1
|
Rosati D, Pradhan A, van Heck JIP, Helder L, Jaeger M, Gow NAR, Joosten LAB, Williams DL, Brown AJP, Bruno M, Netea MG. Candida albicans N-Linked Mannans Potentiate the Induction of Trained Immunity via Dectin-2. J Infect Dis 2024; 230:768-777. [PMID: 38446996 PMCID: PMC11420807 DOI: 10.1093/infdis/jiae112] [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: 07/28/2023] [Revised: 01/23/2024] [Accepted: 02/28/2024] [Indexed: 03/08/2024] Open
Abstract
The interaction between the Candida albicans cell wall and pattern recognition receptors is crucial for the initiation of host immune responses, which, ultimately, contribute to the clearance of this pathogenic fungus. In the present study, we investigate the ability of C. albicans mannans to modulate immune response and induce innate immune memory (also termed trained immunity). Using mutants of C. albicans that are defective in or lack mannosyl residues, we show that alterations in the mannosylation of the C. albicans cell wall affect the innate cytokine response and strongly reduce the secretion of T-cell-derived cytokines. Subsequently, we demonstrate that the branching of N-linked mannan, but not O-linked mannan, is essential to potentiate the induction of trained immunity, a process mediated by dectin 2. In conclusion, N-linked mannan is needed, in addition to β-glucans, for an effective induction of trained immunity by C. albicans.
Collapse
Affiliation(s)
- Diletta Rosati
- Department of Internal Medicine and Radboud Center for Infectious Diseases, Radboud University Medical Center, Geert Grooteplein Zuid 10, 6525 GA, Nijmegen, the Netherlands
| | - Arnab Pradhan
- Medical Research Council Centre for Medical Mycology, University of Exeter, Exeter EX4 4QD, United Kingdom
| | - Julia I P van Heck
- Department of Internal Medicine and Radboud Center for Infectious Diseases, Radboud University Medical Center, Geert Grooteplein Zuid 10, 6525 GA, Nijmegen, the Netherlands
| | - Leonie Helder
- Department of Internal Medicine and Radboud Center for Infectious Diseases, Radboud University Medical Center, Geert Grooteplein Zuid 10, 6525 GA, Nijmegen, the Netherlands
| | - Martin Jaeger
- Department of Internal Medicine and Radboud Center for Infectious Diseases, Radboud University Medical Center, Geert Grooteplein Zuid 10, 6525 GA, Nijmegen, the Netherlands
| | - Neil A R Gow
- Medical Research Council Centre for Medical Mycology, University of Exeter, Exeter EX4 4QD, United Kingdom
| | - Leo A B Joosten
- Department of Internal Medicine and Radboud Center for Infectious Diseases, Radboud University Medical Center, Geert Grooteplein Zuid 10, 6525 GA, Nijmegen, the Netherlands
- Department of Medical Genetics, Iuliu Hatieganu University of Medicine and Pharmacy Cluj-Napoca, Romania
| | - David L Williams
- Departments of Surgery, Biomedical Sciences and Center of Excellence in Inflammation, Infectious Disease and Immunity, Quillen College of Medicine, East Tennessee State University, Johnson City, Tennessee, USA
| | - Alistair J P Brown
- Medical Research Council Centre for Medical Mycology, University of Exeter, Exeter EX4 4QD, United Kingdom
| | - Mariolina Bruno
- Department of Internal Medicine and Radboud Center for Infectious Diseases, Radboud University Medical Center, Geert Grooteplein Zuid 10, 6525 GA, Nijmegen, the Netherlands
| | - Mihai G Netea
- Department of Internal Medicine and Radboud Center for Infectious Diseases, Radboud University Medical Center, Geert Grooteplein Zuid 10, 6525 GA, Nijmegen, the Netherlands
- Department for Immunology and Metabolism, Life and Medical Sciences Institute (LIMES), University of Bonn, Bonn, Germany
| |
Collapse
|
2
|
López-Collazo E, del Fresno C. Endotoxin tolerance and trained immunity: breaking down immunological memory barriers. Front Immunol 2024; 15:1393283. [PMID: 38742111 PMCID: PMC11089161 DOI: 10.3389/fimmu.2024.1393283] [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: 02/28/2024] [Accepted: 04/09/2024] [Indexed: 05/16/2024] Open
Abstract
For decades, innate immune cells were considered unsophisticated first responders, lacking the adaptive memory of their T and B cell counterparts. However, mounting evidence demonstrates the surprising complexity of innate immunity. Beyond quickly deploying specialized cells and initiating inflammation, two fascinating phenomena - endotoxin tolerance (ET) and trained immunity (TI) - have emerged. ET, characterized by reduced inflammatory response upon repeated exposure, protects against excessive inflammation. Conversely, TI leads to an enhanced response after initial priming, allowing the innate system to mount stronger defences against subsequent challenges. Although seemingly distinct, these phenomena may share underlying mechanisms and functional implications, blurring the lines between them. This review will delve into ET and TI, dissecting their similarities, differences, and the remaining questions that warrant further investigation.
Collapse
Affiliation(s)
- Eduardo López-Collazo
- The Innate Immune Response Group, Hospital la Paz Institute for Health Research (IdiPAZ), La Paz University Hospital, Madrid, Spain
- Tumour Immunology Laboratory, IdiPAZ, La Paz University Hospital, Madrid, Spain
- Centro de Investigación Biomédica en Red (CIBER), Respiratory Diseases (CIBRES), Madrid, Spain
| | - Carlos del Fresno
- The Innate Immune Response Group, Hospital la Paz Institute for Health Research (IdiPAZ), La Paz University Hospital, Madrid, Spain
- Immunomodulation Laboratory, IdiPAZ, La Paz University Hospital, Madrid, Spain
| |
Collapse
|
3
|
McBride MA, Stothers CL, Fensterheim BA, Caja KR, Owen AM, Hernandez A, Bohannon JK, Patil NK, Ali S, Dalal S, Rahim M, Trenary IA, Young JD, Williams DL, Sherwood ER. Bacteria- and fungus-derived PAMPs induce innate immune memory via similar functional, metabolic, and transcriptional adaptations. J Leukoc Biol 2024; 115:358-373. [PMID: 37793181 PMCID: PMC10872320 DOI: 10.1093/jleuko/qiad120] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2023] [Revised: 08/28/2023] [Accepted: 09/15/2023] [Indexed: 10/06/2023] Open
Abstract
Exposure to pathogen-associated molecular patterns (PAMPs) induces an augmented, broad-spectrum antimicrobial response to subsequent infection, a phenomenon termed innate immune memory. This study examined the effects of treatment with β-glucan, a fungus-derived dectin-1 ligand, or monophosphoryl lipid A (MPLA), a bacteria-derived Toll-like receptor 4 ligand, on innate immune memory with a focus on identifying common cellular and molecular pathways activated by these diverse PAMPs. Treatment with either PAMP prepared the innate immune system to respond more robustly to Pseudomonas aeruginosa infection in vivo by facilitating mobilization of innate leukocytes into blood, recruitment of leukocytes to the site of infection, augmentation of microbial clearance, and attenuation of cytokine production. Examination of macrophages ex vivo showed amplification of metabolism, phagocytosis, and respiratory burst after treatment with either agent, although MPLA more robustly augmented these activities and more effectively facilitated killing of bacteria. Both agents activated gene expression pathways in macrophages that control inflammation, antimicrobial functions, and protein synthesis and suppressed pathways regulating cell division. β-glucan treatment minimally altered macrophage differential gene expression in response to lipopolysaccharide (LPS) challenge, whereas MPLA attenuated the magnitude of the LPS-induced transcriptional response, especially cytokine gene expression. These results show that β-glucan and MPLA similarly augment the innate response to infection in vivo. Yet, MPLA more potently induces alterations in macrophage metabolism, antimicrobial functions, gene transcription and the response to LPS.
Collapse
Affiliation(s)
- Margaret A. McBride
- Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, 1211 Medical Center Drive, Nashville 37232, Tennessee
| | - Cody L. Stothers
- Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, 1211 Medical Center Drive, Nashville 37232, Tennessee
| | - Benjamin A. Fensterheim
- Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, 1211 Medical Center Drive, Nashville 37232, Tennessee
| | - Katherine R. Caja
- Department of Anesthesiology, Vanderbilt University Medical Center, 1211 Medical Center Drive, Nashville 37232, Tennessee
| | - Allison M. Owen
- Department of Anesthesiology, Vanderbilt University Medical Center, 1211 Medical Center Drive, Nashville 37232, Tennessee
| | - Antonio Hernandez
- Department of Anesthesiology, Vanderbilt University Medical Center, 1211 Medical Center Drive, Nashville 37232, Tennessee
| | - Julia K. Bohannon
- Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, 1211 Medical Center Drive, Nashville 37232, Tennessee
- Department of Anesthesiology, Vanderbilt University Medical Center, 1211 Medical Center Drive, Nashville 37232, Tennessee
| | - Naeem K. Patil
- Department of Anesthesiology, Vanderbilt University Medical Center, 1211 Medical Center Drive, Nashville 37232, Tennessee
| | - Sabah Ali
- Department of Anesthesiology, Vanderbilt University Medical Center, 1211 Medical Center Drive, Nashville 37232, Tennessee
| | - Sujata Dalal
- Department of Anesthesiology, Vanderbilt University Medical Center, 1211 Medical Center Drive, Nashville 37232, Tennessee
| | - Mohsin Rahim
- Department of Chemical and Biomolecular Engineering, Vanderbilt University, 2301 Vanderbilt Place, Nashville 37235, Tennessee
| | - Irina A. Trenary
- Department of Chemical and Biomolecular Engineering, Vanderbilt University, 2301 Vanderbilt Place, Nashville 37235, Tennessee
| | - Jamey D. Young
- Department of Chemical and Biomolecular Engineering, Vanderbilt University, 2301 Vanderbilt Place, Nashville 37235, Tennessee
- Department of Molecular Physiology and Biophysics, Vanderbilt University, 2215 Garland Avenue, Nashville 37232, Tennessee
| | - David L. Williams
- Department of Surgery, Quillen College of Medicine, East Tennessee State University, 325 North State of Franklin Road, Johnson City 37604, Tennessee
- Center for Inflammation, Infectious Disease and Immunity, Quillen College of Medicine, East Tennessee State University, 325 North State of Franklin Road, Johnson City 37604, Tennessee
| | - Edward R. Sherwood
- Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, 1211 Medical Center Drive, Nashville 37232, Tennessee
- Department of Anesthesiology, Vanderbilt University Medical Center, 1211 Medical Center Drive, Nashville 37232, Tennessee
- Department of Surgery, Quillen College of Medicine, East Tennessee State University, 325 North State of Franklin Road, Johnson City 37604, Tennessee
- Center for Inflammation, Infectious Disease and Immunity, Quillen College of Medicine, East Tennessee State University, 325 North State of Franklin Road, Johnson City 37604, Tennessee
| |
Collapse
|
4
|
Cheng QJ, Farrell K, Fenn J, Ma Z, Makanani SK, Siemsen J. Dectin-1 ligands produce distinct training phenotypes in human monocytes through differential activation of signaling networks. Sci Rep 2024; 14:1454. [PMID: 38228717 PMCID: PMC10791629 DOI: 10.1038/s41598-024-51620-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2023] [Accepted: 01/08/2024] [Indexed: 01/18/2024] Open
Abstract
Cells of the innate immune system retain memory of prior exposures through a process known as innate immune training. β-glucan, a Dectin-1 ligand purified from the Candida albicans cell wall, has been one of the most widely utilized ligands for inducing innate immune training. However, many Dectin-1 ligands exist, and it is not known whether these all produce the same phenotype. Using a well-established in vitro model of innate immune training, we compared two commercially available Dectin-1 agonists, zymosan and depleted zymosan, with the gold standard β-glucan in the literature. We found that depleted zymosan, a β-glucan purified from Saccharomyces cerevisiae cell wall through alkali treatment, produced near identical effects as C. albicans β-glucan. However, untreated zymosan produced a distinct training effect from β-glucans at both the transcript and cytokine level. Training with zymosan diminished, rather than potentiated, induction of cytokines such as TNF and IL-6. Zymosan activated NFκB and AP-1 transcription factors more strongly than β-glucans. The addition of the toll-like receptor (TLR) ligand Pam3CSK4 was sufficient to convert the training effect of β-glucans to a phenotype resembling zymosan. We conclude that differential activation of TLR signaling pathways determines the phenotype of innate immune training induced by Dectin-1 ligands.
Collapse
Affiliation(s)
- Quen J Cheng
- Department of Medicine, Division of Infectious Diseases, David Geffen School of Medicine, University of California, Los Angeles, CA, USA.
- Molecular Biology Institute, University of California, Los Angeles, CA, USA.
| | - Kylie Farrell
- Department of Medicine, Division of Infectious Diseases, David Geffen School of Medicine, University of California, Los Angeles, CA, USA
- Department of Microbiology, Immunology, and Molecular Genetics, University of California, Los Angeles, CA, USA
| | - Jeffrey Fenn
- Department of Microbiology, Immunology, and Molecular Genetics, University of California, Los Angeles, CA, USA
| | - Zuchao Ma
- Department of Surgery, Center of Excellence in Inflammation, Infectious Disease and Immunity, Quillen College of Medicine, East Tennessee State University, Johnson City, TN, USA
| | - Sara K Makanani
- Molecular Biology Institute, University of California, Los Angeles, CA, USA
- Department of Microbiology, Immunology, and Molecular Genetics, University of California, Los Angeles, CA, USA
| | - Jonathan Siemsen
- Department of Microbiology, Immunology, and Molecular Genetics, University of California, Los Angeles, CA, USA
| |
Collapse
|
5
|
Hasan A, Roome T, Wahid M, Ansari SA, Khan JA, Jilani SNA, Jawed A, Kiyani A. Expression of Toll-like receptor 2, Dectin-1, and Osteopontin in murine model of pulpitis. Clin Oral Investig 2023; 27:1177-1192. [PMID: 36205788 DOI: 10.1007/s00784-022-04732-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2022] [Accepted: 10/01/2022] [Indexed: 11/03/2022]
Abstract
OBJECTIVES This in vivo animal study aimed to develop a murine model of pulpitis induced by pulp exposure with or without application of zymosan in Naval Medical Research Institute (NMRI) mice and observe expressions of Toll-like receptor (TLR)-2, TLR-4, Dectin-1, Osteopontin (OPN), tumor necrosis factor alpha (TNF-α), interleukin (IL)-6, and IL-1ß. MATERIAL AND METHODS A total of 168 NMRI mice were divided into two groups, i.e., group A (n = 84) (pulpitis induced by pulp exposure only) and group B (n = 84) (pulpitis induced by pulp exposure and zymosan application). Right maxillary molar pulps were exposed with ¼ round bur, and animals were sacrificed at 0, 6, 9, 12, 24, 48, and 72 h. The exposed teeth were obtained for real-time polymerase chain reaction (qRT-PCR) analysis and histological and immunohistochemistry (IHC) analysis. RESULTS Histological evaluation revealed a time-dependent steady increase in inflammation. Similar time-dependent increase in the expression of inflammatory cytokines was noted. Group A exhibited an increase in TLR-4, Dectin-1, and OPN at 6 h, while TLR-2 was expressed at 24 h. Group B expressed TLR-2, Dectin-1, and OPN at 9, 48, and 72 h, respectively (p ≤ 0.05). Expression of OPN and TNF-α exhibited a similar pattern in both groups. IHC also detected expression of TLR-2, Dectin-1, TLR4, and CD68 in some cells at 6 and 9 h. CONCLUSIONS NMRI mice provided for a stable pulp inflammation model. Zymosan may be used to develop pulp inflammation model and study inflammatory response towards fungal antigens. Dental pulp expressed Dectin-1 receptor. OPN and TNF-α exhibited a similar expression pattern. CLINICAL RELEVANCE Innate immunity of dental pulp is capable of detecting fungal pathogens.
