1
|
Zhong J, Li J, Wu D, Deng X, Lu Y, Yu XQ. Characterization and functional analysis of Spodoptera litura galectins. PEST MANAGEMENT SCIENCE 2025. [PMID: 39898411 DOI: 10.1002/ps.8685] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/24/2024] [Revised: 12/20/2024] [Accepted: 01/14/2025] [Indexed: 02/04/2025]
Abstract
BACKGROUND Insects rely on their effective innate immune system to defend against pathogen infection, and pattern recognition receptors (PRRs) play a crucial role in insect immunity. Galectins, a family of animal β-galactoside-binding lectins, may serve as PRRs. Mammalian galectins are involved in diverse physiological processes; however, less is known about functions of insect galectins. RESULT In this study, we focused on an agricultural pest Spodoptera litura and identified a total of 13 Slgalectins, and a functional study of Slgalectin-4 and Slgalectin-7 was conducted. Both recombinant Slgalectin-4 and Slgalectin-7 proteins directly bound to Bacillus thuringiensis and an entomopathogenic fungus Metarhizium rileyi. Importantly, supplementing recombinant Slgalectin-4 to S. litura larvae significantly increased larval survival after M. rileyi infection but accelerated larvae death after B. thuringiensis infection, whereas supplementing Slgalectin-7 increased larval survival after B. thuringiensis infection but did not influence larval survival after M. rileyi infection. Supplementing both Slgalectin-4 and Slgalectin-7 altered the expression of some antimicrobial peptide genes in larval hemocytes after microbial infection, which may partly account for the roles of Slgalectin-4 and -7 in larval survival. CONCLUSION Our findings revealed that Slgalectin-4 and -7 may serve as PRRs to bind different pathogens and alter expression of immune effector genes such as antimicrobial peptide genes, eventually modulating immune responses of S. litura larvae to pathogens such as B. thuringiensis and M. rileyi. © 2025 Society of Chemical Industry.
Collapse
Affiliation(s)
- Jielai Zhong
- Guangdong Provincial Key Laboratory of Insect Developmental Biology and Applied Technology, Guangzhou Key Laboratory of Insect Development Regulation and Application Research, Institute of Insect Science and Technology, School of Life Sciences, South China Normal University, Guangzhou, China
| | - Jin Li
- Guangdong Provincial Key Laboratory of Insect Developmental Biology and Applied Technology, Guangzhou Key Laboratory of Insect Development Regulation and Application Research, Institute of Insect Science and Technology, School of Life Sciences, South China Normal University, Guangzhou, China
| | - Denghui Wu
- Guangdong Provincial Key Laboratory of Insect Developmental Biology and Applied Technology, Guangzhou Key Laboratory of Insect Development Regulation and Application Research, Institute of Insect Science and Technology, School of Life Sciences, South China Normal University, Guangzhou, China
| | - Xinyue Deng
- Guangdong Provincial Key Laboratory of Insect Developmental Biology and Applied Technology, Guangzhou Key Laboratory of Insect Development Regulation and Application Research, Institute of Insect Science and Technology, School of Life Sciences, South China Normal University, Guangzhou, China
| | - Yuzhen Lu
- Guangdong Provincial Key Laboratory of Insect Developmental Biology and Applied Technology, Guangzhou Key Laboratory of Insect Development Regulation and Application Research, Institute of Insect Science and Technology, School of Life Sciences, South China Normal University, Guangzhou, China
| | - Xiao-Qiang Yu
- Guangdong Provincial Key Laboratory of Insect Developmental Biology and Applied Technology, Guangzhou Key Laboratory of Insect Development Regulation and Application Research, Institute of Insect Science and Technology, School of Life Sciences, South China Normal University, Guangzhou, China
| |
Collapse
|
2
|
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
|
3
|
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
|
4
|
Song Q, Li Q, Yang Y, Gao H, Han F. Antimicrobial Functions of Galectins from Fish, Mollusks, and Crustaceans: A Review. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2024; 72:24895-24907. [PMID: 39471068 DOI: 10.1021/acs.jafc.4c05412] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/01/2024]
Abstract
Galectins are a member of the β-galactoside binding protein family, which play a pivotal role in the immune defense of vertebrates as a pattern recognition receptor and occupy an important position in the innate immune system of invertebrates. The study of galectins in aquatic organisms has only recently emerged. Galectins in aquatic animals exhibit agglutination activity toward bacteria, inhibit bacterial growth, and enhance phagocytosis of immune cells. Additionally, some galectins contribute to the antiviral immune defenses of aquatic animals. This review aims to review recent advancements in the antimicrobial mechanisms, molecular structures, and evolution of galectins from fish, mollusks, and crustaceans. The antimicrobial galectins, as crucial components in the innate immune defense, pave new avenues for developing innovative disease control strategies in aquaculture.
Collapse
Affiliation(s)
- Qing Song
- Strait Institute of Flexible Electronics (SIFE, Future Technologies), Fujian Key Laboratory of Flexible Electronics, Fujian Normal University and Strait Laboratory of Flexible Electronics (SLoFE), Fuzhou 350117, Fujian, China
| | - Qiaoying Li
- State Key Laboratory of Mariculture Breeding, Key Laboratory of Healthy Mariculture for the East China Sea, Fisheries College, Jimei University, Xiamen, Fujian 361021, China
| | - Yao Yang
- State Key Laboratory of Mariculture Breeding, Key Laboratory of Healthy Mariculture for the East China Sea, Fisheries College, Jimei University, Xiamen, Fujian 361021, China
| | - Haijun Gao
- Strait Institute of Flexible Electronics (SIFE, Future Technologies), Fujian Key Laboratory of Flexible Electronics, Fujian Normal University and Strait Laboratory of Flexible Electronics (SLoFE), Fuzhou 350117, Fujian, China
| | - Fang Han
- State Key Laboratory of Mariculture Breeding, Key Laboratory of Healthy Mariculture for the East China Sea, Fisheries College, Jimei University, Xiamen, Fujian 361021, China
| |
Collapse
|
5
|
Huang Y, Wang Y, Huang X, Yu X. Unveiling the overlooked fungi: the vital of gut fungi in inflammatory bowel disease and colorectal cancer. Gut Pathog 2024; 16:59. [PMID: 39407244 PMCID: PMC11481806 DOI: 10.1186/s13099-024-00651-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/16/2024] [Accepted: 09/27/2024] [Indexed: 10/19/2024] Open
Abstract
The fungi of the human microbiota play important roles in the nutritional metabolism and immunological balance of the host. Recently, research has increasingly emphasised the role of fungi in modulating inflammation in intestinal diseases and maintaining health in this environment. It is therefore necessary to understand more clearly the interactions and mechanisms of the microbiota/pathogen/host relationship and the resulting inflammatory processes, as well as to offer new insights into the prevention, diagnosis and treatment of inflammatory bowel disease (IBD), colorectal cancer (CRC) and other intestinal pathologies. In this review, we comprehensively elucidate the fungal-associated pathogenic mechanisms of intestinal inflammation in IBD and related CRC, with an emphasis on three main aspects: the direct effects of fungi and their metabolites on the host, the indirect effects mediated by interactions with other intestinal microorganisms and the immune regulation of the host. Understanding these mechanisms will enable the development of innovative approaches based on the use of fungi from the resident human microbiota such as dietary interventions, fungal probiotics and faecal microbiota transplantation in the prevention, diagnosis and treatment of intestinal diseases.
Collapse
Affiliation(s)
- Yilin Huang
- School of Basic Medical Sciences, Jiangxi Medical College, Nanchang University, Nanchang, 330006, China
- Huankui Academy, Jiangxi Medical College, Nanchang University, Nanchang, 330031, China
| | - Yang Wang
- School of Basic Medical Sciences, Jiangxi Medical College, Nanchang University, Nanchang, 330006, China
| | - Xiaotian Huang
- School of Basic Medical Sciences, Jiangxi Medical College, Nanchang University, Nanchang, 330006, China.
| | - Xiaomin Yu
- School of Basic Medical Sciences, Jiangxi Medical College, Nanchang University, Nanchang, 330006, China.
| |
Collapse
|
6
|
Pinho SS, Alves I, Gaifem J, Rabinovich GA. Immune regulatory networks coordinated by glycans and glycan-binding proteins in autoimmunity and infection. Cell Mol Immunol 2023; 20:1101-1113. [PMID: 37582971 PMCID: PMC10541879 DOI: 10.1038/s41423-023-01074-1] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2023] [Accepted: 07/26/2023] [Indexed: 08/17/2023] Open
Abstract
The immune system is coordinated by an intricate network of stimulatory and inhibitory circuits that regulate host responses against endogenous and exogenous insults. Disruption of these safeguard and homeostatic mechanisms can lead to unpredictable inflammatory and autoimmune responses, whereas deficiency of immune stimulatory pathways may orchestrate immunosuppressive programs that contribute to perpetuate chronic infections, but also influence cancer development and progression. Glycans have emerged as essential components of homeostatic circuits, acting as fine-tuners of immunological responses and potential molecular targets for manipulation of immune tolerance and activation in a wide range of pathologic settings. Cell surface glycans, present in cells, tissues and the extracellular matrix, have been proposed to serve as "self-associated molecular patterns" that store structurally relevant biological data. The responsibility of deciphering this information relies on different families of glycan-binding proteins (including galectins, siglecs and C-type lectins) which, upon recognition of specific carbohydrate structures, can recalibrate the magnitude, nature and fate of immune responses. This process is tightly regulated by the diversity of glycan structures and the establishment of multivalent interactions on cell surface receptors and the extracellular matrix. Here we review the spatiotemporal regulation of selected glycan-modifying processes including mannosylation, complex N-glycan branching, core 2 O-glycan elongation, LacNAc extension, as well as terminal sialylation and fucosylation. Moreover, we illustrate examples that highlight the contribution of these processes to the control of immune responses and their integration with canonical tolerogenic pathways. Finally, we discuss the power of glycans and glycan-binding proteins as a source of immunomodulatory signals that could be leveraged for the treatment of autoimmune inflammation and chronic infection.
Collapse
Affiliation(s)
- Salomé S Pinho
- i3S - Institute for Research and Innovation in Health, University of Porto, 4200-135, Porto, Portugal.
- ICBAS-School of Medicine and Biomedical Sciences, University of Porto, 4050-313, Porto, Portugal.
- Faculty of Medicine, University of Porto, 4200-319, Porto, Portugal.
| | - Inês Alves
- i3S - Institute for Research and Innovation in Health, University of Porto, 4200-135, Porto, Portugal
| | - Joana Gaifem
- i3S - Institute for Research and Innovation in Health, University of Porto, 4200-135, Porto, Portugal
| | - Gabriel A Rabinovich
- Laboratorio de Glicomedicina, Instituto de Biología y Medicina Experimental (IBYME), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), C1428, Ciudad de Buenos Aires, Argentina.
- Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, C1428, Ciudad de Buenos Aires, Argentina.
| |
Collapse
|
7
|
Wagner AS, Lumsdaine SW, Mangrum MM, Reynolds TB. Caspofungin-induced β(1,3)-glucan exposure in Candida albicans is driven by increased chitin levels. mBio 2023; 14:e0007423. [PMID: 37377417 PMCID: PMC10470516 DOI: 10.1128/mbio.00074-23] [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: 01/09/2023] [Accepted: 05/04/2023] [Indexed: 06/29/2023] Open
Abstract
To successfully induce disease, Candida albicans must effectively evade the host immune system. One mechanism used by C. albicans to achieve this is to mask immunogenic β(1,3)-glucan epitopes within its cell wall under an outer layer of mannosylated glycoproteins. Consequently, induction of β(1,3)-glucan exposure (unmasking) via genetic or chemical manipulation increases fungal recognition by host immune cells in vitro and attenuates disease during systemic infection in mice. Treatment with the echinocandin caspofungin is one of the most potent drivers of β(1,3)-glucan exposure. Several reports using murine infection models suggest a role for the immune system, and specifically host β(1,3)-glucan receptors, in mediating the efficacy of echinocandin treatment in vivo. However, the mechanism by which caspofungin-induced unmasking occurs is not well understood. In this report, we show that foci of unmasking co-localize with areas of increased chitin within the yeast cell wall in response to caspofungin, and that inhibition of chitin synthesis via nikkomycin Z attenuates caspofungin-induced β(1,3)-glucan exposure. Furthermore, we find that both the calcineurin and Mkc1 mitogen-activated protein kinase pathways work synergistically to regulate β(1,3)-glucan exposure and chitin synthesis in response to drug treatment. When either of these pathways are interrupted, it results in a bimodal population of cells containing either high or low chitin content. Importantly, increased unmasking correlates with increased chitin content within these cells. Microscopy further indicates that caspofungin-induced unmasking correlates with actively growing cells. Collectively, our work presents a model in which chitin synthesis induces unmasking within the cell wall in response to caspofungin in growing cells. IMPORTANCE Systemic candidiasis has reported mortality rates ranging from 20% to 40%. The echinocandins, including caspofungin, are first-line antifungals used to treat systemic candidiasis. However, studies in mice have shown that echinocandin efficacy relies on both its cidal impacts on Candida albicans, as well as a functional immune system to successfully clear invading fungi. In addition to direct C. albicans killing, caspofungin increases exposure (unmasking) of immunogenic β(1,3)-glucan moieties. To evade immune detection, β(1,3)-glucan is normally masked within the C. albicans cell wall. Consequently, unmasked β(1,3)-glucan renders these cells more visible to the host immune system and attenuates disease progression. Therefore, discovery of how caspofungin-induced unmasking occurs is needed to elucidate how the drug facilitates host immune system-mediated clearance in vivo. We report a strong and consistent correlation between chitin deposition and unmasking in response to caspofungin and propose a model in which altered chitin synthesis drives increased unmasking during drug exposure.
Collapse
Affiliation(s)
- Andrew S. Wagner
- Department of Microbiology, University of Tennessee, Knoxville, Tennessee, USA
| | | | - Mikayla M. Mangrum
- Department of Microbiology, University of Tennessee, Knoxville, Tennessee, USA
| | - Todd B. Reynolds
- Department of Microbiology, University of Tennessee, Knoxville, Tennessee, USA
| |
Collapse
|
8
|
Yang Y, Wu B, Li W, Han F. Molecular Characterization of Galectin-3 in Large Yellow Croaker Larimichthys crocea Functioning in Antibacterial Activity. Int J Mol Sci 2023; 24:11539. [PMID: 37511297 PMCID: PMC10380712 DOI: 10.3390/ijms241411539] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2023] [Revised: 07/13/2023] [Accepted: 07/15/2023] [Indexed: 07/30/2023] Open
Abstract
Galectins are proteins that play a crucial role in the innate immune response against pathogenic microorganisms. Previous studies have suggested that Galectin-3 could be a candidate gene for antibacterial immunity in the large yellow croaker Larimichthys crocea. In this study, we cloned the Galectin-3 gene in the large yellow croaker, and named it LcGal-3. The deduced amino acid sequence of LcGal-3 contains a carbohydrate recognition domain with two conserved β-galactoside binding motifs. Quantitative reverse transcription PCR (qRT-PCR) analysis revealed that LcGal-3 was expressed in all the organs/tissues that were tested, with the highest expression level in the gill. In Larimichthys crocea kidney cell lines, LcGal-3 protein was distributed in both the cytoplasm and nucleus. Moreover, we found that the expression of LcGal-3 was significantly upregulated upon infection with Pseudomonas plecoglossicida, as demonstrated by qRT-PCR analyses. We also purified the LcGal-3 protein that was expressed in prokaryotes, and found that it has the ability to agglutinate large yellow croaker red blood cells in a Ca2+-independent manner. The agglutination activity of LcGal-3 was inhibited by lipopolysaccharides (LPS) in a concentration-dependent manner, as shown in the sugar inhibition test. Additionally, LcGal-3 exhibited agglutination and antibacterial activities against three Gram-negative bacteria, including P. plecoglossicida, Vibrio parahaemolyticus, and Vibrio harveyi. Furthermore, we studied the agglutination mechanism of the LcGal-3 protein using blood coagulation tests with LcGal-3 deletion and point mutation proteins. Our results indicate that LcGal-3 protein plays a critical role in the innate immunity of the large yellow croaker, providing a basis for further studies on the immune mechanism and disease-resistant breeding in L. crocea and other marine fish.
Collapse
Affiliation(s)
- Yao Yang
- Key Laboratory of Healthy Mariculture for the East China Sea, Minsistry of Agriculture and Rural Affairs Fujian Provincial Key Laboratory of Marine Fishery Resources and Eco-Environment, Jimei University, Xiamen 361021, China
| | - Baolan Wu
- Key Laboratory of Healthy Mariculture for the East China Sea, Minsistry of Agriculture and Rural Affairs Fujian Provincial Key Laboratory of Marine Fishery Resources and Eco-Environment, Jimei University, Xiamen 361021, China
| | - Wanbo Li
- Key Laboratory of Healthy Mariculture for the East China Sea, Minsistry of Agriculture and Rural Affairs Fujian Provincial Key Laboratory of Marine Fishery Resources and Eco-Environment, Jimei University, Xiamen 361021, China
| | - Fang Han
- Key Laboratory of Healthy Mariculture for the East China Sea, Minsistry of Agriculture and Rural Affairs Fujian Provincial Key Laboratory of Marine Fishery Resources and Eco-Environment, Jimei University, Xiamen 361021, China
| |
Collapse
|
9
|
Ramadan A, Cao Z, Hassan M, Zetterberg F, Nilsson UJ, Gadjeva M, Rathinam V, Panjwani N. Galectin-8 Downmodulates TLR4 Activation and Impairs Bacterial Clearance in a Mouse Model of Pseudomonas aeruginosa Keratitis. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2023; 210:398-407. [PMID: 36603009 PMCID: PMC9898164 DOI: 10.4049/jimmunol.2200706] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/20/2022] [Accepted: 11/15/2022] [Indexed: 01/06/2023]
Abstract
Pseudomonas aeruginosa provokes a painful, sight-threatening corneal infection. It progresses rapidly and is difficult to treat. In this study, using a mouse model of P. aeruginosa keratitis, we demonstrate the importance of a carbohydrate-binding protein, galectin-8 (Gal-8), in regulation of the innate immune response. First, using two distinct strains of P. aeruginosa, we established that Gal-8-/- mice are resistant to P. aeruginosa keratitis. In contrast, mice deficient in Gal-1, Gal-3, and Gal-9 were fully susceptible. Second, the addition of exogenous rGal-8 to LPS (TLR4 ligand)-stimulated bone marrow-derived macrophages suppressed 1) the activation of the NF-κB pathway, and 2) formation of the MD-2/TLR4 complex. Additionally, the expression level of reactive oxygen species was substantially higher in infected Gal-8-/- bone marrow neutrophils (BMNs) compared with the Gal-8+/+ BMNs, and the P. aeruginosa killing capacity of Gal-8-/- BMNs was considerably higher compared with that of Gal-8+/+ BMNs. In the bacterial killing assays, the addition of exogenous rGal-8 almost completely rescued the phenotype of Gal-8-/- BMNs. Finally, we demonstrate that a subconjunctival injection of a Gal-8 inhibitor markedly reduces the severity of infection in the mouse model of P. aeruginosa keratitis. These data lead us to conclude that Gal-8 downmodulates the innate immune response by suppressing activation of the TLR4 pathway and clearance of P. aeruginosa by neutrophils. These findings have broad implications for developing novel therapeutic strategies for treatment of conditions resulting from the hyperactive immune response both in ocular as well as nonocular tissues.
Collapse
Affiliation(s)
- Abdulraouf Ramadan
- New England Eye Center/Department of Ophthalmology, Tufts University School of Medicine, Boston, MA. 02111, USA
| | - Zhiyi Cao
- New England Eye Center/Department of Ophthalmology, Tufts University School of Medicine, Boston, MA. 02111, USA
| | - Mujtaba Hassan
- Centre for Analysis and Synthesis, Department of Chemistry, Lund University, Lund, Sweden
| | | | - Ulf J. Nilsson
- Centre for Analysis and Synthesis, Department of Chemistry, Lund University, Lund, Sweden
| | - Mihaela Gadjeva
- Department of Medicine, Division of Infectious Diseases, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA. 02115, USA
| | - Vijay Rathinam
- Department of Immunology, UConn Health School of Medicine, Farmington, CT
| | - Noorjahan Panjwani
- New England Eye Center/Department of Ophthalmology, Tufts University School of Medicine, Boston, MA. 02111, USA
- Department of Developmental, Molecular and Chemical Biology, Tufts University School of Medicine, Boston, MA, 02111, USA
| |
Collapse
|
10
|
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
|
11
|
Extrathymic expression of Aire controls the induction of effective T H17 cell-mediated immune response to Candida albicans. Nat Immunol 2022; 23:1098-1108. [PMID: 35761088 DOI: 10.1038/s41590-022-01247-6] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2019] [Accepted: 05/18/2022] [Indexed: 01/06/2023]
Abstract
Patients with loss of function in the gene encoding the master regulator of central tolerance AIRE suffer from a devastating disorder called autoimmune polyendocrine syndrome type 1 (APS-1), characterized by a spectrum of autoimmune diseases and severe mucocutaneous candidiasis. Although the key mechanisms underlying the development of autoimmunity in patients with APS-1 are well established, the underlying cause of the increased susceptibility to Candida albicans infection remains less understood. Here, we show that Aire+MHCII+ type 3 innate lymphoid cells (ILC3s) could sense, internalize and present C. albicans and had a critical role in the induction of Candida-specific T helper 17 (TH17) cell clones. Extrathymic Rorc-Cre-mediated deletion of Aire resulted in impaired generation of Candida-specific TH17 cells and subsequent overgrowth of C. albicans in the mucosal tissues. Collectively, our observations identify a previously unrecognized regulatory mechanism for effective defense responses against fungal infections.
Collapse
|
12
|
Chen T, Wagner AS, Reynolds TB. When Is It Appropriate to Take Off the Mask? Signaling Pathways That Regulate ß(1,3)-Glucan Exposure in Candida albicans. FRONTIERS IN FUNGAL BIOLOGY 2022; 3:842501. [PMID: 36908584 PMCID: PMC10003681 DOI: 10.3389/ffunb.2022.842501] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Accepted: 01/31/2022] [Indexed: 12/21/2022]
Abstract
Candida spp. are an important source of systemic and mucosal infections in immune compromised populations. However, drug resistance or toxicity has put limits on the efficacy of current antifungals. The C. albicans cell wall is considered a good therapeutic target due to its roles in viability and fungal pathogenicity. One potential method for improving antifungal strategies could be to enhance the detection of fungal cell wall antigens by host immune cells. ß(1,3)-glucan, which is an important component of fungal cell walls, is a highly immunogenic epitope. Consequently, multiple host pattern recognition receptors, such as dectin-1, complement receptor 3 (CR3), and the ephrin type A receptor A (EphA2) are capable of recognizing exposed (unmasked) ß(1,3)-glucan moieties on the cell surface to initiate an anti-fungal immune response. However, ß(1,3)-glucan is normally covered (masked) by a layer of glycosylated proteins on the outer surface of the cell wall, hiding it from immune detection. In order to better understand possible mechanisms of unmasking ß(1,3)-glucan, we must develop a deeper comprehension of the pathways driving this phenotype. In this review, we describe the medical importance of ß(1,3)-glucan exposure in anti-fungal immunity, and highlight environmental stimuli and stressors encountered within the host that are capable of inducing changes in the levels of surface exposed ß(1,3)-glucan. Furthermore, particular focus is placed on how signal transduction cascades regulate changes in ß(1,3)-glucan exposure, as understanding the role that these pathways have in mediating this phenotype will be critical for future therapeutic development.
