1
|
Yu WB, Ye ZH, Shi JJ, Deng WQ, Chen J, Lu JJ. Dual blockade of GSTK1 and CD47 improves macrophage-mediated phagocytosis on cancer cells. Biochem Pharmacol 2025; 236:116898. [PMID: 40147800 DOI: 10.1016/j.bcp.2025.116898] [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: 12/09/2024] [Revised: 03/21/2025] [Accepted: 03/24/2025] [Indexed: 03/29/2025]
Abstract
CD47 is a crucial anti-phagocytic signal in regulating macrophage responses and its manipulation offers the therapeutic potential in cancer treatment. However, in many cases, blockade of CD47 by itself is insufficient to activate macrophage effectively, indicating other unidentified phagocytosis-regulating factors to resist the macrophage activity. In this study, a genome-wide human CRISPR-Cas9 library was developed for comprehensive screening of phagocytosis-regulating factors in the context of CD47 blockade. The screening results identified GSTK1 as a potential anti-phagocytic signal counteracting the efficacy of CD47-based phagocytosis. The disruption of GSTK1 significantly increased the phagocytosis rate of cancer cells by macrophages in combination with anti-CD47 antibody. Further mechanism investigation unveiled that GSTK1 blockade increased the membrane exposure of calreticulin in different cancer cells, which might be the primary mechanism driving enhanced macrophage-mediated phagocytosis. To this end, siGSTK1-loaded nanoparticles (siGSTK1-LNPs) were designed to suppress the GSTK1 expression efficiently. The comparable phagocytosis efficacy was also observed when combining siGSTK1-LNPs with anti-CD47 antibody. Above all, GSTK1 blockade was identified as a promising and feasible stimulus for enhancing the effectiveness of anti-CD47 antibody, introducing a novel and effective combination approach in cancer immunotherapy.
Collapse
Affiliation(s)
- Wei-Bang Yu
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macau, China
| | - Zi-Han Ye
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macau, China
| | - Jia-Jie Shi
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macau, China
| | - Wei-Qing Deng
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macau, China
| | - Jun Chen
- Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Jin-Jian Lu
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macau, China; Department of Pharmaceutical Sciences, Faculty of Health Sciences, University of Macau, Macau, China; MoE Frontiers Science Center for Precision Oncology, University of Macau, Macau, China.
| |
Collapse
|
2
|
Zhang X, Ye XS. A review on the development of sialyltransferase inhibitors. Carbohydr Res 2025; 551:109427. [PMID: 39977976 DOI: 10.1016/j.carres.2025.109427] [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: 01/26/2025] [Revised: 02/12/2025] [Accepted: 02/12/2025] [Indexed: 02/22/2025]
Abstract
Sialylation of terminal glycoconjugates is involved in many important physiological and pathological processes such as tumor metastasis, drug resistance, organismal immunity, and viral infections. Sialyltransferases are enzymes responsible for sialylation modification in organisms, and potent sialyltransferase inhibitors can not only serve as probes for glycobiology studies, but also hold great promise to become agents for tumor therapy and viral infection control in the clinic. This review summarizes the latest progress in the development and application of various sialyltransferase inhibitors. The current challenges and development trends are also discussed.
Collapse
Affiliation(s)
- Xiang Zhang
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, and Chemical Biology Center, Peking University, Beijing, 100191, China
| | - Xin-Shan Ye
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, and Chemical Biology Center, Peking University, Beijing, 100191, China.
| |
Collapse
|
3
|
Yamaguchi SI, Takemura M, Miwa K, Morimoto N, Nakayama M. Siglec-14-Mediated Inflammatory Responses to Carbon Nanomaterials. ACS APPLIED BIO MATERIALS 2025; 8:2927-2937. [PMID: 40099920 DOI: 10.1021/acsabm.4c01736] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/20/2025]
Abstract
Carbon nanomaterials (CNM), including carbon nanotubes (CNT) and graphene nanoplatelets (GNP), are expected to have diverse industrial applications due to their unique physical properties. However, concerns have been raised regarding their toxicity in humans. In this context, risk assessment must include an understanding of the molecular mechanisms underlying human recognition of CNM. We have recently identified human sialic acid-binding immunoglobulin-like lectin (Siglec)-14 as a CNT-recognizing receptor. Since no rodent orthologs for Siglec-14 exist, previous rodent toxicological studies may underestimate CNM toxicity in humans. Therefore, in this study, we investigate Siglec-14 responses to various CNM. Siglec-14 recognizes various types of CNM via its extracellular aromatic cluster with a similar affinity, regardless of size and shape. Ultrathin single-walled CNT (SWCNT) and spherical carbon black nanoparticles (CBNP) activated macrophage Siglec-14 signaling, leading to IL-8 production. Notably, GNP as well as long needle-like MWCNT not only activate this inflammatory signal but also cause phagosomal damage, leading to the release of IL-1β, the most prominent pro-inflammatory cytokine. In mice transduced with Siglec-14, intratracheal injection of GNP or long needle-like MWCNT caused lung inflammation, whereas injection of SWCNT or CBNP did not. Taken together, these results suggest that CNM-induced inflammation requires two processes: macrophage receptor ligation and phagosomal damage. This indicates that CNM may be safe unless they cause damage to the macrophage phagosome.
Collapse
Affiliation(s)
- Shin-Ichiro Yamaguchi
- Laboratory of Immunology and Microbiology, College of Pharmaceutical Sciences, Ritsumeikan University, Kusatsu 525-8577, Japan
| | - Miki Takemura
- Laboratory of Immunology and Microbiology, College of Pharmaceutical Sciences, Ritsumeikan University, Kusatsu 525-8577, Japan
| | - Karen Miwa
- Laboratory of Immunology and Microbiology, College of Pharmaceutical Sciences, Ritsumeikan University, Kusatsu 525-8577, Japan
| | - Nobuyuki Morimoto
- Faculty of Materials for Energy, Shimane University, Matsue, Shimane 690-8504, Japan
| | - Masafumi Nakayama
- Laboratory of Immunology and Microbiology, College of Pharmaceutical Sciences, Ritsumeikan University, Kusatsu 525-8577, Japan
- Research Center for Animal Life Science, Shiga University of Medical Sciences, Otsu 525-0072, Japan
| |
Collapse
|
4
|
Hirane N, Yokoi Y, Kobayashi A, Yamada M, Konno T, Kumeta H, Wakui H, Otaki M, Miura Y, Kakisaka T, Kamiyama T, Taketomi A, Nouso K, Kimura S, Nishimura SI. Site-Specific O-Glycosylation in Oncofetal Fibronectin IIICS Domain Creates Cancer Stage-Specific Biomarkers. J Am Chem Soc 2025; 147:12170-12184. [PMID: 40162478 DOI: 10.1021/jacs.5c00333] [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: 04/02/2025]
Abstract
Despite the pathological importance of oncofetal fibronectin isoforms associated with cancer cell progression and metastasis, our understanding of the structural and molecular details that occurred in this abundant and ubiquitous extracellular matrix component remains largely unknown. Here, we communicate that site-specific O-glycosylation in the oncofetal fibronectin creates cancer stage-specific serum biomarkers for hepatocellular carcinoma (HCC) by quantitative MS-based glycoproteomic approach. Selective reaction monitoring (SRM) using a structure-defined synthetic glycopeptide library enabled absolute quantitation of the targeted label-free tryptic fragments bearing cancer-relevant O-glycans derived from the type III homology connective segment (IIICS) domain of the oncofetal fibronectin (onfFN) in the sera of HCC patients and healthy controls. We found site-specific O-glycoform alteration from T/sialyl T antigens to Tn/sialyl Tn antigens at the consecutive threonine residues 2155Thr-Thr-Ala2157 within this fragment during cancer progression. Surprisingly, this dynamic glycoform alteration is observed specifically in the patient sera diagnosed newly as stage 2-4 groups not in the normal control and stage 1 patient groups. Our results provide compelling evidence that site-specific glycoform changes observed in the onfFN IIICS domain during the tumor proliferation elaborate new class cancer stage-relevant "dynamic epitopes" as highly potential cancer diagnostic and therapeutic targets.
Collapse
Affiliation(s)
- Nozomi Hirane
- Field of Drug Discovery Research, Faculty of Advanced Life Science, and Graduate School of Life Science, Hokkaido University, N21, W11, Kita-ku, Sapporo 001-0021, Japan
- ENU Pharma, Inc. Satellite Laboratory, Hokkaido University, N21, W11, Kita-ku, Sapporo 001-0021, Japan
| | - Yasuhiro Yokoi
- Field of Drug Discovery Research, Faculty of Advanced Life Science, and Graduate School of Life Science, Hokkaido University, N21, W11, Kita-ku, Sapporo 001-0021, Japan
- ENU Pharma, Inc. Satellite Laboratory, Hokkaido University, N21, W11, Kita-ku, Sapporo 001-0021, Japan
| | - Aiko Kobayashi
- Field of Drug Discovery Research, Faculty of Advanced Life Science, and Graduate School of Life Science, Hokkaido University, N21, W11, Kita-ku, Sapporo 001-0021, Japan
| | - Misaki Yamada
- Field of Drug Discovery Research, Faculty of Advanced Life Science, and Graduate School of Life Science, Hokkaido University, N21, W11, Kita-ku, Sapporo 001-0021, Japan
| | - Taiki Konno
- Field of Drug Discovery Research, Faculty of Advanced Life Science, and Graduate School of Life Science, Hokkaido University, N21, W11, Kita-ku, Sapporo 001-0021, Japan
| | - Hiroyuki Kumeta
- Field of Drug Discovery Research, Faculty of Advanced Life Science, and Graduate School of Life Science, Hokkaido University, N21, W11, Kita-ku, Sapporo 001-0021, Japan
| | - Hajime Wakui
- Field of Drug Discovery Research, Faculty of Advanced Life Science, and Graduate School of Life Science, Hokkaido University, N21, W11, Kita-ku, Sapporo 001-0021, Japan
| | - Michiru Otaki
- Field of Drug Discovery Research, Faculty of Advanced Life Science, and Graduate School of Life Science, Hokkaido University, N21, W11, Kita-ku, Sapporo 001-0021, Japan
| | - Yoshiaki Miura
- ENU Pharma, Inc. Satellite Laboratory, Hokkaido University, N21, W11, Kita-ku, Sapporo 001-0021, Japan
| | - Tatsuhiko Kakisaka
- Department of Gastroenterological Surgery I, Graduate School of Medicine, Hokkaido University, N15, W7, Kita-ku, Sapporo 060-8638, Japan
| | - Toshiya Kamiyama
- Department of Gastroenterological Surgery I, Graduate School of Medicine, Hokkaido University, N15, W7, Kita-ku, Sapporo 060-8638, Japan
| | - Akinobu Taketomi
- Department of Gastroenterological Surgery I, Graduate School of Medicine, Hokkaido University, N15, W7, Kita-ku, Sapporo 060-8638, Japan
| | - Kazuhiro Nouso
- Department of Gastroenterology and Hepatology, Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, Okayama University, Okayama 700-8530, Japan
| | - Satoshi Kimura
- Department of Laboratory Medicine and Central Clinical Laboratory, Showa University, Northern Yokohama Hospital, Yokohama 224-8503, Japan
| | - Shin-Ichiro Nishimura
- Field of Drug Discovery Research, Faculty of Advanced Life Science, and Graduate School of Life Science, Hokkaido University, N21, W11, Kita-ku, Sapporo 001-0021, Japan
- ENU Pharma, Inc. Satellite Laboratory, Hokkaido University, N21, W11, Kita-ku, Sapporo 001-0021, Japan
| |
Collapse
|
5
|
Tong Y, Chen R, Lu X, Chen C, Sun G, Yu X, Lyu S, Feng M, Long Y, Gong L, Chen L. A nanobody-enzyme fusion protein targeting PD-L1 and sialic acid exerts anti-tumor effects by C-type lectin pathway-mediated tumor associated macrophages repolarizing. Int J Biol Macromol 2025; 298:139953. [PMID: 39824395 DOI: 10.1016/j.ijbiomac.2025.139953] [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: 09/12/2024] [Revised: 01/06/2025] [Accepted: 01/14/2025] [Indexed: 01/20/2025]
Abstract
Aberrant sialylated glycosylation in the tumor microenvironment is a novel immune suppression pathway, which has garnered significant attention as a targetable glycoimmune checkpoint for cancer immunotherapy to address the dilemma of existing therapies. However, rational drug design and in-depth mechanistic studies are urgently required for tumor sialic acid to become valuable glycoimmune targets. In this study, we explored the positive correlation of PD-L1 and sialyltransferase expression in clinical colorectal cancer tissues and identified their mutual regulation effects in macrophages. Subsequently, we characterized a new sialidase with excellent properties from human oral symbiotic bacteria and then developed a novel nanobody-enzyme fusion protein, designated as Nb16-Sia, to concurrently target the PD-L1 and sialic acid. Results from syngeneic colon tumor models reveal superior efficacy of Nb16-Sia over monotherapy and combinations, which could remodel the tumor immune microenvironment. Mechanistically, Nb16-Sia, which could repolarize macrophages from the tumor-promoting M2 to anti-tumor M1 phenotype via the C-type lectin pathway, exerted its antitumor efficacy mainly by regulating tumor-associated macrophages. Our strategy of nanobody-enzyme fusion protein effectively enables the delivery of sialidase, allows the collaboration between anti-PD-L1 nanobody and sialidase in combating tumors, and holds considerable promise for further development.
Collapse
Affiliation(s)
- Yongliang Tong
- Department of Medical Microbiology, Key Laboratory of Medical Molecular Virology of Ministries of Education and Health, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Runqiu Chen
- Shanghai Institute of Infectious Diseases and Biosecurity, Fudan University, Shanghai, China; Department of Microbiological and Biochemical Pharmacy, School of Pharmacy, Fudan University, Shanghai, China; State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
| | - Xinrong Lu
- Department of Medical Microbiology, Key Laboratory of Medical Molecular Virology of Ministries of Education and Health, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Cuiying Chen
- Department of Research and Development, Sysdiagno (Nanjing) Biotech Co., Ltd, Nanjing, Jiangsu Province, China
| | - Guiqin Sun
- School of Medical Technology and Information Engineering, Zhejiang Chinese Medical University, Hangzhou, Zhejiang Province, China
| | - Xiaolu Yu
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
| | - Shaoxian Lyu
- Department of Medical Microbiology, Key Laboratory of Medical Molecular Virology of Ministries of Education and Health, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Meiqing Feng
- Shanghai Institute of Infectious Diseases and Biosecurity, Fudan University, Shanghai, China; Department of Microbiological and Biochemical Pharmacy, School of Pharmacy, Fudan University, Shanghai, China.
| | - Yiru Long
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China.
| | - Likun Gong
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China.
| | - Li Chen
- Department of Medical Microbiology, Key Laboratory of Medical Molecular Virology of Ministries of Education and Health, School of Basic Medical Sciences, Fudan University, Shanghai, China; Translational Glycomics Research Center, Fudan Zhangjiang Institute, Shanghai, China.
| |
Collapse
|
6
|
Fernandez Alarcon J, Perez Schmidt P, Panini N, Caruso F, Violatto MB, Sukubo NG, Martinez‐Serra A, Ekalle‐Soppo CB, Morelli A, Moscatiello GY, Grasselli C, Corbelli A, Fiordaliso F, Kelk J, Petrosilli L, d'Orazio G, Mateu Ferrando R, Verdaguer Ferrer A, Fornaguera C, Lay L, Fumagalli S, Recchia S, Monopoli MP, Polito L, Bigini P, Sitia G. Functional Polarization of Liver Macrophages by Glyco Gold Nanoparticles. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2407458. [PMID: 39950558 PMCID: PMC12021048 DOI: 10.1002/advs.202407458] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/03/2024] [Revised: 09/24/2024] [Indexed: 04/26/2025]
Abstract
Macrophages are crucial drivers of innate immunity. Reprogramming macrophages to a restorative phenotype in cancer or autoimmune diseases can stop their cancer-promoting activity or trigger anti-inflammatory immunity. Glycans have emerged as key components for immunity as they are involved in many pathophysiological disorders. Previous studies have demonstrated that supraphysiological amounts of mannose (Man) or sialic acid (Sia) can inhibit tumor growth and stimulate differentiation of regulatory T cells. Man is known to affect glucose metabolism in glycolysis by competing for the same intracellular transporters and affecting macrophage polarization, whereas Sia alters macrophage differentiation via signaling through Siglec-1. Herein, this work describes a macrophage targeting platform using gold nanoparticles (GNPs) functionalized with Man and Sia monosaccharides which exhibit high liver tropism. A single dose of glyco-GNPs can convert macrophages to a restorative phenotype in two completely different immune environments. Man promotes tumor-associated macrophages toward an antitumorigenic activity in a MC38 liver colorectal cancer model by secretion of TNF-α, IL -1β, and IL -6 in the tumor microenvironment. However, in a proinflammatory environment, as observed in a mouse model of autoimmune disease, primary biliary cholangitis, Man impairs the production of TNF-α, IL-1β, Arg1, and IL-6 cytokines. The results probe the dual role of Man in macrophage repolarization in response to the immune system. This study is a proof-of-concept that demonstrates that nanomedicine using specific glycans designed to target other immune cells such as myeloid cells, are a promising strategy not only against cancer but also against other pathologies such as autoimmune diseases.
