1
|
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
|
2
|
Drummond-Guy O, Daly J, Wu A, Stewart N, Milne K, Duff C, Nelson BH, Williams KC, Wisnovsky S. Polysialic acid is upregulated on activated immune cells and negatively regulates anticancer immune activity. Front Oncol 2025; 15:1520948. [PMID: 40182033 PMCID: PMC11965634 DOI: 10.3389/fonc.2025.1520948] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2024] [Accepted: 02/20/2025] [Indexed: 04/05/2025] Open
Abstract
Suppression of anticancer immune function is a key driver of tumorigenesis. Identifying molecular pathways that inhibit anticancer immunity is critical for developing novel immunotherapeutics. One such molecule that has recently been identified is the carbohydrate polysialic acid (polySia), whose expression is dramatically upregulated on both cancer cells and immune cells in breast cancer patient tissues. The role of polySia in the anticancer immune response, however, remains incompletely understood. In this study, we profile polySia expression on both healthy primary immune cells and on infiltrating immune cells in the tumour microenvironment (TME). These studies reveal polySia expression on multiple immune cell subsets in patient breast tumors. We find that stimulation of primary T-cells and macrophages in vitro induces a significant upregulation of polySia expression. We subsequently show that polySia is appended to a range of different carrier proteins within these immune cells. Finally, we find that selective removal of polySia can significantly potentiate killing of breast cancer cells by innate immune cells. These studies implicate polySia as a significant negative regulator of anticancer immunity.
Collapse
Affiliation(s)
- Olivia Drummond-Guy
- Faculty of Pharmaceutical Sciences, University of British Columbia, Vancouver, BC, Canada
| | - John Daly
- Faculty of Pharmaceutical Sciences, University of British Columbia, Vancouver, BC, Canada
| | - Angeline Wu
- Faculty of Pharmaceutical Sciences, University of British Columbia, Vancouver, BC, Canada
| | - Natalie Stewart
- Faculty of Pharmaceutical Sciences, University of British Columbia, Vancouver, BC, Canada
| | - Katy Milne
- Deeley Research Centre, British Columbia (BC) Cancer, Victoria, BC, Canada
| | - Chloe Duff
- Deeley Research Centre, British Columbia (BC) Cancer, Victoria, BC, Canada
| | - Brad H. Nelson
- Deeley Research Centre, British Columbia (BC) Cancer, Victoria, BC, Canada
- Department of Medical Genetics, University of British Columbia, Vancouver, BC, Canada
- Department of Biochemistry and Microbiology, University of Victoria, Victoria, BC, Canada
| | - Karla C. Williams
- Faculty of Pharmaceutical Sciences, University of British Columbia, Vancouver, BC, Canada
| | - Simon Wisnovsky
- Faculty of Pharmaceutical Sciences, University of British Columbia, Vancouver, BC, Canada
| |
Collapse
|
3
|
Goode EA, Orozco-Moreno M, Hodgson K, Nabilah A, Murali M, Peng Z, Merx J, Rossing E, Pijnenborg JFA, Boltje TJ, Wang N, Elliott DJ, Munkley J. Sialylation Inhibition Can Partially Revert Acquired Resistance to Enzalutamide in Prostate Cancer Cells. Cancers (Basel) 2024; 16:2953. [PMID: 39272811 PMCID: PMC11393965 DOI: 10.3390/cancers16172953] [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: 07/18/2024] [Revised: 08/08/2024] [Accepted: 08/22/2024] [Indexed: 09/15/2024] Open
Abstract
Prostate cancer is a lethal solid malignancy and a leading cause of cancer-related deaths in males worldwide. Treatments, including radical prostatectomy, radiotherapy, and hormone therapy, are available and have improved patient survival; however, recurrence remains a huge clinical challenge. Enzalutamide is a second-generation androgen receptor antagonist that is used to treat castrate-resistant prostate cancer. Among patients who initially respond to enzalutamide, virtually all acquire secondary resistance, and an improved understanding of the mechanisms involved is urgently needed. Aberrant glycosylation, and, in particular, alterations to sialylated glycans, have been reported as mediators of therapy resistance in cancer, but a link between tumour-associated glycans and resistance to therapy in prostate cancer has not yet been investigated. Here, using cell line models, we show that prostate cancer cells with acquired resistance to enzalutamide therapy have an upregulation of the sialyltransferase ST6 beta-galactoside alpha-2,6-sialyltransferase 1 (ST6GAL1) and increased levels of α2,6-sialylated N-glycans. Furthermore, using the sialyltransferase inhibitor P-SiaFNEtoc, we discover that acquired resistance to enzalutamide can be partially reversed by combining enzalutamide therapy with sialic acid blockade. Our findings identify a potential role for ST6GAL1-mediated aberrant sialylation in acquired resistance to enzalutamide therapy for prostate cancer and suggest that sialic acid blockade in combination with enzalutamide may represent a novel therapeutic approach in patients with advanced disease. Our study also highlights the potential to bridge the fields of cancer biology and glycobiology to develop novel combination therapies for prostate cancer.
Collapse
Affiliation(s)
- Emily Archer Goode
- Newcastle University Centre for Cancer, Newcastle University Institute of Biosciences, Newcastle NE1 3BZ, UK
| | - Margarita Orozco-Moreno
- Newcastle University Centre for Cancer, Newcastle University Institute of Biosciences, Newcastle NE1 3BZ, UK
| | - Kirsty Hodgson
- Newcastle University Centre for Cancer, Newcastle University Institute of Biosciences, Newcastle NE1 3BZ, UK
| | - Amirah Nabilah
- Newcastle University Centre for Cancer, Newcastle University Institute of Biosciences, Newcastle NE1 3BZ, UK
| | - Meera Murali
- Newcastle University Centre for Cancer, Newcastle University Institute of Biosciences, Newcastle NE1 3BZ, UK
| | - Ziqian Peng
- Newcastle University Centre for Cancer, Newcastle University Institute of Biosciences, Newcastle NE1 3BZ, UK
| | - Jona Merx
- Synthetic Organic Chemistry, Institute for Molecules and Materials, Radboud University, 6525 XZ Nijmegen, The Netherlands
| | - Emiel Rossing
- GlycoTherapeutics B.V., 6511 AJ Nijmegen, The Netherlands
| | | | - Thomas J Boltje
- Synthetic Organic Chemistry, Institute for Molecules and Materials, Radboud University, 6525 XZ Nijmegen, The Netherlands
- GlycoTherapeutics B.V., 6511 AJ Nijmegen, The Netherlands
| | - Ning Wang
- The Mellanby Centre for Musculoskeletal Research, Division of Clinical Medicine, The University of Sheffield, Sheffield S10 2TN, UK
- Leicester Cancer Research Centre, Department of Genetics and Genome Biology, University of Leicester, Leicester LE2 7LX, UK
| | - David J Elliott
- Newcastle University Centre for Cancer, Newcastle University Institute of Biosciences, Newcastle NE1 3BZ, UK
| | - Jennifer Munkley
- Newcastle University Centre for Cancer, Newcastle University Institute of Biosciences, Newcastle NE1 3BZ, UK
| |
Collapse
|
4
|
Gao G, Liu R, Wu D, Gao D, Lv Y, Xu X, Fu B, Lin Z, Wang T, He A, Bai J. Risk score constructed with neutrophil extracellular traps-related genes predicts prognosis and immune microenvironment in multiple myeloma. Front Oncol 2024; 14:1365460. [PMID: 38919521 PMCID: PMC11196624 DOI: 10.3389/fonc.2024.1365460] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2024] [Accepted: 05/28/2024] [Indexed: 06/27/2024] Open
Abstract
Background Multiple myeloma (MM) exhibits considerable heterogeneity in treatment responses and survival rates, even when standardized care is administered. Ongoing efforts are focused on developing prognostic models to predict these outcomes more accurately. Recently, neutrophil extracellular traps (NETs) have emerged as a potential factor in MM progression, sparking investigation into their role in prognostication. Methods In this study, a multi-gene risk scoring model was constructed using the intersection of NTEs and differentially expressed genes (DEGs), applying the least absolute shrinkage and selection operator (LASSO) Cox regression model. A nomogram was established, and the prognostic model's effectiveness was determined via Kaplan-Meier survival analysis, receiver operating characteristic (ROC) curve, and decision curve analysis (DCA). The ESTIMATE algorithm and immune-related single-sample gene set enrichment analysis (ssGSEA) were employed to evaluate the level of immune infiltration. The sensitivity of chemotherapy drugs was assessed using the Genomics of Drug Sensitivity in Cancer (GDSC) database. Ultimately, the presence of the detected genes was confirmed through quantitative real-time polymerase chain reaction (qRT-PCR) analysis in MM cell specimens. Results 64 NETs-DEGs were yielded, and through univariate Cox regression and LASSO regression analysis, we constructed a risk score composed of six genes: CTSG, HSPE1, LDHA, MPO, PINK1, and VCAM1. MM patients in three independent datasets were classified into high- and low-risk groups according to the risk score. The overall survival (OS) of patients in the high-risk group was significantly reduced compared to the low-risk group. Furthermore, the risk score was an independent predictive factor for OS. In addition, interactions between the risk score, immune score, and immune cell infiltration were investigated. Further analysis indicated that patients in the high-risk group were more sensitive to a variety of chemotherapy and targeted drugs, including bortezomib. Moreover, the six genes provided insights into the progression of plasma cell disorders. Conclusion This study offers novel insights into the roles of NETs in prognostic prediction, immune status, and drug sensitivity in MM, serving as a valuable supplement and enhancement to existing grading systems.
Collapse
Affiliation(s)
- Gongzhizi Gao
- Department of Hematology, The Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
| | - Rui Liu
- Department of Hematology, The Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
| | - Dong Wu
- Department of Hematology, The Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
| | - Dandan Gao
- Department of Hematology, The Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
| | - Yang Lv
- Department of Hematology, The Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
| | - Xuezhu Xu
- Department of Hematology, The Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
| | - Bingjie Fu
- Department of Hematology, The Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
| | - Zujie Lin
- Department of Hematology, The Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
| | - Ting Wang
- Department of Hematology, The Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
| | - Aili He
- Department of Hematology, The Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
- National-Local Joint Engineering Research Center of Biodiagnostics & Biotherapy, The Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
- Xi’an Key Laboratory of hematological diseases, Xi’an, China
| | - Ju Bai
- Department of Hematology, The Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
- Xi’an Key Laboratory of hematological diseases, Xi’an, China
| |
Collapse
|
5
|
Wang B, Hou C, Yu X, Liu J, Wang J. The prognostic value of sialylation-related long non-coding RNAs in lung adenocarcinoma. Sci Rep 2024; 14:8879. [PMID: 38632255 PMCID: PMC11024174 DOI: 10.1038/s41598-024-59130-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2023] [Accepted: 04/08/2024] [Indexed: 04/19/2024] Open
Abstract
There has been increasing interest in the role of epigenetic modification in cancers recently. Among the various modifications, sialylation has emerged as a dominant subtype implicated in tumor progression, metastasis, immune evasion, and chemoresistance. The prognostic significance of sialylation-related molecules has been demonstrated in colorectal cancer. However, the potential roles and regulatory mechanisms of sialylation in lung adenocarcinoma (LUAD) have not been thoroughly investigated. Through Pearson correlation, univariate Cox hazards proportional regression, and random survival forest model analyses, we identified several prognostic long non-coding RNAs (lncRNAs) associated with aberrant sialylation and tumor progression, including LINC00857, LINC00968, LINC00663, and ITGA9-AS1. Based on the signatures of four lncRNAs, we classified patients into two clusters with different landscapes using a non-negative matrix factorization approach. Collectively, patients in Cluster 1 (C1) exhibited worse prognoses than those in Cluster 2 (C2), as well as heavier tumor mutation burden. Functional enrichment analysis showed the enrichment of several pro-tumor pathways in C1, differing from the upregulated Longevity and programmed cell death pathways in C2. Moreover, we profiled immune infiltration levels of important immune cell lineages in two subgroups using MCPcounter scores and single sample gene set enrichment analysis scores, revealing a relatively immunosuppressive microenvironment in C1. Risk analysis indicated that LINC00857 may serve as a pro-tumor regulator, while the other three lncRNAs may be protective contributors. Consistently, we observed upregulated LINC00857 in C1, whereas increased expressive levels of LINC00968, LINC00663, and ITGA9-AS1 were observed in C2. Finally, drug sensitivity analysis suggested that patients in the two groups may benefit from different therapeutic strategies, contributing to precise treatment in LUAD. By integrating multi-omics data, we identified four core sialylation-related lncRNAs and successfully established a prognostic model to distinguish patients with different characterizations. These findings may provide some insights into the underlying mechanism of sialylation, and offer a new stratification way as well as clinical guidance in LUAD.
