1
|
Wang K, Liu Y, Zhang Z, Zheng Z, Tang W, Teng W, Mu X, Wang J, Zhang Y. Insights into oral lentinan immunomodulation: Dectin-1-mediated lymphatic transport from Peyer's patch M cells to mononuclear phagocytes. Carbohydr Polym 2024; 346:122586. [PMID: 39245482 DOI: 10.1016/j.carbpol.2024.122586] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2024] [Revised: 07/22/2024] [Accepted: 08/06/2024] [Indexed: 09/10/2024]
Abstract
Lentinan (LNT), a natural polysaccharide, has been reported to exhibit immunomodulatory effects in the intestine after oral administration. Herein, we aimed to investigate the lymphatic transport of LNT in Peyer's patches (PPs) by traceable fluorescent labeling and to explore whether/how LNT contacts related immune cells. Near-infrared imaging confirmed the absorption of LNT in the small intestinal segment and its accumulation within PPs after oral administration. Subsequently, tissue imaging confirmed that M cells are the main cells responsible for transporting LNT to PPs, and an M cell model was established to explore the involvement of Dectin-1 in the absorption process. Systematic in vitro and in vivo studies revealed that the Dectin-1 further mediates the uptake of LNT by mononuclear phagocytes in PPs. Moreover, LNT can promote the proliferation and differentiation of mononuclear phagocytes, thereby activating immune responses. In summary, this study elucidates the pharmacokinetic mechanisms by which LNT exerts oral immunomodulatory effects, providing a theoretical basis for the development and application of other polysaccharides.
Collapse
Affiliation(s)
- Kaiping Wang
- Hubei Key Laboratory of Nature Medicinal Chemistry and Resource Evaluation, Tongji Medical College of Pharmacy, Huazhong University of Science and Technology, 430030 Wuhan, China
| | - Yuxuan Liu
- Hubei Key Laboratory of Nature Medicinal Chemistry and Resource Evaluation, Tongji Medical College of Pharmacy, Huazhong University of Science and Technology, 430030 Wuhan, China
| | - Zeming Zhang
- Hubei Key Laboratory of Nature Medicinal Chemistry and Resource Evaluation, Tongji Medical College of Pharmacy, Huazhong University of Science and Technology, 430030 Wuhan, China
| | - Ziming Zheng
- Department of Pharmacy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 430030 Wuhan, China; Hubei Province Clinical Research Center for Precision Medicine for Critical Illness, 430030 Wuhan, PR China
| | - Wenqi Tang
- Department of Pharmacy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 430030 Wuhan, China
| | - Wangtianzi Teng
- Hubei Key Laboratory of Nature Medicinal Chemistry and Resource Evaluation, Tongji Medical College of Pharmacy, Huazhong University of Science and Technology, 430030 Wuhan, China
| | - Xu Mu
- Department of Pharmacy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 430030 Wuhan, China
| | - Jinglin Wang
- Department of Pharmacy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 430030 Wuhan, China; Hubei Province Clinical Research Center for Precision Medicine for Critical Illness, 430030 Wuhan, PR China.
| | - Yu Zhang
- Department of Pharmacy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 430030 Wuhan, China; Hubei Province Clinical Research Center for Precision Medicine for Critical Illness, 430030 Wuhan, PR China.
| |
Collapse
|
2
|
Sahu SR, Dutta A, Peroumal D, Kumari P, Utakalaja BG, Patel SK, Acharya N. Immunogenicity and efficacy of CNA25 as a potential whole-cell vaccine against systemic candidiasis. EMBO Mol Med 2024; 16:1254-1283. [PMID: 38783167 PMCID: PMC11178797 DOI: 10.1038/s44321-024-00080-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2023] [Revised: 05/10/2024] [Accepted: 05/10/2024] [Indexed: 05/25/2024] Open
Abstract
Disseminated fungal infections account for ~1.5 million deaths per year worldwide, and mortality may increase further due to a rise in the number of immunocompromised individuals and drug-resistance fungal species. Since an approved antifungal vaccine is yet to be available, this study explored the immunogenicity and vaccine efficacy of a DNA polymerase mutant strain of Candida albicans. CNA25 is a pol32ΔΔ strain that exhibits growth defects and does not cause systemic candidiasis in mice. Immunized mice with live CNA25 were fully protected against C. albicans and C. parapsilosis but partially against C. tropicalis and C. glabrata infections. CNA25 induced steady expression of TLR2 and Dectin-1 receptors leading to a faster recognition and clearance by the immune system associated with the activation of protective immune responses mostly mediated by neutrophils, macrophages, NK cells, B cells, and CD4+ and CD8+ T cells. Molecular blockade of Dectin-1, IL-17, IFNγ, and TNFα abolished resistance to reinfection. Altogether, this study suggested that CNA25 collectively activates innate, adaptive, and trained immunity to be a promising live whole-cell vaccine against systemic candidiasis.
Collapse
Affiliation(s)
- Satya Ranjan Sahu
- Department of Infectious Disease Biology, Institute of Life Sciences, Bhubaneswar, Odisha, 751023, India
- Regional Center for Biotechnology, Faridabad, Haryana, 751021, India
| | - Abinash Dutta
- Department of Infectious Disease Biology, Institute of Life Sciences, Bhubaneswar, Odisha, 751023, India
| | - Doureradjou Peroumal
- Department of Infectious Disease Biology, Institute of Life Sciences, Bhubaneswar, Odisha, 751023, India
| | - Premlata Kumari
- Department of Infectious Disease Biology, Institute of Life Sciences, Bhubaneswar, Odisha, 751023, India
- Regional Center for Biotechnology, Faridabad, Haryana, 751021, India
| | - Bhabasha Gyanadeep Utakalaja
- Department of Infectious Disease Biology, Institute of Life Sciences, Bhubaneswar, Odisha, 751023, India
- Regional Center for Biotechnology, Faridabad, Haryana, 751021, India
| | - Shraddheya Kumar Patel
- Department of Infectious Disease Biology, Institute of Life Sciences, Bhubaneswar, Odisha, 751023, India
- Regional Center for Biotechnology, Faridabad, Haryana, 751021, India
| | - Narottam Acharya
- Department of Infectious Disease Biology, Institute of Life Sciences, Bhubaneswar, Odisha, 751023, India.
| |
Collapse
|
3
|
Case S, O'Brien T, Ledwith AE, Chen S, Horneck Johnston CJH, Hackett EE, O'Sullivan M, Charles-Messance H, Dempsey E, Yadav S, Wilson J, Corr SC, Nagar S, Sheedy FJ. β-glucans from Agaricus bisporus mushroom products drive Trained Immunity. Front Nutr 2024; 11:1346706. [PMID: 38425482 PMCID: PMC10902450 DOI: 10.3389/fnut.2024.1346706] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2023] [Accepted: 01/29/2024] [Indexed: 03/02/2024] Open
Abstract
Introduction Macrofungi, such as edible mushrooms, have been used as a valuable medical resource for millennia as a result of their antibacterial and immuno-modulatory components. Mushrooms contain dietary fibers known as β-glucans, a class of polysaccharides previously linked to the induction of Trained Immunity. However, little is known about the ability of mushroom-derived β-glucans to induce Trained Immunity. Methods & results Using various powdered forms of the white button mushroom (Agaricus bisporus), we found that mouse macrophages pre-treated with whole mushroom powder (WMP) displayed enhanced responses to restimulation with TLR ligands, being particularly sensitive to Toll-like receptor (TLR)-2 stimulation using synthetic lipopeptides. This trained response was modest compared to training observed with yeast-derived β-glucans and correlated with the amount of available β-glucans in the WMP. Enriching for β-glucans content using either a simulated in-vitro digestion or chemical fractionation retained and boosted the trained response with WMP, respectively. Importantly, both WMP and digested-WMP preparations retained β-glucans as identified by nuclear magnetic resonance analysis and both displayed the capacity to train human monocytes and enhanced responses to restimulation. To determine if dietary incorporation of mushroom products can lead to Trained Immunity in myeloid cells in vivo, mice were given a regimen of WMP by oral gavage prior to sacrifice. Flow cytometric analysis of bone-marrow progenitors indicated alterations in hematopoietic stem and progenitor cells population dynamics, with shift toward myeloid-committed multi-potent progenitor cells. Mature bone marrow-derived macrophages derived from these mice displayed enhanced responses to restimulation, again particularly sensitive to TLR2. Discussion Taken together, these data demonstrate that β-glucans from common macrofungi can train innate immune cells and could point to novel ways of delivering bio-available β-glucans for education of the innate immune system.
Collapse
Affiliation(s)
- Sarah Case
- School of Biochemistry and Immunology, Trinity College, Dublin, Ireland
| | - Tara O'Brien
- School of Biochemistry and Immunology, Trinity College, Dublin, Ireland
| | - Anna E. Ledwith
- School of Biochemistry and Immunology, Trinity College, Dublin, Ireland
| | - Shilong Chen
- NatPro Centre, School of Pharmacy and Pharmaceutical Sciences, Trinity College, Dublin, Ireland
| | | | - Emer E. Hackett
- School of Biochemistry and Immunology, Trinity College, Dublin, Ireland
| | | | | | - Elaine Dempsey
- School of Genetics and Microbiology, Trinity College, Dublin, Ireland
| | | | | | - Sinead C. Corr
- School of Genetics and Microbiology, Trinity College, Dublin, Ireland
- APC Microbiome Ireland, University College Cork, Cork, Ireland
| | - Shipra Nagar
- NatPro Centre, School of Pharmacy and Pharmaceutical Sciences, Trinity College, Dublin, Ireland
| | - Frederick J. Sheedy
- School of Biochemistry and Immunology, Trinity College, Dublin, Ireland
- NatPro Centre, School of Pharmacy and Pharmaceutical Sciences, Trinity College, Dublin, Ireland
| |
Collapse
|
4
|
Wan T, Wang Y, He K, Zhu S. Microbial sensing in the intestine. Protein Cell 2023; 14:824-860. [PMID: 37191444 PMCID: PMC10636641 DOI: 10.1093/procel/pwad028] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2023] [Accepted: 05/04/2023] [Indexed: 05/17/2023] Open
Abstract
The gut microbiota plays a key role in host health and disease, particularly through their interactions with the immune system. Intestinal homeostasis is dependent on the symbiotic relationships between the host and the diverse gut microbiota, which is influenced by the highly co-evolved immune-microbiota interactions. The first step of the interaction between the host and the gut microbiota is the sensing of the gut microbes by the host immune system. In this review, we describe the cells of the host immune system and the proteins that sense the components and metabolites of the gut microbes. We further highlight the essential roles of pattern recognition receptors (PRRs), the G protein-coupled receptors (GPCRs), aryl hydrocarbon receptor (AHR) and the nuclear receptors expressed in the intestinal epithelial cells (IECs) and the intestine-resident immune cells. We also discuss the mechanisms by which the disruption of microbial sensing because of genetic or environmental factors causes human diseases such as the inflammatory bowel disease (IBD).
Collapse
Affiliation(s)
- Tingting Wan
- Division of Life Sciences and Medicine, The CAS Key Laboratory of Innate Immunity and Chronic Disease, Institute of Immunology, School of Basic Medical Sciences, University of Science and Technology of China, Hefei 230027, China
| | - Yalong Wang
- Division of Life Sciences and Medicine, The CAS Key Laboratory of Innate Immunity and Chronic Disease, Institute of Immunology, School of Basic Medical Sciences, University of Science and Technology of China, Hefei 230027, China
| | - Kaixin He
- Division of Life Sciences and Medicine, The CAS Key Laboratory of Innate Immunity and Chronic Disease, Institute of Immunology, School of Basic Medical Sciences, University of Science and Technology of China, Hefei 230027, China
| | - Shu Zhu
- Division of Life Sciences and Medicine, The CAS Key Laboratory of Innate Immunity and Chronic Disease, Institute of Immunology, School of Basic Medical Sciences, University of Science and Technology of China, Hefei 230027, China
- Department of Digestive Disease, Division of Life Sciences and Medicine, The First Affiliated Hospital of USTC, University of Science and Technology of China, Hefei 230001, China
- Institute of Health and Medicine, Hefei Comprehensive National Science Center, Hefei 230601, China
| |
Collapse
|
5
|
Harasawa A, Ishiyama S, Mochizuki K. Fructo-oligosaccharide-mediated alteration in claudin expression in small intestinal absorptive Caco-2 cells is positively associated with the induction of inflammatory genes and the glucan receptor gene CLEC7A. Nutrition 2023; 115:112140. [PMID: 37481839 DOI: 10.1016/j.nut.2023.112140] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2023] [Revised: 06/02/2023] [Accepted: 06/18/2023] [Indexed: 07/25/2023]
Abstract
OBJECTIVES Indigestible carbohydrates may strengthen tight junctions (TJs) independently of intestinal bacteria. This study investigated whether indigestible carbohydrates (i.e., fructo-oligosaccharides [FOS]) promote TJs directly to intestinal absorptive Caco-2 cells and examined the association between the expression of genes constructing TJs and other genes using mRNA microarray analysis. METHODS Caco-2 cells at 1.0 × 105/mL were seeded in a type I collagen plate and cultured in high-glucose Dulbecco's modified Eagle medium (DMEM) supplemented with 10% fetal calf serum (FCS); the cells reached confluence at 7 d after seeding. Ten days after the cells reached confluency, they were cultured for 24 h in 10% FCS-containing DMEM medium supplemented with 0%, 5%, or 10% FOS. We performed mRNA microarray to identify the genes whose expression was altered by FOS. Subsequently, quantitative reverse transcription polymerase chain reaction was performed for these altered genes, including CLEC7A encoding the glucan receptor, and for the claudin (CLDN) family genes. The expression of CLDN2, CLDN4, and CLEC7A proteins was assessed using western blot analysis. RESULTS FOS decreased the mRNA and protein expression of CLDN2, which weakens TJs, and increased those of CLDN4, which strengthens TJs, in Caco-2 cells. FOS treatment (10%) reduced the mRNA expression of antioxidative genes and induced the expression of immune response-related genes, including CLEC7A, CCL2, and ITGA2. Furthermore, the expression of CLEC7A protein was enhanced by FOS. CONCLUSIONS Induction of TJ-strengthening CLDN4 and reduction of TJ-weakening CLDN2 by FOS treatment in small intestinal absorptive Caco-2 cells is positively associated with the induction of inflammatory genes, including CLEC7A.
Collapse
Affiliation(s)
- Aya Harasawa
- Department of Integrated Applied Life Science, Integrated Graduate School of Medicine, Engineering, and Agricultural Sciences, University of Yamanashi, Kofu, Japan
| | - Shiori Ishiyama
- Laboratory of Food and Nutritional Sciences, Department of Local Produce and Food Sciences, Faculty of Life and Environmental Sciences, University of Yamanashi, Kofu, Japan
| | - Kazuki Mochizuki
- Laboratory of Food and Nutritional Sciences, Department of Local Produce and Food Sciences, Faculty of Life and Environmental Sciences, University of Yamanashi, Kofu, Japan.
| |
Collapse
|
6
|
Al B, Suen TK, Placek K, Netea MG. Innate (learned) memory. J Allergy Clin Immunol 2023; 152:551-566. [PMID: 37385546 DOI: 10.1016/j.jaci.2023.06.014] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2023] [Revised: 05/24/2023] [Accepted: 06/01/2023] [Indexed: 07/01/2023]
Abstract
With the growing body of evidence, it is now clear that not only adaptive immune cells but also innate immune cells can mount a more rapid and potent nonspecific immune response to subsequent exposures. This process is known as trained immunity or innate (learned) immune memory. This review discusses the different immune and nonimmune cell types of the central and peripheral immune systems that can develop trained immunity. This review highlights the intracellular signaling and metabolic and epigenetic mechanisms underlying the formation of innate immune memory. Finally, this review explores the health implications together with the potential therapeutic interventions harnessing trained immunity.
