1
|
Zhang P, Wang L, Liu H, Lin S, Guo D. Unveiling the crucial role of glycosylation modification in lung adenocarcinoma metastasis through artificial neural network-based spatial multi-omics single-cell analysis and Mendelian randomization. BMC Cancer 2025; 25:249. [PMID: 39948531 PMCID: PMC11823056 DOI: 10.1186/s12885-025-13650-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2024] [Accepted: 02/05/2025] [Indexed: 02/17/2025] Open
Abstract
BACKGROUND Investigations into the intricacies of glycosylation modifications, a prevalent post-translational alteration observed in neoplasms, especially remain elusive in the context of lung adenocarcinoma. Through the integration of multiple omics approaches, the investigation aimed to delineate the significance of glycosylation in lung adenocarcinoma, with an objective to pinpoint viable biological targets. METHODS Initial steps involved the identification of genes differentially expressed in relation to glycosylation at the aggregate transcriptome level within lung adenocarcinoma tissues. This was followed by analyses of localization and function employing both single-cell and spatial transcriptomics to provide a more nuanced understanding. In pursuit of elucidating functional disparities in glycosylation patterns, a predictive framework employing artificial neural networks was constructed. To ascertain causal relationships between specific genes and lung adenocarcinoma, Mendelian randomization was applied, culminating in the experimental validation of these genes' roles. RESULTS Analysis at the single-cell level uncovered marked glycosylation modification expressions in metastatic tissues of lung adenocarcinoma. Moreover, tissues of lung adenocarcinoma with elevated expression of genes associated with glycosylation displayed enhanced differentiation and activation across signaling pathways including TGF-β, oxidative stress, and WNT. Through spatial transcriptomics, zones of intense glycosylation modification were pinpointed within tumor nests and proximate to tumor-associated blood vessels. An artificial neural network-derived prognostic model demonstrated outstanding predictive capability, with AUC scores achieving 0.84, 0.83, and 0.89 for 1, 3, and 5-year forecasts, respectively. The group identified as high-risk was characterized by pronounced immunosuppression and diminished responsiveness to immunotherapy. Mendelian randomization analysis pinpointed GLANT2 (OR = 1.3654, p < 0.05) and GYS1 (OR = 1.2668, p < 0.05) as genes contributing to the pathogenesis of lung adenocarcinoma. Cell assays have reaffirmed that the inhibition of GYS1 significantly reduces proliferation and invasion in lung adenocarcinoma cell lines, while also decreasing glycogen storage and the formation of glycosylation end products, indicating suppression of glycosylation processes. These findings identify GYS1 as a prospective glycosylation-linked biological target for lung adenocarcinoma therapy.
Collapse
Affiliation(s)
- Penngcheng Zhang
- Department of General Surgery, The Third Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
- The First Affiliated Hospital of Zhejiang Chinese Medical University (Zheiiang Provincial Hospital of Chinese Medicine), Hangzhou, Zhejiang, China
| | - Lexin Wang
- General Hospital of Ningxia Medical University, Yinchuan, Ningxia, China
- Western Institute of Digital-Intelligent Medicine, Chongqing, China
| | - Hanwen Liu
- Department of General Surgery, The Third Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
| | - Shengyou Lin
- The First Affiliated Hospital of Zhejiang Chinese Medical University (Zheiiang Provincial Hospital of Chinese Medicine), Hangzhou, Zhejiang, China.
| | - Dechao Guo
- Department of General Surgery, The Third Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China.
| |
Collapse
|
2
|
Lee H, Ko N, Namgoong S, Ham S, Koo J. Recent advances in and applications of ex vivo drug sensitivity analysis for blood cancers. Blood Res 2024; 59:37. [PMID: 39503808 PMCID: PMC11541977 DOI: 10.1007/s44313-024-00032-8] [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: 07/31/2024] [Accepted: 09/06/2024] [Indexed: 11/09/2024] Open
Abstract
Blood cancers, including leukemia, multiple myeloma, and lymphoma, pose significant challenges owing to their heterogeneous nature and the limitations of traditional treatments. Precision medicine has emerged as a transformative approach that offers tailored therapeutic strategies based on individual patient profiles. Ex vivo drug sensitivity analysis is central to this advancement, which enables testing of patient-derived cancer cells against a panel of therapeutic agents to predict clinical responses. This review provides a comprehensive overview of the latest advancements in ex vivo drug sensitivity analyses and their application in blood cancers. We discuss the development of more comprehensive drug response metrics and the evaluation of drug combinations to identify synergistic interactions. Additionally, we present evaluation of the advanced therapeutics such as antibody-drug conjugates using ex vivo assays. This review describes the critical role of ex vivo drug sensitivity analyses in advancing precision medicine by examining technological innovations and clinical applications. Ultimately, these innovations are paving the way for more effective and individualized treatments, improving patient outcomes, and establishing new standards for the management of blood cancers.
Collapse
Affiliation(s)
- Haeryung Lee
- Department of Chemical Engineering, Hongik University, Seoul, 04066, Republic of Korea
| | - Nahee Ko
- Department of Chemical Engineering, Hongik University, Seoul, 04066, Republic of Korea
| | - Sujin Namgoong
- Department of Chemical Engineering, Hongik University, Seoul, 04066, Republic of Korea
| | - Seunghyok Ham
- ImpriMedKorea, Inc., Seoul, 03920, Republic of Korea
| | - Jamin Koo
- Department of Chemical Engineering, Hongik University, Seoul, 04066, Republic of Korea.
- ImpriMedKorea, Inc., Seoul, 03920, Republic of Korea.
| |
Collapse
|
3
|
Visconti A, Rossi N, Bondt A, Ederveen AH, Thareja G, Koeleman CAM, Stephan N, Halama A, Lomax-Browne HJ, Pickering MC, Zhou XJ, Wuhrer M, Suhre K, Falchi M. The genetics and epidemiology of N- and O-immunoglobulin A glycomics. Genome Med 2024; 16:96. [PMID: 39123268 PMCID: PMC11312925 DOI: 10.1186/s13073-024-01369-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2024] [Accepted: 07/26/2024] [Indexed: 08/12/2024] Open
Abstract
BACKGROUND Immunoglobulin (Ig) glycosylation modulates the immune response and plays a critical role in ageing and diseases. Studies have mainly focused on IgG glycosylation, and little is known about the genetics and epidemiology of IgA glycosylation. METHODS We generated, using a novel liquid chromatography-mass spectrometry method, the first large-scale IgA glycomics dataset in serum from 2423 twins, encompassing 71 N- and O-glycan species. RESULTS We showed that, despite the lack of a direct genetic template, glycosylation is highly heritable, and that glycopeptide structures are sex-specific, and undergo substantial changes with ageing. We observe extensive correlations between the IgA and IgG glycomes, and, exploiting the twin design, show that they are predominantly influenced by shared genetic factors. A genome-wide association study identified eight loci associated with both the IgA and IgG glycomes (ST6GAL1, ELL2, B4GALT1, ABCF2, TMEM121, SLC38A10, SMARCB1, and MGAT3) and two novel loci specifically modulating IgA O-glycosylation (C1GALT1 and ST3GAL1). Validation of our findings in an independent cohort of 320 individuals from Qatar showed that the underlying genetic architecture is conserved across ancestries. CONCLUSIONS Our study delineates the genetic landscape of IgA glycosylation and provides novel potential functional links with the aetiology of complex immune diseases, including genetic factors involved in IgA nephropathy risk.
Collapse
Affiliation(s)
- Alessia Visconti
- Department of Twin Research and Genetic Epidemiology, King's College London, London, UK
- Center for Biostatistics, Epidemiology and Public Health, Department of Clinical and Biological Sciences, University of Turin, Turin, Italy
| | - Niccolò Rossi
- Department of Twin Research and Genetic Epidemiology, King's College London, London, UK
| | - Albert Bondt
- Center for Proteomics and Metabolomics, Leiden University Medical Center, Leiden, The Netherlands
| | - Agnes Hipgrave Ederveen
- Center for Proteomics and Metabolomics, Leiden University Medical Center, Leiden, The Netherlands
| | - Gaurav Thareja
- Department of Biophysics and Physiology, Weill Cornell Medicine-Qatar, Doha, Qatar
| | - Carolien A M Koeleman
- Center for Proteomics and Metabolomics, Leiden University Medical Center, Leiden, The Netherlands
| | - Nisha Stephan
- Department of Biophysics and Physiology, Weill Cornell Medicine-Qatar, Doha, Qatar
| | - Anna Halama
- Department of Biophysics and Physiology, Weill Cornell Medicine-Qatar, Doha, Qatar
| | - Hannah J Lomax-Browne
- Centre for Inflammatory Disease, Department of Immunology and Inflammation, Imperial College London, London, UK
| | - Matthew C Pickering
- Centre for Inflammatory Disease, Department of Immunology and Inflammation, Imperial College London, London, UK
| | - Xu-Jie Zhou
- Renal Division, Peking University First Hospital, Beijing, China
- Peking University Institute of Nephrology, Beijing, China
- Key Laboratory of Renal Disease, Ministry of Health of China, Beijing, China
- Key Laboratory of Chronic Kidney Disease Prevention and Treatment, Ministry of Education, Peking University, Beijing, China
| | - Manfred Wuhrer
- Center for Proteomics and Metabolomics, Leiden University Medical Center, Leiden, The Netherlands
| | - Karsten Suhre
- Department of Biophysics and Physiology, Weill Cornell Medicine-Qatar, Doha, Qatar
| | - Mario Falchi
- Department of Twin Research and Genetic Epidemiology, King's College London, London, UK.
| |
Collapse
|
4
|
Hu D, Kobayashi N, Ohki R. FUCA1: An Underexplored p53 Target Gene Linking Glycosylation and Cancer Progression. Cancers (Basel) 2024; 16:2753. [PMID: 39123480 PMCID: PMC11311387 DOI: 10.3390/cancers16152753] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2024] [Revised: 07/26/2024] [Accepted: 07/31/2024] [Indexed: 08/12/2024] Open
Abstract
Cancer is a difficult-to-cure disease with high worldwide incidence and mortality, in large part due to drug resistance and disease relapse. Glycosylation, which is a common modification of cellular biomolecules, was discovered decades ago and has been of interest in cancer research due to its ability to influence cellular function and to promote carcinogenesis. A variety of glycosylation types and structures regulate the function of biomolecules and are potential targets for investigating and treating cancer. The link between glycosylation and carcinogenesis has been more recently revealed by the role of p53 in energy metabolism, including the p53 target gene alpha-L-fucosidase 1 (FUCA1), which plays an essential role in fucosylation. In this review, we summarize roles of glycan structures and glycosylation-related enzymes to cancer development. The interplay between glycosylation and tumor microenvironmental factors is also discussed, together with involvement of glycosylation in well-characterized cancer-promoting mechanisms, such as the epidermal growth factor receptor (EGFR), phosphatidylinositol-3-kinase/protein kinase B (PI3K/Akt) and p53-mediated pathways. Glycan structures also modulate cell-matrix interactions, cell-cell adhesion as well as cell migration and settlement, dysfunction of which can contribute to cancer. Thus, further investigation of the mechanistic relationships among glycosylation, related enzymes and cancer progression may provide insights into potential novel cancer treatments.
Collapse
Affiliation(s)
- Die Hu
- Department of Pharmacology and Toxicology, University of Toronto, Toronto, ON M5S 1A8, Canada;
| | - Naoya Kobayashi
- Laboratory of Fundamental Oncology, National Cancer Center Research Institute, Tsukiji 5-1-1, Chuo-ku, Tokyo 104-0045, Japan;
- Department of NCC Cancer Science, Graduate School of Medical and Dental Science, Tokyo Medical and Dental University (TMDU), 1-5-45 Yushima, Bunkyo-ku, Tokyo 113-8510, Japan
| | - Rieko Ohki
- Laboratory of Fundamental Oncology, National Cancer Center Research Institute, Tsukiji 5-1-1, Chuo-ku, Tokyo 104-0045, Japan;
| |
Collapse
|
5
|
Gu J, Isaji T. Specific sialylation of N-glycans and its novel regulatory mechanism. Glycoconj J 2024; 41:175-183. [PMID: 38958800 PMCID: PMC11329402 DOI: 10.1007/s10719-024-10157-8] [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: 11/29/2022] [Revised: 06/04/2024] [Accepted: 06/19/2024] [Indexed: 07/04/2024]
Abstract
Altered glycosylation is a common feature of cancer cells. Some subsets of glycans are found to be frequently enriched on the tumor cell surface and implicated in different tumor phenotypes. Among these, changes in sialylation have long been associated with metastatic cell behaviors such as invasion and enhanced cell survival. Sialylation typically exists in three prominent linkages: α2,3, α2,6, and α2,8, catalyzed by a group of sialyltransferases. The aberrant expression of all three linkages has been related to cancer progression. The increased α2,6 sialylation on N-glycans catalyzed by β-galactoside α2,6 sialyltransferase 1 (ST6Gal1) is frequently observed in many cancers. In contrast, functions of α2,3 sialylation on N-glycans catalyzed by at least three β-galactoside α2,3-sialyltransferases, ST3Gal3, ST3Gal4, and ST3Gal6 remain elusive due to a possibility of compensating for one another. In this minireview, we briefly describe functions of sialylation and recent findings that different α2,3 sialyltransferases specifically modify target proteins, as well as sialylation regulatory mechanisms vis a complex formation among integrin α3β1, Golgi phosphoprotein 3 (GOLPH3), phosphatidylinositol 4-kinase IIα (PI4KIIα), focal adhesion kinase (FAK) and sialyltransferase, which suggests a new concept for the regulation of glycosylation in cell biology.
Collapse
Affiliation(s)
- Jianguo Gu
- Division of Regulatory Glycobiology, Institute of Molecular Biomembrane and Glycobiology, Tohoku Medical and Pharmaceutical University, 4-4-1 Komatsushima, Aoba-ku, Sendai, Miyagi, 981-8558, Japan.
| | - Tomoya Isaji
- Division of Regulatory Glycobiology, Institute of Molecular Biomembrane and Glycobiology, Tohoku Medical and Pharmaceutical University, 4-4-1 Komatsushima, Aoba-ku, Sendai, Miyagi, 981-8558, Japan
| |
Collapse
|
6
|
Giannakoulas A, Nikolaidis M, Amoutzias GD, Giannakoulas N. A comparative analysis of transcriptomics of newly diagnosed multiple myeloma: exploring drug repurposing. Front Oncol 2024; 14:1390105. [PMID: 38690165 PMCID: PMC11058662 DOI: 10.3389/fonc.2024.1390105] [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: 02/22/2024] [Accepted: 04/01/2024] [Indexed: 05/02/2024] Open
Abstract
Multiple myeloma (MM) is an incurable malignant plasma cell disorder characterized by the infiltration of clonal plasma cells in the bone marrow compartment. Gene Expression Profiling (GEP) has emerged as a powerful investigation tool in modern myeloma research enabling the dissection of the molecular background of MM and allowing the identification of gene products that could potentially serve as targets for therapeutic intervention. In this study we investigated shared transcriptomic abnormalities across newly diagnosed multiple myeloma (NDMM) patient cohorts. In total, publicly available transcriptomic data of 7 studies from CD138+ cells from 281 NDMM patients and 44 healthy individuals were integrated and analyzed. Overall, we identified 28 genes that were consistently differentially expressed (DE) between NDMM patients and healthy donors (HD) across various studies. Of those, 9 genes were over/under-expressed in more than 75% of NDMM patients. In addition, we identified 4 genes (MT1F, PURPL, LINC01239 and LINC01480) that were not previously considered to participate in MM pathogenesis. Meanwhile, by mining three drug databases (ChEMBL, IUPHAR/BPS and DrugBank) we identified 31 FDA-approved and 144 experimental drugs that target 8 of these 28 over/under-expressed MM genes. Taken together, our study offers new insights in MM pathogenesis and importantly, it reveals potential new treatment options that need to be further investigated in future studies.
