1
|
Habeeb IF, Alao TE, Delgado D, Buffone A. When a negative (charge) is not a positive: sialylation and its role in cancer mechanics and progression. Front Oncol 2024; 14:1487306. [PMID: 39628991 PMCID: PMC11611868 DOI: 10.3389/fonc.2024.1487306] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2024] [Accepted: 10/10/2024] [Indexed: 12/06/2024] Open
Abstract
Sialic acids and sialoglycans are critical actors in cancer progression and metastasis. These terminal sugar residues on glycoproteins and glycolipids modulate key cellular processes such as immune evasion, cell adhesion, and migration. Aberrant sialylation is driven by overexpression of sialyltransferases, resulting in hypersialylation on cancer cell surfaces as well as enhancing tumor aggressiveness. Sialylated glycans alter the structure of the glycocalyx, a protective barrier that fosters cancer cell detachment, migration, and invasion. This bulky glycocalyx also increases membrane tension, promoting integrin clustering and downstream signaling pathways that drive cell proliferation and metastasis. They play a critical role in immune evasion by binding to Siglecs, inhibitory receptors on immune cells, which transmit signals that protect cancer cells from immune-mediated destruction. Targeting sialylation pathways presents a promising therapeutic opportunity to understand the complex roles of sialic acids and sialoglycans in cancer mechanics and progression, which is crucial for developing novel diagnostic and therapeutic strategies that can disrupt these processes and improve cancer treatment outcomes.
Collapse
Affiliation(s)
- Issa Funsho Habeeb
- Department of Biomedical Engineering, New Jersey Institute of Technlogy, Newark, NJ, United States
| | - Toheeb Eniola Alao
- Department of Biomedical Engineering, New Jersey Institute of Technlogy, Newark, NJ, United States
| | - Daniella Delgado
- Department of Biomedical Engineering, New Jersey Institute of Technlogy, Newark, NJ, United States
| | - Alexander Buffone
- Department of Biomedical Engineering, New Jersey Institute of Technlogy, Newark, NJ, United States
- Chemical and Materials Engineering, New Jersey Institute of Technlogy, Newark, NJ, United States
| |
Collapse
|
2
|
Zhang SZ, Lobo A, Li PF, Zhang YF. Sialylated glycoproteins and sialyltransferases in digestive cancers: Mechanisms, diagnostic biomarkers, and therapeutic targets. Crit Rev Oncol Hematol 2024; 197:104330. [PMID: 38556071 DOI: 10.1016/j.critrevonc.2024.104330] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2023] [Revised: 03/12/2024] [Accepted: 03/19/2024] [Indexed: 04/02/2024] Open
Abstract
Sialic acid (SA), as the ultimate epitope of polysaccharides, can act as a cap at the end of polysaccharide chains to prevent their overextension. Sialylation is the enzymatic process of transferring SA residues onto polysaccharides and is catalyzed by a group of enzymes known as sialyltransferases (SiaTs). It is noteworthy that the sialylation level of glycoproteins is significantly altered when digestive cancer occurs. And this alteration exhibits a close correlation with the progression of these cancers. In this review, from the perspective of altered SiaTs expression levels and changed glycoprotein sialylation patterns, we summarize the pathogenesis of gastric cancer (GC), colorectal cancer (CRC), pancreatic ductal adenocarcinoma (PDAC), and hepatocellular carcinoma (HCC). Furthermore, we propose potential early diagnostic biomarkers and prognostic indicators for different digestive cancers. Finally, we summarize the therapeutic value of sialylation in digestive system cancers.
Collapse
Affiliation(s)
- Shao-Ze Zhang
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Qingdao 266021, China
| | - Amara Lobo
- Department of Critical Care Medicine Holy Family Hospital, St Andrew's Road, Bandra (West), Mumbai 400050, India
| | - Pei-Feng Li
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Qingdao 266021, China.
| | - Yin-Feng Zhang
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Qingdao 266021, China.
| |
Collapse
|
3
|
An SY, Kim KS, Lee YC, Kim SH. Transcription of human β-galactoside α2,6-sialyltransferase (hST6Gal I) is downregulated by curcumin through AMPK signaling in human colon carcinoma HCT116 cells. Genes Genomics 2023; 45:901-909. [PMID: 37231294 DOI: 10.1007/s13258-023-01398-2] [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: 01/12/2023] [Accepted: 05/08/2023] [Indexed: 05/27/2023]
Abstract
BACKGROUND In this study, we observed that in human colon carcinoma HCT116 cells mRNA level of the human β-galactoside α2,6-sialyltransferase (hST6Gal I) was decreased by curcumin. FACS analysis using the α2,6-sialyl-specific lectin (SNA) also showed a noticeable decrease in binding to SNA by curcumin. OBJECTIVE To investigate the mechanism for curcumin-triggered downregulation of hST6Gal I transcription. METHODS The mRNA levels of nine kinds of hST genes were assessed by RT-PCR after curcumin was treated in HCT116 cells. The level of hST6Gal I product on cell surface was examined by flow cytometry analysis. Luciferase reporter plasmids with 5'-deleted constructs and mutants of the hST6Gal I promoter were transiently transfected into HCT116 cells, and the luciferase activity was measured after treatment with curcumin. RESULTS Curcumin led to significant transcriptional repression of the hST6Gal I promoter. Promoter analysis using deletion mutants proved that the - 303 to - 189 region of the hST6Gal I promoter is required for transcriptional repression in response to curcumin. Among putative binding sites for transcription factors IK2, GATA1, TCF12, TAL1/E2A, SPT, and SL1 in this region, by site-directed mutagenesis analysis the TAL/E2A binding site (nucleotides - 266/- 246) was proved to be crucial for curcumin-triggered downregulation of hST6Gal I transcription in HCT116 cells. The transcription activity of hST6Gal I gene in HCT116 cells was markedly suppressed by compound C, an AMP-activated protein kinase (AMPK) inhibitor. CONCLUSION These indicate that gene expression of hST6Gal I in HCT116 cells is controlled through AMPK/TAL/E2A signal pathway.
Collapse
Affiliation(s)
- So-Young An
- Department of Medicinal Biotechnology, College of Health Sciences, Dong-A University, Busan, 49315, South Korea
| | - Kyoung-Sook Kim
- Department of Medicinal Biotechnology, College of Health Sciences, Dong-A University, Busan, 49315, South Korea
| | - Young-Choon Lee
- Department of Medicinal Biotechnology, College of Health Sciences, Dong-A University, Busan, 49315, South Korea.
| | - Seok-Ho Kim
- Department of Medicinal Biotechnology, College of Health Sciences, Dong-A University, Busan, 49315, South Korea.
| |
Collapse
|
4
|
Boyaval F, Dalebout H, Van Zeijl R, Wang W, Fariña-Sarasqueta A, Lageveen-Kammeijer GSM, Boonstra JJ, McDonnell LA, Wuhrer M, Morreau H, Heijs B. High-Mannose N-Glycans as Malignant Progression Markers in Early-Stage Colorectal Cancer. Cancers (Basel) 2022; 14:1552. [PMID: 35326703 PMCID: PMC8945895 DOI: 10.3390/cancers14061552] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2022] [Revised: 03/09/2022] [Accepted: 03/16/2022] [Indexed: 02/05/2023] Open
Abstract
The increase incidence of early colorectal cancer (T1 CRC) last years is mainly due to the introduction of population-based screening for CRC. T1 CRC staging based on histological criteria remains challenging and there is high variability among pathologists in the scoring of these criteria. It is crucial to unravel the biology behind the progression of adenoma into T1 CRC. Glycomic studies have reported extensively on alterations of the N-glycomic pattern in CRC; therefore, investigating these alterations may reveal new insights into the development of T1 CRC. We used matrix-assisted laser desorption ionization (MALDI) mass spectrometry imaging (MSI) to spatially profile the N-glycan species in a cohort of pT1 CRC using archival formalin-fixed and paraffin-embedded (FFPE) material. To generate structural information on the observed N-glycans, CE-ESI-MS/MS was used in conjunction with MALDI-MSI. Relative intensities and glycosylation traits were calculated based on a panel of 58 N-glycans. Our analysis showed pronounced differences between normal epithelium, dysplastic, and carcinoma regions. High-mannose-type N-glycans were higher in the dysplastic region than in carcinoma, which correlates to increased proliferation of the cells. We observed changes in the cancer invasive front, including higher expression of α2,3-linked sialic acids which followed the glycosylation pattern of the carcinoma region.
Collapse
Affiliation(s)
- Fanny Boyaval
- Department of Pathology, Leiden University Medical Center, Albinusdreef, 2333 ZA Leiden, The Netherlands;
- Center for Proteomics & Metabolomics, Leiden University Medical Center, Albinusdreef, 2333 ZA Leiden, The Netherlands; (H.D.); (R.V.Z.); (W.W.); (G.S.M.L.-K.); (M.W.)
| | - Hans Dalebout
- Center for Proteomics & Metabolomics, Leiden University Medical Center, Albinusdreef, 2333 ZA Leiden, The Netherlands; (H.D.); (R.V.Z.); (W.W.); (G.S.M.L.-K.); (M.W.)
| | - René Van Zeijl
- Center for Proteomics & Metabolomics, Leiden University Medical Center, Albinusdreef, 2333 ZA Leiden, The Netherlands; (H.D.); (R.V.Z.); (W.W.); (G.S.M.L.-K.); (M.W.)
| | - Wenjun Wang
- Center for Proteomics & Metabolomics, Leiden University Medical Center, Albinusdreef, 2333 ZA Leiden, The Netherlands; (H.D.); (R.V.Z.); (W.W.); (G.S.M.L.-K.); (M.W.)
| | - Arantza Fariña-Sarasqueta
- Department of Pathology, Cancer Center Amsterdam, Amsterdam University Medical Centers, University of Amsterdam, Meibergdreef 9, 1105 AZ Amsterdam, The Netherlands;
| | - Guinevere S. M. Lageveen-Kammeijer
- Center for Proteomics & Metabolomics, Leiden University Medical Center, Albinusdreef, 2333 ZA Leiden, The Netherlands; (H.D.); (R.V.Z.); (W.W.); (G.S.M.L.-K.); (M.W.)
| | - Jurjen J. Boonstra
- Department of Gastroenterology and Hepatology, Leiden University Medical Centre, 2300 RC Leiden, The Netherlands;
| | - Liam A. McDonnell
- Fondazione Pisana per la Scienza ONLUS, Via Ferruccio Giovannini, 56017 San Giuliano Terme, Italy;
| | - Manfred Wuhrer
- Center for Proteomics & Metabolomics, Leiden University Medical Center, Albinusdreef, 2333 ZA Leiden, The Netherlands; (H.D.); (R.V.Z.); (W.W.); (G.S.M.L.-K.); (M.W.)
| | - Hans Morreau
- Department of Pathology, Leiden University Medical Center, Albinusdreef, 2333 ZA Leiden, The Netherlands;
| | - Bram Heijs
- Center for Proteomics & Metabolomics, Leiden University Medical Center, Albinusdreef, 2333 ZA Leiden, The Netherlands; (H.D.); (R.V.Z.); (W.W.); (G.S.M.L.-K.); (M.W.)
| |
Collapse
|
5
|
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: 66] [Impact Index Per Article: 16.5] [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
|
6
|
Punch PR, Irons EE, Manhardt CT, Marathe H, Lau JTY. The sialyltransferase ST6GAL1 protects against radiation-induced gastrointestinal damage. Glycobiology 2021; 30:446-453. [PMID: 31897489 DOI: 10.1093/glycob/cwz108] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2019] [Revised: 12/19/2019] [Accepted: 12/20/2019] [Indexed: 12/14/2022] Open
Abstract
High-dose irradiation poses extreme risk of mortality from acute damage to the hematopoietic compartment and gastrointestinal tract. While bone marrow transplantation can reestablish the hematopoietic compartment, a more imminent risk of death is posed by gastrointestinal acute radiation syndrome (GI-ARS), for which there are no FDA-approved medical countermeasures. Although the mechanisms dictating the severity of GI-ARS remain incompletely understood, sialylation by ST6GAL1 has been shown to protect against radiation-induced apoptosis in vitro. Here, we used a C57BL/6 St6gal1-KO mouse model to investigate the contribution of ST6GAL1 to susceptibility to total body irradiation in vivo. Twelve gray total body ionizing γ-irradiation (TBI) followed by bone marrow transplant is not lethal to wild-type mice, but St6gal1-KO counterparts succumbed within 7 d. Both St6gal1-KO and wild-type animals exhibited damage to the GI epithelium, diarrhea and weight loss, but these symptoms became progressively more severe in the St6gal1-KO animals while wild-type counterparts showed signs of recovery by 120 h after TBI. Increased apoptosis in the GI tracts of St6gal1-KO mice and the absence of regenerative crypts were also observed. Together, these observations highlight an important role for ST6GAL1 in protection and recovery from GI-ARS in vivo.