Collapse
Affiliation(s)
- Burcu Al
- Department of Molecular Immunology and Cell Biology, Life and Medical Sciences Institute, University of Bonn
| | - Tsz K Suen
- Department of Molecular Immunology and Cell Biology, Life and Medical Sciences Institute, University of Bonn
| | - Katarzyna Placek
- Department of Molecular Immunology and Cell Biology, Life and Medical Sciences Institute, University of Bonn
| | - Mihai G Netea
- Department of Molecular Immunology and Cell Biology, Life and Medical Sciences Institute, University of Bonn; Department of Internal Medicine and Radboud Center for Infectious Diseases, Radboud University Medical Center, Nijmegen.
| |
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
|
Zhou Y, Zhang Y, Yu W, Qin Y, He H, Dai F, Wang Y, Zhu F, Zhou G. Immunomodulatory role of spleen tyrosine kinase in chronic inflammatory and autoimmune diseases. Immun Inflamm Dis 2023; 11:e934. [PMID: 37506139 PMCID: PMC10373573 DOI: 10.1002/iid3.934] [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: 01/26/2023] [Revised: 06/18/2023] [Accepted: 06/19/2023] [Indexed: 07/30/2023] Open
Abstract
BACKGROUND The high prevalence of chronic inflammatory diseases or autoimmune reactions is a major source of concern and affects the quality of life of patients. Chronic inflammatory or autoimmune diseases are associated with many diseases in humans, including asthma, rheumatoid arthritis, systemic lupus erythematosus, inflammatory bowel disease and cancer. Splenic tyrosine kinase (SYK) is a non-receptor tyrosine kinase that plays an important role in immune receptor signalling in immune and inflammatory responses. METHODS This is a review article in which we searched for keywords "splenic tyrosine kinase", "inflammation" and "autoimmune diseases" in published literature such as Pubmed and Web of Science to collect relevant information and then conducted a study focusing on the latest findings on the involvement of SYK in chronic inflammatory or autoimmune diseases. RESULTS This paper reviews the regulation of Fcγ, NF-κB, B cell and T cell-related signalling pathways by SYK, which contributes to disease progression in chronic inflammatory and autoimmune diseases such as airway fibrosis, inflammatory skin disease and inflammatory bowel disease. CONCLUSION This paper shows that SYK plays an important role in chronic inflammatory and autoimmune diseases. syk targets hematological, autoimmune and other inflammatory diseases and therefore, inhibition of SYK expression or blocking its related pathways may provide new ideas for clinical prevention and treatment of inflammatory or autoimmune diseases.
Collapse
Affiliation(s)
- Yaqi Zhou
- Department of Clinical Medicine, Jining Medical University, Jining, Shandong, China
| | - Yaowen Zhang
- Department of Gastroenterology, Affiliated Hospital of Jining Medical University, Jining Medical University, Jining, Shandong, China
| | - Wei Yu
- Department of Gastroenterology, Affiliated Hospital of Jining Medical University, Jining Medical University, Jining, Shandong, China
| | - Yufen Qin
- Department of Gastroenterology, Affiliated Hospital of Jining Medical University, Jining Medical University, Jining, Shandong, China
| | - Heng He
- Department of Gastroenterology, Affiliated Hospital of Jining Medical University, Jining Medical University, Jining, Shandong, China
| | - Fengxian Dai
- Department of Gastroenterology, Affiliated Hospital of Jining Medical University, Jining Medical University, Jining, Shandong, China
| | - Yibo Wang
- Department of Gastroenterology, Affiliated Hospital of Jining Medical University, Jining Medical University, Jining, Shandong, China
| | - Fengqin Zhu
- Department of Gastroenterology, Affiliated Hospital of Jining Medical University, Jining Medical University, Jining, Shandong, China
| | - Guangxi Zhou
- Department of Gastroenterology, Affiliated Hospital of Jining Medical University, Jining Medical University, Jining, Shandong, China
| |
Collapse
|
9
|
Zhang J, Xiao Y, Wang H, Zhang H, Chen W, Lu W. Lactic acid bacteria-derived exopolysaccharide: Formation, immunomodulatory ability, health effects, and structure-function relationship. Microbiol Res 2023; 274:127432. [PMID: 37320895 DOI: 10.1016/j.micres.2023.127432] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2023] [Revised: 06/05/2023] [Accepted: 06/06/2023] [Indexed: 06/17/2023]
Abstract
Exopolysaccharides (EPSs) synthesized by lactic acid bacteria (LAB) have implications for host health and act as food ingredients. Due to the variability of LAB-EPS (lactic acid bacteria-derived exopolysaccharide) gene clusters, especially the glycosyltransferase genes that determine monosaccharide composition, the structure of EPS is very rich. EPSs are synthesized by LAB through the extracellular synthesis pathway and the Wzx/Wzy-dependent pathway. LAB-EPS has a strong immunomodulatory ability. The EPSs produced by different genera of LAB, especially Lactobacillus, Leuconostoc, and Streptococcus, have different immunomodulatory abilities because of their specific structures. LAB-EPS possesses other health effects, including antitumor, antioxidant, intestinal barrier repair, antimicrobial, antiviral, and cholesterol-lowering activities. The bioactivities of LAB-EPS are tightly related to their structures such us monosaccharide composition, glycosidic bonds, and molecular weight (MW). For the excellent physicochemical property, LAB-EPS acts as product improvers in dairy, bakery food, and meat in terms of stability, emulsification, thickening, and gelling. We systematically summarize the detailed process of EPS from synthesis to application, with emphasis on physiological mechanisms of EPS, and specific structure-function relationship, which provides theoretical support for the potential commercial value in the pharmaceutical, chemical, food, and cosmetic industries.
Collapse
Affiliation(s)
- Jie Zhang
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi, Jiangsu 214122, China; School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu 214122, China
| | - Yue Xiao
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi, Jiangsu 214122, China; School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu 214122, China
| | - Hongchao Wang
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi, Jiangsu 214122, China; School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu 214122, China
| | - Hao Zhang
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi, Jiangsu 214122, China; School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu 214122, China; National Engineering Research Center for Functional Food, Jiangnan University, Wuxi, Jiangsu 214122, China
| | - Wei Chen
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi, Jiangsu 214122, China; School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu 214122, China; National Engineering Research Center for Functional Food, Jiangnan University, Wuxi, Jiangsu 214122, China
| | - Wenwei Lu
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi, Jiangsu 214122, China; School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu 214122, China; National Engineering Research Center for Functional Food, Jiangnan University, Wuxi, Jiangsu 214122, China.
| |
Collapse
|
10
|
Wen C, Geervliet M, de Vries H, Fabà L, den Hil PJRV, Skovgaard K, Savelkoul HFJ, Schols HA, Wells JM, Tijhaar E, Smidt H. Agaricus subrufescens fermented rye affects the development of intestinal microbiota, local intestinal and innate immunity in suckling-to-nursery pigs. Anim Microbiome 2023; 5:24. [PMID: 37041617 PMCID: PMC10088699 DOI: 10.1186/s42523-023-00244-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Accepted: 03/23/2023] [Indexed: 04/13/2023] Open
Abstract
BACKGROUND Agaricus subrufescens is considered as one of the most important culinary-medicinal mushrooms around the world. It has been widely suggested to be used for the development of functional food ingredients to promote human health ascribed to the various properties (e.g., anti-inflammatory, antioxidant, and immunomodulatory activities). In this context, the interest in A. subrufescens based feed ingredients as alternatives for antibiotics has also been fuelled during an era of reduced/banned antibiotics use. This study aimed to investigate the effects of a fermented feed additive -rye overgrown with mycelium (ROM) of A. subrufescens-on pig intestinal microbiota, mucosal gene expression and local and systemic immunity during early life. Piglets received ROM or a tap water placebo (Ctrl) perorally every other day from day 2 after birth until 2 weeks post-weaning. Eight animals per treatment were euthanized and dissected on days 27, 44 and 70. RESULTS The results showed ROM piglets had a lower inter-individual variation of faecal microbiota composition before weaning and a lower relative abundance of proteobacterial genera in jejunum (Undibacterium and Solobacterium) and caecum (Intestinibacter and Succinivibrionaceae_UCG_001) on day 70, as compared to Ctrl piglets. ROM supplementation also influenced gut mucosal gene expression in both ileum and caecum on day 44. In ileum, ROM pigs showed increased expression of TJP1/ZO1 but decreased expression of CLDN3, CLDN5 and MUC2 than Ctrl pigs. Genes involved in TLR signalling (e.g., TICAM2, IRAK4 and LY96) were more expressed but MYD88 and TOLLIP were less expressed in ROM pigs than Ctrl animals. NOS2 and HIF1A involved in redox signalling were either decreased or increased in ROM pigs, respectively. In caecum, differentially expressed genes between two groups were mainly shown as increased expression (e.g., MUC2, PDGFRB, TOLLIP, TNFAIP3 and MYD88) in ROM pigs. Moreover, ROM animals showed higher NK cell activation in blood and enhanced IL-10 production in ex vivo stimulated MLN cells before weaning. CONCLUSIONS Collectively, these results suggest that ROM supplementation in early life modulates gut microbiota and (local) immune system development. Consequently, ROM supplementation may contribute to improving health of pigs during the weaning transition period and reducing antibiotics use.
Collapse
Affiliation(s)
- Caifang Wen
- Laboratory of Microbiology, Wageningen University & Research, Wageningen, The Netherlands
- Laboratory of Food Chemistry, Wageningen University & Research, Wageningen, The Netherlands
| | - Mirelle Geervliet
- Cell Biology and Immunology Group, Wageningen University & Research, Wageningen, The Netherlands
| | - Hugo de Vries
- Laboratory of Microbiology, Wageningen University & Research, Wageningen, The Netherlands
- Host-Microbe Interactomics Group, Wageningen University & Research, Wageningen, The Netherlands
| | - Lluís Fabà
- Research and Development, Trouw Nutrition, Amersfoort, The Netherlands
| | - Petra J Roubos-van den Hil
- Research and Development, Trouw Nutrition, Amersfoort, The Netherlands
- DSM Food and Beverages - Fresh Dairy, Wageningen, The Netherlands
| | - Kerstin Skovgaard
- Department of Biotechnology and Biomedicine, Technical University of Denmark, Lyngby, Denmark
| | - Huub F J Savelkoul
- Cell Biology and Immunology Group, Wageningen University & Research, Wageningen, The Netherlands
| | - Henk A Schols
- Laboratory of Food Chemistry, Wageningen University & Research, Wageningen, The Netherlands
| | - Jerry M Wells
- Host-Microbe Interactomics Group, Wageningen University & Research, Wageningen, The Netherlands
| | - Edwin Tijhaar
- Cell Biology and Immunology Group, Wageningen University & Research, Wageningen, The Netherlands
| | - Hauke Smidt
- Laboratory of Microbiology, Wageningen University & Research, Wageningen, The Netherlands.
| |
Collapse
|
11
|
Wang X, Chan YS, Wong K, Yoshitake R, Sadava D, Synold TW, Frankel P, Twardowski PW, Lau C, Chen S. Mechanism-Driven and Clinically Focused Development of Botanical Foods as Multitarget Anticancer Medicine: Collective Perspectives and Insights from Preclinical Studies, IND Applications and Early-Phase Clinical Trials. Cancers (Basel) 2023; 15:701. [PMID: 36765659 PMCID: PMC9913787 DOI: 10.3390/cancers15030701] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2022] [Revised: 01/13/2023] [Accepted: 01/19/2023] [Indexed: 01/25/2023] Open
Abstract
Cancer progression and mortality remain challenging because of current obstacles and limitations in cancer treatment. Continuous efforts are being made to explore complementary and alternative approaches to alleviate the suffering of cancer patients. Epidemiological and nutritional studies have indicated that consuming botanical foods is linked to a lower risk of cancer incidence and/or improved cancer prognosis after diagnosis. From these observations, a variety of preclinical and clinical studies have been carried out to evaluate the potential of botanical food products as anticancer medicines. Unfortunately, many investigations have been poorly designed, and encouraging preclinical results have not been translated into clinical success. Botanical products contain a wide variety of chemicals, making them more difficult to study than traditional drugs. In this review, with the consideration of the regulatory framework of the USFDA, we share our collective experiences and lessons learned from 20 years of defining anticancer foods, focusing on the critical aspects of preclinical studies that are required for an IND application, as well as the checkpoints needed for early-phase clinical trials. We recommend a developmental pipeline that is based on mechanisms and clinical considerations.
Collapse
Affiliation(s)
- Xiaoqiang Wang
- Department of Cancer Biology & Molecular Medicine, Beckman Research Institute, City of Hope, 1500 E. Duarte Rd., Duarte, CA 91010, USA
| | - Yin S. Chan
- Department of Cancer Biology & Molecular Medicine, Beckman Research Institute, City of Hope, 1500 E. Duarte Rd., Duarte, CA 91010, USA
| | - Kelly Wong
- Department of Cancer Biology & Molecular Medicine, Beckman Research Institute, City of Hope, 1500 E. Duarte Rd., Duarte, CA 91010, USA
| | - Ryohei Yoshitake
- Department of Cancer Biology & Molecular Medicine, Beckman Research Institute, City of Hope, 1500 E. Duarte Rd., Duarte, CA 91010, USA
| | - David Sadava
- Department of Cancer Biology & Molecular Medicine, Beckman Research Institute, City of Hope, 1500 E. Duarte Rd., Duarte, CA 91010, USA
| | - Timothy W. Synold
- Department of Medical Oncology & Therapeutics Research, City of Hope Comprehensive Cancer Center, 1500 E. Duarte Rd., Duarte, CA 91010, USA
| | - Paul Frankel
- Department of Computational and Quantitative Medicine, Beckman Research Institute, City of Hope, 1500 E. Duarte Rd., Duarte, CA 91010, USA
| | - Przemyslaw W. Twardowski
- Department of Urologic Oncology, Saint John’s Cancer Institute, 2200 Santa Monica Blvd, Santa Monica, CA 90404, USA
| | - Clayton Lau
- Department of Surgery, City of Hope Comprehensive Cancer Center, 1500 E. Duarte Rd., Duarte, CA 91010, USA
| | - Shiuan Chen
- Department of Cancer Biology & Molecular Medicine, Beckman Research Institute, City of Hope, 1500 E. Duarte Rd., Duarte, CA 91010, USA
| |
Collapse
|
12
|
Cohen-Kedar S, Shaham Barda E, Rabinowitz KM, Keizer D, Abu-Taha H, Schwartz S, Kaboub K, Baram L, Sadot E, White I, Wasserberg N, Wolff-Bar M, Levy-Barda A, Dotan I. Human intestinal epithelial cells can internalize luminal fungi via LC3-associated phagocytosis. Front Immunol 2023; 14:1142492. [PMID: 36969163 PMCID: PMC10030769 DOI: 10.3389/fimmu.2023.1142492] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2023] [Accepted: 02/22/2023] [Indexed: 03/29/2023] Open
Abstract
Background Intestinal epithelial cells (IECs) are the first to encounter luminal microorganisms and actively participate in intestinal immunity. We reported that IECs express the β-glucan receptor Dectin-1, and respond to commensal fungi and β-glucans. In phagocytes, Dectin-1 mediates LC3-associated phagocytosis (LAP) utilizing autophagy components to process extracellular cargo. Dectin-1 can mediate phagocytosis of β-glucan-containing particles by non-phagocytic cells. We aimed to determine whether human IECs phagocytose β-glucan-containing fungal particles via LAP. Methods Colonic (n=18) and ileal (n=4) organoids from individuals undergoing bowel resection were grown as monolayers. Fluorescent-dye conjugated zymosan (β-glucan particle), heat-killed- and UV inactivated C. albicans were applied to differentiated organoids and to human IEC lines. Confocal microscopy was used for live imaging and immuno-fluorescence. Quantification of phagocytosis was carried out with a fluorescence plate-reader. Results zymosan and C. albicans particles were phagocytosed by monolayers of human colonic and ileal organoids and IEC lines. LAP was identified by LC3 and Rubicon recruitment to phagosomes and lysosomal processing of internalized particles was demonstrated by co-localization with lysosomal dyes and LAMP2. Phagocytosis was significantly diminished by blockade of Dectin-1, actin polymerization and NAPDH oxidases. Conclusions Our results show that human IECs sense luminal fungal particles and internalize them via LAP. This novel mechanism of luminal sampling suggests that IECs may contribute to the maintenance of mucosal tolerance towards commensal fungi.