Collapse
Affiliation(s)
- Angelos Giannakoulas
- Department of Hematology, Faculty of Medicine, School of Health Sciences, University of Thessaly, Larissa, Greece
| | - Marios Nikolaidis
- Bioinformatics Laboratory, Department of Biochemistry & Biotechnology, School of Health Sciences, University of Thessaly, Larissa, Greece
| | - Grigorios D. Amoutzias
- Bioinformatics Laboratory, Department of Biochemistry & Biotechnology, School of Health Sciences, University of Thessaly, Larissa, Greece
| | - Nikolaos Giannakoulas
- Department of Hematology, Faculty of Medicine, School of Health Sciences, University of Thessaly, Larissa, Greece
| |
Collapse
|
7
|
Tsai HE, Chen CL, Chang TT, Fu CW, Chen WC, Perez SJLP, Hsiao PW, Tai MH, Li WS. Development of a Novel, Potent, and Selective Sialyltransferase Inhibitor for Suppressing Cancer Metastasis. Int J Mol Sci 2024; 25:4283. [PMID: 38673867 PMCID: PMC11050067 DOI: 10.3390/ijms25084283] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2024] [Revised: 04/01/2024] [Accepted: 04/08/2024] [Indexed: 04/28/2024] Open
Abstract
Sialyltransferase-catalyzed membrane protein and lipid glycosylation plays a vital role as one of the most abundant post-translational modifications and diversification reactions in eukaryotes. However, aberrant sialylation has been associated with cancer malignancy and metastasis. Sialyltransferases thus represent emerging targets for the development of small molecule cancer drugs. Herein, we report the inhibitory effects of a recently discovered lithocholic acid derivative FCW393 on sialyltransferase catalytic activity, integrin sialyation, cancer-associated signal transduction, MDA-MB-231 and B16F10 cell migration and invasion, and in in vivo studies, on tumor growth, metastasis, and angiogenesis. FCW393 showed effective and selective inhibition of the sialyltransferases ST6GAL1 (IC50 = 7.8 μM) and ST3GAL3 (IC50 = 9.45 μM) relative to ST3GAL1 (IC50 > 400 μM) and ST8SIA4 (IC50 > 100 μM). FCW393 reduced integrin sialylation in breast cancer and melanoma cells dose-dependently and downregulated proteins associated with the integrin-regulated FAK/paxillin and GEF/Rho/ROCK pathways, and with the VEGF-regulated Akt/NFκB/HIF-1α pathway. FCW393 inhibited cell migration (IC50 = 2.6 μM) and invasion in in vitro experiments, and in in vivo studies of tumor-bearing mice, FCW393 reduced tumor size, angiogenesis, and metastatic potential. Based on its demonstrated selectivity, cell permeability, relatively low cytotoxicity (IC50 = 55 μM), and high efficacy, FCW393 shows promising potential as a small molecule experimental tool compound and a lead for further development of a novel cancer therapeutic.
Collapse
Grants
- AS-KPQ-110-EIMD, AS-KPQ-109-BioMed, AS-KPQ-110-BioMed and AS-KPQ-111-KNT Academia Sinica
- MOST, Taiwan, MOST 110-0210-01-22-02, MOST-108-3114-Y-001-002, MOST 108-3111-Y-001-056, MOST 106-2113-M-001-011, MOST 103-2325-B-001-001 and MOST108-2314-B-110-003-MY2 Ministry of Science and Technology, TAIWAN
- 108-36 Kaohsiung Armed Forces General Hospital, TAIWAN
Collapse
Affiliation(s)
- Han-En Tsai
- Institute of Chemistry, Academia Sinica, Taipei 115, Taiwan (C.-W.F.); (S.J.L.P.P.)
| | - Chia-Ling Chen
- Institute of Chemistry, Academia Sinica, Taipei 115, Taiwan (C.-W.F.); (S.J.L.P.P.)
| | - Tzu-Ting Chang
- Biomedical Translation Research Center, Academia Sinica, National Biotechnology Research Park, Taipei 115, Taiwan
| | - Chih-Wei Fu
- Institute of Chemistry, Academia Sinica, Taipei 115, Taiwan (C.-W.F.); (S.J.L.P.P.)
- Department of Chemistry, National Central University, Taoyuan 320, Taiwan
| | - Wei-Chia Chen
- Institute of Chemistry, Academia Sinica, Taipei 115, Taiwan (C.-W.F.); (S.J.L.P.P.)
- Department of Chemistry, National Taiwan Normal University, Taipei 106, Taiwan
| | - Ser John Lynon P. Perez
- Institute of Chemistry, Academia Sinica, Taipei 115, Taiwan (C.-W.F.); (S.J.L.P.P.)
- Biomedical Translation Research Center, Academia Sinica, National Biotechnology Research Park, Taipei 115, Taiwan
- Department of Applied Chemistry, National Yang Ming Chiao Tung University, Hsinchu 300, Taiwan
- Sustainable Chemical Science and Technology, Taiwan International Graduate Program, National Yang Ming Chiao Tung University and Academia Sinica, Taipei 115, Taiwan
| | - Pei-Wen Hsiao
- Agricultural Biotechnology Research Center, Academia Sinica, Taipei 115, Taiwan
| | - Ming-Hong Tai
- Doctoral Degree Program in Marine Biotechnology, National Sun Yat-Sen University, Kaohsiung 804, Taiwan
- Center for Neuroscience, National Sun Yat-Sen University, Kaohsiung 804, Taiwan
- Institute of Biomedical Sciences, National Sun Yat-Sen University, Kaohsiung 804, Taiwan
| | - Wen-Shan Li
- Institute of Chemistry, Academia Sinica, Taipei 115, Taiwan (C.-W.F.); (S.J.L.P.P.)
- Biomedical Translation Research Center, Academia Sinica, National Biotechnology Research Park, Taipei 115, Taiwan
- Sustainable Chemical Science and Technology, Taiwan International Graduate Program, National Yang Ming Chiao Tung University and Academia Sinica, Taipei 115, Taiwan
- Doctoral Degree Program in Marine Biotechnology, National Sun Yat-Sen University, Kaohsiung 804, Taiwan
- Ph.D. Program in Drug Discovery and Development Industry, College of Pharmacy, Taipei Medical University, Taipei 110, Taiwan
- Department of Medicinal and Applied Chemistry, College of Life Science, Kaohsiung Medical University, Kaohsiung 807, Taiwan
| |
Collapse
|
8
|
Sanz-Ortega L, Andersson A, Carlsten M. Harnessing upregulated E-selectin while enhancing SDF-1α sensing redirects infused NK cells to the AML-perturbed bone marrow. Leukemia 2024; 38:579-589. [PMID: 38182818 PMCID: PMC10912028 DOI: 10.1038/s41375-023-02126-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2023] [Revised: 12/11/2023] [Accepted: 12/14/2023] [Indexed: 01/07/2024]
Abstract
Increased bone marrow (BM) homing of NK cells is associated with positive outcome in patients with acute myeloid leukemia (AML) treated within adoptive NK cell transfer trials. While most efforts to further improve the efficacy focus on augmenting NK cell persistence and cytotoxicity, few address their ability to home to the tumor. Here, we decipher how AML growth alters the BM niche to impair NK cell infiltration and how insights can be utilized to resolve this issue. We show that AML development gradually impairs the BM homing capacity of infused NK cells, which was tightly linked to loss of SDF-1α in this environment. AML development also triggered up-regulation of E-selectin on BM endothelial cells. Given the poor E-selectin-binding capacity of NK cells, introduction of fucosyltransferase-7 (FUT7) to the NK cells per mRNA transfection resulted in potent E-selectin binding and stronger adhesion to E-selectin+ endothelial cells. Co-introduction of FUT7 and gain-of-function CXCR4 (CXCR4R334X) redirected NK cell homing to the BM of AML-bearing mice nearly to the levels in AML-free mice. This work shows how impaired NK cell homing caused by AML-induced microenvironmental changes can be overcome by genetic engineering. We speculate our insights can help further advance future NK cell immunotherapies.
Collapse
Affiliation(s)
- Laura Sanz-Ortega
- Center for Hematology and Regenerative Medicine, Department of Medicine, Huddinge, Karolinska Institutet, Stockholm, Sweden
| | - Agneta Andersson
- Center for Hematology and Regenerative Medicine, Department of Medicine, Huddinge, Karolinska Institutet, Stockholm, Sweden
| | - Mattias Carlsten
- Center for Hematology and Regenerative Medicine, Department of Medicine, Huddinge, Karolinska Institutet, Stockholm, Sweden.
- Center for Cell Therapy and Allogeneic Stem Cell Transplantation, Karolinska Comprehensive Cancer Center, Karolinska University Hospital, Stockholm, Sweden.
| |
Collapse
|
9
|
Li M, Ma Z, Zhang Y, Feng H, Li Y, Sang W, Zhu R, Huang R, Yan J. Integrative analysis of the ST6GALNAC family identifies GATA2-upregulated ST6GALNAC5 as an adverse prognostic biomarker promoting prostate cancer cell invasion. Cancer Cell Int 2023; 23:141. [PMID: 37468844 DOI: 10.1186/s12935-023-02983-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2023] [Accepted: 06/29/2023] [Indexed: 07/21/2023] Open
Abstract
BACKGROUND ST6GALNAC family members function as sialyltransferases and have been implicated in cancer progression. However, their aberrant expression levels, prognostic values and specific roles in metastatic prostate cancer (PCa) remain largely unclear. METHODS Two independent public datasets (TCGA-PRAD and GSE21032), containing 648 PCa samples in total, were employed to comprehensively examine the mRNA expression changes of ST6GALNAC family members in PCa, as well as their associations with clinicopathological parameters and prognosis. The dysregulation of ST6GALNAC5 was further validated in a mouse PCa model and human PCa samples from our cohort (n = 64) by immunohistochemistry (IHC). Gene Set Enrichment Analysis, Gene Ontology, Kyoto Encyclopedia of Genes and Genomes and drug sensitivity analyses were performed to enrich the biological processes most related to ST6GALNAC5. Sulforhodamine B, transwell, luciferase reporter and chromatin immunoprecipitation (ChIP) assays were used to examine the PCa cell proliferation, invasion and transcriptional regulation, respectively. RESULTS Systematical investigation of six ST6GALNAC family members in public datasets revealed that ST6GALNAC5 was the only gene consistently and significantly upregulated in metastatic PCa, and ST6GALNAC5 overexpression was also positively associated with Gleason score and predicted poor prognosis in PCa patients. IHC results showed that (1) ST6GALNAC5 protein expression was increased in prostatic intraepithelial neoplasia and further elevated in PCa from a PbCre;PtenF/F mouse model; (2) overexpressed ST6GALNAC5 protein was confirmed in human PCa samples comparing with benign prostatic hyperplasia samples from our cohort (p < 0.001); (3) ST6GALNAC5 overexpression was significantly correlated with perineural invasion of PCa. Moreover, we first found transcription factor GATA2 positively and directly regulated ST6GALNAC5 expression at transcriptional level. ST6GALNAC5 overexpression could partially reverse GATA2-depletion-induced inhibition of PCa cell invasion. The GATA2-ST6GALNAC5 signature exhibited better prediction on the poor prognosis in PCa patients than GATA2 or ST6GALNAC5 alone. CONCLUSIONS Our results indicated that GATA2-upregulated ST6GALNAC5 might serve as an adverse prognostic biomarker promoting prostate cancer cell invasion.
Collapse
Affiliation(s)
- Meiqian Li
- Model Animal Research Center, Medical School of Nanjing University, Nanjing University, Nanjing, China
| | - Zhihui Ma
- Center for Drug Safety Evaluation and Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Yuqing Zhang
- Department of Urology, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai, China
| | - Hanyi Feng
- School of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing, China
| | - Yang Li
- Department of Urology, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai, China
| | - Weicong Sang
- Department of Urology, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai, China
| | - Rujian Zhu
- Department of Urology, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai, China.
| | - Ruimin Huang
- Center for Drug Safety Evaluation and Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China.
- University of Chinese Academy of Sciences, Beijing, China.
- School of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing, China.
| | - Jun Yan
- Department of Laboratory Animal Science, Fudan University, Shanghai, China.
- Department of Urology, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai, China.
- Model Animal Research Center, Nanjing University, Nanjing, China.
| |
Collapse
|
10
|
Yousefi H, Bahramy A, Zafari N, Delavar MR, Nguyen K, Haghi A, Kandelouei T, Vittori C, Jazireian P, Maleki S, Imani D, Moshksar A, Bitaraf A, Babashah S. Notch signaling pathway: a comprehensive prognostic and gene expression profile analysis in breast cancer. BMC Cancer 2022; 22:1282. [PMID: 36476410 PMCID: PMC9730604 DOI: 10.1186/s12885-022-10383-z] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2022] [Accepted: 11/29/2022] [Indexed: 12/12/2022] Open
Abstract
Breast cancer is a complex disease exhibiting a great degree of heterogeneity due to different molecular subtypes. Notch signaling regulates the differentiation of breast epithelial cells during normal development and plays a crucial role in breast cancer progression through the abnormal expression of the Notch up-and down-stream effectors. To date, there are only a few patient-centered clinical studies using datasets characterizing the role of Notch signaling pathway regulators in breast cancer; thus, we investigate the role and functionality of these factors in different subtypes using publicly available databases containing records from large studies. High-throughput genomic data and clinical information extracted from TCGA were analyzed. We performed Kaplan-Meier survival and differential gene expression analyses using the HALLMARK_NOTCH_SIGNALING gene set. To determine if epigenetic regulation of the Notch regulators contributes to their expression, we analyzed methylation levels of these factors using the TCGA HumanMethylation450 Array data. Notch receptors and ligands expression is generally associated with the tumor subtype, grade, and stage. Furthermore, we showed gene expression levels of most Notch factors were associated with DNA methylation rate. Modulating the expression levels of Notch receptors and effectors can be a potential therapeutic approach for breast cancer. As we outline herein, elucidating the novel prognostic and regulatory roles of Notch implicate this pathway as an essential mediator controlling breast cancer progression.