Collapse
Grants
- 2022ZD08 National Traditional Chinese Medicine Inheritance and Innovation Center, the First Clinical Medical College of Guangzhou University of Traditional Chinese Medicine, China
- 2022ZD08 National Traditional Chinese Medicine Inheritance and Innovation Center, the First Clinical Medical College of Guangzhou University of Traditional Chinese Medicine, China
- 2022ZD08 National Traditional Chinese Medicine Inheritance and Innovation Center, the First Clinical Medical College of Guangzhou University of Traditional Chinese Medicine, China
- 20241105 Administration of Traditional Chinese Medicine of Guangdong Province, China
- 20241105 Administration of Traditional Chinese Medicine of Guangdong Province, China
- 20221402 Science and Technology Planning Project of Guangdong Province, China
- 20221402 Science and Technology Planning Project of Guangdong Province, China
- 20221402 Science and Technology Planning Project of Guangdong Province, China
Collapse
Affiliation(s)
- Beiru Wang
- First Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou, 510405, Guangdong, China
| | - Chengyu Hou
- First Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou, 510405, Guangdong, China
| | - Xiang Yu
- First Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou, 510405, Guangdong, China
| | - Jiaxin Liu
- First Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou, 510405, Guangdong, China
| | - Jiyong Wang
- Department of Thoracic Surgery, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, 510405, Guangdong, China.
| |
Collapse
|
6
|
O'Dwyer M, Kirkham-McCarthy L, Cerreto M, Foà R, Natoni A. PSGL-1 decorated with sialyl Lewis a/x promotes high affinity binding of myeloma cells to P-selectin but is dispensable for E-selectin engagement. Sci Rep 2024; 14:1756. [PMID: 38243063 PMCID: PMC10798956 DOI: 10.1038/s41598-024-52212-2] [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: 10/31/2023] [Accepted: 01/16/2024] [Indexed: 01/21/2024] Open
Abstract
Dissemination of multiple myeloma into the bone marrow proceeds through sequential steps mediated by a variety of adhesion molecules and chemokines that eventually results in the extravasation of malignant plasma cells into this protective niche. Selectins are a class of C-type lectins that recognize carbohydrate structures exposed on blood borne cells and participate in the first step of the extravasation cascade, serving as brakes to slow down circulating cells enabling them to establish firm adhesion onto the endothelium. Myeloma cells enriched for the expression of selectin ligands present an aggressive disease in vivo that is refractory to bortezomib treatment and can be reverted by small molecules targeting E-selectin. In this study, we have defined the molecular determinants of the selectin ligands expressed on myeloma cells. We show that PSGL-1 is the main protein carrier of sialyl Lewisa/x-related structures in myeloma. PSGL-1 decorated with sialyl Lewisa/x is essential for P-selectin binding but dispensable for E-selectin binding. Moreover, sialylation is required for E-selectin engagement whereas high affinity binding to P-selectin occurs even in the absence of sialic acid. This study provides further knowledge on the biology of selectin ligands in myeloma, opening the way to their clinical application as diagnostic tools and therapeutic targets.
Collapse
Affiliation(s)
- Michael O'Dwyer
- Translational Research Facility, University of Galway, Galway, Ireland
| | - Lucy Kirkham-McCarthy
- Biomedical Sciences, School of Medicine, National University of Ireland Galway, Galway, Ireland
| | - Marina Cerreto
- Hematology, Department of Translational and Precision Medicine, Sapienza University, Rome, Italy
| | - Robin Foà
- Hematology, Department of Translational and Precision Medicine, Sapienza University, Rome, Italy
| | - Alessandro Natoni
- Hematology, Department of Translational and Precision Medicine, Sapienza University, Rome, Italy.
| |
Collapse
|
7
|
Pozzo F, Tissino E, Zucchetto A, Gattei V. CD49d in chronic lymphocytic leukemia: a molecule with multiple regulation layers. Comment to "Sialylation regulates migration in chronic lymphocytic leukemia". Haematologica 2024; 109:362-363. [PMID: 37199129 PMCID: PMC10772489 DOI: 10.3324/haematol.2023.283237] [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: 03/29/2023] [Accepted: 05/10/2023] [Indexed: 05/19/2023] Open
Abstract
Not available.
Collapse
Affiliation(s)
- Federico Pozzo
- Clinical and Experimental Onco-Haematology Unit, Centro di Riferimento Oncologico di Aviano (CRO) IRCCS, Aviano.
| | - Erika Tissino
- Clinical and Experimental Onco-Haematology Unit, Centro di Riferimento Oncologico di Aviano (CRO) IRCCS, Aviano
| | - Antonella Zucchetto
- Clinical and Experimental Onco-Haematology Unit, Centro di Riferimento Oncologico di Aviano (CRO) IRCCS, Aviano
| | - Valter Gattei
- Clinical and Experimental Onco-Haematology Unit, Centro di Riferimento Oncologico di Aviano (CRO) IRCCS, Aviano
| |
Collapse
|
8
|
Moons SJ, Hornikx DLAH, Aasted MKM, Pijnenborg JFA, Calzari M, White PB, Narimatsu Y, Clausen H, Wandall HH, Boltje TJ, Büll C. UV light-induced spatial loss of sialic acid capping using a photoactivatable sialyltransferase inhibitor. RSC Chem Biol 2023; 4:506-511. [PMID: 37415865 PMCID: PMC10320844 DOI: 10.1039/d3cb00006k] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2023] [Accepted: 05/06/2023] [Indexed: 07/08/2023] Open
Abstract
Sialic acids cap glycans displayed on mammalian glycoproteins and glycolipids and mediate many glycan-receptor interactions. Sialoglycans play a role in diseases such as cancer and infections where they facilitate immune evasion and metastasis or serve as cellular receptors for viruses, respectively. Strategies that specifically interfere with cellular sialoglycan biosynthesis, such as sialic acid mimetics that act as metabolic sialyltransferase inhibitors, enable research into the diverse biological functions of sialoglycans. Sialylation inhibitors are also emerging as potential therapeutics for cancer, infection, and other diseases. However, sialoglycans serve many important biological functions and systemic inhibition of sialoglycan biosynthesis can have adverse effects. To enable local and inducible inhibition of sialylation, we have synthesized and characterized a caged sialyltransferase inhibitor that can be selectively activated with UV-light. A photolabile protecting group was conjugated to a known sialyltransferase inhibitor (P-SiaFNEtoc). This yielded a photoactivatable inhibitor, UV-SiaFNEtoc, that remained inactive in human cell cultures and was readily activated through radiation with 365 nm UV light. Direct and short radiation of a human embryonic kidney (HEK293) cell monolayer was well-tolerated and resulted in photoactivation of the inhibitor and subsequent spatial restricted synthesis of asialoglycans. The developed photocaged sialic acid mimetic holds the potential to locally hinder the synthesis of sialoglycans through focused treatment with UV light and may be applied to bypass the adverse effects related to systemic loss of sialylation.
Collapse
Affiliation(s)
- Sam J Moons
- Cluster for Molecular Chemistry, Institute for Molecules and Materials, Radboud University Nijmegen Nijmegen The Netherlands
| | - Daniël L A H Hornikx
- Department of Biomolecular Chemistry, Institute for Molecules and Materials, Radboud University Nijmegen Nijmegen The Netherlands
| | - Mikkel K M Aasted
- Copenhagen Center for Glycomics, Department of Cellular and Molecular Medicine, Faculty of Health Sciences, University of Copenhagen Copenhagen Denmark
| | - Johan F A Pijnenborg
- Cluster for Molecular Chemistry, Institute for Molecules and Materials, Radboud University Nijmegen Nijmegen The Netherlands
| | - Matteo Calzari
- Cluster for Molecular Chemistry, Institute for Molecules and Materials, Radboud University Nijmegen Nijmegen The Netherlands
| | - Paul B White
- Cluster for Molecular Chemistry, Institute for Molecules and Materials, Radboud University Nijmegen Nijmegen The Netherlands
| | - Yoshiki Narimatsu
- Copenhagen Center for Glycomics, Department of Cellular and Molecular Medicine, Faculty of Health Sciences, University of Copenhagen Copenhagen Denmark
| | - Henrik Clausen
- Copenhagen Center for Glycomics, Department of Cellular and Molecular Medicine, Faculty of Health Sciences, University of Copenhagen Copenhagen Denmark
| | - Hans H Wandall
- Copenhagen Center for Glycomics, Department of Cellular and Molecular Medicine, Faculty of Health Sciences, University of Copenhagen Copenhagen Denmark
| | - Thomas J Boltje
- Cluster for Molecular Chemistry, Institute for Molecules and Materials, Radboud University Nijmegen Nijmegen The Netherlands
| | - Christian Büll
- Department of Biomolecular Chemistry, Institute for Molecules and Materials, Radboud University Nijmegen Nijmegen The Netherlands
- Copenhagen Center for Glycomics, Department of Cellular and Molecular Medicine, Faculty of Health Sciences, University of Copenhagen Copenhagen Denmark
| |
Collapse
|
9
|
Natoni A, Cerreto M, De Propris MS, Del Giudice I, Soscia R, Peragine N, Intoppa S, Milani ML, Guarini A, Foà R. Sialylation regulates migration in chronic lymphocytic leukemia. Haematologica 2023; 108:1851-1860. [PMID: 36779594 PMCID: PMC10316253 DOI: 10.3324/haematol.2022.281999] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2020] [Accepted: 01/27/2023] [Indexed: 02/11/2023] Open
Abstract
Sialylation is the terminal addition of sialic acid to underlying glycans. It plays a prominent role in cell adhesion and immune regulation. Sialylated structures found on adhesion molecules, such as CD49d, mediate the interactions between cancer cells and the microenvironment, facilitating metastatic seeding in target organs. Chronic lymphocytic leukemia (CLL) is a clonal B-cell malignancy characterized by the accumulation of CD5-positive B cells in the peripheral blood, bone marrow and lymph nodes. CLL cells proliferate mainly in the lymph node "proliferation centers", where the microenvironment provides pro-survival signals. Thus, migration and homing into these protective niches play a crucial role in CLL biology. In recent years, therapeutic strategies aimed at inducing the egress of CLL cells from the lymph nodes and bone marrow into the circulation have been highly successful. In this study, the sialylation status of 79 untreated and 24 ibrutinib-treated CLL patients was characterized by flow cytometry. Moreover, the effect of sialic acid removal on migration was tested by a transwell assay. Finally, we examined the sialylation status of CD49d by Western blot analysis. We found that CLL cells are highly sialylated, particularly those characterized by an "activated" immune phenotype. Notably, sialylation regulates CLL migration through the post-translational modification of CD49d. Finally, we showed that therapeutic agents that induce CLL mobilization from their protective niches, such as ibrutinib, modulate sialic acid levels. We propose that sialylation is an important regulator of CLL trafficking and may represent a novel target to further improve CLL therapy.
