1
|
Cao Y, Yi W, Zhu Q. Glycosylation in the tumor immune response: the bitter side of sweetness. Acta Biochim Biophys Sin (Shanghai) 2024; 56:1184-1198. [PMID: 38946426 PMCID: PMC11399423 DOI: 10.3724/abbs.2024107] [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: 05/14/2024] [Accepted: 06/04/2024] [Indexed: 07/02/2024] Open
Abstract
Glycosylation is the most structurally diverse form of post-translational modification (PTM) of proteins that affects a myriad of cellular processes. As a pivotal regulator of protein homeostasis, glycosylation notably impacts the function of proteins, spanning from protein localization and stability to protein-protein interactions. Aberrant glycosylation is a hallmark of cancer, and extensive studies have revealed the multifaceted roles of glycosylation in tumor growth, migration, invasion and immune escape Over the past decade, glycosylation has emerged as an immune regulator in the tumor microenvironment (TME). Here, we summarize the intricate interplay between glycosylation and the immune system documented in recent literature, which orchestrates the regulation of the tumor immune response through endogenous lectins, immune checkpoints and the extracellular matrix (ECM) in the TME. In addition, we discuss the latest progress in glycan-based cancer immunotherapy. This review provides a basic understanding of glycosylation in the tumor immune response and a theoretical framework for tumor immunotherapy.
Collapse
Affiliation(s)
- Yuting Cao
- />Department of BiochemistryCollege of Life SciencesZhejiang UniversityHangzhou310058China
| | - Wen Yi
- />Department of BiochemistryCollege of Life SciencesZhejiang UniversityHangzhou310058China
| | - Qiang Zhu
- />Department of BiochemistryCollege of Life SciencesZhejiang UniversityHangzhou310058China
| |
Collapse
|
2
|
Roger I, Montero P, Milara J, Cortijo J. Pirfenidone and nintedanib attenuates pulmonary artery endothelial and smooth muscle cells transformations induced by IL-11. Eur J Pharmacol 2024; 972:176547. [PMID: 38561103 DOI: 10.1016/j.ejphar.2024.176547] [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: 11/02/2023] [Revised: 03/21/2024] [Accepted: 03/27/2024] [Indexed: 04/04/2024]
Abstract
Idiopathic pulmonary fibrosis (IPF) associated to pulmonary hypertension (PH) portends a poor prognosis, characterized by lung parenchyma fibrosis and pulmonary artery remodeling. Serum and parenchyma levels of Interleukin 11 (IL-11) are elevated in IPF-PH patients and contributes to pulmonary artery remodeling and PH. However, the effect of current approved therapies against IPF in pulmonary artery remodeling induced by IL-11 is unknown. The aim of this study is to analyze the effects of nintedanib and pirfenidone on pulmonary artery endothelial and smooth muscle cell remodeling induced by IL-11 in vitro. Our results show that nintedanib (NTD) and pirfenidone (PFD) ameliorates endothelial to mesenchymal transition (EnMT), pulmonary artery smooth muscle cell to myofibroblast-like transformation and pulmonary remodeling in precision lung cut slices. This study provided also evidence of the inhibitory effect of PFD and NTD on IL-11-induced endothelial and muscle cells proliferation and senescence. The inhibitory effect of these drugs on monocyte arrest and angiogenesis was also studied. Finally, we observed that IL-11 induced canonical signal transducer and activator of transcription 3 (STAT3) and non-canonical mitogen-activated protein kinase 1/2 (ERK1/2) phosphorylation, but, PFD and NTD only inhibited ERK1/2 phosphorylation. Therefore, this study provided evidence of the inhibitory effect of NTD and PFD on markers of pulmonary artery remodeling induced by IL-11.
Collapse
Affiliation(s)
- Inés Roger
- Biomedical Research Networking Centre on Respiratory Diseases (CIBERES), Health Institute Carlos III, 28029, Madrid, Spain; Department of Pharmacology, Faculty of Medicine, University of Valencia, 46010, Valencia, Spain; Faculty of Health Sciences, Universidad Europea de Valencia, 46010, Valencia, Spain.
| | - Paula Montero
- Biomedical Research Networking Centre on Respiratory Diseases (CIBERES), Health Institute Carlos III, 28029, Madrid, Spain; Department of Pharmacology, Faculty of Medicine, University of Valencia, 46010, Valencia, Spain; Faculty of Health Sciences, Universidad Europea de Valencia, 46010, Valencia, Spain
| | - Javier Milara
- Biomedical Research Networking Centre on Respiratory Diseases (CIBERES), Health Institute Carlos III, 28029, Madrid, Spain; Department of Pharmacology, Faculty of Medicine, University of Valencia, 46010, Valencia, Spain; Pharmacy Unit, University General Hospital Consortium, 46014, Valencia, Spain
| | - Julio Cortijo
- Biomedical Research Networking Centre on Respiratory Diseases (CIBERES), Health Institute Carlos III, 28029, Madrid, Spain; Department of Pharmacology, Faculty of Medicine, University of Valencia, 46010, Valencia, Spain; Research and Teaching Unit, University General Hospital Consortium, 46014, Valencia, Spain
| |
Collapse
|
3
|
Jang I, Menon S, Indra I, Basith R, Beningo KA. Calpain Small Subunit Mediated Secretion of Galectin-3 Regulates Traction Stress. Biomedicines 2024; 12:1247. [PMID: 38927454 PMCID: PMC11200796 DOI: 10.3390/biomedicines12061247] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2024] [Revised: 05/29/2024] [Accepted: 05/30/2024] [Indexed: 06/28/2024] Open
Abstract
The complex regulation of traction forces (TF) produced during cellular migration remains poorly understood. We have previously found that calpain 4 (Capn4), the small non-catalytic subunit of the calpain 1 and 2 proteases, regulates the production of TF independent of the proteolytic activity of the larger subunits. Capn4 was later found to facilitate tyrosine phosphorylation and secretion of the lectin-binding protein galectin-3 (Gal3). In this study, recombinant Gal3 (rGal3) was added to the media-enhanced TF generated by capn4-/- mouse embryonic fibroblasts (MEFs). Extracellular Gal3 also rescued defects in the distribution, morphology, and adhesive strength of focal adhesions present in capn4-/- MEF cells. Surprisingly, extracellular Gal3 does not influence mechanosensing. c-Abl kinase was found to affect Gal3 secretion and the production of TF through phosphorylation of Y107 on Gal3. Our study also suggests that Gal3-mediated regulation of TF occurs through signaling pathways triggered by β1 integrin but not by focal adhesion kinase (FAK) Y397 autophosphorylation. Our findings provide insights into the signaling mechanism by which Capn4 and secreted Gal3 regulate cell migration through the modulation of TF distinctly independent from a mechanosensing mechanism.
Collapse
Affiliation(s)
| | | | | | | | - Karen A. Beningo
- Department of Biological Sciences, Wayne State University, Detroit, MI 48202, USA; (I.J.)
| |
Collapse
|
4
|
Radziejewska I. Tumor-associated carbohydrate antigens of MUC1 - Implication in cancer development. Biomed Pharmacother 2024; 174:116619. [PMID: 38643541 DOI: 10.1016/j.biopha.2024.116619] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2024] [Revised: 04/12/2024] [Accepted: 04/17/2024] [Indexed: 04/23/2024] Open
Abstract
Glycosylation of cancerous epithelial MUC1 protein is specifically altered in comparison to that which is presented by healthy cells. One of such changes is appearing tumor-associated carbohydrate antigens (TACAs) which are rare in normal tissues and are highly correlated with poor clinical outcomes and cancer progression. This review summarizes and describes the role of Tn, T antigens, their sialylated forms as well as fucosylated Lewis epitopes in different aspects of tumor development, progression, and metastasis. Finally, applications of MUC1 glycan epitopes as potential targets for therapeutic strategy of cancers are notified. One of the novelties of this review is presentation of TACAs as inherently connected with MUC1 mucin.
Collapse
Affiliation(s)
- Iwona Radziejewska
- Department of Medical Chemistry, Medical University of Białystok, ul. Mickiewicza 2, Białystok 15-222, Poland.
| |
Collapse
|
5
|
Iwamoto S, Mori Y, Yamashita T, Ojima K, Akita K, Togano S, Kushiyama S, Yashiro M, Yatera Y, Yamaguchi T, Komiyama A, Sago Y, Itano N, Nakada H. Trophoblast cell surface antigen-2 phosphorylation triggered by binding of galectin-3 drives metastasis through down-regulation of E-cadherin. J Biol Chem 2023; 299:104971. [PMID: 37380081 PMCID: PMC10392139 DOI: 10.1016/j.jbc.2023.104971] [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: 10/11/2022] [Revised: 05/24/2023] [Accepted: 05/26/2023] [Indexed: 06/30/2023] Open
Abstract
The expression of trophoblast cell surface antigen-2 (Trop-2) is enhanced in many tumor tissues and is correlated with increased malignancy and poor survival of patients with cancer. Previously, we demonstrated that the Ser-322 residue of Trop-2 is phosphorylated by protein kinase Cα (PKCα) and PKCδ. Here, we demonstrate that phosphomimetic Trop-2 expressing cells have markedly decreased E-cadherin mRNA and protein levels. Consistently, mRNA and protein of the E-cadherin-repressing transcription factors zinc finger E-Box binding homeobox 1 (ZEB1) were elevated, suggesting transcriptional regulation of E-cadherin expression. The binding of galectin-3 to Trop-2 enhanced the phosphorylation and subsequent cleavage of Trop-2, followed by intracellular signaling by the resultant C-terminal fragment. Binding of β-catenin/transcription factor 4 (TCF4) along with the C-terminal fragment of Trop-2 to the ZEB1 promoter upregulated ZEB1 expression. Of note, siRNA-mediated knockdown of β-catenin and TCF4 increased the expression of E-cadherin through ZEB1 downregulation. Knockdown of Trop-2 in MCF-7 cells and DU145 cells resulted in downregulation of ZEB1 and subsequent upregulation of E-cadherin. Furthermore, wild-type and phosphomimetic Trop-2 but not phosphorylation-blocked Trop-2 were detected in the liver and/or lung of some nude mice bearing primary tumors inoculated intraperitoneally or subcutaneously with wild-type or mutated Trop-2 expressing cells, suggesting that Trop-2 phosphorylation, plays an important role in tumor cell mobility in vivo, too. Together with our previous finding of Trop-2 dependent regulation of claudin-7, we suggest that the Trop-2-mediated cascade involves concurrent derangement of both tight and adherence junctions, which may drive metastasis of epithelial tumor cells.
Collapse
Affiliation(s)
- Shungo Iwamoto
- Department of Molecular Biosciences, Faculty of Life Sciences, Kyoto Sangyo University, Kyoto, Japan
| | - Yugo Mori
- Department of Molecular Biosciences, Faculty of Life Sciences, Kyoto Sangyo University, Kyoto, Japan
| | - Tomoko Yamashita
- Department of Molecular Biosciences, Faculty of Life Sciences, Kyoto Sangyo University, Kyoto, Japan
| | - Kazuki Ojima
- Department of Molecular Biosciences, Faculty of Life Sciences, Kyoto Sangyo University, Kyoto, Japan
| | - Kaoru Akita
- Department of Molecular Biosciences, Faculty of Life Sciences, Kyoto Sangyo University, Kyoto, Japan
| | - Shingo Togano
- Department of Molecular Oncology and Therapeutics, Osaka Metropolitan University Graduate School of Medicine, Osaka, Japan
| | - Shuhei Kushiyama
- Department of Molecular Oncology and Therapeutics, Osaka Metropolitan University Graduate School of Medicine, Osaka, Japan
| | - Masakazu Yashiro
- Department of Molecular Oncology and Therapeutics, Osaka Metropolitan University Graduate School of Medicine, Osaka, Japan
| | - Yuki Yatera
- Department of Molecular Biosciences, Faculty of Life Sciences, Kyoto Sangyo University, Kyoto, Japan
| | - Tomoko Yamaguchi
- Department of Molecular Biosciences, Faculty of Life Sciences, Kyoto Sangyo University, Kyoto, Japan
| | - Akane Komiyama
- Department of Molecular Biosciences, Faculty of Life Sciences, Kyoto Sangyo University, Kyoto, Japan
| | - Yuki Sago
- Department of Molecular Biosciences, Faculty of Life Sciences, Kyoto Sangyo University, Kyoto, Japan
| | - Naoki Itano
- Department of Molecular Biosciences, Faculty of Life Sciences, Kyoto Sangyo University, Kyoto, Japan
| | - Hiroshi Nakada
- Department of Molecular Biosciences, Faculty of Life Sciences, Kyoto Sangyo University, Kyoto, Japan.
| |
Collapse
|
6
|
Radziejewska I. Galectin-3 and Epithelial MUC1 Mucin-Interactions Supporting Cancer Development. Cancers (Basel) 2023; 15:2680. [PMID: 37345016 DOI: 10.3390/cancers15102680] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2023] [Revised: 05/04/2023] [Accepted: 05/05/2023] [Indexed: 06/23/2023] Open
Abstract
Aberrant glycosylation of cell surface proteins is a very common feature of many cancers. One of the glycoproteins, which undergoes specific alterations in the glycosylation of tumor cells is epithelial MUC1 mucin, which is highly overexpressed in the malignant state. Such changes lead to the appearance of tumor associated carbohydrate antigens (TACAs) on MUC1, which are rarely seen in healthy cells. One of these structures is the Thomsen-Friedenreich disaccharide Galβ1-3GalNAc (T or TF antigen), which is typical for about 90% of cancers. It was revealed that increased expression of the T antigen has a big impact on promoting cancer progression and metastasis, among others, due to the interaction of this antigen with the β-galactose binding protein galectin-3 (Gal-3). In this review, we summarize current information about the interactions between the T antigen on MUC1 mucin and Gal-3, and their impact on cancer progression and metastasis.
Collapse
Affiliation(s)
- Iwona Radziejewska
- Department of Medical Chemistry, Medical University of Białystok, ul. Mickiewicza 2a, 15-222 Białystok, Poland
| |
Collapse
|
7
|
Semba R, Horimoto Y, Sakata-Matsuzawa M, Ishizuka Y, Denda-Nagai K, Fujihira H, Noji M, Onagi H, Ichida M, Miura H, Watanabe J, Saito M, Saito T, Arakawa A, Irimura T. Possible correlation of apical localization of MUC1 glycoprotein with luminal A-like status of breast cancer. Sci Rep 2023; 13:5281. [PMID: 37002293 PMCID: PMC10066179 DOI: 10.1038/s41598-023-32579-4] [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: 09/21/2022] [Accepted: 03/29/2023] [Indexed: 04/03/2023] Open
Abstract
Adjuvant chemotherapy has played a major role in the treatment of hormone receptor-positive breast cancer for many years. To better determine which patient subsets need adjuvant chemotherapy, various gene expression analyses have been developed, but cost-effective tools to identify such patients remain elusive. In the present report, we retrospectively investigated immunohistochemical expression and subcellular localization of MUC1 in primary tumors and examined their relationship to tumor malignancy, chemotherapy effect and patient outcomes. We retrospectively examined three patient cohorts with hormone receptor-positive/human epidermal growth factor receptor 2-negative invasive breast cancer: 51 patients who underwent 21-gene expression analysis (multi-gene assay-cohort), 96 patients who received neoadjuvant chemotherapy (neoadjuvant chemotherapy-cohort), and 609 patients whose tumor tissue was used in tissue-microarrays (tissue-microarray-cohort). The immunohistochemical staining pattern of the anti-MUC1 monoclonal antibody, Ma695, was examined in cancer tissues, and subcellular localization was determined as apical, cytoplasmic or negative. In the multi-gene assay-cohort, tumors with apical patterns had the lowest recurrence scores, reflecting lower tumor malignancy, and were significantly lower than MUC1-negative tumors (P = 0.038). In the neoadjuvant chemotherapy-cohort, there was no correlation between MUC1 staining patterns and effects of chemotherapy. Finally, in the tissue-microarray-cohort, we found that patients with apical MUC1 staining patterns had significantly longer disease-free-survival and overall survival than other patterns (P = 0.020 and 0.039, respectively). Our data suggest that an apical MUC1 staining pattern indicates luminal A-likeness. Assessment of the subcellular localization of MUC1 glycoprotein may be useful for identifying patients who can avoid adjuvant chemotherapy.
