1
|
Peiffer AL, Dugan AE, Kiessling LL. Soluble Human Lectins at the Host-Microbe Interface. Annu Rev Biochem 2024; 93:565-601. [PMID: 38640018 PMCID: PMC11296910 DOI: 10.1146/annurev-biochem-062917-012322] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/21/2024]
Abstract
Human lectins are integral to maintaining microbial homeostasis on the skin, in the blood, and at mucosal barriers. These proteins can recognize microbial glycans and inform the host about its microbial status. In accordance with their roles, their production can vary with tissue type. They also can have unique structural and biochemical properties, and they can influence microbial colonization at sites proximal and distal to their tissue of origin. In line with their classification as innate immune proteins, soluble lectins have long been studied in the context of acute infectious disease, but only recently have we begun to appreciate their roles in maintaining commensal microbial communities (i.e., the human microbiota). This review provides an overview of soluble lectins that operate at host-microbe interfaces, their glycan recognition properties, and their roles in physiological and pathological mechanisms.
Collapse
Affiliation(s)
- Amanda L Peiffer
- Department of Chemistry, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA;
| | - A E Dugan
- Department of Chemistry, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA;
| | - L L Kiessling
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
- Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
- Department of Chemistry, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA;
| |
Collapse
|
2
|
Tang X, Xiao Z, Chen M, Jin J, Yan C, Zhu X, Wang Z, Zhang D. A prototype galectin-1 (also known as galecin-2) from large yellow croaker (Larimichthys crocea): Molecular and function study. FISH & SHELLFISH IMMUNOLOGY 2024; 145:109314. [PMID: 38142827 DOI: 10.1016/j.fsi.2023.109314] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/07/2023] [Revised: 12/15/2023] [Accepted: 12/17/2023] [Indexed: 12/26/2023]
Abstract
Galectin-1 (also known as galecin-2), one member of galectins family, has multiple functions as a pattern recognition receptor (PRR) in innate immune defense system. In the present study, LcGal-1, a prototype galectin, was identified and function investigated in large yellow croaker (Larimichthys crocea). LcGal-1 consists of one carbohydrate recognition domain (CRD), which contains two carbohydrate binding motifs HFNPR and WG-E-R. LcGal-1 had a ubiquitous tissues profile with the highest and lowest expression in spleen and muscle, respectively. Moreover, it was in cytoplasm and nucleus of head-kidney cells in large yellow croaker. RT-qRCR showed that P. plecoglossicida induced LcGal-1 up-regulated expression in liver and gills, and the results were validated by immunohistochemistry analysis. Additionally, the recombinant LcGal-1 (rLcGal-1) showed agglutinate activity on erythrocytes, and the histidine (His) in the HFNPR motif was a key locus to the activity. The agglutination effect of rLcGal-1 on erythrocytes could be inhibited by LPS, α-lactase and d-galactose. The rLcGal-1 was able to bind and agglutinate Gram+ and Gram-bacteria, and damage bacterial membrane as confirmed by PI staining and SEM observation. Transcriptome analysis showed that the overexpressed LcGal-1 in HEK 293T cells could induce 176 DGEs, including 172 boosting genes and 4 falling genes. Collectively, LcGal-1 was a key immune gene involved in the recognition, conjunction, and elimination of pathogens in L. crocea, as well as multiple physiological and pathological regulatory processes.
Collapse
Affiliation(s)
- Xin Tang
- Key Laboratory of Healthy Mariculture for the East China Sea, Ministry of Agriculture and Rural Affairs, Jimei University, Xiamen, China
| | - Zhiqun Xiao
- Key Laboratory of Healthy Mariculture for the East China Sea, Ministry of Agriculture and Rural Affairs, Jimei University, Xiamen, China
| | - Meiling Chen
- Key Laboratory of Healthy Mariculture for the East China Sea, Ministry of Agriculture and Rural Affairs, Jimei University, Xiamen, China
| | - Jian Jin
- Key Laboratory of Healthy Mariculture for the East China Sea, Ministry of Agriculture and Rural Affairs, Jimei University, Xiamen, China
| | - Chunmei Yan
- Key Laboratory of Healthy Mariculture for the East China Sea, Ministry of Agriculture and Rural Affairs, Jimei University, Xiamen, China
| | - Xingcheng Zhu
- Key Laboratory of Healthy Mariculture for the East China Sea, Ministry of Agriculture and Rural Affairs, Jimei University, Xiamen, China
| | - Zhiyong Wang
- Key Laboratory of Healthy Mariculture for the East China Sea, Ministry of Agriculture and Rural Affairs, Jimei University, Xiamen, China; Laboratory for Marine Fisheries Science and Food Production Processes, Qingdao National Laboratory for Marine Science and Technology, Qingdao, China
| | - Dongling Zhang
- Key Laboratory of Healthy Mariculture for the East China Sea, Ministry of Agriculture and Rural Affairs, Jimei University, Xiamen, China.
| |
Collapse
|
3
|
Jiang X, Xiao X, Li H, Gong Y, Wang M, Yang H, Zhao L, Jiang Y, Wei Y, Zhao C, Li J, Chen Y, Feng S, Deng H, Ma S, Xu Y, Liu Y, Tsokos GC, Jiang M, Zhang X. Oxidized galectin-1 in SLE fails to bind the inhibitory receptor VSTM1 and increases reactive oxygen species levels in neutrophils. Cell Mol Immunol 2023; 20:1339-1351. [PMID: 37737309 PMCID: PMC10616122 DOI: 10.1038/s41423-023-01084-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2023] [Accepted: 09/01/2023] [Indexed: 09/23/2023] Open
Abstract
Inhibitory immune receptors set thresholds for immune cell activation, and their deficiency predisposes a person to autoimmune responses. However, the agonists of inhibitory immune receptors remain largely unknown, representing untapped sources of treatments for autoimmune diseases. Here, we show that V-set and transmembrane domain-containing 1 (VSTM1) is an inhibitory receptor and that its binding by the competent ligand soluble galectin-1 (Gal1) is essential for maintaining neutrophil viability mediated by downregulated reactive oxygen species production. However, in patients with systemic lupus erythematosus (SLE), circulating Gal1 is oxidized and cannot be recognized by VSTM1, leading to increased intracellular reactive oxygen species levels and reduced neutrophil viability. Dysregulated neutrophil function or death contributes significantly to the pathogenesis of SLE by providing danger molecules and autoantigens that drive the production of inflammatory cytokines and the activation of autoreactive lymphocytes. Interestingly, serum levels of glutathione, an antioxidant able to convert oxidized Gal1 to its reduced form, were negatively correlated with SLE disease activity. Taken together, our findings reveal failed inhibitory Gal1/VSTM1 pathway activation in patients with SLE and provide important insights for the development of effective targeted therapies.
Collapse
Affiliation(s)
- Xu Jiang
- Medical Research Center, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital; Department of Rheumatology, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing, China
| | - Xinyue Xiao
- Department of Rheumatology, Key Laboratory of Myositis, China-Japan Friendship Hospital, Beijing, China
| | - Hao Li
- Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Yiyi Gong
- Medical Research Center, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Beijing, China
| | - Min Wang
- Department of Rheumatology, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Clinical Immunology Center, Chinese Academy of Medical Sciences, Beijing, China
| | - Huaxia Yang
- Department of Rheumatology and Clinical Immunology, Peking Union Medical College Hospital, Clinical Immunology Center, Chinese Academy of Medical Sciences and Peking Union Medical College; The Ministry of Education Key Laboratory, Beijing, China
| | - Lidan Zhao
- Department of Rheumatology and Clinical Immunology, Peking Union Medical College Hospital, Clinical Immunology Center, Chinese Academy of Medical Sciences and Peking Union Medical College; The Ministry of Education Key Laboratory, Beijing, China
| | - Ying Jiang
- Department of Rheumatology, Xiangya Hospital, Central South University, Hunan, China
| | - Yanping Wei
- Medical Research Center, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Beijing, China
| | - Chongchong Zhao
- MOE Key Laboratory of Bioinformatics, Center for Synthetic & Systems Biology, School of Life Sciences, Tsinghua University, Beijing, China
| | - Jin Li
- MOE Key Laboratory of Bioinformatics, Center for Synthetic & Systems Biology, School of Life Sciences, Tsinghua University, Beijing, China
| | - Yuling Chen
- MOE Key Laboratory of Bioinformatics, Center for Synthetic & Systems Biology, School of Life Sciences, Tsinghua University, Beijing, China
| | - Shan Feng
- Key Laboratory of Structural Biology of Zhejiang Province, School of Life Sciences, Westlake University, Hangzhou, Zhejiang, China
| | - Haiteng Deng
- MOE Key Laboratory of Bioinformatics, Center for Synthetic & Systems Biology, School of Life Sciences, Tsinghua University, Beijing, China
| | - Shiliang Ma
- Department of Rheumatology, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Clinical Immunology Center, Chinese Academy of Medical Sciences, Beijing, China
| | - Yue Xu
- Department of Rheumatology, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Clinical Immunology Center, Chinese Academy of Medical Sciences, Beijing, China
| | - Yudong Liu
- Department of Rheumatology, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Clinical Immunology Center, Chinese Academy of Medical Sciences, Beijing, China
| | - George C Tsokos
- Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA.
| | - Minghong Jiang
- State Key Laboratory of Common Mechanism Research for Major Diseases, Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Beijing, China.
| | - Xuan Zhang
- Department of Rheumatology, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Clinical Immunology Center, Chinese Academy of Medical Sciences, Beijing, China.
| |
Collapse
|
4
|
Loh KWZ, Hu Z, Soong TW. Modulation of Ca V1.2 Channel Function by Interacting Proteins and Post-Translational Modifications: Implications in Cardiovascular Diseases and COVID-19. Handb Exp Pharmacol 2023. [PMID: 36764970 DOI: 10.1007/164_2023_636] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/12/2023]
Abstract
CaV1.2 calcium channel is the primary conduit for Ca2+ influx into cardiac and smooth muscles that underscores its importance in the pathogenesis of hypertension, atherosclerosis, myocardial infarction, and heart failure. But, a few controversies still remain. Therefore, exploring new ways to modulate CaV1.2 channel activity will augment the arsenal of CaV1.2 channel-based therapeutics for treatment of cardiovascular diseases. Here, we will mainly introduce a couple of emerging CaV1.2 channel interacting proteins, such as Galectin-1 and Cereblon, and discuss their roles in hypertension and heart failure through fine-tuning CaV1.2 channel activity. Of current interest, we will also evaluate the implication of the role of CaV1.2 channel in SARS-CoV-2 infection and the potential treatments of COVID-19-related cardiovascular symptoms.
Collapse
Affiliation(s)
- Kelvin Wei Zhern Loh
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore.,Cardiovascular Diseases Translational Research Programme, National University of Singapore, Singapore, Singapore
| | - Zhenyu Hu
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore.,Cardiovascular Diseases Translational Research Programme, National University of Singapore, Singapore, Singapore
| | - Tuck Wah Soong
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore. .,Cardiovascular Diseases Translational Research Programme, National University of Singapore, Singapore, Singapore. .,Healthy Longevity Translational Research Programme, National University of Singapore, Singapore, Singapore.
| |
Collapse
|
5
|
Abstract
The galectin family consists of carbohydrate (glycan) binding proteins that are expressed by a wide variety of cells and bind to galactose-containing glycans. Galectins can be located in the nucleus or the cytoplasm, or can be secreted into the extracellular space. They can modulate innate and adaptive immune cells by binding to glycans on the surface of immune cells or intracellularly via carbohydrate-dependent or carbohydrate-independent interactions. Galectins expressed by immune cells can also participate in host responses to infection by directly binding to microorganisms or by modulating antimicrobial functions such as autophagy. Here we explore the diverse ways in which galectins have been shown to impact immunity and discuss the opportunities and challenges in the field.