Collapse
Affiliation(s)
- Alessandro Natoni
- Hematology, Department of Translational and Precision Medicine, Sapienza University, Rome.
| | - Marina Cerreto
- Hematology, Department of Translational and Precision Medicine, Sapienza University, Rome
| | | | - Ilaria Del Giudice
- Hematology, Department of Translational and Precision Medicine, Sapienza University, Rome
| | - Roberta Soscia
- Hematology, Department of Translational and Precision Medicine, Sapienza University, Rome
| | - Nadia Peragine
- Hematology, Department of Translational and Precision Medicine, Sapienza University, Rome
| | - Stefania Intoppa
- Hematology, Department of Translational and Precision Medicine, Sapienza University, Rome
| | - Maria Laura Milani
- Hematology, Department of Translational and Precision Medicine, Sapienza University, Rome
| | - Anna Guarini
- Department of Molecular Medicine, Sapienza University, Rome
| | - Robin Foà
- Hematology, Department of Translational and Precision Medicine, Sapienza University, Rome
| |
Collapse
|
10
|
何 旎, 周 文. [Latest Findings on the Mechanism of the Interaction Between Multiple Myeloma Cells and Bone Marrow Microenvironment]. SICHUAN DA XUE XUE BAO. YI XUE BAN = JOURNAL OF SICHUAN UNIVERSITY. MEDICAL SCIENCE EDITION 2023; 54:475-481. [PMID: 37248571 PMCID: PMC10475439 DOI: 10.12182/20230560207] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 01/12/2023] [Indexed: 05/31/2023]
Abstract
Multiple myeloma (MM) is a hematologic malignancy of terminally differentiated plasma cells. The mechanisms of the pathogenesis and progression of MM include genetic abnormalities of the MM cells and the interaction between MM cells and bone marrow microenvironment (BMME). MM cells start malignant proliferation in BMME and contribute to the pathogenesis and progression of MM through direct or indirect interactions between cells and the extracellular matrix. Exploring the mechanism of interaction between MM cells and the microenvironment is crucial to improving our understanding of the pathogenesis and progression of MM and early diagnosis and treatment. In addition, the metabolic reprogramming of tumors is one of the key issues of oncology research. Herein, we summarized published findings on the the altered metabolic reprogramming of MM and the characteristics of MM metabolic-microbial interactions in order to gain an in-depth understanding of MM pathogenesis and progression and drug resistance mechanisms, and ultimately to explore for new strategies for MM treatment.
Collapse
Affiliation(s)
- 旎涵 何
- 中南大学基础医学院 肿瘤研究所 (长沙 412000)Cancer Research Institute, School of Basic Medical Sciences, Central South University, Changsha 412000, China
| | - 文 周
- 中南大学基础医学院 肿瘤研究所 (长沙 412000)Cancer Research Institute, School of Basic Medical Sciences, Central South University, Changsha 412000, China
| |
Collapse
|
11
|
Sugiura T, Hashimoto K, Kikuta K, Anazawa U, Nomura T, Kameyama A. Expression and localisation of MUC1 modified with sialylated core-2 O-glycans in mucoepidermoid carcinoma. Sci Rep 2023; 13:5752. [PMID: 37031283 PMCID: PMC10082819 DOI: 10.1038/s41598-023-32597-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2022] [Accepted: 03/29/2023] [Indexed: 04/10/2023] Open
Abstract
Mucoepidermoid carcinoma (MEC) is the most frequent of the rare salivary gland malignancies. We previously reported high expression of Mucin 1 (MUC1) modified with sialylated core-2 O-glycans in MEC by using tissue homogenates. In this study, we characterised glycan structures of MEC and identified the localisation of cells expressing these distinctive glycans on MUC1. Mucins were extracted from the frozen tissues of three patients with MEC, and normal salivary glands (NSGs) extracted from seven patients, separated by supported molecular matrix electrophoresis (SMME) and the membranes stained with various lectins. In addition, formalin-fixed, paraffin-embedded sections from three patients with MEC were subjected to immunohistochemistry (IHC) with various monoclonal antibodies and analysed for C2GnT-1 expression by in situ hybridisation (ISH). Lectin blotting of the SMME membranes revealed that glycans on MUC1 from MEC samples contained α2,3-linked sialic acid. In IHC, MUC1 was diffusely detected at MEC-affected regions but was specifically detected at apical membranes in NSGs. ISH showed that C2GnT-1 was expressed at the MUC1-positive in MEC-affected regions but not in the NSG. MEC cells produced MUC1 modified with α2,3-linked sialic acid-containing core-2 O-glycans. MUC1 containing these glycans deserves further study as a new potential diagnostic marker of MEC.
Collapse
Affiliation(s)
- Takanori Sugiura
- Department of Oral Oncology, Oral and Maxillofacial Surgery, Ichikawa General Hospital, Tokyo Dental College, 5-11-13 Sugano, Ichikawa-Shi, Chiba, 272-8513, Japan
| | - Kazuhiko Hashimoto
- Department of Pathology and Laboratory Medicine, Ichikawa General Hospital, Tokyo Dental College, 5-11-13 Sugano, Ichikawa-Shi, Chiba, 272-8513, Japan
| | - Kazutaka Kikuta
- Department of Musculoskeletal Oncology and Orthopaedic Surgery, Tochigi Cancer Center, 4-9-13 Yohnan, Utsunomiya, Tochigi, 320-0834, Japan
| | - Ukei Anazawa
- Department of Orthopaedic Surgery, Ichikawa General Hospital, Tokyo Dental College, 5-11-13 Sugano, Ichikawa-Shi, Chiba, 272-8513, Japan
| | - Takeshi Nomura
- Department of Oral Oncology, Oral and Maxillofacial Surgery, Ichikawa General Hospital, Tokyo Dental College, 5-11-13 Sugano, Ichikawa-Shi, Chiba, 272-8513, Japan
- Oral Cancer Center, Tokyo Dental College, 5-11-13 Sugano, Ichikawa-Shi, Chiba, 272-8513, Japan
| | - Akihiko Kameyama
- Cellular and Molecular Biotechnology Research Institute, National Institute of Advanced Industrial Science and Technology (AIST), 1-1-1 Higashi, Tsukuba, Ibaraki, 305-8565, Japan.
| |
Collapse
|
12
|
Natoni A, Cerreto M, De Propris MS, Petrucci MT, Fazio F, Intoppa S, Milani ML, Kirkham-McCarthy L, Henderson R, Swan D, Guarini A, O'Dwyer M, Foà R. Sialofucosylation Enables Platelet Binding to Myeloma Cells via P-Selectin and Suppresses NK Cell-Mediated Cytotoxicity. Cancers (Basel) 2023; 15:cancers15072154. [PMID: 37046814 PMCID: PMC10093642 DOI: 10.3390/cancers15072154] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2023] [Revised: 03/29/2023] [Accepted: 04/03/2023] [Indexed: 04/14/2023] Open
Abstract
Multiple myeloma (MM) is a plasma cell disorder that develops in the bone marrow (BM) and is characterized by uncontrolled proliferation and the ability to disseminate to different sites of the skeleton. Sialofucosylated structures, particularly Sialyl Lewis a/x (SLea/x), facilitate the homing of MM cells into the BM, leading to resistance to bortezomib in vivo. Platelets have been shown to play an important role in tumor metastasis. Platelets can bind to the surface of cancer cells, forming a "cloak" that protects them from the shear stress of the bloodstream and natural killer (NK) cell-mediated cytotoxicity. In this study, we showed that the presence of SLea/x induced a strong binding of MM cells to P-selectin, leading to specific and direct interactions with platelets, which could be inhibited by a P-selectin-blocking antibody. Importantly, platelets surrounded SLea/x-enriched MM cells, protecting them from NK cell-mediated cytotoxicity. The interactions between the platelets and MM cells were also detected in BM samples obtained from MM patients. Platelet binding to SLea/x-enriched MM cells was increased in patients with symptomatic disease and at relapse. These data suggest an important role of SLea/x and platelets in MM disease progression and resistance to therapy.
Collapse
Affiliation(s)
- Alessandro Natoni
- Hematology, Department of Translational and Precision Medicine, Sapienza University, 00161 Rome, Italy
| | - Marina Cerreto
- Hematology, Department of Translational and Precision Medicine, Sapienza University, 00161 Rome, Italy
| | - Maria Stefania De Propris
- Hematology, Department of Translational and Precision Medicine, Sapienza University, 00161 Rome, Italy
| | - Maria Teresa Petrucci
- Hematology, Department of Translational and Precision Medicine, Sapienza University, 00161 Rome, Italy
| | - Francesca Fazio
- Hematology, Department of Translational and Precision Medicine, Sapienza University, 00161 Rome, Italy
| | - Stefania Intoppa
- Hematology, Department of Translational and Precision Medicine, Sapienza University, 00161 Rome, Italy
| | - Maria Laura Milani
- Hematology, Department of Translational and Precision Medicine, Sapienza University, 00161 Rome, Italy
| | - Lucy Kirkham-McCarthy
- Biomedical Sciences, School of Medicine, National University of Ireland Galway, H91 TK33 Galway, Ireland
| | - Robert Henderson
- Department of Haematology, Galway University Hospital, H71 YR71 Galway, Ireland
| | - Dawn Swan
- Department of Haematology, Galway University Hospital, H71 YR71 Galway, Ireland
| | - Anna Guarini
- Department of Molecular Medicine, Sapienza University, 00161 Rome, Italy
| | - Michael O'Dwyer
- Biomedical Sciences, School of Medicine, National University of Ireland Galway, H91 TK33 Galway, Ireland
| | - Robin Foà
- Hematology, Department of Translational and Precision Medicine, Sapienza University, 00161 Rome, Italy
| |
Collapse
|
13
|
Yang B, Yin S, Zhou Z, Huang L, Xi M. Inflammation Control and Tumor Growth Inhibition of Ovarian Cancer by Targeting Adhesion Molecules of E-Selectin. Cancers (Basel) 2023; 15:cancers15072136. [PMID: 37046797 PMCID: PMC10093113 DOI: 10.3390/cancers15072136] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2023] [Revised: 03/27/2023] [Accepted: 04/02/2023] [Indexed: 04/07/2023] Open
Abstract
Objective: The aim is to use E-selectin-binding peptide (ESBP) to actively recognize E-selectin, so allowing a drug delivery system to actively recognize the cells and inhibit the tumor growth of ovarian cancer by targeting adhesion molecules of E-selectin. An ovarian-cancer-directed drug delivery system was designed based on the high affinity of E-selectin-binding peptide (ESBP) to E-selectin. The effects and mechanisms of ESBP-bovine serum albumin (BSA) polymerized nanoparticles were investigated. Methods: BSA polymerized nanoparticles (BSANPs) and ESBP-BSANPs-paclitaxel (PTX) were prepared and their characteristics were measured. The in vitro targetability and cytotoxicity of ESBP-BSANPs-PTX were evaluated through in vitro drug uptake and MTT experiments. The mechanisms of ESBP-BSANPs-PTX were investigated via apoptosis, wound healing and immunohistochemistry assays. The in vivo targeting properties and drug effects were observed in a mouse tumor-bearing model. Results: In vitro experiments revealed an increase in the uptake of ESBP-BSANPs-FITC. The cytotoxicity of ESBP-BSANPs-PTX in A2780/CP70, HUVEC, RAW264.7 and ID8 cells was higher than that of PTX alone. ESBP-BSANPs-PTX increased cell apoptosis in a dose-dependent manner and exhibited a greater ability to inhibit cell migration than BSANPs-PTX. In vivo experiments demonstrated the targetability and good effects of ESBP-BSANPs. Conclusions: ESBP-BSANPs-PTX improve PTX targetability, provide tumor-specific and potent therapeutic activities, and show promise for the development of agents in preclinical epithelial ovarian cancer.
Collapse
Affiliation(s)
- Bowen Yang
- Department of Gynecology and Obstetrics, West China Second University Hospital, Sichuan University, Chengdu 610041, China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, Chengdu 610041, China
| | - Shanmei Yin
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu 610041, China
| | - Zishuo Zhou
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu 610041, China
| | - Luyao Huang
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu 610041, China
| | - Mingrong Xi
- Department of Gynecology and Obstetrics, West China Second University Hospital, Sichuan University, Chengdu 610041, China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, Chengdu 610041, China
| |
Collapse
|
14
|
Stanczak MA, Läubli H. Siglec receptors as new immune checkpoints in cancer. Mol Aspects Med 2023; 90:101112. [PMID: 35948467 DOI: 10.1016/j.mam.2022.101112] [Citation(s) in RCA: 54] [Impact Index Per Article: 27.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2022] [Revised: 07/26/2022] [Accepted: 07/28/2022] [Indexed: 02/08/2023]
Abstract
Cancer immunotherapy in the form of immune checkpoint inhibitors and cellular therapies has improved the treatment and prognosis of many patients. Nevertheless, most cancers are still resistant to currently approved cancer immunotherapies. New approaches and rational combinations are needed to overcome these resistances. There is emerging evidence that Siglec receptors could be regarded as new immune checkpoints and targets for cancer immunotherapy. In this review, we summarize the experimental evidence supporting Siglec receptors as new immune checkpoints in cancer and discuss their mechanisms of action, as well as current efforts to target Siglec receptors and their interactions with sialoglycan Siglec-ligands.
