapse
Affiliation(s)
- Zhuo Yang
- Department of Molecular and Cellular Biology, The Scripps Research Institute, La Jolla, CA, USA
- Department of Chemistry, The Scripps Research Institute, La Jolla, CA, USA
| | - Yingqin Hou
- Department of Molecular and Cellular Biology, The Scripps Research Institute, La Jolla, CA, USA
| | - Geramie Grande
- Department of Chemistry, The Scripps Research Institute, La Jolla, CA, USA
| | - Jong Hyun Cho
- Department of Pathology, Immunology and Laboratory Medicine, Rutgers University-New Jersey Medical School, Newark, NJ, USA
| | - Chao Wang
- Department of Molecular and Cellular Biology, The Scripps Research Institute, La Jolla, CA, USA
| | - Yujie Shi
- Department of Molecular and Cellular Biology, The Scripps Research Institute, La Jolla, CA, USA
| | - Jaroslav Zak
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, CA, USA
| | - Yue Wan
- Department of Chemistry, The Scripps Research Institute, La Jolla, CA, USA
| | - Ke Qin
- Department of Molecular and Cellular Biology, The Scripps Research Institute, La Jolla, CA, USA
| | - Dongfang Liu
- Department of Pathology, Immunology and Laboratory Medicine, Rutgers University-New Jersey Medical School, Newark, NJ, USA
| | - John R Teijaro
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, CA, USA
| | - Richard A Lerner
- Department of Chemistry, The Scripps Research Institute, La Jolla, CA, USA
| | - Peng Wu
- Department of Molecular and Cellular Biology, The Scripps Research Institute, La Jolla, CA, USA.
| |
Collapse
|
28
|
Cieniewicz B, Oliveira E, Saxton M, Torabi D, Bhatta A, Kukutla P, Arballo A, Yang Z, Yu B, Fate M, Ning H, Corey L, Maiti A, Corey D. Therapeutic Targeting of TIM-4-L with Engineered T Cells for Acute Myeloid Leukemia. Clin Cancer Res 2024; 30:1878-1888. [PMID: 38451195 DOI: 10.1158/1078-0432.ccr-23-3044] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2023] [Revised: 01/16/2024] [Accepted: 03/04/2024] [Indexed: 03/08/2024]
Abstract
PURPOSE Disruption of lipid bilayer asymmetry is a common feature observed in cancer cells and offers novel routes for therapeutic targeting. We used the natural immune receptor TIM-4 to interrogate for loss of plasma membrane phospholipid polarity in primary acute myelogenous leukemia (AML) samples and evaluated the anti-leukemic activity of TIM-4-L-directed T-cell therapy in preclinical AML models. EXPERIMENTAL DESIGN We performed FACS analysis on 33 primary AML bone marrow specimens and correlated TIM-4-L expression frequency and intensity with molecular disease characteristics. Using Kasumi-1 and MV-4-11 AML cell lines, we further tested the anti-leukemic effects of TIM-4-L-directed engineered T cells in vitro and in vivo. RESULTS We found that 86% of untreated AML blasts displayed upregulation of cell surface TIM-4-L. These observations were agnostic to AML genetic classification, as samples with mutations in TP53, ASXL1, and RUNX1 displayed TIM-4-L upregulation similar to that seen in favorable and intermediate subtypes. TIM-4-L dysregulation was also stably present in AML cell lines. To evaluate the potential of targeting upregulated TIM-4-L with adoptive T-cell therapy, we constructed TIM-4-L-directed engineered T cells, which demonstrated potent anti-leukemic effects, effectively eliminating AML cell lines with a range of endogenous TIM-4-L expression levels both in vitro and in vivo. CONCLUSIONS These results highlight TIM-4-L as a highly prevalent target on AML across a range of genetic classifications and novel target for T-cell-based therapy in AML. Further investigations into the role of TIM-4-L in AML pathogenesis and its potential as an anti-leukemic target for clinical development are warranted.
Collapse
MESH Headings
- Humans
- Leukemia, Myeloid, Acute/genetics
- Leukemia, Myeloid, Acute/therapy
- Leukemia, Myeloid, Acute/pathology
- Leukemia, Myeloid, Acute/immunology
- Leukemia, Myeloid, Acute/metabolism
- Mice
- Animals
- T-Lymphocytes/immunology
- T-Lymphocytes/metabolism
- Membrane Proteins/genetics
- Membrane Proteins/metabolism
- Cell Line, Tumor
- Xenograft Model Antitumor Assays
- Female
- Male
- Middle Aged
- Adult
- Aged
- Immunotherapy, Adoptive/methods
Collapse
Affiliation(s)
| | | | - Mike Saxton
- CERo Therapeutics Inc., South San Francisco, California
| | - Damoun Torabi
- CERo Therapeutics Inc., South San Francisco, California
| | - Ankit Bhatta
- CERo Therapeutics Inc., South San Francisco, California
| | | | | | - Zhuo Yang
- CERo Therapeutics Inc., South San Francisco, California
| | - Bi Yu
- CERo Therapeutics Inc., South San Francisco, California
| | - Maria Fate
- CERo Therapeutics Inc., South San Francisco, California
| | - Hongxiu Ning
- CERo Therapeutics Inc., South San Francisco, California
| | - Lawrence Corey
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, Washington
| | - Abhishek Maiti
- Department of Leukemia, University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Daniel Corey
- CERo Therapeutics Inc., South San Francisco, California
| |
Collapse
|
29
|
Han R, Lin C, Lu C, Wang Y, Kang J, Hu C, Dou Y, Wu D, He T, Tang H, Zheng J, Li L, He Y. Sialyltransferase ST3GAL4 confers osimertinib resistance and offers strategies to overcome resistance in non-small cell lung cancer. Cancer Lett 2024; 588:216762. [PMID: 38408602 DOI: 10.1016/j.canlet.2024.216762] [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: 09/03/2023] [Revised: 02/17/2024] [Accepted: 02/22/2024] [Indexed: 02/28/2024]
Abstract
The third-generation EGFR-TKI osimertinib is widely used in EGFR-mutated positive non-small cell lung cancer (NSCLC) patients, but drug resistance is inevitable. The currently known mechanisms only explain resistance in a small proportion of patients. For most patients, the mechanism of osimertinib resistance is still unclear, especially for EGFR-independent resistance. Herein, we thoroughly investigated the novel mechanism of osimertinib resistance and treatment strategies. We identified that ST3GAL4, a sialyltransferase, catalyzes terminal glycan sialylation of receptor protein tyrosine kinases, which induces acquired resistance to osimertinib in vitro and in vivo. In addition, ST3GAL4 is generally overexpressed in osimertinib-resistant patients with unknown resistance mechanisms. ST3GAL4 modifies MET glycosylation on N785 with sialylation, which antagonizes K48-related ubiquitin-dependent MET degradation and subsequently activates MET and its downstream proliferation signaling pathways. Meanwhile, ST3GAL4 knockdown or inhibition by brigatinib resensitizes resistant non-small cell lung cancer cells to osimertinib in vitro and in vivo This study suggests that ST3GAL4 can induce acquired resistance to osimertinib, which may be an important EGFR-independent resistance mechanism Furthermore, targeting ST3GAL4 with brigatinib provides new strategies to overcome osimertinib resistance.