Collapse
|
6
|
Sylvén C, Wärdell E, Månsson-Broberg A, Cingolani E, Ampatzis K, Larsson L, Björklund Å, Giacomello S. High cardiomyocyte diversity in human early prenatal heart development. iScience 2022; 26:105857. [PMID: 36624836 PMCID: PMC9823232 DOI: 10.1016/j.isci.2022.105857] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2022] [Revised: 07/19/2022] [Accepted: 12/18/2022] [Indexed: 12/24/2022] Open
Abstract
Cardiomyocytes play key roles during cardiogenesis, but have poorly understood features, especially in prenatal stages. Here, we characterized human prenatal cardiomyocytes, 6.5-7 weeks post-conception, by integrating single-cell RNA sequencing, spatial transcriptomics, and ligand-receptor interaction information. Using a computational workflow developed to dissect cell type heterogeneity, localize cell types, and explore their molecular interactions, we identified eight types of developing cardiomyocyte, more than double compared to the ones identified in the Human Developmental Cell Atlas. These have high variability in cell cycle activity, mitochondrial content, and connexin gene expression, and are differentially distributed in the ventricles, including outflow tract, and atria, including sinoatrial node. Moreover, cardiomyocyte ligand-receptor crosstalk is mainly with non-cardiomyocyte cell types, encompassing cardiogenesis-related pathways. Thus, early prenatal human cardiomyocytes are highly heterogeneous and develop unique location-dependent properties, with complex ligand-receptor crosstalk. Further elucidation of their developmental dynamics may give rise to new therapies.
Collapse
Affiliation(s)
- Christer Sylvén
- Department of Medicine, Karolinska Institute, Huddinge, Sweden,Corresponding author
| | - Eva Wärdell
- Department of Medicine, Karolinska Institute, Huddinge, Sweden
| | | | | | | | - Ludvig Larsson
- Science for Life Laboratory, Department of Gene Technology, KTH Royal Institute of Technology, Stockholm, Sweden
| | - Åsa Björklund
- Department of Cell and Molecular Biology, National Bioinformatics Infrastructure Sweden, Science for Life Laboratory, Uppsala University, Uppsala, Sweden
| | - Stefania Giacomello
- Science for Life Laboratory, Department of Gene Technology, KTH Royal Institute of Technology, Stockholm, Sweden,Corresponding author
| |
Collapse
|
7
|
Mansour AA, Krautter F, Zhi Z, Iqbal AJ, Recio C. The interplay of galectins-1, -3, and -9 in the immune-inflammatory response underlying cardiovascular and metabolic disease. Cardiovasc Diabetol 2022; 21:253. [PMID: 36403025 PMCID: PMC9675972 DOI: 10.1186/s12933-022-01690-7] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/19/2022] [Accepted: 11/08/2022] [Indexed: 11/21/2022] Open
Abstract
Galectins are β-galactoside-binding proteins that bind and crosslink molecules via their sugar moieties, forming signaling and adhesion networks involved in cellular communication, differentiation, migration, and survival. Galectins are expressed ubiquitously across immune cells, and their function varies with their tissue-specific and subcellular location. Particularly galectin-1, -3, and -9 are highly expressed by inflammatory cells and are involved in the modulation of several innate and adaptive immune responses. Modulation in the expression of these proteins accompany major processes in cardiovascular diseases and metabolic disorders, such as atherosclerosis, thrombosis, obesity, and diabetes, making them attractive therapeutic targets. In this review we consider the broad cellular activities ascribed to galectin-1, -3, and -9, highlighting those linked to the progression of different inflammatory driven pathologies in the context of cardiovascular and metabolic disease, to better understand their mechanism of action and provide new insights into the design of novel therapeutic strategies.
Collapse
Affiliation(s)
- Adel Abo Mansour
- Institute of Cardiovascular Sciences (ICVS), College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, King Khalid University, Abha, Saudi Arabia
| | - Franziska Krautter
- Institute of Cardiovascular Sciences (ICVS), College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK
| | - Zhaogong Zhi
- Institute of Cardiovascular Sciences (ICVS), College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK
| | - Asif Jilani Iqbal
- Institute of Cardiovascular Sciences (ICVS), College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK.
| | - Carlota Recio
- Instituto Universitario de Investigaciones Biomédicas y Sanitarias (IUIBS), Farmacología Molecular y Traslacional -BIOPharm, Universidad de Las Palmas de Gran Canaria, Las Palmas de Gran Canaria, Las Palmas, Spain.
| |
Collapse
|
8
|
Shang M, Hu Y, Cao H, Lin Q, Yi N, Zhang J, Gu Y, Yang Y, He S, Lu M, Peng L, Li L. Concordant and Heterogeneity of Single-Cell Transcriptome in Cardiac Development of Human and Mouse. Front Genet 2022; 13:892766. [PMID: 35832197 PMCID: PMC9271823 DOI: 10.3389/fgene.2022.892766] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2022] [Accepted: 05/16/2022] [Indexed: 11/28/2022] Open
Abstract
Normal heart development is vital for maintaining its function, and the development process is involved in complex interactions between different cell lineages. How mammalian hearts develop differently is still not fully understood. In this study, we identified several major types of cardiac cells, including cardiomyocytes (CMs), fibroblasts (FBs), endothelial cells (ECs), ECs/FBs, epicardial cells (EPs), and immune cells (macrophage/monocyte cluster, MACs/MONOs), based on single-cell transcriptome data from embryonic hearts of both human and mouse. Then, species-shared and species-specific marker genes were determined in the same cell type between the two species, and the genes with consistent and different expression patterns were also selected by constructing the developmental trajectories. Through a comparison of the development stage similarity of CMs, FBs, and ECs/FBs between humans and mice, it is revealed that CMs at e9.5 and e10.5 of mice are most similar to those of humans at 7 W and 9 W, respectively. Mouse FBs at e10.5, e13.5, and e14.5 are correspondingly more like the same human cells at 6, 7, and 9 W. Moreover, the e9.5-ECs/FBs of mice are most similar to that of humans at 10W. These results provide a resource for understudying cardiac cell types and the crucial markers able to trace developmental trajectories among the species, which is beneficial for finding suitable mouse models to detect human cardiac physiology and related diseases.
Collapse
Affiliation(s)
- Mengyue Shang
- Key Laboratory of Arrhythmias, Ministry of Education of China, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
- Heart Health Center, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
- Institute of Medical Genetics, Tongji University, Shanghai, China
| | - Yi Hu
- Key Laboratory of Arrhythmias, Ministry of Education of China, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
- Heart Health Center, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
- Institute of Medical Genetics, Tongji University, Shanghai, China
| | - Huaming Cao
- Department of Cardiology, Shanghai Shibei Hospital, Shanghai, China
| | - Qin Lin
- Key Laboratory of Arrhythmias, Ministry of Education of China, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
- Heart Health Center, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
- Institute of Medical Genetics, Tongji University, Shanghai, China
| | - Na Yi
- Key Laboratory of Arrhythmias, Ministry of Education of China, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
- Heart Health Center, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
- Institute of Medical Genetics, Tongji University, Shanghai, China
| | - Junfang Zhang
- Institute of Medical Genetics, Tongji University, Shanghai, China
| | - Yanqiong Gu
- Institute of Medical Genetics, Tongji University, Shanghai, China
| | - Yujie Yang
- Institute of Medical Genetics, Tongji University, Shanghai, China
| | - Siyu He
- Key Laboratory of Arrhythmias, Ministry of Education of China, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
- Heart Health Center, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
- Institute of Medical Genetics, Tongji University, Shanghai, China
| | - Min Lu
- Key Laboratory of Arrhythmias, Ministry of Education of China, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Luying Peng
- Key Laboratory of Arrhythmias, Ministry of Education of China, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
- Heart Health Center, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
- Institute of Medical Genetics, Tongji University, Shanghai, China
- Department of Medical Genetics, Tongji University School of Medicine, Shanghai, China
- Research Units of Origin and Regulation of Heart Rhythm, Chinese Academy of Medical Sciences, Beijing, China
- *Correspondence: Luying Peng, ; Li Li,
| | - Li Li
- Key Laboratory of Arrhythmias, Ministry of Education of China, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
- Heart Health Center, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
- Institute of Medical Genetics, Tongji University, Shanghai, China
- Department of Medical Genetics, Tongji University School of Medicine, Shanghai, China
- Research Units of Origin and Regulation of Heart Rhythm, Chinese Academy of Medical Sciences, Beijing, China
- *Correspondence: Luying Peng, ; Li Li,
| |
Collapse
|
9
|
Stowell SR, Rodrigues LC, Dias-Baruffi M, Cummings RD, Arthur CM. Examining Galectin Binding Specificity Using Glycan Microarrays. METHODS IN MOLECULAR BIOLOGY (CLIFTON, N.J.) 2022; 2442:151-168. [PMID: 35320525 DOI: 10.1007/978-1-0716-2055-7_9] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
Glycan binding proteins (GBPs) possess the unique ability to regulate a wide variety of biological processes through interactions with highly modifiable cell surface glycans. While many studies demonstrate the impact of glycan modification on GBP recognition and activity, the relative contribution of subtle changes in glycan structure on GBP binding can be difficult to define. To overcome limitations in the analysis of GBP-glycan interactions, recent studies utilized glycan microarray platforms containing hundreds of structurally defined glycans. These studies not only provided important information regarding GBP-glycan interactions in general but have also resulted in significant insight into binding specificity and biological activity of the galectin family. We will describe the methods used when employing glycan microarray platforms to examine galectin-glycan binding specificity and function.
Collapse
Affiliation(s)
- Sean R Stowell
- Joint Program in Transfusion Medicine, Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA.,Harvard Glycomics Center, Harvard Medical School, Boston, MA, USA
| | - Lilian C Rodrigues
- Department of Clinical Analysis, Toxicological and Bromatological, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Ribeirão Preto, Brazil
| | - Marcelo Dias-Baruffi
- Department of Clinical Analysis, Toxicological and Bromatological, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Ribeirão Preto, Brazil
| | | | - Connie M Arthur
- Joint Program in Transfusion Medicine, Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA. .,Harvard Glycomics Center, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
10
|
Cerri DG, Rodrigues LC, Fermino ML, Papoti M, Cummings RD, Stowell SR, Dias-Baruffi M. Investigation of Galectins in Frozen Tissue and Mammalian Cell Culture Using Confocal Miccroscopy. Methods Mol Biol 2022; 2442:289-306. [PMID: 35320532 DOI: 10.1007/978-1-0716-2055-7_16] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Galectins are multifunctional glycan-binding proteins present in various tissues that participate in multiple physiological and pathological processes and are considered as not only biomarkers of human diseases but also molecular targets for treating cancer and inflammatory illnesses in many organs. In the glycobiology field, it is crucial to determine the pattern of galectin expression and location in cells and tissues. Confocal microscopy is a powerful imaging technology that represents a unique approach to investigate the expression and location of biomolecules in various tissues and cells. The confocal microscope acquires images of the specimen through the reflected or fluorescent light from the objective's focal plane, using laser light focused on a small spot inside the tissue or cell. This technique provides high-resolution and high-contrast images without artifacts generated by conventional microscopy and enables reconstruction of virtual tridimensional images by acquiring multiple sections from several focal planes, which makes it possible to obtain the precise spatial location of any cellular structure or molecule. Furthermore, confocal microscopy is a non-invasive tissue imaging strategy used in clinical practices. We describe herein the immunofluorescence confocal method for examining galectins in frozen tissue sections and mammalian cell culture.