Collapse
Affiliation(s)
- Michal A Stanczak
- The Bloomberg-Kimmel Institute for Cancer Immunotherapy at Johns Hopkins, Baltimore, MD, 21287, USA
| | - Heinz Läubli
- Laboratory for Cancer Immunotherapy, Department of Biomedicine, University of Basel, Division of Oncology, University Hospital Basel, Switzerland.
| |
Collapse
|
15
|
Miró L, López J, Guerrero PE, Martínez-Bosch N, Manero-Rupérez N, Moreno M, Ortiz MR, Llop E, Navarro P, Peracaula R. Sialyltransferase Inhibitor Ac 53F axNeu5Ac Reverts the Malignant Phenotype of Pancreatic Cancer Cells, and Reduces Tumor Volume and Favors T-Cell Infiltrates in Mice. Cancers (Basel) 2022; 14:cancers14246133. [PMID: 36551619 PMCID: PMC9776040 DOI: 10.3390/cancers14246133] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2022] [Revised: 12/07/2022] [Accepted: 12/09/2022] [Indexed: 12/15/2022] Open
Abstract
Hypersialylation is a feature of pancreatic ductal adenocarcinoma (PDA) and it has been related to tumor malignancy and immune suppression. In this work, we have evaluated the potential of the sialyltransferase inhibitor, Ac53FaxNeu5Ac, to decrease tumor sialoglycans in PDA and to revert its malignant phenotype. Sialoglycans on PDA cells were evaluated by flow cytometry, and the functional impact of Ac53FaxNeu5Ac was assessed using E-selectin adhesion, migration, and invasion assays. PDA tumors were generated in syngeneic mice from KC cells and treated with Ac53FaxNeu5Ac to evaluate tumor growth, mice survival, and its impact on blocking sialic acid (SA) and on the tumor immune component. Ac53FaxNeu5Ac treatment on human PDA cells decreased α2,3-SA and sialyl-Lewisx, which resulted in a reduction in their E-selectin adhesion, and in their migratory and invasive capabilities. Subcutaneous murine tumors treated with Ac53FaxNeu5Ac reduced their volume, their SA expression, and modified their immune component, with an increase in CD8+ T-lymphocytes and NK cells. In conclusion, Ac53FaxNeu5Ac treatment weakened PDA cells' malignant phenotype, thereby reducing tumor growth while favoring anti-tumor immune surveillance. Altogether, these results show the positive impact of reducing SA expression by inhibiting cell sialyltransferases and open the way to use sialyltransferase inhibitors to target this dismal disease.
Collapse
Affiliation(s)
- Laura Miró
- Biochemistry and Molecular Biology Unit, Department of Biology, University of Girona, 17003 Girona, Spain
- Girona Biomedical Research Institute (IDIBGI), 17190 Girona, Spain
| | - Júlia López
- Biochemistry and Molecular Biology Unit, Department of Biology, University of Girona, 17003 Girona, Spain
- Girona Biomedical Research Institute (IDIBGI), 17190 Girona, Spain
| | - Pedro E. Guerrero
- Biochemistry and Molecular Biology Unit, Department of Biology, University of Girona, 17003 Girona, Spain
- Girona Biomedical Research Institute (IDIBGI), 17190 Girona, Spain
| | - Neus Martínez-Bosch
- Cancer Research Program, Hospital del Mar Medical Research Institute (IMIM), Unidad Asociada IIBB-CSIC, 08003 Barcelona, Spain
| | - Noemí Manero-Rupérez
- Cancer Research Program, Hospital del Mar Medical Research Institute (IMIM), Unidad Asociada IIBB-CSIC, 08003 Barcelona, Spain
| | - Mireia Moreno
- Cancer Research Program, Hospital del Mar Medical Research Institute (IMIM), Unidad Asociada IIBB-CSIC, 08003 Barcelona, Spain
| | - M. Rosa Ortiz
- Pathology Department, Josep Trueta University Hospital, 17007 Girona, Spain
| | - Esther Llop
- Biochemistry and Molecular Biology Unit, Department of Biology, University of Girona, 17003 Girona, Spain
- Girona Biomedical Research Institute (IDIBGI), 17190 Girona, Spain
| | - Pilar Navarro
- Cancer Research Program, Hospital del Mar Medical Research Institute (IMIM), Unidad Asociada IIBB-CSIC, 08003 Barcelona, Spain
- Institute of Biomedical Research of Barcelona (IIBB)-CSIC, 08036 Barcelona, Spain
- Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), 08036 Barcelona, Spain
| | - Rosa Peracaula
- Biochemistry and Molecular Biology Unit, Department of Biology, University of Girona, 17003 Girona, Spain
- Girona Biomedical Research Institute (IDIBGI), 17190 Girona, Spain
- Correspondence: ; Tel.: +34-972418370
| |
Collapse
|
16
|
Peru S, Prochazkova-Carlotti M, Cherrier F, Velazquez J, Richard E, Idrissi Y, Cappellen D, Azzi-Martin L, Pham-Ledard A, Beylot-Barry M, Merlio JP, Poglio S. Cutaneous Lymphocyte Antigen Is a Potential Therapeutic Target in Cutaneous T-Cell Lymphoma. J Invest Dermatol 2022; 142:3243-3252.e10. [PMID: 35850209 DOI: 10.1016/j.jid.2022.06.016] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2021] [Revised: 06/08/2022] [Accepted: 06/10/2022] [Indexed: 01/05/2023]
Abstract
Cutaneous T-cell lymphoma (CTCL) such as Sézary syndrome or mycosis fungoides corresponds to an abnormal infiltration of T lymphocytes in the skin. CTCL cells have a heterogeneous phenotype and express cell adhesion molecules such as cutaneous lymphocyte antigen (CLA) supporting skin homing. The use of a mAb (HECA-452) against CLA significantly decreased transendothelial migration and survival of CTCL cells from patient samples and My-La cell line. The decrease of CLA expression by inhibition of its maturation enzyme, ST3 β-galactoside α-2,3-sialyltransferase 4, also impaired CTCL cell migration, proliferation, and survival. We confirmed in vivo that treatment with anti-CLA mAb decreased the tumorigenicity as well as dissemination of CTCL cells in different tissues compared with the control group. Our findings provide evidence of the involvement of CLA in CTCL cell migration and survival, supporting that CLA inhibition could represent an actionable therapy in patients with CTCL.
Collapse
Affiliation(s)
- Sara Peru
- BRIC (BoRdeaux Institute of onCology), UMR1312, INSERM, Univ. Bordeaux, Bordeaux, France
| | | | - Floriane Cherrier
- BRIC (BoRdeaux Institute of onCology), UMR1312, INSERM, Univ. Bordeaux, Bordeaux, France
| | - Joanne Velazquez
- BRIC (BoRdeaux Institute of onCology), UMR1312, INSERM, Univ. Bordeaux, Bordeaux, France
| | - Elodie Richard
- BRIC (BoRdeaux Institute of onCology), UMR1312, INSERM, Univ. Bordeaux, Bordeaux, France
| | - Yamina Idrissi
- BRIC (BoRdeaux Institute of onCology), UMR1312, INSERM, Univ. Bordeaux, Bordeaux, France
| | - David Cappellen
- BRIC (BoRdeaux Institute of onCology), UMR1312, INSERM, Univ. Bordeaux, Bordeaux, France; Tumor Bank and Tumor Biology Laboratory, Bordeaux University Hospital, Pessac, France
| | - Lamia Azzi-Martin
- BRIC (BoRdeaux Institute of onCology), UMR1312, INSERM, Univ. Bordeaux, Bordeaux, France; UFR des Sciences Médicales, Bordeaux University, Bordeaux, France
| | - Anne Pham-Ledard
- BRIC (BoRdeaux Institute of onCology), UMR1312, INSERM, Univ. Bordeaux, Bordeaux, France; Dermatology Department, Bordeaux University Hospital, Bordeaux, France
| | - Marie Beylot-Barry
- BRIC (BoRdeaux Institute of onCology), UMR1312, INSERM, Univ. Bordeaux, Bordeaux, France; Dermatology Department, Bordeaux University Hospital, Bordeaux, France
| | - Jean-Philippe Merlio
- BRIC (BoRdeaux Institute of onCology), UMR1312, INSERM, Univ. Bordeaux, Bordeaux, France; Tumor Bank and Tumor Biology Laboratory, Bordeaux University Hospital, Pessac, France
| | - Sandrine Poglio
- BRIC (BoRdeaux Institute of onCology), UMR1312, INSERM, Univ. Bordeaux, Bordeaux, France.
| |
Collapse
|
17
|
Patil V, Bohara R, Winter C, Kilcoyne M, McMahon S, Pandit A. An insight into new glycotherapeutics in glial inflammation: Understanding the role of glycosylation in mitochondrial function and acute to the chronic phases of inflammation. CNS Neurosci Ther 2022; 29:429-444. [PMID: 36377513 PMCID: PMC9804060 DOI: 10.1111/cns.14016] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2022] [Revised: 09/16/2022] [Accepted: 09/29/2022] [Indexed: 11/16/2022] Open
Abstract
INTRODUCTION Glycosylation plays a critical role during inflammation and glial scar formation upon spinal cord injury (SCI) disease progression. Astrocytes and microglia are involved in this cascade to modulate the inflammation and tissue remodeling from acute to chronic phases. Therefore, understating the glycan changes in these glial cells is paramount. METHOD AND RESULTS A lectin microarray was undertaken using a cytokine-driven inflammatory mixed glial culture model, revealing considerable differential glycosylation from the acute to the chronic phase in a cytokine-combination generated inflamed MGC model. It was found that several N- and O-linked glycans associated with glia during SCI were differentially regulated. Pearson's correlation hierarchical clustering showed that groups were separated into several clusters, illustrating the heterogenicity among the control, cytokine combination, and LPS treated groups and the day on which treatment was given. Control and LPS treatments were observed to be in dense clusters. This was further confirmed with lectin immunostaining in which GalNAc, GlcNAc, mannose, fucose and sialic acid-binding residues were detected in astrocytes and microglia. However, the sialyltransferase inhibitor inhibited this modification (upregulation of the sialic acid expression), which indeed modulates the mitochondrial functions. CONCLUSIONS The present study is the first functional investigation of glycosylation modulation in a mixed glial culture model, which elucidates the role of the glycome in neuroinflammation in progression and identified potential therapeutic targets for future glyco therapeutics in neuroinflammation.
Collapse
Affiliation(s)
- Vaibhav Patil
- CÚRAM, SFI Research Centre for Medical DevicesUniversity of GalwayGalwayIreland
| | - Raghvendra Bohara
- CÚRAM, SFI Research Centre for Medical DevicesUniversity of GalwayGalwayIreland
| | - Carla Winter
- CÚRAM, SFI Research Centre for Medical DevicesUniversity of GalwayGalwayIreland
| | - Michelle Kilcoyne
- CÚRAM, SFI Research Centre for Medical DevicesUniversity of GalwayGalwayIreland,MicrobiologyUniversity of GalwayGalwayIreland
| | - Siobhan McMahon
- CÚRAM, SFI Research Centre for Medical DevicesUniversity of GalwayGalwayIreland,AnatomyGalwayIreland
| | - Abhay Pandit
- CÚRAM, SFI Research Centre for Medical DevicesUniversity of GalwayGalwayIreland
| |
Collapse
|
18
|
GC S, Tuy K, Rickenbacker L, Jones R, Chakraborty A, Miller CR, Beierle EA, Hanumanthu VS, Tran AN, Mobley JA, Bellis SL, Hjelmeland AB. α2,6 Sialylation mediated by ST6GAL1 promotes glioblastoma growth. JCI Insight 2022; 7:e158799. [PMID: 36345944 PMCID: PMC9675560 DOI: 10.1172/jci.insight.158799] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2022] [Accepted: 09/20/2022] [Indexed: 11/09/2022] Open
Abstract
One of the least-investigated areas of brain pathology research is glycosylation, which is a critical regulator of cell surface protein structure and function. β-Galactoside α2,6-sialyltransferase (ST6GAL1) is the primary enzyme that α2,6 sialylates N-glycosylated proteins destined for the plasma membrane or secretion, thereby modulating cell signaling and behavior. We demonstrate a potentially novel, protumorigenic role for α2,6 sialylation and ST6GAL1 in the deadly brain tumor glioblastoma (GBM). GBM cells with high α2,6 sialylation exhibited increased in vitro growth and self-renewal capacity and decreased mouse survival when orthotopically injected. α2,6 Sialylation was regulated by ST6GAL1 in GBM, and ST6GAL1 was elevated in brain tumor-initiating cells (BTICs). Knockdown of ST6GAL1 in BTICs decreased in vitro growth, self-renewal capacity, and tumorigenic potential. ST6GAL1 regulates levels of the known BTIC regulators PDGF Receptor β (PDGFRB), Activated Leukocyte Cell Adhesion Molecule, and Neuropilin, which were confirmed to bind to a lectin-recognizing α2,6 sialic acid. Loss of ST6GAL1 was confirmed to decrease PDGFRB α2,6 sialylation, total protein levels, and the induction of phosphorylation by PDGF-BB. Thus, ST6GAL1-mediated α2,6 sialylation of a select subset of cell surface receptors, including PDGFRB, increases GBM growth.