Collapse
Affiliation(s)
- Patrick R Punch
- Department of Molecular & Cellular Biology, Roswell Park Comprehensive Cancer Center, Elm & Carlton Streets, Buffalo, NY 14263, USA
| | - Eric E Irons
- Department of Molecular & Cellular Biology, Roswell Park Comprehensive Cancer Center, Elm & Carlton Streets, Buffalo, NY 14263, USA
| | - Charles T Manhardt
- Department of Molecular & Cellular Biology, Roswell Park Comprehensive Cancer Center, Elm & Carlton Streets, Buffalo, NY 14263, USA
| | - Himangi Marathe
- Department of Molecular & Cellular Biology, Roswell Park Comprehensive Cancer Center, Elm & Carlton Streets, Buffalo, NY 14263, USA
| | - Joseph T Y Lau
- Department of Molecular & Cellular Biology, Roswell Park Comprehensive Cancer Center, Elm & Carlton Streets, Buffalo, NY 14263, USA
| |
Collapse
|
7
|
Glycobiology of the Epithelial to Mesenchymal Transition. Biomedicines 2021; 9:biomedicines9070770. [PMID: 34356834 PMCID: PMC8301408 DOI: 10.3390/biomedicines9070770] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2021] [Revised: 06/30/2021] [Accepted: 06/30/2021] [Indexed: 12/20/2022] Open
Abstract
Glycosylation consists in the covalent, enzyme mediated, attachment of sugar chains to proteins and lipids. A large proportion of membrane and secreted proteins are indeed glycoproteins, while glycolipids are fundamental component of cell membranes. The biosynthesis of sugar chains is mediated by glycosyltransferases, whose level of expression represents a major factor of regulation of the glycosylation process. In cancer, glycosylation undergoes profound changes, which often contribute to invasion and metastasis. Epithelial to mesenchymal transition (EMT) is a key step in metastasis formation and is intimately associated with glycosylation changes. Numerous carbohydrate structures undergo up- or down-regulation during EMT and often regulate the process. In this review, we will discuss the relationship with EMT of the N-glycans, of the different types of O-glycans, including the classical mucin-type, O-GlcNAc, O-linked fucose, O-linked mannose and of glycolipids. Finally, we will discuss the role in EMT of galectins, a major class of mammalian galactoside-binding lectins. While the expression of specific carbohydrate structures can be used as a marker of EMT and of the propensity to migrate, the manipulation of the glycosylation machinery offers new perspectives for cancer treatment through inhibition of EMT.
Collapse
|
8
|
Grabenstein S, Barnard KN, Anim M, Armoo A, Weichert WS, Bertozzi CR, Parrish CR, Willand-Charnley R. Deacetylated sialic acids modulates immune mediated cytotoxicity via the sialic acid-Siglec pathway. Glycobiology 2021; 31:1279-1294. [PMID: 34192335 DOI: 10.1093/glycob/cwab068] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2020] [Revised: 06/09/2021] [Accepted: 06/20/2021] [Indexed: 12/11/2022] Open
Abstract
Cancers utilize glycans to evade the immune system via the Sialic acid (Sia)-Siglec (Sialic-acid-binding immunoglobulin-like lectins) pathway. Specifically, atypical structural forms of sialic acid bind to inhibitory Siglec receptors on Natural Killer (NK) cells resulting in the suppression of immune cell mediated cytotoxicity. The mechanism of action that governs the Sia-Siglec pathway in cancers is not understood. Specifically, how deviations from the typical form of Sia mechanistically contribute. Here we focused on modulating 9-O and 7,9-O-acetylation of Neu5Ac, via CRISPR-Cas9 gene editing, a functional group that is absent from Sias on many types of cancer cells. The two genes that are responsible for regulating the level of acetylation on Neu5Ac, are Sialic acid acetylesterase (SIAE) and Sialic acid acetyltransferase (CASD1). These genes modulated Siglec binding in colon, lung, and a non-cancerous kidney cell line. In the absence of SIAE, Neu5Ac is acetylated, engagement of cancer associated Siglecs is reduced while binding was increased when the ability to acetylate was removed via CASD1 knock out. In the absence of SIAE NK mediated cytotoxicity increased in both colon and lung cancer cells. In addition to modulating Siglec binding, SIAE expression modulates the level of Sias in a cell, and the α2-6-linkage of Sias - which is specifically upregulated and associated with cancers. Uncovering how functional group alterations on Neu5Ac contribute mechanistically to both Siglec receptor binding, the Sia-Siglec immune evasion pathway, and the production of cancer associated glycosidic linkages -offers a promising avenue for targeted cancer immune therapies in the future.
Collapse
Affiliation(s)
- Susan Grabenstein
- Department of Chemistry & Biochemistry, South Dakota State University, Brookings, SD 57007
| | - Karen N Barnard
- Basic Sciences Division, Fred Hutchinson Cancer Research Center, Seattle WA, 98109
| | - Mathias Anim
- Department of Chemistry & Biochemistry, South Dakota State University, Brookings, SD 57007
| | - Albert Armoo
- Department of Chemistry & Biochemistry, South Dakota State University, Brookings, SD 57007
| | - Wendy S Weichert
- Baker Institute for Animal Health, Department of Microbiology and Immunology College of Veterinary Medicine, Cornell University, Ithaca, NY 14853
| | - Carolyn R Bertozzi
- Department of Chemistry, Stanford University, Stanford, CA 94305.,Howard Hughes Medical Institute, Stanford University, Stanford, CA, USA
| | - Colin R Parrish
- Baker Institute for Animal Health, Department of Microbiology and Immunology College of Veterinary Medicine, Cornell University, Ithaca, NY 14853
| | | |
Collapse
|
9
|
Glycosyltransferase B4GALNT2 as a Predictor of Good Prognosis in Colon Cancer: Lessons from Databases. Int J Mol Sci 2021; 22:ijms22094331. [PMID: 33919332 PMCID: PMC8122605 DOI: 10.3390/ijms22094331] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Revised: 04/16/2021] [Accepted: 04/19/2021] [Indexed: 12/27/2022] Open
Abstract
BACKGROUND glycosyltransferase B4GALNT2 and its cognate carbohydrate antigen Sda are highly expressed in normal colon but strongly downregulated in colorectal carcinoma (CRC). We previously showed that CRC patients expressing higher B4GALNT2 mRNA levels displayed longer survival. Forced B4GALNT2 expression reduced the malignancy and stemness of colon cancer cells. METHODS Kaplan-Meier survival curves were determined in "The Cancer Genome Atlas" (TCGA) COAD cohort for several glycosyltransferases, oncogenes, and tumor suppressor genes. Whole expression data of coding genes as well as miRNA and methylation data for B4GALNT2 were downloaded from TCGA. RESULTS the prognostic potential of B4GALNT2 was the best among the glycosyltransferases tested and better than that of many oncogenes and tumor suppressor genes; high B4GALNT2 expression was associated with a lower malignancy gene expression profile; differential methylation of an intronic B4GALNT2 gene position and miR-204-5p expression play major roles in B4GALNT2 regulation. CONCLUSIONS high B4GALNT2 expression is a strong predictor of good prognosis in CRC as a part of a wider molecular signature that includes ZG16, ITLN1, BEST2, and GUCA2B. Differential DNA methylation and miRNA expression contribute to regulating B4GALNT2 expression during colorectal carcinogenesis.
Collapse
|
10
|
Fernandes Â, Dias AM, Silva MC, Gaifem J, Azevedo CM, Carballo I, Pinho SS. The Role of Glycans in Chronic Inflammatory Gastrointestinal and Liver Disorders and Cancer. COMPREHENSIVE GLYCOSCIENCE 2021:444-470. [DOI: 10.1016/b978-0-12-819475-1.00036-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
11
|
Dorsett KA, Marciel MP, Hwang J, Ankenbauer KE, Bhalerao N, Bellis SL. Regulation of ST6GAL1 sialyltransferase expression in cancer cells. Glycobiology 2020; 31:530-539. [PMID: 33320246 DOI: 10.1093/glycob/cwaa110] [Citation(s) in RCA: 57] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2020] [Revised: 11/07/2020] [Accepted: 11/25/2020] [Indexed: 02/06/2023] Open
Abstract
The ST6GAL1 sialyltransferase, which adds α2-6 linked sialic acids to N-glycosylated proteins, is overexpressed in a wide range of human malignancies. Recent studies have established the importance of ST6GAL1 in promoting tumor cell behaviors such as invasion, resistance to cell stress and chemoresistance. Furthermore, ST6GAL1 activity has been implicated in imparting cancer stem cell characteristics. However, despite the burgeoning interest in the role of ST6GAL1 in the phenotypic features of tumor cells, insufficient attention has been paid to the molecular mechanisms responsible for ST6GAL1 upregulation during neoplastic transformation. Evidence suggests that these mechanisms are multifactorial, encompassing genetic, epigenetic, transcriptional and posttranslational regulation. The purpose of this review is to summarize current knowledge regarding the molecular events that drive enriched ST6GAL1 expression in cancer cells.
Collapse
Affiliation(s)
- Kaitlyn A Dorsett
- Department of Cell, Developmental and Integrative Biology, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Michael P Marciel
- Department of Cell, Developmental and Integrative Biology, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Jihye Hwang
- Department of Cell, Developmental and Integrative Biology, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Katherine E Ankenbauer
- Department of Cell, Developmental and Integrative Biology, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Nikita Bhalerao
- Department of Cell, Developmental and Integrative Biology, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Susan L Bellis
- Department of Cell, Developmental and Integrative Biology, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| |
Collapse
|
12
|
An SY, Lee M, Yoon HK, Abekura F, Kim KS, Kim DH, Kim HJ, Lee K, Kim CH, Lee YC. Regulation of human β-galactoside α2,6-sialyltransferase (hST6Gal I) gene expression during differentiation of human osteoblastic MG-63 cells. Glycoconj J 2020; 37:681-690. [PMID: 33108606 DOI: 10.1007/s10719-020-09959-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2020] [Revised: 08/27/2020] [Accepted: 10/20/2020] [Indexed: 10/23/2022]
Abstract
In this study, we found that gene expression of the human β-galactoside α2,6-sialyltransferase (hST6Gal I) was specifically increased during differentiation of human MG-63 osteoblastic cells by serum starvation (SS). In parallel, a distinct increase in binding to SNA, the α2,6-sialyl-specific lectin, was observed in serum-starved cells, as demonstrated by FACS analysis. 5'-Rapid amplification of cDNA ends analysis demonstrated that the increase of hST6Gal I transcript by SS is mediated by P1 promoter. To elucidate transcriptional regulation of hST6Gal I in SS-induced MG-63 cells, we functionally characterized the P1 promoter region of the hST6Gal I gene. The 5'-deletion analysis of P1 promoter region revealed that the 189 bp upstream region of transcription start site is critical for transcriptional activity of hST6Gal I gene in SS-induced MG-63 cells. This region contains the predicted binding sites for several transcription factors, including AREB6, FOXP1, SIX3, HNF1, YY2, and MOK2. The mutagenesis analysis for these sites and chromatin immunoprecipitation assay demonstrated that the YY2 binding site at -98 to -77 was essential for the SS-induced hST6Gal I gene expression during differentiation of MG-63 cells.
Collapse
Affiliation(s)
- So-Young An
- Department of Medicinal Biotechnology, College of Health Sciences, Dong-A University, Busan, 49315, South Korea
| | - Miri Lee
- Department of Medicinal Biotechnology, College of Health Sciences, Dong-A University, Busan, 49315, South Korea
| | - Hyun-Kyoung Yoon
- Department of Medicinal Biotechnology, College of Health Sciences, Dong-A University, Busan, 49315, South Korea
| | - Fukushi Abekura
- Molecular and Cellular Glycobiology Unit, Department of Biological Sciences, SungKyunKwan University, Kyunggi-Do, 16419, South Korea
| | - Kyoung-Sook Kim
- Department of Medicinal Biotechnology, College of Health Sciences, Dong-A University, Busan, 49315, South Korea
| | - Dong-Hyun Kim
- Department of Medicinal Biotechnology, College of Health Sciences, Dong-A University, Busan, 49315, South Korea
| | - Hyeon-Jun Kim
- Department of Orthopaedic Surgery, College of Medicine, Dong-A University, Busan, 49201, South Korea
| | - Kichoon Lee
- Functional Genomics Laboratory, Department of Animal Sciences, the Ohio State University, 2029 Fyffe Court, Columbus, OH, 43210, USA
| | - Cheorl-Ho Kim
- Molecular and Cellular Glycobiology Unit, Department of Biological Sciences, SungKyunKwan University, Kyunggi-Do, 16419, South Korea.
| | - Young-Choon Lee
- Department of Medicinal Biotechnology, College of Health Sciences, Dong-A University, Busan, 49315, South Korea.
| |
Collapse
|
13
|
Läubli H, Kawanishi K, George Vazhappilly C, Matar R, Merheb M, Sarwar Siddiqui S. Tools to study and target the Siglec-sialic acid axis in cancer. FEBS J 2020; 288:6206-6225. [PMID: 33251699 DOI: 10.1111/febs.15647] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2020] [Revised: 11/18/2020] [Accepted: 11/23/2020] [Indexed: 12/16/2022]
Abstract
Siglecs are widely expressed on leucocytes and bind to ubiquitously presented glycans containing sialic acids (sialoglycans). Most Siglecs carry an immunoreceptor tyrosine-based inhibition motif (ITIM) and elicit an inhibitory intracellular signal upon ligand binding. A few Siglec receptors can, however, recruit immunoreceptor tyrosine-based activation motif (ITAM)-containing factors, which activate cells. The role of hypersialylation (the enhanced expression of sialoglycans) has recently been explored in cancer progression. Mechanistic studies have shown that hypersialylation on cancer cells can engage inhibitory Siglecs on the surface of immune cells and induce immunosuppression. These recent studies strongly suggest that the Siglec-sialic acid axis can act as a potential target for cancer immunotherapy. Moreover, the use of new tools and techniques is facilitating these studies. In this review, we summarise techniques used to study Siglecs, including different mouse models, monoclonal antibodies, Siglec fusion proteins, and sialoglycan arrays. Furthermore, we discuss the recent major developments in the study of Siglecs in cancer immunosuppression, tools, and techniques used in targeting the Siglec-sialic acid axis and the possibility of clinical intervention.
