1
|
Coccimiglio M, Chiodo F, van Kooyk Y. The sialic acid-Siglec immune checkpoint: an opportunity to enhance immune responses and therapy effectiveness in melanoma. Br J Dermatol 2024; 190:627-635. [PMID: 38197441 DOI: 10.1093/bjd/ljad517] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2023] [Revised: 12/13/2023] [Accepted: 12/17/2023] [Indexed: 01/11/2024]
Abstract
Modulation of immune responses through immune checkpoint blockade has revolutionized cutaneous melanoma treatment. However, it is still the case that not all patients respond successfully to these therapies, indicating the presence of as yet unknown resistance mechanisms. Hence, it is crucial to find novel targets to improve therapy efficacy. One of the described resistance mechanisms is regulated by immune inhibitory Siglec receptors, which are engaged by the carbohydrates sialic acids expressed on tumour cells, contributing to programmed cell death protein-1 (PD1)-like immune suppression mechanisms. In this review, we provide an overview on the regulation of sialic acid synthesis, its expression in melanoma, and the contribution of the sialic acid-Siglec axis to tumour development and immune suppressive mechanisms in the tumour microenvironment. Finally, we highlight potential sialic acid-Siglec axis-related therapeutics to improve the treatment of melanoma.
Collapse
Affiliation(s)
- Magali Coccimiglio
- Amsterdam University Medical Center, Vrije Universiteit Amsterdam, Department of Molecular Cell Biology and Immunology, Amsterdam, the Netherlands
- Cancer Center Amsterdam, Amsterdam, the Netherlands
- Amsterdam Institute for Infection and Immunity, Amsterdam, the Netherlands
| | - Fabrizio Chiodo
- Amsterdam University Medical Center, Vrije Universiteit Amsterdam, Department of Molecular Cell Biology and Immunology, Amsterdam, the Netherlands
- Cancer Center Amsterdam, Amsterdam, the Netherlands
- Amsterdam Institute for Infection and Immunity, Amsterdam, the Netherlands
- Italian National Research Council, Institute of Biomolecular Chemistry, Pozzuoli, Naples, Italy
| | - Yvette van Kooyk
- Amsterdam University Medical Center, Vrije Universiteit Amsterdam, Department of Molecular Cell Biology and Immunology, Amsterdam, the Netherlands
- Cancer Center Amsterdam, Amsterdam, the Netherlands
- Amsterdam Institute for Infection and Immunity, Amsterdam, the Netherlands
| |
Collapse
|
2
|
Lectin-Based Study Reveals the Presence of Disease-Relevant Glycoepitopes in Bladder Cancer Cells and Ectosomes. Int J Mol Sci 2022; 23:ijms232214368. [PMID: 36430846 PMCID: PMC9699364 DOI: 10.3390/ijms232214368] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2022] [Revised: 11/15/2022] [Accepted: 11/17/2022] [Indexed: 11/22/2022] Open
Abstract
Bladder cancer is a malignancy that remains a therapeutic challenge and requires the identification of new biomarkers and mechanisms of progression. Several studies showed that extracellular vesicles promote angiogenesis, migration and metastasis, and inhibit apoptosis in bladder cancer. This effect may depend on their glycosylation status. Thus, the aim of this study was to compare glycosylation profiles of T-24 urothelial bladder cancer cells, HCV-29 normal ureter epithelial cells, and ectosomes released by both cell lines using lectin blotting and flow cytometry. Ectosomes displayed distinct total and surface glycosylation profiles with abundance of β-1,6-branched glycans and sialilated structures. Then, it was investigated whether the glycosylation status of the T-24 and HCV-29 cells is responsible for the effect exerted by ectosomes on the proliferation and migration of recipient cells. Stronger proproliferative and promigratory activity of T-24-derived ectosomes was observed in comparison to ectosomes from HCV-29 cells. When ectosomes were isolated from DMJ-treated cells, the aforementioned effects were diminished, suggesting that glycans carried by ectosomes were involved in modulation of recipient cell function. HCV-29- and T-24-derived ectosomes also increased the viability and motility of endothelial HUVEC cells and Hs27 fibroblasts. This supports the hypothesis that ectosomes can modulate the function of various cells present in the tumor microenvironment.