Collapse
Affiliation(s)
- Daniel Giuliano Cerri
- Departamento de Análises Clínicas, Toxicológicas e Bromatológicas da Faculdade de Ciências Farmacênuticas de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, São Paulo, Brasil
| | - Lilian Cataldi Rodrigues
- Departamento de Análises Clínicas, Toxicológicas e Bromatológicas da Faculdade de Ciências Farmacênuticas de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, São Paulo, Brasil
| | - Marise Lopes Fermino
- Departamento de Análises Clínicas, Toxicológicas e Bromatológicas da Faculdade de Ciências Farmacênuticas de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, São Paulo, Brasil
| | - Marcelo Papoti
- Escola de Educação Física e Esporte de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, São Paulo, Brasil
| | - Richard D Cummings
- Department of Surgery, National Center for Functional Glycomics, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Sean R Stowell
- Department of Pathology, Harvard Medical School, Brigham and Women's Hospital, Boston, MA, USA
| | - Marcelo Dias-Baruffi
- Departamento de Análises Clínicas, Toxicológicas e Bromatológicas da Faculdade de Ciências Farmacênuticas de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, São Paulo, Brasil.
| |
Collapse
|
11
|
Stowell SR, Dias-Baruffi M, Cummings RD, Arthur CM. Detection of Phosphatidylserine Exposure on Leukocytes Following Treatment with Human Galectins. Methods Mol Biol 2022; 2442:533-548. [PMID: 35320544 DOI: 10.1007/978-1-0716-2055-7_28] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Cellular turnover represents a fundamental aspect of immunological homeostasis. While many factors appear to regulate leukocyte removal during inflammatory resolution, recent studies suggest that members of the galectin family play a unique role in orchestrating this process. Unlike cellular removal through apoptotic cell death, several members of the galectin family induce surface expression of phosphatidylserine (PS), a phagocytic marker on cells undergoing apoptosis, in the absence of cell death. However, similar to PS on cells undergoing apoptosis, galectin-induced PS exposure sensitizes cells to phagocytic removal. As galectins appear to prepare cells for phagocytic removal without actually inducing apoptotic cell death, this process has recently been coined preaparesis. Given the unique characteristics of galectin-induced PS exposure in the context of preaparesis, we will examine unique considerations when evaluating the potential impact of different galectin family members on PS exposure and cell viability.
Collapse
Affiliation(s)
- Sean R Stowell
- Joint Program in Transfusion Medicine, Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
- Harvard Glycomics Center, Harvard Medical School, Boston, MA, USA
| | - Marcelo Dias-Baruffi
- Department of Clinical Analysis, Toxicological and Bromatological, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Ribeirão Preto, Brazil
| | | | - Connie M Arthur
- Joint Program in Transfusion Medicine, Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA.
- Harvard Glycomics Center, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
12
|
Wu SC, Paul A, Cummings RD, Feasley CL, Arthur CM, Stowell SR. Alkylation of Galectin-1 with Iodoacetamide and Mass Spectrometric Mapping of the Sites of Incorporation. Methods Mol Biol 2022; 2442:75-87. [PMID: 35320520 DOI: 10.1007/978-1-0716-2055-7_4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Galectins can display unique sensitivity to oxidative changes that result in significant conformational alterations that prevent carbohydrate recognition. While a variety of approaches can be utilized to prevent galectin oxidation, several of these require inclusion of reducing agents that not only prevent galectins from undergoing oxidative inactivation but can also interfere with normal redox potentials required for fundamental cellular processes. To overcome the limitations associated with placing cells in an artificial reducing environment, cysteine residues on galectins can be directly alkylated with iodoacetamide to form a stable thioether adduct that is resistant to further modification. Iodoacetamide alkylated galectin remains stable over prolonged periods of time and retains the carbohydrate binding and biological activities of the protein. As a result, this approach allows examination of the biological roles of a stabilized form of galectin-1 without introducing the confounding variables that can occur when typical soluble reducing agents are employed.
Collapse
Affiliation(s)
- Shang-Chuen Wu
- Joint Program in Transfusion Medicine, Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Anu Paul
- Joint Program in Transfusion Medicine, Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | | | | | - Connie M Arthur
- Joint Program in Transfusion Medicine, Brigham and Women's Hospital, National Center for Functional Glycomics, Harvard Medical School, Boston, MA, USA.
| | - Sean R Stowell
- Harvard Glycomics Center, Harvard Medical School, Boston, MA, USA.
- Joint Program in Transfusion Medicine, Department of Pathology, Brigham and Women's Hospital, National Center for Functional Glycomics, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
13
|
Galectin-1 is associated with the severity of coronary artery disease and adverse cardiovascular events in patients undergoing coronary angiography. Sci Rep 2020; 10:20683. [PMID: 33244142 PMCID: PMC7692553 DOI: 10.1038/s41598-020-77804-6] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2020] [Accepted: 11/02/2020] [Indexed: 12/02/2022] Open
Abstract
Galectin-1, a β-galactoside-binding lectin mediating inflammation and neovascularization, is reported to attenuate ventricular remodeling after myocardial infarction. But its role in stable coronary artery disease (CAD) has not been fully elucidated. This study aimed to identify the relationship between the circulating galectin-1 level and the severity of CAD in patients with suspected CAD. Pre-procedure galectin-1 and high-sensitivity C-reactive protein (hs-CRP) concentrations were measured in 834 subjects who underwent scheduled coronary angiography. Subjects were grouped into tertiles of the galectin-1 levels. SYNTAX scores were calculated to evaluate the severity of CAD. All patients were followed until January 2019 or the occurrence of major adverse cardiovascular events (MACE). Patients with higher galectin-1 concentrations were older; had greater prevalence of hypertension, diabetes, chronic kidney disease, and heart failure; and were more likely to present with higher hs-CRP levels and SYNTAX scores. During the follow-up period of 1.3 ± 1.1 years, patients in the highest tertile of galectin-1 were associated with a greater risk of MACE after adjustment for age, sex, comorbidities, co-medications, serum levels of hemoglobin, creatinine, hs-CRP, ejection fraction, SYNTAX scores, and revascularization modalities (adjusted hazard ratio 10.95, 95% confidence interval 2.29–52.47, p = 0.003). Galectin-1 showed better discriminatory performance than hs-CRP, and non-inferior performance to SYNTAX scores, in predicting the incidence of MACE.
Collapse
|
14
|
Tazhitdinova R, Timoshenko AV. The Emerging Role of Galectins and O-GlcNAc Homeostasis in Processes of Cellular Differentiation. Cells 2020; 9:cells9081792. [PMID: 32731422 PMCID: PMC7465113 DOI: 10.3390/cells9081792] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2020] [Revised: 07/24/2020] [Accepted: 07/24/2020] [Indexed: 02/07/2023] Open
Abstract
Galectins are a family of soluble β-galactoside-binding proteins with diverse glycan-dependent and glycan-independent functions outside and inside the cell. Human cells express twelve out of sixteen recognized mammalian galectin genes and their expression profiles are very different between cell types and tissues. In this review, we summarize the current knowledge on the changes in the expression of individual galectins at mRNA and protein levels in different types of differentiating cells and the effects of recombinant galectins on cellular differentiation. A new model of galectin regulation is proposed considering the change in O-GlcNAc homeostasis between progenitor/stem cells and mature differentiated cells. The recognition of galectins as regulatory factors controlling cell differentiation and self-renewal is essential for developmental and cancer biology to develop innovative strategies for prevention and targeted treatment of proliferative diseases, tissue regeneration, and stem-cell therapy.
Collapse
|
15
|
Liu Y, Zhou X, Hu N, Wang C, Zhao L. P311 regulates distal lung development via its interaction with several binding proteins. Mech Dev 2020; 163:103633. [PMID: 32682987 DOI: 10.1016/j.mod.2020.103633] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2020] [Revised: 05/27/2020] [Accepted: 07/03/2020] [Indexed: 01/17/2023]
Abstract
Little is known about the molecular mechanisms underlying alveolar development. P311, a putative neuronal protein originally identified for its high expression during neuronal development, has once been reported to play a potential role in distal lung generation. However, the function of this protein has been poorly understood so far. Hence, we carried out a yeast two-hybrid screen, combining with other protein-protein interaction experiments, to isolate several binding partners of P311 during lung development, which may help us explore its function. We report 7 proteins here, including Gal-1, Loxl-1 and SPARC, etc, that can interact with it. Most of them have similar spatio-temporal expression patterns to P311. In addition, it was also found that P311 could stimulate their expression indirectly in L929 mouse fibroblast. Besides, computational methods were applied to construct a P311 centered protein-protein interaction network during alveolarization, using the 7 binding partners and their protein interaction information provided by public data resources. By analyzing the structure and function of this network, the effects of P311 on lung development were further clarified and all of the bioinformatic predictions from the network could be validated by real experiments. We have found here that P311 can control lung redox events, extracellular matrix and cell cycle progression, which are all crucial to pulmonary morphogenesis. This gives us a novel thought to explore the mechanisms controlling alveolarization.
Collapse
Affiliation(s)
- Yu Liu
- Department of Biochemistry and Molecular Biology, College of Life Sciences, Nankai University, Tianjin 300071, China; Department of Computational Medicine and Bioinformatics, Medical School, University of Michigan, Ann Arbor, MI 48109, USA
| | - Xiaohai Zhou
- Department of Biochemistry and Molecular Biology, College of Life Sciences, Nankai University, Tianjin 300071, China
| | - Naiyue Hu
- Department of Biochemistry and Molecular Biology, College of Life Sciences, Nankai University, Tianjin 300071, China
| | - Chunyan Wang
- Department of Biochemistry and Molecular Biology, College of Life Sciences, Nankai University, Tianjin 300071, China
| | - Liqing Zhao
- Department of Biochemistry and Molecular Biology, College of Life Sciences, Nankai University, Tianjin 300071, China.
| |
Collapse
|
16
|
Zhang T, van Die I, Tefsen B, van Vliet SJ, Laan LC, Zhang J, Ten Dijke P, Wuhrer M, Belo AI. Differential O- and Glycosphingolipid Glycosylation in Human Pancreatic Adenocarcinoma Cells With Opposite Morphology and Metastatic Behavior. Front Oncol 2020; 10:732. [PMID: 32582529 PMCID: PMC7280451 DOI: 10.3389/fonc.2020.00732] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2019] [Accepted: 04/16/2020] [Indexed: 01/15/2023] Open
Abstract
Changes in the glycosylation profile of cancer cells have been strongly associated with cancer progression. To increase our insights into the role of glycosylation in human pancreatic ductal adenocarcinoma (PDAC), we performed a study on O-glycans and glycosphingolipid (GSL) glycans of the PDAC cell lines Pa-Tu-8988T (PaTu-T) and Pa-Tu-8988S (PaTu-S). These cell lines are derived from the same patient, but show an almost opposite phenotype, morphology and capacity to metastasize, and may thus provide an attractive model to study the role of glycosylation in progression of PDAC. Gene-array analysis revealed that 24% of the glycosylation-related genes showed a ≥ 1.5-fold difference in expression level between the two cell lines. Subsequent validation of the data by porous graphitized carbon nano-liquid chromatography coupled to a tandem ion trap mass spectrometry and flow cytometry established major differences in O-glycans and GSL-glycans between the cell lines, including lower levels of T and sialylated Tn (sTn) antigens, neoexpression of globosides (Gb3 and Gb4), and higher levels of gangliosides in the mesenchymal-like PaTu-T cells compared to the epithelial-like PaTu-S. In addition, PaTu-S cells demonstrated a significantly higher binding of the immune-lectins macrophage galactose-type lectin and galectin-4 compared to PaTu-T. In summary, our data provide a comprehensive and differential glycan profile of two PDAC cell lines with disparate phenotypes and metastatic behavior. This will allow approaches to modulate and monitor the glycosylation of these PDAC cell lines, which opens up avenues to study the biology and metastatic behavior of PDAC.
