151
|
Rajput VK, MacKinnon A, Mandal S, Collins P, Blanchard H, Leffler H, Sethi T, Schambye H, Mukhopadhyay B, Nilsson UJ. A Selective Galactose-Coumarin-Derived Galectin-3 Inhibitor Demonstrates Involvement of Galectin-3-glycan Interactions in a Pulmonary Fibrosis Model. J Med Chem 2016; 59:8141-7. [PMID: 27500311 DOI: 10.1021/acs.jmedchem.6b00957] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
Synthesis of doubly 3-O-coumarylmethyl-substituted thiodigalactosides from bis-3-O-propargyl-thiodigalactoside resulted in highly selective and high affinity galectin-3 inhibitors. Mutant studies, structural analysis, and molecular modeling revealed that the coumaryl substituents stack onto arginine side chains. One inhibitor displayed efficacy in a murine model of bleomycin-induced lung fibrosis similar to that of a known nonselective galectin-1/galectin-3 inhibitor, which strongly suggests that blocking galectin-3 glycan recognition is an important antifibrotic drug target.
Collapse
Affiliation(s)
- Vishal K Rajput
- Indian Institute of Science Education and Research-Kolkata (IISER) Kolkata , Mohanpur Campus, P.O. BCKV Campus Main Office, Mohanpur, Nadia 741246, India.,Centre for Analysis and Synthesis, Department of Chemistry, Lund University , POB 124, SE-221 00 Lund, Sweden
| | - Alison MacKinnon
- MRC Centre for Inflammation Research, The Queen's Medical Research Institute, University of Edinburgh , Edinburgh EH16 4TJ, U.K
| | - Santanu Mandal
- Centre for Analysis and Synthesis, Department of Chemistry, Lund University , POB 124, SE-221 00 Lund, Sweden
| | - Patrick Collins
- Institute for Glycomics, Griffith University , Gold Coast Campus, Parklands Southport, Queensland 4222, Australia
| | - Helen Blanchard
- Institute for Glycomics, Griffith University , Gold Coast Campus, Parklands Southport, Queensland 4222, Australia
| | - Hakon Leffler
- Department of Laboratory Medicine, Section MIG, Lund University , BMC-C1228b, Klinikgatan 28, SE-221 84 Lund, Sweden
| | - Tariq Sethi
- Department of Respiratory Medicine and Allergy, Kings College , Denmark Hill Campus, Bessemer Road, London SE5 9RS, U.K
| | - Hans Schambye
- Galecto Biotech ApS, COBIS , Ole Maaloes vej 3, Copenhagen N, DK-2200, Denmark
| | - Balaram Mukhopadhyay
- Indian Institute of Science Education and Research-Kolkata (IISER) Kolkata , Mohanpur Campus, P.O. BCKV Campus Main Office, Mohanpur, Nadia 741246, India
| | - Ulf J Nilsson
- Centre for Analysis and Synthesis, Department of Chemistry, Lund University , POB 124, SE-221 00 Lund, Sweden
| |
Collapse
|
152
|
Campo VL, Marchiori MF, Rodrigues LC, Dias-Baruffi M. Synthetic glycoconjugates inhibitors of tumor-related galectin-3: an update. Glycoconj J 2016; 33:853-876. [PMID: 27526114 DOI: 10.1007/s10719-016-9721-z] [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: 04/04/2016] [Revised: 07/28/2016] [Accepted: 08/02/2016] [Indexed: 01/23/2023]
Abstract
Galectin-3 is associated with the development and malignancy of several types of tumor, mediating important tumor-related functions, such as tumorigenesis, neoplastic transformation, tumor cell survival, angiogenesis, tumor metastasis and regulation of apoptosis. Therefore, synthetic galectin-3 inhibitors are of utmost importance for development of new antitumor therapeutic strategies. In this review we present an updated selection of synthetic glycoconjugates inhibitors of tumor-related galectin-3, properly addressed as monosaccharide- and disaccharide-based inhibitors, and multivalent-based inhibitors, disclosuring relevant methods for their synthesis along with their inhibitory activities towards galectin-3. In general, Cu(I)-assisted 1,3-dipolar azide-alkyne cycloaddition (CuAAC) reactions were predominantly applied for the synthesis of the described inhibitors, which had their inhibitory activities against galectin-3 evaluated by fluorescence polarization, surface plasmon resonance (SPR), hemagglutination, ELISA and cell imaging assays. Overall, the presented synthetic glycoconjugates represent frontline galectin-3 inhibitors, finding important biomedical applications in cancer.
Collapse
Affiliation(s)
- Vanessa Leiria Campo
- Faculdade de Ciências Farmacêuticas de Ribeirão Preto, USP, Av. Café S/N, CEP, Ribeirão Preto, SP, 14040-903, Brazil.
| | - Marcelo Fiori Marchiori
- Faculdade de Ciências Farmacêuticas de Ribeirão Preto, USP, Av. Café S/N, CEP, Ribeirão Preto, SP, 14040-903, Brazil
| | - Lílian Cataldi Rodrigues
- Faculdade de Ciências Farmacêuticas de Ribeirão Preto, USP, Av. Café S/N, CEP, Ribeirão Preto, SP, 14040-903, Brazil
| | - Marcelo Dias-Baruffi
- Faculdade de Ciências Farmacêuticas de Ribeirão Preto, USP, Av. Café S/N, CEP, Ribeirão Preto, SP, 14040-903, Brazil
| |
Collapse
|
153
|
Delaine T, Collins P, MacKinnon A, Sharma G, Stegmayr J, Rajput VK, Mandal S, Cumpstey I, Larumbe A, Salameh BA, Kahl-Knutsson B, van Hattum H, van Scherpenzeel M, Pieters RJ, Sethi T, Schambye H, Oredsson S, Leffler H, Blanchard H, Nilsson UJ. Galectin-3-Binding Glycomimetics that Strongly Reduce Bleomycin-Induced Lung Fibrosis and Modulate Intracellular Glycan Recognition. Chembiochem 2016; 17:1759-70. [PMID: 27356186 DOI: 10.1002/cbic.201600285] [Citation(s) in RCA: 145] [Impact Index Per Article: 16.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2016] [Indexed: 11/12/2022]
Abstract
Discovery of glycan-competitive galectin-3-binding compounds that attenuate lung fibrosis in a murine model and that block intracellular galectin-3 accumulation at damaged vesicles, hence revealing galectin-3-glycan interactions involved in fibrosis progression and in intracellular galectin-3 activities, is reported. 3,3'-Bis-(4-aryltriazol-1-yl)thiodigalactosides were synthesized and evaluated as antagonists of galectin-1, -2, -3, and -4 N-terminal, -4 C-terminal, -7 and -8 N-terminal, -9 N-terminal, and -9 C-terminal domains. Compounds displaying low-nanomolar affinities for galectins-1 and -3 were identified in a competitive fluorescence anisotropy assay. X-ray structural analysis of selected compounds in complex with galectin-3, together with galectin-3 mutant binding experiments, revealed that both the aryltriazolyl moieties and fluoro substituents on the compounds are involved in key interactions responsible for exceptional affinities towards galectin-3. The most potent galectin-3 antagonist was demonstrated to act in an assay monitoring galectin-3 accumulation upon amitriptyline-induced vesicle damage, visualizing a biochemically/medically relevant intracellular lectin-carbohydrate binding event and that it can be blocked by a small molecule. The same antagonist administered intratracheally attenuated bleomycin-induced pulmonary fibrosis in a mouse model with a dose/response profile comparing favorably with that of oral administration of the marketed antifibrotic compound pirfenidone.
Collapse
Affiliation(s)
- Tamara Delaine
- Centre for Analysis and Synthesis, Department of Chemistry, Lund University, P. O. Box 124, 221 00, Lund, Sweden
| | - Patrick Collins
- Institute for Glycomics, Griffith University, Gold Coast Campus, Queensland, 4222, Australia
| | - Alison MacKinnon
- MRC Centre for Inflammation Research, The Queen's Medical Research Institute, University of Edinburgh, 49 Little France Crescent, Edinburgh, EH16 4TJ, UK
| | - G Sharma
- Department of Laboratory Medicine, Section MIG, Lund University, BMC-C1228b, Klinikgatan 28, 221 84, Lund, Sweden
| | - John Stegmayr
- Department of Laboratory Medicine, Section MIG, Lund University, BMC-C1228b, Klinikgatan 28, 221 84, Lund, Sweden
| | - Vishal K Rajput
- Centre for Analysis and Synthesis, Department of Chemistry, Lund University, P. O. Box 124, 221 00, Lund, Sweden
| | - Santanu Mandal
- Centre for Analysis and Synthesis, Department of Chemistry, Lund University, P. O. Box 124, 221 00, Lund, Sweden
| | - Ian Cumpstey
- Centre for Analysis and Synthesis, Department of Chemistry, Lund University, P. O. Box 124, 221 00, Lund, Sweden
| | - Amaia Larumbe
- Centre for Analysis and Synthesis, Department of Chemistry, Lund University, P. O. Box 124, 221 00, Lund, Sweden
| | - Bader A Salameh
- Centre for Analysis and Synthesis, Department of Chemistry, Lund University, P. O. Box 124, 221 00, Lund, Sweden.,Chemistry Department, The Hashemite University, P. O. Box 150459, Zarka, 13115, Jordan
| | - Barbro Kahl-Knutsson
- Department of Laboratory Medicine, Section MIG, Lund University, BMC-C1228b, Klinikgatan 28, 221 84, Lund, Sweden
| | - Hilde van Hattum
- Department of Medicinal Chemistry and Chemical Biology, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, P. O. Box 80082, 3508 TB, Utrecht, Netherlands
| | - Monique van Scherpenzeel
- Department of Medicinal Chemistry and Chemical Biology, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, P. O. Box 80082, 3508 TB, Utrecht, Netherlands.,Translational Metabolic Laboratory, 51 Radboud University Medical Center, P. O. Box 9101, 6500 HB, Nijmegen, Netherlands
| | - Roland J Pieters
- Department of Medicinal Chemistry and Chemical Biology, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, P. O. Box 80082, 3508 TB, Utrecht, Netherlands
| | - Tariq Sethi
- Department of Respiratory Medicine and Allergy, Kings College, 41 Denmark Hill Campus, Bessemer Road, London, SE5 9RJ, UK
| | - Hans Schambye
- Galecto Biotech ApS, COBIS, Ole Maaloes vej 3, Copenhagen N, 2200, Denmark
| | - Stina Oredsson
- Department of Biology, Lund University, P. O. Box 118, 221 00, Lund, Sweden
| | - Hakon Leffler
- Department of Laboratory Medicine, Section MIG, Lund University, BMC-C1228b, Klinikgatan 28, 221 84, Lund, Sweden
| | - Helen Blanchard
- Institute for Glycomics, Griffith University, Gold Coast Campus, Queensland, 4222, Australia.
| | - Ulf J Nilsson
- Centre for Analysis and Synthesis, Department of Chemistry, Lund University, P. O. Box 124, 221 00, Lund, Sweden.
| |
Collapse
|
154
|
Using CRISPR-Cas9 to quantify the contributions of O-glycans, N-glycans and Glycosphingolipids to human leukocyte-endothelium adhesion. Sci Rep 2016; 6:30392. [PMID: 27458028 PMCID: PMC4960646 DOI: 10.1038/srep30392] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2016] [Accepted: 06/30/2016] [Indexed: 01/20/2023] Open
Abstract
There is often interest in dissecting the relative contributions of the N-glycans, O-glycans and glycosphingolipids (GSLs) in regulating complex biological traits like cell signaling, adhesion, development and metastasis. To address this, we developed a CRISPR-Cas9 toolkit to selectively truncate each of these commonly expressed glycan-types. Here, O-glycan biosynthesis was truncated by knocking-out Core 1 β3Gal-T Specific Molecular Chaperone (COSMC), N-glycans by targeting the β1,2 GlcNAc-transferase (MGAT1) and GSLs by deleting UDP-glucose ceramide glucosyltransferase (UGCG). These reagents were applied to reveal the glycoconjugates regulating human myeloid cell adhesion to selectins under physiological shear-flow observed during inflammation. These functional studies show that leukocyte rolling on P- and L-selectin is ablated in cells lacking O-glycans, with N-glycan truncation also increasing cell rolling velocity on L-selectin. All three glycan families contributed to E-selectin dependent cell adhesion with N-glycans contributing to all aspects of the leukocyte adhesion cascade, O-glycans only being important during initial recruitment, and GSLs stabilizing slow cell rolling and the transition to firm arrest. Overall, the genome editing tools developed here may be broadly applied in studies of cellular glycosylation.
Collapse
|
155
|
Chen J, Gao J, Zhang M, Cai M, Xu H, Jiang J, Tian Z, Wang H. Systemic localization of seven major types of carbohydrates on cell membranes by dSTORM imaging. Sci Rep 2016; 6:30247. [PMID: 27453176 PMCID: PMC4958959 DOI: 10.1038/srep30247] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2016] [Accepted: 07/01/2016] [Indexed: 12/13/2022] Open
Abstract
Carbohydrates on the cell surface control intercellular interactions and play a vital role in various physiological processes. However, their systemic distribution patterns are poorly understood. Through the direct stochastic optical reconstruction microscopy (dSTORM) strategy, we systematically revealed that several types of representative carbohydrates are found in clustered states. Interestingly, the results from dual-color dSTORM imaging indicate that these carbohydrate clusters are prone to connect with one another and eventually form conjoined platforms where different functional glycoproteins aggregate (e.g., epidermal growth factor receptor, (EGFR) and band 3 protein). A thorough understanding of the ensemble distribution of carbohydrates on the cell surface paves the way for elucidating the structure-function relationship of cell membranes and the critical roles of carbohydrates in various physiological and pathological cell processes.