Collapse
|
3
|
Marciel MP, Haldar B, Hwang J, Bhalerao N, Bellis SL. Role of tumor cell sialylation in pancreatic cancer progression. Adv Cancer Res 2022; 157:123-155. [PMID: 36725107 PMCID: PMC11342334 DOI: 10.1016/bs.acr.2022.07.003] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is one of the deadliest malignancies and is currently the third leading cause of cancer death. The aggressiveness of PDAC stems from late diagnosis, early metastasis, and poor efficacy of current chemotherapies. Thus, there is an urgent need for effective biomarkers for early detection of PDAC and development of new therapeutic strategies. It has long been known that cellular glycosylation is dysregulated in pancreatic cancer cells, however, tumor-associated glycans and their cognate glycosylating enzymes have received insufficient attention as potential clinical targets. Aberrant glycosylation affects a broad range of pathways that underpin tumor initiation, metastatic progression, and resistance to cancer treatment. One of the prevalent alterations in the cancer glycome is an enrichment in a select group of sialylated glycans including sialylated, branched N-glycans, sialyl Lewis antigens, and sialylated forms of truncated O-glycans such as the sialyl Tn antigen. These modifications affect the activity of numerous cell surface receptors, which collectively impart malignant characteristics typified by enhanced cell proliferation, migration, invasion and apoptosis-resistance. Additionally, sialic acids on tumor cells engage inhibitory Siglec receptors on immune cells to dampen anti-tumor immunity, further promoting cancer progression. The goal of this review is to summarize the predominant changes in sialylation occurring in pancreatic cancer, the biological functions of sialylated glycoproteins in cancer pathogenesis, and the emerging strategies for targeting sialoglycans and Siglec receptors in cancer therapeutics.
Collapse
Affiliation(s)
- Michael P Marciel
- Department of Cell, Developmental and Integrative Biology, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Barnita Haldar
- Department of Cell, Developmental and Integrative Biology, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Jihye Hwang
- Department of Cell, Developmental and Integrative Biology, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Nikita Bhalerao
- Department of Cell, Developmental and Integrative Biology, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Susan L Bellis
- Department of Cell, Developmental and Integrative Biology, University of Alabama at Birmingham, Birmingham, AL, United States.
| |
Collapse
|
4
|
Ziganshina MM, Kulikova GV, Shchegolev AI, Shmakov RG, Kan NE, Sukhikh GT. Comparative Characteristics of Sialoglycans Expression Disorders in the Placental Barrier Structures in Preeclampsia and Fetal Growth Restriction. Bull Exp Biol Med 2022; 173:270-275. [DOI: 10.1007/s10517-022-05532-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2021] [Indexed: 11/28/2022]
|
5
|
Li Z, Zhang J, Ai HW. Genetically Encoded Green Fluorescent Biosensors for Monitoring UDP-GlcNAc in Live Cells. ACS CENTRAL SCIENCE 2021; 7:1763-1770. [PMID: 34729420 PMCID: PMC8554846 DOI: 10.1021/acscentsci.1c00745] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/22/2021] [Indexed: 06/13/2023]
Abstract
Uridine diphosphate N-acetylglucosamine (UDP-GlcNAc) is a nucleotide sugar used by glycosyltransferases to synthesize glycoproteins, glycosaminoglycans, glycolipids, and glycoRNA. UDP-GlcNAc also serves as the donor substrate for forming O-GlcNAc, a dynamic intracellular protein modification involved in diverse signaling and disease processes. UDP-GlcNAc is thus a central metabolite connecting nutrition, metabolism, signaling, and disease. There is a great interest in monitoring UDP-GlcNAc in biological systems. Here, we present the first genetically encoded, green fluorescent UDP-GlcNAc sensor (UGAcS), an optimized insertion of a circularly permuted green fluorescent protein (cpGFP) into an inactive mutant of an Escherichia coli UDP-GlcNAc transferase, for ratiometric monitoring of UDP-GlcNAc dynamics in live mammalian cells. Although UGAcS responds to UDP-GlcNAc quite selectively among various nucleotide sugars, UDP and uridine triphosphate (UTP) interfere with the response. We thus developed another biosensor named UXPS, which is responsive to UDP and UTP but not UDP-GlcNAc. We demonstrated the use of the biosensors to follow UDP-GlcNAc levels in cultured mammalian cells perturbed with nutritional changes, pharmacological inhibition, and knockdown or overexpression of key enzymes in the UDP-GlcNAc synthesis pathway. We further utilized the biosensors to monitor UDP-GlcNAc concentrations in pancreatic MIN6 β-cells under various culture conditions.
