1
|
Zhao Y, Lyu Z, Prather B, Lewis TR, Kang J, Wang RE. Metabolic Probing of Sialylated Glycoconjugates with Fluorine-Selenol Displacement Reaction (FSeDR). ACS BIO & MED CHEM AU 2025; 5:119-130. [PMID: 39990939 PMCID: PMC11843343 DOI: 10.1021/acsbiomedchemau.4c00084] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/27/2024] [Revised: 11/27/2024] [Accepted: 12/02/2024] [Indexed: 02/25/2025]
Abstract
Dysregulated sialic acid biosynthesis is characteristic of the onset and progression of human diseases including hormone-sensitive prostate cancer and breast cancer. The sialylated glycoconjugates involved in this process are therefore important targets for identification and functional studies. To date, one of the most common strategies is metabolic glycoengineering, which utilizes N-acetylmannosamine (ManNAc) analogues such as N-azidoacetylmannosamine (ManNAz) to hijack sialic acid biosynthesis and label the sialylated glycoconjugates with "click chemistry (CuAAC)" tags. Yet, current chemical modifications including those CuAAC-based alkyne/azide tags are still big in size, and the resulting steric hindrance perturbs the mannosamine and sialic acid derivatives' recognition and metabolism by enzymes involved in biosynthetic pathways. As a result, the peracetylated ManNAz has compromised incorporation to sialic acid substrates and manifests cellular growth inhibition and cytotoxicity. Herein, we show that the α-fluorinated peracetylated analogue ManN(F-Ac) displayed a satisfying safety profile in mammalian cell lines at concentrations as high as 500 μM. More importantly, aliphatic selenol-containing probes can efficiently displace α-fluorine in fluoroacetamide-containing substrates including ManN(F-Ac) at a neutral pH range (∼7.2). The combined use of peracetylated ManN(F-Ac) and the dethiobiotin-selenol probe as the fluorine-selenol displacement reaction (FSeDR) toolkit allowed for successful metabolic labeling of sialoglycoproteins in multiple prostate and cancer cell lines, including PC-3 and MDA-MB-231. More sialoglycoproteins in these cell lines were demonstrated to be labeled by FSeDR compared with the traditional CuAAC approach. Lastly, with FSeDR-mediated metabolic labeling, we were able to probe the cellular expression level and spatial distribution of sialylated glycoconjugates during the progression of these hormone-sensitive cancer cells. Taken together, the promising results suggest the potential of the FSeDR strategy to efficiently and systematically identify and study sialic acid substrates and potentially empower metabolic engineering on a diverse set of glycosylated proteins that are vital for human diseases.
Collapse
Affiliation(s)
| | | | - Benjamin Prather
- Department of Chemistry, Temple University, 1901 N. 13th Street, Philadelphia, Pennsylvania 19122, United States
| | - Todd R. Lewis
- Department of Chemistry, Temple University, 1901 N. 13th Street, Philadelphia, Pennsylvania 19122, United States
| | - Jinfeng Kang
- Department of Chemistry, Temple University, 1901 N. 13th Street, Philadelphia, Pennsylvania 19122, United States
| | - Rongsheng E. Wang
- Department of Chemistry, Temple University, 1901 N. 13th Street, Philadelphia, Pennsylvania 19122, United States
| |
Collapse
|
2
|
Cheng P, Hothpet V, Bhat G, Bailey K, Li L, Samuelson DR. Alcohol induces α2-6sialo mucin O-glycans that kill U937 macrophages mediated by sialic acid-binding immunoglobulin-like lectin 7 (Siglec 7). FEBS Open Bio 2025; 15:165-179. [PMID: 39592427 PMCID: PMC11705458 DOI: 10.1002/2211-5463.13919] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2024] [Revised: 09/06/2024] [Accepted: 10/18/2024] [Indexed: 11/28/2024] Open
Abstract
Alcohol misuse increases infections and cancer fatalities, but mechanisms underlying its toxicity are ill-defined. We show that alcohol treatment of human tracheobronchial epithelial cells leads to inactivation of giantin-mediated Golgi targeting of glycosylation enzymes. Loss of core 2 N-acetylglucosaminyltransferase 1, which uses only giantin for Golgi targeting, coupled with shifted targeting of other glycosylation enzymes to Golgi matrix protein 130-Golgi reassembly stacking protein 65, the site normally used by core 1 enzyme, results in loss of sialyl Lewis x and increase of sialyl Lewis a and α2-6sialo mucin O-glycans. The α2-6sialo mucin O-glycans induced by alcohol cause death of U937 macrophages mediated by sialic acid-binding immunoglobulin-like lectin 7. These results provide a mechanistic insight into the cause of the toxic effects of alcohol and might contribute to the development of therapies to alleviate its toxicity.
Collapse
Affiliation(s)
- Pi‐Wan Cheng
- Department of Biochemistry and Molecular Biology, College of MedicineUniversity of Nebraska Medical CenterOmahaNEUSA
- Fred and Pamela Buffett Cancer CenterUniversity of Nebraska Medical CenterOmahaNEUSA
| | - Vishwanath‐Reddy Hothpet
- Department of Biochemistry and Molecular Biology, College of MedicineUniversity of Nebraska Medical CenterOmahaNEUSA
- Present address:
State Forensic LaboratoryBengaluruIndia
| | - Ganapati Bhat
- Department of Biochemistry and Molecular Biology, College of MedicineUniversity of Nebraska Medical CenterOmahaNEUSA
- Present address:
Dayananda Sagar UniversityBengaluruIndia
| | - Kristina Bailey
- Department of Internal Medicine, College of MedicineUniversity of Nebraska Medical CenterOmahaNEUSA
| | - Lei Li
- Department of Chemistry and Center for Diagnostic & TherapeuticsGeorgia State UniversityAtlantaGAUSA
| | - Derrick R. Samuelson
- Department of Internal Medicine, College of MedicineUniversity of Nebraska Medical CenterOmahaNEUSA
| |
Collapse
|
3
|
Brockhausen I, Falconer D, Sara S. Relationships between bacteria and the mucus layer. Carbohydr Res 2024; 546:109309. [PMID: 39549591 DOI: 10.1016/j.carres.2024.109309] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2024] [Revised: 11/05/2024] [Accepted: 11/07/2024] [Indexed: 11/18/2024]
Abstract
The mucus layer on epithelial cells is an essential barrier, as well as a nutrient-rich niche for bacteria, forming a dynamic, functional and symbiotic ecosystem and first line of defense against invading pathogens. Particularly bacteria in biofilms are very difficult to eradicate. The extensively O-glycosylated mucins are the main glycoproteins in mucus that interact with microbes. For example, mucins act as adhesion receptors and nutritional substrates for gut bacteria. Mucins also play important roles in immune responses, and they control the composition of the microbiome, primarily due to the abundance of complex O-glycans. In inflammation or infection, the structures of mucin O-glycans can change and thus affect mucin function, impact biofilm formation and the induction of virulence pathways in bacteria. In turn, bacteria can support host cell growth, mucin production and can stimulate changes in the host immune system and responses leading to healthy tissue function. The external polysaccharides of bacteria are critical for controlling adhesion and biofilm formation. It is therefore important to understand the relationships between the mucus layer and microbes, the mechanisms and regulation of the biosynthesis of mucins, of bacterial surface polysaccharides, and adhesins. This knowledge can provide biomarkers, vaccines and help to develop new approaches for improved therapies, including antibiotic treatments.