Collapse
Affiliation(s)
- Tao Zhang
- Center for Proteomics and Metabolomics, Leiden University Medical Center, Leiden, Netherlands
| | - Irma van Die
- Department of Molecular Cell Biology and Immunology, Cancer Center Amsterdam, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, Netherlands
| | - Boris Tefsen
- Department of Molecular Cell Biology and Immunology, Cancer Center Amsterdam, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, Netherlands.,Department of Biological Sciences, Xi'an Jiaotong-Liverpool University, Suzhou, China
| | - Sandra J van Vliet
- Department of Molecular Cell Biology and Immunology, Cancer Center Amsterdam, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, Netherlands
| | - Lisa C Laan
- Department of Molecular Cell Biology and Immunology, Cancer Center Amsterdam, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, Netherlands
| | - Jing Zhang
- Department of Cell and Chemical Biology and Oncode Institute, Leiden University Medical Center, Leiden, Netherlands
| | - Peter Ten Dijke
- Department of Cell and Chemical Biology and Oncode Institute, Leiden University Medical Center, Leiden, Netherlands
| | - Manfred Wuhrer
- Center for Proteomics and Metabolomics, Leiden University Medical Center, Leiden, Netherlands
| | - Ana I Belo
- Department of Molecular Cell Biology and Immunology, Cancer Center Amsterdam, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, Netherlands
| |
Collapse
|
17
|
Fanfone D, Stanicki D, Nonclercq D, Port M, Vander Elst L, Laurent S, Muller RN, Saussez S, Burtea C. Molecular Imaging of Galectin-1 Expression as a Biomarker of Papillary Thyroid Cancer by Using Peptide-Functionalized Imaging Probes. BIOLOGY 2020; 9:biology9030053. [PMID: 32183292 PMCID: PMC7150867 DOI: 10.3390/biology9030053] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/20/2020] [Revised: 03/03/2020] [Accepted: 03/09/2020] [Indexed: 01/11/2023]
Abstract
Thyroid cancers are the most frequent endocrine cancers and their incidence is increasing worldwide. Thyroid nodules occur in over 19–68% of the population, but only 7–15% of them are diagnosed as malignant. Diagnosis relies on a fine needle aspiration biopsy, which is often inconclusive and about 90% of thyroidectomies are performed for benign lesions. Galectin-1 has been proposed as a confident biomarker for the discrimination of malignant from benign nodules. We previously identified by phage display two peptides (P1 and P7) targeting galectin-1, with the goal of developing imaging probes for non-invasive diagnosis of thyroid cancer. The peptides were coupled to ultra-small superparamagnetic particles of iron oxide (USPIO) or to a near-infrared dye (CF770) for non-invasive detection of galectin-1 expression in a mouse model of papillary thyroid cancer (PTC, as the most frequent one) by magnetic resonance imaging and fluorescence lifetime imaging. The imaging probes functionalized with the two peptides presented comparable image enhancement characteristics. However, those coupled to P7 were more favorable, and showed decreased retention by the liver and spleen (known for their galectin-1 expression) and high sensitivity (75%) and specificity (100%) of PTC detection, which confirm the aptitude of this peptide to discriminate human malignant from benign nodules (80% sensitivity, 100% specificity) previously observed by immunohistochemistry.
Collapse
Affiliation(s)
- Deborah Fanfone
- Department of General, Organic and Biomedical Chemistry, UMONS, Avenue Victor Maistriau 19, 7000 Mons, Belgium; (D.F.); (L.V.E.); (S.L.); (R.N.M.)
| | - Dimitri Stanicki
- Center for Microscopy and Molecular Imaging, Rue Adrienne Bolland, 8, 6041 Charleroi, Belgium;
| | - Denis Nonclercq
- Laboratory of Histology, Faculty of Medicine and Pharmacy, University of Mons–UMONS, Avenue du Champ de Mars 6, 7000 Mons, Belgium;
| | - Marc Port
- Laboratoire de Génomique, Bioinformatique et Chimie Moléculaire (EA 7528), Equipe Chimie Moléculaire, Conservatoire National des Arts et Métiers (CNAM), HESAM Université, 75003 Paris, France;
| | - Luce Vander Elst
- Department of General, Organic and Biomedical Chemistry, UMONS, Avenue Victor Maistriau 19, 7000 Mons, Belgium; (D.F.); (L.V.E.); (S.L.); (R.N.M.)
| | - Sophie Laurent
- Department of General, Organic and Biomedical Chemistry, UMONS, Avenue Victor Maistriau 19, 7000 Mons, Belgium; (D.F.); (L.V.E.); (S.L.); (R.N.M.)
- Center for Microscopy and Molecular Imaging, Rue Adrienne Bolland, 8, 6041 Charleroi, Belgium;
| | - Robert N. Muller
- Department of General, Organic and Biomedical Chemistry, UMONS, Avenue Victor Maistriau 19, 7000 Mons, Belgium; (D.F.); (L.V.E.); (S.L.); (R.N.M.)
- Center for Microscopy and Molecular Imaging, Rue Adrienne Bolland, 8, 6041 Charleroi, Belgium;
| | - Sven Saussez
- Laboratory of Human Anatomy and Experimental Oncology, UMONS, Avenue du Champ de Mars, 6, 7000 Mons, Belgium;
| | - Carmen Burtea
- Department of General, Organic and Biomedical Chemistry, UMONS, Avenue Victor Maistriau 19, 7000 Mons, Belgium; (D.F.); (L.V.E.); (S.L.); (R.N.M.)
- Correspondence: ; Tel.: +32-6537-3814
| |
Collapse
|
18
|
Wang N, Dang M, Zhang W, Lei Y, Liu Z. Galectin-3 is associated with severe heart failure and death: A hospital-based study in Chinese patients. Scand J Immunol 2020; 91:e12826. [PMID: 31514240 DOI: 10.1111/sji.12826] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2019] [Revised: 09/01/2019] [Accepted: 09/04/2019] [Indexed: 12/20/2022]
Abstract
Heart failure (HF) is a serious disease syndrome characterized by elevated pro-inflammatory cytokines and inflammatory mediators presume to have significant contribution on disease progression. Galectins are carbohydrate-binding proteins responsible of various physiological functions. Role of galectins in heart failure has been ill-defined. In the present case-controls study, 136 patients clinically diagnosed with heart failure and 125 healthy Chinese controls were recruited. Levels of galectins (Gal-1, 3 and 9) and cytokines (IFN-γ, IL-17A, IL-4 and TGF-β) were quantified by ELISA. Increased levels of galectin-1 and 3 was observed in HF patients and associated with clinical severity. In addition, pro-inflammatory cytokines such as IFN-γ and IL-17A were increased in patients whereas, anti-inflammatory TGFβ was decreased. Galectin-3 was positively correlated with IFN-γ, IL-17A and inversely with TGF-β. Furthermore, ROC curve analysis suggested galectin-3 as a promising biomarker for diagnosis and HF and clinical severity. Interestingly, a two-year follow-up indicated significant association of elevated galectin-3 with mortality due to HF. In conclusion, galectin-3 associated with HF and clinical manifestations possibly by inducing pro-inflammatory cytokines and could be a possible biomarker of HF and severe clinical conditions.
Collapse
Affiliation(s)
- Na Wang
- Department of Cardiovascular, Zhoukou Central Hospital, Zhoukou City, China
| | - Minyan Dang
- Innoscience Research Sdn Bhd, Subang Jaya, Malaysia
| | - Wenzhi Zhang
- Innoscience Research Sdn Bhd, Subang Jaya, Malaysia
| | - Yan Lei
- Innoscience Research Sdn Bhd, Subang Jaya, Malaysia
| | - Zhaochuan Liu
- Department of Cardiology, Army No.82 Group Military Hospital, Baoding, China
| |
Collapse
|
19
|
Rodrigues LC, Kabeya LM, Azzolini AECS, Cerri DG, Stowell SR, Cummings RD, Lucisano-Valim YM, Dias-Baruffi M. Galectin-1 modulation of neutrophil reactive oxygen species production depends on the cell activation state. Mol Immunol 2019; 116:80-89. [PMID: 31630079 DOI: 10.1016/j.molimm.2019.10.001] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2018] [Revised: 04/01/2019] [Accepted: 10/01/2019] [Indexed: 11/18/2022]
Abstract
Here we report the effects of exogenous and endogenous galectin-1 (Gal-1) in modulating the functional responses of human and murine neutrophils at different stages of activation, i.e. naive, primed, and activated. Exposure to Gal-1 did not induce ROS production in either naive or N-formyl-methionyl-leucyl-phenylalanine-primed (fMLP; 10-9 M) neutrophils. However, Gal-1 elicited a concentration-dependent ROS production in neutrophils activated with fMLP at concentrations ranging from 10-8 M to 10-6 M. Additional fMLP (10-7 M) stimulation of fMLP-activated neutrophils increased ROS production, whose intensity was inversely related to the fMLP concentration used in the first activation step (10-8 M to 10-6 M), and was not influenced by the presence of Gal-1. Naive neutrophils treated with Gal-1 and then exposed to fMLP (10-6 M) or phorbol-12-myristate-13-acetate (10-7 M) produced less ROS, as compared to naive neutrophils not treated with Gal-1. Interestingly, these in vitro Gal-1 effects were associated with Gal-1 carbohydrate-binding activity and the ability to decrease FPR-1 (formyl peptide receptor 1) expression in naive human neutrophils. Conversely, positive ROS modulation by Gal-1 in activated neutrophils was not associated with FPR-1 expression but it was related to its carbohydrate recognition. In vitro, fMLP stimulation of Gal-1-/- mouse neutrophils produced more ROS than fMLP stimulation of Gal-1+/+ neutrophils and this effect may be associated with increased FPR-1 expression. Exogenous Gal-1 induced ROS production in Gal-1-/- mouse neutrophils more effectively than in Gal-1+/+ mouse neutrophils. Compared to Gal-1+/+ mice, Gal-1-/- mice exhibited lower bacterial load in the peritoneal fluid and peripheral blood, thus indicating a greater bactericidal activity in vivo. These findings demonstrate that endogenous Gal-1 restricts ROS generation that correlates with bacterial killing capacity in inflammatory neutrophils. Thus, endogenous and exogenous Gal-1 may either positively or negatively modulate the effector functions of neutrophils according to the cell activation stage.