Collapse
Affiliation(s)
- Ryoko Semba
- Department of Breast Oncology, Juntendo University Faculty of Medicine, Tokyo, 113-0033, Japan
| | - Yoshiya Horimoto
- Department of Breast Oncology, Juntendo University Faculty of Medicine, Tokyo, 113-0033, Japan.
- Department of Human Pathology, Juntendo University Faculty of Medicine, Tokyo, Japan.
| | - Madoka Sakata-Matsuzawa
- Department of Breast Oncology, Juntendo University Faculty of Medicine, Tokyo, 113-0033, Japan
| | - Yumiko Ishizuka
- Department of Breast Oncology, Juntendo University Faculty of Medicine, Tokyo, 113-0033, Japan
| | - Kaori Denda-Nagai
- Intractable Disease Research Center, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Haruhiko Fujihira
- Division of Glycobiologics, Department of Breast Oncology, Juntendo University Faculty of Medicine, Tokyo, Japan
- Glycometabolic Biochemistry Laboratory, RIKEN Cluster for Pioneering Research, RIKEN, Wako, Japan
| | - Miki Noji
- Division of Glycobiologics, Department of Breast Oncology, Juntendo University Faculty of Medicine, Tokyo, Japan
| | - Hiroko Onagi
- Department of Human Pathology, Juntendo University Faculty of Medicine, Tokyo, Japan
| | - Miyu Ichida
- Department of Human Pathology, Juntendo University Faculty of Medicine, Tokyo, Japan
| | - Hiroyoshi Miura
- Department of Surgery, Koshigaya Municipal Hospital, Saitama, Japan
| | - Junichiro Watanabe
- Department of Breast Oncology, Juntendo University Faculty of Medicine, Tokyo, 113-0033, Japan
| | - Mitsue Saito
- Department of Breast Oncology, Juntendo University Faculty of Medicine, Tokyo, 113-0033, Japan
| | - Tsuyoshi Saito
- Department of Human Pathology, Juntendo University Faculty of Medicine, Tokyo, Japan
- Intractable Disease Research Center, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Atsushi Arakawa
- Department of Human Pathology, Juntendo University Faculty of Medicine, Tokyo, Japan
| | - Tatsuro Irimura
- Division of Glycobiologics, Department of Breast Oncology, Juntendo University Faculty of Medicine, Tokyo, Japan
| |
Collapse
|
8
|
Aslanis V, Slack RJ, MacKinnon AC, McClinton C, Tantawi S, Gravelle L, Nilsson UJ, Leffler H, Brooks A, Khindri SK, Marshall RP, Pedersen A, Schambye H, Zetterberg F. Safety and pharmacokinetics of GB1211, an oral galectin-3 inhibitor: a single- and multiple-dose first-in-human study in healthy participants. Cancer Chemother Pharmacol 2023; 91:267-280. [PMID: 36914828 PMCID: PMC10010643 DOI: 10.1007/s00280-023-04513-y] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2022] [Accepted: 02/21/2023] [Indexed: 03/16/2023]
Abstract
PURPOSE Galectin-3, a β-galactoside-binding lectin, plays a key role in several cellular pathways involved in chronic inflammation, heart disease and cancer. GB1211 is an orally bioavailable galectin-3 inhibitor, developed to be systemically active. We report safety and pharmacokinetics (PK) of GB1211 in healthy participants. METHODS This phase 1, double-blind, placebo-controlled, first-in-human study (NCT03809052) included a single ascending-dose phase (with a food-effect cohort) where participants across seven sequential cohorts were randomized 3:1 to receive oral GB1211 (5, 20, 50, 100, 200 or 400 mg) or placebo. In the multiple ascending-dose phase, participants received 50 or 100 mg GB1211 or placebo twice daily for 10 days. All doses were administered in the fasted state except in the food-effect cohort where doses were given 30 min after a high-fat meal. RESULTS All 78 participants received at least one GB1211 dose (n = 58) or placebo (n = 20) and completed the study. No safety concerns were identified. Following single and multiple oral doses under fasted conditions, maximum GB1211 plasma concentrations were reached at 1.75-4 h (median) post-dose; mean half-life was 11-16 h. There was a ~ twofold GB1211 accumulation in plasma with multiple dosing, with steady-state reached within 3 days; 30% of the administered dose was excreted in urine as unchanged drug. Absorption in the fed state was delayed by 2 h but systemic exposure was unaffected. CONCLUSION GB1211 was well tolerated, rapidly absorbed, and displayed favorable PK, indicating a potential to treat multiple disease types. These findings support further clinical development of GB1211. CLINICAL TRIAL REGISTRATION The study was registered with ClinicalTrials.gov (identifier: NCT03809052).
Collapse
Affiliation(s)
| | | | | | | | | | | | - Ulf J Nilsson
- Department of Chemistry, Lund University, 22100, Lund, Sweden
| | - Hakon Leffler
- Department of Laboratory Medicine, Lund University, 22100, Lund, Sweden
| | | | | | | | | | | | | |
Collapse
|
9
|
Huang QF, Cheng YB, Guo QH, Wang Y, Chen YL, Zhang DY, An DW, Li Y, Wang JG. Serum Galectin-3 and Mucin-1 (CA15-3) in Relation to Renal Function in Untreated Chinese Patients. Am J Hypertens 2023; 36:176-182. [PMID: 36226892 DOI: 10.1093/ajh/hpac115] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Revised: 09/21/2022] [Accepted: 10/12/2022] [Indexed: 11/12/2022] Open
Abstract
BACKGROUND Galectin-3 is a multi-functional lectin protein and a ligand of mucin-1 (CA15-3), and has been linked to renal fibrosis in animal models and renal function in humans. However, no population study has ever explored the associations with both ligand and receptor. We therefore investigate the independent association of renal function with serum galectin-3 and mucin-1 (CA15-3) in untreated Chinese patients. METHODS The study participants were outpatients who were suspected of hypertension, but had not been treated with antihypertensive medication. Serum galectin-3 and mucin-1 (CA15-3) concentrations were both measured by the enzyme-linked immunosorbent assay (ELISA) method. Estimated glomerular filtration rate (eGFR) was calculated from serum creatinine by the use of the Chronic Kidney Disease Epidemiology Collaboration (CKD-EPI) equation. RESULTS The 1,789 participants included 848 (47.4%) men. Mean (±SD) age was 51.3 ± 10.7 years. Multiple regression analyses showed that eGFR was significantly associated with serum galectin-3 and mucin-1 (CA15-3) concentration (0.68 and 1.32 ml/min/1.73 m2 decrease per 1-SD increase in log transformed serum galectin-3 and mucin-1 (CA15-3) concentration, respectively; P ≤ 0.006). The association of eGFR with serum mucin-1 (CA15-3) concentration was significantly stronger in the overweight (BMI 24.0-27.9 kg/m2) and obese (BMI ≥ 28.0 kg/m2) than in normal weight subjects (BMI < 24.0 kg/m2, P for interaction 0.018). Path analysis showed that serum galectin-3 concentration had both a direct (P = 0.016) and a mucin-1 mediated indirect effect (P = 0.014) on eGFR. CONCLUSIONS Both circulating galectin-3 and mucin-1 (CA15-3) were significantly associated with renal function. The role of galectin-3 on renal function might be partially via mucin-1.
Collapse
Affiliation(s)
- Qi-Fang Huang
- Department of Cardiovascular Medicine, Center for Epidemiological Studies and Clinical Trials and Center for Vascular Evaluations, State Key Laboratory of Medical Genomics, Shanghai Key Laboratory of Hypertension, The Shanghai Institute of Hypertension, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yi-Bang Cheng
- Department of Cardiovascular Medicine, Center for Epidemiological Studies and Clinical Trials and Center for Vascular Evaluations, State Key Laboratory of Medical Genomics, Shanghai Key Laboratory of Hypertension, The Shanghai Institute of Hypertension, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Qian-Hui Guo
- Department of Cardiovascular Medicine, Center for Epidemiological Studies and Clinical Trials and Center for Vascular Evaluations, State Key Laboratory of Medical Genomics, Shanghai Key Laboratory of Hypertension, The Shanghai Institute of Hypertension, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Ying Wang
- Department of Cardiovascular Medicine, Center for Epidemiological Studies and Clinical Trials and Center for Vascular Evaluations, State Key Laboratory of Medical Genomics, Shanghai Key Laboratory of Hypertension, The Shanghai Institute of Hypertension, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yi-Lin Chen
- Department of Cardiovascular Medicine, Center for Epidemiological Studies and Clinical Trials and Center for Vascular Evaluations, State Key Laboratory of Medical Genomics, Shanghai Key Laboratory of Hypertension, The Shanghai Institute of Hypertension, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Dong-Yan Zhang
- Department of Cardiovascular Medicine, Center for Epidemiological Studies and Clinical Trials and Center for Vascular Evaluations, State Key Laboratory of Medical Genomics, Shanghai Key Laboratory of Hypertension, The Shanghai Institute of Hypertension, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - De-Wei An
- Department of Cardiovascular Medicine, Center for Epidemiological Studies and Clinical Trials and Center for Vascular Evaluations, State Key Laboratory of Medical Genomics, Shanghai Key Laboratory of Hypertension, The Shanghai Institute of Hypertension, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yan Li
- Department of Cardiovascular Medicine, Center for Epidemiological Studies and Clinical Trials and Center for Vascular Evaluations, State Key Laboratory of Medical Genomics, Shanghai Key Laboratory of Hypertension, The Shanghai Institute of Hypertension, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Ji-Guang Wang
- Department of Cardiovascular Medicine, Center for Epidemiological Studies and Clinical Trials and Center for Vascular Evaluations, State Key Laboratory of Medical Genomics, Shanghai Key Laboratory of Hypertension, The Shanghai Institute of Hypertension, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
10
|
Targeting galectin-driven regulatory circuits in cancer and fibrosis. Nat Rev Drug Discov 2023; 22:295-316. [PMID: 36759557 DOI: 10.1038/s41573-023-00636-2] [Citation(s) in RCA: 64] [Impact Index Per Article: 64.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/20/2022] [Indexed: 02/11/2023]
Abstract
Galectins are a family of endogenous glycan-binding proteins that have crucial roles in a broad range of physiological and pathological processes. As a group, these proteins use both extracellular and intracellular mechanisms as well as glycan-dependent and independent pathways to reprogramme the fate and function of numerous cell types. Given their multifunctional roles in both tissue fibrosis and cancer, galectins have been identified as potential therapeutic targets for these disorders. Here, we focus on the therapeutic relevance of galectins, particularly galectin 1 (GAL1), GAL3 and GAL9 to tumour progression and fibrotic diseases. We consider an array of galectin-targeted strategies, including small-molecule carbohydrate inhibitors, natural polysaccharides and their derivatives, peptides, peptidomimetics and biological agents (notably, neutralizing monoclonal antibodies and truncated galectins) and discuss their mechanisms of action, selectivity and therapeutic potential in preclinical models of fibrosis and cancer. We also review the results of clinical trials that aim to evaluate the efficacy of galectin inhibitors in patients with idiopathic pulmonary fibrosis, nonalcoholic steatohepatitis and cancer. The rapid pace of glycobiology research, combined with the acute need for drugs to alleviate fibrotic inflammation and overcome resistance to anticancer therapies, will accelerate the translation of anti-galectin therapeutics into clinical practice.
Collapse
|
11
|
Highlights on the Role of Galectin-3 in Colorectal Cancer and the Preventive/Therapeutic Potential of Food-Derived Inhibitors. Cancers (Basel) 2022; 15:cancers15010052. [PMID: 36612048 PMCID: PMC9817985 DOI: 10.3390/cancers15010052] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2022] [Revised: 12/16/2022] [Accepted: 12/19/2022] [Indexed: 12/24/2022] Open
Abstract
Colorectal cancer (CRC) is a leading cause of death worldwide. Despite advances in surgical and therapeutic management, tumor metastases and resistance to therapy still represent major hurdles. CRC risk is highly modifiable by lifestyle factors, including diet, which strongly influences both cancer incidence and related mortality. Galectin-3 (Gal-3) is a multifaceted protein involved in multiple pathophysiological pathways underlying chronic inflammation and cancer. Its versatility is given by the ability to participate in a wide range of tumor-promoting processes, including cell-cell/cell-matrix interactions, cell growth regulation and apoptosis, and the immunosuppressive tumor microenvironment. This review provides an updated summary of preclinical and observational human studies investigating the pathogenetic role of Gal-3 in intestinal inflammation and CRC, as well as the potential of Gal-3 activity inhibition by plant-source food-derived bioactive compounds to control CRC onset/growth. These studies highlight both direct and immuno-mediated effects of Gal-3 on tumor growth and invasiveness and its potential role as a CRC prognostic biomarker. Substantial evidence indicates natural food-derived Gal-3 inhibitors as promising candidates for CRC prevention and therapy. However, critical issues, such as their bioavailability and efficacy, in controlled human studies need to be addressed to translate research progress into clinical applications.
Collapse
|
12
|
Fujii C, Harumiya S, Sato Y, Kawakubo M, Matoba H, Nakayama J. α1,4-linked N-acetylglucosamine suppresses gastric cancer development by inhibiting MUC1-mediated signaling. Cancer Sci 2022; 113:3852-3863. [PMID: 35959971 PMCID: PMC9633294 DOI: 10.1111/cas.15530] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2022] [Revised: 07/28/2022] [Accepted: 08/06/2022] [Indexed: 11/29/2022] Open
Abstract
Gastric cancer is the second leading cause of cancer deaths worldwide, and more understanding of its molecular basis is urgently needed. Gastric gland mucin secreted from pyloric gland cells, mucous neck cells, and cardiac gland cells of the gastric mucosa harbors unique O‐glycans carrying terminal α1,4‐linked N‐acetylglucosamine (αGlcNAc) residues. We previously reported that αGlcNAc loss correlated positively with poor outcomes for patients with differentiated‐type gastric cancer. However, the molecular mechanisms underlying these outcomes remained poorly understood. Here, we examined the effects of upregulated αGlcNAc expression on malignant phenotypes of the differentiated‐type gastric cancer cell lines, AGS and MKN7. Upregulation of αGlcNAc following ectopic expression of its biosynthetic enzyme attenuated cell proliferation, motility, and invasiveness of AGS and MKN7 cells in vitro. Moreover, AGS cell tumorigenicity was significantly suppressed by αGlcNAc overexpression in a xenograft model. To define the molecular mechanisms underlying these phenotypes, we investigated αGlcNAc binding proteins in AGS cells and identified Mucin‐1 (MUC1) and podocalyxin. Both proteins were colocalized with αGlcNAc on human gastric cancer cells. We also found that αGlcNAc was bound to MUC1 in murine normal gastric mucosa. When we assessed the effects of αGlcNAc binding to MUC1, we found that αGlcNAc blocked galectin‐3 binding to MUC1, phosphorylation of the MUC1 C‐terminus, and recruitment of Src and β‐catenin to that C‐terminus. These results suggest that αGlcNAc regulates cancer cell phenotypes by dampening MUC1 signal transduction.