Collapse
Affiliation(s)
- Sarit Cohen-Kedar
- Division of Gastroenterology, Rabin Medical Center, Petah-Tikva, Israel
- Felsenstein Medical Research Center, Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv, Israel
- *Correspondence: Iris Dotan, ; Sarit Cohen-Kedar,
| | - Efrat Shaham Barda
- Division of Gastroenterology, Rabin Medical Center, Petah-Tikva, Israel
- Felsenstein Medical Research Center, Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv, Israel
| | - Keren Masha Rabinowitz
- Division of Gastroenterology, Rabin Medical Center, Petah-Tikva, Israel
- Felsenstein Medical Research Center, Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv, Israel
| | - Danielle Keizer
- Division of Gastroenterology, Rabin Medical Center, Petah-Tikva, Israel
- Felsenstein Medical Research Center, Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv, Israel
| | - Hanan Abu-Taha
- Division of Gastroenterology, Rabin Medical Center, Petah-Tikva, Israel
- Felsenstein Medical Research Center, Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv, Israel
| | - Shoshana Schwartz
- Division of Gastroenterology, Rabin Medical Center, Petah-Tikva, Israel
- Felsenstein Medical Research Center, Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv, Israel
| | - Kawsar Kaboub
- Division of Gastroenterology, Rabin Medical Center, Petah-Tikva, Israel
- Felsenstein Medical Research Center, Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv, Israel
| | - Liran Baram
- Felsenstein Medical Research Center, Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv, Israel
| | - Eran Sadot
- Division of Surgery, Rabin Medical Center, Petah-Tikva, Israel
| | - Ian White
- Division of Surgery, Rabin Medical Center, Petah-Tikva, Israel
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Nir Wasserberg
- Division of Surgery, Rabin Medical Center, Petah-Tikva, Israel
| | - Meirav Wolff-Bar
- Department of Pathology, Rabin Medical Center, Petah-Tikva, Israel
| | | | - Iris Dotan
- Division of Gastroenterology, Rabin Medical Center, Petah-Tikva, Israel
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
- *Correspondence: Iris Dotan, ; Sarit Cohen-Kedar,
| |
Collapse
|
13
|
Gould SJ, Foey AD, Salih VM. An organotypic oral mucosal infection model to study host-pathogen interactions. J Tissue Eng 2023; 14:20417314231197310. [PMID: 37873034 PMCID: PMC10590543 DOI: 10.1177/20417314231197310] [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] [Subscribe] [Scholar Register] [Received: 05/01/2023] [Accepted: 08/10/2023] [Indexed: 10/25/2023] Open
Abstract
Early in vitro oral mucosal infection models (OMMs) failed to consider the suitability of the model environment to represent the host immune response. Denture stomatitis (DS) is mediated by Candida albicans, but the role of Staphylococcus aureus remains uncertain. A collagen hydrogel-based OMM containing HaCaT and HGF cell types was developed, characterised and employed to study of tissue invasion and pro-inflammatory cytokine production in response to pathogens. Models formed a robust epithelium. Despite their inflammatory baseline, 24-h infection with C. albicans, and/or S. aureus led to tissue invasion, and significantly upregulated IL-6 and IL-8 production by OMMs when compared to the unstimulated control. No significant difference in IL-6 or IL-8 production by OMMs was observed between single and dual infections. These attributes indicate that this newly developed OMM is suitable for the study of DS and could be implemented for the wider study of oral infection.
Collapse
Affiliation(s)
- Samantha J Gould
- Department of Clinical and Biomedical Sciences, University of Exeter, Exeter, Devon, UK
| | - Andrew D Foey
- School of Biomedical Health Sciences, University of Plymouth, Plymouth, Devon, UK
| | - Vehid M Salih
- Peninsula Dental School, University of Plymouth, Plymouth, Devon, UK
| |
Collapse
|
14
|
Wagner AS, Vogel AK, Lumsdaine SW, Phillips EK, Willems HME, Peters BM, Reynolds TB. Mucosal Infection with Unmasked Candida albicans Cells Impacts Disease Progression in a Host Niche-Specific Manner. Infect Immun 2022; 90:e0034222. [PMID: 36374100 PMCID: PMC9753624 DOI: 10.1128/iai.00342-22] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2022] [Accepted: 10/26/2022] [Indexed: 11/16/2022] Open
Abstract
Shielding the immunogenic cell wall epitope β(1, 3)-glucan under an outer layer of mannosylated glycoproteins is an essential virulence factor deployed by Candida albicans during systemic infection. Accordingly, mutants with increased β(1, 3)-glucan exposure (unmasking) display increased immunostimulatory capabilities in vitro and attenuated virulence during systemic infection in mice. However, little work has been done to assess the impact of increased unmasking during the two most common manifestations of candidiasis, namely, oropharyngeal candidiasis (OPC) and vulvovaginal candidiasis (VVC). We have shown previously that the expression of a single hyperactive allele of the MAP3K STE11ΔN467 induces unmasking via the Cek1 MAPK pathway, attenuates fungal burden, and prolongs survival during systemic infection in mice. Here, we expand on these findings and show that infection with an unmasked STE11ΔN467 mutant also impacts disease progression during OPC and VVC murine infection models. Male mice sublingually infected with the STE11ΔN467 mutant showed a significant reduction in tongue fungal burden at 2 days postinfection and a modest reduction at 5 days postinfection. However, we find that selection for STE11ΔN467 suppressor mutants that no longer display increased unmasking occurs within the oral cavity and is likely responsible for the restoration of fungal burden trends to wild-type levels later in the infection. In the VVC infection model, no attenuation in fungal burden was observed. However, polymorphonuclear cell recruitment and interleukin-1β (IL-1β) levels within the vaginal lumen, markers of immunopathogenesis, were increased in mice infected with unmasked STE11ΔN467 cells. Thus, our data suggest a niche-specific impact for unmasking on disease progression.
Collapse
Affiliation(s)
- Andrew S. Wagner
- Department of Microbiology, University of Tennessee, Knoxville, Tennessee, USA
| | - Amanda K. Vogel
- Integrated Program in Biomedical Sciences, College of Graduate Health Sciences, University of Tennessee Health Science Center, Memphis, Tennessee, USA
| | | | - Elise K. Phillips
- Department of Microbiology, University of Tennessee, Knoxville, Tennessee, USA
| | - Hubertine M. E. Willems
- Department of Clinical Pharmacy and Translational Science, College of Pharmacy, University of Tennessee Health Science Center, Memphis, Tennessee, USA
| | - Brian M. Peters
- Department of Clinical Pharmacy and Translational Science, College of Pharmacy, University of Tennessee Health Science Center, Memphis, Tennessee, USA
| | - Todd B. Reynolds
- Department of Microbiology, University of Tennessee, Knoxville, Tennessee, USA
| |
Collapse
|
15
|
Liu X, Luo D, Guan J, Chen J, Xu X. Mushroom polysaccharides with potential in anti-diabetes: Biological mechanisms, extraction, and future perspectives: A review. Front Nutr 2022; 9:1087826. [PMID: 36590224 PMCID: PMC9794872 DOI: 10.3389/fnut.2022.1087826] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2022] [Accepted: 11/29/2022] [Indexed: 12/15/2022] Open
Abstract
Diabetes mellitus (DM) is a global health threat. Searching for anti-diabetic components from natural resources is of intense interest to scientists. Mushroom polysaccharides have received growing attention in anti-diabetes fields due to their advantages in broad resources, structure diversity, and multiple bioactivities, which are considered an unlimited source of healthy active components potentially applied in functional foods and nutraceuticals. In this review, the current knowledge about the roles of oxidative stress in the pathogenesis of DM, the extraction method of mushroom polysaccharides, and their potential biological mechanisms associated with anti-diabetes, including antioxidant, hypolipidemic, anti-inflammatory, and gut microbiota modulatory actions, were summarized based on a variety of in vitro and in vivo studies, with aiming at better understanding the roles of mushroom polysaccharides in the prevention and management of DM and its complications. Finally, future perspectives including bridging the gap between the intervention of mushroom polysaccharides and the modulation of insulin signaling pathway, revealing structure-bioactivity of mushroom polysaccharides, developing synergistic foods, conducting well-controlled clinical trials that may be very helpful in discovering valuable mushroom polysaccharides and better applications of mushroom polysaccharides in diabetic control were proposed.
Collapse
Affiliation(s)
| | | | | | | | - Xiaofei Xu
- College of Food Science and Engineering, Guangdong Ocean University, Yangjiang, China
| |
Collapse
|
16
|
Wagner AS, Lumsdaine SW, Mangrum MM, King AE, Hancock TJ, Sparer TE, Reynolds TB. Cek1 regulates ß(1,3)-glucan exposure through calcineurin effectors in Candida albicans. PLoS Genet 2022; 18:e1010405. [PMID: 36121853 PMCID: PMC9521907 DOI: 10.1371/journal.pgen.1010405] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2022] [Revised: 09/29/2022] [Accepted: 08/30/2022] [Indexed: 11/19/2022] Open
Abstract
In order to successfully induce disease, the fungal pathogen Candida albicans regulates exposure of antigens like the cell wall polysaccharide ß(1,3)-glucan to the host immune system. C. albicans covers (masks) ß(1,3)-glucan with a layer of mannosylated glycoproteins, which aids in immune system evasion by acting as a barrier to recognition by host pattern recognition receptors. Consequently, enhanced ß(1,3)-glucan exposure (unmasking) makes fungal cells more visible to host immune cells and facilitates more robust fungal clearance. However, an understanding of how C. albicans regulates its exposure levels of ß(1,3)-glucan is needed to leverage this phenotype. Signal transduction pathways and their corresponding effector genes mediating these changes are only beginning to be defined. Here, we report that the phosphatase calcineurin mediates unmasking of ß(1,3)-glucan in response to inputs from the Cek1 MAPK pathway and in response to caspofungin exposure. In contrast, calcineurin reduces ß-glucan exposure in response to high levels of extracellular calcium. Thus, depending on the input, calcineurin acts as a switchboard to regulate ß(1,3)-glucan exposure levels. By leveraging these differential ß(1,3)-glucan exposure phenotypes, we identified two novel effector genes in the calcineurin regulon, FGR41 and C1_11990W_A, that encode putative cell wall proteins and mediate masking/unmasking. Loss of either effector caused unmasking and attenuated virulence during systemic infection in mice. Furthermore, immunosuppression restored the colonization decrease seen in mice infected with the fgr41Δ/Δ mutant to wild-type levels, demonstrating a reliance on the host immune system for virulence attenuation. Thus, calcineurin and its downstream regulon are general regulators of unmasking.
Collapse
Affiliation(s)
- Andrew S. Wagner
- Department of Microbiology, University of Tennessee at Knoxville, Knoxville, Tennessee, United States of America
| | - Stephen W. Lumsdaine
- Department of Microbiology, University of Tennessee at Knoxville, Knoxville, Tennessee, United States of America
| | - Mikayla M. Mangrum
- Department of Microbiology, University of Tennessee at Knoxville, Knoxville, Tennessee, United States of America
| | - Ainsley E. King
- Department of Microbiology, University of Tennessee at Knoxville, Knoxville, Tennessee, United States of America
| | - Trevor J. Hancock
- Department of Microbiology, University of Tennessee at Knoxville, Knoxville, Tennessee, United States of America
| | - Timothy E. Sparer
- Department of Microbiology, University of Tennessee at Knoxville, Knoxville, Tennessee, United States of America
| | - Todd B. Reynolds
- Department of Microbiology, University of Tennessee at Knoxville, Knoxville, Tennessee, United States of America
| |
Collapse
|
17
|
Lachat J, Pascault A, Thibaut D, Le Borgne R, Verbavatz JM, Weiner A. Trans-cellular tunnels induced by the fungal pathogen Candida albicans facilitate invasion through successive epithelial cells without host damage. Nat Commun 2022; 13:3781. [PMID: 35773250 PMCID: PMC9246882 DOI: 10.1038/s41467-022-31237-z] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2021] [Accepted: 06/09/2022] [Indexed: 11/09/2022] Open
Abstract
The opportunistic fungal pathogen Candida albicans is normally commensal, residing in the mucosa of most healthy individuals. In susceptible hosts, its filamentous hyphal form can invade epithelial layers leading to superficial or severe systemic infection. Although invasion is mainly intracellular, it causes no apparent damage to host cells at early stages of infection. Here, we investigate C. albicans invasion in vitro using live-cell imaging and the damage-sensitive reporter galectin-3. Quantitative single cell analysis shows that invasion can result in host membrane breaching at different stages and host cell death, or in traversal of host cells without membrane breaching. Membrane labelling and three-dimensional 'volume' electron microscopy reveal that hyphae can traverse several host cells within trans-cellular tunnels that are progressively remodelled and may undergo 'inflations' linked to host glycogen stores. Thus, C. albicans early invasion of epithelial tissues can lead to either host membrane breaching or trans-cellular tunnelling.
Collapse
Affiliation(s)
- Joy Lachat
- Sorbonne Université, Inserm, CNRS, Centre d'Immunologie et des Maladies Infectieuses, Cimi-Paris, 75013, Paris, France
| | - Alice Pascault
- Sorbonne Université, Inserm, CNRS, Centre d'Immunologie et des Maladies Infectieuses, Cimi-Paris, 75013, Paris, France
| | - Delphine Thibaut
- Sorbonne Université, Inserm, CNRS, Centre d'Immunologie et des Maladies Infectieuses, Cimi-Paris, 75013, Paris, France
| | - Rémi Le Borgne
- Université Paris Cité, CNRS, Institut Jacques Monod, 75013, Paris, France
| | | | - Allon Weiner
- Sorbonne Université, Inserm, CNRS, Centre d'Immunologie et des Maladies Infectieuses, Cimi-Paris, 75013, Paris, France.
| |
Collapse
|
18
|
Ishibashi KI, Onaka N, Nishida N, Takahashi M, Adachi Y, Ohno N. Dectin-1 reactivity to paramylon derived from Euglena gracilis EOD-1. Biol Pharm Bull 2022; 45:1394-1397. [PMID: 35753759 DOI: 10.1248/bpb.b22-00247] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Euglena gracilis is a microalga that has recently attracted attention because of its bioactivities. Paramylon (PM), a major β-1,3-glucan, constitutes 70%-80% of the cells of the E. gracilis EOD-1 strain. Dectin-1 is a pattern recognition receptor that recognizes β-glucan. However, it is unclear whether PM binds to dectin-1. In this study, we investigated the reactivity of EOD1PM with dectin-1 by analyzing the binding of soluble murine and human dectin-1-Fc fusion protein (m dectin-1 Fc, h dectin-1 Fc) to EOD1PM using flow cytometry and ELISA.m dectin-1 Fc bound to EOD1PM particles when m dectin-1-Fc is added. Furthermore, the binding specificity was examined in a competitive reaction following addition of a soluble antigen. It was found that the binding of m dectin-1-Fc to EOD1PM was not inhibited by the addition of dextran or ovalbumin but by the addition of solubilized EOD1PM or Candida cell wall- solubilized β-glucan. In addition, the h dectin-1-Fc fusion protein was found to specifically bind to EOD1PM. These results suggest that dectin-1 recognizes and binds to the β-glucan structure of EOD1PM.Dectin-1 is expressed in leukocytes as a β-glucan receptor and is involved in the expression of various biological activities; therefore, the dectin-1 pathway may be involved in the biological activity of EOD1PM.
Collapse
Affiliation(s)
- Ken-Ichi Ishibashi
- Laboratory of Host Defense and Responses, Faculty of Nutrition, Kagawa Nutrition University.,Laboratory for Immunopharmacology of Microbial Products, School of Pharmacy, Tokyo University of Pharmacy and Life Sciences
| | | | | | | | - Yoshiyuki Adachi
- Laboratory for Immunopharmacology of Microbial Products, School of Pharmacy, Tokyo University of Pharmacy and Life Sciences
| | - Naohito Ohno
- Laboratory for Immunopharmacology of Microbial Products, School of Pharmacy, Tokyo University of Pharmacy and Life Sciences
| |
Collapse
|
19
|
Gairola A, Benjamin A, Weatherston JD, Cirillo JD, Wu HJ. Recent Developments in Drug Delivery for Treatment of Tuberculosis by Targeting Macrophages. ADVANCED THERAPEUTICS 2022; 5:2100193. [PMID: 36203881 PMCID: PMC9531895 DOI: 10.1002/adtp.202100193] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2021] [Indexed: 11/10/2022]
Abstract
Tuberculosis (TB) is among the greatest public health and safety concerns in the 21st century, Mycobacterium tuberculosis, which causes TB, infects alveolar macrophages and uses these cells as one of its primary sites of replication. The current TB treatment regimen, which consist of chemotherapy involving a combination of 3-4 antimicrobials for a duration of 6-12 months, is marked with significant side effects, toxicity, and poor compliance. Targeted drug delivery offers a strategy that could overcome many of the problems of current TB treatment by specifically targeting infected macrophages. Recent advances in nanotechnology and material science have opened an avenue to explore drug carriers that actively and passively target macrophages. This approach can increase the drug penetration into macrophages by using ligands on the nanocarrier that interact with specific receptors for macrophages. This review encompasses the recent development of drug carriers specifically targeting macrophages actively and passively. Future directions and challenges associated with development of effective TB treatment is also discussed.