Collapse
Affiliation(s)
- Heinz Läubli
- Laboratory for Cancer Immunotherapy, Department of Biomedicine, and Medical Oncology, Department of Internal Medicine, University Hospital Basel, Switzerland
| | - Kunio Kawanishi
- Kidney and Vascular Pathology, University of Tsukuba, Ibaraki, Japan
| | | | - Rachel Matar
- Department of Biotechnology, American University of Ras Al Khaimah (AURAK), UAE
| | - Maxime Merheb
- Department of Biotechnology, American University of Ras Al Khaimah (AURAK), UAE
| | | |
Collapse
|
14
|
Zhao Y, Chen J, Zhong H, Zhang C, Zhou Y, Mao W, Yu C. Functionalized Ag/Fe-MOFs nanocomposite as a novel endogenous redox mediator for determination of α2,6-sialylated glycans in serum. Mikrochim Acta 2020; 187:649. [DOI: 10.1007/s00604-020-04608-w] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2020] [Accepted: 10/20/2020] [Indexed: 12/11/2022]
|
15
|
Zhu Q, Mao W, Zhang C, Zhou Y, Tang Z, Yu C. Au@BSA microspheres-luminol and a novel luminescent Zeolitic Imidazolate Framework were used for potential-resolved electrochemiluminescence to detect dual targets. Anal Chim Acta 2020; 1140:89-98. [PMID: 33218493 DOI: 10.1016/j.aca.2020.09.057] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2020] [Revised: 09/24/2020] [Accepted: 09/25/2020] [Indexed: 12/21/2022]
Abstract
Here, a novel electrochemiluminescence biosensor based on potential-resolved strategy was firstly prepared for the detection of dual targets α2,3-sialylated glycans and α2,6-sialylated glycans. This is the first time that Au@BSA microsphere was used to connect with luminol to enhance its ECL intensity, and it can generate ECL signals at positive potential. Zeolitic Imidazolate Framework-8 (ZIF-8) and Meso-tetra (4-carboxyphenyl) porphyrin (TCPP) were linked using a one-pot method to synthesize a novel luminescent ZIF (L-ZIF) named TZZ, which can emit ECL signals at negative potential. Moreover, magnetite microspheres were used to construct a sandwich-type biosensor to obtain higher sensitivity and reduce background signals. In addition, the biosensor manufactured directly in solution have a wider linear range than constructed on electrode because it has more available space than the electrode surface. Due to the above advantages, the prepared ECL biosensor exhibited high sensitivity, stability and broader linear range, even for practical analysis. Therefore, the prepared ECL biosensor will become a promising method for determination of α2,3-sialylated glycans and α2,6-sialylated glycans in clinical applications in the future. What is more, it provides a potential method for detection of other multi-targets.
Collapse
Affiliation(s)
- Qihao Zhu
- College of Pharmacy, Chongqing Medical University, Chongqing, 400016, PR China
| | - Weiran Mao
- Chongqing University Cancer Hospital, Chongqing, 400016, PR China
| | - Chengli Zhang
- The First People's Hospital of Zigong, Zigong, Sichuan, 643000, PR China
| | - Yuan Zhou
- College of Pharmacy, Chongqing Medical University, Chongqing, 400016, PR China
| | - Zhiyong Tang
- Department of Pharmacy, Nanchong Central Hospital, The Second Clinical Medical College, North Sichuan Medical College, Nanchong, Sichuan, 637000, PR China
| | - Chao Yu
- College of Pharmacy, Chongqing Medical University, Chongqing, 400016, PR China.
| |
Collapse
|
16
|
Nummela P, Heiskanen A, Kytölä S, Haglund C, Lepistö A, Satomaa T, Ristimäki A. Altered linkage pattern of N-glycan sialic acids in pseudomyxoma peritonei. Glycobiology 2020; 31:211-222. [PMID: 33539510 DOI: 10.1093/glycob/cwaa079] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2020] [Revised: 07/17/2020] [Accepted: 08/13/2020] [Indexed: 11/13/2022] Open
Abstract
Pseudomyxoma peritonei (PMP) is a highly mucinous adenocarcinoma growing in the peritoneal cavity and most commonly originating from the appendix. Glycans play an important role in carcinogenesis, and glycosylation is altered in malignant diseases, including PMP. We have previously demonstrated that fucosylation of N-glycans is increased in PMP, but we did not observe modulation of overall sialylation. As sialic acids can be attached to the rest of the glycan via α2,3- or α2,6-linkage, we have now analyzed the linkage patterns of sialic acids in tissue specimens of normal appendices, low-grade appendiceal mucinous neoplasms (LAMN), low-grade (LG) PMP and high-grade (HG) PMP. For the linkage analysis, the enzymatically released acidic N-glycans were first treated with ethyl esterification or α2,3-sialidase digestion followed by MALDI-TOF mass spectrometry. Significant increase in the relative abundance of α2,6-sialylated and decrease in α2,3-sialylated N-glycans was observed in PMP tumors as compared to the normal appendices (P < 0.025). More specifically, increased α2,6-sialylation (P < 0.05) and decreased α2,3-sialylation (P < 0.01) were detected in afucosylated and monofucosylated N-glycans of PMPs, whereas the less abundant multifucosylated glycans, containing terminal fucose, demonstrated increased α2,3-sialylation (P < 0.01). Importantly, the increase in α2,6-sialylation was also detected between PMP and the appendiceal precursor lesion LAMN (P < 0.01). The identified glycosylation alterations produce ligands for sialic acid-binding immunoglobulin-like lectins (Siglecs) and sialofucosylated glycans binding selectins, which play a role in the peritoneal dissemination and progression of the disease.
Collapse
Affiliation(s)
- Pirjo Nummela
- Applied Tumor Genomics Research Program, Research Programs Unit, Faculty of Medicine, University of Helsinki, Haartmaninkatu 8, FI-00290, Helsinki, Finland
| | | | - Soili Kytölä
- Department of Genetics, HUSLAB, HUS Diagnostic Center, Helsinki University Hospital, Topeliuksenkatu 32, FI-00290 Helsinki, Finland
| | - Caj Haglund
- Department of Surgery, University of Helsinki and Helsinki University Hospital, Stenbäckinkatu 9A, FI-00290 Helsinki, Finland.,Translational Cancer Medicine Research Program, Research Programs Unit, Faculty of Medicine, University of Helsinki, Haartmaninkatu 8, FI-00290, Finland
| | - Anna Lepistö
- Department of Surgery, University of Helsinki and Helsinki University Hospital, Stenbäckinkatu 9A, FI-00290 Helsinki, Finland
| | - Tero Satomaa
- Glykos Finland Ltd, Viikinkaari 6, FI-00790 Helsinki, Finland
| | - Ari Ristimäki
- Applied Tumor Genomics Research Program, Research Programs Unit, Faculty of Medicine, University of Helsinki, Haartmaninkatu 8, FI-00290, Helsinki, Finland.,Department of Pathology, HUSLAB, University of Helsinki and Helsinki University Hospital, Haartmaninkatu 3, FI-00290 Helsinki, Finland
| |
Collapse
|
17
|
Pérez AG, Andrade-Da-Costa J, De Souza WF, De Souza Ferreira M, Boroni M, De Oliveira IM, Freire-Neto CA, Fernandes PV, De Lanna CA, Souza-Santos PT, Morgado-Díaz JA, De-Freitas-Junior JCM. N‑glycosylation and receptor tyrosine kinase signaling affect claudin‑3 levels in colorectal cancer cells. Oncol Rep 2020; 44:1649-1661. [PMID: 32945502 PMCID: PMC7448416 DOI: 10.3892/or.2020.7727] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2019] [Accepted: 07/14/2020] [Indexed: 12/14/2022] Open
Abstract
Changes in protein levels in different components of the apical junctional complex occur in colorectal cancer (CRC). Claudin-3 is one of the main constituents of tight junctions, and its overexpression can increase the paracellular flux of macromolecules, as well as the malignant potential of CRC cells. The aim of this study was to investigate the molecular mechanisms involved in the regulation of claudin-3 and its prognostic value in CRC. In silico evaluation in each of the CRC consensus molecular subtypes (CMSs) revealed that high expression levels of CLDN3 (gene encoding claudin-3) in CMS2 and CMS3 worsened the patients' long-term survival, whereas a decrease in claudin-3 levels concomitant with a reduction in phosphorylation levels of epidermal growth factor receptor (EGFR) and insulin-like growth factor 1 receptor (IGF1R) could be achieved by inhibiting N-glycan biosynthesis in CRC cells. We also observed that specific inactivation of these receptor tyrosine kinases (RTKs) led to a decrease in claudin-3 levels, and this regulation seems to be mediated by phospholipase C (PLC) and signal transducer and activator of transcription 3 (STAT3) in CRC cells. RTKs are modulated by their N-linked glycans, and inhibition of N-glycan biosynthesis decreased the claudin-3 levels; therefore, we evaluated the correlation between N-glycogenes and CLDN3 expression levels in each of the CRC molecular subtypes. The CMS1 (MSI immune) subtype concomitantly exhibited low expression levels of CLDN3 and N-glycogenes (MGAT5, ST6GAL1, and B3GNT8), whereas CMS2 (canonical) exhibited high gene expression levels of CLDN3 and N-glycogenes (ST6GAL1 and B3GNT8). A robust positive correlation was also observed between CLDN3 and B3GNT8 expression levels in all CMSs. These results support the hypothesis of a mechanism integrating RTK signaling and N-glycosylation for the regulation of claudin-3 levels in CRC, and they suggest that CLDN3 expression can be used to predict the prognosis of patients identified as CMS2 or CMS3.
Collapse
Affiliation(s)
- Amelia G Pérez
- Cellular and Molecular Oncobiology Program, National Cancer Institute (INCA), Rio de Janeiro, RJ 20231‑050, Brazil
| | - Jéssica Andrade-Da-Costa
- Cellular and Molecular Oncobiology Program, National Cancer Institute (INCA), Rio de Janeiro, RJ 20231‑050, Brazil
| | - Waldemir F De Souza
- Cellular and Molecular Oncobiology Program, National Cancer Institute (INCA), Rio de Janeiro, RJ 20231‑050, Brazil
| | - Michelle De Souza Ferreira
- Cellular and Molecular Oncobiology Program, National Cancer Institute (INCA), Rio de Janeiro, RJ 20231‑050, Brazil
| | - Mariana Boroni
- Bioinformatics and Computational Biology Laboratory, National Cancer Institute (INCA), Rio de Janeiro, RJ 20231‑050, Brazil
| | - Ivanir M De Oliveira
- Pathology Division, National Cancer Institute (INCA), Rio de Janeiro, RJ 20231‑050, Brazil
| | - Carlos A Freire-Neto
- Cellular and Molecular Oncobiology Program, National Cancer Institute (INCA), Rio de Janeiro, RJ 20231‑050, Brazil
| | - Priscila V Fernandes
- Pathology Division, National Cancer Institute (INCA), Rio de Janeiro, RJ 20231‑050, Brazil
| | - Cristóvão A De Lanna
- Bioinformatics and Computational Biology Laboratory, National Cancer Institute (INCA), Rio de Janeiro, RJ 20231‑050, Brazil
| | | | - José A Morgado-Díaz
- Cellular and Molecular Oncobiology Program, National Cancer Institute (INCA), Rio de Janeiro, RJ 20231‑050, Brazil
| | | |
Collapse
|
18
|
Venturi G, Gomes Ferreira I, Pucci M, Ferracin M, Malagolini N, Chiricolo M, Dall'Olio F. Impact of sialyltransferase ST6GAL1 overexpression on different colon cancer cell types. Glycobiology 2020; 29:684-695. [PMID: 31317190 DOI: 10.1093/glycob/cwz053] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2019] [Revised: 07/11/2019] [Accepted: 07/11/2019] [Indexed: 12/11/2022] Open
Abstract
Cancer-associated glycan structures can be both tumor markers and engines of disease progression. The structure Siaα2,6Galβ1,4GlcNAc (Sia6LacNAc), synthesized by sialyltransferase ST6GAL1, is a cancer-associated glycan. Although ST6GAL1/Sia6LacNAc are often overexpressed in colorectal cancer (CRC), their biological and clinical significance remains unclear. To get insights into the clinical relevance of ST6GAL1 expression in CRC, we interrogated The Cancer Genome Atlas with mRNA expression data of hundreds of clinically characterized CRC and normal samples. We found an association of low ST6GAL1 expression with microsatellite instability (MSI), BRAF mutations and mucinous phenotype but not with stage, response to therapy and survival. To investigate the impact of ST6GAL1 expression in experimental systems, we analyzed the transcriptome and the phenotype of the CRC cell lines SW948 and SW48 after retroviral transduction with ST6GAL1 cDNA. The two cell lines display the two main pathways of CRC transformation: chromosomal instability and MSI, respectively. Constitutive ST6GAL1 expression induced much deeper transcriptomic changes in SW948 than in SW48 and affected different genes in the two cell lines. ST6GAL1 expression affected differentially the tyrosine phosphorylation induced by hepatocyte growth factor, the ability to grow in soft agar, to heal a scratch wound and to invade Matrigel in the two cell lines. These results indicate that the altered expression of a cancer-associated glycosyltransferase impacts the gene expression profile, as well as the phenotype, although in a cancer subtype-specific manner.