Collapse
Affiliation(s)
- Rui Han
- Department of Respiratory Disease, Daping Hospital, Army Medical University, Chongqing, China
| | - Caiyu Lin
- Department of Respiratory Disease, Daping Hospital, Army Medical University, Chongqing, China
| | - Conghua Lu
- Department of Respiratory Disease, Daping Hospital, Army Medical University, Chongqing, China
| | - Yubo Wang
- Department of Respiratory Disease, Chongqing University Jiangjin Hospital, China
| | - Jun Kang
- Department of Respiratory Disease, Daping Hospital, Army Medical University, Chongqing, China
| | - Chen Hu
- Department of Respiratory Disease, Daping Hospital, Army Medical University, Chongqing, China
| | - Yuanyao Dou
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, China
| | - Di Wu
- Department of Respiratory Disease, Daping Hospital, Army Medical University, Chongqing, China
| | - TingTing He
- Department of Respiratory Disease, Daping Hospital, Army Medical University, Chongqing, China
| | - Huan Tang
- Department of Respiratory Disease, Daping Hospital, Army Medical University, Chongqing, China
| | - Jie Zheng
- Department of Respiratory Disease, Daping Hospital, Army Medical University, Chongqing, China
| | - Li Li
- Department of Respiratory Disease, Daping Hospital, Army Medical University, Chongqing, China
| | - Yong He
- Department of Respiratory Disease, Daping Hospital, Army Medical University, Chongqing, China.
| |
Collapse
|
30
|
Chang J, Feng Q, Mao Y, Zhang Z, Xu Y, Chen Y, Zheng P, Lin S, Shen F, Zhang Z, Zhang Z, He G, Xu J, Wei Y. Siglec9 + tumor-associated macrophages predict prognosis and therapeutic vulnerability in patients with colon cancer. Int Immunopharmacol 2024; 130:111771. [PMID: 38430807 DOI: 10.1016/j.intimp.2024.111771] [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: 12/22/2023] [Revised: 02/09/2024] [Accepted: 02/26/2024] [Indexed: 03/05/2024]
Abstract
BACKGROUND Siglec9 has been identified as an immune checkpoint molecule on tumor-associated macrophages (TAMs). Nevertheless, the expression profile and clinical significance of Siglec9 + TAMs in colon cancer (CC) are still not fully understood. METHODS Two clinical cohorts from distinct medical centers were retrospectively enrolled. Immunohistochemistry and immunofluorescence were conducted to evaluate the infiltration of immune cells. Single-cell RNA sequencing and flow cytometry were utilized to identify the impact of Siglec9 + TAMs on the tumor immune environment, which was subsequently validated through bioinformatics analysis of the TCGA database. Prognosis and the benefit of adjuvant chemotherapy (ACT) were also evaluated using Cox regression analysis and the Kaplan-Meier method. RESULTS High infiltration of Siglec9 + TAMs was associated with worse prognosis and better benefit from 6-month ACT. Siglec9 + TAMs contributed to immunoevasion by promoting the infiltration of immunosuppressive cells and the dysfunction process of CD8 + T cells. Additionally, high infiltration of Siglec9 + TAMs was associated with the mesenchymal-featured subtype and overexpression of the VEGF signaling pathway, which was validated by the strongest communication between Siglec9 + TAMs and vascular endothelial cells. CONCLUSIONS Siglec9 + TAMs may serve as a biomarker for prognosis and response to ACT in CC. Furthermore, the immunoevasive contexture and angiogenesis stimulated by Siglec9 + TAMs suggest potential treatment combinations for CC patients.
Collapse
Affiliation(s)
- Jiang Chang
- Colorectal Cancer Center, Zhongshan Hospital, Fudan University, Shanghai, China; Department of General Surgery, Zhongshan Hospital, Fudan University, Shanghai, China; Department of General Surgery, Huadong Hospital, Fudan University, Shanghai, China
| | - Qingyang Feng
- Colorectal Cancer Center, Zhongshan Hospital, Fudan University, Shanghai, China; Department of General Surgery, Zhongshan Hospital, Fudan University, Shanghai, China; Shanghai Engineering Research Center of Colorectal Cancer Minimally Invasive, Shanghai, China
| | - Yihao Mao
- Colorectal Cancer Center, Zhongshan Hospital, Fudan University, Shanghai, China; Department of General Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Zhiyuan Zhang
- Colorectal Cancer Center, Zhongshan Hospital, Fudan University, Shanghai, China; Department of General Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Yuqiu Xu
- Colorectal Cancer Center, Zhongshan Hospital, Fudan University, Shanghai, China; Department of General Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Yijiao Chen
- Colorectal Cancer Center, Zhongshan Hospital, Fudan University, Shanghai, China; Department of General Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Peng Zheng
- Colorectal Cancer Center, Zhongshan Hospital, Fudan University, Shanghai, China; Department of General Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Songbin Lin
- General Surgery Department, Zhongshan Hospital, Fudan University (Xiamen Branch), Xiamen, Fujian Province, China
| | - Feifan Shen
- Colorectal Cancer Center, Zhongshan Hospital, Fudan University, Shanghai, China; Department of General Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Zhuojian Zhang
- Colorectal Cancer Center, Zhongshan Hospital, Fudan University, Shanghai, China; Department of General Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Ziqi Zhang
- Colorectal Cancer Center, Zhongshan Hospital, Fudan University, Shanghai, China; Department of General Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Guodong He
- Colorectal Cancer Center, Zhongshan Hospital, Fudan University, Shanghai, China; Department of General Surgery, Zhongshan Hospital, Fudan University, Shanghai, China; Shanghai Engineering Research Center of Colorectal Cancer Minimally Invasive, Shanghai, China.
| | - Jianmin Xu
- Colorectal Cancer Center, Zhongshan Hospital, Fudan University, Shanghai, China; Department of General Surgery, Zhongshan Hospital, Fudan University, Shanghai, China; Shanghai Engineering Research Center of Colorectal Cancer Minimally Invasive, Shanghai, China.
| | - Ye Wei
- Colorectal Cancer Center, Zhongshan Hospital, Fudan University, Shanghai, China; Department of General Surgery, Zhongshan Hospital, Fudan University, Shanghai, China; Shanghai Engineering Research Center of Colorectal Cancer Minimally Invasive, Shanghai, China; Department of General Surgery, Huadong Hospital, Fudan University, Shanghai, China.
| |
Collapse
|
31
|
Shi Y, Bashian EE, Hou Y, Wu P. Chemical immunology: Recent advances in tool development and applications. Cell Chem Biol 2024; 31:S2451-9456(24)00080-1. [PMID: 38508196 PMCID: PMC11393185 DOI: 10.1016/j.chembiol.2024.02.006] [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: 09/01/2023] [Revised: 02/01/2024] [Accepted: 02/22/2024] [Indexed: 03/22/2024]
Abstract
Immunology was one of the first biological fields to embrace chemical approaches. The development of new chemical approaches and techniques has provided immunologists with an impressive arsenal of tools to address challenges once considered insurmountable. This review focuses on advances at the interface of chemistry and immunobiology over the past two decades that have not only opened new avenues in basic immunological research, but also revolutionized drug development for the treatment of cancer and autoimmune diseases. These include chemical approaches to understand and manipulate antigen presentation and the T cell priming process, to facilitate immune cell trafficking and regulate immune cell functions, and therapeutic applications of chemical approaches to disease control and treatment.
Collapse
Affiliation(s)
- Yujie Shi
- Department of Molecular and Cellular Biology, The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, CA 92037, USA
| | - Eleanor E Bashian
- Department of Molecular and Cellular Biology, The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, CA 92037, USA
| | - Yingqin Hou
- Department of Molecular and Cellular Biology, The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, CA 92037, USA
| | - Peng Wu
- Department of Molecular and Cellular Biology, The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, CA 92037, USA.
| |
Collapse
|
32
|
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
|
33
|
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
|
34
|
McCord K, Wang C, Anhalt M, Poon WW, Gavin AL, Wu P, Macauley MS. Dissecting the Ability of Siglecs To Antagonize Fcγ Receptors. ACS CENTRAL SCIENCE 2024; 10:315-330. [PMID: 38435516 PMCID: PMC10906256 DOI: 10.1021/acscentsci.3c00969] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/01/2023] [Revised: 12/19/2023] [Accepted: 12/21/2023] [Indexed: 03/05/2024]
Abstract
Fcγ receptors (FcγRs) play key roles in the effector function of IgG, but their inappropriate activation plays a role in several disease etiologies. Therefore, it is critical to better understand how FcγRs are regulated. Numerous studies suggest that sialic acid-binding immunoglobulin-type lectins (Siglecs), a family of immunomodulatory receptors, modulate FcγR activity; however, it is unclear of the circumstances in which Siglecs can antagonize FcγRs and which Siglecs have this ability. Using liposomes displaying selective ligands to coengage FcγRs with a specific Siglec, we explore the ability of Siglec-3, Siglec-5, Siglec-7, and Siglec-9 to antagonize signaling downstream of FcγRs. We demonstrate that Siglec-3 and Siglec-9 can fully inhibit FcγR activation in U937 cells when coengaged with FcγRs. Cells expressing Siglec mutants reveal differential roles for the immunomodulatory tyrosine-based inhibitory motif (ITIM) and immunomodulatory tyrosine-based switch motif (ITSM) in this inhibition. Imaging flow cytometry enabled visualization of SHP-1 recruitment to Siglec-3 in an ITIM-dependent manner, while SHP-2 recruitment is more ITSM-dependent. Conversely, both cytosolic motifs of Siglec-9 contribute to SHP-1/2 recruitment. Siglec-7 poorly antagonizes FcγR activation for two reasons: masking by cis ligands and differences in its ITIM and ITSM. A chimera of the Siglec-3 extracellular domains and Siglec-5 cytosolic tail strongly inhibits FcγR when coengaged, providing evidence that Siglec-5 is more like Siglec-3 and Siglec-9 in its ability to antagonize FcγRs. Additionally, Siglec-3 and Siglec-9 inhibited FcγRs when coengaged by cells displaying ligands for both the Siglec and FcγRs. These results suggest a role for Siglecs in mediating FcγR inhibition in the context of an immunological synapse, which has important relevance to the effectiveness of immunotherapies.