Collapse
Affiliation(s)
- Chifumi Fujii
- Department of Molecular Pathology, Shinshu University School of Medicine, 3-1-1 Asahi, Matsumoto 390-8621, Japan.,Department of Biotechnology, Institute for Biomedical Sciences, Interdisciplinary Cluster for Cutting Edge Research, Shinshu University, 390-8621, Matsumoto, Japan
| | - Satoru Harumiya
- Department of Molecular Pathology, Shinshu University School of Medicine, 3-1-1 Asahi, Matsumoto 390-8621, Japan
| | - Yoshiko Sato
- Department of Molecular Pathology, Shinshu University School of Medicine, 3-1-1 Asahi, Matsumoto 390-8621, Japan
| | - Masatomo Kawakubo
- Department of Molecular Pathology, Shinshu University School of Medicine, 3-1-1 Asahi, Matsumoto 390-8621, Japan
| | - Hisanori Matoba
- Department of Molecular Pathology, Shinshu University School of Medicine, 3-1-1 Asahi, Matsumoto 390-8621, Japan
| | - Jun Nakayama
- Department of Molecular Pathology, Shinshu University School of Medicine, 3-1-1 Asahi, Matsumoto 390-8621, Japan
| |
Collapse
|
13
|
Heterogeneous population of macrophages in the development of non-alcoholic fatty liver disease☆. LIVER RESEARCH 2022. [DOI: 10.1016/j.livres.2022.06.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
|
14
|
Supruniuk K, Radziejewska I. MUC1 is an oncoprotein with a significant role in apoptosis (Review). Int J Oncol 2021; 59:68. [PMID: 34278474 PMCID: PMC8360618 DOI: 10.3892/ijo.2021.5248] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2021] [Accepted: 06/29/2021] [Indexed: 01/10/2023] Open
Abstract
Mucin 1 (MUC1) is a membrane-bound, highly glycosylated protein that is overexpressed in all stages of malignant transformation. Overexpression of MUC1 together with loss of polarization and hypoglycosylation are associated with resistance to apoptosis, which is the process that results in efficient removal of damaged cells. Inhibition of the apoptotic process is responsible for tumor development, tumor progression and drug resistance. MUC1 is considered as an oncogenic molecule that is involved in various signaling pathways responsible for the regulation of apoptosis. Based on this, the aim of the present study was to discuss the involvement of MUC1 in the divergent mechanisms regulating programmed cell death.
Collapse
Affiliation(s)
- Katarzyna Supruniuk
- Department of Medical Chemistry, Medical University of Białystok, 15‑222 Białystok, Poland
| | - Iwona Radziejewska
- Department of Medical Chemistry, Medical University of Białystok, 15‑222 Białystok, Poland
| |
Collapse
|
15
|
Seguin L, Odouard S, Corlazzoli F, Haddad SA, Moindrot L, Calvo Tardón M, Yebra M, Koval A, Marinari E, Bes V, Guérin A, Allard M, Ilmjärv S, Katanaev VL, Walker PR, Krause KH, Dutoit V, Sarkaria JN, Dietrich PY, Cosset É. Macropinocytosis requires Gal-3 in a subset of patient-derived glioblastoma stem cells. Commun Biol 2021; 4:718. [PMID: 34112916 PMCID: PMC8192788 DOI: 10.1038/s42003-021-02258-z] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2020] [Accepted: 05/21/2021] [Indexed: 12/11/2022] Open
Abstract
Recently, we involved the carbohydrate-binding protein Galectin-3 (Gal-3) as a druggable target for KRAS-mutant-addicted lung and pancreatic cancers. Here, using glioblastoma patient-derived stem cells (GSCs), we identify and characterize a subset of Gal-3high glioblastoma (GBM) tumors mainly within the mesenchymal subtype that are addicted to Gal-3-mediated macropinocytosis. Using both genetic and pharmacologic inhibition of Gal-3, we showed a significant decrease of GSC macropinocytosis activity, cell survival and invasion, in vitro and in vivo. Mechanistically, we demonstrate that Gal-3 binds to RAB10, a member of the RAS superfamily of small GTPases, and β1 integrin, which are both required for macropinocytosis activity and cell survival. Finally, by defining a Gal-3/macropinocytosis molecular signature, we could predict sensitivity to this dependency pathway and provide proof-of-principle for innovative therapeutic strategies to exploit this Achilles' heel for a significant and unique subset of GBM patients.
Collapse
Affiliation(s)
- Laetitia Seguin
- University Côte d'Azur, CNRS UMR7284, INSERM U1081, Institute for Research on Cancer and Aging (IRCAN), Nice, France
| | - Soline Odouard
- Laboratory of Tumor Immunology, Department of Oncology, Center for Translational Research in Onco-Hematology, Swiss Cancer Center Léman (SCCL), Geneva University Hospitals, University of Geneva, Geneva, Switzerland
| | - Francesca Corlazzoli
- Laboratory of Tumor Immunology, Department of Oncology, Center for Translational Research in Onco-Hematology, Swiss Cancer Center Léman (SCCL), Geneva University Hospitals, University of Geneva, Geneva, Switzerland
| | - Sarah Al Haddad
- Laboratory of Tumor Immunology, Department of Oncology, Center for Translational Research in Onco-Hematology, Swiss Cancer Center Léman (SCCL), Geneva University Hospitals, University of Geneva, Geneva, Switzerland
| | - Laurine Moindrot
- Laboratory of Tumor Immunology, Department of Oncology, Center for Translational Research in Onco-Hematology, Swiss Cancer Center Léman (SCCL), Geneva University Hospitals, University of Geneva, Geneva, Switzerland
| | - Marta Calvo Tardón
- Laboratory of Immunobiology of brain tumors, Center for Translational Research in Onco-Hematology, Geneva University Hospitals, and University of Geneva, Geneva, Switzerland
| | - Mayra Yebra
- Department of Surgery, Moores Cancer Center, University of California San Diego, La Jolla, CA, USA
| | - Alexey Koval
- Department of Cell Physiology and Metabolism, Medical School, University of Geneva, Geneva, Switzerland
| | - Eliana Marinari
- Laboratory of Tumor Immunology, Department of Oncology, Center for Translational Research in Onco-Hematology, Swiss Cancer Center Léman (SCCL), Geneva University Hospitals, University of Geneva, Geneva, Switzerland
| | - Viviane Bes
- Laboratory of Immunobiology of brain tumors, Center for Translational Research in Onco-Hematology, Geneva University Hospitals, and University of Geneva, Geneva, Switzerland
| | - Alexandre Guérin
- Department of Pathology and Immunology, Medical School, University of Geneva, Geneva, Geneva, Switzerland
| | - Mathilde Allard
- Laboratory of Tumor Immunology, Department of Oncology, Center for Translational Research in Onco-Hematology, Swiss Cancer Center Léman (SCCL), Geneva University Hospitals, University of Geneva, Geneva, Switzerland
| | - Sten Ilmjärv
- Department of Pathology and Immunology, Medical School, University of Geneva, Geneva, Geneva, Switzerland
| | - Vladimir L Katanaev
- Department of Cell Physiology and Metabolism, Medical School, University of Geneva, Geneva, Switzerland
| | - Paul R Walker
- Laboratory of Immunobiology of brain tumors, Center for Translational Research in Onco-Hematology, Geneva University Hospitals, and University of Geneva, Geneva, Switzerland
| | - Karl-Heinz Krause
- Department of Pathology and Immunology, Medical School, University of Geneva, Geneva, Geneva, Switzerland
| | - Valérie Dutoit
- Laboratory of Tumor Immunology, Department of Oncology, Center for Translational Research in Onco-Hematology, Swiss Cancer Center Léman (SCCL), Geneva University Hospitals, University of Geneva, Geneva, Switzerland
| | - Jann N Sarkaria
- Department of Radiation Oncology, Mayo Clinic, Rochester, MN, USA
| | - Pierre-Yves Dietrich
- Laboratory of Tumor Immunology, Department of Oncology, Center for Translational Research in Onco-Hematology, Swiss Cancer Center Léman (SCCL), Geneva University Hospitals, University of Geneva, Geneva, Switzerland
| | - Érika Cosset
- Laboratory of Tumor Immunology, Department of Oncology, Center for Translational Research in Onco-Hematology, Swiss Cancer Center Léman (SCCL), Geneva University Hospitals, University of Geneva, Geneva, Switzerland.
| |
Collapse
|
16
|
Overexpression of miRNA-9 enhances galectin-3 levels in oral cavity cancers. Mol Biol Rep 2021; 48:3979-3989. [PMID: 34021445 DOI: 10.1007/s11033-021-06398-7] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2021] [Accepted: 05/03/2021] [Indexed: 12/13/2022]
Abstract
Oral cavity cancer (OCC) is the predominant subtype of head and neck cancer (HNC) and has up to 50% mortality. Genome-wide microRNA (miR) sequencing data indicates overexpression of miR-9-5p in HNC tumours, however, the biological role of miR-9-5p in OCC is complex; it can either act as a tumour suppressor or an oncomir, regulating many target genes at the post-transcriptional level. We have investigated the overexpression of miR-9-5p in three OCC cell lines. We have evaluated its expression levels and Galectin-3 as potential biomarkers in saliva samples collected from controls and OCC patients. We found that over expression of miR-9-5p in OCC cell lines resulted in a significant reduction in cell proliferation and migration, and an increase in apoptosis, which was paralleled by an increase in Galectin-3 secretion and export of Galectin-3 protein. Our data are consistent with miR-9-5p being a modulator of Galectin-3 via the AKT/γ-catenin pathway. In addition, the positive correlation between the levels of miR-9-5p expression and secreted Galectin-3 in saliva reflects a similar relationship in vivo, and supports the utility of their integrative evaluation in OCC. Our findings indicate that both miR-9-5p and Galectin-3 are critical biomolecules in the progression of OCC.
Collapse
|
17
|
Heine V, Hovorková M, Vlachová M, Filipová M, Bumba L, Janoušková O, Hubálek M, Cvačka J, Petrásková L, Pelantová H, Křen V, Elling L, Bojarová P. Immunoprotective neo-glycoproteins: Chemoenzymatic synthesis of multivalent glycomimetics for inhibition of cancer-related galectin-3. Eur J Med Chem 2021; 220:113500. [PMID: 33962190 DOI: 10.1016/j.ejmech.2021.113500] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2021] [Revised: 04/15/2021] [Accepted: 04/16/2021] [Indexed: 02/07/2023]
Abstract
Galectin-3 plays a crucial role in cancerogenesis; its targeting is a prospective pathway in cancer diagnostics and therapy. Multivalent presentation of glycans was shown to strongly increase the affinity of glycoconjugates to galectin-3. Further strengthening of interaction with galectin-3 may be accomplished using artificial glycomimetics with apt aryl substitutions. We established a new, as yet undescribed chemoenzymatic method to produce selective C-3-substituted N,N'-diacetyllactosamine glycomimetics and coupled them to human serum albumin. From a library of enzymes, only β-N-acetylhexosaminidase from Talaromyces flavus was able to efficiently synthesize the C-3-propargylated disaccharide. Various aryl residues were attached to the functionalized N,N'-diacetyllactosamine via click chemistry to assess the impact of the aromatic substitution. In ELISA-type assays with galectin-3, free glycomimetics exhibited up to 43-fold stronger inhibitory potency to Gal-3 than the lactose standard. Coupling to human serum albumin afforded multivalent neo-glycoproteins with up to 4209-fold increased inhibitory potency per glycan compared to the monovalent lactose standard. Surface plasmon resonance brought further information on the kinetics of galectin-3 inhibition. The potential of prepared neo-glycoproteins to target galectin-3 was demonstrated on colorectal adenocarcinoma DLD-1 cells. We investigated the uptake of neo-glycoproteins into cells and observed limited non-specific transport into the cytoplasm. Therefore, neo-glycoproteins primarily act as efficient scavengers of exogenous galectin-3 of cancer cells, inhibiting its interaction with the cell surface, and protecting T-lymphocytes against galectin-3-induced apoptosis. The present neo-glycoproteins combine the advantage of a straightforward synthesis, selectivity, non-toxicity, and high efficiency for targeting exogenous galectin-3, with possible application in the immunomodulatory treatment of galectin-3-overexpressing cancers.
Collapse
Affiliation(s)
- Viktoria Heine
- Institute of Microbiology of the Czech Academy of Sciences, Vídeňská 1083, CZ-14220, Prague 4, Czech Republic; Institute of Biotechnology and Helmholtz Institute for Biomedical Engineering, RWTH Aachen, Pauwelstr. 20, D-52079 Aachen, Germany
| | - Michaela Hovorková
- Institute of Microbiology of the Czech Academy of Sciences, Vídeňská 1083, CZ-14220, Prague 4, Czech Republic; Department of Genetics and Microbiology, Faculty of Science, Charles University, Viničná 5, CZ-12843, Prague 2, Czech Republic
| | - Miluše Vlachová
- Institute of Microbiology of the Czech Academy of Sciences, Vídeňská 1083, CZ-14220, Prague 4, Czech Republic
| | - Marcela Filipová
- Institute of Macromolecular Chemistry of the Czech Academy of Sciences, Heyrovského Nám. 2, CZ-16206 Prague 6, Czech Republic
| | - Ladislav Bumba
- Institute of Microbiology of the Czech Academy of Sciences, Vídeňská 1083, CZ-14220, Prague 4, Czech Republic
| | - Olga Janoušková
- Institute of Macromolecular Chemistry of the Czech Academy of Sciences, Heyrovského Nám. 2, CZ-16206 Prague 6, Czech Republic
| | - Martin Hubálek
- Institute of Organic Chemistry and Biochemistry of the Czech Academy of Sciences, Flemingovo Náměstí 2, CZ-16610 Prague 6, Czech Republic
| | - Josef Cvačka
- Institute of Organic Chemistry and Biochemistry of the Czech Academy of Sciences, Flemingovo Náměstí 2, CZ-16610 Prague 6, Czech Republic
| | - Lucie Petrásková
- Institute of Microbiology of the Czech Academy of Sciences, Vídeňská 1083, CZ-14220, Prague 4, Czech Republic
| | - Helena Pelantová
- Institute of Microbiology of the Czech Academy of Sciences, Vídeňská 1083, CZ-14220, Prague 4, Czech Republic
| | - Vladimír Křen
- Institute of Microbiology of the Czech Academy of Sciences, Vídeňská 1083, CZ-14220, Prague 4, Czech Republic
| | - Lothar Elling
- Institute of Biotechnology and Helmholtz Institute for Biomedical Engineering, RWTH Aachen, Pauwelstr. 20, D-52079 Aachen, Germany
| | - Pavla Bojarová
- Institute of Microbiology of the Czech Academy of Sciences, Vídeňská 1083, CZ-14220, Prague 4, Czech Republic.
| |
Collapse
|
18
|
Ballester B, Milara J, Cortijo J. The role of mucin 1 in respiratory diseases. Eur Respir Rev 2021; 30:30/159/200149. [PMID: 33536260 DOI: 10.1183/16000617.0149-2020] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2020] [Accepted: 09/04/2020] [Indexed: 01/21/2023] Open
Abstract
Recent evidence has demonstrated that mucin 1 (MUC1) is involved in many pathological processes that occur in the lung. MUC1 is a transmembrane protein mainly expressed by epithelial and hematopoietic cells. It has a receptor-like structure, which can sense the external environment and activate intracellular signal transduction pathways through its cytoplasmic domain. The extracellular domain of MUC1 can be released to the external environment, thus acting as a decoy barrier to mucosal pathogens, as well as serving as a serum biomarker for the diagnosis and prognosis of several respiratory diseases such as lung cancer and interstitial lung diseases. Furthermore, bioactivated MUC1-cytoplasmic tail (CT) has been shown to act as an anti-inflammatory molecule in several airway infections and mediates the expression of anti-inflammatory genes in lung diseases such as chronic rhinosinusitis, chronic obstructive pulmonary disease and severe asthma. Bioactivated MUC1-CT has also been reported to interact with several effectors linked to cellular transformation, contributing to the progression of respiratory diseases such as lung cancer and pulmonary fibrosis. In this review, we summarise the current knowledge of MUC1 as a promising biomarker and drug target for lung disease.