Collapse
Affiliation(s)
- Junling Chen
- State Key Laboratory of Electroanalytical Chemistry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilin 130022, P.R. China
- University of Chinese Academy of Sciences, Beijing 100049, P.R. China
| | - Jing Gao
- State Key Laboratory of Electroanalytical Chemistry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilin 130022, P.R. China
- University of Chinese Academy of Sciences, Beijing 100049, P.R. China
| | - Min Zhang
- State Key Laboratory of Electroanalytical Chemistry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilin 130022, P.R. China
- University of Chinese Academy of Sciences, Beijing 100049, P.R. China
| | - Mingjun Cai
- State Key Laboratory of Electroanalytical Chemistry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilin 130022, P.R. China
| | - Haijiao Xu
- State Key Laboratory of Electroanalytical Chemistry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilin 130022, P.R. China
| | - Junguang Jiang
- State Key Laboratory of Electroanalytical Chemistry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilin 130022, P.R. China
| | - Zhiyuan Tian
- School of Chemistry and Chemical Engineering, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Hongda Wang
- State Key Laboratory of Electroanalytical Chemistry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilin 130022, P.R. China
| |
Collapse
|
156
|
Chen J, Gao J, Cai M, Xu H, Jiang J, Tian Z, Wang H. Mechanistic insights into the distribution of carbohydrate clusters on cell membranes revealed by dSTORM imaging. NANOSCALE 2016; 8:13611-13619. [PMID: 27362510 DOI: 10.1039/c6nr02513g] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/06/2023]
Abstract
Cell surface carbohydrates play significant roles in many physiological processes and act as primary markers to indicate various cellular physiological states. The functions of carbohydrates are always associated with their expression and distribution on cell membranes. Based on our previous work, we found that carbohydrates tend to form clusters; however, the underlying mechanism of these clusters remains unknown. Through the direct stochastic optical reconstruction microscopy (dSTORM) strategy, we found that with the contributions of lipid raft as a stable factor and actin cytoskeleton as a restrictive factor, carbohydrate clusters can stably exist with restricted size. Additionally, we revealed that the formation of most carbohydrate clusters (Gal and GlcANc clusters) depended on the carbohydrate-binding proteins (i.e., galectins) cross-linking their specific carbohydrate ligands. Our results clarify the organizational mechanism of carbohydrates on cell surfaces from their formation, stable existence and size-restriction, which promotes a better understanding of the relationship between the function and distribution of carbohydrates, as well as the structure of cell membranes.
Collapse
Affiliation(s)
- Junling Chen
- State Key Laboratory of Electroanalytical Chemistry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilin 130022, P.R. China. and University of Chinese Academy of Sciences, Beijing 100049, P.R. China
| | - Jing Gao
- State Key Laboratory of Electroanalytical Chemistry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilin 130022, P.R. China.
| | - Mingjun Cai
- State Key Laboratory of Electroanalytical Chemistry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilin 130022, P.R. China.
| | - Haijiao Xu
- State Key Laboratory of Electroanalytical Chemistry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilin 130022, P.R. China.
| | - Junguang Jiang
- State Key Laboratory of Electroanalytical Chemistry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilin 130022, P.R. China.
| | - Zhiyuan Tian
- School of Chemistry and Chemical Engineering, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Hongda Wang
- State Key Laboratory of Electroanalytical Chemistry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilin 130022, P.R. China.
| |
Collapse
|
157
|
Friedel M, André S, Goldschmidt H, Gabius HJ, Schwartz-Albiez R. Galectin-8 enhances adhesion of multiple myeloma cells to vascular endothelium and is an adverse prognostic factor. Glycobiology 2016; 26:1048-1058. [PMID: 27287437 DOI: 10.1093/glycob/cww066] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2016] [Revised: 05/24/2016] [Accepted: 05/29/2016] [Indexed: 11/13/2022] Open
Abstract
Multiple myeloma is characterized by abnormal infiltration of malignant plasma cells into bone marrow. Testing the hypothesis that bivalent galectin-8 (Gal-8) may influence homing of myeloma cells to vascular endothelium as a key prerequisite for infiltration, we analyzed the two Gal-8 splice variants (Gal-8S, Gal-8L). They differ in the length of their linker peptide connecting the two lectin domains. Both Gal-8 isoforms bind to cells of the myeloma lines Gal-8+ MOLP-8 and Gal-8- LP-1 in a glycan-inhibitable manner. Both Gal-8 isoforms led to enhanced adhesion of myeloma cells to vascular endothelium under dynamic shear stress conditions, Gal-8L (by more than 40-fold) even stronger than Gal-8S. Additional treatment of endothelial cells with tumour necrosis factor prior to the dynamic shear stress assay entailed an almost 100-fold enhanced adhesion of myeloma cells without addition of Gal-8 variants and a further 1.5-1.7-fold enhancement by addition of Gal-8 variants. We also found that elevated expression of Gal-8 in native multiple myeloma cells is an adverse prognostic factor for overall and event-free survival using patients' gene expression profile data of the total therapy 2 and 3 myeloma studies. Also, elevated concentrations of Gal-8 were detected (45%, 19/42 patients) in sera of multiple myeloma patients compared to those of healthy, age-matched donors. Both experimental and clinical data strongly point to the significance of Gal-8 for multiple myeloma development.
Collapse
Affiliation(s)
- Myriam Friedel
- Clinical Cooperation Unit Applied Tumor Immunity, Deutsches Krebsforschungszentrum, 69120 Heidelberg, Germany
| | - Sabine André
- Institut für Physiologische Chemie, Tierärztliche Fakultät, Ludwig-Maximilians-Universität München, 80539 München, Germany
| | - Hartmut Goldschmidt
- Medizinische Klinik V, Universitätsklinikum Heidelberg und Nationales Centrum für Tumorerkrankungen Heidelberg, 69120 Heidelberg, Germany
| | - Hans-Joachim Gabius
- Institut für Physiologische Chemie, Tierärztliche Fakultät, Ludwig-Maximilians-Universität München, 80539 München, Germany
| | - Reinhard Schwartz-Albiez
- Clinical Cooperation Unit Applied Tumor Immunity, Deutsches Krebsforschungszentrum, 69120 Heidelberg, Germany
| |
Collapse
|
158
|
Takeuchi T, Arata Y, Kasai KI. Galactoseβ1-4fucose: A unique disaccharide unit found inN-glycans of invertebrates including nematodes. Proteomics 2016; 16:3137-3147. [DOI: 10.1002/pmic.201600001] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2016] [Revised: 03/22/2016] [Accepted: 04/12/2016] [Indexed: 11/06/2022]
Affiliation(s)
| | - Yoichiro Arata
- Faculty of Pharmaceutical Sciences; Josai University; Saitama Japan
| | - Ken-ichi Kasai
- School of Pharmaceutical Sciences; Teikyo University; Tokyo Japan
| |
Collapse
|
159
|
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: 6.1] [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
|
160
|
Enzymes for N-Glycan Branching and Their Genetic and Nongenetic Regulation in Cancer. Biomolecules 2016; 6:biom6020025. [PMID: 27136596 PMCID: PMC4919920 DOI: 10.3390/biom6020025] [Citation(s) in RCA: 110] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2016] [Revised: 04/15/2016] [Accepted: 04/21/2016] [Indexed: 02/07/2023] Open
Abstract
N-glycan, a fundamental and versatile protein modification in mammals, plays critical roles in various physiological and pathological events including cancer progression. The formation of N-glycan branches catalyzed by specific N-acetylglucosaminyltransferases [GnT-III, GnT-IVs, GnT-V, GnT-IX (Vb)] and a fucosyltransferase, Fut8, provides functionally diverse N-glycosylated proteins. Aberrations of these branches are often found in cancer cells and are profoundly involved in cancer growth, invasion and metastasis. In this review, we focus on the GlcNAc and fucose branches of N-glycans and describe how their expression is dysregulated in cancer by genetic and nongenetic mechanisms including epigenetics and nucleotide sugar metabolisms. We also survey the roles that these N-glycans play in cancer progression and therapeutics. Finally, we discuss possible applications of our knowledge on basic glycobiology to the development of medicine and biomarkers for cancer therapy.
Collapse
|
161
|
Tamura M, Sasai A, Ozawa R, Saito M, Yamamoto K, Takeuchi T, Ohtake K, Tateno H, Hirabayashi J, Kobayashi J, Arata Y. Identification of the cysteine residue responsible for oxidative inactivation of mouse galectin-2. J Biochem 2016; 160:233-241. [PMID: 27122052 DOI: 10.1093/jb/mvw029] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2016] [Accepted: 03/23/2016] [Indexed: 11/15/2022] Open
Abstract
Galectins are a group of animal lectins characterized by their specificity for β-galactosides. Mouse galectin-2 (mGal-2) is predominantly expressed in the gastrointestinal tract and has been identified as one of the main gastric mucosal proteins that are uniquely sensitive to S-nitrosylation. We have previously reported that oxidation of mGal-2 by hydrogen peroxide (H2O2) resulted in the loss of sugar-binding ability, whereas pre-treatment of mGal-2 with S-nitrosocysteine prevented H2O2-induced inactivation. In this study, we used point-mutated recombinant mGal-2 proteins to study which of the two highly conserved Cys residues in mGal-2 must be S-nitrosylated for protection against oxidative inactivation. Mutation of Cys57 to a Met residue (C57M) did not result in lectin inactivation following H2O2 treatment, whereas Cys75 mutation to Ser (C75S) led to significantly reduced lectin activity, as is the case for wild-type mGal-2. However, pre-treatment of the C75S mutant with S-nitrosocysteine protected the protein from H2O2-induced inactivation. Therefore, Cys57 is suggested to be responsible for oxidative inactivation of the mGal-2 protein, and protection of the sulfhydryl group of the Cys57 in mGal-2 by S-nitrosylation is likely important for maintaining mGal-2 protein function in an oxidative environment such as the gastrointestinal tract.
Collapse
Affiliation(s)
- Mayumi Tamura
- Department of Biochemistry, Faculty of Pharmaceutical Sciences, Josai University, Saitama 350-0295, Japan
| | - Akari Sasai
- Department of Biochemistry, Faculty of Pharmaceutical Sciences, Josai University, Saitama 350-0295, Japan
| | - Rika Ozawa
- Department of Biochemistry, Faculty of Pharmaceutical Sciences, Josai University, Saitama 350-0295, Japan
| | - Masanori Saito
- Department of Biochemistry, Faculty of Pharmaceutical Sciences, Josai University, Saitama 350-0295, Japan
| | - Kaori Yamamoto
- Department of Biochemistry, Faculty of Pharmaceutical Sciences, Josai University, Saitama 350-0295, Japan
| | - Tomoharu Takeuchi
- Department of Biochemistry, Faculty of Pharmaceutical Sciences, Josai University, Saitama 350-0295, Japan
| | - Kazuo Ohtake
- Division of Pathophysiology, Department of Clinical Dietetics and Human Nutrition, Faculty of Pharmaceutical Sciences, Josai University, Saitama 350-0295, Japan
| | - Hiroaki Tateno
- Biotechnology Research Institute for Drug Discovery, National Institute of Advanced Industrial Science and Technology, Ibaraki 305-8568, Japan
| | - Jun Hirabayashi
- Biotechnology Research Institute for Drug Discovery, National Institute of Advanced Industrial Science and Technology, Ibaraki 305-8568, Japan
| | - Jun Kobayashi
- Division of Pathophysiology, Department of Clinical Dietetics and Human Nutrition, Faculty of Pharmaceutical Sciences, Josai University, Saitama 350-0295, Japan
| | - Yoichiro Arata
- Department of Biochemistry, Faculty of Pharmaceutical Sciences, Josai University, Saitama 350-0295, Japan
| |
Collapse
|
162
|
Stegmayr J, Lepur A, Kahl-Knutson B, Aguilar-Moncayo M, Klyosov AA, Field RA, Oredsson S, Nilsson UJ, Leffler H. Low or No Inhibitory Potency of the Canonical Galectin Carbohydrate-binding Site by Pectins and Galactomannans. J Biol Chem 2016; 291:13318-34. [PMID: 27129206 PMCID: PMC4933242 DOI: 10.1074/jbc.m116.721464] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2016] [Indexed: 12/17/2022] Open
Abstract
Some complex plant-derived polysaccharides, such as modified citrus pectins and galactomannans, have been shown to have promising anti-inflammatory and anti-cancer effects. Most reports propose or claim that these effects are due to interaction of the polysaccharides with galectins because the polysaccharides contain galactose-containing side chains that might bind this class of lectin. However, their direct binding to and/or inhibition of the evolutionarily conserved galactoside-binding site of galectins has not been demonstrated. Using a well established fluorescence anisotropy assay, we tested the direct interaction of several such polysaccharides with physiological concentrations of a panel of galectins. The bioactive pectic samples tested were very poor inhibitors of the canonical galactoside-binding site for the tested galectins, with IC50 values >10 mg/ml for a few or in most cases no inhibitory activity at all. The galactomannan Davanat® was more active, albeit not a strong inhibitor (IC50 values ranging from 3 to 20 mg/ml depending on the galectin). Pure synthetic oligosaccharide fragments found in the side chains and backbone of pectins and galactomannans were additionally tested. The most commonly found galactan configuration in pectins had no inhibition of the galectins tested. Galactosylated tri- and pentamannosides, representing the structure of Davanat®, had an inhibitory effect of galectins comparable with that of free galactose. Further evaluation using cell-based assays, indirectly linked to galectin-3 inhibition, showed no inhibition of galectin-3 by the polysaccharides. These data suggest that the physiological effects of these plant polysaccharides are not due to inhibition of the canonical galectin carbohydrate-binding site.
Collapse
Affiliation(s)
- John Stegmayr
- From the Section MIG (Microbiology, Immunology, Glycobiology), Department of Laboratory Medicine, Lund University, 221 00 Lund, Sweden, the Department of Biology and
| | - Adriana Lepur
- From the Section MIG (Microbiology, Immunology, Glycobiology), Department of Laboratory Medicine, Lund University, 221 00 Lund, Sweden
| | - Barbro Kahl-Knutson
- From the Section MIG (Microbiology, Immunology, Glycobiology), Department of Laboratory Medicine, Lund University, 221 00 Lund, Sweden
| | - Matilde Aguilar-Moncayo
- the Department of Biological Chemistry, John Innes Centre, Norwich Research Park, NR4 7UH Norwich, United Kingdom, and
| | | | - Robert A Field
- the Department of Biological Chemistry, John Innes Centre, Norwich Research Park, NR4 7UH Norwich, United Kingdom, and
| | | | - Ulf J Nilsson
- Centre for Analysis and Synthesis, Department of Chemistry, Lund University, Lund, Sweden
| | - Hakon Leffler
- From the Section MIG (Microbiology, Immunology, Glycobiology), Department of Laboratory Medicine, Lund University, 221 00 Lund, Sweden,
| |
Collapse
|
163
|
Yang RY, Xue H, Yu L, Velayos-Baeza A, Monaco AP, Liu FT. Identification of VPS13C as a Galectin-12-Binding Protein That Regulates Galectin-12 Protein Stability and Adipogenesis. PLoS One 2016; 11:e0153534. [PMID: 27073999 PMCID: PMC4830523 DOI: 10.1371/journal.pone.0153534] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2016] [Accepted: 03/30/2016] [Indexed: 01/13/2023] Open
Abstract
Galectin-12, a member of the galectin family of β-galactoside-binding animal lectins, is preferentially expressed in adipocytes and required for adipocyte differentiation in vitro. This protein was recently found to regulate lipolysis, whole body adiposity, and glucose homeostasis in vivo. Here we identify VPS13C, a member of the VPS13 family of vacuolar protein sorting-associated proteins highly conserved throughout eukaryotic evolution, as a major galectin-12-binding protein. VPS13C is upregulated during adipocyte differentiation, and is required for galectin-12 protein stability. Knockdown of Vps13c markedly reduces the steady-state levels of galectin-12 by promoting its degradation through primarily the lysosomal pathway, and impairs adipocyte differentiation. Our studies also suggest that VPS13C may have a broader role in protein quality control. The regulation of galectin-12 stability by VPS13C could potentially be exploited for therapeutic intervention of obesity and related metabolic diseases.