Collapse
Affiliation(s)
- Kelli
A. McCord
- Department
of Chemistry, University of Alberta, 11227 Saskatchewan Drive, Edmonton, Alberta T6G 2G2, Canada
| | - Chao Wang
- Department
of Molecular Medicine, Scripps Research
Institute, 10550 North Torrey Pines Road, La Jolla, California 92037, United States
| | - Mirjam Anhalt
- Department
of Chemistry, University of Alberta, 11227 Saskatchewan Drive, Edmonton, Alberta T6G 2G2, Canada
| | - Wayne W. Poon
- Institute
for Memory Impairments and Neurological Disorders, University of California, Irvine, California 92617, United States
| | - Amanda L. Gavin
- Department
of Immunology and Microbiology, Scripps
Research Institute, 10550 North Torrey Pines Road, La Jolla, California 92037, United States
| | - Peng Wu
- Department
of Molecular Medicine, Scripps Research
Institute, 10550 North Torrey Pines Road, La Jolla, California 92037, United States
| | - Matthew S. Macauley
- Department
of Chemistry, University of Alberta, 11227 Saskatchewan Drive, Edmonton, Alberta T6G 2G2, Canada
- Department
of Medical Microbiology and Immunology, University of Alberta, Edmonton, Alberta T6G 2E1, Canada
| |
Collapse
|
35
|
Kosutova N, Lorencova L, Hires M, Jane E, Orovcik L, Kollar J, Kozics K, Gabelova A, Ukraintsev E, Rezek B, Kasak P, Cernocka H, Ostatna V, Blahutova J, Vikartovska A, Bertok T, Tkac J. Negative Charge-Carrying Glycans Attached to Exosomes as Novel Liquid Biopsy Marker. SENSORS (BASEL, SWITZERLAND) 2024; 24:1128. [PMID: 38400284 PMCID: PMC10892626 DOI: 10.3390/s24041128] [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: 01/09/2024] [Revised: 02/02/2024] [Accepted: 02/05/2024] [Indexed: 02/25/2024]
Abstract
Prostate cancer (PCa) is the second most common cancer. In this paper, the isolation and properties of exosomes as potential novel liquid biopsy markers for early PCa liquid biopsy diagnosis are investigated using two prostate human cell lines, i.e., benign (control) cell line RWPE1 and carcinoma cell line 22Rv1. Exosomes produced by both cell lines are characterised by various methods including nanoparticle-tracking analysis, dynamic light scattering, scanning electron microscopy and atomic force microscopy. In addition, surface plasmon resonance (SPR) is used to study three different receptors on the exosomal surface (CD63, CD81 and prostate-specific membrane antigen-PMSA), implementing monoclonal antibodies and identifying the type of glycans present on the surface of exosomes using lectins (glycan-recognising proteins). Electrochemical analysis is used to understand the interfacial properties of exosomes. The results indicate that cancerous exosomes are smaller, are produced at higher concentrations, and exhibit more nega tive zeta potential than the control exosomes. The SPR experiments confirm that negatively charged α-2,3- and α-2,6-sialic acid-containing glycans are found in greater abundance on carcinoma exosomes, whereas bisecting and branched glycans are more abundant in the control exosomes. The SPR results also show that a sandwich antibody/exosomes/lectins configuration could be constructed for effective glycoprofiling of exosomes as a novel liquid biopsy marker.
Collapse
Affiliation(s)
- Natalia Kosutova
- Institute of Chemistry, Slovak Academy of Sciences, Dubravska cesta 5807/9, 845 38 Bratislava, Slovakia (L.L.); (E.J.)
| | - Lenka Lorencova
- Institute of Chemistry, Slovak Academy of Sciences, Dubravska cesta 5807/9, 845 38 Bratislava, Slovakia (L.L.); (E.J.)
| | - Michal Hires
- Institute of Chemistry, Slovak Academy of Sciences, Dubravska cesta 5807/9, 845 38 Bratislava, Slovakia (L.L.); (E.J.)
| | - Eduard Jane
- Institute of Chemistry, Slovak Academy of Sciences, Dubravska cesta 5807/9, 845 38 Bratislava, Slovakia (L.L.); (E.J.)
| | - Lubomir Orovcik
- Institute of Materials and Machine Mechanics, Slovak Academy of Sciences, Dubravska cesta 9/6319, 845 13 Bratislava, Slovakia
| | - Jozef Kollar
- Polymer Institute, Slovak Academy of Sciences, Dubravska cesta 9, 845 41 Bratislava, Slovakia
| | - Katarina Kozics
- Biomedical Research Centre, Slovak Academy of Sciences, Dubravska cesta 9, 845 05 Bratislava, Slovakia (A.G.)
| | - Alena Gabelova
- Biomedical Research Centre, Slovak Academy of Sciences, Dubravska cesta 9, 845 05 Bratislava, Slovakia (A.G.)
| | - Egor Ukraintsev
- Department of Physics, Faculty of Electrical Engineering, Czech Technical University in Prague, Technicka 2, 166 27 Prague, Czech Republic; (E.U.); (B.R.)
| | - Bohuslav Rezek
- Department of Physics, Faculty of Electrical Engineering, Czech Technical University in Prague, Technicka 2, 166 27 Prague, Czech Republic; (E.U.); (B.R.)
| | - Peter Kasak
- Centre for Advanced Materials, Qatar University, Doha P.O. Box 2713, Qatar;
| | - Hana Cernocka
- Institute of Biophysics, Czech Academy of Sciences, Kralovopolska 135, 61200 Brno, Czech Republic; (H.C.)
| | - Veronika Ostatna
- Institute of Biophysics, Czech Academy of Sciences, Kralovopolska 135, 61200 Brno, Czech Republic; (H.C.)
| | - Jana Blahutova
- Institute of Chemistry, Slovak Academy of Sciences, Dubravska cesta 5807/9, 845 38 Bratislava, Slovakia (L.L.); (E.J.)
| | - Alica Vikartovska
- Institute of Chemistry, Slovak Academy of Sciences, Dubravska cesta 5807/9, 845 38 Bratislava, Slovakia (L.L.); (E.J.)
| | - Tomas Bertok
- Institute of Chemistry, Slovak Academy of Sciences, Dubravska cesta 5807/9, 845 38 Bratislava, Slovakia (L.L.); (E.J.)
| | - Jan Tkac
- Institute of Chemistry, Slovak Academy of Sciences, Dubravska cesta 5807/9, 845 38 Bratislava, Slovakia (L.L.); (E.J.)
| |
Collapse
|
36
|
Ghosh M, Hazarika P, Dhanya SJ, Pooja D, Kulhari H. Exploration of sialic acid receptors as a potential target for cancer treatment: A comprehensive review. Int J Biol Macromol 2024; 257:128415. [PMID: 38029891 DOI: 10.1016/j.ijbiomac.2023.128415] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2023] [Revised: 11/22/2023] [Accepted: 11/22/2023] [Indexed: 12/01/2023]
Abstract
The potential to target anticancer drugs directly to cancer cells is the most difficult challenge in the current scenario. Progressive works are being done on multifarious receptors and are on the horizon, expected to facilitate tailored treatment for cancer. Among several receptors, one is the sialic acid (SA) receptor by which cancer cells can be targeted directly as hyper sialylation is one of the most distinguishing characteristics of cancer cells. SA receptors have shown tremendous potential for tumor targeting because of their elevated expression in a range of human malignancies including prostate, breast, gastric cells, myeloid leukemia, liver, etc. This article reviews the overexpression of SA receptors in various tumors and diverse strategies for targeting these receptors to deliver drugs, enzymes, and genes for therapeutic applications. It also summarizes the diagnostic applications of SA-grafted nanoparticles for imaging various SA-overexpressing cancer cells and technological advances that are propelling sialic acid to the forefront of cancer therapy.