Collapse
Affiliation(s)
- Jennifer Fernandez Alarcon
- Department of Molecular Biochemistry and PharmacologyIstituto di Ricerche Farmacologiche Mario Negri IRCCSVia Mario Negri 2Milano20156Italy
- Grup d'Enginyeria de Materials (GEMAT)Institut Químic de Sarrià (IQS)Universitat Ramon Llull (URL)Via Augusta 390Barcelona08017Spain
| | - Patricia Perez Schmidt
- Istituto di Scienze e Tecnologie Chimiche “Giulio Natta”SCITEC‐CNR, Via G. Fantoli 16/15Milano20138Italy
| | - Nicolo Panini
- Department of OncologyIstituto di Ricerche Farmacologiche Mario Negri IRCCSVia Mario Negri 2Milano20156Italy
| | - Francesca Caruso
- Experimental Hepatology UnitDivision of Immunology, Transplantation and Infectious DiseasesIRCCS San Raffaele Scientific InstituteVia Olgettina 58Milano20132Italy
| | - Martina B. Violatto
- Department of Molecular Biochemistry and PharmacologyIstituto di Ricerche Farmacologiche Mario Negri IRCCSVia Mario Negri 2Milano20156Italy
| | - Naths Grazia Sukubo
- School of Medicine and SurgeryUniversity of Milano‐BicoccaPiazza dell'Ateneo Nuovo1Milano20126Italy
| | - Alberto Martinez‐Serra
- Department of ChemistryRoyal College of Surgeons of Ireland RCSISt Stephens Green 123DublinIreland
| | - Charlotte Blanche Ekalle‐Soppo
- Experimental Hepatology UnitDivision of Immunology, Transplantation and Infectious DiseasesIRCCS San Raffaele Scientific InstituteVia Olgettina 58Milano20132Italy
| | - Annalisa Morelli
- Department of Molecular Biochemistry and PharmacologyIstituto di Ricerche Farmacologiche Mario Negri IRCCSVia Mario Negri 2Milano20156Italy
| | - Giulia Yuri Moscatiello
- Department of Molecular Biochemistry and PharmacologyIstituto di Ricerche Farmacologiche Mario Negri IRCCSVia Mario Negri 2Milano20156Italy
| | - Chiara Grasselli
- Department of OncologyIstituto di Ricerche Farmacologiche Mario Negri IRCCSVia Mario Negri 2Milano20156Italy
| | - Alessandro Corbelli
- Department of Molecular Biochemistry and PharmacologyIstituto di Ricerche Farmacologiche Mario Negri IRCCSVia Mario Negri 2Milano20156Italy
| | - Fabio Fiordaliso
- Department of Molecular Biochemistry and PharmacologyIstituto di Ricerche Farmacologiche Mario Negri IRCCSVia Mario Negri 2Milano20156Italy
| | - Joe Kelk
- Department of NeurosciencesIstituto di Ricerche Farmacologiche Mario Negri IRCCSVia Mario Negri 2Milano20156Italy
| | - Laura Petrosilli
- Department of Organic ChemistryUniversity degli Studi di MilanoVia Golgi 19Milano20133Italy
| | - Giuseppe d'Orazio
- Department of Organic ChemistryUniversity degli Studi di MilanoVia Golgi 19Milano20133Italy
| | - Ruth Mateu Ferrando
- Department of Organic ChemistryUniversity degli Studi di MilanoVia Golgi 19Milano20133Italy
| | - Ariadna Verdaguer Ferrer
- Department of Analytical and Applied ChemistryInstitut Químic de Sarrià (IQS)Universitat Ramon Llull (URL)Via Augusta 390Barcelona08017Spain
| | - Cristina Fornaguera
- Grup d'Enginyeria de Materials (GEMAT)Institut Químic de Sarrià (IQS)Universitat Ramon Llull (URL)Via Augusta 390Barcelona08017Spain
| | - Luigi Lay
- Department of Organic ChemistryUniversity degli Studi di MilanoVia Golgi 19Milano20133Italy
| | - Stefano Fumagalli
- Department of NeurosciencesIstituto di Ricerche Farmacologiche Mario Negri IRCCSVia Mario Negri 2Milano20156Italy
| | - Sandro Recchia
- Department of Science and High TechnologyUniversity of InsubriaVia Valleggio 11Como22100Italy
| | - Marco P. Monopoli
- Department of ChemistryRoyal College of Surgeons of Ireland RCSISt Stephens Green 123DublinIreland
| | - Laura Polito
- Istituto di Scienze e Tecnologie Chimiche “Giulio Natta”SCITEC‐CNR, Via G. Fantoli 16/15Milano20138Italy
| | - Paolo Bigini
- Department of Molecular Biochemistry and PharmacologyIstituto di Ricerche Farmacologiche Mario Negri IRCCSVia Mario Negri 2Milano20156Italy
| | - Giovanni Sitia
- Experimental Hepatology UnitDivision of Immunology, Transplantation and Infectious DiseasesIRCCS San Raffaele Scientific InstituteVia Olgettina 58Milano20132Italy
| |
Collapse
|
7
|
Abreu C, Di Carluccio C, Ječmen T, Skořepa O, Bláha J, Marchetti R, Silipo A, Vaněk O. Insights into stability, dimerisation, and ligand binding properties of Siglec-7: Isotope labelling in HEK293 cells for protein characterisation by NMR spectroscopy. Int J Biol Macromol 2025; 309:142672. [PMID: 40164254 DOI: 10.1016/j.ijbiomac.2025.142672] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2024] [Revised: 03/27/2025] [Accepted: 03/28/2025] [Indexed: 04/02/2025]
Abstract
Siglec-7, an immune checkpoint receptor, has emerged as a promising target for cancer immunotherapy due to its involvement in the regulation of immune and inflammatory responses. However, while its participation in immunoediting and immune evasion is well established, understanding its biological context, relevant ligands, and associated signalling pathways remains limited. Understanding these aspects is crucial for the development of effective immunotherapies targeting Siglec-7. In this study, three expression constructs of Siglec-7 were designed, expressed, and characterised, including an analysis of the oligomeric state of its extracellular domain. The N-terminal V-set Ig carbohydrate recognition domain was also produced in an isotopically double-labelled (13C,15N) mammalian cell growth medium. Two stable constructs suitable for biophysical and structural studies were identified. These findings reveal the noncovalent dimerisation of Siglec-7, offering new insights into its possible ligand interactions, signal transduction mechanisms, or receptor/ligand clustering. The dimerisation of Siglec-7 may be essential to achieve multivalent, high-avidity interactions with glycoconjugates, which may result in enhanced or alternative signalling processes within the NK cell immune synapse. In addition, a detailed protocol for generating double-labelled Siglec-7 in HEK293 cells, which may apply to other proteins under similar conditions, was described. These findings contribute to a better understanding of the biophysical and structural properties of Siglec-7 and are key to the design of more precise and effective cancer immunotherapies targeting Siglec-7.
Collapse
Affiliation(s)
- Celeste Abreu
- Department of Biochemistry, Faculty of Science, Charles University, Hlavova 2030, 12840 Prague, Czech Republic
| | - Cristina Di Carluccio
- Department of Chemical Sciences, University of Naples Federico II, Via Cintia 4, 80126 Napoli, Italy; CEINGE-Biotecnologie Avanzate Franco Salvatore, Via Gaetano Salvatore 486, 80145 Napoli, Italy
| | - Tomáš Ječmen
- Department of Biochemistry, Faculty of Science, Charles University, Hlavova 2030, 12840 Prague, Czech Republic
| | - Ondřej Skořepa
- Department of Biochemistry, Faculty of Science, Charles University, Hlavova 2030, 12840 Prague, Czech Republic
| | - Jan Bláha
- EMBL, Hamburg Unit c/o DESY, Notkestrasse 85, 22607 Hamburg, Germany
| | - Roberta Marchetti
- Department of Chemical Sciences, University of Naples Federico II, Via Cintia 4, 80126 Napoli, Italy
| | - Alba Silipo
- Department of Chemical Sciences, University of Naples Federico II, Via Cintia 4, 80126 Napoli, Italy; CEINGE-Biotecnologie Avanzate Franco Salvatore, Via Gaetano Salvatore 486, 80145 Napoli, Italy; Department of Chemistry, School of Science, Osaka University, 1-1 Machikaneyama, Toyonaka, 560-0043 Osaka, Japan
| | - Ondřej Vaněk
- Department of Biochemistry, Faculty of Science, Charles University, Hlavova 2030, 12840 Prague, Czech Republic.
| |
Collapse
|
8
|
Ye H, Liao W, Pan J, Shi Y, Wang Q. Immune checkpoint blockade for cancer therapy: current progress and perspectives. J Zhejiang Univ Sci B 2025; 26:203-226. [PMID: 40082201 PMCID: PMC11906392 DOI: 10.1631/jzus.b2300492] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/16/2025]
Abstract
Dysfunction of anti-tumor immune responses is crucial for cancer progression. Immune checkpoint blockade (ICB), which can potentiate T cell responses, is an effective strategy for the normalization of host anti-tumor immunity. In recent years, immune checkpoints, expressed on both tumor cells and immune cells, have been identified; some of them have exhibited potential druggability and have been approved by the US Food and Drug Administration (FDA) for clinical treatment. However, limited responses and immune-related adverse events (irAEs) cannot be ignored. This review outlines the development and applications of ICBs, potential strategies for overcoming resistance, and future directions for ICB-based cancer immunotherapy.
Collapse
Affiliation(s)
- Hongying Ye
- Institute of Immunology, Zhejiang University School of Medicine, Hangzhou 310058, China
- The Key Laboratory for Immunity and Inflammatory Diseases of Zhejiang Province, Hangzhou 310058, China
| | - Weijie Liao
- Institute of Immunology, Zhejiang University School of Medicine, Hangzhou 310058, China
- The Key Laboratory for Immunity and Inflammatory Diseases of Zhejiang Province, Hangzhou 310058, China
| | - Jiongli Pan
- Institute of Immunology, Zhejiang University School of Medicine, Hangzhou 310058, China
- The Key Laboratory for Immunity and Inflammatory Diseases of Zhejiang Province, Hangzhou 310058, China
| | - Yin Shi
- Department of Biochemistry, Zhejiang University School of Medicine, Hangzhou 310058, China.
| | - Qingqing Wang
- Institute of Immunology, Zhejiang University School of Medicine, Hangzhou 310058, China.
- The Key Laboratory for Immunity and Inflammatory Diseases of Zhejiang Province, Hangzhou 310058, China.
| |
Collapse
|
9
|
Haubner S, Subklewe M, Sadelain M. Honing CAR T cells to tackle acute myeloid leukemia. Blood 2025; 145:1113-1125. [PMID: 39630061 DOI: 10.1182/blood.2024024063] [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: 09/18/2024] [Accepted: 11/15/2024] [Indexed: 03/14/2025] Open
Abstract
ABSTRACT Acute myeloid leukemia (AML) remains a dismal disease with poor prognosis, particularly in the relapsed/refractory (R/R) setting. Chimeric antigen receptor (CAR) therapy has yielded remarkable clinical results in other leukemias and thus has, in principle, the potential to achieve similar outcomes in R/R AML. Redirecting the approved CD19-specific CAR designs against the myeloid antigens CD33, CD123, or CLEC12A has occasionally yielded morphologic leukemia-free states but has so far been marred by threatening myeloablation and early relapses. These safety and efficacy limitations are largely due to the challenge of identifying suitable target antigens and designing adequate receptors for effective recognition and safe elimination of AML. Building on lessons learned from the initial clinical attempts, a new wave of CAR strategies relying on alternative target antigens and innovative CAR designs is about to enter clinical evaluation. Adapted multiantigen targeting, logic gating, and emerging cell engineering solutions offer new possibilities to better direct T-cell specificity and sensitivity toward AML. Pharmacologic modulation and genetic epitope engineering may extend these approaches by augmenting target expression in AML cells or minimizing target expression in normal hematopoietic cells. On/off switches or CAR T-cell depletion may curb excessive or deleterious CAR activity. Investigation of AML-intrinsic resistance and leukemic microenvironmental factors is poised to reveal additional targetable AML vulnerabilities. We summarize here the findings, challenges, and new developments of CAR therapy for AML. These illustrate the need to specifically adapt CAR strategies to the complex biology of AML to achieve better therapeutic outcomes.
Collapse
Affiliation(s)
- Sascha Haubner
- Columbia Initiative in Cell Engineering and Therapy, Department of Medicine, Columbia University, New York, NY
| | - Marion Subklewe
- Department of Medicine III, University Hospital, Ludwig Maximilian University Munich, Munich, Germany
| | - Michel Sadelain
- Columbia Initiative in Cell Engineering and Therapy, Department of Medicine, Columbia University, New York, NY
| |
Collapse
|
10
|
Wang Q, He J, Lei T, Li X, Yue S, Liu C, Hu Q. New insights into cancer immune checkpoints landscape from single-cell RNA sequencing. Biochim Biophys Acta Rev Cancer 2025; 1880:189298. [PMID: 40088992 DOI: 10.1016/j.bbcan.2025.189298] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2025] [Revised: 03/07/2025] [Accepted: 03/07/2025] [Indexed: 03/17/2025]
Abstract
Immune checkpoint blockade (ICB) therapy represents a pivotal advancement in tumor immunotherapy by restoring the cytotoxic lymphocytes' anti-tumor activity through the modulation of immune checkpoint functions. Nevertheless, many patients experience suboptimal therapeutic outcomes, likely due to the immunosuppressive tumor microenvironment, drug resistance, and other factors. Single-cell RNA sequencing has assisted to precisely investigate the immune infiltration patterns before and after ICB treatment, enabling a high-resolution depiction of previously unrecognized functional interaction among immune checkpoints. This review addresses the heterogeneity between tumor microenvironments that respond to or resist ICB therapy, highlighting critical factors underlying the variation in immunotherapy efficacy and elucidating treatment failure. Furthermore, a comprehensive examination is provided of how specific ICBs modulate immune and tumor cells to achieve anti-tumor effects and generate treatment resistance, alongside a summary of emerging immune checkpoints identified as promising targets for cancer immunotherapy through single-cell RNA sequencing applications.
Collapse
Affiliation(s)
- Qian Wang
- Department of Oncology, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Jiahui He
- Department of Oncology, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Tianyu Lei
- Department of Oncology, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Xiaohui Li
- Department of Radiation Oncology, Peking University First Hospital, Beijing 100034, China
| | - Shengqin Yue
- Department of Oncology, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Chao Liu
- Department of Radiation Oncology, Peking University First Hospital, Beijing 100034, China.
| | - Qinyong Hu
- Department of Oncology, Renmin Hospital of Wuhan University, Wuhan 430060, China; Renmin Hospital of Wuhan Economic and Technological Development Zone (Hannan), Wuhan 430090, China.
| |
Collapse
|
11
|
Belbasis L, Morris S, van Duijn C, Bennett D, Walters R. Mendelian randomization identifies proteins involved in neurodegenerative diseases. Brain 2025:awaf018. [PMID: 40037332 DOI: 10.1093/brain/awaf018] [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: 02/24/2024] [Revised: 10/26/2024] [Accepted: 12/20/2024] [Indexed: 03/06/2025] Open
Abstract
Proteins are involved in multiple biological functions. High-throughput technologies have allowed the measurement of thousands of proteins in population biobanks. In this study, we aimed to identify proteins related to Alzheimer's disease, Parkinson's disease, multiple sclerosis and amyotrophic lateral sclerosis by leveraging large-scale genetic and proteomic data. We performed a two-sample cis Mendelian randomization study by selecting instrumental variables for the abundance of >2700 proteins measured by either Olink or SomaScan platforms in plasma from the UK Biobank and the deCODE Health Study. We also used the latest publicly available genome-wide association studies for the neurodegenerative diseases of interest. The potentially causal effect of proteins on neurodegenerative diseases was estimated based on the Wald ratio. We tested 13 377 protein-disease associations, identifying 169 associations that were statistically significant (5% false discovery rate). Evidence of co-localization between plasma protein abundance and disease risk (posterior probability > 0.80) was identified for 61 protein-disease pairs, leading to 50 unique protein-disease associations. Notably, 23 of 50 protein-disease associations corresponded to genetic loci not previously reported by genome-wide association studies. The two-sample Mendelian randomization and co-localization analysis also showed that APOE abundance in plasma was associated with three subcortical volumes (hippocampus, amygdala and nucleus accumbens) and white matter hyper-intensities, whereas PILRA and PILRB abundance in plasma was associated with caudate nucleus volume. Our study provided a comprehensive assessment of the effect of the human proteome that is currently measurable through two different platforms on neurodegenerative diseases. The newly associated proteins indicated the involvement of complement (C1S and C1R), microglia (SIRPA, SIGLEC9 and PRSS8) and lysosomes (CLN5) in Alzheimer's disease; the interleukin-6 pathway (CTF1) in Parkinson's disease; lysosomes (TPP1), blood-brain barrier integrity (MFAP2) and astrocytes (TNFSF13) in amyotrophic lateral sclerosis; and blood-brain barrier integrity (VEGFB), oligodendrocytes (PARP1), node of Ranvier and dorsal root ganglion (NCS1, FLRT3 and CDH15) and the innate immune system (CR1, AHSG and WARS) in multiple sclerosis. Our study demonstrates how harnessing large-scale genomic and proteomic data can yield new insights into the role of the plasma proteome in the pathogenesis of neurodegenerative diseases.
Collapse
Affiliation(s)
- Lazaros Belbasis
- Nuffield Department of Population Health, University of Oxford, Oxford OX3 7LF, UK
| | - Sam Morris
- Nuffield Department of Population Health, University of Oxford, Oxford OX3 7LF, UK
| | - Cornelia van Duijn
- Nuffield Department of Population Health, University of Oxford, Oxford OX3 7LF, UK
| | - Derrick Bennett
- Nuffield Department of Population Health, University of Oxford, Oxford OX3 7LF, UK
| | - Robin Walters
- Nuffield Department of Population Health, University of Oxford, Oxford OX3 7LF, UK
| |
Collapse
|
12
|
Rodriguez E. Tumor Glycosylation: A Main Player in the Modulation of Immune Responses. Eur J Immunol 2025; 55:e202451318. [PMID: 40071681 PMCID: PMC11898543 DOI: 10.1002/eji.202451318] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2024] [Revised: 02/13/2025] [Accepted: 02/17/2025] [Indexed: 03/15/2025]
Abstract
Tumor immune escape refers to the process by which cancer cells evade detection and destruction by the immune system. Glycosylation, a post-translational modification that is altered in almost all cancer types, plays a crucial role in this process by modulating immune responses. This review examines our current understanding of how aberrant tumor glycosylation contributes to a tolerogenic microenvironment, focusing on specific glycosylation signatures-fucosylation, truncated O-glycans, and sialylation-and the immune receptors involved. Additionally, the clinical significance of tumor glycosylation is discussed, emphasizing its potential in developing novel therapeutic approaches aimed at improving immune system recognition and targeting of cancer cells. The review underscores the importance of ongoing research in this area to identify effective strategies for countering tumor immune escape and enhancing the efficacy of cancer treatments.
Collapse
Affiliation(s)
- Ernesto Rodriguez
- Amsterdam UMC location Vrije Universiteit AmsterdamMolecular Cell Biology and ImmunologyAmsterdamThe Netherlands
- Cancer Center AmsterdamCancer Biology and ImmunologyAmsterdamThe Netherlands
- Amsterdam Institute for Infection and ImmunityCancer ImmunologyAmsterdamThe Netherlands
| |
Collapse
|
13
|
Wu Y, You Y, Jiang T, He Y, Fan Q, Zeng X, Li T, Lu Y, Qi L, Zhou F, Sun L, Wang D, Zou Y, Zhang G, Yuan Y, Mao Y. Proximity Labeling and Genetic Screening Reveal that DSG2 is a Counter Receptor of Siglec-9 and Suppresses Macrophage Phagocytosis. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2406654. [PMID: 39813162 PMCID: PMC11884560 DOI: 10.1002/advs.202406654] [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: 06/16/2024] [Revised: 12/23/2024] [Indexed: 01/18/2025]
Abstract
Cancer cells present sialylated glycoconjugates that modulate the activity of various immune cells within the tumor microenvironment through trans interaction with immunosuppressive Siglec receptors. Identifying counter receptors for Siglecs can provide valuable targets for cancer immunotherapy, but it presents significant challenges. Here, the identification of DSG2 (Desmoglein 2) as a dominant counter receptor of Siglec-9 in melanoma cells is reported, using a workflow that combines the strength of proximity labeling and the advantage of CRISPR knockout screening. It is further demonstrated that the interaction between DSG2 and Siglec-9 is mainly dependent on sialic acid-bearing N-glycans on DSG2. Importantly, blocking trans interaction between DSG2 and Siglec-9 significantly enhances macrophage phagocytosis of melanoma cells and, to a lesser extent, other cancer cells. The work thus suggests sialylated DSG2 as a potential "don't eat me" signal molecule with therapeutic potentials in cancer immunotherapy.