Collapse
Affiliation(s)
- Hassan Yousefi
- Biochemistry & Molecular Biology, Louisiana State University Health Science Center (LSUHSC), New Orleans, LA, USA
| | - Afshin Bahramy
- Department of Medical Genetics, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Narges Zafari
- Department of Medical Genetics, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Mahsa Rostamian Delavar
- Department of Cell and Molecular Biology and Microbiology, Faculty of Biological Science and Technology, University of Isfahan, Isfahan, Iran
| | - Khoa Nguyen
- Department of Medicine, Tulane University School of Medicine, New Orleans, LA, USA
| | - Atousa Haghi
- Hematology Oncology and Stem Cell Transplantation Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Tahmineh Kandelouei
- Huntsman Cancer Institute, University of Utah School of Medicine, Salt Lake City, Utah, USA
| | - Cecilia Vittori
- Louisiana State University Health Sciences Center (LSUHSC), and Stanley S. Scott Cancer Center, New Orleans, LA, USA
| | - Parham Jazireian
- Department of Biology, University Campus 2, University of Guilan, Rasht, Iran
| | - Sajad Maleki
- Department of Biochemistry, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran
| | - Danyal Imani
- Department of Immunology, School of Public Health, Tehran University of Medical Sciences (TUMS), Tehran, Iran
| | - Amin Moshksar
- Interventional Radiology, University of Texas Medical Branch (UTMB), Galveston, TX, USA
| | - Amirreza Bitaraf
- Department of Molecular Genetics, Faculty of Biological Sciences, Tarbiat Modares University, P.O. Box, Tehran, 14115-154, Iran
| | - Sadegh Babashah
- Department of Molecular Genetics, Faculty of Biological Sciences, Tarbiat Modares University, P.O. Box, Tehran, 14115-154, Iran.
| |
Collapse
|
11
|
Huang J, Huang J, Zhang G. Insights into the Role of Sialylation in Cancer Metastasis, Immunity, and Therapeutic Opportunity. Cancers (Basel) 2022; 14:5840. [PMID: 36497322 PMCID: PMC9737300 DOI: 10.3390/cancers14235840] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2022] [Revised: 11/24/2022] [Accepted: 11/24/2022] [Indexed: 11/29/2022] Open
Abstract
Sialylation is an enzymatic process that covalently attaches sialic acids to glycoproteins and glycolipids and terminates them by creating sialic acid-containing glycans (sialoglycans). Sialoglycans, usually located in the outmost layers of cells, play crucial biological roles, notably in tumor transformation, growth, metastasis, and immune evasion. Thus, a deeper comprehension of sialylation in cancer will help to facilitate the development of innovative cancer therapies. Cancer sialylation-related articles have consistently increased over the last four years. The primary subjects of these studies are sialylation, cancer, immunotherapy, and metastasis. Tumor cells activate endothelial cells and metastasize to distant organs in part by the interactions of abnormally sialylated integrins with selectins. Furthermore, cancer sialylation masks tumor antigenic epitopes and induces an immunosuppressive environment, allowing cancer cells to escape immune monitoring. Cytotoxic T lymphocytes develop different recognition epitopes for glycosylated and nonglycosylated peptides. Therefore, targeting tumor-derived sialoglycans is a promising approach to cancer treatments for limiting the dissemination of tumor cells, revealing immunogenic tumor antigens, and boosting anti-cancer immunity. Exploring the exact tumor sialoglycans may facilitate the identification of new glycan targets, paving the way for the development of customized cancer treatments.
Collapse
Affiliation(s)
- Jianmei Huang
- School of Medicine, University of Electronic Science and Technology of China, Chengdu 610054, China
| | - Jianming Huang
- Biochemistry and Molecular Biology, Sichuan Cancer Institute, Chengdu 610041, China
| | - Guonan Zhang
- School of Medicine, University of Electronic Science and Technology of China, Chengdu 610054, China
- Department of Gynecologic Oncology, Sichuan Cancer Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu 610041, China
| |
Collapse
|
12
|
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
|
13
|
Li J, Long Y, Sun J, Wu J, He X, Wang S, Wang X, Miao X, Huang R, Yan J. Comprehensive landscape of the ST3GAL family reveals the significance of ST3GAL6-AS1/ST3GAL6 axis on EGFR signaling in lung adenocarcinoma cell invasion. Front Cell Dev Biol 2022; 10:931132. [PMID: 36092699 PMCID: PMC9462654 DOI: 10.3389/fcell.2022.931132] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2022] [Accepted: 07/20/2022] [Indexed: 11/20/2022] Open
Abstract
Sialylation aberration has been implicated in lung cancer development by altering signaling pathways. Hence, it is urgent to identify key sialyltransferases in the development of lung adenocarcinoma (LUAD), which is a common malignant subtype of non-small cell lung cancer. Herein, by systematically investigating the expression levels of ST3GAL family members in several public databases, we consistently found the frequent downregulation of ST3GAL6 in LUAD samples. Its downregulation is significantly negatively associated with stage, and significantly reduced in proximal-proliferative molecular subtype and predicts poor clinical outcomes. By protein–protein interaction network analysis and validation, we found that ST3GAL6 deficiency promotes LUAD cell invasiveness with the activated EGFR/MAPK signaling, accompanied by the elevated expression levels of matrix metalloproteinases 2 and 9, which can be partially reversed by EGFR inhibitor, gefitinib. Additionally, the ST3GAL6 level was positively regulated by ST3GAL6-AS1, an antisense long non-coding RNA to its host gene. The downregulation of ST3GAL6-AS1 also heralds a worse prognosis in LUAD patients and promotes LUAD cell invasiveness, recapitulating the function of its host gene, ST3GAL6. Altogether, ST3GAL6-AS1-regulated ST3GAL6 is a frequently downregulated sialyltransferase in LUAD patients and negatively regulates EGFR signaling, which can serve as a promising independent prognostic marker in LUAD patients.
Collapse
Affiliation(s)
- Jiaxuan Li
- Department of Respiratory Medicine, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Yiming Long
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Jingya Sun
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
| | - Jiajun Wu
- Department of Laboratory Animal Science, Fudan University, Shanghai, China
| | - Xiao He
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
- School of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing, China
| | - Simei Wang
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Xiongbiao Wang
- Department of Respiratory Medicine, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Interventional Cancer Institute of Chinese Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Xiayi Miao
- Department of Respiratory Medicine, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- *Correspondence: Xiayi Miao, ; Ruimin Huang, ; Jun Yan,
| | - Ruimin Huang
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
- University of Chinese Academy of Sciences, Beijing, China
- School of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing, China
- *Correspondence: Xiayi Miao, ; Ruimin Huang, ; Jun Yan,
| | - Jun Yan
- Department of Respiratory Medicine, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Department of Laboratory Animal Science, Fudan University, Shanghai, China
- *Correspondence: Xiayi Miao, ; Ruimin Huang, ; Jun Yan,
| |
Collapse
|
14
|
Volkhina IV, Butolin EG. Clinical and Diagnostic Significance of Sialic Acids Determination in Biological Material. BIOCHEMISTRY (MOSCOW) SUPPLEMENT. SERIES B, BIOMEDICAL CHEMISTRY 2022; 16:165-174. [PMID: 35990315 PMCID: PMC9377294 DOI: 10.1134/s199075082203012x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/17/2021] [Revised: 02/14/2022] [Accepted: 02/16/2022] [Indexed: 06/15/2023]
Abstract
Sialic acids (SA) are neuraminic acid derivatives, located at the terminal position in the chains of monosaccharide residues of various glycoconjugates. SA play a dual role: they either mask recognition sites, or, on the contrary, represent biological targets that can be recognized by receptor proteins and serve as ligands. The desialylation/sialylation processes can be considered as a dynamic modification regulated by sialyltransferases and sialidases in response to external or internal stimuli. This review describes the structural and functional diversity and the potential use of SA fractions as biomarkers for various pathological conditions. Almost any extreme impact on the body and inflammatory processes are accompanied by an increase in the level of both total and free SA in the blood and tissues. Possible reasons for the increase of sialoglycoconjugate metabolism indicators in biological material include: (i) activation of the hepatocyte synthesis and secretion of various acute-phase proteins, many of which are sialoglycoproteins, (ii) impaired membrane integrity and destruction of body cells, (iii) high activity of sialidases (neurominidases) and sialyltransferases. Most acute and chronic liver diseases are characterized by the decrease in the total level of SA in the blood serum (because many plasma proteins are synthesized and glycosylated in hepatocytes). Aberrant sialylation results in changes of sialoglycoconjugate structure, its ability to perform biological functions and sialoglycoconjugate half-life. Glycosylation is the most common post-translational modification of proteins in the virus, which not only promotes the formation of specific conformation of viral proteins, but also modulates their interaction with receptors and affects host cell recognition, viral replication and infectivity. Serum total SA concentration increases in some benign and inflammatory conditions, which indicates a lack of specificity and limits their use for early detection and screening of neoplastic diseases. Clinical and diagnostic value of determining the sialoglycoconjugate metabolic indicators, including changes in the content of both SA fractions and specific proteins in various biological fluids and tissues, consists in establishing the causes and mechanisms of biochemical changes in the body in certain diseases. In combination with the measurement of existing markers, they can be used to improve diagnosis, staging and monitoring of therapeutic response in some pathological conditions where the need for specificity is less than for specific diagnostics.
Collapse
Affiliation(s)
- I. V. Volkhina
- Saint Petersburg State Pediatric Medical University, ul. Litovskaya 2, 194100 St.Petersburg, Russia
| | - E. G. Butolin
- Izhevsk State Medical Academy, ul. Kommunarov 201, 426034 Izhevsk, Russia
| |
Collapse
|
15
|
Doostkam A, Malekmakan L, Hosseinpour A, Janfeshan S, Roozbeh J, Masjedi F. Sialic acid: an attractive biomarker with promising biomedical applications. ASIAN BIOMED 2022; 16:153-167. [PMID: 37551166 PMCID: PMC10321195 DOI: 10.2478/abm-2022-0020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
This broad, narrative review highlights the roles of sialic acids as acidic sugars found on cellular membranes. The role of sialic acids in cellular communication and development has been well established. Recently, attention has turned to the fundamental role of sialic acids in many diseases, including viral infections, cardiovascular diseases, neurological disorders, diabetic nephropathy, and malignancies. Sialic acid may be a target for developing new drugs to treat various cancers and inflammatory processes. We recommend the routine measurement of serum sialic acid as a sensitive inflammatory marker in various diseases.
Collapse
Affiliation(s)
- Aida Doostkam
- Shiraz Nephro-Urology Research Center, Shiraz University of Medical Sciences, Shiraz7193635899, Iran
| | - Leila Malekmakan
- Shiraz Nephro-Urology Research Center, Shiraz University of Medical Sciences, Shiraz7193635899, Iran
| | - Alireza Hosseinpour
- Student Research Committee, Shiraz University of Medical Sciences, Shiraz7134853185, Iran
| | - Sahar Janfeshan
- Shiraz Nephro-Urology Research Center, Shiraz University of Medical Sciences, Shiraz7193635899, Iran
| | - Jamshid Roozbeh
- Shiraz Nephro-Urology Research Center, Shiraz University of Medical Sciences, Shiraz7193635899, Iran
| | - Fatemeh Masjedi
- Shiraz Nephro-Urology Research Center, Shiraz University of Medical Sciences, Shiraz7193635899, Iran
| |
Collapse
|
16
|
Ma Z, Yang H, Kessler M, Sperandio M, Mahner S, Jeschke U, von Schönfeldt V. Targeting Aberrantly Elevated Sialyl Lewis A as a Potential Therapy for Impaired Endometrial Selection Ability in Unexplained Recurrent Miscarriage. Front Immunol 2022; 13:919193. [PMID: 35837404 PMCID: PMC9273867 DOI: 10.3389/fimmu.2022.919193] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2022] [Accepted: 06/02/2022] [Indexed: 11/23/2022] Open
Abstract
Background Carbohydrate Lewis antigens including sialyl Lewis A (sLeA), sialyl Lewis X (sLeX), Lewis X (LeX), and Lewis Y (LeY) are the commonest cell surface glycoconjugates that play pivotal roles in multiple biological processes, including cell adhesion and cell communication events during embryogenesis. SLeX, LeY, and associated glycosyltransferases ST3GAL3 and FUT4 have been reported to be involved in human embryo implantation. While the expression pattern of Lewis antigens in the decidua of unexplained recurrent miscarriage (uRM) patients remains unclear. Methods Paraffin-embedded placental tissue slides collected from patients experiencing early miscarriages (6–12 weeks) were analyzed using immunohistochemical (IHC) and immunofluorescent (IF) staining. An in vitro assay was developed using endometrial cell line RL95-2 and trophoblast cell line HTR-8/SVneo. Modulatory effect of potential glycosyltransferase on Lewis antigens expression was investigated by target-specific small interfering RNA (siRNA) knockdown in RL95-2 cells. HTR-8/SVneo cells spheroids adhesion assay was applied to investigate the intrinsic role of Lewis antigens in the abnormal implantation process of uRM. The expression of Lewis antigens in RL95-2 cells in response to the treatment with pro-implantation cytokine IL-1β was further measured by flow cytometry and immunocytochemical (ICC) staining. Results IHC staining revealed that Lewis antigens are mainly expressed in the luminal and glandular epithelium, IF staining further indicated the cellular localization at the apical membrane of the epithelial cells. FUTs, ST3GALs, and NEU1 located in both stromal and epithelial cells. We have found that the expression of sLeA, LeX, FUT3/4, and ST3GAL3/4 are significantly upregulated in the RM group, while FUT1 is downregulated. SLeX, LeY, ST3GAL6, and NEU1 showed no significant differences between groups. FUT3 knockdown in RL95-2 cells significantly decreased the expression of sLeA and the spheroids adhesion to endometrial monolayer. Anti-sLeA antibody can remarkably suppress both the basal and IL-1β induced adhesion of HTR-8/SVneo spheroids to RL95-2 cells monolayer. While further flow cytometry and ICC detection indicated that the treatment of RL95-2 cells with IL-1β significantly increases the surface expression of LeX, but not sLeA. Conclusions SLeA, LeX, and pertinent glycosyltransferase genes FUT1/3/4 and ST3GAL3/4 are notably dysregulated in the decidua of uRM patients. FUT3 accounts for the synthesis of sLeA in RL95-2 cells and affects the endometrial receptivity. Targeting aberrantly elevated sLeA may be a potential therapy for the inappropriate implantation in uRM.