Collapse
Affiliation(s)
- Lílian C Rodrigues
- Department of Clinical Analyses, Toxicology and Food Sciences, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Ribeirão Preto, SP, Brazil
| | - Luciana M Kabeya
- Department of Physics and Chemistry, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Ribeirão Preto, SP, Brazil
| | - Ana Elisa C S Azzolini
- Department of Physics and Chemistry, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Ribeirão Preto, SP, Brazil
| | - Daniel Giuliano Cerri
- Department of Clinical Analyses, Toxicology and Food Sciences, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Ribeirão Preto, SP, Brazil
| | - Sean R Stowell
- Pathology Department, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Richard D Cummings
- Department of Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, 3 Blackfan Circle, Room 11087, Boston, MA, 02115, USA
| | - Yara Maria Lucisano-Valim
- Department of Physics and Chemistry, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Ribeirão Preto, SP, Brazil
| | - Marcelo Dias-Baruffi
- Department of Clinical Analyses, Toxicology and Food Sciences, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Ribeirão Preto, SP, Brazil.
| |
Collapse
|
20
|
Robinson BS, Arthur CM, Evavold B, Roback E, Kamili NA, Stowell CS, Vallecillo-Zúniga ML, Van Ry PM, Dias-Baruffi M, Cummings RD, Stowell SR. The Sweet-Side of Leukocytes: Galectins as Master Regulators of Neutrophil Function. Front Immunol 2019; 10:1762. [PMID: 31440233 PMCID: PMC6693361 DOI: 10.3389/fimmu.2019.01762] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2019] [Accepted: 07/11/2019] [Indexed: 12/13/2022] Open
Abstract
Among responders to microbial invasion, neutrophils represent one of the earliest and perhaps most important factors that contribute to initial host defense. Effective neutrophil immunity requires their rapid mobilization to the site of infection, which requires efficient extravasation, activation, chemotaxis, phagocytosis, and eventual killing of potential microbial pathogens. Following pathogen elimination, neutrophils must be eliminated to prevent additional host injury and subsequent exacerbation of the inflammatory response. Galectins, expressed in nearly every tissue and regulated by unique sensitivity to oxidative and proteolytic inactivation, appear to influence nearly every aspect of neutrophil function. In this review, we will examine the impact of galectins on neutrophils, with a particular focus on the unique biochemical traits that allow galectin family members to spatially and temporally regulate neutrophil function.
Collapse
Affiliation(s)
- Brian S Robinson
- Department of Laboratory Medicine and Pathology, Center for Transfusion Medicine and Cellular Therapies, Emory University School of Medicine, Atlanta, GA, United States
| | - Connie M Arthur
- Department of Laboratory Medicine and Pathology, Center for Transfusion Medicine and Cellular Therapies, Emory University School of Medicine, Atlanta, GA, United States
| | - Birk Evavold
- Department of Laboratory Medicine and Pathology, Center for Transfusion Medicine and Cellular Therapies, Emory University School of Medicine, Atlanta, GA, United States
| | - Ethan Roback
- Department of Laboratory Medicine and Pathology, Center for Transfusion Medicine and Cellular Therapies, Emory University School of Medicine, Atlanta, GA, United States
| | - Nourine A Kamili
- Department of Laboratory Medicine and Pathology, Center for Transfusion Medicine and Cellular Therapies, Emory University School of Medicine, Atlanta, GA, United States
| | - Caleb S Stowell
- Department of Laboratory Medicine and Pathology, Center for Transfusion Medicine and Cellular Therapies, Emory University School of Medicine, Atlanta, GA, United States
| | | | - Pam M Van Ry
- Department of Biochemistry, Brigham Young University, Provo, UT, United States
| | - Marcelo Dias-Baruffi
- Department of Clinical Analyses, Toxicology and Food Sciences, School of Pharmaceutical Sciences of Ribeirao Preto, University of São Paulo, São Paulo, Brazil
| | - Richard D Cummings
- Department of Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, United States
| | - Sean R Stowell
- Department of Laboratory Medicine and Pathology, Center for Transfusion Medicine and Cellular Therapies, Emory University School of Medicine, Atlanta, GA, United States
| |
Collapse
|
21
|
Galectin-1 as an Emerging Mediator of Cardiovascular Inflammation: Mechanisms and Therapeutic Opportunities. Mediators Inflamm 2018; 2018:8696543. [PMID: 30524200 PMCID: PMC6247465 DOI: 10.1155/2018/8696543] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2018] [Accepted: 09/30/2018] [Indexed: 01/07/2023] Open
Abstract
Galectin-1 (Gal-1), an evolutionarily conserved β-galactoside-binding lectin, controls immune cell homeostasis and tempers acute and chronic inflammation by blunting proinflammatory cytokine synthesis, engaging T-cell apoptotic programs, promoting expansion of T regulatory (Treg) cells, and deactivating antigen-presenting cells. In addition, this lectin promotes angiogenesis by co-opting the vascular endothelial growth factor receptor (VEGFR) 2 signaling pathway. Since a coordinated network of immunomodulatory and proangiogenic mediators controls cardiac homeostasis, this lectin has been proposed to play a key hierarchical role in cardiac pathophysiology via glycan-dependent regulation of inflammatory responses. Here, we discuss the emerging roles of Gal-1 in cardiovascular diseases including acute myocardial infarction, heart failure, Chagas cardiomyopathy, pulmonary hypertension, and ischemic stroke, highlighting underlying anti-inflammatory mechanisms and therapeutic opportunities. Whereas Gal-1 administration emerges as a potential novel treatment option in acute myocardial infarction and ischemic stroke, Gal-1 blockade may contribute to attenuate pulmonary arterial hypertension.
Collapse
|
22
|
Carlos CP, Silva AA, Gil CD, Oliani SM. Pharmacological treatment with galectin-1 protects against renal ischaemia-reperfusion injury. Sci Rep 2018; 8:9568. [PMID: 29934646 PMCID: PMC6015078 DOI: 10.1038/s41598-018-27907-y] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2017] [Accepted: 06/07/2018] [Indexed: 11/21/2022] Open
Abstract
Galectin-1 protein (GAL-1) has important anti-inflammatory properties, but related pharmacologic approaches to effectively treat or prevent renal ischaemia and reperfusion injury are highly limited. Here, we investigated the effect of GAL-1 in a renal ischaemia-reperfusion injury rat model and an in vitro hypoxia-reoxygenation model with a proximal renal tubular epithelial cell line. In vivo, pretreatment with GAL-1 attenuated the renal parameters changed by ischaemia-reperfusion/hypoxia-reoxygenation, with recovery of renal function, protecting against influx of leukocytes, cell death and oxidative stress. Ischaemia-reperfusion/hypoxia-reoxygenation was also associated with increased renal endogenous expression of GAL-1 and intercellular adhesion molecule 1 (ICAM-1) plus augmented levels of proinflammatory cytokines IL-1β, TNF-α and MCP-1 and decreased anti-inflammatory IL-10 in urine, all of which were abrogated by GAL-1 treatment. In vitro studies demonstrated renal tubular epithelial cells as an important source of GAL-1 during hypoxia-reoxygenation and confirmed the protective effects of exogenous GAL-1 through downregulation of proinflammatory cytokine release by proximal renal tubular epithelial cells. Collectively, our findings confirm the important anti-inflammatory role of GAL-1 in kidney ischaemia and reperfusion injury and indicate its promising use as a therapeutic approach.
Collapse
Affiliation(s)
- Carla P Carlos
- Department of Biology, Instituto de Biociências, Letras e Ciências Exatas, Sao Paulo State University, UNESP, São José do Rio Preto, SP, Brazil.,Department of Medicine, FACERES School of Medicine, São José do Rio Preto, SP, Brazil
| | - Analice A Silva
- Department of Biology, Instituto de Biociências, Letras e Ciências Exatas, Sao Paulo State University, UNESP, São José do Rio Preto, SP, Brazil
| | - Cristiane D Gil
- Department of Morphology and Genetics, Federal University of Sao Paulo, UNIFESP, São Paulo, SP, Brazil
| | - Sonia M Oliani
- Department of Biology, Instituto de Biociências, Letras e Ciências Exatas, Sao Paulo State University, UNESP, São José do Rio Preto, SP, Brazil. .,Department of Morphology and Genetics, Federal University of Sao Paulo, UNIFESP, São Paulo, SP, Brazil.
| |
Collapse
|
23
|
Paz H, Joo EJ, Chou CH, Fei F, Mayo KH, Abdel-Azim H, Ghazarian H, Groffen J, Heisterkamp N. Treatment of B-cell precursor acute lymphoblastic leukemia with the Galectin-1 inhibitor PTX008. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2018; 37:67. [PMID: 29580262 PMCID: PMC5870532 DOI: 10.1186/s13046-018-0721-7] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/10/2017] [Accepted: 02/25/2018] [Indexed: 02/06/2023]
Abstract
Background Drug resistance of B-cell precursor acute lymphoblastic leukemia (BP-ALL) cells is conferred by both intrinsic and extrinsic factors, which could be targeted to promote chemo-sensitization. Our previous studies showed that Galectin-3, a lectin that clusters galactose-modified glycoproteins and that has both an intracellular and extracellular location, protects different subtypes of BP-ALL cells against chemotherapy. Galectin-1 is related to Galectin-3 and its expression was previously reported to be restricted to the MLL subtype of BP-ALL. Methods and results Here, we report that Galectin-1 is expressed at different levels in and on different subclasses of BP-ALLs. Bone marrow plasma also contains high levels of Galectin-1. PTX008 is an allosteric inhibitor which inhibits Galectin-1 but not Galectin-3-mediated agglutination. The compound reduces migration of BP-ALL cells to CXCL12 and OP9 stromal cells and inhibits fibronectin-mediated adhesion. It also affects cell cycle progression of BCP-ALL cells. PTX008 is cytostatic for BP-ALL cells even when these are co-cultured with protective stroma, and can sensitize ALL cells to vincristine chemotherapy in vitro and in mice. Conclusions PTX008 inhibits multiple functions that contribute to BP-ALL survival. The effects of Galectin-1 inhibition on both BP-ALL cell proliferation and migration suggest both the leukemia cells as well as the microenvironment that protects these cells may be targeted. Electronic supplementary material The online version of this article (10.1186/s13046-018-0721-7) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Helicia Paz
- Section of Molecular Carcinogenesis, Division of Hematology/Oncology and Bone Marrow Transplantation, The Saban Research Institute of Children's Hospital Los Angeles, Los Angeles, CA, 90027, USA.,Department of Surgical Oncology, UCLA, Los Angeles, CA, 90095, USA
| | - Eun Ji Joo
- Department of Systems Biology, Beckman Research Institute City of Hope, Monrovia, CA, USA
| | - Chih-Hsing Chou
- Section of Molecular Carcinogenesis, Division of Hematology/Oncology and Bone Marrow Transplantation, The Saban Research Institute of Children's Hospital Los Angeles, Los Angeles, CA, 90027, USA.,Division of Immunobiology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, 45229, USA
| | - Fei Fei
- Section of Molecular Carcinogenesis, Division of Hematology/Oncology and Bone Marrow Transplantation, The Saban Research Institute of Children's Hospital Los Angeles, Los Angeles, CA, 90027, USA.,Pathology Department, University of Alabama, Birmingham, AL, USA
| | - Kevin H Mayo
- Department of Biochemistry, Molecular Biology & Biophysics, University of Minnesota, Health Sciences Center, 6-155 Jackson Hall, 321 Church Street, Minneapolis, MN, 55455, USA
| | - Hisham Abdel-Azim
- Division of Hematology/Oncology and Bone Marrow Transplantation, Children's Hospital Los Angeles, Los Angeles, CA, 90027, USA
| | - Haike Ghazarian
- Department of Systems Biology, Beckman Research Institute City of Hope, Monrovia, CA, USA
| | - John Groffen
- Section of Molecular Carcinogenesis, Division of Hematology/Oncology and Bone Marrow Transplantation, The Saban Research Institute of Children's Hospital Los Angeles, Los Angeles, CA, 90027, USA
| | - Nora Heisterkamp
- Department of Systems Biology, Beckman Research Institute City of Hope, Monrovia, CA, USA.