Collapse
Affiliation(s)
- Giulia Venturi
- Department of Experimental, Diagnostic and Specialty Medicine, General Pathology Building, University of Bologna Via S. Giacomo 14, Bologna, Italy
| | - Inês Gomes Ferreira
- Department of Experimental, Diagnostic and Specialty Medicine, General Pathology Building, University of Bologna Via S. Giacomo 14, Bologna, Italy
| | - Michela Pucci
- Department of Experimental, Diagnostic and Specialty Medicine, General Pathology Building, University of Bologna Via S. Giacomo 14, Bologna, Italy
| | - Manuela Ferracin
- Department of Experimental, Diagnostic and Specialty Medicine, General Pathology Building, University of Bologna Via S. Giacomo 14, Bologna, Italy
| | - Nadia Malagolini
- Department of Experimental, Diagnostic and Specialty Medicine, General Pathology Building, University of Bologna Via S. Giacomo 14, Bologna, Italy
| | - Mariella Chiricolo
- Department of Experimental, Diagnostic and Specialty Medicine, General Pathology Building, University of Bologna Via S. Giacomo 14, Bologna, Italy
| | - Fabio Dall'Olio
- Department of Experimental, Diagnostic and Specialty Medicine, General Pathology Building, University of Bologna Via S. Giacomo 14, Bologna, Italy
| |
Collapse
|
19
|
Zhang D, Xie Q, Wang Q, Wang Y, Miao J, Li L, Zhang T, Cao X, Li Y. Mass spectrometry analysis reveals aberrant N-glycans in colorectal cancer tissues. Glycobiology 2019; 29:372-384. [PMID: 30698702 DOI: 10.1093/glycob/cwz005] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2018] [Revised: 11/23/2018] [Accepted: 01/26/2019] [Indexed: 12/17/2022] Open
Abstract
Aberrant glycosylation is strongly correlated with the development of various cancers. Tumor-associated carbohydrate antigens, including N-glycans, are predominantly expressed on the tumor cell surface. Because the incidence of colorectal cancer is high in China, we investigated aberrant N-glycans from colorectal cancer tissues (CRC) in Chinese patients. By Linear ion trap quadrupole-electrospray ionization mass spectrometry, we performed glycomic assays on N-glycans obtained from solid CRC tissues and paired peritumoral tissues. In total, aberrant N-glycans were expressed in the colorectal tumor tissues. Specifically, seven bisecting structures (M/Z 9732+, 10602+, 10752+, 11622+, 11772+, 12642+, 13522+) decreased, M/Z 10552+ (two-antennae complex N-glycan) and M/Z 12792+ (three-antennae complex N-glycan) decreased, M/Z 10132+ and M/Z 11162+ (high-mannose N-glycan) increased, and M/Z 12282+ (bifucosylated N-glycan) increased. To evaluate the MS profile data, several statistical tools were applied, including student's t test, orthogonal partial least squares discriminant analysis and receiver operating characteristic curve. The measurement of the degree of bisecting N-glycans had an area under the curve value of 0.823. Interestingly, we observed that the bisecting N-glycans decreased with the tumor stages. This phenomenon was not found in esophageal squamous cell carcinoma, in which the bisecting N-glycans had no change. Thus, the expression of bisecting N-glycans may be an interesting point in the study of colorectal cancer.
Collapse
Affiliation(s)
- Dongmei Zhang
- Jiangsu Key Laboratory of Infection and Immunity, Institutes of Biology and Medical Science, Soochow University, 199 Ren-Ai Road, SuZhou, Jiangsu Province, China
| | - Qing Xie
- Jiangsu Key Laboratory of Infection and Immunity, Institutes of Biology and Medical Science, Soochow University, 199 Ren-Ai Road, SuZhou, Jiangsu Province, China
| | - Qian Wang
- Nanjing First Hospital, Nanjing Medical University, Nanjing, Jiangsu Province, China
| | - Yanping Wang
- Jiangsu Key Laboratory of Infection and Immunity, Institutes of Biology and Medical Science, Soochow University, 199 Ren-Ai Road, SuZhou, Jiangsu Province, China
| | - Jinsheng Miao
- Jiangsu Key Laboratory of Infection and Immunity, Institutes of Biology and Medical Science, Soochow University, 199 Ren-Ai Road, SuZhou, Jiangsu Province, China
| | - Ling Li
- Jiangsu Key Laboratory of Infection and Immunity, Institutes of Biology and Medical Science, Soochow University, 199 Ren-Ai Road, SuZhou, Jiangsu Province, China
| | - Tong Zhang
- Jiangsu Key Laboratory of Infection and Immunity, Institutes of Biology and Medical Science, Soochow University, 199 Ren-Ai Road, SuZhou, Jiangsu Province, China
| | - Xiufeng Cao
- Nanjing First Hospital, Nanjing Medical University, Nanjing, Jiangsu Province, China.,Taikang Xianlin Drum Tower Hospital School of Medicine, Nanjing University, Nanjing, Jiangsu Province, China
| | - Yunsen Li
- Jiangsu Key Laboratory of Infection and Immunity, Institutes of Biology and Medical Science, Soochow University, 199 Ren-Ai Road, SuZhou, Jiangsu Province, China
| |
Collapse
|
20
|
Peng Y, Wang L, Zhang Y, Bao H, Lu H. Stable Isotope Sequential Derivatization for Linkage-Specific Analysis of Sialylated N-Glycan Isomers by MS. Anal Chem 2019; 91:15993-16001. [PMID: 31730330 DOI: 10.1021/acs.analchem.9b04727] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Sialylated N-glycans play pivotal role in several important biological and pathological processes. Their sialyl-linkage isomers, mostly α-2,3- and α-2,6-linked, act differently during the cellular events and several diseases. While mass spectrometry (MS) technology is a powerful tool in N-glycome analysis, it still suffers from an inability to distinguish linkage isomers of native N-glycans. Herein, we described a sequential selective derivatization method, by which α-2,6- and α-2,3-linked sialic acids are sequentially labeled with methylamide incorporated with a different stable isotope. Isobaric labeling avoids inducing bias in ionization efficiency and chromatographic behavior. In optimized reaction conditions, high derivatization selectivity (∼99%) was achieved for both α-2,3- and α-2,6-linked sialic acid. High accuracy of quantitation within a dynamic range of 2 orders of magnitude and high reproducibility (CV < 20%, n = 3) were demonstrated using standard glycans and multisialylated N-glycans. Finally, this method was applied in profiling the N-glycome of serum from CRC patients, where a level of six sialyl-linkage isomers were found to be altered significantly compared with that from healthy individuals.
Collapse
|
21
|
Zhou L, Zhang S, Zou X, Lu J, Yang X, Xu Z, Shan A, Jia W, Liu F, Yan X, Su H, Liang T, Zheng M, Zhang Y, Feng B. The β-galactoside α2,6-sialyltranferase 1 (ST6GAL1) inhibits the colorectal cancer metastasis by stabilizing intercellular adhesion molecule-1 via sialylation. Cancer Manag Res 2019; 11:6185-6199. [PMID: 31308754 PMCID: PMC6613604 DOI: 10.2147/cmar.s208631] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2019] [Accepted: 05/09/2019] [Indexed: 12/24/2022] Open
Abstract
Background: Colorectal cancer (CRC) is one of the most frequent malignancies of the digestive system. Elevated expression of β-galactoside α2,6-sialyltranferase 1 (ST6GAL1) has been observed in multiple cancers. But the mechanism of how ST6GAL1 might affect cancer cells remains to be clarified. Our previous study recognized intercellular adhesion molecule-1(ICAM-1) as a probable substrate of ST6GAL1 through mass spectrometry (MS) analysis. ICAM-1 is related to tumor metastasis in various cancers. Methods: First, ST6GAL1 was overexpressed and knocked down to perform transwell and wound healing assays, and the results were further confirmed in vivo. Based on the results of MS, GO and KEGG analysis were applied to reveal the connection between ST6GAL1 and ICAM-1. Immunoblot and tissue microarrays were administered to investigate the expression of ICAM-1 in different stages of CRC. Next, PCR, lectin precipitation and cycloheximide (CHX) were used to demonstrate the mechanism of ST6GAL1 on ICAM-1. Moreover, we investigated the sialylation on soluble ICAM in serum and its connection to tumor staging. Results: Overexpression of ST6GAL1 inhibited the migratory ability, while knockdown of ST6GAL1 cells had the reverse effect. Moreover, nude mice injected with ST6GAL1-knockdown cells harvested more liver metastases. Based on the GO and KEGG analysis, data from TCGA database showed a positive correlation between ST6GAL1 and ICAM-1. ICAM-1 also demonstrated a significant decrease in stage III/IV compared with stage I/II tumors. Our results revealed that ST6GAL1 could increase the stability of ICAM-1 through sialylation but had little influence on transcriptional level. Additionally, results of serum lectin precipitation revealed a correlation between the level of sialylation on soluble ICAM and CRC staging. Conclusion: This study illustrated that ST6GAL1 inhibited the metastatic ability of CRC by stabilizing ICAM-1 via sialylation and demonstrated a correlation between CRC staging and the sialylation on soluble ICAM-1 in serum.
Collapse
Affiliation(s)
- Leqi Zhou
- Department of General Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China.,Shanghai Minimally Invasive Surgery Center, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China
| | - Sen Zhang
- Department of General Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China.,Shanghai Minimally Invasive Surgery Center, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China
| | - Xia Zou
- Ministry of Education Key Laboratory of Systems Biomedicine, Shanghai Center for Systems Biomedicine (SCSB), Shanghai Jiao Tong University, Shanghai, People's Republic of China
| | - Jishun Lu
- Ministry of Education Key Laboratory of Systems Biomedicine, Shanghai Center for Systems Biomedicine (SCSB), Shanghai Jiao Tong University, Shanghai, People's Republic of China
| | - Xiao Yang
- Department of General Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China.,Shanghai Minimally Invasive Surgery Center, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China
| | - Zhijue Xu
- Ministry of Education Key Laboratory of Systems Biomedicine, Shanghai Center for Systems Biomedicine (SCSB), Shanghai Jiao Tong University, Shanghai, People's Republic of China
| | - Aidong Shan
- Ministry of Education Key Laboratory of Systems Biomedicine, Shanghai Center for Systems Biomedicine (SCSB), Shanghai Jiao Tong University, Shanghai, People's Republic of China
| | - Wenjuan Jia
- Ministry of Education Key Laboratory of Systems Biomedicine, Shanghai Center for Systems Biomedicine (SCSB), Shanghai Jiao Tong University, Shanghai, People's Republic of China
| | - Feng Liu
- Ministry of Education Key Laboratory of Systems Biomedicine, Shanghai Center for Systems Biomedicine (SCSB), Shanghai Jiao Tong University, Shanghai, People's Republic of China
| | - Xialin Yan
- Department of General Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China.,Shanghai Minimally Invasive Surgery Center, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China
| | - Hao Su
- Department of General Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China.,Shanghai Minimally Invasive Surgery Center, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China
| | - Tao Liang
- Ministry of Education Key Laboratory of Systems Biomedicine, Shanghai Center for Systems Biomedicine (SCSB), Shanghai Jiao Tong University, Shanghai, People's Republic of China
| | - Minhua Zheng
- Department of General Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China.,Shanghai Minimally Invasive Surgery Center, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China
| | - Yan Zhang
- Ministry of Education Key Laboratory of Systems Biomedicine, Shanghai Center for Systems Biomedicine (SCSB), Shanghai Jiao Tong University, Shanghai, People's Republic of China
| | - Bo Feng
- Department of General Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China.,Shanghai Minimally Invasive Surgery Center, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China
| |
Collapse
|
22
|
Li J, He J, Zhang C, Chen J, Mao W, Yu C. Dual-type responsive electrochemical biosensor for the detection of α2,6-sialylated glycans based on AuNRs-SA coupled with c-SWCNHs/S-PtNC nanocomposites signal amplification. Biosens Bioelectron 2019; 130:166-173. [PMID: 30735949 DOI: 10.1016/j.bios.2019.01.054] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2018] [Revised: 01/19/2019] [Accepted: 01/22/2019] [Indexed: 02/08/2023]
Abstract
In this study, a dual-type responsive electrochemical biosensor was developed for the quantitative detection of α2,6-sialylated glycans (α2,6-sial-Gs), a potential biomarker of tumors. The gold nanorods (AuNRs), which exhibited great specific surface area, as well as good biocompatibility, was synthesized by the way of seed growth method. Furthermore, a biotin-streptavidin (biotin-SA) system was introduced to improve the immunoreaction efficiency. Accordingly, a label-free biosensor was fabricated based on AuNRs-SA for the quick detection of α2,6-sial-Gs by recording the signal of differential pulse voltammetry (DPV). Furthermore, to expand the ultrasensitive detection of α2,6-sial-Gs, a carboxylated single-walled carbon nanohorns/sulfur-doped platinum nanocluster (c-SWCNHs/S-PtNC) was synthesized for the first time as a novel signal label, which showed an excellent catalytic performance. The usage of c-SWCNHs/S-PtNC could significantly amplify the electrochemical signal recorded by the amperometric i-t curve. Herein, a sandwich type biosensor was constructed by combining the AuNRs-SA on the electrode and c-SWCNHs/S-PtNC (signal amplifier). The label-free biosensor possessed a linear range from 5 ng mL-1 to 5 μg mL-1 with a detection limit of 0.50 ng mL-1, and the sandwich-type biosensor possessed a wide linear range from 1 fg mL-1 to 100 ng mL-1 with a detection limit of 0.69 fg mL-1. Furthermore, the biosensor exhibited excellent recovery and stability, indicating its potential for use in actual samples.