Collapse
Affiliation(s)
- Tian Chen
- Department of Pathogenic Biology, School of Biomedical Sciences, Shandong University, Jinan, China
| | - Andrew S. Wagner
- Department of Microbiology, University of Tennessee, Knoxville, Knoxville, TN, United States
| | - Todd B. Reynolds
- Department of Microbiology, University of Tennessee, Knoxville, Knoxville, TN, United States
| |
Collapse
|
13
|
Lujan AL, Croci DO, Rabinovich GA, Damiani MT. Galectins as potential therapeutic targets in STIs in the female genital tract. Nat Rev Urol 2022; 19:240-252. [PMID: 35105978 DOI: 10.1038/s41585-021-00562-1] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/23/2021] [Indexed: 12/12/2022]
Abstract
Every day, more than one million people worldwide acquire a sexually transmitted infection (STI). This public health problem has a direct effect on women's reproductive and sexual health as STIs can cause irreversible damage to fertility and can have negative consequences associated with discrimination and social exclusion. Infection with one sexually transmitted pathogen predisposes to co-infection with others, suggesting the existence of shared pathways that serve as molecular links between these diseases. Galectins, a family of β-galactoside-binding proteins, have emerged as endogenous mediators that facilitate cell-surface binding, internalization and cell invasion of many sexually transmitted pathogens, including Chlamydia trachomatis, Neisseria gonorrhoeae, Trichomonas vaginalis, Candida albicans, HIV and herpes simplex virus. The ability of certain galectins to dimerize or form multimeric complexes confers the capacity to interact simultaneously with glycosylated ligands on both the pathogen and the cervico-vaginal tissue on these proteins. Galectins can act as a bridge by engaging glycans from the pathogen surface and glycosylated receptors from host cells, which is a mechanism that has been shown to be shared by several sexually transmitted pathogens. In the case of viruses and obligate intracellular bacteria, binding to the cell surface promotes pathogen internalization and cell invasion. Inflammatory responses that occur in cervico-vaginal tissue might trigger secretion of galectins, which in turn control the establishment, evolution and severity of STIs. Thus, galectin-targeted therapies could potentially prevent or decrease STIs caused by a diverse array of pathogenic microorganisms; furthermore, anti-galectin agents might reduce treatment costs of STIs and reach the most vulnerable populations.
Collapse
Affiliation(s)
- Agustin L Lujan
- Laboratorio de Bioquímica e Inmunidad, Instituto de Bioquímica y Biotecnología, Facultad de Ciencias Médicas, Instituto de Medicina y Biología Experimental de Cuyo (IMBECU), Universidad Nacional de Cuyo (UNCUYO), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Mendoza, Argentina.,Centre for Genomic Regulation (CRG), The Barcelona Institute of Science and Technology, Barcelona, Spain
| | - Diego O Croci
- Laboratorio de Inmunopatología, Facultad de Ciencias Exactas y Naturales, Instituto de Histología y Embriología de Mendoza (IHEM), Universidad Nacional de Cuyo (UNCUYO), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Mendoza, Argentina
| | - Gabriel A Rabinovich
- Laboratorio de Glicomedicina, Instituto de Biología y Medicina Experimental (IBYME), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina. .,Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires (UBA), C1428AGE, Buenos Aires, Argentina.
| | - Maria T Damiani
- Laboratorio de Bioquímica e Inmunidad, Instituto de Bioquímica y Biotecnología, Facultad de Ciencias Médicas, Instituto de Medicina y Biología Experimental de Cuyo (IMBECU), Universidad Nacional de Cuyo (UNCUYO), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Mendoza, Argentina.
| |
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
|
Kamili NA, Paul A, Wu SC, Dias-Baruffi M, Cummings RD, Arthur CM, Stowell SR. Evaluation of the Bactericidal Activity of Galectins. Methods Mol Biol 2022; 2442:517-531. [PMID: 35320543 DOI: 10.1007/978-1-0716-2055-7_27] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Over a century ago, Karl Landsteiner discovered that blood group antigens could predict the immunological outcome of red blood cell transfusion. While the discovery of ABO(H) blood group antigens revolutionized transfusion medicine, many questions remain regarding the development and regulation of naturally occurring anti-blood group antibody formation. Early studies suggested that blood group antibodies develop following stimulation by bacteria that express blood group antigens. While this may explain the development of anti-blood group antibodies in blood group-negative individuals, how blood group-positive individuals protect themselves against blood group-positive microbes remained unknown. Recent studies suggest that several members of the galectin family specifically target blood group-positive microbes, thereby providing innate immune protection against blood group antigen-positive microbes regardless of the blood group status of an individual. Importantly, subsequent studies suggest that this unique form of immunity may not be limited to blood group expressing microbes, but may reflect a more generalized form of innate immunity against molecular mimicry. As this form of antimicrobial activity represents a unique and unprecedented form of immunity, we will examine important considerations and methodological approaches that can be used when seeking to ascertain the potential antimicrobial activity of various members of the galectin family.
Collapse
Affiliation(s)
- Nourine A Kamili
- Joint Program in Transfusion Medicine, Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Anu Paul
- Joint Program in Transfusion Medicine, Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Shang-Chuen Wu
- Joint Program in Transfusion Medicine, Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Marcelo Dias-Baruffi
- Department of Clinical Analysis, Toxicological and Bromatological, School of Pharmaceutical Sciences of Ribeirao Preto, University of Sao Paulo, Ribeirao Preto, Brazil
| | | | - Connie M Arthur
- Joint Program in Transfusion Medicine, Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
- Harvard Glycomics Center, Harvard Medical School, Boston, MA, USA
| | - Sean R Stowell
- Joint Program in Transfusion Medicine, Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA.
- Harvard Glycomics Center, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
16
|
Stowell SR, Dias-Baruffi M, Cummings RD, Arthur CM. Detection of Phosphatidylserine Exposure on Leukocytes Following Treatment with Human Galectins. Methods Mol Biol 2022; 2442:533-548. [PMID: 35320544 DOI: 10.1007/978-1-0716-2055-7_28] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Cellular turnover represents a fundamental aspect of immunological homeostasis. While many factors appear to regulate leukocyte removal during inflammatory resolution, recent studies suggest that members of the galectin family play a unique role in orchestrating this process. Unlike cellular removal through apoptotic cell death, several members of the galectin family induce surface expression of phosphatidylserine (PS), a phagocytic marker on cells undergoing apoptosis, in the absence of cell death. However, similar to PS on cells undergoing apoptosis, galectin-induced PS exposure sensitizes cells to phagocytic removal. As galectins appear to prepare cells for phagocytic removal without actually inducing apoptotic cell death, this process has recently been coined preaparesis. Given the unique characteristics of galectin-induced PS exposure in the context of preaparesis, we will examine unique considerations when evaluating the potential impact of different galectin family members on PS exposure and cell viability.
Collapse
Affiliation(s)
- Sean R Stowell
- Joint Program in Transfusion Medicine, Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
- Harvard Glycomics Center, Harvard Medical School, Boston, MA, USA
| | - Marcelo Dias-Baruffi
- Department of Clinical Analysis, Toxicological and Bromatological, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Ribeirão Preto, Brazil
| | | | - Connie M Arthur
- Joint Program in Transfusion Medicine, Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA.
- Harvard Glycomics Center, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
17
|
Leppänen A, Arthur CM, Stowell SR, Cummings RD. Examination of Whole-Cell Galectin Binding by Solid Phase and Flow Cytometric Analysis. Methods Mol Biol 2022; 2442:187-203. [PMID: 35320527 DOI: 10.1007/978-1-0716-2055-7_11] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
We have utilized simple flow cytometric and fluorescence-based solid phase assays to study the interaction of glycan binding proteins (GBP) to cell surface glycoconjugates. These methods utilize commonly employed flow cytometry techniques and commercially available streptavidin-coated microplates to immobilize various biotinylated ligands, such as glycopeptides, oligosaccharides, and whole cells. Using this approach, fluorescently labeled GBPs, in particular, members of the galectin family, can be interrogated for potential interactions with cell surface carbohydrates, including elucidation of the potential impact of alterations in glycosylation on carbohydrate recognition. Using these approaches, we present examples of flow cytometric and fluorescence-based solid phase assays to study galectin-carbohydrate interactions.
Collapse
Affiliation(s)
| | - Connie M Arthur
- Joint Program in Transfusion Medicine, Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
- Harvard Glycomics Center, Harvard Medical School, Boston, MA, USA
| | - Sean R Stowell
- Joint Program in Transfusion Medicine, Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
- Harvard Glycomics Center, Harvard Medical School, Boston, MA, USA
| | | |
Collapse
|
18
|
Bruneau A, Hundertmark J, Guillot A, Tacke F. Molecular and Cellular Mediators of the Gut-Liver Axis in the Progression of Liver Diseases. Front Med (Lausanne) 2021; 8:725390. [PMID: 34650994 PMCID: PMC8505679 DOI: 10.3389/fmed.2021.725390] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2021] [Accepted: 09/01/2021] [Indexed: 12/15/2022] Open
Abstract
The gut-liver axis covers the bidirectional communication between the gut and the liver, and thus includes signals from liver-to-gut (e.g., bile acids, immunoglobulins) and from gut-to-liver (e.g., nutrients, microbiota-derived products, and recirculating bile acids). In a healthy individual, liver homeostasis is tightly controlled by the mostly tolerogenic liver resident macrophages, the Kupffer cells, capturing the gut-derived antigens from the blood circulation. However, disturbances of the gut-liver axis have been associated to the progression of varying chronic liver diseases, such as non-alcoholic fatty liver disease, non-alcoholic steatohepatitis, and primary sclerosing cholangitis. Notably, changes of the gut microbiome, or intestinal dysbiosis, combined with increased intestinal permeability, leads to the translocation of gut-derived bacteria or their metabolites into the portal vein. In the context of concomitant or subsequent liver inflammation, the liver is then infiltrated by responsive immune cells (e.g., monocytes, neutrophils, lymphoid, or dendritic cells), and microbiota-derived products may provoke or exacerbate innate immune responses, hence perpetuating liver inflammation and fibrosis, and potentiating the risks of developing cirrhosis. Similarly, food derived antigens, bile acids, danger-, and pathogen-associated molecular patterns are able to reshape the liver immune microenvironment. Immune cell intracellular signaling components, such as inflammasome activation, toll-like receptor or nucleotide-binding oligomerization domain-like receptors signaling, are potent targets of interest for the modulation of the immune response. This review describes the current understanding of the cellular landscape and molecular pathways involved in the gut-liver axis and implicated in chronic liver disease progression. We also provide an overview of innovative therapeutic approaches and current clinical trials aiming at targeting the gut-liver axis for the treatment of patients with chronic liver and/or intestinal diseases.
Collapse
Affiliation(s)
- Alix Bruneau
- Department of Hepatology & Gastroenterology, Charité Universitätsmedizin Berlin, Campus Virchow-Klinikum (CVK) and Campus Charité Mitte (CCM), Berlin, Germany
| | - Jana Hundertmark
- Department of Hepatology & Gastroenterology, Charité Universitätsmedizin Berlin, Campus Virchow-Klinikum (CVK) and Campus Charité Mitte (CCM), Berlin, Germany
| | - Adrien Guillot
- Department of Hepatology & Gastroenterology, Charité Universitätsmedizin Berlin, Campus Virchow-Klinikum (CVK) and Campus Charité Mitte (CCM), Berlin, Germany
| | - Frank Tacke
- Department of Hepatology & Gastroenterology, Charité Universitätsmedizin Berlin, Campus Virchow-Klinikum (CVK) and Campus Charité Mitte (CCM), Berlin, Germany
| |
Collapse
|
19
|
Sehrawat S, Kaur M. Galectin-3 as a modifier of anti-microbial immunity: Unraveling the unknowns. Glycobiology 2021; 30:418-426. [PMID: 31985798 DOI: 10.1093/glycob/cwaa005] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2019] [Revised: 01/20/2020] [Accepted: 01/20/2020] [Indexed: 12/12/2022] Open
Abstract
Galectins play diverse roles in pathophysiology of infectious diseases and cancers. Galectin-3 is one of the most studied family member and the only chimeric type lectin. Many aspects of its biogenesis, range of activities, and the disease-modifying potential particularly during microbial infections are yet to be known. We review our current understanding of these issues and also highlight gaps in better defining the immune modulatory potential of galectin-3 during different stages of host responsiveness when an infection sets in. Additionally, we discuss commonly used strategies to disrupt galectin-3 functions both extracellulalry and intracellularly. Existing and improved novel strategies could help fine-tune immune responses to achieve better prognosis of infectious diseases.
