1
|
Iqbal M, Feng C, Zong G, Wang LX, Vasta GR. Galectin-3 disrupts tight junctions of airway epithelial cell monolayers by inducing expression and release of matrix metalloproteinases upon influenza A infection. Glycobiology 2025; 35:cwae093. [PMID: 39569730 PMCID: PMC11727335 DOI: 10.1093/glycob/cwae093] [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] [Received: 08/30/2024] [Revised: 11/03/2024] [Accepted: 11/19/2024] [Indexed: 11/22/2024] Open
Abstract
Galectins are β-galactosyl-binding lectins with key roles in early development, immune regulation, and infectious disease. Influenza A virus (IAV) infects the airway epithelia, and in severe cases may lead to bacterial superinfections and hypercytokinemia, and eventually, to acute respiratory distress syndrome (ARDS) through the breakdown of airway barriers. The detailed mechanisms involved, however, remain poorly understood. Our prior in vivo studies in a murine model system revealed that upon experimental IAV and pneumococcal primary and secondary challenges, respectively, galectin-1 and galectin-3 (Gal-3) are released into the airway and bind to the epithelium that has been desialylated by the viral neuraminidase, contributing to secondary bacterial infection and hypercytokinemia leading to the clinical decline and death of the animals. Here we report the results of in vitro studies that reveal the role of the extracellular Gal-3 in additional detrimental effects on the host by disrupting the integrity of the airway epithelial barrier. IAV infection of the human airway epithelia cell line A549 increased release of Gal-3 and its binding to the A549 desialylated cell surface, notably to the transmembrane signaling receptors CD147 and integrin-β1. Addition of recombinant Gal-3 to A549 monolayers resulted in enhanced expression and release of matrix metalloproteinases, leading to disruption of cell-cell tight junctions, and a significant increase in paracellular permeability. This study reveals a critical mechanism involving Gal-3 that may significantly contribute to the severity of IAV infections by promoting disruption of tight junctions and enhanced permeability of the airway epithelia, potentially leading to lung edema and ARDS.
Collapse
Affiliation(s)
- Muddassar Iqbal
- Department of Microbiology and Immunology, Institute of Marine and Environmental Technology, University of Maryland School of Medicine, Colwell Center, 701 East Pratt Street, Baltimore, MD 21202, USA
| | - Chiguang Feng
- Department of Microbiology and Immunology, Institute of Marine and Environmental Technology, University of Maryland School of Medicine, Colwell Center, 701 East Pratt Street, Baltimore, MD 21202, USA
- Current address: Immunity, Inflammation, and Disease Laboratory, National Institute of Environmental Health Sciences, Research Triangle Park, Durham, NC 27709, USA
| | - Guanghui Zong
- Department of Chemistry and Biochemistry,University of Maryland, Chemistry Bldg, 1526, 8051 Regents Dr, College Park, MD 20742, USA
| | - Lai-Xi Wang
- Department of Chemistry and Biochemistry,University of Maryland, Chemistry Bldg, 1526, 8051 Regents Dr, College Park, MD 20742, USA
| | - Gerardo R Vasta
- Department of Microbiology and Immunology, Institute of Marine and Environmental Technology, University of Maryland School of Medicine, Colwell Center, 701 East Pratt Street, Baltimore, MD 21202, USA
| |
Collapse
|
2
|
Sato S, Iwaki J, Hirabayashi J. Decoding the multifaceted roles of galectins in self-defense. Semin Immunol 2024; 77:101926. [PMID: 39721561 DOI: 10.1016/j.smim.2024.101926] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/17/2024] [Revised: 12/13/2024] [Accepted: 12/14/2024] [Indexed: 12/28/2024]
Abstract
In this review, we aim to explore the multifaceted roles of galectins in host defense from a broader perspective, particularly regarding their functions when host integrity is compromised. Numerous comprehensive reviews on galectin functions in immunity have already been published. For researchers new to the field, this wealth of information may create an impression of galectins as proteins involved in a wide array of biological processes. Furthermore, due to the heterogeneity of galectin ligands, glycans, there is a risk of perceiving galectin-specific functions as ambiguous, potentially obscuring their core biological significance. To address this, we revisit foundational aspects, focusing on the significance of the recognition of galactose, a "late-comer" monosaccharide in evolutionary terms, provide an overview of galectin glycan binding specificity, with emphasis on the potential biological importance of each carbohydrate-recognition domain. We also discuss the biological implications of the galectin location paradox wherein these cytosolic lectins function in host defense despite their glycan ligands being synthesized in the secretory pathway. Additionally, we examine the role of galectins in liquid-liquid phase separation on membranes, which may facilitate their diverse functions in cellular responses. Through this approach, we aim to re-evaluate the complex and diverse biological roles of galectins in host defense.
Collapse
Affiliation(s)
- Sachiko Sato
- Axe of Infectious and Immune Diseases, CHU de Quebec-Université Laval Research Centre, Faculty of Medicine, and Research Centre for Infectious Diseases, Laval University, Quebec City, Canada.
| | - Jun Iwaki
- Tokyo Chemical Industry Co., Ltd., Tokyo, Japan.
| | - Jun Hirabayashi
- Institute for Glyco-core Research, Nagoya University, Tokai Higher Education and Research System, Nagoya, Japan.
| |
Collapse
|
3
|
Jia Q, Yang Y, Yao S, Chen X, Hu Z. Emerging Roles of Galectin-3 in Pulmonary Diseases. Lung 2024; 202:385-403. [PMID: 38850292 DOI: 10.1007/s00408-024-00709-y] [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] [Received: 03/22/2024] [Accepted: 05/24/2024] [Indexed: 06/10/2024]
Abstract
Galectin-3 is a multifunctional protein that is involved in various physiological and pathological events. Emerging evidence suggests that galectin-3 also plays a critical role in the pathogenesis of pulmonary diseases. Galectin-3 can be produced and secreted by various cell types in the lungs, and the overexpression of galectin-3 has been found in acute lung injury/acute respiratory distress syndrome (ALI/ARDS), pulmonary hypertension (PH), pulmonary fibrosis diseases, lung cancer, lung infection, chronic obstructive pulmonary disease (COPD), and asthma. Galectin-3 exerts diverse effects on the inflammatory response, immune cell activation, fibrosis and tissue remodeling, and tumorigenesis in these pulmonary disorders, and genetic and pharmacologic modulation of galectin-3 has therapeutic effects on the treatment of pulmonary illnesses. In this review, we summarize the structure and function of galectin-3 and the underlying mechanisms of galectin-3 in pulmonary disease pathologies; we also discuss preclinical and clinical evidence regarding the therapeutic potential of galectin-3 inhibitors in these pulmonary disorders. Additionally, targeting galectin-3 may be a very promising therapeutic approach for the treatment of pulmonary diseases.
Collapse
Affiliation(s)
- Qi Jia
- Department of Anesthesiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Institute of Anesthesia and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Key Laboratory of Anesthesiology and Resuscitation (Huazhong University of Science and Technology), Ministry of Education, Wuhan, China
| | - Yiyi Yang
- Department of Anesthesiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Institute of Anesthesia and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Key Laboratory of Anesthesiology and Resuscitation (Huazhong University of Science and Technology), Ministry of Education, Wuhan, China
| | - Shanglong Yao
- Department of Anesthesiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Institute of Anesthesia and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Key Laboratory of Anesthesiology and Resuscitation (Huazhong University of Science and Technology), Ministry of Education, Wuhan, China
| | - Xiangdong Chen
- Department of Anesthesiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Institute of Anesthesia and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Key Laboratory of Anesthesiology and Resuscitation (Huazhong University of Science and Technology), Ministry of Education, Wuhan, China
| | - Zhiqiang Hu
- Department of Anesthesiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.
- Institute of Anesthesia and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.
- Key Laboratory of Anesthesiology and Resuscitation (Huazhong University of Science and Technology), Ministry of Education, Wuhan, China.
| |
Collapse
|
4
|
Das S, Kaminski TW, Schlegel BT, Bain W, Hu S, Patel A, Kale SL, Chen K, Lee JS, Mallampalli RK, Kagan VE, Rajasundaram D, McVerry BJ, Sundd P, Kitsios GD, Ray A, Ray P. Neutrophils and galectin-3 defend mice from lethal bacterial infection and humans from acute respiratory failure. Nat Commun 2024; 15:4724. [PMID: 38830855 PMCID: PMC11148175 DOI: 10.1038/s41467-024-48796-y] [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] [Received: 08/16/2023] [Accepted: 05/14/2024] [Indexed: 06/05/2024] Open
Abstract
Respiratory infection by Pseudomonas aeruginosa, common in hospitalized immunocompromised and immunocompetent ventilated patients, can be life-threatening because of antibiotic resistance. This raises the question of whether the host's immune system can be educated to combat this bacterium. Here we show that prior exposure to a single low dose of lipopolysaccharide (LPS) protects mice from a lethal infection by P. aeruginosa. LPS exposure trained the innate immune system by promoting expansion of neutrophil and interstitial macrophage populations distinguishable from other immune cells with enrichment of gene sets for phagocytosis- and cell-killing-associated genes. The cell-killing gene set in the neutrophil population uniquely expressed Lgals3, which encodes the multifunctional antibacterial protein, galectin-3. Intravital imaging for bacterial phagocytosis, assessment of bacterial killing and neutrophil-associated galectin-3 protein levels together with use of galectin-3-deficient mice collectively highlight neutrophils and galectin-3 as central players in LPS-mediated protection. Patients with acute respiratory failure revealed significantly higher galectin-3 levels in endotracheal aspirates (ETAs) of survivors compared to non-survivors, galectin-3 levels strongly correlating with a neutrophil signature in the ETAs and a prognostically favorable hypoinflammatory plasma biomarker subphenotype. Taken together, our study provides impetus for harnessing the potential of galectin-3-expressing neutrophils to protect from lethal infections and respiratory failure.
Collapse
Affiliation(s)
- Sudipta Das
- Division of Pulmonary, Allergy, Critical Care, and Sleep Medicine and Acute Lung Injury Center of Excellence, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA, 15213, USA
| | - Tomasz W Kaminski
- VERSITI Blood Research Institute and Medical College of Wisconsin, Milwaukee, WI, 53233, USA
| | - Brent T Schlegel
- Department of Pediatrics, Division of Health Informatics, University of Pittsburgh School of Medicine, Pittsburgh, PA, 15224, USA
| | - William Bain
- Division of Pulmonary, Allergy, Critical Care, and Sleep Medicine and Acute Lung Injury Center of Excellence, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA, 15213, USA
- Veteran's Affairs Pittsburgh Healthcare System, Pittsburgh, PA, 15240, USA
| | - Sanmei Hu
- Division of Pulmonary, Allergy, Critical Care, and Sleep Medicine and Acute Lung Injury Center of Excellence, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA, 15213, USA
| | - Akruti Patel
- Division of Pulmonary, Allergy, Critical Care, and Sleep Medicine and Acute Lung Injury Center of Excellence, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA, 15213, USA
| | - Sagar L Kale
- Division of Pulmonary, Allergy, Critical Care, and Sleep Medicine and Acute Lung Injury Center of Excellence, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA, 15213, USA
| | - Kong Chen
- Division of Pulmonary, Allergy, Critical Care, and Sleep Medicine and Acute Lung Injury Center of Excellence, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA, 15213, USA
| | - Janet S Lee
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Rama K Mallampalli
- Department of Medicine, The Ohio State University (OSU), Columbus, OH, 43210, USA
| | - Valerian E Kagan
- Department of Environmental and Occupational Health, University of Pittsburgh, Pittsburgh, PA, 15261, USA
| | - Dhivyaa Rajasundaram
- Department of Pediatrics, Division of Health Informatics, University of Pittsburgh School of Medicine, Pittsburgh, PA, 15224, USA
| | - Bryan J McVerry
- Division of Pulmonary, Allergy, Critical Care, and Sleep Medicine and Acute Lung Injury Center of Excellence, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA, 15213, USA
| | - Prithu Sundd
- VERSITI Blood Research Institute and Medical College of Wisconsin, Milwaukee, WI, 53233, USA
| | - Georgios D Kitsios
- Division of Pulmonary, Allergy, Critical Care, and Sleep Medicine and Acute Lung Injury Center of Excellence, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA, 15213, USA
| | - Anuradha Ray
- Division of Pulmonary, Allergy, Critical Care, and Sleep Medicine and Acute Lung Injury Center of Excellence, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA, 15213, USA
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA, 15213, USA
| | - Prabir Ray
- Division of Pulmonary, Allergy, Critical Care, and Sleep Medicine and Acute Lung Injury Center of Excellence, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA, 15213, USA.
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA, 15213, USA.
| |
Collapse
|
5
|
Suárez JAG, Calumby RJN, Silva DP, Barbosa VT, Maranhão FCA, Moreira IF, Melhem MSC, Moreira RTF. Neonatal innate immunity response in invasive candidiasis. BRAZ J BIOL 2024; 84:e275155. [PMID: 38808781 DOI: 10.1590/1519-6984.275155] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2023] [Accepted: 03/18/2024] [Indexed: 05/30/2024] Open
Abstract
Infections caused by Candida spp. are frequent in critically hospitalized patients, especially among premature neonates, representing one of the most common healthcare-related infections. Although there is considerable production of current knowledge about the mechanisms of immune response, aspects involved in the newborn's innate defense are not fully understood. The aim of this study was to describe the innate immune mechanisms involved in the defense of neonates against invasive candidiasis. This is an integrative literature review from the Scopus, Scifinder, Medline, Web of Science databases and the electronic libraries ScienceDirect and Scielo, in the period between 2002 and 2020, with rescue based on primary descriptor Immunity Innate plus secondary descriptors Candidiasis Invasive AND Infant Newborn. We have observed the involvement of various mechanisms in the neonatal response against invasive candidiasis, including the recognition, signaling, recruitment, and initiation of an effective immune response. These mechanisms encompass the presence of antimicrobial peptides, phagocytosis, synthesis of reactive oxygen species, inflammatory mediators, and complex cell signaling systems mediated by Pattern Recognition Receptors (PRRs). With this study, it is expected to contribute to the expansion of knowledge about the immunological mechanisms involved in the innate immune response of the newborn against disseminated infections caused by Candida species, and in the same sense, highlight the importance of this knowledge as a reflex in the decrease in mortality in the neonatal period.
Collapse
Affiliation(s)
- J A G Suárez
- Universidade Federal de Ouro Preto - UFOP, Ouro Preto, MG, Brasil
| | - R J N Calumby
- Universidade Federal de Alagoas - UFAL, Campus A. C. Simões, Maceió, AL, Brasil
| | - D P Silva
- Universidade Federal de Alagoas - UFAL, Campus A. C. Simões, Maceió, AL, Brasil
| | - V T Barbosa
- Universidade Federal de Alagoas - UFAL, Campus A. C. Simões, Maceió, AL, Brasil
| | - F C A Maranhão
- Universidade Federal de Alagoas - UFAL, Campus A. C. Simões, Maceió, AL, Brasil
| | - I F Moreira
- Universidade Federal de Alagoas - UFAL, Campus A. C. Simões, Maceió, AL, Brasil
| | - M S C Melhem
- Universidade Federal de Mato Grosso do Sul - UFMS, Faculdade de Medicina, Programa de Pós Graduação em Doenças Infecciosas, Campo Grande, MS, Brasil
| | - R T F Moreira
- Universidade Federal de Alagoas - UFAL, Campus A. C. Simões, Maceió, AL, Brasil
- Universidade Federal de Mato Grosso do Sul - UFMS, Faculdade de Medicina, Programa de Pós Graduação em Doenças Infecciosas, Campo Grande, MS, Brasil
| |
Collapse
|
6
|
Hong S, Kim J, Jung K, Ahn M, Moon C, Nomura Y, Matsuda H, Tanaka A, Jeong H, Shin T. Histopathological evaluation of the lungs in experimental autoimmune encephalomyelitis. J Vet Sci 2024; 25:e35. [PMID: 38834505 PMCID: PMC11156594 DOI: 10.4142/jvs.23302] [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] [Received: 12/01/2023] [Revised: 02/29/2024] [Accepted: 03/07/2024] [Indexed: 06/06/2024] Open
Abstract
IMPORTANCE Experimental autoimmune encephalomyelitis (EAE) is an animal model of multiple sclerosis characterized by inflammation within the central nervous system. However, inflammation in non-neuronal tissues, including the lungs, has not been fully evaluated. OBJECTIVE This study evaluated the inflammatory response in lungs of EAE mice by immunohistochemistry and histochemistry. METHODS Eight adult C57BL/6 mice were injected with myelin oligodendrocyte glycoprotein35-55 to induce the EAE. Lungs and spinal cords were sampled from the experimental mice at the time of sacrifice and used for the western blotting, histochemistry, and immunohistochemistry. RESULTS Histopathological examination revealed inflammatory lesions in the lungs of EAE mice, characterized by infiltration of myeloperoxidase (MPO)- and galectin-3-positive cells, as determined by immunohistochemistry. Increased numbers of collagen fibers in the lungs of EAE mice were confirmed by histopathological analysis. Western blotting revealed significantly elevated level of osteopontin (OPN), cluster of differentiation 44 (CD44), MPO and galectin-3 in the lungs of EAE mice compared with normal controls (p < 0.05). Immunohistochemical analysis revealed both OPN and CD44 in ionized calcium-binding adapter molecule 1-positive macrophages within the lungs of EAE mice. CONCLUSIONS AND RELEVANCE Taken together, these findings suggest that the increased OPN level in lungs of EAE mice led to inflammation; concurrent increases in proinflammatory factors (OPN and galectin-3) caused pulmonary impairment.