Collapse
Affiliation(s)
- Beatriz Ballester
- Division of Pulmonary Sciences and Critical Care Medicine, School of Medicine, University of Colorado, Aurora, CO, USA .,CIBERES, Health Institute Carlos III, Valencia, Spain.,Both authors contributed equally to this work
| | - Javier Milara
- CIBERES, Health Institute Carlos III, Valencia, Spain.,Pharmacy Unit, Consorcio Hospital General de Valencia, Valencia, Spain.,Pharmacology Dept, University Jaume I, Castellon, Spain.,Both authors contributed equally to this work
| | - Julio Cortijo
- CIBERES, Health Institute Carlos III, Valencia, Spain.,Research and teaching Unit, Consorcio Hospital General de Valencia, Valencia, Spain.,Dept of Pharmacology, Faculty of Medicine, University of Valencia, Valencia, Spain
| |
Collapse
|
19
|
Ratan C, Cicily K D D, Nair B, Nath LR. MUC Glycoproteins: Potential Biomarkers and Molecular Targets for Cancer Therapy. Curr Cancer Drug Targets 2021; 21:132-152. [PMID: 33200711 DOI: 10.2174/1568009620666201116113334] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2020] [Revised: 08/02/2020] [Accepted: 10/04/2020] [Indexed: 02/08/2023]
Abstract
MUC proteins have great significance as prognostic and diagnostic markers as well as a potential target for therapeutic interventions in most cancers of glandular epithelial origin. These are high molecular weight glycosylated proteins located in the epithelial lining of several tissues and ducts. Mucins belong to a heterogeneous group of large O-glycoproteins that can be either secreted or membrane-bound. Glycosylation, a post-translational modification affects the biophysical, functional and biochemical properties and provides structural complexity for these proteins. Aberrant expression and glycosylation of mucins contribute to tumour survival and proliferation in many cancers, which in turn activates numerous signalling pathways such as NF-kB, ERα, HIF, MAPK, p53, c-Src, Wnt and JAK-STAT, etc. This subsequently induces cancer cell growth, proliferation and metastasis. The present review mainly demonstrates the functional aspects of MUC glycoproteins along with its unique signalling mechanism and role of aberrant glycosylation in cancer progression and therapeutics. The importance of MUC proteins and its subtypes in a wide spectrum of cancers including but not limited to breast cancer, colorectal cancer, endometrial and cervical cancer, lung cancer, primary liver cancer, pancreatic cancer, prostate cancer and ovarian cancer has been exemplified with significance in targeting the same. Several patents associated with the MUC proteins in the field of cancer therapy are also emphasized in the current review.
Collapse
Affiliation(s)
- Chameli Ratan
- Department of Pharmacognosy, Amrita School of Pharmacy, Amrita Vishwa Vidyapeetham, AIMS Health Science Campus, Ponekkara P. O., Kochi, Kerala 682041, India
| | - Dalia Cicily K D
- Department of Pharmacognosy, Amrita School of Pharmacy, Amrita Vishwa Vidyapeetham, AIMS Health Science Campus, Ponekkara P. O., Kochi, Kerala 682041, India
| | - Bhagyalakshmi Nair
- Department of Pharmacognosy, Amrita School of Pharmacy, Amrita Vishwa Vidyapeetham, AIMS Health Science Campus, Ponekkara P. O., Kochi, Kerala 682041, India
| | - Lekshmi R Nath
- Department of Pharmacognosy, Amrita School of Pharmacy, Amrita Vishwa Vidyapeetham, AIMS Health Science Campus, Ponekkara P. O., Kochi, Kerala 682041, India
| |
Collapse
|
20
|
Radziejewska I, Borzym-Kluczyk M, Leszczyńska K. Luteolin alters MUC1 extracellular domain, sT antigen, ADAM-17, IL-8, IL-10 and NF-κB expression in Helicobacter pylori-infected gastric cancer CRL-1739 cells: A preliminary study. Biomed Rep 2020; 14:19. [PMID: 33335725 PMCID: PMC7739866 DOI: 10.3892/br.2020.1395] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2020] [Accepted: 11/06/2020] [Indexed: 12/13/2022] Open
Abstract
Luteolin is a natural flavonoid possessing certain beneficial pharmacological properties, including anti-oxidant, anti-inflammatory, anti-microbial and anti-cancer properties. The majority of types of gastric cancer with chronic gastritis are caused by infection with Helicobacter pylori (H. pylori). The present study evaluated the effect of luteolin on a number of selected factors that are potentially involved in gastric cancer development. The study was performed using gastric cancer CRL-1739 cells treated with 30 µM luteolin and H. pylori alone or combined. ELISA and reverse transcription PCR were used to assess the expression levels of MUC1, GalNAcα-R (Tn antigen) and NeuAcα2-3Galβ1-3GalNAc-R (sT antigen), ADAM-17, IL-8, IL-10 and NF-κB. H. pylori and luteolin independently and in combination significantly reduced the expression levels of the extracellular domain of MUC1 in gastric cancer cells compared with the untreated control cells. ADAM-17 expression was reduced by treatment with the pathogen and luteolin. Additionally, both factors reduced sT antigen expression. Treatment with 30 ≤M luteolin significantly induced IL-8 expression at the mRNA and protein level, and the mRNA expression levels of IL-10 and NF-κB compared with the control. Both H. pylori and luteolin induced IL-8 protein expression. The present preliminary results suggest that luteolin may be used to treat patients with gastric cancer.
Collapse
Affiliation(s)
- Iwona Radziejewska
- Department of Medical Chemistry, Medical University of Bialystok, Bialystok, 15-222 Podlaskie Voivodeship, Poland
| | - Małgorzata Borzym-Kluczyk
- Department of Pharmaceutical Biochemistry, Medical University of Bialystok, Bialystok, 15-222 Podlaskie Voivodeship, Poland
| | - Katarzyna Leszczyńska
- Department of Microbiology, Medical University of Bialystok, Bialystok, 15-222 Podlaskie Voivodeship, Poland
| |
Collapse
|
21
|
Abstract
Galectin-3 is a glycan-binding protein (GBP) that binds β-galactoside glycan structures to orchestrate a variety of important biological events, including the activation of hepatic stellate cells and regulation of immune responses. While the requisite glycan epitopes needed to bind galectin-3 have long been elucidated, the cellular glycoproteins that bear these glycan signatures remain unknown. Given the importance of the three-dimensional (3D) arrangement of glycans in dictating GBP interactions, strategies that allow the identification of GBP receptors in live cells, where the native glycan presentation and glycoprotein expression are preserved, have significant advantages over static and artificial systems. Here we describe the integration of a proximity labeling method and quantitative mass spectrometry to map the glycan and glycoprotein interactors for galectin-3 in live human hepatic stellate cells and peripheral blood mononuclear cells. Understanding the identity of the glycoproteins and defining the structures of the glycans will empower efforts to design and develop selective therapeutics to mitigate galectin-3-mediated biological events.
Collapse
|
22
|
Distinct Pattern of Endoplasmic Reticulum Protein Processing and Extracellular Matrix Proteins in Functioning and Silent Corticotroph Pituitary Adenomas. Cancers (Basel) 2020; 12:cancers12102980. [PMID: 33066652 PMCID: PMC7650558 DOI: 10.3390/cancers12102980] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2020] [Revised: 09/30/2020] [Accepted: 10/05/2020] [Indexed: 12/30/2022] Open
Abstract
Simple Summary Corticotroph pituitary adenomas present a spectrum of functionality regarding hormonal production, ranging from functioning to silent tumors. Moreover, they show different invasiveness and recurrent behavior profiles, the silent being considered an aggressive type of adenomas. Through analyses of global transcriptome and proteome, we show that both groups expressed genes and protein related to protein synthesis and vesicular transport, and present a distinct pattern of collagen/ extracellular matrix proteins. Endoplasmic reticulum protein processing is a key factor for hormone production in functioning corticotroph adenomas. Furthermore, a distinct cell adhesion profile in silent corticotroph adenomas may explain the aggressive behavior. Together, our findings shed light on the different repertoires of activated signaling pathways in corticotroph pituitary adenomas and may reveal new potential medical targets. Abstract Functioning (FCA) and silent corticotroph (SCA) pituitary adenomas act differently from a clinical perspective, despite both subtypes showing positive TBX19 (TPIT) and/or adrenocorticotropic hormone (ACTH) staining by immunohistochemistry. They are challenging to treat, the former due to functional ACTH production and consequently hypercortisolemia, and the latter due to invasive and recurrent behavior. Moreover, the molecular mechanisms behind their distinct behavior are not clear. We investigated global transcriptome and proteome changes in order to identify signaling pathways that can explain FCA and SCA differences (e.g., hormone production vs. aggressive growth). In the transcriptomic study, cluster analyses of differentially expressed genes revealed two distinct groups in accordance with clinical and histological classification. However, in the proteomic study, a greater degree of heterogeneity within the SCA group was found. Genes and proteins related to protein synthesis and vesicular transport were expressed by both adenoma groups, although different types and a distinct pattern of collagen/extracellular matrix proteins were presented by each group. Moreover, several genes related to endoplasmic reticulum protein processing were overexpressed in the FCA group. Together, our findings shed light on the different repertoires of activated signaling pathways in corticotroph adenomas, namely, the increased protein processing capacity of FCA and a specific pattern of adhesion molecules that may play a role in the aggressiveness of SCA.
Collapse
|
23
|
Bertuzzi S, Quintana JI, Ardá A, Gimeno A, Jiménez-Barbero J. Targeting Galectins With Glycomimetics. Front Chem 2020; 8:593. [PMID: 32850631 PMCID: PMC7426508 DOI: 10.3389/fchem.2020.00593] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2020] [Accepted: 06/08/2020] [Indexed: 01/06/2023] Open
Abstract
Among glycan-binding proteins, galectins, β-galactoside-binding lectins, exhibit relevant biological roles and are implicated in many diseases, such as cancer and inflammation. Their involvement in crucial pathologies makes them interesting targets for drug discovery. In this review, we gather the last approaches toward the specific design of glycomimetics as potential drugs against galectins. Different approaches, either using specific glycomimetic molecules decorated with key functional groups or employing multivalent presentations of lactose and N-acetyl lactosamine analogs, have provided promising results for binding and modulating different galectins. The review highlights the results obtained with these approximations, from the employment of S-glycosyl compounds to peptidomimetics and multivalent glycopolymers, mostly employed to recognize and/or detect hGal-1 and hGal-3.
Collapse
Affiliation(s)
- Sara Bertuzzi
- CIC bioGUNE, Basque Research Technology Alliance, Derio, Spain
| | - Jon I Quintana
- CIC bioGUNE, Basque Research Technology Alliance, Derio, Spain
| | - Ana Ardá
- CIC bioGUNE, Basque Research Technology Alliance, Derio, Spain
| | - Ana Gimeno
- CIC bioGUNE, Basque Research Technology Alliance, Derio, Spain
| | - Jesús Jiménez-Barbero
- CIC bioGUNE, Basque Research Technology Alliance, Derio, Spain.,Ikerbasque, Basque Foundation for Science, Bilbao, Spain.,Department of Organic Chemistry II, Faculty of Science and Technology, University of the Basque Country - UPV-EHU, Leioa, Spain
| |
Collapse
|
24
|
Filipová M, Bojarová P, Rodrigues Tavares M, Bumba L, Elling L, Chytil P, Gunár K, Křen V, Etrych T, Janoušková O. Glycopolymers for Efficient Inhibition of Galectin-3: In Vitro Proof of Efficacy Using Suppression of T Lymphocyte Apoptosis and Tumor Cell Migration. Biomacromolecules 2020; 21:3122-3133. [PMID: 32697592 DOI: 10.1021/acs.biomac.0c00515] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
The development of efficient galectin-3 (Gal-3) inhibitors draws attention in the field of anti-cancer therapy, especially due to the prominent role of extra- and intracellular Gal-3 in vital processes of cancerogenesis, such as immunosuppression, stimulation of tumor cells proliferation, survival, invasion, apoptotic resistance, and metastasis formation and progression. Here, by combining poly-LacNAc (Galβ4GlcNAc)-derived oligosaccharides with N-(2-hydroxypropyl) methacrylamide (HPMA) copolymers, we synthesized multivalent glycopolymer inhibitors with a high potential to target extracellular and intracellular Gal-3. The inhibitory capabilities of the best conjugate in the studied series were in the nanomolar range proving the excellent Gal-3 inhibitory potential. Moreover, thorough investigation of the inhibitory effect in the biological conditions showed that the glycopolymers strongly inhibited Gal-3-induced apoptosis of T lymphocytes and suppressed migration and spreading of colorectal, breast, melanoma, and prostate cancer cells. In sum, the strong inhibitory activity toward Gal-3, combined with favorable pharmacokinetics of HPMA copolymers ensuring enhanced tumor accumulation via the enhanced permeability and retention effect, nominate the glycopolymers containing LacdiNAc-LacNAc (GalNAcβ4GlcNAcβ3Galβ4GlcNAc) tetrasaccharide as promising tools for preclinical in anti-cancer therapy evaluation.
Collapse
Affiliation(s)
- Marcela Filipová
- Institute of Macromolecular Chemistry of the Czech Academy of Sciences, Heyrovského nám. 2, CZ-16206 Prague 6, Czech Republic
| | - Pavla Bojarová
- Institute of Microbiology of the Czech Academy of Sciences, Vídeňská 1083, CZ-14220 Prague 4 Czech Republic.,Department of Health Care Disciplines and Population Protection, Faculty of Biomedical Engineering, Czech Technical University in Prague, Sítná Sq. 3105, CZ-27201 Kladno, Czech Republic
| | - Marina Rodrigues Tavares
- Institute of Macromolecular Chemistry of the Czech Academy of Sciences, Heyrovského nám. 2, CZ-16206 Prague 6, Czech Republic
| | - Ladislav Bumba
- Institute of Microbiology of the Czech Academy of Sciences, Vídeňská 1083, CZ-14220 Prague 4 Czech Republic
| | - Lothar Elling
- Institute of Biotechnology and Helmholtz Institute for Biomedical Engineering, RWTH Aachen, Pauwelstr. 20, D-52079 Aachen, Germany
| | - Petr Chytil
- Institute of Macromolecular Chemistry of the Czech Academy of Sciences, Heyrovského nám. 2, CZ-16206 Prague 6, Czech Republic
| | - Kristýna Gunár
- Institute of Macromolecular Chemistry of the Czech Academy of Sciences, Heyrovského nám. 2, CZ-16206 Prague 6, Czech Republic
| | - Vladimír Křen
- Institute of Microbiology of the Czech Academy of Sciences, Vídeňská 1083, CZ-14220 Prague 4 Czech Republic
| | - Tomáš Etrych
- Institute of Macromolecular Chemistry of the Czech Academy of Sciences, Heyrovského nám. 2, CZ-16206 Prague 6, Czech Republic
| | - Olga Janoušková
- Institute of Macromolecular Chemistry of the Czech Academy of Sciences, Heyrovského nám. 2, CZ-16206 Prague 6, Czech Republic
| |
Collapse
|
25
|
Shimada C, Xu R, Al-Alem L, Stasenko M, Spriggs DR, Rueda BR. Galectins and Ovarian Cancer. Cancers (Basel) 2020; 12:cancers12061421. [PMID: 32486344 PMCID: PMC7352943 DOI: 10.3390/cancers12061421] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Revised: 05/26/2020] [Accepted: 05/28/2020] [Indexed: 02/07/2023] Open
Abstract
Ovarian cancer is known for its aggressive pathological features, including the capacity to undergo epithelial to mesenchymal transition, promoting angiogenesis, metastatic potential, chemoresistance, inhibiting apoptosis, immunosuppression and promoting stem-like features. Galectins, a family of glycan-binding proteins defined by a conserved carbohydrate recognition domain, can modulate many of these processes, enabling them to contribute to the pathology of ovarian cancer. Our goal herein was to review specific galectin members identified in the context of ovarian cancer, with emphasis on their association with clinical and pathological features, implied functions, diagnostic or prognostic potential and strategies being developed to disrupt their negative actions.