Collapse
Affiliation(s)
- Ying Wu
- School of Pharmaceutical SciencesSun Yat‐sen UniversityGuangzhou510006China
| | - Yuyu You
- School of Pharmaceutical SciencesSun Yat‐sen UniversityGuangzhou510006China
| | - Tingsong Jiang
- School of Pharmaceutical SciencesSun Yat‐sen UniversityGuangzhou510006China
| | - Yuqi He
- School of Pharmaceutical SciencesSun Yat‐sen UniversityGuangzhou510006China
| | - Qingchi Fan
- School of Pharmaceutical SciencesSun Yat‐sen UniversityGuangzhou510006China
| | - Xinlei Zeng
- School of Pharmaceutical SciencesSun Yat‐sen UniversityGuangzhou510006China
| | - Ting Li
- School of Pharmaceutical SciencesSun Yat‐sen UniversityGuangzhou510006China
| | - Yuxing Lu
- School of Pharmaceutical SciencesSun Yat‐sen UniversityGuangzhou510006China
| | - Liang Qi
- School of Pharmaceutical SciencesSun Yat‐sen UniversityGuangzhou510006China
| | - Fengxia Zhou
- School of Pharmaceutical SciencesSun Yat‐sen UniversityGuangzhou510006China
| | - Lingyu Sun
- School of Pharmaceutical SciencesSun Yat‐sen UniversityGuangzhou510006China
| | - Danyang Wang
- State Key Laboratory of Oncology in South ChinaCollaborative Innovation Center for Cancer MedicineSun Yat‐sen University Cancer CenterGuangzhou510275China
| | - Yong Zou
- Department of Blood TransfusionThe Third Affiliated Hospital of Sun Yat‐sen UniversityGuangzhou510000China
| | - Guigen Zhang
- Institute of Human VirologyDepartment of Pathogen Biology and Biosecurityand Key Laboratory of Tropical Disease Control of Ministry of EducationZhongshan School of MedicineSun Yat‐sen UniversityGuangzhou510080China
| | - Yanqiu Yuan
- School of Pharmaceutical SciencesSun Yat‐sen UniversityGuangzhou510006China
- State Key Laboratory of Anti‐Infective Drug Discovery and DevelopmentSchool of Pharmaceutical SciencesSun Yat‐sen UniversityGuangzhou510006China
| | - Yang Mao
- School of Pharmaceutical SciencesSun Yat‐sen UniversityGuangzhou510006China
- Guangdong Provincial Key Laboratory of Drug Non‐Clinical Evaluation and ResearchGuangzhou510006China
| |
Collapse
|
14
|
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
|
15
|
Singh S, Islam SMS, Liu R, Adeniji OS, Giron LB, Saini P, Danesh A, Denton PW, Jones B, Xiao H, Abdel-Mohsen M. HIV-Induced Sialoglycans on Infected Cells Promote Immune Evasion from Myeloid Cell-Mediated Killing. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.02.21.639489. [PMID: 40060686 PMCID: PMC11888171 DOI: 10.1101/2025.02.21.639489] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 03/18/2025]
Abstract
Sialic acid-containing glycans (sialoglycans) on pathological cells interact with Siglecs, glycol-immune checkpoint receptors expressed on myeloid cells such as monocytes and neutrophils. This interaction suppresses the cytotoxic functions of these immune cells. We show that HIV infection reprograms the glycosylation machinery of infected cells to increase the expression of specific sialoglycan ligands for Siglecs-3, -7, and -9. These ligands engage Siglecs on myeloid cells, impairing their ability to target HIV-infected cells. Selective disruption of these interactions using 10-1074-Sia, an HIV-specific antibody conjugated to sialidase-an enzyme that removes sialic acids-significantly enhances monocyte- and neutrophil-mediated killing of HIV-infected cells in autologous assays. Treatment with 10-1074-Sia in humanized mice infected with HIV reduces viral load and decreases inflammation. These findings reveal a novel immune evasion mechanism exploited by HIV to evade myeloid cell immune surveillance and highlight the potential of targeting sialoglycan-Siglec interactions to improve immune clearance of HIV-infected cells.
Collapse
Affiliation(s)
| | | | - Rui Liu
- Rice University, Houston, TX, USA
| | | | | | | | - Ali Danesh
- Weill Cornell Medicine, New York, NY, USA
| | | | - Brad Jones
- Weill Cornell Medicine, New York, NY, USA
| | - Han Xiao
- Rice University, Houston, TX, USA
| | | |
Collapse
|
16
|
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
|
17
|
Bertuzzi S, Lete MG, Franconetti A, Diercks T, Delgado S, Oyenarte I, Moure MJ, Nuñez‐Franco R, Valverde P, Lenza MP, Sobczak K, Jiménez‐Osés G, Paulson JC, Ardá A, Ereño‐Orbea J, Jiménez‐Barbero J. Exploring Glycan-Lectin Interactions in Natural-Like Environments: A View Using NMR Experiments Inside Cell and on Cell Surface. Chemistry 2025; 31:e202403102. [PMID: 39588609 PMCID: PMC11833217 DOI: 10.1002/chem.202403102] [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: 11/20/2024] [Accepted: 11/25/2024] [Indexed: 11/27/2024]
Abstract
Glycan-mediated molecular recognition events are essential for life. NMR is widely used to monitor glycan binding to lectins in solution using isolated glycans and lectins. In this context, we herein explore diverse NMR methodologies, from both the receptor and ligand perspectives, to monitor glycan-lectin interactions under experimental conditions mimicking the native milieu inside cells and on cell surface. For the NMR experiments inside cells, galectin-7 is employed as model, since most galectins are soluble and carry out their functions in the cellular micro-environment. Using Danio Rerio oocytes, the 1H-15N HMQC NMR spectrum of a folded galectin has been observed inside cell for the first time, using a glycomimetic ligand (TDG) to overcoming the natural tendency of galectins to bind to numerous galactose-containing receptors within cells. Alternatively, most lectins, other than galectins, are displayed on the cell surface, providing a multivalent presentation to bind their glycan partners in cis (at the same cell) or in trans (on other cells). In this case, ligand-based STD-NMR experiments have been successfully applied to account for the interactions of natural glycans and glycomimetics with Siglec-10. These methodologies provide the proof-of-concept to open the door to the NMR analysis of the recognition of glycans in native-like settings.
Collapse
Affiliation(s)
- Sara Bertuzzi
- Center for Cooperative Research in Biosciences (CIC bioGUNE)Basque Research and Technology Alliance (BRTA)48160Derio, BizkaiaSpain
| | - Marta G. Lete
- Center for Cooperative Research in Biosciences (CIC bioGUNE)Basque Research and Technology Alliance (BRTA)48160Derio, BizkaiaSpain
| | - Antonio Franconetti
- Center for Cooperative Research in Biosciences (CIC bioGUNE)Basque Research and Technology Alliance (BRTA)48160Derio, BizkaiaSpain
| | - Tammo Diercks
- Center for Cooperative Research in Biosciences (CIC bioGUNE)Basque Research and Technology Alliance (BRTA)48160Derio, BizkaiaSpain
| | - Sandra Delgado
- Center for Cooperative Research in Biosciences (CIC bioGUNE)Basque Research and Technology Alliance (BRTA)48160Derio, BizkaiaSpain
| | - Iker Oyenarte
- Center for Cooperative Research in Biosciences (CIC bioGUNE)Basque Research and Technology Alliance (BRTA)48160Derio, BizkaiaSpain
| | - Maria J. Moure
- Center for Cooperative Research in Biosciences (CIC bioGUNE)Basque Research and Technology Alliance (BRTA)48160Derio, BizkaiaSpain
| | - Reyes Nuñez‐Franco
- Center for Cooperative Research in Biosciences (CIC bioGUNE)Basque Research and Technology Alliance (BRTA)48160Derio, BizkaiaSpain
| | - Pablo Valverde
- Center for Cooperative Research in Biosciences (CIC bioGUNE)Basque Research and Technology Alliance (BRTA)48160Derio, BizkaiaSpain
| | - Maria Pia Lenza
- Center for Cooperative Research in Biosciences (CIC bioGUNE)Basque Research and Technology Alliance (BRTA)48160Derio, BizkaiaSpain
| | - Klaudia Sobczak
- Center for Cooperative Research in Biosciences (CIC bioGUNE)Basque Research and Technology Alliance (BRTA)48160Derio, BizkaiaSpain
| | - Gonzalo Jiménez‐Osés
- Center for Cooperative Research in Biosciences (CIC bioGUNE)Basque Research and Technology Alliance (BRTA)48160Derio, BizkaiaSpain
| | - James C. Paulson
- Departments of Molecular Medicine and Immunology & MicrobiologyThe Scripps Research Institute10550 North Torrey Pines RoadLa Jolla, California92037USA
| | - Ana Ardá
- Center for Cooperative Research in Biosciences (CIC bioGUNE)Basque Research and Technology Alliance (BRTA)48160Derio, BizkaiaSpain
- Ikerbasque, Basque Foundation for ScienceBilbaoSpain
| | - June Ereño‐Orbea
- Center for Cooperative Research in Biosciences (CIC bioGUNE)Basque Research and Technology Alliance (BRTA)48160Derio, BizkaiaSpain
- Ikerbasque, Basque Foundation for ScienceBilbaoSpain
| | - Jesús Jiménez‐Barbero
- Center for Cooperative Research in Biosciences (CIC bioGUNE)Basque Research and Technology Alliance (BRTA)48160Derio, BizkaiaSpain
- Ikerbasque, Basque Foundation for ScienceBilbaoSpain
- Department of Organic & Inorganic ChemistryFaculty of Science and TechnologyUniversity of the Basque Country, EHU-UPV48940Leioa, BizkaiaSpain
- Centro de Investigacion Biomedica En Red de Enfermedades Respiratorias28029MadridSpain
| |
Collapse
|
18
|
Krishnamoorthy V, Daly J, Kim J, Piatnitca L, Yuen KA, Kumar B, Taherzadeh Ghahfarrokhi M, Bui TQT, Azadi P, Vu LP, Wisnovsky S. The glycosyltransferase ST3GAL4 drives immune evasion in acute myeloid leukemia by synthesizing ligands for the glyco-immune checkpoint receptor Siglec-9. Leukemia 2025; 39:346-359. [PMID: 39551873 PMCID: PMC11794148 DOI: 10.1038/s41375-024-02454-w] [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: 04/30/2024] [Revised: 10/18/2024] [Accepted: 10/25/2024] [Indexed: 11/19/2024]
Abstract
Immunotherapy has demonstrated promise as a treatment for acute myeloid leukemia (AML). However, there is still an urgent need to identify new molecules that inhibit the immune response to AML. Most prior research in this area has focused on protein-protein interaction interfaces. While carbohydrates also regulate immune recognition, the role of cell-surface glycans in driving AML immune evasion is comparatively understudied. The Siglecs, for example, are an important family of inhibitory, glycan-binding signaling receptors that have emerged as prime targets for cancer immunotherapy in recent years. In this study, we find that AML cells express ligands for the receptor Siglec-9 at high levels. Integrated CRISPR genomic screening and clinical bioinformatic analysis identified ST3GAL4 as a potential driver of Siglec-9 ligand expression in AML. Depletion of ST3GAL4 by CRISPR-Cas9 knockout (KO) dramatically reduced the expression of Siglec-9 ligands in AML cells. Mass spectrometry analysis of cell-surface glycosylation in ST3GAL4 KO cells revealed that Siglec-9 primarily binds N-linked sialoglycans on these cell types. Finally, we found that ST3GAL4 KO enhanced the sensitivity of AML cells to phagocytosis by Siglec-9-expressing macrophages. This work reveals a novel axis of immune evasion and implicates ST3GAL4 as a possible target for immunotherapy in AML.
Collapse
MESH Headings
- Humans
- Leukemia, Myeloid, Acute/immunology
- Leukemia, Myeloid, Acute/pathology
- Leukemia, Myeloid, Acute/metabolism
- Leukemia, Myeloid, Acute/genetics
- Sialyltransferases/metabolism
- Sialyltransferases/genetics
- Sialic Acid Binding Immunoglobulin-like Lectins/metabolism
- Ligands
- Immune Evasion
- Antigens, CD/metabolism
- beta-Galactoside alpha-2,3-Sialyltransferase
- Glycosylation
- Cell Line, Tumor
- CRISPR-Cas Systems
Collapse
Affiliation(s)
- Vignesh Krishnamoorthy
- Faculty of Pharmaceutical Sciences, University of British Columbia, Vancouver, BC, Canada
| | - John Daly
- Faculty of Pharmaceutical Sciences, University of British Columbia, Vancouver, BC, Canada
| | - Jimmy Kim
- Faculty of Pharmaceutical Sciences, University of British Columbia, Vancouver, BC, Canada
| | - Lidia Piatnitca
- Faculty of Pharmaceutical Sciences, University of British Columbia, Vancouver, BC, Canada
| | - Katie A Yuen
- Terry Fox Laboratory, British Columbia Cancer Research Institute, Vancouver, BC, Canada
| | - Bhoj Kumar
- Complex Carbohydrate Research Center, University of Georgia, Athens, GA, USA
| | | | - Tom Q T Bui
- Faculty of Pharmaceutical Sciences, University of British Columbia, Vancouver, BC, Canada
- Terry Fox Laboratory, British Columbia Cancer Research Institute, Vancouver, BC, Canada
| | - Parastoo Azadi
- Complex Carbohydrate Research Center, University of Georgia, Athens, GA, USA
| | - Ly P Vu
- Faculty of Pharmaceutical Sciences, University of British Columbia, Vancouver, BC, Canada
- Terry Fox Laboratory, British Columbia Cancer Research Institute, Vancouver, BC, Canada
| | - Simon Wisnovsky
- Faculty of Pharmaceutical Sciences, University of British Columbia, Vancouver, BC, Canada.
| |
Collapse
|
19
|
Liu Q, Xing H, Xiong M, Zhang XB. Specifically Editing Cancer Sialoglycans for Enhanced In Vivo Immunotherapy through Aptamer-Enzyme Chimeras. Angew Chem Int Ed Engl 2025; 64:e202414327. [PMID: 39324841 DOI: 10.1002/anie.202414327] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2024] [Revised: 09/26/2024] [Accepted: 09/26/2024] [Indexed: 09/27/2024]
Abstract
Immune checkpoint blockade (ICB) therapies have demonstrated remarkable clinical success in treating cancer. However, their objective response rate remains suboptimal because current therapies rely on limited immune checkpoints that fail to cover the multiple immune evasion pathways of cancer. To explore potential ICB strategies, we propose a glycoimmune checkpoint elimination (glycoICE) therapy based on targeted editing of sialoglycans on the tumor cell surface using an aptamer-enzyme chimera (ApEC). The ApEC can be readily generated via a one-step bioorthogonal procedure, allowing for large-scale and uniform production. It specifically targets and desialylates cancer cells, disrupting the sialoglycan-Siglec axis to activate immune cells and enhance immunotherapy efficacy, while its high tumor selectivity minimizes side effects from indiscriminate desialylation of normal tissues. Furthermore, the ApEC has the potential to be a versatile platform for specific editing of sialoglycans in different tumor models by adjusting the aptamer sequences to target specific protein markers. This research not only introduces a novel molecular tool for the effective editing of sialoglycans in complex environments, but also provides valuable insights for advancing DNA-based drugs towards in vivo and clinical applications.
Collapse
Affiliation(s)
- Qin Liu
- Molecular Science and Biomedicine Laboratory, State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, Hunan University, 410082, Changsha, Hunan, P. R. China
| | - Hang Xing
- Institute of Chemical Biology and Nanomedicine, State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, Hunan University, 410082, Changsha, Hunan, P. R. China
| | - Mengyi Xiong
- Molecular Science and Biomedicine Laboratory, State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, Hunan University, 410082, Changsha, Hunan, P. R. China
| | - Xiao-Bing Zhang
- Molecular Science and Biomedicine Laboratory, State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, Hunan University, 410082, Changsha, Hunan, P. R. China
| |
Collapse
|
20
|
Yin J, Lu Y, Liu Y, Shi Q, Shi M, Zhu Z, Fu D, Wang Z, Li C. SIGLEC11 promotes M2 macrophage polarization through AKT-mTOR signaling and facilitates the progression of gastric cancer. J Immunother Cancer 2025; 13:e010162. [PMID: 39755581 PMCID: PMC11748936 DOI: 10.1136/jitc-2024-010162] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2024] [Accepted: 11/21/2024] [Indexed: 01/06/2025] Open
Abstract
BACKGROUND Sialic acid-binding immunoglobulin-like lectins (SIGLECs) are widely expressed on immune cell surfaces, play an important role in maintaining immune homeostasis and regulating inflammatory responses, and are increasingly emerging as potential targets for tumor immunotherapy. However, the expression profile and crucial role of SIGLEC11 in gastric cancer (GC) remain unclear. This study aimed to elucidate the prognostic relevance of SIGLEC11 expression and its role in the immune microenvironment in patients with GC. METHODS SIGLEC11 expression profile was analyzed using bioinformatics, immunohistochemistry, and immunofluorescence staining. Flow cytometry, mouse tumor models, patient-derived tumor organoid models, and RNA sequencing were used to explore the potential functions with the underlying mechanisms of SIGLEC11 in a coculture system of macrophages and GC cells. RESULTS We demonstrated that SIGLEC11 was predominantly expressed in normal tissues. However, tumor-infiltrating SIGLEC11+ cells in the high SIGLEC11 expression subgroups showed poor overall survival, which was associated with the expression of an immunosuppressive regulator. Our results showed that SIGLEC11 was predominantly expressed in monocytes and macrophages and selectively upregulated in tumor-associated macrophages. Furthermore, SIGLEC11 promoted macrophage M2 polarization via AKT-mTOR signaling. In addition, SIGLEC11+ macrophages accelerate GC progression. CONCLUSIONS The abundance of SIGLEC11+ M2-like macrophage-infiltrating tumors may serve as a biomarker for identifying immunosuppressive subtypes of GC. Thus, the potential role of SIGLEC11+ M2 macrophages as therapeutic targets warrants further investigation.