Collapse
Affiliation(s)
- Sungmoo Hong
- College of Veterinary Medicine and Veterinary Medical Research Institute, Jeju National University, Jeju 63243, Korea
| | - Jeongtae Kim
- Department of Anatomy, Kosin University College of Medicine, Busan 49267, Korea
| | - Kyungsook Jung
- Functional Biomaterials Research Center, Korea Research Institute of Bioscience and Biotechnology, Jeongeup 56212, Korea
| | - Meejung Ahn
- Department of Animal Science, College of Life Science, Sangji University, Wonju 26339, Korea
| | - Changjong Moon
- Department of Veterinary Anatomy and Animal Behavior, College of Veterinary Medicine and BK21 FOUR Program, Chonnam National University, Gwangju 61186, Korea
| | - Yoshihiro Nomura
- Scleroprotein and Leather Research Institute, Faculty of Agriculture, Tokyo University of Agriculture and Technology, Tokyo 183-8509, Japan
| | - Hiroshi Matsuda
- Laboratory of Comparative Animal Medicine, Division of Animal Life Science, Faculty of Agriculture, Tokyo University of Agriculture and Technology, Tokyo 183-8509, Japan
| | - Akane Tanaka
- Laboratory of Comparative Animal Medicine, Division of Animal Life Science, Faculty of Agriculture, Tokyo University of Agriculture and Technology, Tokyo 183-8509, Japan
| | - Hyohoon Jeong
- College of Veterinary Medicine and Veterinary Medical Research Institute, Jeju National University, Jeju 63243, Korea.
| | - Taekyun Shin
- College of Veterinary Medicine and Veterinary Medical Research Institute, Jeju National University, Jeju 63243, Korea.
| |
Collapse
|
7
|
Omo-Lamai S, Wang Y, Patel MN, Essien EO, Shen M, Majumdar A, Espy C, Wu J, Channer B, Tobin M, Murali S, Papp TE, Maheshwari R, Wang L, Chase LS, Zamora ME, Arral ML, Marcos-Contreras OA, Myerson JW, Hunter CA, Tsourkas A, Muzykantov V, Brodsky I, Shin S, Whitehead KA, Gaskill P, Discher D, Parhiz H, Brenner JS. Lipid Nanoparticle-Associated Inflammation is Triggered by Sensing of Endosomal Damage: Engineering Endosomal Escape Without Side Effects. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.04.16.589801. [PMID: 38659905 PMCID: PMC11042321 DOI: 10.1101/2024.04.16.589801] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/26/2024]
Abstract
Lipid nanoparticles (LNPs) have emerged as the dominant platform for RNA delivery, based on their success in the COVID-19 vaccines and late-stage clinical studies in other indications. However, we and others have shown that LNPs induce severe inflammation, and massively aggravate pre-existing inflammation. Here, using structure-function screening of lipids and analyses of signaling pathways, we elucidate the mechanisms of LNP-associated inflammation and demonstrate solutions. We show that LNPs' hallmark feature, endosomal escape, which is necessary for RNA expression, also directly triggers inflammation by causing endosomal membrane damage. Large, irreparable, endosomal holes are recognized by cytosolic proteins called galectins, which bind to sugars on the inner endosomal membrane and then regulate downstream inflammation. We find that inhibition of galectins abrogates LNP-associated inflammation, both in vitro and in vivo . We show that rapidly biodegradable ionizable lipids can preferentially create endosomal holes that are smaller in size and reparable by the endosomal sorting complex required for transport (ESCRT) pathway. Ionizable lipids producing such ESCRT-recruiting endosomal holes can produce high expression from cargo mRNA with minimal inflammation. Finally, we show that both routes to non-inflammatory LNPs, either galectin inhibition or ESCRT-recruiting ionizable lipids, are compatible with therapeutic mRNAs that ameliorate inflammation in disease models. LNPs without galectin inhibition or biodegradable ionizable lipids lead to severe exacerbation of inflammation in these models. In summary, endosomal escape induces endosomal membrane damage that can lead to inflammation. However, the inflammation can be controlled by inhibiting galectins (large hole detectors) or by using biodegradable lipids, which create smaller holes that are reparable by the ESCRT pathway. These strategies should lead to generally safer LNPs that can be used to treat inflammatory diseases.
Collapse
|
8
|
Plattner K, Bachmann MF, Vogel M. On the complexity of IgE: The role of structural flexibility and glycosylation for binding its receptors. FRONTIERS IN ALLERGY 2023; 4:1117611. [PMID: 37056355 PMCID: PMC10089267 DOI: 10.3389/falgy.2023.1117611] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2022] [Accepted: 03/13/2023] [Indexed: 03/30/2023] Open
Abstract
It is well established that immunoglobulin E (IgE) plays a crucial role in atopy by binding to two types of Fcε receptors (FcεRI and FcεRII, also known as CD23). The cross-linking of FcεRI-bound IgE on effector cells, such as basophils and mast cells, initiates the allergic response. Conversely, the binding of IgE to CD23 modulates IgE serum levels and antigen presentation. In addition to binding to FcεRs, IgE can also interact with other receptors, such as certain galectins and, in mice, some FcγRs. The binding strength of IgE to its receptors is affected by its valency and glycosylation. While FcεRI shows reduced binding to IgE immune complexes (IgE-ICs), the binding to CD23 is enhanced. There is no evidence that galectins bind IgE-ICs. On the other hand, IgE glycosylation plays a crucial role in the binding to FcεRI and galectins, whereas the binding to CD23 seems to be independent of glycosylation. In this review, we will focus on receptors that bind to IgE and examine how the glycosylation and complexation of IgE impact their binding.
Collapse
Affiliation(s)
- Kevin Plattner
- Department of Immunology, University Clinic for Rheumatology and Immunology, University of Bern, Bern, Switzerland
- Department of Biomedical Research Bern (DBMR), University of Bern, Bern, Switzerland
| | - Martin F. Bachmann
- Department of Immunology, University Clinic for Rheumatology and Immunology, University of Bern, Bern, Switzerland
- Department of Biomedical Research Bern (DBMR), University of Bern, Bern, Switzerland
- Nuffield Department of Medicine, The Jenner Institute, University of Oxford, Oxford, United Kingdom
| | - Monique Vogel
- Department of Immunology, University Clinic for Rheumatology and Immunology, University of Bern, Bern, Switzerland
- Department of Biomedical Research Bern (DBMR), University of Bern, Bern, Switzerland
- Correspondence: Monique Vogel
| |
Collapse
|
9
|
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
|
10
|
Čoma M, Manning JC, Kaltner H, Gál P. The sweet side of wound healing: galectins as promising therapeutic targets in hemostasis, inflammation, proliferation, and maturation/remodeling. Expert Opin Ther Targets 2023; 27:41-53. [PMID: 36716023 DOI: 10.1080/14728222.2023.2175318] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
INTRODUCTION Understanding the molecular and cellular processes involved in skin wound healing may pave the way for the development of innovative approaches to transforming the identified natural effectors into therapeutic tools. Based on the extensive involvement of the ga(lactoside-binding)lectin family in (patho)physiological processes, it has been well established that galectins are involved in a wide range of cell-cell and cell-matrix interactions. AREAS COVERED In the present paper, we provide an overview of the biological role of galectins in repair and regeneration, focusing on four main phases (hemostasis, inflammation, proliferation, and maturation/remodeling) of skin repair using basic wound models (open excision vs. sutured incision). EXPERT OPINION The reported data make a strong case for directing further efforts to treat excisional and incisional wounds differently. Functions of galectins essentially result from their modular presentation. In fact, Gal-1 seems to play a role in the early phases of healing (anti-inflammatory) and wound contraction, Gal-3 accelerates re-epithelization and increases tensile strength (scar inductor). Galectins have also become subject of redesigning by engineering to optimize the activity. Clinically relevant, these new tools derived from the carbohydrate recognition domain platform may also prove helpful for other purposes, such as potent antibacterial agglutinins and opsonins.
Collapse
Affiliation(s)
- Matúš Čoma
- Department of Biomedical Research, East-Slovak Institute of Cardiovascular Diseases Inc, Košice, Slovak Republic.,Department of Pharmacology, Faculty of Medicine, Pavol Jozef Šafárik University, Košice, Slovak Republic
| | - Joachim C Manning
- Institute of Physiological Chemistry, Faculty of Veterinary Medicine, Ludwig-Maximilian University, Munich, Germany
| | - Herbert Kaltner
- Institute of Physiological Chemistry, Faculty of Veterinary Medicine, Ludwig-Maximilian University, Munich, Germany
| | - Peter Gál
- Department of Biomedical Research, East-Slovak Institute of Cardiovascular Diseases Inc, Košice, Slovak Republic.,Department of Pharmacology, Faculty of Medicine, Pavol Jozef Šafárik University, Košice, Slovak Republic.,Prague Burn Center, Third Faculty of Medicine, Charles University and University Hospital Královske Vinohrady, Prague, Czech Republic.,Department of Pharmacognosy and Botany, Faculty of Pharmacy, Comenius University, Bratislava, Slovak Republic
| |
Collapse
|
11
|
Karsli E, Anabarli Metin D, Canacik O, Sabirli R, Kaymaz B, Kurt O, Koseler A. Galectin-3 as a Potential Prognostic Biomarker for COVID-19 Disease: A Case-Control Study. Cureus 2022; 14:e28805. [PMID: 36225452 PMCID: PMC9534518 DOI: 10.7759/cureus.28805] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/05/2022] [Indexed: 12/15/2022] Open
Abstract
Background Recent studies have investigated the importance of Galetin-3 in inflammation, fibrosis, cell proliferation, cardiac disease, diabetes, and tumor formation. Aims This study aims to investigate the role of the Galectin-3 level in the diagnosis of COVID-19 pneumonia and the value of the Galectin-3 level in predicting the clinical course of the patient. Methods This study employed a prospective, case-control study design and was conducted at Bakircay University Ciğli Training and Research Hospital. A total of 100 patients (40 had moderate and 60 had severe/critical COVID-19 disease according to World Health Organisation guidelines) and 50 non-symptomatic healthy volunteers participated in the study. Blood samples were taken from patients at the time of hospital admission, after which serum was isolated. Following the isolation of serum, Galectin-3 levels were evaluated using the enzyme-linked immunosorbent assay (ELISA) method. Results The serum Galectin-3 level was measured as 13.57 (10.9-16.4) ng/mL in the control group, 13.52 (10.69-16.6) ng/mL in the moderate disease group, and 11.65 (6.09-14.33) ng/mL in the severe/critical disease group. Serum Galectin-3 levels were significantly lower in the severe/critical disease group compared to the control and moderate disease groups (p=0.001 and p=0.019, respectively). Using ROC analysis, a larger area under the curve (AUC) for the serum Galectin-3 levels of the control group (AUC=0.622, 95% CI =0.529-0.714; p=0.015) was calculated compared to the COVID-19 patient group for the diagnosis of COVID-19 disease. The Galectin-3 level was found to be 75% sensitive and 50% specific at a cut-off level of 11.3 ng/mL in predicting the need for ICU treatment. Conclusion Galectin-3 levels may be a beneficial biomarker in predicting the clinical severity of COVID-19 disease when used in conjunction with other known biomarkers, at the time of admission to the emergency department (ED).
Collapse
|
12
|
Humphries DC, Mills R, Boz C, McHugh BJ, Hirani N, Rossi AG, Pedersen A, Schambye HT, Slack RJ, Leffler H, Nilsson UJ, Wang W, Sethi T, Mackinnon AC. Galectin-3 inhibitor GB0139 protects against acute lung injury by inhibiting neutrophil recruitment and activation. Front Pharmacol 2022; 13:949264. [PMID: 36003515 PMCID: PMC9393216 DOI: 10.3389/fphar.2022.949264] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2022] [Accepted: 06/30/2022] [Indexed: 11/13/2022] Open
Abstract
Rationale: Galectin-3 (Gal-3) drives fibrosis during chronic lung injury, however, its role in acute lung injury (ALI) remains unknown. Effective pharmacological therapies available for ALI are limited; identifying novel concepts in treatment is essential. GB0139 is a Gal-3 inhibitor currently under clinical investigation for the treatment of idiopathic pulmonary fibrosis. We investigate the role of Gal-3 in ALI and evaluate whether its inhibition with GB0139 offers a protective role. The effect of GB0139 on ALI was explored in vivo and in vitro. Methods: The pharmacokinetic profile of intra-tracheal (i.t.) GB0139 was investigated in C57BL/6 mice to support the daily dosing regimen. GB0139 (1–30 µg) was then assessed following acute i.t. lipopolysaccharide (LPS) and bleomycin administration. Histology, broncho-alveolar lavage fluid (BALf) analysis, and flow cytometric analysis of lung digests and BALf were performed. The impact of GB0139 on cell activation and apoptosis was determined in vitro using neutrophils and THP-1, A549 and Jurkat E6 cell lines. Results: GB0139 decreased inflammation severity via a reduction in neutrophil and macrophage recruitment and neutrophil activation. GB0139 reduced LPS-mediated increases in interleukin (IL)-6, tumor necrosis factor alpha (TNFα) and macrophage inflammatory protein-1-alpha. In vitro, GB0139 inhibited Gal-3-induced neutrophil activation, monocyte IL-8 secretion, T cell apoptosis and the upregulation of pro-inflammatory genes encoding for IL-8, TNFα, IL-6 in alveolar epithelial cells in response to mechanical stretch. Conclusion: These data indicate that Gal-3 adopts a pro-inflammatory role following the early stages of lung injury and supports the development of GB0139, as a potential treatment approach in ALI.
Collapse
Affiliation(s)
- Duncan C. Humphries
- Centre for Inflammation Research, University of Edinburgh, Edinburgh, United Kingdom
- Galecto Inc. Nine Edinburgh BioQuarter, Edinburgh, United Kingdom
| | - Ross Mills
- Centre for Inflammation Research, University of Edinburgh, Edinburgh, United Kingdom
| | - Cecilia Boz
- Centre for Inflammation Research, University of Edinburgh, Edinburgh, United Kingdom
| | - Brian J. McHugh
- Centre for Inflammation Research, University of Edinburgh, Edinburgh, United Kingdom
| | - Nikhil Hirani
- Centre for Inflammation Research, University of Edinburgh, Edinburgh, United Kingdom
| | - Adriano G. Rossi
- Centre for Inflammation Research, University of Edinburgh, Edinburgh, United Kingdom
| | | | | | | | - Hakon Leffler
- Department of Laboratory Medicine, Lund University, Lund, Sweden
| | | | - Wei Wang
- Department of Asthma, Allergy and Respiratory Science, King’s College London, Guy’s Hospital, London, United Kingdom
| | - Tariq Sethi
- Galecto Inc, Copenhagen, Denmark
- Department of Asthma, Allergy and Respiratory Science, King’s College London, Guy’s Hospital, London, United Kingdom
| | - Alison C. Mackinnon
- Centre for Inflammation Research, University of Edinburgh, Edinburgh, United Kingdom
- Galecto Inc. Nine Edinburgh BioQuarter, Edinburgh, United Kingdom
- *Correspondence: Alison C. Mackinnon,
| |
Collapse
|
13
|
Galectin-3 Is a Crucial Immunological Disease Marker in Patients with Fungal Keratitis. DISEASE MARKERS 2022; 2022:1380560. [PMID: 35845133 PMCID: PMC9286934 DOI: 10.1155/2022/1380560] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/24/2021] [Revised: 05/18/2022] [Accepted: 06/01/2022] [Indexed: 11/17/2022]
Abstract
Fungal keratitis, one of the most common infectious eye diseases in China, often results in a poor prognosis due to a delayed diagnosis and the insufficiency of effective therapy. There is an urgent need to identify specific biomarkers for the disease. In this study, we screened out tear proteins in patients with fungal keratitis by microsphere-based immunoassay analysis. Levels of cytokine expression were determined in both human corneal epithelial cell models in vitro and the corneas of patients by western blot, quantitative polymerase chain reaction (qPCR), and immunofluorescence analysis. Neutrophil activation was examined by flow cytometry analysis. The relationship between the cytokine expression and neutrophils was evaluated by immunofluorescence costaining and correlation analysis. These results demonstrated that the galectin-3 expression level was increased in both cell model and patient samples at the early and late stages of fungal keratitis. The neutrophils were significantly activated during the disease course of fungal keratitis. Meanwhile, colocalization and a positive correlation between galectin-3 and neutrophils were observed, suggesting that galectin-3 may play a crucial role in the recruitment of neutrophils and immune regulation of fungal keratitis. In conclusion, galectin-3 could be a key disease marker implying a beneficial immune response in the pathogenesis of fungal keratitis, which might be a target of therapeutic strategy in the future.