Collapse
Affiliation(s)
- Zhi Ma
- Department of Obstetrics and Gynaecology, University Hospital, Ludwig-Maximilians-Universität Munich, Munich, Germany
| | - Huixia Yang
- Department of Obstetrics and Gynaecology, University Hospital, Ludwig-Maximilians-Universität Munich, Munich, Germany
| | - Mirjana Kessler
- Department of Obstetrics and Gynaecology, University Hospital, Ludwig-Maximilians-Universität Munich, Munich, Germany
| | - Markus Sperandio
- Biomedical Center (BMC), Institute for Cardiovascular Physiology and Pathophysiology, Walter Brendel Center for Experimental Medicine (WBex), Faculty of Medicine, Ludwig-Maximilians-Universität Munich, Munich, Germany
| | - Sven Mahner
- Department of Obstetrics and Gynaecology, University Hospital, Ludwig-Maximilians-Universität Munich, Munich, Germany
| | - Udo Jeschke
- Department of Obstetrics and Gynaecology, University Hospital, Ludwig-Maximilians-Universität Munich, Munich, Germany
- Department of Obstetrics and Gynaecology, University Hospital Augsburg, Augsburg, Germany
- *Correspondence: Udo Jeschke,
| | - Viktoria von Schönfeldt
- Department of Obstetrics and Gynaecology, University Hospital, Ludwig-Maximilians-Universität Munich, Munich, Germany
| |
Collapse
|
17
|
Targeting hypersialylation in multiple myeloma represents a novel approach to enhance NK cell-mediated tumor responses. Blood Adv 2022; 6:3352-3366. [PMID: 35294519 PMCID: PMC9198929 DOI: 10.1182/bloodadvances.2021006805] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2021] [Accepted: 03/07/2022] [Indexed: 11/20/2022] Open
Abstract
Hypersialylation in MM facilitates immune evasion of NK cells but can be overcome by targeted desialylation or genetic deletion of Siglec-7. Desialylation unmasks CD38 expression on MM cells, enhancing NK cell–mediated ADCC induced by CD38 targeting of monoclonal antibodies.
Abnormal glycosylation is a hallmark of cancer, and the hypersialylated tumor cell surface facilitates abnormal cell trafficking and drug resistance in several malignancies, including multiple myeloma (MM). Furthermore, hypersialylation has also been implicated in facilitating evasion of natural killer (NK) cell–mediated immunosurveillance but not in MM to date. In this study, we explore the role of hypersialylation in promoting escape from NK cells. We document strong expression of sialic acid-derived ligands for Siglec-7 (Siglec-7L) on primary MM cells and MM cell lines, highlighting the possibility of Siglec-7/Siglec-7L interactions in the tumor microenvironment. Interactomics experiments in MM cell lysates revealed PSGL-1 as the predominant Siglec-7L in MM. We show that desialylation, using both a sialidase and sialyltransferase inhibitor (SIA), strongly enhances NK cell–mediated cytotoxicity against MM cells. Furthermore, MM cell desialylation results in increased detection of CD38, a well-validated target in MM. Desialylation enhanced NK cell cytotoxicity against CD38+ MM cells after treatment with the anti-CD38 monoclonal antibody daratumumab. Additionally, we show that MM cells with low CD38 expression can be treated with all trans-retinoic acid (ATRA), SIA and daratumumab to elicit a potent NK cell cytotoxic response. Finally, we demonstrate that Siglec-7KO potentiates NK cell cytotoxicity against Siglec-7L+ MM cells. Taken together, our work shows that desialylation of MM cells is a promising novel approach to enhance NK cell efficacy against MM, which can be combined with frontline therapies to elicit a potent anti-MM response.
Collapse
|
18
|
Volkhina IV, Butolin EG. [Clinical and diagnostic significance of sialic acids determination in biological material]. BIOMEDITSINSKAIA KHIMIIA 2022; 68:7-17. [PMID: 35221292 DOI: 10.18097/pbmc20226801007] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Sialic acids (SA) are derivatives of neuraminic acid; they are located at the terminal position in the chains of monosaccharide residues of various glycoconjugates. SA play a dual role, they either mask recognition sites, or, on the contrary, represent biological targets that can be recognized by receptor proteins and serve as ligands. The desialylation/sialylation processes can be viewed as a dynamic modification regulated by sialyltransferases and sialidases in response to external or internal stimuli. This review describes the structural and functional diversity and the potential use of SA fractions as biomarkers for various pathological conditions. Almost any extreme effects on the body and inflammatory processes lead to an increase in the level of both total and free SA in the blood and tissues. Possible reasons for the increase of sialoglycoconjugate metabolism indicators in biological material include activation of the hepatocyte synthesis and secretion of various acute-phase proteins, many of which are sialoglycoproteins, violation of the membrane integrity and destruction of body cells, and also high activity of sialidases (neurominidases) and sialyltransferases. Most acute and chronic liver diseases are characterized by the decrease in the total level of SA in the blood serum (because many plasma proteins are synthesized and glycosylated in hepatocytes). Aberrant sialylation results in changes of sialoglycoconjugate structure, its ability to perform biological functions and half-life. Glycosylation is the most common post-translational modification of proteins in the virus, which not only promotes the formation of specific conformation of viral proteins, but also modulates their interaction with receptors and affects host cell recognition, viral replication and infectivity. Serum total SA concentration increases in some benign and inflammatory conditions, which indicates a lack of specificity and limits their use for early detection and screening of neoplastic diseases. Nevertheless, determining blood SA level and measuring concentration of existing biomarkers can be used to improve diagnostic indicators, to stage and monitor therapeutic response in some types of cancer, when the need for specificity is less than for diagnosis. Clinical and diagnostic value of determining the sialoglycoconjugate metabolic indicators, including changes in the content of both SA fractions and specific proteins in various biological fluids and tissues, lies in establishing the causes and mechanisms of biochemical changes in the body in certain diseases.
Collapse
Affiliation(s)
- I V Volkhina
- Saint Petersburg State Pediatric Medical University, Saint Petersburg, Russia
| | - E G Butolin
- Izhevsk State Medical Academy, Izhevsk, Russia
| |
Collapse
|
19
|
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
|
20
|
Gao Z, Xu M, Yue S, Shan H, Xia J, Jiang J, Yang S. Abnormal sialylation and fucosylation of saliva glycoproteins: Characteristics of lung cancer-specific biomarkers. CURRENT RESEARCH IN PHARMACOLOGY AND DRUG DISCOVERY 2021; 3:100079. [PMID: 35005612 PMCID: PMC8718573 DOI: 10.1016/j.crphar.2021.100079] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2021] [Revised: 10/31/2021] [Accepted: 12/15/2021] [Indexed: 12/23/2022] Open
Abstract
Dysregulated surface glycoproteins play an important role in tumor cell proliferation and progression. Abnormal glycosylation of these glycoproteins may activate tumor signal transduction and lead to tumor development. The tumor microenvironment alters its molecular composition, some of which regulate protein glycosylation biosynthesis. The glycosylation of saliva proteins in lung cancer patients is different from healthy controls, in which the glycans of cancer patients are highly sialylated and hyperfucosylated. Most studies have shown that O-glycans from cancer are truncated O-glycans, while N-glycans contain fucoses and sialic acids. Because glycosylation analysis is challenging, there are few reports on how glycosylation of saliva proteins is related to the occurrence or progression of lung cancer. In this review, we discussed glycoenzymes involved in protein glycosylation, their changes in tumor microenvironment, potential tumor biomarkers present in body fluids, and abnormal glycosylation of saliva or lung glycoproteins. We further explored the effect of glycosylation changes on tumor signal transduction, and emphasized the role of receptor tyrosine kinases in tumorigenesis and metastasis.
Collapse
Affiliation(s)
- Ziyuan Gao
- Center for Clinical Mass Spectrometry, School of Pharmaceutical Sciences, Soochow University, Jiangsu, 215123, China
- Department of Respiratory and Critical Care Medicine, Dushu Lake Hospital to Soochow University, Suzhou, Jiangsu, 215125, China
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Soochow University; Suzhou Jiangsu, 215006, China
| | - Mingming Xu
- Center for Clinical Mass Spectrometry, School of Pharmaceutical Sciences, Soochow University, Jiangsu, 215123, China
| | - Shuang Yue
- Center for Clinical Mass Spectrometry, School of Pharmaceutical Sciences, Soochow University, Jiangsu, 215123, China
| | - Huang Shan
- Center for Clinical Mass Spectrometry, School of Pharmaceutical Sciences, Soochow University, Jiangsu, 215123, China
| | - Jun Xia
- Department of Clinical Laboratory Center, Zhejiang Provincial People's Hospital, People's Hospital of Hangzhou Medical College, Hangzhou, Zhejiang, 310014, China
| | - Junhong Jiang
- Department of Respiratory and Critical Care Medicine, Dushu Lake Hospital to Soochow University, Suzhou, Jiangsu, 215125, China
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Soochow University; Suzhou Jiangsu, 215006, China
| | - Shuang Yang
- Center for Clinical Mass Spectrometry, School of Pharmaceutical Sciences, Soochow University, Jiangsu, 215123, China
| |
Collapse
|
21
|
Hugonnet M, Singh P, Haas Q, von Gunten S. The Distinct Roles of Sialyltransferases in Cancer Biology and Onco-Immunology. Front Immunol 2021; 12:799861. [PMID: 34975914 PMCID: PMC8718907 DOI: 10.3389/fimmu.2021.799861] [Citation(s) in RCA: 60] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2021] [Accepted: 12/02/2021] [Indexed: 12/24/2022] Open
Abstract
Aberrant glycosylation is a key feature of malignant transformation. Hypersialylation, the enhanced expression of sialic acid-terminated glycoconjugates on the cell surface, has been linked to immune evasion and metastatic spread, eventually by interaction with sialoglycan-binding lectins, including Siglecs and selectins. The biosynthesis of tumor-associated sialoglycans involves sialyltransferases, which are differentially expressed in cancer cells. In this review article, we provide an overview of the twenty human sialyltransferases and their roles in cancer biology and immunity. A better understanding of the individual contribution of select sialyltransferases to the tumor sialome may lead to more personalized strategies for the treatment of cancer.
Collapse
Affiliation(s)
- Marjolaine Hugonnet
- Institute of Pharmacology, University of Bern, Bern, Switzerland
- Bern Center for Precision Medicine (BCPM), University of Bern, Bern, Switzerland
| | - Pushpita Singh
- Institute of Pharmacology, University of Bern, Bern, Switzerland
| | - Quentin Haas
- Institute of Pharmacology, University of Bern, Bern, Switzerland
| | - Stephan von Gunten
- Institute of Pharmacology, University of Bern, Bern, Switzerland
- Bern Center for Precision Medicine (BCPM), University of Bern, Bern, Switzerland
| |
Collapse
|
22
|
Jin Y, Liang Y, Su Y, Hui L, Liu H, Ding L, Zhou F. Identification of novel combined biomarkers in the diagnosis of multiple myeloma. Hematology 2021; 26:964-969. [PMID: 34871540 DOI: 10.1080/16078454.2021.2003065] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022] Open
Abstract
PURPOSE Multiple myeloma (MM) is a haematological malignant disease with a clonal proliferation of plasma cells, and timely surveillance is helpful to improve the survival rate of patients with MM. However, there is a lack of simple and effective biomarkers for the diagnosis, prognosis, and residual disease evaluation of MM. MATERIAL & METHODS In the detection cohort, we used the samples from six newly diagnosed MM patients and six control subjects. Plasma proteins were labelled with dimethyl reagents and enriched by lectin AANL6, then the deglycosylated peptides were identified by LC-MS/MS. Differentially expressed proteins were used for further exploration. In the validation cohort, we used 90 newly diagnosed patients with MM and 70 cases of unrelated diseases as controls. The diagnosis performance was analysed by ROC analysis using SPSS. RESULTS In this study, we show, using lectin blots with AANL6, that glycosylation levels were higher in MM patients than in controls. After AANL6 enrichment, we detected 58 differentially expressed proteins using quantitative proteomics. We further validated one candidate Fibulin-1 (FBLN1). Using an Elisa assay, we showed that FBLN1 expression was increased in plasma of 90 cases of MM, and which was significantly correlated with DKK1 expression. ROC analysis showed that these two markers had a 95.7% specificity for determining the diagnosis of MM. CONCLUSION These data suggest that the MM cases display increased glycosylation after AANL6 enrichment and that the combined expression of FBLN1 and DKK1 can be used as an effective diagnostic biomarker.
Collapse
Affiliation(s)
- Yanxia Jin
- Department of Haematology, Zhongnan Hospital of Wuhan University, Wuhan, People's Republic of China.,Hubei Key Laboratory of Edible Wild Plants Conservation and Utilization, Hubei Normal University, Huangshi, People's Republic of China
| | - Yuxing Liang
- Department of Haematology, Zhongnan Hospital of Wuhan University, Wuhan, People's Republic of China
| | - Yanting Su
- College of Life Sciences, Wuhan University, Wuhan, People's Republic of China
| | - Lingyun Hui
- Department of Laboratory Medicine, First Affiliated Hospital, Medical School of Xi'an Jiaotong University, Xi'an, People's Republic of China
| | - Hailing Liu
- Department of Clinical Haematology, Second Affiliated Hospital, Medical School of Xi'an Jiaotong University, Xi'an, People's Republic of China
| | - Lu Ding
- Department of Haematology, Zhongnan Hospital of Wuhan University, Wuhan, People's Republic of China
| | - Fuling Zhou
- Department of Haematology, Zhongnan Hospital of Wuhan University, Wuhan, People's Republic of China
| |
Collapse
|
23
|
Glycation Interferes with the Expression of Sialyltransferases in Meningiomas. Cells 2021; 10:cells10123298. [PMID: 34943806 PMCID: PMC8699175 DOI: 10.3390/cells10123298] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2021] [Revised: 11/19/2021] [Accepted: 11/22/2021] [Indexed: 11/16/2022] Open
Abstract
Meningiomas are the most common non-malignant intracranial tumors and prefer, like most tumors, anaerobic glycolysis for energy production (Warburg effect). This anaerobic glycolysis leads to an increased synthesis of the metabolite methylglyoxal (MGO) or glyoxal (GO), which is known to react with amino groups of proteins. This reaction is called glycation, thereby building advanced glycation end products (AGEs). In this study, we investigated the influence of glycation on sialylation in two meningioma cell lines, representing the WHO grade I (BEN-MEN-1) and the WHO grade III (IOMM-Lee). In the benign meningioma cell line, glycation led to differences in expression of sialyltransferases (ST3GAL1/2/3/5/6, ST6GAL1/2, ST6GALNAC2/6, and ST8SIA1/2), which are known to play a role in tumor progression. We could show that glycation of BEN-MEN-1 cells led to decreased expression of ST3Gal5. This resulted in decreased synthesis of the ganglioside GM3, the product of ST3Gal5. In the malignant meningioma cell line, we observed changes in expression of sialyltransferases (ST3GAL1/2/3, ST6GALNAC5, and ST8SIA1) after glycation, which correlates with less aggressive behavior.