Collapse
Affiliation(s)
- Meheli Ghosh
- School of Nano Sciences, Central University of Gujarat, Gandhinagar, Gujarat 382030, India
| | - Priyodarshini Hazarika
- School of Nano Sciences, Central University of Gujarat, Gandhinagar, Gujarat 382030, India
| | - S J Dhanya
- School of Nano Sciences, Central University of Gujarat, Gandhinagar, Gujarat 382030, India
| | - Deep Pooja
- School of Pharmacy, National Forensic Science University, Gandhinagar, Gujarat 382007, India.
| | - Hitesh Kulhari
- School of Nano Sciences, Central University of Gujarat, Gandhinagar, Gujarat 382030, India; Department of Pharmaceutical Technology (Formulations), National Institute of Pharmaceutical Education and Research, Guwahati, Assam 781101, India.
| |
Collapse
|
37
|
Geng Z, Wu L, Wang Q, Ma J, Shi Z. Non B Cell-Derived Immunoglobulins in Intestinal Tract. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2024; 1445:137-149. [PMID: 38967756 DOI: 10.1007/978-981-97-0511-5_11] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/06/2024]
Abstract
Intestinal epithelium constitutes a barrier to the unrestricted movement of pathogens, and other detrimental substances from the external world (gut lumen) into the interstitial environment. Intestinal epithelial cells obstruct harmful substances passing through the epithelium as a physical and chemical barrier; Moreover, the epithelial cells can express Toll-like receptors (TLRs) and cytokines to exert innate immune function. In addition, high levels of immunoglobulin A (IgA) and other antibodies exist in the intestinal mucosa, maintaining intestinal immune homeostasis in conjunction with intestinal probiotics. Traditionally, these antibodies have been deemed to be secreted by submucosal plasma cells. Nonetheless, in recent years, it has been demonstrated that intestinal epithelial cells produce a substantial amount of Igs, especially IgA or free Ig light chains, which are involved in intestinal immune homeostasis and the survival of normal epithelial cells. Furthermore, mounting evidence affirms that many human carcinoma cells, including colorectal cancer (CRC), can overexpress Igs, particularly IgG. Cancer-derived Igs exhibit a unique V(D)J rearrangement pattern distinct from B cell-derived Ig; moreover, this cancer cell-derived IgG also has a unique sialic acid modification on the 162 site of CH1 domain (SIA-IgG). The SIA-IgG plays a crucial role in promoting cancer initiation, progression, metastasis, and tumour immune escape. Simultaneously, CRC cells can also express free Ig light chains, which promote colitis, colitis-associated colon carcinogenesis, and CRC progression. Therefore, Igs expressed by CRC cells could be a potential target for diagnosing and preventing the transformation of inflammation into cancer, as well as treating CRC.
Collapse
Affiliation(s)
- Zihan Geng
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, China.
| | - Lina Wu
- Central Laboratory, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital & Institute, Beijing, China
| | - Qianqian Wang
- School of Food and Drug, Shenzhen Polytechnic University, Shenzhen, China
| | - Junfan Ma
- Department of Clinical Research, Sinocelltech Group Limited, Beijing, China
| | - Zhan Shi
- Department of Ultrasound in Medicine, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
| |
Collapse
|
38
|
Johns SC, Gupta P, Lee YH, Friend J, Fuster MM. Glycocalyx transduces membrane leak in brain tumor cells exposed to sharp magnetic pulsing. Biophys J 2023; 122:4425-4439. [PMID: 37992690 PMCID: PMC10698326 DOI: 10.1016/j.bpj.2023.10.020] [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: 02/24/2023] [Revised: 07/23/2023] [Accepted: 10/19/2023] [Indexed: 11/24/2023] Open
Abstract
Mechanisms by which electric (E) or magnetic (B) fields might be harnessed to affect tumor cell behavior remain poorly defined, presenting a barrier to translation. We hypothesized in early studies that the glycocalyx of lung cancer cells might play a role in mediating plasma membrane leak by low-frequency pulsed magnetic fields (Lf-PMF) generated on a low-energy solenoid platform. In testing glioblastoma and neuroblastoma cells known to overexpress glycoproteins rich in modifications by the anionic glycan sialic acid (Sia), exposure of brain tumor cells on the same platform to a pulse train that included a 5 min 50Hz Lf-PMF (dB/dt ∼ 2 T/s at 10 ms pulse widths) induced a very modest but significant protease leak above that of control nonexposed cells (with modest but significant reductions in long-term tumor cell viability after the 5 min exposure). Using a markedly higher dB/dt system (80 T/s pulses, 70 μs pulse-width at 5.9 cm from a MagVenture coil source) induced markedly greater leak by the same cells, and eliminating Sia by treating cells with AUS sialidase immediately preexposure abrogated the effect entirely in SH-SY5Y neuroblastoma cells, and partially in T98G glioblastoma cells. The system demonstrated significant leak (including inward leak of propidium iodide), with reduced leak at lower dB/dt in a variety of tumor cells. The ability to abrogate Lf-PMF protease leak by pretreatment with sialidase in SH-SY5Y brain tumor cells or with heparin lyase in A549 lung tumor cells indicated the importance of heavy Sia or heparan sulfate glycosaminoglycan glycocalyx modifications as dominant glycan species mediating Lf-PMF membrane leak in respective tumor cells. This "first-physical" Lf-PMF tumor glycocalyx event, with downstream cell stress, may represent a critical and "tunable" transduction mechanism that depends on characteristic anionic glycans overexpressed by distinct malignant tumors.
Collapse
Affiliation(s)
- Scott C Johns
- VA San Diego Healthcare System, San Diego, California; Veterans Medical Research Foundation, San Diego, California
| | - Purva Gupta
- VA San Diego Healthcare System, San Diego, California; Department of Medicine, Division of Pulmonary & Critical Care, University of California San Diego, La Jolla, California
| | - Yi-Hung Lee
- Department of Bioengineering, University of California San Diego, La Jolla, California
| | - James Friend
- Department of Mechanical and Aerospace Engineering, University of California San Diego, La Jolla, California
| | - Mark M Fuster
- VA San Diego Healthcare System, San Diego, California; Veterans Medical Research Foundation, San Diego, California; Department of Medicine, Division of Pulmonary & Critical Care, University of California San Diego, La Jolla, California; Glycobiology Research and Training Center, University of California San Diego, La Jolla, California.
| |
Collapse
|
39
|
Cerella C, Dicato M, Diederich M. Enhancing personalized immune checkpoint therapy by immune archetyping and pharmacological targeting. Pharmacol Res 2023; 196:106914. [PMID: 37714393 DOI: 10.1016/j.phrs.2023.106914] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/22/2023] [Revised: 09/04/2023] [Accepted: 09/06/2023] [Indexed: 09/17/2023]
Abstract
Immune checkpoint inhibitors (ICIs) are an expanding class of immunotherapeutic agents with the potential to cure cancer. Despite the outstanding clinical response in patient subsets, most individuals become refractory or develop resistance. Patient stratification and personalized immunotherapies are limited by the absence of predictive response markers. Recent findings show that dominant patterns of immune cell composition, T-cell status and heterogeneity, and spatiotemporal distribution of immune cells within the tumor microenvironment (TME) are becoming essential determinants of prognosis and therapeutic response. In this context, ICIs also function as investigational tools and proof of concept, allowing the validation of the identified mechanisms. After reviewing the current state of ICIs, this article will explore new comprehensive predictive markers for ICIs based on recent discoveries. We will discuss the recent establishment of a classification of TMEs into immune archetypes as a tool for personalized immune profiling, allowing patient stratification before ICI treatment. We will discuss the developing comprehension of T-cell diversity and its role in shaping the immune profile of patients. We describe the potential of strategies that score the mutual spatiotemporal modulation between T-cells and other cellular components of the TME. Additionally, we will provide an overview of a range of synthetic and naturally occurring or derived small molecules. We will compare compounds that were recently identified by in silico prediction to wet lab-validated drug candidates with the potential to function as ICIs and/or modulators of the cellular components of the TME.