Collapse
Affiliation(s)
- Chisa Shimada
- Department of Obstetrics and Gynecology, Vincent Center for Reproductive Biology, Massachusetts General Hospital, Boston, MA 02114, USA; (C.S.); (R.X.); (L.A.-A.); (D.R.S.)
- Obstetrics, Gynecology, and Reproductive Biology, Harvard Medical School, Boston, MA 02115, USA
| | - Rui Xu
- Department of Obstetrics and Gynecology, Vincent Center for Reproductive Biology, Massachusetts General Hospital, Boston, MA 02114, USA; (C.S.); (R.X.); (L.A.-A.); (D.R.S.)
- Obstetrics, Gynecology, and Reproductive Biology, Harvard Medical School, Boston, MA 02115, USA
| | - Linah Al-Alem
- Department of Obstetrics and Gynecology, Vincent Center for Reproductive Biology, Massachusetts General Hospital, Boston, MA 02114, USA; (C.S.); (R.X.); (L.A.-A.); (D.R.S.)
- Obstetrics, Gynecology, and Reproductive Biology, Harvard Medical School, Boston, MA 02115, USA
| | - Marina Stasenko
- Gynecology Service, Memorial Sloan Kettering Cancer Center, 1275 York Ave, New York City, NY 10065, USA;
| | - David R. Spriggs
- Department of Obstetrics and Gynecology, Vincent Center for Reproductive Biology, Massachusetts General Hospital, Boston, MA 02114, USA; (C.S.); (R.X.); (L.A.-A.); (D.R.S.)
- Department of Hematology/Medical Oncology, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Bo R. Rueda
- Department of Obstetrics and Gynecology, Vincent Center for Reproductive Biology, Massachusetts General Hospital, Boston, MA 02114, USA; (C.S.); (R.X.); (L.A.-A.); (D.R.S.)
- Obstetrics, Gynecology, and Reproductive Biology, Harvard Medical School, Boston, MA 02115, USA
- Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, Massachusetts General Hospital, Boston, MA 02114, USA
- Correspondence:
| |
Collapse
|
26
|
Ballester B, Milara J, Cortijo J. Pirfenidone anti-fibrotic effects are partially mediated by the inhibition of MUC1 bioactivation. Oncotarget 2020; 11:1306-1320. [PMID: 32341751 PMCID: PMC7170494 DOI: 10.18632/oncotarget.27526] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2019] [Accepted: 03/03/2020] [Indexed: 12/19/2022] Open
Abstract
Pirfenidone is a pleiotropic molecule approved to treat idiopathic pulmonary fibrosis (IPF). Pirfenidone has demonstrated to downregulate transforming growth factor-β1 (TGF-β1) cellular effects. However, its anti-fibrotic mechanism remains unclear. Here, we aim to analyze the effects of pirfenidone on the TGF-β1 canonical and non-canonical pathways, as well as, on the most characteristic IPF cellular processes. Results observed in this work showed that TGF-β1-induced canonical SMAD3 and non-canonical ERK1/2 phosphorylations were not inhibited by pirfenidone in alveolar A549 and lung fibroblasts MRC5 cells. In contrast, pirfenidone inhibited TGF-β1-induced MUC1-CT Thr41 (1224) and Tyr46 (1229) phosphorylations, thus reducing the β-catenin activation. Additionally, immunoprecipitation and immunofluorescence studies in ATII cells and lung fibroblasts showed that pirfenidone inhibited the formation and nuclear translocation of the transcriptional fibrotic TGF-β1-induced phospho-SMAD3/MUC1-CT/active-β-catenin complex, and consequently the SMAD-binding element activation (SBE). This study provided also evidence of the inhibitory effect of pirfenidone on the TGF-β1-induced ATII to mesenchymal and fibroblast to myofibroblast transitions, fibroblast proliferation and ATII and fibroblast senescence. Therefore, it indicates that pirfenidone’s inhibitory effect on TGF-β1-induced fibrotic cellular processes is mediated by the inhibition of MUC1-CT phosphorylation, β-catenin activation, nuclear complex formation of phospho-SMAD3/MUC1-CT/active β-catenin and SBE activation, which may be of value to further develop anti-fibrotic IPF therapies.
Collapse
Affiliation(s)
- Beatriz Ballester
- Department of Pharmacology, Faculty of Medicine, University of Valencia, Valencia, Spain.,CIBERES, Health Institute Carlos III, Valencia, Spain.,These authors contributed equally to this work
| | - Javier Milara
- Department of Pharmacology, Faculty of Medicine, University of Valencia, Valencia, Spain.,CIBERES, Health Institute Carlos III, Valencia, Spain.,Health Research Institute INCLIVA, Valencia, Spain.,Pharmacy Unit, Clinic University Hospital, Valencia, Spain.,These authors contributed equally to this work
| | - Julio Cortijo
- Department of Pharmacology, Faculty of Medicine, University of Valencia, Valencia, Spain.,CIBERES, Health Institute Carlos III, Valencia, Spain.,Research and Teaching Unit, University General Hospital Consortium, Valencia, Spain
| |
Collapse
|
27
|
Mito A, Kumazawa-Inoue K, Kojima-Aikawa K. ZG16p, an Animal Homologue of Plant β-Prism Fold Lectins: Purification Methods of Natural and Recombinant ZG16p and Inhibition Assay of Cancer Cell Growth Using ZG16p. Methods Mol Biol 2020; 2132:339-347. [PMID: 32306341 DOI: 10.1007/978-1-0716-0430-4_33] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
ZG16p is a soluble 16-kDa protein abundantly expressed in the pancreas and gut, and has a β-prism fold structure similar to that of mannose-binding Jacalin-related lectins (mJRLs) such as BanLec, Heltuba, and Artocarpin. ZG16p binds to mannose via the well-conserved GXXXD loop among mJRLs and sulfated glycosaminoglycans (e.g., heparin and heparan sulfate) via the basic patch of molecular surface. In addition to the above binding activities, ZG16p has inhibitory activity against proliferation of colon cancer cells. This manuscript describes purification of rat pancreatic ZG16p and recombinant ZG16p expressed in Escherichia coli expression system, and cell growth inhibition assay using ZG16p as an inhibitor.
Collapse
Affiliation(s)
- Akiko Mito
- Natural Science Division, Faculty of Science, Ochanomizu University, Tokyo, Japan
| | - Kaori Kumazawa-Inoue
- Natural Science Division, Faculty of Science, Ochanomizu University, Tokyo, Japan
| | - Kyoko Kojima-Aikawa
- Natural Science Division, Faculty of Science, Ochanomizu University, Tokyo, Japan.
- Natural Science Division, Faculty of Core Research, Ochanomizu University, Tokyo, Japan.
| |
Collapse
|
28
|
Milara J, Ballester B, Montero P, Escriva J, Artigues E, Alós M, Pastor-Clerigues A, Morcillo E, Cortijo J. MUC1 intracellular bioactivation mediates lung fibrosis. Thorax 2019; 75:132-142. [PMID: 31801904 DOI: 10.1136/thoraxjnl-2018-212735] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2018] [Revised: 11/05/2019] [Accepted: 11/15/2019] [Indexed: 12/30/2022]
Abstract
BACKGROUND Serum KL6/mucin 1 (MUC1) has been identified as a potential biomarker in idiopathic pulmonary fibrosis (IPF), but the role of MUC1 intracellular bioactivation in IPF is unknown. OBJECTIVE To characterise MUC1 intracellular bioactivation in IPF. METHODS AND RESULTS The expression and phosphorylation of Thr41 and Tyr46 on the intracellular MUC1-cytoplasmic tail (CT) was increased in patients with IPF (n=22) compared with healthy subjects (n=21) and localised to fibroblasts and hyperplastic alveolar type II cells. Transforming growth factor (TGF)-β1 phosphorylated SMAD3 and thereby increased the phosphorylation of MUC1-CT Thr41 and Tyr46 in lung fibroblasts and alveolar type II cells, activating β-catenin to form a phospho-Smad3/MUC1-CT and MUC1-CT/β-catenin nuclear complex. This nuclear complex promoted alveolar epithelial type II and fibroblast to myofibroblast transitions, as well as cell senescence and fibroblast proliferation. The inhibition of MUC1-CT nuclear translocation using the inhibitor, GO-201 or silencing MUC1 by siRNA, reduced myofibroblast transition, senescence and proliferation in vitro. Bleomycin-induced lung fibrosis was reduced in mice treated with GO-201 and in MUC1-knockout mice. The profibrotic lectin, galectin-3, directly activated MUC1-CT and served as a bridge between the TGF-β receptor and the MUC1-C domain, indicating TGF-β1-dependent and TGF-β1-independent intracellular bioactivation of MUC1. CONCLUSIONS MUC1 intracellular bioactivation is enhanced in IPF and promotes fibrotic processes that could represent potential druggable targets for IPF.
Collapse
Affiliation(s)
- Javier Milara
- Pharmacy, Hospital Clínico Universitario de Valencia, Valencia, Spain
| | - Beatriz Ballester
- Pharmacology, Universitat de Valencia Facultat de Medicina i Odontologia, Valencia, Spain
| | - Paula Montero
- Farmacología, Universitat de Valencia Facultat de Medicina i Odontologia, Valencia, Spain
| | - Juan Escriva
- Thoracic Surgery, Hospital Universitari i Politecnic La Fe, Valencia, Spain
| | - Enrique Artigues
- Surgery, Hospital General Universitari de València, Valencia, Spain
| | - Manuel Alós
- Pharmacy, Hospital Clínico Universitario de Valencia, Valencia, Spain
| | - Alfonso Pastor-Clerigues
- Unidad de Investigación y Docencia, Consorci Hospital General Universitari de Valencia, Valencia, Spain
| | | | - Julio Cortijo
- Pharmacology, University of Valencia, Valencia, Spain
| |
Collapse
|
29
|
Yamashita T, Mori Y, Alzaaqi SM, Yashiro M, Sawada T, Hirakawa K, Nakada H. Induction of Trop-2 expression through the binding of galectin-3 to MUC1. Biochem Biophys Res Commun 2019; 516:44-49. [PMID: 31196625 DOI: 10.1016/j.bbrc.2019.06.003] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2019] [Accepted: 06/01/2019] [Indexed: 02/07/2023]
Abstract
Both mucin 1 (MUC1) and trophoblast cell surface antigen 2 (Trop-2) are overexpressed in various epithelial tumor cells, and their high expression is correlated with a poor prognosis. Both proteins were expressed in a human breast cancer cell line, MCF-7 cells, but neither was in a human colon cancer cell line, HCT116 cells. When MUC1 cDNA was introduced into HCT116 cells (HCT116/MUC1), expression of Trop-2 was induced. Reciprocally, treatment of MCF-7 cells with MUC1 siRNA reduced the level of Trop-2. Mithramycin A, an inhibitor of specificity protein 1 (Sp1) transcription factor, effectively inhibited the expression of Trop-2. Consistently, treatment with Sp1 siRNA reduced the expression of Trop-2. To reveal the relationship between MUC1 and Sp1, coimmunoprecipitation assays were performed. Sp1 was coimmunoprecipitated with MUC1 and the level of coimmunoprecipitated Sp1 increased in relation to the level of induced Trop-2. It is known that galectin-3 is an endogenous ligand of MUC1. Binding of galectin-3 to MUC1 elevated the recruitment of Sp1 to MUC1, and knockdown of galectin-3 reduced the level of Trop-2. These results suggest that the binding of galectin-3 to MUC1 enhances the recruitment of Sp1, leading to promotion of the transcription of Trop-2.
Collapse
Affiliation(s)
- Tomoko Yamashita
- Department of Molecular Biosciences, Faculty of Life Sciences, Kyoto Sangyo University, Kyoto, 603-8555, Japan
| | - Yugo Mori
- Department of Molecular Biosciences, Faculty of Life Sciences, Kyoto Sangyo University, Kyoto, 603-8555, Japan
| | - Shouq M Alzaaqi
- Department of Molecular Biosciences, Faculty of Life Sciences, Kyoto Sangyo University, Kyoto, 603-8555, Japan
| | - Masakazu Yashiro
- Department of Surgical Oncology, Osaka City University Graduate School of Medicine, Osaka, 545-8585, Japan; Molecular Oncology and Therapeutics, Osaka City University Graduate School of Medicine, Osaka, 545-8585, Japan
| | - Tetsuji Sawada
- Department of Surgical Oncology, Osaka City University Graduate School of Medicine, Osaka, 545-8585, Japan
| | - Kosei Hirakawa
- Department of Surgical Oncology, Osaka City University Graduate School of Medicine, Osaka, 545-8585, Japan
| | - Hiroshi Nakada
- Department of Molecular Biosciences, Faculty of Life Sciences, Kyoto Sangyo University, Kyoto, 603-8555, Japan.
| |
Collapse
|
30
|
Fang T, Liu DD, Ning HM, Dan Liu, Sun JY, Huang XJ, Dong Y, Geng MY, Yun SF, Yan J, Huang RM. Modified citrus pectin inhibited bladder tumor growth through downregulation of galectin-3. Acta Pharmacol Sin 2018; 39:1885-1893. [PMID: 29769742 PMCID: PMC6289393 DOI: 10.1038/s41401-018-0004-z] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2017] [Revised: 01/07/2018] [Accepted: 01/08/2018] [Indexed: 01/26/2023] Open
Abstract
Modified citrus pectin (MCP) is a carbohydrate enriched complex, which has been implicated in cancer treatment and prevention. However, the effects of MCP on urinary bladder cancer (UBC) are unknown. In this study, MCP was first tested in T24 and J82 human UBC cells and showed the inhibition of cell viability by the sulforhodamine B (SRB) assay. The MCP-treated UBC cells exhibited G2/M phase arrest with the decrease of Cyclin B1 and phosphorylated Cdc2. Caspase-3 was also activated, leading to the cleavage of Caspase-3 and PARP. We further explored the possible molecular mechanisms upon MCP treatment in UBC cells. Reduction of galectin-3 was observed and followed with the inactivation of Akt signaling pathway. Of note, galectin-3 knockdown by RNA interference recapitulated the MCP-mediated anti-proliferation, cell cycle arrest and apoptosis. Moreover, oral administration of MCP to the T24 xenograft-bearing nude mice inhibited the tumor growth significantly (P < 0.05). Quantification analysis of immunohistochemistry staining for Ki67 and cleaved Caspase-3 confirmed the decrease of proliferation index (P < 0.05) and the increase of apoptosis index (P < 0.01) in 700 mg/kg MCP-fed UBC xenografts. Using the information from TCGA database, we revealed that the overexpression of galectin-3 was associated with high tumor grade with lymph node metastasis, poor overall survival in UBC patients. Considering the remarkable inhibitory effects of MCP on UBC cell proliferation and survival in vitro and in vivo mainly through galectin-3, which is upregulated in UBCs, MCP may become an attractive agent, as a natural dietary fiber, for prevention and therapy of UBCs.