Collapse
|
24
|
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
|
25
|
Toutonji A, Mandava M, Guglietta S, Tomlinson S. Chronic complement dysregulation drives neuroinflammation after traumatic brain injury: a transcriptomic study. Acta Neuropathol Commun 2021; 9:126. [PMID: 34281628 PMCID: PMC8287781 DOI: 10.1186/s40478-021-01226-2] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2021] [Accepted: 07/07/2021] [Indexed: 02/07/2023] Open
Abstract
Activation of the complement system propagates neuroinflammation and brain damage early and chronically after traumatic brain injury (TBI). The complement system is complex and comprises more than 50 components, many of which remain to be characterized in the normal and injured brain. Moreover, complement therapeutic studies have focused on a limited number of histopathological outcomes, which while informative, do not assess the effect of complement inhibition on neuroprotection and inflammation in a comprehensive manner. Using high throughput gene expression technology (NanoString), we simultaneously analyzed complement gene expression profiles with other neuroinflammatory pathway genes at different time points after TBI. We additionally assessed the effects of complement inhibition on neuropathological processes. Analyses of neuroinflammatory genes were performed at days 3, 7, and 28 post injury in male C57BL/6 mice following a controlled cortical impact injury. We also characterized the expression of 59 complement genes at similar time points, and also at 1- and 2-years post injury. Overall, TBI upregulated the expression of markers of astrogliosis, immune cell activation, and cellular stress, and downregulated the expression of neuronal and synaptic markers from day 3 through 28 post injury. Moreover, TBI upregulated gene expression across most complement activation and effector pathways, with an early emphasis on classical pathway genes and with continued upregulation of C2, C3 and C4 expression 2 years post injury. Treatment using the targeted complement inhibitor, CR2-Crry, significantly ameliorated TBI-induced transcriptomic changes at all time points. Nevertheless, some immune and synaptic genes remained dysregulated with CR2-Crry treatment, suggesting adjuvant anti-inflammatory and neurotropic therapy may confer additional neuroprotection. In addition to characterizing complement gene expression in the normal and aging brain, our results demonstrate broad and chronic dysregulation of the complement system after TBI, and strengthen the view that the complement system is an attractive target for TBI therapy.
Collapse
Affiliation(s)
- Amer Toutonji
- Department of Microbiology and Immunology, Medical University of South Carolina, 173 Ashley Avenue, BSB 204, MSC 504, Charleston, SC 29425 USA
| | - Mamatha Mandava
- Department of Microbiology and Immunology, Medical University of South Carolina, 173 Ashley Avenue, BSB 204, MSC 504, Charleston, SC 29425 USA
| | - Silvia Guglietta
- Department of Microbiology and Immunology, Medical University of South Carolina, 173 Ashley Avenue, BSB 204, MSC 504, Charleston, SC 29425 USA
| | - Stephen Tomlinson
- Department of Microbiology and Immunology, Medical University of South Carolina, 173 Ashley Avenue, BSB 204, MSC 504, Charleston, SC 29425 USA
- Ralph Johnson VA Medical Center, Charleston, SC 29401 USA
| |
Collapse
|
26
|
Ip PP, Li Q, Lin WH, Chang CC, Fann CSJ, Chen HY, Liu FT, Lebrilla CB, Yang CC, Liao F. Analysis of site-specific glycan profiles of serum proteins in patients with multiple sclerosis or neuromyelitis optica spectrum disorder - a pilot study. Glycobiology 2021; 31:1230-1238. [PMID: 34132764 DOI: 10.1093/glycob/cwab053] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2020] [Revised: 06/07/2021] [Accepted: 06/08/2021] [Indexed: 11/14/2022] Open
Abstract
Glycosylation is important for biological functions of proteins and greatly affected by diseases. Exploring the glycosylation profile of the protein-specific glycosylation and/or the site-specific glycosylation may help understand disease etiology, differentiate diseases, and ultimately develop therapeutics. Patients with multiple sclerosis (MS) and patients with neuromyelitis optica spectrum disorder (NMOSD) are sometimes difficult to differentiate due to the similarity in their clinical symptoms. The disease-related glycosylation profiles of MS and NMOSD have not yet been well studied. Here, we analyzed site-specific glycan profiles of serum proteins of these patients by using a recently developed mass spectrometry technique. A total of 286 glycopeptides from 49 serum glycoproteins were quantified and compared between healthy controls (n = 6), remitting MS (n = 45) and remitting NMOSD (n = 23) patients. Significant differences in the levels of site-specific N-glycans on inflammation-associated components [IgM, IgG1, IgG2, complement components 8b (CO8B), attractin], central nerve system-damage-related serum proteins [apolipoprotein D (APOD), alpha-1-antitrypsin, plasma kallikrein and ADAMTS-like protein 3] were observed among three study groups. We furthered demonstrated that site-specific N-glycans on APOD on site 98, CO8B on sites 243 and 553 are potential markers to differentiate MS from NMOSD with an area under receiver operating curve value greater than 0.75. All these observations indicate that remitting MS or NMOSD patients possess a unique disease-associated glyco-signature in their serum proteins. We conclude that monitoring one's serum protein glycan profile using this high-throughput analysis may provide an additional diagnostic criterion for differentiating diseases, monitoring disease status and estimating response-to-treatment effect.
Collapse
Affiliation(s)
- Peng Peng Ip
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taipei 115, Taiwan
| | - Qiongyu Li
- Department of Chemistry, University of California Davis, One Shields Avenue, Davis, California 95616, United States
| | - Wei-Han Lin
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taipei 115, Taiwan
| | - Chien-Ching Chang
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taipei 115, Taiwan
| | | | - Huan-Yuan Chen
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taipei 115, Taiwan
| | - Fu-Tong Liu
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taipei 115, Taiwan
| | - Carlito B Lebrilla
- Department of Chemistry, University of California Davis, One Shields Avenue, Davis, California 95616, United States
| | - Chih-Chao Yang
- Department of Neurology, National Taiwan University Hospital, Taipei, Taiwan
| | - Fang Liao
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taipei 115, Taiwan
| |
Collapse
|
27
|
Ma Z, Yang H, Peng L, Kuhn C, Chelariu-Raicu A, Mahner S, Jeschke U, von Schönfeldt V. Expression of the Carbohydrate Lewis Antigen, Sialyl Lewis A, Sialyl Lewis X, Lewis X, and Lewis Y in the Placental Villi of Patients With Unexplained Miscarriages. Front Immunol 2021; 12:679424. [PMID: 34135905 PMCID: PMC8202085 DOI: 10.3389/fimmu.2021.679424] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2021] [Accepted: 05/10/2021] [Indexed: 11/13/2022] Open
Abstract
Background Lewis antigens such as Sialyl Lewis A (sLeA), Sialyl Lewis X (sLeX), Lewis X (LeX), and Lewis Y (LeY) are a class of carbohydrate molecules that are known to mediate adhesion between tumor cells and endothelium by interacting with its selectin ligands. However, their potential role in miscarriage remains enigmatic. This study aims to analyze the expression pattern of sLeA, sLeX, LeX, and LeY in the placental villi tissue of patients with a medical history of unexplained miscarriages. Methods Paraffin-embedded slides originating from placental tissue were collected from patients experiencing a miscarriage early in their pregnancy (6–13 weeks). Tissues collected from spontaneous (n = 20) and recurrent (n = 15) miscarriages were analyzed using immunohistochemical and immunofluorescent staining. Specimens obtained from legally terminated normal pregnancies were considered as control group (n = 18). Assessment of villous vessel density was performed in another cohort (n = 10 each group) of gestation ages-paired placenta tissue. Protein expression was evaluated with Immunoreactive Score (IRS). Statistical analysis was performed by using Graphpad Prism 8. Results Expression of sLeA, sLeX, LeX, and LeY in the syncytiotrophoblast was significantly upregulated in the control group compared with spontaneous and recurrent miscarriage groups. However, no prominent differences between spontaneous and recurrent miscarriage groups were identified. Potential key modulators ST3GAL6 and NEU1 were found to be significantly downregulated in the recurrent miscarriage group and upregulated in the spontaneous group, respectively. Interestingly, LeX and LeY expression was also detected in the endothelial cells of villous vessels in the control group but no significant expression in miscarriage groups. Furthermore, assessment of villous vessel density using CD31 found significantly diminished vessels in all size groups of villi (small villi <200 µm, P = 0.0371; middle villi between 200 and 400 µm, P = 0.0010 and large villi >400 µm, P = 0.0003). Immunofluorescent double staining also indicated the co-localization of LeX/Y and CD31. Conclusions The expression of four mentioned carbohydrate Lewis antigens and their potential modulators, ST3GAL6 and NEU1, in the placenta of patients with miscarriages was significantly different from the normal pregnancy. For the first time, their expression pattern in the placenta was illustrated, which might shed light on a novel understanding of Lewis antigens’ role in the pathogenesis of miscarriages.
Collapse
Affiliation(s)
- Zhi Ma
- Department of Obstetrics and Gynaecology, University Hospital LMU Munich, Munich, Germany
| | - Huixia Yang
- Department of Obstetrics and Gynaecology, University Hospital LMU Munich, Munich, Germany
| | - Lin Peng
- Department of Obstetrics and Gynaecology, University Hospital LMU Munich, Munich, Germany
| | - Christina Kuhn
- Department of Obstetrics and Gynaecology, University Hospital Augsburg, Augsburg, Germany
| | - Anca Chelariu-Raicu
- Department of Obstetrics and Gynaecology, University Hospital LMU Munich, Munich, Germany
| | - Sven Mahner
- Department of Obstetrics and Gynaecology, University Hospital LMU Munich, Munich, Germany
| | - Udo Jeschke
- Department of Obstetrics and Gynaecology, University Hospital LMU Munich, Munich, Germany.,Department of Obstetrics and Gynaecology, University Hospital Augsburg, Augsburg, Germany
| | | |
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
|
Shen Y, Feng Y, Li F, Jia Y, Peng Y, Zhao W, Hu J, He A. lncRNA ST3GAL6‑AS1 promotes invasion by inhibiting hnRNPA2B1‑mediated ST3GAL6 expression in multiple myeloma. Int J Oncol 2021; 58:5. [PMID: 33649796 DOI: 10.3892/ijo.2021.5185] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2020] [Accepted: 12/10/2020] [Indexed: 12/12/2022] Open
Abstract
Multiple myeloma (MM) is an incurable disease caused by the infiltration of malignant plasma B cells into bone marrow, whose pathogenesis remains largely unknown. Long non‑coding RNAs (lncRNAs) have emerged as important factors in pathogenesis. Our previous study validated that lncRNA ST3 β‑galactoside α‑2,3‑sialyltransferase 6 antisense RNA 1 (ST3GAL6‑AS1) was upregulated markedly in MM. Therefore, the aim of the study was to investigate the molecular mechanisms of ST3GAL6‑AS1 in MM cells. ST3GAL6‑AS1 expression levels in MM cells was detected using reverse transcription‑quantitative PCR. ST3GAL6‑AS1 antisense oligonucleotides and small interfering RNAs were transfected into MM cells to downregulate expression. In vitro assays were performed to investigate the functional role of ST3GAL6‑AS1 in MM cells. RNA pull‑down, RNA immunoprecipitation and comprehensive identification of RNA‑binding proteins using mass spectrometry assays were used to determine the mechanism of ST3GAL6‑AS1‑mediated regulation of underlying targets. It was reported that knockdown of ST3GAL6‑AS1 suppressed the adhesion, migration and invasion ability of MM cells in vitro. Expression of ST3GAL6 was significantly reduced when ST3GAL6‑AS1 was knock downed in MM cells. Moreover, mechanistic investigation showed that ST3GAL6‑AS1 could suppress ST3GAL6 mRNA degradation via interacting with heterogeneous nuclear ribonucleoprotein A2B1 (hnRNPA2B1). The present results suggested that upregulated lncRNA ST3GAL6‑AS1 promotes adhesion and invasion of MM cells by binding with hnRNPA2B1 to regulate ST3GAL6 expression.
Collapse
Affiliation(s)
- Ying Shen
- Department of Hematology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710004, P.R. China
| | - Yuandong Feng
- Department of Hematology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710004, P.R. China
| | - Fangmei Li
- Department of Hematology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710004, P.R. China
| | - Yachun Jia
- Department of Hematology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710004, P.R. China
| | - Yue Peng
- Department of Hematology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710004, P.R. China
| | - Wanhong Zhao
- Department of Hematology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710004, P.R. China
| | - Jinsong Hu
- Health Science Center of Xi'an Jiaotong University, Xi'an, Shaanxi 710001, P.R. China
| | - Aili He
- Department of Hematology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710004, P.R. China
| |
Collapse
|
30
|
Abstract
The surfaces of all living organisms and most secreted proteins share a common feature: They are glycosylated. As the outermost-facing molecules, glycans participate in nearly all immunological processes, including driving host-pathogen interactions, immunological recognition and activation, and differentiation between self and nonself through a complex array of pathways and mechanisms. These fundamental immunologic roles are further cast into sharp relief in inflammatory, autoimmune, and cancer disease states in which immune regulation goes awry. Here, we review the broad impact of glycans on the immune system and discuss the changes and clinical opportunities associated with the onset of immunologic disease.
Collapse
Affiliation(s)
- Julie Y Zhou
- Department of Pathology, Case Western Reserve University School of Medicine, Cleveland, Ohio 44106-7288, USA;
| | - Brian A Cobb
- Department of Pathology, Case Western Reserve University School of Medicine, Cleveland, Ohio 44106-7288, USA;
| |
Collapse
|
31
|
Jia Y, Zhou J, Tan TK, Chung TH, Wong RWJ, Chooi JY, Lim JSL, Sanda T, Ooi M, De Mel S, Soekojo C, Chen Y, Zhang E, Cai Z, Shen P, Ruan J, Chng WJ. Myeloma-specific superenhancers affect genes of biological and clinical relevance in myeloma. Blood Cancer J 2021; 11:32. [PMID: 33579893 PMCID: PMC7881003 DOI: 10.1038/s41408-021-00421-7] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2020] [Revised: 12/14/2020] [Accepted: 01/20/2021] [Indexed: 01/09/2023] Open
Abstract
Multiple myeloma (MM) is an aggressive plasma cell neoplasm characterized by genomic heterogeneity. Superenhancers (SEs) are defined as large clusters of enhancers in close genomic proximity, which regulate genes for maintaining cellular identity and promote oncogenic transcription to which cancer cells highly addicted. Here, we analyzed cis-regulatory elements in MM samples with H3K27ac ChIP-seq, to identify novel SE-associated genes involved in the myeloma pathogenesis. SEs and their associated genes in cancerous tissue were compared with the control samples, and we found SE analysis alone uncovered cell-lineage-specific transcription factors and well-known oncogenes ST3GAL6 and ADM. Using a transcriptional CDK7 inhibitor, THZ1, coupled with H3K27ac ChlP-seq, we identified MAGI2 as a novel SE-associated gene of myeloma cells. Elevated MAGI2 was related to myelomagenesis with gradual increased expression from MGUS, SMM to newly diagnosed and relapsed MM. High prevalence of MAGI2 was also associated with poor survival of MM patients. Importantly, inhibition of the SE activity associated with MAGI2 decreased MAGI2 expression, inhibited cell growth and induced cell apoptosis. Mechanistically, we revealed that the oncogenic transcription factor, MAF, directly bound to the SE region and activated gene transcription. In summary, the discoveries of these acquired SEs-associated genes and the novel mechanism by which they are regulated provide new insights into MM biology and MAGI2-MAF-SE regulatory circuit offer potential novel targets for disease treatment.