Collapse
Affiliation(s)
- Inka Brockhausen
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston, Ontario, Canada.
| | - Dylan Falconer
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston, Ontario, Canada
| | - Sara Sara
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston, Ontario, Canada
| |
Collapse
|
4
|
Khorami-Sarvestani S, Hanash SM, Fahrmann JF, León-Letelier RA, Katayama H. Glycosylation in cancer as a source of biomarkers. Expert Rev Proteomics 2024; 21:345-365. [PMID: 39376081 DOI: 10.1080/14789450.2024.2409224] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2024] [Revised: 08/12/2024] [Accepted: 09/17/2024] [Indexed: 10/09/2024]
Abstract
INTRODUCTION Glycosylation, the process of glycan synthesis and attachment to target molecules, is a crucial and common post-translational modification (PTM) in mammalian cells. It affects the protein's hydrophilicity, charge, solubility, structure, localization, function, and protection from proteolysis. Aberrant glycosylation in proteins can reveal new detection and therapeutic Glyco-biomarkers, which help to improve accurate early diagnosis and personalized treatment. This review underscores the pivotal role of glycans and glycoproteins as a source of biomarkers in human diseases, particularly cancer. AREAS COVERED This review delves into the implications of glycosylation, shedding light on its intricate roles in cancer-related cellular processes influencing biomarkers. It is underpinned by a thorough examination of literature up to June 2024 in PubMed, Scopus, and Google Scholar; concentrating on the terms: (Glycosylation[Title/Abstract]) OR (Glycan[Title/Abstract]) OR (glycoproteomics[Title/Abstract]) OR (Proteoglycans[Title/Abstract]) OR (Glycomarkers[Title/Abstract]) AND (Cancer[Title/Abstract]) AND ((Diagno*[Title/Abstract]) OR (Progno*[Title/Abstract])). EXPERT OPINION Glyco-biomarkers enhance early cancer detection, allow early intervention, and improve patient prognoses. However, the abundance and complex dynamic glycan structure may make their scientific and clinical application difficult. This exploration of glycosylation signatures in cancer biomarkers can provide a detailed view of cancer etiology and instill hope in the potential of glycosylation to revolutionize cancer research.
Collapse
Affiliation(s)
- Sara Khorami-Sarvestani
- Department of Clinical Cancer Prevention, the University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Samir M Hanash
- Department of Clinical Cancer Prevention, the University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Johannes F Fahrmann
- Department of Clinical Cancer Prevention, the University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Ricardo A León-Letelier
- Department of Clinical Cancer Prevention, the University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Hiroyuki Katayama
- Department of Clinical Cancer Prevention, the University of Texas MD Anderson Cancer Center, Houston, TX, USA
| |
Collapse
|
5
|
Hao Y, Gu C, Luo W, Shen J, Xie F, Zhao Y, Song X, Han Z, He J. The role of protein post-translational modifications in prostate cancer. PeerJ 2024; 12:e17768. [PMID: 39148683 PMCID: PMC11326433 DOI: 10.7717/peerj.17768] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2024] [Accepted: 06/27/2024] [Indexed: 08/17/2024] Open
Abstract
Involving addition of chemical groups or protein units to specific residues of the target protein, post-translational modifications (PTMs) alter the charge, hydrophobicity, and conformation of a protein, which in turn influences protein function, protein-protein interaction, and protein aggregation. These alterations, which include phosphorylation, glycosylation, ubiquitination, methylation, acetylation, lipidation, and lactylation, are significant biological events in the development of cancer, and play vital roles in numerous biological processes. The processes behind essential functions, the screening of clinical illness signs, and the identification of therapeutic targets all depend heavily on further research into the PTMs. This review outlines the influence of several PTM types on prostate cancer (PCa) diagnosis, therapy, and prognosis in an effort to shed fresh light on the molecular causes and progression of the disease.
Collapse
Affiliation(s)
- Yinghui Hao
- Central Laboratory, The Affiliated Panyu Central Hospital of Guangzhou Medical University, Guangzhou, China
| | - Chenqiong Gu
- Central Laboratory, The Affiliated Panyu Central Hospital of Guangzhou Medical University, Guangzhou, China
| | - Wenfeng Luo
- Central Laboratory, The Affiliated Panyu Central Hospital of Guangzhou Medical University, Guangzhou, China
| | - Jian Shen
- Central Laboratory, The Affiliated Panyu Central Hospital of Guangzhou Medical University, Guangzhou, China
| | - Fangmei Xie
- Central Laboratory, The Affiliated Panyu Central Hospital of Guangzhou Medical University, Guangzhou, China
| | - Ying Zhao
- Central Laboratory, The Affiliated Panyu Central Hospital of Guangzhou Medical University, Guangzhou, China
| | - Xiaoyu Song
- Central Laboratory, The Affiliated Panyu Central Hospital of Guangzhou Medical University, Guangzhou, China
| | - Zeping Han
- Central Laboratory, The Affiliated Panyu Central Hospital of Guangzhou Medical University, Guangzhou, China
| | - Jinhua He
- Central Laboratory, The Affiliated Panyu Central Hospital of Guangzhou Medical University, Guangzhou, China
- Rehabilitation Medicine Institute of Panyu District, Guangzhou, Guangdong, China
| |
Collapse
|
6
|
Radziejewska I. Tumor-associated carbohydrate antigens of MUC1 - Implication in cancer development. Biomed Pharmacother 2024; 174:116619. [PMID: 38643541 DOI: 10.1016/j.biopha.2024.116619] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2024] [Revised: 04/12/2024] [Accepted: 04/17/2024] [Indexed: 04/23/2024] Open
Abstract
Glycosylation of cancerous epithelial MUC1 protein is specifically altered in comparison to that which is presented by healthy cells. One of such changes is appearing tumor-associated carbohydrate antigens (TACAs) which are rare in normal tissues and are highly correlated with poor clinical outcomes and cancer progression. This review summarizes and describes the role of Tn, T antigens, their sialylated forms as well as fucosylated Lewis epitopes in different aspects of tumor development, progression, and metastasis. Finally, applications of MUC1 glycan epitopes as potential targets for therapeutic strategy of cancers are notified. One of the novelties of this review is presentation of TACAs as inherently connected with MUC1 mucin.