Collapse
Affiliation(s)
- Tian Fang
- Department of Comparative Medicine, Jinling Hospital, Clinical School of Medical College of Nanjing University, Nanjing, 210002, China
| | - Dan-Dan Liu
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - He-Ming Ning
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
- Shanghai Tech University, Shanghai, 201210, China
| | - Dan Liu
- State Key Laboratory of Pharmaceutical Biotechnology and MOE Key Laboratory of Model Animals for Disease Study, Model Animal Research Center of Nanjing University, Nanjing, 210061, China
| | - Jing-Ya Sun
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
| | - Xiao-Jing Huang
- State Key Laboratory of Pharmaceutical Biotechnology and MOE Key Laboratory of Model Animals for Disease Study, Model Animal Research Center of Nanjing University, Nanjing, 210061, China
| | - Yu Dong
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
- Shanghai University, Shanghai, 200444, China
| | - Mei-Yu Geng
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
- Shanghai Tech University, Shanghai, 201210, China
| | - Shi-Feng Yun
- Department of Comparative Medicine, Jinling Hospital, Clinical School of Medical College of Nanjing University, Nanjing, 210002, China.
| | - Jun Yan
- State Key Laboratory of Pharmaceutical Biotechnology and MOE Key Laboratory of Model Animals for Disease Study, Model Animal Research Center of Nanjing University, Nanjing, 210061, China.
- Collaborative Innovation Center of Genetics and Development, Shanghai, 200438, China.
| | - Rui-Min Huang
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China.
- University of Chinese Academy of Sciences, Beijing, 100049, China.
| |
Collapse
|
31
|
Lillehoj EP, Guang W, Hyun SW, Liu A, Hegerle N, Simon R, Cross AS, Ishida H, Luzina IG, Atamas SP, Goldblum SE. Neuraminidase 1-mediated desialylation of the mucin 1 ectodomain releases a decoy receptor that protects against Pseudomonas aeruginosa lung infection. J Biol Chem 2018; 294:662-678. [PMID: 30429216 DOI: 10.1074/jbc.ra118.006022] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2018] [Revised: 11/13/2018] [Indexed: 01/19/2023] Open
Abstract
Pseudomonas aeruginosa (Pa) expresses an adhesin, flagellin, that engages the mucin 1 (MUC1) ectodomain (ED) expressed on airway epithelia, increasing association of MUC1-ED with neuraminidase 1 (NEU1) and MUC1-ED desialylation. The MUC1-ED desialylation unmasks both cryptic binding sites for Pa and a protease recognition site, permitting its proteolytic release as a hyperadhesive decoy receptor for Pa. We found here that intranasal administration of Pa strain K (PAK) to BALB/c mice increases MUC1-ED shedding into the bronchoalveolar compartment. MUC1-ED levels increased as early as 12 h, peaked at 24-48 h with a 7.8-fold increase, and decreased by 72 h. The a-type flagellin-expressing PAK strain and the b-type flagellin-expressing PAO1 strain stimulated comparable levels of MUC1-ED shedding. A flagellin-deficient PAK mutant provoked dramatically reduced MUC1-ED shedding compared with the WT strain, and purified flagellin recapitulated the WT effect. In lung tissues, Pa increased association of NEU1 and protective protein/cathepsin A with MUC1-ED in reciprocal co-immunoprecipitation assays and stimulated MUC1-ED desialylation. NEU1-selective sialidase inhibition protected against Pa-induced MUC1-ED desialylation and shedding. In Pa-challenged mice, MUC1-ED-enriched bronchoalveolar lavage fluid (BALF) inhibited flagellin binding and Pa adhesion to human airway epithelia by up to 44% and flagellin-driven motility by >30%. Finally, Pa co-administration with recombinant human MUC1-ED dramatically diminished lung and BALF bacterial burden, proinflammatory cytokine levels, and pulmonary leukostasis and increased 5-day survival from 0% to 75%. We conclude that Pa flagellin provokes NEU1-mediated airway shedding of MUC1-ED, which functions as a decoy receptor protecting against lethal Pa lung infection.
Collapse
Affiliation(s)
| | | | - Sang W Hyun
- Medicine, and.,U.S. Department of Veterans Affairs, Veterans Affairs Medical Center, Baltimore, Maryland 20201, and
| | - Anguo Liu
- Medicine, and.,U.S. Department of Veterans Affairs, Veterans Affairs Medical Center, Baltimore, Maryland 20201, and
| | - Nicolas Hegerle
- Medicine, and.,Institute for Global Health, University of Maryland School of Medicine, Baltimore, Maryland 20201
| | - Raphael Simon
- Medicine, and.,Institute for Global Health, University of Maryland School of Medicine, Baltimore, Maryland 20201
| | - Alan S Cross
- Medicine, and.,Institute for Global Health, University of Maryland School of Medicine, Baltimore, Maryland 20201
| | - Hideharu Ishida
- Department of Applied Bio-organic Chemistry, Gifu University, Gifu 501-1193 Japan
| | - Irina G Luzina
- Medicine, and.,U.S. Department of Veterans Affairs, Veterans Affairs Medical Center, Baltimore, Maryland 20201, and
| | - Sergei P Atamas
- Medicine, and.,U.S. Department of Veterans Affairs, Veterans Affairs Medical Center, Baltimore, Maryland 20201, and
| | - Simeon E Goldblum
- Medicine, and.,U.S. Department of Veterans Affairs, Veterans Affairs Medical Center, Baltimore, Maryland 20201, and.,Pathology and
| |
Collapse
|
32
|
Thomas L, Pasquini LA. Galectin-3-Mediated Glial Crosstalk Drives Oligodendrocyte Differentiation and (Re)myelination. Front Cell Neurosci 2018; 12:297. [PMID: 30258354 PMCID: PMC6143789 DOI: 10.3389/fncel.2018.00297] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2018] [Accepted: 08/17/2018] [Indexed: 12/17/2022] Open
Abstract
Galectin-3 (Gal-3) is the only chimeric protein in the galectin family. Gal-3 structure comprises unusual tandem repeats of proline and glycine-rich short stretches bound to a carbohydrate-recognition domain (CRD). The present review summarizes Gal-3 functions in the extracellular and intracellular space, its regulation and its internalization and secretion, with a focus on the current knowledge of Gal-3 role in central nervous system (CNS) health and disease, particularly oligodendrocyte (OLG) differentiation, myelination and remyelination in experimental models of multiple sclerosis (MS). During myelination, microglia-expressed Gal-3 promotes OLG differentiation by binding glycoconjugates present only on the cell surface of OLG precursor cells (OPC). During remyelination, microglia-expressed Gal-3 favors an M2 microglial phenotype, hence fostering myelin debris phagocytosis through TREM-2b phagocytic receptor and OLG differentiation. Gal-3 is necessary for myelin integrity and function, as evidenced by myelin ultrastructural and behavioral studies from LGALS3-/- mice. Mechanistically, Gal-3 enhances actin assembly and reduces Erk 1/2 activation, leading to early OLG branching. Gal-3 later induces Akt activation and increases MBP expression, promoting gelsolin release and actin disassembly and thus regulating OLG final differentiation. Altogether, findings indicate that Gal-3 mediates the glial crosstalk driving OLG differentiation and (re)myelination and may be regarded as a target in the design of future therapies for a variety of demyelinating diseases.
Collapse
Affiliation(s)
- Laura Thomas
- Department of Biological Chemistry, School of Pharmacy and Biochemistry, University of Buenos Aires, Buenos Aires, Argentina.,Institute of Chemistry and Biological Physicochemistry (IQUIFIB), National Scientific and Technical Research Council (CONICET), Buenos Aires, Argentina
| | - Laura Andrea Pasquini
- Department of Biological Chemistry, School of Pharmacy and Biochemistry, University of Buenos Aires, Buenos Aires, Argentina.,Institute of Chemistry and Biological Physicochemistry (IQUIFIB), National Scientific and Technical Research Council (CONICET), Buenos Aires, Argentina
| |
Collapse
|
33
|
Santarsia S, Grosso AS, Trovão F, Jiménez-Barbero J, Carvalho AL, Nativi C, Marcelo F. Molecular Recognition of a Thomsen-Friedenreich Antigen Mimetic Targeting Human Galectin-3. ChemMedChem 2018; 13:2030-2036. [PMID: 30094951 DOI: 10.1002/cmdc.201800525] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2018] [Indexed: 11/11/2022]
Abstract
Overexpression of the Thomsen-Friedenreich (TF) antigen in cell membrane proteins occurs in 90 % of adenocarcinomas. Additionally, the binding of the TF antigen to human galectin-3 (Gal-3), also frequently overexpressed in malignancy, promotes cancer progression and metastasis. In this context, structures that interfere with this specific interaction have the potential to prevent cancer metastasis. A multidisciplinary approach combining the optimized synthesis of a TF antigen mimetic with NMR, X-ray crystallography methods, and isothermal titration calorimetry assays was used to unravel the molecular structural details that govern the Gal-3/TF mimetic interaction. The TF mimetic has a binding affinity for Gal-3 similar to that of the TF natural antigen and retains the binding epitope and bioactive conformation observed for the native antigen. Furthermore, from a thermodynamic perspective, a decrease in the enthalpic contribution was observed for the Gal-3/TF mimetic complex; however, this behavior is compensated by a favorable gain in entropy. From a structural perspective, these results establish our TF mimetic as a scaffold to design multivalent solutions to potentially interfere with Gal-3 aberrant interactions and for likely use in hampering Gal-3-mediated cancer cell adhesion and metastasis.
Collapse
Affiliation(s)
- Sabrina Santarsia
- Department of Chemistry Ugo Schiff, University of Florence, Via della Lastruccia, 13-50019, Sesto Fiorentino, Italy
| | - Ana Sofia Grosso
- UCIBIO, REQUIMTE, Departamento de Química, Faculdade De Ciências e Tecnologia, Universidade Nova de Lisboa, 2829-516, Caparica, Portugal
| | - Filipa Trovão
- UCIBIO, REQUIMTE, Departamento de Química, Faculdade De Ciências e Tecnologia, Universidade Nova de Lisboa, 2829-516, Caparica, Portugal
| | - Jesús Jiménez-Barbero
- CIC-bioGUNE Bizkaia, 48160, Derio, Spain.,Ikerbasque, Basque Foundation for Science, 48005, Bilbao, Spain.,Department of Organic Chemistry II, EHU-UPV, 48040, Leioa, Spain
| | - Ana Luísa Carvalho
- UCIBIO, REQUIMTE, Departamento de Química, Faculdade De Ciências e Tecnologia, Universidade Nova de Lisboa, 2829-516, Caparica, Portugal
| | - Cristina Nativi
- Department of Chemistry Ugo Schiff, University of Florence, Via della Lastruccia, 13-50019, Sesto Fiorentino, Italy
| | - Filipa Marcelo
- UCIBIO, REQUIMTE, Departamento de Química, Faculdade De Ciências e Tecnologia, Universidade Nova de Lisboa, 2829-516, Caparica, Portugal
| |
Collapse
|
34
|
Xu WW, Zheng CC, Huang YN, Chen WY, Yang QS, Ren JY, Wang YM, He QY, Liao HX, Li B. Synephrine Hydrochloride Suppresses Esophageal Cancer Tumor Growth and Metastatic Potential through Inhibition of Galectin-3-AKT/ERK Signaling. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2018; 66:9248-9258. [PMID: 30113849 DOI: 10.1021/acs.jafc.8b04020] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/08/2023]
Abstract
A library consisting of 429 food-source compounds was used to screen the natural products with anticancer properties in esophageal squamous cell carcinoma (ESCC). We demonstrated for the first time that synephrine, an active compound isolated from leaves of citrus trees, markedly suppressed cell proliferation (inhibition rate with 20 μM synephrine at day 5:71.1 ± 5.8% and 75.7 ± 6.2% for KYSE30 and KYSE270, respectively) and colony formation (inhibition rate with 10 μM synephrine: 86.5 ± 5.9% and 82.3 ± 4.5% for KYSE30 and KYSE270, respectively), as well as migration (inhibition rate with 10 μM synephrine: 76.9 ± 4.4% and 62.2 ± 5.8% for KYSE30 and KYSE270, respectively) and invasion abilities (inhibition rate with 10 μM synephrine: 73.3 ± 7.5% and 75.3 ± 3.4% for KYSE30 and KYSE270, respectively) of ESCC cells in a dose-dependent manner, without significant toxic effect on normal esophageal epithelial cells. Mechanistically, quantitative proteomics and bioinformatics analyses were performed to explore the synephrine-regulated proteins. Western blot and qRT-PCR data indicated that synephrine may downregulate Galectin-3 to inactivate AKT and ERK pathways. In addition, we found that the sensitivity of ESCC to fluorouracil (5-FU) could be enhanced by synephrine. Furthermore, in vivo experiments showed that synephrine had significant antitumor effect on ESCC tumor xenografts in nude mice (inhibition rate with 20 mg/kg synephrine is 61.3 ± 20.5%) without observed side effects on the animals. Taken together, synephrine, a food-source natural product, may be a potential therapeutic strategy in ESCC.
Collapse
Affiliation(s)
- Wen Wen Xu
- Institute of Biomedicine, College of Life Science and Technology , Jinan University , Guangzhou 510632 , P. R. China
- Guangdong Provincial Key Laboratory of Bioengineering Medicine, College of Life Science and Technology , Jinan University , Guangzhou 510632 , P. R. China
- National Engineering Research Center of Genetic Medicine , Jinan University , Guangzhou 510632 , P. R. China
| | - Can-Can Zheng
- Institute of Biomedicine, College of Life Science and Technology , Jinan University , Guangzhou 510632 , P. R. China
- Key Laboratory of Functional Protein Research of Guangdong Higher Education Institutes, Institute of Life and Health Engineering, College of Life Science and Technology , Jinan University , Guangzhou 510632 , P. R. China
| | - Yun-Na Huang
- Institute of Biomedicine, College of Life Science and Technology , Jinan University , Guangzhou 510632 , P. R. China
- Guangdong Provincial Key Laboratory of Bioengineering Medicine, College of Life Science and Technology , Jinan University , Guangzhou 510632 , P. R. China
- National Engineering Research Center of Genetic Medicine , Jinan University , Guangzhou 510632 , P. R. China
| | - Wen-You Chen
- Department of Thoracic Surgery, First Affiliated Hospital , Jinan University , Guangzhou 510632 , P. R. China
| | - Qing-Sheng Yang
- Department of Thoracic Surgery, First Affiliated Hospital , Jinan University , Guangzhou 510632 , P. R. China
| | - Jia-Yi Ren
- School of Traditional Chinese Medicine , Jinan University , Guangzhou 510632 , P. R. China
| | - Yue-Ming Wang
- Institute of Biomedicine, College of Life Science and Technology , Jinan University , Guangzhou 510632 , P. R. China
- Guangdong Provincial Key Laboratory of Bioengineering Medicine, College of Life Science and Technology , Jinan University , Guangzhou 510632 , P. R. China
- National Engineering Research Center of Genetic Medicine , Jinan University , Guangzhou 510632 , P. R. China
| | - Qing-Yu He
- Key Laboratory of Functional Protein Research of Guangdong Higher Education Institutes, Institute of Life and Health Engineering, College of Life Science and Technology , Jinan University , Guangzhou 510632 , P. R. China
| | - Hua-Xin Liao
- Institute of Biomedicine, College of Life Science and Technology , Jinan University , Guangzhou 510632 , P. R. China
- Guangdong Provincial Key Laboratory of Bioengineering Medicine, College of Life Science and Technology , Jinan University , Guangzhou 510632 , P. R. China
- National Engineering Research Center of Genetic Medicine , Jinan University , Guangzhou 510632 , P. R. China
| | - Bin Li
- Key Laboratory of Functional Protein Research of Guangdong Higher Education Institutes, Institute of Life and Health Engineering, College of Life Science and Technology , Jinan University , Guangzhou 510632 , P. R. China
| |
Collapse
|
35
|
Guo M, You C, Dou J. Role of transmembrane glycoprotein mucin 1 (MUC1) in various types of colorectal cancer and therapies: Current research status and updates. Biomed Pharmacother 2018; 107:1318-1325. [PMID: 30257347 DOI: 10.1016/j.biopha.2018.08.109] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2018] [Revised: 08/07/2018] [Accepted: 08/22/2018] [Indexed: 02/07/2023] Open
Abstract
Colorectal carcinoma (CRC) is the third most common malignant tumor in the world. In recent years, the morbidity and mortality of CRC have increased in the world due to increasingly ageing population, modern dietary habits, environmental change, genetic disorders and chronic intestinal inflammation. Despite recent advances in earlier detection and improvements in chemotherapy, the 5-year survival rate of patients with metastatic CRC remains low. Therefore, novel effective treatment strategies for primary or metastatic CRC have emerged to enhance cure rate as well as elongation of patient's survival. Immunotherapy has been proposed for a potentially effective therapeutic approach to the treatment of CRC. Tumor vaccination in preclinical and clinical studies has supported the antitumor activity induced by immunization with CRC cell vaccines. Epithelial cell molecule Mucin 1 (MUC1), a transmembrane glycoprotein aberrantly overexpressed in various cancers including CRC, has been used as a candidate target antigen in the peptide, dendritic cell, and whole tumor vaccines. Several clinical trials in progress reveal the immunogenicity and suitability of MUC1 that acted as immunotherapeutic vaccines for CRC/colorectal cancer stem cells (CCSC). The present review summarizes the potential roles of MUC1 on CRC/CCSC vaccines according to the latest data. Moreover, this review also discusses the novel strategies for targeting CCSC via inducing an immune response against MUC1 to achieve the best prevention and treatment effects in animal models and clinical trails.