Collapse
|
14
|
Ayona D, Zarza SM, Landemarre L, Roubinet B, Decloquement P, Raoult D, Fournier PE, Desnues B. Human galectin-1 and galectin-3 promote Tropheryma whipplei infection. Gut Microbes 2022; 13:1-15. [PMID: 33573443 PMCID: PMC7889132 DOI: 10.1080/19490976.2021.1884515] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Abstract
Tropheryma whipplei, is an actinobacterium that causes different infections in humans, including Whipple's disease. The bacterium infects and replicates in macrophages, leading to a Th2-biased immune response. Previous studies have shown that T. whipplei harbors complex surface glycoproteins with evidence of sialylation. However, the exact contribution of these glycoproteins for infection and survival remains obscure. To address this, we characterized the bacterial glycoprofile and evaluated the involvement of human β-galactoside-binding lectins, Galectin-1 (Gal-1) and Galectin-3 (Gal-3) which are highly expressed by macrophages as receptors for bacterial glycans. Tropheryma whipplei glycoproteins harbor different sugars including glucose, mannose, fucose, β-galactose and sialic acid. Mass spectrometry identification revealed that these glycoproteins were membrane- and virulence-associated glycoproteins. Most of these glycoproteins are highly sialylated and N-glycosylated while some of them are rich in poly-N-acetyllactosamine (Poly-LAcNAc) and bind Gal-1 and Gal-3. In vitro, T. whipplei modulates the expression and cellular distribution of Gal-1 and Gal-3. Although both galectins promote T. whipplei infection by enhancing bacterial cell entry, only Gal-3 is required for optimal bacterial uptake. Finally, we found that serum levels of Gal-1 and Gal-3 were altered in patients with T. whipplei infections as compared to healthy individuals, suggesting that galectins are also involved in vivo. Among T. whipplei membrane-associated proteins, poly-LacNAc rich-glycoproteins promote infection through interaction with galectins. T. whipplei modulates the expression of Gal-1 and Gal-3 both in vitro and in vivo. Drugs interfering with galectin-glycan interactions may provide new avenues for the treatment and diagnosis of T. whipplei infections.
Collapse
Affiliation(s)
- Diyoly Ayona
- Aix Marseille Univ, IRD, APHM, MEPHI, Marseille, France,IHU-Méditerranée Infection, Marseille, France
| | - Sandra Madariaga Zarza
- Aix Marseille Univ, IRD, APHM, MEPHI, Marseille, France,IHU-Méditerranée Infection, Marseille, France
| | | | - Benoît Roubinet
- Glycodiag, Rue De Chartres, BP6759, 45067, Orléans cedex 2, France
| | - Philippe Decloquement
- Aix Marseille Univ, IRD, APHM, MEPHI, Marseille, France,IHU-Méditerranée Infection, Marseille, France
| | - Didier Raoult
- Aix Marseille Univ, IRD, APHM, MEPHI, Marseille, France,IHU-Méditerranée Infection, Marseille, France
| | - Pierre-Edouard Fournier
- IHU-Méditerranée Infection, Marseille, France,Aix Marseille Univ, IRD, APHM, VITROME, Marseille, France,Pierre-Edouard Fournier Aix Marseille Univ, VITROME, IHU - Méditerranée Infection, 19-21 Boulevard Jean Moulin, 13005Marseille, France
| | - Benoit Desnues
- Aix Marseille Univ, IRD, APHM, MEPHI, Marseille, France,IHU-Méditerranée Infection, Marseille, France,CONTACT Benoit Desnues MEPHI, IHU - Méditerranée Infection, Aix Marseille Univ, 19-21 Boulevard Jean Moulin, 13005, Marseille, France
| |
Collapse
|
15
|
Humphries DC, Mills R, Dobie R, Henderson NC, Sethi T, Mackinnon AC. Selective Myeloid Depletion of Galectin-3 Offers Protection Against Acute and Chronic Lung Injury. Front Pharmacol 2021; 12:715986. [PMID: 34526900 PMCID: PMC8435800 DOI: 10.3389/fphar.2021.715986] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2021] [Accepted: 08/18/2021] [Indexed: 12/03/2022] Open
Abstract
Rationale: Galectin-3 (Gal-3) is an immune regulator and an important driver of fibrosis in chronic lung injury, however, its role in acute lung injury (ALI) remains unknown. Previous work has shown that global deletion of galectin-3 reduces collagen deposition in a bleomycin-induced pulmonary fibrosis model (MacKinnon et al., Am. J. Respir. Crit. Care Med., 2012, 185, 537–46). An inhaled Gal-3 inhibitor, GB0139, is undergoing Phase II clinical development for idiopathic pulmonary fibrosis (IPF). This work aims to elucidate the role of Gal-3 in the myeloid and mesenchymal compartment on the development of acute and chronic lung injury. Methods:LgalS3fl/fl mice were generated and crossed with mice expressing the myeloid (LysM) and mesenchymal (Pdgfrb) cre drivers to yield LysM-cre+/-/LgalS3fl/fl and Pdgfrb-cre+/-/LgalS3fl/fl mice. The response to acute (bleomycin or LPS) or chronic (bleomycin) lung injury was compared to globally deficient Gal-3−/− mice. Results: Myeloid depletion of Gal-3 led to a significant reduction in Gal-3 expression in alveolar macrophages and neutrophils and a reduction in neutrophil recruitment into the interstitium but not into the alveolar space. The reduction in interstitial neutrophils corelated with decreased levels of pulmonary inflammation following acute bleomycin and LPS administration. In addition, myeloid deletion decreased Gal-3 levels in bronchoalveolar lavage (BAL) and reduced lung fibrosis induced by chronic bleomycin. In contrast, no differences in BAL Gal-3 levels or fibrosis were observed in Pdgfrb-cre+/-/LgalS3fl/flmice. Conclusions: Myeloid cell derived Galectin-3 drives acute and chronic lung inflammation and supports direct targeting of galectin-3 as an attractive new therapy for lung inflammation.
Collapse
Affiliation(s)
- Duncan C Humphries
- Centre for Inflammation Research, University of Edinburgh, Edinburgh, United Kingdom
| | - Ross Mills
- Centre for Inflammation Research, University of Edinburgh, Edinburgh, United Kingdom
| | - Ross Dobie
- Centre for Inflammation Research, University of Edinburgh, Edinburgh, United Kingdom
| | - Neil C Henderson
- Centre for Inflammation Research, University of Edinburgh, Edinburgh, United Kingdom
| | | | - Alison C Mackinnon
- Centre for Inflammation Research, University of Edinburgh, Edinburgh, United Kingdom.,Galecto Inc, Copenhagen, Denmark
| |
Collapse
|
16
|
Zinserling VA, Swistunov VV, Botvinkin AD, Stepanenko LA, Makarova AE. Lobar (croupous) pneumonia: old and new data. Infection 2021; 50:235-242. [PMID: 34472009 PMCID: PMC8409273 DOI: 10.1007/s15010-021-01689-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2021] [Accepted: 08/23/2021] [Indexed: 01/20/2023]
Abstract
Background and aim Pneumonia remains one of the most frequent death causes worldwide. Among the etiological factors S. pneumoniae-causing lobar pneumonia plays a leading role. According to current textbook knowledge at least three sequential stages of lobar pneumonia are distinguished: congestion, red hepatization and gray hepatization. However, there are no detailed data supporting this stage concept. There are also controversial views on its etiology. In this study, the lung changes in lobar pneumonia were related to the cause and duration of the disease. In addition, the complications of the disease were evaluated. PCR studies verified the etiology of pneumonia. Material and methods Lobar pneumonia was analyzed in 252 post mortem cases examined in a large hospital in Irkutsk. The pathology, etiology of pneumonia, course of disease and cause of death were recorded and correlated to its clinical course and duration. In the second part of the study, the results in 95 patients were analyzed in detail and related to PCR findings. Results Most patients were adult men of low social status who showed signs of severe alcoholism. Lobar pneumonia was observed in 85% of the patients, while the remaining patients showed sublobar (“lobular”, focal) lung involvement. Histologically, three patterns of inflammation were observed, which in most patients occurred concurrently in different parts of the involved lobe: “congestion”, characterized by serous exudation with multiple cocci (41% of cases), “red hepatization” (41% of cases) and “gray hepatization” (100% of cases). The latter pattern was subdivided into three subgroups according to the ratio of fibrin—neutrophils and the presence of macrophages. The mean number of different histological patterns observed per patient was 3.8. There was no correlation between the inflammatory patterns and the duration of the disease. In 23% of the patients, the cause of death was of pulmonary origin, while the remaining patients died of extrapulmonary complications (i.e. acute heart failure 26%, acute vascular insufficiency 15% purulent meningitis 11–24.3%. In 29/95 patients (20 with lobar and 9 with focal pneumonia) pneumococcal etiology of pneumonia was established by PCR. Conclusion Lobar pneumonia is a distinct clinico-pathological entity caused by S. pneumoniae, demonstrated by PCR testing and/or cytological examinations. Bacteriologic studies frequently give falsenegative results. Lobar pneumonia is characterized by three main histopathological patterns (congestion or microbeous edema, and red and gray hepatization) which usually occur side by side and not in chronological order. Early death is often related to heart failure and septic shock, while meningitis is a frequent complication later in the course.
Collapse
Affiliation(s)
- Vsevolod A Zinserling
- Department of Pathomorphology, Institute of Experimental Medicine, V.A. Almazov Research Center, Saint Petersburg, Russia. .,Center of Infectious Pathology, S.P. Botkin Infectious Hospital, Saint-Petersburg, Russia.
| | - Vladimir V Swistunov
- Irkutsk's Cities Municipal Hospital, Irkutsk, Russia.,Irkutsk Medical University, Irkutsk, Russia
| | | | | | - Angelica E Makarova
- Irkutsk's Cities Municipal Hospital, Irkutsk, Russia.,Irkutsk Medical University, Irkutsk, Russia
| |
Collapse
|
17
|
I'm Infected, Eat Me! Innate Immunity Mediated by Live, Infected Cells Signaling To Be Phagocytosed. Infect Immun 2021; 89:IAI.00476-20. [PMID: 33558325 DOI: 10.1128/iai.00476-20] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Innate immunity against pathogens is known to be mediated by barriers to pathogen invasion, activation of complement, recruitment of immune cells, immune cell phagocytosis of pathogens, death of infected cells, and activation of the adaptive immunity via antigen presentation. Here, we propose and review evidence for a novel mode of innate immunity whereby live, infected host cells induce phagocytes to phagocytose the infected cell, thereby potentially reducing infection. We discuss evidence that host cells, infected by virus, bacteria, or other intracellular pathogens (i) release nucleotides and chemokines as find-me signals, (ii) expose on their surface phosphatidylserine and calreticulin as eat-me signals, (iii) release and bind opsonins to induce phagocytosis, and (iv) downregulate don't-eat-me signals CD47, major histocompatibility complex class I (MHC1), and sialic acid. As long as the pathogens of the host cell are destroyed within the phagocyte, then infection can be curtailed; if antigens from the pathogens are cross-presented by the phagocyte, then an adaptive response would also be induced. Phagocytosis of live infected cells may thereby mediate innate immunity.
Collapse
|
18
|
Li M, Chen YB, Liu F, Qu JQ, Ren LC, Chai J, Tang CE. Galectin‑3 facilitates the proliferation and migration of nasopharyngeal carcinoma cells via activation of the ERK1/2 and Akt signaling pathways, and is positively correlated with the inflammatory state of nasopharyngeal carcinoma. Mol Med Rep 2021; 23:370. [PMID: 33760180 PMCID: PMC7986014 DOI: 10.3892/mmr.2021.12009] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2020] [Accepted: 02/23/2021] [Indexed: 01/07/2023] Open
Abstract
Nasopharyngeal carcinoma (NPC) is an epithelial carcinoma originating from the nasopharyngeal mucosal tissue and is highly prevalent in southeast Asia. Galectin-3 (gal-3) serves crucial roles in many cancers but its role in NPC remains to be elucidated. The aim of the present study was to investigate the role of gal-3 in NPC. Immunohistochemistry and ELISA were used to determine the expression level of gal-3 in patients with NPC or chronic rhinitis (CR). Gal-3 short hairpin (sh)RNA was established to knockdown gal-3 in 5–8F and 6–10B cells, allowing for the evaluation of the roles of gal-3 in proliferation, migration and apoptosis in NPC cell lines. Immunohistochemistry staining of IL-6 and IL-8 was applied to access the inflammatory state of tumor tissues, and the correlation between the inflammatory state and gal-3 was analyzed. The results demonstrated that gal-3 was upregulated in patients with NPC compared with patients with CR. Knockdown of gal-3 inhibited proliferation and migration in 5-8F and 6-10B cells, as well as promoted apoptosis in these cells. The expression levels of MMP-9 and IL-8 were also decreased in 5-8F and 6-10B cells after transfection with gal-3 shRNA. A positive correlation was identified between the expression level of gal-3 and the inflammatory state of NPC. The phosphorylation levels of ERK1/2 and Akt were downregulated after knockdown of gal-3 in 5-8F and 6-10B cells. In conclusion, the expression level of gal-3 was upregulated in patients with NPC and was positively correlated with the inflammatory state of NPC. The results suggested that gal-3 promoted the proliferation and migration of 5-8F and 6-10B cells, while inhibiting the apoptosis of these cells. Moreover, activation of ERK1/2 and Akt may be the underlying mechanism of the effects of gal-3 on NPC.
Collapse
Affiliation(s)
- Mei Li
- Institute of Medical Science Research, Xiangya Hospital, Central South University, Changsha, Hunan 410008, P.R. China
| | - Yu Bin Chen
- Department of Cardiac Surgery, Xiangya Hospital, Central South University, Changsha, Hunan 410008, P.R. China
| | - Fen Liu
- Institute of Medical Science Research, Xiangya Hospital, Central South University, Changsha, Hunan 410008, P.R. China
| | - Jia Quan Qu
- Cholestatic Liver Diseases Center and Department of Gastroenterology, Southwest Hospital, Third Military Medical University, Chongqing 400038, P.R. China
| | - Li Cheng Ren
- Department of Burn and Reconstructive Surgery, Xiangya Hospital, Central South University, Changsha, Hunan 410008, P.R. China
| | - Jin Chai
- Cholestatic Liver Diseases Center and Department of Gastroenterology, Southwest Hospital, Third Military Medical University, Chongqing 400038, P.R. China
| | - Can E Tang
- Institute of Medical Science Research, Xiangya Hospital, Central South University, Changsha, Hunan 410008, P.R. China
| |
Collapse
|
19
|
Velickovic M, Arsenijevic A, Acovic A, Arsenijevic D, Milovanovic J, Dimitrijevic J, Todorovic Z, Milovanovic M, Kanjevac T, Arsenijevic N. Galectin-3, Possible Role in Pathogenesis of Periodontal Diseases and Potential Therapeutic Target. Front Pharmacol 2021; 12:638258. [PMID: 33815121 PMCID: PMC8017193 DOI: 10.3389/fphar.2021.638258] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2020] [Accepted: 02/11/2021] [Indexed: 12/11/2022] Open
Abstract
Periodontal diseases are chronic inflammatory diseases that occur due to the imbalance between microbial communities in the oral cavity and the immune response of the host that lead to destruction of tooth supporting structures and finally to alveolar bone loss. Galectin-3 is a β-galactoside-binding lectin with important roles in numerous biological processes. By direct binding to microbes and modulation of their clearence, Galectin-3 can affect the composition of microbial community in the oral cavity. Galectin-3 also modulates the function of many immune cells in the gingiva and gingival sulcus and thus can affect immune homeostasis. Few clinical studies demonstrated increased expression of Galectin-3 in different forms of periodontal diseases. Therefore, the objective of this mini review is to discuss the possible effects of Galectin-3 on the process of immune homeostasis and the balance between oral microbial community and host response and to provide insights into the potential therapeutic targeting of Gal-3 in periodontal disease.