Collapse
Affiliation(s)
- Iwona Radziejewska
- Department of Medical Chemistry, Medical University of Białystok, ul. Mickiewicza 2, Białystok 15-222, Poland.
| |
Collapse
|
7
|
Wang W, de Nier CR, Wuhrer M, Lageveen-Kammeijer GS. In-Depth Glycoproteomic Assay of Urinary Prostatic Acid Phosphatase. ACS MEASUREMENT SCIENCE AU 2024; 4:117-126. [PMID: 38404489 PMCID: PMC10885330 DOI: 10.1021/acsmeasuresciau.3c00055] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/27/2023] [Revised: 11/12/2023] [Accepted: 11/14/2023] [Indexed: 02/27/2024]
Abstract
Prostate-specific antigen (PSA) is a well-known clinical biomarker in prostate cancer (PCa) diagnosis, but a better test is still needed, as the serum-level-based PSA quantification exhibits limited specificity and comes with poor predictive value. Prior to PSA, prostatic acid phosphatase (PAP) was used, but it was replaced by PSA because PSA improved the early detection of PCa. Upon revisiting PAP and its glycosylation specifically, it appears to be a promising new biomarker candidate. Namely, previous studies have indicated that PAP glycoforms differ between PCa and non-PCa individuals. However, an in-depth characterization of PAP glycosylation is still lacking. In this study, we established an in-depth glycoproteomic assay for urinary PAP by characterizing both the micro- and macroheterogeneity of the PAP glycoprofile. For this purpose, PAP samples were analyzed by capillary electrophoresis coupled to mass spectrometry after affinity purification from urine and proteolytic digestion. The developed urinary PAP assay was applied on a pooled DRE (digital rectal examination) urine sample from nine individuals. Three glycosylation sites were characterized, namely N94, N220, and N333, via N-glycopeptide analysis. Taking sialic acid linkage isomers into account, a total of 63, 27, and 4 N-glycan structures were identified, respectively. The presented PAP glycoproteomic assay will enable the determination of potential glycomic biomarkers for the early detection and prognosis of PCa in cohort studies.
Collapse
Affiliation(s)
- Wei Wang
- Leiden
University Medical Center, Center for Proteomics
and Metabolomics, Leiden 2300 RC, The Netherlands
| | - Carmen R. de Nier
- Leiden
University Medical Center, Center for Proteomics
and Metabolomics, Leiden 2300 RC, The Netherlands
| | - Manfred Wuhrer
- Leiden
University Medical Center, Center for Proteomics
and Metabolomics, Leiden 2300 RC, The Netherlands
| | - Guinevere S.M. Lageveen-Kammeijer
- Leiden
University Medical Center, Center for Proteomics
and Metabolomics, Leiden 2300 RC, The Netherlands
- University
of Groningen, Groningen Research
Institute of Pharmacy, Groningen 9713 AV, The Netherlands
| |
Collapse
|
8
|
Weng W, Ren S, Teng C, Guo J, Guo Q, Zhang W, Zong C, Ding N. Chemoenzymatic synthesis and immunological evaluation of sialyl-Thomsen-Friedenreich (sTF) antigen conjugate to CRM197. Bioorg Med Chem 2024; 100:117615. [PMID: 38342079 DOI: 10.1016/j.bmc.2024.117615] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2023] [Revised: 01/25/2024] [Accepted: 01/26/2024] [Indexed: 02/13/2024]
Abstract
sTF (sialyl-Thomsen-Friedenreich) is a type of tumor-associated carbohydrate antigens (TACAs) and is highly expressed in various human malignancies. To validate if sTF could be a valuable molecular target for future cancer vaccine development, in this work the sTF antigen was prepared by adopting a strategy combining chemical and enzymatic methods, and then was covalently conjugated to a carrier protein, CRM197. The preliminary immunological evaluation, performed on BALB/c mice, revealed that the sTF-CRM197 conjugate elicited high titers of specific IgG antibodies. FACS experiments showed that the antisera induced by sTF-CRM197 conjugate could specifically recognize and bind to sTF-positive cancer cells T-47D. Furthermore, the conjugate mediated effective and specific antibody-mediated complement-dependent cytotoxicity (CDC).
Collapse
Affiliation(s)
- Weizhao Weng
- Department of Medicinal Chemistry, School of Pharmacy, Fudan University, 826 Zhangheng Road, Shanghai 201203, China
| | - Sumei Ren
- Research Center of Basic Medicine, Academy of Medical Sciences, Zhengzhou University, Zhengzhou 450001, Henan, China
| | - Changcai Teng
- School of Pharmaceutical Sciences, College of Marine Science, Hainan University, Haikou 570228, China
| | - Jia Guo
- Department of Medicinal Chemistry, School of Pharmacy, Fudan University, 826 Zhangheng Road, Shanghai 201203, China
| | - Qiuyu Guo
- Department of Medicinal Chemistry, School of Pharmacy, Fudan University, 826 Zhangheng Road, Shanghai 201203, China
| | - Wei Zhang
- Department of Medicinal Chemistry, School of Pharmacy, Fudan University, 826 Zhangheng Road, Shanghai 201203, China
| | - Chengli Zong
- School of Pharmaceutical Sciences, College of Marine Science, Hainan University, Haikou 570228, China.
| | - Ning Ding
- Department of Medicinal Chemistry, School of Pharmacy, Fudan University, 826 Zhangheng Road, Shanghai 201203, China.
| |
Collapse
|
9
|
Huynh CM, Mavliutova L, Sparrman T, Sellergren B, Irgum K. Elucidation of the Binding Orientation in α2,3- and α2,6-Linked Neu5Ac-Gal Epitopes toward a Hydrophilic Molecularly Imprinted Monolith. ACS OMEGA 2023; 8:44238-44249. [PMID: 38027366 PMCID: PMC10666243 DOI: 10.1021/acsomega.3c06836] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/08/2023] [Revised: 10/20/2023] [Accepted: 10/23/2023] [Indexed: 12/01/2023]
Abstract
N-Acetylneuraminic acid and its α2,3/α2,6-glycosidic linkages with galactose (Neu5Ac-Gal) are major carbohydrate antigen epitopes expressed in various pathological processes, such as cancer, influenza, and SARS-CoV-2. We here report a strategy for the synthesis and binding investigation of molecularly imprinted polymers (MIPs) toward α2,3 and α2,6 conformations of Neu5Ac-Gal antigens. Hydrophilic imprinted monoliths were synthesized from melamine monomer in the presence of four different templates, namely, N-acetylneuraminic acid (Neu5Ac), N-acetylneuraminic acid methyl ester (Neu5Ac-M), 3'-sialyllactose (3SL), and 6'-sialyllactose (6SL), in a tertiary solvent mixture at temperatures varying from -20 to +80 °C. The MIPs prepared at cryotemperatures showed a preferential affinity for the α2,6 linkage sequence of 6SL, with an imprinting factor of 2.21, whereas the α2,3 linkage sequence of 3SL resulted in nonspecific binding to the polymer scaffold. The preferable affinity for the α2,6 conformation of Neu5Ac-Gal was evident also when challenged by a mixture of other mono- and disaccharides in an aqueous test mixture. The use of saturation transfer difference nuclear magnetic resonance (STD-NMR) on suspensions of crushed monoliths allowed for directional interactions between the α2,3/α2,6 linkage sequences on their corresponding MIPs to be revealed. The Neu5Ac epitope, containing acetyl and polyalcohol moieties, was the major contributor to the sequence recognition for Neu5Ac(α2,6)Gal(β1,4)Glc, whereas contributions from the Gal and Glc segments were substantially lower.