Collapse
Affiliation(s)
- Jia Li
- College of Pharmacy, Institute of Life Science and School of Public Health, Chongqing Medical University, Chongqing 400016, PR China
| | - Junlin He
- College of Pharmacy, Institute of Life Science and School of Public Health, Chongqing Medical University, Chongqing 400016, PR China
| | - Chengli Zhang
- College of Pharmacy, Institute of Life Science and School of Public Health, Chongqing Medical University, Chongqing 400016, PR China
| | - Jun Chen
- College of Pharmacy, Institute of Life Science and School of Public Health, Chongqing Medical University, Chongqing 400016, PR China
| | - Weiran Mao
- College of Pharmacy, Institute of Life Science and School of Public Health, Chongqing Medical University, Chongqing 400016, PR China
| | - Chao Yu
- College of Pharmacy, Institute of Life Science and School of Public Health, Chongqing Medical University, Chongqing 400016, PR China.
| |
Collapse
|
23
|
Rodrigues E, Macauley MS. Hypersialylation in Cancer: Modulation of Inflammation and Therapeutic Opportunities. Cancers (Basel) 2018; 10:cancers10060207. [PMID: 29912148 PMCID: PMC6025361 DOI: 10.3390/cancers10060207] [Citation(s) in RCA: 150] [Impact Index Per Article: 21.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2018] [Revised: 06/13/2018] [Accepted: 06/14/2018] [Indexed: 02/06/2023] Open
Abstract
Cell surface glycosylation is dynamic and often changes in response to cellular differentiation under physiological or pathophysiological conditions. Altered glycosylation on cancers cells is gaining attention due its wide-spread occurrence across a variety of cancer types and recent studies that have documented functional roles for aberrant glycosylation in driving cancer progression at various stages. One change in glycosylation that can correlate with cancer stage and disease prognosis is hypersialylation. Increased levels of sialic acid are pervasive in cancer and a growing body of evidence demonstrates how hypersialylation is advantageous to cancer cells, particularly from the perspective of modulating immune cell responses. Sialic acid-binding receptors, such as Siglecs and Selectins, are well-positioned to be exploited by cancer hypersialylation. Evidence is also mounting that Siglecs modulate key immune cell types in the tumor microenvironment, particularly those responsible for maintaining the appropriate inflammatory environment. From these studies have come new and innovative ways to block the effects of hypersialylation by directly reducing sialic acid on cancer cells or blocking interactions between sialic acid and Siglecs or Selectins. Here we review recent works examining how cancer cells become hypersialylated, how hypersialylation benefits cancer cells and tumors, and proposed therapies to abrogate hypersialylation of cancer.
Collapse
Affiliation(s)
- Emily Rodrigues
- Department of Chemistry, University of Alberta, Edmonton, AB T6G 2G2, Canada.
| | - Matthew S Macauley
- Department of Chemistry, University of Alberta, Edmonton, AB T6G 2G2, Canada.
- Department of Medical Microbiology and Immunology, University of Alberta, Edmonton, AB T6G 2G2, Canada.
| |
Collapse
|
24
|
Zhang Z, Wuhrer M, Holst S. Serum sialylation changes in cancer. Glycoconj J 2018; 35:139-160. [PMID: 29680984 PMCID: PMC5916985 DOI: 10.1007/s10719-018-9820-0] [Citation(s) in RCA: 125] [Impact Index Per Article: 17.9] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2017] [Revised: 02/14/2018] [Accepted: 03/26/2018] [Indexed: 12/17/2022]
Abstract
Cancer is a major cause of death in both developing and developed countries. Early detection and efficient therapy can greatly enhance survival. Aberrant glycosylation has been recognized to be one of the hallmarks of cancer as glycans participate in many cancer-associated events. Cancer-associated glycosylation changes often involve sialic acids which play important roles in cell-cell interaction, recognition and immunological response. This review aims at giving a comprehensive overview of the literature on changes of sialylation in serum of cancer patients. Furthermore, the methods available to measure serum and plasma sialic acids as well as possible underlying biochemical mechanisms involved in the serum sialylation changes are surveyed. In general, total serum sialylation levels appear to be increased with various malignancies and show a potential for clinical applications, especially for disease monitoring and prognosis. In addition to overall sialic acid levels and the amount of sialic acid per total protein, glycoprofiling of specific cancer-associated glycoproteins, acute phase proteins and immunoglobulins in serum as well as the measurements of sialylation-related enzymes such as sialidases and sialyltransferases have been reported for early detection of cancer, assessing cancer progression and improving prognosis of cancer patients. Moreover, sialic-acid containing glycan antigens such as CA19-9, sialyl Lewis X and sialyl Tn on serum proteins have also displayed their value in cancer diagnosis and management whereby increased levels of these factors positively correlated with metastasis or poor prognosis.
Collapse
Affiliation(s)
- Zejian Zhang
- Center for Proteomics and Metabolomics, Leiden University Medical Center, Postzone S3, Postbus 9600, 2300 RC, Leiden, NL, The Netherlands.,Department of Biochemistry and Molecular Biology, Key Laboratory of Glycoconjugate Research Ministry of Public Health, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Manfred Wuhrer
- Center for Proteomics and Metabolomics, Leiden University Medical Center, Postzone S3, Postbus 9600, 2300 RC, Leiden, NL, The Netherlands
| | - Stephanie Holst
- Center for Proteomics and Metabolomics, Leiden University Medical Center, Postzone S3, Postbus 9600, 2300 RC, Leiden, NL, The Netherlands.
| |
Collapse
|
25
|
Wu CY, Huo JP, Zhang XK, Zhang YJ, Hu WM, Yang P, Lu JB, Zhang ZL, Cao Y. Loss of CD15 expression in clear cell renal cell carcinoma is correlated with worse prognosis in Chinese patients. Jpn J Clin Oncol 2018; 47:1182-1188. [PMID: 29036563 DOI: 10.1093/jjco/hyx139] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2017] [Accepted: 09/12/2017] [Indexed: 01/01/2023] Open
Abstract
Purpose To explore the role of CD15 expression in the prognosis of clear cell renal cell carcinoma (ccRCC) in Chinese patients. Methods The study included 301 patients who had undergone surgery for localized ccRCC. All paraffin-embedded tumor sections were collected to make a set of tissue microarrays. CD15 expression was assessed by immunohistochemistry. The relationship between CD15 expression and survival parameters, clinicopathology features was assessed. Kaplan-Meier and Cox proportional hazards model were utilized to determine the correlation between CD15 expression and overall survival (OS). Results The median follow-up time was 54.6 months (range, 3-121 months). The positive rate of CD15 expression was 81.7% (246/301). The cut-off value of CD15 expression was defined as the maximum for Youden index by plotting the receiver operating characteristic curve for survival status. As the threshold was 0.5, all cases were divided into two groups: positive expression group and negative expression group. In correlation analysis, loss of CD15 expression was correlated with female gender, higher Fuhrman nuclear grade, with sarcomatoid differentiation, with necrosis, and with vascular invasion. Kaplan-Meier analysis indicated that the OS time of patients with loss of CD15 expression was shorter than that of patients with positive CD15 expression (P = 0.013). Conclusion CD15 is a significant prognostic factor in clear cell renal cell carcinoma.
Collapse
Affiliation(s)
- Chen-Yan Wu
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center.,Department of Pathology, Sun Yat-sen University Cancer Center
| | - Jian-Ping Huo
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center.,Department of Pathology, Sun Yat-sen University Cancer Center
| | - Xin-Ke Zhang
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center.,Department of Pathology, Sun Yat-sen University Cancer Center
| | - Yi-Jun Zhang
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center.,Department of Pathology, Sun Yat-sen University Cancer Center
| | - Wan-Ming Hu
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center.,Department of Pathology, Sun Yat-sen University Cancer Center
| | - Ping Yang
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center.,Department of Pathology, Sun Yat-sen University Cancer Center
| | - Jia-Bin Lu
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center.,Department of Pathology, Sun Yat-sen University Cancer Center
| | - Zhi-Ling Zhang
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center.,Department of Urology, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Yun Cao
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center.,Department of Pathology, Sun Yat-sen University Cancer Center
| |
Collapse
|
26
|
Mariño-Crespo Ó, Fernández-Briera A, Gil-Martín E. Identification of proteins with the CDw75 epitope in human colorectal cancer. Oncol Lett 2018; 15:580-587. [PMID: 29391890 DOI: 10.3892/ol.2017.7336] [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: 11/07/2016] [Accepted: 07/28/2017] [Indexed: 11/05/2022] Open
Abstract
The CDw75 epitope is an α(2,6) sialylated antigen overexpressed in colorectal cancer (CRC), where its expression correlates with the progression of the disease. The CDw75 epitope is located mainly in N-glycoproteins, whose identity remains unknown. The aim of the present study was to identify proteins with the CDw75 epitope as a strategy to deepen the understanding of molecular pathogenesis of CRC and to identify novel biomarkers for this disease. For this purpose, a two-dimensional electrophoresis approach was employed. Protein spots in the gels were matched to the corresponding CDw75 positive spots in the immunoblotted polyvinylidene difluoride membranes, and further identification of the protein species was performed by mass spectrometry. Additionally, one-dimensional western blotting experiments were performed to verify the expression of these candidate proteins in the colorectal tissue and their coincidence in molecular mass with the CDw75-positive bands. The findings of the present study indicate that haptoglobin and the keratins 8 (K8) and 18 (K18) are proteins with the CDw75 epitope in the colorectal tissue from CRC patients and also suggest novel functions and cellular locations for these proteins in the colorectal tissue and in relation to CRC.
Collapse
Affiliation(s)
- Óscar Mariño-Crespo
- Department of Biochemistry, Genetics and Immunology, Biomedical Research Center (CINBIO, 'Centro Singular de Investigación de Galicia'), University of Vigo, 36310 Vigo, Spain
| | - Almudena Fernández-Briera
- Department of Biochemistry, Genetics and Immunology, Biomedical Research Center (CINBIO, 'Centro Singular de Investigación de Galicia'), University of Vigo, 36310 Vigo, Spain
| | - Emilio Gil-Martín
- Department of Biochemistry, Genetics and Immunology, Biomedical Research Center (CINBIO, 'Centro Singular de Investigación de Galicia'), University of Vigo, 36310 Vigo, Spain
| |
Collapse
|
27
|
Very N, Lefebvre T, El Yazidi-Belkoura I. Drug resistance related to aberrant glycosylation in colorectal cancer. Oncotarget 2018; 9:1380-1402. [PMID: 29416702 PMCID: PMC5787446 DOI: 10.18632/oncotarget.22377] [Citation(s) in RCA: 69] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2017] [Accepted: 10/04/2017] [Indexed: 02/06/2023] Open
Abstract
Colorectal cancer (CRC) is the fourth leading cause of cancer-related deaths in the world. Drug resistance of tumour cells remains the main challenge toward curative treatments efficiency. Several epidemiologic studies link emergence and recurrence of this cancer to metabolic disorders. Glycosylation that modifies more than 80% of human proteins is one of the most widepread nutrient-sensitive post-translational modifications. Aberrant glycosylation participates in the development and progression of cancer. Thus, some of these glycan changes like carbohydrate antigen CA 19-9 (sialyl Lewis a, sLea) or those found on carcinoembryonic antigen (CEA) are already used as clinical biomarkers to detect and monitor CRC. The current review highlights emerging evidences accumulated mainly during the last decade that establish the role played by altered glycosylations in CRC drug resistance mechanisms that induce resistance to apoptosis and activation of signaling pathways, alter drug absorption and metabolism, and led to stemness acquisition. Knowledge in this field of investigation could aid to the development of better therapeutic approaches with new predictive biomarkers and targets tied in with adapted diet.
Collapse
Affiliation(s)
- Ninon Very
- Unité de Glycobiologie Structurale et Fonctionnelle, UGSF-UMR 8576 CNRS, Université de Lille, Lille 59000, France
| | - Tony Lefebvre
- Unité de Glycobiologie Structurale et Fonctionnelle, UGSF-UMR 8576 CNRS, Université de Lille, Lille 59000, France
| | - Ikram El Yazidi-Belkoura
- Unité de Glycobiologie Structurale et Fonctionnelle, UGSF-UMR 8576 CNRS, Université de Lille, Lille 59000, France
| |
Collapse
|
28
|
de Freitas Junior JCM, Morgado-Díaz JA. The role of N-glycans in colorectal cancer progression: potential biomarkers and therapeutic applications. Oncotarget 2017; 7:19395-413. [PMID: 26539643 PMCID: PMC4991391 DOI: 10.18632/oncotarget.6283] [Citation(s) in RCA: 64] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2015] [Accepted: 10/22/2015] [Indexed: 12/12/2022] Open
Abstract
Changes in glycosylation, which is one of the most common protein post-translational modifications, are considered to be a hallmark of cancer. N-glycans can modulate cell migration, cell-cell adhesion, cell signaling, growth and metastasis. The colorectal cancer (CRC) is a leading cause of cancer-related mortality and the correlation between CRC progression and changes in the pattern of expression of N-glycans is being considered in the search for new biomarkers. Here, we review the role of N-glycans in CRC cell biology. The perspectives on emerging N-glycan-related anticancer therapies, along with new insights and challenges, are also discussed.