Collapse
Affiliation(s)
- Sharvan Sehrawat
- Department of Biological Science, Indian Institute of Science Education and Research Mohali, SAS Nagar Knowledge City, PO Manauli, Mohali 140306 India
| | - Manpreet Kaur
- Department of Biological Science, Indian Institute of Science Education and Research Mohali, SAS Nagar Knowledge City, PO Manauli, Mohali 140306 India
| |
Collapse
|
20
|
Niu Y, Lin J, Li C, Peng X, Jiang N, Xu Q, Yin M, Lin H, Gu L, Zhao G. Galectin-3 plays an important pro-inflammatory role in A. fumigatus keratitis by recruiting neutrophils and activating p38 in neutrophils. Int Immunopharmacol 2021; 97:107706. [PMID: 33933850 DOI: 10.1016/j.intimp.2021.107706] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2021] [Revised: 04/15/2021] [Accepted: 04/19/2021] [Indexed: 11/18/2022]
Abstract
PURPOSE To determine the role of galectin-3 (Gal-3) in cornea infected by Aspergillus fumigatus (A. fumigatus). METHODS Gal-3 was tested in normal and infected corneas of C57BL/6 mice. Mice corneas were pretreated with or without rmGal-3 or Gal-3 siRNA and infected with A. fumigatus. Recombinant mouse (rm) Gal-3 stimulated polymorphonuclear neutrophilic leukocytes (PMNs). PMNs were stimulated with 75% ethanol-killed A. fumigatus with or without pretreatment of Gal-3 siRNA. Disease severity was documented by clinical score and photographs with a slit lamp. PCR, Western blot, and ELISA tested expression of Gal-3, interleukin (IL)-1β, IL-6, macrophage inflammatory protein 2 (MIP-2) and p-p38. PMNs infiltration was assessed by flow cytometry and myeloperoxidase (MPO) assay. RESULTS Gal-3 expression was significantly elevated by A. fumigatus in mice corneas. rmGal-3 treatment increased clinical scores, PMNs infiltration, and cytokines expression, which were decreased by Gal-3 siRNA treatment. In PMNs, Gal-3 expression was also significantly increased by A. fumigatus. The rmGal-3 treatment upregulated proinflammatory cytokines secretion and p-p38 expression, which was significantly inhibited by Gal-3 siRNA. CONCLUSION These data proved that A. fumigatus increased Gal-3 expression and elevated disease clinical scores, PMNs infiltration and cytokines expression through Gal-3. In PMNs, A. fumigatus upregulated IL-1β and IL-6 secretion through the Gal-3 / p38 pathway.
Collapse
Affiliation(s)
- Yawen Niu
- Department of Ophthalmology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong Province, China
| | - Jing Lin
- Department of Ophthalmology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong Province, China
| | - Cui Li
- Department of Ophthalmology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong Province, China
| | - Xudong Peng
- Department of Ophthalmology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong Province, China
| | - Nan Jiang
- Department of Ophthalmology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong Province, China
| | - Qiang Xu
- Department of Ophthalmology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong Province, China
| | - Min Yin
- Department of Ophthalmology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong Province, China
| | - Hao Lin
- Department of Ophthalmology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong Province, China
| | - Lingwen Gu
- Department of Ophthalmology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong Province, China
| | - Guiqiu Zhao
- Department of Ophthalmology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong Province, China.
| |
Collapse
|
21
|
Exploration of Galectin Ligands Displayed on Gram-Negative Respiratory Bacterial Pathogens with Different Cell Surface Architectures. Biomolecules 2021; 11:biom11040595. [PMID: 33919637 PMCID: PMC8074145 DOI: 10.3390/biom11040595] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2021] [Revised: 04/13/2021] [Accepted: 04/15/2021] [Indexed: 12/29/2022] Open
Abstract
Galectins bind various pathogens through recognition of distinct carbohydrate structures. In this work, we examined the binding of four human galectins to the Gram-negative bacteria Klebsiella pneumoniae (Kpn) and non-typeable Haemophilus influenzae (NTHi), which display different surface glycans. In particular, Kpn cells are covered by a polysaccharide capsule and display an O-chain-containing lipopolysaccharide (LPS), whereas NTHi is not capsulated and its LPS, termed lipooligosacccharide (LOS), does not contain O-chain. Binding assays to microarray-printed bacteria revealed that galectins-3, -4, and -8, but not galectin-1, bind to Kpn and NTHi cells, and confocal microscopy attested binding to bacterial cells in suspension. The three galectins bound to array-printed Kpn LPS. Moreover, analysis of galectin binding to mutant Kpn cells evidenced that the O-chain is the docking point for galectins on wild type Kpn. Galectins-3, -4, and -8 also bound the NTHi LOS. Microarray-assisted comparison of the binding to full-length and truncated LOSs, as well as to wild type and mutant cells, supported LOS involvement in galectin binding to NTHi. However, deletion of the entire LOS oligosaccharide chain actually increased binding to NTHi cells, indicating the availability of other ligands on the bacterial surface, as similarly inferred for Kpn cells devoid of both O-chain and capsule. Altogether, the results illustrate galectins’ versatility for recognizing different bacterial structures, and point out the occurrence of so far overlooked galectin ligands on bacterial surfaces.
Collapse
|
22
|
Rezende CP, Martins Oliveira Brito PK, Pessoni AM, Da Silva TA, Goldman GH, Almeida F. Altered expression of genes related to innate antifungal immunity in the absence of galectin-3. Virulence 2021; 12:981-988. [PMID: 33779504 PMCID: PMC8009118 DOI: 10.1080/21505594.2021.1903212] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Galectin-3 (Gal-3) is the most studied member of the animal galectin family, which comprises β-galactoside-binding lectins and participates in several cellular events. Its expression in cells involved in innate and adaptive immunity is related to anti- and proinflammatory functions, signaling an important role in inflammatory, infectious, and tumorigenesis processes. Mice deficient in Gal-3 exhibit important phenotypes, but it is unclear whether these phenotypes reflect an impairment of the functions of this protein. Gal-3 plays an important role in modulating the immune response to different pathogenic microorganisms. However, the role of Gal-3 in immunity to infection is still poorly understood. Therefore, we investigated the effects of Gal-3 deletion on the expression of genes involved in the innate immune response in the lungs, spleens, and brains of Gal-3 KO mice. Gene profiling expression analysis suggested that Gal-3 deletion resulted in differentially modulated expression of the genes encoding beta-glucan, mannose and chitin-responsive pattern recognition receptors, signal transduction, inflammation, and phagocytosis. Our data thus suggest the importance of Gal-3 expression in the host innate immune system.
Collapse
Affiliation(s)
- Caroline Patini Rezende
- Department of Biochemistry and Immunology, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, SP, Brazil
| | | | - Andre Moreira Pessoni
- Department of Biochemistry and Immunology, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, SP, Brazil
| | - Thiago Aparecido Da Silva
- Department of Cellular and Molecular Biology, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, SP, Brazil
| | - Gustavo H Goldman
- Departamento De Ciencias Farmaceuticas, Faculdade De Ciencias Farmaceuticas De Ribeirao Preto, Universidade De Sao Paulo, Ribeirao Preto, SP, Brazil
| | - Fausto Almeida
- Department of Biochemistry and Immunology, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, SP, Brazil
| |
Collapse
|
23
|
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
|
24
|
Srejovic IM, Lukic ML. Galectin-3 in T cell-mediated immunopathology and autoimmunity. Immunol Lett 2021; 233:57-67. [PMID: 33753135 DOI: 10.1016/j.imlet.2021.03.009] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2021] [Accepted: 03/17/2021] [Indexed: 01/05/2023]
Abstract
Galectin-3 (Gal-3) is the only member of galectin family able to form pentamers and heterodimers with chemokines. Its presence in various cells and tissues suggests variety of regulatory functions in physiological conditions, but increasing body of evidence indicates involvement of Gal-3 in pathological cascades of many diseases. Gal-3 exerts different, sometimes opposite, effects in various disorders or in different phases of the same disease. These differences in action of Gal-3 are related to the localization of Gal-3 in the cell, types of receptors through which it acts, or the types of cells that secrete it. As a regulator of immune response and T-cell activity, Gal-3 appears to have important role in development of autoimmunity mediated by T cells. Absence of Gal-3 in C57Bl6 mice favors Th2 mediated inflammatory myocarditis but attenuate fibrosis. Recent data also indicate Gal-3 involvement in development atherosclerosis. In pathogenesis of diabetes type 1 and autoimmune components of diabetes type 2 Gal-3 may have detrimental or protective role depending on its intracellular or extracellular localization. Gal-3 mediates autoimmune hepatic damage through activation of T-cells or natural killer T cells. Gal-3 is an important mediator in neurodevelopment, neuropathology and behavior due to its expression both in neurons and glial cells. All together, assessing the role of Gal-3 in immunopathology and autoimmunity it could be concluded that it is an important participant in pathogenesis, as well as promising monitoring marker and therapeutic target.
Collapse
Affiliation(s)
- Ivan M Srejovic
- University of Kragujevac, Faculty of Medical Sciences, Department of Physiology, Svetozara Markovica 69, 34000, Kragujevac, Serbia.
| | - Miodrag L Lukic
- University of Kragujevac, Faculty of Medical Sciences, Department of Physiology, Svetozara Markovica 69, 34000, Kragujevac, Serbia; University of Kragujevac, Faculty of Medical Sciences, Center for Molecular Medicine and Stem Cell Research, Svetozara Markovica 69, 34000, Kragujevac, Serbia.
| |
Collapse
|
25
|
Wu X, Xia Y, He F, Zhu C, Ren W. Intestinal mycobiota in health and diseases: from a disrupted equilibrium to clinical opportunities. MICROBIOME 2021; 9:60. [PMID: 33715629 PMCID: PMC7958491 DOI: 10.1186/s40168-021-01024-x] [Citation(s) in RCA: 71] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/13/2020] [Accepted: 02/04/2021] [Indexed: 05/08/2023]
Abstract
Bacteria, viruses, protozoa, and fungi establish a complex ecosystem in the gut. Like other microbiota, gut mycobiota plays an indispensable role in modulating intestinal physiology. Notably, the most striking characteristics of intestinal fungi are their extraintestinal functions. Here, we provide a comprehensive review of the importance of gut fungi in the regulation of intestinal, pulmonary, hepatic, renal, pancreatic, and brain functions, and we present possible opportunities for the application of gut mycobiota to alleviate/treat human diseases. Video Abstract.