Collapse
Affiliation(s)
- Sajina GC
- Department of Cell, Developmental and Integrative Biology
| | - Kaysaw Tuy
- Department of Cell, Developmental and Integrative Biology
| | | | - Robert Jones
- Department of Cell, Developmental and Integrative Biology
| | | | | | | | | | | | - James A. Mobley
- Department of Anesthesiology, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | | | | |
Collapse
|
19
|
Rossing E, Pijnenborg JFA, Boltje TJ. Chemical tools to track and perturb the expression of sialic acid and fucose monosaccharides. Chem Commun (Camb) 2022; 58:12139-12150. [PMID: 36222364 PMCID: PMC9623448 DOI: 10.1039/d2cc04275d] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2022] [Accepted: 10/05/2022] [Indexed: 11/24/2022]
Abstract
The biosynthesis of glycans is a highly conserved biological process and found in all domains of life. The expression of cell surface glycans is increasingly recognized as a target for therapeutic intervention given the role of glycans in major pathologies such as cancer and microbial infection. Herein, we summarize our contributions to the development of unnatural monosaccharide derivatives to infiltrate and alter the expression of both mammalian and bacterial glycans and their therapeutic application.
Collapse
Affiliation(s)
- Emiel Rossing
- Department of Synthetic Organic Chemistry, Institute for Molecules and Materials, Radboud University, Heyendaalseweg 135, 6525AJ, Nijmegen, The Netherlands.
| | - Johan F A Pijnenborg
- Department of Synthetic Organic Chemistry, Institute for Molecules and Materials, Radboud University, Heyendaalseweg 135, 6525AJ, Nijmegen, The Netherlands.
| | - Thomas J Boltje
- Department of Synthetic Organic Chemistry, Institute for Molecules and Materials, Radboud University, Heyendaalseweg 135, 6525AJ, Nijmegen, The Netherlands.
| |
Collapse
|
20
|
Aberrant Sialylation in Cancer: Therapeutic Opportunities. Cancers (Basel) 2022; 14:cancers14174248. [PMID: 36077781 PMCID: PMC9454432 DOI: 10.3390/cancers14174248] [Citation(s) in RCA: 49] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2022] [Revised: 08/15/2022] [Accepted: 08/23/2022] [Indexed: 11/16/2022] Open
Abstract
The surface of every eukaryotic cell is coated in a thick layer of glycans that acts as a key interface with the extracellular environment. Cancer cells have a different ‘glycan coat’ to healthy cells and aberrant glycosylation is a universal feature of cancer cells linked to all of the cancer hallmarks. This means glycans hold huge potential for the development of new diagnostic and therapeutic strategies. One key change in tumour glycosylation is increased sialylation, both on N-glycans and O-glycans, which leads to a dense forest of sialylated structures covering the cell surface. This hypersialylation has far-reaching consequences for cancer cells, and sialylated glycans are fundamental in tumour growth, metastasis, immune evasion and drug resistance. The development of strategies to inhibit aberrant sialylation in cancer represents an important opportunity to develop new therapeutics. Here, I summarise recent advances to target aberrant sialylation in cancer, including the development of sialyltransferase inhibitors and strategies to inhibit Siglecs and Selectins, and discuss opportunities for the future.
Collapse
|
21
|
Schwestermann J, Besse A, Driessen C, Besse L. Contribution of the Tumor Microenvironment to Metabolic Changes Triggering Resistance of Multiple Myeloma to Proteasome Inhibitors. Front Oncol 2022; 12:899272. [PMID: 35692781 PMCID: PMC9178120 DOI: 10.3389/fonc.2022.899272] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2022] [Accepted: 04/25/2022] [Indexed: 11/13/2022] Open
Abstract
Virtually all patients with multiple myeloma become unresponsive to treatment with proteasome inhibitors over time. Relapsed/refractory multiple myeloma is accompanied by the clonal evolution of myeloma cells with heterogeneous genomic aberrations, diverse proteomic and metabolic alterations, and profound changes of the bone marrow microenvironment. However, the molecular mechanisms that drive resistance to proteasome inhibitors within the context of the bone marrow microenvironment remain elusive. In this review article, we summarize the latest knowledge about the complex interaction of malignant plasma cells with its surrounding microenvironment. We discuss the pivotal role of metabolic reprograming of malignant plasma cells within the tumor microenvironment with a subsequent focus on metabolic rewiring in plasma cells upon treatment with proteasome inhibitors, driving multiple ways of adaptation to the treatment. At the same time, mutual interaction of plasma cells with the surrounding tumor microenvironment drives multiple metabolic alterations in the bone marrow. This provides a tumor-promoting environment, but at the same time may offer novel therapeutic options for the treatment of relapsed/refractory myeloma patients.
Collapse
Affiliation(s)
| | | | | | - Lenka Besse
- Laboratory of Experimental Oncology, Clinics for Medical Hematology and Oncology, Cantonal Hospital St. Gallen, St. Gallen, Switzerland
| |
Collapse
|
22
|
Zerdan MB, Nasr L, Kassab J, Saba L, Ghossein M, Yaghi M, Dominguez B, Chaulagain CP. Adhesion molecules in multiple myeloma oncogenesis and targeted therapy. Int J Hematol Oncol 2022; 11:IJH39. [PMID: 35663420 PMCID: PMC9136637 DOI: 10.2217/ijh-2021-0017] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2021] [Accepted: 04/07/2022] [Indexed: 11/21/2022] Open
Abstract
Every day we march closer to finding the cure for multiple myeloma. The myeloma cells inflict their damage through specialized cellular meshwork and cytokines system. Implicit in these interactions are cellular adhesion molecules and their regulators which include but are not limited to integrins and syndecan-1/CD138, immunoglobulin superfamily cell adhesion molecules, such as CD44, cadherins such as N-cadherin, and selectins, such as E-selectin. Several adhesion molecules are respectively involved in myelomagenesis such as in the transition from the precursor disorder monoclonal gammopathy of undetermined significance to indolent asymptomatic multiple myeloma (smoldering myeloma) then to active multiple myeloma or primary plasma cell leukemia, and in the pathological manifestations of multiple myeloma.
Collapse
Affiliation(s)
- Maroun Bou Zerdan
- Department of Hematology-Oncology, Myeloma & Amyloidosis Program, Maroone Cancer Center, Cleveland Clinic Florida, Weston, FL 33331, USA
| | - Lewis Nasr
- Saint-Joseph University, Faculty of Medicine, Beirut, Lebanon
| | - Joseph Kassab
- Saint-Joseph University, Faculty of Medicine, Beirut, Lebanon
| | - Ludovic Saba
- Saint-Joseph University, Faculty of Medicine, Beirut, Lebanon
| | - Myriam Ghossein
- Department of Medicine & Medical Sciences, University of Balamand, Balamand, Lebanon
| | - Marita Yaghi
- Department of Hematology-Oncology, Myeloma & Amyloidosis Program, Maroone Cancer Center, Cleveland Clinic Florida, Weston, FL 33331, USA
| | - Barbara Dominguez
- Department of Hematology-Oncology, Myeloma & Amyloidosis Program, Maroone Cancer Center, Cleveland Clinic Florida, Weston, FL 33331, USA
| | - Chakra P Chaulagain
- Department of Hematology-Oncology, Myeloma & Amyloidosis Program, Maroone Cancer Center, Cleveland Clinic Florida, Weston, FL 33331, USA
| |
Collapse
|
23
|
Bindeman WE, Fingleton B. Glycosylation as a regulator of site-specific metastasis. Cancer Metastasis Rev 2022; 41:107-129. [PMID: 34967926 PMCID: PMC8930623 DOI: 10.1007/s10555-021-10015-1] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/13/2021] [Accepted: 12/20/2021] [Indexed: 12/17/2022]
Abstract
Metastasis is considered to be responsible for 90% of cancer-related deaths. Although it is clinically evident that metastatic patterns vary by primary tumor type, the molecular mechanisms underlying the site-specific nature of metastasis are an area of active investigation. One mechanism that has emerged as an important player in this process is glycosylation, or the addition of sugar moieties onto protein and lipid substrates. Glycosylation is the most common post-translational modification, occurring on more than 50% of translated proteins. Many of those proteins are either secreted or expressed on the cell membrane, thereby making glycosylation an important mediator of cell-cell interactions, including tumor-microenvironment interactions. It has been recently discovered that alteration of glycosylation patterns influences cancer metastasis, both globally and in a site-specific manner. This review will summarize the current knowledge regarding the role of glycosylation in the tropism of cancer cells for several common metastatic sites, including the bone, lung, brain, and lymph nodes.
Collapse
Affiliation(s)
- Wendy E Bindeman
- Department of Pharmacology, Vanderbilt University, Nashville, TN, USA
| | - Barbara Fingleton
- Department of Pharmacology, Vanderbilt University, Nashville, TN, USA.
| |
Collapse
|
24
|
Thompson Z, Steed AJ, Millot J, Chen Y, Cowan JA. Exploring artificial metalloglycosidases as selective catalysts for the recognition and degradation of the sLe x tetrasaccharide. NEW J CHEM 2022. [DOI: 10.1039/d2nj02144g] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Copper-bound fucose-binding peptide removes cellular sLex antigen that has been linked to cell adhesion and cancer metastasis. Catalytic and mass spectrometry assays further demonstrate degradation of native sLex tetrasaccharide in solution.
Collapse
Affiliation(s)
- Zechariah Thompson
- Department of Chemistry and Biochemistry, The Ohio State University, 100 West 18th Avenue, Columbus, Ohio 43210, USA
| | - Alexander J. Steed
- Department of Chemistry and Biochemistry, The Ohio State University, 100 West 18th Avenue, Columbus, Ohio 43210, USA
| | - Jack Millot
- Department of Chemistry and Biochemistry, The Ohio State University, 100 West 18th Avenue, Columbus, Ohio 43210, USA
| | - Yinghua Chen
- Department of Physiology and Biophysics, Case Western Reserve University, 10900 Euclid Avenue, Cleveland, OH 44106, USA
| | - J. A. Cowan
- Department of Chemistry and Biochemistry, The Ohio State University, 100 West 18th Avenue, Columbus, Ohio 43210, USA
| |
Collapse
|
25
|
Dobie C, Montgomery AP, Szabo R, Yu H, Skropeta D. Synthesis and biological evaluation of selective phosphonate-bearing 1,2,3-triazole-linked sialyltransferase inhibitors. RSC Med Chem 2021; 12:1680-1689. [PMID: 34778769 DOI: 10.1039/d1md00079a] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2021] [Accepted: 06/19/2021] [Indexed: 01/01/2023] Open
Abstract
The critical role of sialyltransferase (ST) enzymes in tumour cell growth and metastasis, as well as links to multi-drug and radiation resistance, has seen STs emerge as a target for potential antimetastatic cancer treatments. One promising class of ST inhibitors that improve upon the pharmacokinetic issues of previous inhibitors is the 1,2,3-triazole-linked transition-state analogues. Herein, we present the design and synthesis of a new generation of 1,2,3-triazole-linked sialyltransferase inhibitors, along with their biological evaluation demonstrating increased potency for phosphonate bearing compounds. The six most promising inhibitors presented in this work exhibited a greater number of binding modes for hST6Gal I over hST3Gal I, with K i ranging from 3-55 μM. This work highlights phosphonate bearing triazole-linked compounds as a promising class of synthetically accessible ST inhibitors that warrant further investigation.