Collapse
Affiliation(s)
- Milica Velickovic
- Department of Dentistry, Faculty of Medical Sciences, University of Kragujevac, Kragujevac, Serbia
| | - Aleksandar Arsenijevic
- Center for Molecular Medicine and Stem Cell Research, Faculty of Medical Sciences, University of Kragujevac, Kragujevac, Serbia
| | - Aleksandar Acovic
- Department of Dentistry, Faculty of Medical Sciences, University of Kragujevac, Kragujevac, Serbia
| | - Dragana Arsenijevic
- Department of Pharmacy, Faculty of Medical Sciences, University of Kragujevac, Kragujevac, Serbia
| | - Jelena Milovanovic
- Center for Molecular Medicine and Stem Cell Research, Faculty of Medical Sciences, University of Kragujevac, Kragujevac, Serbia.,Department of Histology and Embriology, Faculty of Medical Sciences, University of Kragujevac, Kragujevac, Serbia
| | - Jelena Dimitrijevic
- Center for Molecular Medicine and Stem Cell Research, Faculty of Medical Sciences, University of Kragujevac, Kragujevac, Serbia
| | - Zeljko Todorovic
- Department of Internal Medicine, Faculty of Medical Sciences, University of Kragujevac, Kragujevac, Serbia
| | - Marija Milovanovic
- Center for Molecular Medicine and Stem Cell Research, Faculty of Medical Sciences, University of Kragujevac, Kragujevac, Serbia
| | - Tatjana Kanjevac
- Department of Dentistry, Faculty of Medical Sciences, University of Kragujevac, Kragujevac, Serbia
| | - Nebojsa Arsenijevic
- Center for Molecular Medicine and Stem Cell Research, Faculty of Medical Sciences, University of Kragujevac, Kragujevac, Serbia
| |
Collapse
|
20
|
Insulin Receptor Substrate p53 Ameliorates High-Glucose-Induced Activation of NF- κB and Impaired Mobility of HUVECs. BIOMED RESEARCH INTERNATIONAL 2021; 2021:3210586. [PMID: 33506012 PMCID: PMC7806382 DOI: 10.1155/2021/3210586] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/23/2020] [Revised: 11/20/2020] [Accepted: 12/24/2020] [Indexed: 01/14/2023]
Abstract
Diabetes-related macrovascular and microvascular complications lead to poor prognosis. Insulin receptor substrate p53 (IRSp53) is known to act as a substrate for the insulin receptor tyrosine kinase, but its role in endothelial dysfunction remains unclear. Human umbilical vein endothelial cells (HUVECs) treated with D-glucose at different concentrations and a streptozocin-induced rat diabetes mellitus (DM) model were used to investigate the effects of hyperglycemia on the expression levels of IRSp53 and galectin-3 (gal-3) and the inflammatory state and mobility of HUVECs. Thereafter, IRSp53-overexpressing HUVECs and IRSp53-knockdown HUVECs were established using IRSp53-overexpressing lentivirus or IRSp53-siRNA to explore the role of IRSp53 in the HUVEC inflammatory state and HUVEC mobility. D-glucose at high concentration (HG) and hyperglycemia were found to induce downregulation of IRSp53 and upregulation of gal-3 in vitro and in vivo. Treatment with HG resulted in activation of NF-κB in HUVECs and impaired HUVEC mobility. Insulin restored HG-induced changes in the expression levels of IRSp53 and gal-3 in HUVECs and protected the cells from NF-κB activation and impaired mobility. Overexpression of IRSp53 inhibited the activation of NF-κB in HUVECs and strengthened HUVEC migration. Knockdown of IRSp53 facilitated the activation of NF-κB in HUVECs and decreased HUVEC migration. However, neither overexpression nor knockdown of IRSp53 altered the effects of insulin on HG-induced detrimental changes in HUVECs. HG and hyperglycemia resulted in downregulation of IRSp53 in vitro and in vivo. IRSp53 is concluded to inhibit the activation of NF-κB in HUVECs and to strengthen HUVEC migration.
Collapse
|
21
|
Liang ZG, Li L, Chen SN, Mao MG, Nie P. Expression and antibacterial analysis of galectin-8 and -9 genes in mandarin fish, Siniperca chuatsi. FISH & SHELLFISH IMMUNOLOGY 2020; 107:463-468. [PMID: 33152404 DOI: 10.1016/j.fsi.2020.10.028] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/15/2020] [Revised: 10/21/2020] [Accepted: 10/31/2020] [Indexed: 06/11/2023]
Abstract
Galectin-8 and galectin-9 belong to tandem repeat-type galectins, and in the present study, these two genes were cloned in mandarin fish Siniperca chuatsi. The open reading frame (ORF) of the mandarin fish galectin-8 and galectin-9 contains 942, and 1008 bp, encoding 313 and 335 amino acids, respectively. As a conserved feature, an N-terminal carbohydrate recognition domain (CRD), and a C-terminal CRD were observed in each of the two galectins in mandarin fish. In healthy fish, galectin-8 and -9 were constitutively expressed in all organs/tissues examined, and their expression can be induced following the stimulation of LPS and poly(I:C). It is obvious that galectin-8 had a higher increase at mRNA level following the stimulation of poly(I:C). It is further demonstrated that mandarin fish galectin-8 inhibited the growth of Flavobacterium columnare and Streptococcus agalactiae, and in addition to the two species of bacteria, galectin-9 inhibited also the growth of Edwardsiella piscicida, which provides the basis for further understanding their antibacterial role in immune response of mandarin fish.
Collapse
Affiliation(s)
- Zhi Gang Liang
- Dalian Ocean University, Dalian, Liaoning Province, 116023, China; State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, Hubei Province, 430072, China
| | - Li Li
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, Hubei Province, 430072, China
| | - Shan Nan Chen
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, Hubei Province, 430072, China
| | - Ming Guang Mao
- Dalian Ocean University, Dalian, Liaoning Province, 116023, China.
| | - P Nie
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, Hubei Province, 430072, China; Laboratory for Marine Biology and Biotechnology, Pilot National Laboratory for Marine Science and Technology (Qingdao), Qingdao, Shandong Province, 266237, China; School of Marine Science and Engineering, Qingdao Agricultural University, Qingdao, Shandong Province, 266109, China.
| |
Collapse
|
22
|
The therapeutic potential of galectin-3 inhibition in fibrotic disease. Int J Biochem Cell Biol 2020; 130:105881. [PMID: 33181315 DOI: 10.1016/j.biocel.2020.105881] [Citation(s) in RCA: 81] [Impact Index Per Article: 16.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2020] [Revised: 10/29/2020] [Accepted: 10/30/2020] [Indexed: 12/20/2022]
Abstract
Galectin-3 is a beta-galactoside-binding mammalian lectin and part of the 15 member galectin family that are evolutionarily highly conserved. It is the only chimeric protein with a C-terminal carbohydrate recognition domain (CRD) linked to a proline, glycine, and tyrosine rich additional N-terminal domain. Galectin-3 binds several cell surface glycoproteins via its CRD domain as well as undergoing oligomerization, via binding at the N-terminal or the CRD, resulting in the formation of a galectin-3 lattice on the cell surface. The galectin-3 lattice has been regarded as being a crucial mechanism whereby extracellular galectin-3 modulates cellular signalling by prolonging retention time or retarding lateral movement of cell surface receptors in the plasma membrane. As such galectin-3 can regulate various cellular functions such as diffusion, compartmentalization and endocytosis of plasma membrane glycoproteins and glycolipids and the functionality of membrane receptors. In multiple models of organ fibrosis, it has been demonstrated that galectin-3 is potently pro-fibrotic and modulates the activity of fibroblasts and macrophages in chronically inflamed organs. Increased galectin-3 expression also activates myofibroblasts resulting in scar formation and may therefore impact common fibrotic pathways leading to fibrosis in multiple organs. Over the last decade there has been a marked increase in the scientific literature investigating galectin-3 in a range of fibrotic diseases as well as the clinical development of new galectin-3 inhibitors. In this review we will examine the role of galectin-3 in fibrosis, the therapeutic strategies for inhibiting galectin-3 in fibrotic disease and the clinical landscape to date.
Collapse
|
23
|
Mei X, Ye Z, Chang Y, Huang S, Song J, Lu F. Trichinella spiralis co-infection exacerbates Plasmodium berghei malaria-induced hepatopathy. Parasit Vectors 2020; 13:440. [PMID: 32883347 PMCID: PMC7469358 DOI: 10.1186/s13071-020-04309-6] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2020] [Accepted: 08/24/2020] [Indexed: 11/11/2022] Open
Abstract
Background Although Plasmodium parasites and intestinal helminths share common endemic areas, the mechanisms of these co-infections on the host immune response remain not fully understood. Liver involvement in severe Plasmodium falciparum infections is a significant cause of morbidity and mortality. However, the effect of pre-existing Trichinella spiralis infection on the immune response and liver immune-pathogenesis in P. berghei ANKA (PbANKA)-infected mice needs to be elucidated. Methods Outbred Kunming mice were infected with T. spiralis and 9 days later were challenged with P. berghei ANKA (PbANKA), and the investigation occurred at 13 days after co-infection. Results Compared with PbANKA-mono-infected mice, T. spiralis + PbANKA-co-infected mice had similar survival rate but lower PbANKA parasitaemia; however, there were more severe hepatosplenomegaly, increased liver and spleen indexes, and increased liver pathology observed by hematoxylin and eosin staining; higher expression levels of galectin (Gal)-1, Gal-3, CD68+ macrophages, and elastase-positive neutrophils measured by immunohistochemical staining; upregulated mRNA expression levels of Gal-1, Gal-3, cytokines (interferon-gamma (IFNγ) and interleukin (IL)-6), and M1 macrophage polarization marker (inducible nitric oxide synthase (iNOS)) in the liver, and increased expression levels of Gal-1, IFNγ, IL-6, eosinophil cationic protein, eosinophil protein X, and M1 (IL-1β and iNOS) and M2 (Ym1) macrophage polarization markers in the spleen of co-infected mice detected by using quantitative real-time reverse transcription polymerase chain reaction (qRT-PCR). In vitro study showed that compared with PbANKA-mono-infected mice, there were significantly increased expression levels of Gal-1, Gal-3, IL-6, IL-1β, and iNOS in the peritoneal macrophage isolated from co-infected mice detected by using qRT-PCR. Correlation analysis revealed significant positive correlations between Gal-3 and IL-1β in the peritoneal macrophages isolated from PbANKA-mono-infected mice, between Gal-3 and IFNγ in the spleen of co-infected mice, and between Gal-1 and Ym1 in the peritoneal macrophages isolated from co-infected mice. Conclusions Our data indicate that pre-existing infection of T. spiralis may suppress P. berghei parasitaemia and aggravate malaria-induced liver pathology through stimulating Gal-1 and Gal-3 expression, activating macrophages, neutrophils, and eosinophils, and promoting mediator release and cytokine production.![]()
Collapse
Affiliation(s)
- Xu Mei
- Artemisinin Research Center and Institute of Science and Technology, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Zhanhong Ye
- Department of Parasitology, Zhongshan School of Medicine; Key Laboratory of Tropical Disease Control of Ministry of Education, Sun Yat-sen University, Guangzhou, China
| | - Yuqing Chang
- Artemisinin Research Center and Institute of Science and Technology, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Shiguang Huang
- School of Stomatology, Jinan University, Guangzhou, China.
| | - Jianping Song
- Artemisinin Research Center and Institute of Science and Technology, Guangzhou University of Chinese Medicine, Guangzhou, China.
| | - Fangli Lu
- Department of Parasitology, Zhongshan School of Medicine; Key Laboratory of Tropical Disease Control of Ministry of Education, Sun Yat-sen University, Guangzhou, China.
| |
Collapse
|
24
|
Ayona D, Fournier PE, Henrissat B, Desnues B. Utilization of Galectins by Pathogens for Infection. Front Immunol 2020; 11:1877. [PMID: 32973776 PMCID: PMC7466766 DOI: 10.3389/fimmu.2020.01877] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2020] [Accepted: 07/13/2020] [Indexed: 12/22/2022] Open
Abstract
Galectins are glycan-binding proteins which are expressed by many different cell types and secreted extracellularly. These molecules are well-known regulators of immune responses and involved in a broad range of cellular and pathophysiological functions. During infections, host galectins can either avoid or facilitate infections by interacting with host cells- and/or pathogen-derived glycoconjugates and less commonly, with proteins. Some pathogens also express self-produced galectins to interfere with host immune responses. This review summarizes pathogens which take advantage of host- or pathogen-produced galectins to establish the infection.
Collapse
Affiliation(s)
- Diyoly Ayona
- Aix Marseille Univ, IRD, APHM, MEPHI, IHU-Méditerranée Infection, Marseille, France
| | | | - Bernard Henrissat
- Architecture et Fonction des Macromolécules Biologiques, CNRS, Aix-Marseille University, Marseille, France
- USC1408 Architecture et Fonction des Macromolécules Biologiques, Institut National de la Recherche Agronomique, Marseille, France
- Department of Biological Sciences, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Benoit Desnues
- Aix Marseille Univ, IRD, APHM, MEPHI, IHU-Méditerranée Infection, Marseille, France
| |
Collapse
|
25
|
Snarr BD, St-Pierre G, Ralph B, Lehoux M, Sato Y, Rancourt A, Takazono T, Baistrocchi SR, Corsini R, Cheng MP, Sugrue M, Baden LR, Izumikawa K, Mukae H, Wingard JR, King IL, Divangahi M, Satoh MS, Yipp BG, Sato S, Sheppard DC. Galectin-3 enhances neutrophil motility and extravasation into the airways during Aspergillus fumigatus infection. PLoS Pathog 2020; 16:e1008741. [PMID: 32750085 PMCID: PMC7428289 DOI: 10.1371/journal.ppat.1008741] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2020] [Revised: 08/14/2020] [Accepted: 06/24/2020] [Indexed: 12/17/2022] Open
Abstract
Aspergillus fumigatus is an opportunistic mold that infects patients who are immunocompromised or have chronic lung disease, causing significant morbidity and mortality in these populations. While the factors governing the host response to A. fumigatus remain poorly defined, neutrophil recruitment to the site of infection is critical to clear the fungus. Galectin-3 is a mammalian β-galactose-binding lectin with both antimicrobial and immunomodulatory activities, however the role of galectin-3 in the defense against molds has not been studied. Here we show that galectin-3 expression is markedly up-regulated in mice and humans with pulmonary aspergillosis. Galectin-3 deficient mice displayed increased fungal burden and higher mortality during pulmonary infection. In contrast to previous reports with pathogenic yeast, galectin-3 exhibited no antifungal activity against A. fumigatus in vitro. Galectin-3 deficient mice exhibited fewer neutrophils in their airways during infection, despite normal numbers of total lung neutrophils. Intravital imaging studies confirmed that galectin-3 was required for normal neutrophil migration to the airspaces during fungal infection. Adoptive transfer experiments demonstrated that stromal rather than neutrophil-intrinsic galectin-3 was necessary for normal neutrophil entry into the airspaces. Live cell imaging studies revealed that extracellular galectin-3 directly increases neutrophil motility. Taken together, these data demonstrate that extracellular galectin-3 facilitates recruitment of neutrophils to the site of A. fumigatus infection, and reveals a novel role for galectin-3 in host defense against fungal infections. The environmental mold Aspergillus fumigatus commonly causes lung infections in people with impaired immunity or those suffering from a chronic lung disease. While neutrophils are a key cell type necessary for the eradication of this infection, the precise mechanism of their recruitment to the site of infection remains incompletely understood. Here we show that the secreted mammalian protein galectin-3 plays an important role in helping neutrophils reaching the fungus within the airways. We found that both mice and humans produce galectin-3 when infected with A. fumigatus, and mice lacking galectin-3 were more susceptible to infection than normal mice. Galectin-3-deficient mice had impaired neutrophil recruitment to the site of infection. In the absence of galectin-3, neutrophils exhibited reduced motility in mouse lungs and in tissue culture. Our study offers insights into the mechanisms underlying the recruitment of neutrophils to the airways during A. fumigatus infection and reveals a new role for galectin-3 in increasing neutrophil motility.