Collapse
Affiliation(s)
- Jingxin Yin
- Department of General Surgery, Shanghai Key Laboratory of Gastric Neoplasms, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Institute of Digestive Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yang Lu
- Department of General Surgery, Shanghai Key Laboratory of Gastric Neoplasms, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Institute of Digestive Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yihao Liu
- Department of General Surgery, Pancreatic Disease Center, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Research Institute of Pancreatic Diseases, Shanghai Key Laboratory of Translational Research for Pancreatic Neoplasms, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Qimeng Shi
- Department of General Surgery, Shanghai Key Laboratory of Gastric Neoplasms, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Institute of Digestive Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Minmin Shi
- Department of General Surgery, Pancreatic Disease Center, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Research Institute of Pancreatic Diseases, Shanghai Key Laboratory of Translational Research for Pancreatic Neoplasms, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zhenggang Zhu
- Department of General Surgery, Shanghai Key Laboratory of Gastric Neoplasms, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Institute of Digestive Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Da Fu
- Department of General Surgery, Pancreatic Disease Center, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Research Institute of Pancreatic Diseases, Shanghai Key Laboratory of Translational Research for Pancreatic Neoplasms, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zhenqiang Wang
- Department of General Surgery, Shanghai Key Laboratory of Gastric Neoplasms, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Institute of Digestive Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Chen Li
- Department of General Surgery, Shanghai Key Laboratory of Gastric Neoplasms, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Institute of Digestive Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
21
|
Adak AK, Tseng H, Chang S, Chiang Y, Lyu K, Lee Y, Lu W, Kuo W, Angata T, Lin C. Comprehensive Modular Synthesis of Ganglioside Glycans and Evaluation of their Binding Affinities to Siglec-7 and Siglec-9. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2412815. [PMID: 39555730 PMCID: PMC11727393 DOI: 10.1002/advs.202412815] [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: 10/22/2024] [Indexed: 11/19/2024]
Abstract
In the present work, bacterial glycosyltransferases are utilized to construct ganglioside glycans in a convergent approach via a sugar‒nucleotide regeneration system and one-pot multienzyme reactions. Starting from β-lactoside enables the diversification of both the glycan moieties and the linkages in the lower α-arm and upper β-arm. Overall, a comprehensive panel of 24 natural a-series (GM3, GM2, GM1a, GD1a, GT1a, and fucosyl-GM1), b-series (GD3, GD2, GD1b, GT1b, and GQ1b), c-series (GT3, GT2, GT1c, GQ1c, and GP1c), α-series (GM1α, GD1aα, and GT1aα), and o-series (GA2, GA1, GM1b, GalNAc-GM1b, and GD1c) ganglioside glycans are prepared, which are suitable for biological studies and further applications. Moreover, a microarray is constructed with these synthesized ganglioside glycans to investigate their binding specificity with recombinant Fc-fused Siglec-7 and Siglec-9, which are immune checkpoint-like glycan recognition proteins on natural killer cells. The microarray binding results reveal that GD3 and GT1aα are specific ligands for Siglec-7 and Siglec-9, respectively, and this discovery can lead to the identification of appropriate ligands for investigating the roles of these Siglecs in immunomodulation.
Collapse
Affiliation(s)
- Avijit K. Adak
- Department of ChemistryNational Tsing Hua University101 Section 2, Kuang Fu RoadHsinchu30013Taiwan
| | - Hsin‐Kai Tseng
- Department of ChemistryNational Tsing Hua University101 Section 2, Kuang Fu RoadHsinchu30013Taiwan
| | - Shu‐Yen Chang
- Department of ChemistryNational Tsing Hua University101 Section 2, Kuang Fu RoadHsinchu30013Taiwan
| | - Yu‐Ching Chiang
- Department of ChemistryNational Tsing Hua University101 Section 2, Kuang Fu RoadHsinchu30013Taiwan
| | - Ke‐Hong Lyu
- Department of ChemistryNational Tsing Hua University101 Section 2, Kuang Fu RoadHsinchu30013Taiwan
| | - Yun‐Sheng Lee
- Department of ChemistryNational Tsing Hua University101 Section 2, Kuang Fu RoadHsinchu30013Taiwan
| | - Wen Lu
- Department of ChemistryNational Tsing Hua University101 Section 2, Kuang Fu RoadHsinchu30013Taiwan
| | - Wen‐Hua Kuo
- Department of ChemistryNational Tsing Hua University101 Section 2, Kuang Fu RoadHsinchu30013Taiwan
| | - Takashi Angata
- Institute of Biological ChemistryAcademia SinicaTaipei11529Taiwan
| | - Chun‐Cheng Lin
- Department of ChemistryNational Tsing Hua University101 Section 2, Kuang Fu RoadHsinchu30013Taiwan
- Department of Medicinal and Applied ChemistryKaohsiung Medical UniversityKaohsiung80708Taiwan
| |
Collapse
|
22
|
Harrison LC, Stone NL, Bandala-Sanchez E, Huntington ND, McLachlan RI, Rautela J, O’Bryan MK. Soluble CD52 mediates immune suppression by human seminal fluid. Front Immunol 2024; 15:1497889. [PMID: 39737172 PMCID: PMC11682959 DOI: 10.3389/fimmu.2024.1497889] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2024] [Accepted: 11/26/2024] [Indexed: 01/01/2025] Open
Abstract
Seminal fluid provides for the carriage and nutrition of sperm, but also modulates immunity to prevent allo-rejection of sperm by the female. Immune suppression by seminal fluid has been associated with extracellular vesicles, originally termed prostasomes, which contain CD52, a glycosylated glycophosphoinositol-anchored peptide released from testicular epithelial cells. Previously, we reported that human T cell-derived CD52, bound to the danger-associated molecular pattern protein, high mobility group box 1 (HMGB1), suppresses T cell function via the inhibitory sialic acid-binding immunoglobulin-like lectin-10 (Siglec-10) receptor. Here we show that human seminal fluid contains high concentrations of CD52 complexed with HMGB1, which mediates T cell suppression indirectly via Siglec-7 on antigen-presenting cells. Proliferation of natural killer (NK) cells, which express Siglec-7 and play a key role in the immune defence of the uterus, was directly suppressed by seminal fluid CD52. These findings elucidate a critical function of seminal fluid to suppress cellular immunity and facilitate reproduction.
Collapse
Affiliation(s)
- Leonard C. Harrison
- Population Heath and Immunity Division, Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia
- The University of Melbourne, Department of Medical Biology, Parkville, VIC, Australia
| | - Natalie L. Stone
- Population Heath and Immunity Division, Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia
- The University of Melbourne, Department of Medical Biology, Parkville, VIC, Australia
| | - Esther Bandala-Sanchez
- Population Heath and Immunity Division, Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia
- The University of Melbourne, Department of Medical Biology, Parkville, VIC, Australia
| | - Nicholas D. Huntington
- Population Heath and Immunity Division, Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia
- The University of Melbourne, Department of Medical Biology, Parkville, VIC, Australia
| | - Robert I. McLachlan
- Hudson Institute of Medical Research, Monash University, Melbourne, VIC, Australia
| | - Jai Rautela
- Population Heath and Immunity Division, Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia
- The University of Melbourne, Department of Medical Biology, Parkville, VIC, Australia
| | - Moira K. O’Bryan
- School of Biosciences and Bio21 Molecular Science and Biotechnology Institute, Faculty of Science, The University of Melbourne, Melbourne, VIC, Australia
| |
Collapse
|
23
|
Fan Y, Sun L, He J, Chen Y, Ma H, Ding H. Siglec15 in blood system diseases: from bench to bedside. Front Immunol 2024; 15:1490505. [PMID: 39697338 PMCID: PMC11652361 DOI: 10.3389/fimmu.2024.1490505] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2024] [Accepted: 11/13/2024] [Indexed: 12/20/2024] Open
Abstract
Inhibiting the PD-1/PD-L1 pathway using immunomodulators has demonstrated promising outcomes in clinics. Immunomodulators can effectively target immune checkpoints with a strong preference for the tumor microenvironment (TME). Besides, immunomodulators specifically target the recently discovered inhibitory immune checkpoint, sialic acid-binding immunoglobulin-like lectin (Siglec-15). Distinctive in its molecular composition, Siglec-15 has a unique molecular composition and been shown to be highly prevalent in numerous solid tumor tissues and tumor-associated macrophages (TAMs) in human subjects. Notably, Siglec-15 is up-regulated across various cancer types. As a result, Siglec-15 has attracted significant attention due to its exclusive nature concerning PD-L1 expression, suggesting its role in immune evasion in patients lacking PD-L1. Siglec-15 predominantly appears in certain populations and can promote tumor development by repressing T lymphocyte activation and proliferation, thereby facilitating tumor cell immune escape. Furthermore, Siglec-15 is implicated in osteoclast differentiation and bone remodeling, indicating that it is a promising target for next-generation cancer immunotherapies. Additionally, Siglec-15 can modulate immune responses to microbial infections. The current treatment strategies for hematological conditions predominantly include conventional intensive chemotherapy and transplantation methods. However, emerging immunotherapeutic approaches are increasingly recognized for their overall effectiveness, indicating that specific molecular targets should be identified. The expression of Siglec-15 within tumor cells may indicate a novel pathway for treating hematological malignancies. In this study, the biological attributes, expression patterns, and pathogenic mechanisms of Siglec-15 across various diseases were reviewed. The role of Siglec-15 in the pathogenesis and laboratory diagnosis of hematological disorders was also evaluated.
Collapse
Affiliation(s)
- Yujia Fan
- Baotou Medical College of Inner Mongolia University of Science and Technology, Baotou, Inner Mongolia, China
| | - Liangliang Sun
- Clinical Laboratory Medicine Centre, Inner Mongolia Autonomous Region People’s Hospital, Hohhot, Inner Mongolia, China
| | - Juan He
- Clinical Laboratory Medicine Centre, Inner Mongolia Autonomous Region People’s Hospital, Hohhot, Inner Mongolia, China
| | - Yuetong Chen
- Clinical Laboratory Medicine Centre, Inner Mongolia Autonomous Region People’s Hospital, Hohhot, Inner Mongolia, China
| | - Hongli Ma
- Baotou Medical College of Inner Mongolia University of Science and Technology, Baotou, Inner Mongolia, China
| | - Haitao Ding
- Clinical Laboratory Medicine Centre, Inner Mongolia Autonomous Region People’s Hospital, Hohhot, Inner Mongolia, China
| |
Collapse
|
24
|
Guerreiro A, Compañón I, Lazaris FS, Labão-Almeida C, Oroz P, Ghirardello M, Marques MC, Corzana F, Bernardes GJL. Non-Natural MUC1 Glycopeptide Homogeneous Cancer Vaccine with Enhanced Immunogenicity and Therapeutic Activity. Angew Chem Int Ed Engl 2024; 63:e202411009. [PMID: 39275921 DOI: 10.1002/anie.202411009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2024] [Revised: 09/05/2024] [Accepted: 09/13/2024] [Indexed: 09/16/2024]
Abstract
Glycopeptides derived from the glycoprotein mucin-1 (MUC1) have shown potential as tumor-associated antigens for cancer vaccine development. However, their low immunogenicity and non-selective conjugation to carriers present significant challenges for the clinical efficacy of MUC1-based vaccines. Here, we introduce a novel vaccine candidate based on a structure-guided design of an artificial antigen derived from MUC1 glycopeptide. This engineered antigen contains two non-natural amino acids and has an α-S-glycosidic bond, where sulfur replaces the conventional oxygen atom linking the peptide backbone to the sugar N-acetylgalactosamine. The glycopeptide is then specifically conjugated to the immunogenic protein carrier CRM197 (Cross-Reactive Material 197), a protein approved for human use. Conjugation involves selective reduction and re-bridging of a disulfide in CRM197, allowing the attachment of a single copy of MUC1. This strategy results in a chemically defined vaccine while maintaining both the structural integrity and immunogenicity of the protein carrier. The vaccine elicits a robust Th1-like immune response in mice and generates antibodies capable of recognizing human cancer cells expressing tumor-associated MUC1. When tested in mouse models of colon adenocarcinoma and pancreatic cancer, the vaccine is effective both as a prophylactic and therapeutic use, significantly delaying tumor growth. In therapeutic applications, improved outcomes were observed when the vaccine was combined with an anti-programmed cell death protein 1 (anti-PD-1) checkpoint inhibitor. Our strategy reduces batch-to-batch variability and enhances both immunogenicity and therapeutic potential. This site-specific approach disputes a prevailing dogma where glycoconjugate vaccines require multivalent display of antigens.
Collapse
Affiliation(s)
- Ana Guerreiro
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina Universidade de Lisboa, Avenida Professor Egas Moniz, 1649-028, Lisboa, Portugal
- Basinnov Lifesciences, Av. José Malhoa 2, Escritório 3.7, 1070-325, Lisboa, Portugal
| | - Ismael Compañón
- Departamento de Química and Instituto de Investigación en Química de la Universidad de La Rioja (IQUR), Madre de Dios, 53, 26006, Logroño, Spain
| | - Foivos S Lazaris
- Departamento de Química and Instituto de Investigación en Química de la Universidad de La Rioja (IQUR), Madre de Dios, 53, 26006, Logroño, Spain
| | - Carlos Labão-Almeida
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina Universidade de Lisboa, Avenida Professor Egas Moniz, 1649-028, Lisboa, Portugal
| | - Paula Oroz
- Departamento de Química and Instituto de Investigación en Química de la Universidad de La Rioja (IQUR), Madre de Dios, 53, 26006, Logroño, Spain
| | - Mattia Ghirardello
- Departamento de Química and Instituto de Investigación en Química de la Universidad de La Rioja (IQUR), Madre de Dios, 53, 26006, Logroño, Spain
| | - Marta C Marques
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina Universidade de Lisboa, Avenida Professor Egas Moniz, 1649-028, Lisboa, Portugal
| | - Francisco Corzana
- Departamento de Química and Instituto de Investigación en Química de la Universidad de La Rioja (IQUR), Madre de Dios, 53, 26006, Logroño, Spain
| | - Gonçalo J L Bernardes
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina Universidade de Lisboa, Avenida Professor Egas Moniz, 1649-028, Lisboa, Portugal
- Basinnov Lifesciences, Av. José Malhoa 2, Escritório 3.7, 1070-325, Lisboa, Portugal
- Yusuf Hamied Department of Chemistry, University of Cambridge, Lensfield Road, CB2 1EW, Cambridge, UK
| |
Collapse
|
25
|
Yin S, Li C, Shen X, Yu G, Cui L, Wu Y, He Y, Yu S, Chen J, Lu S, Qiu G, Song M, Qian C, Zou Z, Yu Y, Xu S. Siglec-G Suppresses CD8 + T Cells Responses through Metabolic Rewiring and Can be Targeted to Enhance Tumor Immunotherapy. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2403438. [PMID: 39373395 PMCID: PMC11615767 DOI: 10.1002/advs.202403438] [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: 04/02/2024] [Revised: 08/31/2024] [Indexed: 10/08/2024]
Abstract
CD8+ T cells play a critical role in cancer immune-surveillance and pathogen elimination. However, their effector function can be severely impaired by inhibitory receptors such as programmed death-1 (PD-1) and T cell immunoglobulin domain and mucin domain-3 (Tim-3). Here Siglec-G is identified as a coinhibitory receptor that limits CD8+ T cell function. Siglec-G is highly expressed on tumor-infiltrating T cells and is enriched in the exhausted T cell subset. Ablation of Siglec-G enhances the efficacy of adoptively transferred T cells and chimeric antigen receptor (CAR) T cells in suppressing solid tumors growth. Mechanistically, sialoglycan ligands, such as CD24 on tumor cells, activate the Siglec-G-SHP2 axis in CD8+ T cells, impairing metabolic reprogramming from oxidative phosphorylation to glycolysis, which dampens cytotoxic T lymphocyte (CTL) activation, expansion, and cytotoxicity. These findings discover a critical role for Siglec-G in inhibiting CD8+ T cell responses, suggesting its potential therapeutic effect in adoptive T cell therapy and tumor immunotherapy.
Collapse
Affiliation(s)
- Shenhui Yin
- National Key Laboratory of Immunity & InflammationNaval Medical University/Second Military Medical UniversityShanghai200433China
| | - Chunzhen Li
- National Key Laboratory of Immunity & InflammationNaval Medical University/Second Military Medical UniversityShanghai200433China
| | - Xin Shen
- National Key Laboratory of Immunity & InflammationNaval Medical University/Second Military Medical UniversityShanghai200433China
| | - Guanyu Yu
- Department of Colorectal SurgeryChanghai HospitalNaval Medical UniversityShanghai200433China
| | - Likun Cui
- National Key Laboratory of Immunity & InflammationNaval Medical University/Second Military Medical UniversityShanghai200433China
| | - Yunyang Wu
- National Key Laboratory of Immunity & InflammationNaval Medical University/Second Military Medical UniversityShanghai200433China
| | - Yixian He
- National Key Laboratory of Immunity & InflammationNaval Medical University/Second Military Medical UniversityShanghai200433China
| | - Shu Yu
- National Key Laboratory of Immunity & InflammationNaval Medical University/Second Military Medical UniversityShanghai200433China
| | - Jie Chen
- National Key Laboratory of Immunity & InflammationNaval Medical University/Second Military Medical UniversityShanghai200433China
| | - Shaoteng Lu
- National Key Laboratory of Immunity & InflammationNaval Medical University/Second Military Medical UniversityShanghai200433China
| | - Guifang Qiu
- National Key Laboratory of Immunity & InflammationNaval Medical University/Second Military Medical UniversityShanghai200433China
| | - Mengqi Song
- National Key Laboratory of Immunity & InflammationNaval Medical University/Second Military Medical UniversityShanghai200433China
| | - Cheng Qian
- National Key Laboratory of Immunity & InflammationNaval Medical University/Second Military Medical UniversityShanghai200433China
| | - Zui Zou
- School of AnesthesiologyNaval Medical UniversityShanghai200433China
- Faculty of AnesthesiologyChanghai HospitalNaval Medical UniversityShanghai200433China
| | - Yizhi Yu
- National Key Laboratory of Immunity & InflammationNaval Medical University/Second Military Medical UniversityShanghai200433China
| | - Sheng Xu
- National Key Laboratory of Immunity & InflammationNaval Medical University/Second Military Medical UniversityShanghai200433China
| |
Collapse
|
26
|
Qi W, Liu C, Shi L, Li H, Hou X, Du H, Chen L, Gao X, Cao X, Guo N, Dong Y, Li C, Yuan F, Teng Z, Hu H, Zhu F, Zhou X, Guo L, Zhao M, Xia M. CD169+ Macrophages Mediate the Immune Response of Allergic Rhinitis Through the Keap1/Nrf2/HO-1 Axis. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2309331. [PMID: 39435598 PMCID: PMC11615775 DOI: 10.1002/advs.202309331] [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: 12/01/2023] [Revised: 08/21/2024] [Indexed: 10/23/2024]
Abstract
CD169+ macrophages are a newly defined macrophage subpopulation that can recognize and bind with other cells through related ligands, playing an essential role in antigen presentation and immune tolerance. However, its role in Allergic Rhinitis (AR) is still unclear. To investigate the characteristics of CD169+ macrophages in AR, this work first detects their expression patterns in the nasal mucosa of clinical patients. These results show a significant increase in CD169+ macrophages in the nasal mucosa of patients with AR. Subsequently, this work establishes an animal AR model using CD169 transgenic mice and compared the advantages of the two models. Moreover, this work also demonstrates the effects of CD169 knockout on eosinophils, Th cells, Treg cells, and the migration of dendritic cells (DCs). In addition, this metabolomic data shows that CD169+ macrophages can upregulate alanine production and increase reactive oxygen species (ROS) levels. This process may be mediated through the Keap1/Nrf2/HO-1 signaling pathway. In addition, this work also finds that SLC38A2 plays an essential role in the process of CD169+ macrophages promoting alanine uptake by DCs. This study confirms that CD169+ macrophages can upregulate their internal alanine production and increase ROS levels through the Keap1/Nrf2/HO-1 axis, playing an irreplaceable role in AR.