Collapse
Affiliation(s)
- Anirudh Gairola
- Department of Chemical Engineering, Texas A&M University, College Station, Texas, USA
| | - Aaron Benjamin
- Department of Microbial Pathogenesis and Immunology, Texas A&M University Health Science Center, Bryan, Texas, USA
| | - Joshua D Weatherston
- Department of Chemical Engineering, Texas A&M University, College Station, Texas, USA
| | - Jeffrey D Cirillo
- Department of Microbial Pathogenesis and Immunology, Texas A&M University Health Science Center, Bryan, Texas, USA
| | - Hung-Jen Wu
- Department of Chemical Engineering, Texas A&M University, College Station, Texas, USA
| |
Collapse
|
20
|
Chowdhury AS, Geetha Bai R, Islam T, Abir M, Narayan M, Khatun Z, Nurunnabi M. Bile acid linked β-glucan nanoparticles for liver specific oral delivery of biologics. Biomater Sci 2022; 10:2929-2939. [PMID: 35471198 PMCID: PMC9949325 DOI: 10.1039/d2bm00316c] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Oral delivery remains one of the most convenient routes for drug administration compared to intravenous, intramuscular, and via suppositories. However, due to the risk of degradation, and proteolysis of molecules in the acidic gastric medium, as well as the difficulty of transporting large molecules through the intestinal membrane, more than half of the therapeutic molecules are prohibited for oral administration. Moreover, most of the large molecules and biological therapeutics are not available in oral dosage form due to their instability in the stomach and inability of intestinal absorption. To achieve expected bioavailability, an orally administered therapeutic molecule must be protected within the stomach, and transportation facilitated via the small intestine. In this project, we have introduced a hybrid carrier, composed of Taurocholic Acid (TA) and β-Glucan (TAG), that is shown to be effective for the simultaneous protection of the biologics in acidic buffer and simulated gastric juice as well as facilitate enhanced absorption and transportation via the small intestine. In this project, we have used an eGFP encoded plasmid as a model biologic to prepare particles mediated with TAG. TAG show the potential of enhancing transfection and expression of eGFP as we have observed two fold higher expression in the cell upon coincubation for 4 h. In vivo studies on orally dosed mice showed that eGFP expression in the liver was significantly higher in TAG containing particles compared to particles without TAG. The findings suggest that the TAG carrier is capable of not only preserving biologics but also transporting them more efficiently to the liver. As a result, this strategy can be employed for a variety of liver-targeted therapeutic delivery to treat a variety of liver diseases.
Collapse
Affiliation(s)
- Ayreen S Chowdhury
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Texas at El Paso, El Paso, TX 79902, USA.
- Department of Bioscience, School of Science and Technology, Nottingham Trent university, Nottingham, NG11 8NS, UK
| | - Renu Geetha Bai
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Texas at El Paso, El Paso, TX 79902, USA.
- School of Natural Sciences and Health, Tallinn University, Tallinn, 10120, Estonia
| | - Tamanna Islam
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Texas at El Paso, El Paso, TX 79902, USA.
| | - Muhammad Abir
- Aerospace Center (cSETR), University of Texas at El Paso, El Paso, TX 79965, USA
| | - Mahesh Narayan
- Department of Chemistry and Biochemistry, University of Texas at El Paso, El paso, TX 79965, USA
| | - Zehedina Khatun
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Texas at El Paso, El Paso, TX 79902, USA.
| | - Md Nurunnabi
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Texas at El Paso, El Paso, TX 79902, USA.
- Aerospace Center (cSETR), University of Texas at El Paso, El Paso, TX 79965, USA
- Biomedical Engineering, College of Engineering, University of Texas at El Paso, El Paso, TX 79965, USA
| |
Collapse
|
21
|
Li M, Zhang R, Li J, Li J. The Role of C-Type Lectin Receptor Signaling in the Intestinal Microbiota-Inflammation-Cancer Axis. Front Immunol 2022; 13:894445. [PMID: 35619716 PMCID: PMC9127077 DOI: 10.3389/fimmu.2022.894445] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2022] [Accepted: 04/04/2022] [Indexed: 12/13/2022] Open
Abstract
As a subset of pattern recognition receptors (PRRs), C-type lectin-like receptors (CLRs) are mainly expressed by myeloid cells as both transmembrane and soluble forms. CLRs recognize not only pathogen associated molecular patterns (PAMPs), but also damage-associated molecular patterns (DAMPs) to promote innate immune responses and affect adaptive immune responses. Upon engagement by PAMPs or DAMPs, CLR signaling initiates various biological activities in vivo, such as cytokine secretion and immune cell recruitment. Recently, several CLRs have been implicated as contributory to the pathogenesis of intestinal inflammation, which represents a prominent risk factor for colorectal cancer (CRC). CLRs function as an interface among microbiota, intestinal epithelial barrier and immune system, so we firstly discussed the relationship between dysbiosis caused by microbiota alteration and inflammatory bowel disease (IBD), then focused on the role of CLRs signaling in pathogenesis of IBD (including Mincle, Dectin-3, Dectin-1, DCIR, DC-SIGN, LOX-1 and their downstream CARD9). Given that CLRs mediate intricate inflammatory signals and inflammation plays a significant role in tumorigenesis, we finally highlight the specific effects of CLRs on CRC, especially colitis-associated cancer (CAC), hoping to open new horizons on pathogenesis and therapeutics of IBD and CAC.
Collapse
Affiliation(s)
- Muhan Li
- Department of Gastroenterology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.,Key Laboratory of Gut Microbiota Translational Medicine Research, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Runfeng Zhang
- Department of Gastroenterology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.,Key Laboratory of Gut Microbiota Translational Medicine Research, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Ji Li
- Department of Gastroenterology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.,Key Laboratory of Gut Microbiota Translational Medicine Research, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Jingnan Li
- Department of Gastroenterology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.,Key Laboratory of Gut Microbiota Translational Medicine Research, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| |
Collapse
|
22
|
Gutierrez MW, van Tilburg Bernardes E, Changirwa D, McDonald B, Arrieta MC. "Molding" immunity-modulation of mucosal and systemic immunity by the intestinal mycobiome in health and disease. Mucosal Immunol 2022; 15:573-583. [PMID: 35474360 DOI: 10.1038/s41385-022-00515-w] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Revised: 03/24/2022] [Accepted: 04/04/2022] [Indexed: 02/04/2023]
Abstract
Fungi are important yet understudied contributors to the microbial communities of the gastrointestinal tract. Starting at birth, the intestinal mycobiome undergoes a period of dynamic maturation under the influence of microbial, host, and extrinsic influences, with profound functional implications for immune development in early life, and regulation of immune homeostasis throughout life. Candida albicans serves as a model organism for understanding the cross-talk between fungal colonization dynamics and immunity, and exemplifies unique mechanisms of fungal-immune interactions, including fungal dimorphism, though our understanding of other intestinal fungi is growing. Given the prominent role of the gut mycobiome in promoting immune homeostasis, emerging evidence points to fungal dysbiosis as an influential contributor to immune dysregulation in a variety of inflammatory and infectious diseases. Here we review current knowledge on the factors that govern host-fungi interactions in the intestinal tract and immunological outcomes in both mucosal and systemic compartments.
Collapse
Affiliation(s)
- Mackenzie W Gutierrez
- Immunology Research Group, Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada.,Department of Pediatrics, Alberta Children's Hospital Research Institute, University of Calgary, Calgary, AB, Canada.,Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada.,International Microbiome Centre, University of Calgary, Calgary, AB, Canada
| | - Erik van Tilburg Bernardes
- Immunology Research Group, Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada.,Department of Pediatrics, Alberta Children's Hospital Research Institute, University of Calgary, Calgary, AB, Canada.,Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada.,International Microbiome Centre, University of Calgary, Calgary, AB, Canada
| | - Diana Changirwa
- Immunology Research Group, Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada.,International Microbiome Centre, University of Calgary, Calgary, AB, Canada.,Department of Critical Care Medicine, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Braedon McDonald
- Immunology Research Group, Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada.,International Microbiome Centre, University of Calgary, Calgary, AB, Canada.,Department of Critical Care Medicine, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Marie-Claire Arrieta
- Immunology Research Group, Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada. .,Department of Pediatrics, Alberta Children's Hospital Research Institute, University of Calgary, Calgary, AB, Canada. .,Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada. .,International Microbiome Centre, University of Calgary, Calgary, AB, Canada.
| |
Collapse
|
23
|
Goldstein ME, Scull MA. Modeling Innate Antiviral Immunity in Physiological Context. J Mol Biol 2022; 434:167374. [PMID: 34863779 PMCID: PMC8940657 DOI: 10.1016/j.jmb.2021.167374] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2021] [Revised: 11/14/2021] [Accepted: 11/15/2021] [Indexed: 12/16/2022]
Abstract
An effective innate antiviral response is critical for the mitigation of severe disease and host survival following infection. In vivo, the innate antiviral response is triggered by cells that detect the invading pathogen and then communicate through autocrine and paracrine signaling to stimulate the expression of genes that inhibit viral replication, curtail cell proliferation, or modulate the immune response. In other words, the innate antiviral response is complex and dynamic. Notably, in the laboratory, culturing viruses and assaying viral life cycles frequently utilizes cells that are derived from tissues other than those that support viral replication during natural infection, while the study of viral pathogenesis often employs animal models. In recapitulating the human antiviral response, it is important to consider that variation in the expression and function of innate immune sensors and antiviral effectors exists across species, cell types, and cell differentiation states, as well as when cells are placed in different contexts. Thus, to gain novel insight into the dynamics of the host response and how specific sensors and effectors impact infection kinetics by a particular virus, the model system must be selected carefully. In this review, we briefly introduce key signaling pathways involved in the innate antiviral response and highlight how these differ between systems. We then review the application of tissue-engineered or 3D models for studying the antiviral response, and suggest how these in vitro culture systems could be further utilized to assay physiologically-relevant host responses and reveal novel insight into virus-host interactions.
Collapse
Affiliation(s)
- Monty E Goldstein
- Department of Cell Biology and Molecular Genetics, Maryland Pathogen Research Institute, 3134 Bioscience Research Building, University of Maryland, College Park, MD 20742, USA
| | - Margaret A Scull
- Department of Cell Biology and Molecular Genetics, Maryland Pathogen Research Institute, 3134 Bioscience Research Building, University of Maryland, College Park, MD 20742, USA.
| |
Collapse
|
24
|
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: 2.5] [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
|
25
|
Mata-Martínez P, Bergón-Gutiérrez M, del Fresno C. Dectin-1 Signaling Update: New Perspectives for Trained Immunity. Front Immunol 2022; 13:812148. [PMID: 35237264 PMCID: PMC8882614 DOI: 10.3389/fimmu.2022.812148] [Citation(s) in RCA: 58] [Impact Index Per Article: 29.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2021] [Accepted: 01/20/2022] [Indexed: 12/12/2022] Open
Abstract
The C-type lectin receptor Dectin-1 was originally described as the β-glucan receptor expressed in myeloid cells, with crucial functions in antifungal responses. However, over time, different ligands both of microbial-derived and endogenous origin have been shown to be recognized by Dectin-1. The outcomes of this recognition are diverse, including pro-inflammatory responses such as cytokine production, reactive oxygen species generation and phagocytosis. Nonetheless, tolerant responses have been also attributed to Dectin-1, depending on the specific ligand engaged. Dectin-1 recognition of their ligands triggers a plethora of downstream signaling pathways, with complex interrelationships. These signaling routes can be modulated by diverse factors such as phosphatases or tetraspanins, resulting either in pro-inflammatory or regulatory responses. Since its first depiction, Dectin-1 has recently gained a renewed attention due to its role in the induction of trained immunity. This process of long-term memory of innate immune cells can be triggered by β-glucans, and Dectin-1 is crucial for its initiation. The main signaling pathways involved in this process have been described, although the understanding of the above-mentioned complexity in the β-glucan-induced trained immunity is still scarce. In here, we have reviewed and updated all these factors related to the biology of Dectin-1, highlighting the gaps that deserve further research. We believe on the relevance to fully understand how this receptor works, and therefore, how we could harness it in different pathological conditions as diverse as fungal infections, autoimmunity, or cancer.
Collapse
Affiliation(s)
| | | | - Carlos del Fresno
- Immune response and Immunomodulation Group, Hospital La Paz Institute for Health Research (IdiPAZ), Madrid, Spain
| |
Collapse
|
26
|
Yong HM, Gour N, Sharma D, Khalil SM, Lane AP, Lajoie S. Epigenetic regulation of epithelial dectin-1 through an IL-33-STAT3 axis in allergic disease. Allergy 2022; 77:207-217. [PMID: 33982290 PMCID: PMC10580706 DOI: 10.1111/all.14898] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2020] [Revised: 04/10/2021] [Accepted: 04/14/2021] [Indexed: 12/15/2022]
Abstract
Allergic diseases arise in susceptible individuals in part because of decrements in protective pathways. The mechanism by which these anti-inflammatory molecules become repressed remains unclear. We have previously reported that epithelial dectin-1 prevents aberrant type 2 responses and is downregulated in the epithelium of allergic patients. Here, we report that dectin-1 is constitutively expressed by the respiratory epithelium in humans and that IL-33 specifically acts as a repressor of dectin-1. Mechanistically, this occurs via IL-33-dependent STAT3 activation and the subsequent repression of the dectin-1 gene, CLEC7A. We have identified a novel enhancer region upstream of the proximal promoter of CLEC7A that is only accessible in epithelial cells, but not in hematopoietic cells. Epigenetic repression of CLEC7A through this newly identified locus, downstream of an aberrant IL-33-STAT3 axis, occurs in the epithelium of allergic individuals. Collectively, our data identify a mechanism of epigenetic fine-tuning of dectin-1 expression in epithelial cells that may participate in allergenicity.
Collapse
Affiliation(s)
- Hwan Mee Yong
- Department of Otolaryngology, Johns Hopkins School of Medicine, Baltimore, MD
- Department of Environmental Health and Engineering, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD
| | - Naina Gour
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins School of Medicine, Baltimore, MD
| | - Deepika Sharma
- Department of Otolaryngology, Johns Hopkins School of Medicine, Baltimore, MD
| | - Syed Muaz Khalil
- Department of Otolaryngology, Johns Hopkins School of Medicine, Baltimore, MD
| | - Andrew P. Lane
- Department of Environmental Health and Engineering, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD
| | - Stephane Lajoie
- Department of Otolaryngology, Johns Hopkins School of Medicine, Baltimore, MD
- Department of Environmental Health and Engineering, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD
| |
Collapse
|
27
|
Peng Y, Chen Y, Ma J, Zhou W, Wang Y, Wang Y, Zheng H, Shi W. Role and mechanism of the Dectin-1-mediated Syk/NF-κB signaling pathway in Talaromyces marneffei infection. Exp Ther Med 2021; 23:84. [PMID: 34938366 PMCID: PMC8688926 DOI: 10.3892/etm.2021.11007] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2021] [Accepted: 11/05/2021] [Indexed: 12/13/2022] Open
Abstract
Dendritic cell-associated C-type lectin-1 (Dectin-1), a C-type lectin receptor, serves a critical role in host antifungal immunity. However, the molecular mechanism and function of Dectin-1-mediated signaling in response to infection by the pathogenic fungus Talaromyces marneffei remains unclear. To understand the role of Dectin-1 signaling against T. marneffei infection, the phosphorylation of spleen tyrosine kinase (Syk), nuclear factor of κ light polypeptide gene enhancer in B-cells inhibitor, α (IκBα) and NF-κB were analyzed using western blotting, and the secretion of cytokines was detected using ELISA. Upon sporular or hyphal heat-killed T. marneffei stimulation, Dectin-1 in THP-1 macrophages recognized and induced the activation of Syk, and in turn triggered phosphorylation of downstream molecules IκBα and NF-κB, thus increasing the secretion of TNF-α and IL-8. Conversely, knockdown of Dectin-1 in THP-1 macrophages downregulated the phosphorylation of Syk, IκBα and NF-κB molecules, and significantly decreased the production of TNF-α and IL-8. These results indicated that Dectin-1 may have a crucial role in inducing the inflammatory response via increasing levels of TNF-α and IL-8 induced by T. marneffei, whereas NF-κB may be the key downstream molecule involved in the response to T. marneffei infection. Subsequently, THP-1 macrophages could orchestrate the innate immune system by releasing the cytokines TNF-α and IL-8. Therefore, it was hypothesized that regulation of the Dectin-1 signaling pathway may effectively interfere with the defense ability of the host against T. marneffei infection.
Collapse
Affiliation(s)
- Yang Peng
- Department of Clinical Laboratory, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu 213003, P.R. China
| | - Yan Chen
- Department of Clinical Laboratory, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu 213003, P.R. China
| | - Jinhong Ma
- Department of Clinical Laboratory, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu 213003, P.R. China
| | - Wei Zhou
- Department of Clinical Laboratory, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu 213003, P.R. China
| | - Yangyang Wang
- Department of Clinical Laboratory, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu 213003, P.R. China
| | - Yuyue Wang
- Department of Clinical Laboratory, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu 213003, P.R. China
| | - Hui Zheng
- Institutes of Biology and Medical Sciences, Soochow University, Suzhou, Jiangsu 215000, P.R. China
| | - Weifeng Shi
- Department of Clinical Laboratory, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu 213003, P.R. China
| |
Collapse
|
28
|
Stappers MH, Nikolakopoulou C, Wiesner DL, Yuecel R, Klein BS, Willment JA, Brown GD. Characterization of antifungal C-type lectin receptor expression on murine epithelial and endothelial cells in mucosal tissues. Eur J Immunol 2021; 51:2341-2344. [PMID: 34114658 PMCID: PMC8593890 DOI: 10.1002/eji.202149192] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2021] [Revised: 04/19/2021] [Accepted: 06/10/2021] [Indexed: 11/06/2022]
Abstract
Our data reveal that selection of enzymes for generating single cell suspensions from murine tissues influences detection of surface expression of antifungal CLRs. Using a method that most preserves receptor expression, we show that non-myeloid expression of antifungal CLRs is limited to MelLec on endothelial cells in murine mucosal tissues.