Collapse
Affiliation(s)
- Xiaoyan Wu
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-Bioresources, Guangdong Laboratory of Lingnan Modern Agriculture, National Engineering Research Center for Breeding Swine Industry, Guangdong Provincial Key Laboratory of Animal Nutrition Control, College of Animal Science, South China Agricultural University, Guangzhou, 510642 China
| | - Yaoyao Xia
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-Bioresources, Guangdong Laboratory of Lingnan Modern Agriculture, National Engineering Research Center for Breeding Swine Industry, Guangdong Provincial Key Laboratory of Animal Nutrition Control, College of Animal Science, South China Agricultural University, Guangzhou, 510642 China
| | - Fang He
- College of Animal Science and Technology, Southwest University, Chongqing, 400716 China
| | - Congrui Zhu
- College of Veterinary Medicine, Kansas State University, Manhattan, KS USA
| | - Wenkai Ren
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-Bioresources, Guangdong Laboratory of Lingnan Modern Agriculture, National Engineering Research Center for Breeding Swine Industry, Guangdong Provincial Key Laboratory of Animal Nutrition Control, College of Animal Science, South China Agricultural University, Guangzhou, 510642 China
| |
Collapse
|
26
|
Zhang T, Jiang S, Sun L. A Fish Galectin-8 Possesses Direct Bactericidal Activity. Int J Mol Sci 2020; 22:ijms22010376. [PMID: 33396490 PMCID: PMC7796122 DOI: 10.3390/ijms22010376] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2020] [Revised: 12/26/2020] [Accepted: 12/28/2020] [Indexed: 01/02/2023] Open
Abstract
Galectins are a family of animal lectins with high affinity for β-galactosides. Galectins are able to bind to bacteria, and a few mammalian galectins are known to kill the bound bacteria. In fish, no galectins with direct bactericidal effect have been reported. In the present study, we identified and characterized a tandem repeat galectin-8 from tongue sole Cynoglossus semilaevis (designated CsGal-8). CsGal-8 possesses conserved carbohydrate recognition domains (CRDs), as well as the conserved HXNPR and WGXEE motifs that are critical for carbohydrate binding. CsGal-8 was constitutively expressed in nine tissues of tongue sole and up-regulated in kidney, spleen, and blood by bacterial challenge. When expressed in HeLa cells, CsGal-8 protein was detected both in the cytoplasm and in the micro-vesicles secreted from the cells. Recombinant CsGal-8 (rCsGal-8) bound to lactose and other carbohydrates in a dose dependent manner. rCsGal-8 bound to a wide range of gram-positive and gram-negative bacteria and was co-localized with the bound bacteria in animal cells. Lactose, fructose, galactose, and trehalose effectively blocked the interactions between rCsGal-8 and different bacteria. Furthermore, rCsGal-8 exerted potent bactericidal activity against some gram-negative bacterial pathogens by directly damaging the membrane and structure of the pathogens. Taken together, these results indicate that CsGal-8 likely plays an important role in the immune defense against some bacterial pathogens by direct bacterial interaction and killing.
Collapse
Affiliation(s)
- Tengfei Zhang
- CAS Key Laboratory of Experimental Marine Biology, CAS Center for Ocean Mega-Science, Institute of Oceanology, Chinese Academy of Sciences, 7 Nanhai Road, Qingdao 266071, China;
- Laboratory for Marine Biology and Biotechnology, Qingdao National Laboratory for Marine Science and Technology, 1 Wenhai Road, Qingdao 266237, China
- University of Chinese Academy of Sciences, 19 Yuquan Road, Beijing 100049, China
| | - Shuai Jiang
- CAS Key Laboratory of Experimental Marine Biology, CAS Center for Ocean Mega-Science, Institute of Oceanology, Chinese Academy of Sciences, 7 Nanhai Road, Qingdao 266071, China;
- Laboratory for Marine Biology and Biotechnology, Qingdao National Laboratory for Marine Science and Technology, 1 Wenhai Road, Qingdao 266237, China
- University of Chinese Academy of Sciences, 19 Yuquan Road, Beijing 100049, China
- Correspondence: (S.J.); (L.S.); Tel.: +86-532-8289-1027 (S.J.); +86-532-8289-8829 (L.S.)
| | - Li Sun
- CAS Key Laboratory of Experimental Marine Biology, CAS Center for Ocean Mega-Science, Institute of Oceanology, Chinese Academy of Sciences, 7 Nanhai Road, Qingdao 266071, China;
- Laboratory for Marine Biology and Biotechnology, Qingdao National Laboratory for Marine Science and Technology, 1 Wenhai Road, Qingdao 266237, China
- University of Chinese Academy of Sciences, 19 Yuquan Road, Beijing 100049, China
- Correspondence: (S.J.); (L.S.); Tel.: +86-532-8289-1027 (S.J.); +86-532-8289-8829 (L.S.)
| |
Collapse
|
27
|
Bashiri S, Koirala P, Toth I, Skwarczynski M. Carbohydrate Immune Adjuvants in Subunit Vaccines. Pharmaceutics 2020; 12:E965. [PMID: 33066594 PMCID: PMC7602499 DOI: 10.3390/pharmaceutics12100965] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2020] [Revised: 10/08/2020] [Accepted: 10/12/2020] [Indexed: 12/17/2022] Open
Abstract
Modern subunit vaccines are composed of antigens and a delivery system and/or adjuvant (immune stimulator) that triggers the desired immune responses. Adjuvants mimic pathogen-associated molecular patterns (PAMPs) that are typically associated with infections. Carbohydrates displayed on the surface of pathogens are often recognized as PAMPs by receptors on antigen-presenting cells (APCs). Consequently, carbohydrates and their analogues have been used as adjuvants and delivery systems to promote antigen transport to APCs. Carbohydrates are biocompatible, usually nontoxic, biodegradable, and some are mucoadhesive. As such, carbohydrates and their derivatives have been intensively explored for the development of new adjuvants. This review assesses the immunological functions of carbohydrate ligands and their ability to enhance systemic and mucosal immune responses against co-administered antigens. The role of carbohydrate-based adjuvants/delivery systems in the development of subunit vaccines is discussed in detail.
Collapse
Affiliation(s)
- Sahra Bashiri
- School of Chemistry and Molecular Bioscience, The University of Queensland, St Lucia, Brisbane, Queensland 4072, Australia; (S.B.); (P.K.)
| | - Prashamsa Koirala
- School of Chemistry and Molecular Bioscience, The University of Queensland, St Lucia, Brisbane, Queensland 4072, Australia; (S.B.); (P.K.)
| | - Istvan Toth
- School of Chemistry and Molecular Bioscience, The University of Queensland, St Lucia, Brisbane, Queensland 4072, Australia; (S.B.); (P.K.)
- Institute for Molecular Bioscience, The University of Queensland, St Lucia, Brisbane, Queensland 4072, Australia
- School of Pharmacy, The University of Queensland, St Lucia, Brisbane, Queensland 4072, Australia
| | - Mariusz Skwarczynski
- School of Chemistry and Molecular Bioscience, The University of Queensland, St Lucia, Brisbane, Queensland 4072, Australia; (S.B.); (P.K.)
| |
Collapse
|
28
|
Chikina AS, Nadalin F, Maurin M, San-Roman M, Thomas-Bonafos T, Li XV, Lameiras S, Baulande S, Henri S, Malissen B, Lacerda Mariano L, Barbazan J, Blander JM, Iliev ID, Matic Vignjevic D, Lennon-Duménil AM. Macrophages Maintain Epithelium Integrity by Limiting Fungal Product Absorption. Cell 2020; 183:411-428.e16. [PMID: 32970988 PMCID: PMC7646275 DOI: 10.1016/j.cell.2020.08.048] [Citation(s) in RCA: 74] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2020] [Revised: 07/27/2020] [Accepted: 08/26/2020] [Indexed: 12/14/2022]
Abstract
The colon is primarily responsible for absorbing fluids. It contains a large number of microorganisms including fungi, which are enriched in its distal segment. The colonic mucosa must therefore tightly regulate fluid influx to control absorption of fungal metabolites, which can be toxic to epithelial cells and lead to barrier dysfunction. How this is achieved remains unknown. Here, we describe a mechanism by which the innate immune system allows rapid quality check of absorbed fluids to avoid intoxication of colonocytes. This mechanism relies on a population of distal colon macrophages that are equipped with "balloon-like" protrusions (BLPs) inserted in the epithelium, which sample absorbed fluids. In the absence of macrophages or BLPs, epithelial cells keep absorbing fluids containing fungal products, leading to their death and subsequent loss of epithelial barrier integrity. These results reveal an unexpected and essential role of macrophages in the maintenance of colon-microbiota interactions in homeostasis. VIDEO ABSTRACT.
Collapse
Affiliation(s)
- Aleksandra S Chikina
- Institut Curie, PSL Research University, CNRS UMR 144, F-75005 Paris, France; Institut Curie, PSL Research University, INSERM U932, F-75005 Paris, France
| | - Francesca Nadalin
- Institut Curie, PSL Research University, INSERM U932, F-75005 Paris, France
| | - Mathieu Maurin
- Institut Curie, PSL Research University, INSERM U932, F-75005 Paris, France
| | - Mabel San-Roman
- Institut Curie, PSL Research University, INSERM U932, F-75005 Paris, France
| | | | - Xin V Li
- Gastroenterology and Hepatology Division, Joan and Sanford I. Weill Department of Medicine, Weill Cornell Medicine, Cornell University, New York, NY 10021, USA; The Jill Roberts Institute for Research in Inflammatory Bowel Disease, Weill Cornell Medicine, Cornell University, New York, NY 10021, USA
| | - Sonia Lameiras
- Institut Curie, PSL Research University, Next Generation Sequencing Facility, F-75005 Paris, France
| | - Sylvain Baulande
- Institut Curie, PSL Research University, Next Generation Sequencing Facility, F-75005 Paris, France
| | - Sandrine Henri
- Centre d'Immunologie de Marseille-Luminy, Aix Marseille Université, INSERM, CNRS, 13288 Marseille, France
| | - Bernard Malissen
- Centre d'Immunologie de Marseille-Luminy, Aix Marseille Université, INSERM, CNRS, 13288 Marseille, France; Centre d'Immunophénomique, Aix Marseille Université, INSERM, CNRS, 13288 Marseille, France
| | | | - Jorge Barbazan
- Institut Curie, PSL Research University, CNRS UMR 144, F-75005 Paris, France
| | - J Magarian Blander
- The Jill Roberts Institute for Research in Inflammatory Bowel Disease, Weill Cornell Medicine, Cornell University, New York, NY 10021, USA
| | - Iliyan D Iliev
- Gastroenterology and Hepatology Division, Joan and Sanford I. Weill Department of Medicine, Weill Cornell Medicine, Cornell University, New York, NY 10021, USA; The Jill Roberts Institute for Research in Inflammatory Bowel Disease, Weill Cornell Medicine, Cornell University, New York, NY 10021, USA
| | | | | |
Collapse
|
29
|
Vasta GR, Wang JX. Galectin-mediated immune recognition: Opsonic roles with contrasting outcomes in selected shrimp and bivalve mollusk species. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2020; 110:103721. [PMID: 32353466 DOI: 10.1016/j.dci.2020.103721] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/06/2020] [Revised: 04/22/2020] [Accepted: 04/22/2020] [Indexed: 06/11/2023]
Abstract
Galectins are a structurally conserved family of ß-galactoside-binding lectins characterized by a unique sequence motif in the carbohydrate recognition domain, and of wide taxonomic distribution, from fungi to mammals. Their biological functions, initially described as key to embryogenesis and early development via recognition of endogenous ("self") carbohydrate moieties, are currently understood as also encompassing tissue repair, cancer metastasis, angiogenesis, adipogenesis, and regulation of immune homeostasis. More recently, however, numerous studies have contributed to establish a new paradigm by revealing that galectins can also bind to exogenous ("non-self") glycans on the surface of potentially pathogenic virus, bacteria, and eukaryotic parasites, and function both as pathogen recognition receptors (PRRs) and effector factors in innate immunity. Our studies on a galectin from the kuruma shrimp Marsupenaeus japonicus (MjGal), revealed that it functions as a typical PRR. Expression of MjGal is upregulated by infectious challenge, and can recognize both Gram (+) and Gram (-) bacteria. MjGal also recognizes carbohydrates on the shrimp hemocyte surface, and can cross-link microbial pathogens to the hemocytes, promoting their phagocytosis and clearance from circulation. Therefore, MjGal contributes to the shrimp's immune defense against infectious challenge both as a PRR and effector factor. Our studies on galectins from the bivalve mollusks, however, have shown that although they can function in immune defense as MjGal, protistan parasites take advantage of the recognition roles of the host galectins, for successful attachment and host infection. We identified in the eastern oyster Crassostrea virginica two galectins (CvGal1 and CvGal2) that not only recognize a large variety of bacterial species, but also the protozoan parasite Perkinsus marinus. Like the shrimp MjGal, both oyster galectins function as opsonins, and promote parasite adhesion and phagocytosis. However, P. marinus survives intrahemocytic oxidative killing and proliferates, eventually causing systemic infection and death of the oyster host. In the softshell clam Mya arenaria we identified a galectin (MaGal1) that displays carbohydrate specificity and recognition properties for sympatric Perkinsus species (P. marinus and P. chesapeaki), that are different from CvGal1 and CvGal2. Our results suggest that although galectins from bivalves can function as PRRs, Perkinsus parasites have co-evolved with their hosts to subvert the galectins' immune functions for host infection by acquisition of carbohydrate-based mimicry.