Collapse
Affiliation(s)
- Yunlu Jia
- Cancer Science Institute of Singapore, National University of Singapore, 14 Medical Drive, Centre for Translational Medicine, Singapore, 117599, Republic of Singapore
- Department of Medical oncology, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Jianbiao Zhou
- Cancer Science Institute of Singapore, National University of Singapore, 14 Medical Drive, Centre for Translational Medicine, Singapore, 117599, Republic of Singapore
- Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117597, Republic of Singapore
| | - Tze King Tan
- Cancer Science Institute of Singapore, National University of Singapore, 14 Medical Drive, Centre for Translational Medicine, Singapore, 117599, Republic of Singapore
| | - Tae-Hoon Chung
- Cancer Science Institute of Singapore, National University of Singapore, 14 Medical Drive, Centre for Translational Medicine, Singapore, 117599, Republic of Singapore
| | - Regina Wan Ju Wong
- Cancer Science Institute of Singapore, National University of Singapore, 14 Medical Drive, Centre for Translational Medicine, Singapore, 117599, Republic of Singapore
| | - Jing-Yuan Chooi
- Cancer Science Institute of Singapore, National University of Singapore, 14 Medical Drive, Centre for Translational Medicine, Singapore, 117599, Republic of Singapore
| | - Julia Sze Lynn Lim
- Cancer Science Institute of Singapore, National University of Singapore, 14 Medical Drive, Centre for Translational Medicine, Singapore, 117599, Republic of Singapore
| | - Takaomi Sanda
- Cancer Science Institute of Singapore, National University of Singapore, 14 Medical Drive, Centre for Translational Medicine, Singapore, 117599, Republic of Singapore
- Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117597, Republic of Singapore
| | - Melissa Ooi
- Department of Hematology-Oncology, National University Cancer Institute of Singapore (NCIS), The National University Health System (NUHS), 1E, Kent Ridge Road, Singapore, 119228, Republic of Singapore
| | - Sanjay De Mel
- Department of Hematology-Oncology, National University Cancer Institute of Singapore (NCIS), The National University Health System (NUHS), 1E, Kent Ridge Road, Singapore, 119228, Republic of Singapore
| | - Cinnie Soekojo
- Department of Hematology-Oncology, National University Cancer Institute of Singapore (NCIS), The National University Health System (NUHS), 1E, Kent Ridge Road, Singapore, 119228, Republic of Singapore
| | - Yongxia Chen
- Department of Surgical Oncology, Sir Run Run Shaw Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | - Enfan Zhang
- Bone Marrow Transplantation Center, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
| | - Zhen Cai
- Bone Marrow Transplantation Center, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
| | - Peng Shen
- Department of Medical oncology, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Jian Ruan
- Department of Medical oncology, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Wee-Joo Chng
- Cancer Science Institute of Singapore, National University of Singapore, 14 Medical Drive, Centre for Translational Medicine, Singapore, 117599, Republic of Singapore.
- Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117597, Republic of Singapore.
- Department of Hematology-Oncology, National University Cancer Institute of Singapore (NCIS), The National University Health System (NUHS), 1E, Kent Ridge Road, Singapore, 119228, Republic of Singapore.
| |
Collapse
|
32
|
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
|
33
|
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
|
34
|
Su H, Wang M, Pang X, Guan F, Li X, Cheng Y. When Glycosylation Meets Blood Cells: A Glance of the Aberrant Glycosylation in Hematological Malignancies. Rev Physiol Biochem Pharmacol 2021; 180:85-117. [PMID: 34031738 DOI: 10.1007/112_2021_60] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Among neoplasia-associated epigenetic alterations, changes in cellular glycosylation have recently received attention as a key component of hematological malignancy progression. Alterations in glycosylation appear to not only directly impact cell growth and survival, but also alter the adhesion of tumor cells and their interactions with the microenvironment, facilitating cancer-induced immunomodulation and eventual metastasis. Changes in glycosylation arise from altered expression of glycosyltransferases, enzymes that catalyze the transfer of saccharide moieties to a wide range of acceptor substrates, such as proteins, lipids, and other saccharides in the endoplasmic reticulum (ER) and Golgi apparatus. Novel glycan structures in hematological malignancies represent new targets for the diagnosis and treatment of blood diseases. This review summarizes studies of the aberrant expression of glycans commonly found in hematological malignancies and their potential mechanisms and defines the specific roles of glycans as drivers or passengers in the development of hematological malignancies.
Collapse
Affiliation(s)
- Huining Su
- Center for Mitochondrial Biology and Medicine, The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, China
| | - Mimi Wang
- Center for Mitochondrial Biology and Medicine, The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, China
| | - Xingchen Pang
- Key Laboratory of Resource Biology and Biotechnology Western China, College of Life Science, Northwest University, Xi'an, China
| | - Feng Guan
- Key Laboratory of Resource Biology and Biotechnology Western China, College of Life Science, Northwest University, Xi'an, China
| | - Xiang Li
- Key Laboratory of Resource Biology and Biotechnology Western China, College of Life Science, Northwest University, Xi'an, China.
| | - Ying Cheng
- Center for Mitochondrial Biology and Medicine, The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, China.
| |
Collapse
|
35
|
Barbato A, Scandura G, Puglisi F, Cambria D, La Spina E, Palumbo GA, Lazzarino G, Tibullo D, Di Raimondo F, Giallongo C, Romano A. Mitochondrial Bioenergetics at the Onset of Drug Resistance in Hematological Malignancies: An Overview. Front Oncol 2020; 10:604143. [PMID: 33409153 PMCID: PMC7779674 DOI: 10.3389/fonc.2020.604143] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2020] [Accepted: 11/13/2020] [Indexed: 12/11/2022] Open
Abstract
The combined derangements in mitochondria network, function and dynamics can affect metabolism and ATP production, redox homeostasis and apoptosis triggering, contributing to cancer development in many different complex ways. In hematological malignancies, there is a strong relationship between cellular metabolism, mitochondrial bioenergetics, interconnections with supportive microenvironment and drug resistance. Lymphoma and chronic lymphocytic leukemia cells, e.g., adapt to intrinsic oxidative stress by increasing mitochondrial biogenesis. In other hematological disorders such as myeloma, on the contrary, bioenergetics changes, associated to increased mitochondrial fitness, derive from the adaptive response to drug-induced stress. In the bone marrow niche, a reverse Warburg effect has been recently described, consisting in metabolic changes occurring in stromal cells in the attempt to metabolically support adjacent cancer cells. Moreover, a physiological dynamic, based on mitochondria transfer, between tumor cells and their supporting stromal microenvironment has been described to sustain oxidative stress associated to proteostasis maintenance in multiple myeloma and leukemia. Increased mitochondrial biogenesis of tumor cells associated to acquisition of new mitochondria transferred by mesenchymal stromal cells results in augmented ATP production through increased oxidative phosphorylation (OX-PHOS), higher drug resistance, and resurgence after treatment. Accordingly, targeting mitochondrial biogenesis, electron transfer, mitochondrial DNA replication, or mitochondrial fatty acid transport increases therapy efficacy. In this review, we summarize selected examples of the mitochondrial derangements in hematological malignancies, which provide metabolic adaptation and apoptosis resistance, also supported by the crosstalk with tumor microenvironment. This field promises a rational design to improve target-therapy including the metabolic phenotype.
Collapse
Affiliation(s)
- Alessandro Barbato
- Department of Clinical and Experimental Medicine, University of Catania, Catania, Italy
| | - Grazia Scandura
- Department of General Surgery and Medical-Surgical Specialties, University of Catania, Catania, Italy
| | - Fabrizio Puglisi
- Department of Clinical and Experimental Medicine, University of Catania, Catania, Italy
| | - Daniela Cambria
- Department of Clinical and Experimental Medicine, University of Catania, Catania, Italy
| | - Enrico La Spina
- Department of General Surgery and Medical-Surgical Specialties, University of Catania, Catania, Italy
| | - Giuseppe Alberto Palumbo
- Department of Medical, Surgical Sciences and Advanced Technologies G.F. Ingrassia, University of Catania, Catania, Italy
| | - Giacomo Lazzarino
- Saint Camillus International University of Health and Medical Sciences, Rome, Italy
| | - Daniele Tibullo
- Department of Biotechnological and Biomedical Sciences, University of Catania, Catania, Italy
| | - Francesco Di Raimondo
- Department of General Surgery and Medical-Surgical Specialties, University of Catania, Catania, Italy
| | - Cesarina Giallongo
- Department of Medical, Surgical Sciences and Advanced Technologies G.F. Ingrassia, University of Catania, Catania, Italy
| | - Alessandra Romano
- Department of Surgery and Medical Specialties, University of Catania, Catania, Italy
| |
Collapse
|
36
|
Dalangood S, Zhu Z, Ma Z, Li J, Zeng Q, Yan Y, Shen B, Yan J, Huang R. Identification of glycogene-type and validation of ST3GAL6 as a biomarker predicts clinical outcome and cancer cell invasion in urinary bladder cancer. Theranostics 2020; 10:10078-10091. [PMID: 32929335 PMCID: PMC7481430 DOI: 10.7150/thno.48711] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2020] [Accepted: 07/27/2020] [Indexed: 12/11/2022] Open
Abstract
Background: Urinary bladder cancer (UBC) is one of the most common causes of morbidity and mortality worldwide characterized by a high risk of invasion and metastasis; however, the molecular classification biomarkers and underlying molecular mechanisms for UBC patient stratification on clinical outcome need to be investigated. Methods: A systematic transcriptomic analysis of 185 glycogenes in the public UBC datasets with survival information and clinicopathological parameters were performed using unsupervised hierarchical clustering. The gene signature for glycogene-type classification was identified using Limma package in R language, and correlated to 8 known molecular features by Gene Set Variation Analysis (GSVA). The clinical relevance and function of a glycogene was characterized by immunohistochemistry in UBC patient samples, and quantitative RT-PCR, Western blotting, promoter activity, MAL II blotting, immunofluorescence staining, wound healing, and transwell assays in UBC cells. Results: A 14-glycogene signature for glycogene-type classification was identified. Among them, ST3GAL6, a glycotransferase to transfer sialic acid to 3'-hydroxyl group of a galactose residue, showed a significant negative association with the subtype with luminal feature in UBC patients (n=2,130 in total). Increased ST3GAL6 was positively correlated to tumor stage, grade, and survival in UBCs from public datasets or our cohort (n=52). Transcription factor GATA3, a luminal-specific marker for UBC, was further identified as a direct upstream regulator of ST3GAL6 to negatively regulate its transactivation. ST3GAL6 depletion decreased MAL II level, cell invasion and migration in 5637 and J82 UBC cells. ST3GAL6 could reverse the effects of GATA3 on global sialylation and cell invasion in SW780 cells. Conclusions: Herein, we successfully identified a novel 14-gene signature for glycogene-type classification of UBC patients. ST3GAL6 gene, from this signature, was demonstrated as a potential biomarker for poor outcomes and cell invasion in UBCs.
Collapse
Affiliation(s)
- Sumiya Dalangood
- MOE Key Laboratory of Model Animals for Disease Study, Model Animal Research Center of Nanjing University, Nanjing 210061, China
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Zhen Zhu
- MOE Key Laboratory of Model Animals for Disease Study, Model Animal Research Center of Nanjing University, Nanjing 210061, China
| | - Zhihui Ma
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Jiaxuan Li
- MOE Key Laboratory of Model Animals for Disease Study, Model Animal Research Center of Nanjing University, Nanjing 210061, China
| | - Qinghe Zeng
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Yilin Yan
- Department of Urology, Shanghai General Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200080, China
| | - Bing Shen
- Department of Urology, Shanghai General Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200080, China
| | - Jun Yan
- Department of Laboratory Animal Science, Fudan University, Shanghai 200032, China
- Model Animal Research Center of Nanjing University, Nanjing 210061, China
| | - Ruimin Huang
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| |
Collapse
|
37
|
Ronchetti D, Todoerti K, Vinci C, Favasuli V, Agnelli L, Manzoni M, Pelizzoni F, Chiaramonte R, Platonova N, Giuliani N, Tassone P, Amodio N, Neri A, Taiana E. Expression Pattern and Biological Significance of the lncRNA ST3GAL6-AS1 in Multiple Myeloma. Cancers (Basel) 2020; 12:cancers12040782. [PMID: 32218309 PMCID: PMC7225964 DOI: 10.3390/cancers12040782] [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: 02/12/2020] [Revised: 03/16/2020] [Accepted: 03/23/2020] [Indexed: 11/16/2022] Open
Abstract
The biological impact of long non-coding RNAs (lncRNAs) in multiple myeloma (MM) is becoming an important aspect of investigation, which may contribute to the understanding of the complex pathobiology of the disease whilst also providing novel potential therapeutic targets. Herein, we investigated the expression pattern and the biological significance of the lncRNA ST3 beta-galactoside alpha-2,3 sialyltransferase 6 antisense RNA 1 (ST3GAL6-AS1) in MM. We documented a high ST3GAL6-AS1 expression level in MM compared to normal plasma cells (PCs) or other hematological malignancies. Transcriptome analyses of MM PCs from patients included in the CoMMpass database indicated a potential involvement of ST3GAL6-AS1 in MAPK signaling and ubiquitin-mediated proteolysis pathways. ST3GAL6-AS1 silencing by LNA-gapmeR antisense oligonucleotides inhibits cell proliferation and triggers apoptosis in MM cell line. Notably, ST3GAL6-AS1 silencing in vitro displayed the down-regulation of the MAPK pathway and protein ubiquitination. These data suggest that ST3GAL6-AS1 deregulation may play a pathogenetic role in MM by affecting both proliferation pathways and circuits fundamental for PC survival. However, ST3GAL6-AS1 expression levels seem not to be significantly associated with clinical outcome and its targeting appears to exert antagonistic effects with proteasome inhibitors used in MM. These findings strongly urge the need for further studies investigating the relevance of ST3GAL6-AS1 in MM.
Collapse
Affiliation(s)
- Domenica Ronchetti
- Department of Oncology and Hemato-Oncology, University of Milan, 20122 Milan, Italy; (D.R.); (C.V.); (V.F.); (L.A.); (M.M.); (E.T.)
- Hematology, Fondazione Cà Granda IRCCS Policlinico, 20122 Milan, Italy; (K.T.); (F.P.)
| | - Katia Todoerti
- Hematology, Fondazione Cà Granda IRCCS Policlinico, 20122 Milan, Italy; (K.T.); (F.P.)
| | - Cristina Vinci
- Department of Oncology and Hemato-Oncology, University of Milan, 20122 Milan, Italy; (D.R.); (C.V.); (V.F.); (L.A.); (M.M.); (E.T.)
| | - Vanessa Favasuli
- Department of Oncology and Hemato-Oncology, University of Milan, 20122 Milan, Italy; (D.R.); (C.V.); (V.F.); (L.A.); (M.M.); (E.T.)
- Hematology, Fondazione Cà Granda IRCCS Policlinico, 20122 Milan, Italy; (K.T.); (F.P.)
| | - Luca Agnelli
- Department of Oncology and Hemato-Oncology, University of Milan, 20122 Milan, Italy; (D.R.); (C.V.); (V.F.); (L.A.); (M.M.); (E.T.)
| | - Martina Manzoni
- Department of Oncology and Hemato-Oncology, University of Milan, 20122 Milan, Italy; (D.R.); (C.V.); (V.F.); (L.A.); (M.M.); (E.T.)