Collapse
Affiliation(s)
- Arshad Hasan
- Department of Operative Dentistry, Dow Dental College, Dow University of Health Sciences, Baba-e-Urdu Road, Karachi, 74200, Pakistan.
| | - Talat Roome
- Department of Pathology, Section Molecular Pathology, Dow International Medical College, Ojha Campus, Gulzar-e-Hijri Karachi, Pakistan.,Dow Institute for Advanced Biological and Animal Research, Dow University of Health Sciences, Ojha Campus, Gulzar-e-Hijri Karachi, Pakistan
| | - Mohsin Wahid
- Department of Pathology, Dow International Medical College, Dow University of Health Sciences, Ojha Campus, Gulzar-e-Hijri Karachi, Pakistan.,Dow Research Institute of Biotechnology and Biomedical Sciences, Dow University of Health Sciences, Ojha Campus, Gulzar-e-Hijri Karachi, Pakistan
| | - Shazia Akbar Ansari
- Department of Oral Pathology, Dow Dental College, Dow University of Health Sciences, Baba-e-Urdu Road, Karachi, 74200, Pakistan
| | - Javeria Ali Khan
- Department of Operative Dentistry, Dow Dental College, Dow University of Health Sciences, Baba-e-Urdu Road, Karachi, 74200, Pakistan
| | - Syeda Neha Ahmed Jilani
- Dow Institute for Advanced Biological and Animal Research, Dow University of Health Sciences, Ojha Campus, Gulzar-e-Hijri Karachi, Pakistan
| | - Abira Jawed
- Department of Oral Pathology, Dow Dental College, Dow University of Health Sciences, Baba-e-Urdu Road, Karachi, 74200, Pakistan
| | - Amber Kiyani
- Department of Oral Diagnosis and Medicine, Islamic International Dental College, Riphah International University, 7th Avenue G-7/4, Islamabad, Pakistan
| |
Collapse
|
6
|
Achmad H, Saleh Ibrahim Y, Mohammed Al-Taee M, Gabr GA, Waheed Riaz M, Hamoud Alshahrani S, Alexis Ramírez-Coronel A, Turki Jalil A, Setia Budi H, Sawitri W, Elena Stanislavovna M, Gupta J. Nanovaccines in cancer immunotherapy: Focusing on dendritic cell targeting. Int Immunopharmacol 2022; 113:109434. [DOI: 10.1016/j.intimp.2022.109434] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2022] [Revised: 10/27/2022] [Accepted: 11/03/2022] [Indexed: 11/17/2022]
|
7
|
Fan F, Liu Y, Liu Y, Lv R, Sun W, Ding W, Cai Y, Li W, Liu X, Qu W. Candida albicans biofilms: antifungal resistance, immune evasion, and emerging therapeutic strategies. Int J Antimicrob Agents 2022; 60:106673. [PMID: 36103915 DOI: 10.1016/j.ijantimicag.2022.106673] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Revised: 08/24/2022] [Accepted: 09/09/2022] [Indexed: 12/14/2022]
Abstract
Candida albicans is a fungal pathogen that can form biofilms on medical devices and host tissue, resulting in serious, life-threatening infections. These fungal biofilms are inherently resistant to traditional antifungal therapies and the host immune system; therefore, biofilm-associated infections are a huge clinical challenge. This review summarizes the most important insights into C. albicans biofilm-associated antifungal drug resistance mechanisms and immune evasion strategies. In addtion, this review also discusses the strategies for antifungal drug use to combat these processes, providing further evidence for novel drugs research and clinical therapies.
Collapse
Affiliation(s)
- FangMei Fan
- Department of Laboratory Medicine, Guiyang Maternity & Child Health Hospital, Guiyang, 550003, China
| | - Yi Liu
- Department of Laboratory Medicine, Guiyang Maternity & Child Health Hospital, Guiyang, 550003, China
| | - YiQing Liu
- Department of Laboratory Medicine, Guiyang Maternity & Child Health Hospital, Guiyang, 550003, China
| | - RuiXue Lv
- Department of Laboratory Medicine, Guiyang Maternity & Child Health Hospital, Guiyang, 550003, China
| | - Wei Sun
- Department of Laboratory Medicine, Guiyang Maternity & Child Health Hospital, Guiyang, 550003, China
| | - WenJing Ding
- Department of Laboratory Medicine, Guiyang Maternity & Child Health Hospital, Guiyang, 550003, China
| | - YanXing Cai
- Department of Laboratory Medicine, Guiyang Maternity & Child Health Hospital, Guiyang, 550003, China
| | - WeiWei Li
- Department of Laboratory Medicine, Guiyang Maternity & Child Health Hospital, Guiyang, 550003, China
| | - Xing Liu
- Department of Laboratory Medicine, Guiyang Maternity & Child Health Hospital, Guiyang, 550003, China
| | - Wei Qu
- Department of Laboratory Medicine, Guiyang Maternity & Child Health Hospital, Guiyang, 550003, China.
| |
Collapse
|
8
|
Wang Y, Zou Y, Chen X, Li H, Yin Z, Zhang B, Xu Y, Zhang Y, Zhang R, Huang X, Yang W, Xu C, Jiang T, Tang Q, Zhou Z, Ji Y, Liu Y, Hu L, Zhou J, Zhou Y, Zhao J, Liu N, Huang G, Chang H, Fang W, Chen C, Zhou D. Innate immune responses against the fungal pathogen Candida auris. Nat Commun 2022; 13:3553. [PMID: 35729111 PMCID: PMC9213489 DOI: 10.1038/s41467-022-31201-x] [Citation(s) in RCA: 35] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Accepted: 06/08/2022] [Indexed: 12/12/2022] Open
Abstract
Candida auris is a multidrug-resistant human fungal pathogen responsible for nosocomial outbreaks worldwide. Although considerable progress has increased our understanding of the biological and clinical aspects of C. auris, its interaction with the host immune system is only now beginning to be investigated in-depth. Here, we compare the innate immune responses induced by C. auris BJCA001 and Candida albicans SC5314 in vitro and in vivo. Our results indicate that C. auris BJCA001 appears to be less immunoinflammatory than C. albicans SC5314, and this differential response correlates with structural features of the cell wall. Candida auris is a multidrug-resistant human fungal pathogen responsible for nosocomial outbreaks worldwide. Here, the authors identify differential innate immune responses induced by C. auris and Candida albicans in vitro and in vivo, which correlate with structural features of the cell wall.
Collapse
Affiliation(s)
- Yuanyuan Wang
- The Center for Microbes, Development and Health, CAS Key Laboratory of Molecular Virology and Immunology, Institut Pasteur of Shanghai, Chinese Academy of Sciences, Shanghai, 200031, China.,University of Chinese Academy of Sciences, Beijing, China.,Nanjing Advanced Academy of Life and Health, Nanjing, 211135, China
| | - Yun Zou
- The Center for Microbes, Development and Health, CAS Key Laboratory of Molecular Virology and Immunology, Institut Pasteur of Shanghai, Chinese Academy of Sciences, Shanghai, 200031, China.,University of Chinese Academy of Sciences, Beijing, China.,Nanjing Advanced Academy of Life and Health, Nanjing, 211135, China
| | - Xiaoqing Chen
- The Center for Microbes, Development and Health, CAS Key Laboratory of Molecular Virology and Immunology, Institut Pasteur of Shanghai, Chinese Academy of Sciences, Shanghai, 200031, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Hao Li
- Department of General Surgery, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200080, China
| | - Zhe Yin
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing, 100071, China
| | - Baocai Zhang
- State Key Laboratory of Plant Genomics, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, 100101, China
| | - Yongbin Xu
- Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Yiquan Zhang
- Wuxi School of Medicine, Jiangnan University, Wuxi, 214122, Jiangsu, China
| | - Rulin Zhang
- Department of Laboratory Medicine, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 20008, China
| | - Xinhua Huang
- The Center for Microbes, Development and Health, CAS Key Laboratory of Molecular Virology and Immunology, Institut Pasteur of Shanghai, Chinese Academy of Sciences, Shanghai, 200031, China
| | - Wenhui Yang
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing, 100071, China
| | - Chaoyue Xu
- The Center for Microbes, Development and Health, CAS Key Laboratory of Molecular Virology and Immunology, Institut Pasteur of Shanghai, Chinese Academy of Sciences, Shanghai, 200031, China.,Nanjing Advanced Academy of Life and Health, Nanjing, 211135, China.,College of Life Science, Shanghai University, Shanghai, China
| | - Tong Jiang
- The Center for Microbes, Development and Health, CAS Key Laboratory of Molecular Virology and Immunology, Institut Pasteur of Shanghai, Chinese Academy of Sciences, Shanghai, 200031, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Qinyu Tang
- Department of Dermatology, Tongji Hospital, Tongji University School of Medicine, Shanghai, 200065, China
| | - Zili Zhou
- The Center for Microbes, Development and Health, CAS Key Laboratory of Molecular Virology and Immunology, Institut Pasteur of Shanghai, Chinese Academy of Sciences, Shanghai, 200031, China
| | - Ying Ji
- The Center for Microbes, Development and Health, CAS Key Laboratory of Molecular Virology and Immunology, Institut Pasteur of Shanghai, Chinese Academy of Sciences, Shanghai, 200031, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Yingqi Liu
- School of Basic Medicine, Gannan Medical University, Ganzhou, China
| | - Lingfei Hu
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing, 100071, China
| | - Jia Zhou
- Center for Single-Cell Omics, School of Public Health, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Yao Zhou
- National Engineering Research Center for Non-Food Biorefinery, Guangxi Academy of Sciences, Nanning, 530007, Guangxi, China
| | - Jingjun Zhao
- Department of Dermatology, Tongji Hospital, Tongji University School of Medicine, Shanghai, 200065, China
| | - Ningning Liu
- Center for Single-Cell Omics, School of Public Health, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Guanghua Huang
- Department of Infectious Disease, Huashan Hospital and State Key Laboratory of Genetic Engineering, School of Life Sciences, Fudan University, Shanghai, China
| | - Haishuang Chang
- Shanghai Institute of Precision Medicine, Shanghai Ninth People's Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Wenxia Fang
- National Engineering Research Center for Non-Food Biorefinery, Guangxi Academy of Sciences, Nanning, 530007, Guangxi, China
| | - Changbin Chen
- The Center for Microbes, Development and Health, CAS Key Laboratory of Molecular Virology and Immunology, Institut Pasteur of Shanghai, Chinese Academy of Sciences, Shanghai, 200031, China. .,Nanjing Advanced Academy of Life and Health, Nanjing, 211135, China.
| | - Dongsheng Zhou
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing, 100071, China.
| |
Collapse
|
9
|
Gutierrez MW, van Tilburg Bernardes E, Changirwa D, McDonald B, Arrieta MC. "Molding" immunity-modulation of mucosal and systemic immunity by the intestinal mycobiome in health and disease. Mucosal Immunol 2022; 15:573-583. [PMID: 35474360 DOI: 10.1038/s41385-022-00515-w] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Revised: 03/24/2022] [Accepted: 04/04/2022] [Indexed: 02/04/2023]
Abstract
Fungi are important yet understudied contributors to the microbial communities of the gastrointestinal tract. Starting at birth, the intestinal mycobiome undergoes a period of dynamic maturation under the influence of microbial, host, and extrinsic influences, with profound functional implications for immune development in early life, and regulation of immune homeostasis throughout life. Candida albicans serves as a model organism for understanding the cross-talk between fungal colonization dynamics and immunity, and exemplifies unique mechanisms of fungal-immune interactions, including fungal dimorphism, though our understanding of other intestinal fungi is growing. Given the prominent role of the gut mycobiome in promoting immune homeostasis, emerging evidence points to fungal dysbiosis as an influential contributor to immune dysregulation in a variety of inflammatory and infectious diseases. Here we review current knowledge on the factors that govern host-fungi interactions in the intestinal tract and immunological outcomes in both mucosal and systemic compartments.
Collapse
Affiliation(s)
- Mackenzie W Gutierrez
- Immunology Research Group, Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada.,Department of Pediatrics, Alberta Children's Hospital Research Institute, University of Calgary, Calgary, AB, Canada.,Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada.,International Microbiome Centre, University of Calgary, Calgary, AB, Canada
| | - Erik van Tilburg Bernardes
- Immunology Research Group, Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada.,Department of Pediatrics, Alberta Children's Hospital Research Institute, University of Calgary, Calgary, AB, Canada.,Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada.,International Microbiome Centre, University of Calgary, Calgary, AB, Canada
| | - Diana Changirwa
- Immunology Research Group, Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada.,International Microbiome Centre, University of Calgary, Calgary, AB, Canada.,Department of Critical Care Medicine, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Braedon McDonald
- Immunology Research Group, Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada.,International Microbiome Centre, University of Calgary, Calgary, AB, Canada.,Department of Critical Care Medicine, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Marie-Claire Arrieta
- Immunology Research Group, Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada. .,Department of Pediatrics, Alberta Children's Hospital Research Institute, University of Calgary, Calgary, AB, Canada. .,Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada. .,International Microbiome Centre, University of Calgary, Calgary, AB, Canada.
| |
Collapse
|
10
|
Sobiepanek A, Kuryk Ł, Garofalo M, Kumar S, Baran J, Musolf P, Siebenhaar F, Fluhr JW, Kobiela T, Plasenzotti R, Kuchler K, Staniszewska M. The Multifaceted Roles of Mast Cells in Immune Homeostasis, Infections and Cancers. Int J Mol Sci 2022; 23:2249. [PMID: 35216365 PMCID: PMC8875910 DOI: 10.3390/ijms23042249] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2022] [Revised: 02/05/2022] [Accepted: 02/07/2022] [Indexed: 02/07/2023] Open
Abstract
Mast cells (MCs) play important roles in normal immune responses and pathological states. The location of MCs on the boundaries between tissues and the external environment, including gut mucosal surfaces, lungs, skin, and around blood vessels, suggests a multitude of immunological functions. Thus, MCs are pivotal for host defense against different antigens, including allergens and microbial pathogens. MCs can produce and respond to physiological mediators and chemokines to modulate inflammation. As long-lived, tissue-resident cells, MCs indeed mediate acute inflammatory responses such as those evident in allergic reactions. Furthermore, MCs participate in innate and adaptive immune responses to bacteria, viruses, fungi, and parasites. The control of MC activation or stabilization is a powerful tool in regulating tissue homeostasis and pathogen clearance. Moreover, MCs contribute to maintaining the homeostatic equilibrium between host and resident microbiota, and they engage in crosstalk between the resident and recruited hematopoietic cells. In this review, we provide a comprehensive overview of the functions of MCs in health and disease. Further, we discuss how mouse models of MC deficiency have become useful tools for establishing MCs as a potential cellular target for treating inflammatory disorders.
