1
|
Chin CL, Lin YL, Cheng PY, Lee P, Chiang BL. Effective Mucosal Adjuvantation of the Intranasal Enterovirus A71 Vaccine With Zymosan. Immunology 2025; 174:349-362. [PMID: 39780346 DOI: 10.1111/imm.13895] [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: 09/16/2024] [Revised: 11/27/2024] [Accepted: 12/23/2024] [Indexed: 01/11/2025] Open
Abstract
Enterovirus A71 (EV-A71) has caused hand, foot, and mouth disease with an increased prevalence of neurological complications and acute mortality, threatening young children around the globe. By provoking mucosal immunity, intranasal vaccination has been suggested to prevent EV-A71 infection. However, antigens delivered via the nasal route usually fail to induce a protective memory response. Zymosan has been identified to activate multiple pattern recognition receptors to orchestrate innate and adaptive immunity. Herein, we aimed to investigate the capacity of zymosan to strengthen the vaccine response induced by an intranasal EV-A71 vaccine. First, we confirmed its remarkable capacity to ignite innate signaling by upregulating cytokine production in primary DCs in vitro. Second, we verified its capacity to promote the vaccine immunogenicity in vivo after triple vaccination with EV-A71, especially with the notable induction of virus-specific IgA at multiple mucosae and the IL-17-producing splenic population after antigen reencounter. Lastly, we validated its capacity to improve vaccine efficacy in vivo after dual vaccination by furnishing neonatal protection against lethal infection. Our findings show that zymosan, at a preferable dosage, could augment the benefits of the intranasal vaccination to tackle EV-A71 infection. This research provides a feasible strategy for preventing EV-A71 infection with severe complications and contributes to the development of nasal spray vaccination.
Collapse
Affiliation(s)
- Chiao-Li Chin
- Graduate Institute of Immunology, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Yu-Li Lin
- Department of Medical Research, National Taiwan University Hospital, Taipei, Taiwan
| | - Pei-Yun Cheng
- Department of Medical Research, National Taiwan University Hospital, Taipei, Taiwan
| | - Ping Lee
- Department of Medical Research, National Taiwan University Hospital, Taipei, Taiwan
| | - Bor-Luen Chiang
- Graduate Institute of Immunology, College of Medicine, National Taiwan University, Taipei, Taiwan
- Department of Medical Research, National Taiwan University Hospital, Taipei, Taiwan
- Genome and Systems Biology Degree Program, College of Life Science, National Taiwan University, Taipei, Taiwan
| |
Collapse
|
2
|
Medhasi S, Sriwarom A, Permpalung N, Torvorapanit P, Plongla R, Chindamporn A, Worasilchai N. Pythium insidiosum-antigen enhances neutrophil-mediated killing of zoospores. Sci Rep 2025; 15:5210. [PMID: 39939657 PMCID: PMC11822016 DOI: 10.1038/s41598-025-88962-w] [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: 06/10/2024] [Accepted: 02/03/2025] [Indexed: 02/14/2025] Open
Abstract
Pythium insidiosum-antigen (PIA) immunotherapy has been used to treat human pythiosis. This study compared PIA-stimulated and unstimulated neutrophils on zoospore viability of P. insidiosum strains. We cultured and collected zoospores of 6 P. insidiosum strains, CBS 777.84, ATCC 58643, ATCC 90586, PEC1, PC10, and CBS 101039. PIA concentrations of 0.01, 0.1, 1, and 10 µg/ml were prepared and were used to stimulate neutrophils isolated from healthy volunteers. The MTT (3-(4, 5-dimethylthiazolyl-2)-2, 5-diphenyltetrazolium bromide) assay was used to calculate the zoospore viability of P. insidiosum strains. Neutrophils stimulated with 0.01 or 0.1 µg/ml PIA, or both, showed a significant reduction in the viability of zoospores of CBS 777.84, ATCC 58643, CBS 101039, PC10, and PEC1 strains. Furthermore, 1 µg/ml PIA-induced neutrophils elicited a significant decrease in the viability of zoospores of ATCC 58643, CBS 101039, and PC10 strains. However, a higher dose of PIA (10 µg/ml) did not demonstrate superiority in reducing the zoospore viability of all six strains. Our findings suggest that PIA immunotherapy improves the zoospore-killing activity of neutrophils, and neutrophils might be involved in the forefront mechanism responsible for the beneficial effects of PIA immunotherapy.
Collapse
Affiliation(s)
- Sadeep Medhasi
- Department of Transfusion Medicine and Clinical Microbiology, Faculty of Allied Health Sciences, Chulalongkorn University, 154 Soi Chula12, Phayathai Road, Pathumwan, Bangkok, 10330, Thailand
- Research Unit of Medical Mycology Diagnosis, Chulalongkorn University, Bangkok, Thailand
| | - Apichaya Sriwarom
- Medical Microbiology, Interdisciplinary Program, Graduate School, Chulalongkorn University, Bangkok, Thailand, Bangkok, Thailand
| | - Nitipong Permpalung
- Department of Microbiology, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Pattama Torvorapanit
- Department of Medicine, Faculty of Medicine, King Chulalongkorn Memorial Hospital, Chulalongkorn University, Bangkok, Thailand
- Thai Red Cross Emerging Infectious Diseases Clinical Center, King Chulalongkorn Memorial Hospital, Thai Red Cross Society, Bangkok, Thailand
| | - Rongpong Plongla
- Department of Medicine, Faculty of Medicine, King Chulalongkorn Memorial Hospital, Chulalongkorn University, Bangkok, Thailand
| | - Ariya Chindamporn
- Department of Microbiology, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| | - Navaporn Worasilchai
- Department of Transfusion Medicine and Clinical Microbiology, Faculty of Allied Health Sciences, Chulalongkorn University, 154 Soi Chula12, Phayathai Road, Pathumwan, Bangkok, 10330, Thailand.
- Research Unit of Medical Mycology Diagnosis, Chulalongkorn University, Bangkok, Thailand.
| |
Collapse
|
3
|
Jaecklein E, Papavinasasundaram K, Ostroff GR, Sassetti C, Soto ER. Targeted delivery of antitubercular drugs using glucan lipid particles. Microbiol Spectr 2025:e0274424. [PMID: 39912634 DOI: 10.1128/spectrum.02744-24] [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: 10/30/2024] [Accepted: 01/11/2025] [Indexed: 02/07/2025] Open
Abstract
Simpler, safer, and faster chemotherapeutic regimens for tuberculosis and other respiratory mycobacterial infections are an urgent need. Many current therapies suffer from suboptimal drug exposure and dose-limiting systemic adverse effects, challenges that could be addressed via controlled delivery of drugs to the primary site of infection. We sought to address this need by designing a flexible formulation platform that targets drugs to the lung macrophages that concentrate at infectious foci. Our approach is based on an encapsulation strategy in which drugs or specifically designed prodrugs are captured in the hydrophobic core of a glucan-lipid particle (GLP). We show chemically diverse antimycobacterial drugs can be efficiently and stably encapsulated within GLP and that these microparticles can be engineered to release drugs upon low pH or reducing conditions that occur upon phagocytosis by macrophages. Encapsulated formulations of clofazimine, isoniazid, and linezolid retain activity against intracellular Mycobacterium tuberculosis (Mtb) in an ex vivo model, demonstrating efficient drug delivery and release. Intranasal administration of GLP-clofazimine to Mtb-infected mice effectively concentrates the drug in the lung and reduces bacterial burden, whereas GLP-delivered linezolid was systemically distributed and failed to inhibit bacterial growth in the lung. This work establishes GLPs as a promising platform for targeted antibiotic delivery to the lung and also illustrates pharmacokinetic parameters that must be considered in future development. IMPORTANCE Tuberculosis (TB) causes an estimated 10.8 million cases each year and remains one of the leading causes of infectious death. Effective treatment is complicated due to the lengthy drug regimen required to prevent relapse and treatment failure. A primary challenge is delivering drugs effectively to lung granulomas, where TB bacteria can persist. Here, we developed yeast-derived glucan lipid microparticles (GLPs) as a novel delivery system to efficiently encapsulate and deliver TB drugs directly to lung tissue via intranasal administration. Of the formulations evaluated, GLP-encapsulated clofazimine achieved increased lung drug levels and reduced bacterial burden in TB-infected mice. The use of GLPs offers a promising approach to improve TB treatment by enabling targeted drug delivery to infection sites within the lungs.
Collapse
Affiliation(s)
- Eleni Jaecklein
- Department of Microbiology and Physiological Systems, University of Massachusetts Chan Medical School, Worcester, USA
| | - Kadamba Papavinasasundaram
- Department of Microbiology and Physiological Systems, University of Massachusetts Chan Medical School, Worcester, USA
| | - Gary R Ostroff
- Program in Molecular Medicine, University of Massachusetts Chan Medical School, Worcester, USA
| | - Christopher Sassetti
- Department of Microbiology and Physiological Systems, University of Massachusetts Chan Medical School, Worcester, USA
| | - Ernesto R Soto
- Program in Molecular Medicine, University of Massachusetts Chan Medical School, Worcester, USA
| |
Collapse
|
4
|
Wagner AS, Smith FM, Bennin DA, Votava JA, Datta R, Giese MA, Zhao W, Skala MC, Fan J, Keller NP, Huttenlocher A. GATA1-deficient human pluripotent stem cells generate neutrophils with improved antifungal immunity that is mediated by the integrin CD18. PLoS Pathog 2025; 21:e1012654. [PMID: 39899622 PMCID: PMC11825098 DOI: 10.1371/journal.ppat.1012654] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2024] [Revised: 02/13/2025] [Accepted: 01/15/2025] [Indexed: 02/05/2025] Open
Abstract
Neutrophils are critical for host defense against fungi. However, the short life span and lack of genetic tractability of primary human neutrophils has limited in vitro analysis of neutrophil-fungal interactions. Human induced pluripotent stem cell (iPSC)-derived neutrophils (iNeutrophils) provide a genetically tractable system to study host defense responses of human neutrophils. Here, we show that deletion of the transcription factor GATA1 from human iPSCs results in iNeutrophils with improved antifungal activity against Aspergillus fumigatus. GATA1-knockout (KO) iNeutrophils have increased maturation, antifungal pattern recognition receptor expression and have improved neutrophil effector functions compared to wild-type iNeutrophils. iNeutrophils also show a shift in their metabolism following stimulation with fungal β-glucan to the pentose phosphate pathway (PPP), similar to primary human neutrophils. Furthermore, we show that deletion of the integrin CD18 attenuates the ability of GATA1-KO iNeutrophils to kill A. fumigatus but is not necessary for the metabolic shift. Collectively, these findings support iNeutrophils as a robust system to study human neutrophil antifungal immunity and has identified specific roles for CD18 in the defense response.
Collapse
Affiliation(s)
- Andrew S. Wagner
- Department of Medical Microbiology and Immunology, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin, United States of America
| | - Frances M. Smith
- Department of Medical Microbiology and Immunology, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin, United States of America
| | - David A. Bennin
- Department of Pediatrics, University of Wisconsin-Madison School of Medicine and Public Health, Madison, Wisconsin, United States of America
| | - James A. Votava
- Morgridge Institute for Research, Madison, Wisconsin, United States of America
| | - Rupsa Datta
- Morgridge Institute for Research, Madison, Wisconsin, United States of America
| | - Morgan A. Giese
- Department of Medical Microbiology and Immunology, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin, United States of America
| | - Wenxuan Zhao
- Morgridge Institute for Research, Madison, Wisconsin, United States of America
| | - Melissa C. Skala
- Morgridge Institute for Research, Madison, Wisconsin, United States of America
| | - Jing Fan
- Department of Medical Microbiology and Immunology, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin, United States of America
- Morgridge Institute for Research, Madison, Wisconsin, United States of America
| | - Nancy P. Keller
- Department of Medical Microbiology and Immunology, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin, United States of America
- Department of Plant Pathology, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
| | - Anna Huttenlocher
- Department of Medical Microbiology and Immunology, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin, United States of America
- Department of Pediatrics, University of Wisconsin-Madison School of Medicine and Public Health, Madison, Wisconsin, United States of America
| |
Collapse
|
5
|
Glass E, Robinson SL, Rosowski EE. Zebrafish use conserved CLR and TLR signaling pathways to respond to fungal PAMPs in zymosan. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2025; 162:105286. [PMID: 39536806 PMCID: PMC11740225 DOI: 10.1016/j.dci.2024.105286] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/18/2024] [Revised: 11/05/2024] [Accepted: 11/05/2024] [Indexed: 11/16/2024]
Abstract
Pattern recognition receptors (PRRs) such as C-type lectin receptors (CLRs) and Toll-like receptors (TLRs) are used by hosts to recognize pathogen-associated molecular patterns (PAMPs) in microorganisms and to initiate innate immune responses. While PRRs exist across invertebrate and vertebrate species, the functional homology of many of these receptors is still unclear. In this study, we investigate the innate immune response of zebrafish larvae to zymosan, a β-glucan-containing particle derived from fungal cell walls. Macrophages and neutrophils robustly respond to zymosan and are required for zymosan-induced activation of the NF-κB transcription factor. Full activation of NF-κB in response to zymosan depends on Card9/Syk and Myd88, conserved CLR and TLR adaptor proteins, respectively. Two putative CLRs, Clec4c and Sclra, are both required for maximal sensing of zymosan and NF-κB activation but not required for inflammatory gene expression. Altogether, we identify conserved PRRs and PRR signaling pathways in larval zebrafish that promote recognition of fungal PAMPs. These results inform modeling of human fungal infections in zebrafish and increase our knowledge of the evolution and conservation of PRR pathways in vertebrates.
Collapse
Affiliation(s)
- Erin Glass
- Department of Biological Sciences, Clemson University, Clemson, SC, USA; Eukaryotic Pathogens Innovation Center, Clemson University, Clemson, SC, USA
| | - Stephan L Robinson
- Eukaryotic Pathogens Innovation Center, Clemson University, Clemson, SC, USA; School of Medicine Greenville, University of South Carolina, Greenville, SC, USA
| | - Emily E Rosowski
- Department of Biological Sciences, Clemson University, Clemson, SC, USA; Eukaryotic Pathogens Innovation Center, Clemson University, Clemson, SC, USA.
| |
Collapse
|
6
|
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
|
7
|
Guo Y, Aufiero MA, Mills KAM, Grassmann SA, Kim H, Zumbo P, Gjonbalaj M, Billips A, Mar KB, Yu Y, Betel D, Sun JC, Hohl TM. An IFN-STAT1-CYBB Axis Defines Protective Plasmacytoid DC to Neutrophil Crosstalk During Aspergillus fumigatus Infection. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.24.620079. [PMID: 39484591 PMCID: PMC11527108 DOI: 10.1101/2024.10.24.620079] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/03/2024]
Abstract
Aspergillus fumigatus is the most common cause of invasive aspergillosis (IA), a devastating infection in immunocompromised patients. Plasmacytoid dendritic cells (pDCs) regulate host defense against IA by enhancing neutrophil antifungal properties in the lung. Here, we define the pDC activation trajectory during A. fumigatus infection and the molecular events that underlie the protective pDC - neutrophil crosstalk. Fungus-induced pDC activation begins after bone marrow egress and results in pDC-dependent regulation of lung type I and type III IFN levels. These pDC-derived products act on type I and type III IFN receptor-expressing neutrophils and control neutrophil fungicidal activity and reactive oxygen species production via STAT1 signaling in a cell-intrinsic manner. Mechanistically, neutrophil STAT1 signaling regulates the transcription and expression of Cybb, which encodes one of five NADPH oxidase subunits. Thus, pDCs regulate neutrophil-dependent immunity against inhaled molds by controlling the local expression of a subunit required for NADPH oxidase assembly and activity in the lung.
Collapse
|
8
|
Wagner AS, Smith FM, Bennin DA, Votava JA, Datta R, Giese MA, Zhao W, Skala MC, Fan J, Keller NP, Huttenlocher A. GATA1-deficient human pluripotent stem cells generate neutrophils with improved antifungal immunity that is mediated by the integrin CD18. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.11.617742. [PMID: 39416161 PMCID: PMC11482877 DOI: 10.1101/2024.10.11.617742] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/19/2024]
Abstract
Neutrophils are critical for host defense against fungi. However, the short life span and lack of genetic tractability of primary human neutrophils has limited in vitro analysis of neutrophil-fungal interactions. Human induced pluripotent stem cell (iPSC)-derived neutrophils (iNeutrophils) are a genetically tractable alternative to primary human neutrophils. Here, we show that deletion of the transcription factor GATA1 from human iPSCs results in iNeutrophils with improved antifungal activity against Aspergillus fumigatus. GATA1 knockout (KO) iNeutrophils have increased maturation, antifungal pattern recognition receptor expression and more readily execute neutrophil effector functions compared to wild-type iNeutrophils. iNeutrophils also show a shift in their metabolism following stimulation with fungal β-glucan, including an upregulation of the pentose phosphate pathway (PPP), similar to primary human neutrophils in vitro. Furthermore, we show that deletion of the integrin CD18 attenuates the ability of GATA1-KO iNeutrophils to kill A. fumigatus but is not necessary for the upregulation of PPP. Collectively, these findings support iNeutrophils as a robust system to study human neutrophil antifungal immunity and has identified specific roles for CD18 in the defense response.