Collapse
Affiliation(s)
- Claudia Cerella
- Laboratoire de Biologie Moléculaire et Cellulaire du Cancer (LBMCC), Fondation Recherche sur le Cancer et les Maladies du Sang, Pavillon 2, 6A rue Barblé, L-1210 Luxembourg, Luxembourg
| | - Mario Dicato
- Laboratoire de Biologie Moléculaire et Cellulaire du Cancer (LBMCC), Fondation Recherche sur le Cancer et les Maladies du Sang, Pavillon 2, 6A rue Barblé, L-1210 Luxembourg, Luxembourg
| | - Marc Diederich
- Department of Pharmacy, College of Pharmacy, Seoul National University, Gwanak-ro, Gwanak-gu, Seoul 08826, Korea.
| |
Collapse
|
40
|
Wang Y, Liu S, Li J, Yin T, Liu Y, Wang Q, Liu X, Cheng L. Comprehensive serum N-glycan profiling identifies a biomarker panel for early diagnosis of non-small-cell lung cancer. Proteomics 2023; 23:e2300140. [PMID: 37474491 DOI: 10.1002/pmic.202300140] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2023] [Revised: 07/01/2023] [Accepted: 07/04/2023] [Indexed: 07/22/2023]
Abstract
Aberrant serum N-glycan profiles have been observed in multiple cancers including non-small-cell lung cancer (NSCLC), yet the potential of N-glycans in the early diagnosis of NSCLC remains to be determined. In this study, serum N-glycan profiles of 275 NSCLC patients and 309 healthy controls were characterized by MALDI-TOF-MS. The levels of serum N-glycans and N-glycosylation patterns were compared between NSCLC and control groups. In addition, a panel of N-glycan biomarkers for NSCLC diagnosis was established and validated using machine learning algorithms. As a result, a total of 54 N-glycan structures were identified in human serum. Compared with healthy controls, 29 serum N-glycans were increased or decreased in NSCLC patients. N-glycan abundance in different histological types or clinical stages of NSCLC presented differentiated changes. Furthermore, an optimal biomarker panel of eight N-glycans was constructed based on logistic regression, with an AUC of 0.86 in the validation set. Notably, this model also showed a desirable capacity in distinguishing early-stage patients from healthy controls (AUC = 0.88). In conclusion, our work highlights the abnormal N-glycan profiles in NSCLC and provides supports potential application of N-glycan biomarker panel in clinical NSCLC detection.
Collapse
Affiliation(s)
- Yi Wang
- Department of Laboratory Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Si Liu
- The Key Laboratory for Biomedical Photonics of MOE at Wuhan National Laboratory for Optoelectronics-Hubei Bioinformatics & Molecular Imaging Key Laboratory, Systems Biology Theme, Department of Biomedical Engineering, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, China
| | - Jiaoyuan Li
- Department of Laboratory Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Tongxin Yin
- Department of Laboratory Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yuanyuan Liu
- The Key Laboratory for Biomedical Photonics of MOE at Wuhan National Laboratory for Optoelectronics-Hubei Bioinformatics & Molecular Imaging Key Laboratory, Systems Biology Theme, Department of Biomedical Engineering, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, China
| | - Qiankun Wang
- Department of Laboratory Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xin Liu
- The Key Laboratory for Biomedical Photonics of MOE at Wuhan National Laboratory for Optoelectronics-Hubei Bioinformatics & Molecular Imaging Key Laboratory, Systems Biology Theme, Department of Biomedical Engineering, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, China
| | - Liming Cheng
- Department of Laboratory Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
41
|
Scott E, Archer Goode E, Garnham R, Hodgson K, Orozco-Moreno M, Turner H, Livermore K, Putri Nangkana K, Frame FM, Bermudez A, Jose Garcia Marques F, McClurg UL, Wilson L, Thomas H, Buskin A, Hepburn A, Duxfield A, Bastian K, Pye H, Arredondo HM, Hysenaj G, Heavey S, Stopka-Farooqui U, Haider A, Freeman A, Singh S, Johnston EW, Punwani S, Knight B, McCullagh P, McGrath J, Crundwell M, Harries L, Heer R, Maitland NJ, Whitaker H, Pitteri S, Troyer DA, Wang N, Elliott DJ, Drake RR, Munkley J. ST6GAL1-mediated aberrant sialylation promotes prostate cancer progression. J Pathol 2023; 261:71-84. [PMID: 37550801 DOI: 10.1002/path.6152] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Revised: 05/05/2023] [Accepted: 06/02/2023] [Indexed: 08/09/2023]
Abstract
Aberrant glycosylation is a universal feature of cancer cells, and cancer-associated glycans have been detected in virtually every cancer type. A common change in tumour cell glycosylation is an increase in α2,6 sialylation of N-glycans, a modification driven by the sialyltransferase ST6GAL1. ST6GAL1 is overexpressed in numerous cancer types, and sialylated glycans are fundamental for tumour growth, metastasis, immune evasion, and drug resistance, but the role of ST6GAL1 in prostate cancer is poorly understood. Here, we analyse matched cancer and normal tissue samples from 200 patients and verify that ST6GAL1 is upregulated in prostate cancer tissue. Using MALDI imaging mass spectrometry (MALDI-IMS), we identify larger branched α2,6 sialylated N-glycans that show specificity to prostate tumour tissue. We also monitored ST6GAL1 in plasma samples from >400 patients and reveal ST6GAL1 levels are significantly increased in the blood of men with prostate cancer. Using both in vitro and in vivo studies, we demonstrate that ST6GAL1 promotes prostate tumour growth and invasion. Our findings show ST6GAL1 introduces α2,6 sialylated N-glycans on prostate cancer cells and raise the possibility that prostate cancer cells can secrete active ST6GAL1 enzyme capable of remodelling glycans on the surface of other cells. Furthermore, we find α2,6 sialylated N-glycans expressed by prostate cancer cells can be targeted using the sialyltransferase inhibitor P-3FAX -Neu5Ac. Our study identifies an important role for ST6GAL1 and α2,6 sialylated N-glycans in prostate cancer progression and highlights the opportunity to inhibit abnormal sialylation for the development of new prostate cancer therapeutics. © 2023 The Authors. The Journal of Pathology published by John Wiley & Sons Ltd on behalf of The Pathological Society of Great Britain and Ireland.