Collapse
Affiliation(s)
- Chau Minh Huynh
- Department
of Chemistry, Umeå University, S-90187 Umeå, Sweden
| | - Liliia Mavliutova
- Department
of Biomedical Sciences, Faculty of Health and Society, Malmö University, SE-20506 Malmö, Sweden
| | - Tobias Sparrman
- Department
of Chemistry, Umeå University, S-90187 Umeå, Sweden
| | - Börje Sellergren
- Department
of Biomedical Sciences, Faculty of Health and Society, Malmö University, SE-20506 Malmö, Sweden
| | - Knut Irgum
- Department
of Chemistry, Umeå University, S-90187 Umeå, Sweden
| |
Collapse
|
10
|
Boelaars K, Goossens-Kruijssen L, Wang D, de Winde CM, Rodriguez E, Lindijer D, Springer B, van der Haar Àvila I, de Haas A, Wehry L, Boon L, Mebius RE, van Montfoort N, Wuhrer M, den Haan JMM, van Vliet SJ, van Kooyk Y. Unraveling the impact of sialic acids on the immune landscape and immunotherapy efficacy in pancreatic cancer. J Immunother Cancer 2023; 11:e007805. [PMID: 37940346 PMCID: PMC10632901 DOI: 10.1136/jitc-2023-007805] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/17/2023] [Indexed: 11/10/2023] Open
Abstract
BACKGROUND Pancreatic ductal adenocarcinoma (PDAC) is one of the deadliest cancers. Despite the successful application of immune checkpoint blockade in a range of human cancers, immunotherapy in PDAC remains unsuccessful. PDAC is characterized by a desmoplastic, hypoxic and highly immunosuppressive tumor microenvironment (TME), where T-cell infiltration is often lacking (immune desert), or where T cells are located distant from the tumor islands (immune excluded). Converting the TME to an immune-inflamed state, allowing T-cell infiltration, could increase the success of immunotherapy in PDAC. METHOD In this study, we use the KPC3 subcutaneous PDAC mouse model to investigate the role of tumor-derived sialic acids in shaping the tumor immune landscape. A sialic acid deficient KPC3 line was generated by genetic knock-out of the CMAS (cytidine monophosphate N-acetylneuraminic acid synthetase) enzyme, a critical enzyme in the synthesis of sialic acid-containing glycans. The effect of sialic acid-deficiency on immunotherapy efficacy was assessed by treatment with anti-programmed cell death protein 1 (PD-1) and agonistic CD40. RESULT The absence of sialic acids in KPC3 tumors resulted in increased numbers of CD4+ and CD8+ T cells in the TME, and reduced frequencies of CD4+ regulatory T cells (Tregs) within the T-cell population. Importantly, CD8+ T cells were able to infiltrate the tumor islands in sialic acid-deficient tumors. These favorable alterations in the immune landscape sensitized sialic acid-deficient tumors to immunotherapy, which was ineffective in sialic acid-expressing KPC3 tumors. In addition, high expression of sialylation-related genes in human pancreatic cancer correlated with decreased CD8+ T-cell infiltration, increased presence of Tregs, and poorer survival probability. CONCLUSION Our results demonstrate that tumor-derived sialic acids mediate T-cell exclusion within the PDAC TME, thereby impairing immunotherapy efficacy. Targeting sialic acids represents a potential strategy to enhance T-cell infiltration and improve immunotherapy outcomes in PDAC.
Collapse
Affiliation(s)
- Kelly Boelaars
- Molecular Cell Biology & Immunology, Amsterdam institute for Infection and Immunity, Cancer Center Amsterdam, Amsterdam UMC Location VUmc, Amsterdam, The Netherlands
| | - Laura Goossens-Kruijssen
- Molecular Cell Biology & Immunology, Amsterdam institute for Infection and Immunity, Cancer Center Amsterdam, Amsterdam UMC Location VUmc, Amsterdam, The Netherlands
| | - Di Wang
- Center for Proteomics and Metabolomics, Leiden University Medical Center, Leiden, The Netherlands
| | - Charlotte M de Winde
- Molecular Cell Biology & Immunology, Amsterdam institute for Infection and Immunity, Cancer Center Amsterdam, Amsterdam UMC Location VUmc, Amsterdam, The Netherlands
| | - Ernesto Rodriguez
- Molecular Cell Biology & Immunology, Amsterdam institute for Infection and Immunity, Cancer Center Amsterdam, Amsterdam UMC Location VUmc, Amsterdam, The Netherlands
| | - Dimitri Lindijer
- Molecular Cell Biology & Immunology, Amsterdam institute for Infection and Immunity, Cancer Center Amsterdam, Amsterdam UMC Location VUmc, Amsterdam, The Netherlands
| | - Babet Springer
- Molecular Cell Biology & Immunology, Amsterdam institute for Infection and Immunity, Cancer Center Amsterdam, Amsterdam UMC Location VUmc, Amsterdam, The Netherlands
| | - Irene van der Haar Àvila
- Molecular Cell Biology & Immunology, Amsterdam institute for Infection and Immunity, Cancer Center Amsterdam, Amsterdam UMC Location VUmc, Amsterdam, The Netherlands
| | - Aram de Haas
- Molecular Cell Biology & Immunology, Amsterdam institute for Infection and Immunity, Cancer Center Amsterdam, Amsterdam UMC Location VUmc, Amsterdam, The Netherlands
| | - Laetitia Wehry
- Molecular Cell Biology & Immunology, Amsterdam institute for Infection and Immunity, Cancer Center Amsterdam, Amsterdam UMC Location VUmc, Amsterdam, The Netherlands
| | | | - Reina E Mebius
- Molecular Cell Biology & Immunology, Amsterdam institute for Infection and Immunity, Cancer Center Amsterdam, Amsterdam UMC Location VUmc, Amsterdam, The Netherlands
| | - Nadine van Montfoort
- Department of Gastroenterology and Hepatology, Leiden University Medical Center, Leiden, The Netherlands
| | - Manfred Wuhrer
- Center for Proteomics and Metabolomics, Leiden University Medical Center, Leiden, The Netherlands
| | - Joke M M den Haan
- Molecular Cell Biology & Immunology, Amsterdam institute for Infection and Immunity, Cancer Center Amsterdam, Amsterdam UMC Location VUmc, Amsterdam, The Netherlands
| | - Sandra J van Vliet
- Molecular Cell Biology & Immunology, Amsterdam institute for Infection and Immunity, Cancer Center Amsterdam, Amsterdam UMC Location VUmc, Amsterdam, The Netherlands
| | - Yvette van Kooyk
- Molecular Cell Biology & Immunology, Amsterdam institute for Infection and Immunity, Cancer Center Amsterdam, Amsterdam UMC Location VUmc, Amsterdam, The Netherlands
| |
Collapse
|
11
|