Collapse
Affiliation(s)
- Wenwen Qi
- Department of OtolaryngologyShandong Provincial Hospital Affiliated to Shandong First Medical UniversityJinanChina
| | - Chengcheng Liu
- Department of Central LaboratoryShandong Provincial Hospital Affiliated to Shandong First Medical UniversityJinanChina
- Medical Science and Technology Innovation CenterShandong First Medical University & Shandong Academy of Medical SciencesJinanChina
| | - Lei Shi
- Department of OtolaryngologyShandong Provincial Hospital Affiliated to Shandong First Medical UniversityJinanChina
| | - Hui Li
- Department of OtolaryngologyShandong Provincial Hospital Affiliated to Shandong First Medical UniversityJinanChina
| | - Xiaozhi Hou
- Department of OtolaryngologyShandong Provincial Hospital Affiliated to Shandong First Medical UniversityJinanChina
| | - Hongjie Du
- Department of OtolaryngologyShandong Provincial Hospital Affiliated to Shandong First Medical UniversityJinanChina
| | - Luqiu Chen
- Department of Pediatric SurgeryQilu HospitalCheeloo College of MedicineShandong UniversityJinanShandong250012China
| | - Xiaochen Gao
- Department of OtolaryngologyShandong Provincial Hospital Affiliated to Shandong First Medical UniversityJinanChina
- Medical Science and Technology Innovation CenterShandong First Medical University & Shandong Academy of Medical SciencesJinanChina
| | - Xue Cao
- Department of OtolaryngologyShandong Provincial HospitalShandong UniversityJinanChina
| | - Na Guo
- Department of OtolaryngologyShandong Provincial Hospital Affiliated to Shandong First Medical UniversityJinanChina
- Medical Science and Technology Innovation CenterShandong First Medical University & Shandong Academy of Medical SciencesJinanChina
| | - Yuhan Dong
- Department of OtolaryngologyShandong Provincial HospitalShandong UniversityJinanChina
| | - Chengzhilin Li
- Department of OtolaryngologyShandong Provincial Hospital Affiliated to Shandong First Medical UniversityJinanChina
- Medical Science and Technology Innovation CenterShandong First Medical University & Shandong Academy of Medical SciencesJinanChina
| | - Fanyu Yuan
- Department of OtolaryngologyShandong Provincial Hospital Affiliated to Shandong First Medical UniversityJinanChina
- Medical Science and Technology Innovation CenterShandong First Medical University & Shandong Academy of Medical SciencesJinanChina
| | - Zhenxiao Teng
- Department of OtolaryngologyShandong Provincial Hospital Affiliated to Shandong First Medical UniversityJinanChina
| | - Houyang Hu
- Department of OtolaryngologyShandong Provincial Hospital Affiliated to Shandong First Medical UniversityJinanChina
- Medical Science and Technology Innovation CenterShandong First Medical University & Shandong Academy of Medical SciencesJinanChina
| | - Fangyuan Zhu
- Department of OtolaryngologyShandong Provincial Hospital Affiliated to Shandong First Medical UniversityJinanChina
| | - Xuanchen Zhou
- Department of OtolaryngologyShandong Provincial Hospital Affiliated to Shandong First Medical UniversityJinanChina
| | - Lulu Guo
- Advanced Medical Research InstituteCheeloo College of MedicineNHC Key Laboratory of OtorhinolaryngologyShandong UniversityJinanChina
| | - Miaoqing Zhao
- Department of PathologyShandong Cancer Hospital and InstituteShandong First Medical University and Shandong Academy of Medical SciencesJinanChina
| | - Ming Xia
- Department of OtolaryngologyShandong Provincial Hospital Affiliated to Shandong First Medical UniversityJinanChina
- Medical Science and Technology Innovation CenterShandong First Medical University & Shandong Academy of Medical SciencesJinanChina
- Department of OtolaryngologyShandong Provincial HospitalShandong UniversityJinanChina
- NHC Key Laboratory of OtorhinolaryngologyJinanChina
| |
Collapse
|
27
|
Sirini C, De Rossi L, Moresco MA, Casucci M. CAR T cells in solid tumors and metastasis: paving the way forward. Cancer Metastasis Rev 2024; 43:1279-1296. [PMID: 39316265 DOI: 10.1007/s10555-024-10213-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/31/2024] [Accepted: 09/10/2024] [Indexed: 09/25/2024]
Abstract
CAR T cell therapy, hailed as a breakthrough in cancer treatment due to its remarkable outcomes in hematological malignancies, encounters significant hurdles when applied to solid tumors. While notable responses to CAR T cells remain sporadic in these patients, challenges persist due to issues such as on-target off-tumor toxicity, difficulties in their trafficking and infiltration into the tumor, and the presence of a hostile and immunosuppressive microenvironment. This review aims to explore recent endeavors aimed at overcoming these obstacles in CAR T cell therapy for solid tumors. Specifically, we will delve into promising strategies for enhancing tumor specificity through antigen targeting, addressing tumor heterogeneity, overcoming physical barriers, and counteracting the immune-suppressive microenvironment.
Collapse
Affiliation(s)
- Camilla Sirini
- Innovative Immunotherapies Unit, Division of Immunology, Transplantation and Infectious Diseases, IRCCS San Raffaele Scientific Institute, 20132, Milan, Italy
- Vita-Salute San Raffaele University, Milan, Italy
| | - Laura De Rossi
- Innovative Immunotherapies Unit, Division of Immunology, Transplantation and Infectious Diseases, IRCCS San Raffaele Scientific Institute, 20132, Milan, Italy
- Vita-Salute San Raffaele University, Milan, Italy
| | - Marta Angiola Moresco
- Innovative Immunotherapies Unit, Division of Immunology, Transplantation and Infectious Diseases, IRCCS San Raffaele Scientific Institute, 20132, Milan, Italy
- Vita-Salute San Raffaele University, Milan, Italy
| | - Monica Casucci
- Innovative Immunotherapies Unit, Division of Immunology, Transplantation and Infectious Diseases, IRCCS San Raffaele Scientific Institute, 20132, Milan, Italy.
| |
Collapse
|
28
|
Ghosh P. Deciphering the Cell Surface Sugar-Coating via Biochemical Pathways. Chemistry 2024; 30:e202401983. [PMID: 39215611 DOI: 10.1002/chem.202401983] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2024] [Revised: 08/28/2024] [Accepted: 08/29/2024] [Indexed: 09/04/2024]
Abstract
Cell surface components, specifically glycans, play a significant role in several biological functions like cell structure, crosstalk between cells, and eventual target recognition of the cells for therapeutics. The dense layer of glycans, i. e., glycocalyx, could differ in taxon, species, and cell type. Glycans are coupled with lipids and proteins to form glycolipids, glycoproteins, proteoglycans, and glycosylphosphatidylinositol-anchored proteins, making their study challenging. However, understanding glycosylation at the cellular level is vital for fundamental research and advancing glycan-targeted therapy. Among different pathways, metabolic glycan labelling uses the natural metabolic processes of the cell to introduce abiotic functionality into glycan residues. The Bertozzi group pioneered metabolic oligosaccharide engineering using glycan salvage pathways to convert monosaccharides with unnatural modifications. This eventually results in the probe becoming part of the complex cellular glycan structures via click chemistry using copper. On the other hand, the boronic acid-based probe can recognise carbohydrates in a single step without any chemical modification of the surface. This review discusses the significance of glycans as biomarkers for different diseases and the necessity to evaluate them in situ within the physiological environment. The review also discusses the prospect of this field and its potential applications.
Collapse
Affiliation(s)
- Pritam Ghosh
- Department of Chemistry, Humboldt-Universität zu Berlin, Brook-Taylor-Str. 2, 12489, Berlin, Germany
| |
Collapse
|
29
|
Lu J, Zhang Y, Wen H, Li J, Chen C, Xiao L. Siglec-15 as a potential molecule involved in osteoclast differentiation and bone metabolism. Heliyon 2024; 10:e38537. [PMID: 39524871 PMCID: PMC11550040 DOI: 10.1016/j.heliyon.2024.e38537] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2023] [Revised: 08/29/2024] [Accepted: 09/25/2024] [Indexed: 11/16/2024] Open
Abstract
Sialic acid-binding immunoglobulin-like lectin 15 (Siglec-15) is a well-conserved type I transmembrane protein of the Siglecs family, distributed in macrophages and dendritic cells in the human spleen and lymph nodes. As an immune receptor, Siglec-15 is expressed in almost all branches of the spinal cord. Siglec-15 participates in the metabolism of the skeleton by regulating osteoclast activity and differentiation and has an influential role in dynamic bone remodelling. The binding of DNAX activation protein of 12 kDa (DAP12), which contains the immunoreceptor tyrosine-based activation motif (ITAM) activation domain, to the Siglec-15 receptor provides a positive stimulatory signal for osteoclast growth, with the involvement of the receptor activator of nuclear factor-κB (RANK)/RANK ligand (RANKL) signalling. Recently, Siglec-15 antibodies have been shown to effectively prevent bone resorption in mouse models of osteoporosis and accelerate fracture healing to some extent. Therefore, exploring the molecular characteristics and functions of Siglec-15 may lead to new therapeutic strategies for common clinical skeletal diseases.
Collapse
Affiliation(s)
- Jiaqi Lu
- Department of Orthodontics, Medical Center of Stomatology, The Second Xiangya Hospital, Central South University, Furong District, Changsha, Hunan, 410011, China
| | - Yinyin Zhang
- Department of Orthodontics, Medical Center of Stomatology, The Second Xiangya Hospital, Central South University, Furong District, Changsha, Hunan, 410011, China
| | - Huiyu Wen
- Department of Orthodontics, Medical Center of Stomatology, The Second Xiangya Hospital, Central South University, Furong District, Changsha, Hunan, 410011, China
| | - Junlin Li
- Department of Orthodontics, Medical Center of Stomatology, The Second Xiangya Hospital, Central South University, Furong District, Changsha, Hunan, 410011, China
| | - Chen Chen
- Department of Orthodontics, Medical Center of Stomatology, The Second Xiangya Hospital, Central South University, Furong District, Changsha, Hunan, 410011, China
| | - Liwei Xiao
- Department of Orthodontics, Medical Center of Stomatology, The Second Xiangya Hospital, Central South University, Furong District, Changsha, Hunan, 410011, China
| |
Collapse
|
30
|
Hashimoto N, Ito S, Harazono A, Tsuchida A, Mouri Y, Yamamoto A, Okajima T, Ohmi Y, Furukawa K, Kudo Y, Kawasaki N, Furukawa K. Bidirectional signals generated by Siglec-7 and its crucial ligand tri-sialylated T to escape of cancer cells from immune surveillance. iScience 2024; 27:111139. [PMID: 39507251 PMCID: PMC11539641 DOI: 10.1016/j.isci.2024.111139] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2024] [Revised: 08/05/2024] [Accepted: 10/07/2024] [Indexed: 11/08/2024] Open
Abstract
Siglec-7, an inhibitory receptor expressed on natural killer (NK) cells, recognizes sialic acid-containing glycans. However, the ligand glycan structures of Siglec-7 and its carrier proteins have not been comprehensively investigated. Here, we identified four sialyltransferases that are used for the synthesis of ligand glycans of Siglec-7 and two ligand O-glycan-carrier proteins, PODXL and MUC13, using a colon cancer line. Upon binding of these ligand glycans, Siglec-7-expressing immune cells showed reduced cytotoxic activity, whereas cancer cells expressing ligand glycans underwent signal activation, leading to enhanced invasion activity. To clarify the structure of the ligand glycan, podoplanin (PDPN) identified as a Siglec-7 ligand-carrier protein, was transfected into HEK293T cells using sialyltransferase cDNAs. Mass spectrometry of the products revealed a ligand glycan, tri-sialylated T antigen. These results indicate that Siglec-7 interaction with its ligand generates bidirectional signals in NK and cancer cells, leading to the efficient escape of cancers from host immune surveillance.
Collapse
Affiliation(s)
- Noboru Hashimoto
- Biochemistry II, Nagoya University Graduate School of Medicine, Nagoya 466-0065, Japan
- Tissue Regeneration, Tokushima University Graduate School of Biomedical Sciences, Tokushima 770-8504, Japan
| | - Shizuka Ito
- Biochemistry II, Nagoya University Graduate School of Medicine, Nagoya 466-0065, Japan
| | - Akira Harazono
- Biological Chemistry and Biologicals, National Institute of Health Sciences, Kanagawa 210-9501, Japan
| | - Akiko Tsuchida
- Laboratory of Glycobiology, The Noguchi Institute, Itabashi 173-0003, Japan
| | - Yasuhiro Mouri
- Oral Bioscience, Tokushima University Graduate School of Biomedical Sciences, Tokushima 770-8504, Japan
| | - Akihito Yamamoto
- Tissue Regeneration, Tokushima University Graduate School of Biomedical Sciences, Tokushima 770-8504, Japan
| | - Tetsuya Okajima
- Biochemistry II, Nagoya University Graduate School of Medicine, Nagoya 466-0065, Japan
| | - Yuhsuke Ohmi
- Clinical Engineering, Chubu University College of Life and Health Science, Aichi 487-8501, Japan
| | - Keiko Furukawa
- Biomedical Sciences, Chubu University College of Life and Health Sciences, Aichi 487-8501, Japan
| | - Yasusei Kudo
- Oral Bioscience, Tokushima University Graduate School of Biomedical Sciences, Tokushima 770-8504, Japan
| | - Nana Kawasaki
- Biopharmaceutical and Regenerative Sciences, Graduate School of Medical Life Science, Yokohama City University, Yokohama 230-0045, Japan
| | - Koichi Furukawa
- Biochemistry II, Nagoya University Graduate School of Medicine, Nagoya 466-0065, Japan
- Biomedical Sciences, Chubu University College of Life and Health Sciences, Aichi 487-8501, Japan
| |
Collapse
|
31
|
Xiang Z, Chu C, Xu D, Chen S. Tuning the Protonation Sensitivity of Weak Acidic Groups on a Zwitterionic Dendrimer for Selectively Targeting GD2-Overexpressed Tumor Cells in an Acidic Tumor Microenvironment. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2024; 40:24106-24116. [PMID: 39481027 DOI: 10.1021/acs.langmuir.4c03503] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/02/2024]
Abstract
Disialoganglioside (GD2) is one of the most popular overexpressed antigens for tumor cell targeting. However, GD2-specific antibodies often show unintended targeting to GD2-expressing health-maintaining cells due to the comparable binding affinities both at physiological pH and in a slightly acidic tumor microenvironment (TME). In this work, an affinity-switchable zwitterionic PAMAM G5 dendrimer (G5-3S) is developed for selective binding to GD2 only in a slightly acidic TME. It has 3 sulfonic groups, 128 carboxylic groups, and 125 amino groups on the surface. This affinity switch is realized by multiple hydrogen bond (H-bond) formation between protonated carboxylic groups surrounding a sulfonic group and overexpressed GD2 clusters on the tumor cell membrane in the slightly acidic TME, whereas there is no stable H-bond formation at physiological pH. Thus, G5-3S shows superior selectivity to GD2-overexpressed tumor cells over anti-GD2 antibodies by avoiding targeting GD2-expressing health-maintaining cells at physiological pH. This suggests that G5-3S is a promising candidate for GD2-overexpressed cancer treatment.
Collapse
Affiliation(s)
- Ziyin Xiang
- Key Laboratory of Biomass Chemical Engineering of Ministry of Education and Zhejiang Key Lab of Smart Biomaterial, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou 310027, China
| | - Chengchao Chu
- Key Laboratory of Biomass Chemical Engineering of Ministry of Education and Zhejiang Key Lab of Smart Biomaterial, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou 310027, China
| | - Da Xu
- Key Laboratory of Biomass Chemical Engineering of Ministry of Education and Zhejiang Key Lab of Smart Biomaterial, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou 310027, China
| | - Shengfu Chen
- Key Laboratory of Biomass Chemical Engineering of Ministry of Education and Zhejiang Key Lab of Smart Biomaterial, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou 310027, China
| |
Collapse
|
32
|
Yang Y, Zeng Q, Liu Y, Li P, Tang Y, Wei Z, Zhou Z. Functional characterization of Siglec14 in facilitating innate defenses in Carassius cuvieri × Carassius auratus red var. FISH & SHELLFISH IMMUNOLOGY 2024; 155:110017. [PMID: 39522706 DOI: 10.1016/j.fsi.2024.110017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/15/2024] [Revised: 10/25/2024] [Accepted: 11/07/2024] [Indexed: 11/16/2024]
Abstract
Sialic acid-binding Ig-like lectin 14 (Siglec14) is a member of the Siglecs family, a group of immune system receptors characterized by their ability to recognize sialic acids, which are a type of carbohydrate commonly found on the surfaces of cells in mammals. However, the role of Siglec14 in the innate immunity of teleosts has not been extensively explored. For this purpose, our study focused on the identification and characterization of a classical Siglec14, designated as WR-Siglec14, derived from the hybrid fish Carassius cuvieri × Carassius auratus red var. Sequence analysis reveals WR-Siglec14 has the considerable homology with Siglec14 orthologs across various species, featuring the conserved immunoglobulin domains. We further investigated the expression patterns of WR-Siglec14 in response to pathogen exposure and observed significant upregulation in key immune-related tissues upon infection with Aeromonas hydrophila. Additionally, the study demonstrated that WR-Siglec14 enhanced the bactericidal activity and interacted with WR-DAP12. Moreover, we found treatment with WR-Siglec14 maintained higher numbers of goblet cells and mitigated apoptosis in the gut tissue during A. hydrophila infection. WR-Siglec14 also stimulated antimicrobial gene expression and reduced bacterial burdens in the fish, significantly improving survival rates against A. hydrophila infection. These results indicate that WR-Siglec14 plays the critical role in immune defense and gut barrier function against pathogen invasion in fish.