Collapse
Affiliation(s)
- Mark H.T. Stappers
- Aberdeen Fungal GroupInstitute of Medical SciencesUniversity of AberdeenAberdeenUK
- Department of BiosciencesMedical Research Council Centre for Medical Mycology at the University of ExeterExeterUK
| | - Christina Nikolakopoulou
- Aberdeen Fungal GroupInstitute of Medical SciencesUniversity of AberdeenAberdeenUK
- Department of BiosciencesMedical Research Council Centre for Medical Mycology at the University of ExeterExeterUK
| | - Darin L. Wiesner
- Department of PediatricsSchool of Medicine and Public HealthUniversity of Wisconsin‐MadisonMadisonWIUSA
| | - Raif Yuecel
- Iain Fraser Cytometry CentreInstitute of Medical SciencesUniversity of AberdeenAberdeenUK
- Exeter Centre for Cytomics (EXCC)Department of BiosciencesCollege of Life and Environmental SciencesUniversity of ExeterExeterEX4 4QDUK
| | - Bruce S. Klein
- Department of PediatricsSchool of Medicine and Public HealthUniversity of Wisconsin‐MadisonMadisonWIUSA
| | - Janet A. Willment
- Aberdeen Fungal GroupInstitute of Medical SciencesUniversity of AberdeenAberdeenUK
- Department of BiosciencesMedical Research Council Centre for Medical Mycology at the University of ExeterExeterUK
| | - Gordon D. Brown
- Aberdeen Fungal GroupInstitute of Medical SciencesUniversity of AberdeenAberdeenUK
- Department of BiosciencesMedical Research Council Centre for Medical Mycology at the University of ExeterExeterUK
| |
Collapse
|
29
|
Zajta E, Csonka K, Tóth A, Tiszlavicz L, Németh T, Orosz A, Novák Á, Csikós M, Vágvölgyi C, Mócsai A, Gácser A. Signaling through Syk or CARD9 Mediates Species-Specific Anti- Candida Protection in Bone Marrow Chimeric Mice. mBio 2021; 12:e0160821. [PMID: 34465030 PMCID: PMC8406149 DOI: 10.1128/mbio.01608-21] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2021] [Accepted: 08/05/2021] [Indexed: 01/12/2023] Open
Abstract
The spleen tyrosine kinase (Syk) and the downstream adaptor protein CARD9 are crucial signaling molecules in antimicrobial immunity. Candida parapsilosis is an emerging fungal pathogen with a high incidence in neonates, while Candida albicans is the most common agent of candidiasis. While signaling through Syk/CARD9 promotes protective host mechanisms in response to C. albicans, its function in immunity against C. parapsilosis remains unclear. Here, we generated Syk-/- and CARD9-/- bone marrow chimeric mice to study the role of Syk/CARD9 signaling in immune responses to C. parapsilosis compared to C. albicans. We demonstrate various functions of this pathway (e.g., phagocytosis, phagosome acidification, and killing) in Candida-challenged, bone marrow-derived macrophages with differential involvement of Syk and CARD9 along with species-specific differences in cytokine production. We report that Syk-/- or CARD9-/- chimeras rapidly display high susceptibility to C. albicans, while C. parapsilosis infection exacerbates over a prolonged period in these animals. Thus, our results establish that Syk and CARD9 contribute to systemic resistance to C. parapsilosis and C. albicans differently. Additionally, we confirm prior studies but also detail new insights into the fundamental roles of both proteins in immunity against C. albicans. Our data further suggest that Syk has a more prominent influence on anti-Candida immunity than CARD9. Therefore, this study reinforces the Syk/CARD9 pathway as a potential target for anti-Candida immune therapy. IMPORTANCE While C. albicans remains the most clinically significant Candida species, C. parapsilosis is an emerging pathogen with increased affinity to neonates. Syk/CARD9 signaling is crucial in immunity to C. albicans, but its role in in vivo responses to other pathogenic Candida species is largely unexplored. We used mice with hematopoietic systems deficient in Syk or CARD9 to comparatively study the function of these proteins in anti-Candida immunity. We demonstrate that Syk/CARD9 signaling has a protective role against C. parapsilosis differently than against C. albicans. Thus, this study is the first to reveal that Syk can exert immune responses during systemic Candida infections species specifically. Additionally, Syk-dependent immunity to a nonalbicans Candida species in an in vivo murine model has not been reported previously. We highlight that the contribution of Syk and CARD9 to fungal infections are not identical and underline this pathway as a promising immune-therapeutic target to fight Candida infections.
Collapse
Affiliation(s)
- Erik Zajta
- HCEMM-USZ Fungal Pathogens Research Group, Department of Microbiology, Faculty of Science and Informatics, University of Szeged, Szeged, Hungary
| | - Katalin Csonka
- HCEMM-USZ Fungal Pathogens Research Group, Department of Microbiology, Faculty of Science and Informatics, University of Szeged, Szeged, Hungary
| | - Adél Tóth
- HCEMM-USZ Fungal Pathogens Research Group, Department of Microbiology, Faculty of Science and Informatics, University of Szeged, Szeged, Hungary
| | | | - Tamás Németh
- Department of Physiology, Semmelweis University School of Medicine, Budapest, Hungary
- Department of Rheumatology and Clinical Immunology, Semmelweis University, Budapest, Hungary
| | - Anita Orosz
- Department of Physiology, Semmelweis University School of Medicine, Budapest, Hungary
| | - Ádám Novák
- HCEMM-USZ Fungal Pathogens Research Group, Department of Microbiology, Faculty of Science and Informatics, University of Szeged, Szeged, Hungary
| | - Máté Csikós
- HCEMM-USZ Fungal Pathogens Research Group, Department of Microbiology, Faculty of Science and Informatics, University of Szeged, Szeged, Hungary
| | - Csaba Vágvölgyi
- HCEMM-USZ Fungal Pathogens Research Group, Department of Microbiology, Faculty of Science and Informatics, University of Szeged, Szeged, Hungary
| | - Attila Mócsai
- Department of Physiology, Semmelweis University School of Medicine, Budapest, Hungary
| | - Attila Gácser
- HCEMM-USZ Fungal Pathogens Research Group, Department of Microbiology, Faculty of Science and Informatics, University of Szeged, Szeged, Hungary
- MTA-SZTE “Lendület” Mycobiome Research Group, University of Szeged, Szeged, Hungary
| |
Collapse
|
30
|
Gong W, Liu P, Zheng T, Wu X, Zhao Y, Ren J. The ubiquitous role of spleen tyrosine kinase (Syk) in gut diseases: From mucosal immunity to targeted therapy. Int Rev Immunol 2021; 41:552-563. [PMID: 34355656 DOI: 10.1080/08830185.2021.1962860] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
Spleen tyrosine kinase (Syk) is a cytoplasmic non-receptor protein tyrosine kinase expressed in a variety of cells and play crucial roles in signal transduction. Syk mediates downstream signaling by recruiting to the dually phosphorylated immunoreceptor tyrosine-based activation motifs (ITAMs) of the transmembrane adaptor molecule or the receptor chain itself. In gut diseases, Syk is observed to be expressed in intestinal epithelial cells, monocytes/macrophages, dendritic cells and mast cells. Activation of Syk in these cells can modulate intestinal mucosal immune response by promoting inflammatory cytokines and chemokines production, thus regulating gut homeostasis. Due to the restriction of specificity and selectivity for the development of Syk inhibitors, only a few such inhibitors are available in gut diseases, including intestinal ischemia/reperfusion damage, infectious disease, inflammatory bowel disease, etc. The promising outcomes of Syk inhibitors from both preclinical and clinical studies have shown to attenuate the progression of gut diseases thereby indicating a great potential in the development of Syk targeted therapy for treatment of gut diseases. This review depicts the characterization of Syk, summarizes the signal pathways of Syk, and discusses its potential targeted therapy for gut diseases.
Collapse
Affiliation(s)
- Wenbin Gong
- School of Medicine, Research Institute of General Surgery, Southeast University, Jinling Hospital, Nanjing, P.R. China
| | - Peizhao Liu
- Research Institute of General Surgery, Jinling Hospital, Nanjing, P.R. China
| | - Tao Zheng
- Department of General Surgery, BenQ Medical Center, The Affiliated BenQ Hospital of Nanjing Medical University, Nanjing, P.R. China
| | - Xiuwen Wu
- Research Institute of General Surgery, Jinling Hospital, Nanjing, P.R. China
| | - Yun Zhao
- Department of General Surgery, BenQ Medical Center, The Affiliated BenQ Hospital of Nanjing Medical University, Nanjing, P.R. China
| | - Jianan Ren
- School of Medicine, Research Institute of General Surgery, Southeast University, Jinling Hospital, Nanjing, P.R. China.,Research Institute of General Surgery, Jinling Hospital, Nanjing, P.R. China
| |
Collapse
|
31
|
d'Enfert C, Kaune AK, Alaban LR, Chakraborty S, Cole N, Delavy M, Kosmala D, Marsaux B, Fróis-Martins R, Morelli M, Rosati D, Valentine M, Xie Z, Emritloll Y, Warn PA, Bequet F, Bougnoux ME, Bornes S, Gresnigt MS, Hube B, Jacobsen ID, Legrand M, Leibundgut-Landmann S, Manichanh C, Munro CA, Netea MG, Queiroz K, Roget K, Thomas V, Thoral C, Van den Abbeele P, Walker AW, Brown AJP. The impact of the Fungus-Host-Microbiota interplay upon Candida albicans infections: current knowledge and new perspectives. FEMS Microbiol Rev 2021; 45:fuaa060. [PMID: 33232448 PMCID: PMC8100220 DOI: 10.1093/femsre/fuaa060] [Citation(s) in RCA: 142] [Impact Index Per Article: 47.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2020] [Accepted: 11/18/2020] [Indexed: 12/11/2022] Open
Abstract
Candida albicans is a major fungal pathogen of humans. It exists as a commensal in the oral cavity, gut or genital tract of most individuals, constrained by the local microbiota, epithelial barriers and immune defences. Their perturbation can lead to fungal outgrowth and the development of mucosal infections such as oropharyngeal or vulvovaginal candidiasis, and patients with compromised immunity are susceptible to life-threatening systemic infections. The importance of the interplay between fungus, host and microbiota in driving the transition from C. albicans commensalism to pathogenicity is widely appreciated. However, the complexity of these interactions, and the significant impact of fungal, host and microbiota variability upon disease severity and outcome, are less well understood. Therefore, we summarise the features of the fungus that promote infection, and how genetic variation between clinical isolates influences pathogenicity. We discuss antifungal immunity, how this differs between mucosae, and how individual variation influences a person's susceptibility to infection. Also, we describe factors that influence the composition of gut, oral and vaginal microbiotas, and how these affect fungal colonisation and antifungal immunity. We argue that a detailed understanding of these variables, which underlie fungal-host-microbiota interactions, will present opportunities for directed antifungal therapies that benefit vulnerable patients.
Collapse
Affiliation(s)
- Christophe d'Enfert
- Unité Biologie et Pathogénicité Fongiques, Département de Mycologie, Institut Pasteur, USC 2019 INRA, 25, rue du Docteur Roux, 75015 Paris, France
| | - Ann-Kristin Kaune
- Aberdeen Fungal Group, Institute of Medical Sciences, University of Aberdeen, Ashgrove Road West, Foresterhill, Aberdeen AB25 2ZD, UK
| | - Leovigildo-Rey Alaban
- BIOASTER Microbiology Technology Institute, 40 avenue Tony Garnier, 69007 Lyon, France
- Université de Paris, Sorbonne Paris Cité, 25, rue du Docteur Roux, 75015 Paris, France
| | - Sayoni Chakraborty
- Microbial Immunology Research Group, Emmy Noether Junior Research Group Adaptive Pathogenicity Strategies, and the Department of Microbial Pathogenicity Mechanisms, Leibniz Institute for Natural Product Research and Infection Biology – Hans Knöll Institute, Beutenbergstraße 11a, 07745 Jena, Germany
- Institute of Microbiology, Friedrich Schiller University, Neugasse 25, 07743 Jena, Germany
| | - Nathaniel Cole
- Gut Microbiology Group, Rowett Institute, University of Aberdeen, Ashgrove Road West, Foresterhill, Aberdeen AB25 2ZD, UK
| | - Margot Delavy
- Unité Biologie et Pathogénicité Fongiques, Département de Mycologie, Institut Pasteur, USC 2019 INRA, 25, rue du Docteur Roux, 75015 Paris, France
- Université de Paris, Sorbonne Paris Cité, 25, rue du Docteur Roux, 75015 Paris, France
| | - Daria Kosmala
- Unité Biologie et Pathogénicité Fongiques, Département de Mycologie, Institut Pasteur, USC 2019 INRA, 25, rue du Docteur Roux, 75015 Paris, France
- Université de Paris, Sorbonne Paris Cité, 25, rue du Docteur Roux, 75015 Paris, France
| | - Benoît Marsaux
- ProDigest BV, Technologiepark 94, B-9052 Gent, Belgium
- Center for Microbial Ecology and Technology (CMET), Department of Biotechnology, Faculty of Bioscience Engineering, Ghent University, Coupure Links, 9000 Ghent, Belgium
| | - Ricardo Fróis-Martins
- Immunology Section, Vetsuisse Faculty, University of Zurich, Winterthurerstrasse 266a, Zurich 8057, Switzerland
- Institute of Experimental Immunology, University of Zurich, Winterthurerstrasse 190, Zürich 8057, Switzerland
| | - Moran Morelli
- Mimetas, Biopartner Building 2, J.H. Oortweg 19, 2333 CH Leiden, The Netherlands
| | - Diletta Rosati
- Department of Internal Medicine and Radboud Center for Infectious Diseases, Radboud University Medical Center, Geert Grooteplein 28, 6525 GA Nijmegen, The Netherlands
| | - Marisa Valentine
- Microbial Immunology Research Group, Emmy Noether Junior Research Group Adaptive Pathogenicity Strategies, and the Department of Microbial Pathogenicity Mechanisms, Leibniz Institute for Natural Product Research and Infection Biology – Hans Knöll Institute, Beutenbergstraße 11a, 07745 Jena, Germany
| | - Zixuan Xie
- Gut Microbiome Group, Vall d'Hebron Institut de Recerca (VHIR), Vall d'Hebron Hospital Universitari, Vall d'Hebron Barcelona Hospital Campus, Passeig Vall d'Hebron 119–129, 08035 Barcelona, Spain
| | - Yoan Emritloll
- Unité Biologie et Pathogénicité Fongiques, Département de Mycologie, Institut Pasteur, USC 2019 INRA, 25, rue du Docteur Roux, 75015 Paris, France
| | - Peter A Warn
- Magic Bullet Consulting, Biddlecombe House, Ugbrook, Chudleigh Devon, TQ130AD, UK
| | - Frédéric Bequet
- BIOASTER Microbiology Technology Institute, 40 avenue Tony Garnier, 69007 Lyon, France
| | - Marie-Elisabeth Bougnoux
- Unité Biologie et Pathogénicité Fongiques, Département de Mycologie, Institut Pasteur, USC 2019 INRA, 25, rue du Docteur Roux, 75015 Paris, France
| | - Stephanie Bornes
- Université Clermont Auvergne, INRAE, VetAgro Sup, UMRF0545, 20 Côte de Reyne, 15000 Aurillac, France
| | - Mark S Gresnigt
- Microbial Immunology Research Group, Emmy Noether Junior Research Group Adaptive Pathogenicity Strategies, and the Department of Microbial Pathogenicity Mechanisms, Leibniz Institute for Natural Product Research and Infection Biology – Hans Knöll Institute, Beutenbergstraße 11a, 07745 Jena, Germany
| | - Bernhard Hube
- Microbial Immunology Research Group, Emmy Noether Junior Research Group Adaptive Pathogenicity Strategies, and the Department of Microbial Pathogenicity Mechanisms, Leibniz Institute for Natural Product Research and Infection Biology – Hans Knöll Institute, Beutenbergstraße 11a, 07745 Jena, Germany
| | - Ilse D Jacobsen
- Microbial Immunology Research Group, Emmy Noether Junior Research Group Adaptive Pathogenicity Strategies, and the Department of Microbial Pathogenicity Mechanisms, Leibniz Institute for Natural Product Research and Infection Biology – Hans Knöll Institute, Beutenbergstraße 11a, 07745 Jena, Germany
| | - Mélanie Legrand
- Unité Biologie et Pathogénicité Fongiques, Département de Mycologie, Institut Pasteur, USC 2019 INRA, 25, rue du Docteur Roux, 75015 Paris, France
| | - Salomé Leibundgut-Landmann
- Immunology Section, Vetsuisse Faculty, University of Zurich, Winterthurerstrasse 266a, Zurich 8057, Switzerland
- Institute of Experimental Immunology, University of Zurich, Winterthurerstrasse 190, Zürich 8057, Switzerland
| | - Chaysavanh Manichanh
- Gut Microbiome Group, Vall d'Hebron Institut de Recerca (VHIR), Vall d'Hebron Hospital Universitari, Vall d'Hebron Barcelona Hospital Campus, Passeig Vall d'Hebron 119–129, 08035 Barcelona, Spain
| | - Carol A Munro
- Aberdeen Fungal Group, Institute of Medical Sciences, University of Aberdeen, Ashgrove Road West, Foresterhill, Aberdeen AB25 2ZD, UK
| | - Mihai G Netea
- Department of Internal Medicine and Radboud Center for Infectious Diseases, Radboud University Medical Center, Geert Grooteplein 28, 6525 GA Nijmegen, The Netherlands
| | - Karla Queiroz
- Mimetas, Biopartner Building 2, J.H. Oortweg 19, 2333 CH Leiden, The Netherlands
| | - Karine Roget
- NEXBIOME Therapeutics, 22 allée Alan Turing, 63000 Clermont-Ferrand, France
| | - Vincent Thomas
- BIOASTER Microbiology Technology Institute, 40 avenue Tony Garnier, 69007 Lyon, France
| | - Claudia Thoral
- NEXBIOME Therapeutics, 22 allée Alan Turing, 63000 Clermont-Ferrand, France
| | | | - Alan W Walker
- Gut Microbiology Group, Rowett Institute, University of Aberdeen, Ashgrove Road West, Foresterhill, Aberdeen AB25 2ZD, UK
| | - Alistair J P Brown
- MRC Centre for Medical Mycology, Department of Biosciences, University of Exeter, Geoffrey Pope Building, Stocker Road, Exeter EX4 4QD, UK
| |
Collapse
|
32
|
Desamero MJM, Chung SH, Kakuta S. Insights on the Functional Role of Beta-Glucans in Fungal Immunity Using Receptor-Deficient Mouse Models. Int J Mol Sci 2021; 22:4778. [PMID: 33946381 PMCID: PMC8125483 DOI: 10.3390/ijms22094778] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Revised: 04/21/2021] [Accepted: 04/27/2021] [Indexed: 12/18/2022] Open
Abstract
Understanding the host anti-fungal immunity induced by beta-glucan has been one of the most challenging conundrums in the field of biomedical research. During the last couple of decades, insights on the role of beta-glucan in fungal disease progression, susceptibility, and resistance have been greatly augmented through the utility of various beta-glucan cognate receptor-deficient mouse models. Analysis of dectin-1 knockout mice has clarified the downstream signaling pathways and adaptive effector responses triggered by beta-glucan in anti-fungal immunity. On the other hand, assessment of CR3-deficient mice has elucidated the compelling action of beta-glucans in neutrophil-mediated fungal clearance, and the investigation of EphA2-deficient mice has highlighted its novel involvement in host sensing and defense to oral mucosal fungal infection. Based on these accounts, this review focuses on the recent discoveries made by these gene-targeted mice in beta-glucan research with particular emphasis on the multifaceted aspects of fungal immunity.