- Hematology, Fondazione Cà Granda IRCCS Policlinico, 20122 Milan, Italy; (K.T.); (F.P.)
| | - Francesca Pelizzoni
- Hematology, Fondazione Cà Granda IRCCS Policlinico, 20122 Milan, Italy; (K.T.); (F.P.)
| | - Raffaella Chiaramonte
- Department of Health Sciences, University of Milan, 20142 Milan, Italy; (R.C.); (N.P.)
| | - Natalia Platonova
- Department of Health Sciences, University of Milan, 20142 Milan, Italy; (R.C.); (N.P.)
| | - Nicola Giuliani
- Hematology and BMT Unit, Azienda Ospedaliero-Universitaria di Parma, 43125 Parma, Italy;
| | - Pierfrancesco Tassone
- Department of Experimental and Clinical Medicine, Magna Graecia University of Catanzaro, 88100 Catanzaro, Italy; (P.T.); (N.A.)
| | - Nicola Amodio
- Department of Experimental and Clinical Medicine, Magna Graecia University of Catanzaro, 88100 Catanzaro, Italy; (P.T.); (N.A.)
| | - Antonino Neri
- Department of Oncology and Hemato-Oncology, University of Milan, 20122 Milan, Italy; (D.R.); (C.V.); (V.F.); (L.A.); (M.M.); (E.T.)
- Hematology, Fondazione Cà Granda IRCCS Policlinico, 20122 Milan, Italy; (K.T.); (F.P.)
- Correspondence: ; Tel.: +39-02-5032-0420; Fax: +39-02-5032-0403
| | - Elisa Taiana
- Department of Oncology and Hemato-Oncology, University of Milan, 20122 Milan, Italy; (D.R.); (C.V.); (V.F.); (L.A.); (M.M.); (E.T.)
- Hematology, Fondazione Cà Granda IRCCS Policlinico, 20122 Milan, Italy; (K.T.); (F.P.)
| |
Collapse
|
38
|
Natoni A, Farrell ML, Harris S, Falank C, Kirkham-McCarthy L, Macauley MS, Reagan MR, O’Dwyer M. Sialyltransferase inhibition leads to inhibition of tumor cell interactions with E-selectin, VCAM1, and MADCAM1, and improves survival in a human multiple myeloma mouse model. Haematologica 2020; 105:457-467. [PMID: 31101754 PMCID: PMC7012485 DOI: 10.3324/haematol.2018.212266] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2018] [Accepted: 05/16/2019] [Indexed: 12/21/2022] Open
Abstract
Aberrant glycosylation resulting from altered expression of sialyltransferases, such as ST3 β-galactoside α2-3-sialyltransferase 6, plays an important role in disease progression in multiple myeloma (MM). Hypersialylation can lead to increased immune evasion, drug resistance, tumor invasiveness, and disseminated disease. In this study, we explore the in vitro and in vivo effects of global sialyltransferase inhibition on myeloma cells using the pan-sialyltransferase inhibitor 3Fax-Neu5Ac delivered as a per-acetylated methyl ester pro-drug. Specifically, we show in vivo that 3Fax-Neu5Ac improves survival by enhancing bortezomib sensitivity in an aggressive mouse model of MM. However, 3Fax-Neu5Ac treatment of MM cells in vitro did not reverse bortezomib resistance conferred by bone marrow (BM) stromal cells. Instead, 3Fax-Neu5Ac significantly reduced interactions of myeloma cells with E-selectin, MADCAM1 and VCAM1, suggesting that reduced sialylation impairs extravasation and retention of myeloma cells in the BM. Finally, we showed that 3Fax-Neu5Ac alters the post-translational modification of the α4 integrin, which may explain the reduced affinity of α4β1/α4β7 integrins for their counter-receptors. We propose that inhibiting sialylation may represent a valuable strategy to restrict myeloma cells from entering the protective BM microenvironment, a niche in which they are normally protected from chemotherapeutic agents such as bortezomib. Thus, our work demonstrates that targeting sialylation to increase the ratio of circulating to BM-resident MM cells represents a new avenue that could increase the efficacy of other anti-myeloma therapies and holds great promise for future clinical applications.
Collapse
Affiliation(s)
- Alessandro Natoni
- Apoptosis Research Center, National University of Ireland, Galway, Ireland
| | - Mariah L. Farrell
- Maine Medical Center Research Institute, Scarborough, ME, USA,Tufts University School of Medicine, Boston, MA, USA,Graduate School of Biomedical Science and Engineering, University of Maine, Orono, ME, USA
| | - Sophie Harris
- Maine Medical Center Research Institute, Scarborough, ME, USA,Tufts University School of Medicine, Boston, MA, USA,Graduate School of Biomedical Science and Engineering, University of Maine, Orono, ME, USA
| | - Carolyne Falank
- Maine Medical Center Research Institute, Scarborough, ME, USA,Tufts University School of Medicine, Boston, MA, USA,Graduate School of Biomedical Science and Engineering, University of Maine, Orono, ME, USA
| | | | - Matthew S. Macauley
- Department of Chemistry and Department of Medical Microbiology and Immunology, University of Alberta, Alberta, Canada
| | - Michaela R. Reagan
- Maine Medical Center Research Institute, Scarborough, ME, USA,Tufts University School of Medicine, Boston, MA, USA,Graduate School of Biomedical Science and Engineering, University of Maine, Orono, ME, USA
| | - Michael O’Dwyer
- Apoptosis Research Center, National University of Ireland, Galway, Ireland,Correspondence: MICHAEL O’DWYER,
| |
Collapse
|
39
|
Mehta KA, Patel KA, Pandya SJ, Patel PS. "Aberrant sialylation plays a significant role in oral squamous cell carcinoma progression". J Oral Pathol Med 2020; 49:253-259. [PMID: 31747460 DOI: 10.1111/jop.12976] [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: 07/15/2019] [Revised: 10/05/2019] [Accepted: 11/17/2019] [Indexed: 12/27/2022]
Abstract
OBJECTIVE Aberrant glycosylation, mainly sialylation and fucosylation, is recently considered as a major hallmark of cancer. Aberrant sialylation has long been associated with various neoplastic diseases. However, role of aberrant sialylation in oral cancer is still in its infancy. The present study aimed to examine mRNA expressions of α-2, 3, α-2, 6 sialyltransferase (ST) families and sialidase in 160 human oral cancer tissues. METHODS mRNA expression of ST3GAL1, ST3GAL2, ST3GAL3, ST3GAL4, ST3GAL6, ST6GAL1, and neuraminidase 3 (NEU3) was analyzed by RT-qPCR in 80 paired malignant and adjacent normal tissues from oral cancer patients. RESULTS The results indicated significant (P ≤ .05) down-regulation of various STs (ST3GAL1, ST3GAL2, ST3GAL3, ST3GAL4, ST3GAL6, and ST6GAL1) and sialidases (NEU3) in malignant tissues as compared to adjacent normal tissues. Higher mRNA levels of ST3GAL2 and ST3GAL3 were significantly associated with advanced stage of the disease, lymph node involvement, and perineural invasion, which denote their role in progression and metastasis of oral cancer. Present study also revealed altered sialylation patterns according to anatomical site of the disease and tobacco habit. CONCLUSION The study demonstrated significant role of elevated mRNA levels of ST3GAL2 and ST3GAL3 in disease progression and metastasis of oral carcinoma.
Collapse
Affiliation(s)
- Kruti A Mehta
- Molecular Oncology Laboratory, Cancer Biology Department, The Gujarat Cancer & Research Institute, Ahmedabad, Gujarat, India.,Life Science Department, University School of Sciences, Gujarat University, Ahmedabad, Gujarat, India
| | - Kinjal A Patel
- Molecular Oncology Laboratory, Cancer Biology Department, The Gujarat Cancer & Research Institute, Ahmedabad, Gujarat, India
| | - Shashank J Pandya
- Surgical Oncology Department, The Gujarat Cancer & Research Institute, Ahmedabad, Gujarat, India
| | - Prabhudas S Patel
- Molecular Oncology Laboratory, Cancer Biology Department, The Gujarat Cancer & Research Institute, Ahmedabad, Gujarat, India.,Life Science Department, University School of Sciences, Gujarat University, Ahmedabad, Gujarat, India
| |
Collapse
|
40
|
Sialic acid and biology of life: An introduction. SIALIC ACIDS AND SIALOGLYCOCONJUGATES IN THE BIOLOGY OF LIFE, HEALTH AND DISEASE 2020. [PMCID: PMC7153325 DOI: 10.1016/b978-0-12-816126-5.00001-9] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Sialic acids are important molecule with high structural diversity. They are known to occur in higher animals such as Echinoderms, Hemichordata, Cephalochorda, and Vertebrata and also in other animals such as Platyhelminthes, Cephalopoda, and Crustaceae. Plants are known to lack sialic acid. But they are reported to occur in viruses, bacteria, protozoa, and fungi. Deaminated neuraminic acid although occurs in vertebrates and bacteria, is reported to occur in abundance in the lower vertebrates. Sialic acids are mostly located in terminal ends of glycoproteins and glycolipids, capsular and tissue polysialic acids, bacterial lipooligosaccharides/polysaccharides, and in different forms that dictate their role in biology. Sialic acid play important roles in human physiology of cell-cell interaction, communication, cell-cell signaling, carbohydrate-protein interactions, cellular aggregation, development processes, immune reactions, reproduction, and in neurobiology and human diseases in enabling the infection process by bacteria and virus, tumor growth and metastasis, microbiome biology, and pathology. It enables molecular mimicry in pathogens that allows them to escape host immune responses. Recently sialic acid has found role in therapeutics. In this chapter we have highlighted the (i) diversity of sialic acid, (ii) their occurrence in the diverse life forms, (iii) sialylation and disease, and (iv) sialic acid and therapeutics.
Collapse
|
41
|
Ho M, Bianchi G, Anderson KC. Proteomics-inspired precision medicine for treating and understanding multiple myeloma. EXPERT REVIEW OF PRECISION MEDICINE AND DRUG DEVELOPMENT 2020; 5:67-85. [PMID: 34414281 DOI: 10.1080/23808993.2020.1732205] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
Abstract
Introduction Remarkable progress in molecular characterization methods has led to significant improvements in how we manage multiple myeloma (MM). The introduction of novel therapies has led to significant improvements in overall survival over the past 10 years. However, MM remains incurable and treatment choice is largely based on outdated risk-adaptive strategies that do not factor in improved treatment outcomes in the context of modern therapies. Areas covered This review discusses current risk-adaptive strategies in MM and the clinical application of proteomics in the monitoring of treatment response, disease progression, and minimal residual disease (MRD). We also discuss promising biomarkers of disease progression, treatment response, and chemoresistance. Finally, we will discuss an immunomics-based approach to monoclonal antibody (mAb), vaccine, and CAR-T cell development. Expert opinion It is an exciting era in oncology with basic scientific knowledge translating in novel therapeutic approaches to improve patient outcomes. With the advent of effective immunotherapies and targeted therapies, it has become crucial to identify biomarkers to aid in the stratification of patients based on anticipated sensitivity to chemotherapy. As a paradigm of diseases highly dependent on protein homeostasis, multiple myeloma provides the perfect opportunity to investigate the use of proteomics to aid in precision medicine.
Collapse
Affiliation(s)
- Matthew Ho
- UCD School of Medicine, College of Health and Agricultural Sciences, University College Dublin, Dublin, Ireland
| | - Giada Bianchi
- LeBow Institute for Myeloma Therapeutics and Jerome Lipper Multiple Myeloma Center, Department of Medical Oncology, Dana Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - Kenneth C Anderson
- LeBow Institute for Myeloma Therapeutics and Jerome Lipper Multiple Myeloma Center, Department of Medical Oncology, Dana Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
42
|
Qi F, Isaji T, Duan C, Yang J, Wang Y, Fukuda T, Gu J. ST3GAL3, ST3GAL4, and ST3GAL6 differ in their regulation of biological functions via the specificities for the α2,3-sialylation of target proteins. FASEB J 2019; 34:881-897. [PMID: 31914669 DOI: 10.1096/fj.201901793r] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2019] [Revised: 09/23/2019] [Accepted: 09/30/2019] [Indexed: 12/14/2022]
Abstract
The α2,3-sialylation of N-glycans is considered important but complicated because the functions of the three β-galactoside α2,3-sialyltransferases, ST3GAL3, ST3GAL4, and ST3GAL6, could be compensating for one another. To distinguish their specific functions, we established each individual knockout (KO) cell line. Loss of either the ST3GAL3 or ST3GAL6 genes decreased cell proliferation and colony formation, as opposed to the effect in the ST3GAL4 KO cells. The phosphorylation levels of ERK and AKT were significantly suppressed in the ST3GAL6 KO and ST3GAL3 KO cells, respectively. The cell aggregations were clearly observed in the KO cells, particularly the ST3GAL3 KO and ST3GAL6 KO cells, and the expression levels of E-cadherin and claudin-1 were enhanced in both those cell lines, but were suppressed in the ST3GAL4 KO cells. Those alterations were reversed with an overexpression of each corresponding gene in rescued cells. Of particular interest, the α2,3-sialylation levels of β1 integrin were clearly suppressed in the ST3GAL4 KO cells, but these were increased in the ST3GAL3 KO and ST3GAL6 KO cells, whereas the α2,3-sialylation levels of EGFR were significantly decreased in the ST3GAL6 KO cells. The decrease in α2,3-sialylation increased the α2,6-sialylation on β1, but not EGFR. Furthermore, a cross-restoration of each of the three genes in ST3GAL6 KO cells showed that overexpression of ST3GAL6 sufficiently rescued the total α2,3-sialylation levels, cell morphology, and α2,3-sialylation of EGFR, whereas the α2,3-sialylation levels of β1 were greatly enhanced by an overexpression of ST3GAL4. These results clearly demonstrate that the three α2,3-sialyltransferases modify characteristic target proteins and regulate cell biological functions in different ways.