| |
Collapse
|
24
|
Koo JH, Cho JY. Erratum to: Treadmill Exercise Attenuates α-Synuclein Levels by Promoting Mitochondrial Function and Autophagy Possibly via SIRT1 in the Chronic MPTP/P-Induced Mouse Model of Parkinson's Disease. Neurotox Res 2017; 32:532-533. [PMID: 28741160 DOI: 10.1007/s12640-017-9779-9] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Affiliation(s)
- Jung-Hoon Koo
- Department of Exercise Biochemistry, Korea National Sport University, Seoul, 138-763, Republic of Korea
- Institute of Sport Science, Korea National Sport University, Seoul, 138-763, Republic of Korea
| | - Joon-Yong Cho
- Department of Exercise Biochemistry, Korea National Sport University, Seoul, 138-763, Republic of Korea.
| |
Collapse
|
25
|
Seasonal and flight-related variation of galectin expression in heart, liver and flight muscles of yellow-rumped warblers (Setophaga coronata). Glycoconj J 2017; 34:603-611. [DOI: 10.1007/s10719-017-9779-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2017] [Revised: 05/25/2017] [Accepted: 05/28/2017] [Indexed: 10/19/2022]
|
26
|
SPRED1 Interferes with K-ras but Not H-ras Membrane Anchorage and Signaling. Mol Cell Biol 2016; 36:2612-25. [PMID: 27503857 DOI: 10.1128/mcb.00191-16] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2016] [Accepted: 08/03/2016] [Indexed: 12/13/2022] Open
Abstract
The Ras/mitogen-activated protein kinase (MAPK) signaling pathway is tightly controlled by negative feedback regulators, such as the tumor suppressor SPRED1. The SPRED1 gene also carries loss-of-function mutations in the RASopathy Legius syndrome. Growth factor stimulation translocates SPRED1 to the plasma membrane, triggering its inhibitory activity. However, it remains unclear whether SPRED1 there acts at the level of Ras or Raf. We show that pharmacological or galectin-1 (Gal-1)-mediated induction of B- and C-Raf-containing dimers translocates SPRED1 to the plasma membrane. This is facilitated in particular by SPRED1 interaction with B-Raf and, via its N terminus, with Gal-1. The physiological significance of these novel interactions is supported by two Legius syndrome-associated mutations that show diminished binding to both Gal-1 and B-Raf. On the plasma membrane, SPRED1 becomes enriched in acidic membrane domains to specifically perturb membrane organization and extracellular signal-regulated kinase (ERK) signaling of active K-ras4B (here, K-ras) but not H-ras. However, SPRED1 also blocks on the nanoscale the positive effects of Gal-1 on H-ras. Therefore, a combinatorial expression of SPRED1 and Gal-1 potentially regulates specific patterns of K-ras- and H-ras-dependent signaling output. More broadly, our results open up the possibility that related SPRED and Sprouty proteins act in a similar Ras and Raf isoform-specific manner.
Collapse
|
27
|
O'Sullivan JM, Jenkins PV, Rawley O, Gegenbauer K, Chion A, Lavin M, Byrne B, O'Kennedy R, Preston RJS, Brophy TM, O'Donnell JS. Galectin-1 and Galectin-3 Constitute Novel-Binding Partners for Factor VIII. Arterioscler Thromb Vasc Biol 2016; 36:855-63. [PMID: 27013611 DOI: 10.1161/atvbaha.115.306915] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2015] [Accepted: 03/14/2016] [Indexed: 01/20/2023]
Abstract
OBJECTIVE Recent studies have demonstrated that galectin-1 (Gal-1) and galectin-3 (Gal-3) can bind von Willebrand factor and directly modulate von Willebrand factor-dependent early thrombus formation in vivo. Because the glycans expressed on human factor VIII (FVIII) are similar to those of von Willebrand factor, we investigated whether galectins might also bind and modulate the activity of FVIII. APPROACH AND RESULTS Immunosorbant assays and surface plasmon resonance analysis confirmed that Gal-1 and Gal-3 bound purified FVIII with high affinity. Exoglycosidase removal of FVIII N-linked glycans significantly reduced binding to both Gal-1 and Gal-3. Moreover, combined removal of both the N- and O-glycans of FVIII further attenuated Gal-3 binding. Notably, specific digestion of FVIII high-mannose glycans at N239 and N2118 significantly impaired FVIII affinity for Gal-1. Importantly Gal-1, but not Gal-3, bound to free FVIII in the plasma milieu, and significantly inhibited FVIII functional activity. Interestingly, commercial recombinant FVIII (rFVIII) concentrates are manufactured in different cell lines and differ in their glycosylation profiles. Although the biological mechanism has not been defined, recent studies in previously untreated patients with severe hemophilia A reported significant differences in inhibitor development associated with different rFVIII products. Interestingly, Gal-1 and Gal-3 both displayed enhanced affinity for BHK-rFVIII compared with CHO-rFVIII. Furthermore, binding of Gal-1 and Gal-3 to BDD-FVIII was markedly reduced compared with full-length rFVIII. CONCLUSIONS We have identified Gal-1 and Gal-3 as novel-binding partners for human FVIII and demonstrated that Gal-1 binding can influence the procoagulant activity of FVIII.
Collapse
Affiliation(s)
- Jamie M O'Sullivan
- From the Haemostasis Research Group, Institute of Molecular Medicine, Trinity Centre for Health Sciences (J.M.O., P.V.J., O.R., K.G., A.C., M.L., T.M.B., J.S.O.) and National Centre for Hereditary Coagulation Disorders (J.S.O.), St. James's Hospital, and Department of Clinical Medicine, School of Medicine (R.J.S.P.), Trinity College, Dublin, Ireland; School of Biotechnology and Biomedical Diagnostics Institute, Dublin City University, Dublin, Ireland (B.B., R.O.); and National Children's Research Centre, Our Lady's Children's Hospital, Crumlin, Dublin, Ireland (R.J.S.P.)
| | - P Vince Jenkins
- From the Haemostasis Research Group, Institute of Molecular Medicine, Trinity Centre for Health Sciences (J.M.O., P.V.J., O.R., K.G., A.C., M.L., T.M.B., J.S.O.) and National Centre for Hereditary Coagulation Disorders (J.S.O.), St. James's Hospital, and Department of Clinical Medicine, School of Medicine (R.J.S.P.), Trinity College, Dublin, Ireland; School of Biotechnology and Biomedical Diagnostics Institute, Dublin City University, Dublin, Ireland (B.B., R.O.); and National Children's Research Centre, Our Lady's Children's Hospital, Crumlin, Dublin, Ireland (R.J.S.P.)
| | - Orla Rawley
- From the Haemostasis Research Group, Institute of Molecular Medicine, Trinity Centre for Health Sciences (J.M.O., P.V.J., O.R., K.G., A.C., M.L., T.M.B., J.S.O.) and National Centre for Hereditary Coagulation Disorders (J.S.O.), St. James's Hospital, and Department of Clinical Medicine, School of Medicine (R.J.S.P.), Trinity College, Dublin, Ireland; School of Biotechnology and Biomedical Diagnostics Institute, Dublin City University, Dublin, Ireland (B.B., R.O.); and National Children's Research Centre, Our Lady's Children's Hospital, Crumlin, Dublin, Ireland (R.J.S.P.)
| | - Kristina Gegenbauer
- From the Haemostasis Research Group, Institute of Molecular Medicine, Trinity Centre for Health Sciences (J.M.O., P.V.J., O.R., K.G., A.C., M.L., T.M.B., J.S.O.) and National Centre for Hereditary Coagulation Disorders (J.S.O.), St. James's Hospital, and Department of Clinical Medicine, School of Medicine (R.J.S.P.), Trinity College, Dublin, Ireland; School of Biotechnology and Biomedical Diagnostics Institute, Dublin City University, Dublin, Ireland (B.B., R.O.); and National Children's Research Centre, Our Lady's Children's Hospital, Crumlin, Dublin, Ireland (R.J.S.P.)
| | - Alain Chion
- From the Haemostasis Research Group, Institute of Molecular Medicine, Trinity Centre for Health Sciences (J.M.O., P.V.J., O.R., K.G., A.C., M.L., T.M.B., J.S.O.) and National Centre for Hereditary Coagulation Disorders (J.S.O.), St. James's Hospital, and Department of Clinical Medicine, School of Medicine (R.J.S.P.), Trinity College, Dublin, Ireland; School of Biotechnology and Biomedical Diagnostics Institute, Dublin City University, Dublin, Ireland (B.B., R.O.); and National Children's Research Centre, Our Lady's Children's Hospital, Crumlin, Dublin, Ireland (R.J.S.P.)
| | - Michelle Lavin
- From the Haemostasis Research Group, Institute of Molecular Medicine, Trinity Centre for Health Sciences (J.M.O., P.V.J., O.R., K.G., A.C., M.L., T.M.B., J.S.O.) and National Centre for Hereditary Coagulation Disorders (J.S.O.), St. James's Hospital, and Department of Clinical Medicine, School of Medicine (R.J.S.P.), Trinity College, Dublin, Ireland; School of Biotechnology and Biomedical Diagnostics Institute, Dublin City University, Dublin, Ireland (B.B., R.O.); and National Children's Research Centre, Our Lady's Children's Hospital, Crumlin, Dublin, Ireland (R.J.S.P.)
| | - Barry Byrne
- From the Haemostasis Research Group, Institute of Molecular Medicine, Trinity Centre for Health Sciences (J.M.O., P.V.J., O.R., K.G., A.C., M.L., T.M.B., J.S.O.) and National Centre for Hereditary Coagulation Disorders (J.S.O.), St. James's Hospital, and Department of Clinical Medicine, School of Medicine (R.J.S.P.), Trinity College, Dublin, Ireland; School of Biotechnology and Biomedical Diagnostics Institute, Dublin City University, Dublin, Ireland (B.B., R.O.); and National Children's Research Centre, Our Lady's Children's Hospital, Crumlin, Dublin, Ireland (R.J.S.P.)
| | - Richard O'Kennedy
- From the Haemostasis Research Group, Institute of Molecular Medicine, Trinity Centre for Health Sciences (J.M.O., P.V.J., O.R., K.G., A.C., M.L., T.M.B., J.S.O.) and National Centre for Hereditary Coagulation Disorders (J.S.O.), St. James's Hospital, and Department of Clinical Medicine, School of Medicine (R.J.S.P.), Trinity College, Dublin, Ireland; School of Biotechnology and Biomedical Diagnostics Institute, Dublin City University, Dublin, Ireland (B.B., R.O.); and National Children's Research Centre, Our Lady's Children's Hospital, Crumlin, Dublin, Ireland (R.J.S.P.)
| | - Roger J S Preston
- From the Haemostasis Research Group, Institute of Molecular Medicine, Trinity Centre for Health Sciences (J.M.O., P.V.J., O.R., K.G., A.C., M.L., T.M.B., J.S.O.) and National Centre for Hereditary Coagulation Disorders (J.S.O.), St. James's Hospital, and Department of Clinical Medicine, School of Medicine (R.J.S.P.), Trinity College, Dublin, Ireland; School of Biotechnology and Biomedical Diagnostics Institute, Dublin City University, Dublin, Ireland (B.B., R.O.); and National Children's Research Centre, Our Lady's Children's Hospital, Crumlin, Dublin, Ireland (R.J.S.P.)
| | - Teresa M Brophy
- From the Haemostasis Research Group, Institute of Molecular Medicine, Trinity Centre for Health Sciences (J.M.O., P.V.J., O.R., K.G., A.C., M.L., T.M.B., J.S.O.) and National Centre for Hereditary Coagulation Disorders (J.S.O.), St. James's Hospital, and Department of Clinical Medicine, School of Medicine (R.J.S.P.), Trinity College, Dublin, Ireland; School of Biotechnology and Biomedical Diagnostics Institute, Dublin City University, Dublin, Ireland (B.B., R.O.); and National Children's Research Centre, Our Lady's Children's Hospital, Crumlin, Dublin, Ireland (R.J.S.P.)