Collapse
Affiliation(s)
- Ye Yang
- State Key Laboratory of Developmental Biology of Freshwater Fish, Engineering Research Center of Polyploid Fish Reproduction and Breeding of the State Education Ministry, College of Life Sciences, Hunan Normal University, Changsha, 410081, Hunan, China
| | - Qiongyao Zeng
- State Key Laboratory of Developmental Biology of Freshwater Fish, Engineering Research Center of Polyploid Fish Reproduction and Breeding of the State Education Ministry, College of Life Sciences, Hunan Normal University, Changsha, 410081, Hunan, China
| | - Yujun Liu
- State Key Laboratory of Developmental Biology of Freshwater Fish, Engineering Research Center of Polyploid Fish Reproduction and Breeding of the State Education Ministry, College of Life Sciences, Hunan Normal University, Changsha, 410081, Hunan, China
| | - Pingyuan Li
- State Key Laboratory of Developmental Biology of Freshwater Fish, Engineering Research Center of Polyploid Fish Reproduction and Breeding of the State Education Ministry, College of Life Sciences, Hunan Normal University, Changsha, 410081, Hunan, China
| | - Yiyang Tang
- State Key Laboratory of Developmental Biology of Freshwater Fish, Engineering Research Center of Polyploid Fish Reproduction and Breeding of the State Education Ministry, College of Life Sciences, Hunan Normal University, Changsha, 410081, Hunan, China
| | - Zehong Wei
- State Key Laboratory of Developmental Biology of Freshwater Fish, Engineering Research Center of Polyploid Fish Reproduction and Breeding of the State Education Ministry, College of Life Sciences, Hunan Normal University, Changsha, 410081, Hunan, China.
| | - Zejun Zhou
- State Key Laboratory of Developmental Biology of Freshwater Fish, Engineering Research Center of Polyploid Fish Reproduction and Breeding of the State Education Ministry, College of Life Sciences, Hunan Normal University, Changsha, 410081, Hunan, China; Guangdong Provincial Key Laboratory of Aquatic Animal Disease Control and Healthy Culture, Guangdong Ocean University, Zhanjiang, 524088, Guangdong, China.
| |
Collapse
|
33
|
Pessentheiner AR, Spann NJ, Autran CA, Oh TG, Grunddal KV, Coker JK, Painter CD, Ramms B, Chiang AW, Wang CY, Hsiao J, Wang Y, Quach A, Booshehri LM, Hammond A, Tognaccini C, Latasiewicz J, Willemsen L, Zengler K, de Winther MP, Hoffman HM, Philpott M, Cribbs AP, Oppermann U, Lewis NE, Witztum JL, Yu R, Atkins AR, Downes M, Evans RM, Glass CK, Bode L, Gordts PL. The human milk oligosaccharide 3'sialyllactose reduces low-grade inflammation and atherosclerosis development in mice. JCI Insight 2024; 9:e181329. [PMID: 39325548 PMCID: PMC11601559 DOI: 10.1172/jci.insight.181329] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2024] [Accepted: 09/24/2024] [Indexed: 09/28/2024] Open
Abstract
Macrophages contribute to the induction and resolution of inflammation and play a central role in chronic low-grade inflammation in cardiovascular diseases caused by atherosclerosis. Human milk oligosaccharides (HMOs) are complex unconjugated glycans unique to human milk that benefit infant health and act as innate immune modulators. Here, we identify the HMO 3'sialyllactose (3'SL) as a natural inhibitor of TLR4-induced low-grade inflammation in macrophages and endothelium. Transcriptome analysis in macrophages revealed that 3'SL attenuates mRNA levels of a selected set of inflammatory genes and promotes the activity of liver X receptor (LXR) and sterol regulatory element binding protein-1 (SREBP1). These acute antiinflammatory effects of 3'SL were associated with reduced histone H3K27 acetylation at a subset of LPS-inducible enhancers distinguished by preferential enrichment for CCCTC-binding factor (CTCF), IFN regulatory factor 2 (IRF2), B cell lymphoma 6 (BCL6), and other transcription factor recognition motifs. In a murine atherosclerosis model, both s.c. and oral administration of 3'SL significantly reduced atherosclerosis development and the associated inflammation. This study provides evidence that 3'SL attenuates inflammation by a transcriptional mechanism to reduce atherosclerosis development in the context of cardiovascular disease.
Collapse
Affiliation(s)
- Ariane R. Pessentheiner
- Department of Medicine, UCSD, La Jolla, California, USA
- Institute of Molecular Biosciences, University of Graz, Graz, Austria
| | | | | | - Tae Gyu Oh
- Gene Expression Laboratory, Salk Institute for Biological Studies, La Jolla, California, USA
| | | | - Joanna K.C. Coker
- Department of Medicine, UCSD, La Jolla, California, USA
- Department of Bioengineering at UCSD, La Jolla, California, USA
| | | | - Bastian Ramms
- Department of Medicine, UCSD, La Jolla, California, USA
| | - Austin W.T. Chiang
- Department of Pediatrics at UCSD, La Jolla, California, USA
- Department of Bioengineering at UCSD, La Jolla, California, USA
- Novo Nordisk Foundation Center for Biosustainability, La Jolla, California, USA
| | - Chen-Yi Wang
- Botnar Research Centre, Nuffield Department of Orthopedics, Rheumatology and Musculoskeletal Sciences, NIH Research Oxford Biomedical Research Unit (BRU), and
- Oxford Centre for Translational Myeloma Research University of Oxford, Oxford, United Kingdom
| | - Jason Hsiao
- Department of Medicine, UCSD, La Jolla, California, USA
| | - Yiwen Wang
- Department of Medicine, UCSD, La Jolla, California, USA
| | - Anthony Quach
- Department of Medicine, UCSD, La Jolla, California, USA
| | | | | | | | | | - Lisa Willemsen
- Department of Medical Biochemistry, Experimental Vascular Biology, Amsterdam Cardiovascular Sciences, Amsterdam Infection and Immunity, Amsterdam University Medical Center (UMC), University of Amsterdam, Amsterdam, the Netherlands
| | - Karsten Zengler
- Department of Bioengineering at UCSD, La Jolla, California, USA
- Center for Microbiome Innovation, UCSD, La Jolla, California, USA
| | - Menno P.J. de Winther
- Department of Medical Biochemistry, Experimental Vascular Biology, Amsterdam Cardiovascular Sciences, Amsterdam Infection and Immunity, Amsterdam University Medical Center (UMC), University of Amsterdam, Amsterdam, the Netherlands
| | - Hal M. Hoffman
- Department of Medicine, UCSD, La Jolla, California, USA
- Department of Pediatrics at UCSD, La Jolla, California, USA
- Rady Children’s Hospital of San Diego, San Diego, California, USA
| | - Martin Philpott
- Botnar Research Centre, Nuffield Department of Orthopedics, Rheumatology and Musculoskeletal Sciences, NIH Research Oxford Biomedical Research Unit (BRU), and
| | - Adam P. Cribbs
- Botnar Research Centre, Nuffield Department of Orthopedics, Rheumatology and Musculoskeletal Sciences, NIH Research Oxford Biomedical Research Unit (BRU), and
- Oxford Centre for Translational Myeloma Research University of Oxford, Oxford, United Kingdom
| | - Udo Oppermann
- Botnar Research Centre, Nuffield Department of Orthopedics, Rheumatology and Musculoskeletal Sciences, NIH Research Oxford Biomedical Research Unit (BRU), and
- Oxford Centre for Translational Myeloma Research University of Oxford, Oxford, United Kingdom
| | - Nathan E. Lewis
- Gene Expression Laboratory, Salk Institute for Biological Studies, La Jolla, California, USA
- Department of Bioengineering at UCSD, La Jolla, California, USA
- Novo Nordisk Foundation Center for Biosustainability, La Jolla, California, USA
| | | | - Ruth Yu
- Gene Expression Laboratory, Salk Institute for Biological Studies, La Jolla, California, USA
| | - Annette R. Atkins
- Gene Expression Laboratory, Salk Institute for Biological Studies, La Jolla, California, USA
| | - Michael Downes
- Gene Expression Laboratory, Salk Institute for Biological Studies, La Jolla, California, USA
| | - Ron M. Evans
- Gene Expression Laboratory, Salk Institute for Biological Studies, La Jolla, California, USA
| | - Christopher K. Glass
- Department of Medicine, UCSD, La Jolla, California, USA
- Department of Cellular and Molecular Medicine and
| | - Lars Bode
- Department of Pediatrics at UCSD, La Jolla, California, USA
- Larsson-Rosenquist Foundation Mother-Milk-Infant Center of Research Excellence (MOMI CORE) and
- Glycobiology Research and Training Center, UCSD, La Jolla, California, USA
| | - Philip L.S.M. Gordts
- Department of Medicine, UCSD, La Jolla, California, USA
- Glycobiology Research and Training Center, UCSD, La Jolla, California, USA
| |
Collapse
|
34
|
Zhong X, D’Antona AM, Rouse JC. Mechanistic and Therapeutic Implications of Protein and Lipid Sialylation in Human Diseases. Int J Mol Sci 2024; 25:11962. [PMID: 39596031 PMCID: PMC11594235 DOI: 10.3390/ijms252211962] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2024] [Revised: 10/28/2024] [Accepted: 11/05/2024] [Indexed: 11/28/2024] Open
Abstract
Glycan structures of glycoproteins and glycolipids on the surface glycocalyx and luminal sugar layers of intracellular membrane compartments in human cells constitute a key interface between intracellular biological processes and external environments. Sialic acids, a class of alpha-keto acid sugars with a nine-carbon backbone, are frequently found as the terminal residues of these glycoconjugates, forming the critical components of these sugar layers. Changes in the status and content of cellular sialic acids are closely linked to many human diseases such as cancer, cardiovascular, neurological, inflammatory, infectious, and lysosomal storage diseases. The molecular machineries responsible for the biosynthesis of the sialylated glycans, along with their biological interacting partners, are important therapeutic strategies and targets for drug development. The purpose of this article is to comprehensively review the recent literature and provide new scientific insights into the mechanisms and therapeutic implications of sialylation in glycoproteins and glycolipids across various human diseases. Recent advances in the clinical developments of sialic acid-related therapies are also summarized and discussed.
Collapse
Affiliation(s)
- Xiaotian Zhong
- BioMedicine Design, Discovery and Early Development, Pfizer Research and Development, 610 Main Street, Cambridge, MA 02139, USA;
| | - Aaron M. D’Antona
- BioMedicine Design, Discovery and Early Development, Pfizer Research and Development, 610 Main Street, Cambridge, MA 02139, USA;
| | - Jason C. Rouse
- Analytical Research and Development, Biotherapeutics Pharmaceutical Sciences, Pfizer Inc., Andover, MA 01810, USA;
| |
Collapse
|
35
|
Zhou Y, Wei S, Xu M, Wu X, Dou W, Li H, Zhang Z, Zhang S. CAR-T cell therapy for hepatocellular carcinoma: current trends and challenges. Front Immunol 2024; 15:1489649. [PMID: 39569202 PMCID: PMC11576447 DOI: 10.3389/fimmu.2024.1489649] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2024] [Accepted: 10/18/2024] [Indexed: 11/22/2024] Open
Abstract
Hepatocellular carcinoma (HCC) ranks among the most prevalent cancers worldwide, highlighting the urgent need for improved diagnostic and therapeutic methodologies. The standard treatment regimen generally involves surgical intervention followed by systemic therapies; however, the median survival rates for patients remain unsatisfactory. Chimeric antigen receptor (CAR) T-cell therapy has emerged as a pivotal advancement in cancer treatment. Both clinical and preclinical studies emphasize the notable efficacy of CAR T cells in targeting HCC. Various molecules, such as GPC3, c-Met, and NKG2D, show significant promise as potential immunotherapeutic targets in liver cancer. Despite this, employing CAR T cells to treat solid tumors like HCC poses considerable challenges within the discipline. Numerous innovations have significant potential to enhance the efficacy of CAR T-cell therapy for HCC, including improvements in T cell trafficking, strategies to counteract the immunosuppressive tumor microenvironment, and enhanced safety protocols. Ongoing efforts to discover therapeutic targets for CAR T cells highlight the need for the development of more practical manufacturing strategies for CAR-modified cells. This review synthesizes recent findings and clinical advancements in the use of CAR T-cell therapies for HCC treatment. We elucidate the therapeutic benefits of CAR T cells in HCC and identify the primary barriers to their broader application. Our analysis aims to provide a comprehensive overview of the current status and future prospects of CAR T-cell immunotherapy for HCC.
Collapse
Affiliation(s)
- Yexin Zhou
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China
- The General Hospital of Western Theater Command, Chengdu, China
| | - Shanshan Wei
- Department of Radiation Oncology, Radiation Oncology Key Laboratory of Sichuan Province, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, Affiliated Cancer Hospital of University of Electronic Science and Technology of China, Chengdu, Sichuan, China
| | - Menghui Xu
- The General Hospital of Western Theater Command, Chengdu, China
| | - Xinhui Wu
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China
| | - Wenbo Dou
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China
| | - Huakang Li
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China
| | - Zhonglin Zhang
- Department of Radiation Oncology, Radiation Oncology Key Laboratory of Sichuan Province, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, Affiliated Cancer Hospital of University of Electronic Science and Technology of China, Chengdu, Sichuan, China
| | - Shuo Zhang
- Department of Radiation Oncology, Radiation Oncology Key Laboratory of Sichuan Province, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, Affiliated Cancer Hospital of University of Electronic Science and Technology of China, Chengdu, Sichuan, China
| |
Collapse
|
36
|
Eggel A, Pennington LF, Jardetzky TS. Therapeutic monoclonal antibodies in allergy: Targeting IgE, cytokine, and alarmin pathways. Immunol Rev 2024; 328:387-411. [PMID: 39158477 PMCID: PMC11659931 DOI: 10.1111/imr.13380] [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] [Indexed: 08/20/2024]
Abstract
The etiology of allergy is closely linked to type 2 inflammatory responses ultimately leading to the production of allergen-specific immunoglobulin E (IgE), a key driver of many allergic conditions. At a high level, initial allergen exposure disrupts epithelial integrity, triggering local inflammation via alarmins including IL-25, IL-33, and TSLP, which activate type 2 innate lymphoid cells as well as other immune cells to secrete type 2 cytokines IL-4, IL-5 and IL-13, promoting Th2 cell development and eosinophil recruitment. Th2 cell dependent B cell activation promotes the production of allergen-specific IgE, which stably binds to basophils and mast cells. Rapid degranulation of these cells upon allergen re-exposure leads to allergic symptoms. Recent advances in our understanding of the molecular and cellular mechanisms underlying allergic pathophysiology have significantly shaped the development of therapeutic intervention strategies. In this review, we highlight key therapeutic targets within the allergic cascade with a particular focus on past, current and future treatment approaches using monoclonal antibodies. Specific targeting of alarmins, type 2 cytokines and IgE has shown varying degrees of clinical benefit in different allergic indications including asthma, chronic spontaneous urticaria, atopic dermatitis, chronic rhinosinusitis with nasal polyps, food allergies and eosinophilic esophagitis. While multiple therapeutic antibodies have been approved for clinical use, scientists are still working on ways to improve on current treatment approaches. Here, we provide context to understand therapeutic targeting strategies and their limitations, discussing both knowledge gaps and promising future directions to enhancing clinical efficacy in allergic disease management.
Collapse
Affiliation(s)
- Alexander Eggel
- Department for BioMedical ResearchUniversity of BernBernSwitzerland
- Department of Rheumatology and ImmunologyUniversity Hospital BernBernSwitzerland
| | | | - Theodore S. Jardetzky
- Department of Structural BiologyStanford University School of MedicineStanfordCaliforniaUSA
| |
Collapse
|
37
|
Bernstein JS, Bernstein JA, Lang DM. Chronic Spontaneous Urticaria: Current and Emerging Biologic Agents. Immunol Allergy Clin North Am 2024; 44:595-613. [PMID: 39389712 DOI: 10.1016/j.iac.2024.07.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/12/2024]
Abstract
Antihistamine refractory chronic spontaneous urticaria (CSU) has a prevalence of up to 50%. Anti-immunoglobulin E (IgE) therapies have revolutionized management of CSU, yet refractory cases persist, suggesting a role for biologic agents that impact alternative routes of mast cell stimulation independent of cross-linking at FcεR1. This review addresses anti-IgE and Th2-targeted therapies in the management of CSU. In addition, we explore novel treatments targeting alternative pathways of mast cell activation including MAS-related G protein-coupled receptor-X2 and sialic acid-binding immunoglobulin-like lectin-6, inhibiting intracellular signaling via Bruton's tyrosine kinase, and disrupting KIT activation by SCF.
Collapse
Affiliation(s)
- Joshua S Bernstein
- Division of Rheumatology, Allergy and Immunology, University of Cincinnati, 234 Goodman Street, Cincinnati, OH 45219, USA
| | - Jonathan A Bernstein
- Division of Rheumatology, Allergy and Immunology, University of Cincinnati, 234 Goodman Street, Cincinnati, OH 45219, USA
| | - David M Lang
- Department of Allergy and Clinical Immunology, Cleveland Clinic, 9500 Euclid Avenue, A90, Cleveland, OH 44195, USA.
| |
Collapse
|
38
|
Ma S, Zhang P, Ye J, Tian Y, Tian X, Jung J, Macauley MS, Zhang J, Wu P, Wen L. Enzyme-Sialylation-Controlled Chemical Sulfation of Glycan Epitopes for Decoding the Binding of Siglec Ligands. J Am Chem Soc 2024; 146:29469-29480. [PMID: 39417319 DOI: 10.1021/jacs.4c08817] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2024]
Abstract
Widely distributed in nature, sulfated glycan epitopes play important roles in diverse pathophysiological processes. However, due to their structural complexity, the preparation of glycan epitopes with structurally defined sulfation patterns is challenging, which significantly hampers the detailed elucidation of their biological functions at the molecular level. Here, we introduce a strategy for site-specific chemical sulfation of glycan epitopes, leveraging enzymatic sialylation and desialylation processes to precisely control the regio-specificity of sulfation of disaccharide or trisaccharide glycan backbones. Using this method, a sulfated glycan library covering the most common sialylated glycan epitopes was prepared in high yield and efficiency. By screening a microarray prepared with this glycan library, we systematically probed their binding specificity with human Siglecs (sialic acid-binding immunoglobulin-type lectins), many of which function as glyco-immune checkpoints to suppress immune system activation. Our investigation revealed that sulfation and sialylation patterns serve as important determinants of Siglec binding affinity and specificity. Thus, these findings offer new insights for the development of research tools and potential therapeutic agents targeting glyco-immune checkpoints by modulating the Siglec signaling pathway.