Collapse
Affiliation(s)
- Mark Joseph Maranan Desamero
- Laboratory of Biomedical Science, Graduate School of Agricultural and Life Sciences, The University of Tokyo, 1-1-1 Yayoi, Bunkyo-ku, Tokyo 113-8657, Japan;
- Department of Basic Veterinary Sciences, College of Veterinary Medicine, University of the Philippines Los Baños, Laguna 4031, Philippines
| | - Soo-Hyun Chung
- Division of Experimental Animal Immunology, Research Institute for Biomedical Sciences, Tokyo University of Science, 2669 Yamazaki, Noda, Chiba 278-0022, Japan;
| | - Shigeru Kakuta
- Laboratory of Biomedical Science, Graduate School of Agricultural and Life Sciences, The University of Tokyo, 1-1-1 Yayoi, Bunkyo-ku, Tokyo 113-8657, Japan;
| |
Collapse
|
33
|
The Immunomodulatory Properties of β-2,6 Fructans: A Comprehensive Review. Nutrients 2021; 13:nu13041309. [PMID: 33921025 PMCID: PMC8071392 DOI: 10.3390/nu13041309] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2021] [Revised: 04/05/2021] [Accepted: 04/11/2021] [Indexed: 02/07/2023] Open
Abstract
Polysaccharides such as β-2,1-linked fructans including inulin or fructose oligosaccharides are well-known prebiotics with recognised immunomodulatory properties. In recent years, other fructan types covering β-2,6-linked fructans, particularly microbial levans, have gained increasing interest in the field. β-2,6-linked fructans of different degrees of polymerisation can be synthesised by plants or microbes including those that reside in the gastrointestinal tract. Accumulating evidence suggests a role for these β-2,6 fructans in modulating immune function. Here, we provide an overview of the sources and structures of β-2,6 fructans from plants and microbes and describe their ability to modulate immune function in vitro and in vivo along with the suggested mechanisms underpinning their immunomodulatory properties. Further, we discuss the limitations and perspectives pertinent to current studies and the potential applications of β-2,6 fructans including in gut health.
Collapse
|
34
|
Cohen-Kedar S, Keizer D, Schwartz S, Rabinowitz KM, Kaboub K, Shaham Barda E, Sadot E, Wolff-Bar M, Shaltiel T, Dotan I. Commensal fungi and their cell-wall β-glucans direct differential responses in human intestinal epithelial cells. Eur J Immunol 2021; 51:864-878. [PMID: 33616974 DOI: 10.1002/eji.202048852] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2020] [Revised: 10/13/2020] [Accepted: 11/25/2020] [Indexed: 12/17/2022]
Abstract
Intestinal epithelial cells (IECs) are the first to encounter luminal antigens and play an active role in intestinal immune responses. We previously reported that β-glucans, major fungal cell-wall glycans, induced chemokine secretion by IEC lines in a Dectin-1- and Syk-dependent manner. Here, we show that in contrast to β-glucans, stimulation of IEC lines with Candida albicans and Saccharomyces cerevisiae did not induce secretion of any of the proinflammatory cytokines IL-8, CCL2, CXCL1, and GM-CSF. Commensal fungi and β-glucans activated Syk and ERK in IEC lines. However, only β-glucans activated p38, JNK, and the transcription factors NF-κB p65 and c-JUN, which were necessary for cytokine secretion. Furthermore, costimulation of IEC lines with β-glucans and C. albicans yielded decreased cytokine secretion compared to stimulation with β-glucans alone. Finally, ex vivo stimulation of human colonic mucosal explants with zymosan and C. albicans, leads to epithelial Syk and ERK phosphorylation, implying recognition of fungi and similar initial signaling pathways as in IEC lines. Lack of cytokine secretion in response to commensal fungi may reflect IECs' response to fungal glycans, other than β-glucans, that contribute to mucosal tolerance. Skewed epithelial response to commensal fungi may impair homeostasis and contribute to intestinal inflammation.
Collapse
Affiliation(s)
- Sarit Cohen-Kedar
- Division of Gastroenterology, Rabin Medical Center, Petah Tikva, Israel.,Felsenstein Medical Research Center, Sackler Faculty of Medicine, Tel-Aviv University, Tel Aviv, Israel
| | - Danielle Keizer
- Division of Gastroenterology, Rabin Medical Center, Petah Tikva, Israel.,Felsenstein Medical Research Center, Sackler Faculty of Medicine, Tel-Aviv University, Tel Aviv, Israel
| | - Suzana Schwartz
- Division of Gastroenterology, Rabin Medical Center, Petah Tikva, Israel.,Felsenstein Medical Research Center, Sackler Faculty of Medicine, Tel-Aviv University, Tel Aviv, Israel
| | - Keren M Rabinowitz
- Division of Gastroenterology, Rabin Medical Center, Petah Tikva, Israel.,Felsenstein Medical Research Center, Sackler Faculty of Medicine, Tel-Aviv University, Tel Aviv, Israel
| | - Kawsar Kaboub
- Division of Gastroenterology, Rabin Medical Center, Petah Tikva, Israel.,Felsenstein Medical Research Center, Sackler Faculty of Medicine, Tel-Aviv University, Tel Aviv, Israel
| | - Efrat Shaham Barda
- Division of Gastroenterology, Rabin Medical Center, Petah Tikva, Israel.,Felsenstein Medical Research Center, Sackler Faculty of Medicine, Tel-Aviv University, Tel Aviv, Israel
| | - Eran Sadot
- Department of Surgery, Beilinson Campus, Rabin Medical Center, Petah Tikva, Israel
| | - Meirav Wolff-Bar
- Department of Pathology, Beilinson Campus, Rabin Medical Center, Petah Tikva, Israel
| | - Tali Shaltiel
- Department of Surgery, Beilinson Campus, Rabin Medical Center, Petah Tikva, Israel
| | - Iris Dotan
- Division of Gastroenterology, Rabin Medical Center, Petah Tikva, Israel.,Felsenstein Medical Research Center, Sackler Faculty of Medicine, Tel-Aviv University, Tel Aviv, Israel
| |
Collapse
|
35
|
Kopiasz Ł, Dziendzikowska K, Gajewska M, Oczkowski M, Majchrzak-Kuligowska K, Królikowski T, Gromadzka-Ostrowska J. Effects of Dietary Oat Beta-Glucans on Colon Apoptosis and Autophagy through TLRs and Dectin-1 Signaling Pathways-Crohn's Disease Model Study. Nutrients 2021; 13:nu13020321. [PMID: 33499397 PMCID: PMC7911679 DOI: 10.3390/nu13020321] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2020] [Revised: 01/18/2021] [Accepted: 01/19/2021] [Indexed: 12/11/2022] Open
Abstract
Background: Crohn’s disease (CD) is characterized by chronic inflammation of the gastrointestinal tract with alternating periods of exacerbation and remission. The aim of this study was to determine the time-dependent effects of dietary oat beta-glucans on colon apoptosis and autophagy in the CD rat model. Methods: A total of 150 Sprague–Dawley rats were divided into two main groups: healthy control (H) and a TNBS (2,4,6-trinitrobenzosulfonic acid)-induced colitis (C) group, both including subgroups fed with feed without beta-glucans (βG−) or feed supplemented with low- (βGl) or high-molar-mass oat beta-glucans (βGh) for 3, 7, or 21 days. The expression of autophagy (LC3B) and apoptosis (Caspase-3) markers, as well as Toll-like (TLRs) and Dectin-1 receptors, in the colon epithelial cells, was determined using immunohistochemistry and Western blot. Results: The results showed that in rats with colitis, after 3 days of induction of inflammation, the expression of Caspase-3 and LC3B in intestinal epithelial cells did not change, while that of TLR 4 and Dectin-1 decreased. Beta-glucan supplementation caused an increase in the expression of TLR 5 and Dectin-1 with no changes in the expression of Caspase-3 and LC3B. After 7 days, a high expression of Caspase-3 was observed in the colitis-induced animals without any changes in the expression of LC3B and TLRs, and simultaneously, a decrease in Dectin-1 expression was observed. The consumption of feed with βGl or βGh resulted in a decrease in Caspase-3 expression and an increase in TLR 5 expression in the CβGl group, with no change in the expression of LC3B and TLR 4. After 21 days, the expression of Caspase-3 and TLRs was not changed by colitis, while that of LC3B and Dectin-1 was decreased. Feed supplementation with βGh resulted in an increase in the expression of both Caspase-3 and LC3B, while the consumption of feed with βGh and βGl increased Dectin-1 expression. However, regardless of the type of nutritional intervention, the expression of TLRs did not change after 21 days. Conclusions: Dietary intake of βGl and βGh significantly reduced colitis by time-dependent modification of autophagy and apoptosis, with βGI exhibiting a stronger effect on apoptosis and βGh on autophagy. The mechanism of this action may be based on the activation of TLRs and Dectin-1 receptor and depends on the period of exacerbation or remission of CD.
Collapse
Affiliation(s)
- Łukasz Kopiasz
- Department of Dietetics, Institute of Human Nutrition Sciences, Warsaw University of Life Sciences, Nowoursynowska 159c, 02-776 Warsaw, Poland; (Ł.K.); (M.O.); (T.K.); (J.G.-O.)
| | - Katarzyna Dziendzikowska
- Department of Dietetics, Institute of Human Nutrition Sciences, Warsaw University of Life Sciences, Nowoursynowska 159c, 02-776 Warsaw, Poland; (Ł.K.); (M.O.); (T.K.); (J.G.-O.)
- Correspondence: ; Tel.: +48-2259-37-033
| | - Małgorzata Gajewska
- Department of Physiological Sciences, Institute of Veterinary Medicine, Warsaw University of Life Sciences, Nowoursynowska 159, 02-776 Warsaw, Poland; (M.G.); (K.M.-K.)
| | - Michał Oczkowski
- Department of Dietetics, Institute of Human Nutrition Sciences, Warsaw University of Life Sciences, Nowoursynowska 159c, 02-776 Warsaw, Poland; (Ł.K.); (M.O.); (T.K.); (J.G.-O.)
| | - Kinga Majchrzak-Kuligowska
- Department of Physiological Sciences, Institute of Veterinary Medicine, Warsaw University of Life Sciences, Nowoursynowska 159, 02-776 Warsaw, Poland; (M.G.); (K.M.-K.)
| | - Tomasz Królikowski
- Department of Dietetics, Institute of Human Nutrition Sciences, Warsaw University of Life Sciences, Nowoursynowska 159c, 02-776 Warsaw, Poland; (Ł.K.); (M.O.); (T.K.); (J.G.-O.)
| | - Joanna Gromadzka-Ostrowska
- Department of Dietetics, Institute of Human Nutrition Sciences, Warsaw University of Life Sciences, Nowoursynowska 159c, 02-776 Warsaw, Poland; (Ł.K.); (M.O.); (T.K.); (J.G.-O.)
| |
Collapse
|
36
|
Korotchenko E, Schießl V, Scheiblhofer S, Schubert M, Dall E, Joubert IA, Strandt H, Neuper T, Sarajlic M, Bauer R, Geppert M, Joedicke D, Wildner S, Schaller S, Winkler S, Gadermaier G, Horejs‐Hoeck J, Weiss R. Laser-facilitated epicutaneous immunotherapy with hypoallergenic beta-glucan neoglycoconjugates suppresses lung inflammation and avoids local side effects in a mouse model of allergic asthma. Allergy 2021; 76:210-222. [PMID: 32621318 PMCID: PMC7818228 DOI: 10.1111/all.14481] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2020] [Revised: 05/28/2020] [Accepted: 06/09/2020] [Indexed: 12/13/2022]
Abstract
Background Allergen‐specific immunotherapy via the skin targets a tissue rich in antigen‐presenting cells, but can be associated with local and systemic side effects. Allergen‐polysaccharide neoglycogonjugates increase immunization efficacy by targeting and activating dendritic cells via C‐type lectin receptors and reduce side effects. Objective We investigated the immunogenicity, allergenicity, and therapeutic efficacy of laminarin‐ovalbumin neoglycoconjugates (LamOVA). Methods The biological activity of LamOVA was characterized in vitro using bone marrow‐derived dendritic cells. Immunogenicity and therapeutic efficacy were analyzed in BALB/c mice. Epicutaneous immunotherapy (EPIT) was performed using fractional infrared laser ablation to generate micropores in the skin, and the effects of LamOVA on blocking IgG, IgE, cellular composition of BAL, lung, and spleen, lung function, and T‐cell polarization were assessed. Results Conjugation of laminarin to ovalbumin reduced its IgE binding capacity fivefold and increased its immunogenicity threefold in terms of IgG generation. EPIT with LamOVA induced significantly higher IgG levels than OVA, matching the levels induced by s.c. injection of OVA/alum (SCIT). EPIT was equally effective as SCIT in terms of blocking IgG induction and suppression of lung inflammation and airway hyperresponsiveness, but SCIT was associated with higher levels of therapy‐induced IgE and TH2 cytokines. EPIT with LamOVA induced significantly lower local skin reactions during therapy compared to unconjugated OVA. Conclusion Conjugation of ovalbumin to laminarin increased its immunogenicity while at the same time reducing local side effects. LamOVA EPIT via laser‐generated micropores is safe and equally effective compared to SCIT with alum, without the need for adjuvant.