Collapse
Affiliation(s)
- Christopher Dobie
- Molecular Horizons and School of Chemistry & Molecular Bioscience, Faculty of Science, Medicine and Health, University of Wollongong NSW 2522 Australia
| | - Andrew P Montgomery
- Molecular Horizons and School of Chemistry & Molecular Bioscience, Faculty of Science, Medicine and Health, University of Wollongong NSW 2522 Australia
| | - Rémi Szabo
- Molecular Horizons and School of Chemistry & Molecular Bioscience, Faculty of Science, Medicine and Health, University of Wollongong NSW 2522 Australia
| | - Haibo Yu
- Molecular Horizons and School of Chemistry & Molecular Bioscience, Faculty of Science, Medicine and Health, University of Wollongong NSW 2522 Australia .,Illawarra Health and Medical Research Institute Wollongong NSW 2522 Australia
| | - Danielle Skropeta
- Molecular Horizons and School of Chemistry & Molecular Bioscience, Faculty of Science, Medicine and Health, University of Wollongong NSW 2522 Australia .,Illawarra Health and Medical Research Institute Wollongong NSW 2522 Australia
| |
Collapse
|
26
|
Perez SJLP, Fu CW, Li WS. Sialyltransferase Inhibitors for the Treatment of Cancer Metastasis: Current Challenges and Future Perspectives. Molecules 2021; 26:5673. [PMID: 34577144 PMCID: PMC8470674 DOI: 10.3390/molecules26185673] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Revised: 09/15/2021] [Accepted: 09/15/2021] [Indexed: 01/19/2023] Open
Abstract
Potent, cell-permeable, and subtype-selective sialyltransferase inhibitors represent an attractive family of substances that can potentially be used for the clinical treatment of cancer metastasis. These substances operate by specifically inhibiting sialyltransferase-mediated hypersialylation of cell surface glycoproteins or glycolipids, which then blocks the sialic acid recognition pathway and leads to deterioration of cell motility and invasion. A vast amount of evidence for the in vitro and in vivo effects of sialyltransferase inhibition or knockdown on tumor progression and tumor cell metastasis or colonization has been accumulated over the past decades. In this regard, this review comprehensively discusses the results of studies that have led to the recent discovery and development of sialyltransferase inhibitors, their potential biomedical applications in the treatment of cancer metastasis, and their current limitations and future opportunities.
Collapse
Affiliation(s)
- Ser John Lynon P. Perez
- Institute of Chemistry, Academia Sinica, Taipei 115, Taiwan; (S.J.L.P.P.); (C.-W.F.)
- Sustainable Chemical Science and Technology, Taiwan International Graduate Program, Academia Sinica, Taipei 115, Taiwan
- Sustainable Chemical Science and Technology, Taiwan International Graduate Program, National Yang Ming Chiao Tung University, Hsinchu 30010, Taiwan
- Department of Applied Chemistry, National Yang Ming Chiao Tung University, Hsinchu 30010, Taiwan
| | - Chih-Wei Fu
- Institute of Chemistry, Academia Sinica, Taipei 115, Taiwan; (S.J.L.P.P.); (C.-W.F.)
- Department of Chemistry, National Central University, Taoyuan City 32001, Taiwan
| | - Wen-Shan Li
- Institute of Chemistry, Academia Sinica, Taipei 115, Taiwan; (S.J.L.P.P.); (C.-W.F.)
- Doctoral Degree Program in Marine Biotechnology, National Sun Yat-Sen University, Kaohsiung 804, Taiwan
- Ph.D. Program in Biotechnology Research and Development, College of Pharmacy, Taipei Medical University, Taipei 110, Taiwan
- Department of Medicinal and Applied Chemistry, College of Life Science, Kaohsiung Medical University, Kaohsiung 807, Taiwan
- Department of Chemistry, College of Science, Tamkang University, New Taipei City 251, Taiwan
- Biomedical Translation Research Center (BioTReC), Academia Sinica, Taipei 115, Taiwan
| |
Collapse
|
27
|
Jia H, Zhang X, Liu X, Qiao R, Liu Y, Lv S, Zhu H, Wang J, Kong Q, Zhang H, Zhang Z. FABP5, a Novel Immune-Related mRNA Prognostic Marker and a Target of Immunotherapy for Multiple Myeloma. Front Med (Lausanne) 2021; 8:667525. [PMID: 34249967 PMCID: PMC8266212 DOI: 10.3389/fmed.2021.667525] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2021] [Accepted: 04/29/2021] [Indexed: 12/31/2022] Open
Abstract
Objective: Multiple myeloma is an incurable hematological malignancy. It is imperative to identify immune markers for early diagnosis and therapy. Here, this study analyzed immune-related mRNAs and assessed their prognostic value and therapeutic potential. Methods: Abnormally expressed immune-related mRNAs were screened between multiple myeloma and normal bone marrow specimens in the GSE47552 and GSE6477 datasets. Their biological functions were then explored. Survival analysis was presented for assessing prognosis-related mRNAs. CIBERSORT was utilized for identifying 22 immune cell compositions of each bone marrow specimen. Correlation between FABP5 mRNA and immune cells was then analyzed in multiple myeloma. Results: Thirty-one immune-related mRNAs were abnormally expressed in multiple myeloma, which were primarily enriched in B cells-related biological processes and pathways. Following validation, FABP5 mRNA was a key risk factor of multiple myeloma. Patients with its up-regulation usually experienced unfavorable outcomes. There were distinct differences in the infiltration levels of B cells naïve, B cells memory, plasma cells, T cells CD4 naïve, resting memory CD4 T cells, activated memory CD4 T cells, Tregs, resting NK cells, M0 macrophages, M1 macrophages, M2 macrophages, and neutrophils between multiple myeloma and normal samples. FABP5 mRNA had correlations to B cells memory, B cells naïve, dendritic cells activated, macrophages M0, macrophages M1, macrophages M2, neutrophils, activated NK cells, resting memory CD4 T cells, CD8 T cells and Tregs. Conclusion: Collectively, our data showed that FABP5 mRNA was related to immune microenvironment, which could be a target of immunotherapy and prognostic marker for multiple myeloma.
Collapse
Affiliation(s)
- Haipeng Jia
- Department of Hematology, The Second Affiliated Hospital of Shandong First Medical University, Taian, China
| | - Xiaofen Zhang
- Department of Hematology, The Second Affiliated Hospital of Shandong First Medical University, Taian, China
| | - Xinxin Liu
- Department of Hematology, The Second Affiliated Hospital of Shandong First Medical University, Taian, China
| | - Ruifang Qiao
- Respiratory Intensive Care Unit, Taian City Central Hospital, Taian, China
| | - Yan Liu
- Department of Hematology, The Second Affiliated Hospital of Shandong First Medical University, Taian, China
| | - Sulong Lv
- Department of Hematology, The Second Affiliated Hospital of Shandong First Medical University, Taian, China
| | - Hongbo Zhu
- Department of Hematology, The Second Affiliated Hospital of Shandong First Medical University, Taian, China
| | - Jie Wang
- Department of Hematology, The Second Affiliated Hospital of Shandong First Medical University, Taian, China
| | - Qiuhong Kong
- Department of Hematology, The Second Affiliated Hospital of Shandong First Medical University, Taian, China
| | - Hong Zhang
- Department of Hematology, The Second Affiliated Hospital of Shandong First Medical University, Taian, China
| | - Zhirong Zhang
- Department of Hematology, The Second Affiliated Hospital of Shandong First Medical University, Taian, China
| |
Collapse
|
28
|
Aberrant Sialylation in Cancer: Biomarker and Potential Target for Therapeutic Intervention? Cancers (Basel) 2021; 13:cancers13092014. [PMID: 33921986 PMCID: PMC8122436 DOI: 10.3390/cancers13092014] [Citation(s) in RCA: 60] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2021] [Revised: 04/19/2021] [Accepted: 04/20/2021] [Indexed: 12/14/2022] Open
Abstract
Simple Summary Sialylation is a post-translational modification that consists in the addition of sialic acid to growing glycan chains on glycoproteins and glycolipids. Aberrant sialylation is an established hallmark of several types of cancer, including breast, ovarian, pancreatic, prostate, colorectal and lung cancers, melanoma and hepatocellular carcinoma. Hypersialylation can be the effect of increased activity of sialyltransferases and results in an excess of negatively charged sialic acid on the surface of cancer cells. Sialic acid accumulation contributes to tumor progression by several paths, including stimulation of tumor invasion and migration, and enhancing immune evasion and tumor cell survival. In this review we explore the mechanisms by which sialyltransferases promote cancer progression. In addition, we provide insights into the possible use of sialyltransferases as biomarkers for cancer and summarize findings on the development of sialyltransferase inhibitors as potential anti-cancer treatments. Abstract Sialylation is an integral part of cellular function, governing many biological processes including cellular recognition, adhesion, molecular trafficking, signal transduction and endocytosis. Sialylation is controlled by the levels and the activities of sialyltransferases on glycoproteins and lipids. Altered gene expression of these enzymes in cancer yields to cancer-specific alterations of glycoprotein sialylation. Mounting evidence indicate that hypersialylation is closely associated with cancer progression and metastatic spread, and can be of prognostic significance in human cancer. Aberrant sialylation is not only a result of cancer, but also a driver of malignant phenotype, directly impacting key processes such as tumor cell dissociation and invasion, cell-cell and cell-matrix interactions, angiogenesis, resistance to apoptosis, and evasion of immune destruction. In this review we provide insights on the impact of sialylation in tumor progression, and outline the possible application of sialyltransferases as cancer biomarkers. We also summarize the most promising findings on the development of sialyltransferase inhibitors as potential anti-cancer treatments.
Collapse
|
29
|
Dunphy K, Dowling P, Bazou D, O’Gorman P. Current Methods of Post-Translational Modification Analysis and Their Applications in Blood Cancers. Cancers (Basel) 2021; 13:1930. [PMID: 33923680 PMCID: PMC8072572 DOI: 10.3390/cancers13081930] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2021] [Revised: 04/04/2021] [Accepted: 04/14/2021] [Indexed: 12/12/2022] Open
Abstract
Post-translational modifications (PTMs) add a layer of complexity to the proteome through the addition of biochemical moieties to specific residues of proteins, altering their structure, function and/or localization. Mass spectrometry (MS)-based techniques are at the forefront of PTM analysis due to their ability to detect large numbers of modified proteins with a high level of sensitivity and specificity. The low stoichiometry of modified peptides means fractionation and enrichment techniques are often performed prior to MS to improve detection yields. Immuno-based techniques remain popular, with improvements in the quality of commercially available modification-specific antibodies facilitating the detection of modified proteins with high affinity. PTM-focused studies on blood cancers have provided information on altered cellular processes, including cell signaling, apoptosis and transcriptional regulation, that contribute to the malignant phenotype. Furthermore, the mechanism of action of many blood cancer therapies, such as kinase inhibitors, involves inhibiting or modulating protein modifications. Continued optimization of protocols and techniques for PTM analysis in blood cancer will undoubtedly lead to novel insights into mechanisms of malignant transformation, proliferation, and survival, in addition to the identification of novel biomarkers and therapeutic targets. This review discusses techniques used for PTM analysis and their applications in blood cancer research.