Radziejewska I, Supruniuk K, Jakimiuk K, Tomczyk M, Bielawska A, Galicka A. Tiliroside Combined with Anti-MUC1 Monoclonal Antibody as Promising Anti-Cancer Strategy in AGS Cancer Cells. Int J Mol Sci 2023; 24:13036. [PMID: 37685842 PMCID: PMC10487805 DOI: 10.3390/ijms241713036] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2023] [Revised: 07/27/2023] [Accepted: 08/17/2023] [Indexed: 09/10/2023] Open
Abstract
Specific changes in mucin-type O-glycosylation are common for many cancers, including gastric ones. The most typical alterations include incomplete synthesis of glycan structures, enhanced expression of truncated O-glycans (Tn, T antigens and their sialylated forms), and overexpression of fucosylation. Such altered glycans influence many cellular activities promoting cancer development. Tiliroside is a glycosidic dietary flavonoid with pharmacological properties, including anti-cancer. In this study, we aim to assess the effect of the combined action of anti-MUC1 and tiliroside on some cancer-related factors in AGS gastric cancer cells. Cancer cells were treated with 40, 80, and 160 µM tiliroside, 5 µg/mL anti-MUC1, and flavonoid together with mAb. Real-Time PCR, ELISA, and Western blotting were applied to examine MUC1 expression, specific, tumor-associated antigens, enzymes taking part in their formation, Gal-3, Akt, and NF-κB. MUC1 expression was significantly reduced by mAb action. The combined action of anti-MUC1 and tiliroside was more effective in comparison with monotherapy in the case of C1GalT1, ST3GalT1, FUT4, Gal-3, NF-κB, Akt mRNAs, and Tn antigen, as well as sialyl T antigen expression. The results of our study indicate that applied combined therapy may be a promising anti-gastric cancer strategy.
Collapse
Affiliation(s)
- Iwona Radziejewska
- Department of Medical Chemistry, Medical University of Białystok, ul. Mickiewicza 2a, 15-222 Białystok, Poland;
| | - Katarzyna Supruniuk
- Department of Medical Biology and Genetics, Medical University of Gdańsk, ul. Dębinki 1, 80-211 Gdańsk, Poland;
| | - Katarzyna Jakimiuk
- Department of Pharmacognosy, Faculty of Pharmacy with the Division of Laboratory Medicine, Medical University of Białystok, ul. Mickiewicza 2a, 15-230 Białystok, Poland; (K.J.); (M.T.)
| | - Michał Tomczyk
- Department of Pharmacognosy, Faculty of Pharmacy with the Division of Laboratory Medicine, Medical University of Białystok, ul. Mickiewicza 2a, 15-230 Białystok, Poland; (K.J.); (M.T.)
| | - Anna Bielawska
- Department of Biotechnology, Medical University of Białystok, ul. Kilińskiego 1, 15-089 Białystok, Poland;
| | - Anna Galicka
- Department of Medical Chemistry, Medical University of Białystok, ul. Mickiewicza 2a, 15-222 Białystok, Poland;
| |
Collapse
|
12
|
Zhou X, Chi K, Zhang C, Liu Q, Yang G. Sialylation: A Cloak for Tumors to Trick the Immune System in the Microenvironment. BIOLOGY 2023; 12:832. [PMID: 37372117 DOI: 10.3390/biology12060832] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/10/2023] [Revised: 06/03/2023] [Accepted: 06/06/2023] [Indexed: 06/29/2023]
Abstract
The tumor microenvironment (TME), where the tumor cells incite the surrounding normal cells to create an immune suppressive environment, reduces the effectiveness of immune responses during cancer development. Sialylation, a type of glycosylation that occurs on cell surface proteins, lipids, and glycoRNAs, is known to accumulate in tumors and acts as a "cloak" to help tumor cells evade immunological surveillance. In the last few years, the role of sialylation in tumor proliferation and metastasis has become increasingly evident. With the advent of single-cell and spatial sequencing technologies, more research is being conducted to understand the effects of sialylation on immunity regulation. This review provides updated insights into recent research on the function of sialylation in tumor biology and summarizes the latest developments in sialylation-targeted tumor therapeutics, including antibody-mediated and metabolic-based sialylation inhibition, as well as interference with sialic acid-Siglec interaction.
Collapse
Affiliation(s)
- Xiaoman Zhou
- The Key Laboratory of Carbohydrate Chemistry and Biotechnology, Ministry of Education, School of Biotechnology, Jiangnan University, Wuxi 214122, China
| | - Kaijun Chi
- The Key Laboratory of Carbohydrate Chemistry and Biotechnology, Ministry of Education, School of Biotechnology, Jiangnan University, Wuxi 214122, China
| | - Chairui Zhang
- The Key Laboratory of Carbohydrate Chemistry and Biotechnology, Ministry of Education, School of Biotechnology, Jiangnan University, Wuxi 214122, China
| | - Quan Liu
- Department of Medical Oncology, Affiliated Hospital of Jiangnan University, Wuxi 214122, China
| | - Ganglong Yang
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing 100190, China
| |
Collapse
|
13
|
Radziejewska I. Galectin-3 and Epithelial MUC1 Mucin-Interactions Supporting Cancer Development. Cancers (Basel) 2023; 15:2680. [PMID: 37345016 DOI: 10.3390/cancers15102680] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2023] [Revised: 05/04/2023] [Accepted: 05/05/2023] [Indexed: 06/23/2023] Open
Abstract
Aberrant glycosylation of cell surface proteins is a very common feature of many cancers. One of the glycoproteins, which undergoes specific alterations in the glycosylation of tumor cells is epithelial MUC1 mucin, which is highly overexpressed in the malignant state. Such changes lead to the appearance of tumor associated carbohydrate antigens (TACAs) on MUC1, which are rarely seen in healthy cells. One of these structures is the Thomsen-Friedenreich disaccharide Galβ1-3GalNAc (T or TF antigen), which is typical for about 90% of cancers. It was revealed that increased expression of the T antigen has a big impact on promoting cancer progression and metastasis, among others, due to the interaction of this antigen with the β-galactose binding protein galectin-3 (Gal-3). In this review, we summarize current information about the interactions between the T antigen on MUC1 mucin and Gal-3, and their impact on cancer progression and metastasis.