Collapse
Affiliation(s)
- Emma Scott
- Newcastle University Centre for Cancer, Newcastle University Institute of Biosciences, Newcastle, UK
| | - Emily Archer Goode
- Newcastle University Centre for Cancer, Newcastle University Institute of Biosciences, Newcastle, UK
| | - Rebecca Garnham
- Newcastle University Centre for Cancer, Newcastle University Institute of Biosciences, Newcastle, UK
| | - Kirsty Hodgson
- Newcastle University Centre for Cancer, Newcastle University Institute of Biosciences, Newcastle, UK
| | - Margarita Orozco-Moreno
- Newcastle University Centre for Cancer, Newcastle University Institute of Biosciences, Newcastle, UK
| | - Helen Turner
- Cellular Pathology, The Royal Victoria Infirmary, Newcastle upon Tyne, UK
| | - Karen Livermore
- Newcastle University Centre for Cancer, Newcastle University Institute of Biosciences, Newcastle, UK
| | - Kyla Putri Nangkana
- Newcastle University Centre for Cancer, Newcastle University Institute of Biosciences, Newcastle, UK
| | - Fiona M Frame
- Cancer Research Unit, Department of Biology, University of York, North Yorkshire, UK
| | - Abel Bermudez
- Canary Center at Stanford for Cancer Early Detection, Department of Radiology, Stanford University, Palo Alto, CA, USA
| | - Fernando Jose Garcia Marques
- Canary Center at Stanford for Cancer Early Detection, Department of Radiology, Stanford University, Palo Alto, CA, USA
| | - Urszula L McClurg
- Institute of Systems, Molecular & Integrative Biology, University of Liverpool, Liverpool, UK
| | - Laura Wilson
- Newcastle University Centre for Cancer, Translational and Clinical Research Institute, Paul O'Gorman Building, Newcastle University, Newcastle upon Tyne, UK
| | - Huw Thomas
- Newcastle University Centre for Cancer, Translational and Clinical Research Institute, Paul O'Gorman Building, Newcastle University, Newcastle upon Tyne, UK
| | - Adriana Buskin
- Newcastle University Centre for Cancer, Translational and Clinical Research Institute, Paul O'Gorman Building, Newcastle University, Newcastle upon Tyne, UK
| | - Anastasia Hepburn
- Newcastle University Centre for Cancer, Translational and Clinical Research Institute, Paul O'Gorman Building, Newcastle University, Newcastle upon Tyne, UK
| | - Adam Duxfield
- Newcastle University Centre for Cancer, Newcastle University Institute of Biosciences, Newcastle, UK
| | - Kayla Bastian
- Newcastle University Centre for Cancer, Newcastle University Institute of Biosciences, Newcastle, UK
| | - Hayley Pye
- Molecular Diagnostics and Therapeutics Group, Charles Bell House, Division of Surgery and Interventional Science, University College London, London, UK
| | - Hector M Arredondo
- The Mellanby Centre for Musculoskeletal Research, Department of Oncology and Metabolism, The University of Sheffield, Sheffield, UK
| | - Gerald Hysenaj
- Newcastle University Centre for Cancer, Newcastle University Institute of Biosciences, Newcastle, UK
| | - Susan Heavey
- Molecular Diagnostics and Therapeutics Group, Charles Bell House, Division of Surgery and Interventional Science, University College London, London, UK
| | - Urszula Stopka-Farooqui
- Molecular Diagnostics and Therapeutics Group, Charles Bell House, Division of Surgery and Interventional Science, University College London, London, UK
| | - Aiman Haider
- Department of Pathology, UCLH NHS Foundation Trust, London, UK
| | - Alex Freeman
- Department of Pathology, UCLH NHS Foundation Trust, London, UK
| | - Saurabh Singh
- UCL Centre for Medical Imaging, Charles Bell House, University College London, London, UK
| | - Edward W Johnston
- UCL Centre for Medical Imaging, Charles Bell House, University College London, London, UK
| | - Shonit Punwani
- UCL Centre for Medical Imaging, Charles Bell House, University College London, London, UK
| | - Bridget Knight
- NIHR Exeter Clinical Research Facility, Royal Devon and Exeter NHS Foundation Trust, Exeter, UK
| | - Paul McCullagh
- Department of Pathology, Royal Devon and Exeter NHS Foundation Trust, Exeter, UK
| | - John McGrath
- Exeter Surgical Health Services Research Unit, Royal Devon and Exeter NHS Foundation Trust, Exeter, UK
| | - Malcolm Crundwell
- Exeter Surgical Health Services Research Unit, Royal Devon and Exeter NHS Foundation Trust, Exeter, UK
| | - Lorna Harries
- Institute of Biomedical and Clinical Sciences, Medical School, College of Medicine and Health, University of Exeter, Exeter, UK
| | - Rakesh Heer
- Newcastle University Centre for Cancer, Translational and Clinical Research Institute, Paul O'Gorman Building, Newcastle University, Newcastle upon Tyne, UK
- Department of Urology, Freeman Hospital, The Newcastle upon Tyne Hospitals NHS Foundation Trust, Newcastle upon Tyne, UK
| | - Norman J Maitland
- Cancer Research Unit, Department of Biology, University of York, North Yorkshire, UK
| | - Hayley Whitaker
- Molecular Diagnostics and Therapeutics Group, Charles Bell House, Division of Surgery and Interventional Science, University College London, London, UK
| | - Sharon Pitteri
- Canary Center at Stanford for Cancer Early Detection, Department of Radiology, Stanford University, Palo Alto, CA, USA
| | - Dean A Troyer
- Cancer Biology and Infectious Disease Research Center, Eastern Virginia Medical School, Norfolk, VA, USA
| | - Ning Wang
- The Mellanby Centre for Musculoskeletal Research, Department of Oncology and Metabolism, The University of Sheffield, Sheffield, UK
| | - David J Elliott
- Newcastle University Centre for Cancer, Newcastle University Institute of Biosciences, Newcastle, UK
| | - Richard R Drake
- Department of Cell and Molecular Pharmacology, Medical University of South Carolina, Charleston, SC, USA
| | - Jennifer Munkley
- Newcastle University Centre for Cancer, Newcastle University Institute of Biosciences, Newcastle, UK
| |
Collapse
|
42
|
Makarova N, Lekka M, Gnanachandran K, Sokolov I. Mechanical Way To Study Molecular Structure of Pericellular Layer. ACS APPLIED MATERIALS & INTERFACES 2023; 15:35962-35972. [PMID: 37489588 PMCID: PMC10401571 DOI: 10.1021/acsami.3c06341] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/05/2023] [Accepted: 07/13/2023] [Indexed: 07/26/2023]
Abstract
Atomic force microscopy (AFM) has been used to study the mechanical properties of cells, in particular, malignant cells. Softening of various cancer cells compared to their nonmalignant counterparts has been reported for various cell types. However, in most AFM studies, the pericellular layer was ignored. As was shown, it could substantially change the measured cell rigidity and miss important information on the physical properties of the pericellular layer. Here we take into account the pericellular layer by using the brush model to do the AFM indentation study of bladder epithelial bladder nonmalignant (HCV29) and cancerous (TCCSUP) cells. It allows us to measure not only the quasistatic Young's modulus of the cell body but also the physical properties of the pericellular layer (the equilibrium length and grafting density). We found that the inner pericellular brush was longer for cancer cells, but its grafting density was similar to that found for nonmalignant cells. The outer brush was much shorter and less dense for cancer cells. Furthermore, we demonstrate a method to convert the obtained physical properties of the pericellular layer into biochemical language better known to the cell biology community. It is done by using heparinase I and neuraminidase enzymatic treatments that remove specific molecular parts of the pericellular layer. The presented here approach can also be used to decipher the molecular composition of not only pericellular but also other molecular layers.
Collapse
Affiliation(s)
- Nadezda Makarova
- Department
of Mechanical Engineering, Tufts University, Medford, Massachusetts 02155, United States
| | - Małgorzata Lekka
- Department
of Biophysical Microstructures, Institute
of Nuclear Physics PAN, PL-31342 Kraków, Poland
| | - Kajangi Gnanachandran
- Department
of Biophysical Microstructures, Institute
of Nuclear Physics PAN, PL-31342 Kraków, Poland
| | - Igor Sokolov
- Department
of Mechanical Engineering, Tufts University, Medford, Massachusetts 02155, United States
- Department
of Physics, Tufts University, Medford, Massachusetts 02155, United States
| |
Collapse
|
43
|
Harduin-Lepers A. The vertebrate sialylation machinery: structure-function and molecular evolution of GT-29 sialyltransferases. Glycoconj J 2023; 40:473-492. [PMID: 37247156 PMCID: PMC10225777 DOI: 10.1007/s10719-023-10123-w] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2023] [Revised: 03/09/2023] [Accepted: 05/10/2023] [Indexed: 05/30/2023]
Abstract
Every eukaryotic cell is covered with a thick layer of complex carbohydrates with essential roles in their social life. In Deuterostoma, sialic acids present at the outermost positions of glycans of glycoconjugates are known to be key players in cellular interactions including host-pathogen interactions. Their negative charge and hydrophilic properties enable their roles in various normal and pathological states and their expression is altered in many diseases including cancers. Sialylation of glycoproteins and glycolipids is orchestrated by the regulated expression of twenty sialyltransferases in human tissues with distinct enzymatic characteristics and preferences for substrates and linkages formed. However, still very little is known on the functional organization of sialyltransferases in the Golgi apparatus and how the sialylation machinery is finely regulated to provide the ad hoc sialome to the cell. This review summarizes current knowledge on sialyltransferases, their structure-function relationships, molecular evolution, and their implications in human biology.