Collapse
|
6
|
The Role of Glycosylation in Melanoma Progression. Cells 2021; 10:cells10082136. [PMID: 34440905 PMCID: PMC8393314 DOI: 10.3390/cells10082136] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2021] [Revised: 08/17/2021] [Accepted: 08/18/2021] [Indexed: 01/06/2023] Open
Abstract
Malignant melanoma is the most aggressive form of skin cancer, which originates from the malignant transformation of melanocytes, the melanin-producing cells of the skin. Melanoma progression is typically described as a stepwise process in which metastasis formation ensues late during disease. A large body of evidence has shown that the accumulation of genetic and epigenetic alterations drives melanoma progression through the different steps. Mortality in melanoma is associated with metastatic disease. Accordingly, early-stage melanoma can be cured in the majority of cases by surgical excision, while late-stage melanoma is a highly lethal disease. Glycosylation is a post-translational modification that involves the transfer of glycosyl moieties to specific amino acid residues of proteins to form glycosidic bonds through the activity of glycosyltransferases. Aberrant glycosylation is considered a hallmark of cancer as it occurs in the majority of tumor types, including melanoma. The most widely occurring glycosylation changes in melanoma are represented by sialylation, fucosylation, and N- and I-glycan branching. In this review, we discuss the role of glycosylation in melanoma and provide insights on the mechanisms by which aberrant glycosylation promotes melanoma progression through activation of invasion and metastasis, immune evasion and cell proliferation.
Collapse
|
7
|
Alginate oligosaccharide attenuates α2,6-sialylation modification to inhibit prostate cancer cell growth via the Hippo/YAP pathway. Cell Death Dis 2019; 10:374. [PMID: 31076566 PMCID: PMC6510775 DOI: 10.1038/s41419-019-1560-y] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2019] [Revised: 03/23/2019] [Accepted: 03/26/2019] [Indexed: 12/11/2022]
Abstract
Chitosan oligosaccharides have been reported to inhibit various tumors. However, the water-soluble marine plant oligosaccharide alginate oligosaccharide (AOS) has only rarely been reported to have anti-cancer effects. Moreover, the inhibitory effect of AOS on prostate cancer and the underlying molecular mechanism remain unknown. This study shows that AOS inhibited cell growth, which was consistent with the attenuation of α2,6-sialylation modification. Furthermore, AOS inhibited ST6Gal-1 promoter activity and thus affected transcriptional processes. In addition, AOS could activate the Hippo/YAP pathway and block the recruitment of both the coactivator YAP and c-Jun. Furthermore, YAP interacted with the transcription factor c-Jun and regulated the transcriptional activity of the downstream target ST6Gal-1 gene. Consistent with in vitro data, AOS suppressed the tumorigenicity of prostate cancer cells via the Hippo/YAP pathway in vivo. In summary, these data indicate that AOS slows the proliferation of prostate cancer and provides a basis for the healthy function of kelp in traditional cognition.
Collapse
|
8
|
Tang L, Chen X, Zhang X, Guo Y, Su J, Zhang J, Peng C, Chen X. N-Glycosylation in progression of skin cancer. Med Oncol 2019; 36:50. [PMID: 31037368 DOI: 10.1007/s12032-019-1270-4] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2019] [Accepted: 04/11/2019] [Indexed: 12/29/2022]
Abstract
Skin cancer can be classified as cutaneous malignant melanoma, basal cell carcinoma, and squamous cell carcinoma. Due to the high level of morbidity and mortality, skin cancer has become a global public health issue worldwide while the pathogenesis of skin cancer is still unclear. It is necessary to further identify the pathogenesis of skin cancer and find candidate targets to diagnose and treat skin cancer. A variety of factors are known to be associated with skin cancer including N-glycosylation, which partly explained the malignant behaviors of skin cancer. In this review, we retrieved databases such as PubMed and Web of Science to elucidate its relationship between glycosylation and skin cancer. We summarized some key glycosyltransferases and proteins during the process of N-glycosylation related to skin cancer, which was helpful to unmask the additional mechanism of skin cancer and find some novel targets of skin cancer.
Collapse
Affiliation(s)
- Ling Tang
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, Hunan, China
- Hunan Key Laboratory of Skin Cancer and Psoriasis, Changsha, Hunan, China
- Hunan Engineering Research Center of Skin Health and Disease, Changsha, Hunan, China
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Xiaoping Chen
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Xu Zhang
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, Hunan, China
- Hunan Key Laboratory of Skin Cancer and Psoriasis, Changsha, Hunan, China
- Hunan Engineering Research Center of Skin Health and Disease, Changsha, Hunan, China
| | - Yeye Guo
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, Hunan, China
- Hunan Key Laboratory of Skin Cancer and Psoriasis, Changsha, Hunan, China
- Hunan Engineering Research Center of Skin Health and Disease, Changsha, Hunan, China
| | - Juan Su
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, Hunan, China
- Hunan Key Laboratory of Skin Cancer and Psoriasis, Changsha, Hunan, China
- Hunan Engineering Research Center of Skin Health and Disease, Changsha, Hunan, China
| | - Jianglin Zhang
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, Hunan, China
- Hunan Key Laboratory of Skin Cancer and Psoriasis, Changsha, Hunan, China
- Hunan Engineering Research Center of Skin Health and Disease, Changsha, Hunan, China
| | - Cong Peng
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, Hunan, China.