Collapse
Affiliation(s)
- Iwona Radziejewska
- Department of Medical Chemistry, Medical University of Białystok, ul. Mickiewicza 2a, 15-222 Białystok, Poland
| |
Collapse
|
14
|
Punetha A, Kotiya D. Advancements in Oncoproteomics Technologies: Treading toward Translation into Clinical Practice. Proteomes 2023; 11:2. [PMID: 36648960 PMCID: PMC9844371 DOI: 10.3390/proteomes11010002] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2022] [Revised: 01/03/2023] [Accepted: 01/04/2023] [Indexed: 01/12/2023] Open
Abstract
Proteomics continues to forge significant strides in the discovery of essential biological processes, uncovering valuable information on the identity, global protein abundance, protein modifications, proteoform levels, and signal transduction pathways. Cancer is a complicated and heterogeneous disease, and the onset and progression involve multiple dysregulated proteoforms and their downstream signaling pathways. These are modulated by various factors such as molecular, genetic, tissue, cellular, ethnic/racial, socioeconomic status, environmental, and demographic differences that vary with time. The knowledge of cancer has improved the treatment and clinical management; however, the survival rates have not increased significantly, and cancer remains a major cause of mortality. Oncoproteomics studies help to develop and validate proteomics technologies for routine application in clinical laboratories for (1) diagnostic and prognostic categorization of cancer, (2) real-time monitoring of treatment, (3) assessing drug efficacy and toxicity, (4) therapeutic modulations based on the changes with prognosis and drug resistance, and (5) personalized medication. Investigation of tumor-specific proteomic profiles in conjunction with healthy controls provides crucial information in mechanistic studies on tumorigenesis, metastasis, and drug resistance. This review provides an overview of proteomics technologies that assist the discovery of novel drug targets, biomarkers for early detection, surveillance, prognosis, drug monitoring, and tailoring therapy to the cancer patient. The information gained from such technologies has drastically improved cancer research. We further provide exemplars from recent oncoproteomics applications in the discovery of biomarkers in various cancers, drug discovery, and clinical treatment. Overall, the future of oncoproteomics holds enormous potential for translating technologies from the bench to the bedside.
Collapse
Affiliation(s)
- Ankita Punetha
- Department of Microbiology, Biochemistry and Molecular Genetics, Rutgers New Jersey Medical School, Rutgers University, 225 Warren St., Newark, NJ 07103, USA
| | - Deepak Kotiya
- Department of Pharmacology and Nutritional Sciences, University of Kentucky, 900 South Limestone St., Lexington, KY 40536, USA
| |
Collapse
|
15
|
Harvey DJ. Analysis of carbohydrates and glycoconjugates by matrix-assisted laser desorption/ionization mass spectrometry: An update for 2019-2020. MASS SPECTROMETRY REVIEWS 2022:e21806. [PMID: 36468275 DOI: 10.1002/mas.21806] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/17/2023]
Abstract
This review is the tenth update of the original article published in 1999 on the application of matrix-assisted laser desorption/ionization (MALDI) mass spectrometry to the analysis of carbohydrates and glycoconjugates and brings coverage of the literature to the end of 2020. Also included are papers that describe methods appropriate to analysis by MALDI, such as sample preparation techniques, even though the ionization method is not MALDI. The review is basically divided into three sections: (1) general aspects such as theory of the MALDI process, matrices, derivatization, MALDI imaging, fragmentation, quantification and the use of arrays. (2) Applications to various structural types such as oligo- and polysaccharides, glycoproteins, glycolipids, glycosides and biopharmaceuticals, and (3) other areas such as medicine, industrial processes and glycan synthesis where MALDI is extensively used. Much of the material relating to applications is presented in tabular form. The reported work shows increasing use of incorporation of new techniques such as ion mobility and the enormous impact that MALDI imaging is having. MALDI, although invented nearly 40 years ago is still an ideal technique for carbohydrate analysis and advancements in the technique and range of applications show little sign of diminishing.
Collapse
Affiliation(s)
- David J Harvey
- Nuffield Department of Medicine, Target Discovery Institute, University of Oxford, Oxford, UK
- Department of Chemistry, University of Oxford, Oxford, Oxfordshire, United Kingdom
| |
Collapse
|
16
|
A roadmap for translational cancer glycoimmunology at single cell resolution. J Exp Clin Cancer Res 2022; 41:143. [PMID: 35428302 PMCID: PMC9013178 DOI: 10.1186/s13046-022-02335-z] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2022] [Accepted: 03/17/2022] [Indexed: 11/11/2022] Open
Abstract
Cancer cells can evade immune responses by exploiting inhibitory immune checkpoints. Immune checkpoint inhibitor (ICI) therapies based on anti-CTLA-4 and anti-PD-1/PD-L1 antibodies have been extensively explored over the recent years to unleash otherwise compromised anti-cancer immune responses. However, it is also well established that immune suppression is a multifactorial process involving an intricate crosstalk between cancer cells and the immune systems. The cancer glycome is emerging as a relevant source of immune checkpoints governing immunosuppressive behaviour in immune cells, paving an avenue for novel immunotherapeutic options. This review addresses the current state-of-the-art concerning the role played by glycans controlling innate and adaptive immune responses, while shedding light on available experimental models for glycoimmunology. We also emphasize the tremendous progress observed in the development of humanized models for immunology, the paramount contribution of advances in high-throughput single-cell analysis in this context, and the importance of including predictive machine learning algorithms in translational research. This may constitute an important roadmap for glycoimmunology, supporting careful adoption of models foreseeing clinical translation of fundamental glycobiology knowledge towards next generation immunotherapies.