Collapse
Affiliation(s)
- Andrew S. Wagner
- Department of Medical Microbiology and Immunology, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| | - Frances M. Smith
- Department of Medical Microbiology and Immunology, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| | - David A. Bennin
- Department of Pediatrics, University of Wisconsin-Madison School of Medicine and Public Health, Madison, WI, USA
| | | | - Rupsa Datta
- Morgridge Institute for Research, Madison, WI, USA
| | - Morgan A. Giese
- Department of Medical Microbiology and Immunology, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| | - Wenxuan Zhao
- Morgridge Institute for Research, Madison, WI, USA
| | | | - Jing Fan
- Department of Medical Microbiology and Immunology, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
- Morgridge Institute for Research, Madison, WI, USA
| | - Nancy P. Keller
- Department of Medical Microbiology and Immunology, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
- Department of Plant Pathology, University of Wisconsin-Madison, WI, USA
| | - Anna Huttenlocher
- Department of Medical Microbiology and Immunology, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
- Department of Pediatrics, University of Wisconsin-Madison School of Medicine and Public Health, Madison, WI, USA
| |
Collapse
|
9
|
Charpak-Amikam Y, Kournos M, Kotzur R, Isaacson B, Bagad Brenner T, Gomez-Cesar E, Abou-Kandil A, Ben-Ami R, Korem M, Guerra N, Osherov N, Mandelboim O. The activating receptor NKG2D is an anti-fungal pattern recognition receptor. Nat Commun 2024; 15:8664. [PMID: 39375344 PMCID: PMC11458907 DOI: 10.1038/s41467-024-52913-2] [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: 04/29/2023] [Accepted: 09/23/2024] [Indexed: 10/09/2024] Open
Abstract
NKG2D is a central activating receptor involved in target recognition and killing by Natural Killer and CD8+ T cells. The known role of NKG2D is to recognize a family of self-induced stress ligands that are upregulated on stressed cells such as cancerous or virally infected cells. Fungal pathogens are a major threat to human health, infecting more than a billion patients yearly and becoming more common and drug resistant. Here we show that NKG2D plays a critical role in the immune response against fungal infections. NKG2D can recognize fungal pathogens from most major families including Candida, Cryptococcus and Aspergillus species, and mice lacking NKG2D are extremely sensitive to fungal infections in models of both invasive and mucosal infections, making NKG2D an anti-fungal pattern recognition receptor.
Collapse
Affiliation(s)
- Yoav Charpak-Amikam
- The Concern Foundation Laboratories at the Lautenberg Center for Immunology and Cancer Research, Hebrew University Medical School, IMRIC, Jerusalem, Israel
| | - Mark Kournos
- The Concern Foundation Laboratories at the Lautenberg Center for Immunology and Cancer Research, Hebrew University Medical School, IMRIC, Jerusalem, Israel
| | - Rebecca Kotzur
- The Concern Foundation Laboratories at the Lautenberg Center for Immunology and Cancer Research, Hebrew University Medical School, IMRIC, Jerusalem, Israel
| | - Batya Isaacson
- The Concern Foundation Laboratories at the Lautenberg Center for Immunology and Cancer Research, Hebrew University Medical School, IMRIC, Jerusalem, Israel
| | - Tal Bagad Brenner
- The Concern Foundation Laboratories at the Lautenberg Center for Immunology and Cancer Research, Hebrew University Medical School, IMRIC, Jerusalem, Israel
| | - Elidet Gomez-Cesar
- The Concern Foundation Laboratories at the Lautenberg Center for Immunology and Cancer Research, Hebrew University Medical School, IMRIC, Jerusalem, Israel
| | - Ammar Abou-Kandil
- Department of Clinical Microbiology and Immunology, Sackler School of Medicine, Tel-Aviv University, Ramat-Aviv, Tel-Aviv, Israel
| | - Ronen Ben-Ami
- Infectious Diseases Unit, Tel Aviv Sourasky Medical Center, and the Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Maya Korem
- Department of Clinical Microbiology and Infectious Diseases, Hadassah-Hebrew University Medical Center, Jerusalem, Israel
| | - Nadia Guerra
- Department of Life Sciences, Imperial College London, London, UK
| | - Nir Osherov
- Department of Clinical Microbiology and Immunology, Sackler School of Medicine, Tel-Aviv University, Ramat-Aviv, Tel-Aviv, Israel
| | - Ofer Mandelboim
- The Concern Foundation Laboratories at the Lautenberg Center for Immunology and Cancer Research, Hebrew University Medical School, IMRIC, Jerusalem, Israel.
| |
Collapse
|
10
|
Mashhouri S, Rahmati A, Azimi A, Fava RA, Ismail IH, Walker J, Elahi S. Targeting Dectin-1 and or VISTA enhances anti-tumor immunity in melanoma but not colorectal cancer model. Cell Oncol (Dordr) 2024; 47:1735-1756. [PMID: 38668817 PMCID: PMC11467025 DOI: 10.1007/s13402-024-00950-w] [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] [Accepted: 04/11/2024] [Indexed: 06/27/2024] Open
Abstract
PURPOSE Acquired resistance to immune checkpoint blockers (ICBs) is a major barrier in cancer treatment, emphasizing the need for innovative strategies. Dectin-1 (gene Clec7a) is a C-type lectin receptor best known for its ability to recognize β-glucan-rich structures in fungal cell walls. While Dectin-1 is expressed in myeloid cells and tumor cells, its significance in cancer remains the subject of controversy. METHODS Using Celc7a-/- mice and curdlan administration to stimulate Dectin-1 signaling, we explored its impact. VISTA KO mice were employed to assess VISTA's role, and bulk RNAseq analyzed curdlan effects on neutrophils. RESULTS Our findings reveal myeloid cells as primary Dectin-1 expressing cells in the tumor microenvironment (TME), displaying an activated phenotype. Strong Dectin-1 co-expression/co-localization with VISTA and PD-L1 in TME myeloid cells was observed. While Dectin-1 deletion lacked protective effects, curdlan stimulation significantly curtailed B16-F10 tumor progression. RNAseq and pathway analyses supported curdlan's role in triggering a cascade of events leading to increased production of pro-inflammatory mediators, potentially resulting in the recruitment and activation of immune cells. Moreover, we identified a heterogeneous subset of Dectin-1+ effector T cells in the TME. Similar to mice, human myeloid cells are the prominent cells expressing Dectin-1 in cancer patients. CONCLUSION Our study proposes Dectin-1 as a potential adjunctive target with ICBs, orchestrating a comprehensive engagement of innate and adaptive immune responses in melanoma. This innovative approach holds promise for overcoming acquired resistance to ICBs in cancer treatment, offering avenues for further exploration and development.
Collapse
Affiliation(s)
- Siavash Mashhouri
- Department of Dentistry, Division of Foundational Sciences, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Canada
| | - Amirhossein Rahmati
- Department of Dentistry, Division of Foundational Sciences, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Canada
| | - Ako Azimi
- Department of Dentistry, Division of Foundational Sciences, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Canada
| | - Roy A Fava
- Department of Veterans Affairs Medical Center, Research Service, White River Junction, VT, USA
- Department of Medicine, Geisel School of Medicine at Dartmouth, Dartmouth Hitchcock Medical Center, Lebanon, NH, USA
| | - Ismail Hassan Ismail
- Department of Oncology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Canada
- Biophysics Department, Faculty of Science, Cairo University, Giza, 12613, Egypt
| | - John Walker
- Department of Oncology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Canada
| | - Shokrollah Elahi
- Department of Dentistry, Division of Foundational Sciences, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Canada.
- Department of Oncology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Canada.
- Li Ka Shing Institute of Virology, University of Alberta, Edmonton, Canada.
| |
Collapse
|
11
|
Zakerska-Banaszak O, Zuraszek-Szymanska J, Eder P, Ladziak K, Slomski R, Skrzypczak-Zielinska M. The Role of Host Genetics and Intestinal Microbiota and Metabolome as a New Insight into IBD Pathogenesis. Int J Mol Sci 2024; 25:9589. [PMID: 39273536 PMCID: PMC11394875 DOI: 10.3390/ijms25179589] [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: 07/12/2024] [Revised: 08/30/2024] [Accepted: 09/02/2024] [Indexed: 09/15/2024] Open
Abstract
Inflammatory bowel disease (IBD) is an incurable, chronic disorder of the gastrointestinal tract whose incidence increases every year. Scientific research constantly delivers new information about the disease and its multivariate, complex etiology. Nevertheless, full discovery and understanding of the complete mechanism of IBD pathogenesis still pose a significant challenge to today's science. Recent studies have unanimously confirmed the association of gut microbial dysbiosis with IBD and its contribution to the regulation of the inflammatory process. It transpires that the altered composition of pathogenic and commensal bacteria is not only characteristic of disturbed intestinal homeostasis in IBD, but also of viruses, parasites, and fungi, which are active in the intestine. The crucial function of the microbial metabolome in the human body is altered, which causes a wide range of effects on the host, thus providing a basis for the disease. On the other hand, human genomic and functional research has revealed more loci that play an essential role in gut homeostasis regulation, the immune response, and intestinal epithelial function. This review aims to organize and summarize the currently available knowledge concerning the role and interaction of crucial factors associated with IBD pathogenesis, notably, host genetic composition, intestinal microbiota and metabolome, and immune regulation.
Collapse
Affiliation(s)
| | | | - Piotr Eder
- Department of Gastroenterology, Dietetics and Internal Medicine, Poznan University of Medical Sciences, 60-355 Poznan, Poland
| | - Karolina Ladziak
- Institute of Human Genetics, Polish Academy of Sciences, 60-479 Poznan, Poland
| | - Ryszard Slomski
- Institute of Human Genetics, Polish Academy of Sciences, 60-479 Poznan, Poland
| | | |
Collapse
|
12
|
Kim JH, Song JW, Kim YH, Kim HJ, Kim RH, Park YH, Nam HS, Kang DO, Yoo H, Park K, Kim JW. Multimodal Imaging-Assisted Intravascular Theranostic Photoactivation on Atherosclerotic Plaque. Circ Res 2024; 135:e114-e132. [PMID: 38989585 DOI: 10.1161/circresaha.123.323970] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/10/2023] [Revised: 07/02/2024] [Accepted: 07/03/2024] [Indexed: 07/12/2024]
Abstract
BACKGROUND Atherosclerosis is a chronic inflammatory disease causing a fatal plaque rupture, and its key aspect is a failure to resolve inflammation. We hypothesize that macrophage-targeted near-infrared fluorescence emitting photoactivation could simultaneously assess macrophage/lipid-rich plaques in vivo and facilitate inflammation resolution. METHODS We fabricated a Dectin-1-targeted photoactivatable theranostic agent through the chemical conjugation of the near-infrared fluorescence-emitting photosensitizer chlorin e6 and the Dectin-1 ligand laminarin (laminarin-chlorin e6 [LAM-Ce6]). Intravascular photoactivation by a customized fiber-based diffuser after administration of LAM-Ce6 effectively reduced inflammation in the targeted plaques of atherosclerotic rabbits in vivo as serially assessed by dual-modal optical coherence tomography-near-infrared fluorescence structural-molecular catheter imaging after 4 weeks. RESULTS The number of apoptotic macrophages peaked at 1 day after laser irradiation and then resolved until 4 weeks. Autophagy was strongly augmented 1 hour after the light therapy, with the formation of autophagolysosomes. LAM-Ce6 photoactivation increased the terminal deoxynucleotidyl transferase dUTP (deoxyuridine triphosphate) nick end labeling/RAM11 (rabbit monocyte/macrophage antibody)- and MerTK (c-Mer tyrosine kinase)-positive cells in the plaques, suggesting enhanced efferocytosis. In line with inflammation resolution, photoactivation reduced the plaque burden through fibrotic replacement via the TGF (transforming growth factor)-β/CTGF (connective tissue growth factor) pathway. CONCLUSIONS Optical coherence tomography-near-infrared fluorescence imaging-guided macrophage Dectin-1-targetable photoactivation could induce the transition of macrophage/lipid-rich plaques into collagen-rich lesions through autophagy-mediated inflammation resolution and TGF-β-dependent fibrotic replacement. This novel strategy offers a new opportunity for the catheter-based theranostic strategy.
Collapse
Affiliation(s)
- Jin Hyuk Kim
- BK21 Graduate Program, Department of Biomedical Sciences, Korea University College of Medicine, Seoul, Korea (J.H.K., J.W.K.)
- Multimodal Imaging and Theranostic Laboratory, Cardiovascular Center, Korea University Guro Hospital (J.H.K., J.W.S., H.J.K., R.H.K., Y.H.P., D.O.K., J.W.K.)
| | - Joon Woo Song
- Multimodal Imaging and Theranostic Laboratory, Cardiovascular Center, Korea University Guro Hospital (J.H.K., J.W.S., H.J.K., R.H.K., Y.H.P., D.O.K., J.W.K.)
| | - Yeon Hoon Kim
- Department of Mechanical Engineering, KAIST, Daejeon, Korea (Y.H.K., H.S.N., H.Y.)
| | - Hyun Jung Kim
- Multimodal Imaging and Theranostic Laboratory, Cardiovascular Center, Korea University Guro Hospital (J.H.K., J.W.S., H.J.K., R.H.K., Y.H.P., D.O.K., J.W.K.)
| | - Ryeong Hyun Kim
- Multimodal Imaging and Theranostic Laboratory, Cardiovascular Center, Korea University Guro Hospital (J.H.K., J.W.S., H.J.K., R.H.K., Y.H.P., D.O.K., J.W.K.)
| | - Ye Hee Park
- Multimodal Imaging and Theranostic Laboratory, Cardiovascular Center, Korea University Guro Hospital (J.H.K., J.W.S., H.J.K., R.H.K., Y.H.P., D.O.K., J.W.K.)
| | - Hyeong Soo Nam
- Department of Mechanical Engineering, KAIST, Daejeon, Korea (Y.H.K., H.S.N., H.Y.)
| | - Dong Oh Kang
- Multimodal Imaging and Theranostic Laboratory, Cardiovascular Center, Korea University Guro Hospital (J.H.K., J.W.S., H.J.K., R.H.K., Y.H.P., D.O.K., J.W.K.)
| | - Hongki Yoo
- Department of Mechanical Engineering, KAIST, Daejeon, Korea (Y.H.K., H.S.N., H.Y.)
| | - Kyeongsoon Park
- Department of Systems Biotechnology, Chung-Ang University, Anseong, Gyeonggi, Korea (K.P.)
| | - Jin Won Kim
- BK21 Graduate Program, Department of Biomedical Sciences, Korea University College of Medicine, Seoul, Korea (J.H.K., J.W.K.)
- Multimodal Imaging and Theranostic Laboratory, Cardiovascular Center, Korea University Guro Hospital (J.H.K., J.W.S., H.J.K., R.H.K., Y.H.P., D.O.K., J.W.K.)
| |
Collapse
|
13
|
Wang Y, Nagase H, Tagawa YI, Taki S, Takamoto M. Endogenous IFN-γ facilitates Pneumocystis infection and downregulates carbohydrate receptors in CD4 + T cell-depleted mice. FEBS Lett 2024; 598:1633-1643. [PMID: 38631897 DOI: 10.1002/1873-3468.14875] [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: 11/16/2023] [Revised: 02/09/2024] [Accepted: 02/24/2024] [Indexed: 04/19/2024]
Abstract
IFN-γ plays a critical role in host defense against intracellular pathogens. IFN-γ is produced in the bronchoalveolar lavage fluid of mice infected with Pneumocystis, but the role of IFN-γ in host defense against Pneumocystis remains controversial. It has been previously reported that although exogenous IFN-γ has beneficial effects on eradication of Pneumocystis, endogenous IFN-γ has a negative impact on innate immunity in immunocompromised hosts. Surprisingly, CD4+ T cell-depleted IFN-γ deficient (GKO) mice exhibit resistance to Pneumocystis. Alveolar macrophages (AM) from GKO mice exhibit higher expression of macrophage mannose receptor (MMR) and Dectin-1. Concomitantly, they exhibited greater ability to phagocytize Pneumocystis, and this activity was suppressed by inhibitors of these receptors. Incubation with IFN-γ resulted in a reduction in both the expression of these receptors on AM and their Pneumocystis-phagocytic activity. These results indicate that endogenous IFN-γ facilitates Pneumocystis to escape from host innate immunity by attenuating the phagocytic activity of AM via downregulation of MMR and Dectin-1.