Collapse
Affiliation(s)
- Ri-Yao Yang
- Department of Dermatology, School of Medicine, University of California-Davis, Sacramento, California, 95817, United States of America
| | - Huiting Xue
- Department of Dermatology, School of Medicine, University of California-Davis, Sacramento, California, 95817, United States of America
- School of Life Sciences, Northeast Normal University, Changchun, 130024, People’s Republic of China
| | - Lan Yu
- Department of Dermatology, School of Medicine, University of California-Davis, Sacramento, California, 95817, United States of America
| | | | - Anthony P. Monaco
- Wellcome Trust Centre for Human Genetics, OX3 7BN, Oxford, United Kingdom
| | - Fu-Tong Liu
- Department of Dermatology, School of Medicine, University of California-Davis, Sacramento, California, 95817, United States of America
- Institute of Biomedical Sciences, Academia Sinica, Nankang, Taipei, 115, Taiwan
| |
Collapse
|
164
|
Stanley P. What Have We Learned from Glycosyltransferase Knockouts in Mice? J Mol Biol 2016; 428:3166-3182. [PMID: 27040397 DOI: 10.1016/j.jmb.2016.03.025] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2016] [Revised: 03/22/2016] [Accepted: 03/23/2016] [Indexed: 11/16/2022]
Abstract
There are five major classes of glycan including N- and O-glycans, glycosaminoglycans, glycosphingolipids, and glycophosphatidylinositol anchors, all expressed at the molecular frontier of each mammalian cell. Numerous biological consequences of altering the expression of mammalian glycans are understood at a mechanistic level, but many more remain to be characterized. Mouse mutants with deleted, defective, or misexpressed genes that encode activities necessary for glycosylation have led the way to identifying key functions of glycans in biology. However, with the advent of exome sequencing, humans with mutations in genes involved in glycosylation are also revealing specific requirements for glycans in mammalian development. The aim of this review is to summarize glycosylation genes that are necessary for mouse embryonic development, pathway-specific glycosylation genes whose deletion leads to postnatal morbidity, and glycosylation genes for which effects are mild, but perturbation of the organism may reveal functional consequences. General strategies for generating and interpreting the phenotype of mice with glycosylation defects are discussed in relation to human congenital disorders of glycosylation (CDG).
Collapse
Affiliation(s)
- Pamela Stanley
- Department of Cell Biology, Albert Einstein College of Medicine, New York, NY 10461, USA.
| |
Collapse
|
165
|
Bapu D, Runions J, Kadhim M, Brooks SA. N-acetylgalactosamine glycans function in cancer cell adhesion to endothelial cells: A role for truncated O-glycans in metastatic mechanisms. Cancer Lett 2016; 375:367-374. [PMID: 26994652 DOI: 10.1016/j.canlet.2016.03.019] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2016] [Revised: 03/10/2016] [Accepted: 03/10/2016] [Indexed: 01/09/2023]
Abstract
Failure in O-glycan chain extension exposing Tn antigen (GalNAc-O-Ser/Thr) is clinically associated with cancer metastasis. This study provides evidence of a functional role for aberrant GalNAc-glycans in cancer cell capture from blood flow and/or adhesion to endothelium. Adhesion of breast cancer cells to human umbilical vein endothelial cell monolayers was modelled under sweeping flow. Adhesion of metastatic, GalNAc glycan-rich, MCF7 and ZR 75 1 cells to endothelium increased over timepoints up to 1.5 hour, after which it plateaued. Adhesion was significantly inhibited (p < 0.001) when cell surface GalNAc-glycans were masked, an effect not seen in GalNAc glycan-poor, non-metastatic BT 474 cells. Masking irrelevant galactose- and mannose-glycans had no inhibitory effect. Imaging of cells post-adhesion over a 24 hour time course using confocal and scanning electron microscopy revealed that up to 6 hours post-adhesion, motile, rounded cancer cells featuring lamellipodia-like processes crawled on an intact endothelial monolayer. From 6-12 hours post-adhesion, cancer cells became stationary, adopted a smooth, circular flattened morphology, and endothelial cells retracted from around them leaving cleared zones in which the cancer cells proceeded to form colonies through cell division.
Collapse
Affiliation(s)
- Deepashree Bapu
- Department of Biological & Medical Sciences, Faculty of Health & Life Sciences, Oxford Brookes University, Gipsy Lane, Headington, Oxford OX3 0BP, UK
| | - John Runions
- Department of Biological & Medical Sciences, Faculty of Health & Life Sciences, Oxford Brookes University, Gipsy Lane, Headington, Oxford OX3 0BP, UK
| | - Munira Kadhim
- Department of Biological & Medical Sciences, Faculty of Health & Life Sciences, Oxford Brookes University, Gipsy Lane, Headington, Oxford OX3 0BP, UK
| | - Susan Ann Brooks
- Department of Biological & Medical Sciences, Faculty of Health & Life Sciences, Oxford Brookes University, Gipsy Lane, Headington, Oxford OX3 0BP, UK.
| |
Collapse
|
166
|
Nagae M, Kanagawa M, Morita-Matsumoto K, Hanashima S, Kizuka Y, Taniguchi N, Yamaguchi Y. Atomic visualization of a flipped-back conformation of bisected glycans bound to specific lectins. Sci Rep 2016; 6:22973. [PMID: 26971576 PMCID: PMC4789653 DOI: 10.1038/srep22973] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2015] [Accepted: 02/23/2016] [Indexed: 11/09/2022] Open
Abstract
Glycans normally exist as a dynamic equilibrium of several conformations. A fundamental question concerns how such molecules bind lectins despite disadvantageous entropic loss upon binding. Bisected glycan, a glycan possessing bisecting N-acetylglucosamine (GlcNAc), is potentially a good model for investigating conformational dynamics and glycan-lectin interactions, owing to the unique ability of this sugar residue to alter conformer populations and thus modulate the biological activities. Here we analyzed bisected glycan in complex with two unrelated lectins, Calsepa and PHA-E. The crystal structures of the two complexes show a conspicuous flipped back glycan structure (designated 'back-fold' conformation), and solution NMR analysis also provides evidence of 'back-fold' glycan structure. Indeed, statistical conformational analysis of available bisected and non-bisected glycan structures suggests that bisecting GlcNAc restricts the conformations of branched structures. Restriction of glycan flexibility by certain sugar residues may be more common than previously thought and impinges on the mechanism of glycoform-dependent biological functions.
Collapse
Affiliation(s)
- Masamichi Nagae
- Structural Glycobiology Team, 2-1 Hirosawa, Wako, Saitama 351-0198, Japan
| | - Mayumi Kanagawa
- Structural Glycobiology Team, 2-1 Hirosawa, Wako, Saitama 351-0198, Japan
| | | | - Shinya Hanashima
- Department of Chemistry, Osaka University, Machikaneyama, Toyonaka, Osaka 560-0043, Japan
| | - Yasuhiko Kizuka
- Disease Glycomics Team, Systems Glycobiology Research Group, RIKEN-Max Planck Joint Research Center, RIKEN Global Research Cluster, 2-1 Hirosawa, Wako, Saitama 351-0198, Japan
| | - Naoyuki Taniguchi
- Disease Glycomics Team, Systems Glycobiology Research Group, RIKEN-Max Planck Joint Research Center, RIKEN Global Research Cluster, 2-1 Hirosawa, Wako, Saitama 351-0198, Japan
| | - Yoshiki Yamaguchi
- Structural Glycobiology Team, 2-1 Hirosawa, Wako, Saitama 351-0198, Japan
| |
Collapse
|
167
|
β-Galactoside-mediated tissue organization during islet reconstitution. Regen Ther 2016; 3:11-14. [PMID: 31245467 PMCID: PMC6581812 DOI: 10.1016/j.reth.2016.01.006] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2015] [Revised: 01/05/2016] [Accepted: 01/20/2016] [Indexed: 01/15/2023] Open
Abstract
We have previously reported that multi-cellular heteroaggregates comprising murine pancreatic α (αTC1.6) and β (MIN6-m9) cell lines spontaneously acquired islet-like architecture and displayed higher insulin secretion rates. However, the mechanisms of self-organization remain unclear. The objective of this study is to examine the possibility that a sugar chain participates in the mutual recognition of the cells during reconstitution of the islet-like structure in vitro. Using a lectin-binding assay, we identified Erythrina cristagalli agglutinin (ECA), which particularly recognizes the β-galactoside structure on the surfaces of MIN6-m9 cells. The self-organization of αTC1.6 and MIN6-m9 was obstructed using ECA-bound MIN6-m9 cells. Lactose neutralized the ECA's inhibitory effect on the autonomous rearrangement of αTC1.6 and MIN6-m9 cells, indicating that the inhibition of cell arrangement by ECA was mediated via β-galactoside. We concluded that a β-galactoside sugar chain was central to the reconstitution of the pancreatic islet-like architecture in vitro.
Collapse
Key Words
- ConA, concanavalin A
- DMEM, Dulbecco's Modified Eagle's Medium
- ECA, Erythrina cristagalli agglutinin
- FITC, fluorescein isothiocyanate
- Islet reconstitution
- Islet-like structure
- LCA, Lens culinaris agglutinin, α-d-mannosyl group
- Lectin
- MAA, Maackia amurensis agglutinin
- MC, methylcellulose
- RCA, Ricinus communis agglutinin
- SSA, Sambucus sieboldiana agglutinin
- Sugar chain
- UEA, Ulex europaeus agglutinin
- WGA, wheat germ agglutinin
- β-Galactoside
Collapse
|
168
|
Kakizaki M, Togayachi A, Narimatsu H, Watanabe R. Contribution of Lewis X Carbohydrate Structure to Neuropathogenic Murine Coronaviral Spread. Jpn J Infect Dis 2016; 69:405-13. [PMID: 26902214 DOI: 10.7883/yoken.jjid.2015.499] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Although Lewis X (Le(x)), a carbohydrate structure, is involved in innate immunity through cell-to-cell and pathogen recognition, its expression has not been observed in mouse monocytes/macrophages (Mo/Mas). The Mo/Mas that infiltrate the meninges after infection with the neuropathogenic murine coronavirus strain srr7 are an initial target of infection. Furthermore, higher inflammatory responses were observed in gene-manipulated mice lacking α1,3-fucosyltransferase 9, which determines the expression of the Le(x) structure, than in wild type mice after infection. We investigated Le(x) expression using CD11b-positive peritoneal exudate cells (PECs) and found that Le(x) is inducible in Mo/Mas after infection with srr7, especially in the syncytial cells during the late phase of infection. The number of syncytial cells was reduced after treatment of the infected PECs with anti-Le(x) antibody, during the late phase of infection. In addition, the antibody treatment induced a marked reduction in the number of the infected cells at 24 hours post inoculation, without changing the infected cell numbers during the initial phase of infection. These data indicate that the Le(x) structure could play a role in syncytial formation and cell-to-cell infection during the late phase of infection.
Collapse
Affiliation(s)
- Masatoshi Kakizaki
- Department of Bioinformatics, Graduate School of Engineering, Soka University
| | | | | | | |
Collapse
|
169
|
Hockl PF, Wolosiuk A, Pérez-Sáez JM, Bordoni AV, Croci DO, Toum-Terrones Y, Soler-Illia GJAA, Rabinovich GA. Glyco-nano-oncology: Novel therapeutic opportunities by combining small and sweet. Pharmacol Res 2016; 109:45-54. [PMID: 26855319 DOI: 10.1016/j.phrs.2016.02.005] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/12/2016] [Revised: 02/02/2016] [Accepted: 02/02/2016] [Indexed: 12/28/2022]
Abstract
Recent efforts toward defining the molecular features of the tumor microenvironment have revealed dramatic changes in the expression of glycan-related genes including glycosyltransferases and glycosidases. These changes affect glycosylation of proteins and lipids not only in cancer cells themselves, but also in cancer associated-stromal, endothelial and immune cells. These glycan alterations including increased frequency of β1,6-branched N-glycans and bisecting N-glycans, overexpression of tumor-associated mucins, preferred expression of T, Tn and sialyl-Tn antigen and altered surface sialylation, may contribute to tumor progression by masking or unmasking specific ligands for endogenous lectins, including members of the C-type lectin, siglec and galectin families. Differential expression of glycans or glycan-binding proteins could be capitalized for the identification of novel biomarkers and might provide novel opportunities for therapeutic intervention. This review focuses on the biological relevance of lectin-glycan interactions in the tumor microenvironment (mainly illustrated by the immunosuppressive and pro-angiogenic activities of galectin-1) and the design of functionalized nanoparticles for pharmacological delivery of multimeric glycans, lectins or selective inhibitors of lectin-glycan interactions with antitumor activity.