Collapse
Affiliation(s)
| | - Viktoria Schießl
- Department of Biosciences University of Salzburg Salzburg Austria
| | | | - Mario Schubert
- Department of Biosciences University of Salzburg Salzburg Austria
| | - Elfriede Dall
- Department of Biosciences University of Salzburg Salzburg Austria
| | | | - Helen Strandt
- Department of Biosciences University of Salzburg Salzburg Austria
| | - Theresa Neuper
- Department of Biosciences University of Salzburg Salzburg Austria
| | - Muamera Sarajlic
- Department of Biosciences University of Salzburg Salzburg Austria
| | - Renate Bauer
- Department of Biosciences University of Salzburg Salzburg Austria
| | - Mark Geppert
- Department of Biosciences University of Salzburg Salzburg Austria
| | - David Joedicke
- Research and Development University of Applied Biosciences Upper Austria Hagenberg Austria
| | - Sabrina Wildner
- Department of Biosciences University of Salzburg Salzburg Austria
| | - Susanne Schaller
- Research and Development University of Applied Biosciences Upper Austria Hagenberg Austria
| | - Stephan Winkler
- Research and Development University of Applied Biosciences Upper Austria Hagenberg Austria
| | | | | | - Richard Weiss
- Department of Biosciences University of Salzburg Salzburg Austria
| |
Collapse
|
37
|
Oerlemans MM, Akkerman R, Ferrari M, Walvoort MT, de Vos P. Benefits of bacteria-derived exopolysaccharides on gastrointestinal microbiota, immunity and health. J Funct Foods 2021. [DOI: 10.1016/j.jff.2020.104289] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
|
38
|
Basso AMM, De Castro RJA, de Castro TB, Guimarães HI, Polez VLP, Carbonero ER, Pomin VH, Hoffmann C, Grossi-de-Sa MF, Tavares AH, Bocca AL. Immunomodulatory activity of β-glucan-containing exopolysaccharides from Auricularia auricular in phagocytes and mice infected with Cryptococcus neoformans. Med Mycol 2020; 58:227-239. [PMID: 31095342 DOI: 10.1093/mmy/myz042] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2019] [Revised: 04/01/2019] [Accepted: 04/12/2019] [Indexed: 12/19/2022] Open
Abstract
Current antifungal drugs present poor effectiveness and there is no available vaccine for fungal infections. Thus, novel strategies to treat or prevent invasive mycosis, such as cryptococcosis, are highly desirable. One strategy is the use of immunomodulators of polysaccharide nature isolated from mushrooms. The purpose of the present work was to evaluate the immunostimulatory activity of β-(1,3)-glucan-containing exopolysaccharides (EPS) from the edible mushrooms Auricularia auricula in phagocytes and mice infected with Cryptococcus neoformans. EPS triggered macrophages and dendritic cell activation upon binding to Dectin-1, a pattern recognition receptor of the C-type lectin receptor family. Engagement of Dectin-1 culminated in pro-inflammatory cytokine production and cell maturation via its canonical Syk-dependent pathway signaling. Furthermore, upon EPS treatment, M2-like phenotype macrophages, known to support intracellular survival and replication of C. neoformans, repolarize to M1 macrophage pattern associated with enhanced production of the microbicidal molecule nitric oxide that results in efficient killing of C. neoformans. Treatment with EPS also upregulated transcript levels of genes encoding products associated with host protection against C. neoformans and Dectin-1 mediated signaling in macrophages. Finally, orally administrated β-glucan-containing EPS from A. auricular enhanced the survival of mice infected with C. neoformans. In conclusion, the results demonstrate that EPS from A. auricula exert immunostimulatory activity in phagocytes and induce host protection against C. neoformans, suggesting that polysaccharides from this mushroom may be promising as an adjuvant for vaccines or antifungal therapy.
Collapse
Affiliation(s)
- A M M Basso
- Department of Cell Biology, Institute of Biological Sciences, University of Brasília, UnB, Brasilia, DF, Brazil
| | - R J A De Castro
- Department of Cell Biology, Institute of Biological Sciences, University of Brasília, UnB, Brasilia, DF, Brazil
| | - T B de Castro
- Department of Cell Biology, Institute of Biological Sciences, University of Brasília, UnB, Brasilia, DF, Brazil
| | - H I Guimarães
- Embrapa Genetic Resources and Biotechnology, Brasília, Brazil
| | - V L P Polez
- Embrapa Genetic Resources and Biotechnology, Brasília, Brazil
| | - E R Carbonero
- Department of Chemistry, Federal University of Goiás, Campus Catalão, GO, Brazil
| | - V H Pomin
- Program of Glicobiology, Institute of Medical Biochemistry Leopoldo de Meis, Federal University Federal of Rio de Janeiro, RJ, Brazil.,Department of BioMolecular Sciences, Division of Pharmacognosy and Research Institute of Pharmaceutical Sciences, School of Pharmacy, the University of Mississippi, Oxford, MS 38677-1848, USA
| | - C Hoffmann
- Department of Food Sciences and Experimental Nutrition, School of Pharmaceutical Sciences, University of São Paulo, São Paulo, Brazil
| | - M F Grossi-de-Sa
- Embrapa Genetic Resources and Biotechnology, Brasília, Brazil.,Graduated Program in Genomic Science and Biotechnology, Catholic University of Brasília, Brasília, DF, Brazil
| | - A H Tavares
- Department of Cell Biology, Institute of Biological Sciences, University of Brasília, UnB, Brasilia, DF, Brazil
| | - A L Bocca
- Department of Cell Biology, Institute of Biological Sciences, University of Brasília, UnB, Brasilia, DF, Brazil
| |
Collapse
|
39
|
Ren L, Zhang J, Zhang T. Immunomodulatory activities of polysaccharides from Ganoderma on immune effector cells. Food Chem 2020; 340:127933. [PMID: 32882476 DOI: 10.1016/j.foodchem.2020.127933] [Citation(s) in RCA: 174] [Impact Index Per Article: 43.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2020] [Revised: 07/10/2020] [Accepted: 08/23/2020] [Indexed: 02/04/2023]
Abstract
Polysaccharides are the most abundant bioactive compounds in Ganoderma and have been widely used as dietary supplements in traditional Chinese medicine for thousands of years. Polysaccharides from Ganoderma exhibit unique biological properties, including anti-tumor, anti-inflammatory, and immunomodulatory activities. Herein, the sources and structures of polysaccharides from Ganoderma were presented. This work also reviews the immunomodulatory activities and possible mechanisms of polysaccharides from Ganoderma on different immune effector cells, including lymphocytes and myeloid cells. As an available adjunctive remedy, polysaccharides from Ganoderma can potentially be applied for the modulation of the host immune system, namely the innate immunity, the cellular immunity, and the humoral immunity.
Collapse
Affiliation(s)
- Li Ren
- College of Food Science and Engineering, Jilin University, Changchun 130062, China
| | - Jie Zhang
- College of Food Science and Engineering, Jilin University, Changchun 130062, China.
| | - Tiehua Zhang
- College of Food Science and Engineering, Jilin University, Changchun 130062, China.
| |
Collapse
|
40
|
Liang J, Zhang JJ, Huang HI, Kanayama M, Youssef N, Jin YJ, Reyes EY, Abram CL, Yang S, Lowell CA, Wang D, Shao L, Shinohara ML, Zhang JY, Hammer GE. The Ubiquitin-Modifying Enzyme A20 Terminates C-Type Lectin Receptor Signals and Is a Suppressor of Host Defense against Systemic Fungal Infection. Infect Immun 2020; 88:e00048-20. [PMID: 32540868 PMCID: PMC7440764 DOI: 10.1128/iai.00048-20] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2020] [Accepted: 06/01/2020] [Indexed: 01/02/2023] Open
Abstract
C-type lectin receptors (CLRs) play key roles in antifungal defense. CLR-induced NF-κB is central to CLR functions in immunity, and thus, molecules that control the amplitude of CLR-induced NF-κB could profoundly influence host defense against fungal pathogens. However, little is known about the mechanisms that negatively regulate CLR-induced NF-κB, and molecules which act on the CLR family broadly and which directly regulate acute CLR-signaling cascades remain unidentified. Here, we identify the ubiquitin-editing enzyme A20 as a negative regulator of acute NF-κB activation downstream of multiple CLR pathways. Absence of A20 suppression results in exaggerated CLR responses in cells which are A20 deficient and also cells which are A20 haplosufficient, including multiple primary immune cells. Loss of a single allele of A20 results in enhanced defense against systemic Candida albicans infection and prolonged host survival. Thus, A20 restricts CLR-induced innate immune responses in vivo and is a suppressor of host defense against systemic fungal infection.
Collapse
Affiliation(s)
- Jie Liang
- Department of Immunology, Duke University Medical Center, Durham, North Carolina, USA
| | - Junyi J Zhang
- Department of Immunology, Duke University Medical Center, Durham, North Carolina, USA
| | - Hsin-I Huang
- Department of Immunology, Duke University Medical Center, Durham, North Carolina, USA
| | - Masashi Kanayama
- Department of Immunology, Duke University Medical Center, Durham, North Carolina, USA
| | - Nourhan Youssef
- Department of Immunology, Duke University Medical Center, Durham, North Carolina, USA
| | - Yingai J Jin
- Department of Dermatology, Duke University Medical Center, Durham, North Carolina, USA
| | - Estefany Y Reyes
- Department of Immunology, Duke University Medical Center, Durham, North Carolina, USA
| | - Clare L Abram
- Department of Laboratory Medicine and Immunology Program, University of California, San Francisco, California, USA
| | - Shigao Yang
- Department of Medicine, Duke University Medical Center, Durham, North Carolina, USA
| | - Clifford A Lowell
- Department of Laboratory Medicine and Immunology Program, University of California, San Francisco, California, USA
| | - Donghai Wang
- Department of Immunology, Duke University Medical Center, Durham, North Carolina, USA
- Department of Medicine, Duke University Medical Center, Durham, North Carolina, USA
| | - Ling Shao
- Department of Medicine, University of Southern California, Los Angeles, California, USA
| | - Mari L Shinohara
- Department of Immunology, Duke University Medical Center, Durham, North Carolina, USA
- Department of Molecular Genetics and Microbiology, Duke University Medical Center, Durham, North Carolina, USA
| | - Jennifer Y Zhang
- Department of Dermatology, Duke University Medical Center, Durham, North Carolina, USA
| | - Gianna Elena Hammer
- Department of Immunology, Duke University Medical Center, Durham, North Carolina, USA
- Department of Molecular Genetics and Microbiology, Duke University Medical Center, Durham, North Carolina, USA
| |
Collapse
|
41
|
Euglena Gracilis and β-Glucan Paramylon Induce Ca 2+ Signaling in Intestinal Tract Epithelial, Immune, and Neural Cells. Nutrients 2020; 12:nu12082293. [PMID: 32751743 PMCID: PMC7468862 DOI: 10.3390/nu12082293] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2020] [Revised: 07/24/2020] [Accepted: 07/27/2020] [Indexed: 12/16/2022] Open
Abstract
The intestinal tract contains over half of all immune cells and peripheral nerves and manages the beneficial interactions between food compounds and the host. Paramylon is a β-1,3-glucan storage polysaccharide from Euglena gracilis (Euglena) that exerts immunostimulatory activities by affecting cytokine production. This study investigated the signaling mechanisms that regulate the beneficial interactions between food compounds and the intestinal tract using cell type-specific calcium (Ca2+) imaging in vivo and in vitro. We successfully visualized Euglena- and paramylon-mediated Ca2+ signaling in vivo in intestinal epithelial cells from mice ubiquitously expressing the Yellow Cameleon 3.60 (YC3.60) Ca2+ biosensor. Moreover, in vivo Ca2+ imaging demonstrated that the intraperitoneal injection of both Euglena and paramylon stimulated dendritic cells (DCs) in Peyer’s patches, indicating that paramylon is an active component of Euglena that affects the immune system. In addition, in vitro Ca2+ imaging in dorsal root ganglia indicated that Euglena, but not paramylon, triggers Ca2+ signaling in the sensory nervous system innervating the intestine. Thus, this study is the first to successfully visualize the direct effect of β-1,3-glucan on DCs in vivo and will help elucidate the mechanisms via which Euglena and paramylon exert various effects in the intestinal tract.
Collapse
|
42
|
Kotani H, Koshizuka T, Matsubara K, Nishiyama K, Sugiyama T, Suzutani T. Relationship Between Human β-Defensin 2 and the Vaginal Environment. Jpn J Infect Dis 2020; 73:214-220. [PMID: 31875602 DOI: 10.7883/yoken.jjid.2019.190] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
As one of the main antimicrobial peptides, human β-defensin 2 (HBD2) plays multiple roles in the lower genital tract. Based on the Nugent score as a diagnostic criterion for bacterial vaginosis, we sought to clarify the correlations among the Nugent score and interleukin-6 (IL-6) and HBD2 levels in vaginal secretions in association with various types of infection. Ninety-eight women were recruited for this study. Levels of HBD2 and IL-6 in vaginal wash were measured by enzyme-linked immunosorbent assays. According to the Nugent method, the number of Lactobacillus morphotypes per field of view was well correlated with the HBD2 level. The amount of HBD2 was also well correlated with the presence of Candida spp. (P < 0.01). In vitro experiments revealed that the expression of HBD2 from the human vaginal epithelial cell line, VK2/E6E7, was induced by the addition of heat-killed C. albicans (HKCA). The addition of HKCA induced expression of Dectin-1 mRNA. A luciferase assay for nuclear factor kappa-light-chain-enhancer of activated B cells (NFκB) responsive elements showed that HKCA activated NF-κB signaling. These results suggested that C. albicans induced the activation of Dectin-1 and (NF-κB) signaling, resulting in HBD2 expression. In conclusion, the expression of HBD2 positively correlated with the presence of Lactobacillus and Candida spp.
Collapse
Affiliation(s)
- Hideko Kotani
- Department of Obstetrics and Gynecology, Ehime University Graduate School of Medicine
| | - Tetsuo Koshizuka
- Department of Microbiology, Fukushima Medical University School of Medicine
| | - Keiichi Matsubara
- Department of Obstetrics and Gynecology, Ehime University Graduate School of Medicine
| | - Kyoko Nishiyama
- Department of Microbiology, Fukushima Medical University School of Medicine
| | - Takashi Sugiyama
- Department of Obstetrics and Gynecology, Ehime University Graduate School of Medicine
| | - Tatsuo Suzutani
- Department of Microbiology, Fukushima Medical University School of Medicine
| |
Collapse
|
43
|
Fu J, Zhang Y, Hu Y, Zhao G, Tang Y, Zou L. Concise review: Coarse cereals exert multiple beneficial effects on human health. Food Chem 2020; 325:126761. [PMID: 32387947 DOI: 10.1016/j.foodchem.2020.126761] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2019] [Revised: 02/23/2020] [Accepted: 04/05/2020] [Indexed: 02/08/2023]
Abstract
Coarse cereals (CC) refer to cereal grains except for rice and wheat which are highly-valued as functional foods with nutritional and pharmacological properties. Owing to their diverse positive effect on chronic diseases, coarse cereals exert a vital role in food industry. CC and the main contents prevent tumor pathogenesis through promoting apoptosis, inducing cell cycle arrest as well as modulating metastasis initiation. Meanwhile, CC ameliorates cardiovascular diseases through affecting multiple pathways, such as CaMKII/p-BFAF-3, NF-κB, MAPK, PI3K/Akt, etc. Besides, CC and the main contents have potential as prebiotics which facilitating the activities and growth of probiotics such as Bifidobacteria and Lactobacillus. However, there's a lack of report on CC' beneficial properties and the underlying mechanisms are not fully understood. Here this article explains in detail, the effect and mechanism of CC on chronic diseases like tumor, inflammation and cardiovascular diseases.