Collapse
Affiliation(s)
- Anna Sobiepanek
- Faculty of Chemistry, Warsaw University of Technology, Noakowskiego 3, 00-664 Warsaw, Poland; (A.S.); (J.B.); (P.M.); (T.K.)
| | - Łukasz Kuryk
- National Institute of Public Health NIH—National Institute of Research, 00-791 Warsaw, Poland;
- Clinical Science, Targovax Oy, Lars Sonckin kaari 14, 02600 Espoo, Finland;
| | - Mariangela Garofalo
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, Via F. Marzolo 5, 35131 Padova, Italy;
| | - Sandeep Kumar
- Clinical Science, Targovax Oy, Lars Sonckin kaari 14, 02600 Espoo, Finland;
| | - Joanna Baran
- Faculty of Chemistry, Warsaw University of Technology, Noakowskiego 3, 00-664 Warsaw, Poland; (A.S.); (J.B.); (P.M.); (T.K.)
| | - Paulina Musolf
- Faculty of Chemistry, Warsaw University of Technology, Noakowskiego 3, 00-664 Warsaw, Poland; (A.S.); (J.B.); (P.M.); (T.K.)
| | - Frank Siebenhaar
- Institute of Allergology, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, 10117 Berlin, Germany; (F.S.); (J.W.F.)
- Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, Allergology and Immunology, 12203 Berlin, Germany
| | - Joachim Wilhelm Fluhr
- Institute of Allergology, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, 10117 Berlin, Germany; (F.S.); (J.W.F.)
- Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, Allergology and Immunology, 12203 Berlin, Germany
| | - Tomasz Kobiela
- Faculty of Chemistry, Warsaw University of Technology, Noakowskiego 3, 00-664 Warsaw, Poland; (A.S.); (J.B.); (P.M.); (T.K.)
| | - Roberto Plasenzotti
- Department of Biomedical Research, Medical University of Vienna, Währingergürtel 18-20, 1090 Vienna, Austria;
| | - Karl Kuchler
- Max Perutz Labs Vienna, Center for Medical Biochemistry, Medical University of Vienna, Campus Vienna Biocenter, Dr. Bohr-Gasse 9/2, 1030 Vienna, Austria;
| | - Monika Staniszewska
- Centre for Advanced Materials and Technologies, Warsaw University of Technology, Poleczki 19, 02-822 Warsaw, Poland
| |
Collapse
|
11
|
Mata-Martínez P, Bergón-Gutiérrez M, del Fresno C. Dectin-1 Signaling Update: New Perspectives for Trained Immunity. Front Immunol 2022; 13:812148. [PMID: 35237264 PMCID: PMC8882614 DOI: 10.3389/fimmu.2022.812148] [Citation(s) in RCA: 58] [Impact Index Per Article: 29.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2021] [Accepted: 01/20/2022] [Indexed: 12/12/2022] Open
Abstract
The C-type lectin receptor Dectin-1 was originally described as the β-glucan receptor expressed in myeloid cells, with crucial functions in antifungal responses. However, over time, different ligands both of microbial-derived and endogenous origin have been shown to be recognized by Dectin-1. The outcomes of this recognition are diverse, including pro-inflammatory responses such as cytokine production, reactive oxygen species generation and phagocytosis. Nonetheless, tolerant responses have been also attributed to Dectin-1, depending on the specific ligand engaged. Dectin-1 recognition of their ligands triggers a plethora of downstream signaling pathways, with complex interrelationships. These signaling routes can be modulated by diverse factors such as phosphatases or tetraspanins, resulting either in pro-inflammatory or regulatory responses. Since its first depiction, Dectin-1 has recently gained a renewed attention due to its role in the induction of trained immunity. This process of long-term memory of innate immune cells can be triggered by β-glucans, and Dectin-1 is crucial for its initiation. The main signaling pathways involved in this process have been described, although the understanding of the above-mentioned complexity in the β-glucan-induced trained immunity is still scarce. In here, we have reviewed and updated all these factors related to the biology of Dectin-1, highlighting the gaps that deserve further research. We believe on the relevance to fully understand how this receptor works, and therefore, how we could harness it in different pathological conditions as diverse as fungal infections, autoimmunity, or cancer.
Collapse
Affiliation(s)
| | | | - Carlos del Fresno
- Immune response and Immunomodulation Group, Hospital La Paz Institute for Health Research (IdiPAZ), Madrid, Spain
| |
Collapse
|
12
|
Brandi P, Conejero L, Cueto FJ, Martínez-Cano S, Dunphy G, Gómez MJ, Relaño C, Saz-Leal P, Enamorado M, Quintas A, Dopazo A, Amores-Iniesta J, Del Fresno C, Nistal-Villán E, Ardavín C, Nieto A, Casanovas M, Subiza JL, Sancho D. Trained immunity induction by the inactivated mucosal vaccine MV130 protects against experimental viral respiratory infections. Cell Rep 2022; 38:110184. [PMID: 34986349 PMCID: PMC8755442 DOI: 10.1016/j.celrep.2021.110184] [Citation(s) in RCA: 39] [Impact Index Per Article: 19.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2020] [Revised: 09/11/2021] [Accepted: 12/07/2021] [Indexed: 12/13/2022] Open
Abstract
MV130 is an inactivated polybacterial mucosal vaccine that confers protection to patients against recurrent respiratory infections, including those of viral etiology. However, its mechanism of action remains poorly understood. Here, we find that intranasal prophylaxis with MV130 modulates the lung immune landscape and provides long-term heterologous protection against viral respiratory infections in mice. Intranasal administration of MV130 provides protection against systemic candidiasis in wild-type and Rag1-deficient mice lacking functional lymphocytes, indicative of innate immune-mediated protection. Moreover, pharmacological inhibition of trained immunity with metformin abrogates the protection conferred by MV130 against influenza A virus respiratory infection. MV130 induces reprogramming of both mouse bone marrow progenitor cells and in vitro human monocytes, promoting an enhanced cytokine production that relies on a metabolic shift. Our results unveil that the mucosal administration of a fully inactivated bacterial vaccine provides protection against viral infections by a mechanism associated with the induction of trained immunity.
Collapse
Affiliation(s)
- Paola Brandi
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
| | - Laura Conejero
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
| | - Francisco J Cueto
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
| | - Sarai Martínez-Cano
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain; Inmunotek S.L., Alcalá de Henares, Spain
| | - Gillian Dunphy
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
| | - Manuel J Gómez
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
| | - Carlos Relaño
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
| | - Paula Saz-Leal
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
| | - Michel Enamorado
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
| | - Ana Quintas
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
| | - Ana Dopazo
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
| | | | - Carlos Del Fresno
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain; Instituto de Investigación Biomédica del Hospital Universitario La Paz (IdiPAZ), Madrid, Spain
| | - Estanislao Nistal-Villán
- Microbiology Section, Department Pharmacological and Health Sciences, Facultad de Farmacia, Universidad CEU San Pablo, Madrid, Spain
| | - Carlos Ardavín
- Departamento de Inmunología y Oncología, Centro Nacional de Biotecnología/CSIC, Madrid, Spain
| | - Antonio Nieto
- Pediatric Pulmonology & Allergy Unit, Health Research Institute, La Fe University Hospital, Valencia, Spain
| | | | | | - David Sancho
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain.
| |
Collapse
|
13
|
Stothers CL, Burelbach KR, Owen AM, Patil NK, McBride MA, Bohannon JK, Luan L, Hernandez A, Patil TK, Williams DL, Sherwood ER. β-Glucan Induces Distinct and Protective Innate Immune Memory in Differentiated Macrophages. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2021; 207:2785-2798. [PMID: 34740960 PMCID: PMC8612974 DOI: 10.4049/jimmunol.2100107] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/03/2021] [Accepted: 09/27/2021] [Indexed: 12/14/2022]
Abstract
Bacterial infections are a common and deadly threat to vulnerable patients. Alternative strategies to fight infection are needed. β-Glucan, an immunomodulator derived from the fungal cell wall, provokes resistance to infection by inducing trained immunity, a phenomenon that persists for weeks to months. Given the durability of trained immunity, it is unclear which leukocyte populations sustain this effect. Macrophages have a life span that surpasses the duration of trained immunity. Thus, we sought to define the contribution of differentiated macrophages to trained immunity. Our results show that β-glucan protects mice from Pseudomonas aeruginosa infection by augmenting recruitment of innate leukocytes to the site of infection and facilitating local clearance of bacteria, an effect that persists for more than 7 d. Adoptive transfer of macrophages, trained using β-glucan, into naive mice conferred a comparable level of protection. Trained mouse bone marrow-derived macrophages assumed an antimicrobial phenotype characterized by enhanced phagocytosis and reactive oxygen species production in parallel with sustained enhancements in glycolytic and oxidative metabolism, increased mitochondrial mass, and membrane potential. β-Glucan induced broad transcriptomic changes in macrophages consistent with early activation of the inflammatory response, followed by sustained alterations in transcripts associated with metabolism, cellular differentiation, and antimicrobial function. Trained macrophages constitutively secreted CCL chemokines and robustly produced proinflammatory cytokines and chemokines in response to LPS challenge. Induction of the trained phenotype was independent of the classic β-glucan receptors Dectin-1 and TLR-2. These findings provide evidence that β-glucan induces enhanced protection from infection by driving trained immunity in macrophages.
Collapse
Affiliation(s)
- Cody L Stothers
- Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, TN;
| | - Katherine R Burelbach
- Department of Anesthesiology, Vanderbilt University Medical Center, Nashville, TN; and
| | - Allison M Owen
- Department of Anesthesiology, Vanderbilt University Medical Center, Nashville, TN; and
| | - Naeem K Patil
- Department of Anesthesiology, Vanderbilt University Medical Center, Nashville, TN; and
| | - Margaret A McBride
- Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, TN
| | - Julia K Bohannon
- Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, TN
- Department of Anesthesiology, Vanderbilt University Medical Center, Nashville, TN; and
| | - Liming Luan
- Department of Anesthesiology, Vanderbilt University Medical Center, Nashville, TN; and
| | - Antonio Hernandez
- Department of Anesthesiology, Vanderbilt University Medical Center, Nashville, TN; and
| | - Tazeen K Patil
- Department of Anesthesiology, Vanderbilt University Medical Center, Nashville, TN; and
| | - David L Williams
- Center for Inflammation, Infectious Disease and Immunity, Department of Surgery, James H. Quillen College of Medicine, East Tennessee State University, Johnson City, TN
| | - Edward R Sherwood
- Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, TN
- Department of Anesthesiology, Vanderbilt University Medical Center, Nashville, TN; and
| |
Collapse
|
14
|
Théroude C, Reverte M, Heinonen T, Ciarlo E, Schrijver IT, Antonakos N, Maillard N, Pralong F, Le Roy D, Roger T. Trained Immunity Confers Prolonged Protection From Listeriosis. Front Immunol 2021; 12:723393. [PMID: 34603295 PMCID: PMC8484647 DOI: 10.3389/fimmu.2021.723393] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2021] [Accepted: 09/06/2021] [Indexed: 12/11/2022] Open
Abstract
Trained immunity refers to the ability of the innate immune system exposed to a first challenge to provide an enhanced response to a secondary homologous or heterologous challenge. We reported that training induced with β-glucan one week before infection confers protection against a broad-spectrum of lethal bacterial infections. Whether this protection persists over time is unknown. To tackle this question, we analyzed the immune status and the response to Listeria monocytogenes (L. monocytogenes) of mice trained 9 weeks before analysis. The induction of trained immunity increased bone marrow myelopoiesis and blood counts of Ly6Chigh inflammatory monocytes and polymorphonuclear neutrophils (PMNs). Ex vivo, whole blood, PMNs and monocytes from trained mice produced increased levels of cytokines in response to microbial products and limited the growth of L. monocytogenes. In vivo, following challenge with L. monocytogenes, peripheral blood leukocytes were massively depleted in control mice but largely preserved in trained mice. PMNs were reduced also in the spleen from control mice, and increased in the spleen of trained mice. In transwell experiments, PMNs from trained mice showed increased spontaneous migration and CXCL2/MIP2α-induced chemotaxis, suggesting that training promotes the migration of PMNs in peripheral organs targeted by L. monocytogenes. Trained PMNs and monocytes had higher glycolytic activity and mitochondrial respiration than control cells when exposed to L. monocytogenes. Bacterial burden and dissemination in blood, spleen and liver as well as systemic cytokines and inflammation (multiplex bead assay and bioluminescence imaging) were reduced in trained mice. In full agreement with these results, mice trained 9 weeks before infection were powerfully protected from lethal listeriosis. Altogether, these data suggest that training increases the generation and the antimicrobial activity of PMNs and monocytes, which may confer prolonged protection from lethal bacterial infection.
Collapse
Affiliation(s)
- Charlotte Théroude
- Infectious Diseases Service, Department of Medicine, Lausanne University Hospital and University of Lausanne, Epalinges, Switzerland
| | - Marta Reverte
- Infectious Diseases Service, Department of Medicine, Lausanne University Hospital and University of Lausanne, Epalinges, Switzerland
| | - Tytti Heinonen
- Infectious Diseases Service, Department of Medicine, Lausanne University Hospital and University of Lausanne, Epalinges, Switzerland
| | - Eleonora Ciarlo
- Infectious Diseases Service, Department of Medicine, Lausanne University Hospital and University of Lausanne, Epalinges, Switzerland
| | - Irene T Schrijver
- Infectious Diseases Service, Department of Medicine, Lausanne University Hospital and University of Lausanne, Epalinges, Switzerland
| | - Nikolaos Antonakos
- Infectious Diseases Service, Department of Medicine, Lausanne University Hospital and University of Lausanne, Epalinges, Switzerland
| | - Nicolas Maillard
- Infectious Diseases Service, Department of Medicine, Lausanne University Hospital and University of Lausanne, Epalinges, Switzerland
| | - Florian Pralong
- Infectious Diseases Service, Department of Medicine, Lausanne University Hospital and University of Lausanne, Epalinges, Switzerland
| | - Didier Le Roy
- Infectious Diseases Service, Department of Medicine, Lausanne University Hospital and University of Lausanne, Epalinges, Switzerland
| | - Thierry Roger
- Infectious Diseases Service, Department of Medicine, Lausanne University Hospital and University of Lausanne, Epalinges, Switzerland
| |
Collapse
|
15
|
Ciarlo E, Heinonen T, Théroude C, Asgari F, Le Roy D, Netea MG, Roger T. Trained Immunity Confers Broad-Spectrum Protection Against Bacterial Infections. J Infect Dis 2021; 222:1869-1881. [PMID: 31889191 PMCID: PMC7653089 DOI: 10.1093/infdis/jiz692] [Citation(s) in RCA: 79] [Impact Index Per Article: 26.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2019] [Accepted: 12/30/2019] [Indexed: 12/29/2022] Open
Abstract
Background The innate immune system recalls a challenge to adapt to a secondary challenge, a phenomenon called trained immunity. Training involves cellular metabolic, epigenetic and functional reprogramming, but how broadly trained immunity protects from infections is unknown. For the first time, we addressed whether trained immunity provides protection in a large panel of preclinical models of infections. Methods Mice were trained and subjected to systemic infections, peritonitis, enteritis, and pneumonia induced by Staphylococcus aureus, Listeria monocytogenes, Escherichia coli, Citrobacter rodentium, and Pseudomonas aeruginosa. Bacteria, cytokines, leukocytes, and hematopoietic precursors were quantified in blood, bone marrow, and organs. The role of monocytes/macrophages, granulocytes, and interleukin 1 signaling was investigated using depletion or blocking approaches. Results Induction of trained immunity protected mice in all preclinical models, including when training and infection were initiated in distant organs. Trained immunity increased bone marrow hematopoietic progenitors, blood Ly6Chigh inflammatory monocytes and granulocytes, and sustained blood antimicrobial responses. Monocytes/macrophages and interleukin 1 signaling were required to protect trained mice from listeriosis. Trained mice were efficiently protected from peritonitis and listeriosis for up to 5 weeks. Conclusions Trained immunity confers broad-spectrum protection against lethal bacterial infections. These observations support the development of trained immunity-based strategies to improve host defenses.