Collapse
Affiliation(s)
- Shengzhou Ma
- Carbohydrate-Based Drug Research Center, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
- State Key Laboratory of Chemical Biology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Pengfei Zhang
- Carbohydrate-Based Drug Research Center, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
- State Key Laboratory of Chemical Biology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Jinfeng Ye
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, California 92037, United States
| | - Yinping Tian
- Carbohydrate-Based Drug Research Center, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
- State Key Laboratory of Chemical Biology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Xiao Tian
- Zhongshan Institute for Drug Discovery, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Zhongshan, Guangdong 528400, China
| | - Jaesoo Jung
- Department of Chemistry, University of Alberta, Edmonton, Alberta T6G 2E1, Canada
| | - Matthew S Macauley
- Department of Chemistry, University of Alberta, Edmonton, Alberta T6G 2E1, Canada
- Department of Medical Microbiology and Immunology, University of Alberta, Edmonton, Alberta T6G 2E1, Canada
| | - Jiabin Zhang
- Carbohydrate-Based Drug Research Center, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
- University of Chinese Academy of Sciences, Beijing 100049, China
- Zhongshan Institute for Drug Discovery, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Zhongshan, Guangdong 528400, China
| | - Peng Wu
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, California 92037, United States
| | - Liuqing Wen
- Carbohydrate-Based Drug Research Center, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
- State Key Laboratory of Chemical Biology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| |
Collapse
|
39
|
Liu Y, Lin W, Gu Y, Lu C, Zhou X, Zhao H, Wang G, Shen A. Dysregulation of SIGLEC1 in non-small cell lung cancer: prognostic implications and immunomodulatory role-a multicenter cohort study. J Cancer Res Clin Oncol 2024; 150:481. [PMID: 39470815 PMCID: PMC11522155 DOI: 10.1007/s00432-024-06005-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2023] [Accepted: 10/16/2024] [Indexed: 11/01/2024]
Abstract
PURPOSE To investigate the clinical significance and functional role of SIGLEC1-positive cells in non-small cell lungcancer (NSCLC) patients, focusing on their prognostic impact and therapeutic response. METHODS A multicenter retrospective cohort analysis was conducted, integrating data from multiple sources. Weanalyzed SIGLEC1 expression in NSCLC tissues, clinicopathological features, overall survival outcomes,chemotherapy responsiveness, and sensitivity to targeted therapies. We also developed a prognostic model basedon SIGLEC1 expression and clinical variables. RESULTS SIGLEC1 expression was significantly downregulated in NSCLC tissues, and the density of SIGLEC1-positivecells was inversely correlated with various clinicopathological features. Notably, patients with high infiltration ofSIGLEC1-positive cells exhibited significantly better overall survival outcomes. Furthermore, elevated SIGLEC1expression was associated with improved responsiveness to chemotherapy and demonstrated distinct patterns ofsensitivity to targeted therapies. A robust prognostic model was developed by integrating SIGLEC1 expression andclinical variables. CONCLUSIONS This study highlighted the downregulation of SIGLEC1 in NSCLC tissues and its significant associationwith patient prognosis and therapeutic response. The findings suggested that SIGLEC1 played a critical role inmodulating the tumor immune microenvironment and has potential as both a prognostic biomarker and therapeutictarget in NSCLC.
Collapse
Affiliation(s)
- Yuan Liu
- Cancer Research Center Nantong, Affiliated Tumor Hospital of Nantong University, Nantong, 226006, China
- Department of Neuroscience, Shanghai Key Laboratory of Emotions and Affective Disorders, Songjiang Hospital and Songjiang Research Institute, Shanghai Jiao Tong University School of Medicine, Shanghai, 201600, China
| | - Wei Lin
- Jiangsu Key Laboratory of Neuropsychiatric Diseases and Institute of Neuroscience, Soochow University, Suzhou, Jiangsu, 215123, China
| | - Yang Gu
- Department of Clinical Medicine, General Medical Class 8, Kangda College of Nanjing Medical University, Lianyungang, 222000, China
| | - Chenlin Lu
- Department of Respiratory and Critical Care Medicine, The Affiliated Taizhou People's Hospital of Nanjing Medical University, Taizhou, 214504, China
| | - Xuan Zhou
- Department of Radiotherapy, Affiliated Hospital of Nantong University, Nantong, Jiangsu, 226001, China
| | - Hongyu Zhao
- Department of Radiotherapy, Affiliated Hospital of Nantong University, Nantong, Jiangsu, 226001, China
| | - Gaoren Wang
- Cancer Research Center Nantong, Affiliated Tumor Hospital of Nantong University, Nantong, 226006, China.
| | - Aiguo Shen
- Cancer Research Center Nantong, Affiliated Tumor Hospital of Nantong University, Nantong, 226006, China.
| |
Collapse
|
40
|
Moar P, Bowler S, Landay AL, Gianella S, Ndhlovu LC, Premeaux TA. Alterations in circulating immunoregulatory proteins discriminate poor CD4 T lymphocyte trajectories in people with HIV on suppressive antiretroviral therapy. mBio 2024; 15:e0226524. [PMID: 39287441 PMCID: PMC11481887 DOI: 10.1128/mbio.02265-24] [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: 08/07/2024] [Accepted: 08/19/2024] [Indexed: 09/19/2024] Open
Abstract
Despite the success in the management of HIV with antiretroviral therapy (ART), people with HIV (PWH) have a heightened state of immune activation and inflammation, and an estimated 10%-40% demonstrate poor CD4 T-cell reconstitution, thereby increasing their mortality and morbidity risk burden. Soluble immunoregulatory proteins that function in lymphocyte activation or inhibition are elevated in PWH and associate with T-cell dysfunction, HIV persistence, and are predictive of comorbid outcomes. Here, we measured a panel of 35 circulating immunoregulatory proteins in 116 PWH with variations in CD4 T-cell counts (poor CD4 trajectory: <200 cells/μl, n = 34 or immune competent: CD4 >500 cells/μl, n = 82) by Luminex. Participants were enrolled in the AIDS Clinical Trials Group Longitudinal Linked Randomized Trials cohort, had initiated ART on enrollment, and had been on suppressive ART for 1 year. Using non-parametric analysis, we found that the levels of CD276, ICOSL, BAFF, OX40, galectin-1, and galectin-9 were significantly higher in PWH with poor CD4 trajectories compared to individuals with immune-competent CD4 T-cell count. Notably, in logistic models, ICOSL and OX40 remained significant after adjusting for age and baseline plasma HIV RNA. Furthermore, Extreme Gradient Boosting machine learning models comprising co-stimulatory and inhibitory checkpoint proteins yielded high accuracy in classification of individuals with poor CD4 trajectories. In summary, we identified a novel signature of circulating immunoregulatory proteins indicative of poor CD4 trajectories that may serve as potential targets to monitor and manage immune perturbations more accurately in PWH during suppressive ART. IMPORTANCE It is essential to track immune perturbations related to insufficient CD4 T-cell recovery in PWH on suppressive ART as those with incomplete reconstitution are at a greater risk of non-AIDS-related morbidity and mortality. Several inflammatory soluble mediators have associated with poor immune reconstitution and adverse morbid outcomes in PWH, yet their implementation into routine clinical care to guide management remains inconsistent. Circulating immune checkpoint proteins have been linked to dysregulated immune pathways during suppressive ART and may serve as improved surrogate markers of clinical relevance. Here we investigate soluble lymphocyte-associated immunoregulatory proteins in virally suppressed PWH with no reported co-morbid outcomes and varying CD4 T-cell counts, to reveal underlying pathways that remain perturbed despite ART. This novel signature of immunoregulatory markers pertaining to poor CD4 T-cell trajectories uncover previously overlooked immune checkpoints as important targets for clinical monitoring of PWH in the setting of durable viral suppression by ART.
Collapse
Affiliation(s)
- Preeti Moar
- />Division of Infectious Diseases, Department of Medicine, Weill Cornell Medicine, New York, New York, USA
| | - Scott Bowler
- />Division of Infectious Diseases, Department of Medicine, Weill Cornell Medicine, New York, New York, USA
| | - Alan L. Landay
- School of Medicine, University of Texas Medical Branch, Galveston, Texas, USA
| | - Sara Gianella
- Division of Infectious Diseases and Global Public Health, Department of Medicine, University of California San Diego, San Diego, California, USA
| | - Lishomwa C. Ndhlovu
- />Division of Infectious Diseases, Department of Medicine, Weill Cornell Medicine, New York, New York, USA
| | - Thomas A. Premeaux
- />Division of Infectious Diseases, Department of Medicine, Weill Cornell Medicine, New York, New York, USA
| |
Collapse
|
41
|
Qiao Y, Tian X, Li S, Niu H. Identification and experimental validation of a sialylation-related long noncoding RNA signature for prognosis of bladder cancer. BMC Urol 2024; 24:222. [PMID: 39390546 PMCID: PMC11465731 DOI: 10.1186/s12894-024-01613-6] [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: 02/02/2024] [Accepted: 09/30/2024] [Indexed: 10/12/2024] Open
Abstract
BACKGROUND The dysregulation of sialylation plays a pivotal role in cancer progression and metastasis, impacting various aspects of tumor behavior. This study aimed to investigate the prognostic significance of long non-coding RNAs (lncRNAs) in relation to sialylation. Additionally, we aimed to develop a signature of sialylation-related lncRNAs in the context of bladder cancer. METHODS This study used transcriptomic data and clinical information from the TCGA (the Cancer Genome Atlas) database to screen for sialylation-related lncRNAs and constructed a prognostic model. The relationships between these lncRNAs and biological pathways, immune cell infiltration, drug sensitivity, etc., were analyzed, and the expression of some lncRNAs was validated at the cellular level. RESULTS This study identified 6 prognostic lncRNAs related to sialylation and constructed a risk score model with high predictive accuracy and reliability. The survival period of patients in the high-risk group was significantly lower than that of the low-risk group, and it was related to various biological pathways and immune functions. In addition, this study found differences in the sensitivity of patients in different risk groups to chemotherapy drugs, providing a reference for personalized treatment. CONCLUSION In this study, we examined the relationship between sialylation-related lncRNA and the prognosis of bladder cancer, providing new molecular markers and potential targets for diagnosis and treatment. Our research revealed correlations between sialylation-related lncRNA characteristics and clinicopathological features, potential mechanisms, somatic mutations, immune microenvironment, chemotherapy response, and predicted drug sensitivity in bladder cancer. Additionally, in vitro cellular studies were conducted to validate these findings and lay the groundwork for future clinical applications.
Collapse
Affiliation(s)
- Yi Qiao
- Department of Urology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Xintao Tian
- Department of Urology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Shengxian Li
- Department of Urology, The Affiliated Hospital of Qingdao University, Qingdao, China.
| | - Haitao Niu
- Department of Urology, The Affiliated Hospital of Qingdao University, Qingdao, China.
| |
Collapse
|
42
|
Xie Z, Liao J, Chen J. Targeting macrophages to reprogram the tumor immune microenvironment. BLOOD SCIENCE 2024; 6:e00203. [PMID: 39149533 PMCID: PMC11326473 DOI: 10.1097/bs9.0000000000000203] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2024] [Accepted: 07/30/2024] [Indexed: 08/17/2024] Open
Affiliation(s)
- Zhidong Xie
- Department of Immunology and Microbiology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Jing Liao
- GMU-GIBH Joint School of Life Sciences, The Guangdong-Hong Kong-Macau Joint Laboratory for Cell Fate Regulation and Diseases, Guangzhou Medical University, Guangzhou, China
| | - Jun Chen
- Department of Immunology and Microbiology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
- Guangdong Engineering and Technology Research Center for Disease-Model Animals, Laboratory Animal Center, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
- Key Laboratory of Tropical Disease Control of the Ministry of Education, Sun Yat-sen University, Guangzhou, China
- Jinfeng Laboratory, Chongqing, China
| |
Collapse
|
43
|
Mohamed KA, Kruf S, Büll C. Putting a cap on the glycome: Dissecting human sialyltransferase functions. Carbohydr Res 2024; 544:109242. [PMID: 39167930 DOI: 10.1016/j.carres.2024.109242] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2024] [Revised: 07/24/2024] [Accepted: 08/13/2024] [Indexed: 08/23/2024]
Abstract
Human glycans are capped with sialic acids and these nine-carbon sugars mediate many of the biological functions and interactions of glycans. Structurally diverse sialic acid caps mark human cells as self and they form the ligands for the Siglec immune receptors and other glycan-binding proteins. Sialic acids enable host interactions with the human microbiome and many human pathogens utilize sialic acids to infect host cells. Alterations in sialic acid-carrying glycans, sialoglycans, can be found in every major human disease including inflammatory conditions and cancer. Twenty sialyltransferase family members in the Golgi apparatus of human cells transfer sialic acids to distinct glycans and glycoconjugates. Sialyltransferases catalyze specific reactions to form unique sialoglycans or they have shared functions where multiple family members generate the same sialoglycan product. Moreover, some sialyltransferases compete for the same glycan substrate, but create different sialic acid caps. The redundant and competing functions make it difficult to understand the individual roles of the human sialyltransferases in biology and to reveal the specific contributions to pathobiological processes. Recent insights hint towards the existence of biosynthetic rules formed by the individual functions of sialyltransferases, their interactions, and cues from the local Golgi environment that coordinate sialoglycan biosynthesis. In this review, we discuss the current structural and functional understanding of the human sialyltransferase family and we review recent technological advances that enable the dissection of individual sialyltransferase activities.
Collapse
Affiliation(s)
- Khadra A Mohamed
- Department of Biomolecular Chemistry, Institute for Molecules and Materials, Faculty of Science, Radboud University Nijmegen, Heyendaalseweg 135, 6525AJ, Nijmegen, the Netherlands
| | - Stijn Kruf
- Department of Biomolecular Chemistry, Institute for Molecules and Materials, Faculty of Science, Radboud University Nijmegen, Heyendaalseweg 135, 6525AJ, Nijmegen, the Netherlands
| | - Christian Büll
- Department of Biomolecular Chemistry, Institute for Molecules and Materials, Faculty of Science, Radboud University Nijmegen, Heyendaalseweg 135, 6525AJ, Nijmegen, the Netherlands.
| |
Collapse
|
44
|
Zhang Z, Li X, Huang M, Huang Y, Tan X, Dong Y, Huang Y, Jian J. Siglec7 functions as an inhibitory receptor of non-specific cytotoxic cells and can regulate the innate immune responses in a primitive vertebrate (Oreochromis niloticus). Int J Biol Macromol 2024; 278:134851. [PMID: 39168212 DOI: 10.1016/j.ijbiomac.2024.134851] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2024] [Revised: 07/29/2024] [Accepted: 08/16/2024] [Indexed: 08/23/2024]
Abstract
In mammals, siglec7, an integral component of the siglecs, is principally found on the surface of natural killer (NK) cells, macrophages, and monocytes, where it interacts with various pathogens to perform immunological regulatory activities. Nonetheless, the immune defense and mechanism of siglec7 in early vertebrates remain unknown. In this study, we identified siglec7 from Oreochromis niloticus (OnSiglec7) and revealed its immune functions. Specifically, OnSiglec7 was abundantly expressed in immune-related tissues of healthy tilapia and its transcription level was strongly activated after being challenged with A. hydrophila, S. agalactiae, and Poly: IC. Meanwhile, OnSiglec7 protein was purified and analyzed, which could recognize multiple pathogens through binding and agglutinating activity. Moreover, OnSiglec7-positive cells were mainly distributed in non-specific cytotoxic cells (NCC) of tilapia HKLs and showed cell membrane localization. Furthermore, OnSiglec7 blockage affected multiple innate immune responses (inflammation, apoptosis, and pyroptosis process) by regulating the activation of MAPK, NF-κB, TLR, and JAK-STAT pathways. Finally, OnSiglec7 blockage also greatly enhanced the cytotoxic effect of tilapia NCC. Summarily, this study uncovers immune functions and mechanisms of siglec7 in primitive vertebrates, thereby enhancing our understanding of the systemic evolution and ancient functions of other siglecs within the host's innate immune system (to our knowledge).
Collapse
Affiliation(s)
- Zhiqiang Zhang
- College of Fishery, Guangdong Ocean University, Guangdong Provincial Key Laboratory of Aquatic Animal Disease Control and Healthy Culture, Zhanjiang, China; Laboratory for Marine Biology and Biotechnology, Qingdao National Laboratory for Marine Science and Technology, Qingdao, China
| | - Xing Li
- College of Fishery, Guangdong Ocean University, Guangdong Provincial Key Laboratory of Aquatic Animal Disease Control and Healthy Culture, Zhanjiang, China
| | - Meiling Huang
- College of Fishery, Guangdong Ocean University, Guangdong Provincial Key Laboratory of Aquatic Animal Disease Control and Healthy Culture, Zhanjiang, China
| | - Yongxiong Huang
- College of Fishery, Guangdong Ocean University, Guangdong Provincial Key Laboratory of Aquatic Animal Disease Control and Healthy Culture, Zhanjiang, China
| | - Xuyan Tan
- College of Fishery, Guangdong Ocean University, Guangdong Provincial Key Laboratory of Aquatic Animal Disease Control and Healthy Culture, Zhanjiang, China
| | - Yuhang Dong
- College of Fishery, Guangdong Ocean University, Guangdong Provincial Key Laboratory of Aquatic Animal Disease Control and Healthy Culture, Zhanjiang, China
| | - Yu Huang
- College of Fishery, Guangdong Ocean University, Guangdong Provincial Key Laboratory of Aquatic Animal Disease Control and Healthy Culture, Zhanjiang, China; Laboratory for Marine Biology and Biotechnology, Qingdao National Laboratory for Marine Science and Technology, Qingdao, China; Guangdong Provincial Engineering Research Center for Aquatic Animal Health Assessment, Shenzhen, China.
| | - Jichang Jian
- College of Fishery, Guangdong Ocean University, Guangdong Provincial Key Laboratory of Aquatic Animal Disease Control and Healthy Culture, Zhanjiang, China; Laboratory for Marine Biology and Biotechnology, Qingdao National Laboratory for Marine Science and Technology, Qingdao, China; Guangdong Provincial Engineering Research Center for Aquatic Animal Health Assessment, Shenzhen, China.
| |
Collapse
|
45
|
Park S, Choi S, Shimpi AA, Estroff LA, Fischbach C, Paszek MJ. Collagen Mineralization Decreases NK Cell-Mediated Cytotoxicity of Breast Cancer Cells via Increased Glycocalyx Thickness. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2311505. [PMID: 38279892 PMCID: PMC11471288 DOI: 10.1002/adma.202311505] [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: 10/31/2023] [Revised: 01/14/2024] [Indexed: 01/29/2024]
Abstract
Skeletal metastasis is common in patients with advanced breast cancer and often caused by immune evasion of disseminated tumor cells (DTCs). In the skeleton, tumor cells not only disseminate to the bone marrow but also to osteogenic niches in which they interact with newly mineralizing bone extracellular matrix (ECM). However, it remains unclear how mineralization of collagen type I, the primary component of bone ECM, regulates tumor-immune cell interactions. Here, a combination of synthetic bone matrix models with controlled mineral content, nanoscale optical imaging, and flow cytometry are utilized to evaluate how collagen type I mineralization affects the biochemical and biophysical properties of the tumor cell glycocalyx, a dense layer of glycosylated proteins and lipids decorating their cell surface. These results suggest that collagen mineralization upregulates mucin-type O-glycosylation and sialylation by tumor cells, which increases their glycocalyx thickness while enhancing resistance to attack by natural killer (NK) cells. These changes are functionally linked as treatment with a sialylation inhibitor decreased mineralization-dependent glycocalyx thickness and made tumor cells more susceptible to NK cell attack. Together, these results suggest that interference with glycocalyx sialylation may represent a therapeutic strategy to enhance cancer immunotherapies targeting bone-metastatic breast cancer.