Collapse
Affiliation(s)
- Brendan D. Snarr
- Department of Microbiology and Immunology, McGill University, Montréal, Canada
- Infectious Diseases and Immunity in Global Health Program, Centre for Translational Biology, Research Institute of the McGill University Health Centre, Montréal, Canada
- McGill Interdisciplinary Initiative in Infection and Immunity, Montréal, Canada
| | - Guillaume St-Pierre
- Laboratory of Glycobiology and Bioimaging, Research Centre for Infectious Diseases, Research Centre of CHU de Québec, Faculty of Medicine, Laval University, Québec City, Canada
| | - Benjamin Ralph
- Department of Microbiology and Immunology, McGill University, Montréal, Canada
- Infectious Diseases and Immunity in Global Health Program, Centre for Translational Biology, Research Institute of the McGill University Health Centre, Montréal, Canada
- McGill Interdisciplinary Initiative in Infection and Immunity, Montréal, Canada
| | - Mélanie Lehoux
- Infectious Diseases and Immunity in Global Health Program, Centre for Translational Biology, Research Institute of the McGill University Health Centre, Montréal, Canada
- McGill Interdisciplinary Initiative in Infection and Immunity, Montréal, Canada
| | - Yukiko Sato
- Infectious Diseases and Immunity in Global Health Program, Centre for Translational Biology, Research Institute of the McGill University Health Centre, Montréal, Canada
| | - Ann Rancourt
- Laboratory of Glycobiology and Bioimaging, Research Centre for Infectious Diseases, Research Centre of CHU de Québec, Faculty of Medicine, Laval University, Québec City, Canada
- Laboratory of DNA Damage Responses and Bioimaging, CHU de Québec, Faculty of Medicine, Laval University, Québec city, Canada
| | - Takahiro Takazono
- Department of Infectious Diseases, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
- Department of Respiratory Medicine, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
| | - Shane R. Baistrocchi
- Infectious Diseases and Immunity in Global Health Program, Centre for Translational Biology, Research Institute of the McGill University Health Centre, Montréal, Canada
| | - Rachel Corsini
- Infectious Diseases and Immunity in Global Health Program, Centre for Translational Biology, Research Institute of the McGill University Health Centre, Montréal, Canada
- McGill Interdisciplinary Initiative in Infection and Immunity, Montréal, Canada
| | - Matthew P. Cheng
- Division of Infectious Diseases and Department of Medical Microbiology, McGill University Health Centre, Montréal, Canada
| | - Michele Sugrue
- University of Florida College of Medicine, Gainsville, Florida, United States of America
| | - Lindsey R. Baden
- Harvard University & Brigham & Women’s Hospital, Boston, Massachusetts, United States of America
| | - Koichi Izumikawa
- Department of Infectious Diseases, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
| | - Hiroshi Mukae
- Department of Respiratory Medicine, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
| | - John R. Wingard
- University of Florida College of Medicine, Gainsville, Florida, United States of America
| | - Irah L. King
- Department of Microbiology and Immunology, McGill University, Montréal, Canada
- McGill Interdisciplinary Initiative in Infection and Immunity, Montréal, Canada
- Meakins-Christie Laboratories, Department of Medicine, Department of Pathology, McGill International TB Centre, McGill University Health Centre, Montréal, Canada
| | - Maziar Divangahi
- Department of Microbiology and Immunology, McGill University, Montréal, Canada
- McGill Interdisciplinary Initiative in Infection and Immunity, Montréal, Canada
- Meakins-Christie Laboratories, Department of Medicine, Department of Pathology, McGill International TB Centre, McGill University Health Centre, Montréal, Canada
| | - Masahiko S. Satoh
- Laboratory of DNA Damage Responses and Bioimaging, CHU de Québec, Faculty of Medicine, Laval University, Québec city, Canada
| | - Bryan G. Yipp
- Calvin, Phoebe and Joan Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, Canada
| | - Sachiko Sato
- Laboratory of Glycobiology and Bioimaging, Research Centre for Infectious Diseases, Research Centre of CHU de Québec, Faculty of Medicine, Laval University, Québec City, Canada
- * E-mail: (SS); (DCS)
| | - Donald C. Sheppard
- Department of Microbiology and Immunology, McGill University, Montréal, Canada
- Infectious Diseases and Immunity in Global Health Program, Centre for Translational Biology, Research Institute of the McGill University Health Centre, Montréal, Canada
- McGill Interdisciplinary Initiative in Infection and Immunity, Montréal, Canada
- Division of Infectious Diseases and Department of Medical Microbiology, McGill University Health Centre, Montréal, Canada
- * E-mail: (SS); (DCS)
| |
Collapse
|
26
|
Cassaglia P, Penas F, Betazza C, Fontana Estevez F, Miksztowicz V, Martínez Naya N, Llamosas MC, Noli Truant S, Wilensky L, Volberg V, Cevey ÁC, Touceda V, Cicale E, Berg G, Fernández M, Goren N, Morales C, González GE. Genetic Deletion of Galectin-3 Alters the Temporal Evolution of Macrophage Infiltration and Healing Affecting the Cardiac Remodeling and Function after Myocardial Infarction in Mice. THE AMERICAN JOURNAL OF PATHOLOGY 2020; 190:1789-1800. [PMID: 32473918 DOI: 10.1016/j.ajpath.2020.05.010] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/17/2019] [Revised: 04/22/2020] [Accepted: 05/05/2020] [Indexed: 12/13/2022]
Abstract
We studied the role of galectin-3 (Gal-3) in the expression of alternative activation markers (M2) on macrophage, cytokines, and fibrosis through the temporal evolution of healing, ventricular remodeling, and function after myocardial infarction (MI). C57BL/6J and Gal-3 knockout mice (Lgals3-/-) were subjected to permanent coronary ligation or sham. We studied i) mortality, ii) macrophage infiltration and expression of markers of alternative activation, iii) cytokine, iv) matrix metalloproteinase-2 activity, v) fibrosis, and vi) cardiac function and remodeling. At 1 week post-MI, lack of Gal-3 markedly attenuated F4/80+ macrophage infiltration and significantly increased the expression of Mrc1 and Chil1, markers of M2 macrophages at the MI zone. Levels of IL-10, IL-6, and matrix metalloproteinase-2 were significantly increased, whereas tumor necrosis factor-α, transforming growth factor-β, and fibrosis were remarkably attenuated at the infarct zone. In Gal-3 knockout mice, scar thinning ratio, expansion, and cardiac remodeling and function were severely affected from the onset of MI. At 4 weeks post-MI, the natural evolution of fibrosis in Gal-3 knockout mice was also affected. Our results suggest that Gal-3 is essential for wound healing because it regulates the dynamics of macrophage infiltration, proinflammatory and anti-inflammatory cytokine expression, and fibrosis along the temporal evolution of MI in mice. The deficit of Gal-3 affected the dynamics of wound healing, thus aggravating the evolution of remodeling and function.
Collapse
Affiliation(s)
- Pablo Cassaglia
- Facultad de Medicina-CONICET, Departamento de Patología, Instituto de Fisiopatología Cardiovascular, Buenos Aires, Argentina
| | - Federico Penas
- Facultad de Medicina, Instituto de Investigaciones Biomédicas en Retrovirus y SIDA (INBIRS), Buenos Aires, Argentina
| | - Celeste Betazza
- Facultad de Medicina-CONICET, Departamento de Patología, Instituto de Fisiopatología Cardiovascular, Buenos Aires, Argentina; Facultad de Medicina, Pontificia Universidad Católica Argentina (UCA), Instituto de Investigaciones Biomédicas (UCA-CONICET), Laboratorio de Patología Cardiovascular Experimental e Hipertensi Arterial, Buenos Aires, Argentina
| | - Florencia Fontana Estevez
- Facultad de Medicina, Pontificia Universidad Católica Argentina (UCA), Instituto de Investigaciones Biomédicas (UCA-CONICET), Laboratorio de Patología Cardiovascular Experimental e Hipertensi Arterial, Buenos Aires, Argentina
| | - Verónica Miksztowicz
- Facultad de Medicina, Pontificia Universidad Católica Argentina (UCA), Instituto de Investigaciones Biomédicas (UCA-CONICET), Laboratorio de Patología Cardiovascular Experimental e Hipertensi Arterial, Buenos Aires, Argentina; Facultad de Farmacia y Bioquímica, Departamento de Bioquímica Clínica-INFIBIOC, Laboratorio de Lípidos y Aterosclerosis, Buenos Aires, Argentina
| | - Nadia Martínez Naya
- Facultad de Medicina-CONICET, Departamento de Patología, Instituto de Fisiopatología Cardiovascular, Buenos Aires, Argentina
| | - María Clara Llamosas
- Facultad de Medicina-CONICET, Departamento de Patología, Instituto de Fisiopatología Cardiovascular, Buenos Aires, Argentina
| | - Sofía Noli Truant
- Facultad de Farmacia y Bioquímica-CONICET, Instituto de Estudios de la Inmunidad Humoral (IDEHU), Buenos Aires, Argentina
| | - Luciana Wilensky
- Facultad de Medicina-CONICET, Departamento de Patología, Instituto de Fisiopatología Cardiovascular, Buenos Aires, Argentina
| | - Verónica Volberg
- Facultad de Medicina-CONICET, Departamento de Patología, Instituto de Fisiopatología Cardiovascular, Buenos Aires, Argentina
| | - Ágata C Cevey
- Facultad de Medicina, Instituto de Investigaciones Biomédicas en Retrovirus y SIDA (INBIRS), Buenos Aires, Argentina
| | - Vanessa Touceda
- Facultad de Farmacia y Bioquímica, Departamento de Bioquímica Clínica-INFIBIOC, Laboratorio de Lípidos y Aterosclerosis, Buenos Aires, Argentina
| | - Eliana Cicale
- Facultad de Ciencias Veterinarias, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Gabriela Berg
- Facultad de Farmacia y Bioquímica, Departamento de Bioquímica Clínica-INFIBIOC, Laboratorio de Lípidos y Aterosclerosis, Buenos Aires, Argentina
| | - Marisa Fernández
- Facultad de Farmacia y Bioquímica-CONICET, Instituto de Estudios de la Inmunidad Humoral (IDEHU), Buenos Aires, Argentina
| | - Nora Goren
- Facultad de Medicina, Instituto de Investigaciones Biomédicas en Retrovirus y SIDA (INBIRS), Buenos Aires, Argentina
| | - Celina Morales
- Facultad de Medicina-CONICET, Departamento de Patología, Instituto de Fisiopatología Cardiovascular, Buenos Aires, Argentina
| | - Germán E González
- Facultad de Medicina-CONICET, Departamento de Patología, Instituto de Fisiopatología Cardiovascular, Buenos Aires, Argentina; Facultad de Medicina, Pontificia Universidad Católica Argentina (UCA), Instituto de Investigaciones Biomédicas (UCA-CONICET), Laboratorio de Patología Cardiovascular Experimental e Hipertensi Arterial, Buenos Aires, Argentina.
| |
Collapse
|
27
|
Srejovic I, Selakovic D, Jovicic N, Jakovljević V, Lukic ML, Rosic G. Galectin-3: Roles in Neurodevelopment, Neuroinflammation, and Behavior. Biomolecules 2020; 10:biom10050798. [PMID: 32455781 PMCID: PMC7277476 DOI: 10.3390/biom10050798] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/1970] [Revised: 05/09/2020] [Accepted: 05/13/2020] [Indexed: 12/16/2022] Open
Abstract
There is a plethora of evidence to suggest that Galectin-3 plays an important role in normal functions of mammalian cells, as well as in different pathogenic conditions. This review highlights recent data published by researchers, including our own team, on roles of Galectin-3 in the nervous system. Here, we discuss the roles of Galectin-3 in brain development, its roles in glial cells, as well as the interactions of glial cells with other neural and invading cells in pathological conditions. Galectin-3 plays an important role in the pathogenesis of neuroinflammatory and neurodegenerative disorders, such as multiple sclerosis, Alzheimer’s disease, Parkinson’s disease, and Huntington’s disease. On the other hand, there is also evidence of the protective role of Galectin-3 due to its anti-apoptotic effect in target cells. Interestingly, genetic deletion of Galectin-3 affects behavioral patterns in maturing and adult mice. The results reviewed in this paper and recent development of highly specific inhibitors suggests that Galectin-3 may be an important therapeutic target in pathological conditions including the disorders of the central nervous system.
Collapse
Affiliation(s)
- Ivan Srejovic
- Department of Physiology, Faculty of Medical Sciences, University of Kragujevac, Svetozara Markovica 69, 34000 Kragujevac Serbia; (I.S.); (D.S.); (V.J.)
| | - Dragica Selakovic
- Department of Physiology, Faculty of Medical Sciences, University of Kragujevac, Svetozara Markovica 69, 34000 Kragujevac Serbia; (I.S.); (D.S.); (V.J.)
| | - Nemanja Jovicic
- Department of Histology and Embryology, Faculty of Medical Sciences, University of Kragujevac, Svetozara Markovica 69, 34000 Kragujevac, Serbia;
| | - Vladimir Jakovljević
- Department of Physiology, Faculty of Medical Sciences, University of Kragujevac, Svetozara Markovica 69, 34000 Kragujevac Serbia; (I.S.); (D.S.); (V.J.)
- Department of Human Pathology, 1st Moscow State Medical University IM Sechenov, 119146 Moscow, Russia
| | - Miodrag L. Lukic
- Department of Physiology—Molecular Medicine Unit, Faculty of Medical Sciences, University of Kragujevac, Svetozara Markovica 69, 34000 Kragujevac, Serbia
- Correspondence: (M.L.L.); (G.R.)
| | - Gvozden Rosic
- Department of Physiology, Faculty of Medical Sciences, University of Kragujevac, Svetozara Markovica 69, 34000 Kragujevac Serbia; (I.S.); (D.S.); (V.J.)
- Correspondence: (M.L.L.); (G.R.)
| |
Collapse
|
28
|
Park AM, Khadka S, Sato F, Omura S, Fujita M, Hsu DK, Liu FT, Tsunoda I. Galectin-3 as a Therapeutic Target for NSAID-Induced Intestinal Ulcers. Front Immunol 2020; 11:550366. [PMID: 33072090 PMCID: PMC7539695 DOI: 10.3389/fimmu.2020.550366] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2020] [Accepted: 09/02/2020] [Indexed: 02/05/2023] Open
Abstract
UNLABELLED Non-steroidal anti-inflammatory drugs (NSAIDs) induce ulcers in the gastrointestinal tract, including the stomach and small intestine. NSAID-induced gastric ulcers can be prevented by taking acid-neutralizing/inhibitory drugs and cytoprotective agents. In contrast, there are no medicines to control NSAID-induced small intestinal ulcers, which are accompanied by a mucosal invasion of bacteria and subsequent activation of immune cells. Galectin-3 (Gal3), an endogenous lectin, has anti-microbial and pro-inflammatory functions. In the small intestine, since Gal3 is highly expressed in epithelial cells constitutively and macrophages inducibly, the Gal3 level can affect microbiota composition and macrophage activation. We hypothesized that the modulation of Gal3 expression could be beneficial in NSAID-induced intestinal ulcers. Using Gal3 knockout (Gal3KO) mice, we determined whether Gal3 could be a therapeutic target in NSAID-induced intestinal ulcers. Following the administration of indomethacin, an NSAID, we found that small intestinal ulcers were less severe in Gal3KO mice than in wild-type (WT) mice. We also found that the composition of intestinal microbiota was different between WT and Gal3KO mice and that bactericidal antibiotic polymyxin B treatment significantly suppressed NSAID-induced ulcers. Furthermore, clodronate, a macrophage modulator, attenuated NSAID-induced ulcers. Therefore, Gal3 could be an exacerbating factor in NSAID-induced intestinal ulcers by affecting the intestinal microbiota population and macrophage activity. Inhibition of Gal3 may be a therapeutic strategy in NSAID-induced intestinal ulcers. CLINICAL TRIAL REGISTRATION www.ClinicalTrials.gov, identifier NCT03832946.