Collapse
Affiliation(s)
- Eleonora Ciarlo
- Infectious Diseases Service, Department of Medicine, Lausanne University Hospital and University of Lausanne, Epalinges, Switzerland
| | - Tytti Heinonen
- Infectious Diseases Service, Department of Medicine, Lausanne University Hospital and University of Lausanne, Epalinges, Switzerland
| | - Charlotte Théroude
- Infectious Diseases Service, Department of Medicine, Lausanne University Hospital and University of Lausanne, Epalinges, Switzerland
| | - Fatemeh Asgari
- Department of Biomedical Sciences, Humanitas University, Milan, Italy
| | - Didier Le Roy
- Infectious Diseases Service, Department of Medicine, Lausanne University Hospital and University of Lausanne, Epalinges, Switzerland
| | - Mihai G Netea
- Radboud Center for Infectious Diseases, and Department of Internal Medicine, Radboud University Medical Center, Nijmegen, The Netherlands.,Department for Genomics & Immunoregulation, Life and Medical Sciences Institute, University of Bonn, Bonn, Germany
| | - Thierry Roger
- Infectious Diseases Service, Department of Medicine, Lausanne University Hospital and University of Lausanne, Epalinges, Switzerland
| |
Collapse
|
16
|
The double-sided effects of Mycobacterium Bovis bacillus Calmette-Guérin vaccine. NPJ Vaccines 2021; 6:14. [PMID: 33495451 PMCID: PMC7835355 DOI: 10.1038/s41541-020-00278-0] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2020] [Accepted: 12/14/2020] [Indexed: 01/30/2023] Open
Abstract
Bacillus Calmette-Guérin (BCG), the only vaccine proven to be effective against tuberculosis (TB), is the most commonly used vaccine globally. In addition to its effects on mycobacterial diseases, an increasing amount of epidemiological and experimental evidence accumulated since its introduction in 1921 has shown that BCG also exerts non-specific effects against a number of diseases, such as non-mycobacterial infections, allergies and certain malignancies. Recent Corona Virus Disease 2019 (COVID-19) outbreak has put BCG, a classic vaccine with significant non-specific protection, into the spotlight again. This literature review briefly covers the diverse facets of BCG vaccine, providing new perspectives in terms of specific and non-specific protection mechanisms of this old, multifaceted, and controversial vaccine.
Collapse
|
17
|
Walk J, Keramati F, de Bree LCJ, Arts RJW, Blok B, Netea MG, Stunnenberg HG, Sauerwein RW. Controlled Human Malaria Infection Induces Long-Term Functional Changes in Monocytes. Front Mol Biosci 2020; 7:604553. [PMID: 33324683 PMCID: PMC7726436 DOI: 10.3389/fmolb.2020.604553] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2020] [Accepted: 11/02/2020] [Indexed: 12/21/2022] Open
Abstract
Innate immune memory responses (also termed "trained immunity") have been described in monocytes after BCG vaccination and after stimulation in vitro with microbial and endogenous ligands such as LPS, β-glucan, oxidized LDL, and monosodium urate crystals. However, whether clinical infections are also capable of inducing a trained immunity phenotype remained uncertain. We evaluated whether Plasmodium falciparum infection can induce innate immune memory by measuring monocyte-derived cytokine production from five volunteers undergoing Controlled Human Malaria Infection. Monocyte responses followed a biphasic pattern: during acute infection, monocytes produced lower amounts of inflammatory cytokines upon secondary stimulation, but 36 days after malaria infection they produced significantly more IL-6 and TNF-α in response to various stimuli. Furthermore, transcriptomic and epigenomic data analysis revealed a clear reprogramming of monocytes at both timepoints, with long-term changes of H3K4me3 at the promoter regions of inflammatory genes that remain present for several weeks after parasite clearance. These findings demonstrate an epigenetic basis of trained immunity induced by human malaria in vivo.
Collapse
Affiliation(s)
- Jona Walk
- Department of Medical Microbiology and Radboud Center for Infectious Diseases, Radboud University Medical Center, Nijmegen, Netherlands.,Department of Internal Medicine and Radboud Center for Infectious Diseases, Radboud University Medical Center, Nijmegen, Netherlands
| | - Farid Keramati
- Department of Molecular Biology, Faculty of Science, Radboud University, Nijmegen, Netherlands
| | - L Charlotte J de Bree
- Department of Internal Medicine and Radboud Center for Infectious Diseases, Radboud University Medical Center, Nijmegen, Netherlands.,Research Center for Vitamins and Vaccines, Bandim Health Project, Statens Serum Institut, Copenhagen, Denmark.,Odense Patient Data Explorative Network, University of Southern Denmark/Odense University Hospital, Odense, Denmark
| | - Rob J W Arts
- Department of Internal Medicine and Radboud Center for Infectious Diseases, Radboud University Medical Center, Nijmegen, Netherlands
| | - Bas Blok
- Department of Internal Medicine and Radboud Center for Infectious Diseases, Radboud University Medical Center, Nijmegen, Netherlands.,Research Center for Vitamins and Vaccines, Bandim Health Project, Statens Serum Institut, Copenhagen, Denmark.,Odense Patient Data Explorative Network, University of Southern Denmark/Odense University Hospital, Odense, Denmark
| | - Mihai G Netea
- Department of Internal Medicine and Radboud Center for Infectious Diseases, Radboud University Medical Center, Nijmegen, Netherlands.,Department for Immunology and Metabolism, Life and Medical Sciences Institute (LIMES), University of Bonn, Bonn, Germany
| | - Hendrik G Stunnenberg
- Department of Molecular Biology, Faculty of Science, Radboud University, Nijmegen, Netherlands
| | - Robert W Sauerwein
- Department of Medical Microbiology and Radboud Center for Infectious Diseases, Radboud University Medical Center, Nijmegen, Netherlands
| |
Collapse
|
18
|
Adams K, Weber KS, Johnson SM. Exposome and Immunity Training: How Pathogen Exposure Order Influences Innate Immune Cell Lineage Commitment and Function. Int J Mol Sci 2020; 21:ijms21228462. [PMID: 33187101 PMCID: PMC7697998 DOI: 10.3390/ijms21228462] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2020] [Revised: 10/29/2020] [Accepted: 11/09/2020] [Indexed: 01/02/2023] Open
Abstract
Immune memory is a defining characteristic of adaptive immunity, but recent work has shown that the activation of innate immunity can also improve responsiveness in subsequent exposures. This has been coined “trained immunity” and diverges with the perception that the innate immune system is primitive, non-specific, and reacts to novel and recurrent antigen exposures similarly. The “exposome” is the cumulative exposures (diet, exercise, environmental exposure, vaccination, genetics, etc.) an individual has experienced and provides a mechanism for the establishment of immune training or immunotolerance. It is becoming increasingly clear that trained immunity constitutes a delicate balance between the dose, duration, and order of exposures. Upon innate stimuli, trained immunity or tolerance is shaped by epigenetic and metabolic changes that alter hematopoietic stem cell lineage commitment and responses to infection. Due to the immunomodulatory role of the exposome, understanding innate immune training is critical for understanding why some individuals exhibit protective phenotypes while closely related individuals may experience immunotolerant effects (e.g., the order of exposure can result in completely divergent immune responses). Research on the exposome and trained immunity may be leveraged to identify key factors for improving vaccination development, altering inflammatory disease development, and introducing potential new prophylactic treatments, especially for diseases such as COVID-19, which is currently a major health issue for the world. Furthermore, continued exposome research may prevent many deleterious effects caused by immunotolerance that frequently result in host morbidity or mortality.
Collapse
|
19
|
Ludwig K, Husain RA, Rubio I. mTORC1 Is Not Principally Involved in the Induction of Human Endotoxin Tolerance. Front Immunol 2020; 11:1515. [PMID: 32849516 PMCID: PMC7426365 DOI: 10.3389/fimmu.2020.01515] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2020] [Accepted: 06/09/2020] [Indexed: 11/13/2022] Open
Abstract
Endotoxin tolerance represents a safeguard mechanism for preventing detrimental prolonged inflammation and exaggerated immune/inflammatory responses from innate immune cells to recurrent harmless pathogens. On the other hand, excessive immune tolerance can contribute to pathological immunosuppression, e.g., as present in sepsis. Monocyte activation is accompanied by intracellular metabolic rearrangements that are reportedly orchestrated by the metabolic signaling node mTORC1. mTORC1-dependent metabolic re-wiring plays a major role in monocyte/macrophage polarization, but whether mTORC1 participates in the induction of endotoxin tolerance and other immune adaptive programs, such as immune training, is not clear. This connection has been difficult to test in the past due to the lack of appropriate models of human endotoxin tolerance allowing for the genetic manipulation of mTORC1. We have addressed this shortcoming by investigating monocytes from tuberous sclerosis (TSC) patients that feature a functional loss of the tumor suppressor TSC1/2 and a concomitant hyperactivation of mTORC1. Subjecting these cells to various protocols of immune priming and adaptation showed that the TSC monocytes are not compromised in the induction of tolerance. Analogously, we find that pharmacological mTORC1 inhibition does not prevent endotoxin tolerance induction in human monocytes. Interestingly, neither manipulation affected the capacity of activated monocytes to switch to increased lactic fermentation. In sum, our findings document that mTORC1 is unlikely to be involved in the induction of endotoxin tolerance in human monocytes and argue against a causal link between an mTORC1-dependent metabolic switch and the induction of immune tolerance.
Collapse
Affiliation(s)
- Kristin Ludwig
- Institute of Molecular Cell Biology, Center for Molecular Biomedicine, University Hospital Jena, Jena, Germany
| | - Ralf A Husain
- Department of Neuropediatrics, University Hospital Jena, Jena, Germany
| | - Ignacio Rubio
- Institute of Molecular Cell Biology, Center for Molecular Biomedicine, University Hospital Jena, Jena, Germany.,Clinic of Anaesthesiology and Intensive Care and Center for Sepsis Control and Care (CSCC), University Hospital Jena, Jena, Germany
| |
Collapse
|
20
|
Hatinguais R, Willment JA, Brown GD. PAMPs of the Fungal Cell Wall and Mammalian PRRs. Curr Top Microbiol Immunol 2020; 425:187-223. [PMID: 32180018 DOI: 10.1007/82_2020_201] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Fungi are opportunistic pathogens that infect immunocompromised patients and are responsible for an estimated 1.5 million deaths every year. The antifungal innate immune response is mediated through the recognition of pathogen-associated molecular patterns (PAMPs) by the host's pattern recognition receptors (PRRs). PRRs are immune receptors that ensure the internalisation and the killing of fungal pathogens. They also mount the inflammatory response, which contributes to initiate and polarise the adaptive response, controlled by lymphocytes. Both the innate and adaptive immune responses are required to control fungal infections. The immune recognition of fungal pathogen primarily occurs at the interface between the membrane of innate immune cells and the fungal cell wall, which contains a number of PAMPs. This chapter will focus on describing the main mammalian PRRs that have been shown to bind to PAMPs from the fungal cell wall of the four main fungal pathogens: Candida albicans, Aspergillus fumigatus, Cryptococcus neoformans and Pneumocystis jirovecii. We will describe these receptors, their functions and ligands to provide the reader with an overview of how the immune system recognises fungal pathogens and responds to them.
Collapse
Affiliation(s)
- Remi Hatinguais
- MRC Centre for Medical Mycology at University of Exeter, Geoffrey Pope Building, Stocker Road, Exeter, UK
| | - Janet A Willment
- MRC Centre for Medical Mycology at University of Exeter, Geoffrey Pope Building, Stocker Road, Exeter, UK
| | - Gordon D Brown
- MRC Centre for Medical Mycology at University of Exeter, Geoffrey Pope Building, Stocker Road, Exeter, UK.
| |
Collapse
|
21
|
Saz-Leal P, Del Fresno C, Brandi P, Martínez-Cano S, Dungan OM, Chisholm JD, Kerr WG, Sancho D. Targeting SHIP-1 in Myeloid Cells Enhances Trained Immunity and Boosts Response to Infection. Cell Rep 2019; 25:1118-1126. [PMID: 30380404 PMCID: PMC6226423 DOI: 10.1016/j.celrep.2018.09.092] [Citation(s) in RCA: 56] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2018] [Revised: 08/14/2018] [Accepted: 09/27/2018] [Indexed: 12/18/2022] Open
Abstract
β-Glucan-induced trained immunity in myeloid cells leads to long-term protection against secondary infections. Although previous studies have characterized this phenomenon, strategies to boost trained immunity remain undefined. We found that β-glucan-trained macrophages from mice with a myeloid-specific deletion of the phosphatase SHIP-1 (LysMΔSHIP-1) showed enhanced proinflammatory cytokine production in response to lipopolysaccharide. Following β-glucan training, SHIP-1-deficient macrophages exhibited increased phosphorylation of Akt and mTOR targets, correlating with augmented glycolytic metabolism. Enhanced training in the absence of SHIP-1 relied on histone methylation and acetylation. Trained LysMΔSHIP-1 mice produced increased amounts of proinflammatory cytokines upon rechallenge in vivo and were better protected against Candida albicans infection compared with control littermates. Pharmacological inhibition of SHIP-1 enhanced trained immunity against Candida infection in mouse macrophages and human peripheral blood mononuclear cells. Our data establish proof of concept for improvement of trained immunity and a strategy to achieve it by targeting SHIP-1.
Collapse
Affiliation(s)
- Paula Saz-Leal
- Immunobiology Lab, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Melchor Fernández Almagro 3, Madrid, 28029, Spain
| | - Carlos Del Fresno
- Immunobiology Lab, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Melchor Fernández Almagro 3, Madrid, 28029, Spain
| | - Paola Brandi
- Immunobiology Lab, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Melchor Fernández Almagro 3, Madrid, 28029, Spain
| | - Sarai Martínez-Cano
- Immunobiology Lab, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Melchor Fernández Almagro 3, Madrid, 28029, Spain
| | - Otto M Dungan
- Department of Chemistry, Syracuse University, Syracuse, NY 13210, USA
| | - John D Chisholm
- Department of Chemistry, Syracuse University, Syracuse, NY 13210, USA
| | - William G Kerr
- Department of Chemistry, Syracuse University, Syracuse, NY 13210, USA; Department of Microbiology and Immunology, State University of New York (SUNY) Upstate Medical University, Syracuse, NY 13210, USA; Pediatrics Department, SUNY Upstate Medical University, Syracuse, NY, USA; Centre d'Immunologie de Marseille-Luminy, Marseille, France
| | - David Sancho
- Immunobiology Lab, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Melchor Fernández Almagro 3, Madrid, 28029, Spain.
| |
Collapse
|
22
|
Abstract
Fungal bioactive polysaccharides are well known and have been widely used in Asia as a part of the traditional diet and medicine. In fact, some biopolymers (mainly β-glucans or glycoconjugate) have already made their way to the market as antitumor or immunostimulating drugs. In the last decades, the relationship between structure and activity of polysaccharides and their detailed mode of action have been the core of intense research to understand and utilize their medicinal properties. Most of the antitumor polysaccharides belong to conserved β-glucans, with a linear β-(1→3)-glucan backbone and attached β-(1→6) branch. Structurally different β-glucans appear to have different affinities toward their receptors and thus generate markedly different host responses. However, their antitumor activities are mainly influenced by molecular mass, degree of branching, conformation, and structure modification of the polysaccharides. β-Glucans act on several immune receptors including Dectin-1, complement receptor (CR3) and TLR-2/6, then trigger both innate and adaptive response and enhance opsonic and nonopsonic phagocytosis. Various receptor interactions explain the possible mode of actions of polysaccharides.