Collapse
Affiliation(s)
- Gerardo R Vasta
- Department of Microbiology and Immunology, School of Medicine, University of Maryland Baltimore, Institute of Marine and Environmental Technology, Baltimore, MD, USA.
| | - Jin-Xing Wang
- Shandong Provincial Key Laboratory of Animal Cells and Developmental Biology, School of Life Sciences, Shandong University, Qingdao, Shandong, China
| |
Collapse
|
30
|
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
|
31
|
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
|
32
|
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
|
33
|
Walker LA, Munro CA. Caspofungin Induced Cell Wall Changes of Candida Species Influences Macrophage Interactions. Front Cell Infect Microbiol 2020; 10:164. [PMID: 32528900 PMCID: PMC7247809 DOI: 10.3389/fcimb.2020.00164] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2019] [Accepted: 03/27/2020] [Indexed: 11/13/2022] Open
Abstract
Candida species are known to differ in their ability to cause infection and have been shown to display varied susceptibilities to antifungal drugs. Treatment with the echinocandin, caspofungin, leads to compensatory alterations in the fungal cell wall. This study was performed to compare the structure and composition of the cell walls of different Candida species alone and in response to caspofungin treatment, and to evaluate how changes at the fungal cell surface affects interactions with macrophages. We demonstrated that the length of the outer fibrillar layer varied between Candida species and that, in most cases, reduced fibril length correlated with increased exposure of β-1,3-glucan on the cell surface. Candida glabrata and Candida guilliermondii, which had naturally more β-1,3-glucan exposed on the cell surface, were phagocytosed significantly more efficiently by J774 macrophages. Treatment with caspofungin resulted in increased exposure of chitin and β-1,3-glucan on the surface of the majority of Candida species isolates that were tested, with the exception of C. glabrata and Candida parapsilosis isolates. This increase in exposure of the inner cell wall polysaccharides, in most cases, correlated with reduced uptake by macrophages and in turn, a decrease in production of TNFα. Here we show that differences in the exposure of cell wall carbohydrates and variations in the repertoire of covalently attached surface proteins of different Candida species contributes to their recognition by immune cells.
Collapse
Affiliation(s)
- Louise A Walker
- School of Medicine, Medical Sciences and Nutrition, Institute of Medical Sciences, University of Aberdeen, Aberdeen, United Kingdom
| | - Carol A Munro
- School of Medicine, Medical Sciences and Nutrition, Institute of Medical Sciences, University of Aberdeen, Aberdeen, United Kingdom
| |
Collapse
|
34
|
Vendele I, Willment JA, Silva LM, Palma AS, Chai W, Liu Y, Feizi T, Spyrou M, Stappers MHT, Brown GD, Gow NAR. Mannan detecting C-type lectin receptor probes recognise immune epitopes with diverse chemical, spatial and phylogenetic heterogeneity in fungal cell walls. PLoS Pathog 2020; 16:e1007927. [PMID: 31999794 PMCID: PMC7012452 DOI: 10.1371/journal.ppat.1007927] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2019] [Revised: 02/11/2020] [Accepted: 12/22/2019] [Indexed: 01/09/2023] Open
Abstract
During the course of fungal infection, pathogen recognition by the innate immune system is critical to initiate efficient protective immune responses. The primary event that triggers immune responses is the binding of Pattern Recognition Receptors (PRRs), which are expressed at the surface of host immune cells, to Pathogen-Associated Molecular Patterns (PAMPs) located predominantly in the fungal cell wall. Most fungi have mannosylated PAMPs in their cell walls and these are recognized by a range of C-type lectin receptors (CTLs). However, the precise spatial distribution of the ligands that induce immune responses within the cell walls of fungi are not well defined. We used recombinant IgG Fc-CTLs fusions of three murine mannan detecting CTLs, including dectin-2, the mannose receptor (MR) carbohydrate recognition domains (CRDs) 4-7 (CRD4-7), and human DC-SIGN (hDC-SIGN) and of the β-1,3 glucan-binding lectin dectin-1 to map PRR ligands in the fungal cell wall of fungi grown in vitro in rich and minimal media. We show that epitopes of mannan-specific CTL receptors can be clustered or diffuse, superficial or buried in the inner cell wall. We demonstrate that PRR ligands do not correlate well with phylogenetic relationships between fungi, and that Fc-lectin binding discriminated between mannosides expressed on different cell morphologies of the same fungus. We also demonstrate CTL epitope differentiation during different phases of the growth cycle of Candida albicans and that MR and DC-SIGN labelled outer chain N-mannans whilst dectin-2 labelled core N-mannans displayed deeper in the cell wall. These immune receptor maps of fungal walls of in vitro grown cells therefore reveal remarkable spatial, temporal and chemical diversity, indicating that the triggering of immune recognition events originates from multiple physical origins at the fungal cell surface.
Collapse
Affiliation(s)
- Ingrida Vendele
- MRC Centre for Medical Mycology, Aberdeen Fungal Group, College of Life Sciences and Medicine, Institute of Medical Sciences, University of Aberdeen, Foresterhill, Aberdeen, United Kingdom
- Division of Infection and Immunity, The Roslin Institute and Royal (Dick) School of Veterinary Studies, University of Edinburgh, Edinburgh, United Kingdom
| | - Janet A. Willment
- MRC Centre for Medical Mycology, Aberdeen Fungal Group, College of Life Sciences and Medicine, Institute of Medical Sciences, University of Aberdeen, Foresterhill, Aberdeen, United Kingdom
- School of Biosciences, University of Exeter, Geoffrey Pope Building, Exeter, United Kingdom
| | - Lisete M. Silva
- Glycosciences Laboratory, Department of Metabolism, Digestion and Reproduction, Imperial College London, London, United Kingdom
| | - Angelina S. Palma
- Glycosciences Laboratory, Department of Metabolism, Digestion and Reproduction, Imperial College London, London, United Kingdom
- UCIBIO, Department of Chemistry, Faculty of Science and Technology, NOVA University of Lisbon, Lisbon, Portugal
| | - Wengang Chai
- Glycosciences Laboratory, Department of Metabolism, Digestion and Reproduction, Imperial College London, London, United Kingdom
| | - Yan Liu
- Glycosciences Laboratory, Department of Metabolism, Digestion and Reproduction, Imperial College London, London, United Kingdom
| | - Ten Feizi
- Glycosciences Laboratory, Department of Metabolism, Digestion and Reproduction, Imperial College London, London, United Kingdom
| | - Maria Spyrou
- MRC Centre for Medical Mycology, Aberdeen Fungal Group, College of Life Sciences and Medicine, Institute of Medical Sciences, University of Aberdeen, Foresterhill, Aberdeen, United Kingdom
- School of Biosciences, University of Exeter, Geoffrey Pope Building, Exeter, United Kingdom
| | - Mark H. T. Stappers
- MRC Centre for Medical Mycology, Aberdeen Fungal Group, College of Life Sciences and Medicine, Institute of Medical Sciences, University of Aberdeen, Foresterhill, Aberdeen, United Kingdom
- School of Biosciences, University of Exeter, Geoffrey Pope Building, Exeter, United Kingdom
| | - Gordon D. Brown
- MRC Centre for Medical Mycology, Aberdeen Fungal Group, College of Life Sciences and Medicine, Institute of Medical Sciences, University of Aberdeen, Foresterhill, Aberdeen, United Kingdom
- School of Biosciences, University of Exeter, Geoffrey Pope Building, Exeter, United Kingdom
| | - Neil A. R. Gow
- MRC Centre for Medical Mycology, Aberdeen Fungal Group, College of Life Sciences and Medicine, Institute of Medical Sciences, University of Aberdeen, Foresterhill, Aberdeen, United Kingdom
- School of Biosciences, University of Exeter, Geoffrey Pope Building, Exeter, United Kingdom
| |
Collapse
|
35
|
Galectins in Host-Pathogen Interactions: Structural, Functional and Evolutionary Aspects. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1204:169-196. [PMID: 32152947 DOI: 10.1007/978-981-15-1580-4_7] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Galectins are a family of ß-galactoside-binding lectins characterized by a unique sequence motif in the carbohydrate recognition domain, and evolutionary and structural conservation from fungi to invertebrates and vertebrates, including mammals. Their biological roles, initially understood as limited to recognition of endogenous ("self") carbohydrate ligands in embryogenesis and early development, dramatically expanded in later years by the discovery of their roles in tissue repair, cancer, adipogenesis, and regulation of immune homeostasis. In recent years, however, evidence has also accumulated to support the notion that galectins can bind ("non-self") glycans on the surface of potentially pathogenic microbes, and function as recognition and effector factors in innate immunity. Thus, this evidence has established a new paradigm by which galectins can function not only as pattern recognition receptors but also as effector factors, by binding to the microbial surface and inhibiting adhesion and/or entry into the host cell, directly killing the potential pathogen by disrupting its surface structures, or by promoting phagocytosis, encapsulation, autophagy, and pathogen clearance from circulation. Strikingly, some viruses, bacteria, and protistan parasites take advantage of the aforementioned recognition roles of the vector/host galectins, for successful attachment and invasion. These recent findings suggest that galectin-mediated innate immune recognition and effector mechanisms, which throughout evolution have remained effective for preventing or fighting viral, bacterial, and parasitic infection, have been "subverted" by certain pathogens by unique evolutionary adaptations of their surface glycome to gain host entry, and the acquisition of effective mechanisms to evade the host's immune responses.
Collapse
|
36
|
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
|
37
|
Pineda M, Corvo L, Callejas-Hernández F, Fresno M, Bonay P. Trypanosoma cruzi cleaves galectin-3 N-terminal domain to suppress its innate microbicidal activity. Clin Exp Immunol 2019; 199:216-229. [PMID: 31593356 DOI: 10.1111/cei.13379] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/03/2019] [Indexed: 12/01/2022] Open
Abstract
Galectin-3 is the best-characterized member of galectins, an evolutionary conserved family of galactoside-binding proteins that play central roles in infection and immunity, regulating inflammation, cell migration and cell apoptosis. Differentially expressed by cells and tissues with immune privilege, they bind not only to host ligands, but also to glycans expressed by pathogens. In this regard, we have previously shown that human galectin-3 recognizes several genetic lineages of the protozoan parasite Trypanosoma cruzi, the causal agent of Chagas' disease or American trypanosomiasis. Herein we describe a molecular mechanism developed by T. cruzi to proteolytically process galectin-3 that generates a truncated form of the protein lacking its N-terminal domain - required for protein oligomerization - but still conserves a functional carbohydrate recognition domain (CRD). Such processing relies on specific T. cruzi proteases, including Zn-metalloproteases and collagenases, and ultimately conveys profound changes in galectin-3-dependent effects, as chemical inhibition of parasite proteases allows galectin-3 to induce parasite death in vitro. Thus, T. cruzi might have established distinct mechanisms to counteract galectin-3-mediated immunity and microbicide properties. Interestingly, non-pathogenic T. rangeli lacked the ability to cleave galectin-3, suggesting that during evolution two genetically similar organisms have developed different molecular mechanisms that, in the case of T. cruzi, favoured its pathogenicity, highlighting the importance of T. cruzi proteases to avoid immune mechanisms triggered by galectin-3 upon infection. This study provides the first evidence of a novel strategy developed by T. cruzi to abrogate signalling mechanisms associated with galectin-3-dependent innate immunity.