Collapse
MESH Headings
- Animals
- Mice
- CD4-Positive T-Lymphocytes/immunology
- CD4-Positive T-Lymphocytes/metabolism
- Down-Regulation
- Immunity, Innate
- Interferon-gamma/metabolism
- Interferon-gamma/immunology
- Interferon-gamma/genetics
- Lectins, C-Type/metabolism
- Lectins, C-Type/genetics
- Lectins, C-Type/immunology
- Lymphocyte Depletion
- Macrophages, Alveolar/immunology
- Macrophages, Alveolar/metabolism
- Macrophages, Alveolar/microbiology
- Mannose Receptor
- Mannose-Binding Lectins/metabolism
- Mannose-Binding Lectins/genetics
- Mice, Inbred C57BL
- Mice, Knockout
- Phagocytosis
- Pneumocystis/immunology
- Pneumocystis Infections/immunology
- Pneumocystis Infections/metabolism
- Pneumocystis Infections/microbiology
- Pneumocystis Infections/genetics
- Receptors, Cell Surface/genetics
- Receptors, Cell Surface/metabolism
- Receptors, Cell Surface/immunology
Collapse
Affiliation(s)
- Yi Wang
- Department of Infection and Host Defense, Shinshu University School of Medicine, Matsumoto, Japan
| | - Hisashi Nagase
- Department of Infection and Host Defense, Shinshu University School of Medicine, Matsumoto, Japan
| | - Yoh-Ichi Tagawa
- Department of Biomolecular Functional Engineering, Graduate School of Bioscience and Engineering, Tokyo Institute of Technology, Yokohama, Japan
| | - Shinsuke Taki
- Department of Molecular and Cellular Immunology, Shinshu University School of Medicine, Matsumoto, Japan
| | - Masaya Takamoto
- Department of Infection and Host Defense, Shinshu University School of Medicine, Matsumoto, Japan
- Community Health Care Research Center, Nagano University of Health and Medicine, Japan
| |
Collapse
|
14
|
Glass E, Robinson SL, Rosowski EE. Zebrafish use conserved CLR and TLR signaling pathways to respond to fungal PAMPs in zymosan. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.24.600417. [PMID: 38979385 PMCID: PMC11230284 DOI: 10.1101/2024.06.24.600417] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/10/2024]
Abstract
Pattern recognition receptors (PRRs) such as C-type lectin receptors (CLRs) and Toll-like receptors (TLRs) are used by hosts to recognize pathogen-associated molecular patterns (PAMPs) in microorganisms and to initiate innate immune responses. While PRRs exist across invertebrate and vertebrate species, the functional homology of many of these receptors is still unclear. In this study, we investigate the innate immune response of zebrafish larvae to zymosan, a β-glucan-containing particle derived from fungal cell walls. Macrophages and neutrophils robustly respond to zymosan and are required for zymosan-induced activation of the NF-κB transcription factor. Full activation of NF-κB in response to zymosan depends on Card9/Syk and Myd88, conserved CLR and TLR adaptor proteins, respectively. Two putative CLRs, Clec4c and Sclra, are both required for maximal sensing of zymosan and NF-κB activation. Altogether, we identify conserved PRRs and PRR signaling pathways in larval zebrafish that promote recognition of fungal PAMPs. These results inform modeling of human fungal infections in zebrafish and increase our knowledge of the evolution and conservation of PRR pathways in vertebrates.
Collapse
Affiliation(s)
- Erin Glass
- Department of Biological Sciences, Clemson University, Clemson, SC
- Eukaryotic Pathogens Innovation Center, Clemson University, Clemson, SC
| | - Stephan L Robinson
- Eukaryotic Pathogens Innovation Center, Clemson University, Clemson, SC
- School of Medicine Greenville, University of South Carolina, Greenville, SC
| | - Emily E Rosowski
- Department of Biological Sciences, Clemson University, Clemson, SC
- Eukaryotic Pathogens Innovation Center, Clemson University, Clemson, SC
| |
Collapse
|
15
|
Wzorek-Łyczko K, Woźniak W, Piwowarczyk A, Kuchar E. The anti-infective effect of β-glucans in children. INT J VITAM NUTR RES 2024; 94:296-307. [PMID: 37779363 DOI: 10.1024/0300-9831/a000793] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/03/2023]
Abstract
Background: β-glucans are bioactive β-D-glucose polysaccharides of natural origin, presenting antimicrobial and immunomodulation properties, with a low risk of toxicity. Objectives: This scoping review aims to present the current knowledge on the anti-infective properties of β-glucans in the pediatric population. Methods: We used the PRISMA Extension for Scoping Reviews Checklist to prepare this review. Studies were identified by electronic searches of Pubmed, Embase, and Cochrane databases up to May 2021. Results: The primary search allowed us to find 6232 studies, twelve of which were finally included in the analysis. Eight studies were designed as randomized, placebo-controlled trials, while in four studies the intervention outcome was compared with the pre-intervention period in the same group. The type of preparation and doses varied between studies: in five trials pleuran was administered (in dose 10 mg/5 kg of body weight/day), and in one study baker's yeast β-glucan was used (in two doses: 35 mg/day and 75 mg/day). In six other studies, the analyzed preparation comprised β-glucan and other substances. The shortest study lasted seven days, while the most prolonged intervention lasted six months, followed by six months of follow-up. Ten out of twelve trials demonstrated the effectiveness of β-glucans in reducing respiratory tract infection incidence or alleviation of upper respiratory tract infection symptoms. Ten out of twelve studies have reported a good tolerance and safety profile. Conclusions: Good tolerance of β-glucans shows a favorable benefit-risk ratio of this type of intervention. Nevertheless, further monitoring of their efficacy and safety in high-quality research is necessary.
Collapse
Affiliation(s)
- Katarzyna Wzorek-Łyczko
- The Department of Pediatrics with Clinical Assessment Unit, Medical University of Warsaw, Poland
| | - Weronika Woźniak
- The Department of Pediatrics with Clinical Assessment Unit, Medical University of Warsaw, Poland
| | - Anna Piwowarczyk
- The Department of Pediatrics with Clinical Assessment Unit, Medical University of Warsaw, Poland
| | - Ernest Kuchar
- The Department of Pediatrics with Clinical Assessment Unit, Medical University of Warsaw, Poland
| |
Collapse
|
16
|
Zhou X, Zhang X, Yu J. Gut mycobiome in metabolic diseases: Mechanisms and clinical implication. Biomed J 2024; 47:100625. [PMID: 37364760 PMCID: PMC11332988 DOI: 10.1016/j.bj.2023.100625] [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: 03/29/2023] [Revised: 05/22/2023] [Accepted: 06/21/2023] [Indexed: 06/28/2023] Open
Abstract
Obesity, type 2 diabetes mellitus (T2DM) and non-alcoholic fatty liver disease (NAFLD) are three common metabolic diseases with high prevalence worldwide. Emerging evidence suggests that gut dysbiosis may influence the development of metabolic diseases, in which gut fungal microbiome (mycobiome) is actively involved. In this review, we summarize the studies exploring the composition changes of gut mycobiome in metabolic diseases and mechanisms by which fungi affect the development of metabolic diseases. The current mycobiome-based therapies, including probiotic fungi, fungal products, anti-fungal agents and fecal microbiota transplantation (FMT), and their implication in treating metabolic diseases are discussed. We highlight the unique role of gut mycobiome in metabolic diseases, providing perspectives for future research on gut mycobiome in metabolic diseases.
Collapse
Affiliation(s)
- Xingyu Zhou
- Department of Medicine and Therapeutics, Institute of Digestive Disease, State Key Laboratory of Digestive Disease, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Xiang Zhang
- Department of Medicine and Therapeutics, Institute of Digestive Disease, State Key Laboratory of Digestive Disease, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Jun Yu
- Department of Medicine and Therapeutics, Institute of Digestive Disease, State Key Laboratory of Digestive Disease, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong SAR, China.
| |
Collapse
|
17
|
Chen T, Gao C. Innate immune signal transduction pathways to fungal infection: Components and regulation. CELL INSIGHT 2024; 3:100154. [PMID: 38464417 PMCID: PMC10924179 DOI: 10.1016/j.cellin.2024.100154] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/19/2023] [Revised: 01/30/2024] [Accepted: 01/30/2024] [Indexed: 03/12/2024]
Abstract
Candida species are significant causes of mucosal and systemic infections in immune compromised populations, including HIV-infected individuals and cancer patients. Drug resistance and toxicity have limited the use of anti-fungal drugs. A good comprehension of the nature of the immune responses to the pathogenic fungi will aid in the developing of new approaches to the treatment of fungal diseases. In recent years, extensive research has been done to understand the host defending systems to fungal infections. In this review, we described how pattern recognition receptors senses the cognate fungal ligands and the cellular and molecular mechanisms of anti-fungal innate immune responses. Furthermore, particular focus is placed on how anti-fungal signal transduction cascades are being activated for host defense and being modulated to better treat the infections in terms of immunotherapy. Understanding the role that these pathways have in mediating host anti-fungal immunity will be crucial for future therapeutic development.
Collapse
Affiliation(s)
- Tian Chen
- Key Laboratory of Infection and Immunity of Shandong Province & Key Laboratory for Experimental Teratology of Ministry of Education, Shandong University, Jinan, 250012, Shandong, China
- Department of Pathogenic Biology, School of Biomedical Sciences, Shandong University, Jinan, 250012, Shandong, China
| | - Chengjiang Gao
- Key Laboratory of Infection and Immunity of Shandong Province & Key Laboratory for Experimental Teratology of Ministry of Education, Shandong University, Jinan, 250012, Shandong, China
- Department of Immunology, School of Biomedical Sciences, Shandong University, Jinan, 250012, Shandong, China
| |
Collapse
|
18
|
Chen Y, Li H, Zhu L, Yang Q, Zhou J. β-Glucan Subverts the Function of Myeloid Cells in Neonates. J Immunol Res 2024; 2024:2765001. [PMID: 38774603 PMCID: PMC11108693 DOI: 10.1155/2024/2765001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2023] [Revised: 03/12/2024] [Accepted: 04/29/2024] [Indexed: 05/24/2024] Open
Abstract
β-Glucan is the main component of the cell wall of pathogen-associated molecular patterns (PAMPs) including various yeast, fungi, or certain bacteria. Previous reports demonstrated that β-glucan was widely investigated as a potent immunomodulators to stimulate innate and adaptive immune responses, which indicated that it could be recommended as an effective adjuvant in immunotherapy. However, the detailed effects of β-glucan on neonatal immunity are still largely unknown. Here, we found that β-glucan did not affect the frequencies and numbers of myeloid cells in the spleen and bone marrow from neonates. Functional assay revealed that β-glucan from neonates compromised the immunosuppressive function of immature myeloid cells, which were myeloid-derived suppressor cells (MDSCs). Flow cytometry or gene expression analysis revealed that β-glucan-derived polymorphonuclear (PMN)-MDSCs produced lower level of reactive oxygen species (ROS) and arginase-1 (Arg1) in neonatal mice. Furthermore, β-glucan administration significantly decreased the frequency and ROS level of PMN-MDSCs in vitro. These observations suggest that β-glucan facilitates the maturation of myeloid cells in early life, which may contribute to its beneficial effects against immune disorders later in life.
Collapse
Affiliation(s)
- Yingying Chen
- Key Laboratory of Immunology, Sino-French Hoffmann Institute, School of Basic Medical Sciences, Guangdong Provincial Key Laboratory of Allergy and Clinical Immunology, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou 511436, China
- Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510000, China
- Department of Clinical Laboratory, The Key Laboratory of Advanced Interdisciplinary Studies Center, The First Affiliated Hospital of Guangzhou Medical University, National Center for Respiratory Medicine, National Clinical Research Center for Respiratory Disease, Guangzhou, Guangdong 510120, China
| | - Hui Li
- Tianjin Institute of Immunology, Key Laboratory of Immune Microenvironment and Disease of the Ministry of Education, Department of Immunology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin 300070, China
| | - Lin Zhu
- Tianjin Institute of Immunology, Key Laboratory of Immune Microenvironment and Disease of the Ministry of Education, Department of Immunology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin 300070, China
| | - Quan Yang
- Key Laboratory of Immunology, Sino-French Hoffmann Institute, School of Basic Medical Sciences, Guangdong Provincial Key Laboratory of Allergy and Clinical Immunology, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou 511436, China
| | - Jie Zhou
- Tianjin Institute of Immunology, Key Laboratory of Immune Microenvironment and Disease of the Ministry of Education, Department of Immunology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin 300070, China
| |
Collapse
|
19
|
Carroll SL, Pasare C, Barton GM. Control of adaptive immunity by pattern recognition receptors. Immunity 2024; 57:632-648. [PMID: 38599163 PMCID: PMC11037560 DOI: 10.1016/j.immuni.2024.03.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2024] [Revised: 03/12/2024] [Accepted: 03/13/2024] [Indexed: 04/12/2024]
Abstract
One of the most significant conceptual advances in immunology in recent history is the recognition that signals from the innate immune system are required for induction of adaptive immune responses. Two breakthroughs were critical in establishing this paradigm: the identification of dendritic cells (DCs) as the cellular link between innate and adaptive immunity and the discovery of pattern recognition receptors (PRRs) as a molecular link that controls innate immune activation as well as DC function. Here, we recount the key events leading to these discoveries and discuss our current understanding of how PRRs shape adaptive immune responses, both indirectly through control of DC function and directly through control of lymphocyte function. In this context, we provide a conceptual framework for how variation in the signals generated by PRR activation, in DCs or other cell types, can influence T cell differentiation and shape the ensuing adaptive immune response.
Collapse
Affiliation(s)
- Shaina L Carroll
- Division of Immunology & Molecular Medicine, Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA USA
| | - Chandrashekhar Pasare
- Division of Immunobiology and Center for Inflammation and Tolerance, Cincinnati Children's Hospital Medical Center, Department of Pediatrics, University of Cincinnati, College of Medicine, Cincinnati, OH USA
| | - Gregory M Barton
- Division of Immunology & Molecular Medicine, Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA USA; Howard Hughes Medical Institute, University of California, Berkeley, Berkeley, CA 94720 USA.
| |
Collapse
|
20
|
Boulifa A, Raftery MJ, Franzén AS, Radecke C, Stintzing S, Blohmer JU, Pecher G. Role of beta-(1→3)(1→6)-D-glucan derived from yeast on natural killer (NK) cells and breast cancer cell lines in 2D and 3D cultures. BMC Cancer 2024; 24:339. [PMID: 38486205 PMCID: PMC10938759 DOI: 10.1186/s12885-024-11979-3] [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: 08/25/2023] [Accepted: 02/07/2024] [Indexed: 03/18/2024] Open
Abstract
BACKGROUND Beta-(1,3)(1,6)-D-glucan is a complex polysaccharide, which is found in the cell wall of various fungi, yeasts, bacteria, algae, barley, and oats and has immunomodulatory, anticancer and antiviral effects. In the present study, we investigated the effect of beta-(1,3)(1,6)-D-glucan derived from yeast on the proliferation of primary NK cells and breast cancer cell lines in 2D and 3D models, and on the cytotoxicity of primary NK cells against breast cancer cell lines in 2D and 3D models. METHODS In this study, we investigated the effects of different concentrations of yeast-derived beta-(1→3)(1→6)-D-glucan on the proliferation and cytotoxicity of human NK cells and breast cancer cell lines in 2D and 3D models using the XTT cell proliferation assay and the CellTiter-Glo® 2.0 assay to determine the cytotoxicity of human NK cells on breast cancer cell lines in 2D and 3D models. RESULTS We found that the co-incubation of NK cells with beta-glucan in the absence of IL2 at 48 h significantly increased the proliferation of NK cells, whereas the co-incubation of NK cells with beta-glucan in the presence of IL2 (70 U/ml) increased the proliferation of NK cells but not significantly. Moreover, beta-glucan significantly inhibited the proliferation of breast cancer cell lines in 2D model and induced a weak, non-significant growth inhibitory effect on breast cancer multicellular tumor spheroids (3D). In addition, the cytotoxicity of NK cells against breast cancer cell lines was examined in 2D and 3D models, and beta-glucan significantly increased the cytotoxicity of NK cells against MCF-7 (in 2D). CONCLUSIONS Yeast derived beta-(1,3)(1,6)-D-glucan could contribute to the treatment of cancer by enhancing NK cell immune response as well as contributing to inhibition of breast cancer cell growth.
Collapse
Affiliation(s)
- Abdelhadi Boulifa
- Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Charitéplatz 1, Berlin, 10117, Germany
- Competence Center of Immuno-Oncology and Translational Cell Therapy (KITZ), Department of Hematology, Oncology and Tumor Immunology, CCM, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Charitéplatz 1, Berlin, 10117, Germany
| | - Martin J Raftery
- Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Charitéplatz 1, Berlin, 10117, Germany
- Competence Center of Immuno-Oncology and Translational Cell Therapy (KITZ), Department of Hematology, Oncology and Tumor Immunology, CCM, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Charitéplatz 1, Berlin, 10117, Germany
| | - Alexander Sebastian Franzén
- Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Charitéplatz 1, Berlin, 10117, Germany
- Competence Center of Immuno-Oncology and Translational Cell Therapy (KITZ), Department of Hematology, Oncology and Tumor Immunology, CCM, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Charitéplatz 1, Berlin, 10117, Germany
| | - Clarissa Radecke
- Competence Center of Immuno-Oncology and Translational Cell Therapy (KITZ), Department of Hematology, Oncology and Tumor Immunology, CCM, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Charitéplatz 1, Berlin, 10117, Germany
| | - Sebastian Stintzing
- Competence Center of Immuno-Oncology and Translational Cell Therapy (KITZ), Department of Hematology, Oncology and Tumor Immunology, CCM, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Charitéplatz 1, Berlin, 10117, Germany
| | - Jens-Uwe Blohmer
- Department of Gynecology with Breast Center Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Charitéplatz 1, Berlin, 10117, Germany
| | - Gabriele Pecher
- Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Charitéplatz 1, Berlin, 10117, Germany.