Collapse
Affiliation(s)
- Ah-Mee Park
- Department of Microbiology, Faculty of Medicine, Kindai University, Osaka, Japan
- *Correspondence: Ah-Mee Park,
| | - Sundar Khadka
- Department of Microbiology, Faculty of Medicine, Kindai University, Osaka, Japan
| | - Fumitaka Sato
- Department of Microbiology, Faculty of Medicine, Kindai University, Osaka, Japan
| | - Seiichi Omura
- Department of Microbiology, Faculty of Medicine, Kindai University, Osaka, Japan
| | - Mitsugu Fujita
- Department of Microbiology, Faculty of Medicine, Kindai University, Osaka, Japan
| | - Daniel K. Hsu
- Department of Dermatology, University of California Davis Health System, Sacramento, CA, United States
| | - Fu-Tong Liu
- Department of Dermatology, University of California Davis Health System, Sacramento, CA, United States
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
| | - Ikuo Tsunoda
- Department of Microbiology, Faculty of Medicine, Kindai University, Osaka, Japan
| |
Collapse
|
29
|
Li FY, Wang SF, Bernardes ES, Liu FT. Galectins in Host Defense Against Microbial Infections. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1204:141-167. [DOI: 10.1007/978-981-15-1580-4_6] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
30
|
Cardiac Chagas Disease: MMPs, TIMPs, Galectins, and TGF- β as Tissue Remodelling Players. DISEASE MARKERS 2019; 2019:3632906. [PMID: 31885735 PMCID: PMC6899287 DOI: 10.1155/2019/3632906] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/16/2019] [Accepted: 11/01/2019] [Indexed: 01/13/2023]
Abstract
A century after the discovery of Chagas disease, studies are still needed to establish the complex pathophysiology of this disease. However, it is known that several proteins and molecules are related to the establishment of this disease, its evolution, and the appearance of its different clinical forms. Metalloproteinases and their tissue inhibitors, galectins, and TGF-β are involved in the process of infection and consequently the development of myocarditis, tissue remodeling, and fibrosis upon infection with Trypanosoma cruzi. Thus, considering that the heart is one of the main target organs in Chagas disease, knowledge regarding the mechanisms of action of these molecules is essential to understand how they interact and trigger local and systemic reactions and, consequently, determine whether they contribute to the development of Chagas' heart disease. In this sense, it is believed that the inflammatory microenvironment caused by the infection alters the expression of these proteins favoring progression of the host-parasite cycle and thereby stimulating cardiac tissue remodeling mechanisms and fibrosis. The aim of this review was to gather information on metalloproteinases and their tissue inhibitors, galectins, and TGF-β and discuss how these molecules and their different interrelationships contribute to the development of Chagas' heart disease.
Collapse
|
31
|
Querol Cano L, Tagit O, Dolen Y, van Duffelen A, Dieltjes S, Buschow SI, Niki T, Hirashima M, Joosten B, van den Dries K, Cambi A, Figdor CG, van Spriel AB. Intracellular Galectin-9 Controls Dendritic Cell Function by Maintaining Plasma Membrane Rigidity. iScience 2019; 22:240-255. [PMID: 31786520 PMCID: PMC6906692 DOI: 10.1016/j.isci.2019.11.019] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2019] [Revised: 10/17/2019] [Accepted: 11/11/2019] [Indexed: 12/24/2022] Open
Abstract
Endogenous extracellular Galectins constitute a novel mechanism of membrane protein organization at the cell surface. Although Galectins are also highly expressed intracellularly, their cytosolic functions are poorly understood. Here, we investigated the role of Galectin-9 in dendritic cell (DC) surface organization and function. By combining functional, super-resolution and atomic force microscopy experiments to analyze membrane stiffness, we identified intracellular Galectin-9 to be indispensable for plasma membrane integrity and structure in DCs. Galectin-9 knockdown studies revealed intracellular Galectin-9 to directly control cortical membrane structure by modulating Rac1 activity, providing the underlying mechanism of Galectin-9-dependent actin cytoskeleton organization. Consequent to its role in maintaining plasma membrane structure, phagocytosis studies revealed that Galectin-9 was essential for C-type-lectin receptor-mediated pathogen uptake by DCs. This was confirmed by the impaired phagocytic capacity of Galectin-9-null murine DCs. Together, this study demonstrates a novel role for intracellular Galectin-9 in modulating DC function, which may be evolutionarily conserved.
Collapse
Affiliation(s)
- Laia Querol Cano
- Department of Tumour Immunology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Geert Grooteplein 26-28, Nijmegen 6525 GA, The Netherlands
| | - Oya Tagit
- Department of Tumour Immunology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Geert Grooteplein 26-28, Nijmegen 6525 GA, The Netherlands
| | - Yusuf Dolen
- Department of Tumour Immunology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Geert Grooteplein 26-28, Nijmegen 6525 GA, The Netherlands
| | - Anne van Duffelen
- Department of Tumour Immunology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Geert Grooteplein 26-28, Nijmegen 6525 GA, The Netherlands
| | - Shannon Dieltjes
- Department of Tumour Immunology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Geert Grooteplein 26-28, Nijmegen 6525 GA, The Netherlands
| | - Sonja I Buschow
- Department of Gastroenterology and Hepatology, Erasmus MC-University Medical Center, Wytemaweg 80, Rotterdam 3015 CN, The Netherlands
| | - Toshiro Niki
- GalPharma Co., Ltd., Takamatsu, Kagawa 761-0301, Japan; Department of Immunology and Immunopathology, Faculty of Medicine, Kagawa University, Takamatsu, Kagawa, 761-0793, Japan
| | - Mitsuomi Hirashima
- GalPharma Co., Ltd., Takamatsu, Kagawa 761-0301, Japan; Department of Immunology and Immunopathology, Faculty of Medicine, Kagawa University, Takamatsu, Kagawa, 761-0793, Japan
| | - Ben Joosten
- Department of Cell Biology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Geert Grooteplein 26-28, 6525 GA Nijmegen, The Netherlands
| | - Koen van den Dries
- Department of Cell Biology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Geert Grooteplein 26-28, 6525 GA Nijmegen, The Netherlands
| | - Alessandra Cambi
- Department of Cell Biology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Geert Grooteplein 26-28, 6525 GA Nijmegen, The Netherlands
| | - Carl G Figdor
- Department of Tumour Immunology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Geert Grooteplein 26-28, Nijmegen 6525 GA, The Netherlands
| | - Annemiek B van Spriel
- Department of Tumour Immunology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Geert Grooteplein 26-28, Nijmegen 6525 GA, The Netherlands.
| |
Collapse
|
32
|
Cockram TOJ, Puigdellívol M, Brown GC. Calreticulin and Galectin-3 Opsonise Bacteria for Phagocytosis by Microglia. Front Immunol 2019; 10:2647. [PMID: 31781126 PMCID: PMC6861381 DOI: 10.3389/fimmu.2019.02647] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2019] [Accepted: 10/25/2019] [Indexed: 12/18/2022] Open
Abstract
Opsonins are soluble, extracellular proteins, released by activated immune cells, and when bound to a target cell, can induce phagocytes to phagocytose the target cell. There are three known classes of opsonin: antibodies, complement factors and secreted pattern recognition receptors, but these have limited access to the brain. We identify here two novel opsonins of bacteria, calreticulin, and galectin-3 (both lectins that can bind lipopolysaccharide), which were released by microglia (brain-resident macrophages) when activated by bacterial lipopolysaccharide. Calreticulin and galectin-3 both bound to Escherichia coli, and when bound increased phagocytosis of these bacteria by microglia. Furthermore, lipopolysaccharide-induced microglial phagocytosis of E. coli bacteria was partially inhibited by: sugars, an anti-calreticulin antibody, a blocker of the calreticulin phagocytic receptor LRP1, a blocker of the galectin-3 phagocytic receptor MerTK, or simply removing factors released from the microglia, indicating this phagocytosis is dependent on extracellular calreticulin and galectin-3. Thus, calreticulin and galectin-3 are opsonins, released by activated microglia to promote clearance of bacteria. This innate immune response of microglia may help clear bacterial infections of the brain.
Collapse
Affiliation(s)
- Tom O J Cockram
- Department of Biochemistry, University of Cambridge, Cambridge, United Kingdom
| | - Mar Puigdellívol
- Department of Biochemistry, University of Cambridge, Cambridge, United Kingdom
| | - Guy C Brown
- Department of Biochemistry, University of Cambridge, Cambridge, United Kingdom
| |
Collapse
|
33
|
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: 6.0] [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
|
34
|
Ayyappan P, Harms RZ, Buckner JH, Sarvetnick NE. Coordinated Induction of Antimicrobial Response Factors in Systemic Lupus Erythematosus. Front Immunol 2019; 10:658. [PMID: 31019506 PMCID: PMC6458289 DOI: 10.3389/fimmu.2019.00658] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2019] [Accepted: 03/11/2019] [Indexed: 12/12/2022] Open
Abstract
Systemic lupus erythematosus (SLE) is a chronic autoimmune disease characterized by dysregulated autoantibody production and complement activation leading to multi-organ damage. The disease is associated with increased intestinal permeability. In this study, we tested the hypothesis that SLE subjects have increased systemic exposure to bacteria. Since bacteria induce the expression of antimicrobial response factors (ARFs), we measured the levels of a series of clinically relevant ARFs in the plasma of SLE subjects. We found that levels of sCD14, lysozyme, and CXCL16 were significantly elevated in SLE subjects. A strong positive correlation was also observed between sCD14 and SELENA-SLEDAI score. Interestingly, the ratio of EndoCAb IgM:total IgM was significantly decreased in SLE and this ratio was negatively correlated with sCD14 levels. Although, there were no significant differences in the levels of lipopolysaccharide binding protein (LBP) and fatty acid binding protein 2 (FABP2), we observed significant positive correlations between lysozyme levels and sCD14, LBP, and FABP2. Moreover, galectin-3 levels also positively correlate with lysozyme, sCD14, and LBP. Since our SLE cohort comprised 43.33% males, we were able to identify gender-specific changes in the levels of ARFs. Overall, these changes in the levels and relationships between ARFs link microbial exposure and SLE. Approaches to reduce microbial exposure or to improve barrier function may provide therapeutic strategies for SLE patients.
Collapse
Affiliation(s)
- Prathapan Ayyappan
- Department of Surgery-Transplant, University of Nebraska Medical Center, Omaha, NE, United States
| | - Robert Z. Harms
- Department of Surgery-Transplant, University of Nebraska Medical Center, Omaha, NE, United States
| | - Jane H. Buckner
- Translational Research Program, Benaroya Research Institute, Seattle, WA, United States
| | - Nora E. Sarvetnick
- Department of Surgery-Transplant, University of Nebraska Medical Center, Omaha, NE, United States
- Mary and Dick Holland Regenerative Medicine Program, University of Nebraska Medical Center, Omaha, NE, United States
| |
Collapse
|
35
|
Verma P, Laforce-Nesbitt SS, Tucker R, Mao Q, De Paepe ME, Bliss JM. Galectin-3 expression and effect of supplementation in neonatal mice with disseminated Candida albicans infection. Pediatr Res 2019; 85:527-532. [PMID: 30679793 PMCID: PMC6397689 DOI: 10.1038/s41390-019-0279-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/18/2018] [Revised: 12/04/2018] [Accepted: 12/31/2018] [Indexed: 01/08/2023]
Abstract
BACKGROUND Invasive candidiasis is an important cause of fungal infections in immunocompromised patients, including premature infants. The S-type lectin, galectin-3 (gal3), is increasingly recognized for its role in antifungal host defense. This study tested the hypothesis that tissue gal3 expression is affected by disseminated infection with Candida albicans and that supplementation with gal3 will provide a benefit in this setting. METHODS To determine the expression of gal3 at the tissue level in response to disseminated infection with C. albicans, adult and neonatal mice were infected using previously established models. End points were chosen that reflected substantive tissue fungal burden but before mortality. RESULTS No differences in gal3 were detected in tissues of adult animals relative to uninfected controls. In neonatal animals, gal3 concentration was lower in the spleen of infected animals compared to uninfected. Pretreatment of neonatal mice with recombinant gal3 was associated with reduced mortality and reduced fungal burden in the kidney, spleen, and lung at 24 h following infection. CONCLUSION These findings suggest that gal3 has an active role in host defense against candidiasis and that neonatal animals can benefit from supplementation with this lectin in the setting of disseminated candidiasis.
Collapse
Affiliation(s)
- Prasoon Verma
- Department of Pediatrics, Women & Infants Hospital of Rhode Island, Alpert Medical School of Brown University, Providence, RI
| | - Sonia S. Laforce-Nesbitt
- Department of Pediatrics, Women & Infants Hospital of Rhode Island, Alpert Medical School of Brown University, Providence, RI
| | - Richard Tucker
- Department of Pediatrics, Women & Infants Hospital of Rhode Island, Alpert Medical School of Brown University, Providence, RI
| | - Quanfu Mao
- Department of Pathology and Laboratory Medicine, Women & Infants Hospital of Rhode Island, Alpert Medical School of Brown University, Providence, RI
| | - Monique E. De Paepe
- Department of Pathology and Laboratory Medicine, Women & Infants Hospital of Rhode Island, Alpert Medical School of Brown University, Providence, RI
| | - Joseph M. Bliss
- Department of Pediatrics, Women & Infants Hospital of Rhode Island, Alpert Medical School of Brown University, Providence, RI,Corresponding Author: Dept. of Pediatrics, Women & Infants Hospital of Rhode Island, 101 Dudley St., Providence, RI 02905, Phone: (401)274-1100, Fax: (401) 453-7571,
| |
Collapse
|
36
|
Zhao QY, Shi SJ, Sun DQ, Zhang SS, Zhou XH. [Correlation between galectin-3 level in bronchoalveolar lavage fluid and cellular immunity in children with refractory Mycoplasma pneumoniae pneumonia]. ZHONGGUO DANG DAI ER KE ZA ZHI = CHINESE JOURNAL OF CONTEMPORARY PEDIATRICS 2019; 21:150-154. [PMID: 30782277 PMCID: PMC7389830 DOI: 10.7499/j.issn.1008-8830.2019.02.008] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 08/01/2018] [Accepted: 11/09/2018] [Indexed: 06/09/2023]
Abstract
OBJECTIVE To study the correlation of galectin-3 level in bronchoalveolar lavage fluid (BALF) with Mycoplasma pneumoniae (MP) load and cellular immunity of neutrophils and macrophages in the airway in children with refractory MP pneumonia (RMPP). METHODS A total of 64 children with RMPP who were hospitalized from January 2013 to January 2017 were enrolled. In addition to the conservative medical treatment, all the 64 children with RMPP were given bronchoalveolar lavage in the acute stage (5-7 days after admission) and 48 out of the 64 children were given bronchoalveolar lavage in the recovery stage (10-14 days after admission). Four milliliters of BALF of the affected lung lobe or segment were collected. ELISA was used to measure the level of galectin-3 in BALF supernatant. RT-PCR was used to measure MP load. Hematoxylin and eosin staining was used to measure the percentage of neutrophils and macrophages. Six children with bronchial foreign bodies were enrolled as the control group. RESULTS The RMPP group had a significantly higher level of galectin-3 in BALF in both the acute and recovery stages than the control group (P<0.01), and the level of galectin-3 in the acute stage was significantly higher than in the recovery stage (P<0.01). The RMPP group had a significantly higher percentage of neutrophils in BALF in both the acute and recovery stages than the control group (P<0.01), and the percentage of neutrophils in the acute stage was significantly higher than in the recovery stage (P<0.01). The RMPP group had a significantly lower percentage of macrophages in BALF in both the acute and recovery stages than the control group (P<0.01), but there was no significant difference in the percentage of macrophages between the acute and recovery stages (P>0.05). The RMPP group had a significantly higher MP load in BALF in both the acute and recovery stages than the control group (P<0.01), and the MP load in the acute stage was significantly higher than in the recovery stage (P<0.01). In the children with RMPP, galectin-3 level in BALF in the acute stage was positively correlated with MP load and the percentage of neutrophils (rs=0.789 and 0.726 respectively; P<0.01). CONCLUSIONS Galectin-3 is involved in the process of airway inflammation in children with RMPP, and the level of galectin-3 in BALF is positively correlated with MP load. RMPP is a cellular immune inflammatory lesion with the increase of neutrophils and the reduction in macrophages. Galectin-3 is closely associated with neutrophil chemotaxis and luminal infiltration in children with RMPP. MP load gradually decreases with the recovery from RMPP, but it is not completely eliminated by the immune system in the recovery stage. MP infection can increase the consumption of macrophages in children with RMPP.