Collapse
Affiliation(s)
- Pablo F Hockl
- Laboratorio de Inmunopatología, Instituto de Biología y Medicina Experimental (IBYME), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Vuelta de Obligado 2490, C1428ADN Buenos Aires, Argentina
| | - Alejandro Wolosiuk
- Gerencia Química, Centro Atómico Constituyentes (CAC), Comisión Nacional de Energía Atómica (CNEA), Avenida General Paz 1499, 1650 San Martín, Argentina
| | - Juan M Pérez-Sáez
- Laboratorio de Inmunopatología, Instituto de Biología y Medicina Experimental (IBYME), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Vuelta de Obligado 2490, C1428ADN Buenos Aires, Argentina
| | - Andrea V Bordoni
- Gerencia Química, Centro Atómico Constituyentes (CAC), Comisión Nacional de Energía Atómica (CNEA), Avenida General Paz 1499, 1650 San Martín, Argentina
| | - Diego O Croci
- Laboratorio de Inmunopatología, Instituto de Biología y Medicina Experimental (IBYME), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Vuelta de Obligado 2490, C1428ADN Buenos Aires, Argentina; Instituto de Histología y Embriología de Mendoza (IHEM), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Casilla de correo 56, 5500 Mendoza, Argentina
| | - Yamili Toum-Terrones
- Gerencia Química, Centro Atómico Constituyentes (CAC), Comisión Nacional de Energía Atómica (CNEA), Avenida General Paz 1499, 1650 San Martín, Argentina
| | - Galo J A A Soler-Illia
- Instituto de Nanosistemas, Universidad Nacional de General San Martín, Av. 25 de Mayo y Francia, 1650 San Martín, Buenos Aires, Argentina; Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Intendente Güiraldes 2160, C1428EGA Buenos Aires, Argentina.
| | - Gabriel A Rabinovich
- Laboratorio de Inmunopatología, Instituto de Biología y Medicina Experimental (IBYME), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Vuelta de Obligado 2490, C1428ADN Buenos Aires, Argentina; Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Intendente Güiraldes 2160, C1428EGA Buenos Aires, Argentina.
| |
Collapse
|
170
|
|
171
|
Nio-Kobayashi J, Hashiba K, Sano M, Okuda K, Duncan WC, Iwanaga T. Expression Profiles and Possible Roles of Galectins in the Corpus Luteum. TRENDS GLYCOSCI GLYC 2016. [DOI: 10.4052/tigg.1416.1j] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Affiliation(s)
- Junko Nio-Kobayashi
- Laboratory of Histology and Cytology, Hokkaido University Graduate School of Medicine
| | - Kazuhisa Hashiba
- Laboratory of Reproductive Physiology, Graduate School of Environmental and Life Science, Okayama University
| | - Masahiro Sano
- Laboratory of Reproductive Physiology, Graduate School of Environmental and Life Science, Okayama University
| | - Kiyoshi Okuda
- Laboratory of Reproductive Physiology, Graduate School of Environmental and Life Science, Okayama University
| | - W. Colin Duncan
- MRC Centre for Reproductive Health, The Queenʼs Medical Research Institute, The University of Edinburgh
| | - Toshihiko Iwanaga
- Laboratory of Histology and Cytology, Hokkaido University Graduate School of Medicine
| |
Collapse
|
172
|
Ferreira JA, Peixoto A, Neves M, Gaiteiro C, Reis CA, Assaraf YG, Santos LL. Mechanisms of cisplatin resistance and targeting of cancer stem cells: Adding glycosylation to the equation. Drug Resist Updat 2016; 24:34-54. [DOI: 10.1016/j.drup.2015.11.003] [Citation(s) in RCA: 117] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2015] [Revised: 11/09/2015] [Accepted: 11/18/2015] [Indexed: 02/06/2023]
|
173
|
Takeuchi T, Nagasaka M, Shimizu M, Tamura M, Arata Y. N-acetylglucosamine suppresses osteoclastogenesis in part through the promotion of O-GlcNAcylation. Bone Rep 2016; 5:15-21. [PMID: 28326343 PMCID: PMC4926832 DOI: 10.1016/j.bonr.2016.02.001] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/07/2016] [Accepted: 02/01/2016] [Indexed: 01/28/2023] Open
Abstract
Osteoclasts are the only cells in an organism capable of resorbing bone. These cells differentiate from monocyte/macrophage lineage cells upon stimulation by receptor activator of NF-κB ligand (RANKL). On the other hand, osteoclastogenesis is reportedly suppressed by glucose via the downregulation of NF-κB activity through suppression of reactive oxygen species generation. To examine whether other sugars might also affect osteoclast development, we compared the effects of monomeric sugars (glucose, galactose, N-acetylglucosamine (GlcNAc), and N-acetylgalactosamine (GalNAc)) on the osteoclastogenesis of murine RAW264 cells. Our results demonstrated that, in addition to glucose, both GlcNAc and GalNAc, which each have little effect on the generation of reactive oxygen species, suppress osteoclastogenesis. We hypothesized that GlcNAc might affect osteoclastogenesis through the upregulation of O-GlcNAcylation and showed that GlcNAc increases global O-GlcNAcylation, thereby suppressing the RANKL-dependent phosphorylation of NF-κB p65. Furthermore, an inhibitor of N-acetyl-β-D-glucosaminidase, O-(2-acetamido-2-deoxy-D-glucopyranosylidene) amino N-phenylcarbamate (PUGNAc), which also increases O-GlcNAcylation, suppressed the osteoclastogenesis of RAW264 cells and that of human peripheral blood mononuclear cells. Together, these data suggest that GlcNAc suppresses osteoclast differentiation in part through the promotion of O-GlcNAcylation. Along with glucose, the monomeric sugars GlcNAc and GalNAc suppress osteoclastic differentiation. Unlike glucose, GlcNAc and GalNAc have little effect on RANKL-induced ROS production. GlcNAc and the N-acetyl-β-D-glucosaminidase inhibitor PUGNAc both increase O-GlcNAcylation and suppress osteoclastogenesis. Upregulation of O-GlcNAcylation suppresses the RANKL-dependent phosphorylation of NF-κB p65. Together, these results suggest that GlcNAc suppresses osteoclastogenesis in part through the promotion of O-GlcNAcylation.
Collapse
Key Words
- Gal, galactose
- GalNAc, N-acetylgalactosamine
- Glc, glucose
- GlcNAc
- GlcNAc, N-acetylglucosamine
- M-CSF, macrophage colony-stimulating factor
- N-acetylglucosamine
- NF-κB
- NF-κB, nuclear factor-κB
- O-GlcNAcylation
- Osteoclast
- PBMC, peripheral blood mononuclear cell
- PUGNAc, O-(2-acetamido-2-deoxy-D-glucopyranosylidene) amino N-phenylcarbamate
- RANKL, receptor activator of nuclear factor-κB ligand
- ROS, reactive oxygen species
- TRAP, tartrate-resistant acid phosphatase
- UDP, uridine diphosphate
- sRANKL, soluble receptor activator of nuclear factor-κB ligand
Collapse
Affiliation(s)
- Tomoharu Takeuchi
- Corresponding author at: Laboratory of Biochemistry, Faculty of Pharmaceutical Sciences, Josai University, 1-1 Keyakidai, Sakado, Saitama 350-0295, Japan.Laboratory of BiochemistryFaculty of Pharmaceutical SciencesJosai University1-1 KeyakidaiSakadoSaitama350-0295Japan
| | | | | | | | | |
Collapse
|
174
|
Analyzing Protein Clusters on the Plasma Membrane: Application of Spatial Statistical Analysis Methods on Super-Resolution Microscopy Images. FOCUS ON BIO-IMAGE INFORMATICS 2016; 219:95-122. [DOI: 10.1007/978-3-319-28549-8_4] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
175
|
Nio-Kobayashi J, Hashiba K, Sano M, Okuda K, Duncan WC, Iwanaga T. Expression Profiles and Possible Roles of Galectins in the Corpus Luteum. TRENDS GLYCOSCI GLYC 2016. [DOI: 10.4052/tigg.1416.1e] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Affiliation(s)
- Junko Nio-Kobayashi
- Laboratory of Histology and Cytology, Hokkaido University Graduate School of Medicine
| | - Kazuhisa Hashiba
- Laboratory of Reproductive Physiology, Graduate School of Environmental and Life Science, Okayama University
| | - Masahiro Sano
- Laboratory of Reproductive Physiology, Graduate School of Environmental and Life Science, Okayama University
| | - Kiyoshi Okuda
- Laboratory of Reproductive Physiology, Graduate School of Environmental and Life Science, Okayama University
| | - W. Colin Duncan
- MRC Centre for Reproductive Health, The Queenʼs Medical Research Institute, The University of Edinburgh
| | - Toshihiko Iwanaga
- Laboratory of Histology and Cytology, Hokkaido University Graduate School of Medicine
| |
Collapse
|
176
|
Kobayakawa Y, Sakumi K, Kajitani K, Kadoya T, Horie H, Kira JI, Nakabeppu Y. Galectin-1 deficiency improves axonal swelling of motor neurones in SOD1(G93A) transgenic mice. Neuropathol Appl Neurobiol 2015; 41:227-44. [PMID: 24707896 DOI: 10.1111/nan.12123] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2013] [Accepted: 01/26/2014] [Indexed: 01/05/2023]
Abstract
AIMS Galectin-1, a member of the β-galactoside-binding lectin family, accumulates in neurofilamentous lesions in the spinal cords of both sporadic and familial amyotrophic lateral sclerosis (ALS) patients with a superoxide dismutase 1 gene (SOD1) mutation (A4V). The aim of this study was to evaluate the roles of endogenous galectin-1 in the pathogenesis of ALS. METHODS Expression of galectin-1 in the spinal cord of mutant SOD1 transgenic (SOD1(G93A) ) mice was examined by pathological analysis, real-time RT-PCR and Western blotting. The effects of galectin-1 deficiency were evaluated by cross-breeding SOD1(G93A) mice with galectin-1 null (Lgals1(-/-) ) mice. RESULTS Before ALS-like symptoms developed in SOD1(G93A) /Lgals1(+/+) mice, strong galectin-1 immunoreactivity was observed in swollen motor axons and colocalized with aggregated neurofilaments. Electron microscopic observations revealed that the diameters of swollen motor axons in the spinal cord were significantly smaller in SOD1(G93A) /Lgals1(-/-) mice, and there was less accumulation of vacuoles compared with SOD1(G93A) /Lgals1(+/+) mice. In symptomatic SOD1(G93A) /Lgals1(+/+) mice, astrocytes surrounding motor axons expressed a high level of galectin-1. CONCLUSIONS Galectin-1 accumulates in neurofilamentous lesions in SOD1(G93A) mice, as previously reported in humans with ALS. Galectin-1 accumulation in motor axons occurs before the development of ALS-like symptoms and is associated with early processes of axonal degeneration in SOD1(G93A) mice. In contrast, galectin-1 expressed in astrocytes may be involved in axonal degeneration during symptom presentation.
Collapse
Affiliation(s)
- Yuko Kobayakawa
- Division of Neurofunctional Genomics, Department of Immunobiology and Neuroscience, Medical Institute of Bioregulation, Kyushu University, Fukuoka, Japan; Department of Neurology, Neurological Institute, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | | | | | | | | | | | | |
Collapse
|
177
|
Karakostis K, Costa C, Zito F, Matranga V. Heterologous expression of newly identified galectin-8 from sea urchin embryos produces recombinant protein with lactose binding specificity and anti-adhesive activity. Sci Rep 2015; 5:17665. [PMID: 26640155 PMCID: PMC4671058 DOI: 10.1038/srep17665] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2015] [Accepted: 11/03/2015] [Indexed: 12/11/2022] Open
Abstract
Galectin family members specifically bind beta-galactoside derivatives and are involved in different cellular events, including cell communication, signalling, apoptosis, and immune responses. Here, we report a tandem-repeat type galectin from the Paracentrotus lividus sea urchin embryo, referred to as Pl-GAL-8. The 933nt sequence encodes a protein of 34.73 kDa, containing the conserved HFNPRF and WGxExR motifs in the two highly similar carbohydrate-recognition domains (CRD). The three-dimensional protein structure model of the N-CRD confirms the high evolutionary conservation of carbohydrate binding sites. The temporal gene expression is regulated during development and transcripts localize at the tip of the archenteron at gastrula stage, in a subset of the secondary mesenchyme cells that differentiate into blastocoelar (immune) cells. Functional studies using a recombinant Pl-GAL-8 expressed in bacteria demonstrate its hemo-agglutinating activity on human red blood cells through the binding to lactose, as well as its ability in inhibiting the adhesion of human Hep-G2 cells to the substrate. The recent implications in autoimmune diseases and inflammatory disorders make Gal-8 an attractive candidate for therapeutic purposes. Our results offer a solid basis for addressing the use of the new Pl-GAL-8 in functional and applicative studies, respectively in the developmental and biomedical fields.
Collapse
Affiliation(s)
- Kostantinos Karakostis
- Consiglio Nazionale delle Ricerche, Istituto di Biomedicina e Immunologia Molecolare “A. Monroy”, Via Ugo La Malfa 153, 90146 Palermo, Italy
| | - Caterina Costa
- Consiglio Nazionale delle Ricerche, Istituto di Biomedicina e Immunologia Molecolare “A. Monroy”, Via Ugo La Malfa 153, 90146 Palermo, Italy
| | - Francesca Zito
- Consiglio Nazionale delle Ricerche, Istituto di Biomedicina e Immunologia Molecolare “A. Monroy”, Via Ugo La Malfa 153, 90146 Palermo, Italy
| | - Valeria Matranga
- Consiglio Nazionale delle Ricerche, Istituto di Biomedicina e Immunologia Molecolare “A. Monroy”, Via Ugo La Malfa 153, 90146 Palermo, Italy
| |
Collapse
|
178
|
Wang X, Zhang S, Lin F, Chu W, Yue S. Elevated Galectin-3 Levels in the Serum of Patients With Alzheimer's Disease. Am J Alzheimers Dis Other Demen 2015; 30:729-32. [PMID: 23823143 PMCID: PMC10852776 DOI: 10.1177/1533317513495107] [Citation(s) in RCA: 70] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
Alzheimer's disease (AD) is a neurodegenerative disorder of the central nervous system. Galectin-3 (Gal-3) is characterized by a conserved sequence within the carbohydrate recognition domain. The effect of Gal-3 in AD is presently unknown. In this study, we found significantly increased Gal-3 serum levels in patients with AD compared to control participants (P=.017). There was no significant difference between patients with mild cognitive impairment (MCI) and healthy controls (P=.143) or between patients with AD and MCI (P=.688). The degree of cognitive impairment, as measured by the Mini-Mental Status Examination score, was found to have a significant correlation with the Gal-3 serum levels in all patients and healthy controls. These data suggest that Gal-3 potentially plays a role in the neuropathogenesis of AD. The Gal-3 found in serum could be a potential candidate for a biomarker panel for AD diagnosis.