Collapse
Affiliation(s)
- Feng Qi
- Division of Regulatory Glycobiology, Institute of Molecular Biomembrane and Glycobiology, Tohoku Medical and Pharmaceutical University, Sendai, Japan.,Department of Pharmacy, The Fourth Affiliated Hospital of Nantong University, Yancheng, China
| | - Tomoya Isaji
- Division of Regulatory Glycobiology, Institute of Molecular Biomembrane and Glycobiology, Tohoku Medical and Pharmaceutical University, Sendai, Japan
| | - Chengwei Duan
- Division of Regulatory Glycobiology, Institute of Molecular Biomembrane and Glycobiology, Tohoku Medical and Pharmaceutical University, Sendai, Japan
| | - Jie Yang
- Division of Regulatory Glycobiology, Institute of Molecular Biomembrane and Glycobiology, Tohoku Medical and Pharmaceutical University, Sendai, Japan
| | - Yuqin Wang
- Department of Pharmacology, Pharmacy College, Nantong University, Nantong, China
| | - Tomohiko Fukuda
- Division of Regulatory Glycobiology, Institute of Molecular Biomembrane and Glycobiology, Tohoku Medical and Pharmaceutical University, Sendai, Japan
| | - Jianguo Gu
- Division of Regulatory Glycobiology, Institute of Molecular Biomembrane and Glycobiology, Tohoku Medical and Pharmaceutical University, Sendai, Japan
| |
Collapse
|
43
|
Munkley J, Scott E. Targeting Aberrant Sialylation to Treat Cancer. MEDICINES (BASEL, SWITZERLAND) 2019; 6:medicines6040102. [PMID: 31614918 PMCID: PMC6963943 DOI: 10.3390/medicines6040102] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/20/2019] [Accepted: 10/10/2019] [Indexed: 04/28/2023]
Abstract
Cell surface carbohydrates (known as glycans) are often aberrantly expressed or found at atypical levels in cancer. Glycans can impact all steps in tumour progression, from malignant transformation to metastasis, and have roles in all the cancer hallmarks. An increased understanding of glycans in the metastatic cascade offers exciting new therapeutic opportunities. Glycan-based targeting strategies are currently being tested in clinical trials and are a rich and untapped frontier for development. As we learn more about cancer glycobiology, new targets will continue to emerge for drug design. One key change in tumour glycosylation is the upregulation of cancer-associated sialylated glycans. Abnormal sialylation is integral to tumour growth, metastasis and immune evasion; therefore, targeting sialic acid moieties in cancer could be of high therapeutic value. Here, we summarise the changes to sialic acid biology in cancer and discuss recent advances and technologies bringing sialic-acid targeting treatments to the forefront of cancer therapeutics.
Collapse
Affiliation(s)
- Jennifer Munkley
- Institute of Genetic Medicine, Newcastle University, Newcastle upon Tyne NE1 3BZ, UK.
| | - Emma Scott
- Institute of Genetic Medicine, Newcastle University, Newcastle upon Tyne NE1 3BZ, UK.
| |
Collapse
|
44
|
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
|
45
|
Bärenwaldt A, Läubli H. The sialoglycan-Siglec glyco-immune checkpoint - a target for improving innate and adaptive anti-cancer immunity. Expert Opin Ther Targets 2019; 23:839-853. [PMID: 31524529 DOI: 10.1080/14728222.2019.1667977] [Citation(s) in RCA: 57] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Introduction: During cancer progression, tumor cells develop several mechanisms to prevent killing and to shape the immune system into a tumor-promoting environment. One of such regulatory mechanism is the overexpression of sialic acid (Sia) on carbohydrates of proteins and lipids on tumor cells. Sia-containing glycans or sialoglycans were shown to inhibit immune effector functions of NK cells and T cells by engaging inhibitory Siglec receptors on the surface of these cells. They can also modulate the differentiation of myeloid cells into tumor-promoting M2 macrophages. Areas covered: We review the role of sialoglycans in cancer and introduce the Siglecs, their expression on different immune cells and their interaction with cancer-associated sialoglycans. The targeting of this sialoglycan-Siglec glyco-immune checkpoint is discussed along with potential therapeutic approaches. Pubmed was searched for publications on Siglecs, sialic acid, and cancer. Expert opinion: The targeting of sialoglycan-Siglec interactions has become a major focus in cancer research. New approaches have been developed that directly target sialic acids in tumor lesions. Targeted sialidases that cleave sialic acid specifically in the tumor, have already shown efficacy; efforts targeting the sialoglycan-Siglec pathway for improvement of CAR T cell therapy are ongoing. The sialoglycan-Siglec immune checkpoint is a promising new target for cancer immunotherapy.
Collapse
Affiliation(s)
- Anne Bärenwaldt
- Division of Medical Oncology, and Laboratory for Cancer Immunotherapy, Department of Biomedicine, University Hospital Basel , Basel , Switzerland
| | - Heinz Läubli
- Division of Medical Oncology, and Laboratory for Cancer Immunotherapy, Department of Biomedicine, University Hospital Basel , Basel , Switzerland
| |
Collapse
|
46
|
Zhang W, Yang M, Wang Y, Wu X, Zhang X, Ding Y, Yin Z. Genomic analysis reveals selection signatures of the Wannan Black pig during domestication and breeding. ASIAN-AUSTRALASIAN JOURNAL OF ANIMAL SCIENCES 2019; 33:712-721. [PMID: 31480149 PMCID: PMC7206397 DOI: 10.5713/ajas.19.0289] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/07/2019] [Accepted: 08/01/2019] [Indexed: 12/31/2022]
Abstract
Objective The Wannan Black pig is a typical Chinese indigenous, disease-resistant pig breed with high fertility, and a crude-feed tolerance that has been bred by artificial selection in the south of Anhui province for a long time. However, genome variation, genetic relationships with other pig breeds, and domestication, remain poorly understood. Here, we focus on elucidating the genetic characteristics of the Wannan Black pig and identifying selection signatures during domestication and breeding. Methods We identified the whole-genome variation in the Wannan Black pig and performed population admixture analyses to determine genetic relationships with other domesticated pig breeds and wild boars. Then, we identified the selection signatures between the Wannan Black pig and Asian wild boars in 100-kb windows sliding in 10 kb steps by using two approaches: the fixation index (FST) and π ratios. Results Resequencing the Wannan Black pig genome yielded 501.52 G of raw data. After calling single-nucleotide variants (SNVs) and insertions/deletions (InDels), we identified 21,316,754 SNVs and 5,067,206 InDels (2,898,582 inserts and 2,168,624 deletions). Additionally, we found genes associated with growth, immunity, and digestive functions. Conclusion Our findings help in explaining the unique genetic and phenotypic characteristics of Wannan Black pigs, which in turn can be informative for future breeding programs of Wannan Black pigs.
Collapse
Affiliation(s)
- Wei Zhang
- College of Animal Science and Technology, Anhui Agricultural University, Hefei 230036, China
| | - Min Yang
- College of Animal Science and Technology, Anhui Agricultural University, Hefei 230036, China
| | - Yuanlang Wang
- College of Animal Science and Technology, Anhui Agricultural University, Hefei 230036, China
| | - Xudong Wu
- College of Animal Science and Technology, Anhui Agricultural University, Hefei 230036, China
| | - Xiaodong Zhang
- College of Animal Science and Technology, Anhui Agricultural University, Hefei 230036, China
| | - Yueyun Ding
- College of Animal Science and Technology, Anhui Agricultural University, Hefei 230036, China
| | - Zongjun Yin
- College of Animal Science and Technology, Anhui Agricultural University, Hefei 230036, China
| |
Collapse
|
47
|
Muz B, Azab F, Fiala M, King J, Kohnen D, Fogler WE, Smith T, Magnani JL, Vij R, Azab AK. Inhibition of E-Selectin (GMI-1271) or E-selectin together with CXCR4 (GMI-1359) re-sensitizes multiple myeloma to therapy. Blood Cancer J 2019; 9:68. [PMID: 31431613 PMCID: PMC6702213 DOI: 10.1038/s41408-019-0227-3] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2018] [Revised: 04/07/2019] [Accepted: 05/14/2019] [Indexed: 12/26/2022] Open
Affiliation(s)
- Barbara Muz
- Department of Radiation Oncology, Cancer Biology Division, Washington University in St. Louis School of Medicine, St. Louis, MO, USA
| | - Feda Azab
- Department of Radiation Oncology, Cancer Biology Division, Washington University in St. Louis School of Medicine, St. Louis, MO, USA
| | - Mark Fiala
- Department of Medicine, Division of Medical Oncology, Washington University in St. Louis School of Medicine, Saint Louis, MO, USA
| | - Justin King
- Department of Medicine, Division of Medical Oncology, Washington University in St. Louis School of Medicine, Saint Louis, MO, USA
| | - Daniel Kohnen
- Department of Medicine, Division of Medical Oncology, Washington University in St. Louis School of Medicine, Saint Louis, MO, USA
| | | | - Ted Smith
- GlycoMimetics Inc., Rockville, MD, USA
| | | | - Ravi Vij
- Department of Medicine, Division of Medical Oncology, Washington University in St. Louis School of Medicine, Saint Louis, MO, USA
| | - Abdel Kareem Azab
- Department of Radiation Oncology, Cancer Biology Division, Washington University in St. Louis School of Medicine, St. Louis, MO, USA.
| |
Collapse
|
48
|
Hillary RF, McCartney DL, Harris SE, Stevenson AJ, Seeboth A, Zhang Q, Liewald DC, Evans KL, Ritchie CW, Tucker-Drob EM, Wray NR, McRae AF, Visscher PM, Deary IJ, Marioni RE. Genome and epigenome wide studies of neurological protein biomarkers in the Lothian Birth Cohort 1936. Nat Commun 2019; 10:3160. [PMID: 31320639 PMCID: PMC6639385 DOI: 10.1038/s41467-019-11177-x] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2019] [Accepted: 06/24/2019] [Indexed: 02/06/2023] Open
Abstract
Although plasma proteins may serve as markers of neurological disease risk, the molecular mechanisms responsible for inter-individual variation in plasma protein levels are poorly understood. Therefore, we conduct genome- and epigenome-wide association studies on the levels of 92 neurological proteins to identify genetic and epigenetic loci associated with their plasma concentrations (n = 750 healthy older adults). We identify 41 independent genome-wide significant (P < 5.4 × 10-10) loci for 33 proteins and 26 epigenome-wide significant (P < 3.9 × 10-10) sites associated with the levels of 9 proteins. Using this information, we identify biological pathways in which putative neurological biomarkers are implicated (neurological, immunological and extracellular matrix metabolic pathways). We also observe causal relationships (by Mendelian randomisation analysis) between changes in gene expression (DRAXIN, MDGA1 and KYNU), or DNA methylation profiles (MATN3, MDGA1 and NEP), and altered plasma protein levels. Together, this may help inform causal relationships between biomarkers and neurological diseases.
Collapse
Affiliation(s)
- Robert F Hillary
- Centre for Genomic and Experimental Medicine, Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh, EH4 2XU, UK
| | - Daniel L McCartney
- Centre for Genomic and Experimental Medicine, Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh, EH4 2XU, UK
| | - Sarah E Harris
- Centre for Cognitive Ageing and Cognitive Epidemiology, University of Edinburgh, Edinburgh, EH8 9JZ, UK
- Department of Psychology, University of Edinburgh, Edinburgh, EH8 9JZ, UK
| | - Anna J Stevenson
- Centre for Genomic and Experimental Medicine, Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh, EH4 2XU, UK
| | - Anne Seeboth
- Centre for Genomic and Experimental Medicine, Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh, EH4 2XU, UK
| | - Qian Zhang
- Institute for Molecular Bioscience, University of Queensland, Brisbane, QLD, 4072, Australia
| | - David C Liewald
- Centre for Cognitive Ageing and Cognitive Epidemiology, University of Edinburgh, Edinburgh, EH8 9JZ, UK
| | - Kathryn L Evans
- Centre for Genomic and Experimental Medicine, Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh, EH4 2XU, UK
- Centre for Cognitive Ageing and Cognitive Epidemiology, University of Edinburgh, Edinburgh, EH8 9JZ, UK
| | - Craig W Ritchie
- Edinburgh Dementia Prevention, Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, EH16 4UX, UK
| | - Elliot M Tucker-Drob
- Department of Psychology, The University of Texas at Austin, Austin, TX, 78712, USA
- Population Research Center, The University of Texas at Austin, Austin, TX, 78712, USA
| | - Naomi R Wray
- Institute for Molecular Bioscience, University of Queensland, Brisbane, QLD, 4072, Australia
| | - Allan F McRae
- Institute for Molecular Bioscience, University of Queensland, Brisbane, QLD, 4072, Australia
| | - Peter M Visscher
- Institute for Molecular Bioscience, University of Queensland, Brisbane, QLD, 4072, Australia
| | - Ian J Deary
- Centre for Cognitive Ageing and Cognitive Epidemiology, University of Edinburgh, Edinburgh, EH8 9JZ, UK
- Department of Psychology, University of Edinburgh, Edinburgh, EH8 9JZ, UK
| | - Riccardo E Marioni
- Centre for Genomic and Experimental Medicine, Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh, EH4 2XU, UK.
- Centre for Cognitive Ageing and Cognitive Epidemiology, University of Edinburgh, Edinburgh, EH8 9JZ, UK.
| |
Collapse
|
49
|
Swan D, Lynch K, Gurney M, O’Dwyer M. Current and emerging immunotherapeutic approaches to the treatment of multiple myeloma. Ther Adv Hematol 2019; 10:2040620719854171. [PMID: 31244984 PMCID: PMC6582283 DOI: 10.1177/2040620719854171] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2019] [Accepted: 05/09/2019] [Indexed: 11/17/2022] Open
Abstract
Multiple myeloma (MM) has a worldwide incidence of 1-5/100,000/year. Outcomes have improved significantly in recent years following incorporation of immunomodulatory drugs and proteasome inhibitors into standard-of-care regimes. MM is profoundly immunosuppressive, enabling immune evasion, proliferation and disease progression. The role of the immune system in MM is becoming increasingly characterized and understood, and numerous therapies are under development or in routine clinical use targeting these elements of MM pathogenesis. In this review we discuss the immunosuppressive effects of MM, then the therapies targeting these defects. Specifically, we review the monoclonal and bispecific antibodies, alongside adoptive cellular therapies currently under investigation.
Collapse
Affiliation(s)
- Dawn Swan
- Clinical Research Facility, University Hospital Galway, Newcastle Road, Galway, H91 YR71, Ireland
| | - Kevin Lynch
- National University of Ireland, Galway, Ireland
| | - Mark Gurney
- University Hospital Galway, Ireland
- National University of Ireland, Galway, Ireland
| | - Michael O’Dwyer
- University Hospital Galway, Ireland
- National University of Ireland, Galway, Ireland
| |
Collapse
|
50
|
I-branched carbohydrates as emerging effectors of malignant progression. Proc Natl Acad Sci U S A 2019; 116:13729-13737. [PMID: 31213534 DOI: 10.1073/pnas.1900268116] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Cell surface carbohydrates, termed "glycans," are ubiquitous posttranslational effectors that can tune cancer progression. Often aberrantly displayed or found at atypical levels on cancer cells, glycans can impact essentially all progressive steps, from malignant transformation to metastases formation. Glycans are structural entities that can directly bind promalignant glycan-binding proteins and help elicit optimal receptor-ligand activity of growth factor receptors, integrins, integrin ligands, lectins, and other type-1 transmembrane proteins. Because glycans play an integral role in a cancer cell's malignant activity and are frequently uniquely expressed, preclinical studies on the suitability of glycans as anticancer therapeutic targets and their promise as biomarkers of disease progression continue to intensify. While sialylation and fucosylation have predominated the focus of cancer-associated glycan modifications, the emergence of blood group I antigens (or I-branched glycans) as key cell surface moieties capable of modulating cancer virulence has reenergized investigations into the role of the glycome in malignant progression. I-branched glycans catalyzed principally by the I-branching enzyme GCNT2 are now indicated in several malignancies. In this Perspective, the putative role of GCNT2/I-branching in cancer progression is discussed, including exciting insights on how I-branches can potentially antagonize the cancer-promoting activity of β-galactose-binding galectins.
Collapse
|