| | - James S O'Donnell
- From the Haemostasis Research Group, Institute of Molecular Medicine, Trinity Centre for Health Sciences (J.M.O., P.V.J., O.R., K.G., A.C., M.L., T.M.B., J.S.O.) and National Centre for Hereditary Coagulation Disorders (J.S.O.), St. James's Hospital, and Department of Clinical Medicine, School of Medicine (R.J.S.P.), Trinity College, Dublin, Ireland; School of Biotechnology and Biomedical Diagnostics Institute, Dublin City University, Dublin, Ireland (B.B., R.O.); and National Children's Research Centre, Our Lady's Children's Hospital, Crumlin, Dublin, Ireland (R.J.S.P.).
| |
Collapse
|
28
|
Timoshenko AV. Towards molecular mechanisms regulating the expression of galectins in cancer cells under microenvironmental stress conditions. Cell Mol Life Sci 2015; 72:4327-40. [PMID: 26245305 PMCID: PMC11113283 DOI: 10.1007/s00018-015-2008-x] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2015] [Revised: 07/12/2015] [Accepted: 07/30/2015] [Indexed: 02/07/2023]
Abstract
Galectins, a family of soluble β-galactoside-binding proteins, serve as mediators of fundamental biological processes, such as cell growth, differentiation, adhesion, migration, survival, and death. The purpose of this review is to summarize the current knowledge regarding the ways in which the expression of individual galectins differs in normal and transformed human cells exposed to various stimuli mimicking physiological and pathological microenvironmental stress conditions. A conceptual point is being made and grounded that the modulation of galectin expression profiles is a key aspect of cellular stress responses. Moreover, this modulation might be precisely regulated at transcriptional and post-transcriptional levels in the context of non-overlapping transcription factors and miRNAs specific to galectins.
Collapse
Affiliation(s)
- Alexander V Timoshenko
- Department of Biology, Western University, 1151 Richmond Street, London, ON, N6A 5B7, Canada.
| |
Collapse
|
29
|
Cerri DG, Arthur CM, Rodrigues LC, Fermino ML, Rocha LB, Stowell SR, Baruffi MD. Examination of galectin localization using confocal microscopy. Methods Mol Biol 2015; 1207:343-54. [PMID: 25253152 DOI: 10.1007/978-1-4939-1396-1_23] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/15/2023]
Abstract
Confocal microscopy provides a unique modality to examine the expression and localization of biomolecules in a variety of settings. Using this technique, an image is acquired from the focal plane of the objective using focused laser light, making it possible to work within the resolution limit of the optical system. In addition, by acquiring multiple images from a variety of focal planes, stacked series of images can provide clear spatial localization of a probed structure or protein. We describe herein the immunofluorescence methods for galectin staining in frozen sections of tissue for galectin localization using confocal microscopy.
Collapse
Affiliation(s)
- Daniel Giuliano Cerri
- Faculty of Pharmaceutical Sciences of Ribeirão Preto, Department of Clinical, Toxicological and Bromatological Analysis, University of Sao Paulo, Ribeirão Preto-S, Brazil
| | | | | | | | | | | | | |
Collapse
|
30
|
Stowell SR, Arthur CM, Cummings RD, Feasley CL. Alkylation of galectin-1 with iodoacetamide and mass spectrometric mapping of the sites of incorporation. Methods Mol Biol 2015; 1207:51-62. [PMID: 25253132 PMCID: PMC5755404 DOI: 10.1007/978-1-4939-1396-1_3] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/03/2023]
Abstract
Galectins can display unique sensitivity to oxidative changes that result in significant conformational alterations that prevent carbohydrate recognition. While a variety of approaches can be utilized to prevent galectin oxidation, several of these require inclusion of reducing agents that not only prevent galectins from undergoing oxidative inactivation, but can also interfere with normal redox potentials required for fundamental cellular processes. To overcome limitations associated with placing cells in an artificial reducing environment, cysteine residues on galectins can be directly alkylated with iodoacetamide to form a stable thioether adduct that is resistant to further modification. Iodoacetamide alkylated galectin remains stable over prolonged periods of time and retains the carbohydrate binding and biological activities of the native protein. As a result, this approach allows examination of the biological roles of a stabilized form of galectin-1 without introducing the confounding variables that can occur when typical soluble reducing agents are employed.
Collapse
Affiliation(s)
- Sean R Stowell
- Center for Transfusion and Cellular Therapies, Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, GA, USA
| | | | | | | |
Collapse
|
31
|
Abstract
Galectins are an evolutionarily ancient family of glycan-binding proteins (GBPs) and are found in all animals. Although they were discovered over 30 years ago, ideas about their biological functions continue to evolve. Current evidence indicates that galectins, which are the only known GBPs that occur free in the cytoplasm and extracellularly, are involved in a variety of intracellular and extracellular pathways contributing to homeostasis, cellular turnover, cell adhesion, and immunity. Here we review evolving insights into galectin biology from a historical perspective and explore current evidence regarding biological roles of galectins.
Collapse
|
32
|
Detection of phosphatidylserine exposure on leukocytes following treatment with human galectins. Methods Mol Biol 2015; 1207:185-200. [PMID: 25253141 DOI: 10.1007/978-1-4939-1396-1_12] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Cellular turnover represents a fundamental aspect of immunological homeostasis. While many factors appear to regulate leukocyte removal during inflammatory resolution, recent studies suggest that members of the galectin family play a unique role in orchestrating this process. Unlike cellular removal through apoptotic cell death, several members of the galectin family induce surface expression of phosphatidylserine (PS), a phagocytic marker on cells undergoing apoptosis, in the absence of cell death. However, similar to PS on cells undergoing apoptosis, galectin-induced PS exposure sensitizes cells to phagocytic removal. As galectins appear to prepare cells for phagocytic removal without actually inducing apoptotic cell death, this process has recently been coined preaparesis. Given the unique characteristics of galectin-induced PS exposure in the context of preaparesis, we will examine important considerations when evaluating the potential impact of different galectin family members on PS exposure and cell viability.
Collapse
|
33
|
Abstract
Glycan binding proteins (GBPs) possess the unique ability to regulate a wide variety of biological processes through interactions with highly modifiable cell surface glycans. While many studies demonstrate the impact of glycan modification on GBP recognition and activity, the relative contribution of subtle changes in glycan structure on GBP binding can be difficult to define. To overcome limitations in the analysis of GBP-glycan interactions, recent studies utilized glycan microarray platforms containing hundreds of structurally defined glycans. These studies not only provided important information regarding GBP-glycan interactions, but have also resulted in significant insight into the binding specificity and biological activity of the galectin family. We will describe the methods used when employing glycan microarray platforms to examine galectin-glycan binding specificity and function.
Collapse
|
34
|
Al-Salam S, Hashmi S. Galectin-1 in early acute myocardial infarction. PLoS One 2014; 9:e86994. [PMID: 24498007 PMCID: PMC3909026 DOI: 10.1371/journal.pone.0086994] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2013] [Accepted: 12/17/2013] [Indexed: 02/02/2023] Open
Abstract
Myocardial infarction (MI) is the most serious manifestation of coronary artery disease and the cause of significant mortality and morbidity worldwide. Galectin-1(GAL-1), a divalent 14.5-kDa protein, is present both inside and outside cells, and has both intracellular and extracellular functions. Hypoxia inducible factor-1 alpha (HIF-1α) is a transcription factor mediating early and late responses to myocardial ischemia. Identification of the pattern of expression of GAL-1 and HIF-1α in the heart during the first 24 hours following acute MI will help in understanding early molecular changes in this event and may provide methods to overcome serious complications. Mouse model of MI was used and heart samples were processed for immunohistochemical and immunofluorescent labeling and Enzyme linked immunosorbent assay to identify GAL-1 and HIF 1α levels in the heart during the first 24 hours following MI. There was significant increase in left ventricular GAL-1 at 20 (p = 0.001) and 30 minutes (p = 0.004) following MI. There was also a significant increase in plasma GAL-1 at 4 hours (p = 0.012) and 24 hours (p = 0.001) following MI. A significant increase in left ventricular HIF-1 α was seen at 20 minutes (p = 0.047) following MI. In conclusion, we show for the first time that GAL-1 level in the left ventricle is increased in early ischemic period. We also report for the first time that HIF-1 α is significantly increased at 20 minutes following MI. In addition we report for the first time that mouse plasma GAL-1 level is significantly raised as early as 4 hours following MI.
Collapse
Affiliation(s)
- Suhail Al-Salam
- Department of Pathology, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain, Abu Dhabi, United Arab Emirates
- * E-mail:
| | - Satwat Hashmi
- Department of Pathology, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain, Abu Dhabi, United Arab Emirates
| |
Collapse
|
35
|
Thijssen VL, Rabinovich GA, Griffioen AW. Vascular galectins: regulators of tumor progression and targets for cancer therapy. Cytokine Growth Factor Rev 2013; 24:547-58. [PMID: 23942184 DOI: 10.1016/j.cytogfr.2013.07.003] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2013] [Revised: 07/13/2013] [Accepted: 07/18/2013] [Indexed: 12/14/2022]
Abstract
Galectins are a family of carbohydrate binding proteins with a broad range of cytokine and growth factor-like functions in multiple steps of cancer progression. They contribute to tumor cell transformation, promote tumor angiogenesis, hamper the anti-tumor immune response, and facilitate tumor metastasis. Consequently, galectins are considered as multifunctional targets for cancer therapy. Interestingly, many of the functions related to tumor progression can be linked to galectins expressed by endothelial cells in the tumor vascular bed. Since the tumor vasculature is an easily accessible target for cancer therapy, understanding how galectins in the tumor endothelium influence cancer progression is important for the translational development of galectin-targeting therapies.
Collapse
Affiliation(s)
- Victor L Thijssen
- Angiogenesis Laboratory, Department of Medical Oncology, VU University Medical Center, Amsterdam, The Netherlands.
| | | | | |
Collapse
|
36
|
Seropian IM, Cerliani JP, Toldo S, Van Tassell BW, Ilarregui JM, González GE, Matoso M, Salloum FN, Melchior R, Gelpi RJ, Stupirski JC, Benatar A, Gómez KA, Morales C, Abbate A, Rabinovich GA. Galectin-1 controls cardiac inflammation and ventricular remodeling during acute myocardial infarction. THE AMERICAN JOURNAL OF PATHOLOGY 2013; 182:29-40. [PMID: 23142379 PMCID: PMC5691326 DOI: 10.1016/j.ajpath.2012.09.022] [Citation(s) in RCA: 78] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/29/2011] [Revised: 09/12/2012] [Accepted: 09/29/2012] [Indexed: 12/11/2022]
Abstract
Galectin-1 (Gal-1), an evolutionarily conserved β-galactoside-binding lectin, plays essential roles in the control of inflammation and neovascularization. Although identified as a major component of the contractile apparatus of cardiomyocytes, the potential role of Gal-1 in modulating heart pathophysiology is uncertain. Here, we aimed to characterize Gal-1 expression and function in the infarcted heart. Expression of Gal-1 was substantially increased in the mouse heart 7 days after acute myocardial infarction (AMI) and in hearts from patients with end-stage chronic heart failure. This lectin was localized mainly in cardiomyocytes and inflammatory infiltrates in peri-infarct areas, but not in remote areas. Both simulated hypoxia and proinflammatory cytokines selectively up-regulated Gal-1 expression in mouse cardiomyocytes, whereas anti-inflammatory cytokines inhibited expression of this lectin or had no considerable effect. Compared with their wild-type counterpart, Gal-1-deficient (Lgals1(-/-)) mice showed enhanced cardiac inflammation, characterized by increased numbers of macrophages, natural killer cells, and total T cells, but reduced frequency of regulatory T cells, leading to impaired cardiac function at baseline and impaired ventricular remodeling 7 days after nonreperfused AMI. Treatment of mice with recombinant Gal-1 attenuated cardiac damage in reperfused AMI. Taken together, our results indicate a protective role for Gal-1 in normal cardiac homeostasis and postinfarction remodeling by preventing cardiac inflammation. Thus, Gal-1 treatment represents a potential novel strategy to attenuate heart failure in AMI.