Collapse
Affiliation(s)
| | - José Andrés Morgado-Díaz
- Cellular Biology Program, Structural Biology Group, Brazilian National Cancer Institute (INCA), Rio de Janeiro, RJ, Brazil
| |
Collapse
|
29
|
Briggs MT, Ho YY, Kaur G, Oehler MK, Everest-Dass AV, Packer NH, Hoffmann P. N-Glycan matrix-assisted laser desorption/ionization mass spectrometry imaging protocol for formalin-fixed paraffin-embedded tissues. RAPID COMMUNICATIONS IN MASS SPECTROMETRY : RCM 2017; 31:825-841. [PMID: 28271569 DOI: 10.1002/rcm.7845] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/30/2016] [Revised: 02/21/2017] [Accepted: 02/23/2017] [Indexed: 06/06/2023]
Abstract
RATIONALE Matrix-assisted laser desorption/ionization mass spectrometry imaging (MALDI-MSI) of the proteome of a tissue has been an established technique for the past decade. In the last few years, MALDI-MSI of the N-glycome has emerged as a novel MALDI-MSI technique. To assess the accuracy and clinical significance of the N-linked glycan spatial distribution, we have developed a method that utilises MALDI-MSI followed by liquid chromatography coupled to tandem mass spectrometry (LC/MS/MS) in order to assign glycan structures to the differentiating MALDI-MSI glycan masses released from the tissue glycoproteins. METHODS AND RESULTS Our workflow presents a comprehensive list of instructions on how to (i) apply MALDI-MSI to spatially map the N-glycome across formalin-fixed paraffin-embedded (FFPE) clinical samples, (ii) structurally characterise N-glycans extracted from consecutive FFPE tissue sections by LC/MS/MS, and (iii) match relevant N-glycan masses from MALDI-MSI with confirmed N-glycan structures determined by LC/MS/MS. CONCLUSIONS Our protocol provides groups that are new to this technique with instructions how to establish N-glycan MALDI-MSI in their laboratory. Furthermore, the method assigns N-glycan structural detail to the masses obtained in the MALDI-MS image. Copyright © 2017 John Wiley & Sons, Ltd.
Collapse
Affiliation(s)
- Matthew T Briggs
- Adelaide Proteomics Centre, School of Biological Sciences, University of Adelaide, Adelaide, Australia, 5005
- Institute for Photonics and Advanced Sensing (IPAS), University of Adelaide, Adelaide, Australia, 5005
| | - Yin Ying Ho
- Adelaide Proteomics Centre, School of Biological Sciences, University of Adelaide, Adelaide, Australia, 5005
| | - Gurjeet Kaur
- Institute for Research in Molecular Medicine (INFORMM), Universiti Sains Malaysia, Pulau Pinang, Malaysia
| | - Martin K Oehler
- Department of Gynaecological Oncology, Royal Adelaide Hospital, Adelaide, South Australia, 5005, Australia
- Robinson Institute, University of Adelaide, Adelaide, Australia, 5005
| | - Arun V Everest-Dass
- ARC Centre for Nanoscale BioPhotonics (CNBP), University of Adelaide, Adelaide, Australia, 5005
- Biomolecular Frontiers Research Centre, Macquarie University, Sydney, Australia, 2109
| | - Nicolle H Packer
- ARC Centre for Nanoscale BioPhotonics (CNBP), University of Adelaide, Adelaide, Australia, 5005
- Biomolecular Frontiers Research Centre, Macquarie University, Sydney, Australia, 2109
| | - Peter Hoffmann
- Adelaide Proteomics Centre, School of Biological Sciences, University of Adelaide, Adelaide, Australia, 5005
- Institute for Photonics and Advanced Sensing (IPAS), University of Adelaide, Adelaide, Australia, 5005
| |
Collapse
|
30
|
Epigenetic Bases of Aberrant Glycosylation in Cancer. Int J Mol Sci 2017; 18:ijms18050998. [PMID: 28481247 PMCID: PMC5454911 DOI: 10.3390/ijms18050998] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2017] [Revised: 04/27/2017] [Accepted: 05/02/2017] [Indexed: 02/07/2023] Open
Abstract
In this review, the sugar portions of glycoproteins, glycolipids, and glycosaminoglycans constitute the glycome, and the genes involved in their biosynthesis, degradation, transport and recognition are referred to as “glycogenes“. The extreme complexity of the glycome requires the regulatory layer to be provided by the epigenetic mechanisms. Almost all types of cancers present glycosylation aberrations, giving rise to phenotypic changes and to the expression of tumor markers. In this review, we discuss how cancer-associated alterations of promoter methylation, histone methylation/acetylation, and miRNAs determine glycomic changes associated with the malignant phenotype. Usually, increased promoter methylation and miRNA expression induce glycogene silencing. However, treatment with demethylating agents sometimes results in silencing, rather than in a reactivation of glycogenes, suggesting the involvement of distant methylation-dependent regulatory elements. From a therapeutic perspective aimed at the normalization of the malignant glycome, it appears that miRNA targeting of cancer-deranged glycogenes can be a more specific and promising approach than the use of drugs, which broad target methylation/acetylation. A very specific type of glycosylation, the addition of GlcNAc to serine or threonine (O-GlcNAc), is not only regulated by epigenetic mechanisms, but is an epigenetic modifier of histones and transcription factors. Thus, glycosylation is both under the control of epigenetic mechanisms and is an integral part of the epigenetic code.
Collapse
|
31
|
Zhang S, Lu J, Xu Z, Zou X, Sun X, Xu Y, Shan A, Lu J, Yan X, Cui Y, Yan W, Du Y, Gu J, Zheng M, Feng B, Zhang Y. Differential expression of ST6GAL1 in the tumor progression of colorectal cancer. Biochem Biophys Res Commun 2017; 486:1090-1096. [PMID: 28377225 DOI: 10.1016/j.bbrc.2017.03.167] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2017] [Accepted: 03/31/2017] [Indexed: 12/21/2022]
Abstract
Elevated expression of β-galactoside α2,6-sialyltranferase 1 (ST6GAL1) has been observed in colorectal cancer (CRC) and demonstrated to be important for its tumorigenesis. Here, we found that ST6GAL1 expression was significantly higher in non-metastatic tumors (stage I and II) than that in metastatic tumors (stage III and IV) using 62 pair-matched tumor/normal tissues. To elucidate the molecular mechanisms of how ST6GAL1 affected the CRC progression, we performed a global identification of the substrates of ST6GAL1 in the colon adenocarcinoma cell line SW480. A total of 318 membrane proteins were identified differentially affected by ST6GAL1 overexpression using metabolic labeling and proteomic analysis. Subsequent bioinformatic analysis revealed a list of potential substrates that might mediate the different functions of ST6GAL1 in CRC including cell movement, cell death and survival. Taken together, these results indicate a dynamic change in the expression of ST6GAL1 during the CRC progression and provide a list of sialylated proteins potentially relevant to the different functions of ST6GAL1 in CRC.
Collapse
Affiliation(s)
- Sen Zhang
- Department of General Surgery, Shanghai Minimally Invasive Surgery Center, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197, Ruijin Er Road, Shanghai 200025, China
| | - Jishun Lu
- Key Laboratory of Systems Biomedicine (Ministry of Education), and Collaborative Innovation Center of Systems Biomedicine, Shanghai Center for Systems Biomedicine (SCSB), Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai 200240, China; Division of Regulatory Glycobiology, Institute of Molecular Biomembrane and Glycobiology, Tohoku Medical and Pharmaceutical University, 4-4-1 Komatsushima, Aobaku, Sendai 981-8558, Miyagi, Japan
| | - Zhijue Xu
- Key Laboratory of Systems Biomedicine (Ministry of Education), and Collaborative Innovation Center of Systems Biomedicine, Shanghai Center for Systems Biomedicine (SCSB), Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai 200240, China
| | - Xia Zou
- Department of General Surgery, Shanghai Minimally Invasive Surgery Center, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197, Ruijin Er Road, Shanghai 200025, China; Key Laboratory of Systems Biomedicine (Ministry of Education), and Collaborative Innovation Center of Systems Biomedicine, Shanghai Center for Systems Biomedicine (SCSB), Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai 200240, China
| | - Xue Sun
- State Key Lab of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, China
| | - Yingjiao Xu
- Key Laboratory of Systems Biomedicine (Ministry of Education), and Collaborative Innovation Center of Systems Biomedicine, Shanghai Center for Systems Biomedicine (SCSB), Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai 200240, China
| | - Aidong Shan
- Key Laboratory of Systems Biomedicine (Ministry of Education), and Collaborative Innovation Center of Systems Biomedicine, Shanghai Center for Systems Biomedicine (SCSB), Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai 200240, China
| | - Jiaoyang Lu
- Department of General Surgery, Shanghai Minimally Invasive Surgery Center, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197, Ruijin Er Road, Shanghai 200025, China
| | - Xialin Yan
- Department of General Surgery, Shanghai Minimally Invasive Surgery Center, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197, Ruijin Er Road, Shanghai 200025, China
| | - Yalu Cui
- Key Laboratory of Systems Biomedicine (Ministry of Education), and Collaborative Innovation Center of Systems Biomedicine, Shanghai Center for Systems Biomedicine (SCSB), Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai 200240, China
| | - Wei Yan
- Key Laboratory of Systems Biomedicine (Ministry of Education), and Collaborative Innovation Center of Systems Biomedicine, Shanghai Center for Systems Biomedicine (SCSB), Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai 200240, China
| | - Yuguo Du
- State Key Lab of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, China
| | - Jianguo Gu
- Division of Regulatory Glycobiology, Institute of Molecular Biomembrane and Glycobiology, Tohoku Medical and Pharmaceutical University, 4-4-1 Komatsushima, Aobaku, Sendai 981-8558, Miyagi, Japan; Department of Pharmacology, Pharmacy College, Nantong University, Nantong 226001, Jiangsu, China
| | - Minhua Zheng
- Department of General Surgery, Shanghai Minimally Invasive Surgery Center, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197, Ruijin Er Road, Shanghai 200025, China
| | - Bo Feng
- Department of General Surgery, Shanghai Minimally Invasive Surgery Center, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197, Ruijin Er Road, Shanghai 200025, China.
| | - Yan Zhang
- Key Laboratory of Systems Biomedicine (Ministry of Education), and Collaborative Innovation Center of Systems Biomedicine, Shanghai Center for Systems Biomedicine (SCSB), Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai 200240, China.
| |
Collapse
|
32
|
Corfield A. Eukaryotic protein glycosylation: a primer for histochemists and cell biologists. Histochem Cell Biol 2017; 147:119-147. [PMID: 28012131 PMCID: PMC5306191 DOI: 10.1007/s00418-016-1526-4] [Citation(s) in RCA: 90] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/25/2016] [Indexed: 12/21/2022]
Abstract
Proteins undergo co- and posttranslational modifications, and their glycosylation is the most frequent and structurally variegated type. Histochemically, the detection of glycan presence has first been performed by stains. The availability of carbohydrate-specific tools (lectins, monoclonal antibodies) has revolutionized glycophenotyping, allowing monitoring of distinct structures. The different types of protein glycosylation in Eukaryotes are described. Following this educational survey, examples where known biological function is related to the glycan structures carried by proteins are given. In particular, mucins and their glycosylation patterns are considered as instructive proof-of-principle case. The tissue and cellular location of glycoprotein biosynthesis and metabolism is reviewed, with attention to new findings in goblet cells. Finally, protein glycosylation in disease is documented, with selected examples, where aberrant glycan expression impacts on normal function to let disease pathology become manifest. The histological applications adopted in these studies are emphasized throughout the text.
Collapse
Affiliation(s)
- Anthony Corfield
- Mucin Research Group, School of Clinical Sciences, Bristol Royal Infirmary, University of Bristol, Bristol, BS2 8HW, UK.
| |
Collapse
|
33
|
Sethi MK, Hancock WS, Fanayan S. Identifying N-Glycan Biomarkers in Colorectal Cancer by Mass Spectrometry. Acc Chem Res 2016; 49:2099-2106. [PMID: 27653471 DOI: 10.1021/acs.accounts.6b00193] [Citation(s) in RCA: 69] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Colorectal cancer (CRC) is one of the most prevalent cancers worldwide. Delineating biological markers (biomarkers) for early detection, when treatment is most effective, is key to prevention and long-term survival of patients. Development of reliable biomarkers requires an increased understanding of the CRC biology and the underlying molecular and cellular mechanisms of the disease. With recent advances in new technologies and approaches, tremendous efforts have been put in proteomics and genomics fields to deliver detailed analysis of the two major biomolecules, genes and proteins, to gain a more complete understanding of cellular systems at both genomic and proteomic levels, allowing a mechanistic understanding of the human diseases, including cancer, and opening avenues for identification of novel gene and protein based prognostic and therapeutic markers. Although the importance of glycosylation in modulating protein function has long been appreciated, glycan analysis has been complicated by the diversity of the glycan structures and the large number of potential glycosylation combinations. Driven by recent technological advances, LC-MS/MS based glycomics is gaining momentum in cancer research and holds considerable potential to deliver new glycan-based markers. In our laboratory, we investigated alterations in N-glycosylation associated with CRC malignancy in a panel of CRC cell lines and CRC patient tissues. In an initial study, LC-MS/MS-based N-glycomics were utilized to map the N-glycome landscape associated with a panel of CRC cell lines (LIM1215, LIM1899, and LIM2405). These studies were subsequently extended to paired tumor and nontumorigenic CRC tissues to validate the findings in the cell line. Our studies in both CRC cell lines and tissues identified a strong representation of high mannose and α2,6-linked sialylated complex N-glycans, which corroborate findings from previous studies in CRC and other cancers. In addition, certain unique glycan determinants such as bisecting β1,4-GlcNAcylation and α2,3-sialylation, identified in the metastatic (LIM1215) and aggressive (LIM2405) CRC cell lines, respectively, were shown to be associated with epidermal growth factor receptor (EGFR) expression status. In this Account, we will describe the mass spectrometry based N-glycomics approach utilized in our laboratory to accurately profile the cell- and tissue-specific N-glycomes associated with CRC. We will highlight altered N-glycosylation observed by our studies, consistent with findings from other cancer studies, and discuss how the observed alterations can provide insights into CRC pathogenesis, opening new avenues to identify novel disease-associated glycan markers.