Collapse
Affiliation(s)
- Xuexin Wang
- Department of Rehabilitation Medicine, Qilu Hospital of Shandong University, Ji'nan, Shandong, People's Republic of China Department of Rehabilitation Medicine, Yuhuangding Hospital, Yantai, Shandong, People's Republic of China
| | - Shuping Zhang
- Department of Clinical Laboratory, Yantai Hospital for Infectious Diseases, Yantai, Shandong, People's Republic of China
| | - Faliang Lin
- Department of Rehabilitation Medicine, Yuhuangding Hospital, Yantai, Shandong, People's Republic of China
| | - Wenzheng Chu
- Department of Neurology, Yuhuangding Hospital, Yantai, Shandong, People's Republic of China
| | - Shouwei Yue
- Department of Rehabilitation Medicine, Qilu Hospital of Shandong University, Ji'nan, Shandong, People's Republic of China
| |
Collapse
|
179
|
González CP, Rodríguez E, Soule S, Fraguas LF, Brena BM, Giacomini C, Irazoqui G. Enzymatic synthesis of 3-aminopropyl-1-O-β-D-galactopyranoside catalyzed byAspergillus oryzaeβ-galactosidase. BIOCATAL BIOTRANSFOR 2015. [DOI: 10.3109/10242422.2015.1095678] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
|
180
|
Entomotoxic and nematotoxic lectins and protease inhibitors from fungal fruiting bodies. Appl Microbiol Biotechnol 2015; 100:91-111. [DOI: 10.1007/s00253-015-7075-2] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2015] [Revised: 10/04/2015] [Accepted: 10/11/2015] [Indexed: 01/26/2023]
|
181
|
Abstract
Spatial organization of the plasma membrane is an essential feature of the cellular response to external stimuli. Receptor organization at the cell surface mediates transmission of extracellular stimuli to intracellular signalling molecules and effectors that impact various cellular processes including cell differentiation, metabolism, growth, migration and apoptosis. Membrane domains include morphologically distinct plasma membrane invaginations such as clathrin-coated pits and caveolae, but also less well-defined domains such as lipid rafts and the galectin lattice. In the present chapter, we will discuss interaction between caveolae, lipid rafts and the galectin lattice in the control of cancer cell signalling.
Collapse
|
182
|
Nagae M, Yamaguchi Y. Sugar recognition and protein-protein interaction of mammalian lectins conferring diverse functions. Curr Opin Struct Biol 2015; 34:108-15. [PMID: 26418728 DOI: 10.1016/j.sbi.2015.08.005] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2015] [Revised: 08/06/2015] [Accepted: 08/10/2015] [Indexed: 11/24/2022]
Abstract
Recent advances in structural analyses of mammalian lectins reveal atomic-level details of their fine specificities toward diverse endogenous and exogenous glycans. Local variations on a common scaffold can enable certain lectins to recognize complex carbohydrate ligands including branched glycans and O-glycosylated peptides. Simultaneous recognition of both glycan and the aglycon moieties enhances the affinity and specificity of lectins such as CLEC-2 and PILRα. Attention has been paid to the roles of galectin and RegIII family of proteins in protein-protein interactions involved in critical biological functions including signal transduction and bactericidal pore formation.
Collapse
Affiliation(s)
- Masamichi Nagae
- Structural Glycobiology Team, Systems Glycobiology Research Group, RIKEN-Max Planck Joint Research Center, RIKEN Global Research Cluster, 2-1 Hirosawa, Wako, Saitama 351-0198, Japan
| | - Yoshiki Yamaguchi
- Structural Glycobiology Team, Systems Glycobiology Research Group, RIKEN-Max Planck Joint Research Center, RIKEN Global Research Cluster, 2-1 Hirosawa, Wako, Saitama 351-0198, Japan.
| |
Collapse
|
183
|
Glycosylation of solute carriers: mechanisms and functional consequences. Pflugers Arch 2015; 468:159-76. [PMID: 26383868 DOI: 10.1007/s00424-015-1730-4] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2015] [Revised: 08/19/2015] [Accepted: 08/21/2015] [Indexed: 12/21/2022]
Abstract
Solute carriers (SLCs) are one of the largest groups of multi-spanning membrane proteins in mammals and include ubiquitously expressed proteins as well as proteins with highly restricted tissue expression. A vast number of studies have addressed the function and organization of SLCs as well as their posttranslational regulation, but only relatively little is known about the role of SLC glycosylation. Glycosylation is one of the most abundant posttranslational modifications of animal proteins and through recent advances in our understanding of protein-glycan interactions, the functional roles of SLC glycosylation are slowly emerging. The purpose of this review is to provide a concise overview of the aspects of glycobiology most relevant to SLCs, to discuss the roles of glycosylation in the regulation and function of SLCs, and to outline the major open questions in this field, which can now be addressed given major technical advances in this and related fields of study in recent years.
Collapse
|
184
|
Zhong Y, Qin Y, Dang L, Jia L, Zhang Z, Wu H, Cui J, Bian H, Li Z. Alteration and localization of glycan-binding proteins in human hepatic stellate cells during liver fibrosis. Proteomics 2015; 15:3283-95. [PMID: 26058380 DOI: 10.1002/pmic.201500030] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2015] [Revised: 04/16/2015] [Accepted: 06/05/2015] [Indexed: 01/10/2023]
Abstract
Glycan-binding proteins (GBPs) play an important role in cell adhesion, bacterial/viral infection, and cellular signaling pathways. However, little is known about the precision alteration of GBPs referred to pathological changes in hepatic stellate cells (HSCs) during liver fibrosis. Here, the carbohydrate microarrays were used to probe the alteration of GBPs in the activated HSCs and quiescent HSCs. As a result, 12 carbohydrates (e.g. Gal, GalNAc, and Man-9Glycan) showed increased signal, while seven carbohydrates (e.g. NeuAc, Lac, and GlcNAc-O-Ser) showed decreased signal in activated HSCs. Three carbohydrates (Gal, GalNAc, and NeuAc) were selected and subsequently used to validate the results of the carbohydrate microarrays as well as assess the distribution and localization of their binding proteins in HSCs and liver tissues by cy/histochemistry; the results showed that GBPs mainly distributed in the cytoplasma membrane and perinuclear region of cytoplasm. The immunocytochemistry was further used to verify some GBPs really exist in Golgi apparatus of the cells. The precision alteration and localization of GBPs referred to pathological changes in HSCs may provide pivotal information to help understand the biological functions of glycans how to exert through their recognition by a wide variety of GBPs. This study could lead to the development of new anti-fibrotic strategies.
Collapse
Affiliation(s)
- Yaogang Zhong
- Laboratory for Functional Glycomics, College of Life Sciences, Northwest University, Xi'an, P. R. China
| | - Yannan Qin
- Laboratory for Functional Glycomics, College of Life Sciences, Northwest University, Xi'an, P. R. China
| | - Liuyi Dang
- Laboratory for Functional Glycomics, College of Life Sciences, Northwest University, Xi'an, P. R. China
| | - Liyuan Jia
- Laboratory for Functional Glycomics, College of Life Sciences, Northwest University, Xi'an, P. R. China
| | - Zhiwei Zhang
- Laboratory for Functional Glycomics, College of Life Sciences, Northwest University, Xi'an, P. R. China
| | - Haoxiang Wu
- Laboratory for Functional Glycomics, College of Life Sciences, Northwest University, Xi'an, P. R. China
| | - Jihong Cui
- Laboratory for Functional Glycomics, College of Life Sciences, Northwest University, Xi'an, P. R. China
| | - Huijie Bian
- Cell Engineering Research Centre and Department of Cell Biology, Fourth Military Medical University, Xi'an, P. R. China
| | - Zheng Li
- Laboratory for Functional Glycomics, College of Life Sciences, Northwest University, Xi'an, P. R. China
| |
Collapse
|
185
|
Immunosuppressive drugs affect high-mannose/hybrid N-glycans on human allostimulated leukocytes. Anal Cell Pathol (Amst) 2015; 2015:324980. [PMID: 26339568 PMCID: PMC4538311 DOI: 10.1155/2015/324980] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2015] [Accepted: 07/21/2015] [Indexed: 02/04/2023] Open
Abstract
N-glycosylation plays an important role in the majority of physiological and pathological processes occurring in the immune system. Alteration of the type and abundance of glycans is an element of lymphocyte differentiation; it is also common in the development of immune-mediated inflammatory diseases. The N-glycosylation process is very sensitive to different environmental agents, among them the pharmacological environment of immunosuppressive drugs. Some results show that high-mannose oligosaccharides have the ability to suppress different stages of the immune response. We evaluated the effects of cyclosporin A (CsA) and rapamycin (Rapa) on high-mannose/hybrid-type glycosylation in human leukocytes activated in a two-way mixed leukocyte reaction (MLR). CsA significantly reduced the number of leukocytes covered by high-mannose/hybrid N-glycans, and the synergistic action of CsA and Rapa led to an increase of these structures on the remaining leukocytes. This is the first study indicating that β1 and β3 integrins bearing high-mannose/hybrid structures are affected by Rapa and CsA. Rapa taken separately and together with CsA changed the expression of β1 and β3 integrins and, by regulating the protein amount, increased the oligomannose/hybrid-type N-glycosylation on the leukocyte surface. We suggest that the changes in the glycosylation profile of leukocytes may promote the development of tolerance in transplantation.
Collapse
|
186
|
Abstract
Despite recent progress in understanding the cancer genome, there is still a relative delay in understanding the full aspects of the glycome and glycoproteome of cancer. Glycobiology has been instrumental in relevant discoveries in various biological and medical fields, and has contributed to the deciphering of several human diseases. Glycans are involved in fundamental molecular and cell biology processes occurring in cancer, such as cell signalling and communication, tumour cell dissociation and invasion, cell-matrix interactions, tumour angiogenesis, immune modulation and metastasis formation. The roles of glycans in cancer have been highlighted by the fact that alterations in glycosylation regulate the development and progression of cancer, serving as important biomarkers and providing a set of specific targets for therapeutic intervention. This Review discusses the role of glycans in fundamental mechanisms controlling cancer development and progression, and their applications in oncology.
Collapse
Affiliation(s)
- Salomé S Pinho
- Instituto de Investigação e Inovação em Saúde (Institute for Research and Innovation in Health), University of Porto, Portugal
- Institute of Molecular Pathology and Immunology of the University of Porto (IPATIMUP), Rua Dr. Roberto Frias s/n, 4200-465 Porto, Portugal
- Institute of Biomedical Sciences Abel Salazar (ICBAS), University of Porto, Rua de Jorge Viterbo Ferreira n.228, 4050-313 Porto, Portugal
| | - Celso A Reis
- Instituto de Investigação e Inovação em Saúde (Institute for Research and Innovation in Health), University of Porto, Portugal
- Institute of Molecular Pathology and Immunology of the University of Porto (IPATIMUP), Rua Dr. Roberto Frias s/n, 4200-465 Porto, Portugal
- Institute of Biomedical Sciences Abel Salazar (ICBAS), University of Porto, Rua de Jorge Viterbo Ferreira n.228, 4050-313 Porto, Portugal
- Faculty of Medicine of the University of Porto, Alameda Prof. Hernâni Monteiro, 4200-319 Porto, Portugal
| |
Collapse
|
187
|
Böcker S, Laaf D, Elling L. Galectin Binding to Neo-Glycoproteins: LacDiNAc Conjugated BSA as Ligand for Human Galectin-3. Biomolecules 2015. [PMID: 26213980 PMCID: PMC4598770 DOI: 10.3390/biom5031671] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Carbohydrate-lectin interactions are relatively weak. As they play an important role in biological recognition processes, multivalent glycan ligands are designed to enhance binding affinity and inhibitory potency. We here report on novel neo-glycoproteins based on bovine serum albumin as scaffold for multivalent presentation of ligands for galectins. We prepared two kinds of tetrasaccharides (N-acetyllactosamine and N,N-diacetyllactosamine terminated) by multi-step chemo-enzymatic synthesis utilizing recombinant glycosyltransferases. Subsequent conjugation of these glycans to lysine groups of bovine serum albumin via squaric acid diethyl ester yielded a set of 22 different neo-glycoproteins with tuned ligand density. The neo-glycoproteins were analyzed by biochemical and chromatographic methods proving various modification degrees. The neo-glycoproteins were used for binding and inhibition studies with human galectin-3 showing high affinity. Binding strength and inhibition potency are closely related to modification density and show binding enhancement by multivalent ligand presentation. At galectin-3 concentrations comparable to serum levels of cancer patients, we detect the highest avidities. Selectivity of N,N-diacetyllactosamine terminated structures towards galectin-3 in comparison to galectin-1 is demonstrated. Moreover, we also see strong inhibitory potency of our scaffolds towards galectin-3 binding. These novel neo-glycoproteins may therefore serve as selective and strong galectin-3 ligands in cancer related biomedical research.
Collapse
Affiliation(s)
- Sophia Böcker
- Laboratory for Biomaterials, Institute for Biotechnology and Helmholtz-Institute for Biomedical Engineering, RWTH Aachen University, Pauwelsstr. 20, 52074 Aachen, Germany.
| | - Dominic Laaf
- Laboratory for Biomaterials, Institute for Biotechnology and Helmholtz-Institute for Biomedical Engineering, RWTH Aachen University, Pauwelsstr. 20, 52074 Aachen, Germany.
| | - Lothar Elling
- Laboratory for Biomaterials, Institute for Biotechnology and Helmholtz-Institute for Biomedical Engineering, RWTH Aachen University, Pauwelsstr. 20, 52074 Aachen, Germany.
| |
Collapse
|
188
|
Sano M, Hashiba K, Nio-Kobayashi J, Okuda K. The luteotrophic function of galectin-1 by binding to the glycans on vascular endothelial growth factor receptor-2 in bovine luteal cells. J Reprod Dev 2015; 61:439-48. [PMID: 26155753 PMCID: PMC4623150 DOI: 10.1262/jrd.2015-056] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
The corpus luteum (CL) is a temporary endocrine gland producing a large amount of progesterone, which is essential for the establishment and maintenance of pregnancy. Galectin-1 is a β-galactose-binding protein that can modify functions of membrane glycoproteins and is expressed in the CL of mice and women. However, the physiological role of galectin-1 in the CL is unclear. In the present study, we investigated the expression and localization of galectin-1 in the bovine CL and the effect of galectin-1 on cultured luteal steroidogenic cells (LSCs) with special reference to its binding to the glycans on vascular endothelial growth factor receptor-2 (VEGFR-2). Galectin-1 protein was highly expressed at the mid and late luteal stages in the membrane fraction of bovine CL tissue and was localized to the surface of LSCs in a carbohydrate-dependent manner. Galectin-1 increased the viability in cultured LSCs. However, the viability of LSCs was decreased by addition of β-lactose, a
competitive carbohydrate inhibitor of galectin-1 binding activity. VEGFR-2 protein, like galectin-1, is also highly expressed in the mid CL, and it was modified by multi-antennary glycans, which can be recognized by galectin-1. An overlay assay using biotinylated galectin-1 revealed that galectin-1 directly binds to asparagine-linked glycans (N-glycans) on VEGFR-2. Enhancement of LSC viability by galectin-1 was suppressed by a selective inhibitor of VEGFR-2. The overall findings suggest that galectin-1 plays a role as a survival factor in the bovine CL, possibly by binding to N-glycans on VEGFR-2.