Collapse
Affiliation(s)
- Qian-Ye Zhao
- Department of Pediatrics, Lianyungang Maternal and Child Health Care Center Affiliated to Yangzhou University, Lianyungang, Jiangsu 222006, China.
| | | | | | | | | |
Collapse
|
37
|
Erriah M, Pabreja K, Fricker M, Baines KJ, Donnelly LE, Bylund J, Karlsson A, Simpson JL. Galectin-3 enhances monocyte-derived macrophage efferocytosis of apoptotic granulocytes in asthma. Respir Res 2019; 20:1. [PMID: 30606211 PMCID: PMC6318889 DOI: 10.1186/s12931-018-0967-9] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2018] [Accepted: 12/16/2018] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND Galectin-3 is a 32 kDa protein secreted by macrophages involved in processes such as cell activation, chemotaxis and phagocytosis. Galectin-3 has previously been shown to improve the ability of airway macrophages to ingest apoptotic cells (efferocytosis) in chronic obstructive pulmonary disease (COPD) and may be of interest in non-eosinophilic asthma (NEA) which is also characterised by impaired efferocytosis. It was hypothesised that the addition of exogenous galectin-3 to monocyte-derived macrophages (MDMs) derived from donors with NEA would enhance their ability to engulf apoptotic granulocytes. METHODS Eligible non-smoking adults with asthma (n = 19), including 7 with NEA and healthy controls (n = 10) underwent a clinical assessment, venepuncture and sputum induction. MDMs were co-cultured with apoptotic granulocytes isolated from healthy donors with or without exogenous recombinant galectin-3 (50 μg/mL) and efferocytosis was assessed by flow cytometry. Galectin-3 expression and localisation in MDMs was visualised by immunofluorescence staining and fluorescence microscopy. Galectin-3, interleukin (IL)-6 and CXCL8 secretion were measured in cell culture supernatants by ELISA and cytometric bead array. RESULTS Baseline efferocytosis (mean (±standard deviation)) was lower in participants with asthma (33.2 (±17.7)%) compared with healthy controls (45.3 (±15.9)%; p = 0.081). Efferocytosis did not differ between the participants with eosinophilic asthma (EA) (31.4 (±19.2)%) and NEA (28.7 (±21.5)%; p = 0.748). Addition of galectin-3 significantly improved efferocytosis in asthma, particularly in NEA (37.8 (±18.1)%) compared with baseline (30.4 (±19.7)%; p = 0.012). Efferocytosis was not associated with any of the clinical outcomes but was negatively correlated with sputum macrophage numbers (Spearman r = - 0.671; p = 0.017). Galectin-3 was diffusely distributed in most MDMs but formed punctate structures in 5% of MDMs. MDM galectin-3 secretion was lower in asthma (9.99 (2.67, 15.48) ng/mL) compared with the healthy controls (20.72 (11.28, 27.89) ng/mL; p = 0.044) while IL-6 and CXCL8 levels were similar. CONCLUSIONS Galectin-3 modulates macrophage function in asthma, indicating a potential role for galectin-3 to reverse impaired efferocytosis in NEA.
Collapse
Affiliation(s)
- Melanie Erriah
- Priority Research Centre for Healthy Lungs, The University of Newcastle, Newcastle, NSW, Australia
| | - Kavita Pabreja
- Priority Research Centre for Healthy Lungs, The University of Newcastle, Newcastle, NSW, Australia
| | - Michael Fricker
- Priority Research Centre for Healthy Lungs, The University of Newcastle, Newcastle, NSW, Australia
| | - Katherine J Baines
- Priority Research Centre for Healthy Lungs, The University of Newcastle, Newcastle, NSW, Australia
| | - Louise E Donnelly
- Airway Disease Section, National Heart and Lung Institute, Imperial College London, London, UK
| | - Johan Bylund
- Department of Oral Microbiology and Immunology, Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
| | - Anna Karlsson
- Department of Rheumatology and Inflammation Research, Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
| | - Jodie L Simpson
- Priority Research Centre for Healthy Lungs, The University of Newcastle, Newcastle, NSW, Australia.
| |
Collapse
|
38
|
Singh V, Phukan UJ. Interaction of host and Staphylococcus aureus protease-system regulates virulence and pathogenicity. Med Microbiol Immunol 2018; 208:585-607. [PMID: 30483863 DOI: 10.1007/s00430-018-0573-y] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2018] [Accepted: 11/22/2018] [Indexed: 02/06/2023]
Abstract
Staphylococcus aureus causes various health care- and community-associated infections as well as certain chronic TH2 driven inflammatory diseases. It is a potent pathogen with serious virulence and associated high morbidity. Severe pathogenicity is accredited to the S. aureus secreted virulence factors such as proteases and host protease modulators. These virulence factors promote adhesion and invasion of bacteria through damage of tight junction barrier and keratinocytes. They inhibit activation and transmigration of various immune cells such as neutrophils (and neutrophil proteases) to evade opsono-phagocytosis and intracellular bacterial killing. Additionally, they protect the bacteria from extracellular killing by disrupting integrity of extracellular matrix. Platelet activation and agglutination is also impaired by these factors. They also block the classical as well as alternative pathways of complement activation and assist in spread of infection through blood and tissue. As these factors are exquisite factors of S. aureus mediated disease development, we have focused on review of diversification of various protease-system associated virulence factors, their structural building, diverse role in disease development and available therapeutic counter measures. This review summarises the role of protease-associated virulence factors during invasion and progression of disease.
Collapse
Affiliation(s)
- Vigyasa Singh
- Molecular Bioprospection Department, CSIR-Central Institute of Medicinal and Aromatic Plants, P.O. CIMAP, Lucknow, 226015, India
| | - Ujjal Jyoti Phukan
- School of Life Science, Jawaharlal Nehru University, New Delhi, 110067, India.
| |
Collapse
|
39
|
Casals C, Campanero-Rhodes MA, García-Fojeda B, Solís D. The Role of Collectins and Galectins in Lung Innate Immune Defense. Front Immunol 2018; 9:1998. [PMID: 30233589 PMCID: PMC6131309 DOI: 10.3389/fimmu.2018.01998] [Citation(s) in RCA: 52] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2018] [Accepted: 08/14/2018] [Indexed: 12/16/2022] Open
Abstract
Different families of endogenous lectins use complementary defense strategies against pathogens. They may recognize non-self glycans typically found on pathogens and/or host glycans. The collectin and galectin families are prominent examples of these two lectin categories. Collectins are C-type lectins that contain a carbohydrate recognition domain and a collagen-like domain. Members of this group include surfactant protein A (SP-A) and D (SP-D), secreted by the alveolar epithelium to the alveolar fluid. Lung collectins bind to several microorganisms, which results in pathogen aggregation and/or killing, and enhances phagocytosis of pathogens by alveolar macrophages. Moreover, SP-A and SP-D influence macrophage responses, contributing to resolution of inflammation, and SP-A is essential for tissue-repair functions of macrophages. Galectins also function by interacting directly with pathogens or by modulating the immune system in response to the infection. Direct binding may result in enhanced or impaired infection of target cells, or can have microbicidal effects. Immunomodulatory effects of galectins include recruitment of immune cells to the site of infection, promotion of neutrophil function, and stimulation of the bactericidal activity of infected macrophages. Moreover, intracellular galectins can serve as danger receptors, promoting autophagy of the invading pathogen. This review will focus on the role of collectins and galectins in pathogen clearance and immune response activation in infectious diseases of the respiratory system.
Collapse
Affiliation(s)
- Cristina Casals
- Centro de Investigación Biomédica en Red de Enfermedades Respiratorias (CIBERES), Instituto de Salud Carlos III, Madrid, Spain.,Departamento de Bioquímica y Biología Molecular, Universidad Complutense de Madrid, Madrid, Spain
| | - María A Campanero-Rhodes
- Centro de Investigación Biomédica en Red de Enfermedades Respiratorias (CIBERES), Instituto de Salud Carlos III, Madrid, Spain.,Instituto de Química Física Rocasolano, CSIC, Madrid, Spain
| | - Belén García-Fojeda
- Centro de Investigación Biomédica en Red de Enfermedades Respiratorias (CIBERES), Instituto de Salud Carlos III, Madrid, Spain.,Departamento de Bioquímica y Biología Molecular, Universidad Complutense de Madrid, Madrid, Spain
| | - Dolores Solís
- Centro de Investigación Biomédica en Red de Enfermedades Respiratorias (CIBERES), Instituto de Salud Carlos III, Madrid, Spain.,Instituto de Química Física Rocasolano, CSIC, Madrid, Spain
| |
Collapse
|
40
|
Sato S. Cytosolic Galectins and Their Release and Roles as Carbohydrate-Binding Proteins in Host–Pathogen Interaction. TRENDS GLYCOSCI GLYC 2018. [DOI: 10.4052/tigg.1739.1se] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Affiliation(s)
- Sachiko Sato
- Glycobiology and Bioimaging Laboratory, Research Centre for Infectious Diseases, Faculty of Medicine, Laval University
| |
Collapse
|
41
|
Robinson BS, Arthur CM, Kamili NA, Stowell SR. Galectin Regulation of Host Microbial Interactions. TRENDS GLYCOSCI GLYC 2018. [DOI: 10.4052/tigg.1738.1se] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Affiliation(s)
- Brian S. Robinson
- Center for Transfusion Medicine and Cellular Therapies, Department of Laboratory Medicine and Pathology, Emory University School of Medicine
| | - Connie M. Arthur
- Center for Transfusion Medicine and Cellular Therapies, Department of Laboratory Medicine and Pathology, Emory University School of Medicine
| | - Nourine A. Kamili
- Center for Transfusion Medicine and Cellular Therapies, Department of Laboratory Medicine and Pathology, Emory University School of Medicine
| | - Sean R. Stowell
- Center for Transfusion Medicine and Cellular Therapies, Department of Laboratory Medicine and Pathology, Emory University School of Medicine
| |
Collapse
|
42
|
Sciacchitano S, Lavra L, Morgante A, Ulivieri A, Magi F, De Francesco GP, Bellotti C, Salehi LB, Ricci A. Galectin-3: One Molecule for an Alphabet of Diseases, from A to Z. Int J Mol Sci 2018; 19:ijms19020379. [PMID: 29373564 PMCID: PMC5855601 DOI: 10.3390/ijms19020379] [Citation(s) in RCA: 249] [Impact Index Per Article: 35.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2017] [Revised: 01/18/2018] [Accepted: 01/22/2018] [Indexed: 02/07/2023] Open
Abstract
Galectin-3 (Gal-3) regulates basic cellular functions such as cell-cell and cell-matrix interactions, growth, proliferation, differentiation, and inflammation. It is not surprising, therefore, that this protein is involved in the pathogenesis of many relevant human diseases, including cancer, fibrosis, chronic inflammation and scarring affecting many different tissues. The papers published in the literature have progressively increased in number during the last decades, testifying the great interest given to this protein by numerous researchers involved in many different clinical contexts. Considering the crucial role exerted by Gal-3 in many different clinical conditions, Gal-3 is emerging as a new diagnostic, prognostic biomarker and as a new promising therapeutic target. The current review aims to extensively examine the studies published so far on the role of Gal-3 in all the clinical conditions and diseases, listed in alphabetical order, where it was analyzed.
Collapse
Affiliation(s)
- Salvatore Sciacchitano
- Department of Clinical and Molecular Medicine, Sapienza University, Policlinico Umberto I, Viale Regina Elena 324, 00161 Rome, Italy.
- Laboratory of Biomedical Research, Niccolò Cusano University Foundation, Via Don Carlo Gnocchi 3, 00166 Rome, Italy.
| | - Luca Lavra
- Laboratory of Biomedical Research, Niccolò Cusano University Foundation, Via Don Carlo Gnocchi 3, 00166 Rome, Italy.
| | - Alessandra Morgante
- Laboratory of Biomedical Research, Niccolò Cusano University Foundation, Via Don Carlo Gnocchi 3, 00166 Rome, Italy.
| | - Alessandra Ulivieri
- Laboratory of Biomedical Research, Niccolò Cusano University Foundation, Via Don Carlo Gnocchi 3, 00166 Rome, Italy.
| | - Fiorenza Magi
- Laboratory of Biomedical Research, Niccolò Cusano University Foundation, Via Don Carlo Gnocchi 3, 00166 Rome, Italy.
| | - Gian Paolo De Francesco
- Department of Oncological Science, Breast Unit, St Andrea University Hospital, Via di Grottarossa, 1035/39, 00189 Rome, Italy.
| | - Carlo Bellotti
- Operative Unit Surgery of Thyroid and Parathyroid, Sapienza University of Rome, S. Andrea Hospital, Via di Grottarossa, 1035/39, 00189 Rome, Italy.
| | - Leila B Salehi
- Laboratory of Biomedical Research, Niccolò Cusano University Foundation, Via Don Carlo Gnocchi 3, 00166 Rome, Italy.
- Department of Biopathology and Diagnostic Imaging, Tor Vergata University, Via Montpellier 1, 00133 Rome, Italy.
| | - Alberto Ricci
- Department of Clinical and Molecular Medicine, Sapienza University, Policlinico Umberto I, Viale Regina Elena 324, 00161 Rome, Italy.
| |
Collapse
|
43
|
Sundqvist M, Welin A, Elmwall J, Osla V, Nilsson UJ, Leffler H, Bylund J, Karlsson A. Galectin-3 type-C self-association on neutrophil surfaces; The carbohydrate recognition domain regulates cell function. J Leukoc Biol 2018; 103:341-353. [PMID: 29345346 DOI: 10.1002/jlb.3a0317-110r] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2017] [Revised: 09/17/2017] [Accepted: 10/16/2017] [Indexed: 11/09/2022] Open
Abstract
Galectin-3 is an endogenous β-galactoside-binding lectin comprising a carbohydrate recognition domain (CRD) linked to a collagen-like N-domain. Both domains are required for galectin-3 to induce cellular effects; a C-terminal fragment of galectin-3, galectin-3C, containing the CRD but lacking the N-domain, binds cell surface glycoconjugates but does not induce cellular effects since cross-linking promoted by the N-domain is thought to be required. Instead, galectin-3C is proposed to antagonize the effects of galectin-3 by competing for binding sites. The aim of this study was to investigate the effects of galectin-3C on galectin-3 interactions with human neutrophils. Recombinant galectin-3C inhibited galectin-3-induced production of reactive oxygen species in primed neutrophils. Surprisingly, this inhibition was not due to competitive inhibition of galectin-3 binding to the cells. In contrast, galectin-3C potentiated galectin-3 binding, in line with emerging evidence that galectin-3 can aggregate not only through the N-domain but also through the CRD. The cell surface interaction between galectin-3C and galectin-3 was corroborated by colocalization of fluorescently labeled galectin-3 and galectin-3C. Galectin-3C can be generated in vivo through cleavage of galectin-3 by proteases. Indeed, in circulation, galectin-3 and galectin-3C were both attached to the cell surface of neutrophils, which displayed great capacity to bind additional galectin-3 and galectin-3C. In conclusion, galectin-3C enhances galectin-3 binding to neutrophils by nonactivating type-C self-association, in parallel to inhibiting neutrophil activation by galectin-3 (induced by type-N self-association). This implicates type-C self-association as a termination system for galectin-3-induced cell activation, with the purpose of avoiding oxidant-dependent tissue damage.
Collapse
Affiliation(s)
- Martina Sundqvist
- Department of Rheumatology and Inflammation Research, Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
| | - Amanda Welin
- Department of Rheumatology and Inflammation Research, Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
| | - Jonas Elmwall
- Department of Rheumatology and Inflammation Research, Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
| | - Veronica Osla
- Department of Rheumatology and Inflammation Research, Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
| | - Ulf J Nilsson
- Centre for Analysis and Synthesis, Department of Chemistry, Lund University, Lund, Sweden
| | - Hakon Leffler
- Department of Laboratory Medicine, Section of Microbiology, Immunology and Glycobiology, Lund University, Lund, Sweden
| | - Johan Bylund
- Department of Oral Microbiology and Immunology, Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
| | - Anna Karlsson
- Department of Rheumatology and Inflammation Research, Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
| |
Collapse
|
44
|
Abstract
Group A streptococcus (GAS) is an important human pathogen that causes a wide variety of cutaneous and systemic infections. Although originally thought to be an extracellular bacterium, numerous studies have demonstrated that GAS can trigger internalization into nonimmune cells to escape from immune surveillance or antibiotic-mediated killing. Epithelial cells possess a defense mechanism involving autophagy-mediated targeting and killing of GAS within lysosome-fused autophagosomes. In endothelial cells, in contrast, we previously showed that autophagy is not sufficient for GAS killing. In the present study, we showed higher galectin-3 (Gal-3) expression and lower Gal-8 expression in endothelial cells than in epithelial cells. The recruitment of Gal-3 to GAS is higher and the recruitment of Gal-8 to GAS is lower in endothelial cells than in epithelial cells. We further showed that Gal-3 promotes GAS replication and diminishes the recruitment of Gal-8 and ubiquitin, the latter of which is a critical protein for autophagy sequestration. After knockdown of Gal-3 in endothelial cells, the colocalization of Gal-8, parkin, and ubiquitin-decorated GAS is significantly increased, as is the interaction of Gal-8 and parkin, an E3 ligase. Furthermore, inhibition of Gal-8 in epithelial cells attenuates recruitment of parkin; both Gal-8 and parkin contribute to ubiquitin recruitment and GAS elimination. Animal studies confirmed that Gal-3-knockout mice develop less-severe skin damage and that GAS replication can be detected only in the air pouch and not in organs and endothelial cells. These results demonstrate that Gal-3 inhibits ubiquitin recruitment by blocking Gal-8 and parkin recruitment, resulting in GAS replication in endothelial cells. In epithelial cells, GAS can be efficiently killed within the lysosome-fused autophaosome compartment. However, we previously showed that, in spite of LC-3 recruitment, the autophagic machinery is not sufficient for GAS killing in endothelial cells. In this report, we provide the first evidence that Gal-3, highly expressed in endothelial cells, blocks the tagging of ubiquitin to GAS by inhibiting recruitment of Gal-8 and parkin, leading to an enhancement of GAS replication. We also provide the first demonstration that Gal-8 can interact with parkin, the critical E3 ligase, for resistance to intracellular bacteria by facilitating the decoration of bacteria with ubiquitin chains. Our findings reveal that differential levels of Gal-3 and Gal-8 expression and recruitment to GAS between epithelial cells and endothelial cells may contribute to the different outcomes of GAS elimination or survival and growth of GAS in these two types of cells.