Collapse
Affiliation(s)
- Ignacio M. Seropian
- VCU Pauley Heart Center, Virginia Commonwealth University, Richmond, Virginia
- School of Pharmacy, Virginia Commonwealth University, Richmond, Virginia
| | - Juan P. Cerliani
- Laboratory of Immunopathology, Institute of Biology and Experimental Medicine (IBYME), National Scientific and Technical Research Council (CONICET), Buenos Aires, Argentina
| | - Stefano Toldo
- VCU Pauley Heart Center, Virginia Commonwealth University, Richmond, Virginia
- VCU Victoria Johnson Center, Virginia Commonwealth University, Richmond, Virginia
| | - Benjamín W. Van Tassell
- VCU Pauley Heart Center, Virginia Commonwealth University, Richmond, Virginia
- School of Pharmacy, Virginia Commonwealth University, Richmond, Virginia
| | - Juan M. Ilarregui
- Laboratory of Immunopathology, Institute of Biology and Experimental Medicine (IBYME), National Scientific and Technical Research Council (CONICET), Buenos Aires, Argentina
| | - Germán E. González
- Institute of Cardiovascular Physiopathology, Faculty of Medicine, University of Buenos Aires, Buenos Aires, Argentina
| | - Mirian Matoso
- Institute of Cardiovascular Physiopathology, Faculty of Medicine, University of Buenos Aires, Buenos Aires, Argentina
| | - Fadi N. Salloum
- VCU Pauley Heart Center, Virginia Commonwealth University, Richmond, Virginia
| | - Ryan Melchior
- VCU Pauley Heart Center, Virginia Commonwealth University, Richmond, Virginia
- School of Pharmacy, Virginia Commonwealth University, Richmond, Virginia
| | - Ricardo J. Gelpi
- Institute of Cardiovascular Physiopathology, Faculty of Medicine, University of Buenos Aires, Buenos Aires, Argentina
| | - Juan C. Stupirski
- Laboratory of Immunopathology, Institute of Biology and Experimental Medicine (IBYME), National Scientific and Technical Research Council (CONICET), Buenos Aires, Argentina
| | - Alejandro Benatar
- Institute of Research in Genetic Engineering and Molecular Biology (INGEBI), National Scientific and Technical Research Council (CONICET), Buenos Aires, Argentina
| | - Karina A. Gómez
- Institute of Research in Genetic Engineering and Molecular Biology (INGEBI), National Scientific and Technical Research Council (CONICET), Buenos Aires, Argentina
| | - Celina Morales
- Institute of Cardiovascular Physiopathology, Faculty of Medicine, University of Buenos Aires, Buenos Aires, Argentina
| | - Antonio Abbate
- VCU Pauley Heart Center, Virginia Commonwealth University, Richmond, Virginia
- VCU Victoria Johnson Center, Virginia Commonwealth University, Richmond, Virginia
| | - Gabriel A. Rabinovich
- Laboratory of Immunopathology, Institute of Biology and Experimental Medicine (IBYME), National Scientific and Technical Research Council (CONICET), Buenos Aires, Argentina
- Department of Biological Chemistry, Faculty of Exact and Natural Sciences, University of Buenos Aires, Buenos Aires, Argentina
| |
Collapse
|
37
|
Negrete-Garcia MC, Jiménez-Torres CY, Alvarado-Vásquez N, Montes-Vizuet AR, Velázquez-Rodriguez JR, Jimenez-Martinez MC, Teran-Juárez LM. Galectin-10 is released in the nasal lavage fluid of patients with aspirin-sensitive respiratory disease. ScientificWorldJournal 2012; 2012:474020. [PMID: 22654612 PMCID: PMC3361262 DOI: 10.1100/2012/474020] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2011] [Accepted: 01/22/2012] [Indexed: 01/19/2023] Open
Abstract
The aim of this work was to determine the presence of galectin-10 in nasal lavage fluid (NLF) of patients with aspirin-sensitive respiratory disease (ASRD) before and after challenge with L-ASA (aspirin) by ELISA. Fifteen ASRD patients, ten aspirin-tolerant asthmatics (ATA), and fifteen healthy controls (HC) were studied. The baseline presence of Galectin-10 in PBMC was determined using real time PCR. Galectin-10 was evaluated in tissue of nasal polyps by western blot. Our results showed a lower expression in PBMC of ASRD patients than in ATA and healthy controls. However, a higher concentration of galectin-10 in NLF was found in ASRD patients before and after L-ASA challenge; western blot confirmed a high expression of galectin-10 in tissue from nasal polyps obtained from ASRD patients. Our results suggest a probable role of galectin-10 in the inflammatory response observed in ASRD patients; however, confirmatory studies are needed.
Collapse
Affiliation(s)
- Ma Cristina Negrete-Garcia
- Department of Immunogenetics and Allergy, Instituto Nacional de Enfermedades Respiratorias, Calzada Tlalpan 4502, 14080 Mexico, DF, Mexico.
| | | | | | | | | | | | | |
Collapse
|
38
|
Saint-Lu N, Oortwijn BD, Pegon JN, Odouard S, Christophe OD, de Groot PG, Denis CV, Lenting PJ. Identification of galectin-1 and galectin-3 as novel partners for von Willebrand factor. Arterioscler Thromb Vasc Biol 2012; 32:894-901. [PMID: 22267483 DOI: 10.1161/atvbaha.111.240309] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
OBJECTIVE Although von Willebrand factor (VWF) is a heavily glycosylated protein, its potential to associate with glycan-binding proteins is poorly investigated. Here, we explored its interaction with the glycan-binding proteins galectin-1 and galectin-3. METHODS AND RESULTS Immunofluorescence analysis using Duolink proximity ligation assays revealed that VWF colocalizes with galectin-1 and galectin-3 in endothelial cells, both before and after stimulation of endothelial cells. Moreover, galectin-1 was found along the typical VWF bundles that are released by endothelial cells. Galectin-1 and galectin-3 could be coprecipitated with VWF from plasma in immunoprecipitation assays, whereas plasma levels of galectin-1 and galectin-3 were significantly reduced in VWF-deficient mice. Binding studies using purified proteins confirmed that VWF could directly interact with both galectins, predominantly via its N-linked glycans. In search of the physiological relevance of the VWF-galectin interaction, we found that inhibition of galectins in in vitro perfusion assays was associated with increased VWF-platelet string formation, a phenomenon that was reproduced in galectin-1/galectin-3 double-deficient mice. These mice were also characterized by a more rapid formation of initial thrombi following ferric chloride-induced injury. CONCLUSIONS We have identified galectin-1 and galectin-3 as novel partners for VWF, and these proteins may modulate VWF-mediated thrombus formation.
Collapse
|
39
|
Cooper D, Iqbal AJ, Gittens BR, Cervone C, Perretti M. The effect of galectins on leukocyte trafficking in inflammation: sweet or sour? Ann N Y Acad Sci 2012; 1253:181-92. [PMID: 22256855 DOI: 10.1111/j.1749-6632.2011.06291.x] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
The trafficking of leukocytes from the blood stream to the surrounding tissue is a fundamental feature of an inflammatory response. Although many of the adhesion molecules and chemokines that direct leukocyte trafficking have been identified, there is still much to be discovered, particularly with regard to the persistence of leukocyte infiltrates in chronic inflammation. Elucidating the molecular mechanisms involved in this process is critical to understanding and treating inflammatory pathologies. Recent studies have identified members of the galectin family as immunoregulatory proteins. Included among the actions of galectins are modulatory effects, both positive and negative, on leukocyte recruitment. The focus of this review is to summarize current knowledge on the role of galectins in leukocyte trafficking during inflammation. A better understanding of the function of this family of endogenous lectins will open new avenues for innovative drug discovery.
Collapse
Affiliation(s)
- Dianne Cooper
- William Harvey Research Institute, Barts and The London School of Medicine, Queen Mary University of London, United Kingdom.
| | | | | | | | | |
Collapse
|
40
|
Vasta GR. Galectins as pattern recognition receptors: structure, function, and evolution. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2012; 946:21-36. [PMID: 21948360 DOI: 10.1007/978-1-4614-0106-3_2] [Citation(s) in RCA: 177] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/03/2022]
Abstract
Galectins constitute an evolutionary conserved family of ß-galactoside-binding proteins, ubiquitous in mammals and other vertebrate taxa, invertebrates, and fungi. Since their discovery in the 1970s, their biological roles, initially understood as limited to recognition of carbohydrate ligands in embryogenesis and development, have expanded in recent years by the discovery of their immunoregulatory activities. A gradual paradigm shift has taken place in the past few years through the recognition that galectins also bind glycans on the surface of potentially pathogenic microbes, and function as recognition and effector factors in innate immunity. Further, an additional level of functional complexity has emerged with the most recent findings that some parasites "subvert" the recognition roles of the vector/host galectins for successful attachment or invasion.
Collapse
Affiliation(s)
- Gerardo R Vasta
- Department of Microbiology and Immunology, School of Medicine, IMET, University of Maryland, Baltimore, MD 21202, USA.
| |
Collapse
|
41
|
Cerliani JP, Stowell SR, Mascanfroni ID, Arthur CM, Cummings RD, Rabinovich GA. Expanding the universe of cytokines and pattern recognition receptors: galectins and glycans in innate immunity. J Clin Immunol 2010; 31:10-21. [PMID: 21184154 DOI: 10.1007/s10875-010-9494-2] [Citation(s) in RCA: 85] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2010] [Accepted: 12/03/2010] [Indexed: 12/31/2022]
Abstract
Effective immunity relies on the recognition of pathogens and tumors by innate immune cells through diverse pattern recognition receptors (PRRs) that lead to initiation of signaling processes and secretion of pro- and anti-inflammatory cytokines. Galectins, a family of endogenous lectins widely expressed in infected and neoplastic tissues have emerged as part of the portfolio of soluble mediators and pattern recognition receptors responsible for eliciting and controlling innate immunity. These highly conserved glycan-binding proteins can control immune cell processes through binding to specific glycan structures on pathogens and tumors or by acting intracellularly via modulation of selective signaling pathways. Recent findings demonstrate that various galectin family members influence the fate and physiology of different innate immune cells including polymorphonuclear neutrophils, mast cells, macrophages, and dendritic cells. Moreover, several pathogens may actually utilize galectins as a mechanism of host invasion. In this review, we aim to highlight and integrate recent discoveries that have led to our current understanding of the role of galectins in host-pathogen interactions and innate immunity. Challenges for the future will embrace the rational manipulation of galectin-glycan interactions to instruct and shape innate immunity during microbial infections, inflammation, and cancer.
Collapse
Affiliation(s)
- Juan P Cerliani
- Laboratorio de Inmunopatología, Instituto de Biología y Medicina Experimental, Consejo Nacional de Investigaciones Científicas y Técnicas, 1428 Buenos Aires, Argentina
| | | | | | | | | | | |
Collapse
|