Collapse
Affiliation(s)
- Mei Guo
- Department of Pathogenic Biology and Immunology, School of Medicine, Southeast University, Nanjing 210009, China
| | - Chengzhong You
- Department of General Surgery, Zhongda Hospital Affiliated to Southeast University, Nanjing 210009, China
| | - Jun Dou
- Department of Pathogenic Biology and Immunology, School of Medicine, Southeast University, Nanjing 210009, China.
| |
Collapse
|
36
|
Mito A, Nakano Y, Saitoh T, Gouraud SSS, Yamaguchi Y, Sato T, Sasaki N, Kojima-Aikawa K. Lectin ZG16p inhibits proliferation of human colorectal cancer cells via its carbohydrate-binding sites. Glycobiology 2018; 28:21-31. [PMID: 29069492 DOI: 10.1093/glycob/cwx088] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2017] [Accepted: 10/19/2017] [Indexed: 12/19/2022] Open
Abstract
Zymogen granule protein 16 (ZG16p) is a soluble lectin that binds to both mannose and heparin/heparan sulfate. It is highly expressed in the human digestive tract and is secreted into the mucus. In this study, we investigated the effect of ZG16p on the proliferation of human colorectal cancer cells. Overexpression of ZG16p in Caco-2 cells decreased cell growth. Recombinant ZG16p markedly inhibited proliferation of Caco-2, LS174T, HCT116 and HCT15 cells. Caco-2 cell growth was not inhibited by two mutated ZG16p proteins, D151A and M5 (K36A, R37A, R53A, R55A and R79A) lacking mannose- and heparin-binding activities, respectively. Immunofluorescent cell staining revealed that ZG16p-D151A maintained its binding to the Caco-2 cell surface, whereas ZG16p-M5 failed to bind to the cells. These results suggest that ZG16p interacts with the cell surface via basic amino acids substituted in ZG16p-M5 and inhibits Caco-2 cell proliferation via Asp151. In addition, growth of patient-derived colorectal tumor organoids in a 3D intestinal stem cell system was suppressed by ZG16p but not by ZG16p-M5. Taken together, our findings indicate that ZG16p inhibits the growth of colorectal cancer cells via its carbohydrate-binding sites in vitro and ex vivo. In this study, a novel pathway in cancer cell growth regulation through cell surface carbohydrate chains is suggested.
Collapse
Affiliation(s)
- Akiko Mito
- Graduate School of Humanities and Sciences, Ochanomizu University, 2-1-1 Otsuka, Bunkyo-ku, Tokyo 112-8610, Japan.,Program for Leading Graduate Schools, Ochanomizu University, 2-1-1 Otsuka, Bunkyo-ku, Tokyo 112-8610, Japan
| | - Yukiko Nakano
- Graduate School of Humanities and Sciences, Ochanomizu University, 2-1-1 Otsuka, Bunkyo-ku, Tokyo 112-8610, Japan
| | - Takako Saitoh
- Graduate School of Humanities and Sciences, Ochanomizu University, 2-1-1 Otsuka, Bunkyo-ku, Tokyo 112-8610, Japan
| | - Sabine S S Gouraud
- Leading Graduate School Promotion Center, Ochanomizu University, 2-1-1 Otsuka, Bunkyo-ku, Tokyo 112-8610, Japan
| | - Yoshiki Yamaguchi
- Structure Glycobiology Team, Systems Glycobiology Research Group, RIKEN Global Research Cluster, 2-1 Hirosawa, Wako, Saitama 351-0198, Japan
| | - Toshiro Sato
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo 160-8582, Japan
| | - Nobuo Sasaki
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo 160-8582, Japan
| | - Kyoko Kojima-Aikawa
- Natural Science Division, Faculty of Core Research, Ochanomizu University, 2-1-1 Otsuka, Bunkyo-ku, Tokyo 112-8610, Japan.,Institute for Human Life Innovation, Ochanomizu University, 2-1-1 Otsuka, Bunkyo-ku, Tokyo 112-8610, Japan
| |
Collapse
|
37
|
Enhanced cortical bone expansion in Lgals3-deficient mice during aging. Bone Res 2018; 6:7. [PMID: 30886760 PMCID: PMC6416267 DOI: 10.1038/s41413-017-0003-6] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2017] [Revised: 07/13/2017] [Accepted: 08/28/2017] [Indexed: 12/12/2022] Open
Abstract
Imbalances between bone formation and bone resorption, which can occur due to aging or sex hormone deprivation, result in decreased bone mass and an increased risk of fracture. Previous studies have suggested that the β-galactoside binding lectin, galectin-3, is involved in bone remodeling. We compared bone parameters of mice having null alleles of the galectin-3 gene (Lgals3-KO) with those of their wild-type littermates. Lgals3 deficiency increased cortical bone expansion at 36 weeks (wk) and preserved or enhanced bone mass in both male and female mutant mice. In addition, female Lgals3-KO mice were protected from age-related loss of trabecular bone. Histomorphometry and ex vivo primary cell differentiation assays showed increased osteoblastogenesis with little-to-no effect on osteoclastogenesis, suggesting the increased bone mass phenotype is primarily due to increased anabolism. Our study identifies galectin-3 as a negative regulator of bone formation and suggests that disruption of galectin-3 may be useful in preventing bone loss during aging. Researchers have identified a promising new drug target to reduce bone loss during aging, a protein called galectin-3. Bones undergo lifelong remodeling via resorption of old bone and generation of new bone. With aging, the balance tips towards resorption, weakening bones. Galectin-3 was known to be involved in bone remodeling and levels increased with age. Bart Williams and co-workers at the Van Andel Research Institute in Grand Rapids, USA, investigated whether the age-related increase in galectin-3 increased bone loss. Using mice lacking the gene encoding galectin-3, the researchers measured bone mass at different ages. In older mice, bone mass was preserved or even enhanced. Further investigation of bone cells showed the increase was probably due to increased bone formation, rather than decreased bone resorption. The researchers conclude that disrupting galectin-3 may help to prevent aging-related bone loss.
Collapse
|
38
|
Interaction of galectin-3 with MUC1 on cell surface promotes EGFR dimerization and activation in human epithelial cancer cells. Cell Death Differ 2017; 24:1937-1947. [PMID: 28731466 PMCID: PMC5635220 DOI: 10.1038/cdd.2017.119] [Citation(s) in RCA: 60] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2017] [Revised: 06/09/2017] [Accepted: 06/20/2017] [Indexed: 01/08/2023] Open
Abstract
Epidermal growth factor receptor (EGFR) is an important regulator of epithelial cell growth and survival in normal and cancerous tissues and is a principal therapeutic target for cancer treatment. EGFR is associated in epithelial cells with the heavily glycosylated transmembrane mucin protein MUC1, a natural ligand of galectin-3 that is overexpressed in cancer. This study reveals that the expression of cell surface MUC1 is a critical enhancer of EGF-induced EGFR activation in human breast and colon cancer cells. Both the MUC1 extracellular and intracellular domains are involved in EGFR activation but the predominant influence comes from its extracellular domain. Binding of galectin-3 to the MUC1 extracellular domain induces MUC1 cell surface polarization and increases MUC1–EGFR association. This leads to a rapid increase of EGFR homo-/hetero-dimerization and subsequently increased, and also prolonged, EGFR activation and signalling. This effect requires both the galectin-3 C-terminal carbohydrate recognition domain and its N-terminal ligand multi-merization domain. Thus, interaction of galectin-3 with MUC1 on cell surface promotes EGFR dimerization and activation in epithelial cancer cells. As MUC1 and galectin-3 are both commonly overexpressed in most types of epithelial cancers, their interaction and impact on EGFR activation likely makes important contribution to EGFR-associated tumorigenesis and cancer progression and may also influence the effectiveness of EGFR-targeted cancer therapy.
Collapse
|
39
|
Taniguchi T, Woodward AM, Magnelli P, McColgan NM, Lehoux S, Jacobo SMP, Mauris J, Argüeso P. N-Glycosylation affects the stability and barrier function of the MUC16 mucin. J Biol Chem 2017; 292:11079-11090. [PMID: 28487369 DOI: 10.1074/jbc.m116.770123] [Citation(s) in RCA: 51] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2016] [Revised: 05/03/2017] [Indexed: 11/06/2022] Open
Abstract
Transmembrane mucins are highly O-glycosylated glycoproteins that coat the apical glycocalyx on mucosal surfaces and represent the first line of cellular defense against infection and injury. Relatively low levels of N-glycans are found on transmembrane mucins, and their structure and function remain poorly characterized. We previously reported that carbohydrate-dependent interactions of transmembrane mucins with galectin-3 contribute to maintenance of the epithelial barrier at the ocular surface. Now, using MALDI-TOF mass spectrometry, we report that transmembrane mucin N-glycans in differentiated human corneal epithelial cells contain primarily complex-type structures with N-acetyllactosamine, a preferred galectin ligand. In N-glycosylation inhibition experiments, we find that treatment with tunicamycin and siRNA-mediated knockdown of the Golgi N-acetylglucosaminyltransferase I gene (MGAT1) induce partial loss of both total and cell-surface levels of the largest mucin, MUC16, and a concomitant reduction in glycocalyx barrier function. Moreover, we identified a distinct role for N-glycans in promoting MUC16's binding affinity toward galectin-3 and in causing retention of the lectin on the epithelial cell surface. Taken together, these studies define a role for N-linked oligosaccharides in supporting the stability and function of transmembrane mucins on mucosal surfaces.
Collapse
Affiliation(s)
- Takazumi Taniguchi
- From the Schepens Eye Research Institute and Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, Boston, Massachusetts 02114
| | - Ashley M Woodward
- From the Schepens Eye Research Institute and Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, Boston, Massachusetts 02114
| | | | - Nicole M McColgan
- From the Schepens Eye Research Institute and Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, Boston, Massachusetts 02114
| | - Sylvain Lehoux
- the Department of Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts 02215
| | - Sarah Melissa P Jacobo
- From the Schepens Eye Research Institute and Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, Boston, Massachusetts 02114
| | - Jérôme Mauris
- From the Schepens Eye Research Institute and Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, Boston, Massachusetts 02114
| | - Pablo Argüeso
- From the Schepens Eye Research Institute and Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, Boston, Massachusetts 02114,
| |
Collapse
|
40
|
mosGCTL-7, a C-Type Lectin Protein, Mediates Japanese Encephalitis Virus Infection in Mosquitoes. J Virol 2017; 91:JVI.01348-16. [PMID: 28250133 DOI: 10.1128/jvi.01348-16] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2016] [Accepted: 01/30/2017] [Indexed: 01/08/2023] Open
Abstract
Japanese encephalitis virus (JEV) is an arthropod-borne flavivirus prevalent in Asia and the Western Pacific and is the leading cause of viral encephalitis. JEV is maintained in a transmission cycle between mosquitoes and vertebrate hosts, but the molecular mechanisms by which the mosquito vector participates in transmission are unclear. We investigated the expression of all C-type lectins during JEV infection in Aedes aegypti The C-type lectin mosquito galactose-specific C-type lectin 7 (mosGCTL-7) (VectorBase accession no. AAEL002524) was significantly upregulated by JEV infection and facilitated infection in vivo and in vitro mosGCTL-7 bound to the N-glycan at N154 on the JEV envelope protein. This recognition of viral N-glycan by mosGCTL-7 is required for JEV infection, and we found that this interaction was Ca2+ dependent. After mosGCTL-7 bound to the glycan, mosPTP-1 bound to mosGCTL-7, promoting JEV entry. The viral burden in vivo and in vitro was significantly decreased by mosPTP-1 double-stranded RNA (dsRNA) treatment, and infection was abolished by anti-mosGCTL-7 antibodies. Our results indicate that the mosGCTL-7/mosPTP-1 pathway plays a key role in JEV infection in mosquitoes. An improved understanding of the mechanisms underlying flavivirus infection in mosquitoes will provide further opportunities for developing new strategies to control viral dissemination in nature.IMPORTANCE Japanese encephalitis virus is a mosquito-borne flavivirus and is the primary cause of viral encephalitis in the Asia-Pacific region. Twenty-four countries in the WHO Southeast Asia and Western Pacific regions have endemic JEV transmission, which exposes >3 billion people to the risks of infection, although JEV primarily affects children. C-type lectins are host factors that play a role in flavivirus infection in humans, swine, and other mammals. In this study, we investigated C-type lectin functions in JEV-infected Aedes aegypti and Culex pipiens pallens mosquitoes and cultured cells. JEV infection changed the expression of almost all C-type lectins in vivo and in vitro, and mosGCTL-7 bound to the JEV envelope protein via an N-glycan at N154. Cell surface mosPTP-1 interacted with the mosGCTL-7-JEV complex to facilitate virus infection in vivo and in vitro Our findings provide further opportunities for developing new strategies to control arbovirus dissemination in nature.
Collapse
|
41
|
Rajabi H, Kufe D. MUC1-C Oncoprotein Integrates a Program of EMT, Epigenetic Reprogramming and Immune Evasion in Human Carcinomas. Biochim Biophys Acta Rev Cancer 2017; 1868:117-122. [PMID: 28302417 DOI: 10.1016/j.bbcan.2017.03.003] [Citation(s) in RCA: 62] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2017] [Revised: 02/17/2017] [Accepted: 03/11/2017] [Indexed: 01/08/2023]
Abstract
The MUC1 gene evolved in mammalian species to provide protection of epithelia. The transmembrane MUC1 C-terminal subunit (MUC1-C) signals stress to the interior of the epithelial cell and, when overexpressed as in most carcinomas, functions as an oncoprotein. MUC1-C induces the epithelial-mesenchymal transition (EMT) by activating the inflammatory NF-κB p65 pathway and, in turn, the EMT-transcriptional repressor ZEB1. Emerging evidence has indicated that MUC1-C drives a program integrating the induction of EMT with activation of stem cell traits, epigenetic reprogramming and immune evasion. This mini-review focuses on the potential importance of this MUC1-C program in cancer progression.