Collapse
Affiliation(s)
- Anne Harduin-Lepers
- Univ. Lille, CNRS, UMR 8576 - UGSF - Unité de Glycobiologie Structurale et Fonctionnelle, F-59000, Lille, France.
| |
Collapse
|
44
|
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
|
45
|
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: 1] [Impact Index Per Article: 0.5] [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
|
46
|
Egan H, Treacy O, Lynch K, Leonard NA, O'Malley G, Reidy E, O'Neill A, Corry SM, De Veirman K, Vanderkerken K, Egan LJ, Ritter T, Hogan AM, Redmond K, Peng L, Che J, Gatlin W, Jayaraman P, Sheehan M, Canney A, Hynes SO, Kerr EM, Dunne PD, O'Dwyer ME, Ryan AE. Targeting stromal cell sialylation reverses T cell-mediated immunosuppression in the tumor microenvironment. Cell Rep 2023; 42:112475. [PMID: 37167967 DOI: 10.1016/j.celrep.2023.112475] [Citation(s) in RCA: 28] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2021] [Revised: 02/03/2023] [Accepted: 04/19/2023] [Indexed: 05/13/2023] Open
Abstract
Immunosuppressive tumor microenvironments (TMEs) reduce the effectiveness of immune responses in cancer. Mesenchymal stromal cells (MSCs), precursors to cancer-associated fibroblasts (CAFs), promote tumor progression by enhancing immune cell suppression in colorectal cancer (CRC). Hyper-sialylation of glycans promotes immune evasion in cancer through binding of sialic acids to their receptors, Siglecs, expressed on immune cells, which results in inhibition of effector functions. The role of sialylation in shaping MSC/CAF immunosuppression in the TME is not well characterized. In this study, we show that tumor-conditioned stromal cells have increased sialyltransferase expression, α2,3/6-linked sialic acid, and Siglec ligands. Tumor-conditioned stromal cells and CAFs induce exhausted immunomodulatory CD8+ PD1+ and CD8+ Siglec-7+/Siglec-9+ T cell phenotypes. In vivo, targeting stromal cell sialylation reverses stromal cell-mediated immunosuppression, as shown by infiltration of CD25 and granzyme B-expressing CD8+ T cells in the tumor and draining lymph node. Targeting stromal cell sialylation may overcome immunosuppression in the CRC TME.
Collapse
Affiliation(s)
- Hannah Egan
- Discipline of Pharmacology and Therapeutics, School of Medicine, College of Medicine, Nursing and Health Sciences, University of Galway, Galway, Ireland; Regenerative Medicine Institute (REMEDI), School of Medicine, College of Medicine, Nursing and Health Sciences, University of Galway, Galway, Ireland; Lambe Institute for Translational Research, School of Medicine, College of Medicine, Nursing and Health Sciences, University of Galway, Galway, Ireland
| | - Oliver Treacy
- Discipline of Pharmacology and Therapeutics, School of Medicine, College of Medicine, Nursing and Health Sciences, University of Galway, Galway, Ireland; Regenerative Medicine Institute (REMEDI), School of Medicine, College of Medicine, Nursing and Health Sciences, University of Galway, Galway, Ireland; Lambe Institute for Translational Research, School of Medicine, College of Medicine, Nursing and Health Sciences, University of Galway, Galway, Ireland
| | - Kevin Lynch
- Discipline of Pharmacology and Therapeutics, School of Medicine, College of Medicine, Nursing and Health Sciences, University of Galway, Galway, Ireland; Regenerative Medicine Institute (REMEDI), School of Medicine, College of Medicine, Nursing and Health Sciences, University of Galway, Galway, Ireland; Lambe Institute for Translational Research, School of Medicine, College of Medicine, Nursing and Health Sciences, University of Galway, Galway, Ireland
| | - Niamh A Leonard
- Discipline of Pharmacology and Therapeutics, School of Medicine, College of Medicine, Nursing and Health Sciences, University of Galway, Galway, Ireland; Regenerative Medicine Institute (REMEDI), School of Medicine, College of Medicine, Nursing and Health Sciences, University of Galway, Galway, Ireland; Lambe Institute for Translational Research, School of Medicine, College of Medicine, Nursing and Health Sciences, University of Galway, Galway, Ireland
| | - Grace O'Malley
- Discipline of Pharmacology and Therapeutics, School of Medicine, College of Medicine, Nursing and Health Sciences, University of Galway, Galway, Ireland; Regenerative Medicine Institute (REMEDI), School of Medicine, College of Medicine, Nursing and Health Sciences, University of Galway, Galway, Ireland; Lambe Institute for Translational Research, School of Medicine, College of Medicine, Nursing and Health Sciences, University of Galway, Galway, Ireland
| | - Eileen Reidy
- Discipline of Pharmacology and Therapeutics, School of Medicine, College of Medicine, Nursing and Health Sciences, University of Galway, Galway, Ireland; Regenerative Medicine Institute (REMEDI), School of Medicine, College of Medicine, Nursing and Health Sciences, University of Galway, Galway, Ireland; Lambe Institute for Translational Research, School of Medicine, College of Medicine, Nursing and Health Sciences, University of Galway, Galway, Ireland; CÚRAM, SFI Research Centre for Medical Devices, University of Galway, Galway, Ireland
| | - Aoise O'Neill
- Discipline of Pharmacology and Therapeutics, School of Medicine, College of Medicine, Nursing and Health Sciences, University of Galway, Galway, Ireland; Regenerative Medicine Institute (REMEDI), School of Medicine, College of Medicine, Nursing and Health Sciences, University of Galway, Galway, Ireland; Lambe Institute for Translational Research, School of Medicine, College of Medicine, Nursing and Health Sciences, University of Galway, Galway, Ireland
| | - Shania M Corry
- Patrick G Johnston Centre for Cancer Research, Queen's University Belfast, Belfast, UK
| | - Kim De Veirman
- Laboratory for Haematology and Immunology (HEIM), Department of Biomedical Sciences, Vrije Universiteit Brussel, Brussels, Belgium
| | - Karin Vanderkerken
- Laboratory for Haematology and Immunology (HEIM), Department of Biomedical Sciences, Vrije Universiteit Brussel, Brussels, Belgium
| | - Laurence J Egan
- Discipline of Pharmacology and Therapeutics, School of Medicine, College of Medicine, Nursing and Health Sciences, University of Galway, Galway, Ireland; Lambe Institute for Translational Research, School of Medicine, College of Medicine, Nursing and Health Sciences, University of Galway, Galway, Ireland
| | - Thomas Ritter
- Regenerative Medicine Institute (REMEDI), School of Medicine, College of Medicine, Nursing and Health Sciences, University of Galway, Galway, Ireland; CÚRAM, SFI Research Centre for Medical Devices, University of Galway, Galway, Ireland
| | - Aisling M Hogan
- Lambe Institute for Translational Research, School of Medicine, College of Medicine, Nursing and Health Sciences, University of Galway, Galway, Ireland; Department of Colorectal Surgery, Galway University Hospital, Galway, Ireland
| | - Keara Redmond
- Patrick G Johnston Centre for Cancer Research, Queen's University Belfast, Belfast, UK
| | - Li Peng
- Palleon Pharmaceuticals, Waltham, MA 02451, USA
| | - Jenny Che
- Palleon Pharmaceuticals, Waltham, MA 02451, USA
| | | | | | - Margaret Sheehan
- Division of Anatomical Pathology, Galway University Hospital, Galway, Ireland
| | - Aoife Canney
- Division of Anatomical Pathology, Galway University Hospital, Galway, Ireland
| | - Sean O Hynes
- Division of Anatomical Pathology, Galway University Hospital, Galway, Ireland; Discipline of Pathology, School of Medicine, College of Medicine, Nursing and Health Sciences, University of Galway, Galway, Ireland
| | - Emma M Kerr
- Patrick G Johnston Centre for Cancer Research, Queen's University Belfast, Belfast, UK
| | - Philip D Dunne
- Patrick G Johnston Centre for Cancer Research, Queen's University Belfast, Belfast, UK; Cancer Research UK Beatson Institute, Glasgow, UK
| | - Michael E O'Dwyer
- Lambe Institute for Translational Research, School of Medicine, College of Medicine, Nursing and Health Sciences, University of Galway, Galway, Ireland; Blood Cancer Network of Ireland (BCNI), Galway, Ireland; Department of Hematology, Galway University Hospital, Galway, Ireland
| | - Aideen E Ryan
- Discipline of Pharmacology and Therapeutics, School of Medicine, College of Medicine, Nursing and Health Sciences, University of Galway, Galway, Ireland; Regenerative Medicine Institute (REMEDI), School of Medicine, College of Medicine, Nursing and Health Sciences, University of Galway, Galway, Ireland; Lambe Institute for Translational Research, School of Medicine, College of Medicine, Nursing and Health Sciences, University of Galway, Galway, Ireland; CÚRAM, SFI Research Centre for Medical Devices, University of Galway, Galway, Ireland.
| |
Collapse
|
47
|
Takatori SC, Son S, Lee DSW, Fletcher DA. Engineered molecular sensors for quantifying cell surface crowding. Proc Natl Acad Sci U S A 2023; 120:e2219778120. [PMID: 37186825 PMCID: PMC10214205 DOI: 10.1073/pnas.2219778120] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2022] [Accepted: 04/13/2023] [Indexed: 05/17/2023] Open
Abstract
Cells mediate interactions with the extracellular environment through a crowded assembly of transmembrane proteins, glycoproteins and glycolipids on their plasma membrane. The extent to which surface crowding modulates the biophysical interactions of ligands, receptors, and other macromolecules is poorly understood due to the lack of methods to quantify surface crowding on native cell membranes. In this work, we demonstrate that physical crowding on reconstituted membranes and live cell surfaces attenuates the effective binding affinity of macromolecules such as IgG antibodies in a surface crowding-dependent manner. We combine experiment and simulation to design a crowding sensor based on this principle that provides a quantitative readout of cell surface crowding. Our measurements reveal that surface crowding decreases IgG antibody binding by 2 to 20 fold in live cells compared to a bare membrane surface. Our sensors show that sialic acid, a negatively charged monosaccharide, contributes disproportionately to red blood cell surface crowding via electrostatic repulsion, despite occupying only ~1% of the total cell membrane by mass. We also observe significant differences in surface crowding for different cell types and find that expression of single oncogenes can both increase and decrease crowding, suggesting that surface crowding may be an indicator of both cell type and state. Our high-throughput, single-cell measurement of cell surface crowding may be combined with functional assays to enable further biophysical dissection of the cell surfaceome.