Collapse
Affiliation(s)
- Zhiyong Xiao
- Beijing Institute of Pharmacology and Toxicology, Beijing, China; State Key Laboratory of Toxicology and Medical Countermeasures, Beijing, China
| | - Wenxia Zhou
- Beijing Institute of Pharmacology and Toxicology, Beijing, China; State Key Laboratory of Toxicology and Medical Countermeasures, Beijing, China
| | - Yongxiang Zhang
- Beijing Institute of Pharmacology and Toxicology, Beijing, China; State Key Laboratory of Toxicology and Medical Countermeasures, Beijing, China.
| |
Collapse
|
23
|
Petit J, Embregts CWE, Forlenza M, Wiegertjes GF. Evidence of Trained Immunity in a Fish: Conserved Features in Carp Macrophages. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2019; 203:216-224. [PMID: 31127029 PMCID: PMC6571502 DOI: 10.4049/jimmunol.1900137] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/01/2019] [Accepted: 04/22/2019] [Indexed: 02/06/2023]
Abstract
Trained immunity is a form of innate immune memory best described in mice and humans. Clear evidence of the evolutionary conservation of trained immunity in teleost fish is lacking. Given the evolutionary position of teleosts as early vertebrates with a fully developed immune system, we hypothesize that teleost myeloid cells show features of trained immunity common to those observed in mammalian macrophages. These would at least include the ability of fish macrophages to mount heightened responses to a secondary stimulus in a nonspecific manner. We established an in vitro model to study trained immunity in fish by adapting a well-described culture system of head kidney-derived macrophages of common carp. A soluble NOD-specific ligand and a soluble β-glucan were used to train carp macrophages, after which cells were rested for 6 d prior to exposure to a secondary stimulus. Unstimulated trained macrophages displayed evidence of metabolic reprogramming as well as heightened phagocytosis and increased expression of the inflammatory cytokines il6 and tnf-α. Stimulated trained macrophages showed heightened production of reactive oxygen and nitrogen species as compared with the corresponding stimulated but untrained cells. We discuss the value of our findings for future studies on trained immunity in teleost fish.
Collapse
Affiliation(s)
- Jules Petit
- Cell Biology and Immunology Group, Wageningen University & Research, 6708 WD Wageningen, the Netherlands; and
| | - Carmen W E Embregts
- Cell Biology and Immunology Group, Wageningen University & Research, 6708 WD Wageningen, the Netherlands; and
| | - Maria Forlenza
- Cell Biology and Immunology Group, Wageningen University & Research, 6708 WD Wageningen, the Netherlands; and
| | - Geert F Wiegertjes
- Cell Biology and Immunology Group, Wageningen University & Research, 6708 WD Wageningen, the Netherlands; and
- Aquaculture and Fisheries Group, Wageningen University & Research, 6708 WD Wageningen, the Netherlands
| |
Collapse
|
24
|
Tamai R, Kiyoura Y. Heat-killed Candida albicans augments synthetic bacterial component-induced proinflammatory cytokine production. Folia Microbiol (Praha) 2019; 64:555-566. [PMID: 30656591 DOI: 10.1007/s12223-019-00679-2] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2018] [Accepted: 01/07/2019] [Indexed: 12/23/2022]
Abstract
Candida albicans can enhance the invasion of oral epithelial cells by Porphyromonas gingivalis, although the fungus is not a periodontal pathogen. In this study, we investigated whether C. albicans augments proinflammatory cytokine production by mouse macrophage-like J774.1 cells incubated with synthetic bacterial components. Mouse macrophage-like J774.1 cells, mouse primary splenocytes, human THP-1 cells, and A549 cells were pretreated with or without heat-killed C. albicans (HKCA) or substitutes for C. albicans cell wall components in 96-well flat-bottomed plates. Cells were then washed and incubated with Pam3CSK4, a Toll-like receptor (TLR) 2 ligand, or lipid A, a TLR4 ligand. Culture supernatants were analyzed by ELISA for secreted IL-6, MCP-1, TNF-α, and IL-8. HKCA augmented TLR ligand-induced proinflammatory cytokine production by J774.1 cells, mouse splenocytes, and THP-1 cells, but not A549 cells. However, IL-6, MCP-1, and TNF-α production induced by Pam3CSK4 or lipid A was not augmented when cells were pretreated with curdlan, a dectin-1 ligand, or mannan, a dectin-2 ligand. In contrast, pretreatment of cells with TLR ligands upregulated the production of IL-6 and TNF-α, but not MCP-1, induced by Pam3CSK4 or lipid A. The results suggest that C. albicans augments synthetic bacterial component-induced cytokine production by J774.1 cells via the TLR pathway, but not the dectin-1 or dectin-2 pathway.
Collapse
Affiliation(s)
- Riyoko Tamai
- Department of Oral Medical Science, Ohu University School of Dentistry, 31-1 Misumido, Tomitamachi, Koriyama, Fukushima, 963-8611, Japan.
| | - Yusuke Kiyoura
- Department of Oral Medical Science, Ohu University School of Dentistry, 31-1 Misumido, Tomitamachi, Koriyama, Fukushima, 963-8611, Japan
| |
Collapse
|
25
|
Role of the small GTPase Rho1 in cell wall integrity, stress response, and pathogenesis of Aspergillus fumigatus. Fungal Genet Biol 2018; 120:30-41. [PMID: 30205199 DOI: 10.1016/j.fgb.2018.09.003] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2018] [Revised: 09/04/2018] [Accepted: 09/08/2018] [Indexed: 11/24/2022]
Abstract
Aspergillus fumigatus is a major pathogen of invasive pulmonary aspergillosis. The small GTPase, Rho1, of A. fumigatus is reported to comprise a potential regulatory subunit of β-1,3-glucan synthase and is indispensable for fungal viability; however, the role of AfRho1 on the growth, cell wall integrity, and pathogenesis of A. fumigatus is still poorly understood. We constructed A. fumigatus mutants with conditional- and overexpression of Rho1 and found that defects of AfRho1 expression led to the reduction of β-1,3-glucan and glucosamine moieties on the cell wall, with down-regulated transcription of genes in the cell wall integrity signaling pathway and a decrease of calcofluor white (CFW)-stimulated mitogen-activated protein kinase (MpkA) phosphorylation and cytoplasmic leakage compared to those of the wild-type strain (WT). In addition, down-regulation of AfRho1 expression caused much higher sensitivity of A. fumigatus to H2O2 and alkaline pH compared to that of WT. Decrease of AfRho1 expression also attenuated the A. fumigatus pathogenicity in Galleria mellonella and inhibited conidial internalization into lung epithelial cells and inflammatory factor release. In contrast, overexpression of Rho1 did not alter A. fumigatus morphology, susceptibility to cell wall stresses, or pathogenicity relative to its parental strain. Taken together, our findings support AfRho1 as an essential regulator of the cell wall integrity, stress response, and pathogenesis of A. fumigatus.
Collapse
|
26
|
Del Fresno C, Iborra S, Saz-Leal P, Martínez-López M, Sancho D. Flexible Signaling of Myeloid C-Type Lectin Receptors in Immunity and Inflammation. Front Immunol 2018; 9:804. [PMID: 29755458 PMCID: PMC5932189 DOI: 10.3389/fimmu.2018.00804] [Citation(s) in RCA: 72] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2017] [Accepted: 04/03/2018] [Indexed: 12/19/2022] Open
Abstract
Myeloid C-type lectin receptors (CLRs) are important sensors of self and non-self that work in concert with other pattern recognition receptors (PRRs). CLRs have been previously classified based on their signaling motifs as activating or inhibitory receptors. However, specific features of the ligand binding process may result in distinct signaling through a single motif, resulting in the triggering of non-canonical pathways. In addition, CLR ligands are frequently exposed in complex structures that simultaneously bind different CLRs and other PRRs, which lead to integration of heterologous signaling among diverse receptors. Herein, we will review how sensing by myeloid CLRs and crosstalk with heterologous receptors is modulated by many factors affecting their signaling and resulting in differential outcomes for immunity and inflammation. Finding common features among those flexible responses initiated by diverse CLR-ligand partners will help to harness CLR function in immunity and inflammation.
Collapse
Affiliation(s)
- Carlos Del Fresno
- Immunobiology Laboratory, Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Madrid, Spain
| | - Salvador Iborra
- Immunobiology Laboratory, Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Madrid, Spain.,Department of Immunology, School of Medicine, Universidad Complutense de Madrid, 12 de Octubre Health Research Institute (imas12), Madrid, Spain
| | - Paula Saz-Leal
- Immunobiology Laboratory, Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Madrid, Spain
| | - María Martínez-López
- Immunobiology Laboratory, Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Madrid, Spain
| | - David Sancho
- Immunobiology Laboratory, Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Madrid, Spain
| |
Collapse
|
27
|
Walachowski S, Tabouret G, Fabre M, Foucras G. Molecular Analysis of a Short-term Model of β-Glucans-Trained Immunity Highlights the Accessory Contribution of GM-CSF in Priming Mouse Macrophages Response. Front Immunol 2017; 8:1089. [PMID: 28955331 PMCID: PMC5601002 DOI: 10.3389/fimmu.2017.01089] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2017] [Accepted: 08/21/2017] [Indexed: 11/16/2022] Open
Abstract
β-Glucans (BGs) are glucose polymers present in the fungal cell wall (CW) and, as such, are recognized by innate immune cells as microbial-associated pattern through Dectin-1 receptor. Recent studies have highlighted the ability of the pathogenic yeast Candida albicans or its CW-derived β(1,3) (1,6)-glucans to increase human monocytes cytokine secretion upon secondary stimulation, a phenomenon now referred as immune training. This ability of monocytes programming confers BGs an undeniable immunotherapeutic potential. Our objective was to determine whether BGs from Saccharomyces cerevisiae, a non-pathogenic yeast, are endowed with such a property. For this purpose, we have developed a short-term training model based on lipopolysaccharide re-stimulation of mouse bone marrow-derived macrophages primed with S. cerevisiae BGs. Through a transcriptome analysis, we demonstrated that BGs induced a specific gene expression signature involving the PI3K/AKT signaling pathway as in human monocytes. Moreover, we showed that over-expression of Csf2 (that encodes for GM-CSF) was a Dectin-1-dependent feature of BG-induced priming of macrophages. Further experiments confirmed that GM-CSF up-regulated Dectin-1 cell surface expression and amplified macrophages response along BG-mediated training. However, the blockade of GM-CSFR demonstrated that GM-CSF was not primarily required for BG-induced training of macrophages although it can substantially improve it. In addition, we found that mouse macrophages trained with BGs upregulated their expression of the four and a half LIM-only protein 2 (Fhl2) in a Dectin-1-dependent manner. Consistently, we observed that intracellular levels of FHL2 increased after stimulation of macrophages with BGs. In conclusion, our experiments provide new insights on GM-CSF contribution to the training of cells from the monocytic lineage and highlights FHL2 as a possible regulator of BG-associated signaling.
Collapse
Affiliation(s)
| | | | - Marion Fabre
- Université de Toulouse, INRA, INP, ENVT, IHAP, Toulouse, France
| | - Gilles Foucras
- Université de Toulouse, INRA, INP, ENVT, IHAP, Toulouse, France
| |
Collapse
|
28
|
Ostrop J, Lang R. Contact, Collaboration, and Conflict: Signal Integration of Syk-Coupled C-Type Lectin Receptors. THE JOURNAL OF IMMUNOLOGY 2017; 198:1403-1414. [PMID: 28167651 DOI: 10.4049/jimmunol.1601665] [Citation(s) in RCA: 53] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/30/2016] [Accepted: 11/14/2016] [Indexed: 12/13/2022]
Abstract
Several spleen tyrosine kinase-coupled C-type lectin receptors (CLRs) have emerged as important pattern recognition receptors for infectious danger. Because encounter with microbial pathogens leads to the simultaneous ligation of several CLRs and TLRs, the signals emanating from different pattern recognition receptors have to be integrated to achieve appropriate biological responses. In this review, we briefly summarize current knowledge about ligand recognition and core signaling by Syk-coupled CLRs. We then address mechanisms of synergistic and antagonistic crosstalk between different CLRs and with TLRs. Emerging evidence suggests that signal integration occurs through 1) direct interaction between receptors, 2) regulation of expression levels and localization, and 3) collaborative or conflicting signaling interference. Accordingly, we aim to provide a conceptual framework for the complex and sometimes unexpected outcome of CLR ligation in bacterial and fungal infection.
Collapse
Affiliation(s)
- Jenny Ostrop
- Center of Molecular Inflammation Research, Norwegian University of Science and Technology, 7491 Trondheim, Norway; .,Department of Cancer Research and Molecular Medicine, Norwegian University of Science and Technology, 7491 Trondheim, Norway; and
| | - Roland Lang
- Mikrobiologisches Institut-Klinische Mikrobiologie, Immunologie und Hygiene, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, 91054 Erlangen, Germany
| |
Collapse
|
29
|
Michalski C, Kan B, Lavoie PM. Antifungal Immunological Defenses in Newborns. Front Immunol 2017; 8:281. [PMID: 28360910 PMCID: PMC5350100 DOI: 10.3389/fimmu.2017.00281] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2017] [Accepted: 02/28/2017] [Indexed: 12/28/2022] Open
Abstract
Newborns are prone to fungal infections, largely due to Candida species. The immunological basis for this vulnerability is not yet fully understood. However, useful insights can be gained from the knowledge of the maturation of immune pathways during ontogeny, particularly when placed in context with how rare genetic mutations in humans predispose to fungal diseases. In this article, we review these most current data on immune functions in human newborns, highlighting pathways most relevant to the response to Candida. While discussing these data, we propose a framework of why deficiencies in these pathways make newborns particularly vulnerable to this opportunistic pathogen.