- Hunan Key Laboratory of Skin Cancer and Psoriasis, Changsha, Hunan, China.
- Hunan Engineering Research Center of Skin Health and Disease, Changsha, Hunan, China.
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, Hunan, China.
| | - Xiang Chen
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, Hunan, China.
- Hunan Key Laboratory of Skin Cancer and Psoriasis, Changsha, Hunan, China.
- Hunan Engineering Research Center of Skin Health and Disease, Changsha, Hunan, China.
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, Hunan, China.
| |
Collapse
|
9
|
Wei A, Fan B, Zhao Y, Zhang H, Wang L, Yu X, Yuan Q, Yang D, Wang S. ST6Gal-I overexpression facilitates prostate cancer progression via the PI3K/Akt/GSK-3β/β-catenin signaling pathway. Oncotarget 2018; 7:65374-65388. [PMID: 27588482 PMCID: PMC5323162 DOI: 10.18632/oncotarget.11699] [Citation(s) in RCA: 60] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2016] [Accepted: 08/24/2016] [Indexed: 11/25/2022] Open
Abstract
ST6Gal-I sialyltransferase adds α2,6-linked sialic acids to the terminal ends of glycan chains of glycoproteins and glycolipids. ST6Gal-I is reportedly upregulated in many cancers, including hepatocellular carcinoma, ovarian cancer and breast cancer. However, the expression and function of ST6Gal-I in prostate cancer (PCa) and the mechanism underlying this function remain largely unknown. In this study, we observed that ST6Gal-I expression was upregulated in human PCa tissues compared to non-malignant prostate tissues. High ST6Gal-I expression was positively correlated with Gleason scores, seminal vesicle involvement and poor survival in patients with PCa. ST6Gal-I knockdown in aggressive prostate cancer PC-3 and DU145 cells significantly inhibited the proliferation, growth, migration and invasion capabilities of these cells. ST6Gal-I knockdown decreased the levels of several PI3K/Akt/GSK-3β/ β-catenin pathway components, such as p-PI3K, (Ser473)p-Akt, (Ser9)p-GSK-3β and β-catenin. Furthermore, targeting this pathway with a PI3K inhibitor or Akt RNA interference decreased p-Akt, p-GSK-3β and β-catenin expression, resulting in decreased PC-3 and DU145 proliferation, migration and invasion. Taken together, these results indicate that ST6Gal-I plays a critical role in cell proliferation and invasion via the PI3K/Akt/GSK-3β/β-catenin signaling pathway during PCa progression and that it might be a promising target for PCa prognosis determination and therapy.
Collapse
Affiliation(s)
- Anwen Wei
- Department of Biochemistry and Molecular Biology, Institute of Glycobiology, Dalian Medical University, Dalian, 116044, Liaoning Province, China
| | - Bo Fan
- Department of Urology, First Affiliated Hospital of Dalian Medical University, Dalian, 116011, Liaoning Province, China
| | - Yujie Zhao
- Department of Biochemistry and Molecular Biology, Institute of Glycobiology, Dalian Medical University, Dalian, 116044, Liaoning Province, China
| | - Han Zhang
- Department of Biochemistry and Molecular Biology, Institute of Glycobiology, Dalian Medical University, Dalian, 116044, Liaoning Province, China
| | - Liping Wang
- Department of Biochemistry and Molecular Biology, Institute of Glycobiology, Dalian Medical University, Dalian, 116044, Liaoning Province, China
| | - Xiao Yu
- Department of Pathology, Dalian Medical University, Dalian, 116044, Liaoning Province, China
| | - Qingmin Yuan
- Department of Biochemistry and Molecular Biology, Institute of Glycobiology, Dalian Medical University, Dalian, 116044, Liaoning Province, China
| | - Deyong Yang
- Department of Urology, First Affiliated Hospital of Dalian Medical University, Dalian, 116011, Liaoning Province, China
| | - Shujing Wang
- Department of Biochemistry and Molecular Biology, Institute of Glycobiology, Dalian Medical University, Dalian, 116044, Liaoning Province, China
| |
Collapse
|
10
|
Yu X, Zhao Y, Wang L, Chen X, Su Z, Zhang H, Yuan Q, Wang S. Sialylated β1, 6 branched N-glycans modulate the adhesion, invasion and metastasis of hepatocarcinoma cells. Biomed Pharmacother 2016; 84:1654-1661. [PMID: 27847205 DOI: 10.1016/j.biopha.2016.10.085] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2016] [Revised: 10/25/2016] [Accepted: 10/25/2016] [Indexed: 12/25/2022] Open
Abstract
The mouse hepatocarcinoma cell lines Hca-F and Hca-P have been derived from hepatocarcinoma in mice and metastasize only to the lymph node. Hca-F cells displayed greater lymphatic metastasis ability than Hca-P cells. When the two cell lines were compared for cell surface sialylated β1,6 branched N-glycans by flow cytometry using L-PHA and SNA, Hca-F cells were found to express significantly higher levels. To explore the effect of increased sialylated β1,6 branched N-glycans on hepatocarcinoma progression, we inhibit their expression in Hca-F cells by using swainsonine treatment and RNA interference. We found that swainsonine treatment or GnT-V-shRNA transfection significantly inhibited the formation of β1,6 branched N-glycans, and partially inhibited the expression of α2,6 sialic acids. Knockdown of sialylated β1,6 branched N-glycans significantly attenuated the invasive and metastatic capability both in vitro and in vivo. Blockade of α2,6 sialic acid expression on Hca-F cell surface by the treatment with neuraminidase caused reduction in cellular adherence to lymph node. In addition, knockdown of sialylated β1,6 branched N-glycans could decrease the expression of Notch1, NICD1, NICD2 and HES1 in Hca-F cells. Collectively, these findings suggest that increased sialylated β1,6 branched N-glycans may contribute to hepatocarcinoma progression by altering the adhesive, invasive and metastatic ability to lymph node via Notch signaling pathway.