Collapse
Affiliation(s)
- Manveen K. Sethi
- Department
of Chemistry and Biomolecular Sciences, Macquarie University, North Ryde, NSW 2109, Australia
| | - William S. Hancock
- Barnett
Institute and Department of Chemistry and Chemical Biology, Northeastern University, Boston, Massachusetts 02115, United States
- Department
of Biomedical Sciences, Macquarie University, North Ryde, NSW 2109, Australia
| | - Susan Fanayan
- Department
of Biomedical Sciences, Macquarie University, North Ryde, NSW 2109, Australia
| |
Collapse
|
34
|
Jung YR, Park JJ, Jin YB, Cao YJ, Park MJ, Kim EJ, Lee M. Silencing of ST6Gal I enhances colorectal cancer metastasis by down-regulating KAI1 via exosome-mediated exportation and thereby rescues integrin signaling. Carcinogenesis 2016; 37:1089-1097. [PMID: 27559112 DOI: 10.1093/carcin/bgw091] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2016] [Revised: 07/31/2016] [Accepted: 08/20/2016] [Indexed: 12/24/2022] Open
Abstract
Aberrant sialylation has long been correlated with human cancer. Increased ST6 Gal I (β-galactoside α 2, 6 sialyltransferase) and consequently higher levels of cell-surface α 2, 6 sialylation has been associated with human colorectal cancer (CRC) metastasis. We have extensive circumstantial data that sialylation is connected to cancer metastasis, but we do not understand in detail how sialylation can switch on/off multiple steps in cancer metastasis. To investigate the molecular mechanism underlying the ST6Gal I-mediated metastasis of CRC, we silenced the ST6Gal I gene in a metastatic SW620 CRC cell line (SW620-shST6Gal I) and examined the metastatic behavior of the cells. We found that various hallmarks of metastatic ability were considerably enhanced in ST6Gal 1-depleted SW620 clones, as assessed both in vitro and in vivo . In particular, the metastasis suppressor, KAI1, was down-regulated in ST6Gal I-deficient SW620 clones. This reflected the increased exosome-mediated exportation of KAI1, and was associated with a decrease in the KAI1-mediated inhibition of integrin. These findings indicate that gene silencing of ST6Gal I could enhance metastasis of CRC by down-regulating KAI1 activity and rescuing its negative effects on integrin signaling.
Collapse
Affiliation(s)
| | - Jung-Jin Park
- Department of Biochemistry and Medical Research Center , College of Medicine , Chungbuk National University , Cheongju 28644 , Republic of Korea
| | - Yeung Bae Jin
- National Primate Research Center , Korea Research Institute of Bioscience and Biotechnology , Cheongju 28116 , Republic of Korea
| | - Yuan Jie Cao
- Department of Radiation Oncology , Tianjin Medical University Cancer Institute and Hospital , National Clinical Research Center for Cancer and Tianjin Key laboratory of Cancer Prevention and Therapy , Huan-Hu-Xi Road , Ti-Yuan-Bei , He Xi District , Tianjin 300060 , P.R. China and
| | - Myung-Jin Park
- Division of Radiation Cancer Research , Korea Institute of Radiological and Medical Sciences , Seoul 01812 , Republic of Korea
| | | | | |
Collapse
|
35
|
Kuo JCH, Ibrahim AEK, Dawson S, Parashar D, Howat WJ, Guttula K, Miller R, Fearnhead NS, Winton DJ, Neves AA, Brindle KM. Detection of colorectal dysplasia using fluorescently labelled lectins. Sci Rep 2016; 6:24231. [PMID: 27071814 PMCID: PMC4829854 DOI: 10.1038/srep24231] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2015] [Accepted: 03/22/2016] [Indexed: 12/26/2022] Open
Abstract
Colorectal cancer screening using conventional colonoscopy lacks molecular information and can miss dysplastic lesions. We tested here the ability of fluorescently labelled lectins to distinguish dysplasia from normal tissue when sprayed on to the luminal surface epithelium of freshly resected colon tissue from the Apc(min) mouse and when applied to fixed human colorectal tissue sections. Wheat germ agglutinin (WGA) showed significantly decreased binding to adenomas in the mouse tissue and in sections of human colon from 47 patients. Changes in WGA binding to the human surface epithelium allowed regions containing normal epithelium (NE) or hyperplastic polyps (HP) to be distinguished from regions containing low-grade dysplasia (LGD), high-grade dysplasia (HGD) or carcinoma (C), with 81% sensitivity, 87% specificity and 93% positive predictive value (PPV). Helix pomatia agglutinin (HGA) distinguished epithelial regions containing NE from regions containing HP, LGD, HGD or C, with 89% sensitivity, 87% specificity and 97% PPV. The decreased binding of WGA and HPA to the luminal surface epithelium in human dysplasia suggests that these lectins may enable more sensitive detection of disease in the clinic using fluorescence colonoscopy.
Collapse
Affiliation(s)
- Joe Chin-Hun Kuo
- Cancer Research UK Cambridge Institute, University of Cambridge, Li Ka Shing Centre, Cambridge, UK
- Department of Biochemistry, University of Cambridge, Cambridge, UK
| | - Ashraf E. K. Ibrahim
- Department of Pathology, Division of Molecular Histopathology, University of Cambridge, Addenbrooke’s Hospital, Cambridge, UK
- MRC, Laboratory of Molecular Biology, Hills Road, Cambridge, UK
| | - Sarah Dawson
- Cambridge Clinical Trials Unit, University of Cambridge, Cambridge, UK
| | - Deepak Parashar
- Statistics and Epidemiology Unit & Cancer Research Centre, Division of Health Sciences, Warwick Medical School, The University of Warwick, Coventry, UK
| | - William J. Howat
- Cancer Research UK Cambridge Institute, University of Cambridge, Li Ka Shing Centre, Cambridge, UK
| | - Kiran Guttula
- Department of Pathology, Division of Molecular Histopathology, University of Cambridge, Addenbrooke’s Hospital, Cambridge, UK
| | - Richard Miller
- Cambridge Colorectal Unit, Addenbrooke’s Hospital, Cambridge, UK
| | | | - Douglas J. Winton
- Cancer Research UK Cambridge Institute, University of Cambridge, Li Ka Shing Centre, Cambridge, UK
| | - André A. Neves
- Cancer Research UK Cambridge Institute, University of Cambridge, Li Ka Shing Centre, Cambridge, UK
| | - Kevin M. Brindle
- Cancer Research UK Cambridge Institute, University of Cambridge, Li Ka Shing Centre, Cambridge, UK
- Department of Biochemistry, University of Cambridge, Cambridge, UK
| |
Collapse
|
36
|
Sethi MK, Fanayan S. Mass Spectrometry-Based N-Glycomics of Colorectal Cancer. Int J Mol Sci 2015; 16:29278-304. [PMID: 26690136 PMCID: PMC4691109 DOI: 10.3390/ijms161226165] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2015] [Revised: 11/23/2015] [Accepted: 12/01/2015] [Indexed: 12/19/2022] Open
Abstract
Colorectal cancer (CRC) is one of the most prevalent cancers worldwide. An increased molecular understanding of the CRC pathology is warranted to gain insights into the underlying molecular and cellular mechanisms of the disease. Altered protein glycosylation patterns are associated with most diseases including malignant transformation. Recent advances in mass spectrometry and bioinformatics have accelerated glycomics research and present a new paradigm for cancer biomarker discovery. Mass spectrometry (MS)-based glycoproteomics and glycomics, therefore, hold considerable promise to improve the discovery of novel biomarkers with utility in disease diagnosis and therapy. This review focuses on the emerging field of glycomics to present a comprehensive review of advances in technologies and their application in studies aimed at discovering novel glycan-based biomarkers. We will also discuss some of the challenges associated with using glycans as biomarkers.
Collapse
Affiliation(s)
- Manveen K Sethi
- Department of Chemistry and Biomolecular Sciences, Macquarie University, North Ryde, NSW 2109, Australia.
| | - Susan Fanayan
- Department of Biomedical Sciences, Macquarie University, North Ryde, NSW 2109, Australia.
| |
Collapse
|
37
|
Ultrasensitive electrochemical biosensor based on reduced graphene oxide-tetraethylene pentamine-BMIMPF6 hybrids for the detection of α2,6-sialylated glycans in human serum. Biosens Bioelectron 2015; 74:953-9. [DOI: 10.1016/j.bios.2015.07.073] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2015] [Revised: 07/26/2015] [Accepted: 07/30/2015] [Indexed: 01/24/2023]
|
38
|
Li W, Niu Y, Xiong DC, Cao X, Ye XS. Highly Substituted Cyclopentane-CMP Conjugates as Potent Sialyltransferase Inhibitors. J Med Chem 2015; 58:7972-90. [PMID: 26406919 DOI: 10.1021/acs.jmedchem.5b01181] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Sialylconjugates on cell surfaces are involved in many biological events such as cellular recognition, signal transduction, and immune response. It has been reported that aberrant sialylation at the nonreducing end of glycoconjugates and overexpression of sialyltransferases (STs) in cells are correlated with the malignance, invasion, and metastasis of tumors. Therefore, inhibitors of STs would provide valuable leads for the discovery of antitumor drugs. On the basis of the transition state of the enzyme-catalyzed sialylation reaction, we proposed that the cyclopentane skeleton in its two puckered conformations might mimic the planar structure of the donor (CMP-Neu5Ac) in the transition state. A series of cyclopentane-containing compounds were designed and synthesized by coupling different cyclopentane α-hydroxyphosphonates with cytidine phosphoramidite. Their inhibitory activities against recombinant human ST6Gal-I were assayed, and a potent inhibitor 48l with a Ki of 0.028 ± 0.006 μM was identified. The results show that the cyclopentanoid-type compounds could become a new type of sialyltransferase inhibitors as biological probes or drug leads.
Collapse
Affiliation(s)
- Wenming Li
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, and Center for Molecular and Translational Medicine, Peking University , Xue Yuan Road No. 38, Beijing 100191, China
| | - Youhong Niu
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, and Center for Molecular and Translational Medicine, Peking University , Xue Yuan Road No. 38, Beijing 100191, China.,State Key Laboratory of Applied Organic Chemistry and College of Chemistry and Chemical Engineering, Lanzhou University , Lanzhou 730000, China
| | - De-Cai Xiong
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, and Center for Molecular and Translational Medicine, Peking University , Xue Yuan Road No. 38, Beijing 100191, China
| | - Xiaoping Cao
- State Key Laboratory of Applied Organic Chemistry and College of Chemistry and Chemical Engineering, Lanzhou University , Lanzhou 730000, China
| | - Xin-Shan Ye
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, and Center for Molecular and Translational Medicine, Peking University , Xue Yuan Road No. 38, Beijing 100191, China
| |
Collapse
|
39
|
Lu J, Gu J. Significance of β-Galactoside α2,6 Sialyltranferase 1 in Cancers. Molecules 2015; 20:7509-27. [PMID: 25919275 PMCID: PMC6272632 DOI: 10.3390/molecules20057509] [Citation(s) in RCA: 93] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2015] [Revised: 04/17/2015] [Accepted: 04/22/2015] [Indexed: 12/16/2022] Open
Abstract
Altered glycosylation is a common feature of cancer cells. It takes a variety of forms, which includes loss of expression or excessive expression of some structures, the accumulation of precursors, the appearance of novel structures, etc. Notably, these changes in glycan structure do not occur as a random consequence of disorder biology. Only a limited subset of oligosaccharides is found frequently enriched on the tumor cell surface and implicated in different tumor phenotypes. Among these, altered sialylation has long been associated with metastatic cell behaviors such as invasion and enhanced cell survival and accumulating evidence points to the alteration occurring in the sialic acid linkage to other sugars, which normally exists in three main configurations: α2,3, α2,6, and α2,8, catalyzed by a group of sialyltransferases. The aberrant expression of all three configurations has been described in cancer progression. However, the increased α2,6 sialylation catalyzed by β-galactoside α2,6 sialyltranferase 1 (ST6Gal I) is frequently observed in many types of the cancers. In this review, we describe the findings on the role of ST6Gal I in cancer progression, and highlight in particular the knowledge of how ST6Gal I-mediated α2,6 sialylated glycans or sialylated carrier proteins regulate cell signaling to promote the malignant phenotype of human carcinoma.