Collapse
|
17
|
Cheng L, Yang F, Tang L, Qian L, Chen X, Guan F, Zhang J, Li G. Electrochemical Evaluation of Tumor Development via Cellular Interface Supported CRISPR/Cas Trans-Cleavage. RESEARCH 2022; 2022:9826484. [PMID: 35474904 PMCID: PMC9011167 DOI: 10.34133/2022/9826484] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/07/2021] [Accepted: 03/14/2022] [Indexed: 11/06/2022]
Abstract
Evaluating tumor development is of great importance for clinic treatment and therapy. It has been known that the amounts of sialic acids on tumor cell membrane surface are closely associated with the degree of cancerization of the cell. So, in this work, cellular interface supported CRISPR/Cas trans-cleavage has been explored for electrochemical simultaneous detection of two types of sialic acids, i.e., N-glycolylneuraminic acid (Neu5Gc) and N-acetylneuraminic acid (Neu5Ac). Specifically, PbS quantum dot-labeled DNA modified by Neu5Gc antibody is prepared to specifically recognize Neu5Gc on the cell surface, followed by the binding of Neu5Ac through our fabricated CdS quantum dot-labeled DNA modified by Sambucus nigra agglutinin. Subsequently, the activated Cas12a indiscriminately cleaves DNA, resulting in the release of PbS and CdS quantum dots, both of which can be simultaneously detected by anodic stripping voltammetry. Consequently, Neu5Gc and Neu5Ac on cell surface can be quantitatively analyzed with the lowest detection limits of 1.12 cells/mL and 1.25 cells/mL, respectively. Therefore, a ratiometric electrochemical method can be constructed for kinetic study of the expression and hydrolysis of Neu5Gc and Neu5Ac on cell surface, which can be further used as a tool to identify bladder cancer cells at different development stages. Our method to evaluate tumor development is simple and easy to be operated, so it can be potentially applied for the detection of tumor occurrence and development in the future.
Collapse
Affiliation(s)
- Liangfen Cheng
- Center for Molecular Recognition and Biosensing, School of Life Sciences, Shanghai University, Shanghai 200444, China
| | - Fuhan Yang
- Department of Urology, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai 200092, China
| | - Longfei Tang
- Center for Molecular Recognition and Biosensing, School of Life Sciences, Shanghai University, Shanghai 200444, China
| | - Lelin Qian
- Center for Molecular Recognition and Biosensing, School of Life Sciences, Shanghai University, Shanghai 200444, China
| | - Xu Chen
- Center for Molecular Recognition and Biosensing, School of Life Sciences, Shanghai University, Shanghai 200444, China
| | - Feng Guan
- College of Life Science, Northwest University, Xi’an 710127, China
| | - Juan Zhang
- Center for Molecular Recognition and Biosensing, School of Life Sciences, Shanghai University, Shanghai 200444, China
| | - Genxi Li
- Center for Molecular Recognition and Biosensing, School of Life Sciences, Shanghai University, Shanghai 200444, China
- State Key Laboratory of Pharmaceutical Biotechnology and Collaborative Innovation Center of Chemistry for Life Sciences, Department of Biochemistry, Nanjing University, Nanjing 210093, China
| |
Collapse
|
18
|
Berghuis AY, Pijnenborg JFA, Boltje TJ, Pijnenborg JMA. Sialic acids in gynecological cancer development and progression: Impact on diagnosis and treatment. Int J Cancer 2021; 150:678-687. [PMID: 34741527 PMCID: PMC9299683 DOI: 10.1002/ijc.33866] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2021] [Revised: 10/17/2021] [Accepted: 10/18/2021] [Indexed: 11/19/2022]
Abstract
Gynecological cancers are in the top 10 of most common cancers in women. Survival and outcome are strongly related to the stage at diagnosis. Therefore, early diagnosis is essential in reducing morbidity and mortality. The high mortality rate of gynecological cancers can mainly be attributed to ovarian cancer (OC). OC is commonly diagnosed at an advanced stage due to a lack of proper screening tools allowing early detection. Endometrial cancer (EC) on the contrary, is mostly diagnosed at an early stage and has, in general, better outcomes. The incidence of nonendometrioid EC has increased in the last decade, displaying a shared tumor biology with OC and consequently significantly worse outcome. New approaches allowing detection of gynecological cancers in an early stage are therefore desired. Recent studies on cancer biology have shown the relevance of altered glycosylation in the occurrence and progression of cancer. The aberrant expression of sialic acid, a specific carbohydrate terminating glycoproteins and glycolipids on the cell‐surface, is frequently correlated with malignancy. We aimed to determine the current understanding of sialic acid function in different gynecological cancers to identify the gaps in knowledge and its potential use for new diagnostic and therapeutic avenues. Therefore we performed a review on current literature focusing on studies where sialylation was linked to gynecological cancers. The identified studies showed elevated levels of sialic acid in serum, tissue and sialylated antigens in most patients with gynecological cancers, underlining its potential for diagnosis.
What's new?
Recent studies have shown the relevance of altered glycosylation in the occurrence and progression of cancer. In this review, the authors found elevated levels of sialic acid in serum and tissue and high levels of sialylated antigens in most patients with gynaecological cancers, underlining the potential of sialic acid for diagnosis. Elevated levels of sialylation were related with tumour growth, poor differentiation, inhibition of apoptosis, and chemoresistance. Taken together, the studies suggest that sialylation levels could be used to discriminate healthy and benign samples from cancer samples and even early and advanced stages in ovarian, cervical, and endometrial cancer.
Collapse
Affiliation(s)
- Anna Y Berghuis
- Faculty of Science, Radboud University, Nijmegen, The Netherlands
| | - Johan F A Pijnenborg
- Cluster for Molecular Chemistry, Institute for Molecules and Materials, Radboud University, Nijmegen, The Netherlands
| | - Thomas J Boltje
- Cluster for Molecular Chemistry, Institute for Molecules and Materials, Radboud University, Nijmegen, The Netherlands
| | - Johanna M A Pijnenborg
- Department of Obstetrics and Gynecology, Radboud University Medical Center, Nijmegen, The Netherlands.,Radboud Institute of Health Science, Nijmegen, The Netherlands
| |
Collapse
|
19
|
Glycosylation: Rising Potential for Prostate Cancer Evaluation. Cancers (Basel) 2021; 13:cancers13153726. [PMID: 34359624 PMCID: PMC8345048 DOI: 10.3390/cancers13153726] [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: 06/25/2021] [Revised: 07/21/2021] [Accepted: 07/21/2021] [Indexed: 12/15/2022] Open
Abstract
Simple Summary Aberrant protein glycosylation is a well-known hallmark of cancer and is associated with differential expression of enzymes such as glycosyltransferases and glycosidases. The altered expression of the enzymes triggers cancer cells to produce glycoproteins with specific cancer-related aberrations in glycan structures. Increasing number of data indicate that glycosylation patterns of PSA and other prostate-originated proteins exert a potential to distinguish between benign prostate disease and cancer as well as among different stages of prostate cancer development and aggressiveness. This review summarizes the alterations in glycan sialylation, fucosylation, truncated O-glycans, and LacdiNAc groups outlining their potential applications in non-invasive diagnostic procedures of prostate diseases. Further research is desired to develop more general algorithms exploiting glycobiology data for the improvement of prostate diseases evaluation. Abstract Prostate cancer is the second most commonly diagnosed cancer among men. Alterations in protein glycosylation are confirmed to be a reliable hallmark of cancer. Prostate-specific antigen is the biomarker that is used most frequently for prostate cancer detection, although its lack of sensitivity and specificity results in many unnecessary biopsies. A wide range of glycosylation alterations in prostate cancer cells, including increased sialylation and fucosylation, can modify protein function and play a crucial role in many important biological processes in cancer, including cell signalling, adhesion, migration, and cellular metabolism. In this review, we summarize studies evaluating the prostate cancer associated glycosylation related alterations in sialylation, mainly α2,3-sialylation, core fucosylation, branched N-glycans, LacdiNAc group and presence of truncated O-glycans (sTn, sT antigen). Finally, we discuss the great potential to make use of glycans as diagnostic and prognostic biomarkers for prostate cancer.