Collapse
Affiliation(s)
- Xiao Yu
- Department of Pathology, Dalian Medical University, Dalian 116044, Liaoning Province, China
| | - Yujie Zhao
- Department of Biochemistry and Molecular Biology, Institute of Glycobiology, Dalian Medical University, Dalian 116044, Liaoning Province, China
| | - Liping Wang
- Department of Biochemistry and Molecular Biology, Institute of Glycobiology, Dalian Medical University, Dalian 116044, Liaoning Province, China
| | - Xixi Chen
- School of Life Science and Medicine, Dalian University of Technology, Panjin 124221, Liaoning, China
| | - Zhen Su
- Department of Biochemistry and Molecular Biology, Institute of Glycobiology, Dalian Medical University, Dalian 116044, Liaoning Province, China
| | - Han Zhang
- Department of Biochemistry and Molecular Biology, Institute of Glycobiology, Dalian Medical University, Dalian 116044, Liaoning Province, China
| | - Qingmin Yuan
- Department of Biochemistry and Molecular Biology, Institute of Glycobiology, Dalian Medical University, Dalian 116044, Liaoning Province, China
| | - Shujing Wang
- Department of Biochemistry and Molecular Biology, Institute of Glycobiology, Dalian Medical University, Dalian 116044, Liaoning Province, China.
| |
Collapse
|
11
|
Lu J, Isaji T, Im S, Fukuda T, Kameyama A, Gu J. Expression of N-Acetylglucosaminyltransferase III Suppresses α2,3-Sialylation, and Its Distinctive Functions in Cell Migration Are Attributed to α2,6-Sialylation Levels. J Biol Chem 2016; 291:5708-5720. [PMID: 26801611 DOI: 10.1074/jbc.m115.712836] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2015] [Indexed: 11/06/2022] Open
Abstract
N-Acetylglucosaminyltransferase III (GnT-III), which catalyzes the addition of the bisecting GlcNAc branch on N-glycans, is usually described as a metastasis suppressor. Overexpression of GnT-III inhibited migration in multiple types of tumor cells. However, these results seem controversial to the clinical observations for the increased expression of GnT-III in human hepatomas, glioma, and ovarian cancers. Here, we present evidence that these inconsistencies are mainly attributed to the different expression pattern of cell sialylation. In detail, we show that overexpression of GnT-III significantly inhibits α2,3-sialylation but not α2,6-sialylation. The migratory ability of cells without or with a low level of α2,6-sialylation is consistently suppressed after GnT-III overexpression. In contrast, the effects of GnT-III overexpression are variable in tumor cells that are highly α2,6-sialylated. Overexpression of GnT-III promotes the cell migration in glioma cells U-251 and hepatoma cells HepG2, although it has little influence in human breast cancer cell MDA-MB-231 and gastric cancer cell MKN-45. Interestingly, up-regulation of α2,6-sialylation by overexpressing β-galactoside α2,6-sialyltranferase 1 in the α2,6-hyposialylated HeLa-S3 cells abolishes the anti-migratory effects of GnT-III. Conversely, depletion of α2,6-sialylation by knock-out of β-galactoside α2,6-sialyltranferase 1 in α2,6-hypersialylated HepG2 cells endows GnT-III with the anti-migratory ability. Taken together, our data clearly demonstrate that high expression of α2,6-sialylation on the cell surface could affect the anti-migratory role of GnT-III, which provides an insight into the mechanistic roles of GnT-III in tumor metastasis.