Collapse
Affiliation(s)
- M Pineda
- Centro de Biología Molecular 'Severo Ochoa', Universidad Autónoma de Madrid, Nicolás Cabrera 1, Madrid, Spain
| | - L Corvo
- Centro de Biología Molecular 'Severo Ochoa', Universidad Autónoma de Madrid, Nicolás Cabrera 1, Madrid, Spain
| | - F Callejas-Hernández
- Centro de Biología Molecular 'Severo Ochoa', Universidad Autónoma de Madrid, Nicolás Cabrera 1, Madrid, Spain
| | - M Fresno
- Centro de Biología Molecular 'Severo Ochoa', Universidad Autónoma de Madrid, Nicolás Cabrera 1, Madrid, Spain
| | - P Bonay
- Centro de Biología Molecular 'Severo Ochoa', Universidad Autónoma de Madrid, Nicolás Cabrera 1, Madrid, Spain
| |
Collapse
|
38
|
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
|
39
|
Ricci CA, Kamal AHM, Chakrabarty JK, Fuess LE, Mann WT, Jinks LR, Brinkhuis V, Chowdhury SM, Mydlarz LD. Proteomic Investigation of a Diseased Gorgonian Coral Indicates Disruption of Essential Cell Function and Investment in Inflammatory and Other Immune Processes. Integr Comp Biol 2019; 59:830-844. [DOI: 10.1093/icb/icz107] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Abstract
As scleractinian coral cover declines in the face of increased frequency in disease outbreaks, future reefs may become dominated by octocorals. Understanding octocoral disease responses and consequences is therefore necessary if we are to gain insight into the future of ecosystem services provided by coral reefs. In Florida, populations of the octocoral Eunicea calyculata infected with Eunicea black disease (EBD) were observed in the field in the fall of 2011. This disease was recognized by a stark, black pigmentation caused by heavy melanization. Histological preparations of E. calyculata infected with EBD demonstrated granular amoebocyte (GA) mobilization, melanin granules in much of the GA population, and the presence of fungal hyphae penetrating coral tissue. Previous transcriptomic analysis also identified immune trade-offs evidenced by increased immune investment at the expense of growth. Our investigation utilized proteogenomic techniques to reveal decreased investment in general cell signaling while increasing energy production for immune responses. Inflammation was also prominent in diseased E. calyculata and sheds light on factors driving the extreme phenotype observed with EBD. With disease outbreaks continuing to increase in frequency, our results highlight new targets within the cnidarian immune system and provide a framework for understanding transcriptomics in the context of an organismal disease phenotype and its protein expression.
Collapse
Affiliation(s)
- Contessa A Ricci
- Department of Biology, University of Texas at Arlington, Arlington, 501 S Nedderman Dr., TX 76010, USA
| | - Abu Hena Mostafa Kamal
- Department of Chemistry and Biochemistry, University of Texas at Arlington, 700 Planetarium Pl, Arlington, TX 76010, USA
| | - Jayanta Kishor Chakrabarty
- Department of Chemistry and Biochemistry, University of Texas at Arlington, 700 Planetarium Pl, Arlington, TX 76010, USA
| | - Lauren E Fuess
- Department of Ecology and Evolutionary Biology University of Connecticut, Storrs, CT 06269, USA
| | - Whitney T Mann
- Department of Biology, University of Texas at Arlington, Arlington, 501 S Nedderman Dr., TX 76010, USA
| | - Lea R Jinks
- Department of Biology, University of Texas at Arlington, Arlington, 501 S Nedderman Dr., TX 76010, USA
| | - Vanessa Brinkhuis
- Washington State Department of Ecology—Central Regional Office, 1250 Alder Street, Union Gap, WA 98903, USA
| | - Saiful M Chowdhury
- Department of Chemistry and Biochemistry, University of Texas at Arlington, 700 Planetarium Pl, Arlington, TX 76010, USA
| | - Laura D Mydlarz
- Department of Biology, University of Texas at Arlington, Arlington, 501 S Nedderman Dr., TX 76010, USA
| |
Collapse
|
40
|
Huang F, Song Y, Chen W, Liu Q, Wang Q, Liu W, Wang X, Wang W. Effects of Candida albicans infection on defense effector secretion by human oral mucosal epithelial cells. Arch Oral Biol 2019; 103:55-61. [PMID: 31136880 DOI: 10.1016/j.archoralbio.2019.05.013] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2019] [Revised: 05/12/2019] [Accepted: 05/13/2019] [Indexed: 12/28/2022]
Abstract
OBJECTIVE The aim of this study was to investigate the effects of Candida albicans on the production of defense effector molecules by human oral mucosal epithelial cells in vitro. DESIGN Immortalized human oral mucosal epithelial (Leuk-1) cells and C. albicans strain 5314 were cocultured at different cell-to-C. albicans ratios. The viability of Leuk-1 cells was determined by MTT and RTCA measurements. The secretory levels of multiple defense effector molecules were determined by Enzyme-linked immunosorbent assay (ELISA). RESULTS Our results indicated that C. albicans significantly decreased the secretion of IgG, cystatin C, lactoferrin, and TGF-β1 in a dose-dependent manner and remarkably reduced the production of IgA independent of the cell-to-C. albicans ratio. However, C. albicans clearly enhanced the secretion of IgM, galectin-3, P-selectin, granzyme B and perforin. CONCLUSION These results suggest that C. albicans may exert a regulatory role in the defense response of oral mucosal epithelial cells by altering secretory levels of defense effector molecules.
Collapse
Affiliation(s)
- Fan Huang
- Department of Oral Medicine, Nanjing Stomatological Hospital, Medical School of Nanjing University, Nanjing, China
| | - Yuefeng Song
- Department of Oral Medicine, Nanjing Stomatological Hospital, Medical School of Nanjing University, Nanjing, China
| | - Wei Chen
- Department of Oral Medicine, Nanjing Stomatological Hospital, Medical School of Nanjing University, Nanjing, China
| | - Qin Liu
- Department of Oral Medicine, Nanjing Stomatological Hospital, Medical School of Nanjing University, Nanjing, China
| | - Qiong Wang
- Department of Mycology, Institute of Dermatology, Chinese Academy of Medical Sciences (CAMS) & Peking Union Medical College (PUMC), Jiangsu Key Laboratory of Molecular Biology for Skin Disease and STIs, Nanjing, China
| | - Weida Liu
- Department of Mycology, Institute of Dermatology, Chinese Academy of Medical Sciences (CAMS) & Peking Union Medical College (PUMC), Jiangsu Key Laboratory of Molecular Biology for Skin Disease and STIs, Nanjing, China
| | - Xiang Wang
- Department of Oral Medicine, Nanjing Stomatological Hospital, Medical School of Nanjing University, Nanjing, China.
| | - Wenmei Wang
- Department of Oral Medicine, Nanjing Stomatological Hospital, Medical School of Nanjing University, Nanjing, China.
| |
Collapse
|
41
|
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
|
42
|
Tamai R, Kiyoura Y. Heat-killed Candida albicans augments synthetic bacterial component-induced proinflammatory cytokine production. Folia Microbiol (Praha) 2019; 64:555-566. [PMID: 30656591 DOI: 10.1007/s12223-019-00679-2] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2018] [Accepted: 01/07/2019] [Indexed: 12/23/2022]
Abstract
Candida albicans can enhance the invasion of oral epithelial cells by Porphyromonas gingivalis, although the fungus is not a periodontal pathogen. In this study, we investigated whether C. albicans augments proinflammatory cytokine production by mouse macrophage-like J774.1 cells incubated with synthetic bacterial components. Mouse macrophage-like J774.1 cells, mouse primary splenocytes, human THP-1 cells, and A549 cells were pretreated with or without heat-killed C. albicans (HKCA) or substitutes for C. albicans cell wall components in 96-well flat-bottomed plates. Cells were then washed and incubated with Pam3CSK4, a Toll-like receptor (TLR) 2 ligand, or lipid A, a TLR4 ligand. Culture supernatants were analyzed by ELISA for secreted IL-6, MCP-1, TNF-α, and IL-8. HKCA augmented TLR ligand-induced proinflammatory cytokine production by J774.1 cells, mouse splenocytes, and THP-1 cells, but not A549 cells. However, IL-6, MCP-1, and TNF-α production induced by Pam3CSK4 or lipid A was not augmented when cells were pretreated with curdlan, a dectin-1 ligand, or mannan, a dectin-2 ligand. In contrast, pretreatment of cells with TLR ligands upregulated the production of IL-6 and TNF-α, but not MCP-1, induced by Pam3CSK4 or lipid A. The results suggest that C. albicans augments synthetic bacterial component-induced cytokine production by J774.1 cells via the TLR pathway, but not the dectin-1 or dectin-2 pathway.
Collapse
Affiliation(s)
- Riyoko Tamai
- Department of Oral Medical Science, Ohu University School of Dentistry, 31-1 Misumido, Tomitamachi, Koriyama, Fukushima, 963-8611, Japan.
| | - Yusuke Kiyoura
- Department of Oral Medical Science, Ohu University School of Dentistry, 31-1 Misumido, Tomitamachi, Koriyama, Fukushima, 963-8611, Japan
| |
Collapse
|
43
|
Childers DS, Avelar GM, Bain JM, Larcombe DE, Pradhan A, Budge S, Heaney H, Brown AJP. Impact of the Environment upon the Candida albicans Cell Wall and Resultant Effects upon Immune Surveillance. Curr Top Microbiol Immunol 2019; 425:297-330. [PMID: 31781866 DOI: 10.1007/82_2019_182] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The fungal cell wall is an essential organelle that maintains cellular morphology and protects the fungus from environmental insults. For fungal pathogens such as Candida albicans, it provides a degree of protection against attack by host immune defences. However, the cell wall also presents key epitopes that trigger host immunity and attractive targets for antifungal drugs. Rather than being a rigid shield, it has become clear that the fungal cell wall is an elastic organelle that permits rapid changes in cell volume and the transit of large liposomal particles such as extracellular vesicles. The fungal cell wall is also flexible in that it adapts to local environmental inputs, thereby enhancing the fitness of the fungus in these microenvironments. Recent evidence indicates that this cell wall adaptation affects host-fungus interactions by altering the exposure of major cell wall epitopes that are recognised by innate immune cells. Therefore, we discuss the impact of environmental adaptation upon fungal cell wall structure, and how this affects immune recognition, focussing on C. albicans and drawing parallels with other fungal pathogens.
Collapse
Affiliation(s)
- Delma S Childers
- Aberdeen Fungal Group, Institute of Medical Sciences, University of Aberdeen, Foresterhill, Aberdeen, AB25 2ZD, UK
| | - Gabriela M Avelar
- Aberdeen Fungal Group, Institute of Medical Sciences, University of Aberdeen, Foresterhill, Aberdeen, AB25 2ZD, UK
| | - Judith M Bain
- Aberdeen Fungal Group, Institute of Medical Sciences, University of Aberdeen, Foresterhill, Aberdeen, AB25 2ZD, UK
| | - Daniel E Larcombe
- Aberdeen Fungal Group, Institute of Medical Sciences, University of Aberdeen, Foresterhill, Aberdeen, AB25 2ZD, UK
- Medical Research Council Centre for Medical Mycology, University of Exeter, Geoffrey Pope Building, Stocker Road, Exeter, EX4 4QD, UK
| | - Arnab Pradhan
- Aberdeen Fungal Group, Institute of Medical Sciences, University of Aberdeen, Foresterhill, Aberdeen, AB25 2ZD, UK
- Medical Research Council Centre for Medical Mycology, University of Exeter, Geoffrey Pope Building, Stocker Road, Exeter, EX4 4QD, UK
| | - Susan Budge
- Aberdeen Fungal Group, Institute of Medical Sciences, University of Aberdeen, Foresterhill, Aberdeen, AB25 2ZD, UK
| | - Helen Heaney
- Aberdeen Fungal Group, Institute of Medical Sciences, University of Aberdeen, Foresterhill, Aberdeen, AB25 2ZD, UK
| | - Alistair J P Brown
- Aberdeen Fungal Group, Institute of Medical Sciences, University of Aberdeen, Foresterhill, Aberdeen, AB25 2ZD, UK.