- Competence Center of Immuno-Oncology and Translational Cell Therapy (KITZ), Department of Hematology, Oncology and Tumor Immunology, CCM, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Charitéplatz 1, Berlin, 10117, Germany.
| |
Collapse
|
21
|
Ravi S, Martin LC, Krishnan M, Kumaresan M, Manikandan B, Ramar M. Interactions between macrophage membrane and lipid mediators during cardiovascular diseases with the implications of scavenger receptors. Chem Phys Lipids 2024; 258:105362. [PMID: 38006924 DOI: 10.1016/j.chemphyslip.2023.105362] [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: 10/09/2023] [Revised: 11/06/2023] [Accepted: 11/20/2023] [Indexed: 11/27/2023]
Abstract
The onset and progression of cardiovascular diseases with the major underlying cause being atherosclerosis, occur during chronic inflammatory persistence in the vascular system, especially within the arterial wall. Such prolonged maladaptive inflammation is driven by macrophages and their key mediators are generally attributed to a disparity in lipid metabolism. Macrophages are the primary cells of innate immunity, endowed with expansive membrane domains involved in immune responses with their signalling systems. During atherosclerosis, the membrane domains and receptors control various active organisations of macrophages. Their scavenger/endocytic receptors regulate the trafficking of intracellular and extracellular cargo. Corresponding influence on lipid metabolism is mediated by their dynamic interaction with scavenger membrane receptors and their integrated mechanisms such as pinocytosis, phagocytosis, cholesterol export/import, etc. This interaction not only results in the functional differentiation of macrophages but also modifies their structural configurations. Here, we reviewed the association of macrophage membrane biomechanics and their scavenger receptor families with lipid metabolites during the event of atherogenesis. In addition, the membrane structure of macrophages and the signalling pathways involved in endocytosis integrated with lipid metabolism are detailed. This article establishes future insights into the scavenger receptors as potential targets for cardiovascular disease prevention and treatment.
Collapse
Affiliation(s)
- Sangeetha Ravi
- Department of Zoology, University of Madras, Guindy Campus, Chennai 600 025, India
| | | | - Mahalakshmi Krishnan
- Department of Zoology, University of Madras, Guindy Campus, Chennai 600 025, India
| | - Manikandan Kumaresan
- Department of Zoology, University of Madras, Guindy Campus, Chennai 600 025, India
| | - Beulaja Manikandan
- Department of Biochemistry, Annai Veilankanni's College for Women, Chennai 600 015, India
| | - Manikandan Ramar
- Department of Zoology, University of Madras, Guindy Campus, Chennai 600 025, India.
| |
Collapse
|
22
|
Dong QQ, Wu Q, Lu Y, Shi Y, Yang KD, Xu XL, Chen W. Exploring β-glucan as a micro-nano system for oral delivery targeted the colon. Int J Biol Macromol 2023; 253:127360. [PMID: 37827417 DOI: 10.1016/j.ijbiomac.2023.127360] [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: 06/26/2023] [Revised: 09/27/2023] [Accepted: 10/09/2023] [Indexed: 10/14/2023]
Abstract
The critical role of oral colon-specific delivery systems (OCDDS) is important for delivering active agents to the colon and rectum specifically via the oral route. The use of micro/nanostructured OCDDS further improves drug stability, bioavailability, and retention time, leading to enhanced therapeutic effects. However, designing micro/nanoscale OCDDSs is challenging due to pH changes, enzymatic degradation, and systemic absorption and metabolism. Biodegradable natural polysaccharides are a promising solution to these problems, and β-glucan is one of the most promising natural polysaccharides due to its unique structural features, conformational flexibility, and specific processing properties. This review covers the diverse chemical structures of β-glucan, its benefits (biocompatibility, easy modification, and colon-specific degradation), and various β-glucan-based micro/nanosized OCDDSs, as well as their drawbacks. The potential of β-glucan offers exciting new opportunities for colon-specific drug delivery.
Collapse
Affiliation(s)
- Qing-Qing Dong
- Longhua Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai 200032, PR China; Shulan International Medical College, Zhejiang Shuren University, Hangzhou 310015, PR China
| | - Qian Wu
- Longhua Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai 200032, PR China
| | - Yi Lu
- Longhua Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai 200032, PR China
| | - Yi Shi
- Longhua Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai 200032, PR China
| | - Ke-Da Yang
- Shulan International Medical College, Zhejiang Shuren University, Hangzhou 310015, PR China
| | - Xiao-Ling Xu
- Shulan International Medical College, Zhejiang Shuren University, Hangzhou 310015, PR China.
| | - Wei Chen
- Longhua Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai 200032, PR China.
| |
Collapse
|
23
|
Park SY, Kim KJ, Jo SM, Jeon JY, Kim BR, Hwang JE, Kim JY. Euglena gracilis (Euglena) powder supplementation enhanced immune function through natural killer cell activity in apparently healthy participants: A randomized, double-blind, placebo-controlled trial. Nutr Res 2023; 119:90-97. [PMID: 37769481 DOI: 10.1016/j.nutres.2023.09.004] [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: 05/25/2023] [Revised: 09/07/2023] [Accepted: 09/07/2023] [Indexed: 10/03/2023]
Abstract
Euglena gracilis (Euglena) is a microalgae found in most freshwater environments that produces paramylon, an insoluble β-1,3-glucan linked to human immunity. We hypothesized that Euglena powder has effects on immune function in apparently healthy adults. The study included male or female volunteers between the ages of 20 and 70 years who had white blood cell counts ranging from 4 × 103/µL to 10 × 103/µL, a "severe" rating on the stress questionnaire from the Korea National Health and Nutrition Examination Survey, and at least 2 upper respiratory infections with cold-like symptoms in the previous year. Participants received either a placebo or 700 mg of Euglena powder daily for 8 weeks. The study measured natural killer cell activity, cytokine concentrations, and blood lipid profiles to confirm the immune effect of Euglena consumption. In conclusion, Euglena improved immunological function through natural killer cell activity. Safety assessment showed no significant changes in vital signs or clinical chemistry indicators, and there were no adverse events associated with Euglena consumption. Euglena supplementation may help boost the immune systems of healthy individuals.
Collapse
Affiliation(s)
- Soo-Yeon Park
- Department of Food Science and Technology, Seoul National University of Science and Technology, Seoul, 01811, Republic of Korea
| | - Kyeong Jin Kim
- Department of Nano Bio Engineering, Seoul National University of Science and Technology, Seoul, 01811, Republic of Korea
| | - So Min Jo
- Department of Food Science and Technology, Seoul National University of Science and Technology, Seoul, 01811, Republic of Korea
| | - Jin-Young Jeon
- BIO R&D Center, Daesang Corp., Seoul, 07789, Republic of Korea
| | - Bo-Ra Kim
- BIO R&D Center, Daesang Corp., Seoul, 07789, Republic of Korea
| | - Ji Eun Hwang
- BIO R&D Center, Daesang Corp., Seoul, 07789, Republic of Korea
| | - Ji Yeon Kim
- Department of Food Science and Technology, Seoul National University of Science and Technology, Seoul, 01811, Republic of Korea; Department of Nano Bio Engineering, Seoul National University of Science and Technology, Seoul, 01811, Republic of Korea.
| |
Collapse
|
24
|
Generalov E, Yakovenko L. Receptor basis of biological activity of polysaccharides. Biophys Rev 2023; 15:1209-1222. [PMID: 37975017 PMCID: PMC10643635 DOI: 10.1007/s12551-023-01102-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2023] [Accepted: 07/19/2023] [Indexed: 11/19/2023] Open
Abstract
Polysaccharides, the most diverse forms of organic molecules in nature, exhibit a large number of different biological activities, such as immunomodulatory, radioprotective, antioxidant, regenerative, metabolic, signaling, antitumor, and anticoagulant. The reaction of cells to a polysaccharide is determined by its specific interaction with receptors present on the cell surface, the type of cells, and their condition. The effect of many polysaccharides depends non-linearly on their concentration. The same polysaccharide in different conditions can have very different effects on cells and organisms, up to the opposite; therefore, when conducting studies of the biological activity of polysaccharides, both for the purpose of developing new drugs or approaches to the treatment of patients, and in order to clarify the features of intracellular processes, information about already known research results is needed. There is a lot of scattered data on the biological activities of polysaccharides, but there are few reviews that would consider natural polysaccharides from various sources and possible molecular mechanisms of their action. The purpose of this review is to present the main results published at different times in order to facilitate the search for information necessary for conducting relevant studies.
Collapse
Affiliation(s)
- Evgenii Generalov
- Faculty of Physics, M.V. Lomonosov Moscow State University, Moscow, 119991 Russia
| | - Leonid Yakovenko
- Faculty of Physics, M.V. Lomonosov Moscow State University, Moscow, 119991 Russia
| |
Collapse
|
25
|
Zhang Y, Tang W, Zheng Z, Nie G, Zhan Y, Mu X, Liu Y, Wang K. Metabolic degradation of polysaccharides from Lentinus edodes by Kupffer cells via the Dectin-1/Syk signaling pathway. Carbohydr Polym 2023; 317:121108. [PMID: 37364942 DOI: 10.1016/j.carbpol.2023.121108] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2023] [Revised: 06/05/2023] [Accepted: 06/08/2023] [Indexed: 06/28/2023]
Abstract
It had been shown that lentinan (LNT) was mainly distributed in the liver after intravenous administration. The study aimed to investigate the integrated metabolic processes and mechanisms of LNT in the liver, as these have not been thoroughly explored. In current work, 5-([4,6-dichlorotriazin-2-yl] amino) fluorescein and cyanine 7 were used to label LNT for tracking its metabolic behavior and mechanisms. Near-infrared imaging demonstrated that LNT was captured mainly by the liver. Kupffer cell (KC) depletion reduced LNT liver localization and degradation in BALB/c mice. Moreover, experiments with Dectin-1 siRNA and Dectin-1/Syk signaling pathway inhibitors showed that LNT was mainly taken up by KCs via the Dectin-1/Syk pathway and promoted lysosomal maturation in KCs via this same pathway, which in turn promoted LNT degradation. These empirical findings offer novel insights into the metabolism of LNT in vivo and in vitro, which will facilitate the further application of LNT and other β-glucans.
Collapse
Affiliation(s)
- 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, China
| | - Wenqi Tang
- Department of Pharmacy, Union Hospital, Tongji Medical College, 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, China
| | - Gang Nie
- Department of Pharmacy, Wuhan Children's Hospital (Wuhan Maternal and Child Healthcare Hospital), Tongji Medical College, Huazhong University of Science and Technology, 430019 Wuhan, China
| | - Yuxue Zhan
- Department of Pharmacy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 430030 Wuhan, China
| | - Xu Mu
- 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
| | - 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.
| |
Collapse
|
26
|
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
|
27
|
Sung M, Yoon Y, Lee J. The Immunomodulatory Effect of β-Glucan Depends on the Composition of the Gut Microbiota. Foods 2023; 12:3148. [PMID: 37685079 PMCID: PMC10487241 DOI: 10.3390/foods12173148] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2023] [Revised: 08/11/2023] [Accepted: 08/17/2023] [Indexed: 09/10/2023] Open
Abstract
This study aimed to elucidate the relationship between the immunomodulatory effects of β-glucan and the composition of gut microbiota in mice. The mice were fed a diet containing β-glucan for 3 weeks, and feces, blood, and tissues were then collected to analyze the immunomodulatory effect and gut microbiota composition. Based on the results of the analysis of the expression level of immune-associated proteins, the high immunomodulatory effect group (HIE) and low immunomodulatory effect group (LIE) were categorized. Before the β-glucan diet, the proportions of the phylum Bacteroidota, family Muribaculaceae, and family Lactobacillaceae were significantly higher in HIE than in LIE. Furthermore, the genus Akkermansia was absent before the β-glucan diet and increased after β-glucan diet. These microbes had the ability to metabolize β-glucan or were beneficial to health. In conclusion, our findings demonstrate that variation in the composition of gut microbiota among individuals can result in varying expressions of β-glucan functionality. This outcome supports the notion that β-glucan may be metabolized through diverse pathways by gut microbes originally possessed by mice, subsequently producing various metabolites, such as short-chain fatty acids. Alternatively, the viscosity of the intestinal mucosa could be enhanced by β-glucan, potentially promoting the growth of certain bacteria (e.g., the genus Akkermansia). This study provides insights into the intricate interplay between β-glucan, gut microbiota, and immunomodulation.
Collapse
Affiliation(s)
- Miseon Sung
- Department of Food and Nutrition, Sookmyung Women’s University, Seoul 04310, Republic of Korea; (M.S.); (Y.Y.)
| | - Yohan Yoon
- Department of Food and Nutrition, Sookmyung Women’s University, Seoul 04310, Republic of Korea; (M.S.); (Y.Y.)
- Risk Analysis Research Center, Sookmyung Women’s University, Seoul 04310, Republic of Korea
| | - Jeeyeon Lee
- Department of Food & Nutrition, Dong-eui University, Busan 47340, Republic of Korea
| |
Collapse
|
28
|
Chen T, Feng Y, Sun W, Zhao G, Wu H, Cheng X, Zhao F, Zhang L, Zheng Y, Zhan P, Zhao W, Liu B, Gao C. The nucleotide receptor STING translocates to the phagosomes to negatively regulate anti-fungal immunity. Immunity 2023; 56:1727-1742.e6. [PMID: 37379835 DOI: 10.1016/j.immuni.2023.06.002] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2022] [Revised: 04/26/2023] [Accepted: 06/02/2023] [Indexed: 06/30/2023]
Abstract
STING (stimulator of interferon genes) exerts protective cellular responses to viral infection via induction of interferon production and autophagy. Here, we report the role of STING in modulating the immune responses toward fungal infection. Upon Candida albicans stimulation, STING transited alongside the endoplasmic reticulum (ER) to the phagosomes. In phagosomes, STING directly bound with Src via the N-terminal 18 amino acids of STING, and this binding prevented Src from recruiting and phosphorylating Syk. Consistently, Syk-associated signaling and production of pro-inflammatory cytokines and chemokines were increased in mouse BMDCs (bone-marrow-derived dendritic cells) lacking STING with fungal treatment. STING deficiency improved anti-fungal immunity in systemic C. albicans infection. Importantly, administration of the N-terminal 18-aa (amino acid) peptide of STING improved host outcomes in disseminated fungal infection. Overall, our study identifies a previously unrecognized function of STING in negatively regulating anti-fungal immune responses and offers a potential therapeutic strategy for controlling C. albicans infection.
Collapse
Affiliation(s)
- Tian Chen
- Key Laboratory of Infection and Immunity of Shandong Province & Key Laboratory for Experimental Teratology of Ministry of Education, Shandong University, Jinan 250012, Shandong, P.R. China; Department of Pathogenic Biology, School of Biomedical Sciences, Shandong University, Jinan 250012, Shandong, P.R. China
| | - Yiting Feng
- Key Laboratory of Infection and Immunity of Shandong Province & Key Laboratory for Experimental Teratology of Ministry of Education, Shandong University, Jinan 250012, Shandong, P.R. China; Department of Immunology, School of Biomedical Sciences, Shandong University, Jinan 250012, Shandong, P.R. China
| | - Wanwei Sun
- Key Laboratory of Infection and Immunity of Shandong Province & Key Laboratory for Experimental Teratology of Ministry of Education, Shandong University, Jinan 250012, Shandong, P.R. China; Department of Immunology, School of Biomedical Sciences, Shandong University, Jinan 250012, Shandong, P.R. China
| | - Guimin Zhao
- Key Laboratory of Infection and Immunity of Shandong Province & Key Laboratory for Experimental Teratology of Ministry of Education, Shandong University, Jinan 250012, Shandong, P.R. China; Department of Immunology, School of Biomedical Sciences, Shandong University, Jinan 250012, Shandong, P.R. China
| | - Han Wu
- Key Laboratory of Infection and Immunity of Shandong Province & Key Laboratory for Experimental Teratology of Ministry of Education, Shandong University, Jinan 250012, Shandong, P.R. China; Department of Immunology, School of Biomedical Sciences, Shandong University, Jinan 250012, Shandong, P.R. China
| | - Xiaochen Cheng
- Key Laboratory of Infection and Immunity of Shandong Province & Key Laboratory for Experimental Teratology of Ministry of Education, Shandong University, Jinan 250012, Shandong, P.R. China; Department of Immunology, School of Biomedical Sciences, Shandong University, Jinan 250012, Shandong, P.R. China
| | - Fabao Zhao
- Department of Medicinal Chemistry, School of Pharmaceutical Sciences, Shandong University, Jinan 250012, P.R. China
| | - Lei Zhang
- Key Laboratory of Infection and Immunity of Shandong Province & Key Laboratory for Experimental Teratology of Ministry of Education, Shandong University, Jinan 250012, Shandong, P.R. China; Department of Immunology, School of Biomedical Sciences, Shandong University, Jinan 250012, Shandong, P.R. China
| | - Yi Zheng
- Key Laboratory of Infection and Immunity of Shandong Province & Key Laboratory for Experimental Teratology of Ministry of Education, Shandong University, Jinan 250012, Shandong, P.R. China; Department of Immunology, School of Biomedical Sciences, Shandong University, Jinan 250012, Shandong, P.R. China
| | - Peng Zhan
- Department of Medicinal Chemistry, School of Pharmaceutical Sciences, Shandong University, Jinan 250012, P.R. China
| | - Wei Zhao
- Key Laboratory of Infection and Immunity of Shandong Province & Key Laboratory for Experimental Teratology of Ministry of Education, Shandong University, Jinan 250012, Shandong, P.R. China; Department of Pathogenic Biology, School of Biomedical Sciences, Shandong University, Jinan 250012, Shandong, P.R. China
| | - Bingyu Liu
- Key Laboratory of Infection and Immunity of Shandong Province & Key Laboratory for Experimental Teratology of Ministry of Education, Shandong University, Jinan 250012, Shandong, P.R. China; Department of Immunology, School of Biomedical Sciences, Shandong University, Jinan 250012, Shandong, P.R. China
| | - Chengjiang Gao
- Key Laboratory of Infection and Immunity of Shandong Province & Key Laboratory for Experimental Teratology of Ministry of Education, Shandong University, Jinan 250012, Shandong, P.R. China; Department of Immunology, School of Biomedical Sciences, Shandong University, Jinan 250012, Shandong, P.R. China.