Collapse
Affiliation(s)
- Christina Michalski
- British Columbia Children's Hospital Research Institute, Vancouver, BC, Canada; Department of Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Bernard Kan
- British Columbia Children's Hospital Research Institute, Vancouver, BC, Canada; Department of Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Pascal M Lavoie
- British Columbia Children's Hospital Research Institute, Vancouver, BC, Canada; Department of Medicine, University of British Columbia, Vancouver, BC, Canada; Department of Pediatrics, University of British Columbia, Vancouver, BC, Canada
| |
Collapse
|
30
|
Wang W, Zhang T, Wang L, Xu J, Li M, Zhang A, Qiu L, Song L. A new non-phagocytic TLR6 with broad recognition ligands from Pacific oyster Crassostrea gigas. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2016; 65:182-190. [PMID: 27443817 DOI: 10.1016/j.dci.2016.07.010] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/04/2016] [Revised: 07/16/2016] [Accepted: 07/18/2016] [Indexed: 06/06/2023]
Abstract
Toll like receptors (TLRs) are evolutionarily prevalent recognition molecules in the Animalia and Plantae kingdom, which play vital roles in immune defense and homeostasis maintenance. Recently, the expansion of TLRs has been reported in invertebrate genomes, but the characters and immune functions of these expanded TLRs were still not well known. In the present study, a new member of TLR family with five LRR domains was identified in Crassostrea gigas (designated CgTLR6). It shared homology with TLRs from other organisms with the closest phylogenic relationship with molluscan TLRs. The recombinant protein of CgTLR6 (rCgTLR6) displayed direct bind activity to gram-negative bacteria Vibrio anguillarum and Vibrio splendidus, gram-positive bacteria Staphylococci aureus and Micrococcus luteus, and fungi Pichia pastoris, but not to fungi Yarrowia lipolytica. It also exhibited affinity to lipopolysaccharide (LPS) and peptidoglycan (PGN), while no affinity to mannan (MAN). The mRNA of CgTLR6 was mainly detected in hemocytes and hepatopancreas, and was significantly induced (p < 0.01) in hemocytes after the oyster was stimulated with LPS, PGN or bacteria V. splendidus. Immunofluorescence analysis indicated that CgTLR6 was mainly located at the membrane of hemocytes. The blockage of CgTLR6 by anti-rCgTLR6 antibody did not significantly inhibit the phagocytic rates of hemocytes toward recognized gram-negative bacteria V. anguillarum and V. splendidus, and unrecognized fungi Y. lipolytica. These results collectively implied that CgTLR6 was a novel non-phagocytic receptor of C. gigas to mediate humoral immune response by recognizing pathogen-associated molecular patterns on the invaders.
Collapse
Affiliation(s)
- Weilin Wang
- Key Laboratory of Experimental Marine Biology, Institute of Oceanology, Chinese Academy of Sciences, Qingdao 266071, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Tao Zhang
- Key Laboratory of Experimental Marine Biology, Institute of Oceanology, Chinese Academy of Sciences, Qingdao 266071, China
| | - Lingling Wang
- Key Laboratory of Mariculture & Stock Enhancement in North China's Sea, Ministry of Agriculture, Dalian Ocean University, Dalian 116023, China.
| | - Jiachao Xu
- Key Laboratory of Experimental Marine Biology, Institute of Oceanology, Chinese Academy of Sciences, Qingdao 266071, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Meijia Li
- Key Laboratory of Experimental Marine Biology, Institute of Oceanology, Chinese Academy of Sciences, Qingdao 266071, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Anguo Zhang
- Key Laboratory of Mariculture & Stock Enhancement in North China's Sea, Ministry of Agriculture, Dalian Ocean University, Dalian 116023, China
| | - Limei Qiu
- Key Laboratory of Experimental Marine Biology, Institute of Oceanology, Chinese Academy of Sciences, Qingdao 266071, China
| | - Linsheng Song
- Key Laboratory of Mariculture & Stock Enhancement in North China's Sea, Ministry of Agriculture, Dalian Ocean University, Dalian 116023, China
| |
Collapse
|
31
|
Hau CS, Tada Y, Kanda N, Watanabe S. Immunoresponses in dermatomycoses. J Dermatol 2016; 42:236-44. [PMID: 25736316 DOI: 10.1111/1346-8138.12718] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2014] [Accepted: 10/16/2014] [Indexed: 12/19/2022]
Abstract
Contact with fungal pathogens initiates a series of host responses beginning with innate immunity, which leads to fungal recognition and microbial killing. The innate immune system also modulates the adaptive immune responses, leading to the establishment of immunological memory and protection against pathogens. In the case of dimorphic fungi such as Candida albicans and Malassezia, the immune system plays an important role in tolerance and resistance when managing the organisms either as commensal microbiota or invading pathogens, and disruption of this balance can result in pathological consequences for the host. In addition, Malassezia and dermatophytes have immunomodulatory capabilities that allow them to adapt to their environments and they may exert different effects in healthy and diseased skin. Here, we discuss the host immune responses to dermatomycoses caused by dimorphic fungi such as C. albicans and Malassezia as well as dermatophytes such as Trichophyton spp. and Arthroderma benhamiae to gain a better understanding of the mechanisms of the host-dermatomycosis interaction.
Collapse
Affiliation(s)
- Carren Sy Hau
- Department of Dermatology, Teikyo University School of Medicine, Tokyo, Japan
| | | | | | | |
Collapse
|
32
|
Stevens WBC, Netea MG, Kater AP, van der Velden WJFM. 'Trained immunity': consequences for lymphoid malignancies. Haematologica 2016; 101:1460-1468. [PMID: 27903713 DOI: 10.3324/haematol.2016.149252] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2016] [Accepted: 06/29/2016] [Indexed: 12/15/2022] Open
Abstract
In hematological malignancies complex interactions exist between the immune system, microorganisms and malignant cells. On one hand, microorganisms can induce cancer, as illustrated by specific infection-induced lymphoproliferative diseases such as Helicobacter pylori-associated gastric mucosa-associated lymphoid tissue lymphoma. On the other hand, malignant cells create an immunosuppressive environment for their own benefit, but this also results in an increased risk of infections. Disrupted innate immunity contributes to the neoplastic transformation of blood cells by several mechanisms, including the uncontrolled clearance of microbial and autoantigens resulting in chronic immune stimulation and proliferation, chronic inflammation, and defective immune surveillance and anti-cancer immunity. Restoring dysfunction or enhancing responsiveness of the innate immune system might therefore represent a new angle for the prevention and treatment of hematological malignancies, in particular lymphoid malignancies and associated infections. Recently, it has been shown that cells of the innate immune system, such as monocytes/macrophages and natural killer cells, harbor features of immunological memory and display enhanced functionality long-term after stimulation with certain microorganisms and vaccines. These functional changes rely on epigenetic reprogramming and have been termed 'trained immunity'. In this review the concept of 'trained immunity' is discussed in the setting of lymphoid malignancies. Amelioration of infectious complications and hematological disease progression can be envisioned to result from the induction of trained immunity, but future studies are required to prove this exciting new hypothesis.
Collapse
Affiliation(s)
- Wendy B C Stevens
- Department of Hematology, Radboud University Medical Centre, Nijmegen
| | - Mihai G Netea
- Department of Internal Medicine, Radboud University Medical Centre, and Radboud Center for Infectious Diseases, Nijmegen.,Radboud Institute of Health Sciences, Radboud University Medical Center, Nijmegen
| | - Arnon P Kater
- Department of Hematology, Lymphoma and Myeloma Center Amsterdam (LYMMCARE) Academic Medical Center, University of Amsterdam, The Netherlands
| | - Walter J F M van der Velden
- Department of Hematology, Radboud University Medical Centre, Nijmegen .,Radboud Institute of Health Sciences, Radboud University Medical Center, Nijmegen
| |
Collapse
|
33
|
Minato KI, Laan LC, Ohara A, van Die I. Pleurotus citrinopileatus polysaccharide induces activation of human dendritic cells through multiple pathways. Int Immunopharmacol 2016; 40:156-163. [DOI: 10.1016/j.intimp.2016.08.034] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2016] [Revised: 08/23/2016] [Accepted: 08/25/2016] [Indexed: 01/10/2023]
|
34
|
Zhao GQ, Lin J, Hu LT, Yin XN, Wang Q, Xu Q, Li H. The role of Dectin-1/Raf-1 signal cascade in innate immune of human corneal epithelial cells against Aspergillus fumigatus infection. Int J Ophthalmol 2016; 9:1371-1375. [PMID: 27803850 DOI: 10.18240/ijo.2016.10.01] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2016] [Accepted: 04/12/2016] [Indexed: 12/11/2022] Open
Abstract
AIM To investigate the expression of the v-raf-1 murine leukemia viral oncogene homolog 1 (Raf-1) and its role in the innate immune response of human corneal epithelial cells (HCECs) infected by Aspergillus fumigatus. METHODS HCECs were cultured in vitro. They were randomly divided into 4 groups, including control group, Aspergillus fumigatus group, GW5074 (an inhibitor of Raf-1) group and Laminarin [an inhibitor of Dendriti-cell-associated C-type lectin 1 (Dectin-1)] group. The protein expression level of total Raf-1 and p-Raf-1was measured by Western blot. The expression of IL-6 and IL-8 mRNA in each group was detected by real-time polymerase chain reaction. RESULTS In Aspergillus fumigatus group, total Raf-1 protein levels in HCECs remained unchanged at 5, 15, 30 and 45min after infection, while p-Raf-1 expression was significantly enhanced at 30min after infection compared with control group. However, the expression of p-Raf-1 was apparently declined after treated with GW5074 or Laminarin compared with Aspergillus fumigatus group. The expression levels of IL-6, IL-8 mRNA were significantly increased after stimulation with fumigatus compared with control group. Pre-treated with GW5074 significantly inhibited Aspergillus fumigatus-induced upregulation of IL-8 and IL-6. CONCLUSION Aspergillus fumigatus stimulation can elevate the expression of p-Raf-1 in HCECs in vitro. Dectin-1/Raf-1 signal pathway may play a role on regulating the expression of inflammatory cytokines, including IL-6 and IL-8.
Collapse
Affiliation(s)
- Gui-Qiu Zhao
- Department of Ophthalmology, the Affiliated Hospital of Qingdao University, Qingdao 266003, Shandong Province, China
| | - Jing Lin
- Department of Ophthalmology, the Affiliated Hospital of Qingdao University, Qingdao 266003, Shandong Province, China
| | - Li-Ting Hu
- Department of Ophthalmology, the Affiliated Hospital of Qingdao University, Qingdao 266003, Shandong Province, China
| | - Xiao-Ni Yin
- Department of Ophthalmology, the Affiliated Hospital of Qingdao University, Qingdao 266003, Shandong Province, China
| | - Qian Wang
- Department of Ophthalmology, the Affiliated Hospital of Qingdao University, Qingdao 266003, Shandong Province, China
| | - Qiang Xu
- Department of Ophthalmology, the Affiliated Hospital of Qingdao University, Qingdao 266003, Shandong Province, China
| | - Hui Li
- Department of Ophthalmology, the Affiliated Hospital of Qingdao University, Qingdao 266003, Shandong Province, China
| |
Collapse
|
35
|
Rizzetto L, Ifrim DC, Moretti S, Tocci N, Cheng SC, Quintin J, Renga G, Oikonomou V, De Filippo C, Weil T, Blok BA, Lenucci MS, Santos MAS, Romani L, Netea MG, Cavalieri D. Fungal Chitin Induces Trained Immunity in Human Monocytes during Cross-talk of the Host with Saccharomyces cerevisiae. J Biol Chem 2016; 291:7961-72. [PMID: 26887946 DOI: 10.1074/jbc.m115.699645] [Citation(s) in RCA: 75] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2015] [Indexed: 11/06/2022] Open
Abstract
The immune system is essential to maintain the mutualistic homeostatic interaction between the host and its micro- and mycobiota. Living as a commensal,Saccharomyces cerevisiaecould potentially shape the immune response in a significant way. We observed thatS. cerevisiaecells induce trained immunity in monocytes in a strain-dependent manner through enhanced TNFα and IL-6 production upon secondary stimulation with TLR ligands, as well as bacterial and fungal commensals. Differential chitin content accounts for the differences in training properties observed among strains, driving induction of trained immunity by increasing cytokine production and direct antimicrobial activity bothin vitroandin vivo These chitin-induced protective properties are intimately associated with its internalization, identifying a critical role of phagosome acidification to facilitate microbial digestion. This study reveals how commensal and passenger microorganisms could be important in promoting health and preventing mucosal diseases by modulating host defense toward pathogens and thus influencing the host microbiota-immune system interactions.
Collapse
Affiliation(s)
- Lisa Rizzetto
- From the Research and Innovation Centre, Fondazione Edmund Mach, 38010 San Michele all'Adige TN, Italy
| | - Daniela C Ifrim
- the Department of Internal Medicine, Division of Experimental Internal Medicine, Radboud University Medical Center, 6525 GA Nijmegen, The Netherlands
| | - Silvia Moretti
- the Department of Experimental Medicine, University of Perugia, Polo Unico Sant'Andrea delle Fratte, 06123 Perugia PG, Italy
| | - Noemi Tocci
- From the Research and Innovation Centre, Fondazione Edmund Mach, 38010 San Michele all'Adige TN, Italy
| | - Shih-Chin Cheng
- the Department of Internal Medicine, Division of Experimental Internal Medicine, Radboud University Medical Center, 6525 GA Nijmegen, The Netherlands
| | - Jessica Quintin
- the Department of Internal Medicine, Division of Experimental Internal Medicine, Radboud University Medical Center, 6525 GA Nijmegen, The Netherlands
| | - Giorgia Renga
- the Department of Experimental Medicine, University of Perugia, Polo Unico Sant'Andrea delle Fratte, 06123 Perugia PG, Italy
| | - Vasilis Oikonomou
- the Department of Experimental Medicine, University of Perugia, Polo Unico Sant'Andrea delle Fratte, 06123 Perugia PG, Italy
| | - Carlotta De Filippo
- From the Research and Innovation Centre, Fondazione Edmund Mach, 38010 San Michele all'Adige TN, Italy, the Institute of Biometeorology, National Research Council, 50145 Florence, Italy
| | - Tobias Weil
- From the Research and Innovation Centre, Fondazione Edmund Mach, 38010 San Michele all'Adige TN, Italy
| | - Bastiaan A Blok
- the Department of Internal Medicine, Division of Experimental Internal Medicine, Radboud University Medical Center, 6525 GA Nijmegen, The Netherlands
| | - Marcello S Lenucci
- the Dipartimento di Scienze e Tecnologie Biologiche ed Ambientali, Università del Salento, 73100 Lecce LE, Italy
| | - Manuel A S Santos
- the Department of Biology and CESAM (Centro de Estudos do Ambiente e do Mar), University of Aveiro, 3810-193 Aveiro, Portugal, and
| | - Luigina Romani
- the Department of Experimental Medicine, University of Perugia, Polo Unico Sant'Andrea delle Fratte, 06123 Perugia PG, Italy
| | - Mihai G Netea
- the Department of Internal Medicine, Division of Experimental Internal Medicine, Radboud University Medical Center, 6525 GA Nijmegen, The Netherlands
| | - Duccio Cavalieri
- From the Research and Innovation Centre, Fondazione Edmund Mach, 38010 San Michele all'Adige TN, Italy, the Institute of Biometeorology, National Research Council, 50145 Florence, Italy, the Department of Biology, University of Florence, 50019 Sesto Fiorentino FI, Italy
| |
Collapse
|
36
|
Walachowski S, Tabouret G, Foucras G. Triggering Dectin-1-Pathway Alone Is Not Sufficient to Induce Cytokine Production by Murine Macrophages. PLoS One 2016; 11:e0148464. [PMID: 26840954 PMCID: PMC4739705 DOI: 10.1371/journal.pone.0148464] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2015] [Accepted: 01/17/2016] [Indexed: 02/07/2023] Open
Abstract
β-glucans (BG) are abundant polysaccharides of the Saccharomyces cerevisiae cell wall (Sc CW), an industry byproduct. They have immuno-stimulatory properties upon engagement of dectin-1 (Clec7a), their main receptor on particular immune cells, and they actually become of great interest because of their preventive or therapeutic potentials. Zymosan, a crude extract of Sc CW was studied as a prototypic BG, despite its miscellaneous PAMPs content. Here, we examined the response of murine wild type or Clec7a-/- bone marrow-derived macrophages (BMDM) to products with increasing BG content (15, 65 or 75%) and compared their effects with those of other dectin-1 ligands. The enrichment process removed TLR ligands while preserving dectin-1 activity. The most enriched extracts have very low NFκB activity and triggered low amounts of cytokine production in contrast with crude products like zymosan and BG15. Furthermore, MyD88-/- BMDM did not produce TNFα in response to crude Sc CW extracts, whereas their response to BG-enriched extracts was unaffected, suggesting that BG alone are not able to initiate cytokine secretion. Although Sc CW-derived BG stimulated the late and strong expression of Csf2 in a dectin-1-dependent manner, they remain poor inducers of chemokine and cytokine production in murine macrophages.