Collapse
Affiliation(s)
- Jia Fu
- School of Medicine, Chengdu University, No. 2025, Cheng Luo Road, Chengdu 610106, Sichuan, China
| | - Yan Zhang
- School of Medicine, Chengdu University, No. 2025, Cheng Luo Road, Chengdu 610106, Sichuan, China
| | - Yichen Hu
- Key Laboratory of Coarse Cereal Processing, Ministry of Agriculture and Rural Affairs, Chengdu University, No. 2025, Cheng Luo Road, Chengdu 610106, Sichuan, China
| | - Gang Zhao
- Key Laboratory of Coarse Cereal Processing, Ministry of Agriculture and Rural Affairs, Chengdu University, No. 2025, Cheng Luo Road, Chengdu 610106, Sichuan, China
| | - Yong Tang
- School of Acupuncture and Tuina, Chengdu University of Traditional Chinese Medicine, No.1166, Liutai Avenue, Wenjiang District, Chengdu 611137, Sichuan, China.
| | - Liang Zou
- School of Medicine, Chengdu University, No. 2025, Cheng Luo Road, Chengdu 610106, Sichuan, China.
| |
Collapse
|
44
|
Effects of Euglena gracilis EOD-1 Ingestion on Salivary IgA Reactivity and Health-Related Quality of Life in Humans. Nutrients 2019; 11:nu11051144. [PMID: 31121913 PMCID: PMC6566313 DOI: 10.3390/nu11051144] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2019] [Revised: 05/18/2019] [Accepted: 05/20/2019] [Indexed: 02/06/2023] Open
Abstract
Euglena gracilis EOD-1, a microalgal strain known for high yields of the β-1, 3-glucan paramylon, is suggested to function as a dietary fiber and enhance immunity. Here, we aimed to investigate the effects of E. gracilis EOD-1 biomass (EOD1BM) ingestion on immunoglobulin A (IgA) antibody titers in saliva, its reactivity, and the health-related quality of life (QOL) in humans. Reacting human immunoglobulin preparations and saliva with paramylon granules revealed the presence of anti-paramylon antibodies in the blood and saliva. We conducted a placebo-controlled, double-blind, crossover study involving 13 healthy subjects who ingested the placebo or EOD1BM for 4 weeks. Saliva was collected from each subject before and after ingestion, and IgA titers and E. gracilis EOD-1 paramylon (EOD1PM) reactivity were compared. In the EOD1BM Ingestion group, the anti-EOD1PM IgA content and titer increased after EOD1BM ingestion. No such change was observed in the Placebo group. Furthermore, the health-related QOL, especially mental health, increased in the EOD1BM Ingestion group. Thus, EOD1BM ingestion led to the production of paramylon (PM)-specific IgA antibody and increased salivary IgA antibody titers. We demonstrate that EOD1BM ingestion enhanced the immunity in the mucosal surface, evoked an antigen-specific response, and increased the health-related QOL, thereby contributing to health improvement.
Collapse
|
45
|
do Prado SBR, Castro-Alves VC, Ferreira GF, Fabi JP. Ingestion of Non-digestible Carbohydrates From Plant-Source Foods and Decreased Risk of Colorectal Cancer: A Review on the Biological Effects and the Mechanisms of Action. Front Nutr 2019; 6:72. [PMID: 31157230 PMCID: PMC6529955 DOI: 10.3389/fnut.2019.00072] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2018] [Accepted: 04/30/2019] [Indexed: 12/13/2022] Open
Abstract
The hypothesis that links the increase in the intake of plant-source foods to a decrease in colorectal cancer (CRC) risk has almost 50 years. Nowadays, systematic reviews and meta-analysis of case-control and cohort studies confirmed the association between dietary patterns and CRC risk, in which the non-digestible carbohydrates (NDC) from plant-source foods are known to play beneficial effects. However, the mechanisms behind the physicochemical properties and biological effects induced by NDC on the decrease of CRC development and progression remain not fully understood. NDC from plant-source foods consist mainly of complex carbohydrates from plant cell wall including pectin and hemicellulose, which vary among foods in structure and in composition, therefore in both physicochemical properties and biological effects. In the present review, we highlighted the mechanisms and described the recent findings showing how these complex NDC from plant-source foods are related to a decrease in CRC risk through induction of both physicochemical effects in the gastrointestinal tract, fermentation-related effects, and direct effects resulting from the interaction between NDC and cellular components including toll-like receptors and galectin-3. Studies support that the definition of the structure-function relationship-especially regarding the fermentation-related effects of NDC, as well as the direct effects of these complex carbohydrates in cells-is crucial for understanding the possible NDC anticancer effects. The dietary recommendations for the intake of NDC are usually quantitative, describing a defined amount of intake per day. However, as NDC from plant-source foods can exert effects that vary widely according to the NDC structure, the dietary recommendations for the intake of NDC plant-source foods are expected to change from a quantitative to a qualitative perspective in the next few years, as occurred for lipid recommendations. Thus, further studies are necessary to define whether specific and well-characterized NDC from plant-source foods induce beneficial effects related to a decrease in CRC risk, thereby improving nutritional recommendations of healthy individuals and CRC patients.
Collapse
Affiliation(s)
- Samira Bernardino Ramos do Prado
- Department of Food Science and Experimental Nutrition, School of Pharmaceutical Sciences, University of São Paulo, São Paulo, Brazil
- Food Research Center (FoRC), CEPID-FAPESP (Research, Innovation and Dissemination Centers, São Paulo Research Foundation), São Paulo, Brazil
| | - Victor Costa Castro-Alves
- Department of Food Science and Experimental Nutrition, School of Pharmaceutical Sciences, University of São Paulo, São Paulo, Brazil
- Food Research Center (FoRC), CEPID-FAPESP (Research, Innovation and Dissemination Centers, São Paulo Research Foundation), São Paulo, Brazil
| | - Gabrielle Fernandez Ferreira
- Department of Food Science and Experimental Nutrition, School of Pharmaceutical Sciences, University of São Paulo, São Paulo, Brazil
| | - João Paulo Fabi
- Department of Food Science and Experimental Nutrition, School of Pharmaceutical Sciences, University of São Paulo, São Paulo, Brazil
- Food Research Center (FoRC), CEPID-FAPESP (Research, Innovation and Dissemination Centers, São Paulo Research Foundation), São Paulo, Brazil
- Food and Nutrition Research Center (NAPAN), University of São Paulo, São Paulo, Brazil
| |
Collapse
|
46
|
Tang C, Makusheva Y, Sun H, Han W, Iwakura Y. Myeloid C-type lectin receptors in skin/mucoepithelial diseases and tumors. J Leukoc Biol 2019; 106:903-917. [PMID: 30964564 PMCID: PMC6850291 DOI: 10.1002/jlb.2ri0119-031r] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2019] [Revised: 03/11/2019] [Accepted: 03/20/2019] [Indexed: 12/30/2022] Open
Abstract
Myeloid C‐type lectin receptors (CLRs), which consist of an extracellular carbohydrate recognition domain and intracellular signal transducing motif such as the immunoreceptor tyrosine‐based activation motif (ITAM) or immunoreceptor tyrosine‐based inhibitory motif (ITIM), are innate immune receptors primarily expressed on myeloid lineage cells such as dendritic cells (DCs) and Mϕs. CLRs play important roles in host defense against infection by fungi and bacteria by recognizing specific carbohydrate components of these pathogens. However, these immune receptors also make important contributions to immune homeostasis of mucosa and skin in mammals by recognizing components of microbiota, as well as by recognizing self‐components such as alarmins from dead cells and noncanonical non‐carbohydrate ligands. CLR deficiency not only induces hypersensitivity to infection, but also causes dysregulation of muco‐cutaneous immune homeostasis, resulting in the development of allergy, inflammation, autoimmunity, and tumors. In this review, we introduce recent discoveries regarding the roles of myeloid CLRs in the immune system exposed to the environment, and discuss the roles of these lectin receptors in the development of colitis, asthma, psoriasis, atopic dermatitis, and cancer. Although some CLRs are suggested to be involved in the development of these diseases, the function of CLRs and their ligands still largely remain to be elucidated.
Collapse
Affiliation(s)
- Ce Tang
- Center for Animal Disease Models, Research Institute for Biomedical Sciences, Tokyo University of Science, Noda, Japan
| | - Yulia Makusheva
- Center for Animal Disease Models, Research Institute for Biomedical Sciences, Tokyo University of Science, Noda, Japan
| | - Haiyang Sun
- Center for Animal Disease Models, Research Institute for Biomedical Sciences, Tokyo University of Science, Noda, Japan
| | - Wei Han
- Center for Animal Disease Models, Research Institute for Biomedical Sciences, Tokyo University of Science, Noda, Japan
| | - Yoichiro Iwakura
- Center for Animal Disease Models, Research Institute for Biomedical Sciences, Tokyo University of Science, Noda, Japan
| |
Collapse
|
47
|
Hernández-Chávez MJ, Franco B, Clavijo-Giraldo DM, Hernández NV, Estrada-Mata E, Mora-Montes HM. Role of protein phosphomannosylation in the Candida tropicalis-macrophage interaction. FEMS Yeast Res 2019; 18:4989128. [PMID: 29718196 DOI: 10.1093/femsyr/foy053] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2017] [Accepted: 04/26/2018] [Indexed: 12/16/2022] Open
Abstract
Candida tropicalis is an opportunistic fungal pathogen responsible for mucosal and systemic infections. The cell wall is the initial contact point between a fungal cell and the host immune system, and mannoproteins are important components that play key roles when interacting with host cells. In Candida albicans, mannans are modified by mannosyl-phosphate moieties, named phosphomannans, which can work as molecular scaffolds to synthesize β1,2-mannooligosaccharides, and MNN4 is a positive regulator of the phosphomannosylation pathway. Here, we showed that C. tropicalis also displays phosphomannans on the cell surface, but the amount of this cell wall component varies depending on the fungal strain. We also identified a functional ortholog of CaMNN4 in C. tropicalis. Disruption of this gene caused depletion of phosphomannan content. The C. tropicalis mnn4Δ did not show defects in the ability to stimulate cytokine production by human mononuclear cells but displayed virulence attenuation in an insect model of candidiasis. When the mnn4Δ-macrophage interaction was analyzed, results showed that presence of cell wall phosphomannan was critical for C. tropicalis phagocytosis. Finally, our results strongly suggest a differential role for phosphomannans during phagocytosis of C. albicans and C. tropicalis.
Collapse
Affiliation(s)
- Marco J Hernández-Chávez
- Departamento de Biología, Universidad de Guanajuato, Noria Alta s/n, col. Noria Alta, C.P. 36050, Guanajuato, Gto., México
| | - Bernardo Franco
- Departamento de Biología, Universidad de Guanajuato, Noria Alta s/n, col. Noria Alta, C.P. 36050, Guanajuato, Gto., México
| | - Diana M Clavijo-Giraldo
- Departamento de Biología, Universidad de Guanajuato, Noria Alta s/n, col. Noria Alta, C.P. 36050, Guanajuato, Gto., México
| | - Nahúm V Hernández
- Departamento de Biología, Universidad de Guanajuato, Noria Alta s/n, col. Noria Alta, C.P. 36050, Guanajuato, Gto., México
| | - Eine Estrada-Mata
- Departamento de Biología, Universidad de Guanajuato, Noria Alta s/n, col. Noria Alta, C.P. 36050, Guanajuato, Gto., México
| | - Héctor Manuel Mora-Montes
- Departamento de Biología, Universidad de Guanajuato, Noria Alta s/n, col. Noria Alta, C.P. 36050, Guanajuato, Gto., México
| |
Collapse
|
48
|
Goren I, Godny L, Reshef L, Yanai H, Gophna U, Tulchinsky H, Dotan I. Starch Consumption May Modify Antiglycan Antibodies and Fecal Fungal Composition in Patients With Ileo-Anal Pouch. Inflamm Bowel Dis 2019; 25:742-749. [PMID: 30535148 DOI: 10.1093/ibd/izy370] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/30/2018] [Indexed: 02/06/2023]
Abstract
BACKGROUND Inflammatory bowel diseases (IBDs) are characterized by serologic responses to glycans. Patients with ulcerative colitis (UC) after proctocolectomy with ileo-anal anastomosis (pouch surgery) may develop inflammation (pouchitis) that resembles Crohn's disease (CD). We hypothesized that patients' serologic responses were affected by their consumption of dietary sugars. This study analyzed the correlations between antiglycan antibody expression and dietary sugar consumption in patients with UC pouch and the evolution in antibody levels over time. METHODS Patients were followed prospectively for 2 consecutive visits. The following antiglycan carbohydrate antibodies were detected by enzyme-linked immunosorbent assay: antichitobioside (ACCA), antilaminaribioside (ALCA), antimannobioside (AMCA), and anti-Saccharomyces cerevisiae (ASCA) antibodies. Patients completed a food frequency questionnaire. The fungal community in patients' fecal samples was analyzed by sequencing the internal transcribed spacer 2 (ITS2) region of nuclear ribosomal DNA. RESULTS We included 75 UC pouch patients aged 45.2 ± 14 years who underwent pouch surgery 9.8 ± 6.7 years previously. Of these patients, 34.7% (n = 26) showed seropositivity for antiglycan antibodies. Starch consumption was significantly higher in patients with positive serologic responses (P = 0.05). Higher starch consumption was associated with higher AMCA and ACCA titers, which increased by 4.08% (0.8%-7.4%; P = 0.014) and 4.8% (0.7%-9.1%; P = 0.007), respectively, for each 10-g increase of dietary starch. The per-patient change in the relative abundance of Candida albicans in fecal samples correlated positively with changes in starch consumption (Spearman's r = 0.72; P = 0.012). CONCLUSIONS Starch consumption correlated with positive antiglycan serology (ACCA and AMCA), suggesting that increased dietary starch intake may promote a specific immune response in patients with IBD.
Collapse
Affiliation(s)
- Idan Goren
- Division of Gastroenterology, Rabin Medical Center, Petah Tikva, Israel.,Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Lihi Godny
- Division of Gastroenterology, Rabin Medical Center, Petah Tikva, Israel.,Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Leah Reshef
- Department of Molecular Microbiology and Biotechnology, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv, Israel
| | - Henit Yanai
- Division of Gastroenterology, Rabin Medical Center, Petah Tikva, Israel.,Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Uri Gophna
- Department of Molecular Microbiology and Biotechnology, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv, Israel
| | - Hagit Tulchinsky
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel.,Department of Surgery, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel
| | - Iris Dotan
- Division of Gastroenterology, Rabin Medical Center, Petah Tikva, Israel.,Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| |
Collapse
|
49
|
Jin X, Zhang M, Cao GF, Yang YF. Saccharomyces cerevisiae mannan induces sheep beta-defensin-1 expression via Dectin-2-Syk-p38 pathways in ovine ruminal epithelial cells. Vet Res 2019; 50:8. [PMID: 30717800 PMCID: PMC6360771 DOI: 10.1186/s13567-019-0624-4] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2018] [Accepted: 12/03/2018] [Indexed: 12/20/2022] Open
Abstract
The rumen epithelium of sheep serves as an immune interface with the environment and secretes antimicrobial peptides with bactericidal function against various pathogens. Sheep beta-defensin-1 (SBD-1), an antimicrobial peptide, is secreted from ovine ruminal epithelial cells (OREC) in response to microbial infections. Mannan, the main component of the Saccharomyces cerevisiae cell wall can stimulate innate and regulatory immune responses that could improve the gastrointestinal environment. We aimed at investigating the effects of mannan on SBD-1 expression and the downstream signaling pathways stimulated in OREC. We cultured OREC; assessed the effects of mannan on SBD-1 expression by qPCR and ELISA; and then investigated the underlying signaling pathways using qPCR, ELISA, Western blotting, immunohistochemistry, and immunohistofluorescence. Interestingly, mannan markedly upregulated SBD-1 expression in a concentration- and time-dependent manner. Dectin-2 Mouse mAb, Syk specific inhibitor R406, and specific inhibitors of the p38, ERK1/2, JNK, and NF-κB pathways attenuated mannan-induced SBD-1 expression to varying degrees. These results demonstrate that SBD-1 is upregulated by mannan via the Dectin-2-Syk axis, and this is regulated to a large extent through the mitogen-activated protein kinase (MAPK) p38 and less so through the ERK1/2 and JNK or the NF-κB pathway. Our findings highlight the immunomodulatory effects of mannan on OREC in terms of mannan-induced SBD-1 expression.
Collapse
Affiliation(s)
- Xin Jin
- Veterinary Medicine College of Inner Mongolia Agricultural University, Hohhot, 010018, China.,Key Laboratory of Clinical Diagnosis and Treatment Technology in Animal Disease, Ministry of Agriculture, Hohhot, 010018, China
| | - Man Zhang
- Veterinary Medicine College of Inner Mongolia Agricultural University, Hohhot, 010018, China.,Key Laboratory of Clinical Diagnosis and Treatment Technology in Animal Disease, Ministry of Agriculture, Hohhot, 010018, China
| | - Gui-Fang Cao
- Veterinary Medicine College of Inner Mongolia Agricultural University, Hohhot, 010018, China.,Key Laboratory of Clinical Diagnosis and Treatment Technology in Animal Disease, Ministry of Agriculture, Hohhot, 010018, China
| | - Yin-Feng Yang
- Veterinary Medicine College of Inner Mongolia Agricultural University, Hohhot, 010018, China. .,Key Laboratory of Clinical Diagnosis and Treatment Technology in Animal Disease, Ministry of Agriculture, Hohhot, 010018, China.
| |
Collapse
|
50
|
Zhang M, Jin X, Yang YF. β-Glucan from Saccharomyces cerevisiae induces SBD-1 production in ovine ruminal epithelial cells via the Dectin-1–Syk–NF-κB signaling pathway. Cell Signal 2019; 53:304-315. [DOI: 10.1016/j.cellsig.2018.10.018] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2018] [Revised: 10/23/2018] [Accepted: 10/25/2018] [Indexed: 12/26/2022]
|