Collapse
|
20
|
Radziejewska I, Supruniuk K, Bielawska A. Anti-cancer effect of combined action of anti-MUC1 and rosmarinic acid in AGS gastric cancer cells. Eur J Pharmacol 2021; 902:174119. [PMID: 33930385 DOI: 10.1016/j.ejphar.2021.174119] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2021] [Revised: 04/16/2021] [Accepted: 04/20/2021] [Indexed: 12/15/2022]
Abstract
MUC1 seems to be promising target in cancer cells due to its abundant and specifically altered expression as well as differential distribution pattern relative to normal tissues. Rosmarinic acid (RA) is a natural, polyphenolic compound with pharmacological activities, including anti-cancer. Herein, we aim to explore the effect of combined action of anti-MUC1 and RA on selected cancer-related factors in AGS gastric cancer cells. Cancer cells were treated with 100, 200 μM rosmarinic acid, 5 μg/ml anti-MUC1 and acid together with antibody. Western blotting, ELISA and RT-PCR were used to assess the expression of MUC1, selected sugar antigens, enzymes participating in protein glycosylation, Gal-3, p53, pro- and anti-apoptotic factors, and caspases-3,-8,-9 in cancer cells. MUC1 mRNA was significantly suppressed by combined action of anti-MUC1 and RA. Such treatment markedly inhibited expression of cancer-related Tn, T, sialyl Tn, sialyl T, and fucosylated sugar antigens as well as mRNA expression of enzymes participating in their formation: ppGalNAcT2, C1GalT1, ST6GalNAcT2, ST3GalT1 and FUT4. C1GalT1 was suppressed also on protein level. Gal-3, factor likely participating in metastasis, was significantly suppressed on mRNA level by RA administrated with anti-MUC1. Pro-apoptotic Bax protein and Bad mRNA were significantly induced, and anti-apoptotic Bcl-2 mRNA expression was inhibited by such treatment. Combined action of mAb and RA markedly increased caspase-9 mRNA expression. Results of the study indicate that combined action of anti-MUC1 and RA is more effective than monotherapy in relation to examined cancer related factors. Such treatment can be considered as new, promising strategy in gastric cancer therapy.
Collapse
Affiliation(s)
- Iwona Radziejewska
- Department of Medical Chemistry, Medical University of Białystok, ul. Mickiewicza 2a, 15-222, Białystok, Poland.
| | - Katarzyna Supruniuk
- Department of Medical Chemistry, Medical University of Białystok, ul. Mickiewicza 2a, 15-222, Białystok, Poland
| | - Anna Bielawska
- Department of Biotechnology, Medical University of Białystok, ul. Kilińskiego 1, 15-089, Białystok, Poland
| |
Collapse
|
21
|
Brockhausen I, Melamed J. Mucins as anti-cancer targets: perspectives of the glycobiologist. Glycoconj J 2021; 38:459-474. [PMID: 33704667 DOI: 10.1007/s10719-021-09986-8] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2021] [Revised: 02/22/2021] [Accepted: 02/26/2021] [Indexed: 12/11/2022]
Abstract
Mucins are highly O-glycosylated glycoproteins that carry a heterogenous variety of O-glycan structures. Tumor cells tend to overexpress specific mucins, such as the cell surface mucins MUC1 and MUC4 that are engaged in signaling and cell growth, and exhibit abnormal glycosylation. In particular, the Tn and T antigens and their sialylated forms are common in cancer mucins. We review herein methods chosen to use cancer-associated glycans and mucins as targets for the design of anti-cancer immunotherapies. Mucin peptides from the glycosylated and transmembrane domains have been combined with immune-stimulating adjuvants in a wide variety of approaches to produce anti-tumor antibodies and vaccines. These mucin conjugates have been tested on cancer cells in vitro and in mice with significant successes in stimulating anti-tumor responses. The clinical trials in humans, however, have shown limited success in extending survival. It seems critical that the individual-specific epitope expression of cancer mucins is considered in future therapies to result in lasting anti-tumor responses.
Collapse
Affiliation(s)
- Inka Brockhausen
- Biomedical and Molecular Sciences, Queen's University, 18 Stuart St, Kingston, ON, K7L 3N6, Canada.
| | - Jacob Melamed
- Biomedical and Molecular Sciences, Queen's University, 18 Stuart St, Kingston, ON, K7L 3N6, Canada
| |
Collapse
|
22
|
Samaržija I. Post-Translational Modifications That Drive Prostate Cancer Progression. Biomolecules 2021; 11:247. [PMID: 33572160 PMCID: PMC7915076 DOI: 10.3390/biom11020247] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2021] [Revised: 02/04/2021] [Accepted: 02/06/2021] [Indexed: 02/07/2023] Open
Abstract
While a protein primary structure is determined by genetic code, its specific functional form is mostly achieved in a dynamic interplay that includes actions of many enzymes involved in post-translational modifications. This versatile repertoire is widely used by cells to direct their response to external stimuli, regulate transcription and protein localization and to keep proteostasis. Herein, post-translational modifications with evident potency to drive prostate cancer are explored. A comprehensive list of proteome-wide and single protein post-translational modifications and their involvement in phenotypic outcomes is presented. Specifically, the data on phosphorylation, glycosylation, ubiquitination, SUMOylation, acetylation, and lipidation in prostate cancer and the enzymes involved are collected. This type of knowledge is especially valuable in cases when cancer cells do not differ in the expression or mutational status of a protein, but its differential activity is regulated on the level of post-translational modifications. Since their driving roles in prostate cancer, post-translational modifications are widely studied in attempts to advance prostate cancer treatment. Current strategies that exploit the potential of post-translational modifications in prostate cancer therapy are presented.
Collapse
Affiliation(s)
- Ivana Samaržija
- Laboratory for Epigenomics, Division of Molecular Medicine, Ruđer Bošković Institute, 10000 Zagreb, Croatia
| |
Collapse
|