Collapse
Affiliation(s)
- Sarah Walachowski
- Université de Toulouse, INP-ENVT, UMR 1225, IHAP, Toulouse, France
- INRA, UMR1225, IHAP, Toulouse, France
| | - Guillaume Tabouret
- Université de Toulouse, INP-ENVT, UMR 1225, IHAP, Toulouse, France
- INRA, UMR1225, IHAP, Toulouse, France
| | - Gilles Foucras
- Université de Toulouse, INP-ENVT, UMR 1225, IHAP, Toulouse, France
- INRA, UMR1225, IHAP, Toulouse, France
- * E-mail:
| |
Collapse
|
37
|
Da Costa TB, De Morais NG, De Lira JMB, De Almeida TM, Gonçalves-De-Albuquerque SDC, Pereira VRA, De Paiva Cavalcanti M, De Castro CMMB. Relation between neonatal malnutrition and gene expression: inflammasome function in infections caused by Candida Albicans. Eur J Nutr 2015; 56:693-704. [PMID: 26658898 DOI: 10.1007/s00394-015-1113-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2015] [Accepted: 11/22/2015] [Indexed: 10/22/2022]
Abstract
PURPOSE To investigate the effects of neonatal malnutrition followed by nutritional replacement on the signaling mechanisms developed by the inflammasome complex by analyzing the expression of the targeted TLR2, TLR4, NLRP3, caspase-1 and release of IL-1β and IL-18 by alveolar macrophages infected in vitro with Candida albicans. METHODS Male Wistar rats (n = 24), 90-120 days, were suckled by mothers whose diet during lactation contained 17 % protein in the nourish group and 8 % protein in the malnourished group. After weaning, both groups were fed a normal protein diet. Macrophages were obtained after tracheostomy, through the collection of bronchoalveolar lavage fluid. The quantification of the expression levels of targets (TLR2, TLR4, NLRP3 and caspase-1) was performed by real-time RT-PCR. Production of cytokines was performed by ELISA. RESULTS The malnourished animals during lactation showed reduced body weight from the fifth day of life, remaining until adulthood. Further, the model applied malnutrition induced a lower expression of TLR4 and caspase-1. The quantification of the TLR2 and NLRP3, as well as the release of IL-1β and IL-18, was not different between groups of animals nourished and malnourished. The system challenged with Candida albicans showed high expression levels of all targets in the study. CONCLUSIONS The tests demonstrate nutritional restriction during critical periods of development, although nutritional supplementation may compromise defense patterns in adulthood in a timely manner, preserving distinct signaling mechanism, so that the individual does not become widely vulnerable to infections by opportunistic pathogens.
Collapse
Affiliation(s)
- Thacianna Barreto Da Costa
- Department of Tropical Medicine, Federal University of Pernambuco, Recife, Brazil. .,Keizo Asami Laboratory of Immunopathology, Federal University of Pernambuco, Av. Prof. Moraes Rego, 1235, Cidade Universitária, Recife, Pernambuco, 50670-901, Brazil.
| | - Natália Gomes De Morais
- Department of Tropical Medicine, Federal University of Pernambuco, Recife, Brazil.,Keizo Asami Laboratory of Immunopathology, Federal University of Pernambuco, Av. Prof. Moraes Rego, 1235, Cidade Universitária, Recife, Pernambuco, 50670-901, Brazil
| | - Joana Maria Bezerra De Lira
- Keizo Asami Laboratory of Immunopathology, Federal University of Pernambuco, Av. Prof. Moraes Rego, 1235, Cidade Universitária, Recife, Pernambuco, 50670-901, Brazil
| | - Thays Miranda De Almeida
- Department of Immunology, Aggeu Magalhães Research Center, Oswaldo Cruz Foundation, Campus Federal University of Pernambuco, Recife, Brazil
| | | | - Valéria Rêgo Alves Pereira
- Department of Immunology, Aggeu Magalhães Research Center, Oswaldo Cruz Foundation, Campus Federal University of Pernambuco, Recife, Brazil
| | - Milena De Paiva Cavalcanti
- Department of Immunology, Aggeu Magalhães Research Center, Oswaldo Cruz Foundation, Campus Federal University of Pernambuco, Recife, Brazil
| | - Célia Maria Machado Barbosa De Castro
- Department of Tropical Medicine, Federal University of Pernambuco, Recife, Brazil.,Keizo Asami Laboratory of Immunopathology, Federal University of Pernambuco, Av. Prof. Moraes Rego, 1235, Cidade Universitária, Recife, Pernambuco, 50670-901, Brazil
| |
Collapse
|
38
|
Herbst S, Shah A, Mazon Moya M, Marzola V, Jensen B, Reed A, Birrell MA, Saijo S, Mostowy S, Shaunak S, Armstrong-James D. Phagocytosis-dependent activation of a TLR9-BTK-calcineurin-NFAT pathway co-ordinates innate immunity to Aspergillus fumigatus. EMBO Mol Med 2015; 7:240-58. [PMID: 25637383 PMCID: PMC4364943 DOI: 10.15252/emmm.201404556] [Citation(s) in RCA: 139] [Impact Index Per Article: 15.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
Transplant recipients on calcineurin inhibitors are at high risk of invasive fungal infection. Understanding how calcineurin inhibitors impair fungal immunity is a key priority for defining risk of infection. Here, we show that the calcineurin inhibitor tacrolimus impairs clearance of the major mould pathogen Aspergillus fumigatus from the airway, by inhibiting macrophage inflammatory responses. This leads to defective early neutrophil recruitment and fungal clearance. We confirm these findings in zebrafish, showing an evolutionarily conserved role for calcineurin signalling in neutrophil recruitment during inflammation. We find that calcineurin–NFAT activation is phagocytosis dependent and collaborates with NF-κB for TNF-α production. For yeast zymosan particles, activation of macrophage calcineurin–NFAT occurs via the phagocytic Dectin-1–spleen tyrosine kinase pathway, but for A. fumigatus, activation occurs via a phagosomal TLR9-dependent and Bruton's tyrosine kinase-dependent signalling pathway that is independent of MyD88. We confirm the collaboration between NFAT and NF-κB for TNF-α production in primary alveolar macrophages. These observations identify inhibition of a newly discovered macrophage TLR9–BTK–calcineurin–NFAT signalling pathway as a key immune defect that leads to organ transplant-related invasive aspergillosis.
Collapse
Affiliation(s)
- Susanne Herbst
- Department of Infectious Diseases and Immunity, Imperial College London, London, UK
| | - Anand Shah
- Department of Infectious Diseases and Immunity, Imperial College London, London, UK
| | - Maria Mazon Moya
- MRC Centre for Molecular Bacteriology and Infection, Imperial College London, London, UK
| | - Vanessa Marzola
- Department of Infectious Diseases and Immunity, Imperial College London, London, UK
| | - Barbara Jensen
- Department of Infectious Diseases and Immunity, Imperial College London, London, UK
| | - Anna Reed
- Lung Transplant Unit, Royal Brompton and Harefield Hospital, London, UK
| | - Mark A Birrell
- National Heart and Lung Institute, Imperial College London, London, UK
| | - Shinobu Saijo
- Medical Mycology Research Centre, Chiba University, Chiba, Japan
| | - Serge Mostowy
- MRC Centre for Molecular Bacteriology and Infection, Imperial College London, London, UK
| | - Sunil Shaunak
- Department of Infectious Diseases and Immunity, Imperial College London, London, UK
| | - Darius Armstrong-James
- Department of Infectious Diseases and Immunity, Imperial College London, London, UK National Heart and Lung Institute, Imperial College London, London, UK
| |
Collapse
|
39
|
Bryant CE, Orr S, Ferguson B, Symmons MF, Boyle JP, Monie TP. International Union of Basic and Clinical Pharmacology. XCVI. Pattern recognition receptors in health and disease. Pharmacol Rev 2015; 67:462-504. [PMID: 25829385 DOI: 10.1124/pr.114.009928] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Since the discovery of Toll, in the fruit fly Drosophila melanogaster, as the first described pattern recognition receptor (PRR) in 1996, many families of these receptors have been discovered and characterized. PRRs play critically important roles in pathogen recognition to initiate innate immune responses that ultimately link to the generation of adaptive immunity. Activation of PRRs leads to the induction of immune and inflammatory genes, including proinflammatory cytokines and chemokines. It is increasingly clear that many PRRs are linked to a range of inflammatory, infectious, immune, and chronic degenerative diseases. Several drugs to modulate PRR activity are already in clinical trials and many more are likely to appear in the near future. Here, we review the different families of mammalian PRRs, the ligands they recognize, the mechanisms of activation, their role in disease, and the potential of targeting these proteins to develop the anti-inflammatory therapeutics of the future.
Collapse
Affiliation(s)
- Clare E Bryant
- Departments of Veterinary Medicine (C.E.B., J.P.B., T.P.M.), Pathology (B.F.), and Biochemistry (M.F.S., J.P.B.), University of Cambridge, Cambridge, United Kingdom; and Institute of Infection and Immunity, Cardiff University, Cardiff, United Kingdom (S.O.)
| | - Selinda Orr
- Departments of Veterinary Medicine (C.E.B., J.P.B., T.P.M.), Pathology (B.F.), and Biochemistry (M.F.S., J.P.B.), University of Cambridge, Cambridge, United Kingdom; and Institute of Infection and Immunity, Cardiff University, Cardiff, United Kingdom (S.O.)
| | - Brian Ferguson
- Departments of Veterinary Medicine (C.E.B., J.P.B., T.P.M.), Pathology (B.F.), and Biochemistry (M.F.S., J.P.B.), University of Cambridge, Cambridge, United Kingdom; and Institute of Infection and Immunity, Cardiff University, Cardiff, United Kingdom (S.O.)
| | - Martyn F Symmons
- Departments of Veterinary Medicine (C.E.B., J.P.B., T.P.M.), Pathology (B.F.), and Biochemistry (M.F.S., J.P.B.), University of Cambridge, Cambridge, United Kingdom; and Institute of Infection and Immunity, Cardiff University, Cardiff, United Kingdom (S.O.)
| | - Joseph P Boyle
- Departments of Veterinary Medicine (C.E.B., J.P.B., T.P.M.), Pathology (B.F.), and Biochemistry (M.F.S., J.P.B.), University of Cambridge, Cambridge, United Kingdom; and Institute of Infection and Immunity, Cardiff University, Cardiff, United Kingdom (S.O.)
| | - Tom P Monie
- Departments of Veterinary Medicine (C.E.B., J.P.B., T.P.M.), Pathology (B.F.), and Biochemistry (M.F.S., J.P.B.), University of Cambridge, Cambridge, United Kingdom; and Institute of Infection and Immunity, Cardiff University, Cardiff, United Kingdom (S.O.)
| |
Collapse
|
40
|
Yan H, Kamiya T, Suabjakyong P, Tsuji NM. Targeting C-Type Lectin Receptors for Cancer Immunity. Front Immunol 2015; 6:408. [PMID: 26379663 PMCID: PMC4547497 DOI: 10.3389/fimmu.2015.00408] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2015] [Accepted: 07/26/2015] [Indexed: 12/21/2022] Open
Abstract
C-type lectin receptors (CLRs) are a large family of soluble and trans-membrane pattern recognition receptors that are widely and primarily expressed on myeloid cells. CLRs are important for cell-cell communication and host defense against pathogens through the recognition of specific carbohydrate structures. Similar to a family of Toll-like receptors, CLRs signaling are involved in the various steps for initiation of innate immune responses and promote secretion of soluble factors such as cytokines and interferons. Moreover, CLRs contribute to endocytosis and antigen presentation, thereby fine-tune adaptive immune responses. In addition, there may also be a direct activation of acquired immunity. On the other hand, glycans, such as mannose structures, Lewis-type antigens, or GalNAc are components of tumor antigens and ligate CLRs, leading to immunoregulation. Therefore, agonists or antagonists of CLRs signaling are potential therapeutic reagents for cancer immunotherapy. We aim to overview the current knowledge of CLRs signaling and the application of their ligands on tumor-associating immune response.
Collapse
Affiliation(s)
- Huimin Yan
- Immune Homeostasis Laboratory, Biomedical Research Institute, National Institute for Advanced Industrial Science and Technology (AIST) , Tsukuba , Japan ; Institute for Liver Disease, Fifth Hospital of Shijiazhuang , Shijiazhuang , China
| | - Tomomori Kamiya
- Immune Homeostasis Laboratory, Biomedical Research Institute, National Institute for Advanced Industrial Science and Technology (AIST) , Tsukuba , Japan ; Research Institute for Biomedical Sciences, Tokyo University of Science , Noda-shi , Japan
| | - Papawee Suabjakyong
- Immune Homeostasis Laboratory, Biomedical Research Institute, National Institute for Advanced Industrial Science and Technology (AIST) , Tsukuba , Japan ; Department of Clinical and Analytical Biochemistry, Graduate School of Pharmaceutical Sciences, Chiba University , Chiba-shi , Japan
| | - Noriko M Tsuji
- Immune Homeostasis Laboratory, Biomedical Research Institute, National Institute for Advanced Industrial Science and Technology (AIST) , Tsukuba , Japan
| |
Collapse
|
41
|
Masuda Y, Nawa D, Nakayama Y, Konishi M, Nanba H. Soluble β-glucan from Grifola frondosa induces tumor regression in synergy with TLR9 agonist via dendritic cell-mediated immunity. J Leukoc Biol 2015; 98:1015-25. [PMID: 26297795 DOI: 10.1189/jlb.1a0814-415rr] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2014] [Accepted: 07/23/2015] [Indexed: 02/04/2023] Open
Abstract
The maturation of dendritic cells into more-immunostimulatory dendritic cells by stimulation with different combinations of immunologic agents is expected to provide efficient, adoptive immunotherapy against cancer. Soluble β-glucan maitake D-fraction, extracted from the maitake mushroom Grifola frondosa, acts as a potent immunotherapeutic agent, eliciting innate and adoptive immune responses, thereby contributing to its antitumor activity. Here, we evaluated the efficacy of maitake D-fraction, in combination with a Toll-like receptor agonist, to treat tumors in a murine model. Our results showed that maitake D-fraction, in combination with the Toll-like receptor 9 agonist, cytosine-phosphate-guanine oligodeoxynucleotide, synergistically increased the expression of dendritic cell maturation markers and interleukin-12 production in dendritic cells, but it did not increase interleukin-10 production, generating strong effector dendritic cells with an augmented capacity for efficiently priming an antigen-specific, T helper 1-type T cell response. Maitake D-fraction enhances cytosine-phosphate-guanine oligodeoxynucleotide-induced dendritic cell maturation and cytokine responses in a dectin-1-dependent pathway. We further showed that a combination therapy using cytosine-phosphate-guanine oligodeoxynucleotide and maitake D-fraction was highly effective, either as adjuvants for dendritic cell vaccination or by direct administration against murine tumor. Therapeutic responses to direct administration were associated with increased CD11c(+) dendritic cells in the tumor site and the induction of interferon-γ-producing CD4(+) and CD8(+) T cells. Our results indicate that maitake D-fraction and cytosine-phosphate-guanine oligodeoxynucleotide synergistically activated dendritic cells, resulting in tumor regression via an antitumor T helper cell 1-type response. Our findings provide the basis for a potent antitumor therapy using a novel combination of immunologic agents for future clinical immunotherapy studies in patients.