Collapse
Affiliation(s)
- Katie Dunphy
- Department of Biology, National University of Ireland, W23 F2K8 Maynooth, Ireland; (K.D.); (P.D.)
| | - Paul Dowling
- Department of Biology, National University of Ireland, W23 F2K8 Maynooth, Ireland; (K.D.); (P.D.)
| | - Despina Bazou
- Department of Haematology, Mater Misericordiae University Hospital, D07 WKW8 Dublin, Ireland;
| | - Peter O’Gorman
- Department of Haematology, Mater Misericordiae University Hospital, D07 WKW8 Dublin, Ireland;
| |
Collapse
|
30
|
The Role of Tumor Microenvironment in Multiple Myeloma Development and Progression. Cancers (Basel) 2021; 13:cancers13020217. [PMID: 33435306 PMCID: PMC7827690 DOI: 10.3390/cancers13020217] [Citation(s) in RCA: 129] [Impact Index Per Article: 32.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Revised: 01/04/2021] [Accepted: 01/06/2021] [Indexed: 12/13/2022] Open
Abstract
Simple Summary Multiple Myeloma (MM) is a hematologic malignancy caused by aberrant plasma cell proliferation in the bone marrow (BM) and constitutes the second most common hematological disease after non-Hodgkin lymphoma. The disease progression is drastically regulated by the immunosuppressive tumor microenvironment (TME) generated by soluble factors and different cells that naturally reside in the BM. This microenvironment does not remain unchanged and alterations favor cancer dissemination. Despite therapeutic advances over the past 15 years, MM remains incurable and therefore understanding the elements that control the TME in MM would allow better-targeted therapies to cure this disease. In this review, we discuss the main events and changes that occur in the BM milieu during MM development. Abstract Multiple myeloma (MM) is a hematologic cancer characterized by clonal proliferation of plasma cells in the bone marrow (BM). The progression, from the early stages of the disease as monoclonal gammopathy of undetermined significance (MGUS) and smoldering multiple myeloma (SMM) to MM and occasionally extramedullary disease, is drastically affected by the tumor microenvironment (TME). Soluble factors and direct cell–cell interactions regulate MM plasma cell trafficking and homing to the BM niche. Mesenchymal stromal cells, osteoclasts, osteoblasts, myeloid and lymphoid cells present in the BM create a unique milieu that favors MM plasma cell immune evasion and promotes disease progression. Moreover, TME is implicated in malignant cell protection against anti-tumor therapy. This review describes the main cellular and non-cellular components located in the BM, which condition the immunosuppressive environment and lead the MM establishment and progression.
Collapse
|
31
|
Xu C, Wang S, Wu Y, Sun X, Yang D, Wang S. Recent advances in understanding the roles of sialyltransferases in tumor angiogenesis and metastasis. Glycoconj J 2021; 38:119-127. [PMID: 33411077 DOI: 10.1007/s10719-020-09967-3] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2020] [Revised: 10/25/2020] [Accepted: 12/08/2020] [Indexed: 01/04/2023]
Abstract
Abnormal glycosylation is a common characteristic of cancer cells and there is a lot of evidence that glycans can regulate the biological behavior of tumor cells. Sialylation modification, a form of glycosylation modification, plays an important role in cell recognition, cell adhesion and cell signal transduction. Abnormal sialylation on the surface of tumor cells is related to tumor migration and invasion, with abnormal expression of sialyltransferases being one of the main causes of abnormal sialylation. Recent studies provide a better understanding of the importance of the sialyltransferases, and how they influences cancer cell angiogenesis, adhesion and Epithelial-Mesenchymal Transition (EMT). The present review will provide a direction for future studies in determining the roles of sialyltransferases in cancer metastasis, and abnormal sialyltransferases are likely to be potential biomarkers for cancer.
Collapse
Affiliation(s)
- Chunyan Xu
- Department of Biochemistry and Molecular Biology, College of Basic Medical Sciences, Institute of Glycobiology, Dalian Medical University, No.9 West Section Lvshun South Road, Dalian, 116044, Liaoning Province, People's Republic of China
| | - Shidan Wang
- Department of Biochemistry and Molecular Biology, College of Basic Medical Sciences, Institute of Glycobiology, Dalian Medical University, No.9 West Section Lvshun South Road, Dalian, 116044, Liaoning Province, People's Republic of China
| | - Yinshuang Wu
- Department of Biochemistry and Molecular Biology, College of Basic Medical Sciences, Institute of Glycobiology, Dalian Medical University, No.9 West Section Lvshun South Road, Dalian, 116044, Liaoning Province, People's Republic of China
| | - Xiaoxin Sun
- Department of Biochemistry and Molecular Biology, College of Basic Medical Sciences, Institute of Glycobiology, Dalian Medical University, No.9 West Section Lvshun South Road, Dalian, 116044, Liaoning Province, People's Republic of China
| | - Deyong Yang
- Department of Urology, First Affiliated Hospital of Dalian Medical University, Dalian, 116011, Liaoning Province, People's Republic of China.
| | - Shujing Wang
- Department of Biochemistry and Molecular Biology, College of Basic Medical Sciences, Institute of Glycobiology, Dalian Medical University, No.9 West Section Lvshun South Road, Dalian, 116044, Liaoning Province, People's Republic of China.
| |
Collapse
|
32
|
Current Understanding of Myelomatous Mesenchymal Stromal Cells Extended through Advances in Experimental Methods. Cancers (Basel) 2020; 13:cancers13010025. [PMID: 33374627 PMCID: PMC7793501 DOI: 10.3390/cancers13010025] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2020] [Revised: 12/20/2020] [Accepted: 12/21/2020] [Indexed: 01/03/2023] Open
Abstract
Simple Summary As the amount of information available has grown, now it is known that many types of non-hematopoietic cells, including mesenchymal stem/progenitor cells, mature mesenchymal cells, and endothelial cells, as well as mature hematopoietic cells such as monocytes, macrophages, T-cells, and B-cells, have roles in the pathogenesis of multiple myeloma. This review focuses on the role of mesenchymal cells in the microenvironment of multiple myeloma. We summarize the experimental strategies and current understanding of the biological roles in the pathogenesis of myeloma. Furthermore, we discuss the possible clinical applications targeting mesenchymal cells. Abstract Multiple myeloma is an incurable cancer formed by malignant plasma cells. For the proliferation and survival of myeloma cells, as well as the occurrence of the complications, numerous intra- and extra-cellular mechanisms are involved. The interaction of myeloma cells with the microenvironment is known to be one of the most critical mechanisms. A specific microenvironment could affect the progression and growth of tumor cells, as well as drug resistance. Among various microenvironment components, such as hematological and non-hematological cells, and soluble factors (cytokines, chemokines, and extracellular matrix (ECM) proteins), in this review, we focus on the role of mesenchymal cells. We aimed to summarize the experimental strategies used for conducting studies and current understanding of the biological roles in the pathogenesis of myeloma. Furthermore, we discuss the possible clinical applications targeting mesenchymal cells.
Collapse
|
33
|
Fairfield H, Dudakovic A, Khatib CM, Farrell M, Costa S, Falank C, Hinge M, Murphy CS, DeMambro V, Pettitt JA, Lary CW, Driscoll HE, McDonald MM, Kassem M, Rosen C, Andersen TL, van Wijnen AJ, Jafari A, Reagan MR. Myeloma-Modified Adipocytes Exhibit Metabolic Dysfunction and a Senescence-Associated Secretory Phenotype. Cancer Res 2020; 81:634-647. [PMID: 33218968 PMCID: PMC7854508 DOI: 10.1158/0008-5472.can-20-1088] [Citation(s) in RCA: 53] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2020] [Revised: 10/05/2020] [Accepted: 11/09/2020] [Indexed: 11/16/2022]
Abstract
Bone marrow adipocytes (BMAd) have recently been implicated in accelerating bone metastatic cancers, such as acute myelogenous leukemia and breast cancer. Importantly, bone marrow adipose tissue (BMAT) expands with aging and obesity, two key risk factors in multiple myeloma disease prevalence, suggesting that BMAds may influence and be influenced by myeloma cells in the marrow. Here, we provide evidence that reciprocal interactions and cross-regulation of myeloma cells and BMAds play a role in multiple myeloma pathogenesis and treatment response. Bone marrow biopsies from patients with multiple myeloma revealed significant loss of BMAT with myeloma cell infiltration of the marrow, whereas BMAT was restored after treatment for multiple myeloma. Myeloma cells reduced BMAT in different preclinical murine models of multiple myeloma and in vitro using myeloma cell-adipocyte cocultures. In addition, multiple myeloma cells altered adipocyte gene expression and cytokine secretory profiles, which were also associated with bioenergetic changes and induction of a senescent-like phenotype. In vivo, senescence markers were also increased in the bone marrow of tumor-burdened mice. BMAds, in turn, provided resistance to dexamethasone-induced cell-cycle arrest and apoptosis, illuminating a new possible driver of myeloma cell evolution in a drug-resistant clone. Our findings reveal that bidirectional interactions between BMAds and myeloma cells have significant implications for the pathogenesis and treatment of multiple myeloma. Targeting senescence in the BMAd or other bone marrow cells may represent a novel therapeutic approach for treatment of multiple myeloma. SIGNIFICANCE: This study changes the foundational understanding of how cancer cells hijack the bone marrow microenvironment and demonstrates that tumor cells induce senescence and metabolic changes in adipocytes, potentially driving new therapeutic directions.
Collapse
Affiliation(s)
- Heather Fairfield
- Maine Medical Center Research Institute, Scarborough, Maine.,Tufts University School of Medicine, Boston, Massachusetts.,University of Maine Graduate School of Biomedical Science and Engineering, Orono, Maine
| | - Amel Dudakovic
- Departments of Orthopedic Surgery and Biochemistry & Molecular Biology, Mayo Clinic, Rochester, Minnesota
| | - Casper M Khatib
- Department of Cellular and Molecular Medicine, Novo Nordisk Foundation Center for Stem Cell Biology (DanStem), University of Copenhagen, Copenhagen, Denmark
| | - Mariah Farrell
- Maine Medical Center Research Institute, Scarborough, Maine.,Tufts University School of Medicine, Boston, Massachusetts.,University of Maine Graduate School of Biomedical Science and Engineering, Orono, Maine
| | - Samantha Costa
- Maine Medical Center Research Institute, Scarborough, Maine.,Tufts University School of Medicine, Boston, Massachusetts.,University of Maine Graduate School of Biomedical Science and Engineering, Orono, Maine
| | - Carolyne Falank
- Maine Medical Center Research Institute, Scarborough, Maine.,Tufts University School of Medicine, Boston, Massachusetts.,University of Maine Graduate School of Biomedical Science and Engineering, Orono, Maine
| | - Maja Hinge
- Division of Haematology, Department of Internal Medicine, Vejle Hospital, Vejle, Denmark
| | - Connor S Murphy
- Maine Medical Center Research Institute, Scarborough, Maine.,Tufts University School of Medicine, Boston, Massachusetts.,University of Maine Graduate School of Biomedical Science and Engineering, Orono, Maine
| | - Victoria DeMambro
- Maine Medical Center Research Institute, Scarborough, Maine.,Tufts University School of Medicine, Boston, Massachusetts.,University of Maine Graduate School of Biomedical Science and Engineering, Orono, Maine
| | - Jessica A Pettitt
- The Garvan Institute of Medical Research, Sydney, New South Wales, Australia
| | | | | | - Michelle M McDonald
- The Garvan Institute of Medical Research, Sydney, New South Wales, Australia
| | - Moustapha Kassem
- Department of Cellular and Molecular Medicine, Novo Nordisk Foundation Center for Stem Cell Biology (DanStem), University of Copenhagen, Copenhagen, Denmark.,Molecular Endocrinology & Stem Cell Research Unit (KMEB), Department of Endocrinology and Metabolism, Odense University Hospital & University of Southern Denmark, Odense, Denmark
| | - Clifford Rosen
- Maine Medical Center Research Institute, Scarborough, Maine.,Tufts University School of Medicine, Boston, Massachusetts.,University of Maine Graduate School of Biomedical Science and Engineering, Orono, Maine
| | - Thomas L Andersen
- Clinical Cell Biology, Department of Regional Health Research, Vejle/Lillebaelt Hospital, University of Southern Denmark, Vejle, Denmark.,Clinical Cell Biology, Department of Pathology, Odense University Hospital - Department of Clinical Research, University of Southern Denmark, Odense, Denmark
| | - Andre J van Wijnen
- Departments of Orthopedic Surgery and Biochemistry & Molecular Biology, Mayo Clinic, Rochester, Minnesota
| | - Abbas Jafari
- Department of Cellular and Molecular Medicine, Novo Nordisk Foundation Center for Stem Cell Biology (DanStem), University of Copenhagen, Copenhagen, Denmark.
| | - Michaela R Reagan
- Maine Medical Center Research Institute, Scarborough, Maine. .,Tufts University School of Medicine, Boston, Massachusetts.,University of Maine Graduate School of Biomedical Science and Engineering, Orono, Maine
| |
Collapse
|
34
|
Dobie C, Skropeta D. Insights into the role of sialylation in cancer progression and metastasis. Br J Cancer 2020; 124:76-90. [PMID: 33144696 PMCID: PMC7782833 DOI: 10.1038/s41416-020-01126-7] [Citation(s) in RCA: 189] [Impact Index Per Article: 37.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2020] [Revised: 09/11/2020] [Accepted: 10/08/2020] [Indexed: 02/07/2023] Open
Abstract
Upregulation of sialyltransferases—the enzymes responsible for the addition of sialic acid to growing glycoconjugate chains—and the resultant hypersialylation of up to 40–60% of tumour cell surfaces are established hallmarks of several cancers, including lung, breast, ovarian, pancreatic and prostate cancer. Hypersialylation promotes tumour metastasis by several routes, including enhancing immune evasion and tumour cell survival, and stimulating tumour invasion and migration. The critical role of enzymes that regulate sialic acid in tumour cell growth and metastasis points towards targeting sialylation as a potential new anti-metastatic cancer treatment strategy. Herein, we explore insights into the mechanisms by which hypersialylation plays a role in promoting metastasis, and explore the current state of sialyltransferase inhibitor development.