Collapse
Affiliation(s)
- Jishun Lu
- From the Division of Regulatory Glycobiology, Institute of Molecular Biomembrane and Glycobiology, Tohoku Pharmaceutical University, 4-4-1 Komatsushima, Aobaku, Sendai, Miyagi, 981-8558 and
| | - Tomoya Isaji
- From the Division of Regulatory Glycobiology, Institute of Molecular Biomembrane and Glycobiology, Tohoku Pharmaceutical University, 4-4-1 Komatsushima, Aobaku, Sendai, Miyagi, 981-8558 and
| | - Sanghun Im
- From the Division of Regulatory Glycobiology, Institute of Molecular Biomembrane and Glycobiology, Tohoku Pharmaceutical University, 4-4-1 Komatsushima, Aobaku, Sendai, Miyagi, 981-8558 and
| | - Tomohiko Fukuda
- From the Division of Regulatory Glycobiology, Institute of Molecular Biomembrane and Glycobiology, Tohoku Pharmaceutical University, 4-4-1 Komatsushima, Aobaku, Sendai, Miyagi, 981-8558 and
| | - Akihiko Kameyama
- the Department of Life Science and Biotechnology, National Institute of Advanced Industrial Science and Technology, 1-1-1 Umezono, Tsukuba, Ibaraki 305-8568, Japan
| | - Jianguo Gu
- From the Division of Regulatory Glycobiology, Institute of Molecular Biomembrane and Glycobiology, Tohoku Pharmaceutical University, 4-4-1 Komatsushima, Aobaku, Sendai, Miyagi, 981-8558 and.
| |
Collapse
|
12
|
Agarwal AK, Srinivasan N, Godbole R, More SK, Budnar S, Gude RP, Kalraiya RD. Role of tumor cell surface lysosome-associated membrane protein-1 (LAMP1) and its associated carbohydrates in lung metastasis. J Cancer Res Clin Oncol 2015; 141:1563-74. [PMID: 25614122 DOI: 10.1007/s00432-015-1917-2] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2014] [Accepted: 01/12/2015] [Indexed: 01/20/2023]
Abstract
PURPOSE Expression of lysosome-associated membrane protein-1 (LAMP1) on the surface correlates with metastatic potential of B16 melanoma cells. Downregulation of their expression in high metastatic (B16F10) cells reduced their surface expression and metastatic potential. Present investigations explore if overexpression of LAMP1 on the surface of low metastatic (B16F1) cells augment their metastatic ability, and if so, how? METHODS B16F1 cells were transduced with lentiviral vector carrying mutant-LAMP1 (Y386A) (mutLAMP1). Surface expression of LAMP1 and carbohydrates was analyzed by flow cytometry, immunofluorescence and/or immunoprecipitation and Western blotting. Cell spreading and motility were assessed on components of extracellular matrix (ECM) (fibronectin) and basement membrane (BM) (matrigel), and galectin-3-coated coverslips/plates. Metastatic potential was assessed using experimental metastasis assay. RESULTS Pre-incubation with anti-LAMP1 antibodies significantly reduced lung metastasis of B16F10 cells. Overexpression of mutLAMP1 significantly increased its surface expression on B16F1 cells, resulting in increased cellular spreading and motility on fibronectin and matrigel. LAMP1 is the major carrier of poly-N-acetyllactosamine (polyLacNAc) on B16F10 cells. However, significantly higher expression of mutLAMP1 had no effect on galectin-3 binding on cell surface or on spreading or motility of cells on galectin-3-coated coverslips/plates. These cells also failed to show any gain in metastatic ability. This could be because LAMP1 from these cells carried significantly lower levels of polyLacNAc in comparison with B16F10 cells. CONCLUSIONS PolyLacNAc on B16F10 cells and galectin-3 on lungs are the major participants in melanoma metastasis. Although surface LAMP1 promotes interactions with organ ECM and BM, carbohydrates on LAMP1 play a decisive role in dictating lung metastasis.