Collapse
Affiliation(s)
- Yuki Masuda
- Department of Microbial Chemistry, Kobe Pharmaceutical University, Motoyama-kitamachi, Higashinada-ku, Kobe 658-8558, Japan
| | - Daiki Nawa
- Department of Microbial Chemistry, Kobe Pharmaceutical University, Motoyama-kitamachi, Higashinada-ku, Kobe 658-8558, Japan
| | - Yoshiaki Nakayama
- Department of Microbial Chemistry, Kobe Pharmaceutical University, Motoyama-kitamachi, Higashinada-ku, Kobe 658-8558, Japan
| | - Morichika Konishi
- Department of Microbial Chemistry, Kobe Pharmaceutical University, Motoyama-kitamachi, Higashinada-ku, Kobe 658-8558, Japan
| | - Hiroaki Nanba
- Department of Microbial Chemistry, Kobe Pharmaceutical University, Motoyama-kitamachi, Higashinada-ku, Kobe 658-8558, Japan
| |
Collapse
|
42
|
Lemoine S, Jaron B, Tabka S, Ettreiki C, Deriaud E, Zhivaki D, Le Ray C, Launay O, Majlessi L, Tissieres P, Leclerc C, Lo-Man R. Dectin-1 activation unlocks IL12A expression and reveals the TH1 potency of neonatal dendritic cells. J Allergy Clin Immunol 2015; 136:1355-68.e1-15. [PMID: 25865351 DOI: 10.1016/j.jaci.2015.02.030] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2014] [Revised: 02/04/2015] [Accepted: 02/13/2015] [Indexed: 01/17/2023]
Abstract
BACKGROUND Early life is characterized by a high susceptibility to infection and a TH2-biased CD4 T-cell response to vaccines. Toll-like receptor (TLR) agonists are currently being implemented as new vaccine adjuvants for TH1 activation, but their translation to the field of pediatric vaccines is facing the impairment of neonatal innate TLR responses. OBJECTIVE We sought to analyze C-type lectin receptor pathways as an alternative or a coactivator to TLRs for neonatal dendritic cell activation for TH1 polarization. METHODS Neonatal monocyte-derived dendritic cells (moDCs) were exposed to various combinations of TLR agonists with or without Dectin-1 agonist. IL-12 and IL-23 responses were analyzed at the transcriptional and protein levels after stimulation. The intracellular pathways triggered by combined TLR plus Dectin-1 stimulation was determined by using pharmacologic inhibitors. The capacity of neonatal moDCs to differentiate naive CD4 TH cells was evaluated in cocultures with heterologous neonatal naive T cells. Curdlan was finally tested as an adjuvant within a subunit tuberculosis vaccine in neonatal mice. RESULTS Simultaneous coactivation through Dectin-1 and TLRs induced robust secretion of IL-12p70 by neonatal moDCs by unlocking transcriptional control on the p35 subunit of IL-12. Both the spleen tyrosine kinase and Raf-1 pathways were involved in this process, allowing differentiation of neonatal naive T cells toward IFN-γ-producing TH1 cells. In vivo a Dectin-1 agonist as adjuvant was sufficient to induce TH1 responses after vaccination of neonatal mice. CONCLUSION Coactivation of neonatal moDCs through Dectin-1 allows TLR-mediated IL-12p70 secretion and TH1 polarization of neonatal T cells. Dectin-1 agonists represent a promising TH1 adjuvant for pediatric vaccination.
Collapse
Affiliation(s)
- Sébastien Lemoine
- Unité de Régulation Immunitaire et Vaccinologie, Institut Pasteur, Paris, France; INSERM U1041, Paris, France
| | - Barbara Jaron
- Unité de Régulation Immunitaire et Vaccinologie, Institut Pasteur, Paris, France; INSERM U1041, Paris, France
| | - Sabrine Tabka
- Unité de Régulation Immunitaire et Vaccinologie, Institut Pasteur, Paris, France; INSERM U1041, Paris, France
| | - Chourouk Ettreiki
- Unité de Réanimation Pédiatrique et Médecine Néonatale, AP-HP, Hôpitaux Universitaires Paris-Sud, Le Kremlin Bicêtre, France; Groupe Equipe Endotoxine, Structures et Activité, Institut de Génétique et Microbiologie, UMR 8621, Université Paris Sud, Orsay, France
| | - Edith Deriaud
- Unité de Régulation Immunitaire et Vaccinologie, Institut Pasteur, Paris, France; INSERM U1041, Paris, France
| | - Dania Zhivaki
- Unité de Régulation Immunitaire et Vaccinologie, Institut Pasteur, Paris, France; INSERM U1041, Paris, France; Université Paris Diderot, Paris, France
| | - Camille Le Ray
- APHP, Department of Obstetrics and Gynecology, Maternité Port Royal, Paris, France; Université Paris Descartes, Faculté de Médecine, Paris, France
| | - Odile Launay
- Université Paris Descartes, Faculté de Médecine, Paris, France; INSERM CIC1417, Paris, France
| | - Laleh Majlessi
- Unité de Pathogénomique Mycobactérienne Intégrée, Institut Pasteur, Paris, France
| | - Pierre Tissieres
- Unité de Réanimation Pédiatrique et Médecine Néonatale, AP-HP, Hôpitaux Universitaires Paris-Sud, Le Kremlin Bicêtre, France; Groupe Equipe Endotoxine, Structures et Activité, Institut de Génétique et Microbiologie, UMR 8621, Université Paris Sud, Orsay, France; Faculté de Médecine, Université Paris Sud, Le Kremlin Bicêtre, France
| | - Claude Leclerc
- Unité de Régulation Immunitaire et Vaccinologie, Institut Pasteur, Paris, France; INSERM U1041, Paris, France
| | - Richard Lo-Man
- Unité de Régulation Immunitaire et Vaccinologie, Institut Pasteur, Paris, France; INSERM U1041, Paris, France.
| |
Collapse
|
43
|
Loures FV, Araújo EF, Feriotti C, Bazan SB, Calich VLG. TLR-4 cooperates with Dectin-1 and mannose receptor to expand Th17 and Tc17 cells induced by Paracoccidioides brasiliensis stimulated dendritic cells. Front Microbiol 2015; 6:261. [PMID: 25873917 PMCID: PMC4379960 DOI: 10.3389/fmicb.2015.00261] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2014] [Accepted: 03/16/2015] [Indexed: 01/21/2023] Open
Abstract
The concomitant use of diverse pattern recognition receptors (PRRs) by innate immune cells can result in synergistic or inhibitory activities that profoundly influence anti-microbial immunity. Dectin-1 and the mannose receptor (MR) are C-type lectin receptors (CLRs) previously reported to cooperate with toll-like receptors (TLRs) signaling in the initial inflammatory response and in the induction of adaptive Th17 and Tc17 immunity mediated by CD4(+) and CD8(+) T cells, respectively. The protective immunity against paracoccidioidomycosis, the most prevalent fungal infection of Latin America, was previously shown to be influenced by these T cell subsets motivating us to study the contribution of TLRs, Dectin-1, and MR to the development of Th17/Tc17 immunity. First, curdlan a specific Dectin-1 agonist was used to characterize the influence of this receptor in the proliferative response and Th17/Tc17 differentiation of naïve lymphocytes induced by Paracoccidioides brasiliensis activated dendritic cells (DCs) from C57BL/6 mice. Then, wild type (WT), Dectin-1(-/-), TLR-2(-/-), and TLR-4(-/-) DCs treated or untreated with anti-Dectin-1 and anti-MR antibodies were used to investigate the contribution of these receptors in lymphocyte activation and differentiation. We verified that curdlan induces an enhanced lymphocyte proliferation and development of IL-17 producing CD4(+) and CD8(+) T cells. In addition, treatment of WT, TLR-2(-/-), and TLR-4(-/-) DCs by anti-Dectin-1 antibodies or antigen presentation by Dectin-1(-/-) DCs led to decreased lymphoproliferation and impaired Th17 and Tc17 expansion. These responses were also inhibited by anti-MR treatment of DCs, but a synergistic action on Th17/Tc17 differentiation was mediated by TLR-4 and MR. Taken together, our results indicate that diverse TLRs and CLRs are involved in the induction of lymphocyte proliferation and Th17/Tc17 differentiation mediated by P. brasiliensis activated DCs, but a synergist action was restricted to Dectin-1, TLR-4, and MR.
Collapse
Affiliation(s)
- Flávio V Loures
- Departamento de Imunologia, Instituto de Ciências Biomédicas, Universidade de São Paulo , São Paulo, Brazil
| | - Eliseu F Araújo
- Departamento de Imunologia, Instituto de Ciências Biomédicas, Universidade de São Paulo , São Paulo, Brazil
| | - Claudia Feriotti
- Departamento de Imunologia, Instituto de Ciências Biomédicas, Universidade de São Paulo , São Paulo, Brazil
| | - Silvia B Bazan
- Departamento de Imunologia, Instituto de Ciências Biomédicas, Universidade de São Paulo , São Paulo, Brazil
| | - Vera L G Calich
- Departamento de Imunologia, Instituto de Ciências Biomédicas, Universidade de São Paulo , São Paulo, Brazil
| |
Collapse
|
44
|
Ding J, Feng T, Ning Y, Li W, Wu Q, Qian K, Wang Y, Qi C. β-Glucan enhances cytotoxic T lymphocyte responses by activation of human monocyte-derived dendritic cells via the PI3K/AKT pathway. Hum Immunol 2015; 76:146-54. [DOI: 10.1016/j.humimm.2015.01.009] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2014] [Revised: 10/10/2014] [Accepted: 01/14/2015] [Indexed: 12/11/2022]
|
45
|
Paulovičová E, Bujdáková H, Chupáčová J, Paulovičová L, Kertys P, Hrubiško M. Humoral immune responses to Candida albicans complement receptor 3-related protein in the atopic subjects with vulvovaginal candidiasis. Novel sensitive marker for Candida infection. FEMS Yeast Res 2015; 15:fou001. [DOI: 10.1093/femsyr/fou001] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
|
46
|
Baram L, Cohen-Kedar S, Spektor L, Elad H, Guzner-Gur H, Dotan I. Differential stimulation of peripheral blood mononuclear cells in Crohn's disease by fungal glycans. J Gastroenterol Hepatol 2014; 29:1976-84. [PMID: 25092526 DOI: 10.1111/jgh.12701] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 04/20/2014] [Indexed: 12/12/2022]
Abstract
BACKGROUND AND AIM Crohn's disease (CD) is characterized by loss of tolerance to intestinal microorganisms. This is reflected by serological responses to fungal glycans such as mannan and β-glucans. Fungal glycans have various effects on immune cells. However, the evidence for their effects in CD is vague. This study aimed to assess the effects of fungal cell wall glycans on human peripheral blood mononuclear cells (PBMCs) from CD and control patients. METHODS Human PBMCs from CD and control patients were stimulated by fungal cell wall glycans. Cytokine secretion was detected by ELISA and glycan receptor expression by flow cytometry. RESULTS Mannan, β-glucans (curdlan), chitosan, and zymosan induced the secretion of interleukin (IL)-1β, IL-6, IL-23, IL-10, and tumor necrosis factor-α by PBMCs. Spleen tyrosin kinase and Src tyrosine kinase were involved in the response to mannan and β-glucans. Mannan and whole yeast cells induced a significantly higher pro-inflammatory cytokine response in CD compared with control patients. CONCLUSIONS The results may suggest that CD is characterized by hyperresponsiveness to fungal glycans. Thus, glycans may potentially be triggering or perpetuating inflammation.
Collapse
Affiliation(s)
- Liran Baram
- IBD Center, Department of Gastroenterology and Liver Diseases and The Research Center for Digestive Tract and Liver Diseases, Tel Aviv Medical Center, Tel Aviv, Israel; Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | | | | | | | | | | |
Collapse
|
47
|
Rizzetto L, De Filippo C, Cavalieri D. Richness and diversity of mammalian fungal communities shape innate and adaptive immunity in health and disease. Eur J Immunol 2014; 44:3166-81. [DOI: 10.1002/eji.201344403] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2014] [Revised: 09/22/2014] [Accepted: 09/23/2014] [Indexed: 02/06/2023]
Affiliation(s)
- Lisa Rizzetto
- Research and Innovation Centre; Fondazione Edmund Mach; San Michele all'Adige TN Italy
| | - Carlotta De Filippo
- Research and Innovation Centre; Fondazione Edmund Mach; San Michele all'Adige TN Italy
| | - Duccio Cavalieri
- Research and Innovation Centre; Fondazione Edmund Mach; San Michele all'Adige TN Italy
| |
Collapse
|
48
|
Donnarumma G, Molinaro A, Cimini D, De Castro C, Valli V, De Gregorio V, De Rosa M, Schiraldi C. Lactobacillus crispatus L1: high cell density cultivation and exopolysaccharide structure characterization to highlight potentially beneficial effects against vaginal pathogens. BMC Microbiol 2014; 14:137. [PMID: 24884965 PMCID: PMC4054921 DOI: 10.1186/1471-2180-14-137] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2014] [Accepted: 05/21/2014] [Indexed: 01/23/2023] Open
Abstract
BACKGROUND Vaginal lactic acid bacteria defend the host against pathogens through a combination of competitive exclusion, competition for nutrients, production of antimicrobial substances and through the activation of the immune system. A new human isolate named Lactobacillus crispatus L1 was characterized in this work, and a preliminary evaluation of its probiotic potential is described together with a process to obtain a high productivity of viable biomass. RESULTS In a simulated digestion process 1.8⋅10(10) cells∙ml(-1) survived the gastric environment with 80% viability, without being affected by small intestine juices. Experiments on six different C sources were performed to analyze growth and organic acids production and, glucose, provided the best performances. A microfiltration strategy was exploited to improve the cellular yield in 2 L-fermentation processes, reaching 27 g · l(-1) of dry biomass. Moreover, L. crispatus L1 demonstrated a greater stability to high concentrations of lactic acid, compared to other lactobacilli. The specific L. crispatus L1 exopolysaccharide was purified from the fermentation broth and characterized by NMR showing structural features and similarity to exopolysaccharides produced by pathogenic strains. Live L. crispatus L1 cells strongly reduced adhesion of a yeast pathogenic strain, Candida albicans in particular, in adherence assays. Interestingly a higher expression of the human defensin HBD-2 was also observed in vaginal cells treated with the purified exopolysaccharide, indicating a possible correlation with C. albicans growth inhibition. CONCLUSIONS The paper describes the evaluation of L. crispatus L1 as potential vaginal probiotic and the fermentation processes to obtain high concentrations of viable cells.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Chiara Schiraldi
- Department of Experimental Medicine, Section of Biotechnology and Molecular Biology, Second University of Naples, via De Crecchio n°7, Naples 80138, Italy.
| |
Collapse
|