Collapse
Affiliation(s)
- Hasan Rajabi
- Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA 02215, United States
| | - Donald Kufe
- Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA 02215, United States.
| |
Collapse
|
42
|
Nio-Kobayashi J, Abidin HBZ, Brown JK, Iwanaga T, Horne AW, Duncan WC. Cigarette smoking alters sialylation in the Fallopian tube of women, with implications for the pathogenesis of ectopic pregnancy. Mol Reprod Dev 2016; 83:1083-1091. [PMID: 27704662 DOI: 10.1002/mrd.22747] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2016] [Accepted: 10/03/2016] [Indexed: 11/08/2022]
Abstract
Sialylation creates a negative charge on the cell surface that can interfere with blastocyst implantation. For example, α2,6-sialylation on terminal galactose, catalyzed by the sialyltransferase ST6GAL1, inhibits the binding of galectin-1, a β-galactoside-binding lectin. We recently reported the potential involvement of galectin-1 and -3 in the pathogenesis of tubal ectopic pregnancy; however, the precise role of galectins and their ligand glycoconjugates remain unclear. Here, we investigated the expression of the genes encoding α2,3- and α2,6-galactoside sialyltransferases (ST3GAL1-6 and ST6GAL1-2) and the localization of sialic acids in the Fallopian tube of women with or without ectopic implantation. ST6GAL1 expression was higher in the mid-secretory phase than the proliferative phase of non-pregnant women (P < 0.0001), whereas ST6GAL1 (P < 0.0001), ST3GAL3 (P = 0.0029), ST3GAL5 (P = 0.0089), and ST3GAL6 (P = 0.0018) were all lower in Fallopian tubes with ectopic implantations. α2,3- and α2,6-sialic acids, however, both remained enriched on the surface of Fallopian tube epithelium. Cigarette smoking, a major risk factor for tubal ectopic pregnancy, was associated with reduced mid-secretory-phase expression of ST6GAL1 (P = 0.0298), but elevated expression of ST3GAL5 (P = 0.0006), an enzyme known to be involved in ciliogenesis. Indeed, sialic acid-containing ciliated inclusion cysts, which are associated with abnormal ciliogenesis, were observed within the epithelium at a higher frequency in women who smoked (P = 0.0177), suggesting that abnormal ciliogenesis is associated with smoking. Thus, cigarette smoking alters sialylation in the Fallopian tube epithelium, and is potentially a source of decreased tubal transport and increased receptivity for blastocyst in the human Fallopian tube. Mol. Reprod. Dev. 83: 1083-1091, 2016. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Junko Nio-Kobayashi
- Laboratory of Histology and Cytology, Hokkaido University Graduate School of Medicine, Sapporo, Japan
| | - Hazirah B Z Abidin
- MRC Centre for Reproductive Health, The Queen's Medical Research Institute, The University of Edinburgh, Edinburgh, United Kingdom
| | - Jeremy K Brown
- MRC Centre for Reproductive Health, The Queen's Medical Research Institute, The University of Edinburgh, Edinburgh, United Kingdom
| | - Toshihiko Iwanaga
- Laboratory of Histology and Cytology, Hokkaido University Graduate School of Medicine, Sapporo, Japan
| | - Andrew W Horne
- MRC Centre for Reproductive Health, The Queen's Medical Research Institute, The University of Edinburgh, Edinburgh, United Kingdom
| | - W Colin Duncan
- MRC Centre for Reproductive Health, The Queen's Medical Research Institute, The University of Edinburgh, Edinburgh, United Kingdom
| |
Collapse
|
43
|
Cascio S, Finn OJ. Intra- and Extra-Cellular Events Related to Altered Glycosylation of MUC1 Promote Chronic Inflammation, Tumor Progression, Invasion, and Metastasis. Biomolecules 2016; 6:biom6040039. [PMID: 27754373 PMCID: PMC5197949 DOI: 10.3390/biom6040039] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2016] [Revised: 08/29/2016] [Accepted: 09/27/2016] [Indexed: 12/12/2022] Open
Abstract
Altered glycosylation of mucin 1 (MUC1) on tumor cells compared to normal epithelial cells was previously identified as an important antigenic modification recognized by the immune system in the process of tumor immunosurveillance. This tumor form of MUC1 is considered a viable target for cancer immunotherapy. The importance of altered MUC1 glycosylation extends also to its role as a promoter of chronic inflammatory conditions that lead to malignant transformation and cancer progression. We review here what is known about the role of specific cancer-associated glycans on MUC1 in protein-protein interactions and intracellular signaling in cancer cells and in their adhesion to each other and the tumor stroma. The tumor form of MUC1 also creates a different landscape of inflammatory cells in the tumor microenvironment by controlling the recruitment of inflammatory cells, establishing specific interactions with dendritic cells (DCs) and macrophages, and facilitating tumor escape from the immune system. Through multiple types of short glycans simultaneously present in tumors, MUC1 acquires multiple oncogenic properties that control tumor development, progression, and metastasis at different steps of the process of carcinogenesis.
Collapse
Affiliation(s)
- Sandra Cascio
- Department of Immunology, University of Pittsburgh, Pittsburgh, PA 15261, USA.
- Fondazione Ri.Med, via Bandiera 11, Palermo 90133, Italy.
| | - Olivera J Finn
- Department of Immunology, University of Pittsburgh, Pittsburgh, PA 15261, USA.
| |
Collapse
|
44
|
Li Y, Yuan C, Wang L, Lu M, Wang Y, Wen Y, Yan R, Xu L, Song X, Li X. Transmembrane protein 147 (TMEM147): another partner protein of Haemonchus contortus galectin on the goat peripheral blood mononuclear cells (PBMC). Parasit Vectors 2016; 9:355. [PMID: 27337943 PMCID: PMC4918192 DOI: 10.1186/s13071-016-1640-0] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2016] [Accepted: 06/13/2016] [Indexed: 12/03/2022] Open
Abstract
Background Recombinant galectins of male and female Haemonchus contortus (rHco-gal-m/f) have been recognized as significant regulators of the functions of goat peripheral blood mononuclear cells (PBMC). In previous research, transmembrane protein 63A (TMEM63A) was identified as a partner protein in the regulation associated with H. contortus infection. However, in the identification of binding partners for galectins of male and female H. contortus (Hco-gal-m/f) by yeast two-hybrid (YTH) screening, it was found that the transmembrane protein 147 (TMEM147) could also bind to Hco-gal-m/f. In this study, the functions of TMEM147 in the regulations of H. contortus galectin on the goat PBMC were investigated. Methods To identify Hco-gal-m/f-interacting proteins, a yeast two-hybrid system to detect interactions was used. Co-immunoprecipitation and immunoblotting were used to validate the interaction between recombinant galectins of male H. contortus (rHco-gal-m) and candidate binding protein. The localization of TMEM147 in PBMC was explored by immunofluorescence in confocal imaging studies. Flow cytometry was used to determine the distribution of TMEM147 in T cells, B cells and monocytes in PBMC. The modulatory effects of rHco-gal-m and TMEM147 on cell proliferation, phagocytosis, nitric oxide production, migration, apoptosis and cytokine mRNA transcription were observed by co-incubation of rHco-gal-m and knockdown of the tmem147 gene. Results In this research, it was demonstrated that TMEM147 could bind to rHco-gal-m/f. Immunofluorescence assays showed that TMEM147 was localized to the cell membrane and within the cell membrane in goat PBMC. Flow cytometric analysis revealed that TMEM147 was expressed in all B cells and monocytes in goat PBMC. However, 3.8 % of T cells did not express this protein. Knockdown of the tmem147 gene using RNA interference (RNAi) showed that the interaction of galectin with TMEM147 mainly mediated cell proliferation, cell apoptosis, transcription of interleukin-10 (IL-10) and transforming growth factor-β1 (TGF-β1) of goat PBMC. This membrane protein, together with TMEM63A, was also related to the regulation of galectin on phagocytosis and nitric oxide production of goat PBMC. However, it might not be involved in the regulation of galectin on the migration and interferon-γ (IFN-γ) transcription of goat PBMC. Conclusions Our results showed that TMEM147 was a binding partner of Hco-gal-m/f and mediated the immunological regulation of Hco-gal-m/f on goat PBMC in a manner different to that of TMEM63A. Electronic supplementary material The online version of this article (doi:10.1186/s13071-016-1640-0) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Yan Li
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, People's Republic of China
| | - Cheng Yuan
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, People's Republic of China
| | - LiKun Wang
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, People's Republic of China
| | - MingMin Lu
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, People's Republic of China
| | - YuJian Wang
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, People's Republic of China
| | - YuLing Wen
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, People's Republic of China
| | - RuoFeng Yan
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, People's Republic of China
| | - LiXin Xu
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, People's Republic of China
| | - XiaoKai Song
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, People's Republic of China
| | - XiangRui Li
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, People's Republic of China.
| |
Collapse
|
45
|
Cardoso ACF, Andrade LNDS, Bustos SO, Chammas R. Galectin-3 Determines Tumor Cell Adaptive Strategies in Stressed Tumor Microenvironments. Front Oncol 2016; 6:127. [PMID: 27242966 PMCID: PMC4876484 DOI: 10.3389/fonc.2016.00127] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2016] [Accepted: 05/10/2016] [Indexed: 01/25/2023] Open
Abstract
Galectin-3 is a member of the β-galactoside-binding lectin family, whose expression is often dysregulated in cancers. While galectin-3 is usually an intracellular protein found in the nucleus and in the cytoplasm, under certain conditions, galectin-3 can be secreted by an yet unknown mechanism. Under stressing conditions (e.g., hypoxia and nutrient deprivation) galectin-3 is upregulated, through the activity of transcription factors, such as HIF-1α and NF-κB. Here, we review evidence that indicates a positive role for galectin-3 in MAPK family signal transduction, leading to cell proliferation and cell survival. Galectin-3 serves as a scaffold protein, which favors the spatial organization of signaling proteins as K-RAS. Upon secretion, extracellular galectin-3 interacts with a variety of cell surface glycoproteins, such as growth factor receptors, integrins, cadherins, and members of the Notch family, among other glycoproteins, besides different extracellular matrix molecules. Through its ability to oligomerize, galectin-3 forms lectin lattices that act as scaffolds that sustain the spatial organization of signaling receptors on the cell surface, dictating its maintenance on the plasma membrane or their endocytosis. Galectin-3 induces tumor cell, endothelial cell, and leukocyte migration, favoring either the exit of tumor cells from a stressed microenvironment or the entry of endothelial cells and leukocytes, such as monocytes/macrophages into the tumor organoid. Therefore, galectin-3 plays homeostatic roles in tumors, as (i) it favors tumor cell adaptation for survival in stressed conditions; (ii) upon secretion, galectin-3 induces tumor cell detachment and migration; and (iii) it attracts monocyte/macrophage and endothelial cells to the tumor mass, inducing both directly and indirectly the process of angiogenesis. The two latter activities are potentially targetable, and specific interventions may be designed to counteract the protumoral role of extracellular galectin-3.
Collapse
Affiliation(s)
- Ana Carolina Ferreira Cardoso
- Departamento de Radiologia e Oncologia, Faculdade de Medicina, Centro de Investigação Translacional em Oncologia, Instituto do Câncer do Estado de São Paulo, Universidade de São Paulo , São Paulo , Brasil
| | - Luciana Nogueira de Sousa Andrade
- Departamento de Radiologia e Oncologia, Faculdade de Medicina, Centro de Investigação Translacional em Oncologia, Instituto do Câncer do Estado de São Paulo, Universidade de São Paulo , São Paulo , Brasil
| | - Silvina Odete Bustos
- Departamento de Radiologia e Oncologia, Faculdade de Medicina, Centro de Investigação Translacional em Oncologia, Instituto do Câncer do Estado de São Paulo, Universidade de São Paulo , São Paulo , Brasil
| | - Roger Chammas
- Departamento de Radiologia e Oncologia, Faculdade de Medicina, Centro de Investigação Translacional em Oncologia, Instituto do Câncer do Estado de São Paulo, Universidade de São Paulo , São Paulo , Brasil
| |
Collapse
|
46
|
Lambropoulou M, Deftereou TE, Kynigopoulos S, Patsias A, Anagnostopoulos C, Alexiadis G, Kotini A, Tsaroucha A, Nikolaidou C, Kiziridou A, Papadopoulos N, Chatzaki E. Co-expression of galectin-3 and CRIP-1 in endometrial cancer: prognostic value and patient survival. Med Oncol 2015; 33:8. [PMID: 26708131 DOI: 10.1007/s12032-015-0723-7] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2015] [Accepted: 12/14/2015] [Indexed: 12/29/2022]
Abstract
Endometrial cancer is the sixth most common cancer in women. Galectin-3 (GAL-3) and CRIP-1 are multifunctional proteins which seem to be involved in many neoplasias. This study aims to point out correlations between clinicopathological findings and endometrial cancer patient survival to GAL-3 and CRIP-1 expression in order to enfold their diagnostic/prognostic potential. Tissues from 46 patients diagnosed with endometrial cancer were studied by immunohistochemistry, using monoclonal antibodies for GAL-3 and CRIP-1, and expression levels were correlated with clinicopathological findings and survival. Analysis was performed at single protein level or as co-expression. High expression of GAL-3 and CRIP-1 was independently associated with tumor depth and histological grade, respectively. Also, there was a significant correlation between high co-expression of the two proteins and the histological grade (aOR 2.66), the tumor depth (aOR 0.32) and the histological type (aOR 1.32), but not with the patients' age. Moreover, high expression of both proteins was observed in patients with shorter survival times. Interestingly, the co-expression of the two proteins exhibited some degree of monotony (Spearman's ρ = 0.768), indicating a common molecular pathway. This study provides evidence for a prognostic clinical potential of the combined study of GAL-3 and CRIP-1 in endometrial cancer. These factors are poorly studied in endometrium, and their role in the carcinogenetic process and on effective therapy awaits further elucidation.
Collapse
Affiliation(s)
- Maria Lambropoulou
- Laboratories of Histology-Embryology, School of Medicine, Democritus University of Thrace, Dragana, 68 100, Alexandroupolis, Greece.
| | - Theodora-Eleftheria Deftereou
- Laboratories of Histology-Embryology, School of Medicine, Democritus University of Thrace, Dragana, 68 100, Alexandroupolis, Greece
| | - Sryridon Kynigopoulos
- Laboratories of Histology-Embryology, School of Medicine, Democritus University of Thrace, Dragana, 68 100, Alexandroupolis, Greece
| | - Anargyros Patsias
- Laboratories of Histology-Embryology, School of Medicine, Democritus University of Thrace, Dragana, 68 100, Alexandroupolis, Greece
| | - Constantinos Anagnostopoulos
- Laboratories of Biochemistry, School of Medicine, Democritus University of Thrace, 68 100, Alexandroupolis, Greece
| | - Georgios Alexiadis
- Private Radiodiagnostic Center of Alexandroupolis, Theagenio Anticancer Hospital, Thessaloniki, Greece
| | - Athanasia Kotini
- Laboratories of Medical Physics, School of Medicine, Democritus University of Thrace, 68 100, Alexandroupolis, Greece
| | - Alexandra Tsaroucha
- Laboratories of Department of Surgery, School of Medicine, Democritus University of Thrace, 68 100, Alexandroupolis, Greece
| | - Christina Nikolaidou
- Laboratories of Histology-Embryology, School of Medicine, Democritus University of Thrace, Dragana, 68 100, Alexandroupolis, Greece
| | | | - Nikolaos Papadopoulos
- Laboratories of Histology-Embryology, School of Medicine, Democritus University of Thrace, Dragana, 68 100, Alexandroupolis, Greece
| | - Ekaterini Chatzaki
- Laboratories of Pharmacology, School of Medicine, Democritus University of Thrace, 68 100, Alexandroupolis, Greece
| |
Collapse
|