Collapse
Affiliation(s)
- Masahiro Sano
- Laboratory of Reproductive Physiology, Graduate School of Environmental and Life Science, Okayama University, Okayama 700-8530, Japan
| | | | | | | |
Collapse
|
189
|
Vasen G, Battistone MA, Croci DO, Brukman NG, Weigel Muñoz M, Stupirski JC, Rabinovich GA, Cuasnicú PS. The galectin-1-glycan axis controls sperm fertilizing capacity by regulating sperm motility and membrane hyperpolarization. FASEB J 2015; 29:4189-200. [PMID: 26136479 DOI: 10.1096/fj.15-270975] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2015] [Accepted: 06/15/2015] [Indexed: 12/15/2022]
Abstract
Lectin-glycan recognition systems play central roles in many physiologic and pathologic processes. We identified a role for galectin-1 (Gal-1), a highly conserved glycan-binding protein, in the control of sperm function. We found that Gal-1 is expressed in the epididymis and associates with sperm during epididymal maturation. Exposure of sperm to Gal-1 resulted in glycan-dependent modulation of the acrosome reaction (AR), a key event in the fertilization process. Gal-1-deficient (Lgals1(-/-)) mice revealed the essential contribution of this lectin for full sperm fertilizing ability both in vitro and in vivo. Mechanistically, Lgals1(-/-) sperm exhibited defects in their ability to develop hyperactivation, a vigorous motility required for penetration of the egg vestments. Moreover, Lgals1(-/-) sperm showed a decreased ability to control cell volume and to undergo progesterone-induced AR, phenotypes that were rescued by exposure of the cells to recombinant Gal-1. Interestingly, the AR defect was associated with a deficiency in sperm membrane potential hyperpolarization. Our study highlights the relevance of the Gal-1-glycan axis in sperm function with critical implications in mammalian reproductive biology.
Collapse
Affiliation(s)
- Gustavo Vasen
- *Instituto de Biología y Medicina Experimental (IBYME-CONICET), Buenos Aires, Argentina; and Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Maria Agustina Battistone
- *Instituto de Biología y Medicina Experimental (IBYME-CONICET), Buenos Aires, Argentina; and Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Diego O Croci
- *Instituto de Biología y Medicina Experimental (IBYME-CONICET), Buenos Aires, Argentina; and Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Nicolás G Brukman
- *Instituto de Biología y Medicina Experimental (IBYME-CONICET), Buenos Aires, Argentina; and Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Mariana Weigel Muñoz
- *Instituto de Biología y Medicina Experimental (IBYME-CONICET), Buenos Aires, Argentina; and Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Juan C Stupirski
- *Instituto de Biología y Medicina Experimental (IBYME-CONICET), Buenos Aires, Argentina; and Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Gabriel A Rabinovich
- *Instituto de Biología y Medicina Experimental (IBYME-CONICET), Buenos Aires, Argentina; and Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Patricia S Cuasnicú
- *Instituto de Biología y Medicina Experimental (IBYME-CONICET), Buenos Aires, Argentina; and Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Buenos Aires, Argentina
| |
Collapse
|
190
|
Garcin PO, Nabi IR, Panté N. Galectin-3 plays a role in minute virus of mice infection. Virology 2015; 481:63-72. [DOI: 10.1016/j.virol.2015.02.019] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2014] [Revised: 02/07/2015] [Accepted: 02/13/2015] [Indexed: 12/19/2022]
|
191
|
Assembly, organization and regulation of cell-surface receptors by lectin–glycan complexes. Biochem J 2015; 469:1-16. [DOI: 10.1042/bj20150461] [Citation(s) in RCA: 54] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Galectins are a family of β-galactoside-binding lectins carrying at least one consensus sequence in the carbohydrate-recognition domain. Properties of glycosylated ligands, such as N- and O-glycan branching, LacNAc (N-acetyl-lactosamine) content and the balance of α2,3- and α2,6-linked sialic acid dramatically influence galectin binding to a preferential set of counter-receptors. The presentation of specific glycans in galectin-binding partners is also critical, as proper orientation and clustering of oligosaccharide ligands on multiple carbohydrate side chains increase the binding avidity of galectins for particular glycosylated receptors. When galectins are released from the cells, they typically concentrate on the cell surface and the local matrix, raising their local concentration. Thus galectins can form their own multimers in the extracellular milieu, which in turn cross-link glycoconjugates on the cell surface generating galectin–glycan complexes that modulate intracellular signalling pathways, thus regulating cellular processes such as apoptosis, proliferation, migration and angiogenesis. Subtle changes in receptor expression, rates of protein synthesis, activities of Golgi enzymes, metabolite concentrations supporting glycan biosynthesis, density of glycans, strength of protein–protein interactions at the plasma membrane and stoichiometry may modify galectin–glycan complexes. Although galectins are key contributors to the formation of these extended glycan complexes leading to promotion of receptor segregation/clustering, and inhibition of receptor internalization by surface retention, when these complexes are disrupted, some galectins, particularly galectin-3 and -4, showed the ability to drive clathrin-independent mechanisms of endocytosis. In the present review, we summarize the data available on the assembly, hierarchical organization and regulation of conspicuous galectin–glycan complexes, and their implications in health and disease.
Collapse
|
192
|
Abstract
Galectins are a family of widely expressed β-galactoside-binding lectins in metazoans. The 15 mammalian galectins have either one or two conserved carbohydrate recognition domains (CRDs), with galectin-3 being able to pentamerize; they form complexes that crosslink glycosylated ligands to form a dynamic lattice. The galectin lattice regulates the diffusion, compartmentalization and endocytosis of plasma membrane glycoproteins and glycolipids. The galectin lattice also regulates the selection, activation and arrest of T cells, receptor kinase signaling and the functionality of membrane receptors, including the glucagon receptor, glucose and amino acid transporters, cadherins and integrins. The affinity of transmembrane glycoproteins to the galectin lattice is proportional to the number and branching of their N-glycans; with branching being mediated by Golgi N-acetylglucosaminyltransferase-branching enzymes and the supply of UDP-GlcNAc through metabolite flux through the hexosamine biosynthesis pathway. The relative affinities of glycoproteins for the galectin lattice depend on the activities of the Golgi enzymes that generate the epitopes of their ligands and, thus, provide a means to analyze biological function of lectins and of the 'glycome' more broadly.
Collapse
Affiliation(s)
- Ivan R Nabi
- Department of Cellular and Physiological Sciences, Life Sciences Institute, 2350 Health Sciences Mall, University of British Columbia, Vancouver, British Columbia, Canada V6T 1Z3
| | - Jay Shankar
- Department of Cellular and Physiological Sciences, Life Sciences Institute, 2350 Health Sciences Mall, University of British Columbia, Vancouver, British Columbia, Canada V6T 1Z3
| | - James W Dennis
- Department of Medical Genetics and Laboratory Medicine and Pathology, University of Toronto, Toronto, Ontario, Canada M5G 1L5
| |
Collapse
|
193
|
Ochieng J, Nangami GN, Ogunkua O, Miousse IR, Koturbash I, Odero-Marah V, McCawley L, Nangia-Makker P, Ahmed N, Luqmani Y, Chen Z, Papagerakis S, Wolf GT, Dong C, Zhou BP, Brown DG, Colacci A, Hamid RA, Mondello C, Raju J, Ryan EP, Woodrick J, Scovassi I, Singh N, Vaccari M, Roy R, Forte S, Memeo L, Salem HK, Amedei A, Al-Temaimi R, Al-Mulla F, Bisson WH, Eltom SE. The impact of low-dose carcinogens and environmental disruptors on tissue invasion and metastasis. Carcinogenesis 2015; 36 Suppl 1:S128-S159. [PMID: 26106135 PMCID: PMC4565611 DOI: 10.1093/carcin/bgv034] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2014] [Revised: 11/14/2014] [Accepted: 11/19/2014] [Indexed: 12/12/2022] Open
Abstract
The purpose of this review is to stimulate new ideas regarding low-dose environmental mixtures and carcinogens and their potential to promote invasion and metastasis. Whereas a number of chapters in this review are devoted to the role of low-dose environmental mixtures and carcinogens in the promotion of invasion and metastasis in specific tumors such as breast and prostate, the overarching theme is the role of low-dose carcinogens in the progression of cancer stem cells. It is becoming clearer that cancer stem cells in a tumor are the ones that assume invasive properties and colonize distant organs. Therefore, low-dose contaminants that trigger epithelial-mesenchymal transition, for example, in these cells are of particular interest in this review. This we hope will lead to the collaboration between scientists who have dedicated their professional life to the study of carcinogens and those whose interests are exclusively in the arena of tissue invasion and metastasis.
Collapse
Affiliation(s)
- Josiah Ochieng
- *To whom correspondence should be addressed. Tel: +1 615 327 6119; Fax: +1 615 327 6442;
| | - Gladys N. Nangami
- Department of Biochemistry and Cancer Biology, Meharry Medical College, Nashville, TN 37208, USA
- Department of Environmental and Occupational Health, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
- Department of Biology/Center for Cancer Research and Therapeutic Development, Clark Atlanta University, Atlanta, GA 30314, USA
- Department of Cancer Biology, Vanderbilt University, Nashville, TN 37232, USA
- Department of Pathology, Wayne State University, Detroit, MI 48201, USA
- Department of Obstetrics and Gynecology, University of Melbourne, Melbourne, Victoria, Australia
- Faculty of Pharmacy, Department of Pathology, Kuwait University, Safat 13110, Kuwait
- Department of Otolaryngology, University of Michigan Medical College, Ann Arbor, MI 48109, USA
- Department of Molecular & Cellular Biochemistry, University of Kentucky, Lexington, KY 40506, USA
- Department of Environmental and Radiological Health Sciences/Food Science and Human Nutrition, College of Veterinary Medicine and Biomedical Sciences, Colorado State University/Colorado School of Public Health, Fort Collins, CO 80523-1680, USA
- Center for Environmental Carcinogenesis and Risk Assessment, Environmental Protection and Health Prevention Agency, Bologna 40126, Italy
- Faculty of Medicine and Health Sciences, University Putra, Serdang, Selangor 43400, Malaysia
- Istituto di Genetica Molecolare, CNR, via Abbiategrasso 207, 27100 Pavia, Italy
- Toxicology Research Division, Bureau of Chemical Safety Food Directorate, Health Products and Food Branch Health Canada, Ottawa, Ontario K1A0K9, Canada
- Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, DC 20057, USA
- Centre for Advanced Research, King George’s Medical University, Chowk, Lucknow, Uttar Pradesh 226003, India
- Mediterranean Institute of Oncology, Viagrande 95029, Italy
- Urology Department, kasr Al-Ainy School of Medicine, Cairo University, El Manial, Cairo 12515, Egypt
- Department of Experimental and Clinical Medicine, University of Firenze, Firenze 50134, Italy and
- Environmental and Molecular Toxicology, Environmental Health Sciences Center, Oregon State University, Corvallis, OR 97331, USA
| | - Olugbemiga Ogunkua
- Department of Biochemistry and Cancer Biology, Meharry Medical College, Nashville, TN 37208, USA
- Department of Environmental and Occupational Health, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
- Department of Biology/Center for Cancer Research and Therapeutic Development, Clark Atlanta University, Atlanta, GA 30314, USA
- Department of Cancer Biology, Vanderbilt University, Nashville, TN 37232, USA
- Department of Pathology, Wayne State University, Detroit, MI 48201, USA
- Department of Obstetrics and Gynecology, University of Melbourne, Melbourne, Victoria, Australia
- Faculty of Pharmacy, Department of Pathology, Kuwait University, Safat 13110, Kuwait
- Department of Otolaryngology, University of Michigan Medical College, Ann Arbor, MI 48109, USA
- Department of Molecular & Cellular Biochemistry, University of Kentucky, Lexington, KY 40506, USA
- Department of Environmental and Radiological Health Sciences/Food Science and Human Nutrition, College of Veterinary Medicine and Biomedical Sciences, Colorado State University/Colorado School of Public Health, Fort Collins, CO 80523-1680, USA
- Center for Environmental Carcinogenesis and Risk Assessment, Environmental Protection and Health Prevention Agency, Bologna 40126, Italy
- Faculty of Medicine and Health Sciences, University Putra, Serdang, Selangor 43400, Malaysia
- Istituto di Genetica Molecolare, CNR, via Abbiategrasso 207, 27100 Pavia, Italy
- Toxicology Research Division, Bureau of Chemical Safety Food Directorate, Health Products and Food Branch Health Canada, Ottawa, Ontario K1A0K9, Canada
- Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, DC 20057, USA
- Centre for Advanced Research, King George’s Medical University, Chowk, Lucknow, Uttar Pradesh 226003, India
- Mediterranean Institute of Oncology, Viagrande 95029, Italy
- Urology Department, kasr Al-Ainy School of Medicine, Cairo University, El Manial, Cairo 12515, Egypt
- Department of Experimental and Clinical Medicine, University of Firenze, Firenze 50134, Italy and
- Environmental and Molecular Toxicology, Environmental Health Sciences Center, Oregon State University, Corvallis, OR 97331, USA
| | - Isabelle R. Miousse
- Department of Environmental and Occupational Health, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| | - Igor Koturbash
- Department of Environmental and Occupational Health, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| | - Valerie Odero-Marah
- Department of Biology/Center for Cancer Research and Therapeutic Development, Clark Atlanta University, Atlanta, GA 30314, USA
| | - Lisa McCawley
- Department of Cancer Biology, Vanderbilt University, Nashville, TN 37232, USA
| | | | - Nuzhat Ahmed
- Department of Obstetrics and Gynecology, University of Melbourne, Melbourne, Victoria, Australia
| | - Yunus Luqmani
- Faculty of Pharmacy, Department of Pathology, Kuwait University, Safat 13110, Kuwait
| | - Zhenbang Chen
- Department of Biochemistry and Cancer Biology, Meharry Medical College, Nashville, TN 37208, USA
- Department of Environmental and Occupational Health, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