Collapse
Affiliation(s)
- Jishun Lu
- Division of Regulatory Glycobiology, Institute of Molecular Biomembrane and Glycobiology, Tohoku Pharmaceutical University, 4-4-1 Komatsushima, Aobaku, Sendai, Miyagi 981-8558, Japan.
| | - Jianguo Gu
- Division of Regulatory Glycobiology, Institute of Molecular Biomembrane and Glycobiology, Tohoku Pharmaceutical University, 4-4-1 Komatsushima, Aobaku, Sendai, Miyagi 981-8558, Japan.
| |
Collapse
|
40
|
Abstract
Glycans on proteins and lipids are known to alter with malignant transformation. The study of these may contribute to the discovery of biomarkers and treatment targets as well as understanding of cancer biology. We here describe the change of glycosylation specifically defining colorectal cancer with view on N-glycans, O-glycans, and glycosphingolipid glycans in colorectal cancer cells and tissues as well as patient sera. Glycan alterations observed in colon cancer include increased β1,6-branching and correlating higher abundance of (poly-)N-acetyllactosamine extensions of N-glycans as well as an increase in (truncated) high-mannose type glycans, while bisected structures decrease. Colorectal cancer-associated O-glycan changes are predominated by reduced expression of core 3 and 4 glycans, whereas higher levels of core 1 glycans, (sialyl) T-antigen, (sialyl) Tn-antigen, and a generally higher density of O-glycans are observed. Specific changes for glycosphingolipid glycans are lower abundances of disialylated structures as well as globo-type glycosphingolipid glycans with exception of Gb3. In general, alterations affecting all discussed glycan types are increased sialylation, fucosylation as well as (sialyl) Lewis-type antigens and type-2 chain glycans. As a consequence, interactions with glycan-binding proteins can be affected and the biological function and cellular consequences of the altered glycosylation with regard to tumorigenesis, metastasis, modulation of immunity, and resistance to antitumor therapy will be discussed. Finally, analytical approaches aiding in the field of glycomics will be reviewed with focus on binding assays and mass spectrometry.
Collapse
Affiliation(s)
- Stephanie Holst
- Center for Proteomics and Metabolomics, Leiden University Medical Center, Leiden, The Netherlands.
| | - Manfred Wuhrer
- Center for Proteomics and Metabolomics, Leiden University Medical Center, Leiden, The Netherlands; Department of Molecular Cell Biology and Immunology, VU University Medical Center, Amsterdam, The Netherlands; Division of BioAnalytical Chemistry, VU University, Amsterdam, The Netherlands
| | - Yoann Rombouts
- Center for Proteomics and Metabolomics, Leiden University Medical Center, Leiden, The Netherlands
| |
Collapse
|
41
|
Zhang Z, Sun Z, Zhu J, Liu J, Huang G, Ye M, Zou H. High-Throughput Determination of the Site-Specific N-Sialoglycan Occupancy Rates by Differential Oxidation of Glycoproteins Followed with Quantitative Glycoproteomics Analysis. Anal Chem 2014; 86:9830-7. [DOI: 10.1021/ac5024638] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Affiliation(s)
- Zhang Zhang
- CAS Key Laboratory of Separation Sciences for Analytical Chemistry, National Chromatographic R&A Center, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian 116023, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Zhen Sun
- CAS Key Laboratory of Separation Sciences for Analytical Chemistry, National Chromatographic R&A Center, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian 116023, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Jun Zhu
- CAS Key Laboratory of Separation Sciences for Analytical Chemistry, National Chromatographic R&A Center, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian 116023, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Jing Liu
- CAS Key Laboratory of Separation Sciences for Analytical Chemistry, National Chromatographic R&A Center, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian 116023, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Guang Huang
- CAS Key Laboratory of Separation Sciences for Analytical Chemistry, National Chromatographic R&A Center, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian 116023, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Mingliang Ye
- CAS Key Laboratory of Separation Sciences for Analytical Chemistry, National Chromatographic R&A Center, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian 116023, China
| | - Hanfa Zou
- CAS Key Laboratory of Separation Sciences for Analytical Chemistry, National Chromatographic R&A Center, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian 116023, China
| |
Collapse
|
42
|
Dall'Olio F, Malagolini N, Trinchera M, Chiricolo M. Sialosignaling: Sialyltransferases as engines of self-fueling loops in cancer progression. Biochim Biophys Acta Gen Subj 2014; 1840:2752-64. [PMID: 24949982 DOI: 10.1016/j.bbagen.2014.06.006] [Citation(s) in RCA: 99] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2014] [Revised: 05/13/2014] [Accepted: 06/10/2014] [Indexed: 02/03/2023]
|
43
|
Increased expression of MUC1 and sialyl Lewis antigens in different areas of clear renal cell carcinoma. Clin Exp Nephrol 2014; 19:732-7. [DOI: 10.1007/s10157-014-1013-y] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2014] [Accepted: 07/08/2014] [Indexed: 01/26/2023]
|
44
|
Molecular Design of Fluorescent Labeled Glycosides as Acceptor Substrates for Sialyltransferases. Biosci Biotechnol Biochem 2014; 74:2287-92. [DOI: 10.1271/bbb.100505] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
|
45
|
Häuselmann I, Borsig L. Altered tumor-cell glycosylation promotes metastasis. Front Oncol 2014; 4:28. [PMID: 24592356 PMCID: PMC3923139 DOI: 10.3389/fonc.2014.00028] [Citation(s) in RCA: 282] [Impact Index Per Article: 25.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2013] [Accepted: 01/29/2014] [Indexed: 12/14/2022] Open
Abstract
Malignant transformation of cells is associated with aberrant glycosylation presented on the cell-surface. Commonly observed changes in glycan structures during malignancy encompass aberrant expression and glycosylation of mucins; abnormal branching of N-glycans; and increased presence of sialic acid on proteins and glycolipids. Accumulating evidence supports the notion that the presence of certain glycan structures correlates with cancer progression by affecting tumor-cell invasiveness, ability to disseminate through the blood circulation and to metastasize in distant organs. During metastasis tumor-cell-derived glycans enable binding to cells in their microenvironment including endothelium and blood constituents through glycan-binding receptors – lectins. In this review, we will discuss current concepts how tumor-cell-derived glycans contribute to metastasis with the focus on three types of lectins: siglecs, galectins, and selectins. Siglecs are present on virtually all hematopoietic cells and usually negatively regulate immune responses. Galectins are mostly expressed by tumor cells and support tumor-cell survival. Selectins are vascular adhesion receptors that promote tumor-cell dissemination. All lectins facilitate interactions within the tumor microenvironment and thereby promote cancer progression. The identification of mechanisms how tumor glycans contribute to metastasis may help to improve diagnosis, prognosis, and aid to develop clinical strategies to prevent metastasis.
Collapse
Affiliation(s)
- Irina Häuselmann
- Zürich Center for Integrative Human Physiology, Institute of Physiology, University of Zürich , Zürich , Switzerland
| | - Lubor Borsig
- Zürich Center for Integrative Human Physiology, Institute of Physiology, University of Zürich , Zürich , Switzerland
| |
Collapse
|
46
|
Kumar R, Nasi R, Bhasin M, Huan Khieu N, Hsieh M, Gilbert M, Jarrell H, Zou W, Jennings HJ. Sialyltransferase inhibitors: consideration of molecular shape and charge/hydrophobic interactions. Carbohydr Res 2013; 378:45-55. [DOI: 10.1016/j.carres.2012.12.017] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2012] [Revised: 12/10/2012] [Accepted: 12/13/2012] [Indexed: 10/27/2022]
|
47
|
Abstract
Tumor cells exhibit striking changes in cell surface glycosylation as a consequence of dysregulated glycosyltransferases and glycosidases. In particular, an increase in the expression of certain sialylated glycans is a prominent feature of many transformed cells. Altered sialylation has long been associated with metastatic cell behaviors including invasion and enhanced cell survival; however, there is limited information regarding the molecular details of how distinct sialylated structures or sialylated carrier proteins regulate cell signaling to control responses such as adhesion/migration or resistance to specific apoptotic pathways. The goal of this review is to highlight selected examples of sialylated glycans for which there is some knowledge of molecular mechanisms linking aberrant sialylation to critical processes involved in metastasis.
Collapse
Affiliation(s)
- Matthew J Schultz
- Department of Cell, Developmental and Integrative Biology, University of Alabama at Birmingham, MCLM 982A 1918 University Boulevard, Birmingham, AL 35294-0005, USA
| | | | | |
Collapse
|
48
|
Wu J, Xie X, Nie S, Buckanovich RJ, Lubman DM. Altered expression of sialylated glycoproteins in ovarian cancer sera using lectin-based ELISA assay and quantitative glycoproteomics analysis. J Proteome Res 2013; 12:3342-52. [PMID: 23731285 DOI: 10.1021/pr400169n] [Citation(s) in RCA: 55] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Herein, we identify and confirm differentially expressed sialoglycoproteins in the serum of patients with ovarian cancer. On the basis of Sambucus nigra (SNA) lectin enrichment and on an isobaric chemical labeling quantitative strategy, clusterin (CLUS), leucine-rich alpha-2-glycoprotein (LRG1), hemopexin (HEMO), vitamin D-binding protein (VDB), and complement factor H (CFH) were found to be differentially expressed in the serum of patients with ovarian cancer compared to benign diseases. The abnormal sialylation levels of CLUS, CFH, and HEMO in serum of ovarian cancer patients were verified by a lectin-based ELISA assay. ELISA assays were further applied to measure total protein level changes of these glycoproteins. Protein levels of CLUS were found to be down-regulated in the serum of ovarian cancer patients, while protein levels of LRG1 were increased. The combination of CLUS and LRG1 (AUC = 0.837) showed improved performance for distinguishing stage III ovarian cancer from benign diseases compared to CA125 alone (AUC = 0.811). In differentiating early stage ovarian cancer from benign diseases or healthy controls, LRG1 showed comparable performance to CA125. An independent sample set was further used to confirm the ability of these candidate markers to detect patients with ovarian cancer. Our study provides a comprehensive strategy for the identification of candidate biomarkers that show the potential for diagnosis of ovarian cancer. Further studies using a large number of samples are necessary to validate the utility of this panel of proteins.
Collapse
Affiliation(s)
- Jing Wu
- Department of Surgery, University of Michigan, Ann Arbor, Michigan 48109, USA
| | | | | | | | | |
Collapse
|
49
|
Shah P, Yang S, Sun S, Aiyetan P, Yarema KJ, Zhang H. Mass spectrometric analysis of sialylated glycans with use of solid-phase labeling of sialic acids. Anal Chem 2013; 85:3606-13. [PMID: 23445396 PMCID: PMC3681956 DOI: 10.1021/ac3033867] [Citation(s) in RCA: 70] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
The analysis of sialylated glycans is critical for understanding the role of sialic acid in normal biological processes as well as in disease. However, the labile nature of sialic acid typically renders routine analysis of this monosaccharide by mass spectrometric methods difficult. To overcome this difficulty we pursued derivatization methodologies, extending established acetohydrazide approaches to aniline-based methods, and finally to optimized p-toluidine derivatization. This new quantitative glycoform profiling method with use of MALDI-TOF in positive ion mode was validated by first comparing N-glycans isolated from fetuin and serum and was then exploited to analyze the effects of increased metabolic flux through the sialic acid pathway in SW1990 pancreatic cancer cells by using a colabeling strategy with light and heavy toluidine. The latter results established that metabolic flux, in a complementary manner to the more well-known impact of sialyltransferase expression, can critically modulate the sialylation of specific glycans while leaving others virtually unchanged.
Collapse
Affiliation(s)
- Punit Shah
- Department of Pathology, Johns Hopkins University, Baltimore, MD 21231
| | - Shuang Yang
- Department of Pathology, Johns Hopkins University, Baltimore, MD 21231
| | - Shisheng Sun
- Department of Pathology, Johns Hopkins University, Baltimore, MD 21231
| | - Paul Aiyetan
- Department of Pathology, Johns Hopkins University, Baltimore, MD 21231
| | - Kevin J. Yarema
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD 21231
| | - Hui Zhang
- Department of Pathology, Johns Hopkins University, Baltimore, MD 21231
| |
Collapse
|
50
|
Borges CR, Rehder DS, Boffetta P. Multiplexed surrogate analysis of glycotransferase activity in whole biospecimens. Anal Chem 2013; 85:2927-36. [PMID: 23368525 PMCID: PMC3650733 DOI: 10.1021/ac3035579] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Dysregulated glycotransferase enzymes in cancer cells produce aberrant glycans--some of which can help facilitate metastases. Within a cell, individual glycotransferases promiscuously help to construct dozens of unique glycan structures, making it difficult to comprehensively track their activity in biospecimens--especially where they are absent or inactive. Here, we describe an approach to deconstruct glycans in whole biospecimens then analytically pool together resulting monosaccharide-and-linkage-specific degradation products ("glycan nodes") that directly represent the activities of specific glycotransferases. To implement this concept, a reproducible, relative quantitation-based glycan methylation analysis methodology was developed that simultaneously captures information from N-, O-, and lipid linked glycans and is compatible with whole biofluids and homogenized tissues; in total, over 30 different glycan nodes are detectable per gas chromatography-mass spectrometry (GC-MS) run. Numerous nonliver organ cancers are known to induce the production of abnormally glycosylated serum proteins. Thus, following analytical validation, in blood plasma, the technique was applied to a group of 59 lung cancer patient plasma samples and age/gender/smoking-status-matched non-neoplastic controls from the Lung Cancer in Central and Eastern Europe (CEE) study to gauge the clinical utility of the approach toward the detection of lung cancer. Ten smoking-independent glycan node ratios were found that detect lung cancer with individual receiver operating characteristic (ROC) c-statistics ranging from 0.76 to 0.88. Two glycan nodes provided novel evidence for altered ST6Gal-I and GnT-IV glycotransferase activities in lung cancer patients. In summary, a conceptually novel approach to the analysis of glycans in unfractionated human biospecimens has been developed that, upon clinical validation for specific applications, may provide diagnostic and/or predictive information in glycan-altering diseases.
Collapse
Affiliation(s)
- Chad R Borges
- Molecular Biomarkers Unit, The Biodesign Institute at Arizona State University, Tempe, Arizona 85287, United States.
| | | | | |
Collapse
|