| |
Collapse
|
29
|
Hasheminasab SS, Conejeros I, Gärtner U, Kamena F, Taubert A, Hermosilla CR. MCT-Dependent Cryptosporidium parvum-Induced Bovine Monocyte Extracellular Traps (METs) under Physioxia. BIOLOGY 2023; 12:961. [PMID: 37508391 PMCID: PMC10376234 DOI: 10.3390/biology12070961] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/15/2023] [Revised: 06/26/2023] [Accepted: 07/03/2023] [Indexed: 07/30/2023]
Abstract
The apicomplexan protozoan parasite Cryptosporidium parvum is responsible for cryptosporidiosis, which is a zoonotic intestinal illness that affects newborn cattle, wild animals, and people all over the world. Mammalian monocytes are bone marrow-derived myeloid leukocytes with important defense effector functions in early host innate immunity due to their ATP purinergic-, CD14- and CD16-receptors, adhesion, migration and phagocytosis capacities, inflammatory, and anti-parasitic properties. The formation of monocyte extracellular traps (METs) has recently been reported as an additional effector mechanism against apicomplexan parasites. Nonetheless, nothing is known in the literature on METs extrusion neither towards C. parvum-oocysts nor sporozoites. Herein, ATP purinergic receptor P2X1, glycolysis, Notch signaling, and lactate monocarboxylate transporters (MCT) were investigated in C. parvum-exposed bovine monocytes under intestinal physioxia (5% O2) and hyperoxia (21% O2; most commonly used hyperoxic laboratory conditions). C. parvum-triggered suicidal METs were confirmed by complete rupture of exposed monocytes, co-localization of extracellular DNA with myeloperoxidase (MPO) and histones (H1-H4) via immunofluorescence- and confocal microscopy analyses. C. parvum-induced suicidal METs resulted not only in oocyst entrapment but also in hindered sporozoite mobility from oocysts according to scanning electron microscopy (SEM) analyses. Early parasite-induced bovine monocyte activation, accompanied by membrane protrusions toward C. parvum-oocysts/sporozoites, was unveiled using live cell 3D-holotomographic microscopy analysis. The administration of NF449, an inhibitor of the ATP purinergic receptor P2X1, to monocytes subjected to varying oxygen concentrations did not yield a noteworthy decrease in C. parvum-induced METosis. This suggests that the cell death process is not dependent on P2X1. Additionally, blockage of glycolysis in monocyte through 2-deoxy glucose (2-DG) inhibition reduced C. parvum-induced METosis but not significantly. According to monocyte energetic state measurements, C. parvum-exposed cells neither increased extracellular acidification rates (ECAR) nor oxygen consumption rates (OCR). Lactate monocarboxylate transporters (MCT) inhibitor (i.e., AR-C 141990) treatments significantly diminished C. parvum-mediated METs extrusion under physioxic (5% O2) condition. Similarly, treatment with either DAPT or compound E, two selective Notch inhibitors, exhibited no significant suppressive effects on bovine MET production. Overall, for the first time, we demonstrate C. parvum-mediated METosis as P2X1-independent but as an MCT-dependent defense mechanism under intestinal physioxia (5% CO2) conditions. METs findings suggest anti-cryptosporidial effects through parasite entrapment and inhibition of sporozoite excystation.
Collapse
Affiliation(s)
- Seyed Sajjad Hasheminasab
- Institute of Parasitology, Biomedical Research Center Seltersberg (BFS), Justus Liebig University Giessen, 35392 Giessen, Germany
| | - Iván Conejeros
- Institute of Parasitology, Biomedical Research Center Seltersberg (BFS), Justus Liebig University Giessen, 35392 Giessen, Germany
| | - Ulrich Gärtner
- Institute of Anatomy and Cell Biology, Justus Liebig University Giessen, 35392 Giessen, Germany
| | - Faustin Kamena
- Laboratory for Molecular Parasitology, Department of Microbiology and Parasitology, University of Buea, Buea P.O. Box 63, Cameroon
| | - Anja Taubert
- Institute of Parasitology, Biomedical Research Center Seltersberg (BFS), Justus Liebig University Giessen, 35392 Giessen, Germany
| | - Carlos R Hermosilla
- Institute of Parasitology, Biomedical Research Center Seltersberg (BFS), Justus Liebig University Giessen, 35392 Giessen, Germany
| |
Collapse
|
30
|
Chen YH, Yeung F, Lacey KA, Zaldana K, Lin JD, Bee GCW, McCauley C, Barre RS, Liang SH, Hansen CB, Downie AE, Tio K, Weiser JN, Torres VJ, Bennett RJ, Loke P, Graham AL, Cadwell K. Rewilding of laboratory mice enhances granulopoiesis and immunity through intestinal fungal colonization. Sci Immunol 2023; 8:eadd6910. [PMID: 37352372 PMCID: PMC10350741 DOI: 10.1126/sciimmunol.add6910] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2022] [Accepted: 05/31/2023] [Indexed: 06/25/2023]
Abstract
The paucity of blood granulocyte populations such as neutrophils in laboratory mice is a notable difference between this model organism and humans, but the cause of this species-specific difference is unclear. We previously demonstrated that laboratory mice released into a seminatural environment, referred to as rewilding, display an increase in blood granulocytes that is associated with expansion of fungi in the gut microbiota. Here, we find that tonic signals from fungal colonization induce sustained granulopoiesis through a mechanism distinct from emergency granulopoiesis, leading to a prolonged expansion of circulating neutrophils that promotes immunity. Fungal colonization after either rewilding or oral inoculation of laboratory mice with Candida albicans induced persistent expansion of myeloid progenitors in the bone marrow. This increase in granulopoiesis conferred greater long-term protection from bloodstream infection by gram-positive bacteria than by the trained immune response evoked by transient exposure to the fungal cell wall component β-glucan. Consequently, introducing fungi into laboratory mice may restore aspects of leukocyte development and provide a better model for humans and free-living mammals that are constantly exposed to environmental fungi.
Collapse
Affiliation(s)
- Ying-Han Chen
- Kimmel Center for Biology and Medicine at the Skirball Institute
| | - Frank Yeung
- Kimmel Center for Biology and Medicine at the Skirball Institute
| | - Keenan A. Lacey
- Department of Microbiology, New York University Grossman School of Medicine, New York, NY 10016, USA
| | - Kimberly Zaldana
- Kimmel Center for Biology and Medicine at the Skirball Institute
| | - Jian-Da Lin
- Department of Biochemical Science and Technology, College of Life Science, National Taiwan University, Taipei City, Taiwan
| | - Gavyn Chern Wei Bee
- Department of Microbiology, New York University Grossman School of Medicine, New York, NY 10016, USA
| | - Caroline McCauley
- Department of Microbiology, New York University Grossman School of Medicine, New York, NY 10016, USA
| | - Ramya S. Barre
- Department of Ecology and Evolutionary Biology, Princeton University, Princeton, New Jersey, USA
| | - Shen-Huan Liang
- Department of Molecular Microbiology and Immunology, Brown University, Providence, RI, USA
| | - Christina B. Hansen
- Department of Ecology and Evolutionary Biology, Princeton University, Princeton, New Jersey, USA
| | - Alexander E Downie
- Department of Ecology and Evolutionary Biology, Princeton University, Princeton, New Jersey, USA
| | - Kyle Tio
- Kimmel Center for Biology and Medicine at the Skirball Institute
| | - Jeffrey N. Weiser
- Antimicrobial-Resistant Pathogens Program
- Department of Microbiology, New York University Grossman School of Medicine, New York, NY 10016, USA
| | - Victor J Torres
- Antimicrobial-Resistant Pathogens Program
- Department of Microbiology, New York University Grossman School of Medicine, New York, NY 10016, USA
| | - Richard J. Bennett
- Department of Molecular Microbiology and Immunology, Brown University, Providence, RI, USA
| | - P’ng Loke
- Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Andrea L. Graham
- Department of Ecology and Evolutionary Biology, Princeton University, Princeton, New Jersey, USA
| | - Ken Cadwell
- Division of Gastroenterology and Hepatology, Department of Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104
- Department of Systems Pharmacology and Translational Therapeutics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104
- Department of Pathology and Laboratory Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104
| |
Collapse
|
31
|
Zhong X, Wang G, Li F, Fang S, Zhou S, Ishiwata A, Tonevitsky AG, Shkurnikov M, Cai H, Ding F. Immunomodulatory Effect and Biological Significance of β-Glucans. Pharmaceutics 2023; 15:1615. [PMID: 37376063 DOI: 10.3390/pharmaceutics15061615] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2023] [Revised: 05/16/2023] [Accepted: 05/25/2023] [Indexed: 06/29/2023] Open
Abstract
β-glucan, one of the homopolysaccharides composed of D-glucose, exists widely in cereals and microorganisms and possesses various biological activities, including anti-inflammatory, antioxidant, and anti-tumor properties. More recently, there has been mounting proof that β-glucan functions as a physiologically active "biological response modulator (BRM)", promoting dendritic cell maturation, cytokine secretion, and regulating adaptive immune responses-all of which are directly connected with β-glucan-regulated glucan receptors. This review focuses on the sources, structures, immune regulation, and receptor recognition mechanisms of β-glucan.
Collapse
Affiliation(s)
- Xuemei Zhong
- School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus, Sun Yat-sen University, Shenzhen 518107, China
- Medical College, Shaoguan University, Shaoguan 512026, China
| | - Guoqing Wang
- School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus, Sun Yat-sen University, Shenzhen 518107, China
| | - Fu Li
- The Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen 518033, China
| | - Sixian Fang
- School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus, Sun Yat-sen University, Shenzhen 518107, China
| | - Siai Zhou
- School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus, Sun Yat-sen University, Shenzhen 518107, China
| | - Akihiro Ishiwata
- RIKEN Cluster for Pioneering Research, Wako 351-0198, Saitama, Japan
| | - Alexander G Tonevitsky
- Faculty of Biology and Biotechnology, National Research University Higher School of Economics, Moscow 117418, Russia
| | - Maxim Shkurnikov
- Faculty of Biology and Biotechnology, National Research University Higher School of Economics, Moscow 117418, Russia
| | - Hui Cai
- School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus, Sun Yat-sen University, Shenzhen 518107, China
| | - Feiqing Ding
- School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus, Sun Yat-sen University, Shenzhen 518107, China
| |
Collapse
|
32
|
Al-Nakhle HH, Khateb AM. Comprehensive In Silico Characterization of the Coding and Non-Coding SNPs in Human Dectin-1 Gene with the Potential of High-Risk Pathogenicity Associated with Fungal Infections. Diagnostics (Basel) 2023; 13:diagnostics13101785. [PMID: 37238269 DOI: 10.3390/diagnostics13101785] [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: 04/08/2023] [Revised: 05/12/2023] [Accepted: 05/16/2023] [Indexed: 05/28/2023] Open
Abstract
The human C-type lectin domain family 7 member A (CLEC7A) gene encodes a Dectin-1 protein that recognizes beta-1,3-linked and beta-1,6-linked glucans, which form the cell walls of pathogenic bacteria and fungi. It plays a role in immunity against fungal infections through pathogen recognition and immune signaling. This study aimed to explore the impact of nsSNPs in the human CLEC7A gene through computational tools (MAPP, PhD-SNP, PolyPhen-1, PolyPhen-2, SIFT, SNAP, and PredictSNP) to identify the most deleterious and damaging nsSNPs. Further, their effect on protein stability was checked along with conservation and solvent accessibility analysis by I-Mutant 2.0, ConSurf, and Project HOPE and post-translational modification analysis using MusiteDEEP. Out of the 28 nsSNPs that were found to be deleterious, 25 nsSNPs affected protein stability. Some SNPs were finalized for structural analysis with Missense 3D. Seven nsSNPs affected protein stability. Results from this study predicted that C54R, L64P, C120G, C120S, S135C, W141R, W141S, C148G, L155P, L155V, I158M, I158T, D159G, D159R, I167T, W180R, L183F, W192R, G197E, G197V, C220S, C233Y, I240T, E242G, and Y3D were the most structurally and functionally significant nsSNPs in the human CLEC7A gene. No nsSNPs were found in the predicted sites for post-translational modifications. In the 5' untranslated region, two SNPs, rs536465890 and rs527258220, showed possible miRNA target sites and DNA binding sites. The present study identified structurally and functionally significant nsSNPs in the CLEC7A gene. These nsSNPs may potentially be used for further evaluation as diagnostic and prognostic biomarkers.
Collapse
Affiliation(s)
- Hakeemah H Al-Nakhle
- Department of Medical Laboratory Technology, Collage of Applied Medical Science, Taibah University, Medina 42353, Saudi Arabia
| | - Aiah M Khateb
- Department of Medical Laboratory Technology, Collage of Applied Medical Science, Taibah University, Medina 42353, Saudi Arabia
- Special Infectious Agents Unit, King Fahd Medical Research Center, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| |
Collapse
|
33
|
Antunes de Mendonça DE, Franco de Godoy MA, Lucredi NC, Comar JF, Almeida IV, Pimenta Vicentini VE. Toxicogenic effects of the mushroom Ganoderma lucidum on human liver and kidney tumor cells and peripheral blood lymphocytes. JOURNAL OF ETHNOPHARMACOLOGY 2023; 307:116226. [PMID: 36739926 DOI: 10.1016/j.jep.2023.116226] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/07/2022] [Revised: 01/18/2023] [Accepted: 01/30/2023] [Indexed: 06/18/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Ganoderma lucidum (Curtis) P. Karst., a bioactive mushroom with medicinal properties, is known to exert immunomodulatory, anti-inflammatory, hypocholesterolemic, hypoglycemic, and hepatoprotective effects. AIM OF THE STUDY In this study, the effects of the G. lucidum fruiting body dry extract (GLE) on human liver (HepG2/C3A) and kidney (786-O) tumor cells and peripheral blood lymphocytes were evaluated. MATERIALS AND METHODS MTT-based cytotoxicity, trypan blue-based cell viability, comet, and cytokinesis-block micronucleus cytome assays were performed, and the production of reactive oxygen species was evaluated in vitro. RESULTS GLE was toxic to the tumor cells, decreasing their viability by increasing their production of reactive oxygen species and inducing damage to their DNA. By contrast, only high concentrations of GLE were toxic to lymphocytes and decreased their viability, whereas low concentrations increased lymphocyte viability. Moreover, primary DNA damage was induced by GLE only at the highest concentration tested. CONCLUSIONS G. lucidum shows potential antitumor effects against cancerous kidney and liver cells, exhibiting cytotoxic and genotoxic activity at low concentrations, whereas the same effects in lymphocytes are mediated only at high concentrations. This mushroom has the potential to be biotechnologically developed into a therapeutic agent for diseases, such as cancer.
Collapse
Affiliation(s)
| | | | | | | | - Igor Vivian Almeida
- Department of Biotechnology, Genetics and Cell Biology, State University of Maringá. Maringá, Paraná, Brazil; Environmental Toxicogenomics Research Group. Federal Rural University of Amazonia. Capitão Poço, Pará, Brazil.
| | | |
Collapse
|
34
|
Singh RP, Bhardwaj A. β-glucans: a potential source for maintaining gut microbiota and the immune system. Front Nutr 2023; 10:1143682. [PMID: 37215217 PMCID: PMC10198134 DOI: 10.3389/fnut.2023.1143682] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2023] [Accepted: 04/03/2023] [Indexed: 05/24/2023] Open
Abstract
The human gastrointestinal (GI) tract holds a complex and dynamic population of microbial communities, which exerts a marked influence on the host physiology during homeostasis and disease conditions. Diet is considered one of the main factors in structuring the gut microbiota across a lifespan. Intestinal microbial communities play a vital role in sustaining immune and metabolic homeostasis as well as protecting against pathogens. The negatively altered gut bacterial composition has related to many inflammatory diseases and infections. β-glucans are a heterogeneous assemblage of glucose polymers with a typical structure comprising a leading chain of β-(1,4) and/or β-(1,3)-glucopyranosyl units with various branches and lengths as a side chain. β-glucans bind to specific receptors on immune cells and initiate immune responses. However, β-glucans from different sources differ in their structures, conformation, physical properties, and binding affinity to receptors. How these properties modulate biological functions in terms of molecular mechanisms is not known in many examples. This review provides a critical understanding of the structures of β-glucans and their functions for modulating the gut microbiota and immune system.