Collapse
Affiliation(s)
- Akhil Kumar Agarwal
- Advanced Centre for Treatment, Research and Education in Cancer (ACTREC), Tata Memorial Centre, Sector 22, Kharghar, Navi Mumbai, 410210, India
| | | | | | | | | | | | | |
Collapse
|
13
|
Inafuku S, Noda K, Amano M, Nishimura SI, Ishida S. Increase of SialylatedN-Glycansin Eyes with Neovascular Glaucoma Secondary to Proliferative Diabetic Retinopathy. Curr Eye Res 2015; 41:721-4. [DOI: 10.3109/02713683.2015.1068816] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
|
14
|
DRAKULIC DANIJELA, VICENTIC JELENAMARJANOVIC, SCHWIRTLICH MARIJA, TOSIC JELENA, KRSTIC ALEKSANDAR, KLAJN ANDRIJANA, STEVANOVIC MILENA. The overexpression of SOX2 affects the migration of human teratocarcinoma cell line NT2/D1. ACTA ACUST UNITED AC 2015; 87:389-404. [DOI: 10.1590/0001-3765201520140352] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2014] [Accepted: 10/14/2014] [Indexed: 12/15/2022]
Abstract
The altered expression of the SOX2 transcription factor is associated with oncogenic or tumor suppressor functions in human cancers. This factor regulates the migration and invasion of different cancer cells. In this study we investigated the effect of constitutive SOX2 overexpression on the migration and adhesion capacity of embryonal teratocarcinoma NT2/D1 cells derived from a metastasis of a human testicular germ cell tumor. We detected that increased SOX2 expression changed the speed, mode and path of cell migration, but not the adhesion ability of NT2/D1 cells. Additionally, we demonstrated that SOX2 overexpression increased the expression of the tumor suppressor protein p53 and the HDM2 oncogene. Our results contribute to the better understanding of the effect of SOX2 on the behavior of tumor cells originating from a human testicular germ cell tumor. Considering that NT2/D1 cells resemble cancer stem cells in many features, our results could contribute to the elucidation of the role of SOX2 in cancer stem cells behavior and the process of metastasis.
Collapse
Affiliation(s)
| | | | | | - JELENA TOSIC
- University of Belgrade, Serbia; University of Lausanne, Switzerland
| | | | | | | |
Collapse
|
15
|
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
|
16
|
Lu J, Isaji T, Im S, Fukuda T, Hashii N, Takakura D, Kawasaki N, Gu J. β-Galactoside α2,6-sialyltranferase 1 promotes transforming growth factor-β-mediated epithelial-mesenchymal transition. J Biol Chem 2014; 289:34627-41. [PMID: 25344606 DOI: 10.1074/jbc.m114.593392] [Citation(s) in RCA: 80] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
β-Galactoside α2,6-sialyltranferase 1 (ST6GAL1) catalyzes the addition of terminal α2,6-sialylation to N-glycans. Increased expression of ST6GAL1 has been reported in diverse carcinomas and highly correlates with tumor progression. Here, we report that St6gal1 transcription and α2,6-sialylated N-glycans are up-regulated during TGF-β-induced epithelial-mesenchymal transition (EMT) in GE11 cells, requiring the Sp1 element within the St6gal1 promoter. Knockdown of St6gal1 strongly suppressed TGF-β-induced EMT with a concomitant increase in E-cadherin expression, a major determinant of epithelial cell adherens junctions. Conversely, overexpression of ST6GAL1 increased the turnover of cell surface E-cadherin and promoted TGF-β-induced EMT. Overexpressing β-galactoside α2,3-sialyltranferase 4 had little influence on EMT, indicating specificity for α2,6-sialylation. The basal mesenchymal phenotype of MDA-MB-231 human breast cancer cells was partially reversed by ST6GAL1 silencing. Moreover, ST6GAL1 knockdown inhibited the phosphorylation of Akt, but not Smad2, suggesting that ST6GAL1 contributes to EMT through a non-Smad signaling pathway. Taken together, our data indicate that ST6GAL1 promotes TGF-β-dependent EMT as well as maintenance of the mesenchymal state by growth signaling, providing a plausible mechanism whereby up-regulated ST6GAL1 may promote malignant progression.