Collapse
Affiliation(s)
- Christopher Dobie
- School of Chemistry & Molecular Bioscience, Faculty of Science, Medicine & Health; and Molecular Horizons, University of Wollongong, NSW, 2522, Wollongong, Australia
| | - Danielle Skropeta
- School of Chemistry & Molecular Bioscience, Faculty of Science, Medicine & Health; and Molecular Horizons, University of Wollongong, NSW, 2522, Wollongong, Australia. .,Illawarra Health & Medical Research Institute, Wollongong, NSW, 2522, Australia.
| |
Collapse
|
35
|
Hao P, Zhang C, Wang R, Yan P, Peng R. Expression and pathogenesis of VCAM-1 and VLA-4 cytokines in multiple myeloma. Saudi J Biol Sci 2020; 27:1674-1678. [PMID: 32489310 PMCID: PMC7254040 DOI: 10.1016/j.sjbs.2020.04.031] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2020] [Revised: 04/15/2020] [Accepted: 04/16/2020] [Indexed: 12/02/2022] Open
Abstract
Objective The objective of this study is to investigate the expression of Vascular cell adhesion molecule-1 (VCAM-1) and very late appearing antigen-4 (VLA-4) cytokines in MM (multiple Myeloma). Method Forty patients with MM are selected as the experimental group and 30 healthy persons as the control group. Flow cytometry is used to detect the expression of VCAM-1 (CD106), VLA-4 (CD49d), CD38 and CD138 antigens in experimental group and control group. ELISA (Enzyme Linked Immunosorbent Assay) is used to detect the concentration of VCAM-1 in serum of experimental group and control group. RT-PCR is used to detect the expression of VCAM-1. Results The positive rate and antigen expression rate of VACM-1 antigen in the experimental group were significantly higher than those in the control group (P < 0.05). There were statistical differences of VLA-4 and VCAM-1 antigens between the initial diagnosis group and the relapse/refractory group, and between the relapse/refractory group and the platform stage group (P < 0.05). There were significant differences between VLA-4 antigen and VACM-1 antigen, phase I and phase II, and between phase I and phase III (P < 0.05). The concentration of VCAM-1 and the expression of VCAM-1 mRNA in the experimental group were significantly higher than (P < 0.01). In the different stages of ISS (International Staging System) and different disease groups in the experimental group, the concentration of VCAM-1 and the expression level of VCAM-1 mRNA are significantly different among the three groups of stage I, II and III (P < 0.01). There is a significant difference between the initial diagnosis group, the relapse/refractory group and the platform group (P < 0.05). Conclusion There are abnormal expressions of adhesion molecules VCAM-1 and VLA-4 in multiple myeloma patients, which are related to ISS staging.
Collapse
Affiliation(s)
- Pan Hao
- Department of Nuclear Medicine, LuHe Hospital, Capital Medical University, Beijing City 101149, China
| | - Chunli Zhang
- Department of Nuclear Medicine, Peaking University First Hospital, Beijing City 100011, China
| | - Rongfu Wang
- Department of Nuclear Medicine, Peaking University First Hospital, Beijing City 100011, China
| | - Ping Yan
- Department of Nuclear Medicine, Peaking University First Hospital, Beijing City 100011, China
| | - Ruchen Peng
- Department of Medical Imaging Center Nuclear Medicine, LuHe Hospital, Capital Medical University, Beijing City 101149, China
| |
Collapse
|
36
|
The CD38 low natural killer cell line KHYG1 transiently expressing CD16 F158V in combination with daratumumab targets multiple myeloma cells with minimal effector NK cell fratricide. Cancer Immunol Immunother 2020; 69:421-434. [PMID: 31919623 DOI: 10.1007/s00262-019-02477-8] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2019] [Accepted: 12/31/2019] [Indexed: 12/23/2022]
Abstract
Multiple myeloma (MM) is a clonal plasma cell malignancy typically associated with the high and uniform expression of the CD38 transmembrane glycoprotein. Daratumumab is a humanized IgG1κ CD38 monoclonal antibody (MoAb) which has demonstrated impressive single agent activity even in relapsed refractory MM patients as well as strong synergy with other anti-MM drugs. Natural Killer (NK) cells are cytotoxic immune effector cells that mediate in vivo tumour immunosurveillance. NK cells also play an important role during MoAb therapy by inducing antibody dependent cellular cytotoxicity (ADCC) via their FcγRIII (CD16) receptor. Furthermore, 15% of the population express a naturally occurring variant of CD16 harbouring a single-point polymorphism (F158V). However, the contribution of NK cells to the efficacy of daratumumab remains debatable as clinical data clearly indicate the rapid depletion of CD38high peripheral blood NK cells in patients upon daratumumab administration. In contrast, CD38low peripheral blood NK cells have been shown to survive daratumumab mediated fratricide in vivo, while still retaining their potent anti-MM cytolytic effector functions ex vivo. Therefore, we hypothesize that transiently expressing the CD16F158V receptor using a "safe" mRNA electroporation-based approach on CD38low NK cells in combination with daratumumab could represent a novel therapeutic option for treatment of MM. In the present study, we investigate a NK cell line (KHYG-1), derived from a patient with aggressive NK cell leukemia, as a platform for generating CD38low NK cells expressing CD16F158V which can be administered as an "off-the-shelf" therapy to target both CD38high and CD38low tumour clones in patients receiving daratumumab.
Collapse
|
37
|
Fu L, Cheng Z, Dong F, Quan L, Cui L, Liu Y, Zeng T, Huang W, Chen J, Pang Y, Ye X, Wu G, Qian T, Chen Y, Si C. Enhanced expression of FCER1G predicts positive prognosis in multiple myeloma. J Cancer 2020; 11:1182-1194. [PMID: 31956364 PMCID: PMC6959079 DOI: 10.7150/jca.37313] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2019] [Accepted: 11/03/2019] [Indexed: 12/12/2022] Open
Abstract
Background: Multiple myeloma (MM) is the second most common hematologic malignancy worldwide and does not have sufficient prognostic indicators. FCER1G (Fc fragment Of IgE receptor Ig) is located on chromosome 1q23.3 and is involved in the innate immunity. Early studies have shown that FCER1G participates in many immune-related pathways encompassing multiple cell types. Meanwhile, it is associated with many malignancies. However, the relationship between MM and FCER1G has not been studied. Methods: In this study, we integrated nine independent gene expression omnibus (GEO) datasets and analyzed the associations of FCER1G expression and myeloma progression, ISS stage, 1q21 amplification and survival in 2296 myeloma patients and 48 healthy donors. Results: The expression of FCER1G showed a decreasing trend with the advance of myeloma. As ISS stage and 1q21 amplification level increased, the expression of FCER1G decreased (P = 0.0012 and 0.0036, respectively). MM patients with high FCER1G expression consistently had longer EFS and OS across three large sample datasets (EFS: P = 0.0057, 0.0049, OS: P = 0.0014, 0.00065, 0.0019 and 0.0029, respectively). Meanwhile, univariate and multivariate analysis indicated that high FCER1G expression was an independent favorable prognostic factor for EFS and OS in MM patients (EFS: P = 0.006, 0.027, OS: P =0.002,0.025, respectively). Conclusions: The expression level of FCER1G negatively correlated with myeloma progression, and high FCER1G expression may be applied as a favorable biomarker in MM patients.
Collapse
Affiliation(s)
- Lin Fu
- Department of Hematology, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510260, China.,Translational Medicine Center, State Key Laboratory of Respiratory Disease, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou 510260, China.,Department of Hematology, Huaihe Hospital of Henan University, Kaifeng, China
| | - Zhiheng Cheng
- Department of Pathology and Medical Biology, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | - Fen Dong
- Institute of Clinical Medical Sciences, China-Japan Friendship Hospital, Beijing, 100029, China
| | - Liang Quan
- Department of Hematology, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510260, China.,Translational Medicine Center, State Key Laboratory of Respiratory Disease, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou 510260, China
| | - Longzhen Cui
- Translational Medicine Center, Huaihe Hospital of Henan University, Kaifeng, China
| | - Yan Liu
- Translational Medicine Center, Huaihe Hospital of Henan University, Kaifeng, China
| | - Tiansheng Zeng
- Department of Biomedical Sciences, University of Sassari, Sassari, 07100, Italy
| | - Wenhui Huang
- Department of Hematology, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510260, China.,Translational Medicine Center, State Key Laboratory of Respiratory Disease, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou 510260, China
| | - Jinghong Chen
- Translational Medicine Center, State Key Laboratory of Respiratory Disease, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou 510260, China
| | - Ying Pang
- Department of Hematology, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510260, China
| | - Xu Ye
- Department of Hematology, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510260, China
| | - Guangsheng Wu
- Department of Hematology, First Affiliated Hospital, Medical College of Shihezi University, Shihezi 832008, China
| | - Tingting Qian
- Department of Hematology, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510260, China.,Translational Medicine Center, State Key Laboratory of Respiratory Disease, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou 510260, China
| | - Yang Chen
- MOE Key Laboratory of Bioinformatics; Bioinformatics Division and Center for Synthetic & Systems Biology, TNLIST; Department of Automation, Tsinghua University, Beijing, 100084, China
| | - Chaozeng Si
- Department of Operations and Information Management, China-Japan Friendship Hospital, Beijing, 100029, China
| |
Collapse
|
38
|
Natoni A, Bohara R, Pandit A, O'Dwyer M. Targeted Approaches to Inhibit Sialylation of Multiple Myeloma in the Bone Marrow Microenvironment. Front Bioeng Biotechnol 2019; 7:252. [PMID: 31637237 PMCID: PMC6787837 DOI: 10.3389/fbioe.2019.00252] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2019] [Accepted: 09/17/2019] [Indexed: 11/13/2022] Open
Abstract
Aberrant glycosylation modulates different aspects of tumor biology, and it has long been recognized as a hallmark of cancer. Among the different forms of glycosylation, sialylation, the addition of sialic acid to underlying oligosaccharides, is often dysregulated in cancer. Increased expression of sialylated glycans has been observed in many types of cancer, including multiple myeloma, and often correlates with aggressive metastatic behavior. Myeloma, a cancer of plasma cells, develops in the bone marrow, and colonizes multiple sites of the skeleton including the skull. In myeloma, the bone marrow represents an essential niche where the malignant cells are nurtured by the microenvironment and protected from chemotherapy. Here, we discuss the role of hypersialylation in the metastatic process focusing on multiple myeloma. In particular, we examine how increased sialylation modulates homing of malignant plasma cells into the bone marrow by regulating the activity of molecules important in bone marrow cellular trafficking including selectins and integrins. We also propose that inhibiting sialylation may represent a new therapeutic strategy to overcome bone marrow-mediated chemotherapy resistance and describe different targeted approaches to specifically deliver sialylation inhibitors to the bone marrow microenvironment.
Collapse
Affiliation(s)
- Alessandro Natoni
- Apoptosis Research Centre, School of Medicine, National University of Ireland, Galway, Ireland
| | - Raghvendra Bohara
- Centre for Research in Medical Devices (CÚRAM), National University of Ireland, Galway, Ireland
| | - Abhay Pandit
- Centre for Research in Medical Devices (CÚRAM), National University of Ireland, Galway, Ireland
| | - Michael O'Dwyer
- Apoptosis Research Centre, School of Medicine, National University of Ireland, Galway, Ireland
| |
Collapse
|