Collapse
Affiliation(s)
- Ravindra Pal Singh
- Department of Industrial Biotechnology, Gujarat Biotechnology University, Gandhinagar, Gujarat, India
| | | |
Collapse
|
35
|
Coyne V, Mead HL, Mongini PKA, Barker BM. B Cell Chronic Lymphocytic Leukemia Development in Mice with Chronic Lung Exposure to Coccidioides Fungal Arthroconidia. Immunohorizons 2023; 7:333-352. [PMID: 37195872 PMCID: PMC10579974 DOI: 10.4049/immunohorizons.2300013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2023] [Accepted: 04/24/2023] [Indexed: 05/19/2023] Open
Abstract
Links between repeated microbial infections and B cell chronic lymphocytic leukemia (B-CLL) have been proposed but not tested directly. This study examines how prolonged exposure to a human fungal pathogen impacts B-CLL development in Eµ-hTCL1-transgenic mice. Monthly lung exposure to inactivated Coccidioides arthroconidia, agents of Valley fever, altered leukemia development in a species-specific manner, with Coccidioides posadasii hastening B-CLL diagnosis/progression in a fraction of mice and Coccidioides immitis delaying aggressive B-CLL development, despite fostering more rapid monoclonal B cell lymphocytosis. Overall survival did not differ significantly between control and C. posadasii-treated cohorts but was significantly extended in C. immitis-exposed mice. In vivo doubling time analyses of pooled B-CLL showed no difference in growth rates of early and late leukemias. However, within C. immitis-treated mice, B-CLL manifests longer doubling times, as compared with B-CLL in control or C. posadasii-treated mice, and/or evidence of clonal contraction over time. Through linear regression, positive relationships were noted between circulating levels of CD5+/B220low B cells and hematopoietic cells previously linked to B-CLL growth, albeit in a cohort-specific manner. Neutrophils were positively linked to accelerated growth in mice exposed to either Coccidioides species, but not in control mice. Conversely, only C. posadasii-exposed and control cohorts displayed positive links between CD5+/B220low B cell frequency and abundance of M2 anti-inflammatory monocytes and T cells. The current study provides evidence that chronic lung exposure to fungal arthroconidia affects B-CLL development in a manner dependent on fungal genotype. Correlative studies suggest that fungal species differences in the modulation of nonleukemic hematopoietic cells are involved.
Collapse
Affiliation(s)
- Vanessa Coyne
- Pathogen Microbiome Institute, Northern Arizona University, Flagstaff, AZ
| | - Heather L. Mead
- Pathogen Microbiome Institute, Northern Arizona University, Flagstaff, AZ
| | | | - Bridget M. Barker
- Pathogen Microbiome Institute, Northern Arizona University, Flagstaff, AZ
| |
Collapse
|
36
|
Ahmad MF, Ahmad FA, Zeyaullah M, Alsayegh AA, Mahmood SE, AlShahrani AM, Khan MS, Shama E, Hamouda A, Elbendary EY, Attia KAHA. Ganoderma lucidum: Novel Insight into Hepatoprotective Potential with Mechanisms of Action. Nutrients 2023; 15:1874. [PMID: 37111092 PMCID: PMC10146730 DOI: 10.3390/nu15081874] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Revised: 04/03/2023] [Accepted: 04/09/2023] [Indexed: 04/29/2023] Open
Abstract
Ganoderma lucidum (G. lucidum) has been widely used for its health benefits as an edible and traditional medicinal mushroom for thousands of years in Asian countries. It is currently used as a nutraceutical and functional food owing to its major bioactive compounds, polysaccharides and triterpenoids. G. lucidum exhibits a broad range of hepatoprotective impacts in various liver disorders, such as hepatic cancer, nonalcoholic fatty liver disease (NAFLD), alcohol-induced liver disease, hepatitis B, hepatic fibrosis, and liver injury induced by carbon tetrachloride (CCl4) and α-amanitin. G. lucidum protects the liver through a broad range of mechanisms that include the modulation of liver Phase I and II enzymes, the suppression of β-glucuronidase, antifibrotic and antiviral actions, the regulation of the production of nitric oxide (NO), the maintenance of hepatocellular calcium homeostasis, immunomodulatory activity, and scavenging free radicals. G. lucidum could signify an encouraging approach for the management of various chronic hepatopathies, and its potential mechanisms make it a distinctive agent when used alone or with other drugs and applied as a functional food, nutraceutical supplement, or adjuvant to modern medicine. This review summarizes the hepatoprotective properties of G. lucidum with its various mechanisms of action on different liver ailments. Biologically active substances derived from G. lucidum are still being studied for their potential benefits in treating different liver ailments.
Collapse
Affiliation(s)
- Md Faruque Ahmad
- Department of Clinical Nutrition, College of Applied Medical Sciences, Jazan University, Jazan 45142, Saudi Arabia
| | - Fakhruddin Ali Ahmad
- Department Forensic Science, School of Engineering and Science, G.D Goenka University, Gurugram 122103, Haryana, India;
| | - Md. Zeyaullah
- Department of Basic Medical Science, College of Applied Medical Sciences, Khamis Mushayt Campus, King Khalid University (KKU), Abha 62561, Saudi Arabia
| | - Abdulrahman A. Alsayegh
- Department of Clinical Nutrition, College of Applied Medical Sciences, Jazan University, Jazan 45142, Saudi Arabia
| | - Syed Esam Mahmood
- Department of Family and Community Medicine, College of Medicine, King Khalid University, Abha 62529, Saudi Arabia
| | - Abdullah M. AlShahrani
- Department of Basic Medical Science, College of Applied Medical Sciences, Khamis Mushayt Campus, King Khalid University (KKU), Abha 62561, Saudi Arabia
| | - Mohammad Suhail Khan
- Department of Public Health, College of Applied Medical Sciences, Khamis Mushayt Campus, King Khalid University (KKU), Abha 62561, Saudi Arabia
| | - Eman Shama
- Department of Clinical Nutrition, College of Applied Medical Sciences, Jazan University, Jazan 45142, Saudi Arabia
| | - Alshaimaa Hamouda
- Department of Clinical Nutrition, College of Applied Medical Sciences, Jazan University, Jazan 45142, Saudi Arabia
| | - Ehab Y. Elbendary
- Department of Clinical Nutrition, College of Applied Medical Sciences, Jazan University, Jazan 45142, Saudi Arabia
| | - Kandil Abdel Hai Ali Attia
- Department of Clinical Nutrition, College of Applied Medical Sciences, Jazan University, Jazan 45142, Saudi Arabia
| |
Collapse
|
37
|
Yang N, Wang M, Lin K, Wang M, Xu D, Han X, Zhao X, Wang Y, Wu G, Luo W, Liang G, Shan P. Dectin-1 deficiency alleviates diabetic cardiomyopathy by attenuating macrophage-mediated inflammatory response. Biochim Biophys Acta Mol Basis Dis 2023; 1869:166710. [PMID: 37054997 DOI: 10.1016/j.bbadis.2023.166710] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2023] [Revised: 03/25/2023] [Accepted: 03/30/2023] [Indexed: 04/15/2023]
Abstract
Cardiovascular diseases are the primary cause of mortality in patients with diabetes and obesity. Hyperglycemia and hyperlipidemia in diabetes alters cardiac function, which is associated with broader cellular processes such as aberrant inflammatory signaling. Recent studies have shown that a pattern recognition receptor called Dectin-1, expressed on macrophages, mediates pro-inflammatory responses in innate immunity. In the present study, we examined the role of Dectin-1 in the pathogenesis of diabetic cardiomyopathy. We observed increased Dectin-1 expression in heart tissues of diabetic mice and localized the source to macrophages. We then investigated the cardiac function in Dectin-1-deficient mice with STZ-induced type 1 diabetes and high-fat-diet-induced type 2 diabetes. Our results show that Dectin-1 deficient mice are protected against diabetes-induced cardiac dysfunction, cardiomyocyte hypertrophy, tissue fibrosis, and inflammation. Mechanistically, our studies show that Dectin-1 is important for cell activation and induction of inflammatory cytokines in high-concentration glucose and palmitate acid (HG + PA)-challenged macrophages. Deficiency of Dectin-1 generate fewer paracrine inflammatory factors capable of causing cardiomyocyte hypertrophy and fibrotic responses in cardiac fibroblasts. In conclusion, this study provides evidence that Dectin-1 mediates diabetes-induced cardiomyopathy through regulating inflammation. Dectin-1 may be a potential target to combat diabetic cardiomyopathy.
Collapse
Affiliation(s)
- Na Yang
- Department of Cardiology and The Key Laboratory of Cardiovascular Disease of Wenzhou, the First Affiliated Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, China; Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Minxiu Wang
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Ke Lin
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Mengyang Wang
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Diyun Xu
- Department of Cardiology and The Key Laboratory of Cardiovascular Disease of Wenzhou, the First Affiliated Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, China; Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Xue Han
- School of Pharmaceutical Sciences, Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Xia Zhao
- School of Pharmaceutical Sciences, Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Yi Wang
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Gaojun Wu
- Department of Cardiology and The Key Laboratory of Cardiovascular Disease of Wenzhou, the First Affiliated Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Wu Luo
- Department of Cardiology and The Key Laboratory of Cardiovascular Disease of Wenzhou, the First Affiliated Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, China; Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Guang Liang
- Department of Cardiology and The Key Laboratory of Cardiovascular Disease of Wenzhou, the First Affiliated Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, China; Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China; School of Pharmaceutical Sciences, Hangzhou Medical College, Hangzhou, Zhejiang, China.
| | - Peiren Shan
- Department of Cardiology and The Key Laboratory of Cardiovascular Disease of Wenzhou, the First Affiliated Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, China.
| |
Collapse
|
38
|
Ye S, Huang H, Han X, Luo W, Wu L, Ye Y, Gong Y, Zhao X, Huang W, Wang Y, Long X, Fu G, Liang G. Dectin-1 Acts as a Non-Classical Receptor of Ang II to Induce Cardiac Remodeling. Circ Res 2023; 132:707-722. [PMID: 36786193 DOI: 10.1161/circresaha.122.322259] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/15/2023]
Abstract
BACKGROUND Cardiac remodeling in heart failure involves macrophage-mediated immune responses. Recent studies have shown that a PRR (pattern recognition receptor) called dectin-1, expressed on macrophages, mediates proinflammatory responses. Whether dectin-1 plays a role in pathological cardiac remodeling is unknown. Here, we identified a potential role of dectin-1 in this disease. METHODS To model aberrant cardiac remodeling, we utilized mouse models of chronic Ang II (angiotensin II) infusion. In this model, we assessed the potential role of dectin-1 through using D1KO (dectin-1 knockout) mice and bone marrow transplantation chimeric mice. We then used cellular and molecular assays to discover the underlying mechanisms of dectin-1 function. RESULTS We found that macrophage dectin-1 is elevated in mouse heart tissues following chronic Ang II administration. D1KO mice were significantly protected against Ang II-induced cardiac dysfunction, hypertrophy, fibrosis, inflammatory responses, and macrophage infiltration. Further bone marrow transplantation studies showed that dectin-1 deficiency in bone marrow-derived cells prevented Ang II-induced cardiac inflammation and dysfunction. Through detailed molecular studies, we show that Ang II binds directly to dectin-1, causing dectin-1 homodimerization and activating the downstream Syk (spleen tyrosine kinase)/NF-κB (nuclear factor kappa B) signaling pathway to induce expression of inflammatory and chemoattractant factors. Mutagenesis studies identified R184 in the C-type lectin domain to interact with Ang II. Blocking dectin-1 in macrophages suppresses Ang II-induced inflammatory mediators and subsequent intercellular cross talk with cardiomyocytes and fibroblasts. CONCLUSIONS Our study has discovered dectin-1 as a new nonclassical receptor of Ang II and a key player in cardiac remolding and dysfunction. These studies suggest that dectin-1 may be a new target for treating hypertension-related heart failure.
Collapse
Affiliation(s)
- Shiju Ye
- School of Pharmaceutical Sciences, Hangzhou Medical College, Zhejiang, China (S.Y., X.H., W.L., X.Z., G.L.).,Department of Cardiology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, China (S.Y., H.H., L.W., Y.Y., Y.G., G.F.).,Key Laboratory of Cardiovascular Intervention and Regenerative Medicine of Zhejiang Province, Hangzhou, China (S.Y., H.H., L.W., Y.Y., Y.G., G.F.).,Department of Cardiology, The First Affiliated Hospital of Wenzhou Medical University, Zhejiang, China (S.Y., W.H.)
| | - He Huang
- Department of Cardiology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, China (S.Y., H.H., L.W., Y.Y., Y.G., G.F.).,Key Laboratory of Cardiovascular Intervention and Regenerative Medicine of Zhejiang Province, Hangzhou, China (S.Y., H.H., L.W., Y.Y., Y.G., G.F.)
| | - Xue Han
- School of Pharmaceutical Sciences, Hangzhou Medical College, Zhejiang, China (S.Y., X.H., W.L., X.Z., G.L.)
| | - Wu Luo
- School of Pharmaceutical Sciences, Hangzhou Medical College, Zhejiang, China (S.Y., X.H., W.L., X.Z., G.L.).,Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Zhejiang, China (W.L., Y.W., X.L., G.L.)
| | - Lili Wu
- Department of Cardiology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, China (S.Y., H.H., L.W., Y.Y., Y.G., G.F.).,Key Laboratory of Cardiovascular Intervention and Regenerative Medicine of Zhejiang Province, Hangzhou, China (S.Y., H.H., L.W., Y.Y., Y.G., G.F.)
| | - Yang Ye
- Department of Cardiology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, China (S.Y., H.H., L.W., Y.Y., Y.G., G.F.).,Key Laboratory of Cardiovascular Intervention and Regenerative Medicine of Zhejiang Province, Hangzhou, China (S.Y., H.H., L.W., Y.Y., Y.G., G.F.)
| | - Yingchao Gong
- Department of Cardiology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, China (S.Y., H.H., L.W., Y.Y., Y.G., G.F.).,Key Laboratory of Cardiovascular Intervention and Regenerative Medicine of Zhejiang Province, Hangzhou, China (S.Y., H.H., L.W., Y.Y., Y.G., G.F.)
| | - Xia Zhao
- School of Pharmaceutical Sciences, Hangzhou Medical College, Zhejiang, China (S.Y., X.H., W.L., X.Z., G.L.)
| | - Weijian Huang
- Department of Cardiology, The First Affiliated Hospital of Wenzhou Medical University, Zhejiang, China (S.Y., W.H.)
| | - Yi Wang
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Zhejiang, China (W.L., Y.W., X.L., G.L.)
| | - Xiaohong Long
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Zhejiang, China (W.L., Y.W., X.L., G.L.)
| | - Guosheng Fu
- Department of Cardiology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, China (S.Y., H.H., L.W., Y.Y., Y.G., G.F.).,Key Laboratory of Cardiovascular Intervention and Regenerative Medicine of Zhejiang Province, Hangzhou, China (S.Y., H.H., L.W., Y.Y., Y.G., G.F.)
| | - Guang Liang
- School of Pharmaceutical Sciences, Hangzhou Medical College, Zhejiang, China (S.Y., X.H., W.L., X.Z., G.L.).,Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Zhejiang, China (W.L., Y.W., X.L., G.L.)
| |
Collapse
|
39
|
Tang C, Sun H, Kadoki M, Han W, Ye X, Makusheva Y, Deng J, Feng B, Qiu D, Tan Y, Wang X, Guo Z, Huang C, Peng S, Chen M, Adachi Y, Ohno N, Trombetta S, Iwakura Y. Blocking Dectin-1 prevents colorectal tumorigenesis by suppressing prostaglandin E2 production in myeloid-derived suppressor cells and enhancing IL-22 binding protein expression. Nat Commun 2023; 14:1493. [PMID: 36932082 PMCID: PMC10023663 DOI: 10.1038/s41467-023-37229-x] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2022] [Accepted: 03/07/2023] [Indexed: 03/19/2023] Open
Abstract
Dectin-1 (gene Clec7a), a receptor for β-glucans, plays important roles in the host defense against fungi and immune homeostasis of the intestine. Although this molecule is also suggested to be involved in the regulation of tumorigenesis, the role in intestinal tumor development remains to be elucidated. In this study, we find that azoxymethane-dextran-sodium-sulfate-induced and ApcMin-induced intestinal tumorigenesis are suppressed in Clec7a-/- mice independently from commensal microbiota. Dectin-1 is preferentially expressed on myeloid-derived suppressor cells (MDSCs). In the Clec7a-/- mouse colon, the proportion of MDSCs and MDSC-derived prostaglandin E2 (PGE2) levels are reduced, while the expression of IL-22 binding protein (IL-22BP; gene Il22ra2) is upregulated. Dectin-1 signaling induces PGE2-synthesizing enzymes and PGE2 suppresses Il22ra2 expression in vitro and in vivo. Administration of short chain β-glucan laminarin, an antagonist of Dectin-1, suppresses the development of mouse colorectal tumors. Furthermore, in patients with colorectal cancer (CRC), the expression of CLEC7A is also observed in MDSCs and correlated with the death rate and tumor severity. Dectin-1 signaling upregulates PGE2-synthesizing enzyme expression and PGE2 suppresses IL22RA2 expression in human CRC-infiltrating cells. These observations indicate a role of the Dectin-1-PGE2-IL-22BP axis in regulating intestinal tumorigenesis, suggesting Dectin-1 as a potential target for CRC therapy.
Collapse
Affiliation(s)
- Ce Tang
- Department of Gastroenterology and Hepatology, The First Affiliated Hospital, Sun Yat-sen University, No.58, Zhong Shan Er Lu, 510080, Guangzhou, Guangdong Province, China.
- Institute of Precision Medicine, The First Affiliated Hospital, Sun Yat-sen University, No.58, Zhong Shan Er Lu, 510080, Guangzhou, Guangdong Province, China.