Collapse
Affiliation(s)
- Jishun Lu
- From the Division of Regulatory Glycobiology, Institute of Molecular Biomembrane and Glycobiology, Tohoku Pharmaceutical University, 4-4-1 Komatsushima, Aobaku, Sendai, Miyagi, 981-8558, Japan and
| | - Tomoya Isaji
- From the Division of Regulatory Glycobiology, Institute of Molecular Biomembrane and Glycobiology, Tohoku Pharmaceutical University, 4-4-1 Komatsushima, Aobaku, Sendai, Miyagi, 981-8558, Japan and
| | - Sanghun Im
- From the Division of Regulatory Glycobiology, Institute of Molecular Biomembrane and Glycobiology, Tohoku Pharmaceutical University, 4-4-1 Komatsushima, Aobaku, Sendai, Miyagi, 981-8558, Japan and
| | - Tomohiko Fukuda
- From the Division of Regulatory Glycobiology, Institute of Molecular Biomembrane and Glycobiology, Tohoku Pharmaceutical University, 4-4-1 Komatsushima, Aobaku, Sendai, Miyagi, 981-8558, Japan and
| | - Noritaka Hashii
- the National Institute of Health Sciences, 1-18-1, Kamiyoga, Setagaya, Tokyo, 158-8501, Japan
| | - Daisuke Takakura
- the National Institute of Health Sciences, 1-18-1, Kamiyoga, Setagaya, Tokyo, 158-8501, Japan
| | - Nana Kawasaki
- the National Institute of Health Sciences, 1-18-1, Kamiyoga, Setagaya, Tokyo, 158-8501, Japan
| | - Jianguo Gu
- From the Division of Regulatory Glycobiology, Institute of Molecular Biomembrane and Glycobiology, Tohoku Pharmaceutical University, 4-4-1 Komatsushima, Aobaku, Sendai, Miyagi, 981-8558, Japan and
| |
Collapse
|
17
|
Invasive potential of melanoma cells correlates with the expression of MT1-MMP and regulated by modulating its association with motility receptors via N-glycosylation on the receptors. BIOMED RESEARCH INTERNATIONAL 2014; 2014:804680. [PMID: 25180193 PMCID: PMC4144153 DOI: 10.1155/2014/804680] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/27/2014] [Accepted: 07/23/2014] [Indexed: 01/01/2023]
Abstract
Matrix remodeling and invasion of basement membrane are the major determinants of malignant progression. Matrix degrading enzymes play a pivotal role in this process and have been shown to be regulated at multiple levels. Using high metastatic B16F10 and its invasive variant B16BL6 cells, we previously demonstrated that the expression of β1,6 branched N-oligosaccharides promotes cellular adhesion on different matrix components which in turn induces secretion of MMP9. The present investigations report that although the two cell lines do not differ in the expression of uPAR, expression of MT1-MMP is significantly higher on B16BL6 cells. Analysis of the transcripts of tissue inhibitors of matrix metalloproteinases (TIMPs) showed that expression of both TIMP1 and TIMP2 correlates negatively with the invasive potential of cells. CD44 and β1 integrin, the two important receptors involved in motility, were identified to carry β1,6 branched N-oligosaccharides in an invasive potential dependent manner. However, their glycosylation status did not appear to influence their surface expression. Although glycosylation on CD44 had no effect, that on β1 integrin significantly affected association of β1 integrin with MT1-MMP. The results thus demonstrate that the cancer cells use multiple mechanisms for degradation of matrix in a controlled manner to couple it with movement for effective invasion.
Collapse
|
18
|
Agarwal AK, Gude RP, Kalraiya RD. Regulation of melanoma metastasis to lungs by cell surface Lysosome Associated Membrane Protein-1 (LAMP1) via galectin-3. Biochem Biophys Res Commun 2014; 449:332-7. [PMID: 24845565 DOI: 10.1016/j.bbrc.2014.05.028] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2014] [Accepted: 05/11/2014] [Indexed: 12/11/2022]
Abstract
Lysosome Associated Membrane Protein-1 (LAMP1), which lines the lysosomes, is often found to be expressed on surface of metastatic cells. We previously demonstrated that its surface expression on B16 melanoma variants correlates with metastatic potential. To establish the role of cell surface LAMP1 in metastasis and to understand the possible mechanism by which it facilitates lung colonization, LAMP1 was downregulated in high metastatic B16F10 cells using shRNAs cloned in a doxycycline inducible vector. This also resulted in significantly decreased LAMP1 on the cell surface. Being a major carrier of poly-N-acetyllactosamine (polyLacNAc) substituted β1,6 branched N-oligosaccharides, the high affinity ligands for galectin-3, LAMP1 down regulation also resulted in appreciably decreased binding of galectin-3 to the cell surface. LAMP1 has been shown to bind to Extracellular Matrix (ECM), Basement Membrane (BM) components and also to galectin-3 (via carbohydrates) which is known to get incorporated into the ECM and BM. Although, LAMP1 downregulation had a marginal effect on cellular spreading and motility on fibronectin and matrigel, it significantly altered the same on galectin-3, and ultimately leading to notably reduced lung metastasis. The results thus for the first time provide direct evidence that cell surface LAMP1 facilitates lung metastasis by providing ligands for galectin-3 which has been shown to be expressed in highest amounts on lungs and constitutively on its vascular endothelium.
Collapse
Affiliation(s)
- Akhil Kumar Agarwal
- Advanced Centre for Treatment Research and Education in Cancer (ACTREC), Tata Memorial Centre, Sector 22, Kharghar, Navi Mumbai 410210, India
| | - Rajiv P Gude
- Advanced Centre for Treatment Research and Education in Cancer (ACTREC), Tata Memorial Centre, Sector 22, Kharghar, Navi Mumbai 410210, India
| | - Rajiv D Kalraiya
- Advanced Centre for Treatment Research and Education in Cancer (ACTREC), Tata Memorial Centre, Sector 22, Kharghar, Navi Mumbai 410210, India.
| |
Collapse
|