- Medical Research Council Centre for Medical Mycology, University of Exeter, Geoffrey Pope Building, Stocker Road, Exeter, EX4 4QD, UK.
| |
Collapse
|
44
|
Nguyen TNY, Padungros P, Wongsrisupphakul P, Sa-Ard-Iam N, Mahanonda R, Matangkasombut O, Choo MK, Ritprajak P. Cell wall mannan of Candida krusei mediates dendritic cell apoptosis and orchestrates Th17 polarization via TLR-2/MyD88-dependent pathway. Sci Rep 2018; 8:17123. [PMID: 30459422 PMCID: PMC6244250 DOI: 10.1038/s41598-018-35101-3] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2018] [Accepted: 10/23/2018] [Indexed: 12/26/2022] Open
Abstract
Dendritic cells (DCs) abundantly express diverse receptors to recognize mannans in the outer surface of Candida cell wall, and these interactions dictate the host immune responses that determine disease outcomes. C. krusei prevalence in candidiasis worldwide has increased since this pathogen has developed multidrug resistance. However, little is known how the immune system responds to C. krusei. Particularly, the molecular mechanisms of the interplay between C. krusei mannan and DCs remain to be elucidated. We investigated how C. krusei mannan affected DC responses in comparison to C. albicans, C. tropicalis and C. glabrata mannan. Our results showed that only C. krusei mannan induced massive cytokine responses in DCs, and led to apoptosis. Although C. krusei mannan-activated DCs underwent apoptosis, they were still capable of initiating Th17 response. C. krusei mannan-mediated DC apoptosis was obligated to the TLR2 and MyD88 pathway. These pathways also controlled Th1/Th17 switching possibly by virtue of the production of the polarizing cytokines IL-12 and IL-6 by the C. krusei mannan activated-DCs. Our study suggests that TLR2 and MyD88 pathway in DCs are dominant for C. krusei mannan recognition, which differs from the previous reports showing a crucial role of C-type lectin receptors in Candida mannan sensing.
Collapse
Affiliation(s)
- Thu Ngoc Yen Nguyen
- Graduate program in Oral Biology, Faculty of Dentistry, Chulalongkorn University, Bangkok, 10330, Thailand
| | - Panuwat Padungros
- Organic Synthesis Research Unit, Department of Chemistry, Faculty of Science, Chulalongkorn University, Phayathai Road, Patumwan, Bangkok, 10330, Thailand
| | - Panachai Wongsrisupphakul
- Organic Synthesis Research Unit, Department of Chemistry, Faculty of Science, Chulalongkorn University, Phayathai Road, Patumwan, Bangkok, 10330, Thailand
| | - Noppadol Sa-Ard-Iam
- Immunology Laboratory, Faculty of Dentistry, Chulalongkorn University, Bangkok, 10330, Thailand
| | - Rangsini Mahanonda
- Immunology Laboratory, Faculty of Dentistry, Chulalongkorn University, Bangkok, 10330, Thailand
- Department of Periodontology, Faculty of Dentistry, Chulalongkorn University, Bangkok, 10330, Thailand
| | - Oranart Matangkasombut
- Laboratory of Biotechnology, Chulabhorn Research Institute, Bangkok, 10210, Thailand
- Research Unit on Oral Microbiology and Immunology and Department of Microbiology, Faculty of Dentistry, Chulalongkorn University, Bangkok, 10330, Thailand
| | - Min-Kyung Choo
- Cutaneous Biology Research Center, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA, 02129, USA
| | - Patcharee Ritprajak
- Research Unit on Oral Microbiology and Immunology and Department of Microbiology, Faculty of Dentistry, Chulalongkorn University, Bangkok, 10330, Thailand.
- Oral Biology Research Center, Faculty of Dentistry, Chulalongkorn University, Bangkok, Thailand.
| |
Collapse
|
45
|
Ferreira RG, Rodrigues LC, Nascimento DC, Kanashiro A, Melo PH, Borges VF, Gozzi A, da Silva Prado D, Borges MC, Ramalho FS, Stowell SR, Cummings RD, Dias-Baruffi M, Cunha FQ, Alves-Filho JC. Galectin-3 aggravates experimental polymicrobial sepsis by impairing neutrophil recruitment to the infectious focus. J Infect 2018; 77:391-397. [DOI: 10.1016/j.jinf.2018.06.010] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2017] [Revised: 05/31/2018] [Accepted: 06/21/2018] [Indexed: 12/29/2022]
|
46
|
Lehto M, Wolff H, Leino R, Alenius H, Savolainen J. A novel glycocluster molecule prevents timothy-induced allergic airway inflammation in mice. Allergy 2018; 73:1700-1706. [PMID: 29377154 DOI: 10.1111/all.13419] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/17/2018] [Indexed: 11/30/2022]
Abstract
BACKGROUND Allergen-specific immunotherapy (SIT) effectively alleviates type I allergic diseases characterized by T helper (Th)2-type immunity. Our recent studies have shown that a synthetic trivalent glycocluster, triacedimannose (TADM), suppresses the Th2-type allergic inflammation. The aim of this study was to compare TADM with two well-known adjuvants, unmethylated cytosine-phosphate-guanine oligodeoxynucleotide (CpG) and monophosphoryl lipid A (MPLA) in a grass allergen-induced chronic allergic inflammation model in mice. METHODS Female BALB/c mice were intranasally sensitized with 50 μL of timothy grass pollen extract (TE) twice a week for a period of 15 weeks. Therapeutic intranasal treatments were then performed once a week after the tenth intranasal TE instillation using TADM (10 or 25 μg/50 μL), CpG-ODN (20 μg/50 μL) or MPLA (2 μg/50 μL). Groups of 9-10 animals per treatment were killed 24 hours after the last timothy dosage. Blood, bronchoalveolar lavage (BAL) fluids and lung biopsies were taken for subsequent analysis. RESULTS When mice were repeatedly exposed to TE for 15 weeks, the number of eosinophils and lymphocytes increased in the BAL fluids. The eosinophil and lymphocyte counts decreased dose-dependently and were practically abolished in the mice treated with TADM. Treatments with MPLA or CpG significantly increased the numbers of neutrophils, while CpG nonsignificantly decreased eosinophilia compared to timothy exposure. CONCLUSIONS A novel synthetic glycocluster molecule inhibited the development of grass-induced eosinophilic pulmonary inflammation in mice when administrated in the airways. This compound could be a candidate to be used either as an adjuvant in SIT or as a topical anti-inflammatory treatment.
Collapse
Affiliation(s)
- M. Lehto
- Department of Occupational Medicine; Finnish Institute of Occupational Health; Helsinki Finland
| | - H. Wolff
- Department of Pathology; Finnish Institute of Occupational Health; Helsinki Finland
| | - R. Leino
- Johan Gadolin Process Chemistry Centre; Laboratory of Organic Chemistry; Åbo Akademi University; Turku Finland
| | - H. Alenius
- Karolinska Institutet; Institute of Environmental Medicine; Stockholm Sweden
- Medical Faculty; University of Helsinki; Helsinki Finland
| | - J. Savolainen
- Department of Pulmonary Diseases and Clinical Allergology; University of Turku; Turku University Hospital; Turku Finland
| |
Collapse
|
47
|
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
|
48
|
Tamai R, Kobayashi-Sakamoto M, Kiyoura Y. Extracellular galectin-1 enhances adhesion to and invasion of oral epithelial cells by Porphyromonas gingivalis. Can J Microbiol 2018; 64:465-471. [PMID: 29544077 DOI: 10.1139/cjm-2017-0461] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Galectin-1 and galectin-3 are C-type lectin receptors that bind to lipopolysaccharide in the cell wall of gram-negative bacteria. In this study, we investigated the effects of galectin-1 and galectin-3 on adhesion to and invasion of the human gingival epithelial cell line Ca9-22 by Porphyromonas gingivalis, a periodontal pathogenic gram-negative bacterium. Recombinant galectin-1, but not galectin-3, enhanced P. gingivalis adhesion and invasion, although both galectins bound similarly to P. gingivalis. Flow cytometry also revealed that Ca9-22 cells express low levels of galectin-1 and moderate levels of galectin-3. Ca9-22 cells in which galectin-3 was knocked-down did not exhibit enhanced P. gingivalis adhesion and invasion. Similarly, specific antibodies to galectin-1 and galectin-3 did not inhibit P. gingivalis adhesion and invasion. These results suggest that soluble galectin-1, but not galectin-3, may exacerbate periodontal disease by enhancing the adhesion to and invasion of host cells by periodontal pathogenic bacteria.
Collapse
Affiliation(s)
- Riyoko Tamai
- Department of Oral Medical Science, Ohu University School of Dentistry, 31-1 Misumido, Tomitamachi, Koriyama, Fukushima 963-8611, Japan.,Department of Oral Medical Science, Ohu University School of Dentistry, 31-1 Misumido, Tomitamachi, Koriyama, Fukushima 963-8611, Japan
| | - Michiyo Kobayashi-Sakamoto
- Department of Oral Medical Science, Ohu University School of Dentistry, 31-1 Misumido, Tomitamachi, Koriyama, Fukushima 963-8611, Japan.,Department of Oral Medical Science, Ohu University School of Dentistry, 31-1 Misumido, Tomitamachi, Koriyama, Fukushima 963-8611, Japan
| | - Yusuke Kiyoura
- Department of Oral Medical Science, Ohu University School of Dentistry, 31-1 Misumido, Tomitamachi, Koriyama, Fukushima 963-8611, Japan.,Department of Oral Medical Science, Ohu University School of Dentistry, 31-1 Misumido, Tomitamachi, Koriyama, Fukushima 963-8611, Japan
| |
Collapse
|
49
|
Weng IC, Chen HL, Lo TH, Lin WH, Chen HY, Hsu DK, Liu FT. Cytosolic galectin-3 and -8 regulate antibacterial autophagy through differential recognition of host glycans on damaged phagosomes. Glycobiology 2018; 28:392-405. [DOI: 10.1093/glycob/cwy017] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2018] [Accepted: 02/23/2018] [Indexed: 12/23/2022] Open
Affiliation(s)
- I-Chun Weng
- Institute of Biomedical Sciences, Academia Sinica, Taipei 115, Taiwan
| | - Hung-Lin Chen
- Institute of Biomedical Sciences, Academia Sinica, Taipei 115, Taiwan
| | - Tzu-Han Lo
- Institute of Biomedical Sciences, Academia Sinica, Taipei 115, Taiwan
| | - Wei-Han Lin
- Institute of Biomedical Sciences, Academia Sinica, Taipei 115, Taiwan
| | - Huan-Yuan Chen
- Institute of Biomedical Sciences, Academia Sinica, Taipei 115, Taiwan
| | - Daniel K Hsu
- Department of Dermatology, School of Medicine, University of California-Davis, Sacramento, CA 95817, USA
| | - Fu-Tong Liu
- Institute of Biomedical Sciences, Academia Sinica, Taipei 115, Taiwan
- Department of Dermatology, School of Medicine, University of California-Davis, Sacramento, CA 95817, USA
| |
Collapse
|
50
|
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
|