- Center for Animal Disease Models, Research Institute for Biomedical Sciences, Tokyo University of Science, Yamazaki 2669, Noda-shi, Chiba, 278-0022, Japan.
| | - Haiyang Sun
- Institute of Precision Medicine, The First Affiliated Hospital, Sun Yat-sen University, No.58, Zhong Shan Er Lu, 510080, Guangzhou, Guangdong Province, China
- Center for Animal Disease Models, Research Institute for Biomedical Sciences, Tokyo University of Science, Yamazaki 2669, Noda-shi, Chiba, 278-0022, Japan
| | - Motohiko Kadoki
- Center for Animal Disease Models, Research Institute for Biomedical Sciences, Tokyo University of Science, Yamazaki 2669, Noda-shi, Chiba, 278-0022, Japan
| | - Wei Han
- Center for Animal Disease Models, Research Institute for Biomedical Sciences, Tokyo University of Science, Yamazaki 2669, Noda-shi, Chiba, 278-0022, Japan
| | - Xiaoqi Ye
- Department of Gastroenterology and Hepatology, The First Affiliated Hospital, Sun Yat-sen University, No.58, Zhong Shan Er Lu, 510080, Guangzhou, Guangdong Province, China
- Center for Animal Disease Models, Research Institute for Biomedical Sciences, Tokyo University of Science, Yamazaki 2669, Noda-shi, Chiba, 278-0022, Japan
| | - Yulia Makusheva
- Center for Animal Disease Models, Research Institute for Biomedical Sciences, Tokyo University of Science, Yamazaki 2669, Noda-shi, Chiba, 278-0022, Japan
| | - Jianping Deng
- Institute of Precision Medicine, The First Affiliated Hospital, Sun Yat-sen University, No.58, Zhong Shan Er Lu, 510080, Guangzhou, Guangdong Province, China
| | - Bingbing Feng
- Institute of Precision Medicine, The First Affiliated Hospital, Sun Yat-sen University, No.58, Zhong Shan Er Lu, 510080, Guangzhou, Guangdong Province, China
| | - Ding Qiu
- Institute of Precision Medicine, The First Affiliated Hospital, Sun Yat-sen University, No.58, Zhong Shan Er Lu, 510080, Guangzhou, Guangdong Province, China
| | - Ying Tan
- Institute of Precision Medicine, The First Affiliated Hospital, Sun Yat-sen University, No.58, Zhong Shan Er Lu, 510080, Guangzhou, Guangdong Province, China
| | - Xinying Wang
- Institute of Precision Medicine, The First Affiliated Hospital, Sun Yat-sen University, No.58, Zhong Shan Er Lu, 510080, Guangzhou, Guangdong Province, China
| | - Zehao Guo
- Department of Gastroenterology and Hepatology, The First Affiliated Hospital, Sun Yat-sen University, No.58, Zhong Shan Er Lu, 510080, Guangzhou, Guangdong Province, China
| | - Chanyan Huang
- Department of Anesthesiology, The First Affiliated Hospital, Sun Yat-sen University, No.58, Zhong Shan Er Lu, 510080, Guangzhou, Guangdong Province, China
| | - Sui Peng
- Department of Gastroenterology and Hepatology, The First Affiliated Hospital, Sun Yat-sen University, No.58, Zhong Shan Er Lu, 510080, Guangzhou, Guangdong Province, China
- Institute of Precision Medicine, The First Affiliated Hospital, Sun Yat-sen University, No.58, Zhong Shan Er Lu, 510080, Guangzhou, Guangdong Province, China
| | - Minhu Chen
- Department of Gastroenterology and Hepatology, The First Affiliated Hospital, Sun Yat-sen University, No.58, Zhong Shan Er Lu, 510080, Guangzhou, Guangdong Province, China
| | - Yoshiyuki Adachi
- Laboratory for Immunopharmacology of Microbial Products, School of Pharmacy, Tokyo University of Pharmacy and Life Sciences, Hachioji, Tokyo, 192-0392, Japan
| | - Naohito Ohno
- Laboratory for Immunopharmacology of Microbial Products, School of Pharmacy, Tokyo University of Pharmacy and Life Sciences, Hachioji, Tokyo, 192-0392, Japan
| | - Sergio Trombetta
- Boehringer Ingelheim USA, 900 Ridgebury Rd, Ridgefield, CT, 06877, USA
| | - Yoichiro Iwakura
- Center for Animal Disease Models, Research Institute for Biomedical Sciences, Tokyo University of Science, Yamazaki 2669, Noda-shi, Chiba, 278-0022, Japan.
| |
Collapse
|
40
|
Liu Y, Li P, Pan W, Zhao J, Olnood CG, Liu Y, Xu YJ. Salecan confers anti-inflammatory effects in liver injury via regulating gut microbiota and its metabolites. Carbohydr Polym 2023; 302:120418. [PMID: 36604080 DOI: 10.1016/j.carbpol.2022.120418] [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: 09/18/2022] [Revised: 11/22/2022] [Accepted: 11/23/2022] [Indexed: 11/29/2022]
Abstract
Salecan, a natural β-glucan and one of the novel food ingredients approved in China, has been shown a variety of positive health effects, yet the mechanism of liver injury remains poorly understood. In addition, β-glucan could induce the shifts in gut microbiota, however, whether modulation of gut microbiota by β-glucan is associated with their positive health effects remain elusive. Here, the anti-inflammatory effects and the underlying mechanism of Salecan supplementation in CCl4-induced liver injury were investigated. After 8 weeks of treatment, we observed that Salecan alleviated liver injury by regulating inflammatory response and M2 macrophage polarization. In addition, Salecan treatment modulated the composition of gut microbiota and antibiotic cocktail treatment indicated that the hepatoprotective effect of Salecan was dependent on the gut microbiota. Fecal microbiota transplantation was used to further verify the mechanism, and we confirmed that microbial colonization partially alleviated liver injury. Besides, microbiota-derived metabolites of Salecan also contributed to the hepatoprotective and anti-inflammatory effect of Salecan against liver injury. These findings supported that Salecan intervention attenuated liver injury by regulating gut microbiota and its metabolites.
Collapse
Affiliation(s)
- Yanjun Liu
- State Key Laboratory of Food Science and Technology, School of Food Science and Technology, National Engineering Research Center for Functional Food, National Engineering Research Center of Cereal Fermentation and Food Biomanufacturing, Collaborative Innovation Center of Food Safety and Quality Control in Jiangsu Province, Jiangnan University, 1800 Lihu Road, Wuxi 214122, Jiangsu, China
| | - Panpan Li
- State Key Laboratory of Food Science and Technology, School of Food Science and Technology, National Engineering Research Center for Functional Food, National Engineering Research Center of Cereal Fermentation and Food Biomanufacturing, Collaborative Innovation Center of Food Safety and Quality Control in Jiangsu Province, Jiangnan University, 1800 Lihu Road, Wuxi 214122, Jiangsu, China
| | - Wenjie Pan
- State Key Laboratory of Food Science and Technology, School of Food Science and Technology, National Engineering Research Center for Functional Food, National Engineering Research Center of Cereal Fermentation and Food Biomanufacturing, Collaborative Innovation Center of Food Safety and Quality Control in Jiangsu Province, Jiangnan University, 1800 Lihu Road, Wuxi 214122, Jiangsu, China
| | - Juan Zhao
- Sichuan Synlight Biotech Ltd., 88 Keyuan South Road, Chengdu 610000, Sichuan, China
| | - Chen Guang Olnood
- Sichuan Synlight Biotech Ltd., 88 Keyuan South Road, Chengdu 610000, Sichuan, China
| | - Yuanfa Liu
- State Key Laboratory of Food Science and Technology, School of Food Science and Technology, National Engineering Research Center for Functional Food, National Engineering Research Center of Cereal Fermentation and Food Biomanufacturing, Collaborative Innovation Center of Food Safety and Quality Control in Jiangsu Province, Jiangnan University, 1800 Lihu Road, Wuxi 214122, Jiangsu, China
| | - Yong-Jiang Xu
- State Key Laboratory of Food Science and Technology, School of Food Science and Technology, National Engineering Research Center for Functional Food, National Engineering Research Center of Cereal Fermentation and Food Biomanufacturing, Collaborative Innovation Center of Food Safety and Quality Control in Jiangsu Province, Jiangnan University, 1800 Lihu Road, Wuxi 214122, Jiangsu, China.
| |
Collapse
|
41
|
Eswar K, Mukherjee S, Ganesan P, Kumar Rengan A. Immunomodulatory Natural Polysaccharides: An Overview of the Mechanisms Involved. Eur Polym J 2023. [DOI: 10.1016/j.eurpolymj.2023.111935] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/22/2023]
|
42
|
Zymosan Particle-Induced Hemodynamic, Cytokine and Blood Cell Changes in Pigs: An Innate Immune Stimulation Model with Relevance to Cytokine Storm Syndrome and Severe COVID-19. Int J Mol Sci 2023; 24:ijms24021138. [PMID: 36674654 PMCID: PMC9863690 DOI: 10.3390/ijms24021138] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Revised: 12/09/2022] [Accepted: 01/03/2023] [Indexed: 01/11/2023] Open
Abstract
Hemodynamic disturbance, a rise in neutrophil-to-lymphocyte ratio (NLR) and release of inflammatory cytokines into blood, is a bad prognostic indicator in severe COVID-19 and other diseases involving cytokine storm syndrome (CSS). The purpose of this study was to explore if zymosan, a known stimulator of the innate immune system, could reproduce these changes in pigs. Pigs were instrumented for hemodynamic analysis and, after i.v. administration of zymosan, serial blood samples were taken to measure blood cell changes, cytokine gene transcription in PBMC and blood levels of inflammatory cytokines, using qPCR and ELISA. Zymosan bolus (0.1 mg/kg) elicited transient hemodynamic disturbance within minutes without detectable cytokine or blood cell changes. In contrast, infusion of 1 mg/kg zymosan triggered maximal pulmonary hypertension with tachycardia, lasting for 30 min. This was followed by a transient granulopenia and then, up to 6 h, major granulocytosis, resulting in a 3-4-fold increase in NLR. These changes were paralleled by massive transcription and/or rise in IL-6, TNF-alpha, CCL-2, CXCL-10, and IL-1RA in blood. There was significant correlation between lymphopenia and IL-6 gene expression. We conclude that the presented model may enable mechanistic studies on late-stage COVID-19 and CSS, as well as streamlined drug testing against these conditions.
Collapse
|
43
|
Kruk L, Braun A, Cosset E, Gudermann T, Mammadova-Bach E. Galectin functions in cancer-associated inflammation and thrombosis. Front Cardiovasc Med 2023; 10:1052959. [PMID: 36873388 PMCID: PMC9981828 DOI: 10.3389/fcvm.2023.1052959] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2022] [Accepted: 01/12/2023] [Indexed: 02/19/2023] Open
Abstract
Galectins are carbohydrate-binding proteins that regulate many cellular functions including proliferation, adhesion, migration, and phagocytosis. Increasing experimental and clinical evidence indicates that galectins influence many steps of cancer development by inducing the recruitment of immune cells to the inflammatory sites and modulating the effector function of neutrophils, monocytes, and lymphocytes. Recent studies described that different isoforms of galectins can induce platelet adhesion, aggregation, and granule release through the interaction with platelet-specific glycoproteins and integrins. Patients with cancer and/or deep-venous thrombosis have increased levels of galectins in the vasculature, suggesting that these proteins could be important contributors to cancer-associated inflammation and thrombosis. In this review, we summarize the pathological role of galectins in inflammatory and thrombotic events, influencing tumor progression and metastasis. We also discuss the potential of anti-cancer therapies targeting galectins in the pathological context of cancer-associated inflammation and thrombosis.
Collapse
Affiliation(s)
- Linus Kruk
- Walther-Straub-Institute for Pharmacology and Toxicology, Ludwig-Maximilians-University, Munich, Germany.,Division of Nephrology, Department of Medicine IV, Ludwig-Maximilians-University Hospital, Munich, Germany
| | - Attila Braun
- Walther-Straub-Institute for Pharmacology and Toxicology, Ludwig-Maximilians-University, Munich, Germany
| | - Erika Cosset
- CRCL, UMR INSERM 1052, CNRS 5286, Centre Léon Bérard, Lyon, France
| | - Thomas Gudermann
- Walther-Straub-Institute for Pharmacology and Toxicology, Ludwig-Maximilians-University, Munich, Germany.,German Center for Lung Research (DZL), Munich, Germany
| | - Elmina Mammadova-Bach
- Walther-Straub-Institute for Pharmacology and Toxicology, Ludwig-Maximilians-University, Munich, Germany.,Division of Nephrology, Department of Medicine IV, Ludwig-Maximilians-University Hospital, Munich, Germany
| |
Collapse
|
44
|
Walachowski S, Breyne K, Secher T, Cougoule C, Guzylack-Piriou L, Meyer E, Foucras G, Tabouret G. Oral supplementation with yeast β-glucans improves the resolution of Escherichia coli-associated inflammatory responses independently of monocyte/macrophage immune training. Front Immunol 2022; 13:1086413. [PMID: 36605196 PMCID: PMC9809295 DOI: 10.3389/fimmu.2022.1086413] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2022] [Accepted: 12/02/2022] [Indexed: 12/24/2022] Open
Abstract
Introduction Confronted with the emerging threat of antimicrobial resistance, the development of alternative strategies to limit the use of antibiotics or potentiate their effect through synergy with the immune system is urgently needed. Many natural or synthetic biological response modifiers have been investigated in this context. Among them, β-glucans, a type of soluble or insoluble polysaccharide composed of a linear or branched string of glucose molecules produced by various cereals, bacteria, algae, and inferior (yeast) and superior fungi (mushrooms) have garnered interest in the scientific community, with not less than 10,000 publications over the last two decades. Various biological activities of β-glucans have been reported, such as anticancer, antidiabetic and immune-modulating effects. In vitro, yeast β-glucans are known to markedly increase cytokine secretion of monocytes/macrophages during a secondary challenge, a phenomenon called immune training. Methods Here, we orally delivered β-glucans derived from the yeast S. cerevisiae to mice that were further challenged with Escherichia coli. Results β-glucan supplementation protected the mice from E. coli intraperitoneal and intra-mammary infections, as shown by a lower bacterial burden and greatly diminished tissue damage. Surprisingly, this was not associated with an increased local immune response. In addition, granulocyte recruitment was transient and limited, as well as local cytokine secretion, arguing for faster resolution of the inflammatory response. Furthermore, ex-vivo evaluation of monocytes/macrophages isolated or differentiated from β-glucan-supplemented mice showed these cells to lack a trained response versus those from control mice. Conclusion In conclusion, dietary β-glucans can improve the outcome of Escherichia coli infections and dampen tissue damages associated to excessive inflammatory response. The mechanisms associated with such protection are not necessarily linked to immune system hyper-activation or immune training.
Collapse
Affiliation(s)
- Sarah Walachowski
- Interactions Hôtes-Agents Pathogènes (IHAP), Université de Toulouse, ENVT, Institut National de la Recherche Agronomique et Environnement (INRAE), Toulouse, France
| | - Koen Breyne
- Molecular Neurogenetics Unit, Neurology and Radiology Department, Massachusetts General Hospital - Harvard Medical School, Charlestown, MA, United States
| | - Thomas Secher
- INSERM, Centre d’Etude des Pathologies Respiratoires, Tours, France,Faculté de Médecine Université de Tours, Tours, France
| | - Céline Cougoule
- Institut de Pharmacologie et de Biologie Structurale, IPBS, Université de Toulouse, Centre National de la Recherche Scientifique (CNRS), Université Paul Sabatier (UPS), Toulouse, France
| | - Laurence Guzylack-Piriou
- Interactions Hôtes-Agents Pathogènes (IHAP), Université de Toulouse, ENVT, Institut National de la Recherche Agronomique et Environnement (INRAE), Toulouse, France
| | - Evelyne Meyer
- Ghent, Belgium Laboratory of Biochemistry, Department of Veterinary and Biosciences, Faculty of Veterinary Medicine, Ghent University, Gent, Belgium
| | - Gilles Foucras
- Interactions Hôtes-Agents Pathogènes (IHAP), Université de Toulouse, ENVT, Institut National de la Recherche Agronomique et Environnement (INRAE), Toulouse, France
| | - Guillaume Tabouret
- Interactions Hôtes-Agents Pathogènes (IHAP), Université de Toulouse, ENVT, Institut National de la Recherche Agronomique et Environnement (INRAE), Toulouse, France,*Correspondence: Guillaume Tabouret,
| |
Collapse
|
45
|
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: 2.7] [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
|
46
|
Seif M, Kakoschke TK, Ebel F, Bellet MM, Trinks N, Renga G, Pariano M, Romani L, Tappe B, Espie D, Donnadieu E, Hünniger K, Häder A, Sauer M, Damotte D, Alifano M, White PL, Backx M, Nerreter T, Machwirth M, Kurzai O, Prommersberger S, Einsele H, Hudecek M, Löffler J. CAR T cells targeting Aspergillus fumigatus are effective at treating invasive pulmonary aspergillosis in preclinical models. Sci Transl Med 2022; 14:eabh1209. [PMID: 36170447 DOI: 10.1126/scitranslmed.abh1209] [Citation(s) in RCA: 38] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Abstract
Aspergillus fumigatus is a ubiquitous mold that can cause severe infections in immunocompromised patients, typically manifesting as invasive pulmonary aspergillosis (IPA). Adaptive and innate immune cells that respond to A. fumigatus are present in the endogenous repertoire of patients with IPA but are infrequent and cannot be consistently isolated and expanded for adoptive immunotherapy. Therefore, we gene-engineered A. fumigatus-specific chimeric antigen receptor (Af-CAR) T cells and demonstrate their ability to confer antifungal reactivity in preclinical models in vitro and in vivo. We generated a CAR targeting domain AB90-E8 that recognizes a conserved protein antigen in the cell wall of A. fumigatus hyphae. T cells expressing the Af-CAR recognized A. fumigatus strains and clinical isolates and exerted a direct antifungal effect against A. fumigatus hyphae. In particular, CD8+ Af-CAR T cells released perforin and granzyme B and damaged A. fumigatus hyphae. CD8+ and CD4+ Af-CAR T cells produced cytokines that activated macrophages to potentiate the antifungal effect. In an in vivo model of IPA in immunodeficient mice, CD8+ Af-CAR T cells localized to the site of infection, engaged innate immune cells, and reduced fungal burden in the lung. Adoptive transfer of CD8+ Af-CAR T cells conferred greater antifungal efficacy compared to CD4+ Af-CAR T cells and an improvement in overall survival. Together, our study illustrates the potential of gene-engineered T cells to treat aggressive infectious diseases that are difficult to control with conventional antimicrobial therapy and support the clinical development of Af-CAR T cell therapy to treat IPA.