Collapse
Affiliation(s)
- Sho C. Takatori
- Department of Bioengineering, University of California, Berkeley, CA94720
- Department of Chemical Engineering, University of California, Santa Barbara, CA93106
| | - Sungmin Son
- Department of Bioengineering, University of California, Berkeley, CA94720
- Department of Bio and Brain Engineering, Korea Advanced Institute of Science and Technology, Daejeon, Republic of Korea
| | - Daniel S. W. Lee
- Department of Bioengineering, University of California, Berkeley, CA94720
| | - Daniel A. Fletcher
- Department of Bioengineering, University of California, Berkeley, CA94720
- University of California, Berkeley/University of California, San Francisco Graduate Group in Bioengineering, Berkeley, CA94720
- Division of Biological Systems and Engineering, Lawrence Berkeley National Laboratory, Berkeley, CA94720
- Chan Zuckerberg Biohub, San Francisco, CA94158
| |
Collapse
|
48
|
Mondal SK, Ahmed MT, Jinka S, Sarkar S, Shukla R, Banerjee R. Progesterone-Cationic Lipid Conjugate-Based Self-Aggregates for Cancer Cell-Selective Uptake through Macropinocytosis and the Antitumour Effect. Mol Pharm 2023. [PMID: 37134112 DOI: 10.1021/acs.molpharmaceut.2c00887] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/04/2023]
Abstract
Progesterone (PR) is an endogenous steroid hormone that activates the progesterone receptor (PgR) and is known to play a critical role in cancer progression. Herein, we report the development of cationic lipid-conjugated PR derivatives by covalently conjugating progesterone with cationic lipids of varying hydrocarbon chain lengths (n = 6-18) through a succinate linker. Cytotoxicity studies performed on eight different cancer cell lines reveal that PR10, one of the lead derivatives, exerts notable toxicity (IC50 = 4-12 μM) in cancer cells irrespective of their PgR expression status and remains largely nontoxic to noncancerous cells. Mechanistic studies show that PR10 induces G2/M-phase cell cycle arrest in cancer cells, leading to apoptosis and cell death by inhibiting the PI3K/AKT cell survival pathway and p53 upregulation. Further, in vivo study shows that PR10 treatment significantly reduces melanoma tumor growth and prolongs the overall survival of melanoma tumor-bearing C57BL/6J mice. Interestingly, PR10 readily forms stable self-aggregates of ∼190 nm size in an aqueous environment and exhibits selective uptake into cancerous cell lines. In vitro uptake mechanism studies in various cell lines (cancerous cell lines B16F10, MCF7, PC3, and noncancerous cell line HEK293) using endocytosis inhibition proves that PR10 nanoaggregates enter selectively into the cancer cells predominantly using macropinocytosis and/or caveolae-mediated endocytosis. Overall, this study highlights the development of a self-aggregating cationic derivative of progesterone with anticancer activity, and its cancer cell-selective accumulation in nanoaggregate form holds great potential in the field of targeted drug delivery.
Collapse
Affiliation(s)
- Sujan Kumar Mondal
- Applied Biology Division, CSIR-Indian Institute of Chemical Technology, Hyderabad 500007, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Mohammed Tanveer Ahmed
- Applied Biology Division, CSIR-Indian Institute of Chemical Technology, Hyderabad 500007, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
- School of Science, STEM College, RMIT University, Melbourne, Victoria 3001, Australia
| | - Sudhakar Jinka
- Applied Biology Division, CSIR-Indian Institute of Chemical Technology, Hyderabad 500007, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Sampa Sarkar
- School of Science, STEM College, RMIT University, Melbourne, Victoria 3001, Australia
| | - Ravi Shukla
- School of Science, STEM College, RMIT University, Melbourne, Victoria 3001, Australia
- NanoBiotechnology Research Laboratory, Centre for Advanced Materials & Industrial Chemistry, RMIT University, Melbourne, Victoria 3001, Australia
| | - Rajkumar Banerjee
- Applied Biology Division, CSIR-Indian Institute of Chemical Technology, Hyderabad 500007, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| |
Collapse
|
49
|
Čaval T, Alisson-Silva F, Schwarz F. Roles of glycosylation at the cancer cell surface: opportunities for large scale glycoproteomics. Theranostics 2023; 13:2605-2615. [PMID: 37215580 PMCID: PMC10196828 DOI: 10.7150/thno.81760] [Citation(s) in RCA: 28] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2022] [Accepted: 04/13/2023] [Indexed: 05/24/2023] Open
Abstract
Cell surface glycosylation has a variety of functions, and its dysregulation in cancer contributes to impaired signaling, metastasis and the evasion of the immune responses. Recently, a number of glycosyltransferases that lead to altered glycosylation have been linked to reduced anti-tumor immune responses: B3GNT3, which is implicated in PD-L1 glycosylation in triple negative breast cancer, FUT8, through fucosylation of B7H3, and B3GNT2, which confers cancer resistance to T cell cytotoxicity. Given the increased appreciation of the relevance of protein glycosylation, there is a critical need for the development of methods that allow for an unbiased interrogation of cell surface glycosylation status. Here we provide an overview of the broad changes in glycosylation at the surface of cancer cell and describe selected examples of receptors with aberrant glycosylation leading to functional changes, with emphasis on immune checkpoint inhibitors, growth-promoting and growth-arresting receptors. Finally, we posit that the field of glycoproteomics has matured to an extent where large-scale profiling of intact glycopeptides from the cell surface is feasible and is poised for discovery of new actionable targets against cancer.
Collapse
|
50
|
Li Y, Wang M, Hong S. Live-Cell Glycocalyx Engineering. Chembiochem 2023; 24:e202200707. [PMID: 36642971 DOI: 10.1002/cbic.202200707] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2022] [Revised: 01/14/2023] [Accepted: 01/14/2023] [Indexed: 01/17/2023]
Abstract
A heavy layer of glycans forms a brush matrix bound to the outside of all the cells in our bodies; it is referred to as the "sugar forest" or glycocalyx. Beyond the increased appreciation of the glycocalyx over the past two decades, recent advances in engineering the glycocalyx on live cells have spurred the creation of cellular drugs and novel medical treatments. The development of new tools and techniques has empowered scientists to manipulate the structures and functions of cell-surface glycans on target cells and endow target cells with desired properties. Herein, we provide an overview of live-cell glycocalyx engineering strategies for controlling the cell-surface molecular repertory to suit therapeutic applications, even though the realm of this field remains young and largely unexplored.
Collapse
Affiliation(s)
- Yuxin Li
- State Key Laboratory of Natural and Biomimetic Drugs, Chemical Biology Center, and School of Pharmaceutical Sciences, Peking University, Health Science Center, Beijing, 100191, China
| | - Mingzhen Wang
- State Key Laboratory of Natural and Biomimetic Drugs, Chemical Biology Center, and School of Pharmaceutical Sciences, Peking University, Health Science Center, Beijing, 100191, China
| | - Senlian Hong
- State Key Laboratory of Natural and Biomimetic Drugs, Chemical Biology Center, and School of Pharmaceutical Sciences, Peking University, Health Science Center, Beijing, 100191, China
| |
Collapse
|