- Department of Biology/Center for Cancer Research and Therapeutic Development, Clark Atlanta University, Atlanta, GA 30314, USA
- Department of Cancer Biology, Vanderbilt University, Nashville, TN 37232, USA
- Department of Pathology, Wayne State University, Detroit, MI 48201, USA
- Department of Obstetrics and Gynecology, University of Melbourne, Melbourne, Victoria, Australia
- Faculty of Pharmacy, Department of Pathology, Kuwait University, Safat 13110, Kuwait
- Department of Otolaryngology, University of Michigan Medical College, Ann Arbor, MI 48109, USA
- Department of Molecular & Cellular Biochemistry, University of Kentucky, Lexington, KY 40506, USA
- Department of Environmental and Radiological Health Sciences/Food Science and Human Nutrition, College of Veterinary Medicine and Biomedical Sciences, Colorado State University/Colorado School of Public Health, Fort Collins, CO 80523-1680, USA
- Center for Environmental Carcinogenesis and Risk Assessment, Environmental Protection and Health Prevention Agency, Bologna 40126, Italy
- Faculty of Medicine and Health Sciences, University Putra, Serdang, Selangor 43400, Malaysia
- Istituto di Genetica Molecolare, CNR, via Abbiategrasso 207, 27100 Pavia, Italy
- Toxicology Research Division, Bureau of Chemical Safety Food Directorate, Health Products and Food Branch Health Canada, Ottawa, Ontario K1A0K9, Canada
- Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, DC 20057, USA
- Centre for Advanced Research, King George’s Medical University, Chowk, Lucknow, Uttar Pradesh 226003, India
- Mediterranean Institute of Oncology, Viagrande 95029, Italy
- Urology Department, kasr Al-Ainy School of Medicine, Cairo University, El Manial, Cairo 12515, Egypt
- Department of Experimental and Clinical Medicine, University of Firenze, Firenze 50134, Italy and
- Environmental and Molecular Toxicology, Environmental Health Sciences Center, Oregon State University, Corvallis, OR 97331, USA
| | - Silvana Papagerakis
- Department of Otolaryngology, University of Michigan Medical College, Ann Arbor, MI 48109, USA
| | - Gregory T. Wolf
- Department of Otolaryngology, University of Michigan Medical College, Ann Arbor, MI 48109, USA
| | - Chenfang Dong
- Department of Molecular & Cellular Biochemistry, University of Kentucky, Lexington, KY 40506, USA
| | - Binhua P. Zhou
- Department of Molecular & Cellular Biochemistry, University of Kentucky, Lexington, KY 40506, USA
| | - Dustin G. Brown
- Department of Environmental and Radiological Health Sciences/Food Science and Human Nutrition, College of Veterinary Medicine and Biomedical Sciences, Colorado State University/Colorado School of Public Health, Fort Collins, CO 80523-1680, USA
| | - Annamaria Colacci
- Center for Environmental Carcinogenesis and Risk Assessment, Environmental Protection and Health Prevention Agency, Bologna 40126, Italy
| | - Roslida A. Hamid
- Faculty of Medicine and Health Sciences, University Putra, Serdang, Selangor 43400, Malaysia
| | - Chiara Mondello
- Istituto di Genetica Molecolare, CNR, via Abbiategrasso 207, 27100 Pavia, Italy
| | - Jayadev Raju
- Toxicology Research Division, Bureau of Chemical Safety Food Directorate, Health Products and Food Branch Health Canada, Ottawa, Ontario K1A0K9, Canada
| | - Elizabeth P. Ryan
- Department of Environmental and Radiological Health Sciences/Food Science and Human Nutrition, College of Veterinary Medicine and Biomedical Sciences, Colorado State University/Colorado School of Public Health, Fort Collins, CO 80523-1680, USA
| | - Jordan Woodrick
- Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, DC 20057, USA
| | - Ivana Scovassi
- Istituto di Genetica Molecolare, CNR, via Abbiategrasso 207, 27100 Pavia, Italy
| | - Neetu Singh
- Centre for Advanced Research, King George’s Medical University, Chowk, Lucknow, Uttar Pradesh 226003, India
| | - Monica Vaccari
- Center for Environmental Carcinogenesis and Risk Assessment, Environmental Protection and Health Prevention Agency, Bologna 40126, Italy
| | - Rabindra Roy
- Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, DC 20057, USA
| | - Stefano Forte
- Mediterranean Institute of Oncology, Viagrande 95029, Italy
| | - Lorenzo Memeo
- Mediterranean Institute of Oncology, Viagrande 95029, Italy
| | - Hosni K. Salem
- Urology Department, kasr Al-Ainy School of Medicine, Cairo University, El Manial, Cairo 12515, Egypt
| | - Amedeo Amedei
- Department of Experimental and Clinical Medicine, University of Firenze, Firenze 50134, Italy and
| | - Rabeah Al-Temaimi
- Faculty of Pharmacy, Department of Pathology, Kuwait University, Safat 13110, Kuwait
| | - Fahd Al-Mulla
- Faculty of Pharmacy, Department of Pathology, Kuwait University, Safat 13110, Kuwait
| | - William H. Bisson
- Environmental and Molecular Toxicology, Environmental Health Sciences Center, Oregon State University, Corvallis, OR 97331, USA
| | - Sakina E. Eltom
- Department of Biochemistry and Cancer Biology, Meharry Medical College, Nashville, TN 37208, USA
- Department of Environmental and Occupational Health, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
- Department of Biology/Center for Cancer Research and Therapeutic Development, Clark Atlanta University, Atlanta, GA 30314, USA
- Department of Cancer Biology, Vanderbilt University, Nashville, TN 37232, USA
- Department of Pathology, Wayne State University, Detroit, MI 48201, USA
- Department of Obstetrics and Gynecology, University of Melbourne, Melbourne, Victoria, Australia
- Faculty of Pharmacy, Department of Pathology, Kuwait University, Safat 13110, Kuwait
- Department of Otolaryngology, University of Michigan Medical College, Ann Arbor, MI 48109, USA
- Department of Molecular & Cellular Biochemistry, University of Kentucky, Lexington, KY 40506, USA
- Department of Environmental and Radiological Health Sciences/Food Science and Human Nutrition, College of Veterinary Medicine and Biomedical Sciences, Colorado State University/Colorado School of Public Health, Fort Collins, CO 80523-1680, USA
- Center for Environmental Carcinogenesis and Risk Assessment, Environmental Protection and Health Prevention Agency, Bologna 40126, Italy
- Faculty of Medicine and Health Sciences, University Putra, Serdang, Selangor 43400, Malaysia
- Istituto di Genetica Molecolare, CNR, via Abbiategrasso 207, 27100 Pavia, Italy
- Toxicology Research Division, Bureau of Chemical Safety Food Directorate, Health Products and Food Branch Health Canada, Ottawa, Ontario K1A0K9, Canada
- Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, DC 20057, USA
- Centre for Advanced Research, King George’s Medical University, Chowk, Lucknow, Uttar Pradesh 226003, India
- Mediterranean Institute of Oncology, Viagrande 95029, Italy
- Urology Department, kasr Al-Ainy School of Medicine, Cairo University, El Manial, Cairo 12515, Egypt
- Department of Experimental and Clinical Medicine, University of Firenze, Firenze 50134, Italy and
- Environmental and Molecular Toxicology, Environmental Health Sciences Center, Oregon State University, Corvallis, OR 97331, USA
| |
Collapse
|
194
|
Vinik Y, Shatz-Azoulay H, Vivanti A, Hever N, Levy Y, Karmona R, Brumfeld V, Baraghithy S, Attar-Lamdar M, Boura-Halfon S, Bab I, Zick Y. The mammalian lectin galectin-8 induces RANKL expression, osteoclastogenesis, and bone mass reduction in mice. eLife 2015; 4:e05914. [PMID: 25955862 PMCID: PMC4424493 DOI: 10.7554/elife.05914] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2014] [Accepted: 04/16/2015] [Indexed: 01/20/2023] Open
Abstract
Skeletal integrity is maintained by the co-ordinated activity of osteoblasts, the bone-forming cells, and osteoclasts, the bone-resorbing cells. In this study, we show that mice overexpressing galectin-8, a secreted mammalian lectin of the galectins family, exhibit accelerated osteoclasts activity and bone turnover, which culminates in reduced bone mass, similar to cases of postmenopausal osteoporosis and cancerous osteolysis. This phenotype can be attributed to a direct action of galectin-8 on primary cultures of osteoblasts that secrete the osteoclastogenic factor RANKL upon binding of galectin-8. This results in enhanced differentiation into osteoclasts of the bone marrow cells co-cultured with galectin-8-treated osteoblasts. Secretion of RANKL by galectin-8-treated osteoblasts can be attributed to binding of galectin-8 to receptor complexes that positively (uPAR and MRC2) and negatively (LRP1) regulate galectin-8 function. Our findings identify galectins as new players in osteoclastogenesis and bone remodeling, and highlight a potential regulation of bone mass by animal lectins. DOI:http://dx.doi.org/10.7554/eLife.05914.001 The forces applied to the body during daily activities cause bones to be constantly remodeled, which is essential for keeping them healthy. In most adult organisms, new bone is created at the same rate at which old bone is destroyed. This means that overall bone mass remains the same. But, in diseases such as osteoporosis or bone cancer, bone is destroyed more rapidly than at which new bone is made. This leads to brittle bones that are more likely to fracture. Understanding how to increase the rate of bone renewal might therefore help scientists develop new treatments for bone diseases. Bone is created by cells called osteoblasts and destroyed by other cells called osteoclasts. Both of these types of cells develop from stem cells in the bone marrow. The activity of these cells is controlled by a number of factors, including the matrix of proteins that holds bone together. A group of proteins called galectins are known to act as a bridge between some of the matrix proteins and molecules on the surface of the cells. Vinik et al. took osteoblasts from a mouse skull, grew them in the laboratory, and then exposed them to a galectin protein called galectin-8. This made the osteoblasts release a protein called RANKL, which is known to boost osteoclast activity. When osteoblasts that had been exposed to galectin-8 were grown alongside bone marrow stem cells, more of the stem cells developed into the bone-destroying osteoclasts. Mice that were genetically engineered to produce more galectin-8 than normal mice develop brittle bones, despite also creating new bone at a higher rate than do normal mice. This is because osteoclast activity increases at a greater rate, resulting in an overall loss of bone in these animals. This is similar to what occurs in some individuals with osteoporosis. These experiments therefore suggest that galectin-8 plays an important role in bone remodeling and that it may be a potential target for drugs that treat diseases that weaken bones. DOI:http://dx.doi.org/10.7554/eLife.05914.002
Collapse
Affiliation(s)
- Yaron Vinik
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel
| | - Hadas Shatz-Azoulay
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel
| | - Alessia Vivanti
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel
| | - Navit Hever
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel
| | - Yifat Levy
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel
| | - Rotem Karmona
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel
| | - Vlad Brumfeld
- Department of Chemical Research Support, Weizmann Institute of Science, Rehovot, Israel
| | - Saja Baraghithy
- Bone Laboratory, The Hebrew University of Jerusalem, Jerusalem, Israel
| | | | - Sigalit Boura-Halfon
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel
| | - Itai Bab
- Bone Laboratory, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Yehiel Zick
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel
| |
Collapse
|
195
|
Hitting the sweet spot-glycans as targets of fungal defense effector proteins. Molecules 2015; 20:8144-67. [PMID: 25955890 PMCID: PMC6272156 DOI: 10.3390/molecules20058144] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2015] [Revised: 04/29/2015] [Accepted: 04/30/2015] [Indexed: 11/16/2022] Open
Abstract
Organisms which rely solely on innate defense systems must combat a large number of antagonists with a comparably low number of defense effector molecules. As one solution of this problem, these organisms have evolved effector molecules targeting epitopes that are conserved between different antagonists of a specific taxon or, if possible, even of different taxa. In order to restrict the activity of the defense effector molecules to physiologically relevant taxa, these target epitopes should, on the other hand, be taxon-specific and easily accessible. Glycans fulfill all these requirements and are therefore a preferred target of defense effector molecules, in particular defense proteins. Here, we review this defense strategy using the example of the defense system of multicellular (filamentous) fungi against microbial competitors and animal predators.
Collapse
|
196
|
Lhuillier C, Barjon C, Niki T, Gelin A, Praz F, Morales O, Souquere S, Hirashima M, Wei M, Dellis O, Busson P. Impact of Exogenous Galectin-9 on Human T Cells: CONTRIBUTION OF THE T CELL RECEPTOR COMPLEX TO ANTIGEN-INDEPENDENT ACTIVATION BUT NOT TO APOPTOSIS INDUCTION. J Biol Chem 2015; 290:16797-811. [PMID: 25947381 DOI: 10.1074/jbc.m115.661272] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2015] [Indexed: 11/06/2022] Open
Abstract
Galectin-9 (gal-9) is a multifunctional β-galactoside-binding lectin, frequently released in the extracellular medium, where it acts as a pleiotropic immune modulator. Despite its overall immunosuppressive effects, a recent study has reported bimodal action of gal-9 on human resting blood T cells with apoptosis occurring in the majority of them, followed by a wave of activation and expansion of Th1 cells in the surviving population. Our knowledge of the signaling events triggered by exogenous gal-9 in T cells remains limited. One of these events is cytosolic calcium (Ca(2+)) release reported in some murine and human T cells. The aim of this study was to investigate the contribution of Ca(2+) mobilization to apoptotic and nonapoptotic effects of exogenous gal-9 in human T cells. We found that the T cell receptor (TCR)-CD3 complex and the Lck kinase were required for Ca(2+) mobilization but not for apoptosis induction in Jurkat cells. These data were confirmed in human CD4(+) T cells from peripheral blood as follows: a specific Lck chemical inhibitor abrogated Ca(2+) mobilization but not apoptosis induction. Moreover, Lck activity was also required for the production of Th1-type cytokines, i.e. interleukin-2 and interferon-γ, which resulted from gal-9 stimulation in peripheral CD4(+) T cells. These findings indicate that gal-9 acts on T cells by two distinct pathways as follows: one mimicking antigen-specific activation of the TCR with a mandatory contribution of proximal elements of the TCR complex, especially Lck, and another resulting in apoptosis that is independent of this complex.