Collapse
Affiliation(s)
- Michelle Seif
- Medizinische Klinik und Poliklinik II und Lehrstuhl für Zelluläre Immuntherapie, Medizinische Klinik und Poliklinik II, Universitätsklinikum Würzburg, 97080 Würzburg, Germany
| | - Tamara Katharina Kakoschke
- Klinik und Poliklinik für Mund-, Kiefer- und Gesichtschirurgie, Klinikum der Universität München, LMU, 80337 München, Germany.,Institut für Infektionsmedizin und Zoonosen, Medizinische Fakultät, LMU, 80539 München, Germany
| | - Frank Ebel
- Institut für Infektionsmedizin und Zoonosen, Medizinische Fakultät, LMU, 80539 München, Germany
| | - Marina Maria Bellet
- Dipartimento di Medicina e Chirurgia, Università degli Studi di Perugia, 06132 Perugia, Italy
| | - Nora Trinks
- Lehrstuhl für Biotechnologie und Biophysik, Biozentrum und RVZ - Center for Integrative and Translational Bioimaging, Julius-Maximilians-Universität Würzburg, 97074 Würzburg, Germany
| | - Giorgia Renga
- Dipartimento di Medicina e Chirurgia, Università degli Studi di Perugia, 06132 Perugia, Italy
| | - Marilena Pariano
- Dipartimento di Medicina e Chirurgia, Università degli Studi di Perugia, 06132 Perugia, Italy
| | - Luigina Romani
- Dipartimento di Medicina e Chirurgia, Università degli Studi di Perugia, 06132 Perugia, Italy
| | - Beeke Tappe
- Medizinische Klinik und Poliklinik II und Lehrstuhl für Zelluläre Immuntherapie, Medizinische Klinik und Poliklinik II, Universitätsklinikum Würzburg, 97080 Würzburg, Germany
| | - David Espie
- Université de Paris, Institut Cochin, INSERM, CNRS, 75014 Paris, France.,CAR-T Cells Department, Invectys, 75013 Paris, France
| | - Emmanuel Donnadieu
- Université de Paris, Institut Cochin, INSERM, CNRS, 75014 Paris, France.,Equipe labellisée Ligue Contre le Cancer, 75014 Paris, France
| | - Kerstin Hünniger
- Institut für Hygiene und Mikrobiologie, Julius-Maximilians-Universität Würzburg, 97080 Würzburg, Germany.,Fungal Septomics Research Group, Leibniz-Institut für Naturstoff-Forschung und Infektionsbiologie - Hans-Knöll-Institut (HKI), 07743 Jena, Germany
| | - Antje Häder
- Fungal Septomics Research Group, Leibniz-Institut für Naturstoff-Forschung und Infektionsbiologie - Hans-Knöll-Institut (HKI), 07743 Jena, Germany
| | - Markus Sauer
- Lehrstuhl für Biotechnologie und Biophysik, Biozentrum und RVZ - Center for Integrative and Translational Bioimaging, Julius-Maximilians-Universität Würzburg, 97074 Würzburg, Germany
| | - Diane Damotte
- Department of Pathology, Paris Centre University Hospitals, AP-HP, 75014 Paris, France.,INSERM U1138, Cordeliers Research Center, Team Cancer, Immune Control and Escape, Paris, France; University Pierre and Marie Curie, 75006 Paris, France
| | - Marco Alifano
- Department of Thoracic Surgery, Paris Centre University Hospitals, AP-HP, Paris, France; University Paris Descartes, 75014 Paris, France
| | - P Lewis White
- Public Health Wales, Microbiology Cardiff, UHW, CF14 4XW Cardiff, UK
| | - Matthijs Backx
- Public Health Wales, Microbiology Cardiff, UHW, CF14 4XW Cardiff, UK
| | - Thomas Nerreter
- Medizinische Klinik und Poliklinik II und Lehrstuhl für Zelluläre Immuntherapie, Medizinische Klinik und Poliklinik II, Universitätsklinikum Würzburg, 97080 Würzburg, Germany
| | - Markus Machwirth
- Medizinische Klinik und Poliklinik II und Lehrstuhl für Zelluläre Immuntherapie, Medizinische Klinik und Poliklinik II, Universitätsklinikum Würzburg, 97080 Würzburg, Germany
| | - Oliver Kurzai
- Institut für Hygiene und Mikrobiologie, Julius-Maximilians-Universität Würzburg, 97080 Würzburg, Germany.,Fungal Septomics Research Group, Leibniz-Institut für Naturstoff-Forschung und Infektionsbiologie - Hans-Knöll-Institut (HKI), 07743 Jena, Germany
| | - Sabrina Prommersberger
- Medizinische Klinik und Poliklinik II und Lehrstuhl für Zelluläre Immuntherapie, Medizinische Klinik und Poliklinik II, Universitätsklinikum Würzburg, 97080 Würzburg, Germany
| | - Hermann Einsele
- Medizinische Klinik und Poliklinik II und Lehrstuhl für Zelluläre Immuntherapie, Medizinische Klinik und Poliklinik II, Universitätsklinikum Würzburg, 97080 Würzburg, Germany
| | - Michael Hudecek
- Medizinische Klinik und Poliklinik II und Lehrstuhl für Zelluläre Immuntherapie, Medizinische Klinik und Poliklinik II, Universitätsklinikum Würzburg, 97080 Würzburg, Germany
| | - Jürgen Löffler
- Medizinische Klinik und Poliklinik II und Lehrstuhl für Zelluläre Immuntherapie, Medizinische Klinik und Poliklinik II, Universitätsklinikum Würzburg, 97080 Würzburg, Germany
| |
Collapse
|
47
|
Al Madhoun A, Kochumon S, Al-Rashed F, Sindhu S, Thomas R, Miranda L, Al-Mulla F, Ahmad R. Dectin-1 as a Potential Inflammatory Biomarker for Metabolic Inflammation in Adipose Tissue of Individuals with Obesity. Cells 2022; 11:2879. [PMID: 36139454 PMCID: PMC9496833 DOI: 10.3390/cells11182879] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2022] [Revised: 09/02/2022] [Accepted: 09/10/2022] [Indexed: 11/17/2022] Open
Abstract
In obesity, macrophage activation and infiltration in adipose tissue (AT) underlie chronic low-grade inflammation-induced insulin resistance. Although dectin-1 is primarily a pathogen recognition receptor and innate immune response modulator, its role in metabolic syndromes remains to be clarified. This study aimed to investigate the dectin-1 gene expression in subcutaneous AT in the context of obesity and associated inflammatory markers. Subcutaneous AT biopsies were collected from 59 nondiabetic (lean/overweight/obese) individuals. AT gene expression levels of dectin-1 and inflammatory markers were determined via real-time reverse transcriptase-quantitative polymerase chain reaction. Dectin-1 protein expression was assessed using immunohistochemistry. Plasma lipid profiles were measured by ELISA. AT dectin-1 transcripts and proteins were significantly elevated in obese as compared to lean individuals. AT dectin-1 transcripts correlated positively with body mass index and fat percentage (r ≥ 0.340, p ≤ 0.017). AT dectin-1 RNA levels correlated positively with clinical parameters, including plasma C-reactive protein and CCL5/RANTES, but negatively with that of adiponectin. The expression of dectin-1 transcripts was associated with that of various proinflammatory cytokines, chemokines, and their cognate receptors (r ≥ 0.300, p ≤ 0.05), but not with anti-inflammatory markers. Dectin-1 and members of the TLR signaling cascade were found to be significantly associated, suggesting an interplay between the two pathways. Dectin-1 expression was correlated with monocyte/macrophage markers, including CD16, CD68, CD86, and CD163, suggesting its monocytes/macrophage association in an adipose inflammatory microenvironment. Dectin-1 expression was independently predicted by CCR5, CCL20, TLR2, and MyD88. In conclusion, dectin-1 may be regarded as an AT biomarker of metabolic inflammation in obesity.
Collapse
Affiliation(s)
- Ashraf Al Madhoun
- Animal and Imaging Core Facilities, Dasman Diabetes Institute, Dasman 15462, Kuwait
- Genetics and Bioinformatics, Dasman Diabetes Institute, Dasman 15462, Kuwait
| | - Shihab Kochumon
- Immunology and Microbiology Department, Dasman Diabetes Institute, Dasman 15462, Kuwait
| | - Fatema Al-Rashed
- Immunology and Microbiology Department, Dasman Diabetes Institute, Dasman 15462, Kuwait
| | - Sardar Sindhu
- Animal and Imaging Core Facilities, Dasman Diabetes Institute, Dasman 15462, Kuwait
- Immunology and Microbiology Department, Dasman Diabetes Institute, Dasman 15462, Kuwait
| | - Reeby Thomas
- Immunology and Microbiology Department, Dasman Diabetes Institute, Dasman 15462, Kuwait
| | - Lavina Miranda
- Genetics and Bioinformatics, Dasman Diabetes Institute, Dasman 15462, Kuwait
| | - Fahd Al-Mulla
- Genetics and Bioinformatics, Dasman Diabetes Institute, Dasman 15462, Kuwait
| | - Rasheed Ahmad
- Immunology and Microbiology Department, Dasman Diabetes Institute, Dasman 15462, Kuwait
| |
Collapse
|
48
|
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
|
49
|
Maldonado SD, Dai J, Dutta O, Hurley HJ, Singh S, Gittens-Williams L, Kalyoussef E, Edelblum KL, Rivera A, Fitzgerald-Bocarsly P. Human Plasmacytoid Dendritic Cells Express C-Type Lectin Receptors and Attach and Respond to Aspergillus fumigatus. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2022; 209:675-683. [PMID: 35879099 DOI: 10.4049/jimmunol.2000632] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/04/2020] [Accepted: 06/03/2022] [Indexed: 09/06/2024]
Abstract
Plasmacytoid dendritic cells (pDCs) have been implicated as having a role in antifungal immunity, but mechanisms of their interaction with fungi and the resulting cellular responses are not well understood. In this study, we identify the direct and indirect biological response of human pDCs to the fungal pathogen Aspergillus fumigatus and characterize the expression and regulation of antifungal receptors on the pDC surface. Results indicate pDCs do not phagocytose Aspergillus conidia, but instead bind hyphal surfaces and undergo activation and maturation via the upregulation of costimulatory and maturation markers. Measuring the expression of C-type lectin receptors dectin-1, dectin-2, dectin-3, and mannose receptor on human pDCs revealed intermediate expression of each receptor compared with monocytes. The specific dectin-1 agonist curdlan induced pDC activation and maturation in a cell-intrinsic and cell-extrinsic manner. The indirect activation of pDCs by curdlan was much stronger than direct stimulation and was mediated through cytokine production by other PBMCs. Overall, our data indicate pDCs express various C-type lectin receptors, recognize and respond to Aspergillus hyphal Ag, and serve as immune enhancers or modulators in the overarching fungal immune response.
Collapse
Affiliation(s)
- Samuel D Maldonado
- Department of Pathology, Immunology and Laboratory Medicine, Rutgers New Jersey Medical School, Newark, NJ
- Rutgers School of Graduate Studies, Newark, NJ
| | - Jihong Dai
- Department of Pathology, Immunology and Laboratory Medicine, Rutgers New Jersey Medical School, Newark, NJ
| | - Orchi Dutta
- Rutgers School of Graduate Studies, Newark, NJ
| | - Harry J Hurley
- Department of Pathology, Immunology and Laboratory Medicine, Rutgers New Jersey Medical School, Newark, NJ
- Rutgers School of Graduate Studies, Newark, NJ
| | - Sukhwinder Singh
- Department of Pathology, Immunology and Laboratory Medicine, Rutgers New Jersey Medical School, Newark, NJ
| | - Lisa Gittens-Williams
- Department of Obstetrics, Gynecology & Women's Health, Rutgers New Jersey Medical School, Newark, NJ
| | - Evelyne Kalyoussef
- Department of Otolaryngology, Rutgers New Jersey Medical School, Newark, NJ
| | - Karen L Edelblum
- Department of Pathology, Immunology and Laboratory Medicine, Rutgers New Jersey Medical School, Newark, NJ
- Center for Immunity and Inflammation, Rutgers New Jersey Medical School, Newark, NJ; and
| | - Amariliz Rivera
- Center for Immunity and Inflammation, Rutgers New Jersey Medical School, Newark, NJ; and
- Department of Pediatrics, Rutgers New Jersey Medical School, Newark, NJ
| | - Patricia Fitzgerald-Bocarsly
- Department of Pathology, Immunology and Laboratory Medicine, Rutgers New Jersey Medical School, Newark, NJ
- Center for Immunity and Inflammation, Rutgers New Jersey Medical School, Newark, NJ; and
| |
Collapse
|
50
|
Jing Z, Wang S, Xu K, Tang Q, Li W, Zheng W, Shi H, Su K, Liu Y, Hong Z. A Potent Micron Neoantigen Tumor Vaccine GP-Neoantigen Induces Robust Antitumor Activity in Multiple Tumor Models. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2022; 9:e2201496. [PMID: 35712770 PMCID: PMC9403634 DOI: 10.1002/advs.202201496] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/15/2022] [Revised: 05/05/2022] [Indexed: 05/28/2023]
Abstract
Therapeutic tumor neoantigen vaccines have been widely studied given their good safety profile and ability to avoid central thymic tolerance. However, targeting antigen-presenting cells (APCs) and inducing robust neoantigen-specific cellular immunity remain challenges. Here, a safe and broad-spectrum neoantigen vaccine delivery system is proposed (GP-Neoantigen) based on β-1,3-glucan particles (GPs) derived from Saccharomyces cerevisiae and coupling peptide antigens with GPs through convenient click chemistry. The prepared system has a highly uniform particle size and high APC targeting specificity. In mice, the vaccine system induced a robust specific CD8+ T cell immune response and humoral immune response against various conjugated peptide antigens and showed strong tumor growth inhibitory activity in EG7·OVA lymphoma, B16F10 melanoma, 4T1 breast cancer, and CT26 colon cancer models. The combination of the toll-like receptors (TLRs) agonist PolyI:C and CpG 2395 further enhanced the antitumor response of the particle system, achieving complete tumor clearance in multiple mouse models and inducing long-term rejection of reinoculated tumors. These results provide the broad possibility for its further clinical promotion and personalized vaccine treatment.
Collapse
Affiliation(s)
- Zhe Jing
- State Key Laboratory of Medicinal Chemical BiologyTianjin Key Laboratory of Protein SciencesCollege of Life SciencesNankai UniversityTianjin300071P. R. China
| | - Shuqing Wang
- State Key Laboratory of Medicinal Chemical BiologyTianjin Key Laboratory of Protein SciencesCollege of Life SciencesNankai UniversityTianjin300071P. R. China
| | - Keyuan Xu
- State Key Laboratory of Medicinal Chemical BiologyTianjin Key Laboratory of Protein SciencesCollege of Life SciencesNankai UniversityTianjin300071P. R. China
| | - Qian Tang
- State Key Laboratory of Medicinal Chemical BiologyTianjin Key Laboratory of Protein SciencesCollege of Life SciencesNankai UniversityTianjin300071P. R. China
| | - Wenjing Li
- State Key Laboratory of Medicinal Chemical BiologyTianjin Key Laboratory of Protein SciencesCollege of Life SciencesNankai UniversityTianjin300071P. R. China
| | - Wei Zheng
- State Key Laboratory of Medicinal Chemical BiologyTianjin Key Laboratory of Protein SciencesCollege of Life SciencesNankai UniversityTianjin300071P. R. China
| | - Haobo Shi
- State Key Laboratory of Medicinal Chemical BiologyTianjin Key Laboratory of Protein SciencesCollege of Life SciencesNankai UniversityTianjin300071P. R. China
| | - Kailing Su
- State Key Laboratory of Medicinal Chemical BiologyTianjin Key Laboratory of Protein SciencesCollege of Life SciencesNankai UniversityTianjin300071P. R. China
| | - Yanting Liu
- State Key Laboratory of Medicinal Chemical BiologyTianjin Key Laboratory of Protein SciencesCollege of Life SciencesNankai UniversityTianjin300071P. R. China
- Department of OncologyThe First Affiliated Hospital of Xinxiang Medical UniversityWeihuiHenan Province453100P. R. China
| | - Zhangyong Hong
- State Key Laboratory of Medicinal Chemical BiologyTianjin Key Laboratory of Protein SciencesCollege of Life SciencesNankai UniversityTianjin300071P. R. China
| |
Collapse
|