Collapse
|
45
|
Ho JE, Gao W, Levy D, Santhanakrishnan R, Araki T, Rosas IO, Hatabu H, Latourelle JC, Nishino M, Dupuis J, Washko GR, O'Connor GT, Hunninghake GM. Galectin-3 Is Associated with Restrictive Lung Disease and Interstitial Lung Abnormalities. Am J Respir Crit Care Med 2017; 194:77-83. [PMID: 26771117 DOI: 10.1164/rccm.201509-1753oc] [Citation(s) in RCA: 65] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
RATIONALE Galectin-3 (Gal-3) has been implicated in the development of pulmonary fibrosis in experimental studies, and Gal-3 levels have been found to be elevated in small studies of human pulmonary fibrosis. OBJECTIVES We sought to study whether circulating Gal-3 concentrations are elevated early in the course of pulmonary fibrosis. METHODS We examined 2,596 Framingham Heart Study participants (mean age, 57 yr; 54% women; 14% current smokers) who underwent Gal-3 assessment using plasma samples and pulmonary function testing between 1995 and 1998. Of this sample, 1,148 underwent subsequent volumetric chest computed tomography. MEASUREMENTS AND MAIN RESULTS Higher Gal-3 concentrations were associated with lower lung volumes (1.4% decrease in percentage of predicted FEV1 per 1 SD increase in log Gal-3; 95% confidence interval [CI], 0.8-2.0%; P < 0.001; 1.2% decrease in percentage of predicted FVC; 95% CI, 0.6-1.8%; P < 0.001) and decreased diffusing capacity of the lung for carbon monoxide (2.1% decrease; 95% CI, 1.3-2.9%; P < 0.001). These associations remained significant after multivariable adjustment (P ≤ 0.008 for all). Compared with the lowest quartile, participants in the highest Gal-3 quartile were more than twice as likely to have interstitial lung abnormalities visualized by computed tomography (multivariable-adjusted odds ratio, 2.67; 95% CI, 1.49-4.76; P < 0.001). CONCLUSIONS Elevated Gal-3 concentrations are associated with interstitial lung abnormalities coupled with a restrictive pattern, including decreased lung volumes and altered gas exchange. These findings suggest a potential role for Gal-3 in early stages of pulmonary fibrosis.
Collapse
Affiliation(s)
- Jennifer E Ho
- 1 Division of Cardiology, Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts.,2 Framingham Heart Study, NHLBI, Framingham, Massachusetts
| | - Wei Gao
- 3 Department of Biostatistics, Boston University School of Public Health, Boston, Massachusetts
| | - Daniel Levy
- 2 Framingham Heart Study, NHLBI, Framingham, Massachusetts.,4 Population Sciences Branch, Division of Intramural Research, NHLBI, Bethesda, Maryland
| | | | - Tetsuro Araki
- 6 Center for Pulmonary Functional Imaging.,7 Department of Radiology, and
| | - Ivan O Rosas
- 8 Pulmonary and Critical Care Division, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Hiroto Hatabu
- 6 Center for Pulmonary Functional Imaging.,7 Department of Radiology, and
| | - Jeanne C Latourelle
- 9 Pulmonary Center, Department of Medicine, and.,10 Department of Neurology, Boston University School of Medicine, Boston, Massachusetts; and
| | - Mizuki Nishino
- 6 Center for Pulmonary Functional Imaging.,7 Department of Radiology, and
| | - Josée Dupuis
- 2 Framingham Heart Study, NHLBI, Framingham, Massachusetts.,3 Department of Biostatistics, Boston University School of Public Health, Boston, Massachusetts
| | - George R Washko
- 6 Center for Pulmonary Functional Imaging.,8 Pulmonary and Critical Care Division, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| | - George T O'Connor
- 2 Framingham Heart Study, NHLBI, Framingham, Massachusetts.,9 Pulmonary Center, Department of Medicine, and
| | - Gary M Hunninghake
- 6 Center for Pulmonary Functional Imaging.,8 Pulmonary and Critical Care Division, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| |
Collapse
|
46
|
Kalinec GM, Lomberk G, Urrutia RA, Kalinec F. Resolution of Cochlear Inflammation: Novel Target for Preventing or Ameliorating Drug-, Noise- and Age-related Hearing Loss. Front Cell Neurosci 2017; 11:192. [PMID: 28736517 PMCID: PMC5500902 DOI: 10.3389/fncel.2017.00192] [Citation(s) in RCA: 86] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2017] [Accepted: 06/20/2017] [Indexed: 12/11/2022] Open
Abstract
A significant number of studies support the idea that inflammatory responses are intimately associated with drug-, noise- and age-related hearing loss (DRHL, NRHL and ARHL). Consequently, several clinical strategies aimed at reducing auditory dysfunction by preventing inflammation are currently under intense scrutiny. Inflammation, however, is a normal adaptive response aimed at restoring tissue functionality and homeostasis after infection, tissue injury and even stress under sterile conditions, and suppressing it could have unintended negative consequences. Therefore, an appropriate approach to prevent or ameliorate DRHL, NRHL and ARHL should involve improving the resolution of the inflammatory process in the cochlea rather than inhibiting this phenomenon. The resolution of inflammation is not a passive response but rather an active, highly controlled and coordinated process. Inflammation by itself produces specialized pro-resolving mediators with critical functions, including essential fatty acid derivatives (lipoxins, resolvins, protectins and maresins), proteins and peptides such as annexin A1 and galectins, purines (adenosine), gaseous mediators (NO, H2S and CO), as well as neuromodulators like acetylcholine and netrin-1. In this review article, we describe recent advances in the understanding of the resolution phase of inflammation and highlight therapeutic strategies that might be useful in preventing inflammation-induced cochlear damage. In particular, we emphasize beneficial approaches that have been tested in pre-clinical models of inflammatory responses induced by recognized ototoxic drugs such as cisplatin and aminoglycoside antibiotics. Since these studies suggest that improving the resolution process could be useful for the prevention of inflammation-associated diseases in humans, we discuss the potential application of similar strategies to prevent or mitigate DRHL, NRHL and ARHL.
Collapse
Affiliation(s)
- Gilda M Kalinec
- Laboratory of Auditory Cell Biology, Department of Head and Neck Surgery, David Geffen School of Medicine, University of CaliforniaLos Angeles, Los Angeles, CA, United States
| | - Gwen Lomberk
- Epigenetics and Chromatin Dynamics Laboratory, Translational Epigenomic Program, Center for Individualized Medicine (CIM) Mayo ClinicRochester, MN, United States
| | - Raul A Urrutia
- Epigenetics and Chromatin Dynamics Laboratory, Translational Epigenomic Program, Center for Individualized Medicine (CIM) Mayo ClinicRochester, MN, United States
| | - Federico Kalinec
- Laboratory of Auditory Cell Biology, Department of Head and Neck Surgery, David Geffen School of Medicine, University of CaliforniaLos Angeles, Los Angeles, CA, United States
| |
Collapse
|
47
|
Galectin-3 Is a Target for Proteases Involved in the Virulence of Staphylococcus aureus. Infect Immun 2017; 85:IAI.00177-17. [PMID: 28438975 DOI: 10.1128/iai.00177-17] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2017] [Accepted: 04/13/2017] [Indexed: 01/21/2023] Open
Abstract
Staphylococcus aureus is a major cause of skin and soft tissue infection. The bacterium expresses four major proteases that are emerging as virulence factors: aureolysin (Aur), V8 protease (SspA), staphopain A (ScpA), and staphopain B (SspB). We hypothesized that human galectin-3, a β-galactoside-binding lectin involved in immune regulation and antimicrobial defense, is a target for these proteases and that proteolysis of galectin-3 is a novel immune evasion mechanism. Indeed, supernatants from laboratory strains and clinical isolates of S. aureus caused galectin-3 degradation. Similar proteolytic capacities were found in Staphylococcus epidermidis isolates but not in Staphylococcus saprophyticus Galectin-3-induced activation of the neutrophil NADPH oxidase was abrogated by bacterium-derived proteolysis of galectin-3, and SspB was identified as the major protease responsible. The impact of galectin-3 and protease expression on S. aureus virulence was studied in a murine skin infection model. In galectin-3+/+ mice, SspB-expressing S. aureus caused larger lesions and resulted in higher bacterial loads than protease-lacking bacteria. No such difference in bacterial load or lesion size was detected in galectin-3-/- mice, which overall showed smaller lesion sizes than the galectin-3+/+ animals. In conclusion, the staphylococcal protease SspB inactivates galectin-3, abrogating its stimulation of oxygen radical production in human neutrophils and increasing tissue damage during skin infection.
Collapse
|
48
|
Leone DA, Peschel A, Brown M, Schachner H, Ball MJ, Gyuraszova M, Salzer-Muhar U, Fukuda M, Vizzardelli C, Bohle B, Rees AJ, Kain R. Surface LAMP-2 Is an Endocytic Receptor That Diverts Antigen Internalized by Human Dendritic Cells into Highly Immunogenic Exosomes. THE JOURNAL OF IMMUNOLOGY 2017; 199:531-546. [DOI: 10.4049/jimmunol.1601263] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/21/2016] [Accepted: 05/10/2017] [Indexed: 12/13/2022]
|
49
|
Dicker AJ, Crichton ML, Pumphrey EG, Cassidy AJ, Suarez-Cuartin G, Sibila O, Furrie E, Fong CJ, Ibrahim W, Brady G, Einarsson GG, Elborn JS, Schembri S, Marshall SE, Palmer CNA, Chalmers JD. Neutrophil extracellular traps are associated with disease severity and microbiota diversity in patients with chronic obstructive pulmonary disease. J Allergy Clin Immunol 2017; 141:117-127. [PMID: 28506850 PMCID: PMC5751731 DOI: 10.1016/j.jaci.2017.04.022] [Citation(s) in RCA: 196] [Impact Index Per Article: 24.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2016] [Revised: 03/28/2017] [Accepted: 04/05/2017] [Indexed: 11/25/2022]
Abstract
Background Neutrophil extracellular traps (NETs) have been observed in the airway in patients with chronic obstructive pulmonary disease (COPD), but their clinical and pathophysiologic implications have not been defined. Objective We sought to determine whether NETs are associated with disease severity in patients with COPD and how they are associated with microbiota composition and airway neutrophil function. Methods NET protein complexes (DNA-elastase and histone-elastase complexes), cell-free DNA, and neutrophil biomarkers were quantified in soluble sputum and serum from patients with COPD during periods of disease stability and during exacerbations and compared with clinical measures of disease severity and the sputum microbiome. Peripheral blood and airway neutrophil function were evaluated by means of flow cytometry ex vivo and experimentally after stimulation of NET formation. Results Sputum NET complexes were associated with the severity of COPD evaluated by using the composite Global Initiative for Obstructive Lung Disease scale (P < .0001). This relationship was due to modest correlations between NET complexes and FEV1, symptoms evaluated by using the COPD assessment test, and higher levels of NET complexes in patients with frequent exacerbations (P = .002). Microbiota composition was heterogeneous, but there was a correlation between NET complexes and both microbiota diversity (P = .009) and dominance of Haemophilus species operational taxonomic units (P = .01). Ex vivo airway neutrophil phagocytosis of bacteria was reduced in patients with increased sputum NET complexes. Consistent results were observed regardless of the method of quantifying sputum NETs. Failure of phagocytosis could be induced experimentally by incubating healthy control neutrophils with soluble sputum from patients with COPD. Conclusion NET formation is increased in patients with severe COPD and associated with more frequent exacerbations and a loss of microbiota diversity.
Collapse
Affiliation(s)
- Alison J Dicker
- Scottish Centre for Respiratory Research, University of Dundee, Ninewells Hospital and Medical School, Dundee, United Kingdom
| | - Megan L Crichton
- Scottish Centre for Respiratory Research, University of Dundee, Ninewells Hospital and Medical School, Dundee, United Kingdom
| | - Eleanor G Pumphrey
- Scottish Centre for Respiratory Research, University of Dundee, Ninewells Hospital and Medical School, Dundee, United Kingdom
| | - Andrew J Cassidy
- Scottish Centre for Respiratory Research, University of Dundee, Ninewells Hospital and Medical School, Dundee, United Kingdom
| | - Guillermo Suarez-Cuartin
- Respiratory Department, Hospital de la Santa Creu i Sant Pau, Institut d'Invesitgacio Biomedica (IIB) Sant Pau, Barcelona, Spain
| | - Oriol Sibila
- Respiratory Department, Hospital de la Santa Creu i Sant Pau, Institut d'Invesitgacio Biomedica (IIB) Sant Pau, Barcelona, Spain
| | - Elizabeth Furrie
- Scottish Centre for Respiratory Research, University of Dundee, Ninewells Hospital and Medical School, Dundee, United Kingdom
| | - Christopher J Fong
- Scottish Centre for Respiratory Research, University of Dundee, Ninewells Hospital and Medical School, Dundee, United Kingdom
| | - Wasyla Ibrahim
- Scottish Centre for Respiratory Research, University of Dundee, Ninewells Hospital and Medical School, Dundee, United Kingdom
| | - Gill Brady
- Scottish Centre for Respiratory Research, University of Dundee, Ninewells Hospital and Medical School, Dundee, United Kingdom
| | - Gisli G Einarsson
- Centre for Infection and Immunity, School of Medicine, Dentistry and Biomedical Sciences, Queen's University Belfast, Belfast, United Kingdom
| | - J Stuart Elborn
- Centre for Infection and Immunity, School of Medicine, Dentistry and Biomedical Sciences, Queen's University Belfast, Belfast, United Kingdom; National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | - Stuart Schembri
- Scottish Centre for Respiratory Research, University of Dundee, Ninewells Hospital and Medical School, Dundee, United Kingdom
| | - Sara E Marshall
- Division of Molecular & Clinical Medicine, School of Medicine, University of Dundee, and the Wellcome Trust, London, United Kingdom
| | - Colin N A Palmer
- Scottish Centre for Respiratory Research, University of Dundee, Ninewells Hospital and Medical School, Dundee, United Kingdom
| | - James D Chalmers
- Scottish Centre for Respiratory Research, University of Dundee, Ninewells Hospital and Medical School, Dundee, United Kingdom.
| |
Collapse
|
50
|
The Many Roles of Galectin-3, a Multifaceted Molecule, in Innate Immune Responses against Pathogens. Mediators Inflamm 2017; 2017:9247574. [PMID: 28607536 PMCID: PMC5457773 DOI: 10.1155/2017/9247574] [Citation(s) in RCA: 128] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2017] [Revised: 04/08/2017] [Accepted: 04/18/2017] [Indexed: 01/06/2023] Open
Abstract
Galectins are a group of evolutionarily conserved proteins with the ability to bind β-galactosides through characteristic carbohydrate-recognition domains (CRD). Galectin-3 is structurally unique among all galectins as it contains a C-terminal CRD linked to an N-terminal protein-binding domain, being the only chimeric galectin. Galectin-3 participates in many functions, both intra- and extracellularly. Among them, a prominent role for Galectin-3 in inflammation has been recognized. Galectin-3 has also been shown to directly bind to pathogens and to have various effects on the functions of the cells of the innate immune system. Thanks to these two properties, Galectin-3 participates in several ways in the innate immune response against invading pathogens. Galectin-3 has been proposed to function not only as a pattern-recognition receptor (PRR) but also as a danger-associated molecular pattern (DAMP). In this review, we analyze the various roles that have been assigned to Galectin-3, both as a PRR and as a DAMP, in the context of immune responses against pathogenic microorganisms.
Collapse
|