Collapse
Affiliation(s)
- Sangwoo Park
- Graduate Field of Biophysics, Cornell University, Ithaca, NY 14853, USA
- Robert Frederick Smith School of Chemical and Biomolecular Engineering, Cornell University, Ithaca, NY 14853, USA
| | - Siyoung Choi
- Nancy E. and Peter C. Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY 14853, USA
| | - Adrian A. Shimpi
- Nancy E. and Peter C. Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY 14853, USA
| | - Lara A. Estroff
- Department of Materials Science and Engineering, Cornell University, Ithaca, NY, 14853, USA
- Kavli Institute at Cornell for Nanoscale Science, Cornell University, Ithaca, NY, 14853, USA
| | - Claudia Fischbach
- Nancy E. and Peter C. Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY 14853, USA
- Kavli Institute at Cornell for Nanoscale Science, Cornell University, Ithaca, NY, 14853, USA
| | - Matthew J. Paszek
- Graduate Field of Biophysics, Cornell University, Ithaca, NY 14853, USA
- Robert Frederick Smith School of Chemical and Biomolecular Engineering, Cornell University, Ithaca, NY 14853, USA
- Kavli Institute at Cornell for Nanoscale Science, Cornell University, Ithaca, NY, 14853, USA
| |
Collapse
|
46
|
Cao C, Fu G, Xu R, Li N. Coupling of Alzheimer's Disease Genetic Risk Factors with Viral Susceptibility and Inflammation. Aging Dis 2024; 15:2028-2050. [PMID: 37962454 PMCID: PMC11346407 DOI: 10.14336/ad.2023.1017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Accepted: 10/17/2023] [Indexed: 11/15/2023] Open
Abstract
Alzheimer's disease (AD) is a neurodegenerative disease characterized by persistent cognitive decline. Amyloid plaque deposition and neurofibrillary tangles are the main pathological features of AD brain, though mechanisms leading to the formation of lesions remain to be understood. Genetic efforts through genome-wide association studies (GWAS) have identified dozens of risk genes influencing the pathogenesis and progression of AD, some of which have been revealed in close association with increased viral susceptibilities and abnormal inflammatory responses in AD patients. In the present study, we try to present a list of AD candidate genes that have been shown to affect viral infection and inflammatory responses. Understanding of how AD susceptibility genes interact with the viral life cycle and potential inflammatory pathways would provide possible therapeutic targets for both AD and infectious diseases.
Collapse
Affiliation(s)
| | | | - Ruodan Xu
- Department of Biomedical Engineering and Technology, Institute of Basic Theory for Chinese Medicine, China Academy of Chinese Medical Sciences, Beijing, 100700, China.
| | - Ning Li
- Department of Biomedical Engineering and Technology, Institute of Basic Theory for Chinese Medicine, China Academy of Chinese Medical Sciences, Beijing, 100700, China.
| |
Collapse
|
47
|
Murphy PV, Dhara A, Fitzgerald LS, Hever E, Konda S, Mandal K. Small lectin ligands as a basis for applications in glycoscience and glycomedicine. Chem Soc Rev 2024; 53:9428-9445. [PMID: 39162695 DOI: 10.1039/d4cs00642a] [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: 08/21/2024]
Abstract
Glycan recognition by lectins mediates important biological events. This Tutorial Review aims to introduce lectin-ligand interactions and show how these molecular recognition events inspire innovations such as: (i) glycomimetic ligands; (ii) multivalent ligand agonists/antagonists; (iii) ligands for precision delivery of therapies to cells, where therapies include vaccines, siRNA and LYTACs (iv) development of diagnostics. A small number of case studies are selected to demonstrate principles for development of new ligands for applications inspired by knowledge of natural glycan ligand structure and function.
Collapse
Affiliation(s)
- Paul V Murphy
- School of Biological and Chemical Sciences, Galway, H91TK33, Ireland.
- SSPC, SFI Research Centre for Pharmaceuticals, Galway, H91TK33, Ireland
- CÚRAM, SFI Research Centre for Medical Devices, University of Galway, University Road, Galway, H91TK33, Ireland
| | - Ashis Dhara
- School of Biological and Chemical Sciences, Galway, H91TK33, Ireland.
- SSPC, SFI Research Centre for Pharmaceuticals, Galway, H91TK33, Ireland
| | - Liam S Fitzgerald
- School of Biological and Chemical Sciences, Galway, H91TK33, Ireland.
- SSPC, SFI Research Centre for Pharmaceuticals, Galway, H91TK33, Ireland
- CÚRAM, SFI Research Centre for Medical Devices, University of Galway, University Road, Galway, H91TK33, Ireland
| | - Eoin Hever
- School of Biological and Chemical Sciences, Galway, H91TK33, Ireland.
| | - Saidulu Konda
- School of Biological and Chemical Sciences, Galway, H91TK33, Ireland.
| | - Kishan Mandal
- School of Biological and Chemical Sciences, Galway, H91TK33, Ireland.
| |
Collapse
|
48
|
Lensch V, Johnson JA, Kiessling LL. Glycoconjugate vaccines: platforms and adjuvants for directed immunity. Glycobiology 2024; 34:cwae092. [PMID: 39593193 PMCID: PMC11604072 DOI: 10.1093/glycob/cwae092] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2024] [Revised: 11/03/2024] [Accepted: 11/23/2024] [Indexed: 11/28/2024] Open
Abstract
Central to immune recognition is the glycocalyx, a glycan-rich coat on all cells that plays a crucial role in interactions that enable pathogen detection and activation of immune defenses. Pathogens and cancerous cells often display distinct glycans on their surfaces, making these saccharide antigens prime targets for vaccine development. However, carbohydrates alone generally serve as poor immunogens due to their often weak binding affinities, inability to effectively recruit T cell help, and reliance on adjuvants to iboost immune activation. The introduction of glycoconjugate vaccines, initially involving the covalent coupling of carbohydrate antigens to carrier proteins, marked a pivotal advancement by facilitating neutralizing antibody production against carbohydrate targets. Despite successes in generating glycoconjugate vaccines against certain bacterial diseases, challenges persist in creating effective vaccines against numerous intracellular pathogens and non-communicable diseases such as cancer. In this review, we highlight new developments in conjugate vaccine platforms aim to overcome these limitations by optimizing the display of glycan and T cell epitopes as well as incorporating defined carbohydrate adjuvants to direct tailored immune responses. These advancements promise to improve the effectiveness of carbohydrate-based vaccines and broaden their coverage against a wide range of diseases.
Collapse
Affiliation(s)
- Valerie Lensch
- Department of Chemistry, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, MA 02139, United States
| | - Jeremiah A Johnson
- Department of Chemistry, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, MA 02139, United States
- Koch Institute for Integrative Cancer Research,Massachusetts Institute of Technology, 500 Main Street, Cambridge, MA 02139, United States
- Broad Institute of MIT and Harvard, 415 Main Street, Cambridge, MA 02142, United States
| | - Laura L Kiessling
- Department of Chemistry, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, MA 02139, United States
- Koch Institute for Integrative Cancer Research,Massachusetts Institute of Technology, 500 Main Street, Cambridge, MA 02139, United States
- Broad Institute of MIT and Harvard, 415 Main Street, Cambridge, MA 02142, United States
| |
Collapse
|
49
|
Yang M, Jiang D, Lai W, Chen K, Xu S, Yu R, Li L, Zhang L, Lu L, Xu Y, Liu Y, Jiang J. Condensed tannin improves growth and alleviates intestinal inflammation of juvenile largemouth bass (Micropterus salmoides) fed with high cottonseed protein concentrate diet. Int J Biol Macromol 2024; 280:135874. [PMID: 39307492 DOI: 10.1016/j.ijbiomac.2024.135874] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2024] [Revised: 09/07/2024] [Accepted: 09/19/2024] [Indexed: 09/30/2024]
Abstract
The objective of this study was to examine the potential of condensed tannin (CT) in mitigating the adverse effects on growth and intestinal health induced by high cottonseed concentrate protein (CPC) diets in juvenile largemouth bass (Micropterus salmoides). Largemouth bass were respectively fed with the basic diet, the high CPC diet, and the CPC + CT diet (incorporated 3.75 g/kg CT into the high CPC diet) for a duration of 8 weeks. Results indicated that the high CPC diet resulted in decreased growth performance and compromised intestinal health. Dietary CT enhanced the growth of fish, improved intestinal function, and optimized intestinal microbiota. Additionally, intestinal transcriptome analysis revealed that dietary CT might mitigate intestinal inflammation by downregulating the related gene expression in the cell adhesion molecule pathway. Furthermore, the gene expression of cd22 and mhc2 was positively correlated with the relative abundance of the Geodermatophilus, an indicator species of intestinal microbiota in high CPC treatment. Our research suggests that the inclusion of CT (3.75 g/kg) in the high CPC diet of largemouth bass can stimulate growth and alleviate negative impacts on intestinal health, indicating that CT can be utilized to enhance the utilization of CPC in fish nutrition.
Collapse
Affiliation(s)
- Manqi Yang
- College of Chemical Engineering, Huaqiao University, Xiamen 361021, China; Academy of Advanced Carbon Conversion Technology, Fujian Provincial Key Laboratory of Biomass Low-Carbon Conversion, Huaqiao University, Xiamen 361021, China
| | - Dahai Jiang
- College of Chemical Engineering, Huaqiao University, Xiamen 361021, China; Academy of Advanced Carbon Conversion Technology, Fujian Provincial Key Laboratory of Biomass Low-Carbon Conversion, Huaqiao University, Xiamen 361021, China
| | - Weibin Lai
- College of Chemical Engineering, Huaqiao University, Xiamen 361021, China; Academy of Advanced Carbon Conversion Technology, Fujian Provincial Key Laboratory of Biomass Low-Carbon Conversion, Huaqiao University, Xiamen 361021, China
| | - Kai Chen
- College of Chemical Engineering, Huaqiao University, Xiamen 361021, China; Academy of Advanced Carbon Conversion Technology, Fujian Provincial Key Laboratory of Biomass Low-Carbon Conversion, Huaqiao University, Xiamen 361021, China
| | - Shuwen Xu
- College of Chemical Engineering, Huaqiao University, Xiamen 361021, China
| | - Ran Yu
- College of Chemical Engineering, Huaqiao University, Xiamen 361021, China
| | - Linyi Li
- College of Chemical Engineering, Huaqiao University, Xiamen 361021, China
| | - Liangliang Zhang
- College of Chemical Engineering, Huaqiao University, Xiamen 361021, China; Academy of Advanced Carbon Conversion Technology, Fujian Provincial Key Laboratory of Biomass Low-Carbon Conversion, Huaqiao University, Xiamen 361021, China.
| | - Liming Lu
- College of Chemical Engineering, Huaqiao University, Xiamen 361021, China; Academy of Advanced Carbon Conversion Technology, Fujian Provincial Key Laboratory of Biomass Low-Carbon Conversion, Huaqiao University, Xiamen 361021, China
| | - Yong Xu
- College of Chemical Engineering, Nanjing Forestry University, Nanjing 210000, China
| | - Yiwen Liu
- Wufeng Chicheng Biotech Co., Ltd., Yichang 443000, China
| | - Jianchun Jiang
- College of Chemical Engineering, Huaqiao University, Xiamen 361021, China; Academy of Advanced Carbon Conversion Technology, Fujian Provincial Key Laboratory of Biomass Low-Carbon Conversion, Huaqiao University, Xiamen 361021, China; Institute of Chemical Industry of Forest Products, CAF, Nanjing 210042, China
| |
Collapse
|
50
|
van Bruggen JAC, Peters FS, Mes M, Rietveld JM, Cerretani E, Cretenet G, van Kampen R, Jongejan A, Moerland PD, Melenhorst JJ, van der Windt GJW, Eldering E, Kater AP. T-cell dysfunction in CLL is mediated through expression of Siglec-10 ligands CD24 and CD52 on CLL cells. Blood Adv 2024; 8:4633-4646. [PMID: 39042920 PMCID: PMC11401197 DOI: 10.1182/bloodadvances.2023011934] [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: 10/16/2023] [Revised: 07/09/2024] [Accepted: 07/11/2024] [Indexed: 07/25/2024] Open
Abstract
ABSTRACT Autologous T-cell-based therapies, such as chimeric antigen receptor (CAR) T-cell therapy, exhibit low success rates in chronic lymphocytic leukemia (CLL) and correlate with a dysfunctional T-cell phenotype observed in patients. Despite various proposed mechanisms of T-cell dysfunction in CLL, the specific CLL-derived factors responsible remain unidentified. This study aimed to investigate the mechanisms through which CLL cells suppress CAR T-cell activation and function. We found that CLL-derived T cells get activated, albeit in a delayed fashion, and specifically that restimulation of CAR T cells in the presence of CLL cells causes impaired cytokine production and reduced proliferation. Notably, coculture of T cells with CD40-activated CLL cells did not lead to T-cell dysfunction, and this required direct cell contact between the CD40-stimulated CLL cells and T cells. Inhibition of kinases involved in the CD40 signaling cascade revealed that the Spare Respiratory Capacity (SRC) kinase inhibitor dasatinib prevented rescue of T-cell function independent of CD40-mediated increased levels of costimulatory and adhesion ligands on CLL cells. Transcriptome profiling of CD40-stimulated CLL cells with or without dasatinib identified widespread differential gene expression. Selecting for surface receptor genes revealed CD40-mediated downregulation of the Sialic acid-binding Ig-like lectin 10 (Siglec-10) ligands CD24 and CD52, which was prevented by dasatinib, suggesting a role for these ligands in functional T-cell suppression in CLL. Indeed, blocking CD24 and/or CD52 markedly reduced CAR T-cell dysfunction upon coculture with resting CLL cells. These results demonstrated that T cells derived from CLL patients can be reinvigorated by manipulating CLL-T-cell interactions. Targeting CD24- and CD52-mediated CLL-T-cell interaction could be a promising therapeutic strategy to enhance T-cell function in CLL.
Collapse
MESH Headings
- Humans
- Leukemia, Lymphocytic, Chronic, B-Cell/metabolism
- Leukemia, Lymphocytic, Chronic, B-Cell/immunology
- Leukemia, Lymphocytic, Chronic, B-Cell/therapy
- CD52 Antigen/metabolism
- T-Lymphocytes/metabolism
- T-Lymphocytes/immunology
- CD24 Antigen/metabolism
- Lymphocyte Activation/immunology
- Ligands
- Receptors, Chimeric Antigen/metabolism
Collapse
Affiliation(s)
- Jaco A. C. van Bruggen
- Department of Hematology, Cancer Center Amsterdam, Lymphoma and Myeloma Center Amsterdam, Amsterdam University Medical Center, University of Amsterdam, Amsterdam, The Netherlands
- Department of Experimental Immunology, Amsterdam Infection & Immunity Institute, Amsterdam University Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | - Fleur S. Peters
- Department of Hematology, Cancer Center Amsterdam, Lymphoma and Myeloma Center Amsterdam, Amsterdam University Medical Center, University of Amsterdam, Amsterdam, The Netherlands
- Department of Experimental Immunology, Amsterdam Infection & Immunity Institute, Amsterdam University Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | - Morris Mes
- Department of Hematology, Cancer Center Amsterdam, Lymphoma and Myeloma Center Amsterdam, Amsterdam University Medical Center, University of Amsterdam, Amsterdam, The Netherlands
- Department of Experimental Immunology, Amsterdam Infection & Immunity Institute, Amsterdam University Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | - Joanne M. Rietveld
- Department of Hematology, Cancer Center Amsterdam, Lymphoma and Myeloma Center Amsterdam, Amsterdam University Medical Center, University of Amsterdam, Amsterdam, The Netherlands
- Department of Experimental Immunology, Amsterdam Infection & Immunity Institute, Amsterdam University Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | - Elisa Cerretani
- Department of Hematology, Cancer Center Amsterdam, Lymphoma and Myeloma Center Amsterdam, Amsterdam University Medical Center, University of Amsterdam, Amsterdam, The Netherlands
- Department of Experimental Immunology, Amsterdam Infection & Immunity Institute, Amsterdam University Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | - Gaspard Cretenet
- Department of Hematology, Cancer Center Amsterdam, Lymphoma and Myeloma Center Amsterdam, Amsterdam University Medical Center, University of Amsterdam, Amsterdam, The Netherlands
- Department of Experimental Immunology, Amsterdam Infection & Immunity Institute, Amsterdam University Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | | | - Aldo Jongejan
- Department of Epidemiology and Data Science, Amsterdam University Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | - Perry D. Moerland
- Department of Epidemiology and Data Science, Amsterdam University Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | - J. Joseph Melenhorst
- Cleveland Clinic, Lerner Research Institute, Center for Immunotherapy & Precision Immuno-Oncology, Cleveland, OH
| | - Gerritje J. W. van der Windt
- Department of Hematology, Cancer Center Amsterdam, Lymphoma and Myeloma Center Amsterdam, Amsterdam University Medical Center, University of Amsterdam, Amsterdam, The Netherlands
- Department of Experimental Immunology, Amsterdam Infection & Immunity Institute, Amsterdam University Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | - Eric Eldering
- Department of Hematology, Cancer Center Amsterdam, Lymphoma and Myeloma Center Amsterdam, Amsterdam University Medical Center, University of Amsterdam, Amsterdam, The Netherlands
- Department of Experimental Immunology, Amsterdam Infection & Immunity Institute, Amsterdam University Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | - Arnon P. Kater
- Department of Hematology, Cancer Center Amsterdam, Lymphoma and Myeloma Center Amsterdam, Amsterdam University Medical Center, University of Amsterdam, Amsterdam, The Netherlands
- Department of Experimental Immunology, Amsterdam Infection & Immunity Institute, Amsterdam University Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| |
Collapse
|