Collapse
Affiliation(s)
- Claire Lhuillier
- From the Université Paris-Sud, 15 Rue Georges Clémenceau, 91400, Orsay, France, the CNRS, UMR 8126, Institut Gustave Roussy, 114 Rue Edouard Vaillant, 94805 Villejuif Cedex, France, the Cellvax, Ecole Nationale Vétérinaire d'Alfort, 7 Avenue du Général de Gaulle, 94704 Maisons-Alfort Cedex, France
| | - Clément Barjon
- From the Université Paris-Sud, 15 Rue Georges Clémenceau, 91400, Orsay, France, the CNRS, UMR 8126, Institut Gustave Roussy, 114 Rue Edouard Vaillant, 94805 Villejuif Cedex, France, the Cellvax, Ecole Nationale Vétérinaire d'Alfort, 7 Avenue du Général de Gaulle, 94704 Maisons-Alfort Cedex, France
| | - Toshiro Niki
- the Department of Immunology and Immunopathology, Faculty of Medicine, Kagawa University, Kagawa 761-0793, Japan, the GalPharma Co., Ltd., Takamatsu, Kagawa 761-0301, Japan
| | - Aurore Gelin
- the CNRS, UMR 8126, Institut Gustave Roussy, 114 Rue Edouard Vaillant, 94805 Villejuif Cedex, France
| | - Françoise Praz
- INSERM, UMR-S 938, Centre de Recherche Saint-Antoine, 75012, Paris, France, the Sorbonne Universités, UPMC Université Paris 06, UMR-S 938, Centre de Recherche Saint-Antoine, 75012, Paris, France
| | - Olivier Morales
- CNRS, UMR 8161 Groupe IRCV, Institut de Biologie de Lille, 1 Rue du Pr. Calmette, 59021 Lille, France
| | - Sylvie Souquere
- From the Université Paris-Sud, 15 Rue Georges Clémenceau, 91400, Orsay, France, UMR 8122, Institut Gustave Roussy, 114 Rue Edouard Vaillant, 94805 Villejuif Cedex, France, and
| | - Mitsuomi Hirashima
- the Department of Immunology and Immunopathology, Faculty of Medicine, Kagawa University, Kagawa 761-0793, Japan, the GalPharma Co., Ltd., Takamatsu, Kagawa 761-0301, Japan
| | - Ming Wei
- the Cellvax, Ecole Nationale Vétérinaire d'Alfort, 7 Avenue du Général de Gaulle, 94704 Maisons-Alfort Cedex, France
| | - Olivier Dellis
- From the Université Paris-Sud, 15 Rue Georges Clémenceau, 91400, Orsay, France, INSERM, UMR-S 757, Bâtiment 440/443, Rue des Adèles, 91405 Orsay, France
| | - Pierre Busson
- From the Université Paris-Sud, 15 Rue Georges Clémenceau, 91400, Orsay, France, the CNRS, UMR 8126, Institut Gustave Roussy, 114 Rue Edouard Vaillant, 94805 Villejuif Cedex, France,
| |
Collapse
|
197
|
Takeuchi T, Tamura M, Ishii N, Ishikida H, Sugimoto S, Suzuki D, Nishiyama K, Takahashi H, Natsugari H, Arata Y. Purification of galectin-1 mutants using an immobilized Galactoseβ1-4Fucose affinity adsorbent. Protein Expr Purif 2015; 111:82-6. [PMID: 25858314 DOI: 10.1016/j.pep.2015.04.001] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2015] [Revised: 03/31/2015] [Accepted: 04/01/2015] [Indexed: 11/19/2022]
Abstract
Galectins are a family of lectins characterized by their carbohydrate recognition domains containing eight conserved amino acid residues, which allows the binding of galectin to β-galactoside sugars such as Galβ1-4GlcNAc. Since galectin-glycan interactions occur extracellularly, recombinant galectins are often used for the functional analysis of these interactions. Although it is relatively easy to purify galectins via affinity to Galβ1-4GlcNAc using affinity adsorbents such as asialofetuin-Sepharose, it could be difficult to do so with mutated galectins, which may have reduced affinity towards their endogenous ligands. However, this is not the case with Caenorhabditis elegans galectin LEC-6; binding to its endogenous recognition unit Galβ1-4Fuc, a unique disaccharide found only in invertebrates, is not necessarily affected by point mutations of the eight well-conserved amino acids. In this study, we constructed mutants of mouse galectin-1 carrying substitutions of each of the eight conserved amino acid residues (H44F, N46D, R48H, V59A, N61D, W68F, E71Q, and R73H) and examined their affinity for Galβ1-4GlcNAc and Galβ1-4Fuc. These mutants, except W68F, had very low affinity for asialofetuin-Sepharose; however, most of them (with the exception of H44F and R48H) could be purified using Galβ1-4Fuc-Sepharose. The affinity of the purified mutant galectins for glycans containing Galβ1-4Fuc or Galβ1-4GlcNAc moieties was quantitatively examined by frontal affinity chromatography, and the results indicated that the mutants retained the affinity only for Galβ1-4Fuc. Given that other mammalian galectins are known to bind Galβ1-4Fuc, our data suggest that immobilized Galβ1-4Fuc ligands could be generally used for easy one-step affinity purification of mutant galectins.
Collapse
Affiliation(s)
- Tomoharu Takeuchi
- Laboratory of Biochemistry, Faculty of Pharmaceutical Sciences, Josai University, 1-1 Keyakidai, Sakado, Saitama 350-0295, Japan.
| | - Mayumi Tamura
- Laboratory of Biochemistry, Faculty of Pharmaceutical Sciences, Josai University, 1-1 Keyakidai, Sakado, Saitama 350-0295, Japan
| | - Nobuaki Ishii
- Laboratory of Biochemistry, Faculty of Pharmaceutical Sciences, Josai University, 1-1 Keyakidai, Sakado, Saitama 350-0295, Japan
| | - Hiroki Ishikida
- Laboratory of Biochemistry, Faculty of Pharmaceutical Sciences, Josai University, 1-1 Keyakidai, Sakado, Saitama 350-0295, Japan
| | - Saori Sugimoto
- Laboratory of Biochemistry, Faculty of Pharmaceutical Sciences, Josai University, 1-1 Keyakidai, Sakado, Saitama 350-0295, Japan
| | - Daichi Suzuki
- Laboratory of Biochemistry, Faculty of Pharmaceutical Sciences, Josai University, 1-1 Keyakidai, Sakado, Saitama 350-0295, Japan
| | - Kazusa Nishiyama
- Laboratory of Synthetic Organic and Medicinal Chemistry, School of Pharmaceutical Sciences, Teikyo University, 2-11-1 Kaga, Itabashi-ku, Tokyo 173-8605, Japan
| | - Hideyo Takahashi
- Laboratory of Synthetic Organic and Medicinal Chemistry, School of Pharmaceutical Sciences, Teikyo University, 2-11-1 Kaga, Itabashi-ku, Tokyo 173-8605, Japan
| | - Hideaki Natsugari
- Laboratory of Synthetic Organic and Medicinal Chemistry, School of Pharmaceutical Sciences, Teikyo University, 2-11-1 Kaga, Itabashi-ku, Tokyo 173-8605, Japan
| | - Yoichiro Arata
- Laboratory of Biochemistry, Faculty of Pharmaceutical Sciences, Josai University, 1-1 Keyakidai, Sakado, Saitama 350-0295, Japan
| |
Collapse
|
198
|
Yang Q, Yang Y, Li L, Sun W, Zhu X, Huang Y. Polymeric nanomedicine for tumor-targeted combination therapy to elicit synergistic genotoxicity against prostate cancer. ACS APPLIED MATERIALS & INTERFACES 2015; 7:6661-6673. [PMID: 25775367 DOI: 10.1021/am509204u] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/04/2023]
Abstract
To improve the therapeutic efficacy of anticancer combination therapy, we designed a nanoplatform based on N-(2-hydroxypropyl) methacrylamide (HPMA) copolymers that allows covalent bonding of two chemotherapeutics acting via different anticancer mechanisms and that can enter target cells by receptor-mediated endocytosis. Doxorubicin (DOX) was covalently conjugated to a nanosized HPMA copolymer using a pH-sensitive hydrazone bond and 5-fluorouracil (5-Fu) was conjugated to the same backbone using an enzymatically degradable oligopeptide Gly-Phe-Leu-Gly sequence. Then, the conjugate was decorated with galectin-3 targeting peptide G3-C12 [P-(G3-C12)-DOX-Fu]. The two drugs showed similar in vitro release profiles, suggesting that they may be able to work synergistically in the codelivery system. In galectin-3 overexpressed PC-3 human prostate carcinoma cells, P-(G3-C12)-DOX-Fu surprisingly exhibited comparable cytotoxicity to free DOX at high concentration by increasing cell internalization and exerting synergistic genotoxic effects of cell cycle arrest, caspase-3 activation, and DNA damage. In mice bearing PC-3 tumor xenografts, the use of tumor-targeting ligand substantially enhanced the intracellular delivery of P-(G3-C12)-DOX-Fu in tumors. The targeted dual drug-loaded conjugate inhibited tumor growth to a greater extent (tumor inhibition of 81.6%) than did nontargeted P-DOX-Fu (71.2%), P-DOX (63%), DOX·HCl (40.5%), P-Fu (32.0%), or 5-Fu (14.6%), without inducing any obvious side effects. These results demonstrate the potential of synergistic combination therapy using targeted nanocarriers for efficient treatment of prostate cancer.
Collapse
Affiliation(s)
- Qingqing Yang
- Key Laboratory of Drug Targeting and Drug Delivery System, Ministry of Education, West China School of Pharmacy, Sichuan University, No. 17, Block 3, Southern Renmin Road, Chengdu 610041, People's Republic of China
| | - Yang Yang
- Key Laboratory of Drug Targeting and Drug Delivery System, Ministry of Education, West China School of Pharmacy, Sichuan University, No. 17, Block 3, Southern Renmin Road, Chengdu 610041, People's Republic of China
| | - Lian Li
- Key Laboratory of Drug Targeting and Drug Delivery System, Ministry of Education, West China School of Pharmacy, Sichuan University, No. 17, Block 3, Southern Renmin Road, Chengdu 610041, People's Republic of China
| | - Wei Sun
- Key Laboratory of Drug Targeting and Drug Delivery System, Ministry of Education, West China School of Pharmacy, Sichuan University, No. 17, Block 3, Southern Renmin Road, Chengdu 610041, People's Republic of China
| | - Xi Zhu
- Key Laboratory of Drug Targeting and Drug Delivery System, Ministry of Education, West China School of Pharmacy, Sichuan University, No. 17, Block 3, Southern Renmin Road, Chengdu 610041, People's Republic of China
| | - Yuan Huang
- Key Laboratory of Drug Targeting and Drug Delivery System, Ministry of Education, West China School of Pharmacy, Sichuan University, No. 17, Block 3, Southern Renmin Road, Chengdu 610041, People's Republic of China
| |
Collapse
|
199
|
Argüeso P, Mauris J, Uchino Y. Galectin-3 as a regulator of the epithelial junction: Implications to wound repair and cancer. Tissue Barriers 2015; 3:e1026505. [PMID: 26451339 DOI: 10.1080/21688370.2015.1026505] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2014] [Revised: 02/27/2015] [Accepted: 02/27/2015] [Indexed: 10/23/2022] Open
Abstract
Epithelial cells are closely connected to each other and to the extracellular matrix by a set of adhesive contacts that provide tissues with unique barrier properties and play a prominent role in cell morphology, tissue physiology, and cell signaling. This review highlights advances made in understanding the contributions of galectin-3, a carbohydrate-binding protein with affinity toward β-galactosides, as a modulator of epithelial junction assembly and function. The interactions of galectin-3 within adhesive structures are discussed in relation to wound healing and tumor progression.
Collapse
Affiliation(s)
- Pablo Argüeso
- Schepens Eye Research Institute and Massachusetts Eye and Ear; Department of Ophthalmology ; Harvard Medical School; Boston, MA USA
| | - Jerome Mauris
- Schepens Eye Research Institute and Massachusetts Eye and Ear; Department of Ophthalmology ; Harvard Medical School; Boston, MA USA
| | - Yuichi Uchino
- Schepens Eye Research Institute and Massachusetts Eye and Ear; Department of Ophthalmology ; Harvard Medical School; Boston, MA USA
| |
Collapse
|
200
|
Gendronneau G, Sanii S, Dang T, Deshayes F, Delacour D, Pichard E, Advedissian T, Sidhu SS, Viguier M, Magnaldo T, Poirier F. Overexpression of galectin-7 in mouse epidermis leads to loss of cell junctions and defective skin repair. PLoS One 2015; 10:e0119031. [PMID: 25741714 PMCID: PMC4351092 DOI: 10.1371/journal.pone.0119031] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2014] [Accepted: 01/12/2015] [Indexed: 01/10/2023] Open
Abstract
BACKGROUND The proteins of the galectin family are implicated in many cellular processes, including cell interactions, polarity, intracellular trafficking, and signal transduction. In human and mouse, galectin-7 is almost exclusively expressed in stratified epithelia, notably in the epidermis. Galectin-7 expression is also altered in several human tumors of epithelial origin. This study aimed at dissecting the consequences of galectin-7 overexpression on epidermis structure and functions in vivo. METHODS We established transgenic mice specifically overexpressing galectin-7 in the basal epidermal keratinocytes and analyzed the consequences on untreated skin and after UVB irradiation or mechanical injury. RESULTS The intercellular cohesion of the epidermis is impaired in transgenic animals, with gaps developing between adjacent keratinocytes, associated with loss of adherens junctions. The epidermal architecture is aberrant with perturbations in the multilayered cellular organisation of the tissue, and structural defects in the basement membrane. These transgenic animals displayed a reduced re-epithelialisation potential following superficial wound, due to a defective collective migration of keratinocytes. Finally, a single mild dose of UVB induced an abnormal apoptotic response in the transgenic epidermis. CONCLUSION These results indicate that an excess of galectin-7 leads to a destabilisation of adherens junctions associated with defects in epidermal repair. As this phenotype shares similarities with that of galectin-7 null mutant mice, we conclude that a critical level of this protein is required for maintaining proper epidermal homeostasis. This study brings new insight into the mode of action of galectins in normal and pathological situations.
Collapse
Affiliation(s)
- Gaëlle Gendronneau
- Institut Jacques Monod, UMR CNRS 7592, Paris-Diderot University, Paris, France
| | - Sadaf Sanii
- Institut Jacques Monod, UMR CNRS 7592, Paris-Diderot University, Paris, France
| | - Tien Dang
- Institut Jacques Monod, UMR CNRS 7592, Paris-Diderot University, Paris, France
| | - Frédérique Deshayes
- Institut Jacques Monod, UMR CNRS 7592, Paris-Diderot University, Paris, France
| | - Delphine Delacour
- Institut Jacques Monod, UMR CNRS 7592, Paris-Diderot University, Paris, France
| | - Evelyne Pichard
- Institut Jacques Monod, UMR CNRS 7592, Paris-Diderot University, Paris, France
| | - Tamara Advedissian
- Institut Jacques Monod, UMR CNRS 7592, Paris-Diderot University, Paris, France
| | - Sukhvinder S. Sidhu
- Institut Jacques Monod, UMR CNRS 7592, Paris-Diderot University, Paris, France
| | - Mireille Viguier
- Institut Jacques Monod, UMR CNRS 7592, Paris-Diderot University, Paris, France
| | | | - Francoise Poirier
- Institut Jacques Monod, UMR CNRS 7592, Paris-Diderot University, Paris, France
- * E-mail:
| |
Collapse
|